Research Feeds

View All
1H NMR- based metabolomics approaches as non-invasive tools for diagnosis of endometriosis A Comparative Study of Blood Levels of Manganese, Some Macroelements and Heavy Metals in Obese and Non-Obese Polycystic Ovary Syndrome Patients A Comparative Study of the Gut Microbiota Associated With Immunoglobulin a Nephropathy and Membranous Nephropathy A comparative study of the gut microbiota in immune-mediated inflammatory diseases-does a common dysbiosis exist? A comprehensive analysis of breast cancer microbiota and host gene expression A comprehensive analysis of breast cancer microbiota and host gene expression A cross-sectional analysis about bacterial vaginosis, high-risk human papillomavirus infection, and cervical intraepithelial neoplasia in Chinese women A cross-sectional pilot study of birth mode and vaginal microbiota in reproductive-age women A metabonomics approach as a means for identification of potentialbiomarkers for early diagnosis of endometriosis A More Diverse Cervical Microbiome Associates with Better Clinical Outcomes in Patients with Endometriosis: A Pilot Study A Multi-Omic Systems-Based Approach Reveals Metabolic Markers of Bacterial Vaginosis and Insight into the Disease A New Approach to Polycystic Ovary Syndrome: The Gut Microbiota A Review of the Anti-inflammatory Properties of Clindamycin in the Treatment of Acne Vulgaris A Systematic Review and Meta-Analysis of Premenstrual Syndrome with Special Emphasis on Herbal Medicine and Nutritional Supplements. Adherence to the Mediterranean Diet, Dietary Patterns and Body Composition in Women with Polycystic Ovary Syndrome (PCOS)

Research Feeds

Our team reviews, analyzes, and summarizes the latest health and nutrition studies to help you be healthier. Updated daily.

Premium Content

We are an entirely self-funded, with no third-party ads or ties to food and supplement companies.

MS and MDD stem from neuroendocrine and neurochemical imbalances, especially altered allopregnanolone and ABA activity. Hormonal and neuroactive therapies improve symptoms, while microbiome and neuroinflammation represent promising research areas.

Category View Feed View

Table of Contents

Women’s Health

201 studies

Endometriosis

54 studies

Metabolomic Signature

3 studies

Breast Cancer

11 studies

Infertility

3 studies

Bacterial Vaginosis

53 studies

Clindamycin

3 studies

Premenstrual Syndrome (PMS)

9 studies

Nickel

4 studies

Premenstrual Dysphoric Disorder (PMDD)

13 studies

Irritable Bowel Syndrome (IBS)

23 studies

Autoimmune Diseases

18 studies

Hashimoto’s Thyroiditis

4 studies

Cardiovascular Health

5 studies

Heart Failure

1 studies

Rheumatoid Arthritis

5 studies

Microbes

24 studies

Metals

34 studies

Essential Oils

2 studies

Brain Health

7 studies

Polycystic ovary syndrome (PCOS)

5 studies

STOPs

3 studies

Anemia

1 studies

Tinidazole

2 studies

Endometriomas

1 studies

Lactoferrin

1 studies

Parkinson’s Disease

2 studies

Alzheimer’s Dementia

1 studies

Metallomic Signatures

1 studies

Low‑Nickel Diet (LNiD)

1 studies

Lipopolysaccharides (LPS)

1 studies

Hyperbaric Oxygen Therapy (HBOT)

1 studies

Autism spectrum disorder (ASD)

1 studies

Multiple Sclerosis (MS)

1 studies

Microbiome Signatures

1 studies

Microbiome-Targeted Interventions (MBTIs)

1 studies

Chronic Pelvic Pain (CPP)

2 studies

Women’s Health

Go to Category Page

1H NMR- based metabolomics approaches as non-invasive tools for diagnosis of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metabolomic Signature
    Metabolomic Signature

    Metabolomic signatures are unique metabolite patterns linked to specific biological conditions, identified through metabolomics. They reveal underlying biochemical activities, aiding in disease diagnosis, biomarker development, and personalized medicine. The microbiome significantly affects these signatures, influencing health and disease outcomes through metabolic interactions.

This study demonstrates the potential of ¹H-NMR metabolomics to diagnose endometriosis non-invasively by identifying metabolic biomarkers and disrupted pathways. Quadratic Discriminant Analysis outperformed Artificial Neural Networks in diagnostic accuracy.

What was studied?

This study investigated the application of metabolomics, specifically through proton nuclear magnetic resonance (¹H-NMR) spectroscopy, to identify non-invasive biomarkers for diagnosing endometriosis. The researchers developed computational models using Quadratic Discriminant Analysis (QDA) and Artificial Neural Networks (ANNs) to analyze metabolic changes in serum samples and assess their utility in early diagnosis of the disease.

Who was studied?

The study analyzed serum samples from 31 infertile women diagnosed with stage II or III endometriosis confirmed via laparoscopy and 15 healthy women without any signs of endometriosis. The participants were aged 22–44 years and were recruited from an infertility center in Iran. Exclusion criteria included recent medical or hormonal treatments, prior gynecological surgeries, or other pelvic inflammatory conditions.

What were the most important findings?

The study revealed significant metabolic differences between women with endometriosis and healthy controls. Key findings included elevated levels of 2-methoxyestrone, 2-methoxyestradiol, androstenedione, aldosterone, dehydroepiandrosterone, and deoxycorticosterone in the endometriosis group, alongside decreased cholesterol and primary bile acids. These metabolic changes are linked to disruptions in steroid hormone biosynthesis and bile acid metabolism, indicating underlying hyperestrogenism and impaired hepatic estrogen clearance. The QDA model achieved a correct classification rate of 76%, with 71% positive predictive value and 78% negative predictive value, outperforming the ANN model, which had lower sensitivity and specificity. Metabolic pathway analyses highlighted altered steroid hormone and bile acid biosynthesis, which are critical in the pathophysiology of endometriosis.

What are the greatest implications of this study?

This study underscores the potential of ¹H-NMR-based metabolomics as a minimally invasive diagnostic tool for endometriosis, reducing reliance on invasive laparoscopy. The identification of specific biomarkers and disrupted pathways could facilitate earlier diagnosis, improved patient stratification, and targeted therapeutic interventions. The findings also demonstrate the utility of computational modeling, particularly QDA, in translating complex metabolomics data into clinically actionable insights. This approach represents a significant advancement in bridging diagnostic gaps for endometriosis.

A Comparative Study of Blood Levels of Manganese, Some Macroelements and Heavy Metals in Obese and Non-Obese Polycystic Ovary Syndrome Patients

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study shows women with PCOS have elevated blood lead and cadmium and reduced magnesium, calcium, and manganese—disrupting hormone levels through oxidative stress and possibly altering gut microbiota.

What was studied?

This study examined the blood levels of trace elements and heavy metals—specifically manganese (Mn), magnesium (Mg), calcium (Ca), cadmium (Cd), and lead (Pb)—in women diagnosed with polycystic ovary syndrome (PCOS), with a comparison between obese and non-obese subgroups. The study also explored correlations between these elements and sex hormones (FSH, LH, testosterone, TSH, prolactin) to assess whether these minerals and toxic metals could play a pathophysiological role in PCOS. The study aimed to evaluate how both beneficial and harmful mineral profiles interact with hormonal dysregulation in PCOS, particularly through oxidative stress pathways.

Who was studied?

The study included 82 Iraqi women divided into four groups: 27 obese women with PCOS (BMI ≥ 30), 27 non-obese women with PCOS (BMI < 30), 14 obese healthy controls, and 14 non-obese healthy controls. Diagnosis of PCOS followed the revised Rotterdam criteria, requiring two of three features—hyperandrogenism, oligo-- or anovulation, and polycystic ovaries—while excluding other endocrine disorders. Blood and serum samples were collected for mineral and hormonal profiling using atomic absorption spectrophotometry and immunoassays.

What were the most important findings?

Both obese and non-obese women with PCOS had significantly higher blood concentrations of lead and cadmium and significantly lower serum concentrations of manganese, magnesium, and calcium compared to their respective control groups. Notably, no significant differences in trace element levels were found between obese and non-obese PCOS groups, indicating that mineral and heavy metal imbalances are intrinsic features of PCOS regardless of BMI.

Correlations between trace elements and hormones provided mechanistic insights. In non-obese PCOS women, blood lead levels positively correlated with serum TSH, suggesting a thyroid-disrupting effect of lead. Cadmium levels positively correlated with total testosterone in obese PCOS women, implying a potential role in hyperandrogenism. Additionally, magnesium levels were inversely correlated with LH in non-obese PCOS women, highlighting magnesium’s regulatory role in gonadotropin release.

These elemental imbalances are tightly linked to oxidative stress, a central driver of PCOS pathogenesis. Lead and cadmium generate reactive oxygen species (ROS), impair mitochondrial function, and deplete antioxidants like glutathione. Manganese deficiency disrupts mitochondrial superoxide dismutase (MnSOD) activity, while low magnesium intensifies ROS generation and impairs glucose metabolism, exacerbating insulin resistance. From a microbiome perspective, excess lead and cadmium may enrich inflammatory genera like Proteobacteria, while deficiencies in magnesium and manganese reduce populations of SCFA-producing species like Faecalibacterium prausnitzii and Roseburia, contributing to systemic inflammation and endocrine disruption.

What are the greatest implications of this study?

This study emphasizes that disruptions in essential and toxic trace elements are fundamental to the hormonal and metabolic disturbances observed in PCOS. Elevated levels of lead and cadmium, coupled with deficiencies in manganese, magnesium, and calcium, suggest a common oxidative and endocrine-disrupting profile in PCOS patients, independent of obesity status. Clinically, these findings support the integration of trace element and heavy metal screening into PCOS evaluation and management. Therapeutic strategies that focus on detoxification (reducing cadmium and lead burden) and repletion of deficient minerals may restore oxidative balance and improve hormonal regulation. Moreover, these mineral imbalances may be influencing gut microbiota composition, suggesting an underexplored link between environmental exposure, trace element status, microbial dysbiosis, and PCOS. Future research should prioritize longitudinal and interventional studies to assess whether correcting these elemental imbalances can modulate the gut–hormone axis and lead to improved reproductive and metabolic outcomes in PCOS.

A comprehensive analysis of breast cancer microbiota and host gene expression

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

The study analyzed breast tumor and adjacent tissues, linking microbiota composition to cancer pathways. Key findings implicate specific microbes in breast cancer progression.

What Was Studied?

This study investigated the microbial composition of breast tumor tissues compared to non-cancerous adjacent (NCA) tissues, focusing on identifying specific microbiota associated with different breast cancer subtypes. The research utilized RNA sequencing data from The Cancer Genome Atlas (TCGA), analyzing microbial reads and their association with host gene expression profiles to explore the role of the tumor microbiota in breast cancer pathogenesis.

Who Was Studied?

The study involved 668 breast tumor tissue samples and 72 NCA samples. The samples were filtered to exclude male patients, metastatic cases, and individuals with a history of breast cancer or neoadjuvant therapy, ensuring a robust cohort for microbial and host gene analysis.

What Were the Most Important Findings?

The study identified distinct microbial signatures between tumor and NCA tissues. Proteobacteria were significantly enriched in tumor samples, while Actinobacteria were more prevalent in NCA tissues. Specific microbial taxa, such as Haemophilus influenzae, were associated with genes involved in tumor-promoting pathways, including the G2M checkpoint, E2F transcription factors, and mitotic spindle assembly. Similarly, Listeria fleischmannii correlated with epithelial-to-mesenchymal transition pathways, a hallmark of cancer metastasis.

Twelve of the most abundant species, including Escherichia coli, Mycobacterium fortuitum, and Salmonella enterica, showed significant differential abundance between tumor and NCA tissues. These species are notable for their potential roles in DNA damage and estrogen metabolism, contributing to genomic instability and hormonal dysregulation in breast cancer. The findings also revealed that less prevalent taxa often showed the most significant differential abundance, highlighting the challenges of detecting meaningful microbial shifts in underpowered studies.

What Are the Greatest Implications of This Study?

This research underscores the complex interplay between the tumor microbiota and host gene expression in breast cancer. The enrichment of specific microbial taxa in tumor tissues and their associations with oncogenic pathways suggest that the microbiota may play an active role in breast cancer progression. These findings open avenues for microbiota-targeted interventions and diagnostic tools based on microbial markers. Furthermore, the study highlights the need for large-scale, well-controlled cohorts to accurately characterize the tumor microbiome and its clinical relevance.

A metabonomics approach as a means for identification of potentialbiomarkers for early diagnosis of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metabolomic Signature
    Metabolomic Signature

    Metabolomic signatures are unique metabolite patterns linked to specific biological conditions, identified through metabolomics. They reveal underlying biochemical activities, aiding in disease diagnosis, biomarker development, and personalized medicine. The microbiome significantly affects these signatures, influencing health and disease outcomes through metabolic interactions.

This study used 1H-NMR metabonomics to identify serum biomarkers for early endometriosis diagnosis. Elevated lactate, alanine, and reduced glucose levels highlight metabolic disruptions, offering a minimally invasive diagnostic tool.

What was studied?

This study investigated the identification of predictive biomarkers for early diagnosis of endometriosis using a minimally invasive, serum-based approach. The researchers utilized proton nuclear magnetic resonance (1H-NMR) metabonomics to analyze serum samples, aiming to distinguish endometriosis patients from healthy controls. The study particularly focused on differences in metabolite profiles to identify markers indicative of the condition.

Who was studied?

The study included 45 women aged under 40, divided into two groups. The first group comprised 22 women diagnosed with early-stage endometriosis (Stages I–II) via laparoscopy, while the control group consisted of 23 age- and BMI-matched healthy women with normal menstrual cycles and proven fertility. Participants with recent hormone therapy or irregular menstrual cycles were excluded. Serum samples were collected during the secretory phase of the menstrual cycle.

What were the most important findings?

The study identified several metabolites with significantly altered levels in women with endometriosis compared to controls. Increased levels of lactate, 3-hydroxybutyrate, alanine, leucine, valine, threonine, lysine, glycerophosphatidylcholine, succinic acid, and 2-hydroxybutyrate were observed in the serum of endometriosis patients, while glucose, isoleucine, arginine, and lipid levels were decreased. Multivariate analysis using Partial Least Squares-Discriminant Analysis (PLS-DA) demonstrated strong sensitivity (81.8%) and specificity (91.3%) in distinguishing endometriosis from controls, with an area under the ROC curve of 0.96. Pathway analysis highlighted arginine and proline metabolism disruptions, glycine, serine, and threonine metabolism, pyruvate metabolism, and lysine biosynthesis and degradation. These findings provide a potential non-invasive diagnostic framework and insights into the metabolic disturbances in endometriosis.

What are the greatest implications of this study?

This study offers a promising step toward non-invasive diagnostic methods for endometriosis, reducing reliance on invasive laparoscopy. The identification of metabolite alterations linked to the disease enhances the understanding of its pathophysiology, emphasizing oxidative stress, anaerobic glycolysis, and metabolic reprogramming similar to malignancies. These findings could lead to better clinical tools for early diagnosis and a deeper understanding of the metabolic underpinnings of endometriosis.

A More Diverse Cervical Microbiome Associates with Better Clinical Outcomes in Patients with Endometriosis: A Pilot Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

This study links cervical microbiome diversity with endometriosis severity. Findings reveal microbial imbalances, particularly in advanced stages, correlate with pain, infertility, and inflammatory pathways. The cervical microbiome may serve as a diagnostic and therapeutic target for improving outcomes in endometriosis, highlighting its role in reproductive health and disease progression.

What Was Studied?

This pilot study investigated the cervical microbiome in patients with endometriosis and its association with clinical outcomes. The research focused on the microbial diversity, composition, and functional roles in cervical mucus, analyzed using 16S rRNA sequencing. The study included healthy women and patients diagnosed with endometriosis to compare microbial profiles and explore the connection between microbiome alterations, disease progression, and associated symptoms like pain, CA125 levels, and infertility.

Who Was Studied?

The study involved 33 women: 10 healthy controls and 23 patients diagnosed with endometriosis (classified by severity into stages I-II and III-IV). The cervical microbiome was analyzed to assess its correlation with clinical features, such as deep infiltrating endometriosis (DIE), CA125 biomarker levels, pain severity, and infertility.

What Were the Most Important Findings?

The study revealed that cervical microbiome diversity is significantly associated with clinical outcomes in endometriosis patients. Specifically, a higher microbial diversity was linked to better outcomes, while notable microbial imbalances characterized advanced disease stages and severe symptoms. Patients with advanced stages of endometriosis exhibited a microbial shift, with an increase in Firmicutes and a decrease in Actinobacteria and Bacteroidetes. Unique microbial profiles were observed, such as elevated Lactobacillus jensenii and Streptococcus agalactiae (GBS), alongside reduced Atopobium vaginae in patients with advanced stages.

Patients presenting severe symptoms, including elevated CA125 biomarker levels, infertility, and higher pain scores, showed significantly reduced microbial richness and diversity. Infertility, a common complication of endometriosis, was associated with an increased Firmicutes/Bacteroidetes ratio. Notably, infertility treatments appeared to reverse these imbalances, restoring microbial diversity and community structure to resemble that of fertile individuals. Additionally, deep infiltrating endometriosis (DIE), a severe form of the condition, was correlated with an overrepresentation of Streptococcus and Prevotella at the genus level.

The study’s functional analyses provided insight into the role of the cervical microbiome in disease progression. Pathways associated with microbial alterations, such as signal transduction, secondary bile acid biosynthesis, and nutrient metabolism, were identified. These pathways may contribute to inflammation, immune dysregulation, and potentially malignancy in severe cases. Such findings underscore the intricate relationship between cervical microbial composition and the pathophysiology of endometriosis. This research positions the cervical microbiome as a critical factor in both the diagnosis and management of endometriosis, offering potential for therapeutic interventions targeting microbial imbalances.

What Are the Greatest Implications of This Study?

The findings suggest that cervical microbiome diversity may serve as a biomarker for diagnosing and monitoring endometriosis progression and complications. The research highlights the therapeutic potential of targeting microbial imbalances to improve clinical outcomes, particularly in infertility. It also underscores the potential link between microbiome alterations and malignancy risks in severe cases, paving the way for preventive and precision medicine approaches in endometriosis management.

A New Approach to Polycystic Ovary Syndrome: The Gut Microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review examines the link between PCOS and gut microbiota dysbiosis, highlighting how dietary and microbial interventions may improve clinical outcomes. The study underscores the potential of prebiotics, probiotics, and synbiotics as treatments.

What was studied?

The study explored the relationship between polycystic ovary syndrome (PCOS) and gut microbiota composition, focusing on the role of microbial dysbiosis in contributing to the metabolic and reproductive dysfunctions characteristic of PCOS. The authors reviewed recent research on the gut microbiome's role in PCOS pathogenesis, along with potential therapeutic approaches targeting gut microbiota, such as prebiotics, probiotics, and synbiotics.

Who was studied?

This review does not focus on a specific patient cohort but rather consolidates findings from multiple studies that examine the gut microbiota of women with PCOS. The studies involved women diagnosed with PCOS according to established diagnostic criteria (NIH, Rotterdam, AE-PCOS), with varying phenotypes including obesity, insulin resistance, and hyperandrogenism.

What were the most important findings?

The review highlighted the growing body of evidence linking dysbiosis of the gut microbiota to PCOS. Studies suggest that changes in the composition of the gut microbiome could contribute to the metabolic and reproductive issues observed in PCOS. Specifically, a reduction in microbial diversity and shifts in the relative abundance of beneficial bacteria (e.g., Faecalibacterium prausnitzii and Blautia) were commonly reported. Additionally, gut microbiota dysbiosis in PCOS appears to influence clinical features such as insulin resistance, hyperandrogenism, and inflammation. The authors also explored how gut microbiota modifications through the use of prebiotics, probiotics, and synbiotics may improve clinical outcomes in PCOS, though the mechanisms remain under investigation. Prebiotics and synbiotics showed some promise in improving insulin sensitivity and reducing hyperandrogenism, but further randomized controlled trials are necessary to establish these interventions as standard treatment options.

What are the greatest implications of this study?

This review underscores the importance of understanding the gut microbiome in the context of PCOS, opening avenues for microbiome-based therapies as adjunct treatments for this condition. The findings suggest that managing gut dysbiosis through dietary modifications (e.g., high-fiber diets), probiotics, prebiotics, and synbiotics could help alleviate metabolic disturbances and reproductive issues associated with PCOS. Clinically, these insights could lead to the development of personalized treatment strategies that address the underlying microbial imbalances, providing a more holistic approach to managing PCOS. However, the review calls for further randomized controlled studies to clarify the causality of the relationship between gut microbiota and PCOS and to determine the most effective therapeutic interventions.

A Systematic Review and Meta-Analysis of Premenstrual Syndrome with Special Emphasis on Herbal Medicine and Nutritional Supplements.

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This systematic review and meta-analysis explores the effectiveness of herbal medicine and nutritional supplements in reducing PMS symptoms, highlighting significant reductions in physical, mood, and behavioral symptoms.

What was reviewed?

This article presents a systematic review and meta-analysis focusing on the effectiveness of herbal medicine and nutritional supplements in managing Premenstrual Syndrome (PMS). It reviews randomized controlled trials (RCTs) assessing the impact of various herbal remedies and supplements on both somatic and psycho-behavioral symptoms of PMS. The review highlights the safety, efficacy, and mechanisms of action of these treatments, while also addressing the limitations of current evidence.

Who was reviewed?

The review concentrates on women experiencing PMS, particularly those with both physical and psychological symptoms that significantly affect their daily functioning. It includes trials involving a wide range of herbal and nutritional interventions, aiming to provide clinicians with evidence of alternative therapies for managing PMS symptoms.

What were the most important findings?

The review reveals that herbal medicines and nutritional supplements can significantly reduce the severity of PMS symptoms, including physical, mood, and behavioral issues. It highlights specific interventions such as Vitex agnus castus (chaste tree), Zingiber officinale (ginger), and Crocus sativus (saffron) as particularly effective. The meta-analysis demonstrated a substantial reduction in Premenstrual Symptoms Screening Tool (PSST) scores, indicating notable symptom improvement. Additionally, the review found that certain interventions, such as Ginkgo biloba and vitamin B1, offered improvements in psychological symptoms like anxiety and depression.

Secondary findings also revealed positive effects on mood, physical symptoms, and behavior, with significant reductions in scores for physical symptoms and mood-related symptoms. The study concluded that herbal treatments, particularly when used over multiple cycles, have the potential to alleviate PMS symptoms significantly. However, it emphasized the need for high-quality trials to confirm these results and establish optimal treatment protocols. Importantly, the review also addressed the safety profile of these treatments, with most studies reporting mild adverse effects such as nausea or digestive discomfort.

What are the greatest implications of this review?

The greatest implication of this review is that herbal medicine and nutritional supplements represent a viable, non-pharmacological option for managing PMS symptoms, offering a safer alternative to traditional treatments with fewer side effects. This study provides clinicians with evidence-based recommendations for integrating these alternative therapies into PMS management, especially for patients who prefer natural treatments or experience adverse reactions to conventional medications. The review also underscores the need for more robust, high-quality studies to refine treatment guidelines, improve symptom assessment tools, and explore the mechanisms underlying the therapeutic effects of herbal interventions. Clinicians can consider incorporating these findings into patient care plans while awaiting further evidence from future studies.

Adherence to the Mediterranean Diet, Dietary Patterns and Body Composition in Women with Polycystic Ovary Syndrome (PCOS)

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Women with PCOS had lower Mediterranean diet adherence and worse metabolic, inflammatory, and hormonal profiles. Poor MD adherence correlated with higher testosterone, CRP, and HOMA-IR, suggesting a microbiome-linked dietary influence on PCOS severity.

What was studied?

This cross-sectional, case-controlled study investigated the relationship between adherence to the Mediterranean diet (MD), dietary intake patterns, body composition, and clinical severity of polycystic ovary syndrome (PCOS) in treatment-naïve women. The primary aim was to determine whether the quality of dietary intake, specifically adherence to the MD, was associated with hyperandrogenism, insulin resistance (IR), inflammation, and altered body composition in women with PCOS compared to BMI- and age-matched controls. Using validated methods, including the PREDIMED score and seven-day food records, the study examined the dietary quality and macronutrient composition, while bioelectrical impedance analysis (BIA) was employed to assess body composition, including phase angle (PhA) as a marker of cellular health and inflammation.

Who was studied?

The study involved 112 treatment-naïve women with PCOS and 112 healthy, age- and BMI-matched controls, all recruited from the same geographical region in Italy. Inclusion criteria limited the population to premenopausal women aged 18–40 with BMIs up to 39.9 kg/m². Participants had no other endocrine or metabolic diseases, had not followed any special diet or taken nutritional supplements in the preceding three months, and had not taken medications affecting metabolism. Dietary assessments were conducted via face-to-face interviews using seven-day food records and the PREDIMED questionnaire, while hormonal, inflammatory, and metabolic parameters were evaluated through fasting blood draws.

What were the most important findings?

Women with PCOS exhibited significantly lower adherence to the Mediterranean diet, consuming less extra-virgin olive oil, fish, legumes, and nuts compared to controls, despite similar total caloric intake. Their diets were higher in simple carbohydrates and saturated fatty acids (SFA), and lower in complex carbohydrates, fiber, monounsaturated fatty acids (MUFA), and omega-3 polyunsaturated fatty acids (n-3 PUFA). These nutritional differences were directly associated with higher serum testosterone levels, increased CRP (a marker of inflammation), higher HOMA-IR values, and worse anthropometric profiles, including increased waist circumference. Body composition analysis revealed significantly lower fat-free mass, phase angle (PhA), and intracellular water, and higher fat mass and extracellular water in PCOS patients, indicating poorer cellular health and hydration status.

From a microbiome perspective, the Mediterranean diet’s high content of fiber, polyphenols, and MUFA promotes the growth of beneficial microbes such as Faecalibacterium prausnitzii, Bifidobacterium spp., and Lactobacillus spp., which are known to produce short-chain fatty acids (SCFAs) like butyrate. SCFAs enhance insulin sensitivity, reduce systemic inflammation, and improve gut barrier function. In contrast, diets high in SFA and simple sugars promote dysbiosis and increase pro-inflammatory taxa such as Proteobacteria. This study indirectly supports the role of microbiota in mediating the diet-PCOS relationship through the systemic metabolic and endocrine improvements associated with MD adherence.

What are the implications of this study?

This study underscores the central role of dietary quality, particularly adherence to the Mediterranean diet, in modulating the severity of PCOS. It provides strong evidence that beyond caloric intake, the types of fat and carbohydrates consumed significantly impact inflammation, insulin sensitivity, and androgen levels. The findings establish a direct link between poor MD adherence and more severe hyperandrogenemia and inflammatory status. Clinically, this reinforces the necessity of nutritional counseling as a first-line intervention in PCOS management. The Mediterranean diet offers a microbiome-friendly strategy, rich in fiber and MUFA, capable of reducing systemic inflammation and potentially improving microbiota composition. Furthermore, phase angle (PhA) emerges as a promising biomarker of PCOS severity and nutritional status. For clinicians and researchers, these findings support incorporating nutritional pattern assessments and body composition analysis into standard PCOS evaluation, emphasizing the gut–diet–hormone axis as a therapeutic target.

Adverse Events Due to Suspected Nickel Hypersensitivity in Patients with Essure Micro-Inserts

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

The study reviewed adverse events linked to nickel hypersensitivity in Essure implants, noting rare reactions and the role of the menstrual cycle in modulating hypersensitivity.

What was studied?

This study aimed to review adverse events associated with suspected nickel hypersensitivity in patients implanted with Essure micro-inserts, a device used for hysteroscopic sterilization. The researchers specifically sought to determine the correlation between reported symptoms and positive results of nickel patch testing, providing insights into the relevance of nickel sensitivity in clinical outcomes. The role of the menstrual cycle in modulating nickel hypersensitivity was also examined to better understand potential confounding factors.

Who was studied?

The study utilized data from adverse event reports collected between 2001 and July 2010, including 63 patients with suspected nickel hypersensitivity. These data were drawn from the MAUDE database, direct manufacturer reports, and published clinical trials involving 650 patients. Patch testing, performed at the discretion of treating physicians, was reported for 20 patients, with 13 testing positive and 7 testing negative.

What were the most important findings?

The study found that the incidence of adverse events potentially related to nickel hypersensitivity in Essure users was exceptionally low, at 0.01%. Among the 13 patients with positive nickel patch tests, symptom resolution occurred in only 4 cases after device removal, with symptoms such as rash, itching, and asthma attributed to nickel allergy. However, two cases demonstrated unresolved symptoms, and the remaining showed no definitive link to nickel hypersensitivity. For the 7 patients with negative patch tests, none of their symptoms were deemed related to the implants. Notably, nickel ion release from Essure devices was minimal, with leaching rates 2,143 times lower than daily dietary nickel intake.

The findings highlight inconsistencies between patch test results and clinical symptoms, questioning the predictive reliability of these tests for implant-related nickel hypersensitivity. The report underscores that nickel-sensitive individuals did not universally experience symptoms, and adverse reactions were rare compared to the prevalence of nickel allergy in the general population.

What are the greatest implications of this study?

The findings suggest that nickel hypersensitivity, as determined by patch testing, is not a clinically significant contraindication for Essure device placement. Despite the presence of nickel in these implants, adverse reactions were rare, and many suspected symptoms lacked a clear link to nickel sensitivity. The study emphasizes the importance of careful evaluation before attributing symptoms to nickel hypersensitivity and suggests that device removal should be reserved for confirmed cases. The data also support the continued use of nickel-containing implants with appropriate monitoring, providing reassurance to clinicians and patients about their safety.

Allopregnanolone in premenstrual dysphoric disorder (PMDD)

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This review identifies impaired sensitivity of GABA-A receptors to the neurosteroid allopregnanolone as central to PMDD, linking receptor plasticity and stress dysregulation to mood symptoms, and highlights promising treatments targeting this pathway.

What was studied?

This review comprehensively examined the role of the neuroactive steroid allopregnanolone (ALLO), a potent positive allosteric modulator of the GABA-A receptor (GABAA-R), in the pathophysiology of premenstrual dysphoric disorder (PMDD). It focused on the evidence supporting altered sensitivity or dysregulation of GABAA-Rs in response to ALLO fluctuations across the menstrual cycle, linking these neurobiological changes to the characteristic mood symptoms and stress sensitivity of PMDD.

Who was studied?

As a review article, this paper synthesized findings from both human clinical studies and animal models, particularly rodents, to elucidate mechanisms underlying PMDD. Human studies included neuroendocrine and neurophysiological investigations of women diagnosed with PMDD compared to controls, focusing on hormonal dynamics, receptor sensitivity, stress response, and symptomatology. Rodent models primarily involved progesterone or ALLO withdrawal paradigms to mimic PMDD symptoms and investigate GABAA-R subunit changes and behavior.

What were the most important findings?

The review highlighted that PMDD is not caused by abnormal circulating hormone levels but rather by impaired CNS sensitivity to normal fluctuations of ALLO. In rodent models, rapid withdrawal from progesterone or ALLO induces anxiety- and depression-like behaviors linked to upregulation of the GABAA-R α4 subunit, implicating receptor plasticity in symptom manifestation. Clinical studies in women with PMDD demonstrated altered GABAA-R function, such as lack of ALLO-induced sedation during the luteal phase and elevated anxiety-potentiated startle responses, indicating dysfunctional receptor adaptation to hormonal changes. The review also detailed how ALLO modulates the hypothalamic-pituitary-adrenal (HPA) axis, with women with PMDD showing altered stress responsivity likely due to impaired ALLO-GABAA-R interaction, leading to heightened stress sensitivity during the luteal phase. Importantly, treatments effective in PMDD, including selective serotonin reuptake inhibitors (SSRIs) and novel GABA-modulating drugs appear to normalize ALLO-GABA signaling, further supporting this pathophysiological model.

What are the greatest implications of this study?

This review consolidates strong evidence that PMDD is fundamentally a disorder of impaired neurosteroid modulation of GABAA-Rs, rather than hormone level abnormalities alone, positioning GABAA-R plasticity and ALLO sensitivity as central to its pathophysiology. Understanding this mechanism clarifies why PMDD symptoms cyclically align with hormonal fluctuations and why patients experience heightened stress sensitivity. Clinically, this suggests that future therapeutic strategies should target the neurosteroid-GABAergic system directly to restore receptor function or stabilize neurosteroid levels, promising more rapid and effective symptom relief than traditional antidepressants. Moreover, this framework encourages the development and testing of novel GABAergic agents tailored to PMDD and related reproductive mood disorders, enhancing personalized medicine for affected women worldwide.

Alteration in gut mycobiota of patients with polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study reveals that women with PCOS have altered gut fungal communities, marked by enriched Saccharomyces and depleted Aspergillus. These shifts may contribute to inflammation, metabolic stress, and endocrine disruption, highlighting fungi as critical players in PCOS pathophysiology.

What Was Studied?

This study examined the composition and function of the gut mycobiota in women with polycystic ovary syndrome (PCOS), a condition primarily explored through bacterial profiling in prior research. The authors aimed to fill a critical knowledge gap by focusing on the fungal component of the gut microbiome, which has increasingly been recognized as a modulator of immune responses and metabolic regulation. Using internal transcribed spacer (ITS) sequencing, they compared fecal fungal communities in 17 PCOS patients and 17 age-matched healthy controls. Functional profiling through PICRUSt2 enabled prediction of metabolic pathways associated with the altered mycobiota. The study’s objective was to determine whether PCOS is associated with fungal dysbiosis and to identify fungal genera that may influence metabolic, immune, or hormonal dysregulation in PCOS pathophysiology.

Who Was Studied?

Seventeen women diagnosed with PCOS based on the 2003 Rotterdam criteria were recruited from a hospital in Northeast China and compared with 17 healthy controls. The two groups were matched for age but differed significantly in body weight and BMI, consistent with typical PCOS presentations. PCOS participants had elevated levels of luteinizing hormone, testosterone, fasting insulin, triglycerides, and other markers indicative of endocrine and metabolic dysfunction. Fecal samples from all participants were collected and analyzed for fungal composition, and serum hormone profiles were assessed to correlate gut mycobiota shifts with systemic alterations.

What Were the Most Important Findings?

This study revealed that women with PCOS exhibit marked fungal dysbiosis in their gut microbiota. PCOS patients had significantly lower alpha diversity, as shown by Shannon and Simpson indices, and distinct fungal community structures as demonstrated by β-diversity analyses. At the phylum level, there was a consistent increase in Ascomycota and a reduction in Basidiomycota in PCOS patients. At the genus level, Saccharomyces, Candida, Zygosaccharomyces, Monascus, and Lentinula were significantly enriched, while Aspergillus, Asterotremella, Trichomonascus, and Cryptococcus were depleted. Notably, Saccharomyces and Lentinula were the dominant fungal taxa in PCOS, whereas Aspergillus, which may exert anti-inflammatory and probiotic-supporting effects, was significantly underrepresented.

Functionally, the fungal taxa enriched in PCOS patients contributed to altered pathways involving ceramide glucosyltransferase, uroporphyrinogen-III synthase, and dextransucrase, among others. These functional predictions suggest that gut fungi in PCOS may be involved in modulating host metabolism, immune signaling, and gut barrier function. Saccharomyces overgrowth, previously associated with immune activation and increased intestinal permeability, may exacerbate inflammation and metabolic stress in PCOS. Lentinula, typically a source of immune-stimulating β-glucans, may shift from immunomodulatory to pro-inflammatory roles under dysregulated conditions. In contrast, the depletion of Aspergillus—a β-galactosidase-producing genus linked to probiotic growth and anti-diabetic activity—may remove protective influences from the gut environment.

What Are the Greatest Implications of This Study?

This study introduces a compelling new dimension to PCOS pathogenesis by demonstrating that fungal dysbiosis, not just bacterial, plays a potentially significant role in disease expression. For clinicians, the findings underscore the need to expand microbiome diagnostics beyond bacterial sequencing, particularly in metabolically complex conditions like PCOS. The identification of Saccharomyces and Lentinula as enriched taxa, and Aspergillus as depleted, offers potential fungal biomarkers for disease stratification or treatment response. These fungal alterations may drive systemic inflammation, disrupt gut barrier integrity, and interfere with hormonal and metabolic signaling.

Therapeutically, the results open up new avenues for interventions aimed at modulating the gut mycobiome. Strategies may include antifungal probiotics, dietary modifications to reduce fungal overgrowth, or fungal metabolite targeting. Additionally, the study paves the way for interkingdom microbiome approaches that account for fungal-bacterial interactions, which may be particularly important in developing combination therapies for PCOS. These findings encourage integrative research into the gut–ovary axis that includes fungi as primary actors rather than passive bystanders.

Alterations of bacteriome, mycobiome and metabolome characteristics in PCOS patients with normal/overweight individuals

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The study explores how alterations in gut bacteria, fungi, and serum metabolites are linked to polycystic ovary syndrome (PCOS), revealing potential diagnostic markers and the role of hyperandrogenemia in disease development.

What was studied?

The study investigated the alterations in the bacteriome, mycobiome, and metabolome of patients with polycystic ovary syndrome (PCOS) compared to healthy individuals, specifically focusing on normal and overweight participants. The aim was to evaluate the potential for developing microbiota-related diagnostic markers for PCOS through integrated multi-omics approaches.

Who was studied?

The study involved 88 fecal samples from PCOS patients and healthy controls, including both normal-weight and overweight individuals. Additionally, 87 serum samples were analyzed to investigate the metabolic profiles of these groups.

What were the most important findings?

The study found significant differences in the gut microbiota, mycobiome, and serum metabolome between PCOS patients and healthy controls. Several bacterial genera, such as Ruminococcus torques, Escherichia/Shigella, and Lactobacillus, were identified as distinctive for PCOS patients, with notable differences between normal and overweight participants. PCOS patients exhibited a distinct fungal profile, with an overrepresentation of genera like Candida, Malassezia, and Kazachstania. Fungal diversity was lower in PCOS patients compared to healthy individuals, particularly in those with obesity.

Serum metabolite analysis revealed significant differences between PCOS and healthy groups, particularly in metabolites linked to androgen levels, insulin resistance, and lipids. These metabolites showed strong associations with the clinical markers of PCOS, such as the free androgen index (FAI) and other hormonal and metabolic parameters. The study developed diagnostic models based on serum metabolites, fungal taxa, and bacterial taxa. The metabolite-based model was found to be more accurate than the microbiota-based model in distinguishing between PCOS and healthy controls, especially for patients with normal BMI.

What are the implications of this study?

The study highlights the critical role of hyperandrogenemia in driving gut microbial dysbiosis and metabolic alterations in PCOS patients. This finding suggests that dysbiosis in both the gut bacteriome and mycobiome could be a significant factor in the pathophysiology of PCOS, independent of BMI. The identification of specific microbial signatures and serum metabolites offers a promising avenue for developing more accurate diagnostic methods for PCOS, potentially aiding in earlier diagnosis and personalized treatment approaches. Additionally, the findings may prompt further research into how gut fungi, particularly Candida, contribute to the disease, given their interaction with metabolic and hormonal pathways.

Altered Microbiome-Derived Extracellular Vesicles in Peritoneal Fluid of Women with Endometriosis: Implications for Pathogenesis and Therapy

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies microbiota alterations in ovarian endometrioma, showing distinct microbial shifts in peritoneal fluid extracellular vesicles. Enrichment of Pseudomonas and Acinetobacter, alongside depletion of Propionibacterium and Actinomyces, suggests inflammatory contributions to pathogenesis. Findings highlight the diagnostic potential of microbiota-derived EVs in endometrioma management.

What was studied?

This study examined the microbiota composition in the peritoneal fluid of women with ovarian endometrioma, focusing specifically on microbiome analyses of extracellular vesicles (EVs). Extracellular vesicles are nanometer-sized particles released by cells, including bacteria, that carry microbial DNA and signaling molecules. The research aimed to determine if women with ovarian endometrioma exhibit distinct microbiota profiles in their peritoneal fluid compared to women without endometriosis. Microbial DNA was sequenced using next-generation sequencing (NGS) of the 16S rDNA V3–V4 regions, allowing for detailed taxonomic identification and comparative analysis.

Who was studied?

The study included 45 women diagnosed with histological evidence of ovarian endometrioma and 45 surgical controls confirmed to be free of endometriosis. Participants were recruited from Asan Medical Center, and peritoneal fluid samples were collected during laparoscopic procedures. Women with endometriosis were classified as having advanced-stage disease, and none of the participants had taken antibiotics, probiotics, or hormonal treatments for 12 weeks prior to sample collection.

What were the most important findings?

The microbiota composition of extracellular vesicles in peritoneal fluid was markedly different between women with ovarian endometrioma and controls. Alpha diversity analysis showed no significant differences in species richness between groups, but beta diversity analysis revealed distinct microbial community shifts in the endometriosis group (p < 0.001). Taxonomic profiling demonstrated increased abundances of Acinetobacter, Pseudomonas, Streptococcus, and Enhydrobacter in women with ovarian endometrioma. Conversely, Propionibacterium, Actinomyces, and Rothia were significantly decreased in the endometriosis group (p < 0.05).

At the family level, Pseudomonadaceae and Moraxellaceae were notably enriched in the endometriosis samples, while Veillonellaceae, Propionibacteriaceae, and Actinomycetaceae were reduced. The data also indicated a significant increase in Pseudomonadales and a decline in Actinomycetales at the order level (p < 0.05). These findings suggest that the altered microbiota composition in extracellular vesicles of the peritoneal fluid may contribute to the inflammatory microenvironment observed in ovarian endometrioma.

Microbial GroupOvarian EndometriomaClinical Implications
AcinetobacterIncreasedLinked to inflammation and immune response in the peritoneal cavity
PseudomonasIncreasedAssociated with pathogenic processes in ovarian endometrioma
StreptococcusIncreasedPotential contributor to local inflammation and immune modulation
EnhydrobacterIncreasedMay play a role in extracellular signaling and immune responses
PropionibacteriumDecreasedLoss may disrupt protective anti-inflammatory effects
ActinomycesDecreasedReduced presence suggests compromised mucosal defenses
RothiaDecreasedMay contribute to a disrupted microbial ecosystem
Pseudomonadaceae (Family)EnrichedSuggests pathogenic influence in peritoneal fluid
Moraxellaceae (Family)EnrichedAssociated with peritoneal inflammation
Veillonellaceae, Propionibacteriaceae, Actinomycetaceae (Families)ReducedIndicates loss of protective and commensal populations

What are the greatest implications of this study?

The study provides compelling evidence that women with ovarian endometrioma possess distinct microbial communities in the peritoneal environment, carried via extracellular vesicles. The enrichment of pathogenic genera such as Pseudomonas and Acinetobacter, alongside the depletion of protective taxa like Propionibacterium and Actinomyces, suggests that these microbial imbalances could play a role in local inflammation and disease progression. These findings underscore the potential of microbiota-derived EVs as non-invasive biomarkers for ovarian endometrioma and open the door for targeted microbiome-modulating therapies to alleviate inflammatory responses and halt disease progression.

Anti-Endometriotic Effects of Pueraria Flower Extract: A Novel Therapeutic Approach

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

The study explored the effects of Pueraria Flower Extract (PFE) on human endometriotic cells and mice, showing that PFE inhibits cell adhesion, migration, and MMP expression, and reduces lesion formation. Highlighting PFE's potential as a non-invasive treatment alternative, the research provides insights into molecular targets for future therapies in managing endometriosis, a condition with limited current treatments.

What was studied?

This study investigated the anti-endometriotic effects of Pueraria flower extract (PFE) on human endometriotic cells and a mouse model of endometriosis. It evaluated the extract's impact on cellular adhesion, migration, and the expression of matrix metalloproteinases (MMPs), key factors in the establishment of endometriotic lesion.

Who was studied?

The research focused on human-immortalized endometriotic cell lines (11Z and 12Z) and mesothelial Met5A cells in vitro. Additionally, a mouse model of induced endometriosis was used to evaluate the effects of PFE in vivo.

What were the most important findings?

Inhibition of Cell Adhesion and Migration: PFE significantly suppressed the adhesion of endometriotic cells to mesothelial cells and reduced cell migration in wound-healing and transwell assays.

Reduction in MMP Expression: PFE decreased both mRNA and protein levels of MMP-2 and MMP-9, enzymes crucial for tissue invasion and lesion establishment in endometriosis.

ERK1/2 Signaling Activation: The study demonstrated that PFE activates the ERK1/2 pathway, which played a role in inhibiting cell migration. This effect was reversed when an ERK1/2 inhibitor was introduced.

Lesion Suppression in Mice: Oral administration of PFE to mice significantly reduced the number of endometriotic lesions without causing toxicity or weight loss.

Role of Isoflavones: Major isoflavones such as tectorigenin were identified as active compounds in PFE, contributing to its anti-endometriotic effects.

What are the greatest implications of this study?

The findings suggest that PFE and its active compounds, particularly tectorigenin, could serve as potential therapeutic agents for endometriosis. By targeting matrix metalloproteinase (MMP) activity and the ERK1/2 pathway, PFE may provide a novel, non-hormonal intervention to mitigate lesion formation and progression. This research highlights the potential for plant-derived compounds in developing treatments that reduce the recurrence and side effects associated with conventional endometriosis therapies.

Antibiotic therapy with metronidazole reduces endometriosis disease progression in mice: a potential role for gut microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Antibiotic treatment in mice with surgically-induced endometriosis reduced disease progression by targeting gut bacteria, significantly decreasing lesion size and inflammation. This study suggests a potential gut microbiota link to endometriosis and indicates a novel treatment approach, warranting further investigation for human application.

What was studied?

The research investigated the impact of antibiotic treatment on the progression of endometriosis, specifically examining how altering the gut microbiota with antibiotics affects the development of endometriotic lesions in a mouse model.

 

Who was studied?

Mice subjected to surgically induced endometriosis were studied. These mice were treated with either broad-spectrum antibiotics or metronidazole to assess the effects of these treatments on the progression of endometriosis.

 

What were the most important findings?

The study found that antibiotic therapy, especially broad-spectrum antibiotics, significantly reduced the size and proliferation of endometriotic lesions compared to vehicle-treated mice. It also reduced inflammatory responses within the lesions. Treatment with metronidazole alone, unlike neomycin, resulted in significantly smaller lesions. Furthermore, fecal microbiota transfer from mice with endometriosis could restore lesion growth and inflammation in metronidazole-treated mice.

 

What are the greatest implications of this study?

These results suggest a crucial role of gut bacteria in promoting endometriosis progression in mice, indicating that manipulating the gut microbiota could offer a new therapeutic strategy for managing endometriosis. If these findings apply to humans, they could lead to the development of improved diagnostic tools and personalized treatment strategies, potentially with fewer side effects than current hormone therapy and surgical options. Further research is needed to understand the mechanisms involved and to explore the translatability of these findings to human patients.

Antibiotic therapy with metronidazole reduces endometriosis disease progression in mice: a potential role for gut microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study shows that antibiotic therapy with metronidazole reduces endometriotic lesion growth and inflammation in mice by targeting the gut microbiota, particularly Bacteroidetes. Findings suggest that microbiota-targeted treatments may offer new therapeutic avenues for endometriosis management.

What Was Studied?

This study examined the impact of antibiotic therapy with metronidazole on endometriosis disease progression in a mouse model, exploring its potential effects on gut microbiota and inflammation. Researchers induced endometriosis in mice through surgical transplantation of uterine tissue onto the peritoneal wall. Mice were then treated with either broad-spectrum antibiotics (vancomycin, neomycin, metronidazole, and ampicillin) or metronidazole alone, with control groups receiving vehicle-only treatment. The primary goal was to determine if modulating gut bacteria through antibiotic therapy could reduce endometriotic lesion growth and inflammation, potentially revealing gut microbiota as a therapeutic target.

Who Was Studied?

The study utilized a well-established mouse model of surgically induced endometriosis, where uterine tissue from estrus-stage mice was autologously transplanted onto the peritoneal wall. Mice were separated into groups receiving either broad-spectrum antibiotics, metronidazole alone, or vehicle treatments. To assess the effect of gut microbiota on disease progression, fecal transplantation experiments were performed, where feces from endometriosis-induced mice were gavaged into metronidazole-treated mice to observe the restoration of lesion growth and inflammation.

What Were the Most Important Findings?

The findings demonstrated that antibiotic therapy, particularly with metronidazole, significantly reduced the size and volume of endometriotic lesions in mice. Mice treated with broad-spectrum antibiotics showed a five-fold reduction in lesion size and markedly fewer proliferating cells and macrophages within the lesions compared to vehicle-treated controls (p < 0.01). Metronidazole-treated mice specifically exhibited smaller ectopic lesions than those receiving neomycin or vehicle, suggesting a unique sensitivity of certain gut bacteria to metronidazole's antimicrobial activity. Importantly, inflammation markers, including IL-1β, TNF-α, IL-6, and TGF-β1, were significantly reduced in the peritoneal fluid of metronidazole-treated mice, indicating a diminished inflammatory response.

Additionally, fecal transplantation experiments highlighted the role of gut microbiota in lesion progression. Oral gavage of feces from endometriosis-induced mice restored lesion growth and inflammation in metronidazole-treated mice, implicating gut bacteria as contributors to disease persistence. 16S rRNA sequencing of fecal samples showed that Bacteroidetes were enriched in endometriosis-induced mice and nearly absent in metronidazole-treated mice, suggesting that the suppression of specific microbial populations might underlie the observed therapeutic effects. This reduction in Bacteroidetes correlated with decreased inflammatory responses and smaller lesion sizes, underscoring the interplay between gut microbiota and endometriosis pathology.

What Are the Greatest Implications of This Study

The study provides compelling evidence that targeting the gut microbiota with antibiotics like metronidazole can effectively reduce endometriotic lesion growth and inflammation in a mouse model. The findings suggest that Bacteroidetes may contribute to lesion persistence and immune activation, and their depletion through metronidazole treatment alleviates these pathological effects. This raises the possibility of microbiota-targeted therapies as a novel approach to managing endometriosis, potentially offering a non-hormonal alternative to traditional treatments. Furthermore, the study highlights the significance of gut microbiota modulation in controlling systemic and local inflammatory responses, paving the way for research into gut-mediated mechanisms of endometriosis and microbiome-based therapeutic strategies.

Antioxidant status in relation to heavy metals induced oxidative stress in patients with polycystic ovarian syndrome (PCOS)

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study links elevated arsenic, cadmium, lead, and mercury levels to oxidative stress and low antioxidant defenses in PCOS patients, highlighting heavy metal toxicity as a key contributor to endocrine dysfunction and microbiome imbalance.

What was studied?

This prospective case-control study examined the association between heavy metal exposure and oxidative stress in women with polycystic ovary syndrome (PCOS). Specifically, the study measured serum levels of arsenic (As), cadmium (Cd), lead (Pb), and mercury (Hg), alongside antioxidant markers—superoxide dismutase (SOD) and glutathione (GSH)—to explore how these toxic metals contribute to oxidative damage and PCOS pathogenesis. By analyzing both the toxicant burden and oxidative biomarkers, the study aimed to clarify whether metal-induced oxidative stress plays a pivotal role in the disease process.

Who was studied?

A total of 106 women aged 19–35 participated in the study: 50 were diagnosed with PCOS according to the Rotterdam criteria, and 56 served as healthy controls. Participants were matched in age and excluded for conditions that might confound oxidative or endocrine measurements, including diabetes, cardiovascular disease, and infectious or metabolic disorders. Clinical and demographic data, including menstrual irregularities, acne, BMI, and blood pressure, were collected. Blood samples were analyzed for fasting glucose, HbA1c, lipid profile, luteinizing hormone (LH), antioxidant status (SOD and GSH), and serum heavy metal concentrations using inductively coupled plasma mass spectrometry (ICP-MS).

What were the most important findings?

The study revealed that women with PCOS had significantly elevated serum levels of arsenic, cadmium, lead, and mercury compared to controls. Concurrently, antioxidant defense markers were notably reduced in the PCOS group—SOD and GSH levels were both significantly lower. There were strong negative correlations between heavy metal levels and antioxidant markers: arsenic, lead, and mercury negatively correlated with GSH; arsenic and lead also negatively correlated with SOD. These findings support a mechanistic link between heavy metal burden and reduced antioxidant capacity in PCOS.

From a microbiome perspective, the accumulation of heavy metals like Cd, Pb, and Hg is known to promote dysbiosis. Specifically, these metals reduce beneficial gut bacteria such as Faecalibacterium prausnitzii and Bifidobacterium spp., while promoting pro-inflammatory taxa like Proteobacteria. The oxidative stress induced by metals may increase gut permeability (“leaky gut”), exacerbating systemic inflammation, a hallmark of PCOS. The decline in antioxidant defenses further allows these oxidative effects to persist, creating a vicious cycle of endocrine disruption and microbiome imbalance.

What are the greatest implications of this study?

This study presents compelling evidence that heavy metal exposure significantly contributes to oxidative stress and potentially accelerates PCOS pathogenesis. The diminished antioxidant defenses in PCOS patients exposed to elevated levels of As, Cd, Pb, and Hg suggest that environmental toxicants act as endocrine-disrupting chemicals, impairing reproductive and metabolic health. Clinically, this underscores the necessity of monitoring both oxidative biomarkers and heavy metal burden in PCOS diagnostics and management. Moreover, therapeutic strategies aimed at detoxification—whether via chelation, dietary interventions, or antioxidant supplementation—could help restore oxidative balance and potentially benefit hormonal and microbiome health. Given the tight interplay between oxidative stress, endocrine signaling, and gut microbial composition, the findings advocate for a more integrative approach to PCOS care that includes environmental toxicology and gut microbiome modulation.

Are Heavy Metal Exposure and Trace Element Levels Related to Metabolic and Endocrine Problems in Polycystic Ovary Syndrome?

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study found that women with PCOS had elevated cadmium, antimony, lead, and mercury levels, alongside reduced zinc and copper. These imbalances correlated with insulin resistance, oxidative stress, and inflammation, suggesting that metal exposure contributes to PCOS pathogenesis and potentially alters the gut microbiome.

What was studied?

This prospective clinical study investigated the relationship between serum levels of heavy metals and trace elements and their association with metabolic and endocrine parameters in women with polycystic ovary syndrome (PCOS). Specifically, the study evaluated arsenic, chromium, cadmium, lead, mercury, antimony (Sb), zinc (Zn), and copper (Cu), and how these elements influence oxidative stress, inflammation, insulin resistance, and clinical features like hirsutism in PCOS patients. The goal was to determine if exposure to toxic metals and altered micronutrient profiles could contribute to PCOS pathophysiology via oxidative and inflammatory pathways.

Who was studied?

The study involved 154 women, 84 diagnosed with PCOS according to the Rotterdam criteria and 70 age-matched healthy controls. All participants were screened to exclude confounding endocrine, metabolic, and inflammatory disorders. Clinical assessments included BMI, waist-hip ratio, Ferriman-Gallwey score (FGS) for hirsutism, and metabolic markers such as fasting glucose, insulin, HOMA-IR, and lipid profiles. Blood samples were analyzed for hormonal parameters, oxidative stress indicators (MDA, TOS, TAS, SOD, OSI), inflammatory markers (TNFα, HsCRP), and serum levels of heavy metals and trace elements using inductively coupled plasma mass spectrometry.

What were the most important findings?

Women with PCOS exhibited significantly elevated levels of cadmium, antimony, mercury, and lead, and significantly reduced serum levels of copper and zinc compared to controls. Importantly, the heavy metals cadmium, lead, and antimony positively correlated with fasting glucose and insulin resistance (HOMA-IR), as well as oxidative stress (MDA, TOS) and inflammation (TNFα, HsCRP), while showing negative correlations with antioxidant defense markers (TAS, SOD, OSI). Zinc and copper levels were significantly lower in the PCOS group and correlated with critical markers: zinc negatively with MDA and TNFα, and positively with TAS, suggesting a protective role against oxidative damage.

From a microbiome standpoint, the implications are striking. Elevated cadmium and lead promote gut dysbiosis by favoring inflammatory taxa such as Proteobacteria and reducing SCFA-producing genera like Faecalibacterium prausnitzii. Zinc deficiency suppresses beneficial microbes like Bifidobacterium, while lower copper levels impair mucosal immunity and reduce microbial diversity. These shifts likely exacerbate systemic inflammation and metabolic dysfunction in PCOS, further reinforcing the microbiome–trace element–endocrine axis.

What are the greatest implications of this study?

This study provides robust evidence that environmental heavy metal exposure and trace element imbalance contribute significantly to the oxidative stress and low-grade inflammation underlying PCOS. The data support the role of cadmium, antimony, and lead as endocrine disruptors and metabolic toxins that may worsen insulin resistance and hirsutism. In contrast, reduced zinc and copper levels reflect compromised antioxidant defense and immune regulation. Clinically, these findings justify the integration of trace element and toxic metal screening into PCOS diagnostics. Furthermore, targeted therapies, such as zinc supplementation, chelation strategies, or dietary interventions to limit metal exposure, may enhance treatment outcomes by reducing oxidative burden and restoring microbiome balance. This multifactorial view of PCOS, incorporating toxicology, endocrinology, and gut ecology, opens new opportunities for personalized care and prevention strategies.

Association Between Dietary Patterns and Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

A diet high in processed foods and sugar increases bacterial vaginosis (BV) risk, while a plant-based diet lowers it. This study highlights the importance of dietary choices in vaginal health, providing insights for clinicians on how nutrition influences the vaginal microbiome.

What was Studied?

This study investigated the relationship between dietary patterns and bacterial vaginosis (BV) in women. Researchers analyzed how different diets influenced BV risk, focusing on five major dietary patterns: "Healthy diet," "Unhealthy diet," "Ovo-vegetarian diet," "Pseudo-Mediterranean diet," and "Western diet."

Who was Studied?

The study included 144 women diagnosed with BV and 151 healthy controls. Participants were recruited from a gynecology clinic in Tehran, Iran, between November 2020 and June 2021. Researchers assessed dietary intake using a food frequency questionnaire and diagnosed BV using the Amsel criteria.

Most Important Findings

Women who followed an "Unhealthy diet" high in sugar, solid oils, red meat, sweets, fried potatoes, and refined grains had a significantly higher risk of BV. Those in the highest tertile of this diet were more than three times as likely to have BV compared to those in the lowest tertile.

Conversely, the "Ovo-vegetarian diet," rich in vegetables, beans, whole grains, and eggs, was strongly associated with a lower BV risk. Women in the highest adherence group for this diet had an 84% lower chance of BV compared to those in the lowest adherence group.

The study also observed a protective but not statistically significant effect of the "Pseudo-Mediterranean diet," which includes nuts, fish, olives, and olive oil. No clear association was found between BV and the "Healthy diet" or "Western diet."

Microbiome analysis linked the "Unhealthy diet" with a disruption in vaginal flora, favoring BV-associated bacteria like Gardnerella vaginalis, Bacteroides spp., Mobiluncus spp., and Mycoplasma hominis. In contrast, the "Ovo-vegetarian diet" promoted conditions favorable for Lactobacillus dominance, which helps maintain vaginal health.

Implications of the Study

This study reinforces the role of diet in vaginal microbiome balance and BV risk. Clinicians should encourage patients to reduce processed foods, refined sugars, and saturated fats while promoting a plant-based diet rich in fiber, whole grains, and essential nutrients. Future research should explore whether dietary modifications can serve as an effective strategy for BV prevention and treatment.

Association between endometriosis and risk of histological subtypes of ovarian cancer: a pooled analysis of case–control studies

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

Endometriosis significantly increases the risk of clear-cell, low-grade serous, and endometrioid ovarian cancers. This study highlights the need for subtype-specific ovarian cancer surveillance and prevention strategies.

DOI: https://doi.org/10.1016%2FS1470-2045(11)70404-1

What Was Studied?

This study examined the association between endometriosis and the risk of specific histological subtypes of ovarian cancer. It involved a pooled analysis of 13 Ovarian Cancer Association Consortium case-control studies. The research aimed to clarify whether the increased risk associated with endometriosis extended to all invasive histological subtypes of ovarian cancer or was limited to specific subtypes.

Who Was Studied?

The study analyzed data from 13,226 controls and 9,818 women with ovarian cancer (7,911 with invasive and 1,907 with borderline ovarian cancer). Among these, 738 women with invasive cancer and 168 with borderline cancer reported a history of endometriosis. The data collection spanned multiple international sites and included self-reported histories of endometriosis.

What Were the Most Important Findings?

The study found that a history of endometriosis significantly increased the risk for three specific subtypes of invasive ovarian cancer: clear-cell, low-grade serous, and endometrioid cancers. The odds ratios for these associations were 3.05, 2.11, and 2.04, respectively. In contrast, no significant association was observed with high-grade serous or mucinous subtypes, nor with borderline ovarian cancers. The findings suggest that endometriosis acts as a precursor lesion for clear-cell and endometrioid ovarian cancers, with molecular similarities supporting this hypothesis. Notably, the association with low-grade serous cancer requires further study, as this subtype showed distinct molecular characteristics, such as a higher likelihood of KRAS or BRAF mutations compared to TP53 mutations in high-grade serous cancers.

What Are the Greatest Implications of This Study?

This study underscores the need for clinicians to recognize the increased risk of specific ovarian cancer subtypes in women with endometriosis. Understanding the mechanisms behind the malignant transformation of endometriosis could enable the identification of high-risk individuals for tailored surveillance or preventive strategies, such as risk-reducing surgery. The study also highlights the importance of considering histological subtypes in ovarian cancer research, reflecting its heterogeneous nature and the need for subtype-specific prevention, screening, and treatment approaches.

Association between Gut Microbiota and Breast Cancer: Diet as a Potential Modulating Factor

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study links reduced gut microbial diversity and specific taxa (e.g., Acidaminococcus, Hungatella) to breast cancer, influenced by diet. Findings suggest microbiome-targeted interventions and dietary strategies could mitigate breast cancer risk.

What Was Studied?

This study examined the association between gut microbiota composition and breast cancer, focusing on the role of diet as a potential modulating factor. Researchers conducted a case-control study involving 42 newly diagnosed, treatment-naïve BCa patients and 44 age-matched cancer-free controls. The gut microbiome was analyzed through 16S rRNA sequencing, and dietary patterns were assessed using the National Cancer Institute Diet History Questionnaire.

Who Was Studied?

Participants included females aged 20–89 years from the Oregon Health & Science University. breast cancer patients were diagnosed through biopsy and had not yet undergone any treatment. Cancer-free controls were matched by age and underwent recent mammograms with non-suspicious results. The study collected fecal samples, dietary data, and comprehensive lifestyle information to ensure robust comparisons.

Most Important Findings

The study identified significant differences in the gut microbiome composition between breast cancer cases and controls, including reduced microbial diversity among breast cancer patients, indicative of dysbiosis. Specifically, the genera Acidaminococcus, Hungatella, and Tyzzerella were enriched, while controls exhibited enrichment of genera such as Christensenellaceae and Dialister. These findings were linked to dietary patterns: Acidaminococcus correlated with lower fruit intake, Hungatella with reduced dairy intake but increased vegetable consumption, and Tyzzerella was not significantly associated with dietary variables. Importantly, the reduced diversity and altered microbial profiles in breast cancer patients align with previous evidence suggesting a role for gut dysbiosis in cancer progression via immune modulation and microbial metabolite production.

Greatest Implications

This study highlights the gut microbiome's potential as a biomarker for breast cancer risk and emphasizes the role of diet in modulating microbial composition. Dysbiosis, characterized by an imbalance in gut microbiota, is linked to breast cancer, suggesting that microbiome-targeted dietary interventions could aid in prevention and management. For example, increased consumption of whole fruits may help reduce levels of Acidaminococcus, a genus enriched in breast cancer patients, while higher dairy intake could lower the abundance of Hungatella, a genus associated with TMAO production and cancer-promoting pathways. Interestingly, the study also found that greater vegetable consumption was linked to higher levels of Hungatella, which has been associated with increased risks of both breast and colorectal cancer. These findings underscore the complexity of dietary influences on the gut microbiome and their potential role in cancer prevention.

Association Between Heavy Metal Exposure and Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This cross-sectional study identified a strong link between elevated serum lead and cadmium levels and increased risk of bacterial vaginosis. It suggests that heavy metal exposure may disrupt vaginal microbiota stability and immunity, contributing to BV susceptibility and pointing to new environmental factors in BV prevention strategies.

What was Studied?

The study examined the association between exposure to heavy metals, specifically lead, cadmium, and mercury, and the risk of bacterial vaginosis (BV) among American women. Using a cross-sectional design, the researchers analyzed data from 2,493 women aged 18 to 49 years who participated in the 2001–2004 cycles of the National Health and Nutrition Examination Survey (NHANES). They measured serum levels of these heavy metals and assessed BV status using Nugent scoring, aiming to clarify whether environmental exposure to heavy metals correlates with BV prevalence.

Who was Studied?

The study included 2,493 American women aged between 18 and 49 years. All participants were selected from NHANES datasets, which provide a representative sample of the U.S. population. The researchers collected vaginal swabs to diagnose BV using the Nugent score and measured serum concentrations of lead, cadmium, and mercury. They controlled for several covariates such as age, body mass index, socioeconomic factors, cholesterol levels, and physical activity to ensure reliable statistical analysis.

Most important findings

The study found a significant positive association between serum lead and cadmium levels and the risk of developing bacterial vaginosis. Specifically, women with the highest serum lead concentrations had a 35% increased risk of BV compared to those with the lowest levels. Similarly, higher cadmium levels were associated with a 41% increased risk of BV in fully adjusted models. However, the researchers found no significant association between serum mercury levels and BV risk.

Stratified analyses revealed that the positive association between lead exposure and BV was more pronounced in women aged 37 to 49 years, those with lower education levels, and those with a higher body mass index. For cadmium, the risk was especially higher among women aged 18 to 24 and 37 to 49 years, and among those of non-Hispanic white and black ethnicity. These results suggest that lead and cadmium may influence vaginal microbiota stability, possibly through immunotoxic or endocrine-disrupting mechanisms, contributing to vaginal dysbiosis and increased BV susceptibility.

Implications of this Study

This study provides the first epidemiological evidence linking heavy metal exposure to increased risk of bacterial vaginosis. The findings suggest that environmental pollutants may act as overlooked risk factors in BV pathogenesis by compromising host immune function, disrupting hormonal balance, and potentially altering the vaginal microbiome. Clinicians and public health officials should consider environmental heavy metal exposure as part of BV risk assessment and prevention strategies. Reducing heavy metal exposure through regulatory policies and patient education could offer an additional layer of protection against BV and its associated reproductive health risks. These results highlight the importance of integrating environmental factors into the broader framework of microbiome-related disease prevention.

Association between Polycystic Ovary Syndrome and Gut Microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study demonstrates that gut dysbiosis in PCOS is not just correlative but causative. FMT and Lactobacillus transplantation restored hormonal balance and ovarian morphology in PCOS rats, confirming gut microbiota as a viable therapeutic target.

What Was Studied?

This study examined the causal relationship between gut microbiota dysbiosis and the pathophysiology of polycystic ovary syndrome (PCOS) using a letrozole-induced rat model. The researchers aimed to determine whether changes in gut microbiota are not merely consequences of PCOS but actively contribute to its development and progression. They evaluated bacterial composition, estrous cycles, sex hormone levels, and ovarian morphology in rats treated with letrozole to induce PCOS. They then investigated whether modulating the gut microbiota through fecal microbiota transplantation (FMT) or Lactobacillus transplantation could reverse PCOS phenotypes. This intervention-based design enabled them to assess the therapeutic potential of microbiota manipulation.

Who Was Studied?

The study used 32 female Sprague-Dawley rats, divided into four groups: a control group, a PCOS group induced by daily oral administration of letrozole, a PCOS group treated with Lactobacillus transplantation, and a PCOS group treated with FMT from healthy rats. The authors collected fecal, serum, and ovarian tissue samples at baseline and post-intervention to evaluate microbiota composition and systemic hormonal effects. This preclinical model allowed for a mechanistic investigation of the microbiota-hormone interaction and its role in reproductive dysfunction.

What Were the Most Important Findings?

The study found that letrozole-induced PCOS rats exhibited classic PCOS phenotypes: disrupted estrous cycles, elevated androgen levels, and cystic ovarian morphology with diminished granulosa layers. These rats also showed marked gut microbiota dysbiosis, characterized by decreased abundance of Lactobacillus, Ruminococcus, and Clostridium, and increased levels of Prevotella. Quantitative PCR confirmed these microbial shifts, while DGGE and sequence analysis further identified species such as Prevotella melaninogenica, Pseudomonas monteilii, and Roseburia intestinalis as more abundant in PCOS rats, and Lactobacillus johnsonii and Ruminococcus torques as depleted.

Following treatment, both FMT and Lactobacillus transplantation improved estrous cycling and normalized ovarian morphology. Hormonal analysis showed that these interventions decreased testosterone and androstenedione while increasing estradiol and estrone levels. FMT produced more pronounced effects than Lactobacillus alone. Importantly, microbial restoration accompanied hormonal normalization, particularly with increased Lactobacillus and Clostridium and decreased Prevotella. These findings provide direct evidence that gut microbial composition can influence endocrine pathways central to PCOS, potentially through mechanisms involving modulation of estrogen biosynthesis and androgen metabolism.

What Are the Greatest Implications of This Study?

This study provides strong preclinical evidence that gut microbiota dysbiosis is not merely a byproduct of PCOS but actively contributes to its endocrine and reproductive features. The observed reversal of PCOS symptoms through FMT and Lactobacillus transplantation suggests that targeted microbial therapies may offer a novel, non-hormonal strategy for treating PCOS. Clinically, these findings support the inclusion of microbiota analysis in the diagnostic and therapeutic planning for PCOS, especially in patients resistant to standard hormonal therapies or those with gastrointestinal symptoms. The identification of Prevotella as a potentially pathogenic genus and Lactobacillus as beneficial aligns with emerging microbiota-based therapeutic models across endocrine disorders. The broader implication is that gut microbiota modulation, through FMT, probiotics, or dietary interventions, could become a cornerstone in managing PCOS by targeting its underlying metabolic and inflammatory components rather than solely addressing reproductive symptoms

Association Between Smoking and Premenstrual Syndrome: A Meta-Analysis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This meta-analysis identifies a significant association between smoking and increased risk of PMS and PMDD. Women who smoke are more likely to experience these disorders, with stronger effects in PMDD.

What was reviewed?

This meta-analysis systematically reviewed studies investigating the association between smoking and premenstrual syndrome (PMS). The authors sought to quantify the relationship between smoking behaviors and the risk of developing PMS or its more severe form, premenstrual dysphoric disorder (PMDD). Thirteen studies involving 25,828 participants were included in this analysis, which aimed to synthesize the results from multiple cohorts, case-control, and cross-sectional studies. The review specifically examined the effect of smoking on the likelihood of developing PMS, with a focus on the severity of symptoms and potential biological mechanisms.

Who was reviewed?

The studies reviewed in this meta-analysis involved a range of populations, including university students, general populations, and patients from different regions. Participants were primarily women of reproductive age, including those diagnosed with PMS or PMDD. The reviewed studies used varying methods for assessing smoking (e.g., self-reported smoking status, smoking quantity) and PMS (e.g., standardized questionnaires, prospective symptom tracking). The sample sizes ranged from smaller case-control studies with fewer than 100 participants to larger cross-sectional studies with over 3,000 participants.

What were the most important findings?

The meta-analysis found a statistically significant association between smoking and the increased risk of PMS. Specifically, smoking was linked to a moderate increase in the odds of developing PMS. This relationship was even stronger for PMDD. The results indicated that women who smoke are more likely to experience PMS, with a stronger association observed in women with the more severe form of PMS, PMDD. Interestingly, the study also identified that smoking behavior during the luteal phase of the menstrual cycle may be influenced by hormonal fluctuations, with nicotine intake potentially exacerbating mood disturbances commonly associated with PMS. Nicotine’s effects on the hypothalamic-pituitary-adrenal (HPA) axis, which is already compromised in PMS, may worsen the stress response, further complicating both PMS and Tobacco Use Disorder in this population.

What are the greatest implications of this review?

The findings suggest that smoking should be considered a modifiable risk factor for PMS and PMDD. The moderate increase in risk, particularly for PMDD, highlights the need for targeted interventions in women who smoke, particularly those suffering from PMS. Clinicians should be aware of the potential exacerbating effects of smoking on menstrual health and consider integrating smoking cessation strategies into the management plans for women with PMS or PMDD. Additionally, the review emphasizes the need for further research into the underlying biological mechanisms, including the role of nicotine in neurocircuitry and stress responses, to improve treatment strategies for both PMS and smoking dependence.

Association of the Cervical Microbiota With Pregnancy Outcome in a Subfertile Population Undergoing In Vitro Fertilization: A Case-Control Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The study investigated the link between cervical microbiota and IVF outcomes in subfertile women. It found that higher microbial diversity and specific bacterial abundances correlated with successful pregnancy. These insights suggest potential for microbiota-based diagnostics and treatments to enhance IVF success.

What Was Studied?

The study focused on the association between the composition of cervical microbiota and pregnancy outcomes in a subfertile population undergoing in vitro fertilization (IVF). Specifically, it aimed to characterize the cervical microbiota of patients undergoing embryo transfer (ET) and assess whether the composition of these microbiota is associated with the outcomes of the IVF treatments. The study utilized high-throughput sequencing technology to analyze the cervical microbiota and explored factors contributing to the observed compositions.

 

Who Was Studied?

The participants in this study were infertile female patients undergoing IVF treatment at the Reproductive Center of Shengjing Hospital of China Medical University. The study included 100 patients who met specific inclusion criteria: aged between 20 to 40, undergoing assisted reproductive technology with their own gametes, and transferring two cleavage-stage embryos. Patients with autoimmune, endocrine, cervical, or endometrial diseases or blood contamination of collected samples were excluded.

 

What Were the Most Important Findings?

Diversity of Microbiota: Higher α diversity in the cervical microbiota was observed in the clinical pregnancy groups compared to non-pregnancy groups, especially in fresh IVF-ET cycles. This suggests a richer microbial environment may be associated with positive pregnancy outcomes.

Microbial Composition: Significant differences in the β diversity (overall microbiota composition) were noted between pregnancy outcomes in both fresh and frozen-thawed cycles, with fresh cycles showing a more pronounced difference.

Specific Microorganisms: In fresh cycles, certain bacterial genera such as Lactobacillus, Akkermansia, Desulfovibrio, Atopobium, and Gardnerella showed different abundances between pregnant and non-pregnant groups. Lactobacillus, in particular, was found to be negatively correlated with other bacteria but positively correlated with serum estradiol levels, which are critical for pregnancy.

Predictive Analysis: Logistic regression analysis indicated that the composition of the cervical microbiota on the day of ET is significantly associated with clinical pregnancy outcomes.

What Are the Greatest Implications of This Study?

Clinical Implications: The study implies that understanding and potentially manipulating the cervical microbiota could enhance IVF success rates. This could lead to new diagnostic tools or treatment strategies, such as prebiotic or probiotic interventions to optimize the cervical microbial environment before embryo transfer.

Research Implications: The findings stress the need for more in-depth research into the role of the microbiota in fertility and pregnancy outcomes. It calls for longitudinal studies to explore how cervical microbiota changes over time and its interactions with hormonal levels and other physiological factors during fertility treatments.

Therapeutic Implications: Given the association between specific bacterial populations and pregnancy success, there might be potential for developing microbial-based therapies or supplements to support IVF procedures, enhancing the uterine environment for embryo implantation and pregnancy continuation.

This study opens up new pathways for both understanding and improving reproductive health interventions in subfertile populations undergoing assisted reproductive technologies.

Association of Trace Elements with Polycystic Ovary Syndrome in Women—A Case-Control Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This case–control study links high copper and low molybdenum levels to metabolic, hormonal, and inflammatory changes in PCOS. Dietary habits influenced these levels, suggesting potential for targeted nutritional interventions.

What was studied?

This case–control study investigated the association between trace element levels and polycystic ovary syndrome (PCOS) in reproductive-aged women. Researchers measured concentrations of both essential trace elements including manganese (Mn), copper (Cu), zinc (Zn), selenium (Se), and molybdenum (Mo), and non-essential trace elements, arsenic (As), cadmium (Cd), mercury (Hg), and lead (Pb) in urine, serum, and whole blood. The study sought to examine how these elements might influence kidney and liver function, metabolic and endocrine parameters, and potential environmental or dietary exposure.

Who was studied?

The study population included 70 women, divided evenly between 35 diagnosed PCOS patients and 35 healthy controls. Participants were aged 20–39 years and selected based on strict criteria to exclude comorbidities such as diabetes, cardiovascular or autoimmune diseases, and recent hormonal treatments. All participants underwent clinical assessments, including anthropometrics, ultrasonography, and hormonal profiling. Dietary, environmental, and lifestyle exposures were gathered using detailed questionnaires, while biological samples were analyzed using high-precision inductively coupled plasma mass spectrometry.

What were the most important findings?

The most significant findings showed that women with PCOS had elevated serum copper (Cu) and reduced whole blood and serum molybdenum (Mo) levels compared to controls. Although these differences lost statistical significance after adjusting for BMI, age, and hematocrit, other associations remained clinically relevant. Cu levels positively correlated with leukocyte count, suggesting an inflammatory link. Conversely, Mo levels negatively correlated with luteinizing hormone (LH), urinary bilirubin, and markers of kidney function such as proteinuria.

From a microbiome perspective, altered copper and molybdenum levels are particularly important. Elevated Cu can promote oxidative stress, disrupt mucosal immunity, and reduce microbial diversity, especially suppressing beneficial bacteria such as Faecalibacterium prausnitzii. Meanwhile, low Mo levels can impair molybdoenzyme function necessary for detoxification and redox regulation, which may result in the accumulation of inflammatory metabolites that disturb the gut barrier. These changes may exacerbate the chronic low-grade inflammation and hormonal dysregulation characteristic of PCOS. The study also identified that beef consumption correlated positively with Cu levels, and cereal and boiled vegetable consumption correlated positively with Mo levels, linking dietary sources directly to trace element concentrations.

What are the greatest implications of this study?

This study provides new insight into the potential role of Mo as well as supporting existing findings on Cu. It suggests that nutritional and environmental factors significantly influence the body’s trace element status, which in turn may modulate inflammation, liver and kidney function, and reproductive hormone levels in women with PCOS. Clinically, these findings support incorporating dietary assessments and trace element screening in the management of PCOS. More importantly, this study underscores a microbiome-relevant pathway: alterations in Cu and Mo not only influence host metabolism but likely affect gut microbial balance, further perpetuating systemic metabolic dysfunction. This integrative perspective could pave the way for dietary or supplemental interventions targeting TE imbalances to improve reproductive and metabolic outcomes in PCOS.

Associations Between Endometriosis and Gut Microbiota

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This case control study explores the gut microbiota's association with endometriosis in women, comparing 66 patients to 198 controls. Using 16S rRNA sequencing, it was found that patients have lower diversity in their gut bacteria and significant differences in the abundance of 12 bacterial types, suggesting that endometriosis may influence gut microbiota composition.

What was studied?

The study examined the gut microbiota in women with endometriosis compared to healthy controls. It aimed to explore differences based on disease localization, symptoms, or treatment and assess the gut microbiota’s potential role in the pathogenesis of endometriosis.

 

Who was studied?

66 women diagnosed with endometriosis at Skåne University Hospital were studied alongside 198 matched controls from the Malmö Offspring Study, assessing their gut microbiota through 16S rRNA sequencing.

 

What were the most important findings?

Significant findings include higher overall microbial diversity in controls compared to endometriosis patients, with specific differences in the abundance of 12 bacteria types between the two groups. After adjusting for false discovery rates, no significant microbiota differences were found within the endometriosis cohort.

 

What are the greatest implications of this study?

The study implies that gut microbiota may be altered in individuals with endometriosis, suggesting a possible link between gut microbiota and the pathogenesis or symptomatology of endometriosis. These findings highlight the need for further research on the gut microbiota’s role in endometriosis, potentially leading to new diagnostic and treatment strategies.

Associations Between Endometriosis and Gut Microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

The gut microbiota has been associated with many diseases, including endometriosis. However, very few studies have been conducted on this topic in human. This study aimed to investigate the association between endometriosis and gut microbiota. Women with endometriosis (N=66) were identified at the Department of Gynaecology and each patient was matched with three controls (N=198) from the general population. All participants answered questionnaires about socioeconomic data, medical history, and gastrointestinal symptoms and passed stool samples. Gut bacteria were analyzed using 16S ribosomal RNA sequencing, and in total, 58 bacteria were observed at genus level in both patients with endometriosis and controls. Comparisons of the microbiota between patients and controls and within the endometriosis cohort were performed. Both alpha and beta diversities were higher in controls than in patients. With the false discovery rate q<0.05, abundance of 12 bacteria belonging to the classes Bacilli, Bacteroidia, Clostridia, Coriobacteriia, and Gammaproteobacter differed significantly between patients and controls. Differences observed between patients with or without isolated ovarian endometriosis, involvement of the gastrointestinal tract, gastrointestinal symptoms, or hormonal treatment disappeared after calculation with false discovery rate. These findings indicate that the gut microbiota may be altered in endometriosis patients.

What Was Studied?

This study investigated the association between endometriosis and gut microbiota. Conducted at Skåne University Hospital in Sweden, the research aimed to understand how the gut microbiome differs in women diagnosed with endometriosis compared to healthy controls. The study included 66 women with endometriosis confirmed through laparoscopy or laparotomy and 198 age, BMI, and smoking-matched controls. Stool samples from both groups were analyzed using 16S ribosomal RNA sequencing to identify bacterial composition at the genus level. The primary objective was to compare the diversity and abundance of gut microbiota between the two groups and explore any microbiome changes correlated with endometriosis characteristics such as disease localization, gastrointestinal symptoms, or hormonal treatment.

Who Was Studied?

The study examined 66 women diagnosed with endometriosis recruited from the Department of Gynaecology at Skåne University Hospital. These participants were matched with 198 controls from the Malmö Offspring Study (MOS), ensuring similarities in age, BMI, and smoking status. Women in the endometriosis group were diagnosed based on clinical criteria, confirmed through surgical procedures, and were excluded if they had comorbid gastrointestinal conditions like Crohn's disease, ulcerative colitis, or irritable bowel syndrome (IBS). The control group, drawn from a population-based cohort, also passed stool samples and completed questionnaires about their medical history and gastrointestinal symptoms.

What Were the Most Important Findings?

The study found significant differences in gut microbiota diversity and composition between women with endometriosis and healthy controls. Notably, alpha and beta diversities were higher in the control group, suggesting a richer and more varied microbial population compared to endometriosis patients. At the genus level, 12 bacterial genera belonging to the classes Bacteroidia, Clostridia, Coriobacteriia, Bacilli, and Gammaproteobacteria differed significantly between groups. For instance, Bacteroides and Parabacteroides were elevated in endometriosis patients, while Paraprevotella and Lachnospira were found in lower abundance compared to controls. Additionally, there was a distinct alteration in the microbial community within the endometriosis cohort based on disease localization and the presence of gastrointestinal symptoms. Patients with isolated ovarian endometriosis exhibited higher levels of Lachnobacterium and Adlercreutzia compared to those with widespread lesions. Furthermore, the presence of gastrointestinal symptoms correlated with lower levels of SMB53 (Clostridia) and Odoribacter (Bacteroidia), while Prevotella was more abundant. Interestingly, hormone treatment was associated with higher levels of Blautia and Ruminococcus in the Clostridia class, along with Butyricimonas in the Bacteroidia class. These findings support the hypothesis that gut microbiota may be altered in endometriosis patients, with distinct microbial signatures linked to hormonal therapy and gastrointestinal involvement.

Increased in Endometriosis PatientsDecreased in Endometriosis Patients
Bacteroides (Bacteroidia)Paraprevotella (Bacteroidia)
Parabacteroides (Bacteroidia)Lachnospira (Clostridia)
Blautia (Clostridia) with hormone treatmentOdoribacter (Bacteroidia) with GI symptoms
Ruminococcus (Clostridia) with hormone treatmentSMB53 (Clostridia) with GI symptoms
Butyricimonas (Bacteroidia) with hormone treatment

What Are the Greatest Implications of This Study?

The study's findings suggest that endometriosis is associated with specific alterations in gut microbiota, which could play a role in the pathophysiology of the disease. The reduced microbial diversity in endometriosis patients points towards a potential dysbiosis that may exacerbate inflammation and modulate estrogen metabolism, both of which are critical in the pathogenesis of endometriosis. Furthermore, specific bacterial shifts linked to hormone treatment indicate that gut microbiota could be influenced by estrogen-related therapies, potentially affecting symptom severity and disease progression. Understanding these microbial associations opens the door to novel therapeutic strategies, such as targeted probiotics or microbiome-based interventions, to alleviate gastrointestinal symptoms and modulate disease activity in endometriosis patients. This research also underlines the need for further studies to explore the bidirectional relationship between gut microbiota and estrogen regulation in estrogen-dependent conditions like endometriosis.

Bacterial infection linked to endometriosis

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Study links Fusobacterium to endometriosis via inflammation and lesion formation. Antibiotics (metronidazole and chloramphenicol) may offer therapeutic potential.

New title: Fusobacterium Infection: A New Pathogenic Insight into Endometriosis and Microbiome-Targeted Therapy Potential

DOI: https://doi.org/10.1016/S2666-5247(23)00221-5

What Was Studied?

This translational study investigated the role of Fusobacterium infection in the pathogenesis of endometriosis. Researchers sought to determine whether bacterial infection, specifically by Fusobacterium nucleatum, contributes to inflammatory alterations in endometrial fibroblasts, potentially leading to the development of endometriosis. The study encompassed molecular analyses, in vitro experiments, and a mouse model to establish causality and mechanism.

Who Was Studied?

The study analyzed uterine tissue samples from 79 patients in two Japanese hospitals, divided into cases with endometriosis and controls without the condition. Further, a mouse model was used to test the infectivity and pathogenic potential of Fusobacterium nucleatum compared to other microbes.

What Were the Most Important Findings?

Fusobacterium nucleatum was found to be significantly more prevalent in the endometrial and endometriotic tissues of patients with endometriosis (64.3%) compared to controls (7.1%), while Erysipelothrix, another potential candidate, was not abundant. Fusobacterium infection was shown to upregulate transgelin (TAGLN) expression in fibroblasts, enhancing their motility, adhesion, and migration through the activation of TGF-β signaling, a pathway known to be critical in the progression of endometriosis. In an animal model, mice inoculated with Fusobacterium-infected uterine tissue developed endometriotic lesions, whereas treatment of donor mice with antibiotics (metronidazole and chloramphenicol) significantly reduced lesion formation in recipient mice. These findings suggest that targeting Fusobacterium with antibiotics holds potential to mitigate the progression of endometriosis, underscoring the importance of further exploration into microbiome-targeted therapies.

What Are the Greatest Implications of This Study?

This research provides evidence that Fusobacterium infection may play a direct role in the etiology of endometriosis. The identification of a bacterial trigger opens avenues for antibiotic-based interventions and highlights the need for clinical trials targeting endometrial infections. Additionally, it underscores the importance of microbial profiling in endometriosis diagnosis and management, potentially redefining treatment paradigms to include microbiome-targeted interventions (MBTIs).

Bacterial Vaginosis - A Brief Synopsis of the Literature

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explores bacterial vaginosis, emphasizing its recurrence, microbial associations, and treatment challenges. It highlights the need for microbiome-based therapies, standardized diagnostic criteria, and potential partner treatment to reduce reinfection. The findings underscore the importance of improved strategies for long-term BV management.

What Was Reviewed?

This review provides a comprehensive synopsis of the current literature on bacterial vaginosis (BV), focusing on its epidemiology, recurrence, persistence, and treatment challenges. The authors examine the impact of BV on reproductive and sexual health, highlighting its association with sexually transmitted infections (STIs) and adverse pregnancy outcomes. Additionally, the review explores the role of the vaginal microbiota in BV pathogenesis, emphasizing the need for more effective long-term treatment options and standardized definitions for recurrent and persistent BV.

Who Was Reviewed?

The review synthesizes studies on women of reproductive age diagnosed with BV, including those with recurrent infections. It also explores research on the vaginal microbiota, sexual partners' role in BV transmission, and the effectiveness of current treatments. By analyzing epidemiological data from various regions, it highlights differences in BV prevalence and risk factors.

Key Findings and Microbial Associations

BV shifts the vaginal microbiota by depleting Lactobacillus species and allowing anaerobic bacteria like Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., and Mobiluncus spp. to overgrow. This microbial imbalance disrupts the vaginal ecosystem and increases susceptibility to STIs, including Neisseria gonorrhoeae, Chlamydia trachomatis, and HIV. Even with metronidazole or clindamycin treatment, BV recurs in up to 80% of cases within three months. The review explores BV recurrence, showing how reinfection, an inability to restore a Lactobacillus-dominant microbiota, and bacterial biofilms contribute to persistent infections. Clinicians rely on Amsel’s criteria and Nugent scoring for diagnosis, but inconsistent definitions of recurrent and persistent BV complicate management. The review also examines partner treatment as a strategy to reduce BV recurrence, though past studies show mixed results.

Implications of the Review

BV remains a significant clinical challenge due to its high recurrence rates, unclear etiology, and association with reproductive health complications. This review calls for more research into microbiome-based therapies, improved diagnostic tools, and standardized definitions of recurrent BV. The findings suggest that future treatment approaches should not only target BV-associated bacteria but also focus on restoring a stable vaginal microbiota. Additionally, reconsidering partner treatment as part of BV management could be an avenue for reducing recurrence rates, provided that future studies can confirm its effectiveness.

Bacterial vaginosis and biofilms: Therapeutic challenges and innovations

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review links BV recurrence to resilient biofilms formed by Gardnerella vaginalis. Probiotics and biofilm disruptors (e.g., Astodrimer gel) improve outcomes by restoring Lactobacillus dominance. Current antibiotics fail to penetrate biofilms, necessitating multimodal therapies. Future research should explore VMT and microbiome-targeted interventions for sustained BV remission.

What was Reviewed?

This narrative review examined the role of biofilms in bacterial vaginosis (BV), focusing on their contribution to treatment resistance and recurrence. The authors synthesized evidence from clinical studies and trials to evaluate the limitations of current antibiotic therapies and explored emerging solutions, such as biofilm-disrupting agents and probiotics, to improve BV management.

Who was Reviewed?

The review analyzed data from diverse patient populations in clinical studies, including women with recurrent BV. It incorporated findings from trials investigating biofilm-targeted therapies, such as enzymatic disruptors (e.g., dispersin B) and probiotics (e.g., Lactobacillus crispatus), to assess their efficacy in restoring vaginal microbiota balance.

What were the most Important Findings?

The review highlighted that BV-associated biofilms, primarily formed by Gardnerella vaginalis and Atopobium vaginae, shield pathogenic bacteria from antibiotics, driving recurrence. Major microbial associations (MMA) included polymicrobial anaerobic communities displacing protective Lactobacillus species. Probiotics and biofilm-disrupting agents (e.g., boric acid, Astodrimer gel) showed promise in clinical trials, with probiotics delaying recurrence by 51% and Astodrimer gel significantly reducing recurrence rates. Notably, Lactobacillus crispatus-based therapies were emphasized for restoring vaginal acidity and inhibiting biofilm formation.

What are the Implications of this Review?

The findings emphasize the need to shift from antibiotic-only approaches to multimodal strategies targeting biofilms. Clinicians should consider adjunct therapies like probiotics and biofilm disruptors to enhance treatment efficacy and reduce recurrence. The review also calls for further research into vaginal microbiome transplantation (VMT) and personalized therapies to address biofilm resilience.

Bacterial Vaginosis and Chlamydia in Tubal Infertility

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

Study links BV and past chlamydial infection to tubal infertility, with 87.5% of BV-positive women having tubal damage. Both infections were often asymptomatic. IVF pregnancy rates were unaffected, but BV showed lower implantation trends. Findings highlight BV’s role in infertility, urging early screening to prevent tubal damage.


What was Studied?

This cross-sectional study investigated the association between bacterial vaginosis (BV), past chlamydial infection, and tubal infertility in women undergoing IVF. The researchers analyzed vaginal swabs and serologic data from 286 women undergoing 344 IVF cycles to determine whether these infections impacted pregnancy rates or were linked to specific infertility causes.

Who was Studied?

The study included 286 women undergoing IVF treatment at a tertiary care infertility referral center in Glasgow, Scotland. Participants provided high vaginal and endocervical swab samples before oocyte retrieval, with serologic testing for Chlamydia trachomatis and BV diagnosis based on Gram staining and anaerobic culture.

What were the most Important Findings?

The study found strong, independent associations between tubal infertility and both BV (87.5% of BV-positive women had tubal damage) and past chlamydial infection (91.2% seropositivity in tubal infertility cases). Notably, BV and chlamydial infections were frequently asymptomatic, with no active chlamydial infections detected. Major microbial associations (MMA) included reduced Lactobacillus dominance in BV-positive women, alongside overgrowth of anaerobic bacteria. Despite these associations, pregnancy rates after IVF were unaffected by BV or past chlamydial infection, though BV-positive women had numerically lower implantation rates (15.2% vs. 31.0% in chlamydia-seropositive women).

What are the Implications of this Study?

The findings underscore BV as a potential pelvic pathogen contributing to tubal damage, independent of chlamydial infection. While IVF success rates remained comparable across groups, the high prevalence of tubal infertility in BV-positive women suggests that early screening and treatment of asymptomatic BV could prevent long-term reproductive complications. Clinicians should consider BV as a modifiable risk factor in infertility workups, particularly in cases of unexplained tubal pathology.

Bacterial Vaginosis and its Association with Infertility, Endometritis, and Pelvic Inflammatory Disease

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

BV disrupts Lactobacillus dominance, increasing infertility risk via inflammation, PID, and endometritis. L. crispatus probiotics reduce BV recurrence; CE treatment boosts IVF success. Early screening and microbiome-targeted therapies are vital to prevent reproductive complications.

What was Reviewed?

This expert review examined the associations between bacterial vaginosis (BV), endometritis, pelvic inflammatory disease (PID), and infertility, synthesizing evidence from clinical studies, microbiome research, and treatment outcomes. The authors explored how BV-related dysbiosis contributes to upper genital tract infections and reproductive complications, while evaluating diagnostic challenges and emerging therapeutic strategies.

Who was Reviewed?

The review analyzed data from diverse populations of reproductive-age women, including those with infertility, recurrent BV, or PID. It incorporated findings from studies on vaginal and endometrial microbiota, clinical trials on BV treatments (e.g., antibiotics, probiotics), and research on immune and inflammatory responses linked to infertility.

What were the most Important Findings?

BV, characterized by reduced Lactobacillus dominance and overgrowth of anaerobes like Gardnerella vaginalis and Atopobium vaginae, was strongly associated with tubal infertility (3.3-fold higher prevalence in infertile women) and PID. Major microbial associations (MMA) included elevated levels of proinflammatory cytokines (IL-1β, IL-6, IL-8) in BV-positive women, which disrupt endometrial receptivity. Subclinical PID, often linked to BV, reduced pregnancy likelihood by 40%. Notably, Lactobacillus crispatus probiotics reduced BV recurrence by 15% compared to placebo, while endometrial microbiota dominated by non-lactobacilli correlated with lower IVF success rates. Chronic endometritis (CE), prevalent in 34%–66% of unexplained infertility cases, improved fertility outcomes post-antibiotic treatment, with cured CE showing a 76.3% pregnancy rate versus 20% in persistent cases.

What are the Implications of this Review?

The findings underscore BV as a modifiable risk factor for infertility, emphasizing the need for early screening and treatment to prevent PID and CE. Clinicians should consider Lactobacillus-based probiotics and biofilm-disrupting agents for recurrent BV. For infertility workups, endometrial microbiota analysis and CE testing are critical, particularly in cases of repeated implantation failure. Future research should prioritize longitudinal studies to clarify causal links between BV dysbiosis and infertility, while optimizing personalized therapies targeting the vaginal microbiome.

Bacterial Vaginosis Is Associated with Variation in Dietary Indices

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

A high glycemic load diet increases bacterial vaginosis (BV) risk, while nutrient-rich diets lower it. This study highlights the link between diet quality and vaginal microbiome balance, providing insights for clinicians on how dietary interventions may help prevent BV.

What was Studied?

This study examined how dietary indices, including glycemic load (GL), glycemic index (GI), the Healthy Eating Index (HEI), and the Naturally Nutrient Rich (NNR) score, influence bacterial vaginosis (BV) prevalence, progression, and persistence. Researchers aimed to determine whether overall dietary quality, rather than just individual nutrients, affects vaginal microbiome balance.

Who was Studied?

The study analyzed data from 1,735 nonpregnant women aged 15 – 44, primarily African American (85.5%), recruited from health clinics in Birmingham, Alabama. Researchers assessed annual dietary intake using the Block98 food frequency questionnaire and classified vaginal flora using Nugent Gram-stain criteria.

Most Important Findings

A higher glycemic load significantly increased the risk of BV. For every 10-unit increase in GL, the likelihood of BV progression and persistence rose. High-GL diets, which result in frequent blood sugar spikes, may contribute to vaginal flora imbalances and increased oxidative stress, reducing the body's ability to maintain a protective microbiome.

Conversely, women with higher NNR scores, which reflect greater nutrient density per calorie, had a lower risk of BV. This suggests that diets rich in vitamins, minerals, and fiber may help support a healthy vaginal microbiome. HEI scores above 70, indicating greater adherence to dietary guidelines, were associated with a reduced BV risk, but this association was only borderline significant after adjusting for confounding factors.

Unlike GL, which considers both carbohydrate quality and quantity, glycemic index (GI) showed no clear link to BV. Because GI only measures how quickly food raises blood sugar without accounting for quantity, it may not fully capture how diet affects vaginal health.

Implications of the Study

This study highlights the role of diet quality in vaginal microbiome health. Clinicians should counsel patients on the risks of high-GL diets and emphasize nutrient-dense food choices to lower BV risk. Future research should explore how dietary modifications influence BV outcomes and whether interventions targeting glycemic load can serve as preventive measures.

Biological rhythms in premenstrual syndrome and premenstrual dysphoric disorder: a systematic review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Women with PMS/PMDD experience disrupted biological rhythms, notably lower melatonin, higher nighttime temperature, and poor sleep quality. These circadian disturbances contribute to symptom severity, suggesting chronobiological targets for improved diagnosis and treatment.

What was reviewed?

This paper systematically reviewed the existing literature on biological rhythm disruptions in women with premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD). The focus was on circadian and other biological rhythms, including sleep–wake cycles, melatonin secretion, core body temperature, cortisol, prolactin, and thyroid-stimulating hormone levels. The review synthesized findings from 25 studies that compared women diagnosed with PMS/PMDD to healthy controls, assessing both subjective and objective markers of biological rhythms to clarify their association with premenstrual symptoms and the underlying pathophysiology.

Who was reviewed?

The review analyzed studies published between 1989 and 2022 across multiple countries, involving women aged 18 to 45 diagnosed with PMS or PMDD using standardized criteria, mostly DSM-III-R to DSM-5. The total sample sizes varied widely, with some studies including over 600 participants. Healthy control groups consisted of women without PMS/PMDD or psychiatric disorders. Studies included diverse methodologies such as polysomnography, actimetry, hormonal assays, core body temperature measurements, and subjective sleep quality assessments, enabling comprehensive evaluation of biological rhythms in the premenstrual context.

What were the most important findings?

The review found consistent evidence that women with PMS/PMDD exhibit significant disruptions in biological rhythms compared to healthy controls. Notably, they present with lower nocturnal melatonin levels, elevated nighttime core body temperature, and poorer subjective sleep quality, all indicating altered circadian regulation. While objective sleep parameters and activity rhythms showed mixed or nonsignificant differences, hormonal rhythms such as cortisol and prolactin demonstrated inconsistent findings across studies. These rhythm disturbances likely contribute to the psychological and physiological symptoms experienced during the luteal phase. The review highlights melatonin dysregulation as a potential key factor in PMS/PMDD pathophysiology and calls for further research into circadian-based mechanisms and their therapeutic implications.

What are the greatest implications of this review?

This review highlights the importance of biological rhythm disruptions in PMS and PMDD, positioning circadian dysfunction, especially melatonin alterations, as a promising target for understanding symptom development and designing novel interventions. Clinicians should recognize that sleep complaints and temperature regulation abnormalities in these disorders reflect deeper circadian disturbances rather than isolated symptoms. The review advocates for integrating chronobiological assessments into clinical evaluations and exploring circadian-modulating treatments, such as light therapy or melatonin supplementation, to improve patient outcomes. It also calls for future research to clarify inconsistent findings in hormonal rhythms and to investigate the potential of personalized circadian therapies tailored to premenstrual symptom profiles.

Boric acid vaginal suppositories: a brief review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review discusses the effectiveness of boric acid vaginal suppositories in treating azole-resistant vaginitis, including its benefits and limitations.

What was reviewed?

This was a review of boric acid vaginal suppositories and their applications, particularly in the treatment of recurrent vulvovaginal candidiasis (vaginal infections caused by yeast) and other forms of vaginitis.

Who was reviewed?

The review examined several studies and case reports that evaluated the effectiveness and safety of boric acid vaginal suppositories in treating various types of vaginal infections, particularly those caused by Candida species.

What were the most important findings?

The review highlighted boric acid as a potentially useful treatment for recurrent vulvovaginal candidiasis, especially in cases where infections are resistant to common antifungal treatments, such as azoles. It was found that boric acid could be effective in treating infections caused by azole-resistant strains, including Candida glabrata and non-Candida albicans species. The antifungal activity of boric acid is thought to be due to its ability to disrupt the fungal cell membrane, although the exact mechanism remains unclear. In addition, while boric acid is generally well-tolerated in short-term use, its long-term safety remains uncertain. Studies indicated that it is not recommended as a first-line treatment, especially in uncomplicated cases, but could serve as an alternative for chronic or resistant infections. The review also found that boric acid has a low risk of systemic absorption when used intravaginally, with blood boron levels remaining low and within safe limits after typical treatment courses.

What are the greatest implications of this review?

The review suggests that boric acid could be a valuable option for treating chronic and azole-resistant forms of vaginitis, particularly when other antifungal treatments fail. This has important clinical implications for managing recurrent vulvovaginal candidiasis, as the increasing resistance to conventional antifungals presents a significant challenge. However, clinicians are advised to use boric acid cautiously, particularly in pregnant and lactating women, due to the lack of sufficient safety data. While it may not be suitable as a first-line treatment for all cases of vaginitis, it presents an alternative for more difficult-to-treat infections, especially in immunocompromised patients or those with antibiotic resistance.

Breast cancer but not the menopausal status is associated with small changes of the gut microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study shows that breast cancer, rather than menopausal status, drives subtle gut microbiota changes. Dysbiosis in BC patients included reduced Blautia obeum and Bifidobacterium. Functional impacts, such as downregulated NAD pathways, suggest gut microbiota's potential role in cancer progression.

What Was Studied?

This study investigated the relationship between gut microbiota composition and breast cancer (BC), focusing on the potential impact of menopausal status on microbiota variations. The researchers used shotgun metagenomics to compare the gut microbiota of 88 newly diagnosed BC patients (47 premenopausal and 41 postmenopausal) with 86 cancer-free controls, stratified by menopausal status.

Who Was Studied?

The participants included Polish women divided into two groups: BC patients and controls. The BC group was further divided into premenopausal and postmenopausal subgroups. Fecal samples were collected before systemic cancer treatment, and patients with prior antibiotic use, inflammatory bowel disease, or a history of cancer (for controls) were excluded.

Most Important Findings

The study showed that menopausal status had no significant impact on the overall gut microbiota composition or diversity. However, breast cancer (BC) patients exhibited gut dysbiosis compared to controls. Premenopausal BC patients demonstrated lower abundances of taxa such as Bifidobacterium and Collinsella massiliensis but higher abundances of the genus Gemmiger. In postmenopausal BC patients, taxa such as Blautia obeum, Dorea formicigenerans, and Bacteroides thetaiotaomicron were reduced, while Faecalibacterium prausnitzii showed an overrepresentation, potentially indicating a protective or prognostic role. Functional alterations were minimal, with the NAD salvage pathway downregulated in premenopausal BC patients, possibly affecting DNA repair. Enterotype analysis revealed that Bacteroides-dominated enterotypes were more common in controls, while Prevotella and Alistipes were enriched in BC patients. Additionally, bacterial diversity was notably lower in postmenopausal BC patients compared to controls, emphasizing the role of gut dysbiosis in BC pathology rather than menopausal status.

GroupMicrobial ChangesFunctional Changes
Premenopausal BC PatientsLower abundances: Bifidobacterium, Collinsella massiliensis. Higher abundances: Gemmiger.Downregulation of NAD salvage pathway, possibly affecting DNA repair.
Postmenopausal BC PatientsReduced levels: Blautia obeum, Dorea formicigenerans, Bacteroides thetaiotaomicron. Overrepresentation: Faecalibacterium prausnitzii.Minimal functional alterations.
Controls vs. BC PatientsBacteroides enterotypes prevalent in controls; Prevotella and Alistipes enriched in BC patients.N/A
Postmenopausal BC Patients (Alpha-Diversity)Lower bacterial diversity compared to controls.N/A

Greatest Implications

The study underscores the importance of gut microbiota in BC development, suggesting that dysbiosis may not be directly related to menopausal status but rather to BC pathology itself. These findings have potential diagnostic implications, as machine learning models using gut microbiota profiles demonstrated an ability to distinguish BC patients from controls with high accuracy (AUC > 0.8). The study highlights the need for further research to explore the mechanisms linking microbiota alterations and BC progression, particularly focusing on key taxa like Faecalibacterium prausnitzii and Bifidobacterium, as well as geographic and lifestyle factors influencing microbiota composition.

Breast cancer in postmenopausal women is associated with an altered gut metagenome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

The study revealed altered gut microbiota in postmenopausal breast cancer patients, with enriched inflammation-associated species and depleted butyrate producers. Functional gene changes suggest links to systemic inflammation and metabolic imbalance, providing insights into microbiota's role in cancer progression.

What Was Studied?

This study investigated the differences in the composition and functional capacities of gut microbiota between postmenopausal breast cancer patients and postmenopausal healthy controls. The researchers conducted a comprehensive shotgun metagenomic analysis to assess microbial diversity, taxonomic abundance, functional gene profiles, and potential associations with clinical indices.

Who Was Studied?

The study involved 44 postmenopausal breast cancer patients and 46 postmenopausal healthy controls, as well as 18 premenopausal breast cancer patients and 25 premenopausal healthy controls. All participants were treatment-naive and free from other conditions such as diabetes or inflammatory bowel diseases, which could confound the microbiome analysis.

What Were the Most Important Findings?

The study found significant differences in gut microbial diversity and composition between postmenopausal breast cancer patients and healthy controls. Microbial diversity was higher in breast cancer patients. Forty-five microbial species exhibited significant differences in abundance; 38 species were enriched in breast cancer patients, including Escherichia coli, Klebsiella sp., and Prevotella amnii, while 7 species, such as Eubacterium eligens and Lactobacillus vaginalis, were depleted. Functionally, the gut metagenomes of patients were enriched in genes linked to lipopolysaccharide (LPS) biosynthesis, iron transport, and secretion systems, which may contribute to systemic inflammation and metabolic alterations. Importantly, butyrate-producing bacteria like Roseburia inulinivorans were reduced in patients, potentially affecting anti-inflammatory processes.

What Are the Greatest Implications of This Study?

This study highlights the potential role of gut microbiota in influencing systemic inflammation, estrogen metabolism, and immune regulation in postmenopausal breast cancer. The enrichment of LPS biosynthesis and iron transport genes points to mechanisms that may drive inflammation and tumorigenesis. The depletion of butyrate producers suggests a loss of anti-inflammatory microbiota functions, underscoring the gut microbiota’s importance in maintaining immune homeostasis. These findings suggest that gut microbiota could serve as biomarkers for breast cancer and potential therapeutic targets to mitigate disease progression.

Breast cancer patients from the Midwest region of the United States have reduced levels of short-chain fatty acid-producing gut bacteria

May 20, 2025
  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study identifies gut dysbiosis in breast cancer patients, highlighting reduced SCFA-producing bacteria and altered microbial pathways. Findings suggest microbiome-targeted interventions could aid breast cancer treatment.

What was studied?

This study investigated the gut microbiome composition in breast cancer (BC) patients from the Midwest region of the United States, focusing on its taxonomic composition and functional profiling. Using 16S ribosomal RNA sequencing, the study examined the bacterial microbiome, specifically targeting short-chain fatty acid (SCFA)-producing bacteria. It aimed to identify microbial dysbiosis and its potential role in breast cancer pathobiology, emphasizing regional differences in microbiome signatures.

Who was studied?

The study included 22 breast cancer patients and 19 healthy controls, all recruited from the University of Iowa. Participants were matched by race, body mass index (BMI), and sex. Inclusion criteria required BC patients to have invasive breast cancer, with exclusion criteria such as antibiotic use during sample collection. Healthy controls were similarly screened for factors that might impact gut microbiota, like recent antibiotic or laxative use.

What were the most important findings?

The study identified significant gut microbiome differences between breast cancer patients and healthy controls, particularly in alpha and beta diversity measures. Breast cancer (BC) patients showed evidence of gut dysbiosis, including a decrease in beneficial SCFA-producing bacteria and an enrichment of pro-inflammatory taxa. These alterations suggest a microbiome imbalance that may contribute to inflammation and disease progression. Furthermore, the study highlighted functional disruptions in microbiome pathways, with reduced production of SCFAs such as propionate and acetate, which are essential for maintaining gut health and modulating immune responses. These findings underscore the importance of microbiome-targeted interventions to restore microbial balance and support breast cancer treatment.

FindingBreast Cancer Patients (BC)Healthy Controls (HC)Relevance
SCFA-Producing BacteriaReduced Faecalibacterium prausnitzii, Alistipes, Parabacteroides merdae, Lachnospira pectinoschizaHigher levelsSCFA reduction contributes to inflammation and impaired gut motility.
Pro-Inflammatory BacteriaEnriched Eggerthella lenta, Blautia speciesReduced levelsLinked to inflammation and cancer progression.
Functional PathwaysDecreased SCFA pathways (propionate, acetate)Intact pathwaysDysbiosis may exacerbate systemic inflammation and disrupt gut homeostasis.
Beta Diversity ClusteringSignificant clustering distinct from HCNo significant clusteringIndicates an altered microbiome composition in BC.

What are the greatest implications of this study?

The findings underscore the role of gut microbial dysbiosis in breast cancer, with SCFA-producing bacteria depletion linked to inflammation and cancer pathogenesis. This highlights potential avenues for microbiome-targeted therapies, such as probiotics or dietary interventions, aimed at restoring SCFA production and microbial balance. Moreover, the study emphasizes the need for region-specific microbiome research to tailor interventions effectively.

Causal effects of gut microbiome on endometriosis: a two-sample Mendelian randomization study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This Mendelian randomization study found a causal relationship between gut microbiota and endometriosis. Porphyromonadaceae and Anaerotruncus increased risk, while Clostridiales_vadin_BB60_group, Oxalobacteraceae, Desulfovibrio, Haemophilus, and Holdemania were protective. These findings support microbiome-targeted interventions as a potential treatment strategy for endometriosis.

What was studied?

This study investigated the causal relationship between gut microbiome composition and endometriosis using a two-sample Mendelian randomization (MR) approach. The researchers aimed to determine whether specific gut microbiota taxa have a direct causal effect on endometriosis risk rather than a mere observational association.

Who was studied?

The study utilized genome-wide association study (GWAS) summary statistics from two major datasets to investigate the causal relationship between gut microbiota and endometriosis. Exposure data on the gut microbiome were obtained from the MiBioGen consortium, which included 18,340 individuals across 24 cohorts from multiple countries. Outcome data for endometriosis were sourced from the FinnGen consortium, comprising 13,456 endometriosis cases and 100,663 controls, all of European ancestry.

Key Findings

The study identified five bacterial taxa with a protective effect against endometriosis and two taxa associated with increased risk. Protective taxa included Clostridiales_vadin_BB60_group, Oxalobacteraceae, Desulfovibrio, Haemophilus, and Holdemania, all of which exhibited odds ratios (OR) below 1, indicating a reduced likelihood of endometriosis in individuals with higher genetic abundance of these bacteria. In contrast, Porphyromonadaceae and Anaerotruncus were associated with increased endometriosis risk, with ORs above 1, suggesting their potential involvement in disease progression. Sensitivity analyses confirmed the robustness of these findings, as no evidence of pleiotropy or heterogeneity was detected, reinforcing the reliability of the causal associations.

Bacterial TaxaP-ValueEffect
Clostridiales_vadin_BB60_group
< 0.01
Protective
Oxalobacteraceae0.014Protective
Desulfovibrio0.046Protective
Haemophilus0.039Protective
Holdemania0.025Protective
Porphyromonadaceae0.027Risk
Anaerotruncus< 0.01Risk

Greatest Implications

The findings of this study provide genetic evidence supporting a causal relationship between gut microbiota and endometriosis, reinforcing previous observational research. This suggests that targeting the gut microbiome through interventions such as probiotics, dietary modifications, or microbiome-targeted interventions (MBTIs) could be a novel approach to managing or preventing endometriosis. The identification of specific bacterial taxa that either increase or decrease endometriosis risk offers a foundation for developing microbiome-targeted interventions tailored to patient needs. Additionally, these results support the estrobolome hypothesis, which proposes that gut microbiota influence estrogen metabolism, potentially contributing to the pathophysiology of endometriosis. By establishing a causal link, this study highlights the importance of gut microbiota in the broader endocrine and inflammatory mechanisms underlying the disease, paving the way for further research into microbiome-based therapeutic strategies.

Causal relationships between gut microbiota and polycystic ovarian syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This Mendelian randomization study identifies a bidirectional causal link between specific gut microbes and PCOS. Bacilli and Burkholderiales increase PCOS risk, while Blautia and Cyanobacteria show protective effects. Findings reveal gut microbial signatures that may be targeted for PCOS treatment.

What Was Studied?

This study investigated the causal relationship between gut microbiota and polycystic ovary syndrome (PCOS) using a bidirectional two-sample Mendelian randomization (MR) design. Researchers used genome-wide association study (GWAS) summary statistics from the MiBioGen consortium to represent gut microbiota composition and the FinnGen cohort to define PCOS cases and controls. The goal was to determine whether specific bacterial taxa causally influence the risk of developing PCOS and, conversely, whether PCOS causally alters gut microbial abundance. By leveraging genetic variants as instrumental variables, this study minimized biases commonly seen in observational research, including reverse causation and confounding. The analytical approach included several MR methods and sensitivity analyses to verify the robustness of findings.

Who Was Studied?

The study utilized summary-level genetic data rather than individual-level clinical cohorts. Gut microbiota data were derived from 18,340 individuals across 24 cohorts, primarily of European ancestry, through the MiBioGen meta-analysis, which identified 196 microbial taxa with sufficient abundance. PCOS outcome data came from 118,870 participants in the FinnGen cohort, including 642 clinically diagnosed PCOS cases and 118,228 controls. The PCOS diagnosis was based on ICD codes in hospital registries and aligned with the Rotterdam criteria. Importantly, both datasets involved participants of European descent, ensuring consistency for MR assumptions and reducing potential population stratification biases.

What Were the Most Important Findings?

The most significant finding was that specific gut microbiota taxa demonstrated a clear causal relationship with PCOS. Notably, the taxa Bacilli, Burkholderiales, and Lachnospiraceae showed a positive causal association with PCOS risk. In contrast, taxa such as Blautia, Bilophila, Cyanobacteria, Alphaproteobacteria, Holdemania, and Candidatus Soleaferrea exhibited a protective causal relationship. Among these, Blautia and Cyanobacteria retained their protective associations across all MR methods used, including the robust cML-MA approach. The study also found evidence for bidirectional causality in two taxa: Alphaproteobacteria and Lachnospiraceae. PCOS reduced the abundance of these microbes, while alterations in these microbes also contributed to PCOS risk.

These results are highly relevant to microbiome signatures, as they move beyond correlation to genetic causation. The major microbial associations (MMAs) emerging from this study establish Blautia and Bilophila as potential protective taxa and Burkholderiales and Bacilli as risk enhancers for PCOS. These taxa interact with PCOS through mechanisms related to insulin resistance, chronic inflammation, bile acid metabolism, SCFA production, and hormonal modulation, including the gut–brain axis. The presence of bidirectional effects particularly strengthens the hypothesis that gut microbiota and PCOS are engaged in a feedback loop that can worsen or potentially mitigate the disease course depending on microbial composition.

What Are the Implications of This Study?

This study provides the strongest evidence to date for a genetically mediated, bidirectional causal link between gut microbiota and PCOS. It significantly advances the field by identifying specific microbial taxa that not only influence PCOS risk but are also altered by the disease itself. For clinicians, these findings suggest that targeting certain microbial taxa could represent a therapeutic avenue for PCOS prevention or management. Modulating the abundance of Blautia, Bilophila, and Alphaproteobacteria through diet, prebiotics, probiotics, or even microbiota transplantation could potentially alter disease risk or symptom severity. The study’s use of MR methodology also sets a benchmark for establishing causal inference in microbiome research, paving the way for precision interventions rooted in genetic evidence. This work redefines PCOS not only as an endocrine-metabolic disorder but also as one with a microbial signature that may be clinically actionable.

Characteristics of gut microbiota in patients with asthenozoospermia: a Chinese pilot study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The study explored gut microbiota differences in men with asthenozoospermia (AS) compared to healthy controls, finding significant disparities in microbial diversity and composition. Key bacteria correlated negatively with sperm motility, suggesting gut microbiota may influence male fertility, potentially offering new targets for diagnosing and treating AS.

What was studied?

The study investigated the characteristics of gut microbiota in patients with asthenozoospermia (AS), a common cause of male infertility characterized by reduced sperm motility. It aimed to identify differences in gut microbiota composition between AS patients and healthy controls and explore the potential links between gut microbiota and the development of AS.

 

Who was studied?

The study involved 108 men, including 60 patients diagnosed with isolated AS and 48 healthy men serving as controls. These participants were recruited from the outpatient department of Tianjin Medical University General Hospital. Men with AS were compared against healthy controls with normal semen parameters.

 

What were the most important findings?

Diversity and Composition: AS patients exhibited significantly lower alpha diversity in their gut microbiota and distinct beta diversity compared to healthy controls. Specific bacteria such as Escherichia_Shigella and various members of the Prevotellaceae family were notably different between the groups.

Correlation with Sperm Mobility: Certain key genera identified were negatively correlated with sperm motility parameters in AS patients, suggesting a potential impact of these microbial communities on sperm function.

Predicted Metabolic Pathways: The study predicted significant differences in metabolic pathways such as steroid biosynthesis and meiosis, potentially influencing the physiological processes related to sperm motility and overall reproductive health.

 

What are the greatest implications of this study?

The findings suggest that gut microbiota dysbiosis might be linked to the development of asthenozoospermia, possibly through effects on systemic inflammation, nutrient synthesis, and hormonal balance. This could lead to new diagnostic and therapeutic approaches, where modulation of gut microbiota could become part of treating or managing male infertility. The study supports the concept of the gut-testis axis, highlighting the broader systemic impact of gut health. Future research could focus on confirming these findings and exploring gut microbiota modification as a therapeutic strategy for AS.

Characteristics of the Gut Microbiota in Japanese Patients with Premenstrual Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This study explores the gut microbiota in Japanese women with Premenstrual Syndrome (PMS), finding significant differences in microbial composition and highlighting the potential role of Collinsella in PMS pathophysiology.

What was studied?

This study investigated the characteristics of the gut microbiota in Japanese women with Premenstrual Syndrome (PMS), aiming to compare the microbial composition between PMS-affected individuals and healthy controls. By analyzing stool samples using 16S rRNA gene sequencing, the study examined the diversity of the microbiome and sought to identify microbial genera potentially associated with the severity of PMS symptoms.

Who was studied?

The study involved a total of 168 women, including 24 diagnosed with PMS and 144 healthy controls. The participants were between the ages of 24 and 49, with the control group selected to match the PMS group in terms of age and other relevant demographic factors. Women with other health conditions, those who had recently used antibiotics, or those with major dietary restrictions were excluded from the study. The aim was to understand how differences in the gut microbiome between the two groups might correlate with PMS symptoms.

What were the most important findings?

The study found that the gut microbiota of women with PMS differed significantly from that of healthy controls. The PMS group exhibited higher α-diversity, which was assessed using the Simpson index, indicating a more varied microbial community. When comparing the overall microbial composition between the groups, there were notable differences in β-diversity and statistical tests. The study also identified several microbial genera that were more abundant in women with PMS. Notably, Collinsella was found to be significantly more abundant in the PMS group, particularly among women aged 30–40 years. This genus was found to be 4.5 times more abundant in the PMS group compared to the controls, and its presence correlated with the severity of PMS symptoms. Bifidobacterium and Blautia, known for their roles in carbohydrate metabolism, were also more prevalent in the PMS group, potentially linking microbiome composition with metabolic processes that could influence PMS symptoms.

What are the greatest implications of this study?

The findings from this study suggest that the gut microbiota may play a significant role in the development and severity of PMS. The identification of Collinsella as a key player in the microbial composition of PMS-affected individuals opens new potential avenues for intervention. The findings imply that microbiome modulation, particularly through dietary adjustments or prebiotics targeting Collinsella, could offer a therapeutic strategy for managing PMS symptoms. The study also emphasizes the importance of further research to confirm these associations and explore the underlying mechanisms connecting microbiome imbalances with PMS.

Characteristics of the gut microbiota in women with premenstrual symptoms: A cross-sectional study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This study explores the association between gut microbiota composition and the severity of premenstrual symptoms, suggesting a potential role for microbiome-based therapies in managing PMS.

What was studied?

The study investigated the characteristics of the gut microbiota in women experiencing premenstrual symptoms, focusing on the potential association between gut microbial composition and the severity of these symptoms. The research aimed to identify microbial differences between women with premenstrual disorders (PMDs) and healthy controls, and to explore how these differences might correlate with the severity of premenstrual syndrome (PMS) symptoms.

Who was studied?

The study involved 56 women, including 27 women with self-reported premenstrual symptoms and 29 women without significant symptoms. Among the 27 women reporting PMS, 21 were further identified as having premenstrual disorders (PMDs) due to the significant interference of their symptoms with their social life. The control group consisted of 22 women with no significant premenstrual symptoms. These participants were matched by age and other demographic factors.

What were the most important findings?

The study found that there were notable differences in the gut microbiota between women experiencing premenstrual disorders (PMDs) and healthy controls. At the phylum level, the abundance of Bacteroidetes was significantly lower in the PMDs group. At the genus level, several bacteria were found to be either more or less prevalent in the PMDs group. Specifically, the PMDs group had lower levels of beneficial bacteria such as Butyricicoccus, Megasphaera, and Parabacteroides, while Anaerotaenia was more prevalent in this group. However, after applying false discovery rate correction, these differences were no longer statistically significant. The abundance of certain microbes, such as Anaerotaenia, correlated positively with the severity of PMS as measured by the Premenstrual Symptoms Questionnaire (PSQ), while Parabacteroides and Megasphaera were negatively associated with symptom severity.

These findings suggest that the gut microbiota may play a role in the pathophysiology of premenstrual disorders, potentially influencing the severity of symptoms. However, the study also noted that there were no significant differences in inflammatory markers (such as CRP, LBP, and sCD14) between the PMDs and control groups, which means that the microbiota’s role might not be directly related to inflammation but may involve other mechanisms such as gut-brain communication.

What are the greatest implications of this study?

This study provides preliminary evidence that the gut microbiota could be linked to the severity of premenstrual symptoms, suggesting that microbiome-based interventions might be a potential therapeutic strategy for managing PMS and PMDs. However, due to the cross-sectional nature of the study, it is not possible to establish causality between microbiota characteristics and the severity of premenstrual symptoms. The study's findings highlight the need for future longitudinal studies to confirm these associations and determine the potential therapeutic role of gut microbiota modulation. Furthermore, the identification of specific microbial imbalances could help develop microbiome-based biomarkers for the diagnosis and treatment of PMS and PMDs, providing a more personalized approach to care.

Characteristics of the gut microbiota in women with premenstrual symptoms: A cross-sectional study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This study identified specific gut microbiota alterations, including reduced butyrate- and GABA-producing bacteria, associated with premenstrual disorder severity, offering new insights into PMD pathophysiology and potential microbiome-targeted treatments.

What was studied?

This cross-sectional pilot study examined the gut microbiota characteristics in women experiencing premenstrual disorders (PMDs) compared to healthy controls, aiming to uncover microbial associations with the severity of premenstrual symptoms and to evaluate inflammatory markers indicative of bacterial translocation.

Who was studied?

The study involved 43 Japanese women aged 20 to 45 years, with 21 women experiencing PMDs severe enough to disrupt social functioning and 22 controls without significant premenstrual symptoms, all selected to exclude confounding factors like recent medication use, neuropsychiatric disorders, and gastrointestinal diseases.

What were the most important findings?

The study revealed that although overall gut microbial diversity did not differ significantly, women with PMDs exhibited lower levels of the Bacteroidetes phylum and reduced abundance of butyrate-producing genera such as Butyricicoccus and Megasphaera, alongside decreased Parabacteroides, a GABA-related genus, while Anaerotaenia was elevated; these microbial shifts correlated with symptom severity, but inflammatory markers linked to endotoxemia showed no group differences, suggesting unique microbiome alterations in PMDs distinct from major depressive disorder.

What are the greatest implications of this study?

These findings suggest that specific gut microbiota alterations, particularly reductions in beneficial butyrate- and GABA-producing bacteria, may underlie premenstrual symptom severity through the gut-brain axis, highlighting potential microbiome-based biomarkers and therapeutic targets for PMDs, and warranting further longitudinal and intervention research to establish causality and clinical applications.

Characterization of the gut microbiota in polycystic ovary syndrome with dyslipidemia

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study explores the gut microbiota in women with PCOS and dyslipidemia, revealing significant microbial imbalances associated with lipid metabolism and hormonal dysregulation, offering potential targets for future therapies.

What Was Studied?

This research focused on the gut microbiota in patients with Polycystic Ovary Syndrome (PCOS), particularly those with dyslipidemia (PCOS.D). The study aimed to identify and compare gut microbial compositions in individuals with PCOS, those with PCOS and dyslipidemia, and healthy controls. It also explored how these microbiota imbalances correlate with metabolic conditions such as lipid profiles and sex hormone levels.

Who Was Studied?

The study enrolled 52 participants, including 18 patients diagnosed with PCOS, 18 with PCOS and dyslipidemia (PCOS.D), and 16 healthy women serving as controls. These individuals were recruited from the First Affiliated Hospital of Anhui Medical University, ensuring the participants met the criteria for each group based on their clinical and metabolic parameters.

What Were the Most Important Findings?

The key findings of this study were related to the gut microbiota dysbiosis observed in patients with PCOS. The results showed significant microbial differences compared to both PCOS-only patients and healthy controls. Specifically, the PCOS.D group exhibited a higher abundance of Clostridium while Faecalibacterium and Holdemanella were notably lower in abundance. This imbalance was linked to lipid metabolism dysfunctions, such as elevated triglycerides (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) levels. In contrast, Faecalibacterium, a genus producing butyrate, was negatively correlated with TG and TC levels, suggesting its potential role in regulating lipid metabolism. Additionally, Pseudomonas, a genus associated with steroid metabolism, was negatively correlated with luteinizing hormone (LH) and LDL-C levels, further suggesting a complex interaction between gut microbiota and metabolic health in PCOS patients.

What Are the Implications of This Study?

The findings suggest that gut microbiota imbalances, especially in the abundance of Faecalibacterium and Clostridium, may play a crucial role in the pathogenesis of dyslipidemia in PCOS patients. This opens the possibility of targeting the gut microbiota for therapeutic interventions, particularly for managing lipid metabolism and improving metabolic health in these individuals. The study also highlights the need to explore further the mechanisms by which specific microbial genera influence lipid profiles and sex hormone levels, potentially guiding future precision medicine approaches for PCOS treatment.

Characterization of Vaginal Microbiota in Women With Recurrent Spontaneous Abortion That Can Be Modified by Drug Treatment

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study explores the link between vaginal microbiota and recurrent spontaneous abortion (RSA), showing that drug treatment, including metformin combined with aspirin, can alter microbiota composition and potentially improve pregnancy outcomes by increasing Lactobacillus abundance.

What was Studied?

This study examined the role of vaginal microbiota in women experiencing recurrent spontaneous abortion (RSA). The researchers aimed to characterize the vaginal microbiota in these women and explore how drug treatments could influence its composition. The study used 16S rRNA gene sequencing to analyze microbial communities, identifying key differences between women with RSA and healthy controls. The study also evaluated the effects of metformin, metformin plus aspirin, and other drug treatments on the vaginal microbiota.

Who was Studied?

The study included women who had experienced recurrent spontaneous abortions (RSA) and healthy controls who had live births without any history of spontaneous abortion. Women with RSA were divided into different groups based on their treatment regimen: untreated, metformin-treated, metformin plus aspirin-treated, and other drug-treated groups. Vaginal samples were collected and analyzed for microbial diversity and community structure using 16S rRNA gene sequencing.

What were the most Important Findings?

The most significant finding of this study was that the vaginal microbiota of women with RSA exhibited reduced microbial richness compared to healthy women. Specifically, the study found that the vaginal microbiota of RSA patients had higher abundances of certain genera such as Megasphaera and Sneathia sanguinegens, both of which have been previously associated with adverse pregnancy outcomes, including spontaneous abortion and preterm birth. Furthermore, the study revealed that the composition of the vaginal microbiota in RSA patients was significantly altered after drug treatment. Metformin alone or in combination with aspirin led to a marked increase in the abundance of Lactobacillus species, which are considered protective against infections and important for maintaining vaginal health. This shift in microbiota composition could be beneficial in preventing RSA and improving pregnancy outcomes.

What are the Implications of this Study?

This study highlights the potential for modifying the vaginal microbiome as a therapeutic strategy for women with recurrent spontaneous abortion. By treating RSA patients with metformin combined with aspirin, the researchers significantly increased the abundance of Lactobacillus species, potentially restoring a more balanced and protective vaginal microbiota.

The identification of specific microbial taxa such as Sneathia sanguinegens and Megasphaera opens new possibilities for targeted interventions. These microbial associations could guide future microbiome-based therapies aimed at improving pregnancy outcomes. Furthermore, the study calls for more research to better understand how vaginal microbiota composition influences pregnancy outcomes. Modifying the vaginal microbiota could become an important aspect of enhancing reproductive health and preventing recurrent spontaneous abortions in affected women.

Circulating bile acid profile characteristics in PCOS patients and the role of bile acids in predicting the pathogenesis of PCOS

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study identified significant bile acid profile changes in PCOS patients and highlighted CDCA and LCA as promising biomarkers, especially when combined with testosterone. The results suggest a bile acid–androgen–microbiome axis in PCOS pathophysiology.

What was studied?

This original research investigated the alterations in circulating bile acid (BA) profiles in women with polycystic ovary syndrome (PCOS) and evaluated the potential of specific bile acid metabolites as biomarkers for predicting PCOS pathogenesis. The study analyzed serum samples from 408 women diagnosed with PCOS and 204 healthy controls matched for age and BMI. The team quantified 15 bile acid fractions using liquid chromatography–tandem mass spectrometry (LC-MS/MS), aiming to discern significant differences in bile acid composition between the two groups. The researchers further assessed the correlation of these bile acids with metabolic parameters, especially glucose metabolism and hyperandrogenism, and examined their diagnostic potential using multivariate statistical models such as OPLS-DA and XGBoost.

Who was studied?

The study included 408 women with PCOS diagnosed according to the Rotterdam 2003 criteria and 204 age- and BMI-matched non-PCOS controls. All participants were between 18–45 years old, with exclusions applied for conditions or medications that could affect bile acid metabolism or mimic PCOS symptoms. Comprehensive clinical, hormonal, metabolic, and imaging assessments were conducted to ensure accurate phenotyping. Importantly, the PCOS cohort was further divided into subgroups based on glucose tolerance (normal vs. impaired) and androgen status (hyperandrogenic vs. non-hyperandrogenic) to explore how bile acid profiles vary across PCOS phenotypes.

What were the most important findings?

The study identified significant alterations in circulating bile acid profiles in women with PCOS, particularly elevated levels of primary (CDCA) and secondary (LCA, DCA) unconjugated bile acids. Among 15 measured metabolites, five, including CDCA and LCA, were significantly different in PCOS patients and showed diagnostic potential. CDCA emerged as the most discriminatory metabolite. Notably, DCA correlated with insulin secretion markers such as fasting and postprandial insulin, though this relationship weakened after adjusting for testosterone, suggesting androgen modulation of these effects. Combined analysis of CDCA, LCA, and testosterone improved PCOS prediction over testosterone alone. These findings highlight a possible interaction between bile acid metabolism, gut microbiota (especially with DCA), and androgen excess in PCOS pathophysiology.

What are the greatest implications of this study?

This study underscores the potential of specific bile acids, particularly CDCA and LCA, as novel biomarkers for PCOS diagnosis and stratification, especially when used in combination with serum testosterone. It reveals that PCOS pathogenesis involves distinct alterations in bile acid metabolism, possibly through activation of alternative synthesis pathways and interaction with gut microbiota. The research contributes to a growing body of evidence supporting the integration of metabolic and microbial markers into PCOS diagnostics. Clinically, the findings advocate for more nuanced, biochemically informed approaches to PCOS diagnosis beyond current criteria, which do not capture metabolic and microbial heterogeneity. Future research should validate these biomarkers longitudinally and explore therapeutic interventions targeting bile acid pathways or microbiome modulation.

Clinicians’ use of Intravaginal Boric Acid Maintenance Therapy for Recurrent Vulvovaginal Candidiasis and Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study evaluates the use of intravaginal boric acid for recurrent vulvovaginal candidiasis and bacterial vaginosis, highlighting its long-term tolerability, high patient satisfaction, and rare side effects.

What was Studied?

This study examined the use of intravaginal boric acid (BA) maintenance therapy in women with recurrent vulvovaginal candidiasis (rVVC) and recurrent bacterial vaginosis (rBV). The researchers performed a retrospective chart review to evaluate clinicians' approaches to prescribing BA for these conditions, focusing on dosage, duration of use, patient satisfaction, and side effects. The study aimed to assess the effectiveness, tolerability, and satisfaction of long-term BA therapy in real-world clinical settings.

Who was Studied?

The study reviewed the medical records of 78 patients from a Johns Hopkins University-affiliated outpatient gynecology clinic. These patients were prescribed intravaginal BA for either rVVC, rBV, or both conditions. The patients were selected based on specific criteria, including multiple visits where BA usage was documented, and those who were prescribed a long-term BA regimen (more than a month). Patients were excluded if there was insufficient documentation regarding the initiation or duration of BA use.

What were the Most Important Findings?

The study revealed that maintenance therapy with intravaginal boric acid was commonly prescribed for rVVC and rBV, with an average duration of use estimated at 13.3 months. A significant portion of patients (37.2%) used BA for a year or more, with some patients continuing therapy for more than three years. The treatment regimen typically included a 7-14 day induction phase with BA, followed by a maintenance phase where patients used 300mg or 600mg of BA 2-3 times per week.

Despite the lack of long-term safety data, the study found high patient satisfaction with BA therapy (76.9%), though a small number of patients (16.7%) were dissatisfied, typically due to continued or worsening symptoms. The study also indicated that patients with rVVC were more likely to receive BA as part of an antifungal induction regimen, while patients with rBV were often prescribed antibiotics in addition to BA. Side effects were rare, with a few patients reporting vaginal irritation or leaking, but these effects were generally manageable.

What are the Implications of this Study?

This study provides real-world evidence supporting the use of intravaginal boric acid as a long-term treatment for recurrent vulvovaginal candidiasis and bacterial vaginosis. Despite the absence of large-scale prospective studies, the findings suggest that BA is well-tolerated over extended periods and that it may be an effective option for women with azole-resistant infections. This study's insights into patient satisfaction, side effects, and clinical practice could inform future treatment guidelines and clinical trials for rVVC and rBV. However, more robust, prospective studies are needed to confirm the efficacy and long-term safety of BA maintenance therapy and to compare it with other available treatments.

Coincidence of uterine malformations and endometriosis: a clinically relevant problem?

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This retrospective study analyzed 279 patients with uterine malformations and found a high prevalence of endometriosis (74.9%), especially among those with non-obstructive malformations. The findings suggest that endometriosis should be proactively investigated in patients with any type of uterine malformation.

What was studied?

The retrospective study explored the prevalence of endometriosis in patients with uterine malformations. It specifically investigated the relationship between different types of uterine malformations (obstructive, non-obstructive, and various classifications under the American Fertility Society (AFS)) and the incidence of endometriosis, which is characterized by the presence of endometrial tissue outside the uterus. The study aimed to determine if obstructive malformations, which could potentially lead to increased retrograde menstruation, have a higher coincidence with endometriosis compared to non-obstructive ones.

 

Who was studied?

The subjects of the study were patients with uterine malformations who were admitted to the hospital between December 1, 2014, and November 30, 2019. A total of 279 cases were analyzed after excluding certain cases where no laparoscopy was performed. The study group comprised a diverse array of uterine malformations, including non-obstructive malformations, obstructive malformations, and cases of uterine agenesia/hypoplasia.

 

What were the most important findings?

The study revealed a high prevalence of endometriosis among patients with uterine malformations, particularly in those with obstructive malformations. Key findings include:

Key Findings Details
Overall Prevalence of Endometriosis 74.9% of the patients had histologically confirmed endometriosis, which included peritoneal, ovarian, and deep infiltrating endometriosis.
Prevalence in Obstructive Uterine Malformations A particularly high correlation with endometriosis was observed in obstructive uterine malformations, with an incidence of 87.5% in women who had an obstructive malformation with an active endometrium.
Prevalence in Septate Uterus High rates of endometriosis were also found in cases of septate uterus, a common uterine malformation, with an overall prevalence of 77.1%.

What are the greatest implications of this study?

The findings suggest significant clinical implications for the management of patients with uterine malformations, particularly regarding the diagnosis and treatment of endometriosis. The implications include:

Diagnostic Approach: There should be a high index of suspicion for endometriosis in patients presenting with uterine malformations, especially those with obstructive types. This could lead to more targeted screening and earlier diagnosis.

Integrated Treatment: For patients with both endometriosis and uterine malformations, comprehensive treatment plans that address both conditions simultaneously might improve clinical outcomes.

Fertility Management: Since both endometriosis and uterine malformations can impact fertility, understanding the link between these conditions is crucial for offering appropriate fertility advice and interventions.

Surgical Considerations: The study supports the use of combined diagnostic procedures, such as laparoscopy and hysteroscopy, in the evaluation of such patients to ensure thorough assessment and treatment planning.

 

Conclusion

Overall, the study underscores the need for specific attention to endometriosis in the presence of uterine malformations to enhance diagnostic accuracy and improve therapeutic strategies, which could ultimately enhance reproductive outcomes and quality of life in affected women.

Combatting antibiotic resistance in Gardnerella vaginalis: A comparative in silico investigation for drug target identification

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study identifies phospho-2-dehydro-3-deoxyheptonate aldolase as a drug target in Gardnerella vaginalis and highlights five FDA-approved compounds as potential treatments for bacterial vaginosis.

Who was Studied?

This study focused on combating antibiotic resistance in Gardnerella vaginalis, a bacterium frequently associated with bacterial vaginosis (BV). The researchers employed an in silico approach to identify potential drug targets and therapeutic strategies. By utilizing subtractive genomics and comparative genomics, they analyzed the G. vaginalis proteome to find unique proteins crucial for bacterial survival and virulence, which could serve as drug targets. The study further explored FDA-approved compounds using virtual screening techniques to identify potential inhibitors of these targets. Additionally, the study performed detailed protein structural modeling, docking, and ADMET profiling of the shortlisted compounds to assess their suitability for therapeutic use.

Who was Studied?

The study focused on Gardnerella vaginalis, which is often linked to bacterial vaginosis. It did not study individuals directly but rather the bacterial proteome, applying computational methods to identify druggable targets within the pathogen. The proteome of G. vaginalis was retrieved from the UniProt database, and various bioinformatics tools were used to identify potential drug targets based on their essentiality, uniqueness, and non-homology to human proteins.

What are the Most Important Findings?

The study identified phospho-2-dehydro-3-deoxyheptonate aldolase (PDA) as a promising drug target for G. vaginalis. This enzyme plays a critical role in the shikimate pathway, which is essential for producing aromatic amino acids and other metabolites. The identification of this enzyme is significant because it is non-homologous to human proteins, reducing the risk of off-target effects. The researchers found five compounds from the DrugBank database that could inhibit PDA effectively. The compounds demonstrated strong binding affinities to the target protein, suggesting their potential as effective treatments. Virtual screening results showed that these compounds have favorable pharmacokinetic profiles, including good bioavailability, and do not inhibit key enzymes responsible for drug metabolism, making them promising candidates for repurposing to treat G. vaginalis infections.

What are the Implications of the Study?

This study presents a significant step toward identifying new therapeutic strategies for treating G. vaginalis infections, particularly in the face of rising antibiotic resistance. The identification of PDA as a drug target opens up new possibilities for developing treatments that are more effective than current antibiotics, which often face resistance issues. The use of in silico methods, such as molecular docking and virtual screening, allows for the rapid identification of promising drug candidates, saving time and resources compared to traditional experimental approaches. By repurposing FDA-approved compounds, the study suggests a faster route to clinical application, potentially providing affordable treatments for bacterial vaginosis. This approach also highlights the potential of computational tools in addressing antibiotic resistance and discovering new uses for existing drugs.

Combination therapy of curcumin and fecal microbiota transplant: Potential treatment of polycystic ovarian syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review proposes a combination of fecal microbiota transplantation (FMT) and curcumin as a potential treatment for Polycystic Ovary Syndrome (PCOS). The hypothesis suggests these therapies could restore gut eubiosis, reduce inflammation, and improve metabolic dysfunction in PCOS patients.

What was studied?

This study proposed a combination therapy of curcumin and fecal microbiota transplantation (FMT) as a potential treatment for Polycystic Ovary Syndrome (PCOS). PCOS is a common endocrine disorder affecting women of reproductive age, characterized by symptoms like anovulation, hyperandrogenism, hirsutism, and infertility. The study explored the role of gut dysbiosis in the development and exacerbation of PCOS and hypothesized that combining FMT and curcumin could help restore gut eubiosis, thereby alleviating the symptoms of PCOS. FMT is considered a holistic therapeutic approach, as it targets not only the gut microbiota but also the virome, fungome, and other microbiota domains. Curcumin, known for its anti-inflammatory and antioxidant properties, was suggested to complement FMT by maintaining a healthy microbiome and reducing the chronic inflammation typical in PCOS.

Who was studied?

This was a review and hypothesis-driven study, not a clinical trial. The study reviewed preclinical data and existing literature on the role of FMT and curcumin in PCOS treatment. It focused on understanding the influence of gut dysbiosis on PCOS pathophysiology, and while no direct human participants were involved in this review, the authors proposed a combination therapy for future clinical trials. The target population discussed in the review is women diagnosed with PCOS, as well as those affected by the symptoms and metabolic abnormalities associated with the condition.

What were the most important findings?

The review highlighted significant evidence linking gut dysbiosis to PCOS, suggesting that alterations in the gut microbiota contribute to the development and progression of the syndrome. Studies indicated that FMT has been effective in addressing gut dysbiosis, leading to improved metabolic profiles and hormonal balance in PCOS patients. However, the need for repetitive FMT treatments and the associated challenges with its standardization and patient acceptance were noted as limitations. To overcome these issues, the combination of FMT with curcumin was proposed. Curcumin’s anti-inflammatory and antioxidant properties are well-documented, and it was shown to reduce oxidative stress and improve insulin resistance, key features of PCOS. The combination of these therapies is hypothesized to restore microbiome balance more sustainably, avoiding the need for repeated FMT sessions.

From a microbiome perspective, curcumin’s ability to modulate the gut microbiota is also relevant. It enhances microbial consumption of nutrients like sugars and polyphenols, which could have positive effects on gut health and potentially improve PCOS symptoms. The review suggests that curcumin can support the beneficial effects of FMT by preventing relapse of dysbiosis and enhancing long-term treatment outcomes.

What are the greatest implications of this study?

The greatest implication of this hypothesis is the potential for a novel, sustainable treatment for PCOS that combines the strengths of FMT and curcumin. Given the role of gut dysbiosis in PCOS pathophysiology, this combination therapy could offer a more holistic approach to treatment. It could not only help in managing the metabolic and endocrine features of PCOS but also reduce the risks associated with long-term pharmacological treatments. Additionally, this approach may help in restoring the gut microbiota, which is increasingly recognized as playing a significant role in the regulation of inflammation and insulin resistance. By leveraging both FMT and curcumin, clinicians might be able to offer a more effective, lower-risk alternative to conventional PCOS treatments that focus mainly on symptomatic relief.

Comorbid Premenstrual Dysphoric Disorder in Women with Bipolar Disorder: Management Challenges

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Management of comorbid PMDD and bipolar disorder requires mood stabilization first, cautious use of antidepressants, hormonal therapy, and adjunctive psychotherapy. Evidence is limited, highlighting the need for personalized, multidisciplinary care and further research.

What was studied?

This review analyzed existing literature on the management and treatment challenges of comorbid Premenstrual Dysphoric Disorder (PMDD) in women diagnosed with Bipolar Disorder (BD). It focused on identifying pharmacological and non-pharmacological strategies to manage the complex interaction of mood symptoms in this dual-diagnosed population.

Who was studied?

The review included data from a small number of case reports and case series comprising six women with comorbid BD and PMDD. Additionally, it integrated broader findings from separate research on BD and PMDD to inform treatment hypotheses for this specific comorbid group.

What were the most important findings?

The study emphasized that optimal management requires first stabilizing bipolar symptoms using mood stabilizers such as lithium and lamotrigine, followed by targeted treatment of PMDD symptoms, typically through estroprogestins during euthymic BD phases. Antidepressants may be cautiously used during depressive episodes but carry a risk of inducing manic switches. Atypical antipsychotics may help manage manic episodes and some PMDD symptoms. Psychotherapeutic interventions like cognitive behavioral therapy (CBT), including internet-based delivery, show promise as adjuncts, though evidence specific to comorbid BD/PMDD is limited. Alternative treatments such as lifestyle modifications, supplements (calcium, vitamin B6), and mind-body therapies have been reported anecdotally. The complexity of comorbid BD/PMDD necessitates individualized, multimodal treatment approaches, yet randomized controlled trials (RCTs) in this population remain scarce, highlighting a critical gap in evidence.

What are the greatest implications of this study?

This review underscores the clinical challenges of managing comorbid BD and PMDD, a condition marked by more severe mood instability and poorer outcomes than either disorder alone. It advocates for careful mood stabilization before addressing PMDD symptoms to prevent exacerbations and cautions regarding antidepressant use in BD due to manic risk. The limited but encouraging data on hormonal treatments and psychotherapy suggest these as valuable components of integrated care. The paucity of RCTs specific to comorbid BD/PMDD calls for urgent research to establish evidence-based protocols. Clinicians must adopt tailored, multidisciplinary strategies combining pharmacological, psychological, and lifestyle interventions to improve quality of life in this vulnerable population.

Comparison of Vaginal, Cervical and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

In this prospective cohort, women with stage 3/4 endometriosis exhibited a unique microbial profile, characterized by the absence of Atopobium and increased levels of potentially pathogenic genera like Gardnerella, Streptococcus, Escherichia, Shigella, and Ureoplasma in their vaginal and cervical microbiota.

What was studied?

The study focused on exploring the associations between the microbiota of the vaginal, cervical, and gut regions and stage 3–4 endometriosis in women. This research aimed to uncover potential differences in the microbiome composition between women diagnosed with advanced endometriosis and healthy controls, particularly at the genus level of microbial classification.

 

Who was studied?

The participants included 14 women with histologically proven stage 3–4 endometriosis and 14 healthy controls. These women were carefully selected based on strict criteria to ensure the stability of the microbiota, excluding those who had ever been pregnant or had conditions/medications that could affect the microbiome. All participants belonged to the same ethnicity, and the study managed to maintain a balance between samples collected during different phases of the menstrual cycle in both groups.

 

What were the most important findings?

The study revealed that while the overall composition of the vaginal, cervical, and gut microbiota was similar between women with and without endometriosis, there were notable genus-level differences. Specifically, Atopobium was absent in the endometriosis group’s vaginal and cervical microbiota. Increases in Gardnerella in the cervical microbiota and Escherichia/Shigella in the gut were more common among those with endometriosis. Sensitivity analyses excluding Lactobacillus showed significant increases in Sneathia, Gardnerella, Streptococcus, Escherichia/Shigella, and Ureaplasma, and a decrease in Alloprevotella in the cervical microbiota of the endometriosis group.

 

What are the greatest implications of this study?

The findings suggest a potentially significant association between the composition of the female microbiota and the presence of stage 3–4 endometriosis, particularly regarding the absence and presence of specific microbial genera. These differences might offer insights into the pathophysiology of endometriosis and indicate potential diagnostic markers or therapeutic targets. Specifically, the study raises intriguing questions about the direction of causation between altered microbiota and endometriosis. It suggests the microbiome’s potential as both a screening tool for endometriosis and a therapeutic target, depending on whether changes in the microbiome are a cause or a consequence of the disease. The study also underscores the potential utility of gut microbiome analysis as a predictive tool for surgical decisions, such as the need for bowel resection to treat deep infiltrating endometriosis.

Overall, this research lays the groundwork for further studies to clarify the causal relationships between dysbiosis and endometriosis and to explore the microbiome’s role in the disease’s pathogenesis, diagnosis, and treatment.

Composition and Functional Potential of the Human Mammary Microbiota Prior to and Following Breast Tumor Diagnosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study explores the mammary microbiota's composition and function before and after breast cancer diagnosis, revealing dysbiosis and metabolic shifts as early markers.

What was studied?

This study investigated the composition and functional potential of the human mammary microbiota in healthy breast tissues and those associated with breast cancer development. The researchers focused on tissue samples collected before cancer diagnosis (prediagnostic or PD), as well as adjacent normal (AN) and tumor (T) tissues from breast cancer patients. Using 16S rRNA sequencing and functional metagenomic predictions, they aimed to identify bacterial dysbiosis and metabolic changes associated with breast cancer progression.

Who was studied?

A total of 141 women were included in the study, contributing 159 breast tissue samples. These included 49 samples from healthy individuals (H), 15 from prediagnostic cases (PD), 49 from adjacent normal tissues (AN), and 46 from tumor tissues (T). The prediagnostic samples were obtained from women who later developed breast cancer, allowing researchers to explore early microbial changes.

What were the most important findings?

The study revealed significant bacterial dysbiosis and metabolic reprogramming in PD, AN, and T tissues compared to healthy tissues. Prediagnostic tissues exhibited an intermediary bacterial composition between healthy and cancerous tissues. Shifts in specific bacterial families such as Bacillaceae, Streptococcaceae, and Corynebacteriaceae were detected in PD tissues and were more pronounced in AN and T tissues. Functional analysis revealed reduced bacterial metabolic activities, particularly pathways related to xenobiotics degradation, which could otherwise protect against carcinogenesis. Additionally, altered correlations between host gene expression and microbial functions were observed, highlighting potential early microbial responses to tumor microenvironments.

What are the greatest implications of this study?

This research highlights the mammary microbiota's potential as a critical biomarker for early breast cancer detection and risk stratification by revealing bacterial dysbiosis and metabolic reprogramming in prediagnostic tissues, suggesting microbial changes may precede clinical symptoms or histological abnormalities. The identification of an intermediary microbiota composition in prediagnostic tissues supports the microbiome's role in early cancer development, indicating microbial shifts as potential early drivers or responders to tumorigenesis. A significant reduction in metabolic functions, such as xenobiotic degradation, in cancer-associated tissues implies a diminished microbial ability to detoxify carcinogens, increasing susceptibility to tumor formation. Altered correlations between microbial taxa and host gene expression further suggest dynamic interactions influencing immune responses, inflammation, and cellular proliferation, with positive associations between microbial functions and tumor-related genes pointing to potential mechanistic links to cancer progression.

These findings not only enhance understanding of the microbiota's role in breast cancer but also offer clinical translation opportunities, including the development of non-invasive diagnostic tools based on prediagnostic microbial signatures, microbiome-modulating therapies to target dysbiosis, and therapeutic interventions aimed at restoring protective bacterial functions and reducing cancer risk.

Comprehensive profiles and diagnostic value of menopausal-specific gut microbiota in premenopausal breast cancer

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study identifies menopausal-specific gut microbial markers and functional pathways linked to breast cancer, offering diagnostic potential and insights into prevention.

What was studied?

This study investigated the gut microbiota profiles, diagnostic value, and functional pathways specific to premenopausal breast cancer patients. It aimed to identify unique gut microbial markers distinguishing premenopausal breast cancer patients from postmenopausal patients and age-matched controls. The study also explored functional pathways of gut microbiota linked to breast cancer progression and diagnostic potential.

Who was studied?

The study analyzed 267 participants divided into four groups: premenopausal controls (Pre-C, n=50), premenopausal breast cancer patients (Pre-BC, n=100), postmenopausal controls (Post-C, n=17), and postmenopausal breast cancer patients (Post-BC, n=100). All breast cancer patients were newly diagnosed with stage I–II disease and excluded if they had received treatments or medications affecting gut microbiota before fecal sample collection.

What were the most important findings?

The study highlights significant differences in gut microbial diversity, composition, and functional pathways between premenopausal and postmenopausal breast cancer patients. Premenopausal breast cancer patients showed reduced α-diversity and distinct β-diversity compared to controls, with alterations in specific bacterial taxa linked to inflammation and cancer progression. In contrast, postmenopausal patients exhibited a different microbial profile, including an increase in pathogenic bacteria. Functional pathway analyses revealed steroid-related and oncogenic pathways in premenopausal patients, while postmenopausal patients were associated with chemical carcinogenesis and aldosterone-regulated pathways. The findings emphasize the diagnostic potential of gut microbiota in differentiating breast cancer subtypes and guiding prevention strategies.

AspectPremenopausal Breast CancerPostmenopausal Breast CancerUniversal Markers (Both Types)
α-DiversitySignificantly reduced compared to controlsNo reduction observed compared to postmenopausal controls-
β-DiversityDistinct from controlsDistinct from controls-
Enriched MicrobesBacteroides fragilis, Anaerostipes (linked to inflammation and progression)Proteobacteria, Klebsiella pneumoniae (pathogenic bacteria)Haemophilus parainfluenzae (increased in both)
Reduced MicrobesBifidobacterium spp. (tumor suppressor)Akkermansia muciniphila (beneficial microbe)Faecalibacterium prausnitzii (decreased in both)
Functional PathwaysSteroid-related pathways; Oncogenic pathways (e.g., Notch/Wnt signaling)Chemical carcinogenesis; Aldosterone-regulated pathways-
Diagnostic PotentialStrong microbial markers for distinguishing premenopausal breast cancerStrong microbial markers for distinguishing postmenopausal breast cancer-

What are the greatest implications of this study?

The findings underscore the diagnostic potential of microbial markers for early, non-invasive breast cancer detection based on menopausal status. Identifying these microbial and functional pathways expands the understanding of breast cancer pathogenesis, especially in premenopausal women. Moreover, the study highlights the gut microbiota as a modifiable factor, suggesting potential interventions like probiotics or dietary changes to mitigate breast cancer risk.

Correlation of fecal metabolomics and gut microbiota in mice with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study explores the relationship between fecal metabolomics and gut microbiota in endometriosis (EMS) mice, finding key metabolic changes and decreased microbiota diversity. Significant pathways, like bile acid biosynthesis and ALA metabolism, were identified, suggesting fecal metabolites affected by dysbacteriosis as potential EMS markers.

What was studied?

The study investigated the interaction between fecal metabolomics and gut microbiota in mice with endometriosis (EMS), aiming to identify metabolic changes and microbiota diversity associated with the disease.

 

Who was studied?

Female C57BL/6J mice, utilized to construct an EMS model, were the subjects of this research, allowing for the examination of fecal metabolites and gut microbiota composition.

 

What were the most important findings?

Significant findings included the identification of 156 differential metabolites, decreasing the diversity and abundance of gut microbiota in EMS mice, and involving key metabolic pathways such as bile acid biosynthesis and alpha-linolenic acid (ALA) metabolism. Notably, increased levels of chenodeoxycholic and ursodeoxycholic acids and decreased levels of ALA and 12,13-EOTrE were found in EMS mice feces.

 

What are the greatest implications of this study?

The study suggests that the identified abnormal fecal metabolites, influenced by gut dysbiosis, may be potential markers for diagnosing EMS. This finding opens new avenues for understanding EMS pathogenesis and developing non-invasive diagnostic tools based on fecal metabolite profiles.

Correlation of fecal metabolomics and gut microbiota in mice with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies gut microbiota and metabolomic shifts in endometriosis, with altered bile acid biosynthesis and ALA metabolism. Elevated CDCA and UDCA levels, coupled with microbial changes, suggest potential biomarkers and therapeutic targets for managing inflammation in endometriosis.

What Was Studied?

This study investigated the correlation between fecal metabolomics and gut microbiota in mice with endometriosis. Using a controlled experimental design, researchers constructed an endometriosis (EMS) mouse model with female C57BL/6J mice and analyzed fecal samples through non-targeted metabolomics and 16S rRNA sequencing. The primary objective was to identify differential metabolites and microbial compositions that could serve as biomarkers for endometriosis and provide insight into the metabolic pathways affected by gut dysbiosis in EMS. Functional prediction of the gut microbiota was performed using PICRUSt, and metabolite-microbiota correlations were assessed through Spearman correlation coefficients.

Who Was Studied

The study involved female C57BL/6J mice, which were divided into two groups: an EMS group and a control group. Endometriosis was induced in the EMS group through intraperitoneal injection of endometrial fragments, while the control group received saline injections with adipose tissue. Fecal samples were collected from both groups, processed for liquid chromatography-mass spectrometry (LC-MS), and subjected to 16S rRNA sequencing to map microbial diversity and metabolic profiles. The study aimed to simulate the inflammatory and microbiome-related characteristics of endometriosis in humans by using this established animal model.

What Were the Most Important Findings?

The study identified significant shifts in both fecal metabolomics and gut microbiota composition in mice with endometriosis compared to controls. A total of 156 named differential metabolites were screened, with key changes observed in pathways linked to secondary bile acid biosynthesis and alpha-linolenic acid (ALA) metabolism. Notably, there was an increased abundance of chenodeoxycholic acid (CDCA) and ursodeoxycholic acid (UDCA) alongside a decreased presence of ALA and 12,13-EOTrE in the EMS mice. Microbial diversity was reduced in the EMS group, with a specific loss in Bacteroides and Firmicutes, contrasted by increases in Proteobacteria and Verrucomicrobia. At the genus level, there was a marked increase in Allobaculum, Akkermansia, Parasutterella, and Rikenella, with significant decreases in Lachnospiraceae, Lactobacillus, and Bacteroides. Functional predictions revealed alterations in oxidative phosphorylation, alanine, aspartate, glutamate metabolism, and starch and sucrose metabolism. Importantly, the study identified Sphingobium and Pseudomonas viridiflava as consistently enriched in EMS mice, suggesting their potential role in inflammation and metabolic disruption. The correlation analysis demonstrated strong associations between specific metabolites (like CDCA and ALA) and microbial shifts, indicating a complex interaction between gut dysbiosis and metabolic imbalances in endometriosis.

ParameterFindings in EMS Mice
Metabolomic Changes156 differential metabolites identified, with key changes in secondary bile acid biosynthesis and alpha-linolenic acid (ALA) metabolism.
Increased MetabolitesChenodeoxycholic acid (CDCA) and Ursodeoxycholic acid (UDCA).
Decreased MetabolitesAlpha-linolenic acid (ALA) and 12,13-EOTrE.
Microbial DiversityOverall reduction in diversity; significant losses in Bacteroides and Firmicutes.
Phylum-Level ShiftsProteobacteria and Verrucomicrobia significantly increased in the EMS group.
Genus-Level IncreasesAllobaculum, Akkermansia, Parasutterella, and Rikenella.
Genus-Level DecreasesLachnospiraceae, Lactobacillus, and Bacteroides.
Functional Pathway AlterationsDisrupted oxidative phosphorylation, alanine, aspartate, glutamate metabolism, and starch and sucrose metabolism.
Unique EnrichmentsSphingobium and Pseudomonas viridiflava enriched in EMS mice, indicating roles in inflammation and metabolic disruption.
Metabolite-Microbiota CorrelationsStrong correlations between CDCA, ALA, and microbial shifts, suggesting complex interactions contributing to dysbiosis and inflammation.

What Are the Greatest Implications of This Study?

The findings suggest that endometriosis is associated with profound shifts in gut microbiota and fecal metabolomics, which may contribute to chronic inflammation and disease persistence. The increased levels of chenodeoxycholic acid (CDCA) and ursodeoxycholic acid (UDCA), combined with reductions in ALA, indicate that bile acid metabolism and fatty acid dysregulation are central to the pathogenesis of endometriosis. The enrichment of Allobaculum, Akkermansia, Parasutterella, and Rikenella in the gut microbiota suggests these species could be contributing to local and systemic inflammation, disrupting gut barrier integrity. These microbial and metabolomic signatures could serve as non-invasive biomarkers for diagnosing endometriosis and may offer new therapeutic targets focused on restoring microbial balance and metabolic homeostasis. Furthermore, the study highlights the critical role of gut microbiota in modulating immune responses and metabolic pathways, paving the way for microbiome-targeted treatments in endometriosis management.

Critical Review on Zeolite Clinoptilolite Safety and Medical Applications in vivo

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The review highlights clinoptilolite's detoxifying, immunomodulatory, and antioxidant properties, showing its safety and potential for in vivo use in humans and animals. It can adsorb toxic metals like nickel, support gut health, and modulate immune responses, making it valuable for conditions like systemic nickel allergy syndrome and microbiome imbalances, with promising applications in both human supplements and veterinary care.

What was reviewed?

The review focused on the safety and medical applications of clinoptilolite, a naturally occurring zeolite, for in vivo use. The analysis encompassed the physical and chemical properties of clinoptilolite, its detoxifying capabilities, and its potential therapeutic effects in both human and veterinary medicine. The review also considered various studies on clinoptilolite's role in detoxification, immune modulation, oxidative stress, and its impact on body homeostasis. The safety of clinoptilolite for internal consumption, including concerns about the potential release of heavy metals like lead during use, was also addressed.

Who was reviewed?

The review examined the available scientific literature concerning the effects of clinoptilolite in various contexts, specifically in animal studies and human trials. It included research from both clinical studies and preclinical evaluations that utilized clinoptilolite as a dietary supplement or therapeutic agent. The literature encompassed studies on humans, including clinical trials and case studies, as well as extensive research on animals such as dairy cows, rats, mice, and poultry, assessing both the therapeutic benefits and safety profiles of clinoptilolite-based products.

What were the most important findings of this review?

  1. Detoxification Properties: Clinoptilolite exhibits high cation-exchange capacity, making it effective in adsorbing toxic metals such as lead, cadmium, and nickel from the body. This property makes it valuable in reducing the load of toxic elements in animals and humans.
  2. Safety Profile: Clinoptilolite, particularly in its micronized and purified forms, has been shown to be generally safe for in vivo use at varying dosages, with no significant toxic effects observed in animals or humans. The European Food Safety Authority (EFSA) has classified it as safe for consumption in animal feeds at specified doses.
  3. Immunomodulatory and Antioxidant Effects: Clinoptilolite can modulate immune responses, as evidenced by its effects on immune cell counts and antioxidant enzyme activity in various studies. This includes improved responses in immunodeficient conditions and a reduction in markers of oxidative stress.
  4. Impact on Microbiota and Gastrointestinal Health: Clinoptilolite has shown positive effects on gut health, potentially through its detoxifying role, the maintenance of intestinal wall integrity, and its ability to modulate the microbiota composition. These effects suggest potential therapeutic roles in conditions like gastrointestinal disorders, dysbiosis, and enhanced resistance to pathogens.

What are the greatest implications of this review?

  1. Potential for Human Therapeutics: The review highlights the potential use of clinoptilolite as a nutraceutical for detoxifying heavy metals and improving immune responses in humans. Its safety profile supports further exploration into its use in supplements aimed at mitigating conditions related to heavy metal exposure, such as systemic nickel allergy syndrome (SNAS) and other metal-induced toxicities.
  2. Veterinary Applications: Clinoptilolite's effectiveness in improving animal health—particularly its ability to enhance reproductive health, detoxify nitrates, and manage mycotoxins—suggests that it could serve as a valuable supplement in livestock production. Its role in reducing ammonia and supporting gut health is particularly important for animal productivity and welfare.
  3. Integration in Microbiome-Targeted Therapies: Given its impact on gastrointestinal health and the microbiome, clinoptilolite holds promise as a component of microbiome-targeted interventions (MBTIs). It could serve as an adjunctive therapy in managing conditions like irritable bowel syndrome (IBS) and other dysbiosis-associated disorders.
  4. Safety Considerations and Future Research: The review underlines the importance of rigorous quality control and characterization of clinoptilolite materials for in vivo applications. Variability in clinoptilolite's physical and chemical properties across different sources necessitates careful evaluation before therapeutic use. Future studies are needed to delineate the specific mechanisms by which clinoptilolite exerts its effects on immune modulation and detoxification, as well as to confirm its efficacy and safety in long-term human applications.

Current Treatment of Bacterial Vaginosis—Limitations and Need for Innovation

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review reveals high BV recurrence rates after metronidazole or clindamycin treatment due to microbial biofilms and potential sexual transmission. While both antibiotics show similar short-term efficacy, they differ in resistance patterns. Biofilm disruptors and partner treatment may improve outcomes, but better diagnostics and combination therapies are urgently needed.

What was Reviewed?

This comprehensive review critically examines the current limitations in bacterial vaginosis (BV) treatment, with particular focus on the high recurrence rates following standard antibiotic therapies, including both metronidazole and clindamycin. The authors analyze the microbial factors contributing to treatment failure, specifically the role of polymicrobial biofilms and antimicrobial resistance patterns in Gardnerella vaginalis and other BV-associated bacteria. The review also explores emerging evidence for sexual transmission of BV-associated microorganisms and evaluates novel therapeutic approaches targeting biofilm disruption and partner treatment strategies.

Who was Reviewed?

The review synthesizes data from multiple clinical trials and observational studies involving women with recurrent BV across diverse populations. It incorporates microbiological research on vaginal and penile microbiota, including studies demonstrating the presence of BV-associated bacteria in male sexual partners. The analysis also examines in vitro studies of biofilm formation and disruption, as well as limited clinical trials of adjunctive therapies like boric acid and probiotics.

Key Findings and Microbial Associations

The review highlights that BV represents a profound dysbiosis where protective Lactobacillus species, particularly L. crispatus, are replaced by a polymicrobial consortium including Gardnerella vaginalis, Atopobium vaginae, and various Clostridiales species. These pathogens form resilient biofilms that protect them from both metronidazole and clindamycin, the two first-line antibiotics for BV. While short-term cure rates approach 80% for both medications, recurrence rates exceed 50% within 6-12 months. The review notes important differences between the antibiotics: clindamycin appears more effective against certain biofilm-embedded pathogens like A. vaginae but may promote clindamycin-resistant anaerobic gram-negative rods, while metronidazole faces challenges with intrinsically resistant G. vaginalis clades. Both antibiotics fail to address the potential sexual transmission of BV-associated bacteria, which are detectable in male partners' genital microbiota and may contribute to reinfection.

Implications of the Review

The review underscores that current antibiotic regimens, whether using metronidazole or clindamycin, are insufficient for long-term BV control due to biofilm persistence and potential sexual transmission. Clinicians should continue following treatment guidelines but recognize these limitations when managing recurrent cases. The findings suggest several important considerations: vaginal clindamycin may be preferable for certain BV subtypes or in pregnancy, while metronidazole remains the most widely studied option. For recurrent BV, adjunctive approaches like biofilm disruptors (boric acid, DNase) or partner treatment may be worth considering, though more research is needed. The review emphasizes the need for improved diagnostics to identify BV subtypes and resistance patterns, as well as the development of combination therapies targeting both pathogens and biofilms. Public health measures promoting condom use and further research into sexual transmission dynamics could help reduce BV recurrence at the population level.

Determinants of premenstrual dysphoric disorder and associated factors among regular undergraduate students at Hawassa University Southern, Ethiopia

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Among Ethiopian female university students, PMDD affects nearly two-thirds and links strongly to severe menstrual pain, irregular cycles, low social support, and contraceptive use. Early identification and comprehensive care are essential to alleviate its academic and psychological impacts.

What was studied?

This institutional-based cross-sectional study investigated the prevalence and determinants of premenstrual dysphoric disorder (PMDD) among regular undergraduate female students at Hawassa University, Ethiopia. It aimed to quantify PMDD prevalence using a standardized premenstrual symptoms screening tool and to identify socio-demographic, clinical, psychological, gynecological, and behavioral factors associated with PMDD in this population.

Who was studied?

The study included 374 regular female undergraduate students aged 18 and above from the College of Medicine and Health Sciences at Hawassa University. Participants were selected using stratified random sampling and completed a self-administered questionnaire assessing premenstrual symptoms, menstrual characteristics, social support, contraceptive use, and behavioral factors. Students with illnesses or absent during data collection were excluded.

What were the most important findings?

The study found a high PMDD prevalence of approximately 63% among participants, significantly higher than many global estimates. Key factors independently associated with PMDD included severe menstrual pain, irregular menstrual cycles, poor or moderate social support, and contraceptive use. Severe dysmenorrhea exacerbated emotional and behavioral symptoms, while social support appeared protective. These findings indicate that PMDD substantially impairs daily functioning, including academic performance, and is influenced by a complex interplay of physiological, psychological, and social factors. The study emphasizes the importance of early screening and tailored interventions to mitigate PMDD's impact on student well-being and success.

What are the greatest implications of this study?

This study highlights PMDD as a prevalent and underrecognized condition with significant negative effects on young women’s mental health and academic performance in a low-resource setting. It underscores the urgent need for integrating PMDD screening and psychosocial support into university health services, especially focusing on managing menstrual pain, providing social support, and carefully evaluating contraceptive use. These findings advocate for multidisciplinary interventions that address both physical and psychological determinants of PMDD, aiming to improve quality of life and academic outcomes. Moreover, the research supports policymakers and educators in developing targeted health promotion programs and facilitating access to effective treatment for PMDD in similar contexts.

Dietary Intake of Selected Nutrients Affects Bacterial Vaginosis in Women

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

Higher dietary fat intake increases the risk of bacterial vaginosis (BV), while folate, vitamin E, and calcium may lower severe BV risk. This study suggests dietary interventions could play a role in managing BV, offering insights for clinicians on how nutrition affects vaginal microbiome balance.

What was Studied?

This study examined the relationship between dietary intake and the presence of bacterial vaginosis (BV) in women. Researchers assessed how macronutrient and micronutrient consumption, particularly fat intake and essential vitamins, influenced the risk of BV and severe BV.

Who was Studied?

The study analyzed data from 1,521 non-pregnant women, aged 15–45, who were part of a larger longitudinal study of vaginal flora in Birmingham, Alabama. The majority (86%) were African American. Participants underwent clinical assessments and completed a food frequency questionnaire.

Most Important Findings

The study found a significant association between dietary fat intake and the risk of BV. Women consuming higher amounts of total fat, saturated fat, and monounsaturated fat had an increased likelihood of BV and severe BV. Total fat intake was linked to a 50% higher risk of BV, while saturated fat and monounsaturated fat were particularly associated with severe BV.

Conversely, higher intakes of folate, vitamin E, and calcium were associated with a reduced risk of severe BV. These nutrients may support local immune function, potentially counteracting the microbial imbalance seen in BV. Energy intake had a marginal association with BV, while carbohydrate and protein intake showed no significant links. The study suggests that a high-fat diet may alter vaginal microflora, increase vaginal pH, and contribute to BV development.

Implications of the Study

Diet plays a crucial role in vaginal health, with fat consumption significantly influencing BV risk. Clinicians can reduce BV risk by advising patients to lower fat intake and increase folate, vitamin E, and calcium. Researchers should further explore how dietary fat disrupts vaginal microbiota and whether targeted nutritional changes can serve as effective prevention strategies.

Dietary Patterns and Bacterial Vaginosis: is there any association?

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

A nutrient-rich diet lowers bacterial vaginosis (BV) risk, while processed foods increase it. This study highlights the importance of dietary choices in vaginal microbiome balance, offering insights for clinicians on how diet influences BV risk.

What was Studied?

Researchers investigated how dietary acid load and adherence to the Alternative Healthy Eating Index (AHEI) affect bacterial vaginosis (BV) risk. They aimed to determine whether dietary patterns and acid-producing foods influence the vaginal microbiome and BV prevalence.

Who was Studied?

The study included 143 women diagnosed with BV and 151 healthy controls, aged 18–45, from a gynecology clinic in Tehran, Iran. Researchers assessed dietary intake using a validated food frequency questionnaire and diagnosed BV using the Amsel criteria.

Most Important Findings

A high AHEI score significantly lowered BV risk. Women in the highest AHEI tertile had a 75% lower chance of developing BV. Consuming more vegetables, nuts, legumes, and unprocessed meats further reduced BV odds. In contrast, high intakes of sugar-sweetened beverages, trans fats, and sodium increased BV risk by up to three times.

Dietary acid load, measured by potential renal acid load (PRAL) and net endogenous acid production (NEAP), showed no significant link to BV. This suggests that overall diet quality, rather than dietary acid-base balance, plays a more critical role in BV risk.

Microbiome analysis linked high AHEI adherence to a Lactobacillus-dominant vaginal environment, which protects against BV. In contrast, poor dietary choices promoted the growth of BV-associated bacteria such as Gardnerella vaginalis, Atopobium spp., and Prevotella spp..

Implications of the Study

Diet quality directly affects vaginal microbiome health. Clinicians should encourage patients to adopt a plant-based diet rich in vegetables, nuts, and legumes while limiting sugar-sweetened beverages and trans fats. Future research should explore whether dietary interventions can serve as an effective strategy for BV prevention and management.

Dioxin-like PCB 126 increases intestinal inflammation and disrupts gut microbiota and metabolic homeostasis.

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study investigated the effects of PCB 126 on gut microbiota and metabolic health in mice. It revealed significant shifts, including a 49-fold increase in the Firmicutes to Bacteroidetes ratio, disruptions in gut diversity, elevated inflammation, and altered metabolic hormone levels. These findings underscore the need for protective health policies and further research.

What was studied?

The research focused on examining the effects of PCB 126, a dioxin-like pollutant, on gut health and microbiota as well as metabolic homeostasis in a mouse model predisposed to cardiometabolic diseases. The study investigated how exposure to this pollutant influences the gut microbiome, intestinal and systemic inflammation, metabolic hormones, and the development of metabolic disease markers like atherosclerosis.

 

Who was studied?

The subjects of this study were male LDL receptor-deficient (Ldlr -/-) mice, which are genetically predisposed to cardiometabolic diseases. These mice were used to model the impact of PCB 126 exposure on the progression of diseases such as diabetes and atherosclerosis, reflecting potential effects in human populations exposed to similar environmental contaminants.

 

What were the most important findings?

 

Category Findings
Microbiota and Metabolic Disruption PCB 126 exposure led to significant shifts in gut microbiota, notably decreased alpha diversity and increased Firmicutes to Bacteroidetes ratio, which are changes associated with gut dysbiosis and metabolic diseases.
Inflammatory Responses The study observed increased systemic and intestinal inflammation in PCB-exposed mice, evidenced by elevated levels of various inflammatory cytokines and metabolic hormones such as insulin and glucagon-like peptide-1 (GLP-1).
Metabolic Hormone Alterations There were increased levels of insulin and insulin-related peptides, indicating potential disruptions in glucose homeostasis and insulin sensitivity due to the toxicant exposure.
Atherosclerosis and Cardiovascular Risk While the study found trends towards increased atherosclerosis in PCB 126 exposed mice, the findings at later stages were less pronounced, suggesting a complex interaction over time between pollutant exposure and cardiovascular disease progression.

 

What are the greatest implications of this study?

The study explored the significant health risks posed by exposure to dioxin-like pollutants such as PCB 126, focusing on its implications for metabolic and inflammatory diseases. The research demonstrated how PCB 126 disrupts gut microbiota, elevates systemic and intestinal inflammation, and impacts metabolic hormone levels, potentially exacerbating conditions like diabetes and atherosclerosis. These findings advocate for the implementation of stringent environmental regulations and public health policies to minimize exposure to such toxicants.

Moreover, the results underline the critical need for clinical and therapeutic interventions that leverage our growing understanding of the interaction between environmental pollutants and gut microbiota. Developing strategies to preserve or restore gut microbiota balance could serve as preventive measures or treatments for those affected by or at risk of pollutant-induced health conditions. Additionally, the study prompts further research into the exact mechanisms through which pollutants affect health, which could guide the development of comprehensive disease management and treatment solutions that integrate environmental considerations.

Dioxin-like PCBs and Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The study reviews the impact of dioxin-like PCBs on endometriosis, highlighting inconsistent epidemiological findings and the greater harm of early life exposures. It underscores the need for stricter regulations and further research to understand PCBs' effects on reproductive health and develop effective interventions.

What was studied?

The study focused on the potential impact of environmental exposure to dioxin-like polychlorinated biphenyls (PCBs) on the development of endometriosis. It specifically reviewed the complexities and challenges of determining the effects of such toxicants on human health, with an emphasis on reproductive tract diseases.

 

Who was studied?

The research predominantly referenced studies involving both human and animal populations. It synthesized findings from various epidemiological studies to evaluate the correlation between PCB exposure and the risk of developing endometriosis in humans, while also considering animal studies for understanding early life exposures and their long-term effects.

 

What were the most important findings?

Category Description
Varying Epidemiological Results Epidemiological studies have shown inconsistent results regarding the relationship between PCB exposure and the development of endometriosis. Some studies suggest a potential association, while others do not.
Importance of Early Life Exposures The research highlighted that exposures to PCBs during early life stages are more detrimental and could be critical in determining the risk of developing diseases like endometriosis later in life.
Mechanisms of Toxicity PCBs, particularly those that are dioxin-like, have significant effects on endocrine and immune functions which could potentially disrupt normal reproductive tract development and function.

What are the greatest implications of this study?

The study emphasizes the necessity of implementing stringent public health policies to limit exposure to PCBs, recognizing their potential for significant long-term effects on reproductive health. It suggests that a deeper understanding of how PCBs affect endometrial physiology could lead to more effective clinical strategies for mitigating the impacts of these unavoidable environmental exposures. Additionally, the research identifies an urgent need for more comprehensive studies that incorporate detailed exposure histories. These studies are essential to elucidate the connections between PCB exposure and endometriosis, and to develop targeted interventions aimed at reducing the risk of disease associated with environmental toxicants.

Does Exposure of Lead and Cadmium Affect the Endometriosis?

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

The study links lead and cadmium exposure to increased endometriosis risk, emphasizing lead's role at low blood levels and synergistic effects with cadmium. It advocates for strict monitoring and preventive measures to minimize exposure.

What Was Studied?

This study investigated the association between occupational exposure to lead and cadmium and the risk of developing endometriosis (EM) among South Korean female workers. Utilizing medical and biological data from over 26,000 individuals who underwent lead-associated medical examinations between 2000 and 2004, the study examined blood lead levels (BLLs), co-exposure to cadmium, and their relationship with hospital admissions for EM.

Who Was Studied?

The study focused on South Korean female workers exposed to lead as part of their occupation. These individuals underwent specialized medical examinations. A total of 26,542 workers were included, with the study comparing EM admissions in lead-exposed workers against the general population and noise-exposed workers as control groups.

What Were the Most Important Findings?

The study found that lead exposure, even at relatively low levels (BLLs < 5 µg/dL), was significantly associated with an increased risk of EM. The standard admission rate (SAR) for EM in lead-exposed workers was 1.24 times higher than the general population, and for workers with BLLs < 5 µg/dL, it was 1.44 times higher. Co-exposure to lead and cadmium demonstrated a synergistic effect, amplifying the risk of EM beyond what could be expected from exposure to either metal alone. While cadmium exposure alone did not show a statistically significant association with EM, the relative excess risk due to interaction (RERI) was 0.33, indicating a notable combined impact of these metals. The study also highlighted that oxidative stress induced by heavy metal exposure likely underpins these effects, with mechanisms involving the disruption of antioxidant defenses and cellular damage.

What Are the Greatest Implications of This Study?

This research underscores the need to minimize exposure to heavy metals, particularly lead and cadmium, among female workers. It also calls for rigorous monitoring of blood lead and cadmium levels in workplaces to mitigate their combined effects. The findings are critical for understanding the environmental and occupational contributors to EM and suggest that policies limiting heavy metal exposure could have a substantial public health impact, especially for at-risk populations.

Dysbiosis of Gut Microbiota (DOGMA) – A novel theory for the development of Polycystic Ovarian Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review introduces the DOGMA theory linking gut dysbiosis and leaky gut to insulin resistance, inflammation, and hyperandrogenism in PCOS. It outlines a compelling rationale for microbiome-targeted therapies using prebiotics and probiotics.

What Was Reviewed?

This paper proposed a novel etiological framework termed the “Dysbiosis of Gut Microbiota (DOGMA) theory" to explain the development of polycystic ovary syndrome (PCOS). It reviewed both experimental and observational evidence linking gut microbiota imbalances to PCOS’s hallmark features: insulin resistance, hyperandrogenism, and anovulation. The authors synthesized findings from microbiology, endocrinology, gastroenterology, and immunology to argue that diet-induced dysbiosis triggers increased intestinal permeability, enabling the systemic translocation of lipopolysaccharides (LPS) from gram-negative bacteria. This metabolic endotoxemia, in turn, activates inflammatory pathways and disrupts insulin signaling, which they propose is the central mechanism leading to the hormonal and reproductive manifestations of PCOS. The paper also examined how probiotics, prebiotics, and synbiotics may represent novel, gut-targeted therapies for PCOS.

Who Was Reviewed?

The authors aggregated evidence from previous studies involving women with PCOS, obese individuals, those with irritable bowel syndrome (IBS) or chronic fatigue syndrome (CFS), and various animal models. These studies consistently demonstrated that individuals with PCOS tend to have poor dietary patterns, high in saturated fats and refined sugars but low in fiber, which promote gut dysbiosis and increased gut permeability. Although the paper did not report new microbiome sequencing data, it drew from prior literature that had established correlations between reduced levels of beneficial bacteria such as Bifidobacterium and Lactobacillus, and increased abundance of pro-inflammatory gram-negative species such as Enterobacteriaceae and Bacteroides.

What Were the Most Important Findings?

The paper’s central hypothesis, DOGMA, asserts that dysbiosis-induced increases in intestinal permeability initiate a cascade of systemic inflammation and insulin resistance that ultimately disrupts ovarian function. Specifically, the increased translocation of LPS into the bloodstream activates macrophages, leading to overproduction of TNF-α and IL-6, which impair insulin receptor signaling and raise systemic insulin levels. This hyperinsulinemia enhances androgen production by ovarian thecal cells and suppresses hepatic SHBG production, thereby increasing circulating free androgens. In parallel, insulin resistance blocks ovulatory follicle maturation, leading to menstrual irregularity and the characteristic polycystic ovarian morphology. The major microbial associations (MMA) discussed include a decline in Bifidobacterium and Lactobacillus, increased Escherichia coli and Bacteroides, and decreased production of short-chain fatty acids like butyrate, which are critical for maintaining mucosal barrier integrity. These findings collectively map a plausible causal pathway from gut microbial imbalance to endocrine dysfunction in PCOS, even in lean individuals.

What Are the Implications of This Review?

The DOGMA hypothesis marks a paradigm shift in PCOS pathophysiology, positioning the gut microbiome not as a secondary player, but as a central driver of disease onset and progression. For clinicians, this reframing has significant implications. It suggests that microbial screening could become part of diagnostic protocols for PCOS, especially in cases not explained by obesity or conventional metabolic syndrome criteria. Therapeutically, this review underscores the potential of prebiotics and probiotics to restore microbial balance, reduce metabolic endotoxemia, and reverse insulin resistance. While empirical support for these interventions in PCOS remains preliminary, the mechanistic rationale is robust, especially given successful outcomes in adjacent conditions like gestational diabetes and obesity.

Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study reveals significant dysbiosis of gut microbiota in women with PCOS, particularly in obese individuals. The altered microbial composition correlates with metabolic and hormonal imbalances, offering potential therapeutic targets for improving PCOS-related conditions.

What Was Studied?

This study focused on the dysbiosis of gut microbiota in women with Polycystic Ovary Syndrome (PCOS) and its correlation with clinical and metabolic parameters. The researchers examined how gut microbiota composition differs between women with PCOS, both obese and non-obese, and healthy controls. Specifically, the study analyzed the relationships between microbial diversity, sex hormones, metabolic markers, and brain-gut axis mediators like serotonin, ghrelin, and peptide YY (PYY).

Who Was Studied?

The study included 48 premenopausal women aged 17–45 years who were divided into four groups: obese women with PCOS, non-obese women with PCOS, obese control women, and non-obese control women. Participants were selected based on their clinical history, and those with conditions like thyroid disorders, hypertension, and lipid dysregulation were excluded from the study.

What Were the Most Important Findings?

The study revealed significant differences in the gut microbiota between PCOS patients and healthy controls, with notable changes linked to obesity. Specifically, PCOS women, especially those who were obese, exhibited a significant decrease in the diversity of their gut microbiota, particularly with a reduction in beneficial species like Akkermansia and Clostridium. Conversely, harmful microbes like Bacteroides and Escherichia/Shigella were more prevalent in the gut microbiota of PCOS patients, particularly those with obesity.

Additionally, the study identified several co-abundance groups (CAGs) of microbes that were associated with various clinical parameters of PCOS, such as waist circumference, testosterone levels, and hirsutism scores. The altered microbiota composition in PCOS was also correlated with metabolic disturbances, including insulin resistance and abnormal lipid profiles. Importantly, the study found that certain gut microbiota were linked to the secretion of brain-gut axis peptides (serotonin, ghrelin, and PYY), which play a role in regulating metabolism and psychological well-being. The changes in microbial communities, such as the abundance of Bacteroides and the scarcity of Akkermansia, suggested a potential mechanism linking gut dysbiosis with the metabolic and hormonal imbalances characteristic of PCOS.

What Are the Implications of This Study?

The findings suggest that gut microbiota dysbiosis may play a significant role in the pathogenesis of PCOS, particularly in modulating metabolic dysfunctions such as insulin resistance, hyperandrogenism, and obesity. This opens new avenues for potential therapeutic interventions, such as dietary modifications, probiotics, or fecal microbiota transplantation, to restore microbial balance and improve the clinical outcomes of PCOS. The study also underscores the importance of considering the gut microbiota as a potential target for managing metabolic diseases associated with PCOS. However, further research with larger sample sizes and causal studies is needed to confirm these associations and explore the mechanistic pathways involved.

Dysbiosis of the Saliva Microbiome in Patients With Polycystic Ovary Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study reveals disrupted diurnal rhythms and significant dysbiosis in the salivary microbiota of PCOS patients, notably elevated Fusobacterium and reduced Actinobacteria. These oral microbial patterns may influence systemic metabolic and hormonal dysregulation.

What Was Studied?

This study investigated the composition, diversity, and diurnal oscillation of the salivary microbiome in women with polycystic ovary syndrome (PCOS) compared to healthy controls. The researchers sought to determine whether salivary microbiota differ in PCOS patients at different time points during the day and to assess how disruptions in these microbial patterns might contribute to metabolic and endocrine dysregulation. Using 16S rRNA gene sequencing, the authors analyzed salivary samples collected every six hours over 24 hours, along with fecal samples, from 10 PCOS patients and 10 age-matched healthy women. They evaluated microbial diversity, taxonomic differences at multiple phylogenetic levels, and predicted metabolic pathway alterations using PICRUSt. A key objective was to identify whether PCOS disrupts the circadian rhythm of oral microbiota, which may contribute to systemic disease processes.

Who Was Studied?

The study recruited 10 women with PCOS diagnosed via the Rotterdam criteria and 10 healthy controls, all aged between 18 and 45, and matched for body mass index (BMI), diet, and lifestyle factors. Participants underwent strict dietary controls before sample collection, and the study excluded individuals with recent antibiotic or probiotic use, hormonal treatment, or oral or systemic diseases. Saliva was sampled at four Zeitgeber time (ZT) points, and stool samples were collected to compare oral and gut microbiota profiles. Blood samples were also taken to assess hormonal and metabolic biomarkers, allowing for correlation with microbial changes.

What Were the Most Important Findings?

PCOS patients showed clear evidence of salivary microbiota dysbiosis. At ZT0, alpha diversity was significantly lower in PCOS participants, indicating reduced species richness and diversity. Beta diversity analysis revealed significant structural shifts in the microbial community at ZT0 and ZT18. Notably, the relative abundance of the phylum Fusobacteria and the genus Fusobacterium was consistently higher in PCOS patients at all time points, while beneficial taxa such as Actinobacteria and Leptotrichia were diminished, particularly at night.

Functionally, PCOS samples exhibited disrupted KEGG pathways. “Methane metabolism” and “butanoate metabolism” were consistently upregulated, both of which have been associated with metabolic disorders and gut permeability. Conversely, pathways related to protein folding, secretion systems, and structural molecule synthesis were downregulated. Critically, the study found that the circadian rhythm of bacterial abundance, observable in healthy individuals for phyla like Proteobacteria, Bacteroidetes, and orders such as Lactobacillales, was largely absent in PCOS patients. This disruption may contribute to both oral health problems and broader metabolic dysfunction via chronobiological misalignment.

While no major differences were detected in gut microbiota composition or diversity between the PCOS and control groups, the oral microbiome showed profound alterations. This suggests that salivary bacteria may offer more accessible and sensitive biomarkers for PCOS-related dysbiosis than fecal bacteria, at least in early or non-obese phenotypes.

What Are the Implications of This Study?

This study provides the first robust evidence that the salivary microbiota in PCOS not only differs significantly from that of healthy controls but also lacks a normal diurnal rhythm. These findings underscore the relevance of the oral microbiome as both a diagnostic window and a potential therapeutic target in PCOS. Clinicians should consider the possibility that microbial timing is as important as microbial composition. Elevated Fusobacterium levels and reduced Actinobacteria may serve as early biomarkers for PCOS-linked dysbiosis, while their functional consequences, such as increased methane production and impaired protein metabolism, suggest mechanistic links to systemic inflammation, insulin resistance, and metabolic syndrome.

From a clinical standpoint, the convenience of saliva sampling, combined with time-sensitive microbial signatures, could facilitate non-invasive PCOS monitoring and risk stratification. This opens the door for chrono-microbiome interventions, timed probiotic delivery, circadian-aligned dietary changes, or salivary microbiome modulation as adjunct therapies. These findings highlight the need for further metagenomic and metatranscriptomic studies to validate and expand the functional understanding of these dysbiotic patterns.

Effect of endometriosis on the fecal bacteriota composition of mice during the acute phase of lesion formation

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study investigated the impact of induced endometriotic lesions on gut microbiota in mice, using GFP+ uterine tissue transplantation. Despite successful lesion formation, no significant changes in gut microbiota composition were observed in the acute phase, suggesting endometriosis may not cause pronounced dysbiosis during early lesion development.

What was studied?

The study investigated whether the induction of endometriosis in mice affects the composition of their gut microbiota. It tested this by transplanting uterine tissue fragments into mice and analyzing changes in the gut microbiota before and after endometriosis induction.

 

Who was studied?

Female C57BL/6 wild-type mice and GFP+ transgenic donor mice were used. Uterine tissue from the donor mice was transplanted into the peritoneal cavity of the wild-type mice to induce endometriosis, with sham-transplanted mice serving as controls.

 

What were the most important findings?

Endometriotic lesions successfully developed in the mice, but the study found no significant alterations in the gut microbiota composition within the 21-day observation period. The bacterial community remained stable, indicating no early-phase intestinal dysbiosis due to endometriosis induction.

 

What are the greatest implications of this study?

The study hypothesizes that there is a bi-directional relationship between gut dysbiosis and endometriosis, where alterations in the gut microbiota may influence the development and progression of endometriosis and vice versa. Although this particular study did not find significant changes in the gut microbiota composition within the early phase of endometriosis induction in mice, it suggests the possibility that the gut microbiota could be involved in hormone-related, inflammatory, angiogenic, and vasculogenic processes associated with endometriosis.

Other studies’ findings, which reported dysbiosis following endometriosis induction, further support the idea of a complex interaction between endometriosis and the gut microbiota. This interaction could potentially impact estrogen metabolism, systemic inflammation, and stem cell homeostasis, all of which are implicated in the pathogenesis of endometriosis. However, the study calls for more research to clarify this relationship, including studies on microbial activity and metabolic function, to understand how gut microbiota might affect endometriosis fully.

Effect of endometriosis on the fecal bacteriota composition of mice during the acute phase of lesion formation

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study found that endometriosis does not induce significant changes in the fecal bacteriota composition during the acute phase of lesion formation. Findings suggest that gut microbial shifts in endometriosis may emerge only during chronic disease stages, highlighting the need for targeted microbiome interventions over prolonged periods.

What Was Studied?

This study investigated the effect of endometriosis on the fecal bacteriota composition of mice during the acute phase of lesion formation. Researchers aimed to understand whether the establishment of endometriotic lesions would influence gut microbial communities, potentially contributing to systemic inflammation or metabolic disruptions associated with the disease. Uterine tissue fragments from GFP+ donor mice were transplanted into the peritoneal cavity of GFP- wild-type mice, inducing endometriotic lesions. Sham-transplanted mice served as controls. Fecal samples were collected three days before, and at 7 and 21 days after lesion induction, and analyzed through 16S rRNA gene sequencing to map changes in microbial composition.


Who Was Studied?

The study involved C57BL/6 wild-type mice as the experimental model for endometriosis. The model was established by transplanting uterine tissue fragments from GFP+ donor mice into the peritoneal cavity of GFP- recipient mice, allowing for easy visualization of endometriotic lesions. Sham-transplanted animals, which received physiological saline solution instead of tissue fragments, served as controls. The study analyzed fecal samples collected at specific time points to assess microbiota changes during the acute phase of endometriosis development.

What Were the Most Important Findings?

The study found that the induction of endometriosis did not produce significant changes in the composition of the fecal bacteriota during the acute phase of lesion formation (7 and 21 days post-transplantation). Despite the successful establishment of endometriotic lesions and typical histomorphology observed under fluorescence microscopy, alpha and beta diversity analyses showed no substantial differences between the endometriosis-induced group and sham controls. Detailed sequencing revealed a highly diverse microbial community dominated by Bacteroidales S24-7 group, Lactobacillus, Prevotellaceae UCG-001 group, and Lachnospiraceae NK4A136 group in both experimental and control mice. Notably, contrary to previous studies suggesting dysbiosis following endometriosis induction, this investigation showed microbial stability throughout the acute phase of lesion formation. The researchers speculated that gut microbiota disturbances may become apparent only in the chronic stages of the disease, reflecting long-term inflammation and tissue remodeling. Furthermore, the study emphasized that strict statistical controls, including the removal of singleton OTUs and application of false discovery rate (FDR) corrections, were applied to prevent false positives. These rigorous controls could have contributed to the observed stability of gut microbiota composition, challenging earlier reports of rapid dysbiosis post-endometriosis induction.

ParameterFindings in Endometriosis-Induced Mice
Microbiota CompositionNo significant changes in the overall composition of fecal bacteriota during the acute phase (7 and 21 days post-transplantation).
Alpha DiversityNo substantial differences observed between endometriosis-induced mice and sham controls, indicating microbial richness and evenness remained stable.
Beta DiversityAnalysis showed no significant shifts in microbial community structure between experimental and control groups.
Dominant GeneraMicrobiota was dominated by Bacteroidales S24-7 group, Lactobacillus, Prevotellaceae UCG-001 group, and Lachnospiraceae NK4A136 group in both groups.
Impact of Lesion FormationInduction of endometriosis did not disrupt gut microbiota composition during the acute phase of lesion establishment.
Statistical Controls AppliedStrict controls, including false discovery rate (FDR) corrections and removal of singleton OTUs, were applied to enhance result reliability.
Hypothesized Long-Term EffectsAuthors suggest that gut dysbiosis may only emerge in chronic stages of endometriosis, not during initial lesion establishment.

What Are the Greatest Implications of This Study?

The study challenges prevailing hypotheses that endometriosis immediately disrupts gut microbiota during the early phases of lesion formation. The findings suggest that intestinal dysbiosis may not occur in the acute phase but could instead be a consequence of chronic inflammation and prolonged disease progression. This insight implies that gut microbial changes observed in patients with endometriosis might reflect long-term disease dynamics rather than initial lesion establishment. These results underscore the need for longitudinal studies to distinguish between acute and chronic microbiome shifts in endometriosis. The findings also highlight the importance of standardized microbiome analysis protocols and strict statistical measures to accurately assess microbial composition in endometriosis models. Understanding the timeline of microbiome alterations in endometriosis could guide therapeutic strategies targeting microbial populations in chronic disease stages rather than acute phases.

Effects of Probiotic Supplementation on Pancreatic β-cell Function and C-reactive Protein in Women with Polycystic Ovary Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study evaluates the impact of probiotic supplementation on insulin resistance and inflammation in women with PCOS. The results suggest that probiotics may help improve insulin sensitivity but have limited effects on inflammation markers.

What was studied?

This randomized, double-blind, placebo-controlled clinical trial studied the effects of probiotic supplementation on pancreatic β-cell function and C-reactive protein (CRP) levels in women with polycystic ovary syndrome (PCOS). The aim was to explore how probiotics might influence insulin sensitivity, metabolic parameters, and inflammation markers in PCOS, which is often associated with insulin resistance, inflammation, and hyperandrogenism.

Who was studied?

The study involved 72 women diagnosed with PCOS based on the Rotterdam criteria. These women were aged between 15 and 40 years and were randomly assigned to receive either probiotic supplementation (n=36) or a placebo (n=36) for 8 weeks. The study excluded participants with other chronic diseases, thyroid disorders, or those who had recently used medications such as antibiotics, insulin, or corticosteroids. All participants underwent fasting blood tests before and after the 8-week intervention to measure fasting blood sugar (FBS), serum insulin, HOMA-IR, and CRP levels.

What were the most important findings?

The primary findings of the study suggest that while probiotic supplementation did not significantly affect CRP or pancreatic β-cell function in the PCOS women, there were some beneficial effects on insulin metabolism. Specifically, serum insulin levels were significantly reduced in the probiotic group after adjusting for covariates, such as age, BMI, and physical activity. There was also a non-significant reduction in fasting blood sugar (FBS) and HOMA-IR in the probiotic group, suggesting potential improvements in insulin sensitivity. However, the study did not find significant changes in CRP levels, indicating that the probiotics may have had a limited impact on inflammation in this cohort.

From a microbiome perspective, probiotics are known to modulate gut microbiota, which plays a crucial role in regulating insulin sensitivity and inflammation. The positive changes in serum insulin levels and HOMA-IR suggest that the probiotics may have helped restore balance in the gut microbiome, potentially reducing insulin resistance, a hallmark of PCOS. However, the lack of significant changes in CRP levels suggests that probiotics alone may not be enough to significantly modulate systemic inflammation in PCOS patients, or a longer supplementation period may be required for more pronounced effects.

What are the greatest implications of this study?

This study provides valuable insights into the potential role of probiotics in managing metabolic and endocrine dysfunctions associated with PCOS. While the effects on insulin resistance were promising, the lack of significant impact on inflammation (as measured by CRP) indicates that probiotics may need to be combined with other therapeutic interventions to fully address the multifactorial nature of PCOS. Clinically, probiotics could be considered as a supplementary treatment for improving insulin sensitivity in PCOS, particularly in patients with insulin resistance. However, further studies with larger sample sizes and longer treatment durations are necessary to confirm the benefits and establish specific probiotic strains and dosages for PCOS management.

Efficacy and Safety of Different Drugs for the Treatment of Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This meta-analysis compared BV treatments, identifying ornidazole as the most effective oral drug and sucrose/probiotics as top non-antibiotic options. Restoring Lactobacillus dominance is key, with vaginal probiotics and sucrose showing high cure rates.

What was reviewed?

This systematic review and network meta-analysis examined the efficacy and safety of multiple treatments for bacterial vaginosis (BV), a common vaginal dysbiosis characterized by the overgrowth of anaerobic bacteria and a decline in protective Lactobacillus species. The study compared antibiotics (metronidazole, clindamycin, tinidazole, secnidazole, ornidazole, ofloxacin) with non-antibiotic therapies (sucrose, probiotics) to determine the most effective and safest options for clinical use. The analysis incorporated both direct and indirect comparisons across studies, providing a comprehensive ranking of treatments based on cure rates and adverse effects.

Who was reviewed?

The meta-analysis included 42 randomized controlled trials (RCTs) and observational studies, encompassing patients diagnosed with BV. The studies were sourced from PubMed and Embase, ensuring a broad evaluation of existing evidence. Participants were treated with either oral or vaginal formulations of the studied drugs, allowing subgroup analyses to assess differences in administration routes.

Most Important Findings

The review highlighted that BV, characterized by a shift from Lactobacillus-dominant vaginal microbiota to an overgrowth of anaerobic bacteria like Gardnerella vaginalisAtopobium vaginae, and Bacteroides spp., responds differently to treatments. Ornidazole emerged as highly effective, with a clinical cure rate superior to clindamycin and secnidazole. Sucrose and probiotics also showed promise, with sucrose ranking highest in clinical cure probability and probiotics demonstrating fewer adverse effects compared to metronidazole. Notably, metronidazole and secnidazole had higher adverse reaction rates than placebo, while probiotics and sucrose were safer alternatives. The study underscored the importance of restoring Lactobacillus dominance to rebalance vaginal microbiota, as sucrose promotes Lactobacillus growth by lowering vaginal pH, and probiotics directly reintroduce beneficial bacteria.

Implications of the Review

The findings suggest that ornidazole could be a superior alternative to traditional BV treatments like metronidazole, particularly for oral administration. Non-antibiotic options like sucrose and probiotics offer effective and safer alternatives, aligning with microbiome-focused therapies. Clinicians should consider these options, especially for patients with recurrent BV or those prone to adverse effects from antibiotics. The study also calls for more high-quality trials to validate these results and explore long-term outcomes.

Efficacy of Vitamin C Vaginal Tablets in Preventing Recurrence of Bacterial Vaginosis: A Randomized Controlled Trial

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study demonstrates that 250 mg vitamin C vaginal tablets reduce the recurrence of bacterial vaginosis, offering an effective and safe prophylactic treatment.

What was Studied?

This study evaluated the efficacy of vitamin C vaginal tablets as a prophylactic treatment for recurrent bacterial vaginosis (rBV). The research was a randomized, double-blind, placebo-controlled clinical trial involving 142 women who had been successfully treated for a recent episode of BV using either metronidazole or clindamycin. These women were randomly assigned to receive either vitamin C or a placebo for six months, to prevent the recurrence of BV.

Who was Studied?

The study involved 142 women aged between 18 and 50 years who had a history of recurrent episodes of bacterial vaginosis, defined as at least two acute episodes in the past year. After successful treatment of a BV episode, participants were randomized into two groups: one receiving 250 mg of vitamin C vaginal tablets and the other a placebo. The study participants were monitored for recurrence of BV for six months.

What were the Most Important Findings?

The study demonstrated that the use of vitamin C vaginal tablets significantly reduced the recurrence rate of BV in women compared to the placebo group. After three months of treatment, the recurrence rate in the vitamin C group was 6.8%, whereas the placebo group had a recurrence rate of 14.7%. After six months, the recurrence rate was 16.2% in the vitamin C group, compared to 32.4% in the placebo group, which was statistically significant (P = 0.024). The vitamin C treatment also showed a significant pH-lowering effect, which is thought to contribute to the inhibition of pathogen overgrowth. Additionally, the Kaplan-Meier survival analysis indicated that the vitamin C group had a significantly lower probability of experiencing a BV relapse (P = 0.029). The treatment was well tolerated, with minimal adverse events, mostly local reactions like itching and burning.

What are the Implications of this Study?

The study highlights the potential of vitamin C vaginal tablets as an effective prophylactic treatment for recurrent bacterial vaginosis. By lowering vaginal pH, vitamin C helps re-establish a more acidic environment, preventing the overgrowth of harmful anaerobic bacteria, such as Gardnerella vaginalis. This finding is significant for women with recurrent BV, particularly those with antibiotic-resistant strains or those who experience frequent recurrences after antibiotic therapy. Given the safety profile and efficacy of vitamin C, it presents a promising alternative or complementary approach to current BV treatments, offering a simple and non-invasive way to manage this chronic condition.

Elevated Lactoferrin and Anti-Lactoferrin Antibodies in Endometriosis: Autoimmune and Microbiome Insights

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This study confirms elevated lactoferrin and anti-lactoferrin antibody levels in endometriosis, suggesting autoimmune involvement. Anti-lactoferrin drops post-surgery, hinting at a biomarker role, while lactoferrin ties to inflammation and potential microbiome links.

What Was Studied?

This study, conducted by Mori-Yamanaka et al. and published in Tohoku J. Exp. Med. in 2023, definitively explored serum lactoferrin (LTF) and anti-lactoferrin antibody (aLF) levels in patients with endometriosis. Endometriosis, a chronic inflammatory condition marked by ectopic endometrial-like tissue, remains poorly understood in terms of its underlying mechanisms. The researchers aimed to determine whether LTF, an iron-binding glycoprotein with antimicrobial and anti-inflammatory properties, and aLF, an autoantibody tied to immune dysregulation, play roles in the disease’s pathology. By measuring these markers in the blood of endometriosis patients compared to controls and assessing changes after surgical intervention, the study sought to uncover potential links to inflammation and autoimmunity. Although the study did not directly investigate microbiome signatures, LTF’s known role in modulating microbial environments suggests a possible indirect connection to gut or pelvic microbiome alterations in endometriosis.

Who Was Studied?

The research focused on 68 Japanese women undergoing surgery at Shiga University of Medical Science Hospital between November 2020 and May 2022. Of these, 51 had surgically and histopathologically confirmed endometriosis, spanning all stages (I-IV) per the revised American Society for Reproductive Medicine classification. The remaining 17 women, who underwent surgery for other gynecological issues like uterine myomas or benign ovarian tumors, served as controls without endometriosis. This cohort provided a robust sample to compare LTF and aLF levels across disease states and post-treatment outcomes, offering clinicians a clear demographic context for interpreting the findings.

What Were the Most Important Findings?

The study conclusively demonstrated that serum LTF and aLF levels are significantly elevated in endometriosis patients compared to controls, with p-values of 0.016 and 0.028, respectively. These elevations were particularly striking in advanced stages (III and IV), showing stronger statistical significance (LTF: p = 0.024; aLF: p = 0.016) compared to controls. Following surgery in 21 patients, aLF levels dropped markedly (p < 0.001), while LTF levels showed no significant change (p = 0.102). Notably, 43% of endometriosis patients exhibited aLF levels above the reference range, a prevalence akin to autoimmune conditions. Although microbiome data wasn’t directly assessed, LTF’s antimicrobial properties hint at potential microbial associations, possibly involving dysbiosis in the pelvic or gut microbiome, which could exacerbate inflammation in endometriosis. These findings position LTF and aLF as key players in the disease’s inflammatory and possibly autoimmune landscape.

What Are the Greatest Implications of This Study?

This study’s implications are profound for clinicians managing endometriosis. The elevated aLF levels, mirroring patterns in autoimmune diseases, strongly suggest that endometriosis involves an autoimmune component, potentially driven by immune responses to microbial or endogenous triggers. This insight could shift treatment paradigms toward immune-modulating therapies. Moreover, the significant post-surgical decline in aLF levels establishes it as a promising biomarker for monitoring disease activity and treatment success, offering a practical tool for clinical decision-making. While LTF’s role remains less clear, its persistence post-surgery and antimicrobial function imply a complex interplay with inflammation and possibly the microbiome, warranting further investigation into microbial signatures like those of Lactobacillus or Prevotella, known to influence pelvic health. Despite the study’s limitations—its small sample and surgical focus—these findings pave the way for innovative diagnostics and therapies, urging clinicians to consider immune and microbial factors in endometriosis care.

Elevated Lead, Nickel, and Bismuth Levels in the Peritoneal Fluid of a Peritoneal Endometriosis Patient without Toxic Habits or Occupational Exposure following a Vegetarian Diet

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

A case study links elevated lead, nickel, and bismuth in peritoneal fluid with endometriosis, highlighting potential dietary and environmental exposures as contributors. Further research may identify these potentially toxic elements (PTEs) as diagnostic biomarkers and therapeutic targets.

What Was Studied?

This case report investigated the multielemental profile of peritoneal fluid (PF) in a 22-year-old woman diagnosed with peritoneal endometriosis. The study aimed to evaluate the concentrations of potentially toxic elements (PTEs), including lead (Pb), nickel (Ni), bismuth (Bi), and cobalt (Co), and to compare them with levels in an age-matched control without endometriosis. The patient had no toxic habits, occupational exposure, or documented environmental exposure, and adhered to a vegetarian diet, raising questions about the dietary and environmental sources of PTEs and their role in the pathogenesis of endometriosis.

Who Was Studied?

The primary subject was a 22-year-old woman diagnosed with peritoneal endometriosis during laparoscopic surgery, where her PF was analyzed. The comparison group included an age-matched control and a reference cohort of ten women diagnosed with non-hormonally dependent benign ovarian cysts.

Most Important Findings

The study revealed significantly elevated levels of Pb (75 µg/L, 90:1 ratio), Ni (40.4 µg/L, 4:1 ratio), Bi (33.3 µg/L, 1.5:1 ratio), and Co (1.39 µg/L, 5:1 ratio) in the PF of the endometriosis patient compared to the control. These findings suggest potential contributions of dietary and environmental exposures to PTEs. Nickel, a cofactor for metalloenzymes, was noted to be higher potentially due to the patient’s vegetarian diet, which is associated with increased nickel intake from plant-based foods such as nuts and legumes. Elevated Pb levels were striking, with concentrations much higher than typical dietary or environmental exposures in industrialized settings. While cobalt and bismuth also showed elevated levels, their specific roles in endometriosis remain unclear. The findings support the hypothesis that environmental and dietary PTE exposure may contribute to the pathogenesis of endometriosis by inducing oxidative stress or endocrine disruption.

Greatest Implications

This study highlights the need to explore PTEs as potential biomarkers for endometriosis diagnosis and as contributors to its etiology. Elevated PTE levels in PF may result from dietary habits, such as a vegetarian diet, or unidentified environmental exposures. This study emphasizes the importance of further investigations into environmental toxicology and dietary patterns in endometriosis patients. Understanding these associations could inform preventative strategies, dietary guidelines, and therapeutic interventions for endometriosis management.

Elevated levels of whole blood nickel in a group of Sri Lankan women with endometriosis: a case control study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study identified elevated blood nickel levels in women with endometriosis, suggesting a potential role of nickel as a metalloestrogen in its pathogenesis.

What Was Studied?

This study investigated the association between whole blood levels of nickel, cadmium, and lead in women with and without endometriosis. Specifically, it aimed to determine whether these heavy metals, known to have estrogenic properties, could be linked to the pathogenesis of endometriosis. The research involved analyzing the whole blood levels of these metals in 50 women with endometriosis and 50 age-matched controls who were confirmed to be free of the condition via laparoscopy or laparotomy.

Who Was Studied?

The study focused on a group of Sri Lankan women of reproductive age who underwent laparotomy or laparoscopy. The participants were divided into two groups: cases (women diagnosed with endometriosis, n=50) and controls (women without endometriosis, n=50). None of the participants were current smokers, and the groups were matched for age and body mass index.

Most Important Findings

The study revealed significantly elevated levels of nickel in the whole blood of women with endometriosis compared to controls (2.6 μg/L vs. 0.8 μg/L, p=0.016). This finding aligns with previous evidence that nickel, a potent metalloestrogen, can activate estrogen receptors and may contribute to the persistence of ectopic endometrial tissue. In contrast, the blood levels of cadmium and lead did not show statistically significant differences between the two groups. The presence of nickel in ectopic endometrial tissue, previously demonstrated by the researchers, supports the hypothesis that hematogenous routes could transport nickel to ectopic sites. Despite these findings, the study's small sample size limits the ability to conclude definitively that nickel is an etiological factor for endometriosis.

Greatest Implications

The discovery of higher nickel levels in women with endometriosis introduces a novel avenue for understanding the role of environmental pollutants, particularly metalloestrogens, in the condition's pathogenesis. It emphasizes the need for larger-scale studies to explore nickel's potential as a biomarker or contributor to endometriosis. Furthermore, this research underscores the importance of addressing environmental and occupational exposures to nickel, especially for women of reproductive age, as part of preventive strategies for endometriosis.

Endometrial whole metabolome profile at the receptive phase: influence of Mediterranean Diet and infertility

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study analyzed the endometrial metabolome of 45 infertile women, revealing 925 metabolites with a dominance of PUFAs. It found that Mediterranean Diet adherence impacts the endometrial environment, suggesting diet modifications could enhance fertility.

What was studied?

The study focused on analyzing the receptive-phase endometrial metabolome profiles among women with infertility and examining their associations with adherence to the Mediterranean Diet (MD). It aimed to identify metabolomic signatures that could influence the intrauterine environment and impact reproductive outcomes.

 

Who was studied?

The study involved 45 women of infertile couples, recruited at the Reproductive Unit of Virgen de las Nieves University Hospital, Granada. These women were diagnosed with various types of infertility, including endometriosis, recurrent implantation failure, unexplained infertility, and male factor infertility.

 

What were the most important findings?

Key findings from the study revealed that 925 distinct metabolites were identified in the endometrial tissue, with lipids, particularly polyunsaturated fatty acids (PUFAs), constituting the largest group. Women diagnosed with endometriosis and recurrent implantation failure exhibited lower levels of PUFAs compared to those with male factor or unexplained infertility. Additionally, adherence to the Mediterranean Diet (MD) was linked to specific changes in the endometrial metabolomic profile, suggesting that diet could influence the uterine environment depending on the individual’s health status.

 

What are the greatest implications of this study?

The implications of this study are significant for reproductive health. The findings suggest that the Mediterranean Diet may positively influence the endometrial environment and potentially enhance reproductive outcomes through its effects on specific metabolomic profiles. Metabolomic profiling emerges as a powerful tool for identifying biomarkers of endometrial health and designing targeted interventions to support successful embryo implantation and fertility. Additionally, further research could develop diet-based therapeutic strategies that improve endometrial receptivity and manage infertility, underscoring the crucial role of lifestyle factors in reproductive health.

Endometriosis and adenomyosis: shared pathophysiology

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study investigates the genetic and epigenetic mechanisms of endometriosis and adenomyosis, highlighting KRAS mutations and abnormal estrogen and progesterone receptor expressions as key factors. It underscores a shared pathophysiology between the conditions, providing insights into treatment targets and the molecular basis of disease progression.

 

What was studied?

The study focused on the pathophysiology, genetic variants, and epigenetic abnormalities underlying endometriosis and adenomyosis. It emphasized the mechanisms through which these conditions develop, the somatic mutations and epigenetic changes in endometrial and adenomyotic tissues, and the clinical implications of these findings. The primary genetic alterations studied include mutations in the KRAS gene and epigenetic modifications affecting estrogen and progesterone receptor expressions. The role of estrogen in promoting these conditions and the concept of progesterone resistance were also critically evaluated.

 

Who was studied?

While the published study does not explicitly mention specific patient groups or demographics, it implicitly refers to women diagnosed with endometriosis and adenomyosis. These conditions affect women of reproductive age, with endometriosis symptoms often starting in the adolescent years and extending to menopause and adenomyosis typically being diagnosed later, often in women in their 40s and 50s. The study involves an analysis of endometrial and adenomyotic tissues from these patients, examining genetic mutations and epigenetic changes within these tissues.

 

What were the most important findings?

Shared Pathophysiology: Both endometriosis and adenomyosis originate from endometrial cells, with KRAS mutations prevalent in both conditions’ epithelial cells. This points to a common genetic predisposition underpinning both diseases.

Epigenetic Abnormalities: Both conditions are characterized by epigenetic defects that lead to abnormal estrogen production and action, particularly through the overexpression of estrogen receptor-β and underexpression of progesterone receptors, resulting in progesterone resistance.

Clinical Implications of Genetic Mutations: The presence of specific genetic mutations, especially in KRAS, not only elucidates the pathogenic pathways of these conditions but also suggests that these mutations provide a survival advantage to ectopic endometrial tissues.

Estrogen’s Role: The study highlights estrogen’s critical role in the establishment and progression of endometriosis and adenomyosis, with implications for treatment strategies aimed at blocking estrogen synthesis.

 

What are the greatest implications of this study?

Therapeutic Targets: Identifying KRAS mutations and epigenetic markers offers new targets for therapeutic intervention, possibly allowing for more personalized treatment approaches for patients with endometriosis and adenomyosis.

Understanding of Disease Mechanism: Elucidating the genetic and epigenetic mechanisms underlying these conditions helps in understanding their pathogenesis, potentially leading to early detection and preventive strategies.

Progesterone Resistance: The insight into the molecular basis of progesterone resistance opens avenues for addressing this challenge in treating endometriosis and adenomyosis, potentially improving the efficacy of current therapies.

Research Directions: These findings pave the way for future research into the molecular and cellular biology of gynecological disorders, encouraging the development of innovative treatments that can address the root causes of these conditions rather than merely managing symptoms.

Endometriosis induces gut microbiota alterations in mice

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study used high-throughput DNA sequencing to show that murine endometriosis alters gut microbiota, notably increasing the Firmicutes/Bacteroidetes ratio and Bifidobacterium levels by day 42, indicating disease-specific dysbiosis. It underscores the need for further research on the long-term effects of endometriosis on gut microbiota and its bidirectional interaction with the host.

What was studied?

The research focused on investigating the impact of murine endometriosis on gut microbiota composition using high-throughput DNA sequencing to explore how the disease affects intestinal microbial communities.

 

Who was studied?

The study subjects were mice. These animals were divided into two groups: one group with induced endometriosis through the intraperitoneal injection of endometrial tissues and a mock group that served as a control.

 

What were the most important findings?

The study’s key findings include the emergence of a distinct gut microbiota composition in mice with endometriosis by day 42 post-modeling, highlighted by an increased Firmicutes/Bacteroidetes ratio and elevated levels of Bifidobacterium. These changes suggest a specific dysbiosis associated with endometriosis.

 

What are the greatest implications of this study?

The study’s most significant implications lie in its pioneering use of high-throughput DNA sequencing to link endometriosis with specific changes in gut microbiota, highlighting the disease’s potential to induce dysbiosis. It suggests the importance of further research to understand the long-term effects of endometriosis on gut microbiota and the bidirectional interactions between the host and its microbiota. This could lead to novel insights into the pathophysiology of endometriosis and inform new therapeutic strategies targeting the gut microbiome.

Endometriosis induces gut microbiota alterations in mice

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study demonstrates that endometriosis induces gut microbiota alterations in a murine model, particularly increasing the Firmicutes/Bacteroidetes ratio and enriching Bifidobacterium and Parasutterella. These shifts suggest dysbiosis as a contributing factor to inflammation and immune dysregulation, supporting the potential for microbiota-targeted therapies in endometriosis management.

What Was Studied?

This study investigated the effects of endometriosis on gut microbiota composition in a murine model, specifically evaluating microbial shifts during the progression of endometriosis. Researchers employed a prospective and randomized design, inducing endometriosis in mice through intraperitoneal injection of endometrial tissues. The primary aim was to characterize changes in gut microbiota over time, utilizing 16S ribosomal-RNA gene sequencing to assess microbial diversity and composition at 7, 14, 28, and 42 days post-induction. The experiment included mock groups as controls, which received saline injections instead of endometrial tissue, to account for any procedural effects.

Who Was Studied?

The study involved C57BL6 mice, a commonly used murine model, to mimic endometriosis development. Mice were divided into two groups: those receiving endometrial tissue injections to induce endometriosis, and mock controls receiving only saline. The animals were sacrificed at four different time points (7, 14, 28, and 42 days) for fecal sample collection and microbiota analysis. Researchers conducted 16S rRNA sequencing on these samples to evaluate alterations in microbial communities associated with endometriosis progression.

What Were the Most Important Findings?

The study revealed that endometriosis induced significant alterations in gut microbiota composition, particularly at 42 days post-induction. Beta diversity analysis demonstrated that the microbial community structure diverged substantially from the mock controls, indicating dysbiosis. At the phylum level, there was an increased Firmicutes/Bacteroidetes ratio, a hallmark often linked to inflammatory conditions. Furthermore, Actinobacteria and Betaproteobacteria were more abundant in the endometriosis group, whereas Bacteroidetes was more dominant in the control group. At the genus level, the study identified increases in Ruminococcaceae-UGG-014, Bifidobacterium, and Parasutterella among endometriosis mice. These microbial shifts suggest that endometriosis disrupts normal gut microbial homeostasis, potentially influencing systemic inflammation and immune modulation. The researchers noted that while alpha diversity remained similar between groups, the specific microbial composition shifted dramatically over the 42-day period. This timeline suggests that gut dysbiosis in endometriosis is progressive and may exacerbate immune system imbalances over time.

Taxonomic LevelMicrobiota Findings in Endometriosis-Induced Mice
Phylum LevelIncreased Firmicutes/Bacteroidetes ratio. Elevated levels of Actinobacteria and Betaproteobacteria.
Class LevelEnhanced representation of Clostridia and Actinobacteria classes.
Order LevelNotable increase in Lactobacillales and Clostridiales.
Family LevelSignificant enrichment of Ruminococcaceae and Bifidobacteriaceae.
Genus LevelMarked increases in Ruminococcaceae-UGG-014, Bifidobacterium, and Parasutterella.
Alpha DiversityNo significant difference in microbial richness or evenness compared to controls.
Beta DiversitySignificant divergence from mock controls, indicating altered microbial community structure.
Inflammatory AssociationsAltered microbiota profile is linked to systemic inflammation and immune modulation, suggesting a role in endometriosis progression.

What Are the Greatest Implications of This Study

The findings underscore the role of gut microbiota dysbiosis in the progression of endometriosis, revealing distinct shifts in microbial populations, especially an elevated Firmicutes/Bacteroidetes ratio. These changes mirror dysbiosis seen in other inflammatory diseases, suggesting that gut microbiota may contribute to systemic inflammation and immune dysfunction in endometriosis. The study highlights the potential for microbiota-targeted therapies to restore gut microbial balance as a therapeutic approach. Additionally, the identification of enriched genera such as Bifidobacterium and Parasutterella suggests potential biomarkers for non-invasive diagnostics. The progressive nature of microbiota alteration observed at 42 days further indicates that early intervention targeting microbial communities could mitigate inflammatory responses and possibly slow disease progression. This research provides a mechanistic link between gut dysbiosis and endometriosis pathology, paving the way for microbiome-based therapeutic strategies.

Endometriosis of the lung: A case report and brief review of the literature

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The study analyzed 12 cases of pulmonary endometriosis, identifying two distinct groups based on lesion characteristics and reproductive history, exploring potential embolic and pleural origins, and emphasizing the need for further research into the pathogenesis and clinical management of this rare condition.

What was studied?

The study focused on pulmonary endometriosis, a rare condition characterized by the presence of endometrial tissue in the lungs. It analyzed a series of historical and recent cases, dividing them into two distinct groups based on the characteristics of the lung lesions and their association with pregnancy and endometrial glands. The study aimed to explore the pathology and potential pathogenesis of these lesions in the lungs, investigating whether they could be of embolic origin and considering their relationship with the pleura.

 

Who was studied?

The individuals studied were women who had cases of histologically confirmed pulmonary endometriosis. This included a total of 12 cases, with the subjects ranging in status from pregnant to post-menopausal. Six of these cases involved women who died during pregnancy or shortly after delivery, presenting with small decidua deposits in the lungs. The other six involved patients with larger lung lesions containing endometrial glands, observed in surgically resected lung specimens. Among these, at least five had a history of previous pregnancies.

 

What were the most important findings?

The study revealed several significant findings regarding pulmonary endometriosis. First, it identified two distinct groups of lesions, categorized based on their characteristics and the reproductive history of the patients. The first group comprised smaller lesions known as decidual deposits. These were likely embolic in origin and were characterized by their lack of contact with the pleura. Conversely, the second group consisted of larger lesions situated directly beneath the pleura and containing endometrial glands, suggesting a potential pleural origin or growth extending from the pleura into the lung substance. Additionally, it was observed that the larger and older lesions in this group were more likely to contain endometrial glands, unlike the smaller, more recent lesions. This differentiation in lesion characteristics provides critical insights into the pathogenesis and clinical presentation of pulmonary endometriosis.

 

What are the greatest implications of this study?

The implications of this study are significant in understanding the etiology and pathogenesis of pulmonary endometriosis:

 

Category Implication
Pathogenetic Insights The study suggests different pathways for the formation of endometrial tissue in the lungs, possibly linked to embolic phenomena or metaplastic transformation. These pathways could vary based on the patient’s reproductive history and the timing of lesion formation.
Diagnostic Considerations The identification of two distinct groups of lesions aids in understanding their clinical presentations, which can guide diagnostic and treatment strategies. This is particularly important in distinguishing the origins and nature of lung lesions in women with or without a clear history of endometriosis.
Clinical Management The study emphasizes the need for heightened awareness of this rare condition in the differential diagnosis of lung lesions. This is especially crucial for women with a history of endometriosis or those experiencing unusual pulmonary symptoms correlated with menstrual cycles.
Research Directions The findings highlight existing gaps in understanding and underscore the need for further research into the mechanisms of endometrial tissue migration and implantation within the lung. This research could provide insights into other forms of extrapelvic endometriosis.

 

Epidemiology of Premenstrual Syndrome (PMS)-A Systematic Review and Meta-Analysis Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This systematic review and meta-analysis estimate the global prevalence of PMS, revealing wide variations across countries and highlighting the need for standardized diagnostic approaches.

What was reviewed?

This paper presents a systematic review and meta-analysis of the global prevalence of Premenstrual Syndrome (PMS). It focuses on synthesizing existing studies to determine the overall rate of PMS occurrence among women, analyzing factors influencing its prevalence, and exploring variations in prevalence rates across different countries and regions. The review compiles data from multiple sources to estimate the global burden of PMS and identify trends over time, employing meta-regression to examine factors that might affect PMS prevalence.

Who was reviewed?

The review examines data from studies involving women of reproductive age, specifically those diagnosed with PMS based on various symptom screening tools, such as the Premenstrual Symptoms Screening Tool (PSST), and other diagnostic scales. The included studies span different regions and countries, offering a broad view of PMS prevalence across diverse populations.

What were the most important findings?

The systematic review and meta-analysis found that the pooled prevalence of PMS across 17 studies was 47.8%, with substantial variation between different countries. The lowest prevalence was reported in France (12%), while Iran had the highest (98%). This wide range of prevalence is indicative of various factors, such as different diagnostic criteria, sample populations, and cultural or environmental influences on PMS reporting and diagnosis. The review also highlighted a trend of increasing PMS prevalence between 1996 and 2011, though the correlation with the year of study was not statistically significant.

Meta-regression analysis revealed a significant correlation between the sample size and the reported prevalence of PMS, with larger studies tending to report lower prevalence rates. The review also noted that the differences in measurement tools used to diagnose PMS across studies could contribute to the observed variability in prevalence. The results underscore the need for standardized diagnostic criteria and more comprehensive studies to better understand the factors driving PMS prevalence globally.

What are the greatest implications of this review?

The findings of this review have significant implications for public health and clinical practice. The high global prevalence of PMS, with nearly half of reproductive-aged women affected, underscores the need for effective diagnostic and management strategies. Clinicians should be aware of the significant variation in PMS prevalence, influenced by geographical and methodological factors, which can impact patient care and treatment approaches. The review emphasizes the importance of further research to standardize diagnostic tools and explore the role of environmental, cultural, and genetic factors in PMS. Moreover, the findings suggest that larger, high-quality studies are needed to provide more reliable data on PMS prevalence, which can inform public health policies and interventions aimed at improving women's reproductive health globally.

Epigenetics of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This paper reviewed the role of epigenetics in endometriosis, exploring DNA methylation and histone modifications as key factors. It highlighted potential diagnostic, prognostic, and therapeutic applications, including non-surgical treatments like histone deacetylase inhibitors, suggesting a promising shift towards targeted molecular approaches in managing the disease.

What was studied?

The review focused on the role of epigenetic changes in endometriosis, exploring how these modifications influence the disease’s etiopathogenesis, diagnosis, and therapeutic approaches. It reviewed existing evidence up to June 2009, linking epigenetic aberrations such as DNA methylation and histone modification to endometriosis, and discussed the potential of these aberrations in developing non-surgical medical therapies for the condition.

 

Who was studied?

The review examined accumulated research data from various studies on endometriosis. This included an analysis of epigenetic changes in the DNA and histone modifications of cells from women affected by endometriosis. It highlighted findings from in vitro studies and also drew insights from broader genetic studies related to hormone functions and immune responses that contribute to endometriosis.

 

What were the most important findings?

Epigenetic Basis of Endometriosis: There is substantial evidence suggesting that epigenetic mechanisms play a significant role in the development and progression of endometriosis, particularly through DNA methylation and histone modifications.

Diagnostic and Prognostic Potential: DNA methylation markers and microRNA (miRNA) profiles offer promising avenues for early diagnosis and prognosis of endometriosis, potentially allowing for better management of the disease.

Therapeutic Innovations: Histone deacetylase inhibitors (HDACIs) have shown promise in in vitro studies as potential treatments for endometriosis by reversing epigenetic aberrations.

 

What are the greatest implications of this review?

The review posits that understanding and manipulating epigenetic changes could lead to significant advances in treating endometriosis. Specifically, epigenetic therapies could provide new, non-surgical options for managing the disease, potentially reducing the need for repetitive surgeries and improving the quality of life for patients. Additionally, epigenetic markers may revolutionize the diagnosis and monitoring of endometriosis, making it possible to detect the disease earlier and tailor treatments more effectively. Overall, the study suggests a shift towards more targeted molecular therapies and diagnostics in endometriosis care, highlighting the need for further research into epigenetic mechanisms.

Evaluation of the relationship between polycystic ovary syndrome and intestinal inflammation as measured by fecal calprotectin levels

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

PCOS is linked with higher gut inflammation, as shown by elevated fecal calprotectin. This may help guide diagnosis and treatment.

What was studied?

The authors investigated whether intestinal inflammation plays a role in polycystic ovary syndrome (PCOS) by analyzing fecal calprotectin levels, a noninvasive biomarker that reflects neutrophil-driven gut inflammation. Given the increasing evidence that inflammation and gut dysbiosis contribute to PCOS, the researchers aimed to determine if elevated calprotectin could serve as an additional indicator of disease presence or severity.

Who was studied?

The study included 54 adult women: 27 with PCOS and 27 healthy controls. All participants were of reproductive age and had a body mass index within the normal range. The authors excluded individuals with gastrointestinal disorders, systemic illness, or recent antibiotic use to isolate the relationship between PCOS and gut inflammation.

What were the most important findings?

Women with PCOS had significantly higher fecal calprotectin levels compared to healthy controls, suggesting greater intestinal inflammation. Interestingly, systemic inflammation, measured by standard markers like hs-CRP, was similar across groups, indicating that the inflammation in PCOS may be localized to the gut. Although calprotectin wasn’t an independent predictor of PCOS in statistical models, it showed excellent specificity. This means it could help differentiate between PCOS and non-PCOS individuals in clinical settings. The findings support that microbial shifts and increased intestinal permeability—hallmarks of gut dysbiosis—may underlie some aspects of PCOS. Elevated calprotectin levels point toward neutrophil activity in the intestinal lining, often triggered by changes in gut microbiota.

What are the greatest implications of this study?

The study underscores the potential role of intestinal inflammation in PCOS and highlights fecal calprotectin as a promising, low-cost marker that could aid diagnosis or monitoring. These findings open the door to new interventions, such as microbiome-targeted therapies, to manage PCOS symptoms. If confirmed in future studies, strategies that reduce gut inflammation might improve hormonal balance and fertility outcomes in PCOS patients. It also reinforces the value of including microbiome-related biomarkers in gynecological evaluations.

Evidence for statin therapy in polycystic ovary syndrome. 

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

This review highlights the potential benefits of statin therapy in women with polycystic ovary syndrome (PCOS), showing improvements in metabolic dysfunction, hyperandrogenism, and cardiovascular risk factors. Statins may offer a dual benefit for PCOS management, though further research is needed for long-term clinical outcomes.

What was reviewed?

This paper provides a review of the evidence supporting statin therapy for managing polycystic ovary syndrome (PCOS), focusing on its potential to reduce cardiovascular risks and address some of the metabolic complications associated with PCOS. The review discusses both the lipid-lowering effects of statins and their pleiotropic effects, including improvements in insulin resistance, hyperandrogenemia, and systemic inflammation. These secondary benefits may offer additional therapeutic value for women with PCOS, a condition commonly linked with metabolic and cardiovascular disturbances.

Who was reviewed?

The review examined existing studies and clinical trials investigating the use of statins in PCOS patients. The studies reviewed explored the effectiveness of statins like atorvastatin and simvastatin in reducing various metabolic and biochemical markers in women with PCOS, such as testosterone levels, insulin resistance, and inflammation. The review focused on understanding how statins could be beneficial in managing the hormonal and metabolic dysfunctions seen in PCOS.

What were the most important findings?

The review revealed promising evidence supporting statin therapy for women with PCOS, particularly due to its pleiotropic effects. Statins were shown to improve the lipid profile in women with PCOS, reducing LDL cholesterol levels, which is crucial given the elevated cardiovascular risks associated with the condition. Beyond lipid-lowering, statins also contributed to significant reductions in hyperandrogenemia, insulin resistance, and markers of systemic inflammation such as C-reactive protein (CRP). These findings suggest that statins could offer a dual benefit by improving both metabolic and reproductive parameters in women with PCOS.

Moreover, the review highlighted that statins like atorvastatin and simvastatin have comparable effects on testosterone reduction, an important aspect of managing hyperandrogenism in PCOS. The use of statins led to a decrease in testosterone levels that was similar to the effects of established antiandrogens. This effect was observed independently of the improvement in lipid profiles, which underscores the potential of statins to address some of the hormonal imbalances seen in PCOS.

What are the greatest implications of this review?

The findings from this review suggest that statins may be a valuable addition to the treatment options for PCOS, especially for women who are at high risk of cardiovascular disease due to the metabolic disturbances commonly seen in the condition. The reduction in hyperandrogenemia, improvement in insulin sensitivity, and decrease in inflammation could provide significant therapeutic benefits, particularly for those who have not responded well to other treatments like insulin sensitizers or antiandrogens. However, the review also emphasized the need for further large-scale studies to validate the long-term efficacy of statins in improving fertility outcomes and reducing cardiovascular events in women with PCOS. The potential teratogenic risks of statins, particularly during pregnancy, warrant caution and a careful approach to their use in reproductive-age women.

Exploring the link between dietary zinc intake and endometriosis risk: insights from a cross-sectional analysis of American women

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • STOPs
    STOPs

    A STOP (Suggested Termination Of Practices) is a recommendation that advocates for the discontinuation of certain medical interventions, treatments, or practices based on emerging evidence indicating that these may be ineffective, harmful, or counterproductive in the management of specific conditions.

This study links higher dietary zinc intake with increased endometriosis risk among American women, highlighting zinc’s complex role in immune modulation and estrogen-related pathways. Findings emphasize the importance of balanced intake for managing endometriosis risk.

What was studied?

This study investigated the association between dietary zinc intake and the risk of endometriosis among American women. Using cross-sectional data from the National Health and Nutrition Examination Survey (NHANES) collected between 1999 and 2006, the researchers aimed to evaluate whether zinc intake, as a key nutritional factor, was linked to the prevalence of endometriosis. Zinc is known for its essential roles in immune modulation, antioxidative defense, and regulation of matrix metalloproteinases (MMPs), all of which are implicated in endometriosis progression.

Who was studied?

The study included 4,315 American women aged 20–54 years, of whom 331 were diagnosed with endometriosis based on self-reported doctor diagnoses. Participants’ dietary zinc intake was assessed using 24-hour dietary recall interviews, with additional data on demographics, lifestyle, and health covariates collected. Women with extreme caloric intakes or incomplete data were excluded to ensure robustness of results.

What were the most important findings?

The study revealed a positive correlation between higher dietary zinc intake and the risk of endometriosis. Women consuming over 14 mg/day of zinc had a significantly higher adjusted odds ratio (1.60, 95% CI: 1.12–2.27, p = 0.009) compared to those with intake ≤8 mg/day. Zinc’s dual role in immune modulation and antioxidative defense was emphasized, particularly its regulation of matrix metalloproteinases (MMPs) like MMP-2 and MMP-9, which are key enzymes in tissue remodeling and endometriotic lesion invasion. Interestingly, despite zinc’s known antioxidative and anti-inflammatory roles, excessive intake appeared to have a counterproductive effect. These nuanced findings highlight zinc’s complex role in endometriosis pathophysiology.

What are the greatest implications of this study?

This research underscores the potential for dietary zinc as both a marker and modifiable factor in endometriosis risk. It raises questions about zinc’s dualistic effects, where optimal levels may support immune health, but excess intake could exacerbate estrogen-related pathways in endometriosis. Clinicians should be cautious when recommending zinc supplementation for reproductive health, particularly in populations at risk for endometriosis. Furthermore, this study strengthens the biological plausibility of microbiome involvement in endometriosis, as zinc is a crucial cofactor for microbial activity, and its imbalance may alter the gut and pelvic microbiota implicated in the disease.

Fecal Microbiota Transplantation: A Potential Tool for Treatment of Human Female Reproductive Tract Diseases

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study evaluates the therapeutic potential of fecal microbiota transplantation (FMT) for treating female reproductive tract diseases, including PCOS, by restoring microbiota balance.

What was studied?

This study investigated the potential of Fecal Microbiota Transplantation (FMT) as a therapeutic tool for treating female reproductive tract diseases, particularly polycystic ovary syndrome (PCOS), endometriosis, and bacterial vaginosis (BV). The research explored the relationship between gut and female genital microbiota, evaluating whether microbiota alterations in the gut could influence female reproductive tract health. The therapeutic potential of FMT for restoring microbiota balance was examined through preclinical and clinical evidence supporting its application in treating these disorders.

Who was studied?

The study focused on the human female reproductive tract, with an emphasis on the gut–female tract microbiota connection. It also involved exploring the use of FMT for managing reproductive diseases in women. The study reviewed the microbiota composition in the female reproductive system, including the vagina and uterus, and its impact on conditions like PCOS, endometriosis, and BV. While not directly involving human subjects in this particular research, the review draws on existing preclinical models and clinical studies.

What were the most important findings?

The most important findings of this study point to the significant influence of gut microbiota on female reproductive health. The gut and female reproductive tract microbiota are interconnected, with specific bacterial patterns associated with reproductive disorders like PCOS, endometriosis, and BV. This suggests that alterations in gut microbiota composition can influence the health of the female reproductive system. Additionally, FMT has shown promise as a potential treatment for restoring microbiota balance in these conditions. Studies indicate that FMT can be an effective therapy for Clostridium difficile infections and may extend to other systemic diseases, including reproductive tract diseases.

From a microbiome perspective, the study highlighted the critical role of microbial modulation in regulating immune responses, particularly in the female reproductive tract. The therapeutic effect of FMT could involve rebalancing the vaginal and uterine microbiota, potentially alleviating symptoms associated with PCOS and BV, and addressing the chronic inflammation and hormonal dysregulation in these conditions.

What are the greatest implications of this study?

The study's findings open a promising new therapeutic avenue for managing female reproductive disorders through FMT. Given the close relationship between the gut microbiota and the female reproductive tract, FMT could represent a groundbreaking treatment option for diseases such as PCOS, endometriosis, and BV. The research also suggests that restoring a healthy microbiota balance via FMT could help alleviate oxidative stress, inflammatory markers, and other metabolic dysfunctions commonly seen in these conditions. This discovery may pave the way for more targeted, microbiome-based treatments, enhancing clinical outcomes for women with reproductive health issues. It also highlights the importance of personalized care, considering the unique microbial signature of each patient.

Fruit and vegetable consumption and risk of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • STOPs
    STOPs

    A STOP (Suggested Termination Of Practices) is a recommendation that advocates for the discontinuation of certain medical interventions, treatments, or practices based on emerging evidence indicating that these may be ineffective, harmful, or counterproductive in the management of specific conditions.

Higher fruit intake, especially citrus fruits, was inversely associated with laparoscopically confirmed endometriosis, suggesting a protective effect potentially linked to beta-cryptoxanthin. In contrast, cruciferous vegetables were linked to increased risk, highlighting the complex interplay between diet and endometriosis risk factors.

What Was Studied

This study explored the potential link between the consumption of fruits and vegetables and the risk of laparoscopically confirmed endometriosis. Using data collected from the Nurses' Health Study II, the researchers analyzed dietary habits over a 22-year period, investigating whether certain food groups and nutrients influenced the likelihood of developing endometriosis.

Who Was Studied

Participants included premenopausal women aged 25–42 years who were enrolled in the Nurses' Health Study II cohort. These women completed biennial surveys assessing health status, lifestyle factors, and dietary intake. Those with a history of endometriosis, cancer, infertility, or hysterectomy were excluded from the analysis, ensuring a focused evaluation of diet and disease development.

Most Important Findings

The study found an inverse relationship between fruit consumption, particularly citrus fruits, and the risk of endometriosis. Women who consumed citrus fruits frequently were less likely to develop endometriosis. Conversely, no significant association was found between total vegetable intake and the disease. Cruciferous vegetables, however, were unexpectedly linked to an increased risk. Beta-cryptoxanthin, a nutrient found in citrus fruits, appeared to play a protective role, and the beneficial effects of fruit consumption were especially notable among participants who had a history of smoking. These findings suggest a potential role for specific dietary components in either mitigating or exacerbating the risk of endometriosis.

Implications

The findings highlight the importance of dietary considerations in understanding endometriosis risk. The protective association of citrus fruits underscores the potential of targeted nutritional interventions to reduce risk. The increased risk observed with cruciferous vegetables raises questions about the role of gastrointestinal symptoms, as these vegetables are high in fermentable oligosaccharides, which could exacerbate symptoms and lead to increased diagnosis rates. Future studies exploring these dietary patterns in greater depth are warranted to clarify the underlying mechanisms and to guide dietary recommendations for those at risk.

GS2 Gallium Complex: A Novel Inhibitor of MMP-14 for Anti-Metastatic Cancer Therapy

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Research on GS2, a new gallium complex, showed potent inhibition of cell invasion and MMP activity in cancer cells, particularly MMP-14. These findings indicate GS2's strong potential as a therapeutic agent against metastatic cancers by interfering with key processes of cancer cell invasion and matrix degradation.

What Was Studied?

This study examined the effects of [N-(5-chloro-2-hydroxyphenyl)-L-aspartato] chlorogallate (GS2), a water-soluble gallium complex, on tumor cell invasion and the activity and expression of matrix metalloproteinases (MMPs). Specifically, it evaluated GS2's anti-invasive properties and its regulatory effects on MMP-2, MMP-9, and MMP-14 in two human cancer cell lines: metastatic HT-1080 fibrosarcoma and MDA-MB231 breast carcinoma cells.

Who Was Studied?

The research utilized human cell lines HT-1080 (fibrosarcoma) and MDA-MB231 (breast carcinoma). Additionally, MCF7 cells transfected with MMP-14 and non-transfected fibroblast cells (F40) were used for supplemental experiments.

Most Important Findings

GS2 demonstrated significant anti-invasive and anti-MMP activities at non-cytotoxic concentrations. The compound inhibited MMP-2, MMP-9, and MMP-14 activities in a dose-dependent manner, with IC50 values of 168 µM, 82 µM, and 20 µM, respectively. GS2 reduced the mRNA expression of MMP-14 in both cell lines and inhibited MMP-2 and MMP-9 expression exclusively in MDA-MB231 cells. Western blotting confirmed decreased MMP-14 protein expression in response to GS2. Importantly, GS2 significantly inhibited cell invasion through a type-I collagen-coated matrix, correlating with the downregulation of MMP-14, a critical regulator of the extracellular matrix and tumor invasion. Notably, GS2's inhibition of MMP-14 showed specificity for cells expressing higher MMP-14 levels, a hallmark of invasive cancer phenotypes.

Greatest Implications

The findings suggest GS2 is a promising candidate for anti-metastatic therapy targeting MMP-14. This is particularly relevant for cancers characterized by elevated MMP-14 expression, such as type II endometrial adenocarcinoma and invasive pituitary adenomas. GS2’s ability to selectively inhibit MMP-14 and reduce cancer cell invasion positions it as a potential therapeutic for limiting tumor metastasis. Moreover, its low cytotoxicity at effective concentrations highlights its clinical applicability.

Gut and oral microbial compositional differences in women with breast cancer, women with ductal carcinoma in situ, and healthy women

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study reveals distinct gut microbiota profiles in breast cancer and DCIS patients, with reduced alpha diversity and functional shifts linked to inflammation. Major microbial associations, including enriched Bacteroides guilds, underscore potential microbiome-targeted interventions. Oral microbiota showed minimal differences, highlighting the gut's critical role in breast cancer progression.

What was studied?

This study investigated and compared the gut and oral microbiota in three distinct groups: women with breast cancer (BC), women with ductal carcinoma in situ (DCIS), and healthy women. Fecal and oral samples were collected and analyzed using 16S rRNA sequencing to assess microbial diversity, composition, and predicted functional potential.

Who was studied?

The study analyzed samples from 154 women, comprising 73 with BC, 32 with DCIS, and 49 healthy controls. Samples were collected before any therapy to ensure no treatment effects influenced the microbiota.

What were the most important findings?

The study found significant differences in gut microbiota composition and diversity between groups, while the oral microbiota exhibited fewer variations. Women with BC had lower gut microbial alpha diversity compared to healthy women. Beta diversity analysis revealed distinct microbial profiles for the BC and DCIS groups compared to healthy controls. Taxonomic analysis identified several major microbial associations (MMAs) in the gut: the Bacteroides and Enterobacteriaceae guilds were enriched in BC patients, while the Clostridiales guild was more prevalent in healthy women. Functionally, the gut microbiota of BC patients showed increased pathways for lipopolysaccharide (LPS) biosynthesis, glycan metabolism, and sphingolipid metabolism, which are linked to systemic inflammation and cancer progression. Conversely, the oral microbiota showed minimal variation across cohorts, with no significant differences in functional pathways or microbial guilds.

What are the greatest implications of this study?

The findings highlight the role of gut microbiota in breast cancer development and progression. The identification of distinct microbial signatures and functional pathways provides a basis for developing microbiome-targeted interventions aimed at improving treatment outcomes and prognosis. Notably, the lack of significant findings in oral microbiota suggests that gut microbiota might have a more critical role in breast cancer etiology. These results pave the way for further research on microbiome-based diagnostic tools and therapeutic strategies for breast cancer.

Gut and Vaginal Microbiomes in PCOS: Implications for Women’s Health

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review links dysbiosis of gut and vaginal microbiota to PCOS pathogenesis, highlighting reduced Lactobacillus, elevated Bacteroides, and hormonal imbalance. It supports microbiome-targeted therapies like FMT and probiotics as promising interventions, emphasizing the role of microbiota in regulating hormones, immunity, and metabolic function.

What Was Reviewed?

This review paper examined the interrelationship between gut and vaginal microbiota and polycystic ovary syndrome (PCOS), offering a multidimensional analysis of how microbial dysbiosis contributes to the endocrine, metabolic, reproductive, and immune disturbances seen in PCOS. The authors summarized both experimental and clinical studies, with a particular focus on microbiota diversity, microbial shifts in composition, and their functional consequences. The review discussed the role of microbiota in regulating sex hormones, immune homeostasis, insulin sensitivity, gut permeability, inflammation, and neuroendocrine communication via the gut–brain axis. Additionally, the paper explored therapeutic strategies such as fecal microbiota transplantation (FMT) and probiotic interventions, aiming to identify translational opportunities for clinical application.

Who Was Reviewed?

The review compiled findings from human studies, animal models, and in vitro mechanistic research. In human studies, both gut and vaginal microbial profiles were compared between women with PCOS and healthy controls using sequencing techniques such as 16S rRNA analysis. Animal models, including rodent studies, were also incorporated to explore the causal mechanisms by which microbial interventions impact hormonal regulation, insulin resistance, and reproductive health. The reviewed cohorts varied across reproductive age, BMI, and hormonal phenotypes, with many studies focusing on women with hyperandrogenism and menstrual irregularities, the hallmarks of PCOS.

What Were the Most Important Findings?

The review identified significant microbial dysbiosis in both the gut and vaginal microbiota of PCOS patients. In the gut, PCOS was consistently associated with reduced alpha and beta diversity, and increased abundance of pro-inflammatory and metabolically detrimental taxa such as Bacteroides vulgatus, Prevotella copri, and Escherichia/Shigella. Simultaneously, beneficial microbes such as Akkermansia muciniphila and members of Ruminococcaceae were diminished. The vaginal microbiota in PCOS showed decreased Lactobacillus species and increased colonization by pathogens like Gardnerella vaginalis, Prevotella, and Chlamydia trachomatis. These microbial patterns are linked to infertility, implantation failure, and adverse pregnancy outcomes.

Mechanistically, the review described how microbial alterations exacerbate insulin resistance through increased branched-chain amino acids and inflammatory cytokines. It also outlined how gut-derived short-chain fatty acids (SCFAs), bile acids, and estrogen-metabolizing enzymes modulate host endocrine and metabolic functions. Importantly, the review explored the gut–brain axis, implicating microbial metabolites in the modulation of the hypothalamic–pituitary–gonadal axis, contributing to anxiety and reproductive dysfunction in PCOS. These major microbial associations (MMAs) anchor PCOS within a broader systems biology framework, suggesting that dysbiosis impacts not just metabolic markers, but also immune balance, hormone regulation, and reproductive health.

What Are the Implications of This Review?

The implication of this review is its positioning of microbiota as a core regulatory system in the etiology and progression of PCOS, rather than a secondary contributor. For clinicians, this reframing encourages the integration of microbiota profiling into PCOS diagnostics, especially in patients with atypical presentations. The consistent loss of microbial diversity and protective Lactobacillus species, combined with enrichment of inflammatory and hormone-disrupting taxa, provides a microbiome-based signature of PCOS. Therapeutically, the review highlights emerging interventions, including FMT and targeted probiotics as viable approaches to restore microbial equilibrium. The evidence supports the concept that modulating gut and vaginal microbiota could lead to improvements in insulin sensitivity, hormone balance, and fertility outcomes. However, the authors note that most mechanistic insights stem from animal studies, calling for rigorous human trials to validate these strategies in clinical practice.

Gut microbiome in PCOS associates to serum metabolomics

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study shows that gut microbiota dysbiosis in PCOS correlates with altered serum metabolites, including elevated LPCs and reduced citric acid. Roseburia and Prevotella_9 were linked to protective metabolic profiles, while Escherichia-Shigella was associated with inflammation and lipid imbalance.

What Was Studied?

This cross-sectional study investigated the relationship between gut microbiota composition and serum metabolomic profiles in women with polycystic ovary syndrome (PCOS), using an integrative approach combining 16S rRNA gene sequencing with untargeted serum metabolomics. The researchers aimed to determine how specific microbial taxa in the gut correlate with alterations in circulating metabolites and whether these associations may help explain the metabolic and endocrine disturbances observed in PCOS. This integrative analysis focused on identifying major microbial associations (MMAs) linked with lipid metabolism, energy homeostasis, and inflammatory markers, shedding light on potential mechanisms underlying PCOS pathophysiology.

Who Was Studied?

The study enrolled 20 women with PCOS diagnosed using the Rotterdam criteria and 20 healthy controls from Chengdu, China. Participants were matched in age but differed significantly in body mass index (BMI), serum testosterone, luteinizing hormone (LH), LH/FSH ratio, and fasting insulin levels. All subjects provided blood and stool samples, and filled out SF-36 quality of life assessments. Strict exclusion criteria were applied to control for confounders such as recent antibiotic, probiotic, or hormonal therapy use, and all participants resided in the same geographical region to minimize environmental variability.

What Were the Most Important Findings?

Women with PCOS exhibited marked dysbiosis in their gut microbiota alongside distinct changes in serum metabolites. Microbial alpha diversity was significantly reduced in the PCOS group, and beta diversity analyses confirmed community-level differences. At the phylum level, PCOS patients showed increased Proteobacteria, Verrucomicrobia, and Fusobacteria, while Firmicutes, Bacteroidetes, and Actinobacteria were decreased. Genera such as Escherichia-Shigella, Alistipes, and Megamonas were enriched in PCOS, whereas beneficial taxa like Roseburia and Bifidobacterium were diminished.

Serum metabolomic analysis identified 15 significantly altered metabolites in the PCOS group. Elevated metabolites included various lysophosphatidylcholines, phosphatidylcholine (PC), ganglioside GA2, and 1-linoleoylglycerophosphocholine—all of which are associated with glycerophospholipid metabolism. Meanwhile, levels of nicotinate beta-d-ribonucleotide and citric acid, markers of nicotinamide and TCA cycle metabolism, respectively, were reduced, indicating impaired energy homeostasis and redox imbalance.

Correlation analyses showed that Prevotella_9 was positively associated with beneficial metabolites like citric acid and nicotinate beta-d-ribonucleotide, and negatively correlated with pro-inflammatory LPCs. Roseburia, a known butyrate producer, was negatively correlated with LPC (20:4). These MMAs suggest that reductions in SCFA-producing microbes and expansions of pro-inflammatory taxa contribute to insulin resistance, oxidative stress, and mood disturbances in PCOS.

What Are the Implications of This Study?

This study provides compelling evidence that PCOS is characterized by a dual disturbance: gut microbial dysbiosis and metabolic dysfunction, which are closely intertwined. Clinically, this suggests the potential for microbiota-informed diagnostics and therapeutic strategies in PCOS. The association of key taxa such as Roseburia and Escherichia-Shigella with specific metabolic pathways, including glycerophospholipid and TCA cycle metabolism, provides biological plausibility for microbial modulation of systemic insulin sensitivity and inflammation. Moreover, the co-occurrence of mental health symptoms and microbial shifts tied to indole and serotonin pathways suggests that the gut–brain–ovary axis should be further explored in PCOS.

From a therapeutic standpoint, restoring beneficial microbes and correcting lipid and energy metabolism through targeted probiotics, dietary modulation, or even bile acid-focused interventions may offer a new avenue for comprehensive PCOS management. Future studies using metagenomics, metatranscriptomics, and intervention trials are needed to validate the causality and clinical utility of these microbiome–metabolome signatures.

Gut microbiome in PCOS associates to serum metabolomics: a cross‑sectional study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study identifies a strong link between gut microbiota dysbiosis and altered serum metabolites in PCOS patients. Specific microbial shifts, including elevated Escherichia-Shigella and reduced Roseburia, correlate with lipid and energy metabolism markers, offering insights into PCOS-related insulin resistance and mood disturbances.

What was studied?

This research investigated the relationship between gut microbiota composition and serum metabolites in women diagnosed with polycystic ovary syndrome (PCOS). By employing a cross-sectional study design, the researchers combined untargeted serum metabolomics and 16S rRNA gene sequencing to explore the microbial and metabolic profiles of PCOS patients compared to healthy controls. The primary aim was to understand whether specific gut microbiota shifts were associated with altered metabolic patterns in PCOS and whether these patterns might explain aspects of the syndrome’s pathophysiology, particularly around insulin resistance and mood disorders.

Who was studied?

The study involved 20 women with PCOS and 20 age-matched healthy controls from the Pixian area of Chengdu, China. All participants were carefully screened to exclude confounding variables such as recent use of antibiotics, probiotics, contraceptives, or hormone treatments. The PCOS group was diagnosed using the Rotterdam criteria, a widely accepted diagnostic standard. Clinical characteristics confirmed that PCOS patients showed higher BMI, elevated testosterone, LH, LH/FSH ratios, and fasting insulin levels compared to controls. Their quality of life scores, measured by the SF-36 questionnaire, were notably lower, suggesting a tangible psychosocial impact likely linked to both metabolic and microbial disturbances.

What were the most important findings?

This study revealed two core findings: a distinct gut microbiome signature and a correlated serum metabolite profile in PCOS patients. The gut microbiome of the PCOS group exhibited lower microbial diversity and a specific taxonomic shift marked by higher abundances of Escherichia-Shigella and Alistipes. Conversely, beneficial genera such as Roseburia and Prevotella were reduced. These shifts in microbial populations were significantly correlated with alterations in serum metabolites, especially within the glycerophospholipid metabolism and energy metabolism pathways.

Specifically, PCOS patients demonstrated elevated levels of lysophosphatidylcholine (LPC) variants, phosphatidylcholine (PC), ganglioside GA2, and 1-linoleoylglycerophosphocholine. Meanwhile, metabolites associated with energy metabolism — including citric acid and nicotinate beta-d-ribonucleotide — were significantly reduced. Correlation analyses highlighted that reduced Prevotella_9 was linked to lower levels of these beneficial metabolites and higher levels of the LPC family, suggesting a mechanistic connection between microbial dysbiosis and metabolic dysfunction.

These microbial-metabolite associations potentially contribute to two hallmark features of PCOS: insulin resistance and mood changes. For instance, higher Escherichia-Shigella and Alistipes levels have previously been linked to depression, while increased LPC concentrations are implicated in inflammation and cardiovascular risk, both common comorbidities in PCOS patients.

What are the greatest implications of this study?

This study underscores the potential of microbiome-metabolome interplay as both a diagnostic and therapeutic target for PCOS. The distinct microbial and metabolic profiles identified in this research offer clues about the biological mechanisms underlying PCOS, particularly the role of gut microbiota in modulating lipid metabolism, energy balance, insulin resistance, and mental health. The identification of Escherichia-Shigella and Alistipes as major microbial markers alongside metabolites like LPCs and citric acid opens pathways for non-invasive biomarkers, enabling earlier diagnosis and monitoring of disease progression.

More importantly, the findings pave the way for microbiome-targeted interventions such as targeted probiotics, dietary interventions, or even fecal microbiota transplantation (FMT) to correct dysbiosis, improve metabolic health, and potentially alleviate mood disorders in PCOS patients. Clinicians should consider the gut microbiome as a central component in the metabolic and psychological management of PCOS, especially in cases where standard endocrine treatments provide incomplete relief.

Gut microbiota alterations reveal potential gut–brain axis changes in polycystic ovary syndrome (PCOS)

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study shows reduced gut microbiota diversity and increased GABA-producing bacteria in PCOS, linking microbial shifts to elevated LH levels and supporting a gut–brain axis mechanism. The findings highlight microbial contributions to neuroendocrine dysfunction and suggest therapeutic avenues through microbiome-targeted interventions.

What Was Studied?

This study investigated the alterations in gut microbiota among women with polycystic ovary syndrome (PCOS) and how these changes may relate to neuroendocrine disturbances, particularly through the gut–brain axis. Researchers examined 40 Han Chinese women divided into lean and overweight subgroups. By controlling for diet and anthropometrics, the authors aimed to isolate microbiota-specific differences. Fecal samples were collected for 16S rRNA gene sequencing to evaluate microbial composition, and blood samples were analyzed for metabolic and hormonal parameters, including insulin, glucose, lipid profiles, inflammatory markers, and reproductive hormones. The study also included a dietary intake survey to assess macronutrients and micronutrients. The primary objective was to determine specific microbial taxa associated with PCOS, especially those involved in gamma-aminobutyric acid (GABA) production, and to correlate these microbial shifts with clinical and endocrine markers such as luteinizing hormone (LH) and LH:FSH ratios.

Who Was Studied?

The study examined 40 women of reproductive age, all of Han ethnicity, and recruited from the same geographic region in Southern China. Twenty participants met the revised Rotterdam criteria for PCOS and were further stratified into lean and overweight groups. The remaining 20 participants were healthy controls matched by age and BMI and similarly stratified. All subjects underwent comprehensive assessments, including anthropometric measurements, hormonal profiling, inflammatory markers, glucose tolerance tests, and dietary intake evaluations. Participants had not taken antibiotics, probiotics, hormonal therapies, or insulin sensitizers for at least three months before the study. Fecal microbiota was analyzed via 16S rRNA gene sequencing to compare microbial diversity and species-level abundance across groups.

What Were the Most Important Findings?

This study provided robust evidence of gut microbiota dysbiosis in women with PCOS, revealing a significant reduction in overall microbial richness and diversity compared to controls. The lean control group exhibited the highest alpha diversity, followed by lean PCOS, overweight controls, and overweight PCOS, suggesting a gradient of microbial health associated with both BMI and PCOS status. Importantly, the study identified increased abundance of specific GABA-producing bacteria in women with PCOS. These bacteria positively correlated with elevated LH levels and LH:FSH ratios, which are key endocrine features of PCOS. Notably, Parabacteroides distasonis was significantly increased even in lean women with PCOS, suggesting that this microbial shift is independent of obesity.

The correlation between these microbial species and neuroendocrine markers supports the existence of a gut–brain axis in PCOS pathophysiology. For instance, Parabacteroides distasonis has previously been shown to increase GABA levels in the brain in murine models, and GABA is known to stimulate GnRH neurons and increase LH secretion. The link between elevated GABA-producing microbes and hypersecretion of LH adds biological plausibility to the idea that microbial metabolites may modulate reproductive hormone axes. In addition, Escherichia coli positively correlated with 2-hour postprandial insulin and negatively with HDL-C, aligning with its association with metabolic dysfunction. These major microbial associations (MMA) reveal microbial targets relevant to both metabolic and reproductive pathways in PCOS.

What Are the Greatest Implications of This Study?

This study introduces a novel and biologically compelling mechanism linking gut microbiota with the neuroendocrine dysregulation seen in PCOS. By identifying GABA-producing microbes as potential modulators of LH secretion through the gut–brain axis, the findings extend current understanding beyond metabolic inflammation and insulin resistance. For clinicians, these results underscore the importance of considering gut microbial signatures when evaluating PCOS, particularly for patients whose symptoms do not align neatly with traditional metabolic phenotypes. This study also suggests that microbial modulation, via diet, probiotics, or targeted microbiome interventions, may eventually serve as a therapeutic strategy to influence both reproductive and metabolic outcomes. Finally, this work lays the groundwork for future studies exploring causality and therapeutic manipulation, including fecal microbiota transplantation or metabolomic profiling of microbial products like GABA in PCOS contexts.

Gut microbiota imbalance and its correlations with hormone and inflammatory factors in patients with stage 3/4 endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This case control study explored the gut microbiota in stage 3/4 endometriosis (EM) by comparing fecal and blood samples from 12 EM patients and 12 controls using 16S rRNA sequencing. Results showed reduced α diversity and an increased Firmicutes/Bacteroidetes ratio in EM patients, with notable taxonomic differences and elevated estradiol and IL-8 levels. The study suggests microbiota-related pathways may influence EM, indicating directions for further research.

What was studied?

The study investigated the role of gut microbiota in endometriosis (EM), focusing on its differences between individuals with stage 3/4 EM and healthy controls and how these differences correlate with serum hormone levels and inflammatory cytokines.

 

Who was studied?

The research involved 12 patients diagnosed with stage 3/4 endometriosis and 12 healthy control subjects. The researchers compared their gut microbiota compositions and measured serum levels of hormones and inflammatory cytokines.

 

What were the most important findings?

Key findings included a lower α diversity of gut microbiota and a higher Firmicutes/Bacteroidetes ratio in the EM group compared to controls. Significant differences in the abundances of various taxa were observed, along with higher serum levels of estradiol (E2) and interleukin-8 (IL-8) in the EM group. The study also identified correlations between specific microbial abundances and levels of estradiol and IL-8.

 

What are the greatest implications of this study?

The study’s implications suggest that the gut microbiota may play a significant role in the pathophysiology of endometriosis through its influence on hormonal and inflammatory pathways. These findings open potential avenues for novel therapeutic strategies targeting the gut microbiota in endometriosis management and highlight the need for further research to verify and expand upon these preliminary observations.

Gut microbiota imbalance and its correlations with hormone and inflammatory factors in patients with stage 3/4 endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study explores the gut microbiota imbalance and its correlations with hormone and inflammatory factors in stage 3/4 endometriosis. Key findings reveal distinct microbial shifts linked to hormonal dysregulation and inflammation, offering insights into disease mechanisms and potential microbiome-targeted therapeutic approaches.

What Was Studied?

This study explored the associations between gut microbiota imbalances and hormone and inflammatory factors in patients with stage 3/4 endometriosis (EM). Conducted at Changhai Hospital, Shanghai, the research aimed to determine how gut microbiome alterations correlate with hormone levels and inflammatory markers in women suffering from moderate to severe endometriosis. Using 16S rRNA high-throughput sequencing, researchers analyzed stool samples to compare the gut microbial composition between 12 women diagnosed with stage 3/4 EM and 12 healthy controls. Blood samples were collected to measure serum hormone levels, including estradiol (E2), and inflammatory cytokines, notably IL-8. The primary objective was to identify microbial shifts associated with EM and understand their correlation with hormone imbalances and inflammation, key factors in the pathogenesis of endometriosis.

Who Was Studied?

The study recruited 12 women with a histological diagnosis of stage 3/4 endometriosis from Changhai Hospital and 12 healthy controls, matched for age (18–40 years) and menstrual regularity. Inclusion criteria for the EM group required confirmed diagnoses of moderate to severe endometriosis per the American Fertility Society Revised Classification (1997). All participants were Han women living in Shanghai, with strict exclusion criteria including recent antibiotic or probiotic use, hormonal therapy, pregnancy, and any comorbid gastrointestinal conditions like inflammatory bowel disease. To minimize confounding factors, participants followed a uniform carbohydrate-based diet three days before sampling, and stool samples were collected within three to five days post-menstruation to account for hormonal fluctuation.

What Were the Most Important Findings?

The study found that women with stage 3/4 endometriosis exhibited a significantly altered gut microbiota profile compared to healthy controls. Notably, the EM group had lower α diversity, indicating reduced microbial richness and variation. At the phylum level, the ratio of Firmicutes to Bacteroidetes was markedly increased in endometriosis patients (3.55 vs. 1.99 in controls), suggesting dysbiosis. The abundance of Actinobacteria, Cyanobacteria, Saccharibacteria, Fusobacteria, and Acidobacteria was significantly higher in the EM group, while Tenericutes were significantly reduced. At the genus level, Bifidobacterium, Blautia, Dorea, Streptococcus, and [Eubacterium] hallii_group showed notable increases, whereas Lachnospira and [Eubacterium] eligens_group were depleted in endometriosis patients. Among the unique genera, Prevotella_7 dominated the EM group, while Coprococcus_2 was prevalent in controls.

Additionally, serum analyses revealed that estradiol (E2) and IL-8 levels were significantly higher in endometriosis patients. Correlation analysis indicated that Blautia and Dorea were positively correlated with elevated E2 levels, while Subdoligranulum abundance inversely correlated with IL-8 levels. These microbial shifts also corresponded with enhanced expression of microbial pathways related to "environmental information processing," "endocrine system," and "immune system," highlighting potential links between gut microbiota and hormonal regulation in endometriosis.

What Are the Greatest Implications of This Study?

The findings of this study suggest that gut microbiota imbalances are closely linked with hormone and inflammatory dysregulation in patients with stage 3/4 endometriosis. The observed microbial shifts, particularly the elevated Firmicutes/Bacteroidetes ratio and increased levels of Bifidobacterium, Blautia, Dorea, and Streptococcus, indicate a state of dysbiosis that may exacerbate inflammatory responses and hormonal imbalances. The positive correlation between Blautia and Dorea with estradiol levels points to the gut microbiome's role in modulating estrogen, potentially influencing the development and progression of endometriosis. Furthermore, the association of Subdoligranulum with IL-8 levels suggests a microbial influence on inflammatory cytokine production, which is known to contribute to endometriosis pathophysiology. These insights provide a foundation for exploring microbiome-targeted therapies aimed at restoring microbial balance and modulating hormonal and inflammatory responses in endometriosis patients. This study also underscores the need for further clinical investigations to validate these microbial markers as diagnostic or therapeutic targets.

Heavy Metals and Essential Elements in Association with Oxidative Stress in Women with Polycystic Ovary Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This systematic review links heavy metal exposure and trace element deficiencies to oxidative stress, inflammation, and hormonal imbalance in PCOS, with implications for metabolic dysfunction and gut microbiome disruption.

What was reviewed?

This systematic review synthesized findings from 15 human studies to evaluate the association between heavy metals, essential trace elements, and oxidative stress (OS) in women with polycystic ovary syndrome (PCOS). The review aimed to determine whether elevated toxic metal exposure and imbalances in essential micronutrients contribute to PCOS pathophysiology through mechanisms involving oxidative damage and inflammation. The authors used PubMed to identify literature from January 2008 to April 2023 and included studies that examined both heavy metals and essential elements in relation to markers of oxidative stress and metabolic and endocrine function in PCOS.

Who was reviewed?

The review encompassed studies involving women of reproductive age diagnosed with PCOS, compared to healthy controls. Across the 15 studies, sample sizes varied from small clinical trials to larger observational cohorts (up to 150 participants). The review focused on blood-based assessments (serum or plasma) of both toxic metals and essential elements, and linked these exposures to metabolic parameters, inflammatory biomarkers (e.g., hs-CRP, TNF-α), and OS markers (e.g., MDA, TAC, SOD, GSH).

What were the most important findings?

The review consistently found that women with PCOS exhibit elevated levels of heavy metals such as cadmium (Cd), lead (Pb), mercury (Hg), arsenic (As), antimony (Sb), tellurium (Te), thallium (Tl), and osmium (Os), while having significantly lower levels of essential elements like zinc (Zn), selenium (Se), and magnesium (Mg). The data reveal that these toxic metals are positively associated with markers of OS and inflammation, and negatively associated with antioxidant capacity. Specifically, Cd, Pb, and Sb levels were strongly correlated with higher fasting blood glucose, HOMA-IR, and pro-inflammatory markers, pointing to a direct metabolic and inflammatory insult. In contrast, supplementation with zinc, selenium, magnesium, and chromium showed protective effects, improving TAC levels, reducing oxidative stress markers, and lowering serum levels of androgens like DHEA and testosterone.

From a microbiome perspective, many of these toxicants, particularly cadmium and lead, are known to disrupt gut microbial balance by suppressing beneficial SCFA-producing bacteria such as Faecalibacterium prausnitzii and Bifidobacterium. This dysbiosis can fuel systemic inflammation and insulin resistance, exacerbating PCOS symptoms. Zinc and selenium support mucosal immunity and microbial diversity, and their deficiency may further impair gut barrier integrity and host–microbiome interactions.

What are the greatest implications of this review?

This review confirms that environmental exposure to heavy metals, alongside deficiencies in essential micronutrients, contributes to oxidative stress, inflammation, and metabolic dysfunction in PCOS. These findings underscore the importance of incorporating toxicological and nutritional evaluations into PCOS management. Clinically, there is a rationale for screening PCOS patients for metal burden and micronutrient status. Therapeutic strategies such as targeted supplementation (e.g., zinc, magnesium, selenium) or chelation, as well as dietary interventions to reduce toxicant exposure, may not only alleviate metabolic and endocrine symptoms but also support gut microbiome restoration. The integration of environmental health with endocrinology and microbiome research provides a promising, systems-level approach for improving outcomes in women with PCOS.

High-Fiber Diet or Combined With Acarbose Alleviates Heterogeneous Phenotypes of Polycystic Ovary Syndrome by Regulating Gut Microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

A high-fiber diet combined with acarbose improved hormonal, metabolic, and inflammatory markers in women with PCOS by remodeling the gut microbiota. Key microbial shifts included increases in Bifidobacterium and reductions in Bacteroides vulgatus, correlating with decreased insulin resistance and hyperandrogenism.

What was studied?

This randomized controlled clinical trial investigated the effects of a high-fiber diet, alone or combined with the alpha-glucosidase inhibitor acarbose, on the clinical phenotypes of polycystic ovary syndrome (PCOS) through modulation of the gut microbiota. The study specifically aimed to determine how dietary fibers and delayed carbohydrate absorption impact hormonal, metabolic, and inflammatory markers, as well as gut microbiome composition, in women with PCOS.

Who was studied?

Twenty-five women diagnosed with PCOS according to the Rotterdam criteria were recruited and randomly assigned into two groups. Fourteen participants received a whole-grain, traditional Chinese medicinal, and prebiotic-rich high-fiber diet (WTP diet), while eleven received the same diet combined with acarbose. The intervention lasted 12 weeks. All participants were treatment-naïve and not on hormone therapy, insulin sensitizers, or antibiotics for at least three months prior to enrollment. The study population spanned a reproductive age range (15–41 years), and measurements included hormonal, glycolipid, inflammatory, and microbiota parameters at weeks 0, 4, 8, and 12.

What were the most important findings?

Both interventions improved PCOS clinical phenotypes, but the combination of a high-fiber diet and acarbose yielded significantly better outcomes in lowering testosterone, LH/FSH ratio, fasting insulin, and HOMA-IR. Moreover, participants in the combination group experienced a more pronounced reduction in ovarian volume, hirsutism score, and immature follicles. These improvements paralleled significant shifts in the gut microbiota.

Gut microbiota analysis revealed enrichment of beneficial taxa, including Bifidobacterium and Lactobacillus, which were negatively associated with PCOS-related markers such as testosterone, LH/FSH ratio, fasting insulin, leptin, and a-AGP, while positively correlated with anti-inflammatory markers like adiponectin and spexin. In contrast, CAGs rich in Bacteroides vulgatus, Alistipes, Bilophila, Lachnospira, and Roseburia were significantly inhibited, particularly in the combination group, and positively associated with hyperandrogenism, insulin resistance, and inflammatory markers. These microbial changes suggest that the beneficial effects were mediated through enhanced SCFA production, gut-brain peptide modulation, and reduced LPS-related inflammation.

What are the greatest implications of this study?

This study provides compelling clinical evidence that gut microbiota-targeted nutritional therapy, particularly high-fiber intake enhanced with acarbose, can modulate endocrine and metabolic disturbances in PCOS. By enriching SCFA-producing probiotics like Bifidobacterium and Lactobacillus, and suppressing pro-inflammatory taxa such as Bacteroides vulgatus and Alistipes, the intervention directly impacted core pathogenic mechanisms: hyperandrogenism and insulin resistance. Additionally, changes in gut–brain peptides like leptin, spexin, and orexin highlight a robust gut-brain axis involvement in PCOS pathophysiology. For clinicians, this trial supports incorporating microbiome-informed dietary strategies, including the use of prebiotic-rich foods and agents like acarbose, into PCOS management. While limited by small sample size, the mechanistic depth, hormonal modulation, and microbial specificity make a strong case for larger multicenter trials to validate this therapeutic paradigm.

I Am the 1 in 10—What Should I Eat? A Research Review of Nutrition in Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review explores how dietary modifications impact endometriosis progression. Antioxidants, omega-3s, and anti-inflammatory diets show promise in symptom relief and hormonal regulation. Personalized nutrition emerges as a pivotal tool for improving patient outcomes.

What Was Reviewed?

The paper titled "I Am the 1 in 10—What Should I Eat? A Research Review of Nutrition in Endometriosis" provides an extensive review of the role of nutrition in the management and progression of endometriosis. The authors systematically explore various dietary factors and interventions, including antioxidants, polyphenols, omega-3 fatty acids, a low-nickel diet, and the Mediterranean diet, among others, in relation to their effects on inflammation, hormonal modulation, and oxidative stress in endometriosis patients.

Who Was Reviewed?

The review primarily evaluated research studies involving women diagnosed with endometriosis. It integrated findings from human clinical trials, observational studies, and in vitro research to synthesize current evidence on nutritional influences on endometriosis-related symptoms and disease progression.

What Were the Most Important Findings?

The review highlighted that endometriosis is a chronic inflammatory and estrogen-dependent condition where dietary modifications can play a pivotal role. It emphasized the following:

Antioxidants and Polyphenols: Foods rich in antioxidants, such as fruits, vegetables, and specific compounds like resveratrol, demonstrated anti-inflammatory and pro-apoptotic effects in reducing endometriosis severity. Polyphenols, especially phytoestrogens, can modulate estrogen activity, impacting endometriotic lesion growth.

Dietary Fats: Omega-3 fatty acids were shown to reduce inflammation and dysmenorrhea, while high consumption of omega-6 and trans fats increased risks.

Specific Diets: The Mediterranean diet, with its anti-inflammatory properties, low-FODMAP and low-nickel diets showed potential benefits in reducing gastrointestinal and systemic symptoms. Gluten-free diets also alleviated pain in a subset of patients.

Dairy and Vitamin D: Dairy consumption, particularly calcium- and vitamin D-rich products, was associated with reduced endometriosis risk. Vitamin D showed immunomodulatory effects, improving inflammatory responses.

Red Meat and Iron Overload: Excessive red meat consumption elevated estrogen and prostaglandin levels, exacerbating endometriosis. The condition was also linked to iron overload in peritoneal fluid, contributing to oxidative stress and infertility.

What Are the Greatest Implications of This Review?

The findings underscore the potential for personalized dietary interventions in endometriosis management, emphasizing the integration of anti-inflammatory, low-toxin, and nutrient-rich foods. Clinicians can leverage these insights to recommend diets tailored to reduce inflammation, regulate estrogen metabolism, and mitigate oxidative stress, thereby improving quality of life and fertility outcomes for patients. Furthermore, the review reinforces the importance of microbiome-targeted dietary strategies in addressing endometriosis-related dysbiosis.

Identification of distinct stool metabolites in women with endometriosis for non-invasive diagnosis and potential for microbiota-based therapies

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies stool-based biomarkers for endometriosis diagnosis and highlights the therapeutic potential of 4-hydroxyindole, a microbiota-derived metabolite, in reducing inflammation and lesion progression.

What Was Studied?

The study examined the stool metabolome of women with endometriosis compared to healthy controls to identify microbiota-derived metabolites with diagnostic and therapeutic potential. Researchers employed metabolomics and microbiota profiling to investigate how altered gut microbiota and their metabolites, specifically 4-hydroxyindole (4HI), affect endometriosis development and progression.

Who Was Studied?

The study included stool samples from 18 women with clinically confirmed endometriosis and 31 healthy control women. The participants’ metabolomic and microbiota profiles were analyzed, followed by functional validation of key metabolites, such as 4HI, in murine and human xenograft models of endometriosis.

What Were the Most Important Findings?

The study identified a distinct stool metabolome in women with endometriosis, characterized by reduced levels of specific microbiota-derived metabolites, including 4HI. Reduced 4HI was linked to a decreased abundance of beneficial gut bacteria like Faecalibacterium and Lachnospiraceae. 4HI showed remarkable therapeutic potential, as it inhibited the initiation and progression of endometriotic lesions, reduced lesion size and volume, and alleviated inflammation and pain in murine models. Moreover, 4HI was effective in regressing well-developed lesions in pre-clinical models. The metabolic signature also highlighted overlap between endometriosis and inflammatory bowel disease (IBD), suggesting common inflammatory pathways and potential misdiagnosis risks.

What Are the Greatest Implications of This Study?

This research revolutionizes the understanding of endometriosis by identifying stool-based biomarkers, particularly 4HI, for non-invasive diagnosis. It highlights 4HI as a promising therapeutic metabolite capable of reducing inflammation and lesion development. These findings pave the way for microbiota-targeted therapies and emphasize the diagnostic overlap with IBD, offering potential for dual-disease screening. Integrating microbiota and metabolomic data offers a novel paradigm for managing endometriosis and related conditions.

Identification of polycystic ovary syndrome potential drug targets based on pathobiological similarity in the protein-protein interaction network

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study identifies potential drug targets for PCOS, focusing on genes involved in insulin resistance and hormone signaling pathways. Findings could help develop more targeted treatments.

What was studied?

This study aimed to identify potential drug targets for Polycystic Ovary Syndrome (PCOS) by leveraging pathobiological similarity with Type 2 Diabetes (T2D). The researchers used computational methods to identify PCOS potential drug targets by analyzing the protein-protein interaction network (PPIN) of PCOS and T2D genes. This network analysis was used to identify overlapping drug targets and modules associated with both diseases, offering insights into potential therapeutic interventions for PCOS.

Who was studied?

The study involved analysis of protein-protein interaction networks (PPIN) related to PCOS and T2D. It did not focus on specific individuals, but instead, it reviewed a set of PCOS-related disease genes, T2D disease genes, and drug targets available in databases. The data was obtained from multiple sources, including disease gene databases like GAD, OMIM, and the Gene Expression Omnibus (GEO).

What were the most important findings?

The study identified 22 potential drug targets for PCOS through a systematic examination of PPIN. Among these, several genes (such as ESR1, RXRA, NCOA1, and PPARG) were shown to play central roles in both the pathogenesis of PCOS and T2D, suggesting their potential as therapeutic targets. The researchers used a computational approach that integrated PCOS and T2D data, successfully identifying overlapping disease genes and known drug targets. PPDT-Module 2, a key module in the analysis, was shown to significantly contribute to PCOS pathogenesis and could be a promising therapeutic target. Furthermore, 42 drugs targeting 13 identified PCOS drug targets were investigated, revealing potential treatments such as pioglitazone and clomiphene, already used in clinical settings.

The study also highlighted how the genes identified were enriched in functional pathways associated with hormone signaling and lipid metabolism, which are critical areas in PCOS and T2D. The strong overlap in functional categories related to steroid hormone receptor signaling, lipid binding, and insulin resistance suggests that drugs targeting these pathways might improve both metabolic and reproductive health in PCOS patients.

From a microbiome perspective, the identified pathways and genes could influence microbial communities. For example, genes associated with lipid metabolism and insulin resistance could alter gut microbial composition, promoting dysbiosis and exacerbating metabolic dysfunctions. The therapeutic targeting of these pathways could help restore microbiome balance, potentially improving clinical outcomes in PCOS patients.

What are the greatest implications of this study?

The study provides valuable insights into the drug targets for PCOS, revealing that certain genes, such as ESR1, RXRA, and PPARG, could serve as promising therapeutic targets. These findings not only contribute to understanding the pathogenesis of PCOS but also offer a framework for developing targeted treatments for this condition. The use of systems biology approaches in this study could pave the way for more personalized medicine in PCOS, with a focus on drugs that target the core pathways of insulin resistance, lipid metabolism, and hormone imbalance.

Additionally, the identification of overlapping drug targets for PCOS and T2D emphasizes the potential of dual-purpose treatments, which could address both conditions simultaneously. Given the shared metabolic disturbances between PCOS and T2D, these findings open the door for novel combination therapies aimed at improving both metabolic and reproductive health in women with PCOS. Furthermore, the study provides a model for investigating other complex diseases through pathobiological similarities, potentially aiding in the identification of new therapeutic targets and improving drug development strategies.

Identifying Bacterial Vaginosis-Associated Bacteria

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study identifies three key bacterial vaginosis-associated species using DNA sequencing and phylogenetics. By classifying BVAB-1, BVAB-2, and BVAB-3, the findings advance understanding of BV’s microbial landscape and highlight new opportunities for targeted diagnostics and treatment.

What Was Studied?

This study analyzed bacterial vaginosis-associated bacteria (BVAB) at the species level using DNA sequencing and phylogenetic analysis. Researchers sought to identify the exact species of BVAB-1, BVAB-2, and BVAB-3, which were previously unknown and only classified by molecular signatures. By applying metagenomic sequencing and comparative analysis, the study aimed to provide clarity on the taxonomy of these bacteria and their role in bacterial vaginosis (BV) pathogenesis.

Who Was Studied?

The study analyzed bacterial DNA extracted from vaginal specimens of women diagnosed with BV. Using genomic data from previous metagenomic studies, the researchers compared BVAB sequences to known bacterial genomes, identifying their closest relatives and evolutionary relationships.

Key Findings and Microbial Associations

The study successfully identified the species of three previously unclassified BV-associated bacteria. BVAB-1 was found to be Clostridiales genomosp. BVAB-1 (later renamed Candidatus Lachnocurva vaginae), BVAB-2 was classified as Oscillospiraceae bacterium strain CHIC02, and BVAB-3 was identified as Mageeibacillus indolicus. These species, previously unknown, are strictly anaerobic and uncultivable in standard laboratory conditions. Their identification provides a more detailed understanding of the microbial shifts in BV and offers new insights into their role in vaginal dysbiosis.

The findings reinforce the idea that BV is not caused by a single pathogen but by a complex shift in the vaginal microbiome. The presence of BVAB-1, BVAB-2, and BVAB-3 in women with BV suggests they may contribute to the condition's persistence and recurrence. By using metagenomic sequencing and phylogenetic analysis, the study clarifies the taxonomy of these bacteria and their evolutionary relationships. This species-level identification allows for improved diagnostics and targeted research into BV-associated microbial interactions.

Implications of the Study

Identifying the specific species associated with BV significantly improves diagnostic and treatment strategies. Clinicians currently use broad-spectrum antibiotics to treat BV, but discovering these species enables the development of more targeted therapies. This study also emphasizes the need for continued metagenomic research, as many BV-associated bacteria are difficult to culture and study in traditional lab settings. With more precise species identification, researchers can better understand how these bacteria interact with the vaginal microbiome and contribute to BV recurrence.

Improved cure of bacterial vaginosis with single dose of tinidazole (2g), Lactobacillus rhamnosus GR-1, and Lactobacillus reuteri RC-14

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

  • Tinidazole
    Tinidazole

    Tinidazole is a nitroimidazole antimicrobial that selectively targets anaerobic bacteria and protozoa, reshaping the gut ecosystem by depleting pathogenic anaerobes while preserving commensals. Clinically validated for giardiasis, bacterial vaginosis, and colorectal surgery prophylaxis. Its DNA-disrupting and biofilm-penetrating actions reduce inflammatory triggers and create niches for healthy microbiota to rebound.

This study shows that oral probiotics significantly boost cure rates of bacterial vaginosis when used with tinidazole, restoring a healthy vaginal microbiome.

What Was Studied?

This randomized, double-blind, placebo-controlled clinical trial investigated whether the combination of a single 2g dose of tinidazole and daily oral probiotics (Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14) improved bacterial vaginosis (BV) cure rates in women, compared to tinidazole alone. BV, a condition characterized by dysbiosis in the vaginal microbiome and reduction of protective Lactobacillus species, has shown poor long-term response to antibiotic treatment alone. The study aimed to determine if probiotic supplementation could enhance the therapeutic efficacy of tinidazole by restoring a more favorable microbial balance.

Who Was Studied?

Sixty-four Brazilian women diagnosed with BV based on Amsel’s criteria and Nugent scoring participated in the study. The participants were randomly assigned to either a placebo group or a probiotic group, both of which received the same tinidazole dose. The probiotic group also received daily capsules containing L. rhamnosus GR-1 and L. reuteri RC-14 for four weeks. Women with other vaginal infections, recent antibiotic use, or immunosuppression were excluded. The trial assessed both subjective symptoms and microbiological cure at the end of the treatment period.

What Were the Most Important Findings?

At the end of the four-week treatment, the probiotic group exhibited a significantly higher cure rate compared to the placebo group as measured by Amsel’s criteria and Nugent score. Women in the probiotic group were assessed with “normal” vaginal microbiota based on Gram stain, compared to the placebo group. Importantly, the study also noted reduced BV-associated microbial morphotypes (Gram-variable rods, curved anaerobes) and a statistically significant improvement in key clinical indicators, including pH, discharge, and odor in the probiotic group. While both groups used tinidazole, the probiotics played a key role in enhancing microbiota restoration. Notably, the probiotic strains used in the study are known for producing biosurfactants, bacteriocins, and signaling molecules that can disrupt pathogenic biofilms, particularly those formed by Gardnerella vaginalis. This mechanism may explain their strong microbiome-modulating effect.

What Are the Greatest Implications of This Study?

This study provides robust clinical evidence supporting the adjunctive use of probiotics with antibiotics to treat BV and improve microbiota restoration. By demonstrating that oral administration of specific Lactobacillus strains significantly improves cure rates and promotes a return to healthy vaginal flora, the study bridges microbiome science with practical gynecological care. Clinicians managing recurrent or treatment-resistant BV can consider integrating targeted probiotic strains to reduce recurrence and enhance long-term remission. Additionally, the study underscores the need for strain-specific probiotic selection, given the inconsistent outcomes with nonspecific lactobacilli. The use of probiotics also holds promise in preserving drug efficacy and reducing the need for prolonged antibiotic exposure, which aligns with antimicrobial stewardship principles and microbiome health preservation.

Increased circulating conjugated primary bile acids are associated with hyperandrogenism in women with polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study links elevated conjugated primary bile acids to hyperandrogenism in PCOS, suggesting bile acid metabolism plays a direct role in endocrine dysfunction and opens new therapeutic perspectives for managing PCOS.

What was studied?

This study explored the relationship between circulating bile acid profiles and hyperandrogenism in women diagnosed with polycystic ovary syndrome (PCOS). Specifically, the researchers investigated whether changes in the concentration and composition of serum bile acids, particularly conjugated primary bile acids, were associated with elevated androgen levels in PCOS patients. Utilizing ultra-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS), the study quantified individual bile acid species in the serum and examined their statistical associations with androgen markers such as total testosterone and androstenedione.

Who was studied?

The study population consisted of 37 women diagnosed with PCOS and 35 age- and BMI-matched healthy control subjects, all recruited from the Endocrinology Department of Drum Tower Hospital, Nanjing, China. The diagnosis of PCOS was based on the Rotterdam criteria, ensuring that participants met at least two of the following: hyperandrogenism, oligo/anovulation, or polycystic ovarian morphology. Subjects were carefully screened to exclude confounding conditions such as diabetes, liver disease, and recent antibiotic use. This rigorous matching and exclusion criteria strengthen the internal validity of the findings.

What were the most important findings?

The researchers identified that women with PCOS had significantly elevated levels of circulating conjugated primary bile acids, specifically glycine-conjugated (GCA and GCDCA) and taurine-conjugated (TCA and TCDCA) species, compared to healthy controls. Interestingly, the total bile acid levels and the total primary bile acid pool were both elevated in PCOS, while secondary bile acids showed no significant difference. The elevated conjugated bile acids also displayed strong positive associations with serum androgen levels, including total testosterone and androstenedione. These associations remained statistically significant even after adjusting for potential confounders such as age, BMI, and insulin resistance (HOMA-IR).

The study also observed that the relative percentage composition of the bile acid pool shifted in PCOS patients, with higher contributions from conjugated primary bile acids like GCA, TCA, and TCDCA and a reduced proportion of secondary bile acids like DCA, LCA, and GLCA. These patterns suggest an altered bile acid metabolism that could be functionally linked to androgen excess in PCOS, potentially through mechanisms involving bile acid receptors such as the farnesoid X receptor (FXR), which has been shown in other research to regulate steroid metabolism.

What are the greatest implications of this study?

This study highlights a novel and potentially important role for bile acid metabolism in the endocrine dysfunction characteristic of PCOS, especially hyperandrogenism. The findings suggest that the gut-liver axis and bile acid signaling could contribute to the development or perpetuation of elevated androgen levels in PCOS, potentially via FXR-mediated inhibition of androgen-to-estrogen conversion in ovarian granulosa cells. If validated in future longitudinal studies, these insights open new avenues for biomarker discovery and therapeutic intervention. Modulating bile acid composition—whether through dietary strategies, microbiome-based therapies, or bile acid sequestrants—could represent a future direction in managing hyperandrogenism and its associated reproductive and metabolic complications in PCOS.

Inflammatory cytokines IL-6, IL-10, IL-13, TNF-α and peritoneal fluid flora were associated with infertility in patients with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This cross-sectional observational study examined the link between inflammatory markers (IL-6, IL-10, IL-13, TNF-α), peritoneal fluid bacterial flora, and infertility in endometriosis patients. Results showed significantly higher white cell counts and elevated levels of inflammatory markers in endometriosis patients compared to controls. Logistic regression confirmed significant associations between these inflammatory markers and infertility.

What was studied?

The study investigated the relationship between inflammatory markers (IL-6, IL-10, IL-13, and TNF-α), the composition of bacterial flora in peritoneal fluid, and infertility in patients with endometriosis.

 

Who was studied?

The participants included 55 patients diagnosed with endometriosis and infertility (observation group) attending a Gynecology Clinic from June 2014 to July 2017 and 30 individuals without endometriosis or infertility issues (control group).

 

What were the most important findings?

The study found elevated white cell counts (monocytes, neutrophils, eosinophils, and basophils) and higher levels of inflammatory cytokines (IL-6, IL-10, IL-13, and TNF-α) in the peritoneal fluid of endometriosis patients with infertility compared to controls. A significant correlation between these inflammatory markers and endometriosis associated with infertility was also established.

 

What are the greatest implications of this study?

The study implies that inflammatory factors in peritoneal fluid play a crucial role in the pathophysiology of infertility associated with endometriosis. These findings suggest that inflammatory cytokines (IL-6, IL-10, IL-13, and TNF-α) could serve as significant biomarkers for diagnosing and understanding the mechanisms of endometriosis-related infertility.

Inflammatory cytokines IL-6, IL-10, IL-13, TNF-α and peritoneal fluid flora were associated with infertility in patients with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study reveals that inflammatory cytokines (IL-6, IL-10, IL-13, and TNF-α) are significantly elevated in the peritoneal fluid of endometriosis patients with infertility. These findings suggest their potential role as diagnostic biomarkers and therapeutic targets for managing reproductive complications in endometriosis.

What Was Studied?

This study investigated the association of inflammatory cytokines and peritoneal fluid flora with infertility in endometriosis patients. Researchers aimed to explore the correlations between elevated inflammatory markers—IL-6, IL-10, IL-13, and TNF-α—and changes in microbial communities within the peritoneal fluid of women suffering from endometriosis-related infertility. Using Ion Torrent PGM platform sequencing, the microbial composition of the peritoneal fluid was mapped, while ELISA assays were employed to quantify cytokine levels. Logistic regression analysis was conducted to determine the relationship between inflammatory factors and infertility severity.

Who Was Studied?

The study included 55 women diagnosed with endometriosis and infertility and a control group of 30 women without endometriosis or infertility. All participants were treated at a gynecology clinic between June 2014 and July 2017. Peritoneal fluid samples were collected laparoscopically from the vesicouterine and rectovaginal pouches. Women receiving hormonal therapy or antibiotics within two months of surgery were excluded to avoid confounding microbial and inflammatory responses. This patient population allowed for a focused analysis of microbial and cytokine-driven inflammation linked to infertility in endometriosis.

What Were the Most Important Findings?

The study found that endometriosis patients with infertility exhibited significantly higher levels of IL-6, IL-10, IL-13, and TNF-α in peritoneal fluid compared to controls (p < 0.05). White blood cell counts, including monocytes, neutrophils, eosinophils, and basophils, were also elevated in the peritoneal fluid of endometriosis patients, supporting the inflammatory hypothesis of disease pathogenesis. Analysis of peritoneal fluid microbiota revealed a predominance of Proteobacteria and Firmicutes, with secondary representation from Actinobacteria, Bacteroidetes, Fusobacterium, and Tenericutes. However, there were no significant differences in the abundance of these phyla between endometriosis patients and controls, suggesting that bacteria themselves are not the primary drivers of infertility in endometriosis. Instead, the study highlighted the role of inflammatory cytokines as critical mediators of infertility. Logistic regression analysis confirmed that IL-6, IL-10, IL-13, and TNF-α were significantly correlated with infertility severity in endometriosis patients, suggesting these cytokines may serve as diagnostic biomarkers for assessing infertility risk. Receiver Operating Characteristic (ROC) analysis showed that IL-6 and TNF-α had the highest diagnostic sensitivity, with AUC values of 0.893 for both, indicating their strong predictive value for identifying infertility risk in endometriosis.

ParameterFindings in Endometriosis Patients with Infertility
Inflammatory CytokinesElevated levels of IL-6, IL-10, IL-13, and TNF-α in peritoneal fluid (p < 0.05).
Immune Cell CountsIncreased monocytes, neutrophils, eosinophils, and basophils in peritoneal fluid.
Peritoneal Fluid MicrobiotaDominated by Proteobacteria and Firmicutes, with secondary presence of Actinobacteria, Bacteroidetes, Fusobacterium, and Tenericutes.
Microbial ShiftsNo significant differences in bacterial phyla between endometriosis patients and controls.
Diagnostic BiomarkersIL-6 and TNF-α showed the highest diagnostic sensitivity for infertility with AUC values of 0.893.
Inflammatory ImplicationsCytokines linked to immune activation, peritoneal inflammation, and possible inhibition of sperm motility.
Therapeutic PotentialAnti-inflammatory strategies targeting IL-6 and TNF-α could improve fertility outcomes in endometriosis patients.

What Are the Greatest Implications of This Study?

This study underscores the critical role of inflammatory cytokines in endometriosis-associated infertility, particularly IL-6, IL-10, IL-13, and TNF-α. These cytokines appear to contribute to peritoneal inflammation, immune cell activation, and possibly sperm motility inhibition, collectively reducing fertility. Although peritoneal fluid microbiota did not significantly differ between endometriosis and control groups, the elevated cytokine levels suggest that microbial translocation and immune responses within the peritoneal cavity may exacerbate inflammation. These findings highlight IL-6 and TNF-α as potential biomarkers for diagnosing infertility in endometriosis patients and suggest that anti-inflammatory therapies targeting these cytokines could improve fertility outcomes. Additionally, the study indicates that monitoring cytokine levels in peritoneal fluid could serve as a non-invasive diagnostic tool for assessing infertility risk in endometriosis, offering a novel approach to reproductive management in affected women.

Influence of heavy metal exposure on gut microbiota: Recent advances

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review highlights that heavy metal exposure disrupts gut microbiota composition and function, leading to dysbiosis and various health implications. Heavy metals alter gene expression in both the host and microbiota, affecting immune regulation and oxidative stress pathways. Dysbiosis induced by heavy metals can lead to inflammation, gut motility disturbances, and increased susceptibility to diseases like inflammatory bowel diseases. The study underscores the importance of understanding heavy metal-induced dysbiosis and its implications for human and environmental health, advocating for further research in this area.

What was studied?
The study investigated the impact of heavy metal exposure on the composition and function of gut microbiota. Specifically, it looked at how heavy metals such as arsenic, cadmium, lead, and mercury affect the diversity, richness, and metabolism of gut bacteria.

 

Who was studied?
The research encompassed various model organisms, including humans, mice, rats, chickens, fish, crayfish, and asiatic toad. Studies were conducted on both adult and juvenile stages of these organisms to understand the effects of heavy metal exposure on gut microbiota across different life stages.

 

What were the most important findings?

Several significant findings have emerged from the study: Heavy metal exposure has been shown to induce dysbiosis in the gut microbiota, manifesting in alterations in microbial composition, gene expression, metabolism, and immune response. Moreover, the gut microbiota play a pivotal role in the detoxification and elimination of heavy metals, facilitated through enzymatic reactions, bioaccumulation, and methylation processes. Notably, exposure to heavy metals results in shifts in the abundance of specific bacterial phyla within the gut microbiome, including Proteobacteria, Firmicutes, and Bacteroidetes. These alterations in gut microbiota composition and function have far-reaching health implications, including oxidative stress, neurobehavioral damage, disrupted lipid metabolism, compromised immune function, and heightened susceptibility to inflammatory bowel diseases.

 

What are the greatest implications of this study?

The study underscores several important implications: It stresses the criticality of comprehending how heavy metals affect gut microbiota, as this knowledge is essential for evaluating the environmental and public health risks linked to heavy metal exposure. Moreover, the findings advocate for the development of guidelines and interventions aimed at mitigating heavy metal-induced toxicity and safeguarding gut microbiota health. Further research is imperative to uncover the mechanisms behind heavy metal-induced dysbiosis of gut microbiota and its repercussions on human health, necessitating investigations into microbial alterations at the species and strain levels using advanced sequencing methodologies like metagenomics. Additionally, epidemiological studies involving human populations are warranted to directly assess the health consequences of heavy metal exposure on gut microbiota and to guide the formulation of preventive measures and public health policies.

Insulin Resistance and the Polycystic Ovary Syndrome Revisited: An Update on Mechanisms and Implications

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review explores insulin resistance as a primary driver of PCOS, detailing molecular signaling defects and their clinical consequences. It provides critical insight into how hyperinsulinemia fuels both metabolic dysfunction and androgen excess, reinforcing PCOS as a systemic endocrine-metabolic disorder, with implications for microbiome-linked disease signatures.

What Was Reviewed?

This review paper revisits the pathophysiological mechanisms linking insulin resistance to polycystic ovary syndrome (PCOS), integrating molecular, clinical, and genetic insights. It offers an updated synthesis of the complex metabolic and reproductive disturbances in PCOS, focusing particularly on how insulin resistance plays a central role in the syndrome’s development. The paper builds on findings from the original 1997 Endocrine Reviews article and incorporates two decades of advances in endocrinology, genetics, and metabolic signaling. The authors evaluate the metabolic and mitogenic effects of insulin, molecular mechanisms like post-binding receptor signaling defects, and explore insulin’s role as a reproductive hormone. The review also emphasizes the impact of hyperinsulinemia and androgen excess on glucose metabolism, ovarian steroidogenesis, and ovulatory dysfunction. Genetic predisposition and developmental programming through intrauterine androgen exposure are also considered contributing factors.

Who Was Reviewed?

The review synthesizes findings from studies involving both lean and obese women with PCOS, alongside control groups without the condition. It integrates data from in vivo human metabolic studies, tissue-specific analyses of adipocytes and skeletal muscle, and molecular experiments using cultured fibroblasts. The reviewed cohorts span racially and ethnically diverse populations, including women from the United States, Europe, and Asia, offering insight into the universality and variability of insulin resistance in PCOS. Importantly, the authors highlight that while insulin resistance is nearly universal in obese women with PCOS, its presence in lean women depends on PCOS phenotype and diagnostic criteria. Studies of first-degree relatives also reveal inherited metabolic and reproductive traits, affirming the role of genetic and familial influences.

What Were the Most Important Findings?

The review reinforces that insulin resistance is a hallmark of PCOS and a central pathogenic factor, even in the absence of obesity. It identifies a post-binding defect in insulin receptor signaling, particularly an increase in serine phosphorylation of the insulin receptor and insulin receptor substrate-1, that impairs insulin’s metabolic actions while leaving mitogenic pathways largely unaffected. This selective insulin resistance may allow hyperinsulinemia to persistently drive androgen overproduction in ovarian theca cells, exacerbating symptoms like anovulation and hirsutism. In skeletal muscle and adipocytes, insulin-mediated glucose uptake is significantly impaired, comparable to levels seen in type 2 diabetes. This dysfunction is not solely due to fat distribution or visceral adiposity but appears intrinsic to PCOS pathophysiology.

From a microbiome perspective, while this review does not directly address gut microbial composition, it offers mechanistic insight into the downstream metabolic disruptions that have been consistently linked in other studies to altered microbiota. These disruptions could correspond with major microbial associations (MMA) observed in PCOS, such as decreased diversity and elevated LPS-producing gram-negative bacteria that amplify systemic inflammation and insulin resistance. Thus, the mechanistic pathways elucidated in this review form a critical biological foundation that helps explain how gut microbiota may further exacerbate PCOS symptoms.

What Are the Implications of This Review?

This paper decisively frames PCOS as a multifactorial metabolic disorder with deep-seated insulin resistance at its core. For clinicians, this calls for an expanded diagnostic and therapeutic lens, one that considers insulin sensitivity as a key biomarker in both lean and obese PCOS patients. The authors strongly advocate for early screening of glucose intolerance and type 2 diabetes in all PCOS phenotypes using a 2-hour OGTT, emphasizing that hemoglobin A1c alone may miss postprandial dysglycemia. Furthermore, the review’s findings justify the use of insulin-sensitizing agents such as metformin and thiazolidinediones not only for metabolic control but also for improving ovulatory function and reducing androgen excess. This review also encourages deeper exploration into how metabolic dysfunction and reproductive impairment intersect in PCOS, providing a roadmap for future studies on gut microbiota and systemic insulin signaling.

Intestinal microbiota influences clinical outcome and side effects of early breast cancer treatment

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

Gut microbiota influences early breast cancer prognosis and treatment side effects, with specific commensals correlating to outcomes. Chemotherapy alters microbiota, favoring beneficial species and improving immune modulation and neuroprotection.

What was studied?

This study examined the impact of intestinal microbiota on the clinical outcomes and side effects of early breast cancer (BC) treatments. Shotgun metagenomics was used to analyze fecal microbiota samples from 76 early BC patients, both pre- and post-chemotherapy. The study aimed to identify specific microbial species associated with BC prognosis and the side effects of chemotherapy, focusing on neurological, gastrointestinal, and metabolic complications. It also explored the functional relevance of gut microbiota in immunocompetent mouse models colonized with BC patient microbiota to establish a causal link between gut microbial composition and tumor growth or therapy efficacy.

Who was studied?

The study involved 76 female BC patients from the CANTO trial (NCT01993498), a long-term prospective cohort designed to quantify and prevent treatment-related toxicities. Patients provided fecal samples before and after chemotherapy, and their plasma was also analyzed for metabolomics. A separate analysis included healthy volunteers (54 Italian and 282 samples from public metagenomes) to contrast microbial signatures. Mouse models were humanized with fecal microbiota from patients and healthy individuals to assess the causal relationship between microbiota and BC outcomes.

What were the most important findings?

The study revealed that the gut microbiota composition significantly correlates with BC prognosis and treatment side effects. Patients with more aggressive tumors (larger size, advanced stage, lymph node involvement) had overrepresentation of species like Clostridiaceae, Veillonella, Bacteroides uniformis, and Blautia wexlerae. In contrast, patients with better prognosis had higher levels of Akkermansia muciniphila, Collinsella aerofaciens, and Eubacterium rectale. Chemotherapy shifted microbial diversity, reducing bacteria associated with poor prognosis and increasing favorable commensals like Methanobrevibacter smithii and Blautia obeum. Functionally, favorable microbiota patterns were linked to neuroprotective and immunomodulatory pathways, such as polyamine biosynthesis and ketogenesis, while unfavorable profiles were associated with inflammation and metabolic dysregulation. Humanized mouse models demonstrated that fecal microbiota from healthy volunteers enhanced tumor response to chemotherapy compared to microbiota from BC patients.

What are the greatest implications of this study?

This study underscores the gut microbiota's role as a biomarker and potential therapeutic target in BC management. The findings suggest that monitoring and modulating gut microbiota could optimize chemotherapy efficacy, mitigate side effects, and improve overall prognosis. Strategies like fecal microbiota transplantation, probiotics, or diet interventions targeting specific microbiota shifts may hold promise. The causal evidence provided by mouse models highlights the translational potential of microbiome-targeted interventions (MBTIs) to improve clinical outcomes for breast cancer patients.

Investigation of In Vitro Efficacy of Boric Acid on Pseudomonas aeruginosa Strains Isolated from Diabetic Foot Infections

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study evaluated boric acid’s in vitro effect on Pseudomonas aeruginosa from diabetic foot infections, confirming its inhibitory potential at 25 mg/l. The findings support boric acid as a cost-effective alternative in wound care, offering clinicians a valuable tool against antibiotic-resistant infections.

What was studied?

This study investigated the in vitro efficacy of boric acid as an antiseptic agent against Pseudomonas aeruginosa strains isolated from diabetic foot infections. The research aimed to determine whether boric acid could serve as a viable alternative for local wound treatment, particularly given the rising issue of antibiotic resistance in diabetic foot infections. Researchers prepared various concentrations of boric acid and tested them on 25 P. aeruginosa strains, evaluating bacterial growth suppression across different boric acid levels.

Who was studied?

The study focused on 25 clinical isolates of P. aeruginosa obtained from patients with diabetic foot infections between January 2010 and June 2015. Each isolate came from a distinct patient, ensuring no duplication of data. These strains represented a typical clinical cohort where P. aeruginosa plays a significant role in complicating wound healing due to its robust antibiotic resistance and biofilm-forming abilities.

What were the most important findings?

The study demonstrated that boric acid has a notable inhibitory effect on P. aeruginosa in vitro. Specifically, no bacterial growth was observed at boric acid concentrations of 25 mg/l or higher, establishing this as the minimum inhibitory concentration (MIC) required to suppress P. aeruginosa. At lower concentrations, varying degrees of bacterial growth persisted, with complete growth seen at 1.6 mg/l and partial suppression at intermediary levels. Importantly, the P. aeruginosa strains exhibited high resistance to common antibiotics such as cefepime, ceftazidime, ciprofloxacin, amikacin, and netilmicin, underscoring the challenge of treating these infections. This research adds to the growing recognition that specific microbial pathogens like P. aeruginosa, often part of the wound microbiome in diabetic foot infections, can be targeted with non-antibiotic antiseptic strategies. The study reinforces boric acid’s broad antimicrobial properties, which extend to inhibiting bacterial growth and potentially biofilm formation, an essential consideration in chronic wound care.

What are the greatest implications of this study?

The study’s findings suggest that boric acid could offer a cost-effective and practical alternative for local wound care, particularly in settings where antibiotic resistance is prevalent and access to advanced antimicrobial agents is limited. With its confirmed MIC of 25 mg/l against P. aeruginosa, boric acid emerges as a promising agent for reducing bacterial load in diabetic foot infections, potentially improving healing outcomes and reducing reliance on systemic antibiotics. The implications are particularly significant for resource-constrained environments and for addressing the growing crisis of antibiotic resistance. Additionally, because boric acid is well tolerated on intact skin and has a long history of use, its reintroduction into wound care protocols could complement current treatment regimens, helping bridge the gap between conventional antibiotics and effective wound management strategies.

Iron Overload and Endometriosis: Mechanisms, Implications, and Therapeutic Targets

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Endometriomas
    Endometriomas

    An endometrioma is a type of ovarian cyst filled with old blood, arising from endometrial tissue outside the uterus, typically causing pain and potentially impacting fertility.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

Iron overload in endometriosis contributes to oxidative stress, inflammation, and tissue damage, driving lesion persistence and subfertility. Ferroptosis resistance and dysregulated iron metabolism highlight therapeutic opportunities using iron chelators and modulators.

What Was Reviewed?

This systematic review evaluated the role of iron in the pathophysiology of endometriosis. The review synthesized findings from 53 studies, including both human and animal research, to provide a comprehensive understanding of how excess iron contributes to oxidative stress, inflammation, and tissue damage in endometriosis. It also explored iron-related mechanisms such as ferroptosis and the implications for subfertility, symptom severity, and potential malignant transformation.

Who Was Reviewed?

The review included a total of 53 studies: 47 human studies involving 3,556 participants and 6 animal studies. The human studies primarily examined women diagnosed with endometriosis, and the included research utilized various bio-samples such as ovarian endometriomas, peritoneal fluid, and ectopic endometrial lesions. Animal studies focused on endometriosis models to explore systemic and local iron mechanics.

Key Findings

Iron overload is consistently found in endometriotic tissues and peritoneal fluid but not in systemic circulation. This localized iron accumulation stems from repeated bleeding within lesions, leading to oxidative stress and inflammation that perpetuates the ectopic growth of endometrial tissue. Dysregulated iron transport and the failure of homeostatic mechanisms contribute to this pathology, with increased expression of proteins such as divalent metal transporter-1 (DMT1) and decreased ferroportin expression in affected tissues.

Markers of oxidative stress, including lipid peroxidation and DNA damage, were significantly elevated in endometriotic lesions. Aberrant resistance to ferroptosis, an iron-dependent form of cell death, was identified as a key mechanism supporting lesion persistence. Additionally, iron-induced ferroptosis was linked to the production of pro-inflammatory and angiogenic factors like IL-8 and VEGFA, exacerbating inflammation and lesion vascularization.

Iron overload was implicated in subfertility, as higher iron concentrations in ovarian follicles and endometriomas were associated with impaired oocyte quality and development. These findings suggest that iron mechanics might influence folliculogenesis and embryo viability. Importantly, the review highlighted the therapeutic potential of iron chelators and ferroptosis modulators for managing endometriosis.

Implications of the Review

This review underscores the central role of aberrant iron metabolism in the pathogenesis of endometriosis, providing a mechanistic basis for its persistence, progression, and associated complications such as subfertility and chronic pain. Iron-related oxidative stress emerges as a critical driver of inflammation and tissue damage, making it a promising target for therapeutic intervention. Future research should explore the efficacy of iron-targeted treatments, such as chelators, and further elucidate the role of ferroptosis in endometriosis. These insights could lead to novel strategies for mitigating symptom severity and improving fertility outcomes in affected women.

Irritable Bowel Syndrome-Like Disorders in Endometriosis: Prevalence of Nickel Sensitivity and Effects of a Low-Nickel Diet. An Open-Label Pilot Study.

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

Nickel allergic contact mucositis was identified in over 90% of endometriosis patients with IBS-like symptoms. A low-nickel diet significantly reduced gastrointestinal, extra-intestinal, and gynecological symptoms, revealing nickel sensitivity as a key driver of endometriosis symptomatology.

What was studied?

This open-label pilot study investigated the prevalence of nickel allergic contact mucositis (Ni ACM) in women with endometriosis who presented with irritable bowel syndrome (IBS)-like symptoms and assessed the efficacy of a low-nickel diet (LNiD) in ameliorating gastrointestinal, extra-intestinal, and gynecological symptoms. The study aimed to determine whether nickel (Ni) sensitivity contributes to the symptom burden in endometriosis and whether dietary nickel restriction can serve as a therapeutic intervention.

Who was studied?

The study initially screened 83 women of reproductive age diagnosed with endometriosis via imaging or laparoscopy who also experienced at least three gastrointestinal symptoms scoring ≥5 on the Gastrointestinal Symptom Rating Scale (GSRS). After applying exclusion criteria (e.g., celiac disease, IgE-mediated food allergies), 47 patients remained eligible. Sixteen dropped out due to the dietary restrictions, leaving 31 who completed the study. Each participant underwent a nickel oral mucosa patch test (omPT) to identify Ni ACM. Of the 31 patients, 28 (90.3%) tested positive. All participants followed a low-Ni diet for three months, with symptoms reassessed using the GSRS at baseline (T0) and after the intervention (T1).

What were the most important findings?

Nickel ACM was highly prevalent among women with endometriosis and IBS-like symptoms, with 90.3% of study completers testing positive via omPT. All patients who adhered to a three-month low-nickel diet experienced statistically significant reductions in all 15 gastrointestinal symptoms, including bloating, abdominal pain, diarrhea, and constipation. Additionally, the LNiD led to meaningful improvements in seven extra-intestinal symptoms such as headache, fatigue, and joint pain, as well as in hallmark gynecological symptoms of endometriosis: chronic pelvic pain, dysmenorrhea, and dyspareunia. These improvements underscore a systemic role of nickel sensitivity in the symptomatology of endometriosis beyond localized pelvic pathology.

From a microbiome perspective, Ni ACM reflects a low-grade inflammatory response that disrupts mucosal immune regulation and intestinal barrier integrity—two key mechanisms implicated in microbial dysbiosis. Though microbiome composition was not directly assessed, the systemic inflammatory profile induced by dietary nickel may favor enrichment of nickel-tolerant pathobionts, including certain Gammaproteobacteria and urease-producing bacteria, while impairing barrier-supporting commensals. This aligns with broader hypotheses on the metallomic drivers of endometriosis-associated dysbiosis.

What are the greatest implications of this study?

This study provides compelling evidence that nickel hypersensitivity may be a clinically significant and previously underrecognized contributor to the gastrointestinal and systemic symptom burden in endometriosis. It positions Ni ACM not only as a comorbidity but as a potential driver of symptom exacerbation, offering a new lens through which to understand IBS-like manifestations in endometriosis. Importantly, the successful use of a targeted dietary intervention based on objective testing (omPT) introduces a personalized medicine framework that could improve quality of life while minimizing unnecessary dietary restrictions, such as those imposed by low-FODMAP diets. If validated in larger, randomized cohorts, the incorporation of nickel testing and dietary counseling into standard endometriosis management could represent a low-risk, high-reward clinical advance.

Ketogenic diet improves fertility in patients with polycystic ovary syndrome: A brief report

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

A ketogenic diet restored menstrual cycles and improved pregnancy rates in obese women with PCOS, even without metformin or ovulation induction. Findings suggest KD enhances fertility through metabolic and microbiome-linked pathways beyond weight loss.

What was studied?

This retrospective study examined the effects of a ketogenic diet (KD) on menstrual regularity and pregnancy rates in women with polycystic ovary syndrome (PCOS). Conducted at the Cleveland Clinic, the study evaluated 30 women diagnosed with PCOS who followed a KD for at least three months as part of a multidisciplinary weight management program. The objective was to determine whether KD could restore ovulatory function and improve fertility, particularly in the context of insulin resistance, obesity, and hyperandrogenism that characterize PCOS.

Who was studied?

The study population included 30 women with PCOS and obesity, aged around 31 years. All participants were enrolled in a structured program that combined nutritional counseling, exercise guidance, and endocrine evaluation. Among them, 18 desired pregnancy, and some were concurrently using metformin or ovulation induction agents. Importantly, participants had no other major endocrine or metabolic conditions, ensuring the focus remained on PCOS-related infertility. The KD protocol restricted daily carbohydrate intake to ≤20g, moderate protein based on weight, and up to 40g of fat, with calorie intake ranging from 1000–1200 kcal/day.

What were the most important findings?

The ketogenic diet led to complete restoration of regular menstrual cycles in all participants with irregular periods, with 92% achieving this within six months. Among those desiring pregnancy, 55.6% (10) became pregnant, five without any ovulation induction or metformin. Women in the non-metformin group had a 100% pregnancy rate, compared to 38.5% in the metformin group. Importantly, there was no statistically significant difference in weight loss between those who conceived and those who did not, suggesting that mechanisms beyond weight reduction, such as improved insulin sensitivity, may be central to the fertility benefits observed. While the study did not directly measure microbiome changes, the KD is known to influence microbial composition, particularly by reducing pro-inflammatory species and enhancing Akkermansia muciniphila and SCFA-producing microbes such as Bacteroides and Faecalibacterium prausnitzii. These microbial shifts are closely associated with improved insulin signaling, reduced systemic inflammation, and improved endocrine profiles—all relevant in PCOS.

What are the implications of this study?

This study provides compelling evidence that the ketogenic diet can be a highly effective non-pharmacologic intervention for improving fertility in women with PCOS. By normalizing menstrual cycles and significantly improving pregnancy rates, especially among those not on pharmacologic ovulation aids, the KD presents a viable alternative or adjunct to current fertility treatments. The results suggest that mechanisms like enhanced insulin sensitivity and reduced inflammatory signaling play a greater role than weight loss alone. For clinicians, the KD offers a microbiome-relevant strategy that addresses the root metabolic and endocrine disturbances of PCOS. Given its high efficacy in restoring ovulation and supporting conception, especially in obese patients with insulin resistance, KD merits consideration in personalized fertility management plans. Larger prospective studies are now needed to further investigate long-term outcomes, microbiome shifts, and to standardize KD protocols for PCOS patients.

Lactobacillus crispatus inhibits growth of Gardnerella vaginalis and Neisseria gonorrhoeae on a porcine vaginal mucosa model.

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

The study shows that Lactobacillus crispatus inhibits the growth of Gardnerella vaginalis and Neisseria gonorrhoeae by lowering pH and producing lactic acid, offering insights into microbiome-targeted interventions for bacterial vaginosis and sexually transmitted infections.

What was Studied?

The study investigated the effects of Lactobacillus crispatus on the growth of Gardnerella vaginalis and Neisseria gonorrhoeae using a porcine vaginal mucosa (PVM) model. It aimed to explore how Lactobacillus crispatus influences the growth of these pathogens and whether it could help prevent or inhibit infection through mechanisms such as the production of lactic acid and pH reduction.

Who was Studied?

The study focused on human clinical isolates of Lactobacillus crispatus, Gardnerella vaginalis, and Neisseria gonorrhoeae. The researchers inoculated these isolates into the ex vivo PVM to observe their colonization, biofilm formation, and interactions.

What were the Most Important Findings?

The study revealed that Lactobacillus crispatus significantly inhibited the growth of both Gardnerella vaginalis and Neisseria gonorrhoeae on the porcine vaginal mucosa model. This inhibition occurred primarily due to the lactic acid production by L. crispatus, which lowered the vaginal pH to levels hostile to these pathogens. The results showed that both G. vaginalis and N. gonorrhoeae grew and formed biofilms at clinically relevant densities on PVM. In particular, the biofilm formation by G. vaginalis and N. gonorrhoeae was evident, and the presence of L. crispatus hindered this process. The production of lactic acid by L. crispatus was crucial for reducing the pH below 5.5, which subsequently inhibited pathogen growth. Conditioned media (CM) from L. crispatus cultures inhibited the growth of N. gonorrhoeae, even when the pH was adjusted to levels conducive for its growth.

What are the Implications of this Study?

The study demonstrates that Lactobacillus crispatus, a key member of the vaginal microbiota, plays a significant protective role against the colonization of harmful pathogens like Gardnerella vaginalis and Neisseria gonorrhoeae. It exerts direct antimicrobial effects and modulates vaginal pH through lactic acid production. By lowering pH, L. crispatus shows potential as both a therapeutic agent and a preventive measure against bacterial vaginosis and sexually transmitted infections, including gonorrhea. This finding supports the importance of maintaining a healthy vaginal microbiota dominated by Lactobacillus species to reduce susceptibility to infections. The PVM model serves as a valuable tool for studying the complex interactions between vaginal microbiota and pathogens, offering insights into the development of targeted microbiome-based interventions.

Levels of Trace Elements in Erythrocytes as Endocrine Disruptors in Obese and Nonobese Women with Polycystic Ovary Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study identified elevated erythrocyte nickel in obese women with PCOS and revealed trace element–hormone correlations linked to reproductive dysfunction. These findings point to micronutrient-driven endocrine disruption and potential gut microbiota imbalances in PCOS.

What was studied?

This study examined how trace elements in erythrocytes function as endocrine disruptors in obese and nonobese women with polycystic ovary syndrome (PCOS). Researchers focused on six trace elements, zinc (Zn), nickel (Ni), iron (Fe), manganese (Mn), copper (Cu), and magnesium (Mg), and their relationship with the hormonal profiles of PCOS patients. Given the limitations of serum testing, the study utilized erythrocyte samples to capture long-term mineral status, providing a more stable biomarker for trace element accumulation and hormonal interaction. The aim was to evaluate whether these trace elements correlate with reproductive hormone levels and contribute to PCOS pathogenesis based on obesity status.

Who was studied?

The study included 47 women with PCOS, divided into two subgroups based on body mass index (BMI): 24 obese women (BMI ≥ 30) and 23 nonobese women (BMI < 30). A control group of 16 healthy women with no signs of PCOS and a BMI within the normal range (mean: 23.6) was also evaluated. All participants were of reproductive age and had not used supplements or experienced significant dietary changes before the study. Blood samples were analyzed using inductively coupled plasma atomic emission spectrometry to quantify trace element levels in red blood cells. Hormonal parameters, lipid profiles, and glucose-insulin homeostasis were also assessed through standard biochemical assays.

What were the most important findings?

The key finding was a significantly elevated level of nickel (Ni) in the erythrocytes of obese women with PCOS compared to both nonobese PCOS women and healthy controls. No significant differences were observed in the levels of other trace elements (Zn, Fe, Mn, Cu, Mg) across the groups. However, several important correlations emerged within each PCOS subgroup. In nonobese women, zinc positively correlated with testosterone, while nickel correlated with estradiol and luteinizing hormone (LH). In obese women, zinc positively correlated with prolactin, magnesium with testosterone, and manganese negatively with thyroid-stimulating hormone (TSH). These trace element–hormone interactions suggest a potential modulatory role of trace elements in ovarian function, particularly via endocrine pathways disrupted in PCOS.

From a microbiome lens, elevated nickel levels are known to disturb microbial homeostasis by promoting pro-inflammatory taxa like Proteobacteria and reducing SCFA-producing species like Faecalibacterium prausnitzii. This dysbiosis contributes to chronic inflammation and hormonal imbalance, which are central to PCOS pathophysiology. Similarly, altered manganese and magnesium levels can disrupt antioxidant defenses, further affecting the gut barrier and endocrine signaling. The consistent zinc-prolactin and nickel-estradiol associations underscore trace elements as potential mediators of both hormonal dysregulation and microbiota shifts.

What are the implications of this study?

This study reveals that trace element imbalances may act as endocrine disruptors and contribute to the progression of PCOS. These mineral–hormone correlations offer a mechanistic explanation for how metabolic obesity intensifies reproductive dysfunction in PCOS through trace element–mediated oxidative stress and inflammation. The erythrocyte-based approach strengthens the clinical relevance, as it reflects chronic exposure rather than transient serum fluctuations. Clinically, monitoring erythrocyte nickel, zinc, magnesium, and manganese could help stratify PCOS patients based on metabolic risk and guide personalized nutritional or detoxification therapies. The implications for the microbiome are profound, suggesting that micronutrient-driven dysbiosis could be an underrecognized trigger of endocrine dysfunction in PCOS.

Long-term use of gonadotropin-releasing hormone analogs and hormone replacement therapy in the management of endometriosis: a randomized trial with a 6-year follow-up

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study investigated the long-term effects of gonadotropin-releasing hormone agonists (GnRHa), with and without hormone replacement therapy (HRT), on bone mineral density (BMD) in women with endometriosis, following up to six years after treatment.

Who was studied?

The study included forty-nine symptomatic women diagnosed with endometriosis via laparoscopy, who volunteered to participate in the randomized trial. These women were undergoing treatment with long-acting GnRH agonists and were subsequently monitored for up to six years.

 

What were the most important findings?

The study’s key findings indicate that long-term use of GnRH agonists results in a reduction in bone mineral density (BMD) at the lumbar spine and hip, a reduction that was not fully recovered even up to six years post-treatment. Additionally, the inclusion of hormone replacement therapy (HRT) did not significantly influence bone mineral density when compared to those who did not receive HRT. The results also demonstrated a considerable range of individual variability in BMD response among the participants, suggesting that the effects of GnRH agonists on BMD can vary significantly among different women.

 

What are the greatest implications of this study?

The implications of these findings are substantial for the management of endometriosis. The study underscores the potential risks associated with long-term GnRH agonist therapy, particularly concerning bone health, which may significantly influence treatment decisions and patient counseling. It also highlights the critical need for monitoring bone mineral density (BMD) in women undergoing prolonged GnRH agonist therapy, suggesting that this monitoring should possibly extend beyond the cessation of treatment. Furthermore, the observed variability in BMD response among participants indicates a need for individualized treatment plans, which may include exploring alternative therapies or preventive measures against bone density loss. Additionally, these findings could drive further research aimed at optimizing ‘add-back’ therapy to balance the effectiveness of GnRH agonists in managing endometriosis symptoms with the mitigation of side effects such as bone loss.

Low intakes of dietary fiber and magnesium are associated with insulin resistance and hyperandrogenism in polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study reveals that low fiber and magnesium intake—not overeating or inactivity—contribute significantly to insulin resistance and hyperandrogenism in PCOS, with strong implications for microbiome-linked interventions.

What was studied?

This study evaluated dietary and lifestyle contributors to insulin resistance (IR) and hyperandrogenism in women with polycystic ovary syndrome (PCOS), specifically focusing on fiber and magnesium intake. Conducted as an observational cohort study at a reproductive medicine center in Canada, researchers enrolled 87 women with PCOS and 50 subfertile women without PCOS. The study aimed to determine if differences in caloric intake or physical activity could explain obesity and metabolic abnormalities in PCOS, and whether specific dietary patterns or micronutrients were associated with PCOS phenotypes and IR.

Who was studied?

The participants included 87 women diagnosed with PCOS based on Rotterdam criteria and 50 control women without PCOS, all aged between 20 and 44. Among the PCOS group, some were classified as having hyperandrogenic PCOS (HA-PCOS) and rest as non-hyperandrogenic. The study population was ethnically diverse, comprising East Asian, European, South Asian, Aboriginal, and South American backgrounds. Participants completed 3-day dietary and activity logs, wore pedometers, and underwent hormonal and metabolic testing, including HOMA-IR calculations for insulin resistance.

What were the most important findings?

The most striking discovery was that women with PCOS, despite having higher BMI and waist-hip ratios, did not consume more calories nor engage in less physical activity than the control group. Instead, they consumed significantly less dietary fiber and magnesium. Within the PCOS group, those with IR had lower fiber and magnesium intakes and a higher glycemic load. Fiber intake was negatively correlated with several metabolic and hormonal markers, including HOMA-IR, fasting insulin, 2-hour glucose, triglycerides, testosterone, and DHEAS levels, while positively correlated with HDL cholesterol. Similarly, magnesium intake was inversely related to IR, C-reactive protein, and testosterone levels, and positively associated with HDL cholesterol.

Fiber and BMI together accounted for 54% of the variance in HOMA-IR, marking fiber intake as a major microbial-modifiable dietary factor. These associations are particularly relevant in the microbiome context since dietary fiber significantly impacts gut microbial composition, increasing short-chain fatty acid (SCFA)-producing bacteria like Faecalibacterium prausnitzii and Roseburia spp., which in turn can enhance insulin sensitivity and modulate systemic inflammation.

What are the greatest implications of this study?

This study challenges the assumption that obesity in PCOS is primarily due to overnutrition or inactivity and instead points clinicians toward specific nutritional targets, fiber and magnesium, as modifiable factors linked to metabolic dysfunction and androgen excess. The clinical implications are significant: increasing fiber and magnesium intake could become a frontline, non-pharmacological strategy in managing IR and hyperandrogenism in PCOS. Moreover, as fiber-rich diets influence microbiome diversity and function, these findings offer a mechanistic basis for future interventions targeting the gut microbiome in PCOS management. Integrating dietary fiber and magnesium monitoring into clinical assessments could better personalize nutrition-based care strategies for women with PCOS.

Lower Fiber Consumption in Women with Polycystic Ovary Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This meta-analysis confirms that women with PCOS consume significantly less dietary fiber than controls, independent of calorie intake. Reduced fiber may worsen PCOS via gut dysbiosis and diminished SCFA production.

What was reviewed?

This meta-analysis synthesized data from 13 observational studies to evaluate whether women with polycystic ovary syndrome (PCOS) consume significantly less dietary fiber than women without PCOS. The analysis sought to determine if reduced fiber intake is an overlooked factor contributing to the metabolic and endocrine dysfunction commonly seen in PCOS. Additionally, the review assessed whether total caloric intake differed between groups and explored how geographic location, dietary assessment methods, and body mass index (BMI) might influence findings. The research incorporated studies conducted across Asia, Europe, North America, and South America, using diverse methodologies such as food diaries, recalls, and food frequency questionnaires (FFQs).

Who was reviewed?

The included studies encompassed a total of 2,469 women, 1,130 with PCOS and 1,339 controls. Participants spanned diverse age groups (adolescent and adult women) and body weight categories (lean and overweight/obese). Most studies used the Rotterdam criteria for PCOS diagnosis. Dietary fiber intake data were extracted using self-reported dietary tools over periods ranging from 1 to 7 days. Nine of the thirteen studies were deemed high quality using the Newcastle–Ottawa Scale. The meta-analysis particularly emphasized studies not adjusted for total energy intake, although two studies did include such adjustments.

What were the most important findings?

The pooled analysis revealed that women with PCOS had significantly lower absolute dietary fiber intake compared to controls, despite no significant difference in total caloric intake. This suggests that the reduction in fiber is independent of overall energy consumption and reflects a dietary quality issue rather than quantity. Subgroup analysis confirmed that this difference persisted across various continents, dietary methods (especially food recall and diary), and study designs (notably case–control). Importantly, the difference was also maintained in adult populations and studies using the Rotterdam criteria for diagnosis.

Though the meta-analysis did not directly assess microbiome composition, the findings carry strong implications. Fiber is a critical prebiotic nutrient known to foster the growth of beneficial microbial taxa such as Faecalibacterium prausnitzii, Bifidobacterium spp., and Lactobacillus spp. These microbes produce short-chain fatty acids—notably butyrate—that regulate systemic inflammation, insulin sensitivity, gut barrier integrity, and even reproductive hormone modulation. A diet deficient in fiber diminishes SCFA production, reduces microbial diversity, and allows expansion of pro-inflammatory taxa like Collinsella and Proteobacteria. These alterations are well aligned with the dysbiosis commonly documented in PCOS, suggesting that low fiber intake may be both a symptom and driver of gut microbiota imbalance in this population.

What are the greatest implications of this review?

This meta-analysis establishes that reduced dietary fiber intake is a consistent dietary pattern in women with PCOS, regardless of caloric intake or body weight. The findings reinforce the need for clinicians to go beyond calorie counting and assess the qualitative aspects of dietary intake, particularly fiber. Given fiber's central role in modulating the gut microbiome and producing SCFAs, inadequate intake could perpetuate insulin resistance, chronic inflammation, and hyperandrogenism in PCOS. These data provide a rationale for integrating dietary fiber intake goals into clinical guidelines for PCOS management. Moreover, the gut–diet–hormone axis illuminated by this review highlights an urgent need for interventional studies targeting fiber intake, either through food-based strategies or supplementation, as a means to correct dysbiosis and metabolic dysfunction in PCOS. Clinicians should consider fiber assessment and counseling as standard practice in the nutritional management of women with PCOS.

Mediterranean Diet Combined With a Low-Carbohydrate Dietary Pattern in the Treatment of Overweight Polycystic Ovary Syndrome Patients

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

A Mediterranean/low-carb diet significantly improved insulin resistance, androgen levels, and weight in overweight PCOS patients. The findings suggest microbiome-mediated metabolic benefits and support clinical adoption of this dietary model over traditional low-fat diets.

What was studied?

This randomized controlled clinical trial evaluated the effects of a combined Mediterranean and low-carbohydrate (MD/LC) dietary pattern compared to a standard low-fat diet in overweight women diagnosed with polycystic ovary syndrome (PCOS). Conducted over 12 weeks, the study aimed to determine whether this novel dietary model could more effectively improve anthropometric, metabolic, and reproductive endocrine parameters. While the low-fat diet restricted fat intake to under 30% of daily calories, the MD/LC model restricted carbohydrate intake to under 100 g/day, increased fat and protein intake, and emphasized traditional Mediterranean components like whole grains, olive oil, vegetables, legumes, and seafood.

Who was studied?

Seventy-two overweight women (BMI ≥ 24 kg/m²), aged 16–45 years, with a PCOS diagnosis based on the Rotterdam criteria were recruited from Changhai Hospital in China. After exclusions and dropouts, 30 participants completed the Mediterranean and low-carbohydrate diet and 29 completed the LF diet. All participants were of Chinese descent, had no other endocrine, cardiovascular, or metabolic conditions, and were not taking medications affecting insulin or lipid metabolism. Dietary intake was closely monitored, and participants were supported with professional dietetic guidance via digital communication tools.

What were the most important findings?

The MD/LC diet group demonstrated significantly greater reductions in body weight, BMI, waist circumference, body fat percentage, and waist-to-hip ratio than the LF group. Notably, the MD/LC group also experienced more profound improvements in metabolic markers: fasting insulin, HOMA-IR, and triglyceride levels dropped more substantially, while insulin sensitivity measured by QUICKI improved to a greater extent. Reproductive hormone profiles also responded better to the MD/LC diet. Total testosterone, luteinizing hormone (LH), and LH/FSH ratio all decreased more prominently in the MD/LC group, suggesting improved ovulatory function and androgen regulation.

Though the study did not measure microbiome outcomes directly, the dietary model has strong implications for gut microbial modulation. The Mediterranean diet is known to promote increased abundance of beneficial microbes such as Faecalibacterium prausnitzii, Bifidobacterium spp., and Lactobacillus spp., while low-carbohydrate diets reduce the fermentable carbohydrate load, possibly altering SCFA-producing bacteria ratios. The observed improvements in insulin sensitivity, inflammation markers, and lipid metabolism align with known microbiome-mediated metabolic pathways, suggesting that the MD/LC diet could drive favorable microbial shifts that improve PCOS outcomes.

What are the implications of this study?

This study highlights the superior efficacy of a calorie-restricted Mediterranean/low-carbohydrate diet in improving the key clinical features of PCOS compared to a traditional low-fat diet. Clinically, this offers a refined, evidence-based dietary model that targets obesity, insulin resistance, and hyperandrogenism—three interlinked hallmarks of PCOS. Importantly, the diet’s structure promotes microbial diversity and metabolite production, suggesting a dual mechanism of action: direct endocrine and metabolic effects, and indirect microbiome-mediated modulation. For clinicians managing PCOS, especially in overweight patients, the MD/LC model provides a practical and sustainable intervention that integrates metabolic restoration with potential microbiome optimization. Future trials with microbiome sequencing are warranted to validate these microbial associations and inform personalized nutrition strategies in PCOS treatment.

Meta-analysis of gut microbiome studies identifies disease-specific and shared responses

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This meta-analysis standardized and re-analyzed data from 28 gut microbiome studies across ten diseases, identifying consistent microbiome signatures associated with specific diseases and a non-specific response common to multiple conditions. Key findings suggest both potential microbial diagnostics and treatments, emphasizing the importance of understanding shared versus disease-specific microbial responses in future research and clinical applications.

What was studied?

The meta-analysis focused on the human gut microbiome’s association with various diseases by analyzing 28 published case-control gut microbiome studies covering ten diseases. The researchers aimed to standardize the processing and analysis of these datasets to identify consistent patterns and shifts in the gut microbiome associated with specific diseases or a generalized health-disease spectrum.

Who was studied?

The participants of the original case-control studies comprised individuals with different diseases, including colorectal cancer, inflammatory bowel disease (IBD), and others, alongside control groups of healthy individuals. The meta-analysis integrated data only from studies with publicly available 16S amplicon sequencing data of stool samples from at least 15 case patients, excluding studies focused solely on children under 5 years old.

 

What were the most important findings?

Consistent Microbial Patterns: The meta-analysis revealed consistent and specific microbiome changes associated with various diseases. For instance, diseases like colorectal cancer showed an enrichment of pathogenic bacteria, while a depletion of health-associated bacteria marked conditions like IBD.

Non-Specific Microbial Responses: A significant finding was that many microbial associations are not disease-specific but rather indicate a non-specific response shared across multiple disease states. Approximately half of the genera identified were common to more than one disease, suggesting a generalized microbial response to disease states rather than unique disease-specific signatures.

Diagnostic and Therapeutic Implications: The study identified distinct categories of dysbiosis (microbial imbalance) that could guide the development of microbiome-based diagnostics and therapeutics. For example, enriching for depleted beneficial microbes could be a strategy for diseases characterized by such depletions.

 

What are the greatest implications of this meta-analysis?

Improved Disease Understanding: By providing a clearer picture of the microbiome’s role in various diseases, the study helps refine our understanding of disease mechanisms and potential microbial contributions to disease processes.

Guidance for Future Research: The findings suggest that future microbiome research in disease contexts should consider the non-specificity of many microbial changes. This realization could influence how researchers design studies and interpret results, potentially focusing on truly disease-specific microbial signatures.

Clinical Applications: The identification of consistent microbial patterns and signatures across diseases opens pathways to developing novel diagnostics and therapies, such as probiotics or fecal microbiota transplants, targeted at restoring healthy microbial communities or addressing specific dysbioses.

Data Sharing and Standardization: The study underscores the value of making raw data and metadata from microbiome studies publicly available and highlights the benefits of using standardized methods for data processing and analysis to compare and integrate results across studies.

Overall, this meta-analysis clarifies the microbiome’s role in disease and sets a framework for future research and clinical applications by demonstrating the importance of understanding both disease-specific and non-specific microbial responses.

Metabolomics reveals perturbations in endometrium and serum of minimal and mild endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metabolomic Signature
    Metabolomic Signature

    Metabolomic signatures are unique metabolite patterns linked to specific biological conditions, identified through metabolomics. They reveal underlying biochemical activities, aiding in disease diagnosis, biomarker development, and personalized medicine. The microbiome significantly affects these signatures, influencing health and disease outcomes through metabolic interactions.

This study highlights metabolic alterations in endometrial tissue and serum of minimal and mild endometriosis patients, identifying amino acid biomarkers with diagnostic potential. A combined serum panel demonstrated 100% sensitivity and 83% specificity for Stage II diagnosis, offering a promising step toward non-invasive early detection of endometriosis.

What Was Studied?

This study investigated the metabolic perturbations in eutopic endometrial tissue and serum of women with minimal and mild endometriosis (Stages I and II) using ^1H Nuclear Magnetic Resonance (NMR)-based metabolomics. The researchers aimed to identify specific metabolites that could be potential biomarkers for the early, non-invasive diagnosis of endometriosis. The study included multivariate and univariate analyses to identify metabolite changes and their diagnostic potential.

Who Was Studied?

The study included 95 women diagnosed with endometriosis (staged using the revised American Society for Reproductive Medicine criteria) and 24 healthy fertile controls. The participants were recruited from Eastern India and Bangladesh, with exclusion criteria ensuring no confounding conditions such as ovarian tumors or pelvic inflammatory disease. Blood and eutopic endometrial tissue samples were collected during the mid-secretory phase of the menstrual cycle.

What Were the Most Important Findings?

Women with minimal and mild endometriosis exhibited significant metabolic alterations, particularly in amino acids. Alanine, lysine, leucine, proline, and phenylalanine levels were notably dysregulated in serum, with tissue samples showing lower levels of these metabolites, except for proline, which positively correlated with serum levels. Alanine alone demonstrated diagnostic potential for Stage I endometriosis, with 90% sensitivity and 58% specificity.

For Stage II, phenylalanine achieved 100% sensitivity but had lower specificity, while a combined panel of metabolites improved diagnostic accuracy, reaching 100% sensitivity and 83% specificity.

In advanced stages, elevated taurine and myo-inositol levels were linked to increased cell proliferation and angiogenesis, highlighting similarities with tumorigenic processes. These findings underscore the critical role of metabolic shifts in endometriosis progression, particularly involving amino acids and nucleotide synthesis, and suggest their utility in early detection and non-invasive diagnostics.

What Are the Greatest Implications of This Study?

This study underscores the potential of metabolomic signatures in elucidating the pathophysiology of endometriosis and developing non-invasive diagnostic tools, especially for early stages where traditional biomarkers like CA-125 are less effective. By identifying a panel of serum metabolites, the research provides a foundation for improving diagnostic accuracy and reducing the need for invasive laparoscopy. Additionally, the observed metabolic similarities between endometriosis and malignancies could inspire further exploration of shared mechanisms, potentially broadening therapeutic targets.

Metalloestrogens: an emerging class of inorganic xenoestrogens with potential to add to the oestrogenic burden of the human breast

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review opens new avenues in toxicology and endocrine research, identifying metalloestrogens as a critical factor in hormone disruption and breast cancer risk. Further studies are necessary to confirm these findings and develop effective mitigation strategies for human health protection.

What was reviewed?

This study, published in the Journal of Applied Toxicology, reviewed the concept and emerging evidence of metalloestrogens mimicking estrogenic activity. The review focused on how these metals interact with estrogen receptors (ERs) like organic xenoestrogens, potentially contributing to estrogenic activity in human breast tissue and increasing the risk of hormone-related cancers such as breast cancer. The review primarily covered in vitro and in vivo studies of various metal ions, including aluminum, antimony, arsenite, barium, cadmium, chromium (Cr(II)), cobalt, copper, lead, mercury, nickel, selenite, tin, and vanadate. The review also highlights significant research contributions from multiple studies and scholars focusing on the effects of metalloestrogens on human breast cancer cell lines, such as MCF-7 and T47D, as well as their impact on gene expression and cellular proliferation.

Most Important Findings:

Estrogenic Activity of Metals: The review found that various metal ions can act as estrogen agonists by binding to estrogen receptors, particularly ERα, and mimicking the actions of physiological estrogens. This was demonstrated in studies showing that metals such as cadmium, nickel, and aluminum could displace estradiol from the ligand-binding domain of ERα, leading to altered gene expression and increased cell proliferation in breast cancer cells.

Molecular Mechanisms: Metals such as cadmium were shown to bind directly to the ligand-binding domain (LBD) of the estrogen receptor, interfering with the receptor's normal function. This binding alters the receptor’s ability to interact with estrogen response elements (EREs) on DNA, thereby affecting the transcription of estrogen-regulated genes. For instance, cadmium was found to downregulate ER levels and upregulate estrogen-regulated gene expression, driving cell proliferation.

Cooperative Action with Estrogens: The metals did not antagonize estradiol’s action; instead, they often enhanced the agonist actions of estradiol. In some cases, metals like copper and cobalt increased breast cancer cell proliferation when combined with estradiol, indicating a synergistic effect that may exacerbate estrogenic signaling in hormone-dependent cancers.

In Vivo Evidence: The review highlighted evidence of in vivo estrogenic activity in animal models, particularly for cadmium, which was shown to increase uterine weight, induce mammary gland development, and alter gene expression. The estrogenic effects of cadmium were noted at doses relevant to human exposure, raising significant concerns about environmental exposure to these metals.

Environmental and Occupational Exposure: The presence of metalloestrogens such as cadmium and aluminum in everyday consumer products (e.g., antiperspirants) and the environment (e.g., tobacco smoke, and industrial pollutants) implies widespread human exposure. These metals can accumulate in the body, especially in breast tissue, and may contribute to the burden of aberrant estrogen signaling involved in breast cancer development.

Greatest Implications:

Breast Cancer Risk: The review underscores the potential for metalloestrogens to increase the risk of breast cancer by contributing to estrogenic signaling within breast tissue. Given that breast cancer is often driven by estrogen receptor activation, the cumulative burden of environmental estrogens and metalloestrogens could enhance the likelihood of cancer development and progression.

Environmental Health and Toxicology: The widespread presence of these metals in the environment, their ability to accumulate in the body, and their newly recognized estrogenic activity suggest a need for revised regulatory guidelines and risk assessments for human exposure to metalloestrogens. This includes re-evaluating safe exposure levels, especially for metals like cadmium, which is already classified as a human carcinogen.

Endocrine Disruption: The concept of metalloestrogens extends the traditional understanding of endocrine-disrupting chemicals (EDCs) beyond organic compounds, emphasizing the need for further investigation into how inorganic metals may impact hormone-related diseases. This review calls for more research on the long-term effects of chronic exposure to metalloestrogens in both wildlife and humans.

Public Health Awareness: There is a strong implication for public health education regarding the sources of metalloestrogen exposure, such as antiperspirants, diet, cigarette smoke, and industrial pollutants. Raising awareness could lead to better personal care practices and lifestyle choices to reduce individual exposure to these potentially harmful metal ions.

Metformin use in women with polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

This review discusses the role of metformin in treating PCOS-related infertility. It highlights its effectiveness in improving ovulation, reducing insulin resistance, and managing hyperandrogenism. Metformin is recommended as a first-line treatment for non-obese women with PCOS-related infertility, with benefits also seen in IVF and metabolic health.

What was reviewed?

This review examines the role of metformin in managing polycystic ovary syndrome (PCOS), specifically focusing on its effectiveness in treating PCOS-related infertility. The paper reviews evidence from randomized controlled trials (RCTs) and other studies, discussing the use of metformin as an insulin-sensitizing agent for women with PCOS who experience anovulatory infertility. It also explores metformin’s impact on metabolic dysfunctions, hyperandrogenism, and its potential use alongside other treatments like clomiphene for improving fertility outcomes in women with PCOS.

Who was reviewed?

The review considers various studies and clinical trials on the use of metformin in women with PCOS. These studies involve women with varying degrees of obesity and insulin resistance, who are experiencing anovulatory infertility, hyperandrogenism, or both. The review synthesizes results from RCTs that examined the effectiveness of metformin alone or in combination with other treatments like clomiphene citrate and aromatase inhibitors in improving ovulation, fertility, and reducing the metabolic disturbances associated with PCOS.

What were the most important findings?

The review highlights several key findings regarding the use of metformin in treating PCOS-related infertility. Metformin has shown efficacy in improving ovulation rates in women with anovulatory infertility, particularly in non-obese women. A Cochrane review of seven RCTs revealed that metformin significantly increased clinical pregnancy rates compared to placebo. However, while metformin showed promise, it did not outperform clomiphene citrate as a first-line treatment for ovulation induction in women with PCOS, particularly in obese patients. The review also found that metformin, when used in combination with clomiphene, can be effective for women who are resistant to clomiphene alone.

Additionally, the review emphasized that metformin has benefits beyond fertility induction. It helps reduce hyperinsulinemia and insulin resistance, which are common in women with PCOS, and can improve associated metabolic conditions such as dyslipidemia and obesity. Furthermore, metformin was found to reduce the risk of ovarian hyperstimulation syndrome (OHSS) in women undergoing in vitro fertilization (IVF). Although metformin’s role in improving long-term health outcomes, such as the prevention of type 2 diabetes, cardiovascular disease, and endometrial cancer, remains inconclusive, it offers significant short-term reproductive benefits.

What are the greatest implications of this review?

The review suggests that metformin should be considered a suitable first-line treatment for non-obese women with anovulatory infertility due to PCOS. For women who are resistant to clomiphene or prefer an alternative to the oral contraceptive pill (OCP) for managing hyperandrogenic symptoms, metformin can be an effective option. Additionally, metformin’s role in reducing the risk of OHSS during IVF procedures underscores its importance in assisted reproductive treatments. The review also raises the need for further research to better define metformin’s long-term benefits in preventing the metabolic and reproductive complications associated with PCOS, as well as its potential to improve long-term health outcomes like diabetes prevention.

Microbial Dysbiosis Is Associated with Human Breast Cancer

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

Microbial dysbiosis, marked by reduced bacterial load and altered species composition, is linked to breast cancer progression. Enrichment of Methylobacterium radiotolerans in tumors and depletion of Sphingomonas yanoikuyae in normal tissue suggest diagnostic and therapeutic potential for microbiota-based interventions in breast cancer.

What Was Studied?

This study examined the microbiota present in breast tumor tissue compared to paired normal breast tissue from the same individuals, as well as healthy breast tissue from individuals without breast cancer. Using next-generation sequencing and quantitative PCR, the research aimed to identify differences in microbial composition, bacterial load, and their potential impact on the tumor microenvironment and breast cancer progression.

Who Was Studied?

The study included 20 breast cancer patients with estrogen receptor-positive (ER+) tumors, for whom paired tumor and normal adjacent tissue were analyzed. Additional bacterial load analysis included 23 healthy controls undergoing reduction mammoplasty. Gene expression profiling was conducted on tissue from six breast cancer patients and three healthy individuals.

Most Important Findings

The study revealed distinct microbial signatures associated with breast cancer. Methylobacterium radiotolerans was significantly enriched in tumor tissue, while Sphingomonas yanoikuyae was more abundant in paired normal tissue. A strong inverse correlation between the abundance of these two species was observed in normal tissue, but not in tumor tissue. Importantly, bacterial load in tumor tissue was markedly reduced compared to both paired normal and healthy breast tissue, with advanced-stage tumors exhibiting the lowest bacterial counts. This reduction in bacterial load correlated with decreased expression of antibacterial response genes, including Toll-like receptors (TLR2, TLR5, and TLR9) and antimicrobial effectors like IL-12A and BPI.

These findings suggest that microbial dysbiosis and a diminished antibacterial immune response in tumor tissue may contribute to breast cancer progression. Additionally, the results highlight the potential diagnostic value of bacterial load as a marker for breast cancer staging.

Greatest Implications

The association between microbial dysbiosis and breast cancer offers novel insights into the disease’s pathogenesis. The depletion of beneficial bacteria, such as Sphingomonas yanoikuyae, and a reduced immune response may create a permissive environment for tumorigenesis. This study supports the exploration of microbiota as a diagnostic tool and potentially as a therapeutic target to restore a healthy microbial balance and enhance immune surveillance. The inverse correlation between bacterial load and tumor stage underscores its potential utility in disease staging and progression monitoring.

Microbiome of the lower genital tract in Chinese women with endometriosis by 16s-rRNA sequencing technique: a pilot study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This case-control study investigates the relationship between genital tract microbiota and endometriosis using 16s-rRNA sequencing. Findings show Atopobium prevalence in endometriosis with adenomyosis cases, highlighting microbiota's distinct functions.

What was studied?

The study investigated the relationship between the genital tract microbiota and endometriosis, particularly focusing on how microbiota diversity and specific bacteria like Atopobium might be associated with the disease.

 

Who was studied?

68 participants, from whom 134 samples were collected from the cervical canal, posterior fornix, and uterine cavity for 16s-rRNA sequencing, were included in the study.

 

What were the most important findings?

Key findings included no significant differences in alpha diversity between the cervical canal and posterior fornix. However, the microbiota profile of patients with adenomyosis and endometriosis differed markedly from the control group, with Atopobium showing significant prevalence in these patients. While no specific biomarkers were identified, PICRUSt analysis revealed several characteristic microbiota functions.

 

What are the greatest implications of this study?

The study suggests a potentially significant role of microbiota, particularly Atopobium, in the pathogenesis of endometriosis combined with adenomyosis. This finding could lead to new insights into the microbiota-immune-endometriosis system interaction, offering new avenues for understanding and possibly treating endometriosis and adenomyosis. Further research is needed to verify the functions of the microbiota identified and their direct association with the diseases.

Microbiome of the lower genital tract in Chinese women with endometriosis by 16s-rRNA sequencing technique: a pilot study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies microbiome shifts in the lower genital tract of Chinese women with endometriosis, marked by Atopobium enrichment and changes in ribosome biogenesis and immune modulation. Findings suggest potential non-invasive biomarkers and therapeutic targets for endometriosis and adenomyosis.

What Was Studied?

This study investigated the microbiome composition of the lower genital tract in Chinese women with endometriosis using 16S rRNA sequencing. Researchers aimed to explore differences in microbial diversity and specific bacterial populations between women diagnosed with endometriosis and healthy controls. The analysis focused on samples collected from the cervical canal, posterior fornix, and uterine cavity to identify distinct microbial profiles and potential biomarkers associated with endometriosis and adenomyosis. A total of 68 participants were included in the study, with 134 samples collected and processed for microbiome analysis.

Who Was Studied?

The study involved 68 women aged 18 to 45, divided into groups: 20 with endometriosis (EM), 19 with adenomyosis (AM), 7 with both adenomyosis and endometriosis (AMEM), and 36 healthy controls (CT). Samples were collected from the cervical canal, posterior fornix, and uterine cavity using sterile techniques to minimize contamination. Participants were excluded if they had bacterial vaginosis (BV), pelvic inflammatory disease, recent antibiotic use, or other infections that might influence microbial composition. This design allowed for a focused investigation of microbiota differences in the lower genital tract specifically related to endometriosis and adenomyosis.

What Were the Most Important Findings

The study found that Lactobacillus remained the predominant genus in the lower genital tract across all groups, reflecting typical vaginal flora. However, significant microbial shifts were observed in women with adenomyosis-endometriosis (AMEM). At the genus level, Atopobium was notably enriched in AMEM patients, representing the most significant microbial difference. While Lactobacillus dominance persisted, AMEM patients exhibited a substantial increase in Coriobacteriaceae and Campylobacteriaceae, indicating a divergence from healthy microbial communities. The AMEM group also displayed elevated levels of Coriobacteriales at the order level, further distinguishing it from other disease groups and controls. Interestingly, LeFSe analysis did not identify specific biomarkers between the cervical canal and posterior fornix, but PICRUSt analysis suggested functional differences, particularly in ribosome biogenesis and two-component system regulation, which may influence immune modulation in endometriosis. The findings suggest that Atopobium may contribute to the pathogenesis of endometriosis through immune disruption, potentially mediated by increased levels of IL-1β, which has been linked to chronic inflammation. The consistent presence of Coriobacteriaceae and Campylobacteriaceae in AMEM further supports a hypothesis of localized dysbiosis contributing to inflammatory processes.

What Are the Greatest Implications of This Study?

This study provides evidence of distinct microbiome alterations in the lower genital tract of women with endometriosis, particularly among those with concurrent adenomyosis. The enrichment of Atopobium and Coriobacteriaceae suggests that specific microbial communities may influence inflammatory pathways and disease progression in endometriosis. The identification of ribosome biogenesis and two-component system regulation as significant functional pathways indicates that microbial-induced immune modulation could play a role in lesion persistence and symptom severity. These findings propose Atopobium as a potential microbial biomarker for adenomyosis-endometriosis and highlight the possibility of targeted microbiome-based therapies to disrupt pathological microbial communities. The study underscores the importance of microbial profiling as a non-invasive diagnostic tool for identifying endometriosis-related dysbiosis, potentially leading to earlier detection and intervention.

Microbiome Profile of Deep Endometriosis Patients: Comparison of Vaginal Fluid, Endometrium and Lesion

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This case study found that a distinct bacterial composition was observed in deep endometriotic lesions, characterized by a reduced prevalence of Lactobacillus and an increased abundance of Alishewanella, Enterococcus, and Pseudomonas.

What was studied?

The study focused on identifying and comparing the bacterial patterns present in the vaginal fluid, eutopic endometrium, and endometriotic lesions of patients with endometriosis to those found in the vaginal fluid and eutopic endometrium of control patients without the disease. High-throughput DNA sequencing of the 16S rRNA marker gene was utilized to analyze the microbiome profile in these different biological samples from both groups.

 

Who was studied?

Twenty-one patients participated in this study, divided into two groups: eleven in the control group and ten in the endometriotic group. The control group consisted of women who underwent laparoscopic surgery for benign gynecological diseases or elective tubal ligation, where the absence of endometriosis was confirmed during peritoneal cavity inspection. The endometriotic group included only women with deep endometriosis, confirmed by laparoscopic surgery and histopathology analysis.

 

What were the most important findings?

The study found that microbiome sequencing of vaginal fluid, eutopic endometrium, and endometriotic lesions typically showed similar profiles, dominated by Lactobacillus, Gardnerella, Streptococcus, and Prevotella. Despite no significant overall differences in microbiome diversity between control and endometriotic patients, deep endometriotic lesions exhibited a distinct bacterial composition with less Lactobacillus and a higher abundance of Alishewanella, Enterococcus, and Pseudomonas.

 

What are the greatest implications of this study?

The study highlights several implications for endometriosis management: It provides insights into the pathogenesis by showing distinct bacterial compositions in deep lesions, suggesting microbial involvement in lesion development. This leads to the potential for non-invasive diagnostics by identifying specific microbial patterns, opening avenues for biomarker-based detection. Therapeutically, interventions like antibiotics or probiotics could be new treatment strategies if certain bacteria contribute to pathogenesis. The findings emphasize the need for more research to establish causal links between microbiome composition and endometriosis, understand the bacterial influence on the disease, and explore microbiome-based treatments.

Microbiome Profile of Deep Endometriosis Patients: Comparison of Vaginal Fluid, Endometrium and Lesion

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study reveals a distinct microbiome profile in deep endometriosis, with unique bacterial communities in lesion sites, suggesting a role in inflammation and disease progression. Potentially pathogenic genera like Pseudomonas and Alishewanella were abundant, highlighting their possible involvement in endometriotic lesion maintenance and inflammatory responses.

What was studied?

This study explored the microbiome profile in deep endometriosis by comparing the bacterial composition of vaginal fluid, eutopic endometrium, and deep endometriotic lesions. The primary goal was to identify whether distinct microbial patterns exist in these tissue sites of women with deep endometriosis compared to those without the condition. To achieve this, researchers employed high-throughput 16S rRNA sequencing to analyze microbial DNA extracted from tissue samples collected during laparoscopic surgery. The analysis included alpha and beta diversity assessments to determine variations in microbial richness and community structure across different tissue types.

Who was studied?

The study involved 21 participants, including 10 women with histologically confirmed deep endometriosis and 11 healthy controls undergoing laparoscopic surgery for benign gynecological conditions or elective tubal ligation. Samples were obtained from three anatomical sites: vaginal fluid, eutopic endometrium, and deep endometriotic lesions. Participants were carefully screened to exclude those with recent antibiotic, antifungal, or probiotic use, as well as those with autoimmune or inflammatory diseases that could affect microbiome composition.

What were the most important findings?

Microbiome analysis revealed significant differences in bacterial profiles between deep endometriotic lesions, eutopic endometrium, and vaginal fluid. In vaginal fluid and endometrial samples, the microbiome was predominantly composed of Lactobacillus, Gardnerella, Streptococcus, and Prevotella. In contrast, deep endometriotic lesions exhibited a distinct microbial landscape with reduced Lactobacillus and higher relative abundances of Alishewanella, Enterococcus, and Pseudomonas. Notably, Alishewanella and Pseudomonas were almost exclusively found in lesion samples, suggesting these genera may be associated with the inflammatory microenvironment characteristic of deep endometriosis.

Further analysis indicated that deep endometriotic lesions had greater microbial diversity than both vaginal fluid and eutopic endometrium. Beta diversity analysis showed a statistically significant difference in microbial community structure between lesion samples and the other tissue sites (p = 0.036), suggesting that endometriotic tissue supports a unique microbiome that could influence local immune responses and inflammation. These findings point towards a potential role for certain bacteria in the pathogenesis of deep endometriosis, either by promoting inflammation or altering tissue homeostasis.

Microbial GroupDeep Endometriosis FindingsClinical Implications
LactobacillusDecreased in lesion samplesReduction may contribute to loss of protective barrier
AlishewanellaIncreased in lesionsPotential involvement in local inflammation
PseudomonasIncreased in lesionsLinked to immune modulation and tissue inflammation
EnterococcusElevated in lesion samplesKnown for its association with chronic inflammation
Alpha DiversityIncreased in lesions compared to other sitesSuggests a unique microbial community fostering inflammation
Beta DiversitySignificantly different from endometrium and vaginal fluid (p = 0.036)Indicates unique microbial signature in lesions

What are the greatest implications of this study?

The results of this study underscore the presence of a unique microbiome profile in deep endometriotic lesions, characterized by increased microbial diversity and the presence of potentially pathogenic bacteria like Pseudomonas and Alishewanella. These findings suggest that microbiome alterations may contribute to the inflammatory environment observed in endometriosis, potentially influencing disease progression and symptom severity. This study opens avenues for further investigation into microbiome-targeted therapies aimed at modulating bacterial communities in endometriotic tissue to alleviate inflammation and inhibit lesion growth. Additionally, the distinct microbial signatures identified in deep endometriosis lesions may serve as non-invasive biomarkers for early detection and improved clinical management of the disease.

Microbiota composition and distribution along the female reproductive tract of women with endometriosis

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This cross-sectional observational study compared microbiota in various reproductive tract locations finding significant microbiota changes in endometriosis patients, especially a decrease in Lactobacillus and increase in specific bacteria in the cervical area.

What was studied?

The study investigated the microbiota distribution across the entire female reproductive tract of endometriosis (EMS) patients and non-EMS women, aiming to identify EMS-specific bacterial species and examine the relationship between flora and disease development.

 

Who was studied?

Fifty women undergoing laparoscopic surgery for benign gynecological diseases or pelvic endometriosis at Peking University Shenzhen Hospital were studied. They were divided into two groups: 36 with pelvic endometriosis (stages I-IV) and 14 controls without endometriosis symptoms.

 

What were the most important findings?

Significant differences in the microbiota distribution were observed, especially a decrease in Lactobacillus in the upper reproductive tract of EMS patients. Specific Operational Taxonomic Units (OTUs), particularly Sphingobium sp. and Pseudomonas viridiflava, were identified as significantly enriched in the endometrium and peritoneal fluid of EMS patients, suggesting their potential role in EMS pathogenesis.

 

What are the greatest implications of this study?

The study offers a new perspective on the pathogenesis of endometriosis, emphasizing the role of specific bacteria in its development. Identifying microbiota changes associated with EMS could lead to novel diagnostic markers and therapeutic targets, enhancing our understanding of the disease and potentially leading to more effective management strategies for endometriosis and other female reproductive tract diseases.

Microbiota composition and distribution along the female reproductive tract of women with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study maps the microbiota composition along the reproductive tract in endometriosis patients, revealing microbial dysbiosis, reduced Lactobacillus, and increased Pseudomonas and Sphingobium in upper reproductive sites. These findings suggest potential microbial contributions to inflammation, lesion persistence, and new biomarkers for diagnosing endometriosis.

What Was Studied?

This study investigated the microbiota composition and distribution along the female reproductive tract of women with endometriosis (EMS) compared to healthy controls. Using 16S rRNA amplicon sequencing, researchers mapped microbial communities from five distinct anatomical sites: the lower third of the vagina (CL), posterior vaginal fornix (CU), cervical mucus (CV), endometrium (ET), and peritoneal fluid (PF). The primary aim was to identify differences in microbial communities across these regions and understand how these alterations may contribute to the pathogenesis of endometriosis. A total of 36 women with confirmed pelvic endometriosis and 14 healthy controls undergoing surgery for benign gynecological conditions were included in the study.

Who Was Studied?

The study included 50 women aged 23 to 44, divided into two groups: 36 patients diagnosed with pelvic endometriosis and 14 healthy controls undergoing laparoscopic surgery for benign conditions like ovarian teratoma, serous cystadenoma, or uterine fibroids. All participants had regular menstrual cycles and had not used hormonal drugs, antibiotics, or vaginal medications within six months before sample collection. Samples were collected during the early follicular phase to minimize hormonal variability, and five distinct anatomical locations were sampled to comprehensively map microbial distribution.

What Were the Most Important Findings?

The study revealed distinct microbiota compositions along the reproductive tract in women with endometriosis compared to healthy controls. The lower reproductive tract (CL, CU) in both groups was predominantly colonized by Lactobacillus, maintaining a stable microbial community. However, microbial diversity began to shift notably at the cervical mucus (CV) in endometriosis patients, with the appearance of Veillonellaceae and an overall reduction in Lactobacillus. This shift continued upward, with dramatic changes observed in the endometrium (ET) and peritoneal fluid (PF) of endometriosis patients. In the ET, Pseudomonas, Acinetobacter, and Vagococcus emerged as dominant taxa, accompanied by a significant reduction of Lactobacillus. The peritoneal fluid samples further diverged, showing high microbial diversity, including Pseudomonas, Acinetobacter, Shewanella, Vagococcus, and Sphingobium, with minimal presence of Lactobacillus. The distinct microbial signatures in these upper reproductive sites suggest a potential role for these genera in promoting inflammation and lesion persistence in endometriosis. Importantly, Sphingobium and Pseudomonas viridiflava were consistently enriched in both the ET and PF of endometriosis patients, indicating their potential as microbial biomarkers for the disease. The findings support the hypothesis that site-specific microbial shifts contribute to the pathogenesis of endometriosis, likely through immune modulation and local inflammation.

Anatomical SiteMicrobiota Findings in Endometriosis Patients
Lower Reproductive Tract (CL, CU)Predominantly colonized by Lactobacillus, maintaining a stable microbial community.
Cervical Mucus (CV)Notable microbial shift with the appearance of Veillonellaceae and a significant reduction in Lactobacillus.
Endometrium (ET)Dominated by Pseudomonas, Acinetobacter, and Vagococcus with a marked reduction of Lactobacillus.
Peritoneal Fluid (PF)High microbial diversity with Pseudomonas, Acinetobacter, Shewanella, Vagococcus, and Sphingobium. Minimal Lactobacillus presence.
Unique EnrichmentsConsistent enrichment of Sphingobium and Pseudomonas viridiflava in ET and PF, suggesting potential biomarkers.

What Are the Greatest Implications of This Study?

The study’s findings suggest that the microbiota composition along the female reproductive tract in endometriosis patients is markedly different from that of healthy controls, with key disruptions beginning in the cervical mucus and intensifying in the endometrium and peritoneal fluid. The reduction of Lactobacillus and the enrichment of pro-inflammatory taxa such as Pseudomonas, Acinetobacter, Vagococcus, Shewanella, and Sphingobium suggest these species may contribute to local immune activation, chronic inflammation, and potentially lesion maintenance in endometriosis. The identification of Sphingobium and Pseudomonas viridiflava as dominant species in upper reproductive tract samples provides a promising direction for non-invasive biomarkers for diagnosing endometriosis. Furthermore, this microbial dysbiosis may offer therapeutic targets for microbiome-modulating treatments aimed at restoring a healthy reproductive tract microbiota, potentially alleviating symptoms and reducing disease progression. This research underscores the importance of targeting microbial ecosystems in understanding and managing endometriosis.

Molecular detection of intrauterine microbial colonization in women with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study reveals that intrauterine microbial colonization is prevalent in women with endometriosis, particularly with Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae. Findings suggest that GnRHa treatment exacerbates microbial colonization, indicating a possible role for targeted antimicrobial therapies in managing endometriosis-associated inflammation.

What Was Studied?

This study investigated the molecular detection of intrauterine microbial colonization in women with and without endometriosis, utilizing a 16S rDNA metagenome assay to evaluate bacterial presence in endometrial swabs and cystic fluid samples. The primary aim was to explore the role of microbial colonization in the intrauterine environment and its potential contribution to endometriosis pathogenesis, especially in women treated with gonadotropin-releasing hormone agonist (GnRHa). The study compared microbial communities in both endometrial tissue and cystic fluid derived from ovarian endometrioma and non-endometrioma cysts. The analysis aimed to validate the "bacterial contamination hypothesis," which posits that microbial colonization might exacerbate inflammatory responses, contributing to endometriosis progression.

Who Was Studied?

The study included 32 women with endometriosis and 32 women without endometriosis. Among these, half of each group (16 women) received GnRHa treatment for 4–6 months prior to sample collection. Endometrial swabs and cystic fluid samples were collected during laparoscopic procedures at Nagasaki University. Women were included if they were of reproductive age, with regular menstrual cycles, and had not taken antibiotics or immunosuppressants within three months prior to the study. The endometriosis cases were confirmed histologically, and cystic fluids were categorized as either ovarian endometrioma (OE) or non-endometrioma (NE) cysts. The study employed 16S rDNA metagenome sequencing using the Illumina MiSeq system to identify bacterial taxa.

What Were the Most Important Findings?

The study identified a significant alteration in the microbial landscape within the intrauterine environment and ovarian cystic fluid of women with endometriosis, particularly those undergoing GnRHa treatment. Notably, there was a significant decrease in Lactobacillaceae populations (p < 0.01) and a marked increase in Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae (p < 0.05 for each) in GnRHa-treated women with endometriosis. This microbial shift was associated with sub-clinical infection in the uterine cavity and cystic fluid of ovarian endometrioma. Furthermore, the 16S metagenome assay detected higher proportions of Streptococcaceae and Staphylococcaceae in ovarian endometrioma cyst fluid compared to non-endometrioma cysts, suggesting a unique microbial signature linked to inflammatory pathogenesis. Interestingly, traditional bacterial culture methods failed to detect colonies in the cystic fluids, while PCR analysis revealed substantial colonization. This discrepancy indicates that sub-clinical infections in the uterine and ovarian microenvironments may contribute to the chronic inflammatory state characteristic of endometriosis. The study also proposed that GnRHa-induced hypoestrogenism might reduce the expression of antimicrobial peptides in the genitourinary tract, facilitating microbial colonization and chronic inflammation.

ParameterFindings in Endometriosis Patients
Intrauterine Microbial ColonizationSignificant increase in Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae populations in endometrial tissue and ovarian cystic fluid.
GnRHa Treatment EffectsDecrease in Lactobacillaceae populations (p < 0.01) with elevated Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae (p < 0.05).
Cystic Fluid MicrobiomeHigher proportions of Streptococcaceae and Staphylococcaceae in ovarian endometrioma cyst fluid compared to non-endometrioma cysts.
Detection MethodsTraditional bacterial culture failed to detect colonies, but 16S rDNA sequencing revealed substantial colonization.
Hypothesized MechanismGnRHa-induced hypoestrogenism may suppress antimicrobial peptides, facilitating microbial colonization and chronic inflammation.
Pathogenic ImplicationsSuggests sub-clinical infection in uterine and ovarian environments as a contributor to chronic inflammation in endometriosis.
Therapeutic ConsiderationPotential for targeted antimicrobial therapy to mitigate microbial load and reduce inflammation in endometriosis.

What Are the Greatest Implications of This Study?

The findings provide robust evidence that intrauterine microbial colonization—particularly of Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae—is prevalent in women with endometriosis and is significantly heightened with GnRHa treatment. This suggests that silent intrauterine infections could exacerbate inflammatory responses and potentially influence disease progression. Furthermore, the detection of bacterial DNA in ovarian endometrioma cystic fluid indicates that microbial colonization extends beyond the uterine environment, potentially affecting ovarian tissue integrity. These insights propose that targeted antimicrobial therapy might mitigate intrauterine bacterial load, reduce inflammation, and improve disease management. The study challenges the traditional view of the sterile uterine environment, suggesting that the bacterial contamination hypothesis should be revisited as a contributing factor in endometriosis pathogenesis.

Molecular detection of microbial colonization in cervical mucus of women with and without endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This cross-sectional observational study investigates cervical mucus in women with and without endometriosis, finding similar microbial distributions overall. However, bacteria such as Corynebacterium, Enterobacteriaceae, Flavobacterium, Pseudomonas, and Streptococcus were more common in women with endometriosis, with Enterobacteriaceae and Streptococcus showing significant associations

What was studied?

The study investigated the microbiota in the cervical mucus of women with and without endometriosis using next-generation sequencing (NGS) technologies. It aimed to clarify whether cervical mucus in women with endometriosis is contaminated with bacteria, which could potentially transmigrate into the intrauterine cavity, influencing the pathogenesis of endometriosis. The research explored the bacterial population’s diversity, its correlation with the disease, and how these findings align with previous studies on intrauterine microbial colonization and its role in endometriosis through LPS/TLR4 engagement of the innate immune system.

 

Who was studied?

The study’s participants consisted of 30 women diagnosed with endometriosis (confirmed by laparoscopy and classified according to the revised American Society for Reproductive Medicine scoring system for stages III-IV) and 39 women without the condition (control group), all of reproductive age (20-44 years). The control group included women with fibroids or benign ovarian tumors other than endometriosis. All subjects had a normal-appearing cervix, were negative for vaginal culturing tests, and had not received endocrine therapy or antibiotics for at least six months before sample collection.

 

What were the most important findings?

The study highlights several findings about the cervical mucus microbiota in women with and without endometriosis. It found a diverse array of microbiota in both groups, with variations not tied to menstrual cycle phases. The endometriosis group exhibited significantly higher alpha diversity, indicating a more diverse bacterial community. Specific bacteria such as Enterobacteriaceae, Corynebacterium, Pseudomonas, Flavobacterium, and Streptococcus were more prevalent in the endometriosis group, with Enterobacteriaceae and Streptococcus showing significantly higher prevalence.

What are the greatest implications of this study?

The findings suggest that the cervical mucus of women with endometriosis harbors a distinct and more diverse bacterial population than women without the condition. The significant presence of specific bacteria, particularly Enterobacteriaceae and Streptococcus, in women with endometriosis may play a role in the disease’s pathogenesis through mechanisms involving bacterial contamination, immune system engagement, and inflammatory responses. These insights open new avenues for understanding endometriosis’s pathophysiology, potentially leading to novel diagnostic and therapeutic strategies. Further, the study implies a possible link between cervical mucus microbiota and adverse pregnancy outcomes, highlighting the need for additional research to explore the mechanistic connections between microbial colonization and endometriosis and its implications for fertility and pregnancy.

Molecular detection of microbial colonization in cervical mucus of women with and without endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies intrauterine microbial colonization in endometriosis, with enriched Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae. GnRHa therapy exacerbated microbial imbalances, highlighting potential therapeutic targets for managing inflammation and infection in endometriosis. Findings suggest microbial modulation may enhance treatment outcomes.

What Was Studied?

This study investigated the molecular detection of microbial colonization in cervical mucus of women with and without endometriosis using next-generation sequencing (NGS) technology. The primary aim was to explore whether specific microbial populations in the cervical mucus are associated with endometriosis and could contribute to its pathogenesis. Researchers collected cervical mucus samples from 30 women with endometriosis and 39 women without the condition. The microbial communities were analyzed through 16S rRNA gene sequencing, alongside Gram staining and real-time PCR to validate the presence of specific bacterial species. This study sought to identify microbial signatures that may play a role in the inflammation and immune responses linked to endometriosis progression.

Who Was Studied?

The study included 69 women of reproductive age, with 30 diagnosed with endometriosis (all classified as r-ASRM stages III-IV) and 39 serving as healthy controls. All participants underwent laparoscopic surgery for diagnosis or benign gynecological conditions. Cervical mucus samples were collected before surgery under sterile conditions to prevent contamination, ensuring accurate representation of the microbiota present. Women with gynecological malignancies, pelvic inflammatory disease, bacterial vaginosis, or recent antibiotic use were excluded to prevent confounding microbial shifts.

What Were the Most Important Findings?

The analysis revealed that cervical mucus in women with endometriosis harbored distinct microbial communities compared to healthy controls. While Lactobacillus spp. remained dominant across all samples, women with endometriosis showed increased populations of Enterobacteriaceae, Streptococcus, Corynebacterium, Pseudomonas, and Flavobacterium. Notably, Enterobacteriaceae and Streptococcus were significantly enriched in endometriosis patients, as confirmed by real-time PCR analysis (p < 0.05). This elevated presence suggests these bacteria may contribute to inflammatory cascades in the cervix and potentially facilitate disease persistence. Additionally, alpha diversity was significantly higher in the cervical mucus of endometriosis patients, indicating a broader microbial distribution. The study also noted that despite high Lactobacillus prevalence (40–60%), the co-existence of pathogenic bacteria like Enterobacteriaceae and Streptococcus was unique to the endometriosis group, hinting at microbial imbalance. These findings support the hypothesis that cervical microbial colonization could be linked to the development and maintenance of endometriosis through immune modulation and inflammatory responses. The study further postulated that the ascent of these pathogens from the cervix into the uterine cavity may exacerbate inflammatory conditions, potentially triggering endometriosis progression via LPS/TLR4 signaling and innate immune activation.

ParameterFindings in Endometriosis Patients
Dominant GeneraLactobacillus spp. remained dominant in both endometriosis patients and controls, with 40–60% prevalence.
Increased GeneraMarked increases in Enterobacteriaceae, Streptococcus, Corynebacterium, Pseudomonas, and Flavobacterium.
Significant EnrichmentEnterobacteriaceae and Streptococcus were significantly enriched (p < 0.05) in the endometriosis group.
Alpha DiversityHigher alpha diversity observed in endometriosis patients, indicating broader microbial distribution in cervical mucus.
Pathogenic Co-ExistenceDespite high Lactobacillus prevalence, Enterobacteriaceae and Streptococcus co-existed exclusively in the endometriosis group.
Inflammatory AssociationsThese pathogens are suspected to drive inflammatory cascades, contributing to lesion persistence and immune dysregulation.
Migration HypothesisPotential pathogen ascent from the cervix to the uterine cavity may exacerbate inflammation and promote endometriosis via LPS/TLR4 signaling and innate immune activation.

What Are the Greatest Implications of This Study?

The study provides compelling evidence that specific microbial communities in cervical mucus—particularly Enterobacteriaceae and Streptococcus—are associated with endometriosis. This microbial imbalance suggests that the cervical microbiome may serve as both a diagnostic biomarker and a therapeutic target for endometriosis management. The findings highlight the possibility of cervical microbial migration into the uterine cavity as a driver of inflammation and lesion growth, underscoring the need for further exploration into microbiota-targeted therapies. By identifying microbial dysbiosis at the cervical level, this study opens the door to non-invasive diagnostic methods and preventive strategies aimed at reducing microbial-induced inflammation in endometriosis patients.

Müllerian duct anomalies coincident with endometriosis: a review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study reviewed the relationship between Müllerian duct anomalies (MDAs) and endometriosis, focusing on their prevalence, pathophysiology, and classification. It highlighted the connection especially in cases with outflow obstruction and underscored the need for early diagnosis and targeted interventions to improve reproductive outcomes and manage symptoms.

What was studied?

The study reviewed the relationship between Müllerian duct anomalies (MDAs) and endometriosis, focusing on their coexistence and the underlying pathophysiological theories that might explain their association. It extensively covered the embryology, genetics, and pathophysiology of MDAs, alongside the American Society for Reproductive Medicine (ASRM) classification of these anomalies. The review also detailed different types of MDAs, their diagnosis, and their association with endometriosis, considering various factors such as uterine outflow obstruction and genetic predispositions.

 

Who was studied?

The study discussed women who present with MDAs and endometriosis, examining available data on the prevalence and nature of these conditions in this demographic. It synthesized information from various studies that stratified the relationship between MDAs and endometriosis according to specific classes of anomalies, particularly focusing on obstructed and non-obstructed MDAs.

 

What were the most important findings?

Association Between MDAs and Endometriosis: There is an established connection, particularly when the MDA involves outflow obstruction, supporting the theory of retrograde menstruation as a contributing factor to the pathogenesis of endometriosis.

Variability in MDA Prevalence and Impact: The prevalence of MDAs varies widely due to differences in diagnostic techniques and patient populations, with a higher prevalence noted among infertile women and those with recurrent miscarriages.

Complexity of Pathogenesis Theories: The study underscores the complexity of endometriosis pathogenesis, including theories like retrograde menstruation, coelomic metaplasia, and the presence of müllerian remnants, suggesting that different types of endometriosis might arise from different mechanisms.

Genetic Factors: Both conditions are influenced by genetic factors, but no single gene mutation has been directly implicated in causing MDAs or endometriosis, suggesting a multifactorial etiology.

 

What are the greatest implications of this study?

Improved Diagnostic and Interventional Strategies: Recognizing the association between MDAs and endometriosis, especially in the presence of obstructive anomalies, could lead to earlier diagnosis and more targeted interventions, potentially improving reproductive outcomes and managing pain symptoms effectively.

Need for Further Research: The study highlights the need for more detailed and controlled studies to define the relationship between specific types of MDAs and the various forms of endometriosis. This could help develop more personalized treatment plans and understand the underlying mechanisms at a deeper level.

Clinical Practice Changes: The findings encourage the adoption of standardized classification systems for MDAs and suggest that surgical interventions might be beneficial in cases of obstructed flow to manage or even reverse symptoms of endometriosis, although the outcomes may vary.

Awareness and Screening: There is an emphasis on the importance of awareness and careful screening for endometriosis in patients with MDAs, which could lead to better management strategies and reduce the long-term impact of these conditions on women’s health.

 

DOI: 10.1007/s00261-020-02465-y

 

New Insights into Photobiomodulation of the Vaginal Microbiome—A Critical Review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The study reviewed theoretical and emerging research on photobiomodulation (PBM) and its effects on the vaginal microbiome, exploring how PBM might beneficially alter microbial communities to improve vaginal health. It suggested that PBM could be a non-invasive tool to treat conditions like bacterial vaginosis and influence systemic diseases by managing microbiome health, presenting a new potential non-pharmacological intervention in gynecological health.

What was studied?

The study reviewed the potential effects of photobiomodulation (PBM) on the vaginal microbiome, particularly exploring theoretical and emerging research on how PBM may influence vaginal health through its interaction with microbial communities.

 

Who was studied?

The study itself is a review and does not directly study a group of individuals. Instead, it synthesizes research on the vaginal microbiome and how it might be affected by PBM treatments, so the “who” primarily encompasses populations from referenced studies within the field of vaginal health and microbiome research.

 

What were the most important findings?

The review suggests that PBM might influence the vaginal microbiome beneficially by affecting microbial communities, potentially helping in treating conditions like bacterial vaginosis or influencing systemic diseases. It discusses the interaction of light treatment with bacteria and its theoretical applications in managing vaginal microbiome health.

 

What are the greatest implications of this study?

The review highlights a promising but underexplored area in the intersection of PBM and vaginal microbiome management. If PBM can effectively influence the vaginal microbiome, it could offer a non-invasive method to manage and treat a range of vaginal health issues and associated systemic conditions, expanding the scope of non-pharmacological interventions in gynecological health. This could lead to new therapeutic modalities that are more focused on microbiome health, influencing everything from disease prevention to treating existing conditions.

Nickel Sensitivity and Symptom Management in Endometriosis: The Role of a Low-Nickel Diet

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Low‑Nickel Diet (LNiD)
    Low‑Nickel Diet (LNiD)

    A low-nickel diet (LNiD) is a therapeutic dietary intervention that eliminates high-nickel foods, primarily plant-based sources such as legumes, nuts, whole grains, and cocoa, to reduce systemic nickel exposure. It is clinically validated for managing systemic nickel allergy syndrome (SNAS) and nickel-induced eczema. Its relevance is well-established in microbiome modulation, with studies demonstrating clinical benefits in conditions such as endometriosis, fibromyalgia, irritable bowel syndrome, and GERD.

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

This study offers new insights into the potential link between nickel sensitivity and symptom severity in endometriosis, suggesting that a low-nickel diet may be a promising intervention for alleviating associated gastrointestinal and gynecological symptoms.

What Was Studied?

This pilot study investigated the prevalence of nickel (Ni) allergic contact mucositis (ACM) in women with endometriosis who experience gastrointestinal symptoms and evaluated the effects of a low-nickel diet on these symptoms. The study focused on assessing the gastrointestinal, extra-intestinal, and gynecological symptom reductions associated with Ni ACM and dietary interventions.

Who Was Studied?

The study enrolled 84 women of reproductive age diagnosed with endometriosis who reported significant gastrointestinal symptoms. Thirty-one participants completed the study, undergoing a diagnostic nickel oral mucosa patch test (omPT) and a subsequent three-month low-nickel diet intervention. Participants were evaluated using symptom questionnaires both at baseline and after dietary changes.

What Were the Most Important Findings?

The study found that 90.3% of participants tested positive for Ni ACM, suggesting a high prevalence of nickel sensitivity among women with endometriosis. Following three months of adhering to a low-nickel diet, significant reductions in all evaluated symptoms were reported. Gastrointestinal symptoms such as abdominal pain, bloating, and diarrhea showed marked improvement. Extra-intestinal symptoms, including fatigue and headaches, and gynecological symptoms such as pelvic pain and dysmenorrhea, also exhibited statistically significant decreases. These findings indicate that nickel sensitivity may contribute to the symptomatic burden of endometriosis, and dietary interventions targeting nickel can alleviate these issues.

The study suggests a potential mechanistic link between nickel exposure, immune responses, and the exacerbation of endometriosis symptoms. Major microbial associations (MMAs) relevant to this context include those influenced by dietary changes, although specific microbiome alterations were not detailed.

What Are the Greatest Implications of This Study?

This research highlights nickel sensitivity as a significant yet previously under-recognized contributor to gastrointestinal and systemic symptoms in endometriosis patients. The findings suggest that incorporating nickel sensitivity screening and low-nickel dietary recommendations could represent a transformative approach to symptom management in endometriosis. Although the sample size was small, the results offer strong preliminary evidence for revising dietary protocols in clinical practice to include low-nickel guidelines, potentially improving the quality of life for patients.

Oral, Vaginal, and Stool Microbial Signatures in Patients With Endometriosis as Potential Diagnostic Non-Invasive Biomarkers: A Prospective Cohort Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study explores oral, vaginal, and stool microbial signatures in patients with endometriosis, highlighting their potential as non-invasive diagnostic biomarkers. Findings suggest distinct microbial shifts in these sites, offering a promising avenue for early detection and targeted intervention in endometriosis management.

What was studied?

This prospective cohort pilot study examined the oral, vaginal, and stool microbiota of three cohorts: confirmed endometriosis patients (ENDO, n=21), patients with other gynecological conditions but no endometriosis (N-ENDO, n=24), and healthy controls without gynecologic disease (HC, n=19). Using 16S rRNA sequencing, the study sought to identify non-invasive microbial biomarkers that could potentially differentiate individuals with endometriosis from others, with the ultimate goal of developing a diagnostic tool.

Who was studied?

A total of 64 women were studied, all age- and sex-matched. ENDO and N-ENDO participants were recruited from a hospital setting where they underwent laparoscopy with histological confirmation. Healthy controls were recruited from a separate longitudinal study (MothersBabies), with no known gynecological pathology.

Key Findings:

The study revealed significant microbial diversity and compositional differences in oral and stool samples among patients with endometriosis, non-endometriosis gynecologic conditions, and healthy controls, while vaginal samples showed no significant variation. Specifically, alpha diversity was reduced in the stool microbiota of endometriosis patients compared to healthy controls, and beta diversity analysis confirmed that both oral and stool communities were distinctly structured across cohorts. LEfSe analysis identified differentially abundant taxa specific to body site and disease severity. In stool samples, Phascolarctobacterium and Lactobacillus were enriched in endometriosis, with Actinomyces elevated in minimal/mild cases and Paraprevotellaceae in moderate/severe cases.

Oral samples from patients with moderate/severe endometriosis were characterized by a marked increase in Fusobacterium, a genus previously shown to facilitate lesion development in murine models and implicated in human periodontal disease. This is especially relevant given the higher incidence of periodontitis in endometriosis patients. Cardiobacterium was elevated in mild disease. In vaginal samples, the enrichment of Escherichia, Enterococcus, and Tepidimonas supports the bacterial contamination hypothesis, which posits that lipopolysaccharide (LPS)-mediated inflammation may play a role in lesion formation.

Here is a summary of the differentially abundant taxa by body site and disease severity:

Body SiteDifferentially Abundant Taxa
StoolPhascolarctobacteriumLactobacillus ↑ in ENDO; Actinomyces ↑ in minimal/mild; Paraprevotellaceae ↑ in moderate/severe
OralFusobacterium ↑ in moderate/severe ENDO; Cardiobacterium ↑ in minimal/mild ENDO
VaginalEscherichiaEnterococcusTepidimonas ↑ in ENDO

Implications for Microbiome Research and Clinical Practice:

The study underscores the potential for developing a non-invasive diagnostic tool for endometriosis using microbial biomarkers obtained from oral or stool samples. Specific taxa such as Fusobacterium, Escherichia, and Phascolarctobacterium emerged as promising microbial targets for future mechanistic and therapeutic investigations due to their known roles in modulating inflammation and estrogen metabolism. Additionally, the observed enrichment of Lactobacillus in the stool of patients with endometriosis suggests a possible link to estrobolome activity, with implications for enhanced estrogen recycling and disease progression. Furthermore, the detection of overlapping genera in the gut and peritoneal fluid, as reported in other studies, lends support to the hypothesis that intestinal bacterial translocation may contribute to the peritoneal inflammation characteristic of endometriosis.

Persistent Organic Pollutants and Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study links persistent organic pollutants (POPs) to metabolic alterations in deep endometriosis, identifying trans-nonachlor and 2-hydroxybutyrate as key markers.

What Was Studied?

This study explored the relationship between persistent organic pollutants (POPs) and the risk of surgically confirmed deep endometriosis by integrating high-resolution metabolomic profiling. It aimed to characterize metabolic changes associated with POP exposure, focusing on polychlorinated biphenyls (PCBs), organochlorinated pesticides (OCPs), and per-/polyfluoroalkyl substances (PFAS). The researchers utilized advanced analytical techniques such as gas and liquid chromatography coupled with high-resolution mass spectrometry (HRMS) and nuclear magnetic resonance (NMR).

Who Was Studied?

A hospital-based case-control cohort in France was recruited, consisting of women with surgically confirmed deep endometriosis and matched controls without the condition. Serum samples were collected from these participants to measure POP levels and conduct comprehensive metabolomic profiling. The study controlled for confounding variables such as demographic and lifestyle factors, ensuring a robust statistical analysis.

What Were the Most Important Findings?

The study identified significant links between specific POPs and endometriosis risk. Trans-nonachlor, an organochlorinated pesticide, emerged as the most strongly associated pollutant, doubling the risk of deep endometriosis. Other key POPs included PCBs 180 and 167. Metabolomic profiling revealed distinctive metabolic disruptions in women with endometriosis. These included elevated serum levels of lactate, ketone bodies, multiple amino acids, reduced bile acids, phosphatidylcholines (PCs), cortisol, and hippuric acid. A noteworthy finding was the metabolite 2-hydroxybutyrate, which correlated with both trans-nonachlor exposure and endometriosis risk, acting as a potential biomarker of the disease and its environmental exposure.

What Are the Greatest Implications of This Study?

This study is groundbreaking in linking POP exposure to metabolic alterations in deep endometriosis, suggesting an environmental component to the disease's pathogenesis. The findings highlight the potential of metabolomic biomarkers, like 2-hydroxybutyrate, for early diagnosis and monitoring of environmental risk factors. These results emphasize the importance of further research to clarify causal relationships and develop interventions to reduce exposure to harmful pollutants. Clinically, integrating metabolomic and environmental data could improve risk assessment and individualized treatment approaches for endometriosis patients.

Plants as source of new therapies for endometriosis: a review of preclinical and clinical studies

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review highlights the potential of plant-derived therapies for endometriosis. Key findings include anti-inflammatory, anti-proliferative, and anti-angiogenic effects of herbal extracts and bioactive compounds, offering safer long-term alternatives to conventional treatments.

What Was Reviewed?

This review systematically examined plant-derived agents and their potential for treating endometriosis. The authors focused on three main categories: herbal extracts, specific plant-derived bioactive compounds, and Chinese herbal medicine (CHM). By analyzing preclinical and clinical studies, the review assessed the efficacy, mechanisms of action, and clinical potential of these agents, including compounds such as resveratrol, epigallocatechin-3-gallate, curcumin, and cannabinoids. The paper aimed to critically evaluate the relevance of natural therapies as safer, long-term alternatives to conventional treatments for endometriosis.

Who Was Reviewed?

The review covered studies involving various experimental models, including human cell lines, rodent models of endometriosis, and limited clinical trials on human subjects. These studies collectively investigated the effects of plant-derived agents on cellular and molecular markers of endometriosis, such as inflammation, angiogenesis, and apoptosis. The review also discussed findings from clinical trials of Chinese herbal medicine and individual bioactive compounds.

What Were the Most Important Findings?

The review identified several plant-derived agents with significant potential for endometriosis therapy. Herbal extracts such as pueraria flower extract (PFE) and aged black garlic exhibited anti-inflammatory, anti-angiogenic, and anti-proliferative effects in experimental models. Bioactive compounds like resveratrol and curcumin demonstrated pleiotropic effects, targeting processes like estrogen modulation, oxidative stress reduction, and inhibition of vascular endothelial growth factor (VEGF) expression. Chinese herbal medicine formulations were found to alleviate symptoms, reduce lesion size, and prevent recurrence in clinical contexts. Mechanistically, these agents influence key pathways involving cytokines (IL-6, IL-8, TNF-α), transcription factors (NF-κB), and matrix metalloproteinases (MMPs), making them promising candidates for integrative treatment strategies.

What Are the Greatest Implications of This Review?

The findings emphasize the need for standardized protocols and further clinical trials to validate the safety and efficacy of plant-derived therapies in human endometriosis patients. The review underscores the potential of these agents as part of multimodal treatment strategies, offering reduced side effects and improved long-term management compared to conventional hormonal or surgical approaches. Additionally, the pleiotropic action of these agents aligns with the complex pathophysiology of endometriosis, addressing inflammation, angiogenesis, and cellular survival concurrently.

Polycystic Ovary Syndrome: Etiology, Current Management, and Future Therapeutics

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review links PCOS to gut microbiota dysbiosis, showing how LPS leakage, SCFA deficiency, and altered bile acids contribute to insulin resistance and hyperandrogenism. Microbiome-based therapies, including probiotics, prebiotics, and IL-22, show strong promise.

What Was Reviewed?

This review presented a comprehensive analysis of the multifactorial causes, pathophysiology, and therapeutic approaches for polycystic ovary syndrome (PCOS). It placed particular emphasis on the role of gut microbiota dysbiosis and its systemic effects on insulin resistance, hyperandrogenism, and chronic inflammation. In addition to outlining traditional treatments, the paper critically evaluated emerging therapies such as probiotics, prebiotics, fecal microbiota transplantation (FMT), miRNA modulation, and IL-22 therapy. This review serves as a key resource for clinicians seeking a holistic understanding of PCOS, connecting microbiome research with endocrine and metabolic interventions.

Who Was Reviewed?

The review synthesized findings from a wide array of clinical studies, animal models, and experimental trials. It referenced data involving women of reproductive age diagnosed with various phenotypes of PCOS, including both obese and lean individuals. It incorporated rodent models, especially those induced by androgens or letrozole, to simulate PCOS pathology and examine microbiome manipulation outcomes. In its assessment of microbiota, the review drew from sequencing studies and intervention trials using specific probiotic strains such as Lactobacillus acidophilus, L. casei, Bifidobacterium bifidum, and prebiotics like inulin and resistant dextrin. These references grounded its recommendations in translational and mechanistic evidence.

What Were the Most Important Findings?

The review outlined several critical findings that directly connect gut microbiota dysbiosis with the clinical hallmarks of PCOS. A key mechanism involves increased gut permeability due to decreased populations of beneficial bacteria like Lactobacillus and Bifidobacterium, alongside an overgrowth of pro-inflammatory species such as Escherichia coli and Shigella. This dysbiosis allows lipopolysaccharides (LPS) to enter systemic circulation, stimulating immune responses that impair insulin receptor function and exacerbate insulin resistance. Hyperinsulinemia then stimulates androgen production by ovarian theca cells and reduces SHBG levels, increasing free testosterone and fueling PCOS symptoms.

The review also addressed microbial metabolites, particularly short-chain fatty acids (SCFAs) and bile acids. Women with PCOS showed reduced production of SCFAs like butyrate, which are vital for maintaining gut integrity and regulating inflammation. Moreover, altered bile acid profiles—especially reductions in glycodeoxycholic and tauroursodeoxycholic acid—were linked to disrupted hormonal balance and metabolic dysfunction. These major microbial associations (MMAs) illustrate how gut microbiota interact with ovarian steroidogenesis, glucose homeostasis, and the immune axis in PCOS.

Importantly, the review highlighted the therapeutic potential of microbiota restoration. Probiotic supplementation with specific strains led to improvements in insulin sensitivity, lipid profiles, and androgen levels. Prebiotics such as resistant dextrin demonstrated similar metabolic benefits. FMTs in animal models reversed hyperandrogenism and restored menstrual cycles, suggesting strong translational potential. Additionally, novel pathways involving IL-22 and miRNA regulation offer future targets for microbial and metabolic rebalancing in PCOS.

What Are the Implications of This Review?

This review has profound implications for the clinical management of PCOS. It reframes the condition as a microbiota-linked systemic disorder rather than solely an endocrine one. By mapping specific microbial patterns to the hallmarks of PCOS—including hyperandrogenism, insulin resistance, and anovulation—the authors offer a rationale for gut-targeted diagnostics and treatments. Clinicians may soon assess microbiome composition as part of a diagnostic workup, particularly in patients with metabolic dysfunction but unclear hormonal profiles.

Furthermore, the review validates a multi-pronged therapeutic strategy, integrating microbiota restoration with hormonal, metabolic, and reproductive targets. The demonstrated success of Lactobacillus and Bifidobacterium supplementation in improving PCOS biomarkers supports the clinical use of probiotics. Similarly, FMT, while currently limited to preclinical studies, presents a compelling intervention with the potential to reset dysregulated metabolic-hormonal loops. Lastly, novel therapies like IL-22 and miRNA modulation could personalize treatment, especially for patients with inflammatory or resistant phenotypes. Overall, this review builds a clear and actionable bridge between microbiome science and PCOS clinical care.

Polycystic ovary syndrome: pathophysiology and therapeutic opportunities

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review presents PCOS as a multisystem disorder driven by gut dysbiosis, neuroendocrine disruption, and adipose androgen synthesis. Targeting B. vulgatus, IL-22, and AKR1C3 may offer novel interventions.

What Was Reviewed?

This comprehensive review synthesized current evidence on the pathophysiology of polycystic ovary syndrome (PCOS) and emerging therapeutic opportunities. It explored how PCOS results from a combination of polygenic susceptibility, environmental influences, and developmental programming. The review paid particular attention to neuroendocrine disruption, androgen biosynthesis, insulin resistance, and the role of gut microbiota and adipose tissue function. It integrated findings from clinical and experimental models to explain the disease's reproductive and metabolic features and offered a detailed examination of therapeutic agents in development, including kisspeptin agonists, neurokinin 3 receptor antagonists, AKR1C3 inhibitors, GLP-1 receptor agonists, and microbiota-based interventions. These insights provided a modern framework for targeting PCOS not just as a reproductive disorder, but as a systemic, multifactorial syndrome with metabolic and microbial roots.

Who Was Reviewed?

The review analyzed studies involving a broad population of women with PCOS from various ethnic backgrounds and phenotypic subtypes. It included data from clinical cohorts, epidemiological studies, and preclinical models such as letrozole- and androgen-induced rodent models. Patients ranged in metabolic phenotype, encompassing lean and obese subtypes with or without insulin resistance. The review also drew on human genome-wide association studies (GWAS), metagenomic studies, and randomized controlled trials investigating microbiota and hormonal therapies. The diversity of sources allowed the authors to assess pathophysiological mechanisms relevant to both early onset and chronic PCOS progression.

What Were the Most Important Findings?

One of the most critical findings is the multifaceted etiology of PCOS, with gut microbiota emerging as a pivotal player. Women with PCOS exhibit increased levels of Bacteroides vulgatus and decreased concentrations of beneficial bile acids such as glycodeoxycholic acid and tauroursodeoxycholic acid. In mouse models, oral gavage with B. vulgatus or fecal samples from PCOS patients induced insulin resistance, ovarian morphological disruption, and hyperandrogenism. These effects were mediated partly through suppressed interleukin-22 (IL-22) production and altered bile acid metabolism. Supplementation with IL-22 or bile acids reversed some PCOS-like features, reinforcing the causal role of microbial dysbiosis. Other major microbial associations (MMAs) include increased LPS-producing gram-negative bacteria and reduced SCFA-producing species like Faecalibacterium prausnitzii, which impair gut barrier integrity and promote systemic inflammation.

The review also underscored that hyperinsulinemia not only drives androgen production but suppresses SHBG, creating a feedback loop of worsening endocrine dysfunction. Adipose tissue contributes actively to androgen synthesis, particularly via AKR1C3, which converts adrenal-derived precursors into potent 11-oxygenated androgens. These metabolites have comparable potency to testosterone and correlate strongly with metabolic risk in PCOS. Additionally, neuroendocrine irregularities, particularly increased kisspeptin and GnRH pulsatility, exacerbate LH-driven androgen excess. The convergence of these pathways—microbial, hormonal, metabolic—solidifies PCOS as a systemic condition requiring holistic intervention strategies.

What Are the Implications of This Review?

This review reframes PCOS as a neuro-metabolic disorder deeply intertwined with gut microbial composition and function. Clinicians should consider integrating microbiome assessment into PCOS diagnostics, especially in patients with lean phenotypes or atypical presentations. Therapeutically, the review paves the way for microbiota-targeted strategies such as IL-22 modulation, bile acid supplementation, and personalized probiotics. The consistent microbial shifts identified—especially involving Bacteroides vulgatus and SCFA-producing taxa—could serve as biomarkers or therapeutic targets. Moreover, understanding the role of AKR1C3 in adipose-driven androgen synthesis opens a novel therapeutic avenue for endocrine normalization. This work also highlights the value of targeting kisspeptin and neurokinin pathways, providing hormone-specific modulation without systemic suppression. In totality, the review advocates for multi-system treatment strategies that address not just fertility and cosmetic concerns, but long-term cardiometabolic health.

Possible Therapeutic Mechanisms and Future Perspectives of Vaginal Microbiota Transplantation

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The study reviewed Vaginal Microbiome Transplant (VMT) as a treatment for dysbiotic vaginal microbiomes, particularly for recurrent bacterial vaginosis. It discussed the potential of restoring a healthy microbiota dominated by Lactobacillus through VMT. Initial clinical trials showed promising results, with a significant recovery rate among participants. The findings suggest VMT could become a crucial non-pharmacological option for managing and potentially curing conditions linked to vaginal microbiome imbalances, enhancing gynecological and possibly broader systemic health.

What was studied?

The study explored the therapeutic mechanisms and future perspectives of Vaginal Microbiota Transplantation (VMT). It focused on transferring healthy vaginal microbiota from a donor to recipients with dysbiotic vaginal microbiomes to restore normal microbial composition and function.

 

Who was studied?

The study didn’t involve direct experimentation on individuals. Instead, it reviewed existing research and clinical trials regarding VMT, particularly looking at cases involving recipients with recurrent bacterial vaginosis treated via VMT.

 

What were the most important findings?

Key findings highlighted VMT’s potential effectiveness in treating recurrent bacterial vaginosis, with a significant portion of the patients achieving recovery. The study emphasized the role of healthy Lactobacillus-dominated microbiota in restoring vaginal health, though noting the small sample sizes and uncontrolled designs of current studies.

 

What are the greatest implications of this study?

The greatest implications include the potential of VMT as a non-invasive, microbiota-based therapy for managing and potentially curing bacterial vaginosis and similar conditions caused by dysbiosis of the vaginal microbiome. This could lead to advancements in gynecological health treatments, reducing reliance on traditional pharmacological interventions and possibly affecting systemic health conditions linked to vaginal microbiota.

Premenstrual disorders and PMDD - a review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Premenstrual dysphoric disorder (PMDD) significantly impairs women’s lives due to abnormal sensitivity to hormonal fluctuations. Accurate diagnosis relies on prospective symptom tracking. Evidence supports SSRIs and hormonal treatments as effective management strategies. Emerging therapies and multidisciplinary care models promise improved outcomes for this complex and under-recognized disorder.

What was reviewed?

This paper provides a comprehensive review of premenstrual disorders (PMDs), with a specific focus on premenstrual dysphoric disorder (PMDD). It synthesizes current knowledge regarding the definitions, classification, prevalence, diagnosis, etiology, and treatment of PMDs and PMDD. The review covers consensus guidelines from professional bodies such as the International Society for Premenstrual Disorders (ISPMD), diagnostic criteria from DSM-V and ICD-11, and evaluates various therapeutic approaches, including non-pharmacological interventions, pharmacotherapy (particularly SSRIs), hormonal treatments, novel agents targeting neuroactive steroids, and surgical options. It also highlights challenges in diagnosis, the significant impact on quality of life and suicidality risk, and research gaps.

Who was reviewed?

The review critically assesses a wide body of clinical, epidemiological, and mechanistic studies involving women experiencing PMDs and PMDD worldwide. It references population prevalence data, genetic and neurobiological studies, clinical trials evaluating treatments such as SSRIs and combined oral contraceptive pills (COCPs), and guidelines developed by multidisciplinary expert panels. The authors draw upon systematic reviews, randomized controlled trials, observational studies, and consensus statements to present a balanced perspective. The review specifically incorporates data related to symptom measurement tools like the Daily Record of Severity of Problems (DRSP) and discusses patient management strategies applicable in primary and specialist care settings.

What were the most important findings?

The review clarifies that PMDD represents a severe subset of PMDs, affecting approximately 5% of women, and carries significant physical, psychological, and social burdens, including a markedly increased risk of suicide attempts. It emphasizes that PMDD symptoms arise from abnormal sensitivity to normal menstrual hormonal fluctuations, especially allopregnanolone's paradoxical effect on GABA-A receptors, rather than altered hormone levels per se. Genetic factors, serotonergic dysregulation, inflammation, and stress history also contribute to pathophysiology. Accurate diagnosis depends on prospective symptom tracking over at least two menstrual cycles to confirm symptom cyclicity, severity, and functional impact.

Treatment must be multidisciplinary and individualized. SSRIs are the first-line pharmacological treatment, acting rapidly and effectively even when dosed intermittently in the luteal phase or symptom-onset, mitigating side effects associated with continuous dosing. Hormonal treatments, particularly COCPs containing drospirenone, show efficacy, though hormonal sensitivity varies and progestogen intolerance complicates therapy. Emerging therapies targeting allopregnanolone modulation and 5-alpha reductase inhibitors are under investigation but require further evidence. GnRH analogues and surgical oophorectomy remain options for severe refractory cases but carry significant risks, including bone density loss. Non-pharmacological approaches, including cognitive behavioral therapy (CBT), dietary modifications, and supplements (calcium, magnesium, vitamin B6), provide complementary benefits.

What are the greatest implications of this review?

This review bridges the knowledge gap between evolving scientific insights into PMDD's neuroendocrine mechanisms and practical clinical management strategies. It advocates for a precision medicine approach tailored to individual hormonal sensitivities and symptom profiles. By synthesizing current evidence, it empowers clinicians to improve diagnostic accuracy through prospective symptom monitoring, recognize the disorder’s profound impact on mental health, and adopt evidence-based treatments, minimizing side effects. The emphasis on rapid SSRI efficacy and flexible dosing regimens offers clinicians practical tools to enhance adherence and patient quality of life. Moreover, highlighting the multidisciplinary nature of optimal care and emerging pharmacotherapies signals future directions for research and therapeutic innovation.

Premenstrual Dysphoric Disorder and the Brain

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

PMDD involves altered brain activation in prefrontal regions tied to hormone sensitivity. Symptoms appear cyclically with hormonal fluctuations, distinguishing it from other mood disorders. Understanding this brain-hormone interaction aids targeted treatment and improves clinical recognition of PMDD.

What was reviewed?

This paper reviewed the neurological basis of premenstrual dysphoric disorder (PMDD), emphasizing its recognition as a distinct mood disorder linked to menstrual cycle hormonal fluctuations. It summarized advances in brain imaging and neurophysiological studies demonstrating altered brain function in PMDD patients, particularly in prefrontal cortex regions involved in executive function and emotion regulation. The review highlighted the significance of hormone sensitivity, especially to estradiol and progesterone, and how these hormonal changes affect cerebral blood flow and neural activation patterns in women with PMDD compared to controls.

Who was reviewed?

The review focused on women diagnosed with PMDD according to rigorous DSM criteria, including prospective symptom tracking. It integrated findings from neuroimaging studies (fMRI, PET), hormonal manipulation paradigms (gonadotropin-releasing hormone agonist followed by hormone add-back), and psychophysiological assessments conducted on small to moderate cohorts of women with PMDD and matched healthy controls. The studies collectively evaluated brain activation, cerebral blood flow, neurotransmitter activity, and behavioral correlates of hormone-driven mood symptoms.

What were the most important findings?

The review underscored that women with PMDD show abnormal activation in the dorsolateral prefrontal cortex and medial frontal gyrus during cognitive tasks, regardless of hormonal state, suggesting a trait vulnerability. Brain activation differences correlated with symptom severity, especially irritability, which is a hallmark PMDD symptom. The cerebellum also showed heightened activity in PMDD. The disorder’s symptom manifestation requires the fluctuating hormonal environment of the luteal phase, implicating hormone sensitivity as a key pathophysiological factor. Unlike other mood disorders, PMDD’s brain dysfunction is specifically linked to normal hormonal changes rather than baseline abnormalities, explaining the cyclical nature of symptoms.

What are the greatest implications of this review?

This review clarifies that PMDD arises from an interaction between inherent brain vulnerabilities and normal hormonal fluctuations, particularly estradiol and progesterone. It encourages clinicians to view PMDD as a neurobiologically distinct disorder with predictable symptom timing linked to menstrual phases. These insights justify targeted hormonal and neuropharmacological treatments and support ongoing research into brain-based biomarkers and personalized therapies. The findings also highlight the importance of early diagnosis and symptom monitoring to improve patient care and quality of life for affected women.

Premenstrual dysphoric disorder-an undervalued diagnosis? A cross-sectional study in Hungarian women

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Hungarian women with probable PMDD experience high rates of anxio-depressive symptoms and reduced well-being. Retrospective screening suggests higher-than-expected prevalence, highlighting the need for better diagnostic tools and tailored treatment to address this underrecognized disorder.

What was studied?

This cross-sectional study assessed the prevalence of probable premenstrual dysphoric disorder (PMDD) among Hungarian women and examined the relationship between probable PMDD, anxio-depressive symptom severity, and overall well-being. The researchers aimed to validate a retrospective DSM-5-based PMDD screening tool in this population and explore psychological symptom patterns related to PMDD, using standardized questionnaires for depression, anxiety, and well-being.

Who was studied?

The study included 112 women of reproductive age from Hungary with regular menstrual cycles who were not using hormonal contraceptives and had no significant neurological, psychiatric, endocrine, or gynecological disorders. The participants were divided into two groups based on PMDD screening results: a probable PMDD group (n=67) and a non-PMDD group (n=45). They completed validated questionnaires measuring probable PMDD symptoms, anxio-depressive severity, and subjective well-being.

What were the most important findings?

The study revealed a surprisingly high prevalence of probable PMDD at nearly 60%, exceeding international estimates, which the authors attribute partly to retrospective screening limitations and recruitment bias. Women with probable PMDD reported significantly greater depressive and anxiety symptoms and lower well-being than controls, regardless of menstrual cycle phase. Logistic regression confirmed that higher anxiety and depression scores predicted probable PMDD diagnosis. The findings corroborate prior evidence that PMDD involves substantial psychological distress that impacts life quality. Notably, anxiety symptom severity did not vary significantly across cycle phases, suggesting persistent affective symptoms. These results highlight the challenges of accurate PMDD diagnosis, especially given the burden of prospective symptom tracking, and underscore the need for tailored psychological assessment and treatment strategies.

What are the greatest implications of this study?

This study emphasizes that probable PMDD is a prevalent and underrecognized condition that severely affects women's mental health and well-being, even beyond the premenstrual phase. It highlights the utility and limitations of retrospective screening tools in estimating PMDD prevalence and calls for improved diagnostic protocols that balance accuracy with practicality. The findings advocate for personalized mental health support and further research to refine diagnostic tools and treatment approaches tailored to the needs of women with PMDD, especially in underrepresented populations.

Premenstrual Dysphoric Disorder: Epidemiology and Treatment

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This review delineates PMDD’s unique neurobiology, highlighting hormone sensitivity, GABAergic dysfunction, and stress interaction. SSRIs provide rapid symptom relief, with hormonal and behavioral therapies complementing care. It emphasizes precise diagnosis and individualized, multidisciplinary treatment to improve outcomes in this debilitating disorder.

What was reviewed?

This paper presents a thorough review of the epidemiology, pathophysiology, and treatment options for premenstrual dysphoric disorder (PMDD). It examines PMDD’s diagnostic criteria as established by DSM-5, highlighting the importance of mood symptoms and prospective symptom tracking for accurate diagnosis. The review synthesizes current understanding of PMDD’s biological underpinnings, including the role of neurosteroids like allopregnanolone, estrogen’s influence on serotonergic systems, brain-derived neurotrophic factor (BDNF) polymorphisms, and the impact of stress and inflammation. It further explores neuroimaging and psychophysiological findings that differentiate PMDD from other affective disorders. The review then evaluates therapeutic approaches, emphasizing SSRIs as the first-line treatment and discussing intermittent dosing strategies, hormonal therapies, cognitive-behavioral therapy, and alternative treatments.

Who was reviewed?

The authors critically analyzed studies involving women diagnosed with PMDD across community and clinical samples worldwide. The review includes epidemiological data, genetic and neurobiological research, and clinical trials assessing treatment efficacy. It references consensus guidelines from psychiatric and gynecological professional bodies, neuroimaging studies comparing PMDD patients to healthy controls, and meta-analyses evaluating pharmacologic and psychotherapeutic interventions. The paper also integrates findings from animal models of hormone sensitivity and neurosteroid modulation relevant to PMDD pathophysiology.

What were the most important findings?

Women with PMDD do not differ in peripheral hormone levels but show altered GABA_A receptor function and neurosteroid sensitivity, contributing to affective symptoms. Estrogen’s modulation of serotonin receptors and transporters further implicates serotonergic dysregulation in PMDD. Genetic factors such as polymorphisms in estrogen receptor and serotonin transporter genes, as well as BDNF variants, may increase susceptibility. Stress history correlates with PMDD diagnosis and may influence neurosteroid responses and HPA axis regulation. Neuroimaging reveals structural and functional brain differences in areas regulating emotion and cognition, including the amygdala and prefrontal cortex, with altered GABA and glutamate levels detected in PMDD patients.

Regarding treatment, SSRIs demonstrate moderate to large effect sizes in symptom reduction, with rapid onset of action allowing for intermittent or symptom-onset dosing regimens that minimize side effects and improve adherence. Hormonal treatments, particularly combined oral contraceptives containing drospirenone, show some efficacy but with high placebo responses and variable individual tolerance. Cognitive-behavioral therapy provides sustained symptom improvement and complements pharmacotherapy, though combined approaches do not necessarily enhance outcomes beyond monotherapy. Alternative therapies such as calcium supplementation and omega-3 fatty acids offer limited benefits and require further validation.

What are the greatest implications of this review?

This review consolidates the complex neurobiological, genetic, and psychosocial factors contributing to PMDD, underscoring its distinction from other mood disorders and the importance of precision in diagnosis and treatment. It reinforces SSRIs as the cornerstone of pharmacotherapy and advocates for flexible dosing strategies tailored to symptom patterns, enhancing patient quality of life and medication adherence. The emerging understanding of neurosteroid modulation opens promising avenues for novel therapeutics targeting GABAergic pathways. The findings call for multidisciplinary, individualized treatment plans incorporating pharmacological, psychological, and lifestyle interventions. The review highlights gaps in long-term safety data for hormonal therapies and the need for improved diagnostic tools and biomarkers. Overall, it equips clinicians with an evidence-based framework to optimize PMDD management and encourages ongoing research to address unmet clinical needs.

Premenstrual dysphoric disorder: General overview, treatment strategies, and focus on sertraline for symptom-onset dosing

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Symptom-onset dosing of sertraline effectively reduces PMDD symptoms, especially mood-related ones, with fewer side effects and limited drug exposure. This targeted approach offers a promising, patient-friendly alternative to continuous SSRI treatment for premenstrual dysphoric disorder.

What was studied?

This paper studied the efficacy and tolerability of symptom-onset dosing of sertraline, a selective serotonin reuptake inhibitor (SSRI), for the treatment of premenstrual dysphoric disorder (PMDD). The focus was on assessing whether administering sertraline starting at the onset of PMDD symptoms, rather than continuous or luteal-phase dosing, could effectively reduce symptom severity and improve clinical outcomes over six menstrual cycles. The study also explored the side effect profile and discontinuation symptoms associated with this targeted treatment approach.

Who was studied?

The study population comprised 252 women with prospectively confirmed PMDD, aged approximately 34 years on average, predominantly white (around 70%), and without significant medical or psychiatric comorbidities. These participants were randomized into two groups: 125 women received flexible doses of sertraline (50–100 mg/day) beginning at symptom onset and continuing until menstruation began, while 127 women received placebo treatment following the same schedule.

What were the most important findings?

The study demonstrated that symptom-onset treatment with sertraline significantly reduced the severity of PMDD symptoms compared to placebo. Specifically, women treated with sertraline showed statistically significant improvements in depressive symptoms as measured by clinician-rated scales and a significant reduction in the daily record of problem severity, including the anger/irritability subscale. While the reduction in premenstrual tension ratings narrowly missed statistical significance, sertraline outperformed placebo in global improvement ratings and had higher clinical response rates (67% vs. 52%). Noticeably, emission rates were not significantly different. The average duration of sertraline use was only about seven days per menstrual cycle, minimizing exposure to the drug and related side effects. Adverse effects, primarily nausea and insomnia, were more common in the sertraline group, but abrupt discontinuation did not lead to withdrawal symptoms. These findings indicate that targeted, short-term SSRI treatment timed to symptom onset is effective and well tolerated in managing PMDD.

What are the greatest implications of this study?

This research challenges traditional views that antidepressants require continuous administration to be effective in PMDD treatment by demonstrating that symptom-onset dosing with sertraline is both efficacious and has a favorable side effect profile. This approach minimizes medication exposure and associated adverse effects, potentially improving adherence and reducing treatment costs. It provides a practical strategy to manage PMDD symptoms precisely when needed, aligning with the disorder's cyclical nature. Future research is needed to compare symptom-onset dosing directly with luteal-phase and continuous dosing regimens and to explore treatment strategies for non-responders to symptom-onset sertraline. Clinically, this study supports personalized, flexible pharmacotherapy for PMDD, enhancing therapeutic outcomes while mitigating risks.

Premenstrual Syndrome and Exercise: A Narrative Review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This narrative review explores how exercise can help manage the symptoms of Premenstrual Syndrome (PMS), focusing on the physiological and psychological benefits of regular physical activity.

What was reviewed?

This narrative review examines the connection between Premenstrual Syndrome (PMS) and exercise. It explores the potential role of exercise in mitigating PMS symptoms and evaluates existing studies to understand how physical activity might alleviate both the physical and psychological impacts of PMS. The review also critiques the methodology of current research and emphasizes the need for further studies to refine exercise interventions for PMS management.

Who was reviewed?

The review targets women who experience PMS, particularly those suffering from physical, psychological, and behavioral symptoms that impact their quality of life. The study highlights the importance of understanding how exercise may serve as a complementary treatment for PMS. The aim is to improve the health outcomes and daily functioning of affected women.

What were the most important findings?

The review identifies key findings regarding the positive effects of exercise on PMS symptoms. PMS symptoms, which include fatigue, mood swings, bloating, and irritability, significantly impair women’s daily activities. The review suggests that exercise, especially aerobic and resistance exercises, can reduce these symptoms, improving both physical and psychological well-being. Regular physical activity helps decrease fatigue, relieve pain (including breast tenderness), and improve mood by regulating hormonal fluctuations. Studies reviewed show that exercise enhances estrogen and progesterone levels and promotes endorphin release, which further alleviates pain and stress.

Exercise improves overall well-being by stimulating the release of neurochemicals like endorphins, which play a crucial role in mood enhancement and pain reduction. Furthermore, exercise reduces the impact of PMS on daily life, including work performance and social engagement. However, the review highlights the inconsistency across studies regarding the optimal exercise prescription for PMS. It calls for more detailed research to determine the best exercise duration, intensity, and frequency needed to achieve maximum benefit.

What are the greatest implications of this review?

This review suggests that exercise is a cost-effective, accessible, and powerful non-pharmacological approach to managing PMS. It encourages clinicians to incorporate exercise recommendations into treatment plans, considering the physical and psychological benefits it offers. Regular physical activity can serve as a complementary treatment alongside pharmacological options, especially for women seeking a holistic management approach. The review also stresses the importance of personalized exercise prescriptions tailored to individual needs and symptom profiles. As research on exercise and PMS continues to evolve, clearer guidelines will emerge, allowing healthcare providers to better support women with PMS through structured exercise programs.

Premenstrual Syndrome and Premenstrual Dysphoric Disorder as Centrally Based Disorders

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

PMS and PMDD stem from neuroendocrine and neurochemical imbalances, especially altered allopregnanolone and GABA activity. Hormonal and neuroactive therapies improve symptoms, while microbiome and neuroinflammation represent promising research areas.

What was studied?

This review examined the neuroendocrine and neurobiological mechanisms underlying Premenstrual Syndrome (PMS) and Premenstrual Dysphoric Disorder (PMDD), emphasizing their classification as centrally based disorders influenced by hormonal fluctuations. It also discussed current and novel therapeutic strategies targeting neuroactive steroids and neuroinflammation in PMS/PMDD.

Who was studied?

The review synthesized findings from clinical, neuroimaging, pharmacological, and molecular studies involving women diagnosed with PMS and PMDD across various reproductive stages, incorporating prospective symptom tracking and biochemical assessments to explore hormone-neurotransmitter interactions and brain sensitivity.

What were the most important findings?

The review highlighted that PMS and PMDD are neuro-hormonal disorders marked by increased central nervous system sensitivity to normal cyclical fluctuations of estrogens and progesterone, especially its metabolite allopregnanolone. This neurosteroid modulates GABA_A receptor activity, affecting mood regulation, and its altered function correlates with emotional and behavioral symptoms in PMDD. Impairments in opioid and serotonergic systems also contribute. Neuroinflammation via GABAergic pathways and elevated pro-inflammatory markers may play a role. Treatment focuses on stabilizing hormones, mainly with combined hormonal contraception, and modulating neuroactive steroids. SSRIs reduce symptoms by affecting serotonin pathways. Novel therapies targeting neurosteroid pathways, including progesterone receptor modulators, 5α-reductase inhibitors, and GABA_A receptor antagonists, show promise. However, treatment responses vary depending on hormonal regimens and individual profiles. Emerging evidence also suggests the gut-brain axis and microbiome influence symptom severity through neuroimmune interactions, though further study is needed.

What are the greatest implications of this study?

This review consolidates the understanding of PMS/PMDD as disorders rooted in neuroendocrine and neurochemical dysregulation, shifting the clinical perspective from purely gynecological or psychiatric frameworks to integrated neurobiological models. It underscores the necessity for personalized therapeutic approaches that combine hormonal regulation with neuroactive agents. The identification of neuroinflammation and microbiome influences opens novel research pathways and potential non-hormonal interventions. Clinicians should consider both established and emerging treatments to optimize symptom control, and researchers must prioritize elucidating the gut-brain interactions and refining neurosteroid-targeted therapies for improved patient outcomes.

Premenstrual syndrome, a common but underrated entity: review of the clinical literature

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This review explores premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD), focusing on symptoms, prevalence, risk factors, and treatment options.

What was reviewed?

This paper is a review of the clinical literature concerning premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD). It focuses on their symptoms, prevalence, risk factors, etiology, and current diagnostic criteria. The review examines treatment options ranging from lifestyle changes to pharmacological interventions, and it highlights the impact of PMS and PMDD on women's health.

Who was reviewed?

The review focuses on the clinical characteristics and findings related to PMS and PMDD in women of reproductive age. It draws from a wide range of studies to summarize the current understanding of these conditions, examining both epidemiological data and clinical treatments.

What were the most important findings?

The review identifies key characteristics of PMS and PMDD, noting that these disorders manifest during the luteal phase of the menstrual cycle and subside with menstruation. It was found that PMS affects a significant portion of the female population, with the prevalence ranging from 10% to 98%, while PMDD affects 2-8% of women. Symptoms can be physical, such as bloating and breast tenderness, or psychological, including mood swings, irritability, and anxiety. The pathogenesis of PMS and PMDD is linked to hormonal fluctuations, particularly estrogen and progesterone, and the interaction of these hormones with central neurotransmitter systems, notably serotonin, GABA, and beta-endorphins.

The review highlights serotonin’s role in the pathogenesis, with women experiencing PMS showing lower serotonin levels in various bodily fluids. Although the exact cause remains unclear, studies suggest that serotonin may be the key mediator of the mood symptoms seen in these disorders. The review also addresses the controversial role of vitamins and minerals in the treatment of PMS, with limited evidence supporting their efficacy over a placebo. Furthermore, it outlines various therapeutic approaches, such as selective serotonin reuptake inhibitors (SSRIs), combined oral contraceptives (COCs), and lifestyle changes, which have been shown to improve symptoms in many patients.

What are the greatest implications of this review?

This review underscores the importance of recognizing PMS and PMDD as significant health issues that can affect a woman’s quality of life. It suggests that, despite their prevalence, these disorders are often underdiagnosed. The review calls for better recognition and diagnosis, particularly using prospective symptom tracking over multiple cycles, as is recommended by the DSM-5 for diagnosing PMDD. Clinicians should consider both pharmacological and non-pharmacological treatments based on the severity of symptoms, as well as individualized care strategies, including SSRIs and COCs for more severe cases. Furthermore, the review suggests that future research should focus on understanding the complex hormonal and neurotransmitter interactions that underpin PMS and PMDD, potentially offering new avenues for treatment development.

Premenstrual syndrome: New insights into etiology and review of treatment methods

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This review examines the latest insights into the etiology and treatment of Premenstrual Syndrome (PMS) and Premenstrual Dysphoric Disorder (PMDD), focusing on hormonal and neurosteroid imbalances.

What was reviewed?

This paper is a review of the etiology and treatment options for Premenstrual Syndrome (PMS) and Premenstrual Dysphoric Disorder (PMDD). It examines the underlying causes of these conditions, including hormonal fluctuations, neurotransmitter imbalances, and the role of neurosteroids like allopregnanolone. The review also analyzes a variety of treatment options, including pharmacological methods (SSRIs, hormonal therapies, neurosteroid treatments) and non-pharmacological interventions, to provide a comprehensive overview of how PMS and PMDD can be managed.

Who was reviewed?

The review focuses on women of reproductive age who experience PMS and PMDD, with special attention to those who suffer from severe symptoms that significantly impair their daily functioning. The article explores clinical data and findings from various studies to provide a thorough understanding of the disorder's impact on women's health, as well as the varying responses to treatment.

What were the most important findings?

The review highlights several key findings about the etiology and treatment of PMS and PMDD. One of the most significant insights is the role of hormonal fluctuations, particularly the progesterone metabolite allopregnanolone, in the onset of PMS symptoms. It is noted that this metabolite modulates the GABA-A receptor in the central nervous system (CNS), which may explain some of the mood and anxiety-related symptoms of PMS. The review also underscores the complexity of the disorder, as it involves multiple physiological systems, including the hypothalamic-pituitary-adrenal (HPA) axis and neurotransmitter pathways such as serotonin.

SSRIs, commonly used to manage mood symptoms, provide rapid relief and are considered the first-line pharmacological treatment for severe cases. Hormonal therapies, particularly those that stabilize estrogen and progesterone levels, are also effective but require careful selection to avoid exacerbating symptoms. The review suggests that therapies targeting neurosteroids like allopregnanolone may offer new avenues for treatment, although more research is needed. In terms of treatment strategies, the review emphasizes the importance of personalized care, where treatments are tailored to individual symptoms and underlying mechanisms. For instance, oral contraceptives containing drospirenone and ethinylestradiol are effective for controlling physical symptoms, while SSRIs are more beneficial for psychological symptoms.

What are the greatest implications of this review?

The greatest implication of this review is the need for a more nuanced approach to diagnosing and treating PMS and PMDD. By understanding the complex hormonal and neurochemical interactions involved, clinicians can better tailor treatments to individual patients. The review suggests that effective treatment goes beyond symptom alleviation to address the root causes of the disorders. Additionally, the paper points to the importance of considering non-pharmacological interventions alongside medications, particularly for patients who experience mild to moderate symptoms. Future research into the role of neurosteroids and their modulation in the CNS could lead to more targeted treatments with fewer side effects.

Presence of metalloestrogens in ectopic endometrial tissue

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study examined the presence of metalloestrogens in ectopic endometrial tissue from fifty women diagnosed with endometriosis. Cadmium, nickel, and lead were found in all tissue samples, with nickel and lead showing particularly high concentrations. These findings suggest that metalloestrogens play a role in the etiology of endometriosis by interacting with estrogen receptors, emphasizing environmental pollutants' role in endometriosis progression.

What was studied?


This study investigated the presence of metalloestrogens in ectopic endometrial tissue from women with endometriosis. Metalloestrogens, heavy metals that can mimic estrogen and may contribute to estrogen-dependent diseases, were the focus, particularly regarding their potential role in the persistence and pathology of endometriosis. The researchers specifically analyzed levels of cadmium, nickel, and lead in ectopic endometrial samples using advanced metal detection techniques, Total Reflection X-ray Fluorescence (TXRF) and Graphite Furnace Atomic Absorption Spectroscopy (GFASS).

Who was studied?


The study included fifty women of reproductive age diagnosed with endometriosis via laparoscopy or laparotomy at the Professorial Gynecology Unit of the National Hospital, Colombo, Sri Lanka, during 2009-2010. The participants underwent these procedures for diagnosis or treatment, and endometriotic tissue samples were collected during surgery. The participants presented with varied symptoms like infertility, dysmenorrhea, chronic pelvic pain, and endometriomas.

What were the most important findings?


The study found significant levels of cadmium, nickel, and lead in all ectopic endometrial tissue samples. Specifically, geometric mean concentrations were reported as follows: cadmium (2.861 μg/Kg), nickel (17.547 μg/Kg), and lead (25.785 μg/Kg). The concentrations varied by tissue site, with the ovarian endometrioma wall showing higher, though not statistically significant, metal levels than pelvic endometrial patches or nodules in the pouch of Douglas.

Implications


This study is one of the first to identify and quantify metalloestrogens in ectopic endometrial tissue, shedding light on a possible environmental and molecular link to endometriosis. It underscores the mechanism by which these metals could perpetuate endometriosis, given their ability to interact with estrogen receptors in ectopic tissue. The implications are substantial for public health, especially given the widespread environmental exposure to metals such as cadmium, nickel, and lead. These findings suggest that environmental pollution may play a significant role in the etiology and progression of endometriosis, calling for further investigation into the estrogen-mimicking properties of environmental metals and their regulation. Additionally, the study highlights the need for preventive measures to reduce heavy metal exposure to nickel and lead, particularly among women susceptible to estrogen-related diseases.

Presence of metalloestrogens in ectopic endometrial tissue

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study quantified metalloestrogens—cadmium, nickel, and lead—in ectopic endometrial tissue, suggesting their role in endometriosis persistence.

What Was Studied?

This study investigated the presence of metalloestrogens—heavy metals with estrogenic effects—in ectopic endometrial tissue. Metalloestrogens, such as cadmium, nickel, and lead, have been implicated in estrogen-dependent diseases like endometriosis. The study aimed to quantify these metals in ectopic endometrial tissues from women diagnosed with endometriosis, using advanced analytical techniques.

Who Was Studied?

The study included 50 women of reproductive age who had endometriosis confirmed through laparotomy or laparoscopy. The participants were patients from a gynecology unit at a tertiary care hospital in Sri Lanka. Samples of ectopic endometrial tissue were collected from these women during surgical procedures, and the disease severity was classified based on the Revised American Society for Reproductive Medicine classification system.

What Were the Most Important Findings?

The study found significant levels of three metalloestrogens—cadmium (2.861 µg/kg), nickel (17.547 µg/kg), and lead (25.785 µg/kg)—in all ectopic endometrial tissue samples analyzed. Among these, lead exhibited the highest concentration. The study is notable for being the first to report the quantitative detection of metalloestrogens in ectopic endometrial tissue. Notably, the presence of these metals varied slightly depending on the tissue site, such as the wall of an endometrioma or nodules in the pelvic region, though these differences were not statistically significant. The findings suggest a potential role for environmental metalloestrogens in the persistence and progression of endometriosis.

What Are the Greatest Implications of This Study?

The detection of metalloestrogens in ectopic endometrial tissue underscores their role in the etiology and maintenance of endometriosis. These metals may act as endocrine disruptors, binding to estrogen receptors in ectopic tissue and mimicking estrogenic effects, thereby contributing to the persistence of the disease. The findings highlight the need for further research to elucidate the mechanistic pathways by which metalloestrogens influence endometriosis. Clinicians should consider environmental exposures and diet as a factor in managing and preventing this condition.

Prevalence and associated factors of premenstrual dysphoric disorder among high school students in Finote Selam town, northwest Ethiopia

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Among Ethiopian high school girls, PMDD affects one-third, linked to irregular cycles, depression, prolonged menstruation, and stress. Early screening and mental health support are crucial to improving well-being and academic success.

What was studied?

This cross-sectional study examined the prevalence of premenstrual dysphoric disorder (PMDD) and its associated factors among high school female students in Finote Selam town, northwest Ethiopia. Using DSM-5 criteria and self-administered questionnaires, the study aimed to quantify PMDD prevalence and identify clinical, psychosocial, and menstrual-related predictors affecting this population's mental health and academic performance.

Who was studied?

The research included 548 high school female students aged 15 to 22 years with regular menstrual cycles, excluding those with serious illness or recent school transfers. Participants completed validated questionnaires assessing PMDD symptoms, depression, perceived stress, social support, menstrual characteristics, and behavioral factors such as substance use.

What were the most important findings?

The study found a high PMDD prevalence (33%), with physical symptoms like breast tenderness and fatigue being most common. Key factors significantly associated with PMDD included irregular menstrual cycles, depressive symptoms, longer menstruation duration, and high perceived stress. PMDD significantly impacted academic performance, social functioning, and psychological well-being. The findings aligned with prior Ethiopian and African studies but were higher than reports from developed countries, possibly reflecting sociocultural, infrastructural, and menstrual hygiene differences influencing symptom expression and health-seeking behaviors.

What are the greatest implications of this study?

This study highlights PMDD as a prevalent and underrecognized condition adversely affecting adolescent females' mental health and educational outcomes in low-resource settings. It emphasizes the urgent need for early screening, stress reduction interventions, and targeted mental health support within primary healthcare and school systems. Addressing menstrual health education, improving hygiene management, and mitigating psychosocial stressors could reduce PMDD burden and improve quality of life. These insights guide clinicians and policymakers toward culturally sensitive, accessible strategies for PMDD diagnosis and management in similar populations.

Probiotics and Polycystic Ovary Syndrome: A Perspective for Management in Adolescents with Obesity

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review highlights how probiotic and synbiotic supplementation improves insulin resistance, inflammation, and androgen levels in obese adolescents with PCOS. Microbiome modulation offers a promising non-pharmacological therapy.

What was reviewed?

This narrative review explored the potential role of probiotics and synbiotics in managing polycystic ovary syndrome (PCOS) in adolescents with obesity. The authors conducted a non-systematic analysis of meta-analyses, clinical trials, and reviews published in the past two decades, aiming to assess whether probiotic supplementation can improve hormonal balance, metabolic profiles, inflammation, and overall PCOS symptomatology. The review sought to consolidate findings about the interaction between obesity, dysbiosis, and PCOS, especially in adolescents, and how targeting the gut microbiome with probiotics could serve as a preventive or therapeutic intervention.

Who was reviewed?

The review focused on adolescent females aged 10–19 with obesity and PCOS, a group particularly susceptible to metabolic and reproductive dysfunction due to overlapping hormonal, genetic, and environmental factors. The included studies comprised both animal and human trials, with some focusing exclusively on adult women while others incorporated adolescent data. The microbiome's role was assessed through its relationship with hyperandrogenism, insulin resistance, inflammatory markers, and hormonal modulation. The review emphasized evidence from randomized controlled trials and meta-analyses but acknowledged that many studies were conducted in adult populations, underscoring the need for adolescent-specific research.

What were the most important findings?

The review identified a clear link between obesity, PCOS, and gut dysbiosis. PCOS is associated with reduced microbial diversity, an imbalance in Firmicutes/Bacteroidetes ratio, and an overrepresentation of pathogenic bacteria like Escherichia and Shigella, accompanied by a reduction in beneficial Lactobacilli and Bifidobacteria. Dysbiosis appears to impair gut barrier integrity and promote systemic inflammation through increased intestinal permeability and lipopolysaccharide (LPS) translocation. These microbial alterations are connected to heightened insulin resistance, elevated testosterone, and disrupted follicular development.

Probiotic supplementation was shown to improve several PCOS-related outcomes. Studies reported reductions in serum testosterone, free androgen index (FAI), HOMA-IR, weight, and BMI. Simultaneously, increases in SHBG, nitric oxide, glutathione, and anti-inflammatory cytokines (e.g., IL-10) were observed. Notably, probiotic strains such as Lactobacillus acidophilus, L. rhamnosus, L. plantarum, Bifidobacterium bifidum, and B. lactis were associated with improved hormonal and metabolic outcomes. The production of short-chain fatty acids (SCFAs), particularly butyrate, played a key mechanistic role by reducing inflammation, improving insulin sensitivity, and restoring gut-ovary axis balance. Additionally, synbiotic supplementation (combining probiotics with prebiotics like inulin or FOS) yielded superior outcomes in some studies, especially in reducing testosterone levels.

What are the greatest implications of this review?

This review underscores the promising role of microbiome-targeted interventions, particularly probiotics and synbiotics, in managing PCOS among adolescents with obesity. While conventional treatments like metformin and oral contraceptives address insulin resistance and hyperandrogenism, they are often accompanied by side effects and limited adherence, especially in young patients. In contrast, probiotics offer a well-tolerated, non-invasive strategy to modulate gut microbiota, reduce systemic inflammation, and improve endocrine function. Clinicians should consider the gut–brain–ovary axis as a central pathway in PCOS pathophysiology and incorporate microbiome-informed interventions alongside dietary and lifestyle modifications. Given the preventive potential of early microbiome modulation, probiotic use in high-risk adolescent populations may help mitigate long-term metabolic and reproductive complications. However, further longitudinal and adolescent-focused clinical trials are essential to refine strain-specific recommendations, dosage, and duration for optimal therapeutic benefit.

Profile of Bile Acid Metabolomics in the Follicular Fluid of PCOS Patients

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study reveals elevated primary and conjugated bile acids in follicular fluid of PCOS patients, suggesting a novel ovarian micro-environmental role in ovulation dysfunction and potential links to gut microbiota.

What was studied?

This study examined the profile of bile acid metabolomics in the follicular fluid (FF) of women with polycystic ovary syndrome (PCOS). For the first time, researchers evaluated how the composition of bile acids in the ovarian micro-environment differs between PCOS and non-PCOS women, aiming to elucidate the potential roles of bile acids in follicular development and ovulatory dysfunction. Using ultra-performance liquid chromatography coupled with tandem mass spectrometry (UPLC-MS/MS), the study quantified 24 bile acid metabolites and assessed their clinical correlations in relation to hormone levels and ovarian characteristics.

Who was studied?

The study included 35 women diagnosed with PCOS based on the Rotterdam criteria and 31 control women undergoing assisted reproduction for male or tubal factor infertility, all with normal menstrual cycles and ovarian function. All participants were under 40 years old and had no history of endocrine disorders, ovarian surgery, or liver dysfunction. The researchers ensured matched baseline characteristics, such as liver enzyme levels and BMI, to avoid confounding bile acid data with liver metabolism variations. Follicular fluid was collected during oocyte retrieval procedures as part of IVF or ICSI cycles.

What were the most important findings?

The study identified a distinct alteration in bile acid composition in the follicular fluid of PCOS patients. Although the total bile acid concentration was not significantly different, specific metabolites showed statistically significant elevations. Four bile acid metabolites, glycocholic acid (GCA), taurocholic acid (TCA), glycochenodeoxycholic acid (GCDCA), and chenodeoxycholic acid-3-β-D-glucuronide (CDCA-3Gln), were significantly higher in PCOS FF compared to controls. The increase was most notable in conjugated and primary bile acids, while levels of secondary and unconjugated bile acids remained unchanged.

GCDCA demonstrated a positive correlation with serum FSH and LH, suggesting its potential involvement in disrupted folliculogenesis and ovulation associated with PCOS. Similarly, CDCA-3Gln correlated with antral follicle count (AFC), indicating a possible relationship with ovarian reserve status. Importantly, there was no association between these bile acid changes and insulin sensitivity, highlighting a potential ovary-specific bile acid dysregulation in PCOS independent of systemic insulin resistance. These bile acids, particularly the conjugated primary forms, may reflect altered bile acid metabolism influenced by gut microbiota, especially given prior reports linking intestinal flora with circulating bile acid changes in PCOS. Although not directly assessed in this study, the elevated GCA and TCA levels echo findings from serum metabolomics that suggest microbial contributions, possibly involving Bacteroides and Clostridium species known to interact with bile acid pools.

What are the greatest implications of this study?

This study underscores the potential role of bile acid metabolites as contributors to the pathophysiology of PCOS, particularly within the ovarian micro-environment. It suggests that bile acid dysregulation may impact granulosa cell function and follicular development, which could influence ovulation. Identifying FF bile acids as potential biomarkers opens new diagnostic and therapeutic avenues, particularly in targeting the bile acid signaling axis or modulating gut microbiota to restore metabolic balance within the ovary. The findings encourage a shift toward integrating ovarian metabolomics with systemic and microbiome data to better characterize PCOS subtypes and treatment targets.

Promising Drug Candidates for the Treatment of Polycystic Ovary Syndrome (PCOS) as Alternatives to the Classical Medication Metformin

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The study evaluates the efficacy of Irosustat, STX140, and compound 1G as alternative treatments for PCOS, showing improvements in metabolic and hormonal profiles, with promising results on oxidative stress and inflammation markers. These compounds offer potential advantages over current PCOS therapies like metformin.

What was studied?

This study focused on identifying new drug candidates for the treatment of Polycystic Ovary Syndrome (PCOS), with an emphasis on evaluating the effects of various compounds on PCOS pathophysiology. The study investigated the use of Irosustat (STX64), STX140, and compound 1G as potential alternatives to metformin in managing symptoms related to hormonal imbalance, metabolic dysfunction, and oxidative stress commonly seen in PCOS.

Who was studied?

The study utilized female Wistar rats to investigate the therapeutic effects of these drug candidates. PCOS was induced in the rats by administering letrozole (1 mg/kg/day) for 35 days, with the onset of abnormal estrous cycles confirming the induction of the condition. Rats were then divided into treatment groups, with one group receiving metformin (500 mg/kg/day) as a reference drug, while the others received STX64, STX140, or 1G for 30 days. The effects were analyzed through biochemical measurements, oxidative stress markers, and histological studies.

What were the most important findings?

The study found that the drug candidates Irosustat, STX140, and compound 1G all demonstrated promising effects on PCOS-related features. Treatment with these compounds resulted in significant improvements in various biochemical parameters, including lipid profiles, blood glucose levels, and hormone levels (testosterone, progesterone, luteinizing hormone (LH), follicle-stimulating hormone (FSH), and estradiol). These treatments also showed beneficial effects on oxidative stress and inflammation pathways, with improvements in Akt, mTOR, and AMPK-α signaling pathways. Histological studies revealed a reduction in the weight of ovaries and the disappearance of fluid-filled cysts in the treatment groups, suggesting potential for reversing ovarian morphology associated with PCOS. The drug candidates also demonstrated less adverse effect on metabolic parameters compared to untreated PCOS rats, thus highlighting their therapeutic potential as alternatives to metformin.

From a microbiome perspective, these improvements could be linked to the modulation of gut microbiota and reduced systemic inflammation. For example, Irosustat and STX140, by regulating androgen levels and improving metabolic health, may impact the gut's microbial balance, favoring beneficial bacteria that support metabolic functions and reduce inflammation. Additionally, these compounds' effects on oxidative stress markers could influence the gut-brain axis, which is crucial in the pathophysiology of PCOS.

What are the greatest implications of this study?

The greatest implication of this study lies in the identification of promising drug candidates, particularly Irosustat, STX140, and compound 1G, as potential treatments for PCOS, especially for patients who do not tolerate metformin. These drug candidates work by targeting oxidative stress, inflammatory pathways, and hormonal imbalances, which are central to PCOS pathophysiology. The findings suggest that these drugs could offer a more comprehensive treatment approach compared to current options, potentially improving not only the metabolic and hormonal aspects of PCOS but also the quality of life for affected women. The study also opens the door for further exploration into the use of these compounds in human trials, highlighting the need for personalized treatment options for women with PCOS

Recurrent Bacterial Vaginosis Following Metronidazole Treatment is Associated with Microbiota Richness at Diagnosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study links high pre-treatment vaginal microbiota diversity to BV recurrence after metronidazole. Women with sustained clearance had lower richness. Lactobacillus iners improved immune markers temporarily, but no cases achieved L. crispatus dominance. Biofilm-forming taxa like Atopobium persisted, suggesting resistance mechanisms.

What was Studied?

This study investigated the association between pre-treatment vaginal microbiota composition and the likelihood of recurrent bacterial vaginosis (BV) following metronidazole treatment. The researchers analyzed cervicovaginal lavage samples from women diagnosed with BV using 16S rRNA gene sequencing to identify microbial signatures linked to treatment failure or success. The study aimed to determine whether specific microbiota characteristics at diagnosis could predict treatment outcomes, including transient clearance, sustained clearance, or recurrence of BV.

Who was Studied?

The study included 28 women diagnosed with symptomatic BV, confirmed by Nugent scoring, who were enrolled in a clinical trial. Participants were non-pregnant, free of other reproductive tract infections, and had not used antibiotics in the 14 days before enrollment. Samples were collected at baseline (pre-treatment), 7–10 days post-treatment, and 28–32 days post-treatment to assess microbial and immune changes.

What were the most Important Findings?

The study revealed that women who failed to clear BV or experienced recurrence had significantly higher pre-treatment microbial richness and evenness than those with sustained clearance. Significant microbial associations (MMA) included polymicrobial anaerobic taxa such as Gardnerella vaginalisPrevotellaSneathia, and Atopobium, which were dominant at baseline. Notably, Lactobacillus iners (CT2) dominance post-treatment was associated with improved mucosal immune markers, including elevated SLPI and reduced ICAM-1, but these benefits were transient in cases of recurrence. The persistence of diverse, low-abundance taxa and biofilm-forming bacteria like Atopobium and Sneathia post-treatment suggested their role in treatment resistance. Importantly, no participants achieved Lactobacillus crispatus (CT1) dominance, highlighting a gap in current therapeutic efficacy.

What are the Implications of this Study?

The findings underscore the limitations of metronidazole in treating BV, particularly in cases with high pre-treatment microbial diversity. The study suggests that microbiome profiling could help identify women at risk of treatment failure, paving the way for personalized therapies. Future research should explore adjunct treatments, such as Lactobacillus crispatus biotherapeutics or biofilm disruptors, to improve outcomes. Additionally, the transient immune improvements observed with Lactobacillus dominance emphasize the need for sustained microbiome modulation to prevent recurrence and associated complications like STI susceptibility.

Relaxed fibronectin: a potential novel target for imaging endometriotic lesions

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Research on relaxed fibronectin as a target for imaging endometriotic lesions showed that a novel radiotracer binds preferentially to this protein in disease areas. This finding could lead to improved diagnostic techniques for endometriosis, offering a non-invasive method to detect lesions accurately, thereby enhancing treatment planning and patient outcomes.

What was studied?

The study investigated relaxed fibronectin as a novel target for imaging endometriotic lesions. Researchers explored using a preclinical radiotracer, [111In]In-FnBPA5, which binds specifically to relaxed fibronectin, an extracellular matrix protein involved in the pathogenesis of diseases like cancer and fibrosis.

Who was studied?

The study involved preclinical experiments using mice and immunohistochemical analysis on tissue samples from mice and patients diagnosed with endometriosis.

What were the most important findings?

The radiotracer [111In]In-FnBPA5 accumulated in the mouse uterus, with uptake varying according to the estrous cycle, suggesting an increased abundance of relaxed fibronectin during estrogen-dependent phases. Immunohistochemical analysis on patient-derived tissues showed that relaxed fibronectin is preferentially located near the endometriotic stroma, supporting its potential as a target for imaging endometriosis.

What are the greatest implications of this study?

The findings that [111In]In-FnBPA5 uptake varies in the mouse uterus with the estrous cycle, indicating increased relaxed fibronectin during estrogen-dependent phases, hold significant implications for future research on endometriosis.

Biomarker Identification: Understanding the fluctuation of relaxed fibronectin could help identify biomarkers for endometriosis, enabling earlier and more accurate diagnosis.

Pathogenesis Insights: These results suggest that estrogen-driven changes in fibronectin might play a role in the development or exacerbation of endometriosis. This could lead to a better understanding of the disease’s underlying mechanisms.

Targeted Therapies: By highlighting the relationship between estrogen, fibronectin, and endometrial tissue changes, new therapeutic targets may be identified, paving the way for treatments that modulate fibronectin or its pathways.

Diagnostic Imaging: The study suggests that targeting relaxed fibronectin could significantly improve the diagnostic imaging of endometriosis. This approach may lead to developing a specific radiotracer for noninvasive detection of endometriotic lesions, potentially enhancing diagnosis accuracy and aiding in better disease management.

Clinical Application: The researchers also suggest using gallium-68 for potential clinical application, which could further refine imaging techniques and improve patient outcomes.

Reproductive Microbiomes: Using the Microbiome as a Novel Diagnostic Tool for Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This cross-sectional observational study examines how endometriosis affects the bacterial communities of the uterus and cervix, considering the condition's role in inflammation, pain management, and infertility in women.

What was studied?

The study aimed to investigate how endometriosis affects the uterine and cervical bacterial communities. Utilizing next-generation amplicon sequencing of the bacterial 16S rRNA gene, the research sought to identify alterations in these microbiomes associated with endometriosis and to determine if specific bacterial taxa within the cervix could help diagnose active endometriosis, potentially avoiding the need for invasive diagnostic procedures like laparoscopic surgery.

 

Who was studied?

Nineteen pre-menopausal women undergoing laparoscopic surgery for pelvic pain with suspicion or known endometriosis constituted the experimental group (n=10, with endometriosis stages I-IV), while women undergoing surgery for benign uterine or ovarian conditions served as controls (n=9). The control group was examined during surgery to confirm the absence of endometriotic lesions. The staging of endometriosis for patients in the experimental group was performed using the revised American Society for Reproductive Medicine (rASRM) classification scale.

 

Key findings of the study include significant differences in bacterial communities between uterine and cervical samples, both in species diversity and abundance, with the uterus displaying a diverse profile of Bacteroidetes and Firmicutes, and the cervix dominated by Lactobacillus. No significant differences in bacterial communities were noted across different endometriosis stages on the day of surgery. However, a distinct cervical bacterial community in a stage III endometriosis patient suggests a link between disease severity and microbiome alterations. Additionally, notable fluctuations in the cervical microbiome were observed over time in this patient, indicating dynamic microbiome changes associated with disease progression and treatment.

 

What are the greatest implications of this study?

The findings underscore the potential of bacterial community profiling as a diagnostic tool for endometriosis, offering a non-invasive method to identify the disease in asymptomatic, infertile women. This approach could facilitate earlier diagnosis and treatment, potentially improving fertility outcomes and reducing the need for invasive diagnostic surgeries. The study also highlights the dynamic nature of the uterine and cervical microbiomes in relation to endometriosis, suggesting that microbiome alterations could be linked to disease severity and progression. Understanding these microbial community changes opens new avenues for researching endometriosis pathogenesis and developing novel therapeutic strategies that target microbiome modulation. Moreover, the fluctuations observed in the microbiome over time, especially in patients with advanced disease, may offer insights into predicting disease progression and treatment outcomes, including fertility potential post-treatment.

Repurposing new drug candidates and identifying crucial molecules underlying PCOS Pathogenesis Based On Bioinformatics Analysis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study highlights the potential for repurposing FDA-approved drugs to treat PCOS, identifying crucial proteins and pathways linked to the disorder.

What was studied?

This study investigated the potential of repurposing FDA-approved drugs to treat Polycystic Ovary Syndrome (PCOS). Using bioinformatics tools, the authors analyzed protein-protein interactions (PPIs) related to PCOS and explored how certain drugs could interact with these proteins to potentially mitigate the pathogenesis of the disorder. The analysis particularly focused on identifying crucial molecules and drug targets that could offer new therapeutic avenues for managing PCOS, particularly addressing issues related to infertility, hormonal imbalance, and metabolic dysfunction.

Who was studied?

The study did not involve human participants directly, but it analyzed proteomic data from existing datasets. The aim was to examine the molecular mechanisms associated with PCOS by constructing a protein interaction network from proteomics data. The study also considered FDA-approved drugs and their interactions with proteins identified in the PCOS pathway, which are crucial to understanding how these drugs may alter disease progression or improve clinical outcomes.

What were the most important findings?

The study identified several proteins, including VEGF, EGF, TGFB1, AGT, AMBP, and RBP4, that are crucial to the pathophysiology of PCOS. These proteins were shared between the PCOS protein network and the proteins targeted by FDA-approved drugs, such as metformin, pioglitazone, spironolactone, and letrozole. The PI3K/AKT signaling pathway, which plays a critical role in ovarian function and follicular development, was also identified as a major point of convergence between PCOS and the therapeutic drugs. This pathway influences oocyte maturation and granulosa cell proliferation, both of which are affected in PCOS.

The analysis also revealed that repurposing drugs like metformin, pioglitazone, and spironolactone could influence these crucial proteins and pathways. The study suggested that other FDA-approved drugs, such as copper and zinc compounds, could also be considered for further investigation due to their potential role in managing PCOS. These findings suggest that targeting the protein networks identified in the study could lead to more effective treatments for PCOS, particularly for fertility and metabolic issues associated with the condition.

What are the greatest implications of this study?

The greatest implication of this study is the potential to repurpose existing FDA-approved drugs for the treatment of PCOS. By identifying key molecular pathways involved in PCOS and matching them with drugs that already target these pathways, the study paves the way for faster, more affordable therapeutic options. Additionally, it highlights the utility of systems biology and bioinformatics in drug repurposing, providing clinicians with new insights into how existing medications might be leveraged to address PCOS-related infertility, hormonal imbalances, and metabolic dysfunction. Further experimental validation of these drug interactions could lead to more personalized, efficient treatments for women with PCOS.

Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome (PCOS)

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review connects gut microbiota dysbiosis to insulin resistance and hyperandrogenism in PCOS, identifying reduced SCFA producers and increased LPS-producing microbes as key signatures. It supports microbiota-targeted therapies as promising treatments for metabolic and reproductive symptoms in PCOS.

What Was Reviewed?

This paper reviewed the role of gut microbiota in the development of insulin resistance (IR) and its contribution to the pathophysiology of polycystic ovary syndrome (PCOS). The authors synthesized a wide body of literature spanning clinical studies, animal models, and microbial metabolomics to illustrate how gut dysbiosis acts as a central mechanism driving PCOS through metabolic, inflammatory, and hormonal pathways. The review explored multiple axes of gut microbiota influence, including endotoxemia, short-chain fatty acid (SCFA) production, bile acid metabolism, branched-chain amino acid (BCAA) synthesis, the gut-brain axis, and hyperandrogenism. These interconnected pathways ultimately lead to IR, hyperinsulinemia, and hormonal imbalance, all of which underpin the key clinical features of PCOS, including ovulatory dysfunction, endometrial receptivity impairment, obesity, and metabolic syndrome.

Who Was Reviewed?

The review encompassed findings from both human and animal studies. Human studies included women diagnosed with PCOS compared to controls, covering lean, obese, insulin-resistant, and normo-insulinemic phenotypes. The authors also incorporated data from rodent models, particularly letrozole-induced PCOS rats and prenatal androgen exposure models, to investigate microbial composition shifts and their functional impact on reproductive and metabolic phenotypes. Specific microbial taxa were evaluated through 16S rRNA sequencing and metagenomics, while endocrine and metabolic parameters were tracked to map microbial influence on systemic physiology.

What Were the Most Important Findings?

The review identified major microbial associations (MMAs) that characterize PCOS-related dysbiosis. At the phylum level, PCOS patients demonstrated a decreased abundance of Bacteroidetes and increased Firmicutes, often resulting in a higher Firmicutes/Bacteroidetes ratio, which has been linked to obesity and metabolic syndrome. Reductions in Bifidobacterium, Lactobacillus, Faecalibacterium prausnitzii, and Roseburia were consistently reported. These microbial shifts compromise intestinal barrier function, leading to increased translocation of lipopolysaccharides (LPS), a key endotoxin that triggers chronic systemic inflammation via the TLR4-CD14 signaling pathway. This inflammation impairs insulin signaling and exacerbates hyperinsulinemia, which then stimulates ovarian androgen production and suppresses SHBG, intensifying the free androgen burden. Additionally, the review highlighted that BCAA-producing microbes such as Prevotella further aggravate insulin resistance. Bile acid metabolism was also altered, with decreased levels of beneficial bile acids like glycodeoxycholic acid and tauroursodeoxycholic acid. These changes interfere with signaling through FXR and GPBAR1, reducing insulin sensitivity.

What Are the Implications of This Review?

This review reframes PCOS as a condition deeply intertwined with microbiota-related metabolic and endocrine dysregulation. For clinicians, this connection offers actionable insights for diagnosis and treatment. The consistent microbial signatures associated with PCOS, such as reduced SCFA producers and increased LPS-producing gram-negative bacteria, support the potential for gut-targeted therapies. Dietary interventions that promote microbial diversity, particularly high-fiber, low-sugar regimens, may alleviate metabolic and reproductive symptoms. The paper also supports the use of probiotics (e.g., Lactobacillus and Bifidobacterium species), prebiotics (e.g., inulin), and fecal microbiota transplantation (FMT) as novel adjunct therapies. In animal studies, both probiotic and FMT interventions restored estrous cycles and improved ovarian morphology, suggesting that modulating the gut microbiome could directly impact ovulation and fertility. However, the authors emphasize that more randomized controlled trials and functional studies are necessary to validate these treatments and define phenotype-specific microbial targets.

Role of Metformin in Polycystic Ovary Syndrome (PCOS)-Related Infertility

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

This review examines the role of metformin in treating PCOS-related infertility, highlighting its effectiveness in improving insulin sensitivity, reducing hyperandrogenism, and restoring ovulation. Metformin serves as a first-line therapy, offering significant benefits for women with anovulatory infertility due to PCOS.

What was reviewed?

This review explores the role of metformin in treating polycystic ovary syndrome (PCOS)-related infertility. PCOS is a common endocrinological disorder that can lead to infertility, characterized by insulin resistance, hyperandrogenism, and anovulation. The review discusses metformin’s mechanisms, its impact on insulin sensitivity, its role in improving ovulation, and its effectiveness in managing metabolic and hormonal imbalances in women with PCOS. The review also emphasizes the drug's benefits in improving menstrual cyclicity and reducing hyperandrogenism, ultimately aiding in fertility restoration.

Who was reviewed?

The review synthesizes findings from various clinical studies and trials examining the effects of metformin on women with PCOS. It draws on observational studies and randomized controlled trials to evaluate the efficacy of metformin in addressing infertility associated with PCOS. The women studied in these trials typically had anovulatory infertility, hyperandrogenism, and varying degrees of insulin resistance, and they were treated with metformin to assess its impact on ovulation and fertility.

What were the most important findings?

The review found that metformin has significant therapeutic benefits for women with PCOS, particularly in restoring menstrual regularity and improving ovulation rates. Metformin works primarily by improving insulin sensitivity, which reduces hyperinsulinemia—a key factor in the pathogenesis of PCOS. This insulin-sensitizing effect contributes to lower circulating androgen levels, which is crucial in managing symptoms like hirsutism and acne. In several studies, metformin, either alone or in combination with other treatments like clomifene citrate, successfully induced ovulation in women who were resistant to standard treatments.

Furthermore, metformin appears to improve metabolic dysfunctions common in PCOS, including insulin resistance, dyslipidemia, and obesity, all of which contribute to the infertility and long-term health risks associated with the condition. However, the review also noted that while metformin improves metabolic and reproductive outcomes, its efficacy in women with significant obesity is less pronounced. The review also highlights that metformin is generally well-tolerated, although some women may experience gastrointestinal side effects.

What are the greatest implications of this review?

The review underscores metformin’s potential as a first-line treatment for women with PCOS-related infertility, especially for those who are insulin-resistant and non-obese. The findings suggest that metformin could be a safer and more accessible alternative to more invasive fertility treatments like in vitro fertilization (IVF). Moreover, metformin’s role in reducing the risk of ovarian hyperstimulation syndrome during assisted reproductive technology procedures makes it particularly valuable in IVF protocols. The review also emphasizes the need for further studies to determine the optimal dose and long-term benefits of metformin, particularly for women with more severe obesity or metabolic complications.

Secnidazole for the Treatment of Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This systematic review analyzed six trials evaluating secnidazole for bacterial vaginosis treatment. Secnidazole at 2 g significantly improved clinical and microbiologic cure rates, showing comparable efficacy to metronidazole. The single-dose regimen enhances adherence, offering an alternative for patients with recurrent BV or adverse effects from standard therapies.

What was Reviewed?

This systematic review evaluated the clinical efficacy, safety, and microbiological outcomes of secnidazole as a treatment option for bacterial vaginosis (BV). The authors reviewed randomized controlled trials that compared secnidazole at different doses with placebo, standard antibiotic regimens, or combination therapies. The review also considered how secnidazole affected the vaginal microbiota, particularly its ability to reduce the abundance of BV-associated bacteria and restore beneficial Lactobacillus species.

Who was Reviewed?

The review encompassed clinical studies involving adult women diagnosed with bacterial vaginosis, with diagnosis typically based on Amsel criteria or Nugent score. The included studies varied in sample size but consistently targeted non-pregnant women of reproductive age who were experiencing symptomatic or recurrent BV. The population also included women with a history of BV treatment failures or recurrences, a subgroup of particular interest due to the chronic and recurrent nature of the condition.

What were the most Important Findings?

This review demonstrated that secnidazole significantly improved both the clinical and microbiological cure rates of bacterial vaginosis compared to placebo. Specifically, in women with three or fewer BV episodes in the last year, 2 g secnidazole substantially reduced BV risk. In women with four or more episodes, the benefit persisted but with slightly lower magnitude.

The clinical cure rate of 2 g secnidazole was comparable to metronidazole (500 mg), oral metronidazole 2 g single dose, secnidazole combined with vaginal metronidazole, or secnidazole plus vaginal ornidazole. However, the 2 g dose performed better than the 1 g dose.

This review highlighted that probiotic therapy was not the focus, but secnidazole use indirectly supports the concept of restoring vaginal eubiosis by reducing pathogenic bacteria. The review did not explicitly measure microbiome shifts in terms of Lactobacillus species or pathogenic taxa, but the improved microbiologic cure rate reflects pathogen reduction.

The authors also emphasized that a single-dose regimen of secnidazole improved patient adherence compared to multi-dose metronidazole or tinidazole therapies. However, beyond adherence, secnidazole's therapeutic effect was statistically similar to these standard treatments. The review proposed secnidazole as a good alternative for women who experienced adverse effects or recurrence with current BV medications.

What are the Implications of this Review?

This review offers clear clinical guidance: secnidazole at 2 g is an effective, single-dose treatment option for bacterial vaginosis, providing comparable cure rates to metronidazole and combination therapies. It may serve as a valuable alternative, particularly for women with recurrent BV or those who face side effects from standard antibiotics. Additionally, while the review did not analyze microbial signatures in detail, the consistent microbiologic cure rates indirectly support the role of secnidazole in reducing BV-associated dysbiosis. Clinicians should consider secnidazole as a viable option in their therapeutic arsenal, particularly when treatment adherence and recurrence prevention are priorities.

Serum copper assessment in patients with polycystic ovary syndrome and tubal infertility

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This 5-year study found significantly higher serum copper levels in women with PCOS, correlating with triglycerides and BMI but not IVF outcomes. Copper may reflect metabolic dysfunction and gut-related oxidative stress in PCOS.

What was studied?

This retrospective, cross-sectional study assessed the serum copper levels in women with infertility, specifically comparing patients with polycystic ovary syndrome (PCOS) and those with tubal infertility, to evaluate copper’s association with hormonal, metabolic, and in vitro fertilization (IVF) parameters. Over five years, the study explored whether elevated copper levels correlate with PCOS clinical phenotypes and IVF outcomes, and whether copper could be a relevant biomarker in reproductive health.

Who was studied?

A total of 766 Chinese women under age 38 were included, comprising 560 women with tubal infertility (no-PCOS group) and 206 women with PCOS undergoing IVF. The diagnosis of PCOS was based on the modified Rotterdam criteria. All participants underwent their first IVF cycle at the Center of Reproduction and Genetics, Suzhou Municipal Hospital, between January 2018 and December 2022. Baseline clinical, metabolic, hormonal, and trace element data, including fasting glucose, triglycerides, cholesterol, lipoproteins, and serum copper, were collected and analyzed.

What were the most important findings?

Women with PCOS showed significantly elevated serum copper concentrations compared to those with tubal infertility. Within the PCOS group, serum copper positively correlated with body mass index (BMI) and triglycerides (TG), but not with fasting glucose, LDL, or HDL. In contrast, the no-PCOS group demonstrated a broader metabolic correlation with copper, including BMI, TG, TC, LDL, and a negative correlation with HDL. Despite these associations, serum copper levels did not significantly predict IVF outcomes such as oocyte retrieval, MII oocyte rate, fertilization, or embryo quality, once confounding variables were controlled for.

From a microbiome perspective, this study indirectly highlights the importance of copper in modulating oxidative stress and lipid metabolism, both of which influence microbial diversity. Elevated copper is known to disrupt microbial homeostasis by promoting oxidative stress and favoring the growth of pro-inflammatory species such as Desulfovibrio and Proteobacteria. Conversely, lower copper levels may favor microbial populations that support metabolic stability. These shifts may exacerbate the gut dysbiosis observed in PCOS, which is closely linked to insulin resistance, chronic inflammation, and hormonal imbalances.

What are the implications of this study?

This study underscores the clinical relevance of elevated serum copper as a marker of metabolic dysregulation in women with PCOS, though not a direct predictor of IVF outcomes. The strong correlation between copper and triglycerides suggests a metabolic link that may be mediated by oxidative stress or microbiome alterations. Given copper’s known impact on gut microbial balance and inflammatory signaling, these findings reinforce the importance of trace element assessment in PCOS, particularly in the context of metabolic health and infertility. While copper may not serve as a standalone biomarker for fertility success, its elevation in PCOS warrants further exploration in longitudinal studies to assess causality and mechanistic pathways, including copper’s effect on the microbiota–ovary axis. Clinicians should consider monitoring serum copper alongside traditional lipid profiles in PCOS management, especially in cases of treatment-resistant metabolic dysfunction.

Serum Copper Level and Polycystic Ovarian Syndrome: A Meta-Analysis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This meta-analysis confirms that serum copper levels are significantly elevated in women with PCOS. Copper may drive hormonal imbalance and oxidative stress, and could influence gut microbiota, highlighting its potential as a biomarker and therapeutic target in PCOS management.

What was reviewed?

This meta-analysis reviewed the association between serum copper levels and polycystic ovary syndrome (PCOS) by pooling data from nine cross-sectional studies encompassing a total of 2,274 women (1,168 with PCOS and 1,106 healthy controls). The goal was to clarify inconsistent findings in previous literature regarding whether circulating copper levels differ significantly in women with PCOS and whether elevated copper might play a role in the pathophysiology of the disorder.

Who was reviewed?

The reviewed studies included adult women diagnosed with PCOS based on the Rotterdam criteria, alongside matched control participants without PCOS. These studies were conducted across diverse geographical regions, including China, Turkey, India, Iran, Sudan, and the USA, and were published between 2012 and 2020. The studies employed either atomic absorption spectrophotometry or inductively coupled plasma mass spectrometry to quantify serum copper. All were assessed as high quality using the Newcastle–Ottawa Scale.

What were the most important findings?

The meta-analysis found that women with PCOS have significantly higher serum copper levels than healthy controls, with a standardized mean difference (SMD) of 0.51 µg/mL. This effect remained statistically significant even after conducting sensitivity analyses and omitting a single contradictory study, which showed an inverse trend. Subgroup analyses by country (China vs. Western) confirmed that elevated copper was consistently observed in both populations, suggesting a robust association independent of geographic or ethnic background.

Biologically, copper acts as a cofactor in several enzymatic reactions involving oxidative metabolism and plays a role in hormone receptor regulation, including acting as a metalloestrogen capable of activating estrogen receptor alpha. Increased copper levels may contribute to oxidative stress in PCOS through enhanced ROS generation, glutathione depletion, and lipid peroxidation. These processes can disrupt endocrine function and potentially influence ovarian physiology, although more research is needed to clarify copper’s direct role in PCOS pathogenesis.

From a microbiome perspective, copper excess disrupts the gut microbiota by decreasing beneficial taxa such as Bifidobacterium spp. and Faecalibacterium prausnitzii, while favoring pro-inflammatory genera like Proteobacteria. These changes contribute to leaky gut, systemic inflammation, and insulin resistance, which are core features of PCOS. As such, elevated copper may not only be a marker of oxidative and inflammatory stress but also a mediator of microbiome-endocrine dysregulation.

What are the implications of this review?

This meta-analysis provides clear evidence that elevated serum copper is associated with PCOS and may play a mechanistic role in its pathophysiology. The findings support the hypothesis that copper acts as an endocrine disruptor by generating oxidative stress and modulating hormone activity, including via metalloestrogenic pathways. For clinicians, this raises the potential for serum copper to serve as a biomarker for metabolic and inflammatory status in PCOS patients. Moreover, it opens new avenues for therapeutic strategies aimed at modulating copper levels through diet, chelation, or supplementation with competing trace elements like zinc. Given copper’s known effects on microbiota, this study further strengthens the case for including trace element monitoring in microbiome-focused PCOS interventions. Future research should explore longitudinal relationships between copper exposure, microbiota changes, and hormonal dysregulation, as well as whether copper modulation improves clinical outcomes in PCOS.

Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory disease

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study revealed that women with PCOS have higher serum levels of zinc and nickel, and lower manganese, especially when inflammation is present. These imbalances may disrupt folliculogenesis and exacerbate oxidative stress, potentially influencing the microbiome and PCOS severity.

What was studied?

This clinical study investigated the serum levels of essential macro- and microelements, specifically magnesium, copper, manganese, nickel, zinc, chromium, selenium, and vanadium, in women diagnosed with polycystic ovary syndrome (PCOS), both with and without concurrent pelvic inflammatory disease (PID). The primary goal was to determine whether the concentrations of these elements differed in PCOS patients and to evaluate their possible role in the pathophysiology of PCOS, especially concerning oxidative stress, inflammation, and reproductive dysfunction.

Who was studied?

The study population consisted of three distinct groups: 30 women with PCOS and PID (Group I), 22 women with PCOS but no inflammatory pelvic disease (Group II), and 25 healthy controls (Group III). All participants were of reproductive age. The research was conducted in Ukraine and used serum samples analyzed via mass spectrometry following microwave digestion. The analysis focused on comparing element levels across the groups and interpreting their biological significance in the context of PCOS-associated metabolic and inflammatory disturbances.

What were the most important findings?

The study found that women with PCOS, regardless of inflammatory status, had significantly elevated levels of serum zinc and nickel, and decreased levels of manganese, compared to healthy controls. Magnesium, chromium, selenium, and vanadium levels did not differ significantly between groups. Zinc and nickel were especially elevated in women with concurrent PID, pointing to a synergistic relationship between inflammation and trace element dysregulation.

Mechanistically, elevated zinc and nickel levels are implicated in exacerbating oxidative stress through increased lipid peroxidation and suppression of antioxidant systems. Nickel, in particular, may damage cellular membranes, disrupt mitochondrial and DNA integrity, and impair transcription processes, collectively impairing folliculogenesis and ovulation. Elevated zinc levels may also result from the action of pro-inflammatory cytokines (IL-1, IL-6, IL-18, TNF-α), which stimulate zinc accumulation via metallothionein upregulation. Meanwhile, reduced manganese, an essential cofactor for mitochondrial antioxidant enzymes, may compromise cellular redox balance and energy metabolism. These patterns may contribute to the hormonal imbalance, chronic inflammation, and reproductive dysfunction seen in PCOS.

In microbiome terms, such trace element imbalances could promote gut dysbiosis. For example, nickel excess is known to enrich pathobionts like Proteobacteria and suppress beneficial anaerobes like Faecalibacterium prausnitzii. A similar trend with excess zinc may reduce Bifidobacterium spp. populations and increase intestinal permeability. Reduced manganese availability may impair SCFA-producing microbes, further disrupting gut-immune-endocrine signaling.

What are the implications of this study?

This study provides strong evidence that trace element imbalances, particularly elevated serum zinc and nickel and decreased manganese, may play a critical role in the pathogenesis of PCOS, especially in cases compounded by inflammation. The findings suggest that these elements influence oxidative stress, mitochondrial function, and immune activity, potentially contributing to reproductive dysfunction. Clinically, this underscores the value of monitoring serum trace elements in PCOS patients, especially those with comorbid inflammatory pelvic conditions. Moreover, nutritional strategies, such as reducing dietary sources of nickel and zinc while increasing manganese intake, may serve as adjunctive interventions. Given the known link between trace elements and microbiota composition, this study also opens a pathway to integrate trace element modulation as part of microbiome-based therapeutic approaches for PCOS.

Serum trace elements and heavy metals in polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study revealed elevated serum copper and zinc, and reduced manganese and lead, in women with PCOS, suggesting trace element dysregulation may fuel oxidative stress and hormonal imbalance. These imbalances may also impact gut microbiota and insulin resistance.

What was studied?

This clinical observational study evaluated the serum levels of essential trace elements in women with polycystic ovary syndrome (PCOS) compared to healthy controls. The research aimed to determine whether alterations in these micronutrients and toxic metals are associated with PCOS and its hormonal profile, particularly focusing on oxidative stress as a contributing mechanism. This was one of the earliest studies to analyze this specific combination of elements in PCOS patients using atomic absorption spectrophotometry.

Who was studied?

The study involved 35 women diagnosed with PCOS based on the Rotterdam criteria and 30 age- and BMI-matched healthy women serving as controls. All participants were of reproductive age and underwent thorough screening to exclude other endocrine disorders or confounding factors, such as medication use, thyroid dysfunction, or metabolic diseases. Blood samples were collected during the early follicular phase to standardize hormonal status, and serum levels of trace elements and hormones, including total testosterone and DHEAS, were measured using validated biochemical and spectrometric methods.

What were the most important findings?

Serum copper and zinc levels were significantly higher in the PCOS group, while manganese and lead levels were significantly lower. No significant differences were found in magnesium, cadmium, or cobalt concentrations between the two groups. Additionally, serum copper showed a strong negative correlation with BMI, while lead levels inversely correlated with total testosterone among PCOS patients—relationships not seen in the control group. Notably, although zinc levels were elevated in PCOS, they remained within the physiological range, whereas manganese levels in the PCOS group were approximately half those of the control group.

From a mechanistic standpoint, these findings align with the oxidative stress hypothesis of PCOS. Elevated copper can catalyze reactive oxygen species (ROS) formation and deplete intracellular glutathione, leading to mitochondrial dysfunction and inflammation. Zinc, while essential for antioxidant enzymes such as Cu/Zn superoxide dismutase (SOD), may reflect compensatory upregulation in response to inflammation. The reduced manganese levels suggest diminished activity of mitochondrial MnSOD, a critical antioxidant defense enzyme. Similarly, decreased lead levels, though surprising, may indicate redistribution or altered metabolic clearance. Each of these trace element imbalances can modulate the gut microbiome. Excess copper and zinc can suppress beneficial taxa like Bifidobacterium and Faecalibacterium prausnitzii, while manganese deficiency can impair the growth of SCFA-producing organisms that modulate inflammation and insulin signaling, features central to PCOS pathology.

What are the greatest implications of this study?

This study reinforces the hypothesis that PCOS is not only an endocrine and metabolic disorder but also a condition marked by trace element dysregulation and likely gut microbial imbalance. The observed elevations in serum copper and zinc, along with depleted manganese and altered lead levels, suggest that micronutrient homeostasis, particularly involving pro- and anti-oxidative pathways, plays a crucial role in the disease process. These findings highlight the need for clinicians to evaluate trace element status in PCOS patients as part of a broader strategy to manage oxidative stress and inflammation. Moreover, the trace elements measured may serve as noninvasive biomarkers for disease severity or subtyping and could inform targeted interventions involving dietary or supplemental modulation. Future studies should investigate the dynamic interactions between trace elements, microbiota composition, and hormone regulation, as well as whether correcting these imbalances improves metabolic, reproductive, and microbiome outcomes in PCOS.

Somatic stem cells and their dysfunction in endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study explored the involvement of somatic and endometrial stem cells in the pathogenesis of endometriosis through a literature review. Key findings include the stem cell clonality in lesions, dysregulated behaviors contributing to disease progression, and genetic alterations. These insights could lead to improved diagnostic tests and targeted therapies, enhancing disease management and treatment strategies.

What was studied?

The study focused on the role of somatic stem cells (SSCs), including endometrial stem cells (EnSCs) and bone marrow-derived mesenchymal stem cells (MSCs), in the pathogenesis of endometriosis. This was conducted through a comprehensive literature review, including in vitro experiments and studies on animal models and human tissue analyses. The research examined the contributions of these stem cells to the development and growth of endometriosis, exploring their genetic, phenotypic, and functional characteristics as well as their involvement in the disease’s mechanisms.

 

Who was studied?

The study subjects included stem cells derived from human and animal endometrium and stem cells involved in endometriosis pathology sourced from menstrual blood and bone marrow. This research compiled evidence from various sources, including previously conducted experiments and clinical observations focusing on how these stem cells contribute to the formation and progression of endometriosis lesions in women affected by the disease and in relevant animal models.

 

What were the most important findings?

Clonality and Origin of Lesions: Evidence suggests that ovarian endometriotic cysts and peritoneal endometriotic lesions may arise from a clonal origin, indicating a possible stem cell basis for the disease.

Stem Cell Dysregulation: Stem cells, particularly EnSCs and MSCs, exhibit dysregulated behaviors in endometriosis, such as increased proliferation, invasiveness, and altered expression of markers that suggest a stem cell-like undifferentiated state.

Contribution to Lesion Growth: Bone marrow-derived stem cells, including MSCs and endothelial progenitor cells, were found to contribute to the pathogenesis and growth of endometriotic lesions by promoting angiogenesis and possibly by transdifferentiating into endometrial-like cells.

Molecular and Phenotypic Alterations: Stem/progenitor cells in endometriotic lesions display a range of genetic and epigenetic alterations and an increased expression of pro-angiogenic factors, which are crucial for lesion survival and growth.

 

What are the greatest implications of this study?

Diagnostic Advancements: The findings suggest that markers identified in the stem cells associated with endometriosis could potentially be used to develop non-invasive diagnostic tests with higher sensitivity and specificity, aiding in early disease detection.

Therapeutic Interventions: Targeting dysfunctional stem cells or their altered pathways offers a promising strategy for developing more effective treatments. This could include interventions that inhibit the recruitment and proliferation of these cells at ectopic sites or modify their angiogenic and inflammatory behaviors.

Understanding Disease Mechanisms: By integrating stem cell-based insights into the pathogenesis of endometriosis, this study supports a more comprehensive understanding of the disease’s etiology, which can lead to more targeted and effective clinical management strategies.

The emphasis on stem cell contributions to endometriosis not only opens up new avenues for research but also highlights potential therapeutic targets that could significantly improve the management and treatment of endometriosis, potentially improving the quality of life for affected women.

Systems pharmacology to investigate the interaction of berberine and other drugs in treating polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study explores the use of berberine in combination with other drugs for treating PCOS, highlighting its effects on insulin resistance, androgen excess, and metabolic dysfunction.

What was studied?

This study employed systems pharmacology to investigate the potential interaction of berberine with other drugs in treating Polycystic Ovary Syndrome (PCOS). Given that PCOS is a complex condition with various manifestations such as hormonal imbalance, insulin resistance, and metabolic dysfunction, the study aimed to explore the polypharmacological effects of berberine and its capacity to enhance the efficacy of existing clinical drugs. The authors used bioinformatics tools to identify candidate targets related to PCOS and map out the biological pathways involved. The goal was to understand how berberine interacts with these targets and how its combination with other drugs might improve therapeutic outcomes for PCOS patients.

Who was studied?

This study did not directly involve human participants, but instead focused on computational analysis and molecular simulations. The study examined the interaction of berberine with known clinical drugs that are commonly used to treat PCOS, such as combined oral contraceptives, antiandrogens, insulin-sensitizing drugs, and others. Using systems pharmacology approaches, the study identified and validated key targets for PCOS and simulated how these targets interacted with berberine and other medications. The analysis relied on protein interaction networks, molecular docking, and drug-target network construction to predict possible therapeutic effects.

What were the most important findings?

The study identified several critical biological pathways and targets related to PCOS, including the insulin signaling pathway, adipocytokine signaling, and androgen biosynthesis. Berberine was found to interact with key targets such as the androgen receptor (AR), estrogen receptor (ESR1), progesterone receptor (PGR), and insulin receptor (INSR), which are all pivotal in managing PCOS symptoms. The analysis also revealed that berberine could enhance the effects of existing drugs by acting on multiple targets within these pathways. For example, berberine can suppress androgen levels by interacting with AR and PGR, reduce insulin resistance by targeting INSR, and modulate lipid metabolism through its effects on the glucocorticoid receptor and other targets.

Molecular docking simulations confirmed that berberine had strong binding affinities for these targets, with similar binding energies to clinical drugs like cyproterone acetate and metformin. The study further suggested that berberine might help reduce the side effects of conventional therapies by competing for the same receptor sites, thus mitigating adverse drug reactions over long-term treatment. Additionally, berberine's ability to act on multiple targets simultaneously positions it as a promising polypharmacological agent in PCOS management.

What are the greatest implications of this study?

The findings from this study have significant clinical implications, particularly for the treatment of PCOS. The ability of berberine to interact with multiple molecular targets involved in PCOS pathophysiology suggests that it could be an effective adjunctive therapy. By enhancing the effectiveness of other clinical drugs and potentially reducing side effects, berberine presents a viable treatment option for patients with PCOS who require comprehensive care for their metabolic and hormonal imbalances. The study highlights the potential of systems pharmacology and drug repurposing in developing novel, cost-effective treatments for complex disorders like PCOS. Future clinical trials are needed to validate the therapeutic benefits of berberine in combination with other drugs, with a focus on optimizing treatment regimens to address the various facets of PCOS.

The bidirectional relationship between endometriosis and microbiome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review highlights the bidirectional relationship between endometriosis and the microbiome, showcasing dysbiosis as a key factor in inflammation and estrogen metabolism. Emerging microbiome-targeted therapies hold promise for diagnosis and treatment.

What Was Reviewed?

The review article explored the bidirectional relationship between endometriosis and the microbiome, emphasizing the role of dysbiosis in the pathogenesis and progression of this chronic inflammatory condition. It discussed microbiome alterations across different sites, including the gut, peritoneal fluid, and female reproductive tract, and evaluated how these microbial shifts influence inflammation, immune modulation, and estrogen metabolism. Furthermore, it highlighted experimental and clinical evidence supporting the potential of microbiome-targeted interventions as both diagnostic tools and treatments for endometriosis.

Who Was Reviewed?

The review synthesized findings from human and animal studies investigating microbiota composition in patients with endometriosis compared to healthy controls. It included a comprehensive analysis of bacterial, viral, and fungal associations across diverse microbiome sites, focusing on patterns of dysbiosis, enriched taxa, and diminished microbial diversity. Specific populations reviewed included women diagnosed with various stages of endometriosis and animal models with surgically induced disease.

What Were the Most Important Findings?

The most notable findings included alterations in gut, cervical, and peritoneal fluid microbiota in women with endometriosis. In the gut, elevated levels of Proteobacteria and reduced Lactobacillaceae were observed. The peritoneal fluid showed enrichment of Acinetobacter and Pseudomonas, while the cervical and vaginal microbiomes exhibited decreased diversity and increased abundance of pathogenic species from the Gardnerella and Streptococcus genus. Dysbiosis was associated with heightened inflammatory responses mediated by lipopolysaccharide (LPS) from Escherichia coli, potentially driving lesion formation through the NF-κB pathway. The concept of “estrobolomes,” gut bacteria influencing estrogen reabsorption, was linked to the hyperestrogenic state characteristic of endometriosis. Notably, antibiotic and probiotic treatments in animal models reduced lesion size, supporting the potential therapeutic role of microbiome modulation.

What Are the Greatest Implications?

The implications of this review are twofold: first, the microbiome holds promise as a non-invasive diagnostic tool for endometriosis, potentially reducing diagnostic delays. Second, microbiome-targeted interventions (MBTIs), such as probiotics, prebiotics, and dietary modifications, may offer novel therapeutic avenues. The findings underscore the necessity for further research into microbiome signatures and their clinical applications, particularly in differentiating disease stages and addressing infertility associated with endometriosis.

The Comorbidity of Endometriosis and Systemic Lupus Erythematosus: A Systematic Review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This review explores the significant comorbidity between systemic lupus erythematosus (SLE) and endometriosis, emphasizing shared pathological pathways.

DOI: 10.7759/cureus.42362

What Was Reviewed?

This systematic review examined the comorbidity between endometriosis and systemic lupus erythematosus (SLE), two chronic conditions with significant implications for women's health. The review aimed to elucidate the prevalence, shared pathophysiological mechanisms, and risk factors linking these diseases, emphasizing immune dysregulation, genetic predispositions, and hormonal influences. The review synthesized findings from nine studies conducted between 2011 and 2021, including case-control, cohort, and systematic review methodologies.

Who Was Reviewed?

The review focused on studies of females aged 12-60, representing the pubertal to postmenopausal age range. The population comprised patients with diagnosed endometriosis and SLE. The studies predominantly included participants from diverse ethnicities and geographies, screened based on standardized inclusion criteria to establish the prevalence and interaction of these conditions.

What Were the Most Important Findings?

The review confirmed a statistically significant correlation between endometriosis and SLE, with women diagnosed with either condition at a heightened risk of developing the other. The findings implicated immune dysregulation, characterized by diminished cytotoxic T-cell activity and elevated humoral immune responses, as a central mechanism. Notable microbial associations include increased systemic inflammation mediated by cytokines such as interleukin-1, interleukin-6, and tumor necrosis factor (TNF-α). Genetic factors also played a role, with gene loci such as PTPN22 associated with increased susceptibility to both conditions. Surgical interventions like hysterectomy were linked to increased inflammation and subsequent autoimmune activation, while modified surgical techniques showed promise in mitigating risk.

What Are the Greatest Implications of This Review?

This review highlights the necessity for clinicians to adopt an interdisciplinary approach when managing patients with either endometriosis or SLE, as their comorbidity exacerbates disease burden and complicates treatment. It emphasizes the importance of targeted therapies to modulate immune response alongside careful evaluation of surgical and hormonal treatment strategies to minimize adverse outcomes. The findings suggest a potential for incorporating microbial and genetic markers into diagnostic and therapeutic protocols to improve outcomes.

The Comparative Effects of Myo-Inositol and Metformin Therapy on the Clinical and Biochemical Parameters of Women of Normal Weight Suffering from Polycystic Ovary Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study compared the effects of myo-inositol and metformin on insulin resistance, menstrual cycle regularity, and hyperandrogenism in women with PCOS. Both therapies were effective, with fewer side effects noted for myo-inositol.

What was studied?

This study investigated the comparative effects of myo-inositol (MI) and metformin (MET) therapy on clinical and biochemical parameters in women with polycystic ovary syndrome (PCOS). The research focused on evaluating the impact of both therapies on insulin resistance (IR), hyperandrogenism, menstrual cycle regulation, and various metabolic markers in PCOS patients with normal BMI. The objective was to determine which therapy is more effective in improving these parameters.

Who was studied?

The study included 80 women diagnosed with PCOS who had insulin resistance but a normal body mass index (BMI). These participants were randomly assigned to two treatment groups: one group received myo-inositol, while the other group received metformin. The study was designed as a randomized controlled trial and aimed to assess the efficacy of these two insulin-sensitizing therapies.

What were the most important findings?

The results indicated that both myo-inositol and metformin significantly reduced insulin resistance, with a marked decrease in the area under the curve (AUC) of insulin during an oral glucose tolerance test (OGTT) for both groups. Both treatments led to improvements in the regulation of menstrual cycles, with more than 90% of patients experiencing regular cycles. The therapies also resulted in a statistically significant reduction in androgenic hormones (such as testosterone and SHBG), which are critical for managing symptoms like hirsutism. The findings suggest that both myo-inositol and metformin are effective in addressing insulin resistance, menstrual irregularities, and hyperandrogenism in women with PCOS, especially those with normal weight.

From a microbiome perspective, insulin resistance and hormonal imbalances are known to influence gut microbiota composition. Studies have shown that insulin resistance can contribute to an imbalance in the gut microbiome, potentially promoting pro-inflammatory taxa. Moreover, treatments like myo-inositol and metformin may have indirect effects on microbiota, such as modulating gut inflammation or affecting microbial populations associated with metabolic health.

What are the implications of this study?

The study highlights the potential of both myo-inositol and metformin as first-line treatments for managing PCOS in women with normal BMI, specifically targeting insulin resistance and hyperandrogenism. The results suggest that both therapies can be effective in improving metabolic and endocrine outcomes in PCOS, but myo-inositol may offer the advantage of fewer gastrointestinal side effects compared to metformin. This makes myo-inositol a promising alternative, particularly for women who experience adverse effects with metformin. The study also emphasizes the importance of considering personalized treatment options for women with PCOS, as different responses may be observed based on individual phenotypes.

The copper chelator ammonium tetrathiomolybdate inhibits the progression of experimental endometriosis in TNFR1-deficient mice

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

Ammonium tetrathiomolybdate (TM) inhibits endometriosis progression in TNFR1-deficient mice by reducing copper and estradiol levels, lesion growth, angiogenesis, and oxidative stress.

What Was Studied?

This study evaluated the therapeutic potential of ammonium tetrathiomolybdate (TM), a copper chelator, in inhibiting the progression of experimental endometriosis (EDT) in TNFR1-deficient mice. It explored TM's effects on copper and estradiol concentrations, lesion development, angiogenesis, oxidative stress, and inflammatory pathways. The research aimed to address how TM mitigates EDT in a worsened state caused by TNFR1 deficiency, a condition characterized by reduced cell death and increased lesion proliferation.

Who Was Studied?

The subjects were TNFR1-deficient female C57BL/6 mice divided into three groups: sham-operated (KO Sham), EDT-induced (KO EDT), and EDT-induced with TM treatment (KO EDT+TM). EDT was induced via autologous uterine tissue transplantation into the intestinal mesentery, and TM was administered orally postoperatively. Experimental outcomes were evaluated one month after EDT induction.

Most Important Findings

The study revealed several critical findings. First, EDT induction significantly elevated copper and estradiol levels in the peritoneal fluid, both of which were restored to physiological levels with TM treatment. TM also reduced lesion volume and weight, decreased cell proliferation, and suppressed angiogenesis, as evidenced by lower blood vessel counts and reduced expression of Vegfa, Fgf2, and Pdgfb. Furthermore, TM altered oxidative stress markers, decreasing the activity of superoxide dismutase (SOD) and catalase (CAT) while increasing lipid peroxidation, suggesting a pro-oxidative environment conducive to apoptotic signaling.

From a microbiome perspective, copper's role as a metalloestrogen and its involvement in estradiol synthesis underline the relevance of copper chelation in addressing estrogen-dependent diseases like endometriosis. By reducing copper levels, TM may disrupt microbial contributions to oxidative stress and inflammation, though direct microbiome-specific findings were not explored.

Greatest Implications

The study's findings suggest TM's dual role in reducing pro-inflammatory and pro-angiogenic pathways while restoring copper and estradiol homeostasis. These mechanisms are vital for mitigating EDT progression, particularly in the context of TNFR1 deficiency, where pathological signaling is exacerbated. Clinically, TM represents a potential adjunct therapy for managing endometriosis, particularly in cases resistant to conventional hormone treatments. The findings also reinforce the broader therapeutic relevance of targeting trace metals like copper in inflammatory and estrogen-dependent conditions.

The effect of resistant dextrin as a prebiotic on metabolic parameters and androgen level in women with polycystic ovarian syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study demonstrates that resistant dextrin supplementation can improve metabolic and endocrine parameters in women with PCOS, offering a promising alternative to traditional therapies.

What Was Studied?

This study investigated the effects of resistant dextrin, a type of prebiotic, on metabolic parameters and androgen levels in women with polycystic ovary syndrome (PCOS). The goal was to determine if resistant dextrin could improve parameters such as lipid profiles, fasting blood glucose (FBS), high-sensitivity C-reactive protein (hsCRP), and levels of dehydroepiandrosterone sulfate (DHEA-S) and free testosterone, which are associated with androgen excess in PCOS. Additionally, the study explored the impact on clinical manifestations such as menstrual cycle irregularities and hirsutism.

Who Was Studied?

The study involved 62 women diagnosed with PCOS based on the Rotterdam criteria. These participants were randomly divided into a prebiotic group, which consumed 20 grams of resistant dextrin daily, and a placebo group, which consumed an equal amount of maltodextrin. Both groups were observed for three months. The participants were assessed for various metabolic and endocrine parameters, including lipid profiles, blood glucose, hsCRP, DHEA-S, free testosterone, and clinical signs such as hirsutism and menstrual cycle irregularities.

What Were the Most Important Findings?

The study found that, after three months, the prebiotic group experienced significant improvements in several metabolic and endocrine parameters. Specifically, resistant dextrin supplementation led to reductions in LDL-cholesterol, triglycerides, total cholesterol, fasting blood glucose, hsCRP, DHEA-S, and free testosterone. Additionally, HDL-cholesterol levels increased significantly in the prebiotic group compared to the placebo group. The prebiotic group also saw improvements in clinical manifestations of PCOS, including a reduction in the hirsutism score and more regular menstrual cycles.

From a microbiome perspective, the findings are particularly relevant. The consumption of resistant dextrin, a prebiotic fiber, is known to modulate the gut microbiota, potentially increasing beneficial bacteria like Bifidobacterium and Lactobacillus. These bacteria play a crucial role in regulating systemic inflammation and metabolic function, which may explain the improvements observed in this study. Furthermore, the increased production of short-chain fatty acids (SCFAs) due to prebiotic fermentation could enhance insulin sensitivity and reduce inflammation, which are key factors in PCOS pathology.

What Are the Greatest Implications of This Study?

This study underscores the potential of resistant dextrin as a therapeutic intervention for improving metabolic and hormonal imbalances in women with PCOS. Given the lack of effective treatments that address both metabolic and endocrine dysfunction in PCOS, the use of prebiotics offers a promising alternative to pharmacological treatments, which often come with side effects. By modulating the gut microbiota, prebiotics may not only improve lipid profiles and insulin resistance but also address clinical symptoms such as hirsutism and menstrual irregularity. This study suggests that dietary interventions using prebiotics like resistant dextrin could be integrated into the management of PCOS, offering a low-cost, side-effect-free alternative to more invasive treatments.

The effects of a low-carbohydrate, ketogenic diet on the polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

A 24-week ketogenic diet significantly reduced insulin resistance, free testosterone, and LH/FSH ratio in women with PCOS, improving fertility markers. The dietary intervention may influence gut microbiota, enhancing metabolic and hormonal outcomes.

What was studied?

This pilot study examined the effects of a low-carbohydrate, ketogenic diet (LCKD) on metabolic and endocrine parameters in overweight and obese women with polycystic ovary syndrome (PCOS). Given PCOS’s well-documented link with insulin resistance and hyperandrogenism, the study hypothesized that carbohydrate restriction would improve insulin sensitivity and hormonal imbalance. The intervention involved instructing participants to consume fewer than 20 grams of carbohydrates per day over 24 weeks. Researchers assessed changes in body weight, fasting insulin, free testosterone, LH/FSH ratio, and subjective PCOS symptoms.

Who was studied?

Eleven women aged 18 to 45 with clinically diagnosed PCOS and a BMI ≥27 kg/m² were enrolled from the Raleigh-Durham area in North Carolina. The final analysis included five participants who completed the 24-week study. All participants were instructed to follow a strict LCKD and attended biweekly follow-ups for adherence monitoring and biochemical assessments. The group was predominantly Caucasian, and participants reported histories of chronic anovulation or hyperandrogenemia.

What were the most important findings?

Among the five women who completed the study, the LCKD led to a significant mean weight reduction of 12%, with individual weight loss ranging from 4% to 16.4%. More notably, there were substantial metabolic and hormonal improvements: fasting serum insulin dropped by approximately 54%, the LH/FSH ratio fell by 36%, and percent free testosterone decreased by 30%. These findings strongly suggest enhanced insulin sensitivity and reduced ovarian androgen production. Two of the women, previously experiencing infertility, became pregnant during the study period.

From a microbiome perspective, this is noteworthy because a ketogenic diet is known to modulate gut microbial composition. Prior studies associate ketogenic diets with increased abundance of Akkermansia muciniphila and Bacteroides species, both of which are linked to improved metabolic profiles, including insulin sensitivity and gut barrier integrity. Although microbiota were not directly assessed in this study, the improvement in metabolic parameters is consistent with microbial shifts observed in similar dietary interventions. The reduction in insulin likely suppressed hyperinsulinemia-induced androgen synthesis and improved sex hormone-binding globulin (SHBG) levels, thereby reducing circulating free testosterone. This is particularly relevant for clinicians considering microbiome-modulating dietary strategies as adjunctive treatment in PCOS.

What are the greatest implications of this study?

The study underscores that a ketogenic diet may serve as an effective non-pharmacological approach to reduce insulin resistance and hyperandrogenism in women with PCOS. This could potentially translate into improved ovulatory function and fertility. The LCKD demonstrated significant endocrine normalization within six months, which is meaningful considering the limited curative options currently available for PCOS. Clinicians should consider LCKD as a viable dietary strategy, especially for PCOS patients struggling with infertility, elevated androgens, or metabolic dysfunctions. Moreover, the potential microbiome-mediated mechanisms add further value by pointing to gut health as an emerging therapeutic axis in PCOS management. While the small sample size and lack of a control group limit generalizability, the results justify larger trials examining diet–microbiome–hormone interactions in this population.

The effects of probiotics, prebiotics, and synbiotics on polycystic ovarian syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This overview confirms probiotics, particularly Lactobacillus and Bifidobacterium, improve insulin resistance, inflammation, and hormone balance in PCOS. Synbiotics showed less consistent effects.

What was reviewed?

This overview synthesized evidence from eight systematic reviews and meta-analyses investigating the effects of probiotics, prebiotics, and synbiotics on the management of polycystic ovarian syndrome (PCOS). The reviews collectively evaluated randomized controlled trials (RCTs) focusing on metabolic, hormonal, and inflammatory markers to clarify how microbiota-modulating interventions influence PCOS-related outcomes. The analysis spanned data from over 4,000 women with PCOS and assessed diverse probiotic strains, prebiotic compounds (like inulin and fructooligosaccharides), and their combinations (synbiotics), typically over 8–12 week periods.

Who was reviewed?

The population under review consisted of women with clinically diagnosed PCOS from multiple RCTs conducted predominantly in Iran and China. The trials included in the systematic reviews examined interventions using specific bacterial strains and dosages. Outcomes measured included anthropometrics (BMI, weight), glycemic indices (fasting glucose, insulin, HOMA-IR), lipid profiles, inflammatory biomarkers (CRP, hsCRP), oxidative stress markers (TAC, MDA), and hormone levels (testosterone, SHBG).

What were the most important findings?

The compiled reviews demonstrated that probiotic supplementation led to modest yet statistically significant reductions in fasting plasma glucose, fasting insulin, HOMA-IR, total cholesterol, triglycerides, VLDL-C, and BMI in women with PCOS. Some reviews noted increased levels of SHBG and improved insulin sensitivity indices such as QUICKI.

The microbial associations of clinical relevance include a restoration of beneficial bacteria often depleted in PCOS, such as Faecalibacterium prausnitzii, Bifidobacterium spp., and Lactobacillus spp.—species known for producing short-chain fatty acids (SCFAs) like butyrate. SCFAs modulate insulin sensitivity, reduce gut permeability, and regulate inflammatory responses. Furthermore, synbiotics had mixed effects; while some reviews reported improvements in glucose and lipid profiles, the benefits were generally less consistent or weaker than probiotics alone.

What are the implications of this review?

This review reinforces the emerging role of the gut microbiota in PCOS pathophysiology and highlights the therapeutic potential of microbiota-targeted interventions. Probiotics demonstrated the most consistent benefits across glycemic, inflammatory, and hormonal parameters. These findings support the integration of microbiome-based strategies, such as targeted probiotic supplementation, into clinical practice for PCOS management. Clinicians should be aware that although results are promising, there remains high heterogeneity among studies in terms of strains used, dosages, and intervention durations. Larger, standardized clinical trials are necessary to define optimal regimens. Still, this growing body of evidence supports a microbiome-informed approach to PCOS care, particularly for improving insulin sensitivity, lowering inflammation, and potentially reducing androgen excess.

The effects of synbiotic supplementation on hormonal status, biomarkers of inflammation and oxidative stress in subjects with polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This randomized trial evaluates the effects of synbiotic supplementation on hormonal, inflammatory, and metabolic parameters in women with PCOS. It shows significant improvements in androgen levels, insulin sensitivity, and inflammation.

What was studied?

This randomized, double-blind, placebo-controlled trial studied the effects of synbiotic supplementation on hormonal status, biomarkers of inflammation, and oxidative stress in women with polycystic ovary syndrome (PCOS). Specifically, the research aimed to evaluate the impact of synbiotics, comprising Lactobacillus acidophilus, Lactobacillus casei, Bifidobacterium bifidum, and inulin, on clinical and biochemical markers in women with PCOS over 12 weeks. The primary focus was on assessing changes in hormone levels such as sex hormone-binding globulin (SHBG), free androgen index (FAI), modified Ferriman-Gallwey (mFG) score (for hirsutism), high-sensitivity C-reactive protein (hs-CRP), nitric oxide (NO) levels, and insulin resistance.

Who was studied?

The study involved 60 women diagnosed with PCOS according to the Rotterdam criteria. These women were randomly assigned to receive either synbiotics (n=30) or a placebo (n=30) for 12 weeks. The participants were screened for exclusion factors, including smoking, pregnancy, thyroid disorders, gastrointestinal problems, and the use of probiotics or synbiotics prior to the study. Clinical assessments and biochemical evaluations were performed before and after the intervention to measure the effects of synbiotic supplementation on the hormonal, inflammatory, and oxidative parameters of PCOS.

What were the most important findings?

The most significant findings of this study were that synbiotic supplementation led to notable improvements in several parameters associated with PCOS. Specifically, the synbiotic group showed a significant increase in SHBG and a decrease in the free androgen index (FAI), indicating a reduction in hyperandrogenism. There was also a significant reduction in mFG scores (indicating a reduction in hirsutism), and serum hs-CRP levels, which are associated with inflammation, were significantly reduced. Additionally, plasma NO levels were significantly increased in the synbiotic group, suggesting improved endothelial function. The synbiotic supplementation also resulted in a significant reduction in insulin levels and the HOMA-IR index, indicating improved insulin sensitivity. However, no significant changes were observed in other biomarkers of oxidative stress (such as total antioxidant capacity (TAC), glutathione (GSH), and malondialdehyde (MDA)).

From a microbiome perspective, the synbiotic intervention likely improved gut health and reduced inflammation, which is crucial in managing the systemic effects of PCOS. The improvement in metabolic and hormonal parameters suggests that synbiotics can restore balance in the gut microbiota, reduce systemic inflammation, and improve insulin resistance, all of which are key contributors to PCOS pathophysiology.

What are the greatest implications of this study?

The results of this study have significant clinical implications for the management of PCOS. The use of synbiotics as a treatment offers a promising non-pharmacological intervention for managing PCOS-related metabolic and hormonal disturbances. By improving insulin sensitivity, reducing inflammation, and balancing androgen levels, synbiotic supplementation may help alleviate symptoms like hirsutism and irregular menstruation. Moreover, this study supports the growing body of evidence linking gut microbiota modulation to endocrine health, suggesting that gut-targeted therapies could play a pivotal role in the treatment of PCOS. Clinically, synbiotics could serve as an adjunct to other PCOS treatments, offering a safer, more sustainable solution with fewer side effects compared to traditional medications. However, further studies with larger sample sizes and longer durations are needed to confirm these findings and establish long-term efficacy.

The Endobiota Study: Comparison of Vaginal, Cervical and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies microbiome shifts in vaginal, cervical, and gut sites in stage 3/4 endometriosis. Absence of Atopobium and elevated Gardnerella suggest immune dysregulation, while Escherichia/Shigella dominance in stool correlates with bowel involvement, indicating potential diagnostic biomarkers.

What Was Studied?

This study, titled "The Endobiota Study: Comparison of Vaginal, Cervical, and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls," aimed to evaluate the differences in microbial composition across the vaginal, cervical, and gut microbiomes in women with advanced-stage (3/4) endometriosis compared to healthy controls. Researchers collected and analyzed samples from three anatomical sites—vaginal swabs, cervical swabs, and stool—using 16S rRNA sequencing to determine the diversity and abundance of bacterial genera. The primary objective was to identify specific microbial signatures and dysbiosis patterns associated with advanced endometriosis.

Who Was Studied?

The study included 28 Caucasian women, 14 diagnosed with histologically confirmed stage 3/4 endometriosis and 14 healthy controls. All participants were of reproductive age, with similar age and BMI distributions between groups. Vaginal, cervical, and stool samples were collected from each participant under sterile conditions to prevent contamination. The endometriosis patients were all confirmed to have deep infiltrating endometriosis with extensive lesions, while the control group consisted of asymptomatic women with no clinical or ultrasound evidence of endometriosis.

What Were the Most Important Findings?

The study uncovered notable dysbiosis in the microbiota composition of women with advanced endometriosis compared to healthy controls. In vaginal samples, Gemella and Atopobium were completely absent in the endometriosis group, suggesting a protective role in healthy women. Cervical samples showed a complete loss of Atopobium and Sneathia in endometriosis patients, while Alloprevotella was significantly elevated. This microbial shift in the cervical microbiota is particularly significant given Atopobium's known associations with maintaining vaginal health. In stool samples, Sneathia, Barnesella, and Gardnerella were significantly decreased in endometriosis patients, while Escherichia/Shigella dominance was observed in two women who subsequently required segmental colon resection for severe bowel involvement. Sensitivity analyses excluding Lactobacillus revealed that Gardnerella represented a significantly higher proportion of the remaining microbiota in the vaginal and cervical niches of the endometriosis group compared to controls (72.9% vs. 36.8% in the vagina and 67.7% vs. 36.8% in the cervix, respectively). Furthermore, Escherichia/Shigella, Streptococcus, and Ureaplasma were markedly elevated, while Prevotella, Dialister, and Megasphaera were significantly reduced. These microbial changes suggest an altered immune response and heightened inflammatory state in women with advanced endometriosis, highlighting potential microbial markers of disease progression.

Anatomical SiteMicrobiota Findings in Advanced Endometriosis Patients
Vaginal SamplesGemella and Atopobium completely absent. Gardnerella significantly elevated (72.9% of microbiota, excluding Lactobacillus).
Cervical SamplesComplete loss of Atopobium and Sneathia. Marked increase in Alloprevotella. Gardnerella elevated (67.7% of microbiota, excluding Lactobacillus).
Stool SamplesSignificant decreases in Sneathia, Barnesella, and Gardnerella. Dominance of Escherichia/Shigella observed in two patients requiring bowel resection.
Additional Microbial ShiftsMarked elevation of Escherichia/Shigella, Streptococcus, and Ureaplasma. Reductions in Prevotella, Dialister, and Megasphaera.
Inflammatory AssociationsDysbiosis patterns suggest an altered immune response and heightened inflammatory state in advanced endometriosis.

The Female Vaginal Microbiome in Health and Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review explores vaginal microbiome in health and Bacterial Vaginosis (BV) highlighting the shift from Lactobacillus-dominance to a dysbiotic state, as well as diagnostic advances and new treatments like probiotics and vaginal microbiota transplantation (VMT).

What was reviewed?

The Female Vaginal Microbiome in Health and Bacterial Vaginosis review article comprehensively examines the vaginal microbiome, focusing on its role in health and disease, specifically bacterial vaginosis (BV). The review covered the characteristics of the healthy vaginal microbiome, the alterations associated with BV, the relationship between BV and various diseases, and the current diagnostic and therapeutic strategies for BV.

Who was reviewed?

The review included extensive analysis of studies and research conducted by various scientists and researchers in the field of microbiology, gynecology, and infectious diseases. Notable contributors to the field, such as Baolei Jia, Eva Raphael, Werner Mendling, and Elena Shipitsyna, provided editorial oversight and peer reviews, ensuring the comprehensive nature of the review. The article also referenced significant studies and findings from researchers like Ravel, Fettweis, Fredricks, and many others who have contributed to the understanding of the vaginal microbiome and BV.

What were the most important findings of this review?

Microbial Composition: The vaginal microbiome in healthy women is dominated by Lactobacillus species, which help maintain an acidic pH and protect against pathogens.

BV Characteristics: BV is marked by a significant reduction in Lactobacillus species and an overgrowth of anaerobic bacteria such as Gardnerella vaginalis, Atopobium vaginae, and Prevotella species.

Disease Association: BV is associated with several adverse reproductive outcomes, including increased susceptibility to sexually transmitted infections (STIs), pelvic inflammatory disease (PID), and adverse obstetric outcomes such as preterm birth.

Diagnosis and Treatment: Traditional diagnostic methods like Amsel criteria and Nugent scoring are supplemented by newer molecular techniques that offer higher accuracy. Treatment primarily involves antibiotics, but recurrence is common due to the persistence of biofilms and antibiotic resistance.

Recent Advances: New diagnostic and therapeutic approaches, including high-throughput sequencing, multi-omic techniques, and microbial-based therapies such as probiotics and vaginal microbiota transplantation (VMT), show promise in better managing BV.

What are the greatest implications of this review?

Enhanced Understanding: The Female Vaginal Microbiome in Health and Bacterial Vaginosis review underscores the complex dynamics of the vaginal microbiome and its critical role in maintaining vaginal health. Understanding these dynamics is crucial for developing targeted interventions.

Improved Diagnostics: The identification of specific biomarkers and the use of advanced molecular techniques can lead to more accurate and timely diagnosis of BV, reducing misdiagnosis and inappropriate treatment.

Therapeutic Innovation: Highlighting the limitations of current antibiotic treatments, the review points towards innovative therapies, including biofilm-disrupting agents and microbial-based treatments, which could offer more sustainable and effective solutions.

Public Health Impact: By linking BV with serious reproductive health issues and increased risk of STIs, the review emphasizes the need for public health initiatives to address BV, which could significantly reduce the burden of these associated conditions.

Research Directions: The review calls for further research into the interactions between the vaginal microbiota and host immune responses, which could reveal new therapeutic targets and strategies for maintaining vaginal health and preventing dysbiosis.

The Gut Microbiome in Polycystic Ovary Syndrome and Its Association with Metabolic Traits

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study links the gut microbiome in PCOS to metabolic traits, identifying microbial shifts tied to insulin resistance and prediabetes. Specific taxa like Ruminococcaceae UCG-002 and Dorea correlate with hormonal and glucose metabolism markers, highlighting the gut microbiome’s role in PCOS-related metabolic health.

What was studied?

This study investigated the association between the gut microbiome and polycystic ovary syndrome (PCOS), particularly its metabolic traits, in women approaching the end of their reproductive years. Using a population-based cohort from the Northern Finland Birth Cohort 1966 (NFBC1966), the research team analyzed fecal microbiome profiles via 16S rRNA sequencing and correlated the bacterial composition with clinical, hormonal, and metabolic characteristics, including markers of glucose tolerance and insulin sensitivity. The core objective was to determine whether microbial composition or diversity could distinguish PCOS cases from controls and whether specific taxa were linked to features like insulin resistance and prediabetes, which are commonly observed in PCOS.

Who was studied?

The study enrolled 303 women, including 102 women diagnosed with PCOS and 201 age- and BMI-matched healthy controls, all drawn from the NFBC1966 longitudinal study. These participants provided fecal samples for microbiome analysis and underwent clinical examinations, including hormonal assays, oral glucose tolerance tests (OGTT), and metabolic profiling. The diagnostic criteria for PCOS were based on self-reported oligomenorrhea, hirsutism, or a medical diagnosis of PCOS or polycystic ovaries (PCOs) at ages 31 and 46, respectively. The cohort was ethnically homogenous and geographically stable, which reduced external confounding factors such as diet, genetics, and socioeconomic variability.

What were the most important findings?

The study found that PCOS itself did not significantly alter overall gut microbiome diversity or composition when comparing PCOS women to matched controls. Alpha diversity, which measures the richness and evenness of microbial species, and beta diversity, which compares the community structure between groups, were not significantly different between the two groups. However, significant associations were uncovered between specific microbial taxa and PCOS-related metabolic traits.

Notably, the genus Ruminococcaceae was positively correlated with favorable metabolic markers, including higher sex hormone-binding globulin (SHBG) levels and improved insulin sensitivity (measured via the Matsuda and Disposition indices). The Clostridiales Family XIII AD3011 group also displayed positive correlations with metabolic health indicators and negative correlations with markers of glucose dysregulation, including glycated hemoglobin (HbA1c) and BMI. Furthermore, within the PCOS group, women with prediabetes had significantly reduced microbial diversity and elevated levels of the genus Dorea compared to PCOS women with normal glucose tolerance. This genus has been previously linked to both metabolic dysfunction and increased blood glucose, reinforcing its potential as a microbial biomarker for insulin resistance and prediabetes in PCOS.

Although the overall gut microbiome profile did not drastically differ between PCOS and non-PCOS women, the identified microbial shifts correlated strongly with metabolic impairments common in PCOS, suggesting that metabolic status, rather than PCOS per se, may drive microbiome alterations.

What are the greatest implications of this study?

This study emphasizes the critical role of metabolic health in shaping the gut microbiome profile of women with PCOS rather than PCOS diagnosis alone. The identification of taxa like Ruminococcaceae and Dorea as being closely linked to insulin sensitivity and glucose metabolism offers promising microbial signatures for future diagnostic and therapeutic development. Clinicians managing PCOS should consider metabolic health as a key modulator of gut microbiome composition, especially when evaluating or planning interventions aimed at microbiome modulation.

The findings also point toward the potential utility of microbial markers to predict metabolic complications in PCOS patients, particularly the risk of developing prediabetes or type 2 diabetes. This aligns with a growing understanding of the gut-liver-metabolism axis and highlights microbiome-based diagnostics and interventions as a future component of personalized care in PCOS management.

The gut microbiota and endometriosis: From pathogenesis to diagnosis and treatment

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

The review explored the connection between gut microbiota and endometriosis, highlighting potential influences on disease mechanisms through hormonal, immune, and inflammatory pathways. It suggests that modulating gut microbiota could lead to innovative diagnostic and therapeutic approaches for endometriosis. This review further suggests that via Fecal Microbiota Transplantation (FMT) may provide a novel therapeutic approach for the clinical treatment of endometriosis.

What was reviewed?

The study reviewed the relationship between the gut microbiota and endometriosis, focusing on how the gut microbiota may influence the pathogenesis, diagnosis, and potential treatment of endometriosis through various mechanisms such as estrogen modulation, immune response, and inflammation.

 

Who was reviewed?

The review did not involve specific individuals as subjects of study but instead synthesized findings from various studies that investigate the gut microbiota’s involvement in patients with endometriosis. It includes analysis of microbial profiles and their correlations with the disease.

 

What were the most important findings?

Important findings highlighted that alterations in the gut microbiota are associated with endometriosis and could potentially influence the disease’s pathogenesis through mechanisms linked to hormonal balance, immune modulation, and inflammatory responses. Specifically, changes in bacterial diversity and specific bacterial groups (such as an increase in the Firmicutes/Bacteroidetes ratio) were noted in patients with endometriosis.

 

What are the greatest implications of this review?

The review suggests that targeting the gut microbiota might offer new strategies for the diagnosis and treatment of endometriosis. Understanding the role of the gut microbiota in endometriosis could lead to non-invasive diagnostic biomarkers and novel therapeutic approaches that involve modulating the gut microbiota through diet, probiotics, or even fecal microbiota transplantation.

The Human Microbiome during Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explores bacterial vaginosis, focusing on microbial imbalances, immune responses, and diagnostic challenges. It highlights the need for microbiome-based treatments and improved diagnostics to reduce recurrence and improve BV management.

What Was Reviewed?

This review examines the human microbiome during bacterial vaginosis (BV), focusing on microbial shifts, host immune responses, and diagnostic challenges. It evaluates BV as a polymicrobial condition rather than an infection caused by a single pathogen. The review highlights how microbial imbalances contribute to BV symptoms, persistence, and recurrence. Additionally, it explores epidemiological factors, diagnostic methods, and host-microbiome interactions that influence BV progression and treatment response.

Who Was Reviewed?

The review synthesizes findings from studies involving women diagnosed with BV, including those experiencing recurrent infections. It incorporates data from molecular sequencing studies and microbiological research to assess the composition of the vaginal microbiome during BV. Additionally, it examines host immune responses to BV-related microbial changes and evaluates the link between BV and increased susceptibility to sexually transmitted infections (STIs) and pregnancy complications such as preterm birth.

Most Important Findings

BV disrupts the vaginal microbiome by reducing Lactobacillus species and increasing anaerobic bacteria such as Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., Mobiluncus spp., and Sneathia spp. Unlike infections that trigger a strong inflammatory response, BV presents as a microbial imbalance rather than an acute immune reaction. The review also highlights how bacterial biofilms protect BV-associated bacteria from antibiotic treatment, contributing to high recurrence rates.

The study also discusses BV’s complex interaction with the host immune system. BV-associated bacteria produce virulence factors that degrade the vaginal mucosal barrier, leading to increased inflammatory markers such as interleukin-1β (IL-1β) and tumor necrosis factor-alpha (TNF-α). This immune dysregulation may explain BV’s association with increased STI susceptibility and adverse pregnancy outcomes.

Diagnosing BV remains challenging due to inconsistencies in clinical and laboratory criteria. Amsel’s clinical criteria and Nugent scoring, which rely on symptom assessment and Gram staining, remain the primary diagnostic tools. However, these methods fail to account for BV’s diverse microbial community, leading to inconsistencies in diagnosing recurrent and persistent cases. The review emphasizes the need for molecular sequencing-based diagnostics that provide a more precise understanding of BV-associated bacterial communities.

Implications of the Review

BV presents a significant clinical challenge due to its high recurrence rate, treatment limitations, and association with reproductive health complications. The review underscores the importance of shifting towards microbiome-targeted therapies rather than relying solely on broad-spectrum antibiotics. Future research should focus on developing treatments that restore Lactobacillus-dominant vaginal microbiota and prevent biofilm formation. Refining molecular diagnostic techniques will help clinicians identify BV-associated bacterial communities more accurately, improving treatment strategies and reducing recurrence.

This review highlights the urgent need for improved diagnostic criteria, personalized treatment approaches, and a deeper understanding of the vaginal microbiome’s role in BV persistence. Advancing these areas of research will help clinicians develop more effective, long-term solutions for BV management.

The impact of endometriosis on dietary choices and activities of everyday life: a cross-sectional study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review highlights how gluten-free, Mediterranean, and anti-inflammatory diets improve pain perception in endometriosis by reducing inflammation and modulating the gut microbiome. Probiotics and bioactive nutrients such as curcumin enhance therapeutic outcomes.

What was reviewed?

This systematic review evaluated the impact of dietary interventions on pain perception in women diagnosed with endometriosis. It explored the connections between dietary changes and the alleviation of symptoms, particularly chronic pain, and assessed the potential of specific dietary patterns and nutrients to influence disease progression and symptom severity. The review included evidence from various studies highlighting the role of diets such as gluten-free, Mediterranean, and anti-inflammatory diets, along with the incorporation of specific nutrients and probiotics.

Who was reviewed?

The review included studies examining women diagnosed with endometriosis, focusing on their dietary habits, pain management strategies, and overall quality of life. The population spanned diverse stages of endometriosis and varying symptom severities, with dietary interventions as a common self-management approach.

What were the most important findings?

The review identified several key dietary patterns and nutrients that positively influenced pain perception and symptom management in women with endometriosis. A gluten-free diet was associated with symptom relief in patients experiencing gastrointestinal-related pain, while the Mediterranean diet showed benefits in reducing inflammation and pain severity due to its high content of antioxidants, omega-3 fatty acids, and polyphenols. Anti-inflammatory diets also gained traction, particularly in severe cases of endometriosis, where eliminating saturated fats and processed meats improved symptom management.

Major microbial associations (MMAs) of endometriosis were also highlighted, particularly the role of probiotics like Lactobacillus in alleviating pain and potentially modulating the gut microbiome to reduce systemic inflammation. The findings emphasize the therapeutic potential of dietary supplements such as curcumin, resveratrol, and quercetin, which possess anti-inflammatory and antioxidant properties.

What are the greatest implications of this review?

The findings suggest that dietary interventions and supplements can serve as non-invasive and complementary strategies for managing endometriosis-related symptoms, particularly chronic pain. By modulating systemic inflammation and influencing the gut microbiome, specific dietary patterns and nutrients may provide a tailored approach to alleviating symptoms. The review underscores the importance of integrating nutritional guidance into endometriosis management protocols.

The Implication of Mechanistic Approaches and the Role of the Microbiome in Polycystic Ovary Syndrome (PCOS)

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review connects gut and vaginal microbiome dysbiosis with PCOS-related inflammation, metabolic imbalance, and hormone disruption, highlighting bile acids and SCFAs as key mediators and therapeutic targets.

What was reviewed?

This review explored the mechanistic underpinnings of polycystic ovary syndrome (PCOS) with a particular focus on the role of the microbiome and associated metabolomic changes. The authors consolidated findings from diverse studies involving both human and animal models to highlight how gut and vaginal microbiota dysbiosis contributes to the metabolic, reproductive, and inflammatory manifestations of PCOS. The review emphasized microbial-mediated alterations in bile acids, short-chain fatty acids (SCFAs), ceramides, and trimethylamine N-oxide (TMAO), discussing how these metabolites affect insulin resistance (IR), hormonal regulation, and immune function.

Who was reviewed?

This review drew on a broad spectrum of human clinical studies, animal model research, and in vitro analyses. Studies included comparisons between women with PCOS and healthy controls, often stratified by phenotype, body mass index, insulin sensitivity, or reproductive status. Additionally, some studies involved germ-free or antibiotic-treated animal models to evaluate the causal role of gut microbiota in PCOS-like phenotypes. The review also incorporated data on vaginal microbiota differences in PCOS patients, comparing their microbial communities to those of healthy women to assess potential impacts on fertility and inflammation.

What were the most important findings?

The review identified that women with PCOS consistently exhibit gut microbiome dysbiosis, including reduced levels of Lactobacillus and Bifidobacterium and increased Escherichia, Shigella, Bacteroides vulgatus, and Prevotella. These microbial shifts disrupt bile acid metabolism, especially lowering GDCA and TUDCA, and impair IL-22 signaling, contributing to inflammation and insulin resistance. Decreased short-chain fatty acid (SCFA) production further weakens gut barrier integrity and affects gut hormone levels such as PYY and GLP-1, exacerbating endocrine imbalance.

Importantly, vaginal microbiota dysbiosis, characterized by increased Streptococcus, Gardnerella, Chlamydia, and Mycoplasma, and decreased Lactobacillus, was noted in PCOS, further implicating local immune disturbances and poor reproductive outcomes. The authors highlighted that the microbiome modulates IL-6, IL-10, IL-18, TNF-α, and CRP levels, linking microbial shifts to chronic low-grade inflammation, a hallmark of PCOS.

What are the greatest implications of this review?

This review makes a compelling case for recognizing gut and vaginal microbiota as central players in the pathophysiology of PCOS. Mapping the complex interactions between microbiome composition, immune signaling, metabolic hormones, and reproductive dysfunction provides a robust framework for considering microbiota-targeted therapies. The findings suggest that manipulating gut flora through probiotics, dietary fiber, or even fecal microbiota transplantation (FMT) could mitigate insulin resistance, reduce inflammation, and restore hormonal balance. Clinically, this highlights the potential for integrating microbiome assessments into PCOS diagnosis and personalized management. Moreover, the inclusion of bile acids and SCFAs as biomarkers or therapeutic targets could revolutionize PCOS treatment strategies by addressing metabolic and endocrine dysfunction at their microbial roots.

The Influence of Lactoferrin in Plasma and Peritoneal Fluid on Iron Metabolism in Women with Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study demonstrates that peritoneal fluid-to-plasma ferritin and lactoferrin ratios distinguish endometriosis stage and severity. Iron overload and shifting iron-binding protein profiles reveal a localized dysregulation that may influence disease progression and potentially pathogenic microbiome selection.

What was studied?

This study investigated the role of lactoferrin (LF) in relation to iron metabolism in women with and without endometriosis by measuring levels of LF, ferritin (FT), transferrin (TF), and iron (Fe) simultaneously in plasma and peritoneal fluid. The authors specifically explored whether the concentrations and ratios of these iron-related proteins in the two biological compartments could distinguish the presence and progression of endometriosis. The goal was to identify noninvasive or minimally invasive biomarkers that may aid in diagnosing or staging the disease based on iron metabolism, especially given endometriosis’ pro-inflammatory, iron-rich microenvironment.

Who was studied?

The study cohort included 90 women of reproductive age undergoing diagnostic laparoscopy, of whom 57 had histologically confirmed endometriosis (stages I–IV) and 33 did not. Plasma and peritoneal fluid samples were collected pre- and intra-operatively. Subjects were classified based on endometriosis diagnosis and stage, and specimens were evaluated for levels of LF, FT, TF, and Fe using ELISA, immunoturbidimetric assay, and colorimetric methods.

What were the most important findings?

Key findings highlight that ferritin and iron concentrations were significantly elevated in peritoneal fluid compared to plasma, especially in patients with advanced-stage endometriosis. In contrast, transferrin was consistently lower in peritoneal fluid. Notably, lactoferrin levels did not significantly differ between women with and without endometriosis when evaluated independently in plasma or peritoneal fluid, but the peritoneal fluid/plasma lactoferrin ratio decreased progressively with increasing disease severity, significantly distinguishing stage I from stage IV. The ferritin ratio was markedly higher in the endometriosis group, underscoring its potential as a disease marker. Correlation analyses revealed that in severe endometriosis, lactoferrin was significantly associated with ferritin and iron in the peritoneal fluid, suggesting a disrupted iron regulation mechanism localized to the disease microenvironment. Importantly, the elevated ferritin concentrations in peritoneal fluid may serve a compensatory, protective role to sequester iron and mitigate oxidative stress, while lactoferrin may lose this protective function as disease progresses.

From a microbiome perspective, this study underscores the iron-dependent ecological shifts that may select for siderophilic pathobionts. The iron overload and pro-oxidative milieu likely fosters the expansion of iron-requiring microbial taxa, potentially including Escherichia, Enterobacter, and Fusobacterium, known to be enriched in some endometriosis microbiome signatures. While microbial profiling was not performed, the metallomic dysregulation described supports the hypothesis that iron availability is a crucial factor in shaping pathogenic microbial communities in endometriosis.

What are the greatest implications of this study?

This study provides compelling evidence that iron-binding proteins—particularly ferritin and lactoferrin—play a localized and differential role in the progression of endometriosis. The findings suggest that peritoneal fluid iron metabolism, and especially the ferritin-to-lactoferrin balance, may be a critical axis of disease progression and potentially a therapeutic target. The study introduces the peritoneal fluid/plasma concentration ratio as a novel diagnostic parameter, offering a more granular assessment than conventional plasma markers. The declining lactoferrin ratio and increasing ferritin ratio with disease severity may signal a transition from iron sequestration and immune modulation toward iron-driven oxidative stress and tissue damage. This may serve as a foundation for the development of metallome-targeted diagnostics and therapies, including exogenous lactoferrin supplementation, which the authors suggest could restore iron balance in advanced disease stages. These findings also have implications for understanding how iron dysregulation may foster microbial dysbiosis, providing a mechanistic link between host iron metabolism and the pathophysiological selection of microbial communities in endometriosis.

The interplay between microbiota, metabolites, immunity during BV

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explains how microbial shifts, metabolite production, and immune responses interact in bacterial vaginosis. It highlights the roles of Gardnerella, Atopobium, and other anaerobes in disrupting vaginal health and discusses how their metabolic byproducts and immune modulation drive BV symptoms and persistence.

What was Reviewed?

This review explored the complex interplay between the vaginal microbiota, metabolite production, and local immune responses in the pathogenesis of bacterial vaginosis (BV). The authors synthesized existing research on how shifts in the vaginal microbiome from Lactobacillus-dominant communities to polymicrobial anaerobic communities contribute to BV development, symptoms, and recurrence. The review particularly emphasized the combined role of microbiota composition, bacterial metabolic products, and vaginal immune responses in driving clinical outcomes and disease persistence.

Who was Reviewed?

The review covered a wide body of research focusing on reproductive-age women diagnosed with or at risk for BV. It drew from studies examining the vaginal microbiota, including key bacteria such as Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., Mobiluncus spp., and Sneathia sanguinegens, as well as the protective Lactobacillus species like L. crispatus and L. iners. It also reviewed studies on the vaginal metabolome and immune responses in BV-positive and BV-negative women.

Most Important Findings

The review consolidated evidence that BV is a multifactorial condition characterized by dysbiosis of the vaginal microbiota, metabolic disruption, and altered immune responses. It described how healthy vaginal microbiomes are dominated by Lactobacillus species, particularly L. crispatus, which maintain vaginal acidity and protect against pathogens. In contrast, BV involves a shift toward a polymicrobial anaerobic community, with increased abundance of Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., Mobiluncus spp., and Sneathia spp., collectively referred to as major microbial associations (MMA) of BV.

These BV-associated bacteria produce key metabolites, including short-chain fatty acids (SCFAs) like acetate and propionate, and volatile amines like putrescine and cadaverine, which raise vaginal pH and produce the characteristic fishy odor of BV. Additionally, BV-associated biofilm formation, particularly involving G. vaginalis and A. vaginae, enhances bacterial persistence and resistance to treatment.

The review highlighted that these microbial and metabolic changes interact with the host’s immune system. BV patients exhibit elevated pro-inflammatory cytokines, especially IL-1β, without a corresponding increase in neutrophil recruitment. This unique immune profile likely results from SCFA-mediated suppression of neutrophil chemotaxis and explains why BV lacks overt inflammatory symptoms despite microbial overgrowth.

Implications of this Review

This review emphasizes the need to redefine BV beyond a simple microbial imbalance. It emphasizes that the metabolic products of BV-associated bacteria and their impact on host immunity are central to disease progression and recurrence. Clinicians should consider not only microbial community shifts but also metabolite profiles and immune responses when diagnosing and managing BV. The review calls for the integration of multi-omic data, microbiome, metabolome, and immunome, to develop more accurate diagnostics and targeted therapeutic strategies. Understanding these interactions may guide the design of personalized interventions to restore microbial and metabolic homeostasis, reduce BV recurrence, and mitigate associated reproductive health risks.

The oral microbiome and breast cancer and non-malignant breast disease, and its relationship with the fecal microbiome in the Ghana Breast Health Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

The study linked reduced oral microbiome diversity and altered microbial profiles to breast cancer and non-malignant breast disease, highlighting strong correlations between oral and fecal microbiomes in cases versus controls. Genera such as Porphyromonas showed significant inverse associations with breast cancer risk.

What was studied?

This study investigated the relationship between the oral microbiome, breast cancer, and non-malignant breast disease, as well as the correlation between the oral and fecal microbiomes in a case-control population in Ghana. Researchers analyzed microbiome samples from 881 women, including 369 breast cancer cases, 93 non-malignant cases, and 419 controls, using 16S rRNA gene sequencing.

Who was studied?

The study population included Ghanaian women aged 18–74 years who were recruited from Accra and Kumasi. Participants comprised breast cancer patients, individuals with non-malignant breast disease, and population-based controls. Oral and fecal microbiome samples were collected, and demographic, lifestyle, and medical history data were recorded.

What are the Most important findings?

The study revealed that oral microbiome alpha-diversity was significantly lower in breast cancer and non-malignant breast disease cases compared to controls. For instance, each 10-unit increase in observed amplicon sequence variants (ASVs) corresponded to a reduction in the odds of breast cancer and non-malignant breast disease by 14% and 21%, respectively. Beta-diversity analyses also showed distinct microbial community compositions between cases and controls. Key genera, including Porphyromonas and Fusobacterium, were inversely associated with breast cancer, with their relative abundances being significantly lower in cases than in controls. A notable finding was the strong inverse correlation between oral Porphyromonas and fecal Bacteroides in breast cancer cases. This relationship is particularly relevant as fecal Bacteroides has been implicated in estrogen metabolism and breast cancer risk. Breast cancer cases also exhibited stronger correlations between oral and fecal microbiomes compared to controls, suggesting a potential systemic interaction.

Shockingly, the study also found that breast cancer and non-malignant breast disease cases were more likely to have taken antibiotics within the last 30 days compared to controls. This raises critical questions about the role of antibiotics in microbiome disruption and their potential contribution to systemic microbial changes that could influence breast cancer risk.

What are the greatest implications?

This study is extraordinary in its scope and implications. It bridges the gap between two traditionally separate microbiomes—oral and fecal—and ties these microbial systems to breast cancer, a disease of immense global health importance. The findings reveal striking patterns: the inverse associations of oral microbiome diversity and specific genera, such as Porphyromonas and Fusobacterium, with breast cancer and non-malignant breast disease are compelling. These microbes, often linked to periodontal disease, emerge here as potential protective or systemic markers in a population with distinct environmental and health contexts.

The strong correlation between the oral and fecal microbiomes in breast cancer cases further underscores the interconnectedness of microbial communities and highlights systemic microbial interactions that remain underexplored in cancer research. The inverse relationship between Porphyromonas in the oral microbiome and Bacteroides in the fecal microbiome—key players in estrogen metabolism—provides intriguing clues about the mechanisms underlying breast cancer pathogenesis.

The right bug in the right place: opportunities for bacterial vaginosis treatment

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review explores the role of vaginal microbiota in bacterial vaginosis and highlights emerging microbiome-informed treatments. It emphasizes microbial signatures of BV, the limitations of antibiotics, and the potential of targeted biotherapeutics to restore microbial balance and reduce recurrence.

What was Reviewed?

This review examines the current understanding of the vaginal microbiome and its relationship to bacterial vaginosis (BV). It discusses how microbial dysbiosis contributes to the onset and persistence of BV and evaluates the potential therapeutic strategies that could leverage microbiome science to treat and prevent the condition. The authors explore the complexity of vaginal microbial communities, particularly focusing on the imbalance between health-associated Lactobacillus species and BV-associated anaerobic bacteria. They review both existing antibiotic treatments and emerging microbiome-informed interventions, including live biotherapeutics and vaginal microbiota transplants (VMT).

Who was Reviewed?

The review focused on published research involving women diagnosed with bacterial vaginosis, as well as healthy women with Lactobacillus-dominated vaginal microbiota. The authors synthesized data from clinical studies, in vitro experiments, and microbiome profiling studies that examined microbial composition, treatment responses, and microbial dynamics in BV-affected and healthy populations. They also reviewed preclinical studies exploring potential microbial therapeutics, including specific bacterial strains and vaginal microbiome restoration strategies.

What were the Most Important Findings?

The review highlighted that bacterial vaginosis is characterized by a distinct microbial signature: a depletion of Lactobacillus species (notably L. crispatus, L. jensenii, and L. gasseri) and an overgrowth of anaerobic bacteria. This microbial imbalance leads to elevated vaginal pH and inflammation, contributing to symptoms and increasing susceptibility to other infections.

The authors emphasized that standard antibiotic treatments, like metronidazole and clindamycin, often result in high recurrence rates and can disrupt both pathogenic and beneficial bacterial populations. They reviewed emerging microbiome-based therapies aimed at correcting vaginal dysbiosis without harming commensal microbes. These include probiotic formulations containing Lactobacillus strains, VMT, and precision antimicrobials targeting specific BV-associated pathogens. Notably, they discussed the importance of strain-specific effects, showing that not all Lactobacillus strains equally promote vaginal health, and that strain selection is critical for therapeutic success.

A key finding was that sustained remission from BV is linked to successful re-establishment of a Lactobacillus-dominant community, specifically L. crispatus. The review also addressed how host factors, sexual activity, and antibiotic exposure influence microbial dynamics, indicating the need for personalized, microbiome-informed approaches to BV treatment.

What are the Implications of this Review?

This review carries significant implications for clinicians managing bacterial vaginosis. It highlights the limitations of antibiotic-centric treatments and underscores the need for microbiome-conscious strategies that restore and maintain vaginal microbial balance. The evidence supports moving toward targeted interventions such as live biotherapeutics and VMT, which can selectively suppress BV-associated pathogens while promoting beneficial lactobacilli. Clinicians should consider that effective, long-term BV management may depend not only on pathogen eradication but also on rebuilding a resilient, health-associated vaginal microbiome. The review points to the potential of precision microbial therapies tailored to individual microbial profiles, marking a shift toward personalized vaginal microbiome medicine. For microbiome signatures research, the paper enriches the understanding of the specific bacterial players involved in BV dysbiosis and recovery.

The role of gut and genital microbiota and the estrobolome in endometriosis, infertility and chronic pelvic pain

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Chronic Pelvic Pain (CPP)
    Chronic Pelvic Pain (CPP)

    Chronic Pelvic Pain (CPP) is persistent pain in the pelvic region lasting six months or longer, often multifactorial, impacting physical and emotional well-being, and associated with various medical conditions.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

This review highlights the gut and genital microbiome's roles in estrogen-driven conditions like endometriosis, infertility, and CPP, emphasizing dysbiosis' impact on inflammation and estrogen metabolism.

What was reviewed?

The reviewed manuscript explored the intricate relationship between the gut and genital microbiomes, the estrobolome, and their roles in the pathophysiology of endometriosis, infertility, and chronic pelvic pain (CPP). The authors critically examined 28 clinical and six preclinical studies to understand microbial dysbiosis's contributions to estrogen metabolism, inflammation, and symptomatology in these conditions. This review also identified methodological gaps in microbiome studies and proposed strategies to improve future research.

Who was reviewed?

The review included human and animal studies, examining women diagnosed with endometriosis, infertility, and CPP, alongside healthy controls. Specific focus was placed on microbial associations in the gut, cervicovaginal, and endometrial microbiomes, with emphasis on bacterial vaginosis-associated bacteria, Lactobacillus depletion, and microbial influences on estrogen-driven mechanisms.

What were the most important findings?

Key findings highlighted that dysbiosis in the gut microbiome disrupts the estrobolome, an essential modulator of estrogen metabolism. This disruption contributes to heightened systemic and local inflammation, potentially exacerbating endometriosis symptoms and infertility. Many studies noted an association between bacterial vaginosis-related bacteria and a reduction in Lactobacillus dominance in the cervicovaginal microbiome with the prevalence of endometriosis and infertility. Additionally, the review underscored a bidirectional relationship between gut microbiota and endometriosis progression in animal models, emphasizing the role of gut dysbiosis in increasing b-glucuronidase activity, leading to elevated circulating estrogen levels.

What are the greatest implications of this review?

This review underscores the need for rigorous, standardized methodologies to better delineate causal relationships between microbiota and gynecological conditions like endometriosis and CPP. The findings of this review suggest that targeting the microbiome could lead to novel diagnostics and therapeutics for estrogen-driven diseases. The review also highlights the potential of leveraging microbiome-based biomarkers for non-invasive diagnostics and monitoring of endometriosis progression, bridging a critical translational gap in gynecological health.

The role of the vaginal microbiome in distinguishing female chronic pelvic pain caused by endometriosis/adenomyosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Chronic Pelvic Pain (CPP)
    Chronic Pelvic Pain (CPP)

    Chronic Pelvic Pain (CPP) is persistent pain in the pelvic region lasting six months or longer, often multifactorial, impacting physical and emotional well-being, and associated with various medical conditions.

This study examines the role of the vaginal microbiome in distinguishing chronic pelvic pain caused by endometriosis and adenomyosis. Findings highlight specific microbial signatures associated with pain severity, offering potential non-invasive biomarkers for differential diagnosis and targeted therapeutic strategies.

What was studied?

This study investigated whether the composition of the vaginal microbiome could serve as a diagnostic biomarker to differentiate chronic pelvic pain (CPP) caused by endometriosis or adenomyosis (EM/AM) from other causes of chronic pelvic pain syndrome (CPPS) in women. Using 16S rRNA sequencing (V4 region), the researchers profiled the vaginal microbiota of 37 women with EM/AM-associated CPP, 25 with CPPS from other causes, and 66 healthy controls without CPPS. Additionally, the study explored whether combining vaginal microbial markers with serum CA125 could improve differential diagnostic accuracy.

Who was studied?

The study included 128 premenopausal women attending the gynecology department of Peking Union Medical College Hospital. These were stratified into three groups: 37 women with surgically confirmed EM/AM-associated CPP, 25 women with non-EM/AM CPPS (adhesions, hydrosalpinx, infertility), and 66 women without any chronic pelvic pain. All participants were HPV-negative, had not recently used antibiotics or vaginal products, and were matched for age, gravidity, parity, and contraceptive method to control for confounding variables.

What were the most important findings?

The vaginal microbiome of women with EM/AM-associated CPP exhibited significantly higher alpha diversity than those in the CPPS and healthy control groups. Taxonomic analyses revealed distinct microbial signatures: increased abundance of Clostridium butyricum, Clostridium disporicum, Alloscardovia omnicolens, and Veillonella montpellierensis, alongside a marked depletion of Lactobacillus jensenii, Lactobacillus reuteri, and Lactobacillus iners. These differentially abundant taxa serve as potential microbiome biomarkers.

Diagnostic performance analysis demonstrated that a combination of microbial biomarkers (specifically, a relative abundance of Clostridium disporicum >0.001105% and Lactobacillus reuteri <0.1911349%) yielded 81.08% sensitivity and 52% specificity for identifying EM/AM-associated CPP. When combined with serum CA125 levels, sensitivity increased to 89.19%, although specificity remained unchanged. Functional predictions via PICRUSt revealed enrichment of metabolic pathways such as amino acid metabolism, energy metabolism, and metabolism of cofactors and vitamins in EM/AM patients, along with downregulation of membrane transport and nucleotide metabolism compared to controls. These shifts may reflect microbial contributions to inflammation and pain signaling pathways implicated in EM/AM-associated CPP.

From a microbiome signature standpoint, the enriched taxa—particularly Clostridium disporicum and Alloscardovia omnicolens—emerge as Major Microbial Associations (MMAs) due to their consistent elevation in EM/AM patients. Conversely, Lactobacillus jensenii and L. reuteri, known for their protective, anti-inflammatory properties, are depleted, suggesting their role in maintaining vaginal eubiosis and preventing EM/AM-associated pathogenesis.

What are the greatest implications of this study?

This research provides compelling evidence that the vaginal microbiome harbors discriminative microbial signatures capable of differentiating EM/AM-associated CPP from other forms of chronic pelvic pain. The incorporation of specific microbial biomarkers, particularly when paired with serum CA125, may improve non-invasive diagnostic accuracy, enabling earlier and more targeted therapeutic intervention. Clinically, these findings underscore the potential of microbiome-informed diagnostics for gynecological conditions where conventional markers fall short. More broadly, this study suggests that vaginal dysbiosis, characterized by Lactobacillus depletion and enrichment of saccharolytic and anaerobic species, could be causally linked to EM/AM pathogenesis, possibly via inflammatory or metabolic pathways. Future studies incorporating metagenomic or metabolomic analyses are warranted to functionally validate these microbial associations and to explore the feasibility of microbial modulation as a therapeutic strategy.

The Vaginal Microbiome as a Tool to Predict rASRM Stage of Disease in Endometriosis: a Pilot Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study reveals Anaerococcus as a predictive marker for endometriosis severity and highlights Lactobacillus crispatus’s lactic acid-mediated modulatory role, offering promise for non-invasive diagnostics.

What Was Studied?

This pilot observational cross-sectional study investigated the vaginal and gut microbiome profiles in women with and without endometriosis to evaluate their potential as less invasive diagnostic tools for the disease. Researchers analyzed microbiome samples collected during two menstrual cycle phases—follicular and menstrual—from 35 women with endometriosis and 24 controls. They further explored the microbiome's association with disease severity, categorized by rASRM (revised American Society for Reproductive Medicine) stages, using 16S rRNA sequencing and machine learning models.

Who Was Studied?

The study included 59 women aged 21–49, with 35 participants having surgically confirmed endometriosis and 24 serving as controls. Participants were excluded if they had a history of autoimmune diseases, active infections, or recent use of antibiotics or hormones. Vaginal and rectal samples were obtained from all subjects to evaluate microbial community state types (CSTs) and their variability across menstrual phases.

What Were the Most Important Findings?

The study highlighted significant shifts in vaginal microbiome profiles between the follicular and menstrual phases, particularly in the distribution of community state types (CSTs). During menstruation, CST IV, defined by increased anaerobic bacterial diversity, became more prominent, while CSTs II and V, dominated by Lactobacillus gasseri and Lactobacillus jensenii, respectively, disappeared. Notably, Lactobacillus crispatus was more abundant in endometriosis patients during menstruation, even within the inflammatory environment. This finding suggests a potential immunomodulatory role for L. crispatus, likely tied to its production of lactic acid, which lowers vaginal pH, inhibits pathogenic bacterial growth, and promotes immune homeostasis. By fostering an anti-inflammatory phenotype through the stimulation of cytokines like IL-10 and regulatory immune cells, L. crispatus may help counterbalance the inflammatory state characteristic of endometriosis.

Moreover, the vaginal microbiome during menstruation demonstrated predictive value for endometriosis severity. Specifically, an operational taxonomic unit (OTU) from the genus Anaerococcus strongly correlated with advanced rASRM stages (3–4), marking its potential as a biomarker for disease progression. These findings underscore the diagnostic and therapeutic promise of the vaginal microbiome, particularly L. crispatus and its role in immune modulation. Future research should further investigate these microbial associations, their lactic acid production, and their influence on the immunological environment in endometriosis.

What Are the Greatest Implications of This Study?

This study provides a foundation for using the vaginal microbiome as a non-invasive diagnostic tool for assessing endometriosis severity. The identification of Anaerococcus as a biomarker for disease stage highlights a significant advancement in linking microbiome alterations to gynecological pathology. Additionally, the potential involvement of Lactobacillus crispatus in modulating local immune responses suggests a dual diagnostic and therapeutic role for microbiome-targeted interventions. However, the findings need validation in larger cohorts due to its pilot nature and small sample size.

The Vaginal Microbiome as a Tool to Predict rASRM Stage of Disease in Endometriosis: a Pilot Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This pilot study analyzed gut and vaginal microbiomes in 59 women (35 with endometriosis, 24 controls) using 16S rRNA sequencing and machine learning to explore their diagnostic potential for endometriosis. Findings indicate microbiome variations with the menstrual cycle and disease severity, suggesting that vaginal microbiome profiles could predict endometriosis stages, offering a novel, less-invasive diagnostic method.

What was studied?

The study focused on characterizing the gut and vaginal microbiome profiles of women with endometriosis compared to controls without the disease, exploring the potential of these profiles as less-invasive diagnostic tools for assessing the severity of endometriosis.

 

Who was studied?

Fifty-nine women participated in the study, including 35 with endometriosis and 24 control subjects. Rectal and vaginal samples were collected from all participants at two different periods of their menstrual cycle.

 

What were the most important findings?

Significant findings included variations in the distribution of vaginal community state types (CSTs) across different phases of the menstrual cycle and differences in gut and vaginal microbiome profiles between patients with varying stages of endometriosis as classified by the revised American Society for Reproductive Medicine (rASRM) stages. Machine-learning models could predict the severity of endometriosis (stages 1-2 vs. 3-4) based on these microbiome profiles, with Anaerococcus genus showing the highest predictive value.

 

What are the greatest implications of this study?

The study suggests that analysis of the vaginal microbiome could serve as a novel, less-invasive method to diagnose and predict the stage of endometriosis. This approach could potentially lead to earlier and more accurate diagnoses of endometriosis, improving treatment planning and outcomes for affected women.

The Vaginal Microbiome as a Tool to Predict rASRM Stage of Disease in Endometriosis: a Pilot Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies the vaginal microbiome as a predictor of endometriosis severity, highlighting microbial shifts that correlate with rASRM staging. Anaerococcus emerged as a key biomarker for advanced disease stages, while CST IV dominance during menstruation suggests inflammatory shifts. Findings support the potential of non-invasive microbiome-based diagnosis for endometriosis.

What Was Studied?

This study investigated the potential use of the vaginal microbiome as a diagnostic tool to predict the stage of disease severity in endometriosis, based on the revised American Society for Reproductive Medicine (rASRM) staging system. Conducted as an observational cross-sectional pilot study, researchers characterized the gut and vaginal microbiome profiles of women with and without endometriosis to explore non-invasive biomarkers for disease staging. A total of 59 women participated, 35 with endometriosis and 24 controls. Rectal and vaginal samples were collected at two different points in the menstrual cycle—the menstrual and follicular phases—to assess the microbial composition's correlation with rASRM stages. Illumina sequencing was utilized to analyze 16S rRNA gene amplicons, with community state types (CSTs) assigned to classify the vaginal microbiota. Random forest-based machine-learning models were constructed to evaluate the predictive power of vaginal microbiota profiles during different menstrual phases.

Who Was Studied?

The study included 35 women with a confirmed diagnosis of endometriosis and 24 control subjects without the disease. Participants were recruited from the University of Sao Paulo and the Massachusetts Institute of Technology, with all subjects providing written informed consent. Inclusion criteria required histological confirmation of endometriosis, while controls were women undergoing laparoscopic surgery for other benign gynecological conditions. Key exclusion criteria included recent antibiotic or hormone use, active infections, autoimmune diseases, and any history of sexually transmitted infections. The vaginal and rectal samples were collected during both the follicular and menstrual phases, known to influence microbial community dynamics.

What Were the Most Important Findings?

The study revealed that the vaginal microbiome's composition significantly differed between endometriosis patients and controls, particularly during the menstrual phase. Classification models built from vaginal microbial profiles during menstruation accurately predicted rASRM stage 1–2 versus stage 3–4 endometriosis. The genus Anaerococcus emerged as the top predictive operational taxonomic unit (OTU) for distinguishing between early and advanced stages of the disease. Notably, the transition of community state types (CSTs) also reflected disease severity. During the menstrual phase, there was a marked increase in CST IV (characterized by anaerobic bacteria dominance) among both endometriosis patients (30%) and controls (25%), with a simultaneous loss of CST II and CST V. CST I, typically dominated by Lactobacillus crispatus, appeared more prevalent in endometriosis patients during menstruation, suggesting an immunomodulatory role that may contribute to the disease's local inflammatory environment. These microbial changes were consistent with differences in local immune response and hormonal fluctuations. This pilot study is the first to demonstrate that vaginal microbiome profiles, particularly the presence of Anaerococcus, may serve as a non-invasive biomarker for endometriosis staging, potentially offering a diagnostic tool that bypasses the need for invasive surgical confirmation.

Microbial GroupEndometriosis FindingsClinical Implications
AnaerococcusIncreased in rASRM stages III–IVMarker for advanced disease severity
CST IV (Community State Type)Dominant during menstruationAssociated with low Lactobacillus and high microbial diversity
Lactobacillus spp.Decreased during menstrual phaseReduced protective barrier; potential inflammation driver
Machine Learning PredictionHigh accuracy (AUC = 0.89)Potential for non-invasive staging of endometriosis
Vaginal Microbiome ShiftsCorrelated with menstrual cycle phaseIndicates dynamic microbial changes tied to inflammation

What Are the Greatest Implications of This Study?

The findings from this study suggest that the vaginal microbiome, specifically the composition of community state types and the presence of Anaerococcus, may be harnessed as a non-invasive biomarker to predict the stage of endometriosis severity. This has profound implications for clinical practice, as it could reduce the dependency on invasive laparoscopy for disease staging, which is currently the gold standard. If validated in larger cohorts, this approach could facilitate early detection and better stratification of endometriosis patients, enabling more targeted and personalized therapeutic interventions. Furthermore, the study underscores the significance of microbiome-driven inflammation in the pathophysiology of endometriosis, opening avenues for microbiome-targeted therapies as a novel strategy to mitigate disease progression and symptom severity. The integration of vaginal microbiome profiling into clinical diagnostics could revolutionize the early detection and management of endometriosis, addressing a critical unmet need in gynecological health.

Tobacco consumption and premenstrual syndrome: A case-control study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This study highlights the significant link between tobacco consumption and the increased risk of premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD). Women who smoke are at higher odds of developing these disorders, with a dose-response relationship.

What was studied?

The study aimed to assess the relationship between tobacco smoking and premenstrual syndrome (PMS), including its more severe form, premenstrual dysphoric disorder (PMDD). It focused on understanding how smoking may contribute to the occurrence of these menstrual disorders. The study utilized a case-control design, comparing women with PMS and PMDD to age-matched controls.

Who was studied?

The study population consisted of women diagnosed with PMS, women diagnosed with PMDD, and control groups who did not have PMS or PMDD. Participants were recruited from three major public hospitals and one family counseling center in Santiago de Compostela, Spain, ensuring a sample that included women of various age groups and social backgrounds. All participants completed a self-administered questionnaire that gathered information on their smoking habits, socio-demographic factors, and menstrual health.

What were the most important findings?

The study found that tobacco consumption is significantly associated with both PMS and PMDD. Specifically, current smokers had a higher likelihood of experiencing PMS and PMDD compared to non-smokers. This association was evident even among ex-smokers, although the odds ratio was less precise due to the small sample size of this group. The findings also indicated a dose-response relationship: women who smoked more than 15 cigarettes per day or those with higher pack-years of tobacco consumption had an increased risk of developing PMS and PMDD, suggesting a potential cumulative effect of tobacco exposure. The results were confirmed by a cubic spline model, which further demonstrated a correlation between the amount of tobacco consumed and the likelihood of developing these menstrual disorders.

What are the greatest implications of this study?

The study’s findings suggest that tobacco consumption may be a modifiable risk factor for the development of PMS and PMDD. Given the high prevalence of tobacco use among women globally, particularly in Europe and the United States, these results highlight the need for public health strategies aimed at reducing smoking in women, particularly those in their reproductive years. Health professionals should be aware of the potential link between smoking and these menstrual disorders, as this could inform both prevention and treatment strategies. This study calls for further research to explore the long-term effects of smoking cessation and whether reducing tobacco exposure could lower the risk of developing PMS or PMDD in susceptible women.

Trace Elements and Endometriosis: Insights into Oxidative Stress and Novel Therapies

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review explores the role of trace elements and oxidative stress in endometriosis, highlighting their potential as therapeutic targets. It underscores the need for further research into the trace elements’ roles in endometriotic lesions.

What was reviewed?

The article reviews the role of trace elements in the pathogenesis and management of endometriosis, a chronic, estrogen-dependent inflammatory disease. It synthesizes existing research on the impact of oxidative stress and environmental exposure to trace elements like zinc, nickel, cadmium, and copper, linking these factors to the formation and proliferation of endometrial-like lesions outside the uterus.

Who was reviewed?

The review focuses on studies involving women with confirmed endometriosis, highlighting environmental and biological factors such as trace element concentrations in blood, urine, and peritoneal fluid. Additionally, it incorporates experimental findings, including animal models, to explore the mechanistic roles of trace elements.

What were the most important findings?

The review emphasizes the link between oxidative stress and endometriosis, with trace elements acting as potential modulators of this process. Zinc, for instance, is identified for its antioxidant and anti-inflammatory roles, with lower levels in endometriosis patients potentially contributing to lesion formation. Nickel, on the other hand, has been implicated in the condition as a metalloestrogen, as further evidenced by improved symptoms following a low-nickel diet. Cadmium and lead, known for inducing oxidative stress, show conflicting associations with endometriosis, though some evidence suggests their presence synergistically exacerbates disease severity. Copper's involvement in angiogenesis and its elevated levels in endometriosis patients suggest a role in lesion proliferation. The review also highlights discrepancies in study findings, emphasizing the need for further research on trace elements within endometriotic implants rather than just systemic fluids.

What are the greatest implications of this review?

The review underscores the potential of targeting trace elements and oxidative stress as therapeutic strategies for endometriosis. It calls for more comprehensive research into the specific roles of trace elements within endometriotic tissue, as these could pave the way for novel diagnostic markers and treatments. Additionally, the environmental and dietary implications of trace element exposure warrant further exploration, particularly in the context of prevention and symptom management.

Transvaginal Photobiomodulation for the Treatment of Chronic Pelvic Pain: A Pilot Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The study evaluated transvaginal photobiomodulation as a treatment for women's chronic pelvic pain (CPP). It found significant pain relief sustained for six months, suggesting TVPBM could be an effective treatment option. Further research is needed to explore broader applications of TVPBM.

The study in question focused on the use of transvaginal photobiomodulation (TV-PBM) as a treatment for chronic pelvic pain (CPP) in women. Here are the details based on the key aspects of the study:

 

What was studied?

The research investigated whether transvaginal photobiomodulation could effectively reduce chronic pelvic pain in women. This was a pilot study aiming to explore the potential benefits of a novel therapy approach using light therapy applied transvaginally to manage pain​​.

 

Who was studied?

The study involved women suffering from chronic pelvic pain. These participants had previously experienced ineffective treatments for their condition and were recruited to evaluate the effectiveness of TV-PBM. Thirteen women completed the study, undergoing a series of nine treatments​​.

 

What were the most important findings?

The study found significant and sustained pain relief in participants up to six months after treatment. It reported a notable reduction in pain scores, with 60% of the participants showing improvement shortly after treatment began, and this effect was maintained throughout the six-month follow-up period. The effect sizes were considered large, suggesting the therapy was highly effective for those it helped​​.

 

What are the greatest implications of this study?

The promising results of this pilot study suggest that TV-PBM could be a viable and effective treatment for managing chronic pelvic pain, a condition that often responds poorly to other forms of treatment. This could lead to a new therapeutic option for many women who currently have limited or ineffective choices. However, further research, including larger and controlled studies, is necessary to confirm these findings and fully establish the therapy’s efficacy, safety, and broader applicability​​.

Unveiling Resistance and Virulence Mechanisms under Darwinian Positive Selection for Novel Drug Discovery for Gardnerella vaginalis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

The study analyzed the genetic evolution of Gardnerella vaginalis, focusing on its resistance and virulence under Darwinian positive selection. It identifies new drug targets and emphasizes the pathogen's evolving resistance mechanisms.

What was Studied?

The study focused on Gardnerella vaginalis, a significant pathogen responsible for bacterial vaginosis(BV), examining its mechanisms of resistance and virulence under Darwinian positive selection. The researchers utilized comparative genomic analysis to identify resistance and virulence-related genes and their evolutionary patterns. The study also aimed to discover potential new drug targets by analyzing these genomic features in the context of the pathogen's evolutionary adaptations.

Who was Studied?

The study analyzed 97 genomes of Gardnerella vaginalis strains, representing a diverse collection of isolates obtained from the National Center for Biotechnology Information (NCBI) datasets. The strains were carefully selected to reflect the genetic variability and resistance phenotypes of this important pathogen, enabling a comprehensive understanding of its evolution.

What were the most Important Findings?

The study identified several crucial findings that provide new insights into the evolution and pathogenic potential of G. vaginalis. The pathogen exhibits significant genomic diversity, which plays a role in its survival and adaptation to selective pressures, particularly from antibiotics. The analysis revealed some genes, such as Mef(A), associated with resistance to macrolides, and tet(M) and tet(L), linked to resistance against tetracycline. These resistance genes were found to be positively selected in multiple G. vaginalis lineages, reflecting the evolutionary pressures that have shaped the pathogen's resistance capabilities.

Furthermore, the study highlighted the pathogen’s ability to form biofilms, a feature that enhances its survival in the host and increases its resistance to antibiotic treatment. This biofilm formation is also associated with the pathogen's ability to engage in horizontal gene transfer, further complicating the treatment landscape. The pan-resistome analysis indicated that the pathogen has an "open" resistome, suggesting its high capacity to acquire new resistance genes, making it a continuously evolving threat. The researchers also identified two potential drug targets, sigA, a sigma factor involved in transcription initiation, and UDP-N-acetylenolpyruvoylglucosamine reductase, an enzyme crucial for cell wall synthesis. These proteins are vital to the pathogen's survival and represent promising targets for the development of new therapeutic approaches.

What are the Implications of this Study?

The study’s findings highlight the dynamic nature of Gardnerella vaginalis and its ability to rapidly adapt to environmental pressures, particularly through the acquisition of resistance genes. The evolution of resistance mechanisms and the presence of virulence factors underscore the pathogen's significant role in reproductive and sexual health complications. The open pan-resistome suggests that G. vaginalis can continue to evolve and acquire new resistance traits, posing an ongoing challenge to existing treatments. The identification of novel drug targets like sigA and UDP-N-acetylenolpyruvoylglucosamine reductase offers valuable insights into how future therapies could be designed to combat infections caused by this pathogen. This research calls for continued surveillance of G. vaginalis strains to track resistance trends and refine clinical treatment strategies.

Usefulness of intermittent clomiphene citrate treatment for women with polycystic ovarian syndrome that is resistant to standard clomiphene citrate treatment

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

The study explored intermittent clomiphene citrate treatment (ICT) for women with PCOS who were resistant to standard CC treatment. ICT showed an 80.8% response rate, offering an effective and safer alternative to gonadotropin therapy with no risk of OHSS or multiple pregnancies.

What was studied?

The study investigated the efficacy of intermittent clomiphene citrate treatment (ICT) in women with polycystic ovary syndrome (PCOS) who were resistant to standard clomiphene citrate (CC) treatment. Clomiphene citrate is commonly used to induce ovulation in women with PCOS; however, some women do not respond to the standard treatment. This research aimed to assess whether a modified dosing schedule, using intermittent CC treatment, could help induce follicular growth in these resistant patients.

Who was studied?

The study focused on 42 infertile women diagnosed with PCOS who were resistant to standard CC treatment. Of these, 26 women underwent the intermittent CC treatment protocol, and their response to the treatment was monitored and analyzed. The patient cohort was selected based on their non-response to the standard 50 mg/day, 5-day CC regimen, which is typically used as a first-line treatment for anovulatory PCOS.

What were the most important findings?

The study found that intermittent CC treatment was highly effective for women who were resistant to standard CC treatment. The protocol involved administering 100 mg of CC for 5-day periods across different phases of the menstrual cycle. The results showed that 80.8% of the patients who had previously been resistant to standard CC treatment responded positively to ICT, with follicular growth observed in many of the cases. Notably, the majority of those who responded had only a single mature follicle, which is a safer outcome in terms of reducing the risk of multiple pregnancies or ovarian hyperstimulation syndrome (OHSS). Additionally, no cases of OHSS or multiple pregnancies were reported, making ICT a promising alternative to gonadotropin therapy, which carries higher risks and requires more intensive treatment.

The efficacy of ICT was particularly notable in patients resistant to the 100 mg dose of CC, where up to 78.2% responded positively after subsequent doses. This suggests that ICT may be more effective than simply increasing the dose of standard CC, providing a less invasive and lower-risk alternative for patients who fail the initial treatment.

What are the greatest implications of this study?

The findings suggest that ICT could become a preferred treatment option for women with CC-resistant PCOS before advancing to gonadotropin therapy. ICT offers several advantages: it is less invasive, more cost-effective, and poses a lower risk of complications like OHSS and multiple pregnancies. By maintaining high serum FSH levels through repeated administration of CC, ICT seems to trigger follicular growth effectively without the need for prolonged or more invasive treatments. The study's outcomes could help reshape the treatment protocols for PCOS, offering a safer, more accessible alternative to gonadotropin therapy. However, further research with larger patient populations is necessary to validate these findings and assess the long-term efficacy of ICT in achieving successful pregnancy outcomes.

Vitamin D and probiotic co-supplementation affects mental health, hormonal, inflammatory and oxidative stress parameters in women with polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Vitamin D and probiotic co-supplementation improved depression, reduced testosterone and inflammation, and enhanced antioxidant status in women with PCOS. Results support microbiome-mediated benefits.

What was studied?

This randomized, double-blinded, placebo-controlled clinical trial investigated the effects of co-supplementation with vitamin D and probiotics on mental health, hormonal profiles, inflammatory markers, and oxidative stress in women with polycystic ovary syndrome (PCOS). The study tested whether the combination of these two interventions could provide synergistic benefits in a population known to have both systemic inflammation and frequent vitamin D deficiency. The researchers hypothesized that probiotic-driven microbiota modulation and vitamin D's immunomodulatory properties could jointly improve both mental and metabolic health in PCOS.

Who was studied?

Sixty women with PCOS, aged 18–40 years and with body mass index (BMI) between 17 and 34 kg/m², participated in the study. All subjects were insulin-resistant (HOMA-IR between 1.4–4) and had not been receiving prior vitamin D or probiotic supplementation. They were randomized into two groups: one received 50,000 IU of vitamin D every two weeks plus 8 × 10⁹ CFU/day of a probiotic blend containing Lactobacillus acidophilus, Bifidobacterium bifidum, Lactobacillus reuteri, and Lactobacillus fermentum for 12 weeks, while the control group received matching placebos.

What were the most important findings?

Vitamin D and probiotic co-supplementation resulted in statistically significant reductions in depression, anxiety, and stress scores, indicating improved mental health. There were also significant improvements in hormonal parameters, particularly a reduction in total testosterone and hirsutism, which are hallmark features of PCOS-related hyperandrogenism. Inflammatory and oxidative stress markers showed marked improvement as well: high-sensitivity C-reactive protein (hs-CRP) and malondialdehyde (MDA) decreased, while total antioxidant capacity (TAC) and glutathione (GSH) levels increased.

From a microbiome perspective, the inclusion of multiple Lactobacillus and Bifidobacterium strains supports known major microbial associations (MMAs) relevant to metabolic and hormonal balance. These strains are associated with improved gut barrier function, increased short-chain fatty acid (SCFA) production—especially butyrate—and reduced translocation of lipopolysaccharide (LPS), which contributes to systemic inflammation. Enhanced expression of vitamin D receptors (VDR) by probiotics could further amplify these effects, facilitating vitamin D’s role in reducing oxidative stress and modulating immune response.

What are the implications of this study?

This trial provides compelling evidence that co-supplementation with vitamin D and probiotics can significantly improve mental health, reduce androgen levels, and counteract systemic inflammation and oxidative stress in women with PCOS. These findings are particularly relevant for clinicians exploring microbiome-modifying interventions in PCOS treatment. By targeting both neuroendocrine and metabolic pathways, the combination of vitamin D and probiotics appears to act via the gut-brain-endocrine axis—a crucial interface in PCOS pathophysiology. The results suggest that routine screening for vitamin D deficiency and addressing gut dysbiosis with targeted probiotics could become integral parts of comprehensive PCOS management. Importantly, this synergistic therapy may reduce the need for multiple pharmacologic agents and improve patient adherence and outcomes. Future research should evaluate long-term effects and perform microbiome sequencing to validate microbial shifts and functional changes driving these clinical benefits.

Endometriosis

Go to Category Page

1H NMR- based metabolomics approaches as non-invasive tools for diagnosis of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metabolomic Signature
    Metabolomic Signature

    Metabolomic signatures are unique metabolite patterns linked to specific biological conditions, identified through metabolomics. They reveal underlying biochemical activities, aiding in disease diagnosis, biomarker development, and personalized medicine. The microbiome significantly affects these signatures, influencing health and disease outcomes through metabolic interactions.

This study demonstrates the potential of ¹H-NMR metabolomics to diagnose endometriosis non-invasively by identifying metabolic biomarkers and disrupted pathways. Quadratic Discriminant Analysis outperformed Artificial Neural Networks in diagnostic accuracy.

What was studied?

This study investigated the application of metabolomics, specifically through proton nuclear magnetic resonance (¹H-NMR) spectroscopy, to identify non-invasive biomarkers for diagnosing endometriosis. The researchers developed computational models using Quadratic Discriminant Analysis (QDA) and Artificial Neural Networks (ANNs) to analyze metabolic changes in serum samples and assess their utility in early diagnosis of the disease.

Who was studied?

The study analyzed serum samples from 31 infertile women diagnosed with stage II or III endometriosis confirmed via laparoscopy and 15 healthy women without any signs of endometriosis. The participants were aged 22–44 years and were recruited from an infertility center in Iran. Exclusion criteria included recent medical or hormonal treatments, prior gynecological surgeries, or other pelvic inflammatory conditions.

What were the most important findings?

The study revealed significant metabolic differences between women with endometriosis and healthy controls. Key findings included elevated levels of 2-methoxyestrone, 2-methoxyestradiol, androstenedione, aldosterone, dehydroepiandrosterone, and deoxycorticosterone in the endometriosis group, alongside decreased cholesterol and primary bile acids. These metabolic changes are linked to disruptions in steroid hormone biosynthesis and bile acid metabolism, indicating underlying hyperestrogenism and impaired hepatic estrogen clearance. The QDA model achieved a correct classification rate of 76%, with 71% positive predictive value and 78% negative predictive value, outperforming the ANN model, which had lower sensitivity and specificity. Metabolic pathway analyses highlighted altered steroid hormone and bile acid biosynthesis, which are critical in the pathophysiology of endometriosis.

What are the greatest implications of this study?

This study underscores the potential of ¹H-NMR-based metabolomics as a minimally invasive diagnostic tool for endometriosis, reducing reliance on invasive laparoscopy. The identification of specific biomarkers and disrupted pathways could facilitate earlier diagnosis, improved patient stratification, and targeted therapeutic interventions. The findings also demonstrate the utility of computational modeling, particularly QDA, in translating complex metabolomics data into clinically actionable insights. This approach represents a significant advancement in bridging diagnostic gaps for endometriosis.

A metabonomics approach as a means for identification of potentialbiomarkers for early diagnosis of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metabolomic Signature
    Metabolomic Signature

    Metabolomic signatures are unique metabolite patterns linked to specific biological conditions, identified through metabolomics. They reveal underlying biochemical activities, aiding in disease diagnosis, biomarker development, and personalized medicine. The microbiome significantly affects these signatures, influencing health and disease outcomes through metabolic interactions.

This study used 1H-NMR metabonomics to identify serum biomarkers for early endometriosis diagnosis. Elevated lactate, alanine, and reduced glucose levels highlight metabolic disruptions, offering a minimally invasive diagnostic tool.

What was studied?

This study investigated the identification of predictive biomarkers for early diagnosis of endometriosis using a minimally invasive, serum-based approach. The researchers utilized proton nuclear magnetic resonance (1H-NMR) metabonomics to analyze serum samples, aiming to distinguish endometriosis patients from healthy controls. The study particularly focused on differences in metabolite profiles to identify markers indicative of the condition.

Who was studied?

The study included 45 women aged under 40, divided into two groups. The first group comprised 22 women diagnosed with early-stage endometriosis (Stages I–II) via laparoscopy, while the control group consisted of 23 age- and BMI-matched healthy women with normal menstrual cycles and proven fertility. Participants with recent hormone therapy or irregular menstrual cycles were excluded. Serum samples were collected during the secretory phase of the menstrual cycle.

What were the most important findings?

The study identified several metabolites with significantly altered levels in women with endometriosis compared to controls. Increased levels of lactate, 3-hydroxybutyrate, alanine, leucine, valine, threonine, lysine, glycerophosphatidylcholine, succinic acid, and 2-hydroxybutyrate were observed in the serum of endometriosis patients, while glucose, isoleucine, arginine, and lipid levels were decreased. Multivariate analysis using Partial Least Squares-Discriminant Analysis (PLS-DA) demonstrated strong sensitivity (81.8%) and specificity (91.3%) in distinguishing endometriosis from controls, with an area under the ROC curve of 0.96. Pathway analysis highlighted arginine and proline metabolism disruptions, glycine, serine, and threonine metabolism, pyruvate metabolism, and lysine biosynthesis and degradation. These findings provide a potential non-invasive diagnostic framework and insights into the metabolic disturbances in endometriosis.

What are the greatest implications of this study?

This study offers a promising step toward non-invasive diagnostic methods for endometriosis, reducing reliance on invasive laparoscopy. The identification of metabolite alterations linked to the disease enhances the understanding of its pathophysiology, emphasizing oxidative stress, anaerobic glycolysis, and metabolic reprogramming similar to malignancies. These findings could lead to better clinical tools for early diagnosis and a deeper understanding of the metabolic underpinnings of endometriosis.

A More Diverse Cervical Microbiome Associates with Better Clinical Outcomes in Patients with Endometriosis: A Pilot Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

This study links cervical microbiome diversity with endometriosis severity. Findings reveal microbial imbalances, particularly in advanced stages, correlate with pain, infertility, and inflammatory pathways. The cervical microbiome may serve as a diagnostic and therapeutic target for improving outcomes in endometriosis, highlighting its role in reproductive health and disease progression.

What Was Studied?

This pilot study investigated the cervical microbiome in patients with endometriosis and its association with clinical outcomes. The research focused on the microbial diversity, composition, and functional roles in cervical mucus, analyzed using 16S rRNA sequencing. The study included healthy women and patients diagnosed with endometriosis to compare microbial profiles and explore the connection between microbiome alterations, disease progression, and associated symptoms like pain, CA125 levels, and infertility.

Who Was Studied?

The study involved 33 women: 10 healthy controls and 23 patients diagnosed with endometriosis (classified by severity into stages I-II and III-IV). The cervical microbiome was analyzed to assess its correlation with clinical features, such as deep infiltrating endometriosis (DIE), CA125 biomarker levels, pain severity, and infertility.

What Were the Most Important Findings?

The study revealed that cervical microbiome diversity is significantly associated with clinical outcomes in endometriosis patients. Specifically, a higher microbial diversity was linked to better outcomes, while notable microbial imbalances characterized advanced disease stages and severe symptoms. Patients with advanced stages of endometriosis exhibited a microbial shift, with an increase in Firmicutes and a decrease in Actinobacteria and Bacteroidetes. Unique microbial profiles were observed, such as elevated Lactobacillus jensenii and Streptococcus agalactiae (GBS), alongside reduced Atopobium vaginae in patients with advanced stages.

Patients presenting severe symptoms, including elevated CA125 biomarker levels, infertility, and higher pain scores, showed significantly reduced microbial richness and diversity. Infertility, a common complication of endometriosis, was associated with an increased Firmicutes/Bacteroidetes ratio. Notably, infertility treatments appeared to reverse these imbalances, restoring microbial diversity and community structure to resemble that of fertile individuals. Additionally, deep infiltrating endometriosis (DIE), a severe form of the condition, was correlated with an overrepresentation of Streptococcus and Prevotella at the genus level.

The study’s functional analyses provided insight into the role of the cervical microbiome in disease progression. Pathways associated with microbial alterations, such as signal transduction, secondary bile acid biosynthesis, and nutrient metabolism, were identified. These pathways may contribute to inflammation, immune dysregulation, and potentially malignancy in severe cases. Such findings underscore the intricate relationship between cervical microbial composition and the pathophysiology of endometriosis. This research positions the cervical microbiome as a critical factor in both the diagnosis and management of endometriosis, offering potential for therapeutic interventions targeting microbial imbalances.

What Are the Greatest Implications of This Study?

The findings suggest that cervical microbiome diversity may serve as a biomarker for diagnosing and monitoring endometriosis progression and complications. The research highlights the therapeutic potential of targeting microbial imbalances to improve clinical outcomes, particularly in infertility. It also underscores the potential link between microbiome alterations and malignancy risks in severe cases, paving the way for preventive and precision medicine approaches in endometriosis management.

Altered Microbiome-Derived Extracellular Vesicles in Peritoneal Fluid of Women with Endometriosis: Implications for Pathogenesis and Therapy

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies microbiota alterations in ovarian endometrioma, showing distinct microbial shifts in peritoneal fluid extracellular vesicles. Enrichment of Pseudomonas and Acinetobacter, alongside depletion of Propionibacterium and Actinomyces, suggests inflammatory contributions to pathogenesis. Findings highlight the diagnostic potential of microbiota-derived EVs in endometrioma management.

What was studied?

This study examined the microbiota composition in the peritoneal fluid of women with ovarian endometrioma, focusing specifically on microbiome analyses of extracellular vesicles (EVs). Extracellular vesicles are nanometer-sized particles released by cells, including bacteria, that carry microbial DNA and signaling molecules. The research aimed to determine if women with ovarian endometrioma exhibit distinct microbiota profiles in their peritoneal fluid compared to women without endometriosis. Microbial DNA was sequenced using next-generation sequencing (NGS) of the 16S rDNA V3–V4 regions, allowing for detailed taxonomic identification and comparative analysis.

Who was studied?

The study included 45 women diagnosed with histological evidence of ovarian endometrioma and 45 surgical controls confirmed to be free of endometriosis. Participants were recruited from Asan Medical Center, and peritoneal fluid samples were collected during laparoscopic procedures. Women with endometriosis were classified as having advanced-stage disease, and none of the participants had taken antibiotics, probiotics, or hormonal treatments for 12 weeks prior to sample collection.

What were the most important findings?

The microbiota composition of extracellular vesicles in peritoneal fluid was markedly different between women with ovarian endometrioma and controls. Alpha diversity analysis showed no significant differences in species richness between groups, but beta diversity analysis revealed distinct microbial community shifts in the endometriosis group (p < 0.001). Taxonomic profiling demonstrated increased abundances of Acinetobacter, Pseudomonas, Streptococcus, and Enhydrobacter in women with ovarian endometrioma. Conversely, Propionibacterium, Actinomyces, and Rothia were significantly decreased in the endometriosis group (p < 0.05).

At the family level, Pseudomonadaceae and Moraxellaceae were notably enriched in the endometriosis samples, while Veillonellaceae, Propionibacteriaceae, and Actinomycetaceae were reduced. The data also indicated a significant increase in Pseudomonadales and a decline in Actinomycetales at the order level (p < 0.05). These findings suggest that the altered microbiota composition in extracellular vesicles of the peritoneal fluid may contribute to the inflammatory microenvironment observed in ovarian endometrioma.

Microbial GroupOvarian EndometriomaClinical Implications
AcinetobacterIncreasedLinked to inflammation and immune response in the peritoneal cavity
PseudomonasIncreasedAssociated with pathogenic processes in ovarian endometrioma
StreptococcusIncreasedPotential contributor to local inflammation and immune modulation
EnhydrobacterIncreasedMay play a role in extracellular signaling and immune responses
PropionibacteriumDecreasedLoss may disrupt protective anti-inflammatory effects
ActinomycesDecreasedReduced presence suggests compromised mucosal defenses
RothiaDecreasedMay contribute to a disrupted microbial ecosystem
Pseudomonadaceae (Family)EnrichedSuggests pathogenic influence in peritoneal fluid
Moraxellaceae (Family)EnrichedAssociated with peritoneal inflammation
Veillonellaceae, Propionibacteriaceae, Actinomycetaceae (Families)ReducedIndicates loss of protective and commensal populations

What are the greatest implications of this study?

The study provides compelling evidence that women with ovarian endometrioma possess distinct microbial communities in the peritoneal environment, carried via extracellular vesicles. The enrichment of pathogenic genera such as Pseudomonas and Acinetobacter, alongside the depletion of protective taxa like Propionibacterium and Actinomyces, suggests that these microbial imbalances could play a role in local inflammation and disease progression. These findings underscore the potential of microbiota-derived EVs as non-invasive biomarkers for ovarian endometrioma and open the door for targeted microbiome-modulating therapies to alleviate inflammatory responses and halt disease progression.

Anti-Endometriotic Effects of Pueraria Flower Extract: A Novel Therapeutic Approach

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

The study explored the effects of Pueraria Flower Extract (PFE) on human endometriotic cells and mice, showing that PFE inhibits cell adhesion, migration, and MMP expression, and reduces lesion formation. Highlighting PFE's potential as a non-invasive treatment alternative, the research provides insights into molecular targets for future therapies in managing endometriosis, a condition with limited current treatments.

What was studied?

This study investigated the anti-endometriotic effects of Pueraria flower extract (PFE) on human endometriotic cells and a mouse model of endometriosis. It evaluated the extract's impact on cellular adhesion, migration, and the expression of matrix metalloproteinases (MMPs), key factors in the establishment of endometriotic lesion.

Who was studied?

The research focused on human-immortalized endometriotic cell lines (11Z and 12Z) and mesothelial Met5A cells in vitro. Additionally, a mouse model of induced endometriosis was used to evaluate the effects of PFE in vivo.

What were the most important findings?

Inhibition of Cell Adhesion and Migration: PFE significantly suppressed the adhesion of endometriotic cells to mesothelial cells and reduced cell migration in wound-healing and transwell assays.

Reduction in MMP Expression: PFE decreased both mRNA and protein levels of MMP-2 and MMP-9, enzymes crucial for tissue invasion and lesion establishment in endometriosis.

ERK1/2 Signaling Activation: The study demonstrated that PFE activates the ERK1/2 pathway, which played a role in inhibiting cell migration. This effect was reversed when an ERK1/2 inhibitor was introduced.

Lesion Suppression in Mice: Oral administration of PFE to mice significantly reduced the number of endometriotic lesions without causing toxicity or weight loss.

Role of Isoflavones: Major isoflavones such as tectorigenin were identified as active compounds in PFE, contributing to its anti-endometriotic effects.

What are the greatest implications of this study?

The findings suggest that PFE and its active compounds, particularly tectorigenin, could serve as potential therapeutic agents for endometriosis. By targeting matrix metalloproteinase (MMP) activity and the ERK1/2 pathway, PFE may provide a novel, non-hormonal intervention to mitigate lesion formation and progression. This research highlights the potential for plant-derived compounds in developing treatments that reduce the recurrence and side effects associated with conventional endometriosis therapies.

Antibiotic therapy with metronidazole reduces endometriosis disease progression in mice: a potential role for gut microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study shows that antibiotic therapy with metronidazole reduces endometriotic lesion growth and inflammation in mice by targeting the gut microbiota, particularly Bacteroidetes. Findings suggest that microbiota-targeted treatments may offer new therapeutic avenues for endometriosis management.

What Was Studied?

This study examined the impact of antibiotic therapy with metronidazole on endometriosis disease progression in a mouse model, exploring its potential effects on gut microbiota and inflammation. Researchers induced endometriosis in mice through surgical transplantation of uterine tissue onto the peritoneal wall. Mice were then treated with either broad-spectrum antibiotics (vancomycin, neomycin, metronidazole, and ampicillin) or metronidazole alone, with control groups receiving vehicle-only treatment. The primary goal was to determine if modulating gut bacteria through antibiotic therapy could reduce endometriotic lesion growth and inflammation, potentially revealing gut microbiota as a therapeutic target.

Who Was Studied?

The study utilized a well-established mouse model of surgically induced endometriosis, where uterine tissue from estrus-stage mice was autologously transplanted onto the peritoneal wall. Mice were separated into groups receiving either broad-spectrum antibiotics, metronidazole alone, or vehicle treatments. To assess the effect of gut microbiota on disease progression, fecal transplantation experiments were performed, where feces from endometriosis-induced mice were gavaged into metronidazole-treated mice to observe the restoration of lesion growth and inflammation.

What Were the Most Important Findings?

The findings demonstrated that antibiotic therapy, particularly with metronidazole, significantly reduced the size and volume of endometriotic lesions in mice. Mice treated with broad-spectrum antibiotics showed a five-fold reduction in lesion size and markedly fewer proliferating cells and macrophages within the lesions compared to vehicle-treated controls (p < 0.01). Metronidazole-treated mice specifically exhibited smaller ectopic lesions than those receiving neomycin or vehicle, suggesting a unique sensitivity of certain gut bacteria to metronidazole's antimicrobial activity. Importantly, inflammation markers, including IL-1β, TNF-α, IL-6, and TGF-β1, were significantly reduced in the peritoneal fluid of metronidazole-treated mice, indicating a diminished inflammatory response.

Additionally, fecal transplantation experiments highlighted the role of gut microbiota in lesion progression. Oral gavage of feces from endometriosis-induced mice restored lesion growth and inflammation in metronidazole-treated mice, implicating gut bacteria as contributors to disease persistence. 16S rRNA sequencing of fecal samples showed that Bacteroidetes were enriched in endometriosis-induced mice and nearly absent in metronidazole-treated mice, suggesting that the suppression of specific microbial populations might underlie the observed therapeutic effects. This reduction in Bacteroidetes correlated with decreased inflammatory responses and smaller lesion sizes, underscoring the interplay between gut microbiota and endometriosis pathology.

What Are the Greatest Implications of This Study

The study provides compelling evidence that targeting the gut microbiota with antibiotics like metronidazole can effectively reduce endometriotic lesion growth and inflammation in a mouse model. The findings suggest that Bacteroidetes may contribute to lesion persistence and immune activation, and their depletion through metronidazole treatment alleviates these pathological effects. This raises the possibility of microbiota-targeted therapies as a novel approach to managing endometriosis, potentially offering a non-hormonal alternative to traditional treatments. Furthermore, the study highlights the significance of gut microbiota modulation in controlling systemic and local inflammatory responses, paving the way for research into gut-mediated mechanisms of endometriosis and microbiome-based therapeutic strategies.

Association between endometriosis and risk of histological subtypes of ovarian cancer: a pooled analysis of case–control studies

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

Endometriosis significantly increases the risk of clear-cell, low-grade serous, and endometrioid ovarian cancers. This study highlights the need for subtype-specific ovarian cancer surveillance and prevention strategies.

DOI: https://doi.org/10.1016%2FS1470-2045(11)70404-1

What Was Studied?

This study examined the association between endometriosis and the risk of specific histological subtypes of ovarian cancer. It involved a pooled analysis of 13 Ovarian Cancer Association Consortium case-control studies. The research aimed to clarify whether the increased risk associated with endometriosis extended to all invasive histological subtypes of ovarian cancer or was limited to specific subtypes.

Who Was Studied?

The study analyzed data from 13,226 controls and 9,818 women with ovarian cancer (7,911 with invasive and 1,907 with borderline ovarian cancer). Among these, 738 women with invasive cancer and 168 with borderline cancer reported a history of endometriosis. The data collection spanned multiple international sites and included self-reported histories of endometriosis.

What Were the Most Important Findings?

The study found that a history of endometriosis significantly increased the risk for three specific subtypes of invasive ovarian cancer: clear-cell, low-grade serous, and endometrioid cancers. The odds ratios for these associations were 3.05, 2.11, and 2.04, respectively. In contrast, no significant association was observed with high-grade serous or mucinous subtypes, nor with borderline ovarian cancers. The findings suggest that endometriosis acts as a precursor lesion for clear-cell and endometrioid ovarian cancers, with molecular similarities supporting this hypothesis. Notably, the association with low-grade serous cancer requires further study, as this subtype showed distinct molecular characteristics, such as a higher likelihood of KRAS or BRAF mutations compared to TP53 mutations in high-grade serous cancers.

What Are the Greatest Implications of This Study?

This study underscores the need for clinicians to recognize the increased risk of specific ovarian cancer subtypes in women with endometriosis. Understanding the mechanisms behind the malignant transformation of endometriosis could enable the identification of high-risk individuals for tailored surveillance or preventive strategies, such as risk-reducing surgery. The study also highlights the importance of considering histological subtypes in ovarian cancer research, reflecting its heterogeneous nature and the need for subtype-specific prevention, screening, and treatment approaches.

Associations Between Endometriosis and Gut Microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

The gut microbiota has been associated with many diseases, including endometriosis. However, very few studies have been conducted on this topic in human. This study aimed to investigate the association between endometriosis and gut microbiota. Women with endometriosis (N=66) were identified at the Department of Gynaecology and each patient was matched with three controls (N=198) from the general population. All participants answered questionnaires about socioeconomic data, medical history, and gastrointestinal symptoms and passed stool samples. Gut bacteria were analyzed using 16S ribosomal RNA sequencing, and in total, 58 bacteria were observed at genus level in both patients with endometriosis and controls. Comparisons of the microbiota between patients and controls and within the endometriosis cohort were performed. Both alpha and beta diversities were higher in controls than in patients. With the false discovery rate q<0.05, abundance of 12 bacteria belonging to the classes Bacilli, Bacteroidia, Clostridia, Coriobacteriia, and Gammaproteobacter differed significantly between patients and controls. Differences observed between patients with or without isolated ovarian endometriosis, involvement of the gastrointestinal tract, gastrointestinal symptoms, or hormonal treatment disappeared after calculation with false discovery rate. These findings indicate that the gut microbiota may be altered in endometriosis patients.

What Was Studied?

This study investigated the association between endometriosis and gut microbiota. Conducted at Skåne University Hospital in Sweden, the research aimed to understand how the gut microbiome differs in women diagnosed with endometriosis compared to healthy controls. The study included 66 women with endometriosis confirmed through laparoscopy or laparotomy and 198 age, BMI, and smoking-matched controls. Stool samples from both groups were analyzed using 16S ribosomal RNA sequencing to identify bacterial composition at the genus level. The primary objective was to compare the diversity and abundance of gut microbiota between the two groups and explore any microbiome changes correlated with endometriosis characteristics such as disease localization, gastrointestinal symptoms, or hormonal treatment.

Who Was Studied?

The study examined 66 women diagnosed with endometriosis recruited from the Department of Gynaecology at Skåne University Hospital. These participants were matched with 198 controls from the Malmö Offspring Study (MOS), ensuring similarities in age, BMI, and smoking status. Women in the endometriosis group were diagnosed based on clinical criteria, confirmed through surgical procedures, and were excluded if they had comorbid gastrointestinal conditions like Crohn's disease, ulcerative colitis, or irritable bowel syndrome (IBS). The control group, drawn from a population-based cohort, also passed stool samples and completed questionnaires about their medical history and gastrointestinal symptoms.

What Were the Most Important Findings?

The study found significant differences in gut microbiota diversity and composition between women with endometriosis and healthy controls. Notably, alpha and beta diversities were higher in the control group, suggesting a richer and more varied microbial population compared to endometriosis patients. At the genus level, 12 bacterial genera belonging to the classes Bacteroidia, Clostridia, Coriobacteriia, Bacilli, and Gammaproteobacteria differed significantly between groups. For instance, Bacteroides and Parabacteroides were elevated in endometriosis patients, while Paraprevotella and Lachnospira were found in lower abundance compared to controls. Additionally, there was a distinct alteration in the microbial community within the endometriosis cohort based on disease localization and the presence of gastrointestinal symptoms. Patients with isolated ovarian endometriosis exhibited higher levels of Lachnobacterium and Adlercreutzia compared to those with widespread lesions. Furthermore, the presence of gastrointestinal symptoms correlated with lower levels of SMB53 (Clostridia) and Odoribacter (Bacteroidia), while Prevotella was more abundant. Interestingly, hormone treatment was associated with higher levels of Blautia and Ruminococcus in the Clostridia class, along with Butyricimonas in the Bacteroidia class. These findings support the hypothesis that gut microbiota may be altered in endometriosis patients, with distinct microbial signatures linked to hormonal therapy and gastrointestinal involvement.

Increased in Endometriosis PatientsDecreased in Endometriosis Patients
Bacteroides (Bacteroidia)Paraprevotella (Bacteroidia)
Parabacteroides (Bacteroidia)Lachnospira (Clostridia)
Blautia (Clostridia) with hormone treatmentOdoribacter (Bacteroidia) with GI symptoms
Ruminococcus (Clostridia) with hormone treatmentSMB53 (Clostridia) with GI symptoms
Butyricimonas (Bacteroidia) with hormone treatment

What Are the Greatest Implications of This Study?

The study's findings suggest that endometriosis is associated with specific alterations in gut microbiota, which could play a role in the pathophysiology of the disease. The reduced microbial diversity in endometriosis patients points towards a potential dysbiosis that may exacerbate inflammation and modulate estrogen metabolism, both of which are critical in the pathogenesis of endometriosis. Furthermore, specific bacterial shifts linked to hormone treatment indicate that gut microbiota could be influenced by estrogen-related therapies, potentially affecting symptom severity and disease progression. Understanding these microbial associations opens the door to novel therapeutic strategies, such as targeted probiotics or microbiome-based interventions, to alleviate gastrointestinal symptoms and modulate disease activity in endometriosis patients. This research also underlines the need for further studies to explore the bidirectional relationship between gut microbiota and estrogen regulation in estrogen-dependent conditions like endometriosis.

Bacterial infection linked to endometriosis

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Study links Fusobacterium to endometriosis via inflammation and lesion formation. Antibiotics (metronidazole and chloramphenicol) may offer therapeutic potential.

New title: Fusobacterium Infection: A New Pathogenic Insight into Endometriosis and Microbiome-Targeted Therapy Potential

DOI: https://doi.org/10.1016/S2666-5247(23)00221-5

What Was Studied?

This translational study investigated the role of Fusobacterium infection in the pathogenesis of endometriosis. Researchers sought to determine whether bacterial infection, specifically by Fusobacterium nucleatum, contributes to inflammatory alterations in endometrial fibroblasts, potentially leading to the development of endometriosis. The study encompassed molecular analyses, in vitro experiments, and a mouse model to establish causality and mechanism.

Who Was Studied?

The study analyzed uterine tissue samples from 79 patients in two Japanese hospitals, divided into cases with endometriosis and controls without the condition. Further, a mouse model was used to test the infectivity and pathogenic potential of Fusobacterium nucleatum compared to other microbes.

What Were the Most Important Findings?

Fusobacterium nucleatum was found to be significantly more prevalent in the endometrial and endometriotic tissues of patients with endometriosis (64.3%) compared to controls (7.1%), while Erysipelothrix, another potential candidate, was not abundant. Fusobacterium infection was shown to upregulate transgelin (TAGLN) expression in fibroblasts, enhancing their motility, adhesion, and migration through the activation of TGF-β signaling, a pathway known to be critical in the progression of endometriosis. In an animal model, mice inoculated with Fusobacterium-infected uterine tissue developed endometriotic lesions, whereas treatment of donor mice with antibiotics (metronidazole and chloramphenicol) significantly reduced lesion formation in recipient mice. These findings suggest that targeting Fusobacterium with antibiotics holds potential to mitigate the progression of endometriosis, underscoring the importance of further exploration into microbiome-targeted therapies.

What Are the Greatest Implications of This Study?

This research provides evidence that Fusobacterium infection may play a direct role in the etiology of endometriosis. The identification of a bacterial trigger opens avenues for antibiotic-based interventions and highlights the need for clinical trials targeting endometrial infections. Additionally, it underscores the importance of microbial profiling in endometriosis diagnosis and management, potentially redefining treatment paradigms to include microbiome-targeted interventions (MBTIs).

Beneficial Effects of a Low-Nickel Diet on Relapsing IBS-Like and Extraintestinal Symptoms of Celiac Patients during a Proper Gluten-Free Diet: Nickel Allergic Contact Mucositis in Suspected Non-Responsive Celiac Disease

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study provides strong evidence that nickel-rich foods in a gluten-free diet can trigger or exacerbate IBS-like gastrointestinal and extraintestinal symptoms in a subset of celiac disease patients, even when the disease is in serological and histological remission. The findings highlight the importance of nickel sensitivity in the pathogenesis of these relapsing symptoms, suggesting that a low-nickel diet can be a valuable intervention to improve patient outcomes. The use of the nickel oral mucosa patch test (Ni omPT) was also validated as a reliable diagnostic tool for identifying nickel-sensitive patients.

What was studied?

The study investigated the prevalence and effects of nickel allergic contact mucositis (Ni ACM) in celiac disease (CD) patients who were on a proper gluten-free diet (GFD) but experienced a relapse of Irritable Bowel Syndrome (IBS)-like gastrointestinal and extraintestinal symptoms. The main goal was to determine whether nickel-rich foods in a gluten-free diet could trigger these symptoms and to evaluate the impact of a low-nickel diet (LNiD) on symptom reduction in these patients. This pilot study also explored the use of the nickel oral mucosa patch test (Ni omPT) to diagnose nickel sensitivity in these patients.

Who was studied?

The study involved 102 consecutive adult celiac disease patients (74 females, 28 males, mean age 42.3 ± 7.4 years) who had been on a gluten-free diet for at least 12 months and were in serological and histological remission of their disease. These patients were selected because they reported relapsing or persisting gastrointestinal and extraintestinal symptoms despite proper adherence to a GFD. After exclusions based on comorbid conditions like lactose intolerance and Helicobacter pylori infection, 20 patients (all female, age 23–65 years) were included in the final analysis.

What were the most important findings?

In a cohort of 20 patients with persistent symptoms despite adherence to a gluten-free diet (GFD), all tested positive for nickel sensitivity via the nickel oral mucosa patch test (Ni omPT), confirming a diagnosis of nickel allergic contact mucositis (Ni ACM). Following prolonged GFD, 83.3% of patients experienced a relapse of symptoms, including abdominal pain, bloating, loose stools, and fatigue, coinciding with high dietary nickel intake from nickel-rich gluten-free foods such as corn. Implementing a low-nickel diet (LNiD) for three months improved 83.4% of total symptoms, with 41.7% reaching statistical significance. Notably, 80% of gastrointestinal and 88.9% of extraintestinal symptoms improved, including significant relief from abdominal pain, swelling, fatigue, and dermatitis. Combining LNiD with GFD restored patients' well-being to levels comparable to those previously achieved by GFD alone, strongly implicating nickel sensitivity as the primary driver of symptom relapse.

What are the greatest implications of this study?

This study provides strong evidence that nickel-rich foods in a gluten-free diet can trigger or exacerbate IBS-like gastrointestinal and extraintestinal symptoms in a subset of celiac disease patients, even when the disease is in serological and histological remission. The findings highlight the importance of nickel sensitivity (Ni ACM) in the pathogenesis of these relapsing symptoms, suggesting that a low-nickel diet (LNiD) can be a valuable intervention to improve patient outcomes. The use of the nickel oral mucosa patch test (Ni omPT) was also validated as a reliable diagnostic tool for identifying nickel-sensitive patients.

Clinically, these results imply that gastroenterologists should consider nickel sensitivity as a differential diagnosis in celiac patients who are non-responsive to a GFD and continue to experience symptoms. The integration of a low-nickel dietary approach alongside the GFD may become an essential part of managing non-responsive celiac disease with overlapping IBS-like symptoms. Further large-scale studies are needed to confirm these findings and refine dietary guidelines for managing nickel sensitivity in this population.

Causal effects of gut microbiome on endometriosis: a two-sample Mendelian randomization study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This Mendelian randomization study found a causal relationship between gut microbiota and endometriosis. Porphyromonadaceae and Anaerotruncus increased risk, while Clostridiales_vadin_BB60_group, Oxalobacteraceae, Desulfovibrio, Haemophilus, and Holdemania were protective. These findings support microbiome-targeted interventions as a potential treatment strategy for endometriosis.

What was studied?

This study investigated the causal relationship between gut microbiome composition and endometriosis using a two-sample Mendelian randomization (MR) approach. The researchers aimed to determine whether specific gut microbiota taxa have a direct causal effect on endometriosis risk rather than a mere observational association.

Who was studied?

The study utilized genome-wide association study (GWAS) summary statistics from two major datasets to investigate the causal relationship between gut microbiota and endometriosis. Exposure data on the gut microbiome were obtained from the MiBioGen consortium, which included 18,340 individuals across 24 cohorts from multiple countries. Outcome data for endometriosis were sourced from the FinnGen consortium, comprising 13,456 endometriosis cases and 100,663 controls, all of European ancestry.

Key Findings

The study identified five bacterial taxa with a protective effect against endometriosis and two taxa associated with increased risk. Protective taxa included Clostridiales_vadin_BB60_group, Oxalobacteraceae, Desulfovibrio, Haemophilus, and Holdemania, all of which exhibited odds ratios (OR) below 1, indicating a reduced likelihood of endometriosis in individuals with higher genetic abundance of these bacteria. In contrast, Porphyromonadaceae and Anaerotruncus were associated with increased endometriosis risk, with ORs above 1, suggesting their potential involvement in disease progression. Sensitivity analyses confirmed the robustness of these findings, as no evidence of pleiotropy or heterogeneity was detected, reinforcing the reliability of the causal associations.

Bacterial TaxaP-ValueEffect
Clostridiales_vadin_BB60_group
< 0.01
Protective
Oxalobacteraceae0.014Protective
Desulfovibrio0.046Protective
Haemophilus0.039Protective
Holdemania0.025Protective
Porphyromonadaceae0.027Risk
Anaerotruncus< 0.01Risk

Greatest Implications

The findings of this study provide genetic evidence supporting a causal relationship between gut microbiota and endometriosis, reinforcing previous observational research. This suggests that targeting the gut microbiome through interventions such as probiotics, dietary modifications, or microbiome-targeted interventions (MBTIs) could be a novel approach to managing or preventing endometriosis. The identification of specific bacterial taxa that either increase or decrease endometriosis risk offers a foundation for developing microbiome-targeted interventions tailored to patient needs. Additionally, these results support the estrobolome hypothesis, which proposes that gut microbiota influence estrogen metabolism, potentially contributing to the pathophysiology of endometriosis. By establishing a causal link, this study highlights the importance of gut microbiota in the broader endocrine and inflammatory mechanisms underlying the disease, paving the way for further research into microbiome-based therapeutic strategies.

Correlation of fecal metabolomics and gut microbiota in mice with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies gut microbiota and metabolomic shifts in endometriosis, with altered bile acid biosynthesis and ALA metabolism. Elevated CDCA and UDCA levels, coupled with microbial changes, suggest potential biomarkers and therapeutic targets for managing inflammation in endometriosis.

What Was Studied?

This study investigated the correlation between fecal metabolomics and gut microbiota in mice with endometriosis. Using a controlled experimental design, researchers constructed an endometriosis (EMS) mouse model with female C57BL/6J mice and analyzed fecal samples through non-targeted metabolomics and 16S rRNA sequencing. The primary objective was to identify differential metabolites and microbial compositions that could serve as biomarkers for endometriosis and provide insight into the metabolic pathways affected by gut dysbiosis in EMS. Functional prediction of the gut microbiota was performed using PICRUSt, and metabolite-microbiota correlations were assessed through Spearman correlation coefficients.

Who Was Studied

The study involved female C57BL/6J mice, which were divided into two groups: an EMS group and a control group. Endometriosis was induced in the EMS group through intraperitoneal injection of endometrial fragments, while the control group received saline injections with adipose tissue. Fecal samples were collected from both groups, processed for liquid chromatography-mass spectrometry (LC-MS), and subjected to 16S rRNA sequencing to map microbial diversity and metabolic profiles. The study aimed to simulate the inflammatory and microbiome-related characteristics of endometriosis in humans by using this established animal model.

What Were the Most Important Findings?

The study identified significant shifts in both fecal metabolomics and gut microbiota composition in mice with endometriosis compared to controls. A total of 156 named differential metabolites were screened, with key changes observed in pathways linked to secondary bile acid biosynthesis and alpha-linolenic acid (ALA) metabolism. Notably, there was an increased abundance of chenodeoxycholic acid (CDCA) and ursodeoxycholic acid (UDCA) alongside a decreased presence of ALA and 12,13-EOTrE in the EMS mice. Microbial diversity was reduced in the EMS group, with a specific loss in Bacteroides and Firmicutes, contrasted by increases in Proteobacteria and Verrucomicrobia. At the genus level, there was a marked increase in Allobaculum, Akkermansia, Parasutterella, and Rikenella, with significant decreases in Lachnospiraceae, Lactobacillus, and Bacteroides. Functional predictions revealed alterations in oxidative phosphorylation, alanine, aspartate, glutamate metabolism, and starch and sucrose metabolism. Importantly, the study identified Sphingobium and Pseudomonas viridiflava as consistently enriched in EMS mice, suggesting their potential role in inflammation and metabolic disruption. The correlation analysis demonstrated strong associations between specific metabolites (like CDCA and ALA) and microbial shifts, indicating a complex interaction between gut dysbiosis and metabolic imbalances in endometriosis.

ParameterFindings in EMS Mice
Metabolomic Changes156 differential metabolites identified, with key changes in secondary bile acid biosynthesis and alpha-linolenic acid (ALA) metabolism.
Increased MetabolitesChenodeoxycholic acid (CDCA) and Ursodeoxycholic acid (UDCA).
Decreased MetabolitesAlpha-linolenic acid (ALA) and 12,13-EOTrE.
Microbial DiversityOverall reduction in diversity; significant losses in Bacteroides and Firmicutes.
Phylum-Level ShiftsProteobacteria and Verrucomicrobia significantly increased in the EMS group.
Genus-Level IncreasesAllobaculum, Akkermansia, Parasutterella, and Rikenella.
Genus-Level DecreasesLachnospiraceae, Lactobacillus, and Bacteroides.
Functional Pathway AlterationsDisrupted oxidative phosphorylation, alanine, aspartate, glutamate metabolism, and starch and sucrose metabolism.
Unique EnrichmentsSphingobium and Pseudomonas viridiflava enriched in EMS mice, indicating roles in inflammation and metabolic disruption.
Metabolite-Microbiota CorrelationsStrong correlations between CDCA, ALA, and microbial shifts, suggesting complex interactions contributing to dysbiosis and inflammation.

What Are the Greatest Implications of This Study?

The findings suggest that endometriosis is associated with profound shifts in gut microbiota and fecal metabolomics, which may contribute to chronic inflammation and disease persistence. The increased levels of chenodeoxycholic acid (CDCA) and ursodeoxycholic acid (UDCA), combined with reductions in ALA, indicate that bile acid metabolism and fatty acid dysregulation are central to the pathogenesis of endometriosis. The enrichment of Allobaculum, Akkermansia, Parasutterella, and Rikenella in the gut microbiota suggests these species could be contributing to local and systemic inflammation, disrupting gut barrier integrity. These microbial and metabolomic signatures could serve as non-invasive biomarkers for diagnosing endometriosis and may offer new therapeutic targets focused on restoring microbial balance and metabolic homeostasis. Furthermore, the study highlights the critical role of gut microbiota in modulating immune responses and metabolic pathways, paving the way for microbiome-targeted treatments in endometriosis management.

Does Exposure of Lead and Cadmium Affect the Endometriosis?

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

The study links lead and cadmium exposure to increased endometriosis risk, emphasizing lead's role at low blood levels and synergistic effects with cadmium. It advocates for strict monitoring and preventive measures to minimize exposure.

What Was Studied?

This study investigated the association between occupational exposure to lead and cadmium and the risk of developing endometriosis (EM) among South Korean female workers. Utilizing medical and biological data from over 26,000 individuals who underwent lead-associated medical examinations between 2000 and 2004, the study examined blood lead levels (BLLs), co-exposure to cadmium, and their relationship with hospital admissions for EM.

Who Was Studied?

The study focused on South Korean female workers exposed to lead as part of their occupation. These individuals underwent specialized medical examinations. A total of 26,542 workers were included, with the study comparing EM admissions in lead-exposed workers against the general population and noise-exposed workers as control groups.

What Were the Most Important Findings?

The study found that lead exposure, even at relatively low levels (BLLs < 5 µg/dL), was significantly associated with an increased risk of EM. The standard admission rate (SAR) for EM in lead-exposed workers was 1.24 times higher than the general population, and for workers with BLLs < 5 µg/dL, it was 1.44 times higher. Co-exposure to lead and cadmium demonstrated a synergistic effect, amplifying the risk of EM beyond what could be expected from exposure to either metal alone. While cadmium exposure alone did not show a statistically significant association with EM, the relative excess risk due to interaction (RERI) was 0.33, indicating a notable combined impact of these metals. The study also highlighted that oxidative stress induced by heavy metal exposure likely underpins these effects, with mechanisms involving the disruption of antioxidant defenses and cellular damage.

What Are the Greatest Implications of This Study?

This research underscores the need to minimize exposure to heavy metals, particularly lead and cadmium, among female workers. It also calls for rigorous monitoring of blood lead and cadmium levels in workplaces to mitigate their combined effects. The findings are critical for understanding the environmental and occupational contributors to EM and suggest that policies limiting heavy metal exposure could have a substantial public health impact, especially for at-risk populations.

Effect of endometriosis on the fecal bacteriota composition of mice during the acute phase of lesion formation

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study found that endometriosis does not induce significant changes in the fecal bacteriota composition during the acute phase of lesion formation. Findings suggest that gut microbial shifts in endometriosis may emerge only during chronic disease stages, highlighting the need for targeted microbiome interventions over prolonged periods.

What Was Studied?

This study investigated the effect of endometriosis on the fecal bacteriota composition of mice during the acute phase of lesion formation. Researchers aimed to understand whether the establishment of endometriotic lesions would influence gut microbial communities, potentially contributing to systemic inflammation or metabolic disruptions associated with the disease. Uterine tissue fragments from GFP+ donor mice were transplanted into the peritoneal cavity of GFP- wild-type mice, inducing endometriotic lesions. Sham-transplanted mice served as controls. Fecal samples were collected three days before, and at 7 and 21 days after lesion induction, and analyzed through 16S rRNA gene sequencing to map changes in microbial composition.


Who Was Studied?

The study involved C57BL/6 wild-type mice as the experimental model for endometriosis. The model was established by transplanting uterine tissue fragments from GFP+ donor mice into the peritoneal cavity of GFP- recipient mice, allowing for easy visualization of endometriotic lesions. Sham-transplanted animals, which received physiological saline solution instead of tissue fragments, served as controls. The study analyzed fecal samples collected at specific time points to assess microbiota changes during the acute phase of endometriosis development.

What Were the Most Important Findings?

The study found that the induction of endometriosis did not produce significant changes in the composition of the fecal bacteriota during the acute phase of lesion formation (7 and 21 days post-transplantation). Despite the successful establishment of endometriotic lesions and typical histomorphology observed under fluorescence microscopy, alpha and beta diversity analyses showed no substantial differences between the endometriosis-induced group and sham controls. Detailed sequencing revealed a highly diverse microbial community dominated by Bacteroidales S24-7 group, Lactobacillus, Prevotellaceae UCG-001 group, and Lachnospiraceae NK4A136 group in both experimental and control mice. Notably, contrary to previous studies suggesting dysbiosis following endometriosis induction, this investigation showed microbial stability throughout the acute phase of lesion formation. The researchers speculated that gut microbiota disturbances may become apparent only in the chronic stages of the disease, reflecting long-term inflammation and tissue remodeling. Furthermore, the study emphasized that strict statistical controls, including the removal of singleton OTUs and application of false discovery rate (FDR) corrections, were applied to prevent false positives. These rigorous controls could have contributed to the observed stability of gut microbiota composition, challenging earlier reports of rapid dysbiosis post-endometriosis induction.

ParameterFindings in Endometriosis-Induced Mice
Microbiota CompositionNo significant changes in the overall composition of fecal bacteriota during the acute phase (7 and 21 days post-transplantation).
Alpha DiversityNo substantial differences observed between endometriosis-induced mice and sham controls, indicating microbial richness and evenness remained stable.
Beta DiversityAnalysis showed no significant shifts in microbial community structure between experimental and control groups.
Dominant GeneraMicrobiota was dominated by Bacteroidales S24-7 group, Lactobacillus, Prevotellaceae UCG-001 group, and Lachnospiraceae NK4A136 group in both groups.
Impact of Lesion FormationInduction of endometriosis did not disrupt gut microbiota composition during the acute phase of lesion establishment.
Statistical Controls AppliedStrict controls, including false discovery rate (FDR) corrections and removal of singleton OTUs, were applied to enhance result reliability.
Hypothesized Long-Term EffectsAuthors suggest that gut dysbiosis may only emerge in chronic stages of endometriosis, not during initial lesion establishment.

What Are the Greatest Implications of This Study?

The study challenges prevailing hypotheses that endometriosis immediately disrupts gut microbiota during the early phases of lesion formation. The findings suggest that intestinal dysbiosis may not occur in the acute phase but could instead be a consequence of chronic inflammation and prolonged disease progression. This insight implies that gut microbial changes observed in patients with endometriosis might reflect long-term disease dynamics rather than initial lesion establishment. These results underscore the need for longitudinal studies to distinguish between acute and chronic microbiome shifts in endometriosis. The findings also highlight the importance of standardized microbiome analysis protocols and strict statistical measures to accurately assess microbial composition in endometriosis models. Understanding the timeline of microbiome alterations in endometriosis could guide therapeutic strategies targeting microbial populations in chronic disease stages rather than acute phases.

Elevated Lactoferrin and Anti-Lactoferrin Antibodies in Endometriosis: Autoimmune and Microbiome Insights

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This study confirms elevated lactoferrin and anti-lactoferrin antibody levels in endometriosis, suggesting autoimmune involvement. Anti-lactoferrin drops post-surgery, hinting at a biomarker role, while lactoferrin ties to inflammation and potential microbiome links.

What Was Studied?

This study, conducted by Mori-Yamanaka et al. and published in Tohoku J. Exp. Med. in 2023, definitively explored serum lactoferrin (LTF) and anti-lactoferrin antibody (aLF) levels in patients with endometriosis. Endometriosis, a chronic inflammatory condition marked by ectopic endometrial-like tissue, remains poorly understood in terms of its underlying mechanisms. The researchers aimed to determine whether LTF, an iron-binding glycoprotein with antimicrobial and anti-inflammatory properties, and aLF, an autoantibody tied to immune dysregulation, play roles in the disease’s pathology. By measuring these markers in the blood of endometriosis patients compared to controls and assessing changes after surgical intervention, the study sought to uncover potential links to inflammation and autoimmunity. Although the study did not directly investigate microbiome signatures, LTF’s known role in modulating microbial environments suggests a possible indirect connection to gut or pelvic microbiome alterations in endometriosis.

Who Was Studied?

The research focused on 68 Japanese women undergoing surgery at Shiga University of Medical Science Hospital between November 2020 and May 2022. Of these, 51 had surgically and histopathologically confirmed endometriosis, spanning all stages (I-IV) per the revised American Society for Reproductive Medicine classification. The remaining 17 women, who underwent surgery for other gynecological issues like uterine myomas or benign ovarian tumors, served as controls without endometriosis. This cohort provided a robust sample to compare LTF and aLF levels across disease states and post-treatment outcomes, offering clinicians a clear demographic context for interpreting the findings.

What Were the Most Important Findings?

The study conclusively demonstrated that serum LTF and aLF levels are significantly elevated in endometriosis patients compared to controls, with p-values of 0.016 and 0.028, respectively. These elevations were particularly striking in advanced stages (III and IV), showing stronger statistical significance (LTF: p = 0.024; aLF: p = 0.016) compared to controls. Following surgery in 21 patients, aLF levels dropped markedly (p < 0.001), while LTF levels showed no significant change (p = 0.102). Notably, 43% of endometriosis patients exhibited aLF levels above the reference range, a prevalence akin to autoimmune conditions. Although microbiome data wasn’t directly assessed, LTF’s antimicrobial properties hint at potential microbial associations, possibly involving dysbiosis in the pelvic or gut microbiome, which could exacerbate inflammation in endometriosis. These findings position LTF and aLF as key players in the disease’s inflammatory and possibly autoimmune landscape.

What Are the Greatest Implications of This Study?

This study’s implications are profound for clinicians managing endometriosis. The elevated aLF levels, mirroring patterns in autoimmune diseases, strongly suggest that endometriosis involves an autoimmune component, potentially driven by immune responses to microbial or endogenous triggers. This insight could shift treatment paradigms toward immune-modulating therapies. Moreover, the significant post-surgical decline in aLF levels establishes it as a promising biomarker for monitoring disease activity and treatment success, offering a practical tool for clinical decision-making. While LTF’s role remains less clear, its persistence post-surgery and antimicrobial function imply a complex interplay with inflammation and possibly the microbiome, warranting further investigation into microbial signatures like those of Lactobacillus or Prevotella, known to influence pelvic health. Despite the study’s limitations—its small sample and surgical focus—these findings pave the way for innovative diagnostics and therapies, urging clinicians to consider immune and microbial factors in endometriosis care.

Elevated Lead, Nickel, and Bismuth Levels in the Peritoneal Fluid of a Peritoneal Endometriosis Patient without Toxic Habits or Occupational Exposure following a Vegetarian Diet

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

A case study links elevated lead, nickel, and bismuth in peritoneal fluid with endometriosis, highlighting potential dietary and environmental exposures as contributors. Further research may identify these potentially toxic elements (PTEs) as diagnostic biomarkers and therapeutic targets.

What Was Studied?

This case report investigated the multielemental profile of peritoneal fluid (PF) in a 22-year-old woman diagnosed with peritoneal endometriosis. The study aimed to evaluate the concentrations of potentially toxic elements (PTEs), including lead (Pb), nickel (Ni), bismuth (Bi), and cobalt (Co), and to compare them with levels in an age-matched control without endometriosis. The patient had no toxic habits, occupational exposure, or documented environmental exposure, and adhered to a vegetarian diet, raising questions about the dietary and environmental sources of PTEs and their role in the pathogenesis of endometriosis.

Who Was Studied?

The primary subject was a 22-year-old woman diagnosed with peritoneal endometriosis during laparoscopic surgery, where her PF was analyzed. The comparison group included an age-matched control and a reference cohort of ten women diagnosed with non-hormonally dependent benign ovarian cysts.

Most Important Findings

The study revealed significantly elevated levels of Pb (75 µg/L, 90:1 ratio), Ni (40.4 µg/L, 4:1 ratio), Bi (33.3 µg/L, 1.5:1 ratio), and Co (1.39 µg/L, 5:1 ratio) in the PF of the endometriosis patient compared to the control. These findings suggest potential contributions of dietary and environmental exposures to PTEs. Nickel, a cofactor for metalloenzymes, was noted to be higher potentially due to the patient’s vegetarian diet, which is associated with increased nickel intake from plant-based foods such as nuts and legumes. Elevated Pb levels were striking, with concentrations much higher than typical dietary or environmental exposures in industrialized settings. While cobalt and bismuth also showed elevated levels, their specific roles in endometriosis remain unclear. The findings support the hypothesis that environmental and dietary PTE exposure may contribute to the pathogenesis of endometriosis by inducing oxidative stress or endocrine disruption.

Greatest Implications

This study highlights the need to explore PTEs as potential biomarkers for endometriosis diagnosis and as contributors to its etiology. Elevated PTE levels in PF may result from dietary habits, such as a vegetarian diet, or unidentified environmental exposures. This study emphasizes the importance of further investigations into environmental toxicology and dietary patterns in endometriosis patients. Understanding these associations could inform preventative strategies, dietary guidelines, and therapeutic interventions for endometriosis management.

Elevated levels of whole blood nickel in a group of Sri Lankan women with endometriosis: a case control study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study identified elevated blood nickel levels in women with endometriosis, suggesting a potential role of nickel as a metalloestrogen in its pathogenesis.

What Was Studied?

This study investigated the association between whole blood levels of nickel, cadmium, and lead in women with and without endometriosis. Specifically, it aimed to determine whether these heavy metals, known to have estrogenic properties, could be linked to the pathogenesis of endometriosis. The research involved analyzing the whole blood levels of these metals in 50 women with endometriosis and 50 age-matched controls who were confirmed to be free of the condition via laparoscopy or laparotomy.

Who Was Studied?

The study focused on a group of Sri Lankan women of reproductive age who underwent laparotomy or laparoscopy. The participants were divided into two groups: cases (women diagnosed with endometriosis, n=50) and controls (women without endometriosis, n=50). None of the participants were current smokers, and the groups were matched for age and body mass index.

Most Important Findings

The study revealed significantly elevated levels of nickel in the whole blood of women with endometriosis compared to controls (2.6 μg/L vs. 0.8 μg/L, p=0.016). This finding aligns with previous evidence that nickel, a potent metalloestrogen, can activate estrogen receptors and may contribute to the persistence of ectopic endometrial tissue. In contrast, the blood levels of cadmium and lead did not show statistically significant differences between the two groups. The presence of nickel in ectopic endometrial tissue, previously demonstrated by the researchers, supports the hypothesis that hematogenous routes could transport nickel to ectopic sites. Despite these findings, the study's small sample size limits the ability to conclude definitively that nickel is an etiological factor for endometriosis.

Greatest Implications

The discovery of higher nickel levels in women with endometriosis introduces a novel avenue for understanding the role of environmental pollutants, particularly metalloestrogens, in the condition's pathogenesis. It emphasizes the need for larger-scale studies to explore nickel's potential as a biomarker or contributor to endometriosis. Furthermore, this research underscores the importance of addressing environmental and occupational exposures to nickel, especially for women of reproductive age, as part of preventive strategies for endometriosis.

Endometriosis induces gut microbiota alterations in mice

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study demonstrates that endometriosis induces gut microbiota alterations in a murine model, particularly increasing the Firmicutes/Bacteroidetes ratio and enriching Bifidobacterium and Parasutterella. These shifts suggest dysbiosis as a contributing factor to inflammation and immune dysregulation, supporting the potential for microbiota-targeted therapies in endometriosis management.

What Was Studied?

This study investigated the effects of endometriosis on gut microbiota composition in a murine model, specifically evaluating microbial shifts during the progression of endometriosis. Researchers employed a prospective and randomized design, inducing endometriosis in mice through intraperitoneal injection of endometrial tissues. The primary aim was to characterize changes in gut microbiota over time, utilizing 16S ribosomal-RNA gene sequencing to assess microbial diversity and composition at 7, 14, 28, and 42 days post-induction. The experiment included mock groups as controls, which received saline injections instead of endometrial tissue, to account for any procedural effects.

Who Was Studied?

The study involved C57BL6 mice, a commonly used murine model, to mimic endometriosis development. Mice were divided into two groups: those receiving endometrial tissue injections to induce endometriosis, and mock controls receiving only saline. The animals were sacrificed at four different time points (7, 14, 28, and 42 days) for fecal sample collection and microbiota analysis. Researchers conducted 16S rRNA sequencing on these samples to evaluate alterations in microbial communities associated with endometriosis progression.

What Were the Most Important Findings?

The study revealed that endometriosis induced significant alterations in gut microbiota composition, particularly at 42 days post-induction. Beta diversity analysis demonstrated that the microbial community structure diverged substantially from the mock controls, indicating dysbiosis. At the phylum level, there was an increased Firmicutes/Bacteroidetes ratio, a hallmark often linked to inflammatory conditions. Furthermore, Actinobacteria and Betaproteobacteria were more abundant in the endometriosis group, whereas Bacteroidetes was more dominant in the control group. At the genus level, the study identified increases in Ruminococcaceae-UGG-014, Bifidobacterium, and Parasutterella among endometriosis mice. These microbial shifts suggest that endometriosis disrupts normal gut microbial homeostasis, potentially influencing systemic inflammation and immune modulation. The researchers noted that while alpha diversity remained similar between groups, the specific microbial composition shifted dramatically over the 42-day period. This timeline suggests that gut dysbiosis in endometriosis is progressive and may exacerbate immune system imbalances over time.

Taxonomic LevelMicrobiota Findings in Endometriosis-Induced Mice
Phylum LevelIncreased Firmicutes/Bacteroidetes ratio. Elevated levels of Actinobacteria and Betaproteobacteria.
Class LevelEnhanced representation of Clostridia and Actinobacteria classes.
Order LevelNotable increase in Lactobacillales and Clostridiales.
Family LevelSignificant enrichment of Ruminococcaceae and Bifidobacteriaceae.
Genus LevelMarked increases in Ruminococcaceae-UGG-014, Bifidobacterium, and Parasutterella.
Alpha DiversityNo significant difference in microbial richness or evenness compared to controls.
Beta DiversitySignificant divergence from mock controls, indicating altered microbial community structure.
Inflammatory AssociationsAltered microbiota profile is linked to systemic inflammation and immune modulation, suggesting a role in endometriosis progression.

What Are the Greatest Implications of This Study

The findings underscore the role of gut microbiota dysbiosis in the progression of endometriosis, revealing distinct shifts in microbial populations, especially an elevated Firmicutes/Bacteroidetes ratio. These changes mirror dysbiosis seen in other inflammatory diseases, suggesting that gut microbiota may contribute to systemic inflammation and immune dysfunction in endometriosis. The study highlights the potential for microbiota-targeted therapies to restore gut microbial balance as a therapeutic approach. Additionally, the identification of enriched genera such as Bifidobacterium and Parasutterella suggests potential biomarkers for non-invasive diagnostics. The progressive nature of microbiota alteration observed at 42 days further indicates that early intervention targeting microbial communities could mitigate inflammatory responses and possibly slow disease progression. This research provides a mechanistic link between gut dysbiosis and endometriosis pathology, paving the way for microbiome-based therapeutic strategies.

Exploring the link between dietary zinc intake and endometriosis risk: insights from a cross-sectional analysis of American women

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • STOPs
    STOPs

    A STOP (Suggested Termination Of Practices) is a recommendation that advocates for the discontinuation of certain medical interventions, treatments, or practices based on emerging evidence indicating that these may be ineffective, harmful, or counterproductive in the management of specific conditions.

This study links higher dietary zinc intake with increased endometriosis risk among American women, highlighting zinc’s complex role in immune modulation and estrogen-related pathways. Findings emphasize the importance of balanced intake for managing endometriosis risk.

What was studied?

This study investigated the association between dietary zinc intake and the risk of endometriosis among American women. Using cross-sectional data from the National Health and Nutrition Examination Survey (NHANES) collected between 1999 and 2006, the researchers aimed to evaluate whether zinc intake, as a key nutritional factor, was linked to the prevalence of endometriosis. Zinc is known for its essential roles in immune modulation, antioxidative defense, and regulation of matrix metalloproteinases (MMPs), all of which are implicated in endometriosis progression.

Who was studied?

The study included 4,315 American women aged 20–54 years, of whom 331 were diagnosed with endometriosis based on self-reported doctor diagnoses. Participants’ dietary zinc intake was assessed using 24-hour dietary recall interviews, with additional data on demographics, lifestyle, and health covariates collected. Women with extreme caloric intakes or incomplete data were excluded to ensure robustness of results.

What were the most important findings?

The study revealed a positive correlation between higher dietary zinc intake and the risk of endometriosis. Women consuming over 14 mg/day of zinc had a significantly higher adjusted odds ratio (1.60, 95% CI: 1.12–2.27, p = 0.009) compared to those with intake ≤8 mg/day. Zinc’s dual role in immune modulation and antioxidative defense was emphasized, particularly its regulation of matrix metalloproteinases (MMPs) like MMP-2 and MMP-9, which are key enzymes in tissue remodeling and endometriotic lesion invasion. Interestingly, despite zinc’s known antioxidative and anti-inflammatory roles, excessive intake appeared to have a counterproductive effect. These nuanced findings highlight zinc’s complex role in endometriosis pathophysiology.

What are the greatest implications of this study?

This research underscores the potential for dietary zinc as both a marker and modifiable factor in endometriosis risk. It raises questions about zinc’s dualistic effects, where optimal levels may support immune health, but excess intake could exacerbate estrogen-related pathways in endometriosis. Clinicians should be cautious when recommending zinc supplementation for reproductive health, particularly in populations at risk for endometriosis. Furthermore, this study strengthens the biological plausibility of microbiome involvement in endometriosis, as zinc is a crucial cofactor for microbial activity, and its imbalance may alter the gut and pelvic microbiota implicated in the disease.

Fruit and vegetable consumption and risk of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • STOPs
    STOPs

    A STOP (Suggested Termination Of Practices) is a recommendation that advocates for the discontinuation of certain medical interventions, treatments, or practices based on emerging evidence indicating that these may be ineffective, harmful, or counterproductive in the management of specific conditions.

Higher fruit intake, especially citrus fruits, was inversely associated with laparoscopically confirmed endometriosis, suggesting a protective effect potentially linked to beta-cryptoxanthin. In contrast, cruciferous vegetables were linked to increased risk, highlighting the complex interplay between diet and endometriosis risk factors.

What Was Studied

This study explored the potential link between the consumption of fruits and vegetables and the risk of laparoscopically confirmed endometriosis. Using data collected from the Nurses' Health Study II, the researchers analyzed dietary habits over a 22-year period, investigating whether certain food groups and nutrients influenced the likelihood of developing endometriosis.

Who Was Studied

Participants included premenopausal women aged 25–42 years who were enrolled in the Nurses' Health Study II cohort. These women completed biennial surveys assessing health status, lifestyle factors, and dietary intake. Those with a history of endometriosis, cancer, infertility, or hysterectomy were excluded from the analysis, ensuring a focused evaluation of diet and disease development.

Most Important Findings

The study found an inverse relationship between fruit consumption, particularly citrus fruits, and the risk of endometriosis. Women who consumed citrus fruits frequently were less likely to develop endometriosis. Conversely, no significant association was found between total vegetable intake and the disease. Cruciferous vegetables, however, were unexpectedly linked to an increased risk. Beta-cryptoxanthin, a nutrient found in citrus fruits, appeared to play a protective role, and the beneficial effects of fruit consumption were especially notable among participants who had a history of smoking. These findings suggest a potential role for specific dietary components in either mitigating or exacerbating the risk of endometriosis.

Implications

The findings highlight the importance of dietary considerations in understanding endometriosis risk. The protective association of citrus fruits underscores the potential of targeted nutritional interventions to reduce risk. The increased risk observed with cruciferous vegetables raises questions about the role of gastrointestinal symptoms, as these vegetables are high in fermentable oligosaccharides, which could exacerbate symptoms and lead to increased diagnosis rates. Future studies exploring these dietary patterns in greater depth are warranted to clarify the underlying mechanisms and to guide dietary recommendations for those at risk.

Gut microbiota imbalance and its correlations with hormone and inflammatory factors in patients with stage 3/4 endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study explores the gut microbiota imbalance and its correlations with hormone and inflammatory factors in stage 3/4 endometriosis. Key findings reveal distinct microbial shifts linked to hormonal dysregulation and inflammation, offering insights into disease mechanisms and potential microbiome-targeted therapeutic approaches.

What Was Studied?

This study explored the associations between gut microbiota imbalances and hormone and inflammatory factors in patients with stage 3/4 endometriosis (EM). Conducted at Changhai Hospital, Shanghai, the research aimed to determine how gut microbiome alterations correlate with hormone levels and inflammatory markers in women suffering from moderate to severe endometriosis. Using 16S rRNA high-throughput sequencing, researchers analyzed stool samples to compare the gut microbial composition between 12 women diagnosed with stage 3/4 EM and 12 healthy controls. Blood samples were collected to measure serum hormone levels, including estradiol (E2), and inflammatory cytokines, notably IL-8. The primary objective was to identify microbial shifts associated with EM and understand their correlation with hormone imbalances and inflammation, key factors in the pathogenesis of endometriosis.

Who Was Studied?

The study recruited 12 women with a histological diagnosis of stage 3/4 endometriosis from Changhai Hospital and 12 healthy controls, matched for age (18–40 years) and menstrual regularity. Inclusion criteria for the EM group required confirmed diagnoses of moderate to severe endometriosis per the American Fertility Society Revised Classification (1997). All participants were Han women living in Shanghai, with strict exclusion criteria including recent antibiotic or probiotic use, hormonal therapy, pregnancy, and any comorbid gastrointestinal conditions like inflammatory bowel disease. To minimize confounding factors, participants followed a uniform carbohydrate-based diet three days before sampling, and stool samples were collected within three to five days post-menstruation to account for hormonal fluctuation.

What Were the Most Important Findings?

The study found that women with stage 3/4 endometriosis exhibited a significantly altered gut microbiota profile compared to healthy controls. Notably, the EM group had lower α diversity, indicating reduced microbial richness and variation. At the phylum level, the ratio of Firmicutes to Bacteroidetes was markedly increased in endometriosis patients (3.55 vs. 1.99 in controls), suggesting dysbiosis. The abundance of Actinobacteria, Cyanobacteria, Saccharibacteria, Fusobacteria, and Acidobacteria was significantly higher in the EM group, while Tenericutes were significantly reduced. At the genus level, Bifidobacterium, Blautia, Dorea, Streptococcus, and [Eubacterium] hallii_group showed notable increases, whereas Lachnospira and [Eubacterium] eligens_group were depleted in endometriosis patients. Among the unique genera, Prevotella_7 dominated the EM group, while Coprococcus_2 was prevalent in controls.

Additionally, serum analyses revealed that estradiol (E2) and IL-8 levels were significantly higher in endometriosis patients. Correlation analysis indicated that Blautia and Dorea were positively correlated with elevated E2 levels, while Subdoligranulum abundance inversely correlated with IL-8 levels. These microbial shifts also corresponded with enhanced expression of microbial pathways related to "environmental information processing," "endocrine system," and "immune system," highlighting potential links between gut microbiota and hormonal regulation in endometriosis.

What Are the Greatest Implications of This Study?

The findings of this study suggest that gut microbiota imbalances are closely linked with hormone and inflammatory dysregulation in patients with stage 3/4 endometriosis. The observed microbial shifts, particularly the elevated Firmicutes/Bacteroidetes ratio and increased levels of Bifidobacterium, Blautia, Dorea, and Streptococcus, indicate a state of dysbiosis that may exacerbate inflammatory responses and hormonal imbalances. The positive correlation between Blautia and Dorea with estradiol levels points to the gut microbiome's role in modulating estrogen, potentially influencing the development and progression of endometriosis. Furthermore, the association of Subdoligranulum with IL-8 levels suggests a microbial influence on inflammatory cytokine production, which is known to contribute to endometriosis pathophysiology. These insights provide a foundation for exploring microbiome-targeted therapies aimed at restoring microbial balance and modulating hormonal and inflammatory responses in endometriosis patients. This study also underscores the need for further clinical investigations to validate these microbial markers as diagnostic or therapeutic targets.

I Am the 1 in 10—What Should I Eat? A Research Review of Nutrition in Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review explores how dietary modifications impact endometriosis progression. Antioxidants, omega-3s, and anti-inflammatory diets show promise in symptom relief and hormonal regulation. Personalized nutrition emerges as a pivotal tool for improving patient outcomes.

What Was Reviewed?

The paper titled "I Am the 1 in 10—What Should I Eat? A Research Review of Nutrition in Endometriosis" provides an extensive review of the role of nutrition in the management and progression of endometriosis. The authors systematically explore various dietary factors and interventions, including antioxidants, polyphenols, omega-3 fatty acids, a low-nickel diet, and the Mediterranean diet, among others, in relation to their effects on inflammation, hormonal modulation, and oxidative stress in endometriosis patients.

Who Was Reviewed?

The review primarily evaluated research studies involving women diagnosed with endometriosis. It integrated findings from human clinical trials, observational studies, and in vitro research to synthesize current evidence on nutritional influences on endometriosis-related symptoms and disease progression.

What Were the Most Important Findings?

The review highlighted that endometriosis is a chronic inflammatory and estrogen-dependent condition where dietary modifications can play a pivotal role. It emphasized the following:

Antioxidants and Polyphenols: Foods rich in antioxidants, such as fruits, vegetables, and specific compounds like resveratrol, demonstrated anti-inflammatory and pro-apoptotic effects in reducing endometriosis severity. Polyphenols, especially phytoestrogens, can modulate estrogen activity, impacting endometriotic lesion growth.

Dietary Fats: Omega-3 fatty acids were shown to reduce inflammation and dysmenorrhea, while high consumption of omega-6 and trans fats increased risks.

Specific Diets: The Mediterranean diet, with its anti-inflammatory properties, low-FODMAP and low-nickel diets showed potential benefits in reducing gastrointestinal and systemic symptoms. Gluten-free diets also alleviated pain in a subset of patients.

Dairy and Vitamin D: Dairy consumption, particularly calcium- and vitamin D-rich products, was associated with reduced endometriosis risk. Vitamin D showed immunomodulatory effects, improving inflammatory responses.

Red Meat and Iron Overload: Excessive red meat consumption elevated estrogen and prostaglandin levels, exacerbating endometriosis. The condition was also linked to iron overload in peritoneal fluid, contributing to oxidative stress and infertility.

What Are the Greatest Implications of This Review?

The findings underscore the potential for personalized dietary interventions in endometriosis management, emphasizing the integration of anti-inflammatory, low-toxin, and nutrient-rich foods. Clinicians can leverage these insights to recommend diets tailored to reduce inflammation, regulate estrogen metabolism, and mitigate oxidative stress, thereby improving quality of life and fertility outcomes for patients. Furthermore, the review reinforces the importance of microbiome-targeted dietary strategies in addressing endometriosis-related dysbiosis.

Identification of distinct stool metabolites in women with endometriosis for non-invasive diagnosis and potential for microbiota-based therapies

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies stool-based biomarkers for endometriosis diagnosis and highlights the therapeutic potential of 4-hydroxyindole, a microbiota-derived metabolite, in reducing inflammation and lesion progression.

What Was Studied?

The study examined the stool metabolome of women with endometriosis compared to healthy controls to identify microbiota-derived metabolites with diagnostic and therapeutic potential. Researchers employed metabolomics and microbiota profiling to investigate how altered gut microbiota and their metabolites, specifically 4-hydroxyindole (4HI), affect endometriosis development and progression.

Who Was Studied?

The study included stool samples from 18 women with clinically confirmed endometriosis and 31 healthy control women. The participants’ metabolomic and microbiota profiles were analyzed, followed by functional validation of key metabolites, such as 4HI, in murine and human xenograft models of endometriosis.

What Were the Most Important Findings?

The study identified a distinct stool metabolome in women with endometriosis, characterized by reduced levels of specific microbiota-derived metabolites, including 4HI. Reduced 4HI was linked to a decreased abundance of beneficial gut bacteria like Faecalibacterium and Lachnospiraceae. 4HI showed remarkable therapeutic potential, as it inhibited the initiation and progression of endometriotic lesions, reduced lesion size and volume, and alleviated inflammation and pain in murine models. Moreover, 4HI was effective in regressing well-developed lesions in pre-clinical models. The metabolic signature also highlighted overlap between endometriosis and inflammatory bowel disease (IBD), suggesting common inflammatory pathways and potential misdiagnosis risks.

What Are the Greatest Implications of This Study?

This research revolutionizes the understanding of endometriosis by identifying stool-based biomarkers, particularly 4HI, for non-invasive diagnosis. It highlights 4HI as a promising therapeutic metabolite capable of reducing inflammation and lesion development. These findings pave the way for microbiota-targeted therapies and emphasize the diagnostic overlap with IBD, offering potential for dual-disease screening. Integrating microbiota and metabolomic data offers a novel paradigm for managing endometriosis and related conditions.

Inflammatory cytokines IL-6, IL-10, IL-13, TNF-α and peritoneal fluid flora were associated with infertility in patients with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study reveals that inflammatory cytokines (IL-6, IL-10, IL-13, and TNF-α) are significantly elevated in the peritoneal fluid of endometriosis patients with infertility. These findings suggest their potential role as diagnostic biomarkers and therapeutic targets for managing reproductive complications in endometriosis.

What Was Studied?

This study investigated the association of inflammatory cytokines and peritoneal fluid flora with infertility in endometriosis patients. Researchers aimed to explore the correlations between elevated inflammatory markers—IL-6, IL-10, IL-13, and TNF-α—and changes in microbial communities within the peritoneal fluid of women suffering from endometriosis-related infertility. Using Ion Torrent PGM platform sequencing, the microbial composition of the peritoneal fluid was mapped, while ELISA assays were employed to quantify cytokine levels. Logistic regression analysis was conducted to determine the relationship between inflammatory factors and infertility severity.

Who Was Studied?

The study included 55 women diagnosed with endometriosis and infertility and a control group of 30 women without endometriosis or infertility. All participants were treated at a gynecology clinic between June 2014 and July 2017. Peritoneal fluid samples were collected laparoscopically from the vesicouterine and rectovaginal pouches. Women receiving hormonal therapy or antibiotics within two months of surgery were excluded to avoid confounding microbial and inflammatory responses. This patient population allowed for a focused analysis of microbial and cytokine-driven inflammation linked to infertility in endometriosis.

What Were the Most Important Findings?

The study found that endometriosis patients with infertility exhibited significantly higher levels of IL-6, IL-10, IL-13, and TNF-α in peritoneal fluid compared to controls (p < 0.05). White blood cell counts, including monocytes, neutrophils, eosinophils, and basophils, were also elevated in the peritoneal fluid of endometriosis patients, supporting the inflammatory hypothesis of disease pathogenesis. Analysis of peritoneal fluid microbiota revealed a predominance of Proteobacteria and Firmicutes, with secondary representation from Actinobacteria, Bacteroidetes, Fusobacterium, and Tenericutes. However, there were no significant differences in the abundance of these phyla between endometriosis patients and controls, suggesting that bacteria themselves are not the primary drivers of infertility in endometriosis. Instead, the study highlighted the role of inflammatory cytokines as critical mediators of infertility. Logistic regression analysis confirmed that IL-6, IL-10, IL-13, and TNF-α were significantly correlated with infertility severity in endometriosis patients, suggesting these cytokines may serve as diagnostic biomarkers for assessing infertility risk. Receiver Operating Characteristic (ROC) analysis showed that IL-6 and TNF-α had the highest diagnostic sensitivity, with AUC values of 0.893 for both, indicating their strong predictive value for identifying infertility risk in endometriosis.

ParameterFindings in Endometriosis Patients with Infertility
Inflammatory CytokinesElevated levels of IL-6, IL-10, IL-13, and TNF-α in peritoneal fluid (p < 0.05).
Immune Cell CountsIncreased monocytes, neutrophils, eosinophils, and basophils in peritoneal fluid.
Peritoneal Fluid MicrobiotaDominated by Proteobacteria and Firmicutes, with secondary presence of Actinobacteria, Bacteroidetes, Fusobacterium, and Tenericutes.
Microbial ShiftsNo significant differences in bacterial phyla between endometriosis patients and controls.
Diagnostic BiomarkersIL-6 and TNF-α showed the highest diagnostic sensitivity for infertility with AUC values of 0.893.
Inflammatory ImplicationsCytokines linked to immune activation, peritoneal inflammation, and possible inhibition of sperm motility.
Therapeutic PotentialAnti-inflammatory strategies targeting IL-6 and TNF-α could improve fertility outcomes in endometriosis patients.

What Are the Greatest Implications of This Study?

This study underscores the critical role of inflammatory cytokines in endometriosis-associated infertility, particularly IL-6, IL-10, IL-13, and TNF-α. These cytokines appear to contribute to peritoneal inflammation, immune cell activation, and possibly sperm motility inhibition, collectively reducing fertility. Although peritoneal fluid microbiota did not significantly differ between endometriosis and control groups, the elevated cytokine levels suggest that microbial translocation and immune responses within the peritoneal cavity may exacerbate inflammation. These findings highlight IL-6 and TNF-α as potential biomarkers for diagnosing infertility in endometriosis patients and suggest that anti-inflammatory therapies targeting these cytokines could improve fertility outcomes. Additionally, the study indicates that monitoring cytokine levels in peritoneal fluid could serve as a non-invasive diagnostic tool for assessing infertility risk in endometriosis, offering a novel approach to reproductive management in affected women.

Iron Overload and Endometriosis: Mechanisms, Implications, and Therapeutic Targets

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Endometriomas
    Endometriomas

    An endometrioma is a type of ovarian cyst filled with old blood, arising from endometrial tissue outside the uterus, typically causing pain and potentially impacting fertility.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

Iron overload in endometriosis contributes to oxidative stress, inflammation, and tissue damage, driving lesion persistence and subfertility. Ferroptosis resistance and dysregulated iron metabolism highlight therapeutic opportunities using iron chelators and modulators.

What Was Reviewed?

This systematic review evaluated the role of iron in the pathophysiology of endometriosis. The review synthesized findings from 53 studies, including both human and animal research, to provide a comprehensive understanding of how excess iron contributes to oxidative stress, inflammation, and tissue damage in endometriosis. It also explored iron-related mechanisms such as ferroptosis and the implications for subfertility, symptom severity, and potential malignant transformation.

Who Was Reviewed?

The review included a total of 53 studies: 47 human studies involving 3,556 participants and 6 animal studies. The human studies primarily examined women diagnosed with endometriosis, and the included research utilized various bio-samples such as ovarian endometriomas, peritoneal fluid, and ectopic endometrial lesions. Animal studies focused on endometriosis models to explore systemic and local iron mechanics.

Key Findings

Iron overload is consistently found in endometriotic tissues and peritoneal fluid but not in systemic circulation. This localized iron accumulation stems from repeated bleeding within lesions, leading to oxidative stress and inflammation that perpetuates the ectopic growth of endometrial tissue. Dysregulated iron transport and the failure of homeostatic mechanisms contribute to this pathology, with increased expression of proteins such as divalent metal transporter-1 (DMT1) and decreased ferroportin expression in affected tissues.

Markers of oxidative stress, including lipid peroxidation and DNA damage, were significantly elevated in endometriotic lesions. Aberrant resistance to ferroptosis, an iron-dependent form of cell death, was identified as a key mechanism supporting lesion persistence. Additionally, iron-induced ferroptosis was linked to the production of pro-inflammatory and angiogenic factors like IL-8 and VEGFA, exacerbating inflammation and lesion vascularization.

Iron overload was implicated in subfertility, as higher iron concentrations in ovarian follicles and endometriomas were associated with impaired oocyte quality and development. These findings suggest that iron mechanics might influence folliculogenesis and embryo viability. Importantly, the review highlighted the therapeutic potential of iron chelators and ferroptosis modulators for managing endometriosis.

Implications of the Review

This review underscores the central role of aberrant iron metabolism in the pathogenesis of endometriosis, providing a mechanistic basis for its persistence, progression, and associated complications such as subfertility and chronic pain. Iron-related oxidative stress emerges as a critical driver of inflammation and tissue damage, making it a promising target for therapeutic intervention. Future research should explore the efficacy of iron-targeted treatments, such as chelators, and further elucidate the role of ferroptosis in endometriosis. These insights could lead to novel strategies for mitigating symptom severity and improving fertility outcomes in affected women.

Irritable Bowel Syndrome-Like Disorders in Endometriosis: Prevalence of Nickel Sensitivity and Effects of a Low-Nickel Diet. An Open-Label Pilot Study.

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

Nickel allergic contact mucositis was identified in over 90% of endometriosis patients with IBS-like symptoms. A low-nickel diet significantly reduced gastrointestinal, extra-intestinal, and gynecological symptoms, revealing nickel sensitivity as a key driver of endometriosis symptomatology.

What was studied?

This open-label pilot study investigated the prevalence of nickel allergic contact mucositis (Ni ACM) in women with endometriosis who presented with irritable bowel syndrome (IBS)-like symptoms and assessed the efficacy of a low-nickel diet (LNiD) in ameliorating gastrointestinal, extra-intestinal, and gynecological symptoms. The study aimed to determine whether nickel (Ni) sensitivity contributes to the symptom burden in endometriosis and whether dietary nickel restriction can serve as a therapeutic intervention.

Who was studied?

The study initially screened 83 women of reproductive age diagnosed with endometriosis via imaging or laparoscopy who also experienced at least three gastrointestinal symptoms scoring ≥5 on the Gastrointestinal Symptom Rating Scale (GSRS). After applying exclusion criteria (e.g., celiac disease, IgE-mediated food allergies), 47 patients remained eligible. Sixteen dropped out due to the dietary restrictions, leaving 31 who completed the study. Each participant underwent a nickel oral mucosa patch test (omPT) to identify Ni ACM. Of the 31 patients, 28 (90.3%) tested positive. All participants followed a low-Ni diet for three months, with symptoms reassessed using the GSRS at baseline (T0) and after the intervention (T1).

What were the most important findings?

Nickel ACM was highly prevalent among women with endometriosis and IBS-like symptoms, with 90.3% of study completers testing positive via omPT. All patients who adhered to a three-month low-nickel diet experienced statistically significant reductions in all 15 gastrointestinal symptoms, including bloating, abdominal pain, diarrhea, and constipation. Additionally, the LNiD led to meaningful improvements in seven extra-intestinal symptoms such as headache, fatigue, and joint pain, as well as in hallmark gynecological symptoms of endometriosis: chronic pelvic pain, dysmenorrhea, and dyspareunia. These improvements underscore a systemic role of nickel sensitivity in the symptomatology of endometriosis beyond localized pelvic pathology.

From a microbiome perspective, Ni ACM reflects a low-grade inflammatory response that disrupts mucosal immune regulation and intestinal barrier integrity—two key mechanisms implicated in microbial dysbiosis. Though microbiome composition was not directly assessed, the systemic inflammatory profile induced by dietary nickel may favor enrichment of nickel-tolerant pathobionts, including certain Gammaproteobacteria and urease-producing bacteria, while impairing barrier-supporting commensals. This aligns with broader hypotheses on the metallomic drivers of endometriosis-associated dysbiosis.

What are the greatest implications of this study?

This study provides compelling evidence that nickel hypersensitivity may be a clinically significant and previously underrecognized contributor to the gastrointestinal and systemic symptom burden in endometriosis. It positions Ni ACM not only as a comorbidity but as a potential driver of symptom exacerbation, offering a new lens through which to understand IBS-like manifestations in endometriosis. Importantly, the successful use of a targeted dietary intervention based on objective testing (omPT) introduces a personalized medicine framework that could improve quality of life while minimizing unnecessary dietary restrictions, such as those imposed by low-FODMAP diets. If validated in larger, randomized cohorts, the incorporation of nickel testing and dietary counseling into standard endometriosis management could represent a low-risk, high-reward clinical advance.

Lactoferrin: a secret weapon in the war against pathogenic bacteria

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review definitively showcases lactoferrin's antimicrobial prowess against resistant bacteria, detailing its mechanisms, effects on virulence factors like biofilms, and potential to restore microbial balance, marking it as a vital therapeutic ally for clinicians.

What Was Reviewed?

This definitive review meticulously examined the antimicrobial properties of lactoferrin (Lf), a cationic glycoprotein integral to the innate immune system, and its potential as a therapeutic weapon against pathogenic bacteria—particularly in the face of escalating antimicrobial resistance. The authors delved into Lf's multifaceted mechanisms of action, which include iron sequestration to starve bacteria, direct disruption of bacterial membranes leading to lysis, and the generation of potent antimicrobial peptides like lactoferricins (Lfcins) and lactoferrampin (Lfampin). The review also explored Lf's ability to neutralize bacterial virulence factors, such as lipopolysaccharides (LPS), secretion systems, and biofilms, across a diverse range of Gram-positive and Gram-negative bacteria. Additionally, it highlighted Lf's synergistic potential with antibiotics, positioning it as a promising adjuvant in antimicrobial therapies. By synthesizing evidence from in vitro, in vivo, and clinical studies, the review underscored Lf's versatility and safety, emphasizing its approval as a dietary supplement and its accessibility for clinical use.

Who Was Reviewed?

The review encompassed a broad spectrum of studies investigating lactoferrin's antibacterial activity, primarily focusing on in vitro experiments with clinically relevant bacterial species. Key pathogens included Escherichia coli (notably enteropathogenic E. coli [EPEC] and enteroaggregative E. coli [EAEC]), Salmonella Typhimurium, Klebsiella pneumoniae, Pseudomonas aeruginosa, Streptococcus mutans, Streptococcus pneumoniae, and Helicobacter pylori, among others. It also included research on Lf's effects on bacterial virulence mechanisms, such as biofilm formation and secretion systems, as well as studies exploring its synergistic potential with antibiotics. While the review emphasized laboratory findings, it also touched upon the clinical applications of bovine Lf (bLf), which has been approved as a dietary supplement, highlighting its relevance for both human and veterinary medicine.

What Were the Most Important Findings?

The review conclusively demonstrated that lactoferrin (Lf) and its derived peptides exhibit broad-spectrum antimicrobial and anti-biofilm activity against a wide array of pathogenic bacteria, significantly impacting microbiome-relevant taxa. Specific targets include Escherichia coli, Salmonella Typhimurium, Klebsiella pneumoniae, Pseudomonas aeruginosa, Streptococcus mutans, Streptococcus pneumoniae, Streptococcus agalactiae (GBS) and Helicobacter pylori, alongside others like Bacteroides fragilis and Bacteroides thetaiotaomicron, which are notable in gut microbiota contexts. Lf's mechanisms extend beyond iron sequestration to include direct membrane disruption, inhibition of bacterial adhesion to host cells, and suppression of biofilm formation—a critical factor in chronic infections and dysbiosis. For instance, Lf disrupts biofilms in P. aeruginosa, S. pneumoniae, and S. mutans, key players in microbiome imbalance. It also impairs virulence by targeting secretion systems and proteases in pathogens like H. pylori and Porphyromonas gingivalis. Although not directly focused on microbiome signatures, Lf's ability to modulate bacterial communities—particularly by inhibiting pathogens and biofilms—suggests a pivotal role in maintaining microbial balance in mucosal sites like the gut and oral cavity, reinforced by its presence in breast milk and potential influence on infant microbiome development.

What Are the Greatest Implications of this Review?

The review firmly establishes lactoferrin (Lf) as a versatile, nontoxic therapeutic agent with profound implications for combating antimicrobial resistance, offering clinicians a robust alternative or adjuvant to conventional antibiotics. Its broad-spectrum activity against multidrug-resistant bacteria, without reported resistance development, positions Lf as a critical tool for managing infections where traditional therapies falter. For clinicians focused on microbiome health, Lf's capacity to inhibit biofilms and virulence factors in pathogens like Pseudomonas aeruginosa and Streptococcus pneumoniae suggests it could restore microbial equilibrium in dysbiotic states, such as in the gut or oral cavity. Its safety, affordability, and regulatory approval as a dietary supplement enhance its clinical accessibility for prevention and treatment. Future research into Lf's specific impacts on microbiome signatures and potential synergies with probiotics could further amplify its utility, bridging microbiome research with practical therapeutic applications.

Metabolomics reveals perturbations in endometrium and serum of minimal and mild endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metabolomic Signature
    Metabolomic Signature

    Metabolomic signatures are unique metabolite patterns linked to specific biological conditions, identified through metabolomics. They reveal underlying biochemical activities, aiding in disease diagnosis, biomarker development, and personalized medicine. The microbiome significantly affects these signatures, influencing health and disease outcomes through metabolic interactions.

This study highlights metabolic alterations in endometrial tissue and serum of minimal and mild endometriosis patients, identifying amino acid biomarkers with diagnostic potential. A combined serum panel demonstrated 100% sensitivity and 83% specificity for Stage II diagnosis, offering a promising step toward non-invasive early detection of endometriosis.

What Was Studied?

This study investigated the metabolic perturbations in eutopic endometrial tissue and serum of women with minimal and mild endometriosis (Stages I and II) using ^1H Nuclear Magnetic Resonance (NMR)-based metabolomics. The researchers aimed to identify specific metabolites that could be potential biomarkers for the early, non-invasive diagnosis of endometriosis. The study included multivariate and univariate analyses to identify metabolite changes and their diagnostic potential.

Who Was Studied?

The study included 95 women diagnosed with endometriosis (staged using the revised American Society for Reproductive Medicine criteria) and 24 healthy fertile controls. The participants were recruited from Eastern India and Bangladesh, with exclusion criteria ensuring no confounding conditions such as ovarian tumors or pelvic inflammatory disease. Blood and eutopic endometrial tissue samples were collected during the mid-secretory phase of the menstrual cycle.

What Were the Most Important Findings?

Women with minimal and mild endometriosis exhibited significant metabolic alterations, particularly in amino acids. Alanine, lysine, leucine, proline, and phenylalanine levels were notably dysregulated in serum, with tissue samples showing lower levels of these metabolites, except for proline, which positively correlated with serum levels. Alanine alone demonstrated diagnostic potential for Stage I endometriosis, with 90% sensitivity and 58% specificity.

For Stage II, phenylalanine achieved 100% sensitivity but had lower specificity, while a combined panel of metabolites improved diagnostic accuracy, reaching 100% sensitivity and 83% specificity.

In advanced stages, elevated taurine and myo-inositol levels were linked to increased cell proliferation and angiogenesis, highlighting similarities with tumorigenic processes. These findings underscore the critical role of metabolic shifts in endometriosis progression, particularly involving amino acids and nucleotide synthesis, and suggest their utility in early detection and non-invasive diagnostics.

What Are the Greatest Implications of This Study?

This study underscores the potential of metabolomic signatures in elucidating the pathophysiology of endometriosis and developing non-invasive diagnostic tools, especially for early stages where traditional biomarkers like CA-125 are less effective. By identifying a panel of serum metabolites, the research provides a foundation for improving diagnostic accuracy and reducing the need for invasive laparoscopy. Additionally, the observed metabolic similarities between endometriosis and malignancies could inspire further exploration of shared mechanisms, potentially broadening therapeutic targets.

Microbiome of the lower genital tract in Chinese women with endometriosis by 16s-rRNA sequencing technique: a pilot study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies microbiome shifts in the lower genital tract of Chinese women with endometriosis, marked by Atopobium enrichment and changes in ribosome biogenesis and immune modulation. Findings suggest potential non-invasive biomarkers and therapeutic targets for endometriosis and adenomyosis.

What Was Studied?

This study investigated the microbiome composition of the lower genital tract in Chinese women with endometriosis using 16S rRNA sequencing. Researchers aimed to explore differences in microbial diversity and specific bacterial populations between women diagnosed with endometriosis and healthy controls. The analysis focused on samples collected from the cervical canal, posterior fornix, and uterine cavity to identify distinct microbial profiles and potential biomarkers associated with endometriosis and adenomyosis. A total of 68 participants were included in the study, with 134 samples collected and processed for microbiome analysis.

Who Was Studied?

The study involved 68 women aged 18 to 45, divided into groups: 20 with endometriosis (EM), 19 with adenomyosis (AM), 7 with both adenomyosis and endometriosis (AMEM), and 36 healthy controls (CT). Samples were collected from the cervical canal, posterior fornix, and uterine cavity using sterile techniques to minimize contamination. Participants were excluded if they had bacterial vaginosis (BV), pelvic inflammatory disease, recent antibiotic use, or other infections that might influence microbial composition. This design allowed for a focused investigation of microbiota differences in the lower genital tract specifically related to endometriosis and adenomyosis.

What Were the Most Important Findings

The study found that Lactobacillus remained the predominant genus in the lower genital tract across all groups, reflecting typical vaginal flora. However, significant microbial shifts were observed in women with adenomyosis-endometriosis (AMEM). At the genus level, Atopobium was notably enriched in AMEM patients, representing the most significant microbial difference. While Lactobacillus dominance persisted, AMEM patients exhibited a substantial increase in Coriobacteriaceae and Campylobacteriaceae, indicating a divergence from healthy microbial communities. The AMEM group also displayed elevated levels of Coriobacteriales at the order level, further distinguishing it from other disease groups and controls. Interestingly, LeFSe analysis did not identify specific biomarkers between the cervical canal and posterior fornix, but PICRUSt analysis suggested functional differences, particularly in ribosome biogenesis and two-component system regulation, which may influence immune modulation in endometriosis. The findings suggest that Atopobium may contribute to the pathogenesis of endometriosis through immune disruption, potentially mediated by increased levels of IL-1β, which has been linked to chronic inflammation. The consistent presence of Coriobacteriaceae and Campylobacteriaceae in AMEM further supports a hypothesis of localized dysbiosis contributing to inflammatory processes.

What Are the Greatest Implications of This Study?

This study provides evidence of distinct microbiome alterations in the lower genital tract of women with endometriosis, particularly among those with concurrent adenomyosis. The enrichment of Atopobium and Coriobacteriaceae suggests that specific microbial communities may influence inflammatory pathways and disease progression in endometriosis. The identification of ribosome biogenesis and two-component system regulation as significant functional pathways indicates that microbial-induced immune modulation could play a role in lesion persistence and symptom severity. These findings propose Atopobium as a potential microbial biomarker for adenomyosis-endometriosis and highlight the possibility of targeted microbiome-based therapies to disrupt pathological microbial communities. The study underscores the importance of microbial profiling as a non-invasive diagnostic tool for identifying endometriosis-related dysbiosis, potentially leading to earlier detection and intervention.

Microbiome Profile of Deep Endometriosis Patients: Comparison of Vaginal Fluid, Endometrium and Lesion

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study reveals a distinct microbiome profile in deep endometriosis, with unique bacterial communities in lesion sites, suggesting a role in inflammation and disease progression. Potentially pathogenic genera like Pseudomonas and Alishewanella were abundant, highlighting their possible involvement in endometriotic lesion maintenance and inflammatory responses.

What was studied?

This study explored the microbiome profile in deep endometriosis by comparing the bacterial composition of vaginal fluid, eutopic endometrium, and deep endometriotic lesions. The primary goal was to identify whether distinct microbial patterns exist in these tissue sites of women with deep endometriosis compared to those without the condition. To achieve this, researchers employed high-throughput 16S rRNA sequencing to analyze microbial DNA extracted from tissue samples collected during laparoscopic surgery. The analysis included alpha and beta diversity assessments to determine variations in microbial richness and community structure across different tissue types.

Who was studied?

The study involved 21 participants, including 10 women with histologically confirmed deep endometriosis and 11 healthy controls undergoing laparoscopic surgery for benign gynecological conditions or elective tubal ligation. Samples were obtained from three anatomical sites: vaginal fluid, eutopic endometrium, and deep endometriotic lesions. Participants were carefully screened to exclude those with recent antibiotic, antifungal, or probiotic use, as well as those with autoimmune or inflammatory diseases that could affect microbiome composition.

What were the most important findings?

Microbiome analysis revealed significant differences in bacterial profiles between deep endometriotic lesions, eutopic endometrium, and vaginal fluid. In vaginal fluid and endometrial samples, the microbiome was predominantly composed of Lactobacillus, Gardnerella, Streptococcus, and Prevotella. In contrast, deep endometriotic lesions exhibited a distinct microbial landscape with reduced Lactobacillus and higher relative abundances of Alishewanella, Enterococcus, and Pseudomonas. Notably, Alishewanella and Pseudomonas were almost exclusively found in lesion samples, suggesting these genera may be associated with the inflammatory microenvironment characteristic of deep endometriosis.

Further analysis indicated that deep endometriotic lesions had greater microbial diversity than both vaginal fluid and eutopic endometrium. Beta diversity analysis showed a statistically significant difference in microbial community structure between lesion samples and the other tissue sites (p = 0.036), suggesting that endometriotic tissue supports a unique microbiome that could influence local immune responses and inflammation. These findings point towards a potential role for certain bacteria in the pathogenesis of deep endometriosis, either by promoting inflammation or altering tissue homeostasis.

Microbial GroupDeep Endometriosis FindingsClinical Implications
LactobacillusDecreased in lesion samplesReduction may contribute to loss of protective barrier
AlishewanellaIncreased in lesionsPotential involvement in local inflammation
PseudomonasIncreased in lesionsLinked to immune modulation and tissue inflammation
EnterococcusElevated in lesion samplesKnown for its association with chronic inflammation
Alpha DiversityIncreased in lesions compared to other sitesSuggests a unique microbial community fostering inflammation
Beta DiversitySignificantly different from endometrium and vaginal fluid (p = 0.036)Indicates unique microbial signature in lesions

What are the greatest implications of this study?

The results of this study underscore the presence of a unique microbiome profile in deep endometriotic lesions, characterized by increased microbial diversity and the presence of potentially pathogenic bacteria like Pseudomonas and Alishewanella. These findings suggest that microbiome alterations may contribute to the inflammatory environment observed in endometriosis, potentially influencing disease progression and symptom severity. This study opens avenues for further investigation into microbiome-targeted therapies aimed at modulating bacterial communities in endometriotic tissue to alleviate inflammation and inhibit lesion growth. Additionally, the distinct microbial signatures identified in deep endometriosis lesions may serve as non-invasive biomarkers for early detection and improved clinical management of the disease.

Microbiota composition and distribution along the female reproductive tract of women with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study maps the microbiota composition along the reproductive tract in endometriosis patients, revealing microbial dysbiosis, reduced Lactobacillus, and increased Pseudomonas and Sphingobium in upper reproductive sites. These findings suggest potential microbial contributions to inflammation, lesion persistence, and new biomarkers for diagnosing endometriosis.

What Was Studied?

This study investigated the microbiota composition and distribution along the female reproductive tract of women with endometriosis (EMS) compared to healthy controls. Using 16S rRNA amplicon sequencing, researchers mapped microbial communities from five distinct anatomical sites: the lower third of the vagina (CL), posterior vaginal fornix (CU), cervical mucus (CV), endometrium (ET), and peritoneal fluid (PF). The primary aim was to identify differences in microbial communities across these regions and understand how these alterations may contribute to the pathogenesis of endometriosis. A total of 36 women with confirmed pelvic endometriosis and 14 healthy controls undergoing surgery for benign gynecological conditions were included in the study.

Who Was Studied?

The study included 50 women aged 23 to 44, divided into two groups: 36 patients diagnosed with pelvic endometriosis and 14 healthy controls undergoing laparoscopic surgery for benign conditions like ovarian teratoma, serous cystadenoma, or uterine fibroids. All participants had regular menstrual cycles and had not used hormonal drugs, antibiotics, or vaginal medications within six months before sample collection. Samples were collected during the early follicular phase to minimize hormonal variability, and five distinct anatomical locations were sampled to comprehensively map microbial distribution.

What Were the Most Important Findings?

The study revealed distinct microbiota compositions along the reproductive tract in women with endometriosis compared to healthy controls. The lower reproductive tract (CL, CU) in both groups was predominantly colonized by Lactobacillus, maintaining a stable microbial community. However, microbial diversity began to shift notably at the cervical mucus (CV) in endometriosis patients, with the appearance of Veillonellaceae and an overall reduction in Lactobacillus. This shift continued upward, with dramatic changes observed in the endometrium (ET) and peritoneal fluid (PF) of endometriosis patients. In the ET, Pseudomonas, Acinetobacter, and Vagococcus emerged as dominant taxa, accompanied by a significant reduction of Lactobacillus. The peritoneal fluid samples further diverged, showing high microbial diversity, including Pseudomonas, Acinetobacter, Shewanella, Vagococcus, and Sphingobium, with minimal presence of Lactobacillus. The distinct microbial signatures in these upper reproductive sites suggest a potential role for these genera in promoting inflammation and lesion persistence in endometriosis. Importantly, Sphingobium and Pseudomonas viridiflava were consistently enriched in both the ET and PF of endometriosis patients, indicating their potential as microbial biomarkers for the disease. The findings support the hypothesis that site-specific microbial shifts contribute to the pathogenesis of endometriosis, likely through immune modulation and local inflammation.

Anatomical SiteMicrobiota Findings in Endometriosis Patients
Lower Reproductive Tract (CL, CU)Predominantly colonized by Lactobacillus, maintaining a stable microbial community.
Cervical Mucus (CV)Notable microbial shift with the appearance of Veillonellaceae and a significant reduction in Lactobacillus.
Endometrium (ET)Dominated by Pseudomonas, Acinetobacter, and Vagococcus with a marked reduction of Lactobacillus.
Peritoneal Fluid (PF)High microbial diversity with Pseudomonas, Acinetobacter, Shewanella, Vagococcus, and Sphingobium. Minimal Lactobacillus presence.
Unique EnrichmentsConsistent enrichment of Sphingobium and Pseudomonas viridiflava in ET and PF, suggesting potential biomarkers.

What Are the Greatest Implications of This Study?

The study’s findings suggest that the microbiota composition along the female reproductive tract in endometriosis patients is markedly different from that of healthy controls, with key disruptions beginning in the cervical mucus and intensifying in the endometrium and peritoneal fluid. The reduction of Lactobacillus and the enrichment of pro-inflammatory taxa such as Pseudomonas, Acinetobacter, Vagococcus, Shewanella, and Sphingobium suggest these species may contribute to local immune activation, chronic inflammation, and potentially lesion maintenance in endometriosis. The identification of Sphingobium and Pseudomonas viridiflava as dominant species in upper reproductive tract samples provides a promising direction for non-invasive biomarkers for diagnosing endometriosis. Furthermore, this microbial dysbiosis may offer therapeutic targets for microbiome-modulating treatments aimed at restoring a healthy reproductive tract microbiota, potentially alleviating symptoms and reducing disease progression. This research underscores the importance of targeting microbial ecosystems in understanding and managing endometriosis.

Molecular detection of intrauterine microbial colonization in women with endometriosis

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • STOPs
    STOPs

    A STOP (Suggested Termination Of Practices) is a recommendation that advocates for the discontinuation of certain medical interventions, treatments, or practices based on emerging evidence indicating that these may be ineffective, harmful, or counterproductive in the management of specific conditions.

A case-controlled molecular study on 32 women, half with endometriosis and half without, investigated microbial colonization in the intrauterine environment and ovarian cystic fluid. It found significant bacterial variations, with certain bacteria types increasing or decreasing, particularly after treatment with gonadotropin-releasing hormone agonist (GnRHa). The study suggests GnRHa treatment might promote sub-clinical infections in the intrauterine and ovarian environments.

What was studied?

The research focused on investigating microbial colonization in women’s intrauterine environment and cystic fluid, utilizing a molecular approach to detect bacterial presence. This involved examining variations in bacterial types and their implications in the context of endometriosis and treatment effects.

Who was studied?

The study included 32 women, evenly divided between those diagnosed with endometriosis and those without the condition. Each group was further split, with half receiving gonadotropin-releasing hormone agonist (GnRHa) treatment, to explore the treatment’s impact on microbial colonization.

What were the most important findings?

Key findings revealed a broad range of bacterial presence in both endometrial swabs and cystic fluids, with significant changes in bacterial families (decrease in Lactobacillacae and increase in Streptococcaceae, Staphylococaceae, and Enterobacteriaceae) observed in GnRHa-treated women with endometriosis. The 16S metagenome assay was more effective than traditional culture methods, particularly in identifying bacteria in ovarian endometrioma cystic fluid.

What are the greatest implications of this study?

The study’s findings suggest the presence of sub-clinical infections in the intrauterine environment and ovarian endometrioma cystic fluid, particularly following GnRHa treatment. This raises concerns about the potential for GnRHa therapy to promote silent infections, indicating a need for careful consideration and monitoring of such treatments in women with endometriosis.

Molecular detection of intrauterine microbial colonization in women with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study reveals that intrauterine microbial colonization is prevalent in women with endometriosis, particularly with Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae. Findings suggest that GnRHa treatment exacerbates microbial colonization, indicating a possible role for targeted antimicrobial therapies in managing endometriosis-associated inflammation.

What Was Studied?

This study investigated the molecular detection of intrauterine microbial colonization in women with and without endometriosis, utilizing a 16S rDNA metagenome assay to evaluate bacterial presence in endometrial swabs and cystic fluid samples. The primary aim was to explore the role of microbial colonization in the intrauterine environment and its potential contribution to endometriosis pathogenesis, especially in women treated with gonadotropin-releasing hormone agonist (GnRHa). The study compared microbial communities in both endometrial tissue and cystic fluid derived from ovarian endometrioma and non-endometrioma cysts. The analysis aimed to validate the "bacterial contamination hypothesis," which posits that microbial colonization might exacerbate inflammatory responses, contributing to endometriosis progression.

Who Was Studied?

The study included 32 women with endometriosis and 32 women without endometriosis. Among these, half of each group (16 women) received GnRHa treatment for 4–6 months prior to sample collection. Endometrial swabs and cystic fluid samples were collected during laparoscopic procedures at Nagasaki University. Women were included if they were of reproductive age, with regular menstrual cycles, and had not taken antibiotics or immunosuppressants within three months prior to the study. The endometriosis cases were confirmed histologically, and cystic fluids were categorized as either ovarian endometrioma (OE) or non-endometrioma (NE) cysts. The study employed 16S rDNA metagenome sequencing using the Illumina MiSeq system to identify bacterial taxa.

What Were the Most Important Findings?

The study identified a significant alteration in the microbial landscape within the intrauterine environment and ovarian cystic fluid of women with endometriosis, particularly those undergoing GnRHa treatment. Notably, there was a significant decrease in Lactobacillaceae populations (p < 0.01) and a marked increase in Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae (p < 0.05 for each) in GnRHa-treated women with endometriosis. This microbial shift was associated with sub-clinical infection in the uterine cavity and cystic fluid of ovarian endometrioma. Furthermore, the 16S metagenome assay detected higher proportions of Streptococcaceae and Staphylococcaceae in ovarian endometrioma cyst fluid compared to non-endometrioma cysts, suggesting a unique microbial signature linked to inflammatory pathogenesis. Interestingly, traditional bacterial culture methods failed to detect colonies in the cystic fluids, while PCR analysis revealed substantial colonization. This discrepancy indicates that sub-clinical infections in the uterine and ovarian microenvironments may contribute to the chronic inflammatory state characteristic of endometriosis. The study also proposed that GnRHa-induced hypoestrogenism might reduce the expression of antimicrobial peptides in the genitourinary tract, facilitating microbial colonization and chronic inflammation.

ParameterFindings in Endometriosis Patients
Intrauterine Microbial ColonizationSignificant increase in Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae populations in endometrial tissue and ovarian cystic fluid.
GnRHa Treatment EffectsDecrease in Lactobacillaceae populations (p < 0.01) with elevated Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae (p < 0.05).
Cystic Fluid MicrobiomeHigher proportions of Streptococcaceae and Staphylococcaceae in ovarian endometrioma cyst fluid compared to non-endometrioma cysts.
Detection MethodsTraditional bacterial culture failed to detect colonies, but 16S rDNA sequencing revealed substantial colonization.
Hypothesized MechanismGnRHa-induced hypoestrogenism may suppress antimicrobial peptides, facilitating microbial colonization and chronic inflammation.
Pathogenic ImplicationsSuggests sub-clinical infection in uterine and ovarian environments as a contributor to chronic inflammation in endometriosis.
Therapeutic ConsiderationPotential for targeted antimicrobial therapy to mitigate microbial load and reduce inflammation in endometriosis.

What Are the Greatest Implications of This Study?

The findings provide robust evidence that intrauterine microbial colonization—particularly of Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae—is prevalent in women with endometriosis and is significantly heightened with GnRHa treatment. This suggests that silent intrauterine infections could exacerbate inflammatory responses and potentially influence disease progression. Furthermore, the detection of bacterial DNA in ovarian endometrioma cystic fluid indicates that microbial colonization extends beyond the uterine environment, potentially affecting ovarian tissue integrity. These insights propose that targeted antimicrobial therapy might mitigate intrauterine bacterial load, reduce inflammation, and improve disease management. The study challenges the traditional view of the sterile uterine environment, suggesting that the bacterial contamination hypothesis should be revisited as a contributing factor in endometriosis pathogenesis.

Molecular detection of microbial colonization in cervical mucus of women with and without endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies intrauterine microbial colonization in endometriosis, with enriched Streptococcaceae, Staphylococcaceae, and Enterobacteriaceae. GnRHa therapy exacerbated microbial imbalances, highlighting potential therapeutic targets for managing inflammation and infection in endometriosis. Findings suggest microbial modulation may enhance treatment outcomes.

What Was Studied?

This study investigated the molecular detection of microbial colonization in cervical mucus of women with and without endometriosis using next-generation sequencing (NGS) technology. The primary aim was to explore whether specific microbial populations in the cervical mucus are associated with endometriosis and could contribute to its pathogenesis. Researchers collected cervical mucus samples from 30 women with endometriosis and 39 women without the condition. The microbial communities were analyzed through 16S rRNA gene sequencing, alongside Gram staining and real-time PCR to validate the presence of specific bacterial species. This study sought to identify microbial signatures that may play a role in the inflammation and immune responses linked to endometriosis progression.

Who Was Studied?

The study included 69 women of reproductive age, with 30 diagnosed with endometriosis (all classified as r-ASRM stages III-IV) and 39 serving as healthy controls. All participants underwent laparoscopic surgery for diagnosis or benign gynecological conditions. Cervical mucus samples were collected before surgery under sterile conditions to prevent contamination, ensuring accurate representation of the microbiota present. Women with gynecological malignancies, pelvic inflammatory disease, bacterial vaginosis, or recent antibiotic use were excluded to prevent confounding microbial shifts.

What Were the Most Important Findings?

The analysis revealed that cervical mucus in women with endometriosis harbored distinct microbial communities compared to healthy controls. While Lactobacillus spp. remained dominant across all samples, women with endometriosis showed increased populations of Enterobacteriaceae, Streptococcus, Corynebacterium, Pseudomonas, and Flavobacterium. Notably, Enterobacteriaceae and Streptococcus were significantly enriched in endometriosis patients, as confirmed by real-time PCR analysis (p < 0.05). This elevated presence suggests these bacteria may contribute to inflammatory cascades in the cervix and potentially facilitate disease persistence. Additionally, alpha diversity was significantly higher in the cervical mucus of endometriosis patients, indicating a broader microbial distribution. The study also noted that despite high Lactobacillus prevalence (40–60%), the co-existence of pathogenic bacteria like Enterobacteriaceae and Streptococcus was unique to the endometriosis group, hinting at microbial imbalance. These findings support the hypothesis that cervical microbial colonization could be linked to the development and maintenance of endometriosis through immune modulation and inflammatory responses. The study further postulated that the ascent of these pathogens from the cervix into the uterine cavity may exacerbate inflammatory conditions, potentially triggering endometriosis progression via LPS/TLR4 signaling and innate immune activation.

ParameterFindings in Endometriosis Patients
Dominant GeneraLactobacillus spp. remained dominant in both endometriosis patients and controls, with 40–60% prevalence.
Increased GeneraMarked increases in Enterobacteriaceae, Streptococcus, Corynebacterium, Pseudomonas, and Flavobacterium.
Significant EnrichmentEnterobacteriaceae and Streptococcus were significantly enriched (p < 0.05) in the endometriosis group.
Alpha DiversityHigher alpha diversity observed in endometriosis patients, indicating broader microbial distribution in cervical mucus.
Pathogenic Co-ExistenceDespite high Lactobacillus prevalence, Enterobacteriaceae and Streptococcus co-existed exclusively in the endometriosis group.
Inflammatory AssociationsThese pathogens are suspected to drive inflammatory cascades, contributing to lesion persistence and immune dysregulation.
Migration HypothesisPotential pathogen ascent from the cervix to the uterine cavity may exacerbate inflammation and promote endometriosis via LPS/TLR4 signaling and innate immune activation.

What Are the Greatest Implications of This Study?

The study provides compelling evidence that specific microbial communities in cervical mucus—particularly Enterobacteriaceae and Streptococcus—are associated with endometriosis. This microbial imbalance suggests that the cervical microbiome may serve as both a diagnostic biomarker and a therapeutic target for endometriosis management. The findings highlight the possibility of cervical microbial migration into the uterine cavity as a driver of inflammation and lesion growth, underscoring the need for further exploration into microbiota-targeted therapies. By identifying microbial dysbiosis at the cervical level, this study opens the door to non-invasive diagnostic methods and preventive strategies aimed at reducing microbial-induced inflammation in endometriosis patients.

Nickel Allergy as a Risk Factor for Endometriosis

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies nickel allergy as an independent risk factor for endometriosis, highlighting shared immune dysregulation and estrogenic pathways. Using a population-based cohort, researchers found a 2.5-fold increased odds of nickel allergy in women with endometriosis, emphasizing the role of environmental exposures in its pathogenesis.

What Was Studied?

This study examined the association between nickel allergy and endometriosis using a population-based nested case-control design. The research aimed to determine whether nickel allergy is an independent risk factor for endometriosis by analyzing a cohort dataset provided by the South Korean National Health Insurance Service (NHIS), which included approximately 1 million individuals. The study was designed to assess causality and improve upon previous correlational studies.

Who Was Studied?

The study included 4,985 women divided into two groups: 997 women with endometriosis and 3,988 controls matched by age and socioeconomic status. The endometriosis group was identified using diagnostic codes, surgery records, and drug prescriptions between 2009 and 2013, while nickel allergy cases were identified between 2002 and 2008 using specific diagnostic and patch test codes.

What Were the Most Important Findings?

The findings revealed that women with endometriosis had a significantly higher prevalence of nickel allergy (0.8%) compared to the control group (0.3%), with an adjusted odds ratio of 2.474 (95% CI: 1.023–5.988; p = 0.044). The study highlights the estrogenic and immune-modulating properties of nickel, suggesting that elevated blood nickel levels associated with nickel allergy may contribute to endometriosis pathogenesis. Both conditions share immunological mechanisms, including cell-mediated hypersensitivity and immune dysregulation. The study also emphasizes the activity of nickel as a metalloestrogen, which may influence endometriosis through its interaction with estrogen receptors. Despite its low clinical prevalence, the findings support nickel allergy as a potential contributing factor to endometriosis, particularly in the context of autoimmune and estrogenic pathways.

What Are the Greatest Implications of This Study?

This study underscores the link between environmental factors, such as nickel exposure, and the development of endometriosis. Identifying nickel allergy as a risk factor paves the way for further research into environmental triggers and immune-mediated mechanisms in endometriosis. These findings could lead to targeted prevention strategies, such as reducing nickel exposure in at-risk populations, and inform therapeutic approaches that address immune and estrogenic pathways in endometriosis management.

Nickel Sensitivity and Symptom Management in Endometriosis: The Role of a Low-Nickel Diet

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Low‑Nickel Diet (LNiD)
    Low‑Nickel Diet (LNiD)

    A low-nickel diet (LNiD) is a therapeutic dietary intervention that eliminates high-nickel foods, primarily plant-based sources such as legumes, nuts, whole grains, and cocoa, to reduce systemic nickel exposure. It is clinically validated for managing systemic nickel allergy syndrome (SNAS) and nickel-induced eczema. Its relevance is well-established in microbiome modulation, with studies demonstrating clinical benefits in conditions such as endometriosis, fibromyalgia, irritable bowel syndrome, and GERD.

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

This study offers new insights into the potential link between nickel sensitivity and symptom severity in endometriosis, suggesting that a low-nickel diet may be a promising intervention for alleviating associated gastrointestinal and gynecological symptoms.

What Was Studied?

This pilot study investigated the prevalence of nickel (Ni) allergic contact mucositis (ACM) in women with endometriosis who experience gastrointestinal symptoms and evaluated the effects of a low-nickel diet on these symptoms. The study focused on assessing the gastrointestinal, extra-intestinal, and gynecological symptom reductions associated with Ni ACM and dietary interventions.

Who Was Studied?

The study enrolled 84 women of reproductive age diagnosed with endometriosis who reported significant gastrointestinal symptoms. Thirty-one participants completed the study, undergoing a diagnostic nickel oral mucosa patch test (omPT) and a subsequent three-month low-nickel diet intervention. Participants were evaluated using symptom questionnaires both at baseline and after dietary changes.

What Were the Most Important Findings?

The study found that 90.3% of participants tested positive for Ni ACM, suggesting a high prevalence of nickel sensitivity among women with endometriosis. Following three months of adhering to a low-nickel diet, significant reductions in all evaluated symptoms were reported. Gastrointestinal symptoms such as abdominal pain, bloating, and diarrhea showed marked improvement. Extra-intestinal symptoms, including fatigue and headaches, and gynecological symptoms such as pelvic pain and dysmenorrhea, also exhibited statistically significant decreases. These findings indicate that nickel sensitivity may contribute to the symptomatic burden of endometriosis, and dietary interventions targeting nickel can alleviate these issues.

The study suggests a potential mechanistic link between nickel exposure, immune responses, and the exacerbation of endometriosis symptoms. Major microbial associations (MMAs) relevant to this context include those influenced by dietary changes, although specific microbiome alterations were not detailed.

What Are the Greatest Implications of This Study?

This research highlights nickel sensitivity as a significant yet previously under-recognized contributor to gastrointestinal and systemic symptoms in endometriosis patients. The findings suggest that incorporating nickel sensitivity screening and low-nickel dietary recommendations could represent a transformative approach to symptom management in endometriosis. Although the sample size was small, the results offer strong preliminary evidence for revising dietary protocols in clinical practice to include low-nickel guidelines, potentially improving the quality of life for patients.

Oral, Vaginal, and Stool Microbial Signatures in Patients With Endometriosis as Potential Diagnostic Non-Invasive Biomarkers: A Prospective Cohort Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study explores oral, vaginal, and stool microbial signatures in patients with endometriosis, highlighting their potential as non-invasive diagnostic biomarkers. Findings suggest distinct microbial shifts in these sites, offering a promising avenue for early detection and targeted intervention in endometriosis management.

What was studied?

This prospective cohort pilot study examined the oral, vaginal, and stool microbiota of three cohorts: confirmed endometriosis patients (ENDO, n=21), patients with other gynecological conditions but no endometriosis (N-ENDO, n=24), and healthy controls without gynecologic disease (HC, n=19). Using 16S rRNA sequencing, the study sought to identify non-invasive microbial biomarkers that could potentially differentiate individuals with endometriosis from others, with the ultimate goal of developing a diagnostic tool.

Who was studied?

A total of 64 women were studied, all age- and sex-matched. ENDO and N-ENDO participants were recruited from a hospital setting where they underwent laparoscopy with histological confirmation. Healthy controls were recruited from a separate longitudinal study (MothersBabies), with no known gynecological pathology.

Key Findings:

The study revealed significant microbial diversity and compositional differences in oral and stool samples among patients with endometriosis, non-endometriosis gynecologic conditions, and healthy controls, while vaginal samples showed no significant variation. Specifically, alpha diversity was reduced in the stool microbiota of endometriosis patients compared to healthy controls, and beta diversity analysis confirmed that both oral and stool communities were distinctly structured across cohorts. LEfSe analysis identified differentially abundant taxa specific to body site and disease severity. In stool samples, Phascolarctobacterium and Lactobacillus were enriched in endometriosis, with Actinomyces elevated in minimal/mild cases and Paraprevotellaceae in moderate/severe cases.

Oral samples from patients with moderate/severe endometriosis were characterized by a marked increase in Fusobacterium, a genus previously shown to facilitate lesion development in murine models and implicated in human periodontal disease. This is especially relevant given the higher incidence of periodontitis in endometriosis patients. Cardiobacterium was elevated in mild disease. In vaginal samples, the enrichment of Escherichia, Enterococcus, and Tepidimonas supports the bacterial contamination hypothesis, which posits that lipopolysaccharide (LPS)-mediated inflammation may play a role in lesion formation.

Here is a summary of the differentially abundant taxa by body site and disease severity:

Body SiteDifferentially Abundant Taxa
StoolPhascolarctobacteriumLactobacillus ↑ in ENDO; Actinomyces ↑ in minimal/mild; Paraprevotellaceae ↑ in moderate/severe
OralFusobacterium ↑ in moderate/severe ENDO; Cardiobacterium ↑ in minimal/mild ENDO
VaginalEscherichiaEnterococcusTepidimonas ↑ in ENDO

Implications for Microbiome Research and Clinical Practice:

The study underscores the potential for developing a non-invasive diagnostic tool for endometriosis using microbial biomarkers obtained from oral or stool samples. Specific taxa such as Fusobacterium, Escherichia, and Phascolarctobacterium emerged as promising microbial targets for future mechanistic and therapeutic investigations due to their known roles in modulating inflammation and estrogen metabolism. Additionally, the observed enrichment of Lactobacillus in the stool of patients with endometriosis suggests a possible link to estrobolome activity, with implications for enhanced estrogen recycling and disease progression. Furthermore, the detection of overlapping genera in the gut and peritoneal fluid, as reported in other studies, lends support to the hypothesis that intestinal bacterial translocation may contribute to the peritoneal inflammation characteristic of endometriosis.

Overlap Between Irritable Bowel Syndrome Diagnosis and Endometriosis in Adolescents

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

The study links endometriosis with a fivefold increased risk of IBS in adolescents, emphasizing the role of acyclic pelvic pain severity and inflammation as shared mechanisms. Early screening for IBS and microbiome-targeted interventions could improve outcomes in this group.

What Was Studied?

The study investigated the association between endometriosis and irritable bowel syndrome (IBS) in adolescent females. Specifically, it aimed to determine the prevalence of IBS in those with and without surgically confirmed endometriosis and to explore how pelvic pain severity and other comorbidities influenced this relationship.

Who Was Studied?

The study analyzed data from 323 adolescent females under 21 years of age who participated in the "Women’s Health Study: Adolescence to Adulthood," a longitudinal cohort. Participants were grouped based on the presence or absence of surgically confirmed endometriosis and whether they met the diagnostic criteria for IBS, as defined by Rome IV guidelines or self-reported clinician diagnoses.

What Were the Most Important Findings?

The study found that adolescents with endometriosis were five times more likely to have IBS than those without endometriosis (adjusted odds ratio [aOR], 5.26). Among participants with endometriosis, the odds of IBS increased with the severity of acyclic pelvic pain, with each 1-point increase in pain severity raising the odds by 31% (aOR, 1.31). A significant overlap in pain-related comorbidities, including migraines, sleep disturbances, and urinary symptoms, was observed in individuals with both conditions. Moreover, central pain sensitization, driven by visceral hypersensitivity and alterations in the peripheral and central nervous systems, was identified as a potential shared mechanism between endometriosis and IBS. This study's microbiome associations highlight the role of inflammatory processes in both conditions. Low-grade mucosal inflammation and mast cell activation, often linked to microbiome dysbiosis, were implicated as contributing factors to the development of IBS in the context of endometriosis.

What Are the Greatest Implications of This Study?

This research underscores the need for integrated screening and management strategies for adolescents presenting with endometriosis and/or IBS. Identifying overlapping symptoms early could reduce diagnostic delays and improve patient outcomes. Furthermore, the findings suggest potential pathways for therapeutic interventions targeting the microbiome and immune regulation, such as central sensitization and inflammation. Clinicians should consider IBS in the differential diagnosis for adolescents with severe acyclic pelvic pain, even in the absence of endometriosis.

Persistent Organic Pollutants and Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study links persistent organic pollutants (POPs) to metabolic alterations in deep endometriosis, identifying trans-nonachlor and 2-hydroxybutyrate as key markers.

What Was Studied?

This study explored the relationship between persistent organic pollutants (POPs) and the risk of surgically confirmed deep endometriosis by integrating high-resolution metabolomic profiling. It aimed to characterize metabolic changes associated with POP exposure, focusing on polychlorinated biphenyls (PCBs), organochlorinated pesticides (OCPs), and per-/polyfluoroalkyl substances (PFAS). The researchers utilized advanced analytical techniques such as gas and liquid chromatography coupled with high-resolution mass spectrometry (HRMS) and nuclear magnetic resonance (NMR).

Who Was Studied?

A hospital-based case-control cohort in France was recruited, consisting of women with surgically confirmed deep endometriosis and matched controls without the condition. Serum samples were collected from these participants to measure POP levels and conduct comprehensive metabolomic profiling. The study controlled for confounding variables such as demographic and lifestyle factors, ensuring a robust statistical analysis.

What Were the Most Important Findings?

The study identified significant links between specific POPs and endometriosis risk. Trans-nonachlor, an organochlorinated pesticide, emerged as the most strongly associated pollutant, doubling the risk of deep endometriosis. Other key POPs included PCBs 180 and 167. Metabolomic profiling revealed distinctive metabolic disruptions in women with endometriosis. These included elevated serum levels of lactate, ketone bodies, multiple amino acids, reduced bile acids, phosphatidylcholines (PCs), cortisol, and hippuric acid. A noteworthy finding was the metabolite 2-hydroxybutyrate, which correlated with both trans-nonachlor exposure and endometriosis risk, acting as a potential biomarker of the disease and its environmental exposure.

What Are the Greatest Implications of This Study?

This study is groundbreaking in linking POP exposure to metabolic alterations in deep endometriosis, suggesting an environmental component to the disease's pathogenesis. The findings highlight the potential of metabolomic biomarkers, like 2-hydroxybutyrate, for early diagnosis and monitoring of environmental risk factors. These results emphasize the importance of further research to clarify causal relationships and develop interventions to reduce exposure to harmful pollutants. Clinically, integrating metabolomic and environmental data could improve risk assessment and individualized treatment approaches for endometriosis patients.

Plants as source of new therapies for endometriosis: a review of preclinical and clinical studies

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review highlights the potential of plant-derived therapies for endometriosis. Key findings include anti-inflammatory, anti-proliferative, and anti-angiogenic effects of herbal extracts and bioactive compounds, offering safer long-term alternatives to conventional treatments.

What Was Reviewed?

This review systematically examined plant-derived agents and their potential for treating endometriosis. The authors focused on three main categories: herbal extracts, specific plant-derived bioactive compounds, and Chinese herbal medicine (CHM). By analyzing preclinical and clinical studies, the review assessed the efficacy, mechanisms of action, and clinical potential of these agents, including compounds such as resveratrol, epigallocatechin-3-gallate, curcumin, and cannabinoids. The paper aimed to critically evaluate the relevance of natural therapies as safer, long-term alternatives to conventional treatments for endometriosis.

Who Was Reviewed?

The review covered studies involving various experimental models, including human cell lines, rodent models of endometriosis, and limited clinical trials on human subjects. These studies collectively investigated the effects of plant-derived agents on cellular and molecular markers of endometriosis, such as inflammation, angiogenesis, and apoptosis. The review also discussed findings from clinical trials of Chinese herbal medicine and individual bioactive compounds.

What Were the Most Important Findings?

The review identified several plant-derived agents with significant potential for endometriosis therapy. Herbal extracts such as pueraria flower extract (PFE) and aged black garlic exhibited anti-inflammatory, anti-angiogenic, and anti-proliferative effects in experimental models. Bioactive compounds like resveratrol and curcumin demonstrated pleiotropic effects, targeting processes like estrogen modulation, oxidative stress reduction, and inhibition of vascular endothelial growth factor (VEGF) expression. Chinese herbal medicine formulations were found to alleviate symptoms, reduce lesion size, and prevent recurrence in clinical contexts. Mechanistically, these agents influence key pathways involving cytokines (IL-6, IL-8, TNF-α), transcription factors (NF-κB), and matrix metalloproteinases (MMPs), making them promising candidates for integrative treatment strategies.

What Are the Greatest Implications of This Review?

The findings emphasize the need for standardized protocols and further clinical trials to validate the safety and efficacy of plant-derived therapies in human endometriosis patients. The review underscores the potential of these agents as part of multimodal treatment strategies, offering reduced side effects and improved long-term management compared to conventional hormonal or surgical approaches. Additionally, the pleiotropic action of these agents aligns with the complex pathophysiology of endometriosis, addressing inflammation, angiogenesis, and cellular survival concurrently.

Plants as source of new therapies for endometriosis: a review of preclinical and clinical studies

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review assesses plant-derived agents for endometriosis therapy, focusing on mechanisms like inflammation, angiogenesis, and apoptosis. Key agents such as resveratrol and curcumin show preclinical promise, while Chinese herbal medicine (CHM) demonstrates clinical utility.

What Was Reviewed?

This review comprehensively evaluates plant-derived agents as potential therapies for endometriosis. It focuses on herbal extracts, specific plant bioactive compounds, and Chinese herbal medicine (CHM) formulations, assessing their mechanisms of action, therapeutic potential, and preclinical and clinical evidence supporting their use. The authors aim to establish these agents as alternatives to current treatments with fewer side effects and long-term efficacy.

Who Was Reviewed?

The studies reviewed include human endometriotic cell lines, surgically induced endometriosis models in animals, and clinical trials involving human participants. The scope of the review is broad, encompassing various agents such as Pueraria flower extract (PFE), curcumin, resveratrol, and CHM formulations, along with their effects on biological processes like inflammation, angiogenesis, oxidative stress, and apoptosis.

Summary of Plant-Derived Agents

The table below summarizes the plant-derived agents, their models, mechanisms of action, and key findings.

Plant/CompoundModel StudiedMechanism of ActionKey Findings
Pueraria Flower Extract (PFE)Human endometriotic cell lines, Balb/c miceAnti-proliferative, anti-inflammatoryReduced lesion size, suppressed MMP-2 and MMP-9 expression, and cell migration.
ResveratrolHuman cell lines, murine models, small clinical trialsAnti-angiogenic, anti-inflammatory, pro-apoptoticReduced VEGF expression, lesion size, and inflammatory cytokines (IL-6, TNF-α).
CurcuminHuman cell lines, rat models, clinical trialsAnti-inflammatory, anti-angiogenicReduced IL-6, IL-8, and VEGF; improved oxidative stress and apoptosis markers.
SilymarinRat modelsAnti-proliferative, antioxidantInduced lesion regression, apoptosis, and fibrosis while inhibiting angiogenesis.
Acai ExtractSprague-Dawley ratsAnti-inflammatory, anti-angiogenicReduced lesion size and markers such as VEGF and COX-2.
Chinese Herbal MedicineHuman clinical trialsMulti-targeted (anti-inflammatory, hormonal balance)Reduced pain and postoperative recurrence, improved quality of life.

These agents show promise as complementary treatments due to their multi-targeted actions and potential to address limitations of current therapies.

Mechanisms of Action

Mechanistically, the plant-derived agents influence key pathways involving cytokines (IL-6, IL-8, TNF-α), transcription factors (NF-κB), and matrix metalloproteinases (MMPs), making them promising candidates for integrative treatment strategies. The pleiotropic effects of plant-derived agents on critical processes in endometriosis pathogenesis are summarized in the following table:

Pathological ProcessImpacted MarkersIntervention
AngiogenesisVEGF, MMP-2, MMP-9Resveratrol, curcumin
InflammationIL-6, IL-8, TNF-αResveratrol, acai extract
Apoptosis ResistanceBcl-2Silymarin, curcumin
Oxidative StressROS, catalase, glutathione peroxidaseResveratrol, silymarin
Proliferation of LesionsERK1/2, cyclin D1Pueraria flower extract, curcumin
Hormonal DysregulationAromatase, COX-2Resveratrol, puerarin

Most Important Findings

The review highlights the multi-targeted action profiles of plant-derived therapies. Resveratrol shows strong evidence in reducing lesion size, VEGF expression, and inflammation across preclinical and limited clinical settings. Curcumin exhibits anti-inflammatory and anti-angiogenic properties by reducing IL-6, IL-8, and VEGF levels, while improving oxidative stress markers. Silymarin is another promising agent with pro-apoptotic and anti-proliferative effects, though its clinical potential is limited by poor bioavailability. Chinese herbal medicine demonstrates efficacy in reducing postoperative recurrence rates and pain, but its variability requires standardized formulations.

Greatest Implications

Plant-derived agents represent a promising addition to multimodal endometriosis treatments, offering pleiotropic benefits and potentially fewer side effects compared to current therapies. They address critical mechanisms such as inflammation, angiogenesis, and oxidative stress, which are central to endometriosis pathology. However, challenges like standardization, bioavailability, and limited clinical evidence remain.

Presence of metalloestrogens in ectopic endometrial tissue

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study quantified metalloestrogens—cadmium, nickel, and lead—in ectopic endometrial tissue, suggesting their role in endometriosis persistence.

What Was Studied?

This study investigated the presence of metalloestrogens—heavy metals with estrogenic effects—in ectopic endometrial tissue. Metalloestrogens, such as cadmium, nickel, and lead, have been implicated in estrogen-dependent diseases like endometriosis. The study aimed to quantify these metals in ectopic endometrial tissues from women diagnosed with endometriosis, using advanced analytical techniques.

Who Was Studied?

The study included 50 women of reproductive age who had endometriosis confirmed through laparotomy or laparoscopy. The participants were patients from a gynecology unit at a tertiary care hospital in Sri Lanka. Samples of ectopic endometrial tissue were collected from these women during surgical procedures, and the disease severity was classified based on the Revised American Society for Reproductive Medicine classification system.

What Were the Most Important Findings?

The study found significant levels of three metalloestrogens—cadmium (2.861 µg/kg), nickel (17.547 µg/kg), and lead (25.785 µg/kg)—in all ectopic endometrial tissue samples analyzed. Among these, lead exhibited the highest concentration. The study is notable for being the first to report the quantitative detection of metalloestrogens in ectopic endometrial tissue. Notably, the presence of these metals varied slightly depending on the tissue site, such as the wall of an endometrioma or nodules in the pelvic region, though these differences were not statistically significant. The findings suggest a potential role for environmental metalloestrogens in the persistence and progression of endometriosis.

What Are the Greatest Implications of This Study?

The detection of metalloestrogens in ectopic endometrial tissue underscores their role in the etiology and maintenance of endometriosis. These metals may act as endocrine disruptors, binding to estrogen receptors in ectopic tissue and mimicking estrogenic effects, thereby contributing to the persistence of the disease. The findings highlight the need for further research to elucidate the mechanistic pathways by which metalloestrogens influence endometriosis. Clinicians should consider environmental exposures and diet as a factor in managing and preventing this condition.

Remission of Endometriosis by Hyperbaric Oxygen Treatment in Rats

May 20, 2025
  • Hyperbaric Oxygen Therapy (HBOT)
    Hyperbaric Oxygen Therapy (HBOT)

    Hyperbaric Oxygen Therapy (HBOT) involves breathing pure oxygen in a pressurized chamber, which increases the amount of oxygen dissolved in the blood and delivered to tissues.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study demonstrates hyperbaric oxygen therapy (HBOT) achieves complete remission of endometriotic lesions in a rat model by reducing hypoxia, inflammation, and TNF-α levels. While HBOT shows potential as a non-invasive therapy for endometriosis, further studies are needed to validate its impact on microbiome modulation.

What Was Studied?

This study, conducted by Aydin et al., investigated the effects of long-term hyperbaric oxygen therapy (HBOT) on experimentally induced endometriosis in a rat model. The primary objective was to assess whether HBOT could lead to remission of endometriotic lesions and alleviate inflammation by modulating peritoneal cytokine levels, particularly tumor necrosis factor-alpha (TNF-α). The study evaluated the volume, histopathological changes, and proliferation markers (Ki-67) of endometriotic implants after six weeks of HBOT.

Who Was Studied?

The study was performed on 40 non-pregnant, female Wistar-Albino rats. After surgical induction of endometriosis using an autotransplantation technique, the rats were divided into two groups: one receiving HBOT (20 rats) and a control group (19 rats) without treatment. HBOT was administered for 2 hours daily at 2.5 atm for six weeks. Both groups underwent multiple laparotomies to evaluate lesion volume, histopathological scores, and cytokine levels before and after treatment.

What Were the Most Important Findings?

The study demonstrated that HBOT resulted in complete remission of endometriotic lesions in a rat model. Significant reductions were observed in lesion volume, histopathological scores, Ki-67 proliferation markers, and TNF-α levels in the peritoneal fluid of the HBOT-treated group compared to controls. Specifically, the mean lesion volume decreased by 29.5% (57.4 ± 12.5 mm³ in the HBOT group vs. 94.6 ± 17.2 mm³ in controls). TNF-α levels were significantly lower in the HBOT group (5.33 ± 1.02 pg/mL vs. 8.16 ± 1.76 pg/mL in controls). Reduced Ki-67 staining indicated diminished cellular proliferation within endometriotic lesions. The findings suggest that HBOT alleviates endometriosis-associated inflammation by suppressing NFκB-mediated pro-inflammatory pathways and reducing TNF-α levels, key drivers of inflammation and angiogenesis in endometriosis.

From a microbiome perspective, while the study did not directly assess microbial changes, the reduction in hypoxia and inflammation could indirectly modulate microbial communities. Hypoxia-driven dysbiosis, favoring facultative anaerobes like E. coli and GBS, is a known contributor to endometriosis pathogenesis. By restoring oxygen levels and dampening inflammation, HBOT may reduce the selective advantage for these pathogens, potentially rebalancing the peritoneal microbiome.

What Are the Greatest Implications of This Study?

The study positions HBOT as a potential non-invasive therapeutic strategy for endometriosis, with demonstrated efficacy in reducing lesion size and inflammation. By targeting hypoxia and pro-inflammatory cytokines, HBOT addresses two critical drivers of endometriosis pathophysiology. This has implications for both clinical management and microbiome research, suggesting that HBOT could indirectly modulate microbial dysbiosis in endometriosis. However, the absence of direct microbial analyses leaves a critical gap in validating HBOT as a microbiome-targeted intervention (MBTI). Further studies incorporating microbiome sequencing and metabolomics are essential to establish a direct link between HBOT and microbiome modulation.

The bidirectional relationship between endometriosis and microbiome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review highlights the bidirectional relationship between endometriosis and the microbiome, showcasing dysbiosis as a key factor in inflammation and estrogen metabolism. Emerging microbiome-targeted therapies hold promise for diagnosis and treatment.

What Was Reviewed?

The review article explored the bidirectional relationship between endometriosis and the microbiome, emphasizing the role of dysbiosis in the pathogenesis and progression of this chronic inflammatory condition. It discussed microbiome alterations across different sites, including the gut, peritoneal fluid, and female reproductive tract, and evaluated how these microbial shifts influence inflammation, immune modulation, and estrogen metabolism. Furthermore, it highlighted experimental and clinical evidence supporting the potential of microbiome-targeted interventions as both diagnostic tools and treatments for endometriosis.

Who Was Reviewed?

The review synthesized findings from human and animal studies investigating microbiota composition in patients with endometriosis compared to healthy controls. It included a comprehensive analysis of bacterial, viral, and fungal associations across diverse microbiome sites, focusing on patterns of dysbiosis, enriched taxa, and diminished microbial diversity. Specific populations reviewed included women diagnosed with various stages of endometriosis and animal models with surgically induced disease.

What Were the Most Important Findings?

The most notable findings included alterations in gut, cervical, and peritoneal fluid microbiota in women with endometriosis. In the gut, elevated levels of Proteobacteria and reduced Lactobacillaceae were observed. The peritoneal fluid showed enrichment of Acinetobacter and Pseudomonas, while the cervical and vaginal microbiomes exhibited decreased diversity and increased abundance of pathogenic species from the Gardnerella and Streptococcus genus. Dysbiosis was associated with heightened inflammatory responses mediated by lipopolysaccharide (LPS) from Escherichia coli, potentially driving lesion formation through the NF-κB pathway. The concept of “estrobolomes,” gut bacteria influencing estrogen reabsorption, was linked to the hyperestrogenic state characteristic of endometriosis. Notably, antibiotic and probiotic treatments in animal models reduced lesion size, supporting the potential therapeutic role of microbiome modulation.

What Are the Greatest Implications?

The implications of this review are twofold: first, the microbiome holds promise as a non-invasive diagnostic tool for endometriosis, potentially reducing diagnostic delays. Second, microbiome-targeted interventions (MBTIs), such as probiotics, prebiotics, and dietary modifications, may offer novel therapeutic avenues. The findings underscore the necessity for further research into microbiome signatures and their clinical applications, particularly in differentiating disease stages and addressing infertility associated with endometriosis.

The Comorbidity of Endometriosis and Systemic Lupus Erythematosus: A Systematic Review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This review explores the significant comorbidity between systemic lupus erythematosus (SLE) and endometriosis, emphasizing shared pathological pathways.

DOI: 10.7759/cureus.42362

What Was Reviewed?

This systematic review examined the comorbidity between endometriosis and systemic lupus erythematosus (SLE), two chronic conditions with significant implications for women's health. The review aimed to elucidate the prevalence, shared pathophysiological mechanisms, and risk factors linking these diseases, emphasizing immune dysregulation, genetic predispositions, and hormonal influences. The review synthesized findings from nine studies conducted between 2011 and 2021, including case-control, cohort, and systematic review methodologies.

Who Was Reviewed?

The review focused on studies of females aged 12-60, representing the pubertal to postmenopausal age range. The population comprised patients with diagnosed endometriosis and SLE. The studies predominantly included participants from diverse ethnicities and geographies, screened based on standardized inclusion criteria to establish the prevalence and interaction of these conditions.

What Were the Most Important Findings?

The review confirmed a statistically significant correlation between endometriosis and SLE, with women diagnosed with either condition at a heightened risk of developing the other. The findings implicated immune dysregulation, characterized by diminished cytotoxic T-cell activity and elevated humoral immune responses, as a central mechanism. Notable microbial associations include increased systemic inflammation mediated by cytokines such as interleukin-1, interleukin-6, and tumor necrosis factor (TNF-α). Genetic factors also played a role, with gene loci such as PTPN22 associated with increased susceptibility to both conditions. Surgical interventions like hysterectomy were linked to increased inflammation and subsequent autoimmune activation, while modified surgical techniques showed promise in mitigating risk.

What Are the Greatest Implications of This Review?

This review highlights the necessity for clinicians to adopt an interdisciplinary approach when managing patients with either endometriosis or SLE, as their comorbidity exacerbates disease burden and complicates treatment. It emphasizes the importance of targeted therapies to modulate immune response alongside careful evaluation of surgical and hormonal treatment strategies to minimize adverse outcomes. The findings suggest a potential for incorporating microbial and genetic markers into diagnostic and therapeutic protocols to improve outcomes.

The effect of hyperbaric oxygen therapy in the inflammatory response in a mouse model of endometriosis: An experimental study

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study highlights hyperbaric oxygen therapy's (HBOT) role in reducing inflammation and NFκB expression in a mouse model of endometriosis. The findings suggest HBOT as a promising non-invasive treatment for managing endometriosis-associated inflammation, targeting hypoxia-induced molecular pathways and offering potential microbiome benefits through reduced inflammatory burden.

What Was Studied?

This study investigated the effect of hyperbaric oxygen therapy (HBOT) on the inflammatory response in a mouse model of endometriosis. The authors focused on the potential therapeutic role of HBOT in reducing inflammation and modulating molecular pathways, particularly the nuclear factor kappa beta (NFκB) pathway, which plays a crucial role in the pathophysiology of endometriosis. The research aimed to address hypoxia-induced inflammation in endometriosis by exploring how HBOT, through its oxygen-rich environment, could attenuate the inflammatory cascade.

Who Was Studied?

The study utilized 24 healthy adult female Swiss albino mice. The animals were randomly divided into three groups: a pre-test group (Group I), a post-test group receiving HBOT (Group II), and a post-test group without HBOT (Group III). Endometriosis was induced via xenotransplantation of human endometrial cells into the mice's peritoneum. Group II received HBOT for 10 days (30 minutes, three times daily), while Group III did not undergo HBOT but was evaluated at the same time points.

What Were the Most Important Findings?

The study found that HBOT significantly reduced the degree of inflammation in endometriosis-induced mice. Group II (HBOT) showed the lowest inflammation scores (1.60 ± 0.53), compared to the pre-test group (9.41 ± 1.99) and the post-test group without HBOT (2.42 ± 0.53). This reduction in inflammation was associated with a significant decrease in NFκB expression, a key pro-inflammatory transcription factor, in the HBOT group. NFκB expression levels correlated strongly with the degree of inflammation (r = 0.670, p ≤ 0.001). These findings suggest that HBOT alleviates the hypoxia-induced inflammatory response by modulating NFκB signaling and reducing peritoneal inflammation.

From a microbiome perspective, hypoxia-induced inflammatory conditions, such as those observed in endometriosis, are often associated with microbial dysbiosis. HBOT's role in reducing inflammation and altering the microenvironment may indirectly influence microbial populations in the peritoneal cavity. This warrants further exploration into whether HBOT could restore microbial balance by reducing the inflammatory burden and hypoxia.

What Are the Greatest Implications of This Study?

The study provides strong evidence for HBOT as a potential therapeutic strategy for reducing inflammation in endometriosis. By mitigating the effects of hypoxia and decreasing NFκB activation, HBOT addresses a key molecular mechanism in the pathogenesis of endometriosis. Clinically, these findings support the use of HBOT as a non-invasive, adjunctive therapy to manage endometriosis-related inflammation. Furthermore, this study underscores the importance of targeting the hypoxia-inflammatory axis to improve outcomes for endometriosis patients. However, the findings also highlight the need for additional research to optimize HBOT protocols, including duration and dose, to achieve maximal therapeutic benefits.

The Endobiota Study: Comparison of Vaginal, Cervical and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies microbiome shifts in vaginal, cervical, and gut sites in stage 3/4 endometriosis. Absence of Atopobium and elevated Gardnerella suggest immune dysregulation, while Escherichia/Shigella dominance in stool correlates with bowel involvement, indicating potential diagnostic biomarkers.

What Was Studied?

This study, titled "The Endobiota Study: Comparison of Vaginal, Cervical, and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls," aimed to evaluate the differences in microbial composition across the vaginal, cervical, and gut microbiomes in women with advanced-stage (3/4) endometriosis compared to healthy controls. Researchers collected and analyzed samples from three anatomical sites—vaginal swabs, cervical swabs, and stool—using 16S rRNA sequencing to determine the diversity and abundance of bacterial genera. The primary objective was to identify specific microbial signatures and dysbiosis patterns associated with advanced endometriosis.

Who Was Studied?

The study included 28 Caucasian women, 14 diagnosed with histologically confirmed stage 3/4 endometriosis and 14 healthy controls. All participants were of reproductive age, with similar age and BMI distributions between groups. Vaginal, cervical, and stool samples were collected from each participant under sterile conditions to prevent contamination. The endometriosis patients were all confirmed to have deep infiltrating endometriosis with extensive lesions, while the control group consisted of asymptomatic women with no clinical or ultrasound evidence of endometriosis.

What Were the Most Important Findings?

The study uncovered notable dysbiosis in the microbiota composition of women with advanced endometriosis compared to healthy controls. In vaginal samples, Gemella and Atopobium were completely absent in the endometriosis group, suggesting a protective role in healthy women. Cervical samples showed a complete loss of Atopobium and Sneathia in endometriosis patients, while Alloprevotella was significantly elevated. This microbial shift in the cervical microbiota is particularly significant given Atopobium's known associations with maintaining vaginal health. In stool samples, Sneathia, Barnesella, and Gardnerella were significantly decreased in endometriosis patients, while Escherichia/Shigella dominance was observed in two women who subsequently required segmental colon resection for severe bowel involvement. Sensitivity analyses excluding Lactobacillus revealed that Gardnerella represented a significantly higher proportion of the remaining microbiota in the vaginal and cervical niches of the endometriosis group compared to controls (72.9% vs. 36.8% in the vagina and 67.7% vs. 36.8% in the cervix, respectively). Furthermore, Escherichia/Shigella, Streptococcus, and Ureaplasma were markedly elevated, while Prevotella, Dialister, and Megasphaera were significantly reduced. These microbial changes suggest an altered immune response and heightened inflammatory state in women with advanced endometriosis, highlighting potential microbial markers of disease progression.

Anatomical SiteMicrobiota Findings in Advanced Endometriosis Patients
Vaginal SamplesGemella and Atopobium completely absent. Gardnerella significantly elevated (72.9% of microbiota, excluding Lactobacillus).
Cervical SamplesComplete loss of Atopobium and Sneathia. Marked increase in Alloprevotella. Gardnerella elevated (67.7% of microbiota, excluding Lactobacillus).
Stool SamplesSignificant decreases in Sneathia, Barnesella, and Gardnerella. Dominance of Escherichia/Shigella observed in two patients requiring bowel resection.
Additional Microbial ShiftsMarked elevation of Escherichia/Shigella, Streptococcus, and Ureaplasma. Reductions in Prevotella, Dialister, and Megasphaera.
Inflammatory AssociationsDysbiosis patterns suggest an altered immune response and heightened inflammatory state in advanced endometriosis.

The impact of endometriosis on dietary choices and activities of everyday life: a cross-sectional study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review highlights how gluten-free, Mediterranean, and anti-inflammatory diets improve pain perception in endometriosis by reducing inflammation and modulating the gut microbiome. Probiotics and bioactive nutrients such as curcumin enhance therapeutic outcomes.

What was reviewed?

This systematic review evaluated the impact of dietary interventions on pain perception in women diagnosed with endometriosis. It explored the connections between dietary changes and the alleviation of symptoms, particularly chronic pain, and assessed the potential of specific dietary patterns and nutrients to influence disease progression and symptom severity. The review included evidence from various studies highlighting the role of diets such as gluten-free, Mediterranean, and anti-inflammatory diets, along with the incorporation of specific nutrients and probiotics.

Who was reviewed?

The review included studies examining women diagnosed with endometriosis, focusing on their dietary habits, pain management strategies, and overall quality of life. The population spanned diverse stages of endometriosis and varying symptom severities, with dietary interventions as a common self-management approach.

What were the most important findings?

The review identified several key dietary patterns and nutrients that positively influenced pain perception and symptom management in women with endometriosis. A gluten-free diet was associated with symptom relief in patients experiencing gastrointestinal-related pain, while the Mediterranean diet showed benefits in reducing inflammation and pain severity due to its high content of antioxidants, omega-3 fatty acids, and polyphenols. Anti-inflammatory diets also gained traction, particularly in severe cases of endometriosis, where eliminating saturated fats and processed meats improved symptom management.

Major microbial associations (MMAs) of endometriosis were also highlighted, particularly the role of probiotics like Lactobacillus in alleviating pain and potentially modulating the gut microbiome to reduce systemic inflammation. The findings emphasize the therapeutic potential of dietary supplements such as curcumin, resveratrol, and quercetin, which possess anti-inflammatory and antioxidant properties.

What are the greatest implications of this review?

The findings suggest that dietary interventions and supplements can serve as non-invasive and complementary strategies for managing endometriosis-related symptoms, particularly chronic pain. By modulating systemic inflammation and influencing the gut microbiome, specific dietary patterns and nutrients may provide a tailored approach to alleviating symptoms. The review underscores the importance of integrating nutritional guidance into endometriosis management protocols.

The Influence of Lactoferrin in Plasma and Peritoneal Fluid on Iron Metabolism in Women with Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study demonstrates that peritoneal fluid-to-plasma ferritin and lactoferrin ratios distinguish endometriosis stage and severity. Iron overload and shifting iron-binding protein profiles reveal a localized dysregulation that may influence disease progression and potentially pathogenic microbiome selection.

What was studied?

This study investigated the role of lactoferrin (LF) in relation to iron metabolism in women with and without endometriosis by measuring levels of LF, ferritin (FT), transferrin (TF), and iron (Fe) simultaneously in plasma and peritoneal fluid. The authors specifically explored whether the concentrations and ratios of these iron-related proteins in the two biological compartments could distinguish the presence and progression of endometriosis. The goal was to identify noninvasive or minimally invasive biomarkers that may aid in diagnosing or staging the disease based on iron metabolism, especially given endometriosis’ pro-inflammatory, iron-rich microenvironment.

Who was studied?

The study cohort included 90 women of reproductive age undergoing diagnostic laparoscopy, of whom 57 had histologically confirmed endometriosis (stages I–IV) and 33 did not. Plasma and peritoneal fluid samples were collected pre- and intra-operatively. Subjects were classified based on endometriosis diagnosis and stage, and specimens were evaluated for levels of LF, FT, TF, and Fe using ELISA, immunoturbidimetric assay, and colorimetric methods.

What were the most important findings?

Key findings highlight that ferritin and iron concentrations were significantly elevated in peritoneal fluid compared to plasma, especially in patients with advanced-stage endometriosis. In contrast, transferrin was consistently lower in peritoneal fluid. Notably, lactoferrin levels did not significantly differ between women with and without endometriosis when evaluated independently in plasma or peritoneal fluid, but the peritoneal fluid/plasma lactoferrin ratio decreased progressively with increasing disease severity, significantly distinguishing stage I from stage IV. The ferritin ratio was markedly higher in the endometriosis group, underscoring its potential as a disease marker. Correlation analyses revealed that in severe endometriosis, lactoferrin was significantly associated with ferritin and iron in the peritoneal fluid, suggesting a disrupted iron regulation mechanism localized to the disease microenvironment. Importantly, the elevated ferritin concentrations in peritoneal fluid may serve a compensatory, protective role to sequester iron and mitigate oxidative stress, while lactoferrin may lose this protective function as disease progresses.

From a microbiome perspective, this study underscores the iron-dependent ecological shifts that may select for siderophilic pathobionts. The iron overload and pro-oxidative milieu likely fosters the expansion of iron-requiring microbial taxa, potentially including Escherichia, Enterobacter, and Fusobacterium, known to be enriched in some endometriosis microbiome signatures. While microbial profiling was not performed, the metallomic dysregulation described supports the hypothesis that iron availability is a crucial factor in shaping pathogenic microbial communities in endometriosis.

What are the greatest implications of this study?

This study provides compelling evidence that iron-binding proteins—particularly ferritin and lactoferrin—play a localized and differential role in the progression of endometriosis. The findings suggest that peritoneal fluid iron metabolism, and especially the ferritin-to-lactoferrin balance, may be a critical axis of disease progression and potentially a therapeutic target. The study introduces the peritoneal fluid/plasma concentration ratio as a novel diagnostic parameter, offering a more granular assessment than conventional plasma markers. The declining lactoferrin ratio and increasing ferritin ratio with disease severity may signal a transition from iron sequestration and immune modulation toward iron-driven oxidative stress and tissue damage. This may serve as a foundation for the development of metallome-targeted diagnostics and therapies, including exogenous lactoferrin supplementation, which the authors suggest could restore iron balance in advanced disease stages. These findings also have implications for understanding how iron dysregulation may foster microbial dysbiosis, providing a mechanistic link between host iron metabolism and the pathophysiological selection of microbial communities in endometriosis.

The Main Theories on the Pathogenesis of Endometriosis

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review synthesizes key theories on endometriosis pathogenesis, emphasizing immune, hormonal, and epigenetic interactions in its progression.

What Was Reviewed?

This review explored the primary theories underlying the pathogenesis of endometriosis, a chronic gynecological disorder characterized by the growth of endometrial-like tissue outside the uterus. The review evaluated various mechanisms, including retrograde menstruation, immune dysregulation, coelomic metaplasia, hormonal imbalance, and epigenetic regulation. Additionally, it considered the role of stem cells and environmental factors in disease onset and progression. The synthesis of these theories aimed to illuminate the multifactorial origins of the disease and its systemic implications.

Who Was Reviewed?

The review included a broad analysis of scientific literature, focusing on research involving women with endometriosis, animal models (notably baboons and mice), and cell-based experiments. Integrating data from diverse biological contexts provided a comprehensive overview of the disease's potential mechanisms.

What Were the Most Important Findings?

The review highlighted that endometriosis likely results from an interplay of multiple pathogenic pathways. Immune dysregulation was a central focus, with macrophages, T cells, and dendritic cells contributing to chronic inflammation, suppressed apoptosis, and lesion growth. Hormonal imbalances, particularly estrogen dominance and progesterone resistance, were identified as pivotal in driving lesion persistence and infertility. Epigenetic changes, such as DNA methylation and micro-RNA dysregulation, emerged as significant contributors to disease progression, influencing inflammation, angiogenesis, and cellular proliferation.

This review did not address key microbial associations in endometriosis, but immune-modulated inflammation and altered hormonal environments suggest indirect links to microbiome perturbations. For example, increased inflammatory cytokines (e.g., TNF-α and IL-6) and VEGF in endometriotic lesions may influence local microbial dynamics.

What Are the Greatest Implications of This Review?

Understanding the multifactorial nature of endometriosis provides a foundation for developing targeted therapeutic strategies. The review underscores the need for integrative approaches combining hormonal regulation, immune modulation, and potential epigenetic therapies. Additionally, exploring microbiome interactions in this context may reveal novel interventions, particularly in immune-regulated inflammation and hormonal imbalances.

The role of gut and genital microbiota and the estrobolome in endometriosis, infertility and chronic pelvic pain

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Chronic Pelvic Pain (CPP)
    Chronic Pelvic Pain (CPP)

    Chronic Pelvic Pain (CPP) is persistent pain in the pelvic region lasting six months or longer, often multifactorial, impacting physical and emotional well-being, and associated with various medical conditions.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

This review highlights the gut and genital microbiome's roles in estrogen-driven conditions like endometriosis, infertility, and CPP, emphasizing dysbiosis' impact on inflammation and estrogen metabolism.

What was reviewed?

The reviewed manuscript explored the intricate relationship between the gut and genital microbiomes, the estrobolome, and their roles in the pathophysiology of endometriosis, infertility, and chronic pelvic pain (CPP). The authors critically examined 28 clinical and six preclinical studies to understand microbial dysbiosis's contributions to estrogen metabolism, inflammation, and symptomatology in these conditions. This review also identified methodological gaps in microbiome studies and proposed strategies to improve future research.

Who was reviewed?

The review included human and animal studies, examining women diagnosed with endometriosis, infertility, and CPP, alongside healthy controls. Specific focus was placed on microbial associations in the gut, cervicovaginal, and endometrial microbiomes, with emphasis on bacterial vaginosis-associated bacteria, Lactobacillus depletion, and microbial influences on estrogen-driven mechanisms.

What were the most important findings?

Key findings highlighted that dysbiosis in the gut microbiome disrupts the estrobolome, an essential modulator of estrogen metabolism. This disruption contributes to heightened systemic and local inflammation, potentially exacerbating endometriosis symptoms and infertility. Many studies noted an association between bacterial vaginosis-related bacteria and a reduction in Lactobacillus dominance in the cervicovaginal microbiome with the prevalence of endometriosis and infertility. Additionally, the review underscored a bidirectional relationship between gut microbiota and endometriosis progression in animal models, emphasizing the role of gut dysbiosis in increasing b-glucuronidase activity, leading to elevated circulating estrogen levels.

What are the greatest implications of this review?

This review underscores the need for rigorous, standardized methodologies to better delineate causal relationships between microbiota and gynecological conditions like endometriosis and CPP. The findings of this review suggest that targeting the microbiome could lead to novel diagnostics and therapeutics for estrogen-driven diseases. The review also highlights the potential of leveraging microbiome-based biomarkers for non-invasive diagnostics and monitoring of endometriosis progression, bridging a critical translational gap in gynecological health.

The role of the vaginal microbiome in distinguishing female chronic pelvic pain caused by endometriosis/adenomyosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Chronic Pelvic Pain (CPP)
    Chronic Pelvic Pain (CPP)

    Chronic Pelvic Pain (CPP) is persistent pain in the pelvic region lasting six months or longer, often multifactorial, impacting physical and emotional well-being, and associated with various medical conditions.

This study examines the role of the vaginal microbiome in distinguishing chronic pelvic pain caused by endometriosis and adenomyosis. Findings highlight specific microbial signatures associated with pain severity, offering potential non-invasive biomarkers for differential diagnosis and targeted therapeutic strategies.

What was studied?

This study investigated whether the composition of the vaginal microbiome could serve as a diagnostic biomarker to differentiate chronic pelvic pain (CPP) caused by endometriosis or adenomyosis (EM/AM) from other causes of chronic pelvic pain syndrome (CPPS) in women. Using 16S rRNA sequencing (V4 region), the researchers profiled the vaginal microbiota of 37 women with EM/AM-associated CPP, 25 with CPPS from other causes, and 66 healthy controls without CPPS. Additionally, the study explored whether combining vaginal microbial markers with serum CA125 could improve differential diagnostic accuracy.

Who was studied?

The study included 128 premenopausal women attending the gynecology department of Peking Union Medical College Hospital. These were stratified into three groups: 37 women with surgically confirmed EM/AM-associated CPP, 25 women with non-EM/AM CPPS (adhesions, hydrosalpinx, infertility), and 66 women without any chronic pelvic pain. All participants were HPV-negative, had not recently used antibiotics or vaginal products, and were matched for age, gravidity, parity, and contraceptive method to control for confounding variables.

What were the most important findings?

The vaginal microbiome of women with EM/AM-associated CPP exhibited significantly higher alpha diversity than those in the CPPS and healthy control groups. Taxonomic analyses revealed distinct microbial signatures: increased abundance of Clostridium butyricum, Clostridium disporicum, Alloscardovia omnicolens, and Veillonella montpellierensis, alongside a marked depletion of Lactobacillus jensenii, Lactobacillus reuteri, and Lactobacillus iners. These differentially abundant taxa serve as potential microbiome biomarkers.

Diagnostic performance analysis demonstrated that a combination of microbial biomarkers (specifically, a relative abundance of Clostridium disporicum >0.001105% and Lactobacillus reuteri <0.1911349%) yielded 81.08% sensitivity and 52% specificity for identifying EM/AM-associated CPP. When combined with serum CA125 levels, sensitivity increased to 89.19%, although specificity remained unchanged. Functional predictions via PICRUSt revealed enrichment of metabolic pathways such as amino acid metabolism, energy metabolism, and metabolism of cofactors and vitamins in EM/AM patients, along with downregulation of membrane transport and nucleotide metabolism compared to controls. These shifts may reflect microbial contributions to inflammation and pain signaling pathways implicated in EM/AM-associated CPP.

From a microbiome signature standpoint, the enriched taxa—particularly Clostridium disporicum and Alloscardovia omnicolens—emerge as Major Microbial Associations (MMAs) due to their consistent elevation in EM/AM patients. Conversely, Lactobacillus jensenii and L. reuteri, known for their protective, anti-inflammatory properties, are depleted, suggesting their role in maintaining vaginal eubiosis and preventing EM/AM-associated pathogenesis.

What are the greatest implications of this study?

This research provides compelling evidence that the vaginal microbiome harbors discriminative microbial signatures capable of differentiating EM/AM-associated CPP from other forms of chronic pelvic pain. The incorporation of specific microbial biomarkers, particularly when paired with serum CA125, may improve non-invasive diagnostic accuracy, enabling earlier and more targeted therapeutic intervention. Clinically, these findings underscore the potential of microbiome-informed diagnostics for gynecological conditions where conventional markers fall short. More broadly, this study suggests that vaginal dysbiosis, characterized by Lactobacillus depletion and enrichment of saccharolytic and anaerobic species, could be causally linked to EM/AM pathogenesis, possibly via inflammatory or metabolic pathways. Future studies incorporating metagenomic or metabolomic analyses are warranted to functionally validate these microbial associations and to explore the feasibility of microbial modulation as a therapeutic strategy.

The Vaginal Microbiome as a Tool to Predict rASRM Stage of Disease in Endometriosis: a Pilot Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies the vaginal microbiome as a predictor of endometriosis severity, highlighting microbial shifts that correlate with rASRM staging. Anaerococcus emerged as a key biomarker for advanced disease stages, while CST IV dominance during menstruation suggests inflammatory shifts. Findings support the potential of non-invasive microbiome-based diagnosis for endometriosis.

What Was Studied?

This study investigated the potential use of the vaginal microbiome as a diagnostic tool to predict the stage of disease severity in endometriosis, based on the revised American Society for Reproductive Medicine (rASRM) staging system. Conducted as an observational cross-sectional pilot study, researchers characterized the gut and vaginal microbiome profiles of women with and without endometriosis to explore non-invasive biomarkers for disease staging. A total of 59 women participated, 35 with endometriosis and 24 controls. Rectal and vaginal samples were collected at two different points in the menstrual cycle—the menstrual and follicular phases—to assess the microbial composition's correlation with rASRM stages. Illumina sequencing was utilized to analyze 16S rRNA gene amplicons, with community state types (CSTs) assigned to classify the vaginal microbiota. Random forest-based machine-learning models were constructed to evaluate the predictive power of vaginal microbiota profiles during different menstrual phases.

Who Was Studied?

The study included 35 women with a confirmed diagnosis of endometriosis and 24 control subjects without the disease. Participants were recruited from the University of Sao Paulo and the Massachusetts Institute of Technology, with all subjects providing written informed consent. Inclusion criteria required histological confirmation of endometriosis, while controls were women undergoing laparoscopic surgery for other benign gynecological conditions. Key exclusion criteria included recent antibiotic or hormone use, active infections, autoimmune diseases, and any history of sexually transmitted infections. The vaginal and rectal samples were collected during both the follicular and menstrual phases, known to influence microbial community dynamics.

What Were the Most Important Findings?

The study revealed that the vaginal microbiome's composition significantly differed between endometriosis patients and controls, particularly during the menstrual phase. Classification models built from vaginal microbial profiles during menstruation accurately predicted rASRM stage 1–2 versus stage 3–4 endometriosis. The genus Anaerococcus emerged as the top predictive operational taxonomic unit (OTU) for distinguishing between early and advanced stages of the disease. Notably, the transition of community state types (CSTs) also reflected disease severity. During the menstrual phase, there was a marked increase in CST IV (characterized by anaerobic bacteria dominance) among both endometriosis patients (30%) and controls (25%), with a simultaneous loss of CST II and CST V. CST I, typically dominated by Lactobacillus crispatus, appeared more prevalent in endometriosis patients during menstruation, suggesting an immunomodulatory role that may contribute to the disease's local inflammatory environment. These microbial changes were consistent with differences in local immune response and hormonal fluctuations. This pilot study is the first to demonstrate that vaginal microbiome profiles, particularly the presence of Anaerococcus, may serve as a non-invasive biomarker for endometriosis staging, potentially offering a diagnostic tool that bypasses the need for invasive surgical confirmation.

Microbial GroupEndometriosis FindingsClinical Implications
AnaerococcusIncreased in rASRM stages III–IVMarker for advanced disease severity
CST IV (Community State Type)Dominant during menstruationAssociated with low Lactobacillus and high microbial diversity
Lactobacillus spp.Decreased during menstrual phaseReduced protective barrier; potential inflammation driver
Machine Learning PredictionHigh accuracy (AUC = 0.89)Potential for non-invasive staging of endometriosis
Vaginal Microbiome ShiftsCorrelated with menstrual cycle phaseIndicates dynamic microbial changes tied to inflammation

What Are the Greatest Implications of This Study?

The findings from this study suggest that the vaginal microbiome, specifically the composition of community state types and the presence of Anaerococcus, may be harnessed as a non-invasive biomarker to predict the stage of endometriosis severity. This has profound implications for clinical practice, as it could reduce the dependency on invasive laparoscopy for disease staging, which is currently the gold standard. If validated in larger cohorts, this approach could facilitate early detection and better stratification of endometriosis patients, enabling more targeted and personalized therapeutic interventions. Furthermore, the study underscores the significance of microbiome-driven inflammation in the pathophysiology of endometriosis, opening avenues for microbiome-targeted therapies as a novel strategy to mitigate disease progression and symptom severity. The integration of vaginal microbiome profiling into clinical diagnostics could revolutionize the early detection and management of endometriosis, addressing a critical unmet need in gynecological health.

Trace Elements and Endometriosis: Insights into Oxidative Stress and Novel Therapies

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review explores the role of trace elements and oxidative stress in endometriosis, highlighting their potential as therapeutic targets. It underscores the need for further research into the trace elements’ roles in endometriotic lesions.

What was reviewed?

The article reviews the role of trace elements in the pathogenesis and management of endometriosis, a chronic, estrogen-dependent inflammatory disease. It synthesizes existing research on the impact of oxidative stress and environmental exposure to trace elements like zinc, nickel, cadmium, and copper, linking these factors to the formation and proliferation of endometrial-like lesions outside the uterus.

Who was reviewed?

The review focuses on studies involving women with confirmed endometriosis, highlighting environmental and biological factors such as trace element concentrations in blood, urine, and peritoneal fluid. Additionally, it incorporates experimental findings, including animal models, to explore the mechanistic roles of trace elements.

What were the most important findings?

The review emphasizes the link between oxidative stress and endometriosis, with trace elements acting as potential modulators of this process. Zinc, for instance, is identified for its antioxidant and anti-inflammatory roles, with lower levels in endometriosis patients potentially contributing to lesion formation. Nickel, on the other hand, has been implicated in the condition as a metalloestrogen, as further evidenced by improved symptoms following a low-nickel diet. Cadmium and lead, known for inducing oxidative stress, show conflicting associations with endometriosis, though some evidence suggests their presence synergistically exacerbates disease severity. Copper's involvement in angiogenesis and its elevated levels in endometriosis patients suggest a role in lesion proliferation. The review also highlights discrepancies in study findings, emphasizing the need for further research on trace elements within endometriotic implants rather than just systemic fluids.

What are the greatest implications of this review?

The review underscores the potential of targeting trace elements and oxidative stress as therapeutic strategies for endometriosis. It calls for more comprehensive research into the specific roles of trace elements within endometriotic tissue, as these could pave the way for novel diagnostic markers and treatments. Additionally, the environmental and dietary implications of trace element exposure warrant further exploration, particularly in the context of prevention and symptom management.

Metabolomic Signature

Go to Category Page

1H NMR- based metabolomics approaches as non-invasive tools for diagnosis of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metabolomic Signature
    Metabolomic Signature

    Metabolomic signatures are unique metabolite patterns linked to specific biological conditions, identified through metabolomics. They reveal underlying biochemical activities, aiding in disease diagnosis, biomarker development, and personalized medicine. The microbiome significantly affects these signatures, influencing health and disease outcomes through metabolic interactions.

This study demonstrates the potential of ¹H-NMR metabolomics to diagnose endometriosis non-invasively by identifying metabolic biomarkers and disrupted pathways. Quadratic Discriminant Analysis outperformed Artificial Neural Networks in diagnostic accuracy.

What was studied?

This study investigated the application of metabolomics, specifically through proton nuclear magnetic resonance (¹H-NMR) spectroscopy, to identify non-invasive biomarkers for diagnosing endometriosis. The researchers developed computational models using Quadratic Discriminant Analysis (QDA) and Artificial Neural Networks (ANNs) to analyze metabolic changes in serum samples and assess their utility in early diagnosis of the disease.

Who was studied?

The study analyzed serum samples from 31 infertile women diagnosed with stage II or III endometriosis confirmed via laparoscopy and 15 healthy women without any signs of endometriosis. The participants were aged 22–44 years and were recruited from an infertility center in Iran. Exclusion criteria included recent medical or hormonal treatments, prior gynecological surgeries, or other pelvic inflammatory conditions.

What were the most important findings?

The study revealed significant metabolic differences between women with endometriosis and healthy controls. Key findings included elevated levels of 2-methoxyestrone, 2-methoxyestradiol, androstenedione, aldosterone, dehydroepiandrosterone, and deoxycorticosterone in the endometriosis group, alongside decreased cholesterol and primary bile acids. These metabolic changes are linked to disruptions in steroid hormone biosynthesis and bile acid metabolism, indicating underlying hyperestrogenism and impaired hepatic estrogen clearance. The QDA model achieved a correct classification rate of 76%, with 71% positive predictive value and 78% negative predictive value, outperforming the ANN model, which had lower sensitivity and specificity. Metabolic pathway analyses highlighted altered steroid hormone and bile acid biosynthesis, which are critical in the pathophysiology of endometriosis.

What are the greatest implications of this study?

This study underscores the potential of ¹H-NMR-based metabolomics as a minimally invasive diagnostic tool for endometriosis, reducing reliance on invasive laparoscopy. The identification of specific biomarkers and disrupted pathways could facilitate earlier diagnosis, improved patient stratification, and targeted therapeutic interventions. The findings also demonstrate the utility of computational modeling, particularly QDA, in translating complex metabolomics data into clinically actionable insights. This approach represents a significant advancement in bridging diagnostic gaps for endometriosis.

A metabonomics approach as a means for identification of potentialbiomarkers for early diagnosis of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metabolomic Signature
    Metabolomic Signature

    Metabolomic signatures are unique metabolite patterns linked to specific biological conditions, identified through metabolomics. They reveal underlying biochemical activities, aiding in disease diagnosis, biomarker development, and personalized medicine. The microbiome significantly affects these signatures, influencing health and disease outcomes through metabolic interactions.

This study used 1H-NMR metabonomics to identify serum biomarkers for early endometriosis diagnosis. Elevated lactate, alanine, and reduced glucose levels highlight metabolic disruptions, offering a minimally invasive diagnostic tool.

What was studied?

This study investigated the identification of predictive biomarkers for early diagnosis of endometriosis using a minimally invasive, serum-based approach. The researchers utilized proton nuclear magnetic resonance (1H-NMR) metabonomics to analyze serum samples, aiming to distinguish endometriosis patients from healthy controls. The study particularly focused on differences in metabolite profiles to identify markers indicative of the condition.

Who was studied?

The study included 45 women aged under 40, divided into two groups. The first group comprised 22 women diagnosed with early-stage endometriosis (Stages I–II) via laparoscopy, while the control group consisted of 23 age- and BMI-matched healthy women with normal menstrual cycles and proven fertility. Participants with recent hormone therapy or irregular menstrual cycles were excluded. Serum samples were collected during the secretory phase of the menstrual cycle.

What were the most important findings?

The study identified several metabolites with significantly altered levels in women with endometriosis compared to controls. Increased levels of lactate, 3-hydroxybutyrate, alanine, leucine, valine, threonine, lysine, glycerophosphatidylcholine, succinic acid, and 2-hydroxybutyrate were observed in the serum of endometriosis patients, while glucose, isoleucine, arginine, and lipid levels were decreased. Multivariate analysis using Partial Least Squares-Discriminant Analysis (PLS-DA) demonstrated strong sensitivity (81.8%) and specificity (91.3%) in distinguishing endometriosis from controls, with an area under the ROC curve of 0.96. Pathway analysis highlighted arginine and proline metabolism disruptions, glycine, serine, and threonine metabolism, pyruvate metabolism, and lysine biosynthesis and degradation. These findings provide a potential non-invasive diagnostic framework and insights into the metabolic disturbances in endometriosis.

What are the greatest implications of this study?

This study offers a promising step toward non-invasive diagnostic methods for endometriosis, reducing reliance on invasive laparoscopy. The identification of metabolite alterations linked to the disease enhances the understanding of its pathophysiology, emphasizing oxidative stress, anaerobic glycolysis, and metabolic reprogramming similar to malignancies. These findings could lead to better clinical tools for early diagnosis and a deeper understanding of the metabolic underpinnings of endometriosis.

Metabolomics reveals perturbations in endometrium and serum of minimal and mild endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metabolomic Signature
    Metabolomic Signature

    Metabolomic signatures are unique metabolite patterns linked to specific biological conditions, identified through metabolomics. They reveal underlying biochemical activities, aiding in disease diagnosis, biomarker development, and personalized medicine. The microbiome significantly affects these signatures, influencing health and disease outcomes through metabolic interactions.

This study highlights metabolic alterations in endometrial tissue and serum of minimal and mild endometriosis patients, identifying amino acid biomarkers with diagnostic potential. A combined serum panel demonstrated 100% sensitivity and 83% specificity for Stage II diagnosis, offering a promising step toward non-invasive early detection of endometriosis.

What Was Studied?

This study investigated the metabolic perturbations in eutopic endometrial tissue and serum of women with minimal and mild endometriosis (Stages I and II) using ^1H Nuclear Magnetic Resonance (NMR)-based metabolomics. The researchers aimed to identify specific metabolites that could be potential biomarkers for the early, non-invasive diagnosis of endometriosis. The study included multivariate and univariate analyses to identify metabolite changes and their diagnostic potential.

Who Was Studied?

The study included 95 women diagnosed with endometriosis (staged using the revised American Society for Reproductive Medicine criteria) and 24 healthy fertile controls. The participants were recruited from Eastern India and Bangladesh, with exclusion criteria ensuring no confounding conditions such as ovarian tumors or pelvic inflammatory disease. Blood and eutopic endometrial tissue samples were collected during the mid-secretory phase of the menstrual cycle.

What Were the Most Important Findings?

Women with minimal and mild endometriosis exhibited significant metabolic alterations, particularly in amino acids. Alanine, lysine, leucine, proline, and phenylalanine levels were notably dysregulated in serum, with tissue samples showing lower levels of these metabolites, except for proline, which positively correlated with serum levels. Alanine alone demonstrated diagnostic potential for Stage I endometriosis, with 90% sensitivity and 58% specificity.

For Stage II, phenylalanine achieved 100% sensitivity but had lower specificity, while a combined panel of metabolites improved diagnostic accuracy, reaching 100% sensitivity and 83% specificity.

In advanced stages, elevated taurine and myo-inositol levels were linked to increased cell proliferation and angiogenesis, highlighting similarities with tumorigenic processes. These findings underscore the critical role of metabolic shifts in endometriosis progression, particularly involving amino acids and nucleotide synthesis, and suggest their utility in early detection and non-invasive diagnostics.

What Are the Greatest Implications of This Study?

This study underscores the potential of metabolomic signatures in elucidating the pathophysiology of endometriosis and developing non-invasive diagnostic tools, especially for early stages where traditional biomarkers like CA-125 are less effective. By identifying a panel of serum metabolites, the research provides a foundation for improving diagnostic accuracy and reducing the need for invasive laparoscopy. Additionally, the observed metabolic similarities between endometriosis and malignancies could inspire further exploration of shared mechanisms, potentially broadening therapeutic targets.

Breast Cancer

Go to Category Page

A comprehensive analysis of breast cancer microbiota and host gene expression

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

The study analyzed breast tumor and adjacent tissues, linking microbiota composition to cancer pathways. Key findings implicate specific microbes in breast cancer progression.

What Was Studied?

This study investigated the microbial composition of breast tumor tissues compared to non-cancerous adjacent (NCA) tissues, focusing on identifying specific microbiota associated with different breast cancer subtypes. The research utilized RNA sequencing data from The Cancer Genome Atlas (TCGA), analyzing microbial reads and their association with host gene expression profiles to explore the role of the tumor microbiota in breast cancer pathogenesis.

Who Was Studied?

The study involved 668 breast tumor tissue samples and 72 NCA samples. The samples were filtered to exclude male patients, metastatic cases, and individuals with a history of breast cancer or neoadjuvant therapy, ensuring a robust cohort for microbial and host gene analysis.

What Were the Most Important Findings?

The study identified distinct microbial signatures between tumor and NCA tissues. Proteobacteria were significantly enriched in tumor samples, while Actinobacteria were more prevalent in NCA tissues. Specific microbial taxa, such as Haemophilus influenzae, were associated with genes involved in tumor-promoting pathways, including the G2M checkpoint, E2F transcription factors, and mitotic spindle assembly. Similarly, Listeria fleischmannii correlated with epithelial-to-mesenchymal transition pathways, a hallmark of cancer metastasis.

Twelve of the most abundant species, including Escherichia coli, Mycobacterium fortuitum, and Salmonella enterica, showed significant differential abundance between tumor and NCA tissues. These species are notable for their potential roles in DNA damage and estrogen metabolism, contributing to genomic instability and hormonal dysregulation in breast cancer. The findings also revealed that less prevalent taxa often showed the most significant differential abundance, highlighting the challenges of detecting meaningful microbial shifts in underpowered studies.

What Are the Greatest Implications of This Study?

This research underscores the complex interplay between the tumor microbiota and host gene expression in breast cancer. The enrichment of specific microbial taxa in tumor tissues and their associations with oncogenic pathways suggest that the microbiota may play an active role in breast cancer progression. These findings open avenues for microbiota-targeted interventions and diagnostic tools based on microbial markers. Furthermore, the study highlights the need for large-scale, well-controlled cohorts to accurately characterize the tumor microbiome and its clinical relevance.

Association between Gut Microbiota and Breast Cancer: Diet as a Potential Modulating Factor

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study links reduced gut microbial diversity and specific taxa (e.g., Acidaminococcus, Hungatella) to breast cancer, influenced by diet. Findings suggest microbiome-targeted interventions and dietary strategies could mitigate breast cancer risk.

What Was Studied?

This study examined the association between gut microbiota composition and breast cancer, focusing on the role of diet as a potential modulating factor. Researchers conducted a case-control study involving 42 newly diagnosed, treatment-naïve BCa patients and 44 age-matched cancer-free controls. The gut microbiome was analyzed through 16S rRNA sequencing, and dietary patterns were assessed using the National Cancer Institute Diet History Questionnaire.

Who Was Studied?

Participants included females aged 20–89 years from the Oregon Health & Science University. breast cancer patients were diagnosed through biopsy and had not yet undergone any treatment. Cancer-free controls were matched by age and underwent recent mammograms with non-suspicious results. The study collected fecal samples, dietary data, and comprehensive lifestyle information to ensure robust comparisons.

Most Important Findings

The study identified significant differences in the gut microbiome composition between breast cancer cases and controls, including reduced microbial diversity among breast cancer patients, indicative of dysbiosis. Specifically, the genera Acidaminococcus, Hungatella, and Tyzzerella were enriched, while controls exhibited enrichment of genera such as Christensenellaceae and Dialister. These findings were linked to dietary patterns: Acidaminococcus correlated with lower fruit intake, Hungatella with reduced dairy intake but increased vegetable consumption, and Tyzzerella was not significantly associated with dietary variables. Importantly, the reduced diversity and altered microbial profiles in breast cancer patients align with previous evidence suggesting a role for gut dysbiosis in cancer progression via immune modulation and microbial metabolite production.

Greatest Implications

This study highlights the gut microbiome's potential as a biomarker for breast cancer risk and emphasizes the role of diet in modulating microbial composition. Dysbiosis, characterized by an imbalance in gut microbiota, is linked to breast cancer, suggesting that microbiome-targeted dietary interventions could aid in prevention and management. For example, increased consumption of whole fruits may help reduce levels of Acidaminococcus, a genus enriched in breast cancer patients, while higher dairy intake could lower the abundance of Hungatella, a genus associated with TMAO production and cancer-promoting pathways. Interestingly, the study also found that greater vegetable consumption was linked to higher levels of Hungatella, which has been associated with increased risks of both breast and colorectal cancer. These findings underscore the complexity of dietary influences on the gut microbiome and their potential role in cancer prevention.

Breast cancer but not the menopausal status is associated with small changes of the gut microbiota

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study shows that breast cancer, rather than menopausal status, drives subtle gut microbiota changes. Dysbiosis in BC patients included reduced Blautia obeum and Bifidobacterium. Functional impacts, such as downregulated NAD pathways, suggest gut microbiota's potential role in cancer progression.

What Was Studied?

This study investigated the relationship between gut microbiota composition and breast cancer (BC), focusing on the potential impact of menopausal status on microbiota variations. The researchers used shotgun metagenomics to compare the gut microbiota of 88 newly diagnosed BC patients (47 premenopausal and 41 postmenopausal) with 86 cancer-free controls, stratified by menopausal status.

Who Was Studied?

The participants included Polish women divided into two groups: BC patients and controls. The BC group was further divided into premenopausal and postmenopausal subgroups. Fecal samples were collected before systemic cancer treatment, and patients with prior antibiotic use, inflammatory bowel disease, or a history of cancer (for controls) were excluded.

Most Important Findings

The study showed that menopausal status had no significant impact on the overall gut microbiota composition or diversity. However, breast cancer (BC) patients exhibited gut dysbiosis compared to controls. Premenopausal BC patients demonstrated lower abundances of taxa such as Bifidobacterium and Collinsella massiliensis but higher abundances of the genus Gemmiger. In postmenopausal BC patients, taxa such as Blautia obeum, Dorea formicigenerans, and Bacteroides thetaiotaomicron were reduced, while Faecalibacterium prausnitzii showed an overrepresentation, potentially indicating a protective or prognostic role. Functional alterations were minimal, with the NAD salvage pathway downregulated in premenopausal BC patients, possibly affecting DNA repair. Enterotype analysis revealed that Bacteroides-dominated enterotypes were more common in controls, while Prevotella and Alistipes were enriched in BC patients. Additionally, bacterial diversity was notably lower in postmenopausal BC patients compared to controls, emphasizing the role of gut dysbiosis in BC pathology rather than menopausal status.

GroupMicrobial ChangesFunctional Changes
Premenopausal BC PatientsLower abundances: Bifidobacterium, Collinsella massiliensis. Higher abundances: Gemmiger.Downregulation of NAD salvage pathway, possibly affecting DNA repair.
Postmenopausal BC PatientsReduced levels: Blautia obeum, Dorea formicigenerans, Bacteroides thetaiotaomicron. Overrepresentation: Faecalibacterium prausnitzii.Minimal functional alterations.
Controls vs. BC PatientsBacteroides enterotypes prevalent in controls; Prevotella and Alistipes enriched in BC patients.N/A
Postmenopausal BC Patients (Alpha-Diversity)Lower bacterial diversity compared to controls.N/A

Greatest Implications

The study underscores the importance of gut microbiota in BC development, suggesting that dysbiosis may not be directly related to menopausal status but rather to BC pathology itself. These findings have potential diagnostic implications, as machine learning models using gut microbiota profiles demonstrated an ability to distinguish BC patients from controls with high accuracy (AUC > 0.8). The study highlights the need for further research to explore the mechanisms linking microbiota alterations and BC progression, particularly focusing on key taxa like Faecalibacterium prausnitzii and Bifidobacterium, as well as geographic and lifestyle factors influencing microbiota composition.

Breast cancer in postmenopausal women is associated with an altered gut metagenome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

The study revealed altered gut microbiota in postmenopausal breast cancer patients, with enriched inflammation-associated species and depleted butyrate producers. Functional gene changes suggest links to systemic inflammation and metabolic imbalance, providing insights into microbiota's role in cancer progression.

What Was Studied?

This study investigated the differences in the composition and functional capacities of gut microbiota between postmenopausal breast cancer patients and postmenopausal healthy controls. The researchers conducted a comprehensive shotgun metagenomic analysis to assess microbial diversity, taxonomic abundance, functional gene profiles, and potential associations with clinical indices.

Who Was Studied?

The study involved 44 postmenopausal breast cancer patients and 46 postmenopausal healthy controls, as well as 18 premenopausal breast cancer patients and 25 premenopausal healthy controls. All participants were treatment-naive and free from other conditions such as diabetes or inflammatory bowel diseases, which could confound the microbiome analysis.

What Were the Most Important Findings?

The study found significant differences in gut microbial diversity and composition between postmenopausal breast cancer patients and healthy controls. Microbial diversity was higher in breast cancer patients. Forty-five microbial species exhibited significant differences in abundance; 38 species were enriched in breast cancer patients, including Escherichia coli, Klebsiella sp., and Prevotella amnii, while 7 species, such as Eubacterium eligens and Lactobacillus vaginalis, were depleted. Functionally, the gut metagenomes of patients were enriched in genes linked to lipopolysaccharide (LPS) biosynthesis, iron transport, and secretion systems, which may contribute to systemic inflammation and metabolic alterations. Importantly, butyrate-producing bacteria like Roseburia inulinivorans were reduced in patients, potentially affecting anti-inflammatory processes.

What Are the Greatest Implications of This Study?

This study highlights the potential role of gut microbiota in influencing systemic inflammation, estrogen metabolism, and immune regulation in postmenopausal breast cancer. The enrichment of LPS biosynthesis and iron transport genes points to mechanisms that may drive inflammation and tumorigenesis. The depletion of butyrate producers suggests a loss of anti-inflammatory microbiota functions, underscoring the gut microbiota’s importance in maintaining immune homeostasis. These findings suggest that gut microbiota could serve as biomarkers for breast cancer and potential therapeutic targets to mitigate disease progression.

Breast cancer patients from the Midwest region of the United States have reduced levels of short-chain fatty acid-producing gut bacteria

May 20, 2025
  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This study identifies gut dysbiosis in breast cancer patients, highlighting reduced SCFA-producing bacteria and altered microbial pathways. Findings suggest microbiome-targeted interventions could aid breast cancer treatment.

What was studied?

This study investigated the gut microbiome composition in breast cancer (BC) patients from the Midwest region of the United States, focusing on its taxonomic composition and functional profiling. Using 16S ribosomal RNA sequencing, the study examined the bacterial microbiome, specifically targeting short-chain fatty acid (SCFA)-producing bacteria. It aimed to identify microbial dysbiosis and its potential role in breast cancer pathobiology, emphasizing regional differences in microbiome signatures.

Who was studied?

The study included 22 breast cancer patients and 19 healthy controls, all recruited from the University of Iowa. Participants were matched by race, body mass index (BMI), and sex. Inclusion criteria required BC patients to have invasive breast cancer, with exclusion criteria such as antibiotic use during sample collection. Healthy controls were similarly screened for factors that might impact gut microbiota, like recent antibiotic or laxative use.

What were the most important findings?

The study identified significant gut microbiome differences between breast cancer patients and healthy controls, particularly in alpha and beta diversity measures. Breast cancer (BC) patients showed evidence of gut dysbiosis, including a decrease in beneficial SCFA-producing bacteria and an enrichment of pro-inflammatory taxa. These alterations suggest a microbiome imbalance that may contribute to inflammation and disease progression. Furthermore, the study highlighted functional disruptions in microbiome pathways, with reduced production of SCFAs such as propionate and acetate, which are essential for maintaining gut health and modulating immune responses. These findings underscore the importance of microbiome-targeted interventions to restore microbial balance and support breast cancer treatment.

FindingBreast Cancer Patients (BC)Healthy Controls (HC)Relevance
SCFA-Producing BacteriaReduced Faecalibacterium prausnitzii, Alistipes, Parabacteroides merdae, Lachnospira pectinoschizaHigher levelsSCFA reduction contributes to inflammation and impaired gut motility.
Pro-Inflammatory BacteriaEnriched Eggerthella lenta, Blautia speciesReduced levelsLinked to inflammation and cancer progression.
Functional PathwaysDecreased SCFA pathways (propionate, acetate)Intact pathwaysDysbiosis may exacerbate systemic inflammation and disrupt gut homeostasis.
Beta Diversity ClusteringSignificant clustering distinct from HCNo significant clusteringIndicates an altered microbiome composition in BC.

What are the greatest implications of this study?

The findings underscore the role of gut microbial dysbiosis in breast cancer, with SCFA-producing bacteria depletion linked to inflammation and cancer pathogenesis. This highlights potential avenues for microbiome-targeted therapies, such as probiotics or dietary interventions, aimed at restoring SCFA production and microbial balance. Moreover, the study emphasizes the need for region-specific microbiome research to tailor interventions effectively.

Composition and Functional Potential of the Human Mammary Microbiota Prior to and Following Breast Tumor Diagnosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study explores the mammary microbiota's composition and function before and after breast cancer diagnosis, revealing dysbiosis and metabolic shifts as early markers.

What was studied?

This study investigated the composition and functional potential of the human mammary microbiota in healthy breast tissues and those associated with breast cancer development. The researchers focused on tissue samples collected before cancer diagnosis (prediagnostic or PD), as well as adjacent normal (AN) and tumor (T) tissues from breast cancer patients. Using 16S rRNA sequencing and functional metagenomic predictions, they aimed to identify bacterial dysbiosis and metabolic changes associated with breast cancer progression.

Who was studied?

A total of 141 women were included in the study, contributing 159 breast tissue samples. These included 49 samples from healthy individuals (H), 15 from prediagnostic cases (PD), 49 from adjacent normal tissues (AN), and 46 from tumor tissues (T). The prediagnostic samples were obtained from women who later developed breast cancer, allowing researchers to explore early microbial changes.

What were the most important findings?

The study revealed significant bacterial dysbiosis and metabolic reprogramming in PD, AN, and T tissues compared to healthy tissues. Prediagnostic tissues exhibited an intermediary bacterial composition between healthy and cancerous tissues. Shifts in specific bacterial families such as Bacillaceae, Streptococcaceae, and Corynebacteriaceae were detected in PD tissues and were more pronounced in AN and T tissues. Functional analysis revealed reduced bacterial metabolic activities, particularly pathways related to xenobiotics degradation, which could otherwise protect against carcinogenesis. Additionally, altered correlations between host gene expression and microbial functions were observed, highlighting potential early microbial responses to tumor microenvironments.

What are the greatest implications of this study?

This research highlights the mammary microbiota's potential as a critical biomarker for early breast cancer detection and risk stratification by revealing bacterial dysbiosis and metabolic reprogramming in prediagnostic tissues, suggesting microbial changes may precede clinical symptoms or histological abnormalities. The identification of an intermediary microbiota composition in prediagnostic tissues supports the microbiome's role in early cancer development, indicating microbial shifts as potential early drivers or responders to tumorigenesis. A significant reduction in metabolic functions, such as xenobiotic degradation, in cancer-associated tissues implies a diminished microbial ability to detoxify carcinogens, increasing susceptibility to tumor formation. Altered correlations between microbial taxa and host gene expression further suggest dynamic interactions influencing immune responses, inflammation, and cellular proliferation, with positive associations between microbial functions and tumor-related genes pointing to potential mechanistic links to cancer progression.

These findings not only enhance understanding of the microbiota's role in breast cancer but also offer clinical translation opportunities, including the development of non-invasive diagnostic tools based on prediagnostic microbial signatures, microbiome-modulating therapies to target dysbiosis, and therapeutic interventions aimed at restoring protective bacterial functions and reducing cancer risk.

Comprehensive profiles and diagnostic value of menopausal-specific gut microbiota in premenopausal breast cancer

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study identifies menopausal-specific gut microbial markers and functional pathways linked to breast cancer, offering diagnostic potential and insights into prevention.

What was studied?

This study investigated the gut microbiota profiles, diagnostic value, and functional pathways specific to premenopausal breast cancer patients. It aimed to identify unique gut microbial markers distinguishing premenopausal breast cancer patients from postmenopausal patients and age-matched controls. The study also explored functional pathways of gut microbiota linked to breast cancer progression and diagnostic potential.

Who was studied?

The study analyzed 267 participants divided into four groups: premenopausal controls (Pre-C, n=50), premenopausal breast cancer patients (Pre-BC, n=100), postmenopausal controls (Post-C, n=17), and postmenopausal breast cancer patients (Post-BC, n=100). All breast cancer patients were newly diagnosed with stage I–II disease and excluded if they had received treatments or medications affecting gut microbiota before fecal sample collection.

What were the most important findings?

The study highlights significant differences in gut microbial diversity, composition, and functional pathways between premenopausal and postmenopausal breast cancer patients. Premenopausal breast cancer patients showed reduced α-diversity and distinct β-diversity compared to controls, with alterations in specific bacterial taxa linked to inflammation and cancer progression. In contrast, postmenopausal patients exhibited a different microbial profile, including an increase in pathogenic bacteria. Functional pathway analyses revealed steroid-related and oncogenic pathways in premenopausal patients, while postmenopausal patients were associated with chemical carcinogenesis and aldosterone-regulated pathways. The findings emphasize the diagnostic potential of gut microbiota in differentiating breast cancer subtypes and guiding prevention strategies.

AspectPremenopausal Breast CancerPostmenopausal Breast CancerUniversal Markers (Both Types)
α-DiversitySignificantly reduced compared to controlsNo reduction observed compared to postmenopausal controls-
β-DiversityDistinct from controlsDistinct from controls-
Enriched MicrobesBacteroides fragilis, Anaerostipes (linked to inflammation and progression)Proteobacteria, Klebsiella pneumoniae (pathogenic bacteria)Haemophilus parainfluenzae (increased in both)
Reduced MicrobesBifidobacterium spp. (tumor suppressor)Akkermansia muciniphila (beneficial microbe)Faecalibacterium prausnitzii (decreased in both)
Functional PathwaysSteroid-related pathways; Oncogenic pathways (e.g., Notch/Wnt signaling)Chemical carcinogenesis; Aldosterone-regulated pathways-
Diagnostic PotentialStrong microbial markers for distinguishing premenopausal breast cancerStrong microbial markers for distinguishing postmenopausal breast cancer-

What are the greatest implications of this study?

The findings underscore the diagnostic potential of microbial markers for early, non-invasive breast cancer detection based on menopausal status. Identifying these microbial and functional pathways expands the understanding of breast cancer pathogenesis, especially in premenopausal women. Moreover, the study highlights the gut microbiota as a modifiable factor, suggesting potential interventions like probiotics or dietary changes to mitigate breast cancer risk.

Gut and oral microbial compositional differences in women with breast cancer, women with ductal carcinoma in situ, and healthy women

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

This study reveals distinct gut microbiota profiles in breast cancer and DCIS patients, with reduced alpha diversity and functional shifts linked to inflammation. Major microbial associations, including enriched Bacteroides guilds, underscore potential microbiome-targeted interventions. Oral microbiota showed minimal differences, highlighting the gut's critical role in breast cancer progression.

What was studied?

This study investigated and compared the gut and oral microbiota in three distinct groups: women with breast cancer (BC), women with ductal carcinoma in situ (DCIS), and healthy women. Fecal and oral samples were collected and analyzed using 16S rRNA sequencing to assess microbial diversity, composition, and predicted functional potential.

Who was studied?

The study analyzed samples from 154 women, comprising 73 with BC, 32 with DCIS, and 49 healthy controls. Samples were collected before any therapy to ensure no treatment effects influenced the microbiota.

What were the most important findings?

The study found significant differences in gut microbiota composition and diversity between groups, while the oral microbiota exhibited fewer variations. Women with BC had lower gut microbial alpha diversity compared to healthy women. Beta diversity analysis revealed distinct microbial profiles for the BC and DCIS groups compared to healthy controls. Taxonomic analysis identified several major microbial associations (MMAs) in the gut: the Bacteroides and Enterobacteriaceae guilds were enriched in BC patients, while the Clostridiales guild was more prevalent in healthy women. Functionally, the gut microbiota of BC patients showed increased pathways for lipopolysaccharide (LPS) biosynthesis, glycan metabolism, and sphingolipid metabolism, which are linked to systemic inflammation and cancer progression. Conversely, the oral microbiota showed minimal variation across cohorts, with no significant differences in functional pathways or microbial guilds.

What are the greatest implications of this study?

The findings highlight the role of gut microbiota in breast cancer development and progression. The identification of distinct microbial signatures and functional pathways provides a basis for developing microbiome-targeted interventions aimed at improving treatment outcomes and prognosis. Notably, the lack of significant findings in oral microbiota suggests that gut microbiota might have a more critical role in breast cancer etiology. These results pave the way for further research on microbiome-based diagnostic tools and therapeutic strategies for breast cancer.

Intestinal microbiota influences clinical outcome and side effects of early breast cancer treatment

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

Gut microbiota influences early breast cancer prognosis and treatment side effects, with specific commensals correlating to outcomes. Chemotherapy alters microbiota, favoring beneficial species and improving immune modulation and neuroprotection.

What was studied?

This study examined the impact of intestinal microbiota on the clinical outcomes and side effects of early breast cancer (BC) treatments. Shotgun metagenomics was used to analyze fecal microbiota samples from 76 early BC patients, both pre- and post-chemotherapy. The study aimed to identify specific microbial species associated with BC prognosis and the side effects of chemotherapy, focusing on neurological, gastrointestinal, and metabolic complications. It also explored the functional relevance of gut microbiota in immunocompetent mouse models colonized with BC patient microbiota to establish a causal link between gut microbial composition and tumor growth or therapy efficacy.

Who was studied?

The study involved 76 female BC patients from the CANTO trial (NCT01993498), a long-term prospective cohort designed to quantify and prevent treatment-related toxicities. Patients provided fecal samples before and after chemotherapy, and their plasma was also analyzed for metabolomics. A separate analysis included healthy volunteers (54 Italian and 282 samples from public metagenomes) to contrast microbial signatures. Mouse models were humanized with fecal microbiota from patients and healthy individuals to assess the causal relationship between microbiota and BC outcomes.

What were the most important findings?

The study revealed that the gut microbiota composition significantly correlates with BC prognosis and treatment side effects. Patients with more aggressive tumors (larger size, advanced stage, lymph node involvement) had overrepresentation of species like Clostridiaceae, Veillonella, Bacteroides uniformis, and Blautia wexlerae. In contrast, patients with better prognosis had higher levels of Akkermansia muciniphila, Collinsella aerofaciens, and Eubacterium rectale. Chemotherapy shifted microbial diversity, reducing bacteria associated with poor prognosis and increasing favorable commensals like Methanobrevibacter smithii and Blautia obeum. Functionally, favorable microbiota patterns were linked to neuroprotective and immunomodulatory pathways, such as polyamine biosynthesis and ketogenesis, while unfavorable profiles were associated with inflammation and metabolic dysregulation. Humanized mouse models demonstrated that fecal microbiota from healthy volunteers enhanced tumor response to chemotherapy compared to microbiota from BC patients.

What are the greatest implications of this study?

This study underscores the gut microbiota's role as a biomarker and potential therapeutic target in BC management. The findings suggest that monitoring and modulating gut microbiota could optimize chemotherapy efficacy, mitigate side effects, and improve overall prognosis. Strategies like fecal microbiota transplantation, probiotics, or diet interventions targeting specific microbiota shifts may hold promise. The causal evidence provided by mouse models highlights the translational potential of microbiome-targeted interventions (MBTIs) to improve clinical outcomes for breast cancer patients.

Microbial Dysbiosis Is Associated with Human Breast Cancer

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

Microbial dysbiosis, marked by reduced bacterial load and altered species composition, is linked to breast cancer progression. Enrichment of Methylobacterium radiotolerans in tumors and depletion of Sphingomonas yanoikuyae in normal tissue suggest diagnostic and therapeutic potential for microbiota-based interventions in breast cancer.

What Was Studied?

This study examined the microbiota present in breast tumor tissue compared to paired normal breast tissue from the same individuals, as well as healthy breast tissue from individuals without breast cancer. Using next-generation sequencing and quantitative PCR, the research aimed to identify differences in microbial composition, bacterial load, and their potential impact on the tumor microenvironment and breast cancer progression.

Who Was Studied?

The study included 20 breast cancer patients with estrogen receptor-positive (ER+) tumors, for whom paired tumor and normal adjacent tissue were analyzed. Additional bacterial load analysis included 23 healthy controls undergoing reduction mammoplasty. Gene expression profiling was conducted on tissue from six breast cancer patients and three healthy individuals.

Most Important Findings

The study revealed distinct microbial signatures associated with breast cancer. Methylobacterium radiotolerans was significantly enriched in tumor tissue, while Sphingomonas yanoikuyae was more abundant in paired normal tissue. A strong inverse correlation between the abundance of these two species was observed in normal tissue, but not in tumor tissue. Importantly, bacterial load in tumor tissue was markedly reduced compared to both paired normal and healthy breast tissue, with advanced-stage tumors exhibiting the lowest bacterial counts. This reduction in bacterial load correlated with decreased expression of antibacterial response genes, including Toll-like receptors (TLR2, TLR5, and TLR9) and antimicrobial effectors like IL-12A and BPI.

These findings suggest that microbial dysbiosis and a diminished antibacterial immune response in tumor tissue may contribute to breast cancer progression. Additionally, the results highlight the potential diagnostic value of bacterial load as a marker for breast cancer staging.

Greatest Implications

The association between microbial dysbiosis and breast cancer offers novel insights into the disease’s pathogenesis. The depletion of beneficial bacteria, such as Sphingomonas yanoikuyae, and a reduced immune response may create a permissive environment for tumorigenesis. This study supports the exploration of microbiota as a diagnostic tool and potentially as a therapeutic target to restore a healthy microbial balance and enhance immune surveillance. The inverse correlation between bacterial load and tumor stage underscores its potential utility in disease staging and progression monitoring.

The oral microbiome and breast cancer and non-malignant breast disease, and its relationship with the fecal microbiome in the Ghana Breast Health Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Breast Cancer
    Breast Cancer

    Traditionally linked to genetic predispositions and environmental exposures, emerging evidence highlights the microbiome as a critical and underappreciated factor influencing breast cancer progression, immune response, and treatment outcomes.

The study linked reduced oral microbiome diversity and altered microbial profiles to breast cancer and non-malignant breast disease, highlighting strong correlations between oral and fecal microbiomes in cases versus controls. Genera such as Porphyromonas showed significant inverse associations with breast cancer risk.

What was studied?

This study investigated the relationship between the oral microbiome, breast cancer, and non-malignant breast disease, as well as the correlation between the oral and fecal microbiomes in a case-control population in Ghana. Researchers analyzed microbiome samples from 881 women, including 369 breast cancer cases, 93 non-malignant cases, and 419 controls, using 16S rRNA gene sequencing.

Who was studied?

The study population included Ghanaian women aged 18–74 years who were recruited from Accra and Kumasi. Participants comprised breast cancer patients, individuals with non-malignant breast disease, and population-based controls. Oral and fecal microbiome samples were collected, and demographic, lifestyle, and medical history data were recorded.

What are the Most important findings?

The study revealed that oral microbiome alpha-diversity was significantly lower in breast cancer and non-malignant breast disease cases compared to controls. For instance, each 10-unit increase in observed amplicon sequence variants (ASVs) corresponded to a reduction in the odds of breast cancer and non-malignant breast disease by 14% and 21%, respectively. Beta-diversity analyses also showed distinct microbial community compositions between cases and controls. Key genera, including Porphyromonas and Fusobacterium, were inversely associated with breast cancer, with their relative abundances being significantly lower in cases than in controls. A notable finding was the strong inverse correlation between oral Porphyromonas and fecal Bacteroides in breast cancer cases. This relationship is particularly relevant as fecal Bacteroides has been implicated in estrogen metabolism and breast cancer risk. Breast cancer cases also exhibited stronger correlations between oral and fecal microbiomes compared to controls, suggesting a potential systemic interaction.

Shockingly, the study also found that breast cancer and non-malignant breast disease cases were more likely to have taken antibiotics within the last 30 days compared to controls. This raises critical questions about the role of antibiotics in microbiome disruption and their potential contribution to systemic microbial changes that could influence breast cancer risk.

What are the greatest implications?

This study is extraordinary in its scope and implications. It bridges the gap between two traditionally separate microbiomes—oral and fecal—and ties these microbial systems to breast cancer, a disease of immense global health importance. The findings reveal striking patterns: the inverse associations of oral microbiome diversity and specific genera, such as Porphyromonas and Fusobacterium, with breast cancer and non-malignant breast disease are compelling. These microbes, often linked to periodontal disease, emerge here as potential protective or systemic markers in a population with distinct environmental and health contexts.

The strong correlation between the oral and fecal microbiomes in breast cancer cases further underscores the interconnectedness of microbial communities and highlights systemic microbial interactions that remain underexplored in cancer research. The inverse relationship between Porphyromonas in the oral microbiome and Bacteroides in the fecal microbiome—key players in estrogen metabolism—provides intriguing clues about the mechanisms underlying breast cancer pathogenesis.

Infertility

Go to Category Page

A More Diverse Cervical Microbiome Associates with Better Clinical Outcomes in Patients with Endometriosis: A Pilot Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

This study links cervical microbiome diversity with endometriosis severity. Findings reveal microbial imbalances, particularly in advanced stages, correlate with pain, infertility, and inflammatory pathways. The cervical microbiome may serve as a diagnostic and therapeutic target for improving outcomes in endometriosis, highlighting its role in reproductive health and disease progression.

What Was Studied?

This pilot study investigated the cervical microbiome in patients with endometriosis and its association with clinical outcomes. The research focused on the microbial diversity, composition, and functional roles in cervical mucus, analyzed using 16S rRNA sequencing. The study included healthy women and patients diagnosed with endometriosis to compare microbial profiles and explore the connection between microbiome alterations, disease progression, and associated symptoms like pain, CA125 levels, and infertility.

Who Was Studied?

The study involved 33 women: 10 healthy controls and 23 patients diagnosed with endometriosis (classified by severity into stages I-II and III-IV). The cervical microbiome was analyzed to assess its correlation with clinical features, such as deep infiltrating endometriosis (DIE), CA125 biomarker levels, pain severity, and infertility.

What Were the Most Important Findings?

The study revealed that cervical microbiome diversity is significantly associated with clinical outcomes in endometriosis patients. Specifically, a higher microbial diversity was linked to better outcomes, while notable microbial imbalances characterized advanced disease stages and severe symptoms. Patients with advanced stages of endometriosis exhibited a microbial shift, with an increase in Firmicutes and a decrease in Actinobacteria and Bacteroidetes. Unique microbial profiles were observed, such as elevated Lactobacillus jensenii and Streptococcus agalactiae (GBS), alongside reduced Atopobium vaginae in patients with advanced stages.

Patients presenting severe symptoms, including elevated CA125 biomarker levels, infertility, and higher pain scores, showed significantly reduced microbial richness and diversity. Infertility, a common complication of endometriosis, was associated with an increased Firmicutes/Bacteroidetes ratio. Notably, infertility treatments appeared to reverse these imbalances, restoring microbial diversity and community structure to resemble that of fertile individuals. Additionally, deep infiltrating endometriosis (DIE), a severe form of the condition, was correlated with an overrepresentation of Streptococcus and Prevotella at the genus level.

The study’s functional analyses provided insight into the role of the cervical microbiome in disease progression. Pathways associated with microbial alterations, such as signal transduction, secondary bile acid biosynthesis, and nutrient metabolism, were identified. These pathways may contribute to inflammation, immune dysregulation, and potentially malignancy in severe cases. Such findings underscore the intricate relationship between cervical microbial composition and the pathophysiology of endometriosis. This research positions the cervical microbiome as a critical factor in both the diagnosis and management of endometriosis, offering potential for therapeutic interventions targeting microbial imbalances.

What Are the Greatest Implications of This Study?

The findings suggest that cervical microbiome diversity may serve as a biomarker for diagnosing and monitoring endometriosis progression and complications. The research highlights the therapeutic potential of targeting microbial imbalances to improve clinical outcomes, particularly in infertility. It also underscores the potential link between microbiome alterations and malignancy risks in severe cases, paving the way for preventive and precision medicine approaches in endometriosis management.

Iron Overload and Endometriosis: Mechanisms, Implications, and Therapeutic Targets

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Endometriomas
    Endometriomas

    An endometrioma is a type of ovarian cyst filled with old blood, arising from endometrial tissue outside the uterus, typically causing pain and potentially impacting fertility.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

Iron overload in endometriosis contributes to oxidative stress, inflammation, and tissue damage, driving lesion persistence and subfertility. Ferroptosis resistance and dysregulated iron metabolism highlight therapeutic opportunities using iron chelators and modulators.

What Was Reviewed?

This systematic review evaluated the role of iron in the pathophysiology of endometriosis. The review synthesized findings from 53 studies, including both human and animal research, to provide a comprehensive understanding of how excess iron contributes to oxidative stress, inflammation, and tissue damage in endometriosis. It also explored iron-related mechanisms such as ferroptosis and the implications for subfertility, symptom severity, and potential malignant transformation.

Who Was Reviewed?

The review included a total of 53 studies: 47 human studies involving 3,556 participants and 6 animal studies. The human studies primarily examined women diagnosed with endometriosis, and the included research utilized various bio-samples such as ovarian endometriomas, peritoneal fluid, and ectopic endometrial lesions. Animal studies focused on endometriosis models to explore systemic and local iron mechanics.

Key Findings

Iron overload is consistently found in endometriotic tissues and peritoneal fluid but not in systemic circulation. This localized iron accumulation stems from repeated bleeding within lesions, leading to oxidative stress and inflammation that perpetuates the ectopic growth of endometrial tissue. Dysregulated iron transport and the failure of homeostatic mechanisms contribute to this pathology, with increased expression of proteins such as divalent metal transporter-1 (DMT1) and decreased ferroportin expression in affected tissues.

Markers of oxidative stress, including lipid peroxidation and DNA damage, were significantly elevated in endometriotic lesions. Aberrant resistance to ferroptosis, an iron-dependent form of cell death, was identified as a key mechanism supporting lesion persistence. Additionally, iron-induced ferroptosis was linked to the production of pro-inflammatory and angiogenic factors like IL-8 and VEGFA, exacerbating inflammation and lesion vascularization.

Iron overload was implicated in subfertility, as higher iron concentrations in ovarian follicles and endometriomas were associated with impaired oocyte quality and development. These findings suggest that iron mechanics might influence folliculogenesis and embryo viability. Importantly, the review highlighted the therapeutic potential of iron chelators and ferroptosis modulators for managing endometriosis.

Implications of the Review

This review underscores the central role of aberrant iron metabolism in the pathogenesis of endometriosis, providing a mechanistic basis for its persistence, progression, and associated complications such as subfertility and chronic pain. Iron-related oxidative stress emerges as a critical driver of inflammation and tissue damage, making it a promising target for therapeutic intervention. Future research should explore the efficacy of iron-targeted treatments, such as chelators, and further elucidate the role of ferroptosis in endometriosis. These insights could lead to novel strategies for mitigating symptom severity and improving fertility outcomes in affected women.

The role of gut and genital microbiota and the estrobolome in endometriosis, infertility and chronic pelvic pain

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Chronic Pelvic Pain (CPP)
    Chronic Pelvic Pain (CPP)

    Chronic Pelvic Pain (CPP) is persistent pain in the pelvic region lasting six months or longer, often multifactorial, impacting physical and emotional well-being, and associated with various medical conditions.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

This review highlights the gut and genital microbiome's roles in estrogen-driven conditions like endometriosis, infertility, and CPP, emphasizing dysbiosis' impact on inflammation and estrogen metabolism.

What was reviewed?

The reviewed manuscript explored the intricate relationship between the gut and genital microbiomes, the estrobolome, and their roles in the pathophysiology of endometriosis, infertility, and chronic pelvic pain (CPP). The authors critically examined 28 clinical and six preclinical studies to understand microbial dysbiosis's contributions to estrogen metabolism, inflammation, and symptomatology in these conditions. This review also identified methodological gaps in microbiome studies and proposed strategies to improve future research.

Who was reviewed?

The review included human and animal studies, examining women diagnosed with endometriosis, infertility, and CPP, alongside healthy controls. Specific focus was placed on microbial associations in the gut, cervicovaginal, and endometrial microbiomes, with emphasis on bacterial vaginosis-associated bacteria, Lactobacillus depletion, and microbial influences on estrogen-driven mechanisms.

What were the most important findings?

Key findings highlighted that dysbiosis in the gut microbiome disrupts the estrobolome, an essential modulator of estrogen metabolism. This disruption contributes to heightened systemic and local inflammation, potentially exacerbating endometriosis symptoms and infertility. Many studies noted an association between bacterial vaginosis-related bacteria and a reduction in Lactobacillus dominance in the cervicovaginal microbiome with the prevalence of endometriosis and infertility. Additionally, the review underscored a bidirectional relationship between gut microbiota and endometriosis progression in animal models, emphasizing the role of gut dysbiosis in increasing b-glucuronidase activity, leading to elevated circulating estrogen levels.

What are the greatest implications of this review?

This review underscores the need for rigorous, standardized methodologies to better delineate causal relationships between microbiota and gynecological conditions like endometriosis and CPP. The findings of this review suggest that targeting the microbiome could lead to novel diagnostics and therapeutics for estrogen-driven diseases. The review also highlights the potential of leveraging microbiome-based biomarkers for non-invasive diagnostics and monitoring of endometriosis progression, bridging a critical translational gap in gynecological health.

Bacterial Vaginosis

Go to Category Page

A Multi-Omic Systems-Based Approach Reveals Metabolic Markers of Bacterial Vaginosis and Insight into the Disease

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study used a multi-omic approach to reveal key microbial and metabolic markers of bacterial vaginosis. It identified two distinct symptomatic BV metabotypes, each defined by unique microbial and metabolite associations, underscoring the need for metabolomics-informed clinical diagnostics.

What was studied?

The study investigated the vaginal microbiome and metabolome of reproductive-age women to identify metabolic markers and microbial associations linked to bacterial vaginosis (BV). Researchers used a multi-omic systems-based approach, integrating deep 16S rRNA gene sequencing with metabolomic profiling of vaginal lavage samples collected from 36 women. This study sought to overcome the limitations of traditional diagnostic methods like the Nugent score and Amsel criteria, which have been criticized for inconsistency and inability to accurately capture symptomatic BV cases.

Who was studied?

The study involved 36 women of reproductive age, who varied demographically and behaviorally. Participants were clinically evaluated for BV using Amsel criteria and Nugent scoring. Vaginal lavage samples were collected from these women and subjected to both microbial and metabolic analyses. The cohort included both symptomatic and asymptomatic women, covering a diverse range of Nugent scores and BV symptoms, to enable the identification of associations between microbial taxa, metabolomic profiles, and disease status.

Most Important Findings

The study identified distinct microbial and metabolomic profiles associated with BV. It showed that microbial community composition, as assessed by 16S rRNA gene sequencing, reflected Nugent scores but poorly matched Amsel criteria. In contrast, metabolomic profiles were more aligned with Amsel-defined symptomatic BV, highlighting the potential diagnostic value of metabolic markers.

The researchers distinguished two symptomatic BV metabotypes (SBVI and SBVII), each linked to unique microbial and metabolic features. SBVI correlated with Mobiluncus spp. and Allisonella spp., while SBVII correlated with Hallella spp. Both metabotypes were marked by disruption of epithelial integrity but differed in microbial signatures and metabolic profiles.

Key microbial associations included increased abundance of Gardnerella spp. and Dialister spp. in samples with high Nugent scores. Dialister spp. correlated strongly with elevated levels of putrescine and cadaverine, compounds responsible for BV-associated odor. Mobiluncus spp. were associated with increased 2-methyl-2-hydroxybutanoic acid, linked to vaginal discharge, while Gardnerella spp. were connected to diethylene glycol, associated with vaginal pain. The study also noted that decreases in lactic acid-producing lactobacilli and increases in acetate- and propionate-producing bacteria characterized the BV state. Importantly, the relative abundance of Gardnerella spp. and Dialister spp. was not consistently associated with Amsel criteria, underscoring the complexity of the microbiome-symptom relationship​​​​.

Implications of this Study

This study advances understanding of BV by providing molecular-level evidence that the symptomatic state of BV cannot be attributed solely to microbial composition. Instead, it highlights that metabolic activity and metabolite production, driven by specific bacterial taxa, play a critical role in disease manifestation. The identification of two distinct symptomatic BV metabotypes suggests that BV is not a singular condition but may arise via different microbial and metabolic pathways. These findings imply that clinical diagnostics for BV should integrate metabolomic data alongside microbial profiling to improve accuracy and reduce misclassification based on Nugent score or Amsel criteria alone. These insights open avenues for targeted microbiome-based interventions and the development of metabolite-specific therapeutic strategies.

An Integrated Efficacy and Safety Analysis of Single-Dose Secnidazole 2 g in the Treatment of Bacterial Vaginosis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study confirms secnidazole’s efficacy and safety as a one-dose treatment for BV, improving outcomes while supporting microbiome restoration.

What was studied?

This integrated study analyzed the efficacy and safety of a single-dose 2 g oral formulation of secnidazole (SOLOSEC™) for the treatment of bacterial vaginosis (BV) in women. Drawing from two randomized, double-blind, placebo-controlled pivotal clinical trials, the researchers aimed to evaluate whether this simplified regimen could overcome the adherence challenges commonly associated with the current extended-dose treatments for BV. The study compared clinical cure rates, microbiological outcomes, and adverse event profiles between the secnidazole and placebo groups, providing a comprehensive efficacy and safety assessment to support regulatory approval and clinical use.

Who was studied?

The integrated analysis included 288 women who met all inclusion and exclusion criteria—169 were treated with 2 g of secnidazole, and 119 received a placebo. Participants ranged in age from 18 to 54 years, with a median of 31. The racial composition was diverse, with over 50% identifying as Black or African American. Participants were also stratified by the number of BV episodes in the past year (three or fewer vs. four or more), acknowledging the recurrent nature of BV in many women. All participants had to meet the four Amsel criteria for BV, ensuring consistency with FDA standards.

What were the most important findings?

The integrated analysis demonstrated that single-dose secnidazole significantly improved clinical outcomes compared to placebo. Patients treated with secnidazole were far more likely to experience complete resolution of symptoms, normalization of discharge and odor, and restoration of a healthy vaginal microbiome. Microbiological analysis showed that more patients achieved normal Nugent scores following treatment, which correlates with reduced presence of BV-associated anaerobic bacteria and increased dominance of beneficial Lactobacillus species. The drug was effective across both first-time and recurrent cases and provided consistent benefits regardless of race. Secnidazole targets key BV-associated organisms such as Gardnerella vaginalis, Atopobium vaginae, and Prevotella, while sparing protective lactobacilli. This microbial specificity aligns closely with the recognized dysbiotic profile of BV and suggests secnidazole may facilitate reestablishment of microbiome homeostasis. Adverse effects were mild and infrequent, most commonly involving vaginal yeast overgrowth and transient gastrointestinal discomfort, with no significant safety concerns emerging in the analysis.

What are the implications of this study?

The findings establish secnidazole as a compelling treatment option for BV that addresses both clinical symptoms and the underlying microbial imbalance. The one-dose regimen greatly enhances patient adherence, a crucial factor in reducing recurrence and treatment failure. Because secnidazole selectively targets harmful bacteria while preserving beneficial species, it supports the restoration of a healthy vaginal microbiome, a key goal in microbiome-based therapeutic strategies. The study also reinforces the validity of BV’s microbial signature as a foundation for targeted intervention. As such, secnidazole not only demonstrates therapeutic efficacy but also contributes to a growing paradigm of microbiome-conscious treatment approaches in gynecologic care.

Antimicrobial activity of bovine lactoferrin against Gardnerella species clinical isolates

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

Bovine lactoferrin shows promise as an adjunctive treatment for bacterial vaginosis, inhibiting metronidazole-resistant G. vaginalis strains and potentiating the effects of clindamycin. Its ability to sequester iron and act synergistically with antibiotics could offer an innovative solution to antibiotic resistance in BV.

What was studied?

The study investigated the antimicrobial activity of bovine lactoferrin (MTbLF) against clinical isolates of Gardnerella vaginalis (G. vaginalis), which is a key pathogen in the development of bacterial vaginosis (BV). The study also examined the potential synergistic effects of bovine lactoferrin when combined with commonly used antibiotics, metronidazole and clindamycin. It utilized a range of in vitro experiments to determine the dose-dependent effects of MTbLF and its ability to inhibit the growth of both metronidazole-resistant and susceptible G. vaginalis isolates.

Who was studied?

The study focused on 71 clinical isolates of Gardnerella vaginalis that were presumptively identified from vaginal samples collected from women diagnosed with bacterial vaginosis. The researchers subjected these isolates to antimicrobial susceptibility testing to evaluate their resistance profiles against metronidazole and clindamycin.

What were the most important findings?

The study found that MTbLF exhibited significant antimicrobial activity against G. vaginalis isolates, including those resistant to metronidazole. The inhibitory effect was dose-dependent and not strain-dependent, suggesting that MTbLF could effectively target G. vaginalis, regardless of the strain. Combining MTbLF with clindamycin enhanced the antibiotic's efficacy against G. vaginalis, producing a synergistic effect. This finding highlights the potential of MTbLF as an adjunctive treatment for BV, particularly in cases involving antibiotic-resistant strains. Additionally, the study confirmed that G. vaginalis strains were unable to utilize bovine lactoferrin as an iron source, contrasting with their ability to acquire iron from human lactoferrin, which may contribute to the pathogen’s resilience in the vaginal environment.

What are the implications of this study?

The study highlights the potential of MTbLF as an adjunct or alternative treatment for BV, especially in cases where traditional antibiotics like metronidazole and clindamycin are ineffective due to resistance. Given its iron-binding properties, MTbLF could help disrupt the growth of G. vaginalis by depriving it of essential iron, thereby hindering its ability to proliferate. The observed synergy between MTbLF and clindamycin could pave the way for more effective combination therapies. Furthermore, MTbLF’s ability to inhibit G. vaginalis, even in biofilm-forming states, highlights its potential in managing BV, a condition known for its recurring nature and complexity. These findings warrant further exploration, particularly in clinical settings, to assess the safety and pharmacokinetics of MTbLF in treating and preventing BV recurrence.

Association Between Dietary Patterns and Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

A diet high in processed foods and sugar increases bacterial vaginosis (BV) risk, while a plant-based diet lowers it. This study highlights the importance of dietary choices in vaginal health, providing insights for clinicians on how nutrition influences the vaginal microbiome.

What was Studied?

This study investigated the relationship between dietary patterns and bacterial vaginosis (BV) in women. Researchers analyzed how different diets influenced BV risk, focusing on five major dietary patterns: "Healthy diet," "Unhealthy diet," "Ovo-vegetarian diet," "Pseudo-Mediterranean diet," and "Western diet."

Who was Studied?

The study included 144 women diagnosed with BV and 151 healthy controls. Participants were recruited from a gynecology clinic in Tehran, Iran, between November 2020 and June 2021. Researchers assessed dietary intake using a food frequency questionnaire and diagnosed BV using the Amsel criteria.

Most Important Findings

Women who followed an "Unhealthy diet" high in sugar, solid oils, red meat, sweets, fried potatoes, and refined grains had a significantly higher risk of BV. Those in the highest tertile of this diet were more than three times as likely to have BV compared to those in the lowest tertile.

Conversely, the "Ovo-vegetarian diet," rich in vegetables, beans, whole grains, and eggs, was strongly associated with a lower BV risk. Women in the highest adherence group for this diet had an 84% lower chance of BV compared to those in the lowest adherence group.

The study also observed a protective but not statistically significant effect of the "Pseudo-Mediterranean diet," which includes nuts, fish, olives, and olive oil. No clear association was found between BV and the "Healthy diet" or "Western diet."

Microbiome analysis linked the "Unhealthy diet" with a disruption in vaginal flora, favoring BV-associated bacteria like Gardnerella vaginalis, Bacteroides spp., Mobiluncus spp., and Mycoplasma hominis. In contrast, the "Ovo-vegetarian diet" promoted conditions favorable for Lactobacillus dominance, which helps maintain vaginal health.

Implications of the Study

This study reinforces the role of diet in vaginal microbiome balance and BV risk. Clinicians should encourage patients to reduce processed foods, refined sugars, and saturated fats while promoting a plant-based diet rich in fiber, whole grains, and essential nutrients. Future research should explore whether dietary modifications can serve as an effective strategy for BV prevention and treatment.

Association Between Heavy Metal Exposure and Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This cross-sectional study identified a strong link between elevated serum lead and cadmium levels and increased risk of bacterial vaginosis. It suggests that heavy metal exposure may disrupt vaginal microbiota stability and immunity, contributing to BV susceptibility and pointing to new environmental factors in BV prevention strategies.

What was Studied?

The study examined the association between exposure to heavy metals, specifically lead, cadmium, and mercury, and the risk of bacterial vaginosis (BV) among American women. Using a cross-sectional design, the researchers analyzed data from 2,493 women aged 18 to 49 years who participated in the 2001–2004 cycles of the National Health and Nutrition Examination Survey (NHANES). They measured serum levels of these heavy metals and assessed BV status using Nugent scoring, aiming to clarify whether environmental exposure to heavy metals correlates with BV prevalence.

Who was Studied?

The study included 2,493 American women aged between 18 and 49 years. All participants were selected from NHANES datasets, which provide a representative sample of the U.S. population. The researchers collected vaginal swabs to diagnose BV using the Nugent score and measured serum concentrations of lead, cadmium, and mercury. They controlled for several covariates such as age, body mass index, socioeconomic factors, cholesterol levels, and physical activity to ensure reliable statistical analysis.

Most important findings

The study found a significant positive association between serum lead and cadmium levels and the risk of developing bacterial vaginosis. Specifically, women with the highest serum lead concentrations had a 35% increased risk of BV compared to those with the lowest levels. Similarly, higher cadmium levels were associated with a 41% increased risk of BV in fully adjusted models. However, the researchers found no significant association between serum mercury levels and BV risk.

Stratified analyses revealed that the positive association between lead exposure and BV was more pronounced in women aged 37 to 49 years, those with lower education levels, and those with a higher body mass index. For cadmium, the risk was especially higher among women aged 18 to 24 and 37 to 49 years, and among those of non-Hispanic white and black ethnicity. These results suggest that lead and cadmium may influence vaginal microbiota stability, possibly through immunotoxic or endocrine-disrupting mechanisms, contributing to vaginal dysbiosis and increased BV susceptibility.

Implications of this Study

This study provides the first epidemiological evidence linking heavy metal exposure to increased risk of bacterial vaginosis. The findings suggest that environmental pollutants may act as overlooked risk factors in BV pathogenesis by compromising host immune function, disrupting hormonal balance, and potentially altering the vaginal microbiome. Clinicians and public health officials should consider environmental heavy metal exposure as part of BV risk assessment and prevention strategies. Reducing heavy metal exposure through regulatory policies and patient education could offer an additional layer of protection against BV and its associated reproductive health risks. These results highlight the importance of integrating environmental factors into the broader framework of microbiome-related disease prevention.

Bacterial biota of women with bacterial vaginosis treated with lactoferrin

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study explored the effects of lactoferrin on the vaginal microbiota in women with bacterial vaginosis, showing its potential as a non-antibiotic treatment option.

What was studied?

This study investigated the bacterial biota in women with bacterial vaginosis (BV) and assessed the effects of two different concentrations of vaginal lactoferrin pessaries (100 mg and 200 mg) on the vaginal bacterial composition. The aim was to characterize the vaginal microbiota before, during, and after lactoferrin treatment.

Who was studied?

Sixty sexually active women of reproductive age (18–45 years old) with symptomatic acute BV were studied. The women were randomly assigned to two groups: one group received 200 mg lactoferrin vaginal pessaries, and the other received 100 mg lactoferrin vaginal pessaries.

What were the most important findings?

The study showed that lactoferrin treatment significantly altered the vaginal microbiota in women with BV. During the treatment, both 100 mg and 200 mg doses of lactoferrin reduced the abundance of bacteria commonly associated with BV, such as Gardnerella, Prevotella, and Lachnospira. Concurrently, the levels of Lactobacillus species increased. The most significant changes were with the 200 mg lactoferrin dose, which maintained the bacterial balance up to 2 weeks after treatment. In contrast, the 100 mg dose did not maintain the microbiota balance as effectively post-treatment, with an increase in Gardnerella and Prevotella species observed. The study also highlighted that Lactobacillus helveticus became the dominant species during and after treatment, a species not previously detected in the vaginal microbiome of these participants.

What are the implications of this study?

The findings suggest that lactoferrin could be a viable alternative therapeutic approach for BV, offering a non-antibiotic treatment option. By promoting the growth of Lactobacillus species and reducing pathogenic bacteria, lactoferrin helps restore a healthier vaginal microbiota. This approach may overcome some of the limitations of antibiotic treatments for BV, such as recurrence and resistance development. The emergence of Lactobacillus helveticus during lactoferrin treatment suggests the potential for new probiotic strains for BV management. These results also point toward the need for further research on the role of lactoferrin in maintaining long-term vaginal health.

Bacterial Communities in Women with Bacterial Vaginosis:

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study used high-resolution sequencing to analyze bacterial communities in women with BV. Findings reveal distinct microbial associations, diagnostic inconsistencies, and racial differences in BV prevalence, highlighting the need for microbiome-targeted therapies and improved diagnostic tools.

What Was Studied?

This study investigated the composition and diversity of vaginal bacterial communities in women with bacterial vaginosis (BV) using high-resolution phylogenetic analysis. Researchers aimed to identify specific bacterial species associated with BV and determine their relationship to clinical diagnostic criteria. By employing deep sequencing of the 16S rRNA gene, the study provided a more precise taxonomic classification of BV-associated bacteria.

Who Was Studied?

The study analyzed vaginal swabs from 220 women with and without BV. Researchers diagnosed BV using Amsel’s clinical criteria and confirmed cases with Gram staining. The study examined bacterial communities in diverse participants, including Black and White women, to assess potential differences in microbiome composition by race.

Key Findings and Microbial Associations

Women with BV exhibited highly diverse and heterogeneous vaginal bacterial communities, unlike those without BV, whose microbiomes were dominated by Lactobacillus crispatus or Lactobacillus iners. The study identified Leptotrichia amnionii and Eggerthella sp. as the only bacteria significantly associated with all four Amsel’s diagnostic criteria for BV. Other BV-associated bacteria, including Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., and Sneathia sanguinegens, correlated with specific diagnostic features, such as clue cells and amine odor.

The study also revealed distinct subgroups of BV-associated bacteria that co-occurred, suggesting metabolic interdependencies. Notably, Black women without BV had higher levels of certain BV-associated bacteria than White women, which may contribute to BV’s higher prevalence in this population. Additionally, the findings highlighted discrepancies between Amsel’s criteria and Nugent scoring, emphasizing the need for more refined diagnostic tools.

Implications of the Study

This study highlights the complexity of BV as a polymicrobial condition rather than an infection caused by a single pathogen. Identifying bacterial species at high resolution improves diagnostic accuracy and reveals potential microbial interactions that sustain BV. The research supports the need for microbiome-targeted therapies rather than broad-spectrum antibiotics, which often fail to prevent recurrence. Racial differences in vaginal microbiota composition suggest that BV treatment strategies should account for population-specific variations.

These findings emphasize the importance of advanced sequencing techniques in BV research, providing a foundation for future studies to develop better diagnostic criteria and treatment options.

Bacterial Vaginosis - A Brief Synopsis of the Literature

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explores bacterial vaginosis, emphasizing its recurrence, microbial associations, and treatment challenges. It highlights the need for microbiome-based therapies, standardized diagnostic criteria, and potential partner treatment to reduce reinfection. The findings underscore the importance of improved strategies for long-term BV management.

What Was Reviewed?

This review provides a comprehensive synopsis of the current literature on bacterial vaginosis (BV), focusing on its epidemiology, recurrence, persistence, and treatment challenges. The authors examine the impact of BV on reproductive and sexual health, highlighting its association with sexually transmitted infections (STIs) and adverse pregnancy outcomes. Additionally, the review explores the role of the vaginal microbiota in BV pathogenesis, emphasizing the need for more effective long-term treatment options and standardized definitions for recurrent and persistent BV.

Who Was Reviewed?

The review synthesizes studies on women of reproductive age diagnosed with BV, including those with recurrent infections. It also explores research on the vaginal microbiota, sexual partners' role in BV transmission, and the effectiveness of current treatments. By analyzing epidemiological data from various regions, it highlights differences in BV prevalence and risk factors.

Key Findings and Microbial Associations

BV shifts the vaginal microbiota by depleting Lactobacillus species and allowing anaerobic bacteria like Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., and Mobiluncus spp. to overgrow. This microbial imbalance disrupts the vaginal ecosystem and increases susceptibility to STIs, including Neisseria gonorrhoeae, Chlamydia trachomatis, and HIV. Even with metronidazole or clindamycin treatment, BV recurs in up to 80% of cases within three months. The review explores BV recurrence, showing how reinfection, an inability to restore a Lactobacillus-dominant microbiota, and bacterial biofilms contribute to persistent infections. Clinicians rely on Amsel’s criteria and Nugent scoring for diagnosis, but inconsistent definitions of recurrent and persistent BV complicate management. The review also examines partner treatment as a strategy to reduce BV recurrence, though past studies show mixed results.

Implications of the Review

BV remains a significant clinical challenge due to its high recurrence rates, unclear etiology, and association with reproductive health complications. This review calls for more research into microbiome-based therapies, improved diagnostic tools, and standardized definitions of recurrent BV. The findings suggest that future treatment approaches should not only target BV-associated bacteria but also focus on restoring a stable vaginal microbiota. Additionally, reconsidering partner treatment as part of BV management could be an avenue for reducing recurrence rates, provided that future studies can confirm its effectiveness.

Bacterial vaginosis and biofilms: Therapeutic challenges and innovations

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review links BV recurrence to resilient biofilms formed by Gardnerella vaginalis. Probiotics and biofilm disruptors (e.g., Astodrimer gel) improve outcomes by restoring Lactobacillus dominance. Current antibiotics fail to penetrate biofilms, necessitating multimodal therapies. Future research should explore VMT and microbiome-targeted interventions for sustained BV remission.

What was Reviewed?

This narrative review examined the role of biofilms in bacterial vaginosis (BV), focusing on their contribution to treatment resistance and recurrence. The authors synthesized evidence from clinical studies and trials to evaluate the limitations of current antibiotic therapies and explored emerging solutions, such as biofilm-disrupting agents and probiotics, to improve BV management.

Who was Reviewed?

The review analyzed data from diverse patient populations in clinical studies, including women with recurrent BV. It incorporated findings from trials investigating biofilm-targeted therapies, such as enzymatic disruptors (e.g., dispersin B) and probiotics (e.g., Lactobacillus crispatus), to assess their efficacy in restoring vaginal microbiota balance.

What were the most Important Findings?

The review highlighted that BV-associated biofilms, primarily formed by Gardnerella vaginalis and Atopobium vaginae, shield pathogenic bacteria from antibiotics, driving recurrence. Major microbial associations (MMA) included polymicrobial anaerobic communities displacing protective Lactobacillus species. Probiotics and biofilm-disrupting agents (e.g., boric acid, Astodrimer gel) showed promise in clinical trials, with probiotics delaying recurrence by 51% and Astodrimer gel significantly reducing recurrence rates. Notably, Lactobacillus crispatus-based therapies were emphasized for restoring vaginal acidity and inhibiting biofilm formation.

What are the Implications of this Review?

The findings emphasize the need to shift from antibiotic-only approaches to multimodal strategies targeting biofilms. Clinicians should consider adjunct therapies like probiotics and biofilm disruptors to enhance treatment efficacy and reduce recurrence. The review also calls for further research into vaginal microbiome transplantation (VMT) and personalized therapies to address biofilm resilience.

Bacterial Vaginosis and Chlamydia in Tubal Infertility

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

Study links BV and past chlamydial infection to tubal infertility, with 87.5% of BV-positive women having tubal damage. Both infections were often asymptomatic. IVF pregnancy rates were unaffected, but BV showed lower implantation trends. Findings highlight BV’s role in infertility, urging early screening to prevent tubal damage.


What was Studied?

This cross-sectional study investigated the association between bacterial vaginosis (BV), past chlamydial infection, and tubal infertility in women undergoing IVF. The researchers analyzed vaginal swabs and serologic data from 286 women undergoing 344 IVF cycles to determine whether these infections impacted pregnancy rates or were linked to specific infertility causes.

Who was Studied?

The study included 286 women undergoing IVF treatment at a tertiary care infertility referral center in Glasgow, Scotland. Participants provided high vaginal and endocervical swab samples before oocyte retrieval, with serologic testing for Chlamydia trachomatis and BV diagnosis based on Gram staining and anaerobic culture.

What were the most Important Findings?

The study found strong, independent associations between tubal infertility and both BV (87.5% of BV-positive women had tubal damage) and past chlamydial infection (91.2% seropositivity in tubal infertility cases). Notably, BV and chlamydial infections were frequently asymptomatic, with no active chlamydial infections detected. Major microbial associations (MMA) included reduced Lactobacillus dominance in BV-positive women, alongside overgrowth of anaerobic bacteria. Despite these associations, pregnancy rates after IVF were unaffected by BV or past chlamydial infection, though BV-positive women had numerically lower implantation rates (15.2% vs. 31.0% in chlamydia-seropositive women).

What are the Implications of this Study?

The findings underscore BV as a potential pelvic pathogen contributing to tubal damage, independent of chlamydial infection. While IVF success rates remained comparable across groups, the high prevalence of tubal infertility in BV-positive women suggests that early screening and treatment of asymptomatic BV could prevent long-term reproductive complications. Clinicians should consider BV as a modifiable risk factor in infertility workups, particularly in cases of unexplained tubal pathology.

Bacterial Vaginosis and its Association with Infertility, Endometritis, and Pelvic Inflammatory Disease

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

BV disrupts Lactobacillus dominance, increasing infertility risk via inflammation, PID, and endometritis. L. crispatus probiotics reduce BV recurrence; CE treatment boosts IVF success. Early screening and microbiome-targeted therapies are vital to prevent reproductive complications.

What was Reviewed?

This expert review examined the associations between bacterial vaginosis (BV), endometritis, pelvic inflammatory disease (PID), and infertility, synthesizing evidence from clinical studies, microbiome research, and treatment outcomes. The authors explored how BV-related dysbiosis contributes to upper genital tract infections and reproductive complications, while evaluating diagnostic challenges and emerging therapeutic strategies.

Who was Reviewed?

The review analyzed data from diverse populations of reproductive-age women, including those with infertility, recurrent BV, or PID. It incorporated findings from studies on vaginal and endometrial microbiota, clinical trials on BV treatments (e.g., antibiotics, probiotics), and research on immune and inflammatory responses linked to infertility.

What were the most Important Findings?

BV, characterized by reduced Lactobacillus dominance and overgrowth of anaerobes like Gardnerella vaginalis and Atopobium vaginae, was strongly associated with tubal infertility (3.3-fold higher prevalence in infertile women) and PID. Major microbial associations (MMA) included elevated levels of proinflammatory cytokines (IL-1β, IL-6, IL-8) in BV-positive women, which disrupt endometrial receptivity. Subclinical PID, often linked to BV, reduced pregnancy likelihood by 40%. Notably, Lactobacillus crispatus probiotics reduced BV recurrence by 15% compared to placebo, while endometrial microbiota dominated by non-lactobacilli correlated with lower IVF success rates. Chronic endometritis (CE), prevalent in 34%–66% of unexplained infertility cases, improved fertility outcomes post-antibiotic treatment, with cured CE showing a 76.3% pregnancy rate versus 20% in persistent cases.

What are the Implications of this Review?

The findings underscore BV as a modifiable risk factor for infertility, emphasizing the need for early screening and treatment to prevent PID and CE. Clinicians should consider Lactobacillus-based probiotics and biofilm-disrupting agents for recurrent BV. For infertility workups, endometrial microbiota analysis and CE testing are critical, particularly in cases of repeated implantation failure. Future research should prioritize longitudinal studies to clarify causal links between BV dysbiosis and infertility, while optimizing personalized therapies targeting the vaginal microbiome.

Bacterial Vaginosis Biofilms: Challenges to Current Therapies and Emerging Solutions

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review discusses bacterial vaginosis, biofilm formation, and emerging therapies targeting biofilms for more effective BV treatments.

What was reviewed?

The paper provides a comprehensive review of bacterial vaginosis (BV), its association with biofilm formation, and challenges related to current treatment strategies. The review explores the microbial composition of BV, focusing on the primary pathogen, Gardnerella vaginalis, and the complex nature of BV biofilms, which contribute to the high recurrence rates of the infection. The review presents emerging therapeutic alternatives targeting BV biofilms, including natural antimicrobial agents and biofilm disruptors.

Who was reviewed?

The review examined various studies, clinical trials, and scientific literature that explored the microbial nature of bacterial vaginosis (BV), focusing on biofilm formation and its implications for treatment. It also reviewed the role of G. vaginalis and other anaerobic bacteria in the pathogenesis of BV, along with current and emerging treatment strategies targeting these biofilms. The review synthesized information from studies that investigated the efficacy of traditional therapies, such as metronidazole and clindamycin, as well as novel biofilm-disrupting agents like DNases, probiotics, and plant-derived antimicrobials.

What were the most important findings?

The review emphasizes the polymicrobial nature of bacterial vaginosis, with a marked decrease in beneficial lactobacilli species and an increase in anaerobic bacteria, such as Gardnerella vaginalis, Atopobium vaginae, Mobiluncus spp., Bacteroides spp., and Prevotella spp. A major highlight of the paper is the critical role of biofilms in BV pathogenesis, as these microbial communities exhibit significant resistance to conventional antibiotic treatments like metronidazole. This biofilm formation creates a dense matrix that protects the bacteria from immune system clearance and limits the effectiveness of standard therapies. Biofilms composed primarily of G. vaginalis are particularly resilient, contributing to treatment failure and the recurrence of BV. The review further discusses how researchers are exploring novel therapies, such as DNases, retrocyclins, probiotics, and plant-derived antimicrobials, to overcome biofilm-related antibiotic resistance. The paper also identifies the need for more research into multi-species biofilm interactions to develop more effective treatments for BV.

What are the implications of this review?

The implications of this review are significant for the clinical management of BV. The findings highlight the need for new treatment strategies that can specifically target biofilms, which are a major obstacle to the eradication of BV. Given the high recurrence rates of BV despite current antibiotic therapies, exploring alternative treatments that can disrupt biofilm structures, such as biofilm disruptors and natural antimicrobials, is essential. Clinicians may benefit from being aware of emerging treatments that could offer better outcomes, particularly for recurrent BV cases that do not respond well to standard treatments. Additionally, the review underscores the importance of considering the entire microbiome, including lactobacilli, when developing treatment plans to ensure that therapies do not disrupt the beneficial microbial community, which is crucial for vaginal health.

Bacterial Vaginosis Is Associated with Variation in Dietary Indices

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

A high glycemic load diet increases bacterial vaginosis (BV) risk, while nutrient-rich diets lower it. This study highlights the link between diet quality and vaginal microbiome balance, providing insights for clinicians on how dietary interventions may help prevent BV.

What was Studied?

This study examined how dietary indices, including glycemic load (GL), glycemic index (GI), the Healthy Eating Index (HEI), and the Naturally Nutrient Rich (NNR) score, influence bacterial vaginosis (BV) prevalence, progression, and persistence. Researchers aimed to determine whether overall dietary quality, rather than just individual nutrients, affects vaginal microbiome balance.

Who was Studied?

The study analyzed data from 1,735 nonpregnant women aged 15 – 44, primarily African American (85.5%), recruited from health clinics in Birmingham, Alabama. Researchers assessed annual dietary intake using the Block98 food frequency questionnaire and classified vaginal flora using Nugent Gram-stain criteria.

Most Important Findings

A higher glycemic load significantly increased the risk of BV. For every 10-unit increase in GL, the likelihood of BV progression and persistence rose. High-GL diets, which result in frequent blood sugar spikes, may contribute to vaginal flora imbalances and increased oxidative stress, reducing the body's ability to maintain a protective microbiome.

Conversely, women with higher NNR scores, which reflect greater nutrient density per calorie, had a lower risk of BV. This suggests that diets rich in vitamins, minerals, and fiber may help support a healthy vaginal microbiome. HEI scores above 70, indicating greater adherence to dietary guidelines, were associated with a reduced BV risk, but this association was only borderline significant after adjusting for confounding factors.

Unlike GL, which considers both carbohydrate quality and quantity, glycemic index (GI) showed no clear link to BV. Because GI only measures how quickly food raises blood sugar without accounting for quantity, it may not fully capture how diet affects vaginal health.

Implications of the Study

This study highlights the role of diet quality in vaginal microbiome health. Clinicians should counsel patients on the risks of high-GL diets and emphasize nutrient-dense food choices to lower BV risk. Future research should explore how dietary modifications influence BV outcomes and whether interventions targeting glycemic load can serve as preventive measures.

Bacterial Vaginosis Recurrence: Drivers, Challenges, and Treatment

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explores bacterial vaginosis recurrence, emphasizing microbial persistence, reinfection, and partner treatment. It highlights the need for improved therapeutic strategies, including antimicrobial and microbiome-focused approaches, to reduce recurrence and improve long-term cure rates.

What Was Reviewed?

This review examines the drivers of bacterial vaginosis (BV) recurrence and the challenges and opportunities in partner treatment. BV is a prevalent vaginal condition with a high recurrence rate, often within six months of treatment. The review explores the mechanisms behind recurrence, including reinfection from untreated sexual partners, the persistence of BV-associated bacteria (BVAB), and the role of biofilms. It also discusses past research on whether treating male and female sexual partners can improve BV cure rates and the barriers to implementing partner treatment in clinical settings.

Who Was Reviewed?

The review evaluates studies involving women diagnosed with BV, particularly those who experience recurrent infections. It also incorporates research on the sexual partners of these women, analyzing the microbial composition of the male urethra and penile skin and how bacterial exchange between partners may contribute to reinfection. Clinical trials on partner treatment strategies, including those testing antibiotics like metronidazole and clindamycin, are also assessed.

Key Findings and Microbial Associations

BV recurrence has multiple causes, with reinfection from sexual partners being an overlooked factor. The review shows that BV-associated bacteria like Gardnerella vaginalis, Atopobium vaginae, and Prevotella spp. can transfer between partners, including heterosexual and same-sex couples. This supports the idea that BV may have a sexually transmissible component.

Past studies on partner treatment have had mixed results, leading to skepticism. Many were limited by small sample sizes, inconsistent diagnostic criteria, and short follow-ups. However, newer research suggests that when partners adhere to antibiotics, recurrence rates may drop. The review highlights challenges in partner treatment trials, including low participation and the need for therapies targeting bacterial biofilms and antimicrobial resistance.

Implications of the Review

BV’s high recurrence rate not only causes discomfort but also increases risks like preterm birth and STI susceptibility. This review calls for a fresh approach, including reconsidering partner treatment. While current guidelines do not recommend treating partners, emerging evidence suggests that doing so, alongside therapies that promote a stable, lactobacilli-dominated vaginal microbiota, may improve long-term cure rates. Future studies should refine partner treatment, enhance antimicrobial strategies, and develop microbiome-targeted therapies to prevent recurrence.





Bacterial Vaginosis: Current Diagnostic Avenues and Future Opportunities

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explores bacterial vaginosis diagnostics, highlighting traditional limitations and emerging molecular solutions. Advances in sequencing, metabolomics, and AI-driven analysis offer promising improvements, enhancing diagnostic accuracy and treatment strategies.

What was reviewed?

This review examines the current methods for bacterial vaginosis (BV) diagnosis and explores future opportunities for improving diagnostic accuracy. It provides an in-depth analysis of traditional clinical and microscopic diagnostic methods, their limitations, and the potential of emerging molecular, metabolomic, and proteomic approaches. The review also highlights the microbiome’s role in BV pathogenesis and discusses how advances in sequencing technologies and biomarker discovery could enhance diagnosis and treatment.

Who was reviewed?

The review synthesizes findings from multiple studies on BV diagnosis, including research on bacterial populations associated with BV, clinical diagnostic criteria, and emerging molecular techniques. It draws from a wide range of studies on vaginal microbiome composition, molecular assays, and point-of-care (POC) diagnostic tools.

What were the most important findings?

Bacterial vaginosis occurs when the vaginal microbiome shifts, reducing lactobacilli and increasing anaerobic bacteria like Gardnerella vaginalis and Atopobium vaginae. Traditional diagnostic methods, including Amsel’s criteria and the Nugent score, have been widely used for decades. However, both have limitations, especially in detecting asymptomatic cases.

Amsel’s criteria require at least three of four clinical signs: thin discharge, high vaginal pH, clue cells, and a fishy odor. The Nugent score relies on Gram staining and bacterial morphotypes. Both methods are subjective and prone to interobserver variability, leading to misdiagnosis, particularly in resource-limited settings.

Molecular diagnostic tools offer better sensitivity and specificity. Nucleic acid amplification tests (NAATs) detect multiple BV-associated bacteria in a single test, making diagnoses more accurate. Next-generation sequencing (NGS) has revealed that BV results from a polymicrobial community, not a single pathogen.

New diagnostic approaches include metabolomics and proteomics, which analyze metabolic byproducts and proteins linked to BV. The sialidase enzyme, produced by BV-associated bacteria, is a promising diagnostic marker. Proteomic studies have identified immune-related proteins that change in BV. These molecular markers could improve diagnostic accuracy and enable personalized treatments.

Artificial intelligence (AI) and machine learning are also being explored for BV diagnosis. AI models analyze microbiome data, metabolomic signatures, and patient outcomes to identify patterns. These advancements could enhance diagnostic precision, especially in clinical settings where fast, accurate, and cost-effective tests are essential.

What are the implications of this review?

The findings highlight the urgent need for improved BV diagnostics, especially in resource-limited settings where syndromic management is common. Relying only on symptoms often leads to misdiagnosis and unnecessary antibiotic use, increasing resistance. Shifting to molecular diagnostics and biomarker-based testing could improve accuracy, reduce misdiagnosis, and enhance treatment outcomes. A key takeaway is that BV diagnosis should go beyond bacterial identification. It should include microbial interactions, biofilm presence, metabolic activity, and immune responses. Rapid point-of-care molecular tests, combined with machine learning and biomarker-based approaches, could greatly improve BV diagnosis and management. Understanding microbial communities and biofilms in BV may also lead to better treatments, including microbiome-targeted therapies and potential vaccines. Since BV increases the risk of sexually transmitted infections, preterm birth, and reproductive issues, improving diagnostic accuracy is essential for better patient outcomes.

Bacterial Vaginosis: What Do We Currently Know?

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explores bacterial vaginosis (BV), emphasizing microbial shifts, diagnostic challenges, and treatment strategies. It highlights the role of biofilms, emerging molecular diagnostics, and microbiome-based therapies like probiotics and vaginal microbiota transplantation, advocating for more effective, microbiome-informed approaches to managing BV and its recurrence.

What Was Reviewed?

This review provides an in-depth exploration of bacterial vaginosis (BV), focusing on its etiology, diagnostic challenges, and treatment strategies. It explores the link between the vaginal microbiome and bacterial vaginosis, highlighting the shift from a Lactobacillus-dominated environment to one dominated by anaerobic bacteria like Gardnerella vaginalis and Atopobium vaginae. The review evaluates molecular and clinical diagnostic tools such as Amsel’s criteria, Nugent scoring, and PCR-based methods. Additionally, it highlights the limitations of antibiotic treatments due to high recurrence rates. It also explores emerging therapies, including probiotics, vaginal microbiota transplantation (VMT), and biofilm-targeting strategies​.

Who Was Reviewed?

This review synthesizes data from various studies examining the vaginal microbiome and its role in BV. It considers research on women of reproductive age from different geographic regions and ethnic backgrounds, recognizing the variability in vaginal microbiota composition. The review also addresses the broader clinical implications of BV, notably its links to sexually transmitted infections, pregnancy complications, and reproductive health​.

What Were the Most Important Findings?

The most significant finding is that BV is a polymicrobial shift, not an infection caused by a single pathogen. A healthy vaginal microbiome is dominated by Lactobacillus spp, but BV causes Lactobacilli decline and anaerobe overgrowth, including Gardnerella vaginalis and Prevotella spp. These bacteria form biofilms that contribute to antibiotic resistance and high recurrence rates.

The review highlights the flaws in traditional diagnostic methods. Amsel’s criteria and Nugent scoring are widely used but lack precision. PCR-based molecular diagnostics provide more accuracy and reliability. Emerging enzymatic and nanotechnology-based diagnostic tools offer potential advancements in BV detection.

Treatment challenges are another crucial aspect. Standard antibiotic therapies, including metronidazole and clindamycin, have a 50% recurrence rate within six months. This has driven interest in alternative approaches, including probiotics aimed at restoring Lactobacillus populations, vaginal microbiota transplantation (VMT) as a means of repopulating healthy microbiota, and biofilm-disrupting agents such as DNases and antimicrobial peptides. Additionally, the review explores the role of sexual transmission in BV persistence and the potential benefits of treating male partners​.

What Are the Implications of This Review?

The findings in this review emphasize the need for more effective diagnostic and therapeutic approaches for BV. The recognition of BV as a polymicrobial dysbiosis rather than a traditional infection suggests that future treatments should focus on restoring a healthy microbiome rather than eliminating bacteria. The high recurrence rate associated with antibiotic treatments highlights the need for strategies that address biofilm-associated resistance and microbiome resilience.

Probiotic-based interventions and vaginal microbiota transplantation could redefine BV treatment by offering long-term microbiome stability. Moreover, the identification of novel diagnostic biomarkers and rapid molecular techniques may enhance early detection and targeted interventions. Clinically, incorporating microbiome-focused therapies into gynecologic and obstetric care could improve reproductive health outcomes by reducing BV-related complications significantly. BV complications include increased susceptibility to sexually transmitted infections and adverse pregnancy events, emphasizing the need for innovative microbiome-based treatments urgently. The review ultimately advocates for a shift toward microbiome-informed medical strategies for managing BV.

Boric Acid for the Treatment of Vaginitis: New Possibilities Using an Old Anti-Infective Agent

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

Boric acid offers promising results for treating resistant vulvovaginal candidiasis, bacterial vaginosis, and trichomoniasis. It shows comparable efficacy to oral itraconazole, with fewer adverse events.

What was studied?

The study focused on the use of boric acid as a treatment for various types of microbial vaginitis, specifically vulvovaginal candidiasis (VVC), bacterial vaginosis (BV), and trichomoniasis. Researchers aimed to compare its efficacy with conventional treatments and determine its potential as an alternative or supplementary therapy.

Who was studied?

This review evaluated clinical trials, observational studies, and interventional studies, including case series and reports. It did not focus on a single group of patients but instead summarized findings from various studies involving individuals with VVC, BV, and trichomoniasis. The studies reviewed ranged from those using boric acid for mycotic vaginitis to those evaluating its effect on bacterial vaginosis and trichomoniasis.

What were the most important findings?

The systematic review revealed that boric acid (BA) demonstrated a promising efficacy profile in treating vulvovaginal candidiasis (VVC), particularly in cases caused by Candida glabrata, which is resistant to azole treatments. The review found an average cure rate of 76% for VVC treated with BA. For recurrent bacterial vaginosis, BA combined with 5-nitroimidazole showed effective control, with promising results for reducing relapses. Maintenance therapy with BA also showed similar efficacy to oral itraconazole for VVC and BV, suggesting it may serve as an alternative for managing these conditions. For Trichomonas vaginalis, prolonged boric acid monotherapy cured a substantial portion of patients with recurrent infections, although the exact regimen still requires further research. The study found that the adverse events associated with boric acid treatment were minimal, with a 7.3% occurrence of mild, temporary side effects.

What are the implications of this study?

The rising antimicrobial resistance in vaginitis pathogens, especially those resistant to conventional treatments such as azoles and metronidazole, makes boric acid an appealing alternative. Its broad-spectrum antimicrobial action, including the inhibition of biofilm formation, makes it a strong candidate for treating persistent and recurrent infections. The study suggests that boric acid could be integrated into treatment regimens for patients with recurrent vulvovaginal candidiasis, bacterial vaginosis, and trichomoniasis, offering an option for cases resistant to other therapies.

Cannabidiol (CBD) Acts as an Antioxidant on Gardnerella vaginalis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

CBD shows antibacterial and antibiofilm activity against Gardnerella vaginalis, suggesting its potential as a novel treatment for bacterial vaginosis, particularly in biofilm-related infections.

What was Studied?

This study focused on the antibacterial and antibiofilm effects of cannabidiol (CBD) on Gardnerella vaginalis, a bacterium commonly associated with bacterial vaginosis (BV). CBD, a non-psychoactive compound derived from cannabis, is known for its antioxidant, anti-inflammatory, and antibacterial properties. The researchers explored how CBD affects G. vaginalis, particularly its metabolic activity, survivability, and biofilm formation. The study examined CBD’s potential as a therapeutic agent to combat BV, given the challenges of high recurrence and resistance to conventional treatments.

Who was Studied?

The clinical isolates of Gardnerella vaginalis were the primary subjects of this study. These bacterial strains were obtained from patients with BV and were treated with CBD to assess its impact on their viability, metabolic activity, and biofilm production. The research team studied the bacterium’s response to CBD in vitro, particularly to changes in membrane potential, reactive oxygen species (ROS) levels, and the bacterial ability to form and maintain biofilms.

What were the most Important Findings?

The study demonstrated that CBD has potent antibacterial effects on G. vaginalis, with a minimum inhibitory concentration (MIC) of 2.5 µg/mL. CBD was found to reduce metabolic activity and cause a significant decrease in bacterial survivability. It achieved this by inducing rapid membrane hyperpolarization and cytoplasmic ATP leakage without increasing membrane permeability, which suggests a non-lytic mode of action. CBD exhibited antioxidant properties by reducing intracellular ROS levels in a dose-dependent manner. Notably, CBD not only inhibited the formation of new biofilms but also disrupted preformed mature biofilms of G. vaginalis. The metabolic activity and biomass of these biofilms were significantly reduced following CBD treatment, with up to 90% reduction at higher concentrations. Notably, the free radical scavenger α-tocopherol neutralized CBD's antibacterial effect, highlighting the role of reactive oxygen species in CBD’s action.

What are the Implications of this Study?

This research suggests that CBD could be a novel therapeutic agent for treating BV caused by Gardnerella vaginalis, especially given its dual action on both live bacteria and biofilms. Biofilms are notoriously difficult to treat with conventional antibiotics, and their disruption is a critical step in reducing the recurrence rates of BV. CBD's ability to both prevent biofilm formation and reduce the viability of mature biofilms makes it a promising candidate for overcoming current treatment limitations, such as antibiotic resistance and the persistence of infection due to biofilm protection. Furthermore, the antioxidant properties of CBD may offer additional benefits in managing oxidative stress in the vaginal environment. These findings warrant further exploration into the potential clinical applications of CBD for BV, particularly as a safer and more effective alternative to existing treatments that may disrupt the vaginal microbiota.

Clinicians’ use of Intravaginal Boric Acid Maintenance Therapy for Recurrent Vulvovaginal Candidiasis and Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study evaluates the use of intravaginal boric acid for recurrent vulvovaginal candidiasis and bacterial vaginosis, highlighting its long-term tolerability, high patient satisfaction, and rare side effects.

What was Studied?

This study examined the use of intravaginal boric acid (BA) maintenance therapy in women with recurrent vulvovaginal candidiasis (rVVC) and recurrent bacterial vaginosis (rBV). The researchers performed a retrospective chart review to evaluate clinicians' approaches to prescribing BA for these conditions, focusing on dosage, duration of use, patient satisfaction, and side effects. The study aimed to assess the effectiveness, tolerability, and satisfaction of long-term BA therapy in real-world clinical settings.

Who was Studied?

The study reviewed the medical records of 78 patients from a Johns Hopkins University-affiliated outpatient gynecology clinic. These patients were prescribed intravaginal BA for either rVVC, rBV, or both conditions. The patients were selected based on specific criteria, including multiple visits where BA usage was documented, and those who were prescribed a long-term BA regimen (more than a month). Patients were excluded if there was insufficient documentation regarding the initiation or duration of BA use.

What were the Most Important Findings?

The study revealed that maintenance therapy with intravaginal boric acid was commonly prescribed for rVVC and rBV, with an average duration of use estimated at 13.3 months. A significant portion of patients (37.2%) used BA for a year or more, with some patients continuing therapy for more than three years. The treatment regimen typically included a 7-14 day induction phase with BA, followed by a maintenance phase where patients used 300mg or 600mg of BA 2-3 times per week.

Despite the lack of long-term safety data, the study found high patient satisfaction with BA therapy (76.9%), though a small number of patients (16.7%) were dissatisfied, typically due to continued or worsening symptoms. The study also indicated that patients with rVVC were more likely to receive BA as part of an antifungal induction regimen, while patients with rBV were often prescribed antibiotics in addition to BA. Side effects were rare, with a few patients reporting vaginal irritation or leaking, but these effects were generally manageable.

What are the Implications of this Study?

This study provides real-world evidence supporting the use of intravaginal boric acid as a long-term treatment for recurrent vulvovaginal candidiasis and bacterial vaginosis. Despite the absence of large-scale prospective studies, the findings suggest that BA is well-tolerated over extended periods and that it may be an effective option for women with azole-resistant infections. This study's insights into patient satisfaction, side effects, and clinical practice could inform future treatment guidelines and clinical trials for rVVC and rBV. However, more robust, prospective studies are needed to confirm the efficacy and long-term safety of BA maintenance therapy and to compare it with other available treatments.

Combatting antibiotic resistance in Gardnerella vaginalis: A comparative in silico investigation for drug target identification

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study identifies phospho-2-dehydro-3-deoxyheptonate aldolase as a drug target in Gardnerella vaginalis and highlights five FDA-approved compounds as potential treatments for bacterial vaginosis.

Who was Studied?

This study focused on combating antibiotic resistance in Gardnerella vaginalis, a bacterium frequently associated with bacterial vaginosis (BV). The researchers employed an in silico approach to identify potential drug targets and therapeutic strategies. By utilizing subtractive genomics and comparative genomics, they analyzed the G. vaginalis proteome to find unique proteins crucial for bacterial survival and virulence, which could serve as drug targets. The study further explored FDA-approved compounds using virtual screening techniques to identify potential inhibitors of these targets. Additionally, the study performed detailed protein structural modeling, docking, and ADMET profiling of the shortlisted compounds to assess their suitability for therapeutic use.

Who was Studied?

The study focused on Gardnerella vaginalis, which is often linked to bacterial vaginosis. It did not study individuals directly but rather the bacterial proteome, applying computational methods to identify druggable targets within the pathogen. The proteome of G. vaginalis was retrieved from the UniProt database, and various bioinformatics tools were used to identify potential drug targets based on their essentiality, uniqueness, and non-homology to human proteins.

What are the Most Important Findings?

The study identified phospho-2-dehydro-3-deoxyheptonate aldolase (PDA) as a promising drug target for G. vaginalis. This enzyme plays a critical role in the shikimate pathway, which is essential for producing aromatic amino acids and other metabolites. The identification of this enzyme is significant because it is non-homologous to human proteins, reducing the risk of off-target effects. The researchers found five compounds from the DrugBank database that could inhibit PDA effectively. The compounds demonstrated strong binding affinities to the target protein, suggesting their potential as effective treatments. Virtual screening results showed that these compounds have favorable pharmacokinetic profiles, including good bioavailability, and do not inhibit key enzymes responsible for drug metabolism, making them promising candidates for repurposing to treat G. vaginalis infections.

What are the Implications of the Study?

This study presents a significant step toward identifying new therapeutic strategies for treating G. vaginalis infections, particularly in the face of rising antibiotic resistance. The identification of PDA as a drug target opens up new possibilities for developing treatments that are more effective than current antibiotics, which often face resistance issues. The use of in silico methods, such as molecular docking and virtual screening, allows for the rapid identification of promising drug candidates, saving time and resources compared to traditional experimental approaches. By repurposing FDA-approved compounds, the study suggests a faster route to clinical application, potentially providing affordable treatments for bacterial vaginosis. This approach also highlights the potential of computational tools in addressing antibiotic resistance and discovering new uses for existing drugs.

Computational approach for drug discovery against Gardnerella vaginalis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study identifies DAHP synthase as a promising drug target for Gardnerella vaginalis in the treatment of bacterial vaginosis, presenting potential inhibitors with favorable pharmacokinetics.

What was studied?

This study explored a computational approach for drug discovery to identify effective treatments against Gardnerella vaginalis (G. vaginalis), the primary cause of bacterial vaginosis (BV). The researchers used a combination of subtractive proteomics, molecular docking, molecular dynamics (MD) simulations, and ADMET profiling to identify potential drug targets and screen inhibitor compounds. The target enzyme selected for further analysis was 3-deoxy-7-phosphoheptulonate synthase (DAHP synthase), which plays a vital role in the shikimate pathway, crucial for the biosynthesis of essential aromatic amino acids.

Who was Studied?

The study analyzed the proteome of G. vaginalis strain. Using computational tools, the study identified 11 potential drug targets within the bacterium, with DAHP synthase being the chosen target for subsequent inhibitor screening. This approach leverages bioinformatics to identify non-homologous bacterial proteins that do not share similarities with the human proteome, reducing the risk of potential toxicity or off-target effects.

Most Important Findings

One of the study's major findings is the identification of DAHP synthase as a critical target for drug development against G. vaginalis. This enzyme is central to the shikimate pathway, which is involved in the production of aromatic amino acids like phenylalanine, tyrosine, and tryptophan, as well as secondary metabolites such as antibiotics and toxins. Inhibiting this enzyme could disrupt essential bacterial functions, impairing the pathogen's ability to thrive in the human host.

Additionally, the study highlighted several inhibitors from the ZINC database that showed high binding affinities towards DAHP synthase, surpassing even the control ligand phosphoenolpyruvate in docking simulations. ZINC98088375, in particular, exhibited promising pharmacokinetic properties, such as high bioavailability and solubility, making it a potential candidate for oral drug formulation. The study also examined the pharmacokinetic behavior of these compounds using PBPK modeling, revealing how health conditions can affect drug absorption and systemic circulation.

Implications of this Study?

This study highlights the potential of computational drug design in overcoming the challenges of treating BV, especially in the face of antibiotic resistance and biofilm formation by G. vaginalis. The identified DAHP synthase inhibitors could lead to more effective treatments, offering an alternative to existing therapies, which have limitations such as high recurrence rates and resistance. The study's approach to selecting drug targets based on subtractive proteomics ensures that only bacterial proteins that do not overlap with human proteins are targeted, thus minimizing toxic or off-target effects.

The ADMET profiling and PBPK modeling offer insight into the safety and efficacy of the compounds, making them potential candidates for further development in clinical settings. This integrated-omics approach provides a rational framework for discovering new therapeutics for BV, highlighting the importance of personalized medicine based on individual health conditions.

Contribution of Essential Oils to the Fight against Microbial Biofilms—A Review

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explores the antimicrobial potential of essential oils (EOs) in combating microbial biofilms, highlighting their effectiveness against pathogens like S. aureus and C. albicans. EOs can serve as an alternative or adjunct to conventional antibiotics, particularly in medical device infections.

What was reviewed?

The review paper focuses on the contribution of essential oils (EOs) in combating microbial biofilms, with an emphasis on their antimicrobial properties and the potential application in medical, food, and other industrial sectors. The review explores the growing body of scientific data showing the effectiveness of essential oils against biofilms, which are known to contribute to persistent infections, particularly those associated with medical devices and chronic infections. The paper provides an in-depth analysis of several studies that highlight how EOs, through their diverse chemical compositions, possess the ability to disrupt biofilm formation by bacteria and fungi.

Who was reviewed

The review analyzed various scientific literature and research papers that evaluated the effectiveness of essential oils in combating microbial biofilms. The focus was on assessing how different essential oils have antimicrobial properties that can disrupt biofilm formation and enhance the effectiveness of traditional treatments.

What were the most important findings?

The most important finding of this review is the significant role essential oils (EOs) can play in combating microbial biofilms, which are highly resistant to conventional antibiotics. The review outlines various essential oils, such as those derived from Melaleuca alternifolia (tea tree oil), Lavandula angustifolia (lavender oil), and Cinnamomum zeylanicum (cinnamon oil), that have been proven to inhibit or eradicate biofilms. Specifically, the antimicrobial components of EOs, including terpenes and terpenoids, act by permeabilizing bacterial membranes, which makes biofilms more susceptible to antimicrobial treatments. The review emphasizes that these oils not only affect planktonic bacteria but also disrupt biofilms formed on surfaces, which is crucial in the context of medical devices like catheters and prosthetics.

In terms of microbial associations, the review focuses on EOs' efficacy against a range of pathogenic organisms, including Staphylococcus aureus, Escherichia coli, and Candida species. Biofilms formed by these organisms on medical devices are notably resistant to conventional treatments, and the use of EOs could offer a complementary or alternative approach. The synergistic effect of combining EOs with traditional antibiotics is also highlighted, as this combination increases the efficacy of treatment and reduces the chances of biofilm formation.

What are the implications of this review?

The review reveals the potential of essential oils as a safe and effective alternative to conventional antibiofilm agents, particularly for infections related to medical devices, foodborne illnesses, and oral health. The diverse chemical compositions of EOs reduce the likelihood of microbial resistance, a growing concern with traditional antibiotics. Furthermore, the combination of EOs with other antimicrobial treatments offers promising results for tackling biofilm-related infections more effectively. The study suggests that further clinical trials and the optimization of EO formulations are needed to fully harness their potential in combating biofilms, especially in medical and industrial applications. Incorporating EO-based therapies into current infection control practices could help address the persistent problem of biofilm-associated infections, which are difficult to treat with traditional antibiotics alone.

Current Treatment of Bacterial Vaginosis—Limitations and Need for Innovation

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review reveals high BV recurrence rates after metronidazole or clindamycin treatment due to microbial biofilms and potential sexual transmission. While both antibiotics show similar short-term efficacy, they differ in resistance patterns. Biofilm disruptors and partner treatment may improve outcomes, but better diagnostics and combination therapies are urgently needed.

What was Reviewed?

This comprehensive review critically examines the current limitations in bacterial vaginosis (BV) treatment, with particular focus on the high recurrence rates following standard antibiotic therapies, including both metronidazole and clindamycin. The authors analyze the microbial factors contributing to treatment failure, specifically the role of polymicrobial biofilms and antimicrobial resistance patterns in Gardnerella vaginalis and other BV-associated bacteria. The review also explores emerging evidence for sexual transmission of BV-associated microorganisms and evaluates novel therapeutic approaches targeting biofilm disruption and partner treatment strategies.

Who was Reviewed?

The review synthesizes data from multiple clinical trials and observational studies involving women with recurrent BV across diverse populations. It incorporates microbiological research on vaginal and penile microbiota, including studies demonstrating the presence of BV-associated bacteria in male sexual partners. The analysis also examines in vitro studies of biofilm formation and disruption, as well as limited clinical trials of adjunctive therapies like boric acid and probiotics.

Key Findings and Microbial Associations

The review highlights that BV represents a profound dysbiosis where protective Lactobacillus species, particularly L. crispatus, are replaced by a polymicrobial consortium including Gardnerella vaginalis, Atopobium vaginae, and various Clostridiales species. These pathogens form resilient biofilms that protect them from both metronidazole and clindamycin, the two first-line antibiotics for BV. While short-term cure rates approach 80% for both medications, recurrence rates exceed 50% within 6-12 months. The review notes important differences between the antibiotics: clindamycin appears more effective against certain biofilm-embedded pathogens like A. vaginae but may promote clindamycin-resistant anaerobic gram-negative rods, while metronidazole faces challenges with intrinsically resistant G. vaginalis clades. Both antibiotics fail to address the potential sexual transmission of BV-associated bacteria, which are detectable in male partners' genital microbiota and may contribute to reinfection.

Implications of the Review

The review underscores that current antibiotic regimens, whether using metronidazole or clindamycin, are insufficient for long-term BV control due to biofilm persistence and potential sexual transmission. Clinicians should continue following treatment guidelines but recognize these limitations when managing recurrent cases. The findings suggest several important considerations: vaginal clindamycin may be preferable for certain BV subtypes or in pregnancy, while metronidazole remains the most widely studied option. For recurrent BV, adjunctive approaches like biofilm disruptors (boric acid, DNase) or partner treatment may be worth considering, though more research is needed. The review emphasizes the need for improved diagnostics to identify BV subtypes and resistance patterns, as well as the development of combination therapies targeting both pathogens and biofilms. Public health measures promoting condom use and further research into sexual transmission dynamics could help reduce BV recurrence at the population level.

Dietary Intake of Selected Nutrients Affects Bacterial Vaginosis in Women

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

Higher dietary fat intake increases the risk of bacterial vaginosis (BV), while folate, vitamin E, and calcium may lower severe BV risk. This study suggests dietary interventions could play a role in managing BV, offering insights for clinicians on how nutrition affects vaginal microbiome balance.

What was Studied?

This study examined the relationship between dietary intake and the presence of bacterial vaginosis (BV) in women. Researchers assessed how macronutrient and micronutrient consumption, particularly fat intake and essential vitamins, influenced the risk of BV and severe BV.

Who was Studied?

The study analyzed data from 1,521 non-pregnant women, aged 15–45, who were part of a larger longitudinal study of vaginal flora in Birmingham, Alabama. The majority (86%) were African American. Participants underwent clinical assessments and completed a food frequency questionnaire.

Most Important Findings

The study found a significant association between dietary fat intake and the risk of BV. Women consuming higher amounts of total fat, saturated fat, and monounsaturated fat had an increased likelihood of BV and severe BV. Total fat intake was linked to a 50% higher risk of BV, while saturated fat and monounsaturated fat were particularly associated with severe BV.

Conversely, higher intakes of folate, vitamin E, and calcium were associated with a reduced risk of severe BV. These nutrients may support local immune function, potentially counteracting the microbial imbalance seen in BV. Energy intake had a marginal association with BV, while carbohydrate and protein intake showed no significant links. The study suggests that a high-fat diet may alter vaginal microflora, increase vaginal pH, and contribute to BV development.

Implications of the Study

Diet plays a crucial role in vaginal health, with fat consumption significantly influencing BV risk. Clinicians can reduce BV risk by advising patients to lower fat intake and increase folate, vitamin E, and calcium. Researchers should further explore how dietary fat disrupts vaginal microbiota and whether targeted nutritional changes can serve as effective prevention strategies.

Dietary Patterns and Bacterial Vaginosis: is there any association?

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

A nutrient-rich diet lowers bacterial vaginosis (BV) risk, while processed foods increase it. This study highlights the importance of dietary choices in vaginal microbiome balance, offering insights for clinicians on how diet influences BV risk.

What was Studied?

Researchers investigated how dietary acid load and adherence to the Alternative Healthy Eating Index (AHEI) affect bacterial vaginosis (BV) risk. They aimed to determine whether dietary patterns and acid-producing foods influence the vaginal microbiome and BV prevalence.

Who was Studied?

The study included 143 women diagnosed with BV and 151 healthy controls, aged 18–45, from a gynecology clinic in Tehran, Iran. Researchers assessed dietary intake using a validated food frequency questionnaire and diagnosed BV using the Amsel criteria.

Most Important Findings

A high AHEI score significantly lowered BV risk. Women in the highest AHEI tertile had a 75% lower chance of developing BV. Consuming more vegetables, nuts, legumes, and unprocessed meats further reduced BV odds. In contrast, high intakes of sugar-sweetened beverages, trans fats, and sodium increased BV risk by up to three times.

Dietary acid load, measured by potential renal acid load (PRAL) and net endogenous acid production (NEAP), showed no significant link to BV. This suggests that overall diet quality, rather than dietary acid-base balance, plays a more critical role in BV risk.

Microbiome analysis linked high AHEI adherence to a Lactobacillus-dominant vaginal environment, which protects against BV. In contrast, poor dietary choices promoted the growth of BV-associated bacteria such as Gardnerella vaginalis, Atopobium spp., and Prevotella spp..

Implications of the Study

Diet quality directly affects vaginal microbiome health. Clinicians should encourage patients to adopt a plant-based diet rich in vegetables, nuts, and legumes while limiting sugar-sweetened beverages and trans fats. Future research should explore whether dietary interventions can serve as an effective strategy for BV prevention and management.

Diversity of Vaginal Microbiota Associated with Bacterial Vaginosis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study analyzed vaginal microbiota diversity in women with BV using molecular techniques. It identified key BV-associated bacteria and emphasized the need for molecular diagnostics and microbiome-targeted therapies.

What Was Studied?

This study analyzed the diversity and composition of vaginal microbiota in women with bacterial vaginosis (BV) using molecular techniques. Researchers compared the microbial communities of BV-positive and healthy women to identify bacterial associations with BV and determine potential diagnostic markers.

Who Was Studied?

The study included 50 women diagnosed with BV and 50 healthy women from China. Researchers collected vaginal samples and analyzed bacterial diversity using PCR-denaturing gradient gel electrophoresis (DGGE), 454 pyrosequencing, and quantitative PCR (qPCR).

Key Findings and Microbial Associations

The study revealed a significant increase in bacterial diversity in BV-positive women compared to healthy controls. The dominant bacterial phyla in BV included Bacteroidetes, Actinobacteria, and Fusobacteria, whereas healthy women had microbiomes dominated by Firmicutes, particularly Lactobacillus species.

Several bacterial genera were strongly associated with BV, including Gardnerella, Atopobium, Megasphaera, Eggerthella, Aerococcus, Leptotrichia/Sneathia, Prevotella, and Papillibacter. These bacteria may serve as potential molecular markers for BV diagnosis. While no single bacterium could be used as a definitive BV indicator, the presence of multiple BV-associated genera strongly correlated with the condition.

The study also highlighted the limitations of traditional culture-based methods in capturing the complexity of BV microbiota. High-throughput sequencing provided a more detailed picture of the vaginal bacterial ecosystem, revealing low-abundance taxa that previous studies had overlooked.

Implications of the Study

These findings reinforce that BV is a polymicrobial condition rather than an infection caused by a single pathogen. The increased bacterial diversity in BV underscores the need for diagnostic approaches that consider microbial community shifts rather than relying solely on individual bacterial markers.

The study supports the development of molecular-based diagnostic tools targeting BV-associated bacteria for more accurate detection. Additionally, understanding microbial interactions in BV could lead to microbiome-targeted therapies rather than traditional broad-spectrum antibiotic treatments, which often fail to prevent recurrence.

Dynamics of Vaginal Microbiota Before, During and After Episodes of Bacterial Vaginosis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study analyzed daily changes in vaginal microbiota before, during, and after BV episodes. Findings highlight microbial instability, early dysbiosis markers, and the need for microbiome-focused BV treatments.

What Was Studied?

This study investigated the daily fluctuations of vaginal microbiota before, during, and after bacterial vaginosis (BV) episodes. Researchers aimed to understand how microbial communities shift over time, particularly in symptomatic and asymptomatic BV cases. They sought to identify microbial patterns that could predict BV onset, persistence, and resolution.

Who Was Studied?

The study followed 25 women over 10 weeks, including 15 with symptomatic BV, six with asymptomatic BV, and four without BV. Participants self-collected vaginal samples daily, which researchers analyzed using 16S rRNA sequencing. The study also recorded vaginal symptoms, menstrual cycle phases, and sexual behaviors to assess how these factors influenced microbiota changes.

Key Findings and Microbial Associations

The study revealed that vaginal microbiota exhibit significant daily fluctuations, especially in women with BV. Before BV episodes, microbial diversity increased, with a rise in anaerobes such as Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., and Megasphaera spp.. Women with symptomatic BV showed a notable decline in Lactobacillus crispatus, while Lactobacillus iners persisted at low levels, suggesting its role in transitional microbial states.

During symptomatic BV, bacterial communities became more heterogeneous, with a dominance of strict anaerobes and a rise in vaginal pH. Some women experienced rapid shifts in microbiota, while others maintained a stable but Lactobacillus-depleted state. Interestingly, after antibiotic treatment, most women’s microbiota temporarily shifted toward a Lactobacillus-dominated state, particularly L. iners. However, within 2-4 weeks, many reverted to their pre-treatment microbial profiles, indicating a high risk of BV recurrence.

Implications of the Study

This study highlights the dynamic nature of vaginal microbiota and reinforces the need for improved BV management strategies. Since BV-associated bacteria can persist and re-emerge despite treatment, future therapies should focus on long-term microbial stabilization rather than short-term symptom relief. The findings suggest that monitoring microbiota changes over time could help predict BV recurrence and guide personalized treatment strategies.

Additionally, the study highlights the limitations of current diagnostic methods, such as Amsel’s criteria and Nugent scoring, which do not capture daily microbial shifts. Incorporating molecular techniques into routine diagnostics could improve BV detection and allow for earlier intervention by identifying microbial imbalances before symptoms develop. This approach could also help tailor treatment strategies, reduce recurrence rates, and support long-term vaginal microbiome stability.

Efficacy and Safety of Different Drugs for the Treatment of Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This meta-analysis compared BV treatments, identifying ornidazole as the most effective oral drug and sucrose/probiotics as top non-antibiotic options. Restoring Lactobacillus dominance is key, with vaginal probiotics and sucrose showing high cure rates.

What was reviewed?

This systematic review and network meta-analysis examined the efficacy and safety of multiple treatments for bacterial vaginosis (BV), a common vaginal dysbiosis characterized by the overgrowth of anaerobic bacteria and a decline in protective Lactobacillus species. The study compared antibiotics (metronidazole, clindamycin, tinidazole, secnidazole, ornidazole, ofloxacin) with non-antibiotic therapies (sucrose, probiotics) to determine the most effective and safest options for clinical use. The analysis incorporated both direct and indirect comparisons across studies, providing a comprehensive ranking of treatments based on cure rates and adverse effects.

Who was reviewed?

The meta-analysis included 42 randomized controlled trials (RCTs) and observational studies, encompassing patients diagnosed with BV. The studies were sourced from PubMed and Embase, ensuring a broad evaluation of existing evidence. Participants were treated with either oral or vaginal formulations of the studied drugs, allowing subgroup analyses to assess differences in administration routes.

Most Important Findings

The review highlighted that BV, characterized by a shift from Lactobacillus-dominant vaginal microbiota to an overgrowth of anaerobic bacteria like Gardnerella vaginalisAtopobium vaginae, and Bacteroides spp., responds differently to treatments. Ornidazole emerged as highly effective, with a clinical cure rate superior to clindamycin and secnidazole. Sucrose and probiotics also showed promise, with sucrose ranking highest in clinical cure probability and probiotics demonstrating fewer adverse effects compared to metronidazole. Notably, metronidazole and secnidazole had higher adverse reaction rates than placebo, while probiotics and sucrose were safer alternatives. The study underscored the importance of restoring Lactobacillus dominance to rebalance vaginal microbiota, as sucrose promotes Lactobacillus growth by lowering vaginal pH, and probiotics directly reintroduce beneficial bacteria.

Implications of the Review

The findings suggest that ornidazole could be a superior alternative to traditional BV treatments like metronidazole, particularly for oral administration. Non-antibiotic options like sucrose and probiotics offer effective and safer alternatives, aligning with microbiome-focused therapies. Clinicians should consider these options, especially for patients with recurrent BV or those prone to adverse effects from antibiotics. The study also calls for more high-quality trials to validate these results and explore long-term outcomes.

Efficacy of Vitamin C Vaginal Tablets in Preventing Recurrence of Bacterial Vaginosis: A Randomized Controlled Trial

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study demonstrates that 250 mg vitamin C vaginal tablets reduce the recurrence of bacterial vaginosis, offering an effective and safe prophylactic treatment.

What was Studied?

This study evaluated the efficacy of vitamin C vaginal tablets as a prophylactic treatment for recurrent bacterial vaginosis (rBV). The research was a randomized, double-blind, placebo-controlled clinical trial involving 142 women who had been successfully treated for a recent episode of BV using either metronidazole or clindamycin. These women were randomly assigned to receive either vitamin C or a placebo for six months, to prevent the recurrence of BV.

Who was Studied?

The study involved 142 women aged between 18 and 50 years who had a history of recurrent episodes of bacterial vaginosis, defined as at least two acute episodes in the past year. After successful treatment of a BV episode, participants were randomized into two groups: one receiving 250 mg of vitamin C vaginal tablets and the other a placebo. The study participants were monitored for recurrence of BV for six months.

What were the Most Important Findings?

The study demonstrated that the use of vitamin C vaginal tablets significantly reduced the recurrence rate of BV in women compared to the placebo group. After three months of treatment, the recurrence rate in the vitamin C group was 6.8%, whereas the placebo group had a recurrence rate of 14.7%. After six months, the recurrence rate was 16.2% in the vitamin C group, compared to 32.4% in the placebo group, which was statistically significant (P = 0.024). The vitamin C treatment also showed a significant pH-lowering effect, which is thought to contribute to the inhibition of pathogen overgrowth. Additionally, the Kaplan-Meier survival analysis indicated that the vitamin C group had a significantly lower probability of experiencing a BV relapse (P = 0.029). The treatment was well tolerated, with minimal adverse events, mostly local reactions like itching and burning.

What are the Implications of this Study?

The study highlights the potential of vitamin C vaginal tablets as an effective prophylactic treatment for recurrent bacterial vaginosis. By lowering vaginal pH, vitamin C helps re-establish a more acidic environment, preventing the overgrowth of harmful anaerobic bacteria, such as Gardnerella vaginalis. This finding is significant for women with recurrent BV, particularly those with antibiotic-resistant strains or those who experience frequent recurrences after antibiotic therapy. Given the safety profile and efficacy of vitamin C, it presents a promising alternative or complementary approach to current BV treatments, offering a simple and non-invasive way to manage this chronic condition.

Fighting polymicrobial biofilms in bacterial vaginosis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review examines how polymicrobial biofilms contribute to bacterial vaginosis (BV) treatment failure and explores alternative strategies for improved therapy.

What was studied?

The study focused on the role of polymicrobial biofilms in bacterial vaginosis (BV) and their impact on treatment outcomes. The review highlights the complexity of BV, which is often driven by polymicrobial biofilms consisting of a variety of microorganisms, including Gardnerella vaginalis, Fannyhessea vaginae, Prevotella bivia, and other anaerobic bacteria. The study also explores how these biofilms contribute to BV's persistence and resistance to treatment.

Who was studied?

The review covers various studies that investigated the microbial composition of BV and its associated biofilms, focusing on the microbial species that are involved in these biofilm structures. It includes research on the role of Gardnerella vaginalis and other BV-associated pathogens in forming biofilms that contribute to the persistence of BV in the vaginal environment.

What were the most important findings?

The review underscores that the formation of polymicrobial biofilms is central to BV's persistence and recurrence. These biofilms provide a protective matrix that shields bacteria from the effects of antimicrobial agents like metronidazole and clindamycin. The study highlights that Gardnerella vaginalis and Fannyhessea vaginae are the dominant species within these biofilms, facilitating the growth of other BV-associated bacteria like Prevotella bivia. This synergistic interaction among bacteria enhances their resistance to treatment and increases the likelihood of BV recurrence.

The study also points out that biofilms are more difficult to treat than planktonic bacteria due to their reduced susceptibility to antibiotics, making treatment regimens less effective. Antibiotics can reduce the bacterial load, but biofilms often persist, leading to relapse.

This review also explores promising alternative strategies, such as probiotics, prebiotics, and phage endolysins. These approaches aim to restore the natural vaginal microbiota by promoting the growth of beneficial Lactobacillus species and reducing the pathogenic bacteria that drive BV.

What are the implications of this study?

The study highlights the critical role of polymicrobial biofilms in BV persistence and recurrence. It suggests that addressing the biofilm structure should be a key focus in developing more effective BV treatments. Traditional antibiotic therapies are insufficient in eliminating BV due to biofilm formation, which provides a physical barrier to treatment and contributes to the high rates of recurrence. The review points to the potential for alternative treatments, like probiotics and phage therapy, to improve patient outcomes by targeting these biofilms and restoring a balanced vaginal microbiome. However, the study stresses the need for further research to validate these therapies and establish their long-term effectiveness.

By understanding the polymicrobial nature of BV and its role in antimicrobial resistance, clinicians can better navigate the challenges of recurrent infections. Exploring non-antibiotic treatments and biofilm-targeting therapies offers a promising direction for more sustainable BV management, providing hope for patients who suffer from recurrent episodes that are resistant to conventional therapies.

Guidelines for the treatment of bacterial vaginosis: focus on tinidazole

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review evaluates the use of tinidazole in treating bacterial vaginosis, highlighting its efficacy, favorable pharmacokinetics, and reduced side effects compared to metronidazole, especially for recurrent cases.

What was reviewed?

The study reviewed the guidelines for treating bacterial vaginosis (BV), with a specific focus on using tinidazole. It assessed the effectiveness of tinidazole as an alternative treatment for BV, compared its pharmacokinetic profile with that of metronidazole, and discussed its potential for treating refractory and recurrent BV cases. The review also included data on various treatment regimens, including single and multiple doses, and compared outcomes with other treatment options like clindamycin.

Who was reviewed?

The review focused on the clinical guidelines and available studies regarding the treatment of bacterial vaginosis, specifically examining the role of tinidazole in BV management. It looked at various clinical trials, case studies, and clinical experiences from multiple populations, highlighting the use of tinidazole in both acute and recurrent BV cases.

What were the most important findings?

The review highlighted that tinidazole, a second-generation nitroimidazole antiprotozoal agent, is effective in treating BV, especially in cases that are refractory to metronidazole or in individuals who experience frequent relapses. Tinidazole has a favorable pharmacokinetic profile, including a longer half-life, which allows for reduced dosing frequency compared to metronidazole. The review also noted that tinidazole had fewer gastrointestinal side effects, such as nausea and metallic taste, compared to metronidazole, making it a more tolerable alternative. Tinidazole was effective in a variety of dosing regimens, including a single dose or multiple daily doses.

The study noted that both tinidazole and metronidazole effectively target the pathogens commonly associated with BV, although minimal comparative data exist to definitively establish the superiority of one over the other. Additionally, the review emphasized the importance of considering tinidazole for women with recurrent or difficult-to-treat BV, where metronidazole and other first-line treatments fail.

What are the implications of this study?

The findings emphasize the value of tinidazole as an effective alternative for treating bacterial vaginosis, particularly in cases that are resistant to or relapse after standard metronidazole treatment. Its improved pharmacokinetics and reduced side effects enhance its potential for long-term use in managing BV, offering a viable option for patients with recurrent infections. Given the persistent nature of BV and its association with complications such as preterm birth and pelvic inflammatory disease, the use of tinidazole could provide significant clinical benefits, particularly for those who do not respond to conventional therapies. The review also emphasizes the need for further research into head-to-head comparisons of tinidazole and metronidazole to more conclusively define their relative roles in BV treatment.

Identifying Bacterial Vaginosis-Associated Bacteria

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study identifies three key bacterial vaginosis-associated species using DNA sequencing and phylogenetics. By classifying BVAB-1, BVAB-2, and BVAB-3, the findings advance understanding of BV’s microbial landscape and highlight new opportunities for targeted diagnostics and treatment.

What Was Studied?

This study analyzed bacterial vaginosis-associated bacteria (BVAB) at the species level using DNA sequencing and phylogenetic analysis. Researchers sought to identify the exact species of BVAB-1, BVAB-2, and BVAB-3, which were previously unknown and only classified by molecular signatures. By applying metagenomic sequencing and comparative analysis, the study aimed to provide clarity on the taxonomy of these bacteria and their role in bacterial vaginosis (BV) pathogenesis.

Who Was Studied?

The study analyzed bacterial DNA extracted from vaginal specimens of women diagnosed with BV. Using genomic data from previous metagenomic studies, the researchers compared BVAB sequences to known bacterial genomes, identifying their closest relatives and evolutionary relationships.

Key Findings and Microbial Associations

The study successfully identified the species of three previously unclassified BV-associated bacteria. BVAB-1 was found to be Clostridiales genomosp. BVAB-1 (later renamed Candidatus Lachnocurva vaginae), BVAB-2 was classified as Oscillospiraceae bacterium strain CHIC02, and BVAB-3 was identified as Mageeibacillus indolicus. These species, previously unknown, are strictly anaerobic and uncultivable in standard laboratory conditions. Their identification provides a more detailed understanding of the microbial shifts in BV and offers new insights into their role in vaginal dysbiosis.

The findings reinforce the idea that BV is not caused by a single pathogen but by a complex shift in the vaginal microbiome. The presence of BVAB-1, BVAB-2, and BVAB-3 in women with BV suggests they may contribute to the condition's persistence and recurrence. By using metagenomic sequencing and phylogenetic analysis, the study clarifies the taxonomy of these bacteria and their evolutionary relationships. This species-level identification allows for improved diagnostics and targeted research into BV-associated microbial interactions.

Implications of the Study

Identifying the specific species associated with BV significantly improves diagnostic and treatment strategies. Clinicians currently use broad-spectrum antibiotics to treat BV, but discovering these species enables the development of more targeted therapies. This study also emphasizes the need for continued metagenomic research, as many BV-associated bacteria are difficult to culture and study in traditional lab settings. With more precise species identification, researchers can better understand how these bacteria interact with the vaginal microbiome and contribute to BV recurrence.

Impact of oral metronidazole treatment on the vaginal microbiota and correlates of treatment failure

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study assesses the impact of metronidazole on the vaginal microbiota, revealing that high G. vaginalis and pathobionts correlate with treatment failure, suggesting a need for biofilm-disrupting therapies in some BV patients.

What was studied?

The study examined the effects of a standard 7-day oral metronidazole treatment on the vaginal microbiota of women diagnosed with bacterial vaginosis (BV) and/or Trichomonas vaginalis. The research aimed to evaluate the impact of the treatment on microbiota changes and identify correlates of treatment failure.

Who was studied?

The study involved 68 HIV-negative, nonpregnant women aged 18-45 years, all diagnosed with BV and/or T. vaginalis, although thirteen women were excluded afterwards. The participants were primarily from Rwanda and included women at high risk for BV, such as those with multiple sexual partners or previous treatments for BV or sexually transmitted infections.

What were the most important findings?

The results revealed a modest reduction in BV-associated anaerobes following metronidazole treatment, with only 16.4% of women showing a decrease of more than 50% in bacterial concentration. The treatment increased lactobacilli, particularly Lactobacillus iners, but did not significantly alter the concentrations of pathobionts, such as Gardnerella vaginalis. Treatment failure was notably more common in women who had a higher pretreatment concentration of G. vaginalis or pathobionts. The presence of biofilms in women with high G. vaginalis abundance may contribute to the suboptimal cure rates, which aligns with the hypothesis that biofilm formation protects these pathogens from metronidazole's effects.

What are the implications of this study?

This study provides valuable insight into why metronidazole treatment for BV often results in high recurrence rates. The findings suggest that metronidazole alone may not be sufficient for women with high G. vaginalis abundance or high pathobiont concentrations. These women may benefit from additional treatments targeting biofilm disruption or specific pathogens, which could help improve the effectiveness of BV therapy and reduce recurrence. The study underscores the complexity of vaginal dysbiosis and the importance of considering microbial composition, including the role of lactobacilli and pathobionts, when determining the most effective treatment strategy for BV.

Improved cure of bacterial vaginosis with single dose of tinidazole (2g), Lactobacillus rhamnosus GR-1, and Lactobacillus reuteri RC-14

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

  • Tinidazole
    Tinidazole

    Tinidazole is a nitroimidazole antimicrobial that selectively targets anaerobic bacteria and protozoa, reshaping the gut ecosystem by depleting pathogenic anaerobes while preserving commensals. Clinically validated for giardiasis, bacterial vaginosis, and colorectal surgery prophylaxis. Its DNA-disrupting and biofilm-penetrating actions reduce inflammatory triggers and create niches for healthy microbiota to rebound.

This study shows that oral probiotics significantly boost cure rates of bacterial vaginosis when used with tinidazole, restoring a healthy vaginal microbiome.

What Was Studied?

This randomized, double-blind, placebo-controlled clinical trial investigated whether the combination of a single 2g dose of tinidazole and daily oral probiotics (Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14) improved bacterial vaginosis (BV) cure rates in women, compared to tinidazole alone. BV, a condition characterized by dysbiosis in the vaginal microbiome and reduction of protective Lactobacillus species, has shown poor long-term response to antibiotic treatment alone. The study aimed to determine if probiotic supplementation could enhance the therapeutic efficacy of tinidazole by restoring a more favorable microbial balance.

Who Was Studied?

Sixty-four Brazilian women diagnosed with BV based on Amsel’s criteria and Nugent scoring participated in the study. The participants were randomly assigned to either a placebo group or a probiotic group, both of which received the same tinidazole dose. The probiotic group also received daily capsules containing L. rhamnosus GR-1 and L. reuteri RC-14 for four weeks. Women with other vaginal infections, recent antibiotic use, or immunosuppression were excluded. The trial assessed both subjective symptoms and microbiological cure at the end of the treatment period.

What Were the Most Important Findings?

At the end of the four-week treatment, the probiotic group exhibited a significantly higher cure rate compared to the placebo group as measured by Amsel’s criteria and Nugent score. Women in the probiotic group were assessed with “normal” vaginal microbiota based on Gram stain, compared to the placebo group. Importantly, the study also noted reduced BV-associated microbial morphotypes (Gram-variable rods, curved anaerobes) and a statistically significant improvement in key clinical indicators, including pH, discharge, and odor in the probiotic group. While both groups used tinidazole, the probiotics played a key role in enhancing microbiota restoration. Notably, the probiotic strains used in the study are known for producing biosurfactants, bacteriocins, and signaling molecules that can disrupt pathogenic biofilms, particularly those formed by Gardnerella vaginalis. This mechanism may explain their strong microbiome-modulating effect.

What Are the Greatest Implications of This Study?

This study provides robust clinical evidence supporting the adjunctive use of probiotics with antibiotics to treat BV and improve microbiota restoration. By demonstrating that oral administration of specific Lactobacillus strains significantly improves cure rates and promotes a return to healthy vaginal flora, the study bridges microbiome science with practical gynecological care. Clinicians managing recurrent or treatment-resistant BV can consider integrating targeted probiotic strains to reduce recurrence and enhance long-term remission. Additionally, the study underscores the need for strain-specific probiotic selection, given the inconsistent outcomes with nonspecific lactobacilli. The use of probiotics also holds promise in preserving drug efficacy and reducing the need for prolonged antibiotic exposure, which aligns with antimicrobial stewardship principles and microbiome health preservation.

Lactobacillus crispatus inhibits growth of Gardnerella vaginalis and Neisseria gonorrhoeae on a porcine vaginal mucosa model.

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

The study shows that Lactobacillus crispatus inhibits the growth of Gardnerella vaginalis and Neisseria gonorrhoeae by lowering pH and producing lactic acid, offering insights into microbiome-targeted interventions for bacterial vaginosis and sexually transmitted infections.

What was Studied?

The study investigated the effects of Lactobacillus crispatus on the growth of Gardnerella vaginalis and Neisseria gonorrhoeae using a porcine vaginal mucosa (PVM) model. It aimed to explore how Lactobacillus crispatus influences the growth of these pathogens and whether it could help prevent or inhibit infection through mechanisms such as the production of lactic acid and pH reduction.

Who was Studied?

The study focused on human clinical isolates of Lactobacillus crispatus, Gardnerella vaginalis, and Neisseria gonorrhoeae. The researchers inoculated these isolates into the ex vivo PVM to observe their colonization, biofilm formation, and interactions.

What were the Most Important Findings?

The study revealed that Lactobacillus crispatus significantly inhibited the growth of both Gardnerella vaginalis and Neisseria gonorrhoeae on the porcine vaginal mucosa model. This inhibition occurred primarily due to the lactic acid production by L. crispatus, which lowered the vaginal pH to levels hostile to these pathogens. The results showed that both G. vaginalis and N. gonorrhoeae grew and formed biofilms at clinically relevant densities on PVM. In particular, the biofilm formation by G. vaginalis and N. gonorrhoeae was evident, and the presence of L. crispatus hindered this process. The production of lactic acid by L. crispatus was crucial for reducing the pH below 5.5, which subsequently inhibited pathogen growth. Conditioned media (CM) from L. crispatus cultures inhibited the growth of N. gonorrhoeae, even when the pH was adjusted to levels conducive for its growth.

What are the Implications of this Study?

The study demonstrates that Lactobacillus crispatus, a key member of the vaginal microbiota, plays a significant protective role against the colonization of harmful pathogens like Gardnerella vaginalis and Neisseria gonorrhoeae. It exerts direct antimicrobial effects and modulates vaginal pH through lactic acid production. By lowering pH, L. crispatus shows potential as both a therapeutic agent and a preventive measure against bacterial vaginosis and sexually transmitted infections, including gonorrhea. This finding supports the importance of maintaining a healthy vaginal microbiota dominated by Lactobacillus species to reduce susceptibility to infections. The PVM model serves as a valuable tool for studying the complex interactions between vaginal microbiota and pathogens, offering insights into the development of targeted microbiome-based interventions.

Metabolic Signatures of Bacterial Vaginosis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study revealed how bacterial vaginosis alters vaginal metabolism, linking specific microbial shifts with distinctive metabolite profiles. It showed that BV-associated bacteria drive metabolic changes that increase vaginal pH, disrupt epithelial integrity, and produce characteristic symptoms, offering new diagnostic markers and therapeutic targets.

What was studied?

The study investigated the metabolic signatures associated with bacterial vaginosis (BV) by integrating global metabolomic profiling with microbiome analysis. Specifically, the researchers aimed to identify metabolomic patterns in vaginal fluid linked to the presence and concentration of specific vaginal bacteria, particularly BV-associated bacteria. They used mass spectrometry-based metabolomics and combined it with broad-range 16S rRNA gene sequencing and quantitative PCR (qPCR) to correlate metabolic changes with microbial community composition and clinical diagnostic criteria for BV.

Who was studied?

The study involved two cohorts of reproductive-age women. In the primary cohort, the researchers analyzed cervicovaginal lavage (CVL) samples from 40 women with BV and 20 women without BV, classified using both Amsel criteria and Nugent scoring. They validated their findings in a second cohort of 40 women with BV and 20 women without BV, ensuring reproducibility of metabolite associations across two independent datasets.

What were the most important findings?

The study demonstrated that BV is marked by dramatic shifts in vaginal metabolite profiles, reflecting the transition from a Lactobacillus-dominant microbiome to a polymicrobial anaerobic community. Researchers identified 279 metabolites, of which 62% differed significantly between women with and without BV. Women with BV exhibited lower concentrations of intact amino acids, dipeptides, and sugars, while showing elevated levels of amino acid catabolites, polyamines (putrescine and cadaverine), and short-chain fatty acids like succinate. These metabolic changes reflected enhanced amino acid catabolism and decreased carbohydrate metabolism, indicating fundamental shifts in microbial metabolism.

Major microbial associations (MMA) included higher abundance of BV-associated bacteria such as Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., Megasphaera spp., and BV-associated bacteria types 1-3 (BVAB1-3), which correlated positively with metabolites like putrescine, cadaverine, and succinate. Conversely, protective lactobacilli (Lactobacillus crispatus and L. jensenii) correlated with intact amino acids, sugars, lactate, and antioxidants like glutathione, reflecting their role in maintaining vaginal health.

Importantly, the study also linked specific metabolites to individual Amsel diagnostic criteria. For example, cadaverine and N-acetylputrescine correlated with elevated vaginal pH and amine odor; deoxycarnitine and pipecolate correlated with the presence of clue cells. These findings suggest that metabolite profiles, more than microbiome composition alone, drive clinical manifestations of BV.

What are the implications of this study?

This study provides compelling evidence that BV is a metabolically distinct condition, not solely defined by microbial composition but by functional metabolic activity of the altered microbiome. The researchers demonstrated that BV-associated bacteria actively reshape the vaginal metabolic environment by depleting amino acids and sugars while increasing production of metabolites that elevate vaginal pH, promote epithelial disruption, and produce characteristic BV symptoms. These metabolic signatures offer potential biomarkers for improving BV diagnostics beyond traditional clinical criteria, which often fail to capture asymptomatic or intermediate cases. This work highlights avenues for targeted therapies that address not only microbial imbalance but also metabolic disruptions, such as treatments designed to restore amino acid levels, reduce polyamine production, or inhibit key metabolic pathways associated with BV pathology.

NMR Metabolomics of Symbioses Between Bacterial Vaginosis Associated Bacteria

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study highlights metabolic interactions between Gardnerella vaginalis and Prevotella bivia, revealing their role in bacterial vaginosis and preterm birth risk. Using NMR metabolomics, it identifies acetate as a key metabolite linked to vaginal dysbiosis, offering insights for improving BV diagnostics and potential microbiome-targeted interventions.

What was studied?

This study investigated the metabolic interactions and symbiotic relationships between bacterial vaginosis (BV)-associated bacteria, particularly Gardnerella vaginalis and Prevotella bivia, using NMR metabolomics. It aimed to characterize their metabolic strategies, assess how co-culture influences metabolite production, and explore implications for vaginal microbiome composition and related health outcomes​.

Who was studied?

The study focused on a panel of BV-associated bacterial isolates, including G. vaginalis (multiple strains), P. bivia, Atopobium vaginae, Mobiluncus curtisii, and Peptostreptococcus anaerobius, along with a selection of Lactobacillus species, which are commonly associated with vaginal health​.

Most Important Findings

The study revealed that P. bivia and G. vaginalis exhibit a mutualistic metabolic relationship, with G. vaginalis supplying asparagine and P. bivia providing uracil. This metabolic exchange influences their ability to thrive in the vaginal environment, potentially promoting BV. The metabolic diversity within G. vaginalis was highlighted, distinguishing strains using either the bifid shunt (BS) pathway or mixed acid fermentation (MAF). Notably, MAF strains produced acetate, formate, and ethanol, contributing to alterations in the vaginal chemical environment. The co-culture of P. bivia with MAF G. vaginalis strains increased acetate production, a hallmark metabolite associated with BV and spontaneous preterm birth (sPTB)​.

Additionally, the study confirmed that L. iners cannot produce acetate, meaning that acetate detected in L. iners-dominated microbiomes originates from other BV-associated bacteria like G. vaginalis. This suggests that acetate levels may serve as a microbial marker for vaginal dysbiosis and potential inflammatory conditions. The findings reinforce that lactate production by Lactobacillus species, particularly L. acidophilus, plays a protective role by acidifying the vaginal environment, whereas acetate and succinate contribute to BV-related dysbiosis​.

Implications of this study

This study provides insights into how bacterial interactions shape the vaginal microbiome and contribute to BV and sPTB. The findings suggest that variations in G. vaginalis metabolism influence the severity and persistence of BV, affecting vaginal pH and inflammatory responses. The identification of key metabolic interactions could refine prediction models for BV and sPTB by incorporating metabolite-based biomarkers like acetate, aspartate, and lactate. Clinicians could leverage this information to develop targeted interventions, such as probiotic therapies, to restore a lactate-dominant vaginal microbiome and reduce the risk of adverse reproductive outcomes​.

Pathogenesis of Bacterial Vaginosis: Discussion of Current Hypotheses

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explores the competing hypotheses behind bacterial vaginosis (BV), emphasizing microbial shifts, racial disparities, and sexual transmission. Key findings suggest BV may be sexually transmitted and driven by Gardnerella vaginalis biofilms, necessitating novel treatment strategies beyond standard antibiotics. Understanding BV pathogenesis is essential for improving prevention and care.

What Was Reviewed?

This review examines the competing hypotheses on bacterial vaginosis (BV) pathogenesis. BV is a prevalent vaginal condition linked to adverse health outcomes such as preterm labor, pelvic inflammatory disease, and increased susceptibility to sexually transmitted infections (STIs). Despite its clinical significance, BV’s underlying cause remains controversial. Researchers debate whether BV results from a single pathogen, microbial imbalance, sexual transmission, genetics, or hygiene practices. The review evaluates each hypothesis and identifies research gaps that must be addressed to improve prevention and treatment strategies.

Who Was Reviewed?

The review synthesizes findings from multiple studies, including epidemiological research, microbiome sequencing, and clinical trials. It assesses the variations in vaginal microbial communities and risk factors across different populations. It includes data comparing African American and European ancestry women, evaluating whether BV prevalence differences are due to genetic predisposition, sexual networks, or socioeconomic factors. In addition, it also examines studies on sexual behavior, hygiene, and microbial interactions to understand BV development and persistence.

What Were the Most Important Findings?

BV occurs when Lactobacillus-dominant vaginal microbiota shifts to an overgrowth of anaerobes like Gardnerella vaginalis, Atopobium, and Prevotella. A major debate centers on whether this microbial shift is the result of an exogenous infection or an imbalance within the vaginal ecosystem. One hypothesis suggests BV is a sexually transmitted condition, as studies consistently link new sexual partners and unprotected intercourse to increased BV risk. Condom use has been shown to reduce recurrence, and BV-associated bacteria, including G. vaginalis, have been detected in both male and female partners. However, conflicting data exist, particularly in cases of BV occurring in virginal individuals.

Another hypothesis proposes that G. vaginalis acts as a keystone pathogen, initiating biofilm formation that facilitates the overgrowth of anaerobic bacteria. These biofilms make BV highly resistant to antibiotic treatment, leading to frequent recurrence. This aligns with studies showing that biofilm persistence, rather than reinfection, is responsible for many recurrent BV cases.

Racial disparities in BV prevalence have also been noted, with African American women more likely to have diverse anaerobic vaginal microbiota compared to women of European ancestry. Some studies suggest genetic or environmental factors, such as diet and socioeconomic status, play a role. However, the evidence remains inconclusive.

What Are the Implications of This Review?

A deeper understanding of BV pathogenesis is essential for developing more effective treatment and prevention strategies. If BV is sexually transmitted, partner treatment may be necessary to reduce recurrence. Addressing biofilm resilience could improve antibiotic efficacy, potentially through biofilm-disrupting agents or microbiome restoration therapies. Additionally, racial disparities highlight the need for research into genetic, environmental, and behavioral influences on vaginal microbiota. Future studies should focus on controlled clinical trials and microbiome-based interventions to prevent and manage BV more effectively.

Prebiotic Maltose Gel Can Promote the Vaginal Microbiota From BV-Related Bacteria Dominant to Lactobacillus in Rhesus Macaque

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study explores the impact of prebiotic maltose gel on the vaginal microbiota of rhesus macaques, showing its potential to shift microbial balance from BV-related bacteria to Lactobacillus dominance, presenting an alternative to antibiotic treatments for bacterial vaginosis.

What was studied?

This study examined the effects of prebiotic maltose gel on the vaginal microbiota in rhesus macaques, specifically its ability to promote the transition of the vaginal microbiota from a bacterial vaginosis (BV)-related bacteria-dominant state to a Lactobacillus-dominant state. The researchers tested whether maltose gel, as a non-antibiotic agent, could effectively encourage the proliferation of Lactobacillus species while suppressing the growth of BV-associated bacteria such as Fusobacterium, Parvimonas, and Mobiluncus.

Who was studied?

The study involved eighteen healthy female rhesus macaques, who were randomly divided into two groups. One group received the prebiotic maltose gel treatment, while the other received a placebo gel. The researchers collected vaginal microbiota samples at several time points during the treatment and after the gel withdrawal to observe changes in microbial composition.

What were the most important findings?

The results showed that maltose gel treatment significantly increased the abundance of Lactobacillus in the vaginal microbiota of rhesus macaques. Throughout the treatment, the Lactobacillus levels gradually increased, while the diversity and abundance of BV-associated bacteria, such as Fusobacterium, Parvimonas, Mobiluncus, and others, decreased. However, following the withdrawal of maltose gel, the Lactobacillus levels gradually decreased, although they remained higher than baseline levels at certain time points, indicating a lasting but moderate effect. This shift towards Lactobacillus dominance in the vaginal microbiota supports the potential of maltose gel as a prebiotic treatment for bacterial vaginosis (BV).

In terms of microbial diversity, the alpha diversity indices of the vaginal microbiota decreased significantly during treatment with maltose gel. The treatment caused a marked decrease in diversity, while the placebo group showed no significant changes. After drug withdrawal, the diversity of microbiota in both groups tended to increase, but the effects of maltose gel were more persistent in promoting Lactobacillus proliferation.

What are the implications of this study?

The study suggests that maltose gel may serve as a promising alternative to antibiotics in the treatment and prevention of bacterial vaginosis. Since BV is often recurrent despite antibiotic treatments, which can also disrupt beneficial Lactobacillus species, maltose gel offers a non-antibiotic strategy that can potentially maintain a healthy vaginal microbiota dominated by Lactobacillus. The prebiotic nature of maltose gel promotes the growth of Lactobacillus while reducing the harmful bacteria associated with BV, without inducing antibiotic resistance.

This study highlights the potential of developing prebiotics like maltose gel as adjunct therapies to traditional BV treatments, offering a more sustainable, long-term solution that supports the microbiome's natural composition. However, further studies, including those in human populations, are necessary to assess the long-term effects and feasibility of such treatments.

Probiotics are a Good Choice for the Treatment of Bacterial Vaginosis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This meta-analysis reviewed 12 randomized controlled trials and found that probiotics significantly improve the clinical cure rate of bacterial vaginosis. Probiotic use restored beneficial Lactobacillus populations and reduced pathogenic bacteria, highlighting their potential as an effective treatment option and adjunct therapy alongside antibiotics.

What was Reviewed?

The meta-analysis reviewed the efficacy of probiotics as a treatment option for bacterial vaginosis (BV). The authors systematically analyzed 20 randomized controlled trials (RCTs) involving 2093 participants to assess whether probiotics, used alone or alongside antibiotics, could improve BV cure rates and restore healthy vaginal microbiota. The review evaluated the clinical cure rates, recurrence rates, adverse effects, and microbial outcomes associated with probiotic administration, considering different probiotic strains, dosages, administration routes, and treatment durations.

Who was Reviewed?

The meta-analysis included RCTs that enrolled women diagnosed with bacterial vaginosis. These participants came from diverse populations, including various age groups and geographic locations. The included studies assessed the effects of probiotic therapy compared to placebo or standard antibiotic treatment, with follow-up periods ranging from a few weeks to several months. The reviewed trials involved women with symptomatic BV, recurrent BV, and asymptomatic cases diagnosed based on standard clinical criteria.

What were the most Important Findings?

This meta-analysis demonstrated that probiotics improved the clinical cure rate of bacterial vaginosis and reduced recurrence compared to control groups. The data consistently showed that probiotics, whether administered orally or intravaginally, enhanced treatment outcomes, with intravaginal probiotics showing slightly better performance. Notably, probiotics used alongside antibiotics provided additional clinical benefit beyond antibiotics alone, suggesting a synergistic relationship.

Beyond clinical outcomes, the review emphasized critical microbial shifts. Women receiving probiotics consistently exhibited an increased abundance of beneficial Lactobacillus species, particularly Lactobacillus crispatus and Lactobacillus jensenii. At the same time, pathogenic bacteria associated with BV, such as Gardnerella vaginalis and Atopobium vaginae, decreased. This microbial rebalancing supports the hypothesis that probiotics help restore vaginal eubiosis, reducing both symptoms and the risk of recurrence. The study's microbiome findings are especially valuable for understanding how probiotics can modify the vaginal ecosystem in BV patients.

What are the Implications of this Review?

This meta-analysis provides strong evidence supporting the use of probiotics as a safe and effective adjunct or alternative treatment for bacterial vaginosis. It underscores the clinical benefit of probiotics in enhancing cure rates, reducing recurrence, and promoting a healthy vaginal microbiota dominated by Lactobacillus species. The findings suggest that clinicians should consider integrating probiotic therapy, particularly intravaginal formulations, into BV treatment protocols. Furthermore, this analysis supports the role of microbiome-targeted therapies in restoring microbial balance and reducing the risk of recurrent infections. The microbial associations identified here, particularly the positive shifts toward Lactobacillus dominance, provide valuable microbial signatures for future BV management and intervention strategies.

Recurrent Bacterial Vaginosis Following Metronidazole Treatment is Associated with Microbiota Richness at Diagnosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study links high pre-treatment vaginal microbiota diversity to BV recurrence after metronidazole. Women with sustained clearance had lower richness. Lactobacillus iners improved immune markers temporarily, but no cases achieved L. crispatus dominance. Biofilm-forming taxa like Atopobium persisted, suggesting resistance mechanisms.

What was Studied?

This study investigated the association between pre-treatment vaginal microbiota composition and the likelihood of recurrent bacterial vaginosis (BV) following metronidazole treatment. The researchers analyzed cervicovaginal lavage samples from women diagnosed with BV using 16S rRNA gene sequencing to identify microbial signatures linked to treatment failure or success. The study aimed to determine whether specific microbiota characteristics at diagnosis could predict treatment outcomes, including transient clearance, sustained clearance, or recurrence of BV.

Who was Studied?

The study included 28 women diagnosed with symptomatic BV, confirmed by Nugent scoring, who were enrolled in a clinical trial. Participants were non-pregnant, free of other reproductive tract infections, and had not used antibiotics in the 14 days before enrollment. Samples were collected at baseline (pre-treatment), 7–10 days post-treatment, and 28–32 days post-treatment to assess microbial and immune changes.

What were the most Important Findings?

The study revealed that women who failed to clear BV or experienced recurrence had significantly higher pre-treatment microbial richness and evenness than those with sustained clearance. Significant microbial associations (MMA) included polymicrobial anaerobic taxa such as Gardnerella vaginalisPrevotellaSneathia, and Atopobium, which were dominant at baseline. Notably, Lactobacillus iners (CT2) dominance post-treatment was associated with improved mucosal immune markers, including elevated SLPI and reduced ICAM-1, but these benefits were transient in cases of recurrence. The persistence of diverse, low-abundance taxa and biofilm-forming bacteria like Atopobium and Sneathia post-treatment suggested their role in treatment resistance. Importantly, no participants achieved Lactobacillus crispatus (CT1) dominance, highlighting a gap in current therapeutic efficacy.

What are the Implications of this Study?

The findings underscore the limitations of metronidazole in treating BV, particularly in cases with high pre-treatment microbial diversity. The study suggests that microbiome profiling could help identify women at risk of treatment failure, paving the way for personalized therapies. Future research should explore adjunct treatments, such as Lactobacillus crispatus biotherapeutics or biofilm disruptors, to improve outcomes. Additionally, the transient immune improvements observed with Lactobacillus dominance emphasize the need for sustained microbiome modulation to prevent recurrence and associated complications like STI susceptibility.

Recurrent bacterial vaginosis–an old approach to a new problem

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

The study finds that hydrogen peroxide vaginal washout effectively alleviates recurrent bacterial vaginosis symptoms, restoring pH and eliminating pathogens in most cases, though it does not fully restore Lactobacillus populations.

What was Studied?

The study evaluated the efficacy of 3% hydrogen peroxide as a single vaginal washout treatment for recurrent bacterial vaginosis (rBV). The primary aim was to assess how hydrogen peroxide affects the symptoms and microbiological profile of women with rBV, particularly focusing on the reduction of vaginal malodor, restoration of vaginal pH, and the absence of bacterial pathogens like Gardnerella vaginalis and other anaerobes.

Who was Studied?

The study involved 30 women with clinically confirmed recurrent bacterial vaginosis, defined as experiencing symptoms of vaginal malodour and discharge after having previously been treated for BV with metronidazole. These women were recruited from a clinical setting and included only those who were symptomatic with BV, had mixed anaerobes isolated in their vaginal swabs, and did not have other genital infections such as gonorrhea, chlamydia, or Candida. A total of 23 women completed the study.

What were the Most Important Findings?

The results demonstrated that the hydrogen peroxide vaginal washout was effective in reducing the malodorous vaginal discharge, with 78% of women reporting complete resolution of symptoms after three weeks. The treatment also led to improvements in vaginal pH, with 96% of women returning to a normal acidic pH. All women showed an absence of "clue cells" and a lack of mixed anaerobes in their vaginal cultures post-treatment. The presence of Lactobacillus species also improved, though to a lesser extent. The study found that hydrogen peroxide was well-tolerated by all participants, with no significant adverse effects such as irritation or inflammation.

However, while hydrogen peroxide improved discharge and malodour in most cases, the full restoration of vaginal lactobacilli colonies did not occur in all women, and two women still experienced mild symptoms. The treatment also failed to restore lactobacilli levels to the extent seen in other BV treatments, indicating that while hydrogen peroxide is effective for symptom relief, it may not be sufficient to fully re-establish the normal vaginal microbiota.

What are the Implications of this Study?

This study suggests that hydrogen peroxide vaginal washout could serve as an alternative or adjunct to antibiotics for treating recurrent bacterial vaginosis. It offers a non-antibiotic approach that is well-tolerated and effective in reducing the primary symptom of malodour. Given the lack of significant side effects and the complete absence of bacterial pathogens in post-treatment cultures, hydrogen peroxide may provide a useful option for women who experience frequent recurrences of BV and are reluctant to use antibiotics due to side effects or resistance concerns. However, the study highlights that hydrogen peroxide treatment may not fully restore vaginal microbiota, specifically Lactobacillus populations, which suggests the need for additional interventions to ensure long-term microbiota health and prevent recurrence.

Saccharomyces cerevisiae–based Probiotic as Novel Anti-microbial Agent for Therapy of Bacterial Vaginosis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study evaluated a Saccharomyces cerevisiae-based probiotic for bacterial vaginosis treatment. The probiotic selectively inhibited BV-associated pathogens like G. vaginalis without harming beneficial lactobacilli, offering a microbiome-friendly alternative to antibiotics.

What was Studied?

The study investigated the potential therapeutic effects of a Saccharomyces cerevisiae-based probiotic as a novel antimicrobial agent in the treatment of bacterial vaginosis (BV). The researchers aimed to evaluate whether this yeast-based probiotic could inhibit the growth of BV-associated pathogenic bacteria and restore vaginal microbial balance, offering an alternative to standard antibiotic treatments.

Who was Studied?

The study utilized in vitro models to assess the antimicrobial activity of the S. cerevisiae-based probiotic against a range of bacterial strains associated with bacterial vaginosis, including Gardnerella vaginalis, Atopobium vaginae, Mobiluncus curtisii, and others. No human or animal participants were involved; rather, laboratory strains of pathogenic bacteria were cultured and tested against the probiotic formulation.

What were the most Important Findings?

The study revealed that the S. cerevisiae-based probiotic demonstrated strong antimicrobial activity against key BV-associated pathogens. Specifically, the probiotic effectively inhibited the growth of G. vaginalis, A. vaginae, M. curtisii, and Prevotella bivia in vitro. Notably, the inhibition was dose-dependent, with higher concentrations of the probiotic resulting in greater suppression of these pathogens. Importantly, the probiotic did not affect beneficial Lactobacillus species such as L. crispatus and L. jensenii, which are critical for maintaining vaginal health. This selectivity highlights a significant microbial signature, the probiotic selectively targeted pathogenic bacteria associated with dysbiosis while sparing commensal, health-associated bacteria. Additionally, the study suggested that the probiotic may modulate the vaginal microbiome by reducing the overgrowth of anaerobic pathogens without disrupting the protective lactobacilli.

What are the Implications of this Study?

The findings of this study have significant implications for the management of bacterial vaginosis. Current BV treatments rely heavily on antibiotics, which often lead to recurrence and may disrupt the vaginal microbiota by eliminating beneficial lactobacilli alongside pathogens. The yeast-based probiotic offers a non-antibiotic therapeutic strategy that can selectively inhibit BV-associated pathogens while preserving or even promoting beneficial microbial populations. This approach could potentially reduce recurrence rates, limit the development of antibiotic resistance, and improve vaginal microbiome resilience. For clinicians, this highlights a promising avenue for microbiome-informed interventions in BV management that target dysbiosis while maintaining microbial balance.

Secnidazole for the Treatment of Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This systematic review analyzed six trials evaluating secnidazole for bacterial vaginosis treatment. Secnidazole at 2 g significantly improved clinical and microbiologic cure rates, showing comparable efficacy to metronidazole. The single-dose regimen enhances adherence, offering an alternative for patients with recurrent BV or adverse effects from standard therapies.

What was Reviewed?

This systematic review evaluated the clinical efficacy, safety, and microbiological outcomes of secnidazole as a treatment option for bacterial vaginosis (BV). The authors reviewed randomized controlled trials that compared secnidazole at different doses with placebo, standard antibiotic regimens, or combination therapies. The review also considered how secnidazole affected the vaginal microbiota, particularly its ability to reduce the abundance of BV-associated bacteria and restore beneficial Lactobacillus species.

Who was Reviewed?

The review encompassed clinical studies involving adult women diagnosed with bacterial vaginosis, with diagnosis typically based on Amsel criteria or Nugent score. The included studies varied in sample size but consistently targeted non-pregnant women of reproductive age who were experiencing symptomatic or recurrent BV. The population also included women with a history of BV treatment failures or recurrences, a subgroup of particular interest due to the chronic and recurrent nature of the condition.

What were the most Important Findings?

This review demonstrated that secnidazole significantly improved both the clinical and microbiological cure rates of bacterial vaginosis compared to placebo. Specifically, in women with three or fewer BV episodes in the last year, 2 g secnidazole substantially reduced BV risk. In women with four or more episodes, the benefit persisted but with slightly lower magnitude.

The clinical cure rate of 2 g secnidazole was comparable to metronidazole (500 mg), oral metronidazole 2 g single dose, secnidazole combined with vaginal metronidazole, or secnidazole plus vaginal ornidazole. However, the 2 g dose performed better than the 1 g dose.

This review highlighted that probiotic therapy was not the focus, but secnidazole use indirectly supports the concept of restoring vaginal eubiosis by reducing pathogenic bacteria. The review did not explicitly measure microbiome shifts in terms of Lactobacillus species or pathogenic taxa, but the improved microbiologic cure rate reflects pathogen reduction.

The authors also emphasized that a single-dose regimen of secnidazole improved patient adherence compared to multi-dose metronidazole or tinidazole therapies. However, beyond adherence, secnidazole's therapeutic effect was statistically similar to these standard treatments. The review proposed secnidazole as a good alternative for women who experienced adverse effects or recurrence with current BV medications.

What are the Implications of this Review?

This review offers clear clinical guidance: secnidazole at 2 g is an effective, single-dose treatment option for bacterial vaginosis, providing comparable cure rates to metronidazole and combination therapies. It may serve as a valuable alternative, particularly for women with recurrent BV or those who face side effects from standard antibiotics. Additionally, while the review did not analyze microbial signatures in detail, the consistent microbiologic cure rates indirectly support the role of secnidazole in reducing BV-associated dysbiosis. Clinicians should consider secnidazole as a viable option in their therapeutic arsenal, particularly when treatment adherence and recurrence prevention are priorities.

Single-Dose, Bioadhesive Clindamycin 2% Gel for Bacterial Vaginosis: A Randomized Controlled Trial

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study evaluates the efficacy and safety of a single-dose 2% clindamycin vaginal gel for treating bacterial vaginosis, demonstrating significant clinical and bacteriologic cure rates compared to a placebo.

What was studied?

This study focused on assessing the efficacy and safety of a single-dose, bioadhesive 2% clindamycin vaginal gel in treating bacterial vaginosis (BV). The study was randomized, controlled, and double-blind, comparing clindamycin with a placebo gel.

Who was studied?

The study included women who had a clinical diagnosis of bacterial vaginosis, defined by meeting all four Amsel’s criteria, and with Nugent scores of 7-10. The researchers randomized the participants into two groups: the clindamycin gel group and the placebo group. The study enrolled a racially diverse population, including a high percentage of Black women, and most participants had a history of recurrent BV.

What were the most important findings?

The study demonstrated that the 2% clindamycin vaginal gel was significantly more effective than the placebo in achieving clinical cure, defined as the resolution of three of the four Amsel’s criteria, at the test-of-cure visit (day 21-30). The clinical cure rate was 70.5% for the clindamycin group, compared to 35.6% for the placebo group, with a difference of 34.9%. Additionally, clindamycin showed statistically significant improvements in bacteriologic and therapeutic cure rates. The gel was also well-tolerated, with vulvovaginal candidiasis being the most common adverse event, a known side effect of clindamycin use.

The study highlights the importance of the bioadhesive property of clindamycin gel, which allows for sustained drug release, thus increasing retention and enhancing the treatment’s efficacy. This mechanism is particularly relevant for improving patient compliance and the therapeutic outcomes in BV treatment. The study design adhered to FDA guidance, specifically including only participants with high Nugent scores (7-10), which strengthens the validity of the findings.

What are the implications of this study?

The study’s findings offer a promising new option for treating BV with a single-dose vaginal gel that enhances patient compliance through reduced leakage and prolonged retention time. The significant clinical cure rates observed in patients with recurrent BV are especially important, as recurrent BV is a common and challenging condition to manage. The study demonstrates that clindamycin’s bioadhesive formulation may be more effective than traditional treatment options that require multiple applications. This gel could become an essential treatment in managing BV, especially in women who experience frequent recurrences.

The study supports the need for further research into improving BV treatment strategies. It also reinforces the importance of managing BV to prevent complications such as infertility, pelvic inflammatory disease, and increased susceptibility to sexually transmitted infections, including HIV.

The Human Microbiome during Bacterial Vaginosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explores bacterial vaginosis, focusing on microbial imbalances, immune responses, and diagnostic challenges. It highlights the need for microbiome-based treatments and improved diagnostics to reduce recurrence and improve BV management.

What Was Reviewed?

This review examines the human microbiome during bacterial vaginosis (BV), focusing on microbial shifts, host immune responses, and diagnostic challenges. It evaluates BV as a polymicrobial condition rather than an infection caused by a single pathogen. The review highlights how microbial imbalances contribute to BV symptoms, persistence, and recurrence. Additionally, it explores epidemiological factors, diagnostic methods, and host-microbiome interactions that influence BV progression and treatment response.

Who Was Reviewed?

The review synthesizes findings from studies involving women diagnosed with BV, including those experiencing recurrent infections. It incorporates data from molecular sequencing studies and microbiological research to assess the composition of the vaginal microbiome during BV. Additionally, it examines host immune responses to BV-related microbial changes and evaluates the link between BV and increased susceptibility to sexually transmitted infections (STIs) and pregnancy complications such as preterm birth.

Most Important Findings

BV disrupts the vaginal microbiome by reducing Lactobacillus species and increasing anaerobic bacteria such as Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., Mobiluncus spp., and Sneathia spp. Unlike infections that trigger a strong inflammatory response, BV presents as a microbial imbalance rather than an acute immune reaction. The review also highlights how bacterial biofilms protect BV-associated bacteria from antibiotic treatment, contributing to high recurrence rates.

The study also discusses BV’s complex interaction with the host immune system. BV-associated bacteria produce virulence factors that degrade the vaginal mucosal barrier, leading to increased inflammatory markers such as interleukin-1β (IL-1β) and tumor necrosis factor-alpha (TNF-α). This immune dysregulation may explain BV’s association with increased STI susceptibility and adverse pregnancy outcomes.

Diagnosing BV remains challenging due to inconsistencies in clinical and laboratory criteria. Amsel’s clinical criteria and Nugent scoring, which rely on symptom assessment and Gram staining, remain the primary diagnostic tools. However, these methods fail to account for BV’s diverse microbial community, leading to inconsistencies in diagnosing recurrent and persistent cases. The review emphasizes the need for molecular sequencing-based diagnostics that provide a more precise understanding of BV-associated bacterial communities.

Implications of the Review

BV presents a significant clinical challenge due to its high recurrence rate, treatment limitations, and association with reproductive health complications. The review underscores the importance of shifting towards microbiome-targeted therapies rather than relying solely on broad-spectrum antibiotics. Future research should focus on developing treatments that restore Lactobacillus-dominant vaginal microbiota and prevent biofilm formation. Refining molecular diagnostic techniques will help clinicians identify BV-associated bacterial communities more accurately, improving treatment strategies and reducing recurrence.

This review highlights the urgent need for improved diagnostic criteria, personalized treatment approaches, and a deeper understanding of the vaginal microbiome’s role in BV persistence. Advancing these areas of research will help clinicians develop more effective, long-term solutions for BV management.

The interplay between microbiota, metabolites, immunity during BV

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review explains how microbial shifts, metabolite production, and immune responses interact in bacterial vaginosis. It highlights the roles of Gardnerella, Atopobium, and other anaerobes in disrupting vaginal health and discusses how their metabolic byproducts and immune modulation drive BV symptoms and persistence.

What was Reviewed?

This review explored the complex interplay between the vaginal microbiota, metabolite production, and local immune responses in the pathogenesis of bacterial vaginosis (BV). The authors synthesized existing research on how shifts in the vaginal microbiome from Lactobacillus-dominant communities to polymicrobial anaerobic communities contribute to BV development, symptoms, and recurrence. The review particularly emphasized the combined role of microbiota composition, bacterial metabolic products, and vaginal immune responses in driving clinical outcomes and disease persistence.

Who was Reviewed?

The review covered a wide body of research focusing on reproductive-age women diagnosed with or at risk for BV. It drew from studies examining the vaginal microbiota, including key bacteria such as Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., Mobiluncus spp., and Sneathia sanguinegens, as well as the protective Lactobacillus species like L. crispatus and L. iners. It also reviewed studies on the vaginal metabolome and immune responses in BV-positive and BV-negative women.

Most Important Findings

The review consolidated evidence that BV is a multifactorial condition characterized by dysbiosis of the vaginal microbiota, metabolic disruption, and altered immune responses. It described how healthy vaginal microbiomes are dominated by Lactobacillus species, particularly L. crispatus, which maintain vaginal acidity and protect against pathogens. In contrast, BV involves a shift toward a polymicrobial anaerobic community, with increased abundance of Gardnerella vaginalis, Atopobium vaginae, Prevotella spp., Mobiluncus spp., and Sneathia spp., collectively referred to as major microbial associations (MMA) of BV.

These BV-associated bacteria produce key metabolites, including short-chain fatty acids (SCFAs) like acetate and propionate, and volatile amines like putrescine and cadaverine, which raise vaginal pH and produce the characteristic fishy odor of BV. Additionally, BV-associated biofilm formation, particularly involving G. vaginalis and A. vaginae, enhances bacterial persistence and resistance to treatment.

The review highlighted that these microbial and metabolic changes interact with the host’s immune system. BV patients exhibit elevated pro-inflammatory cytokines, especially IL-1β, without a corresponding increase in neutrophil recruitment. This unique immune profile likely results from SCFA-mediated suppression of neutrophil chemotaxis and explains why BV lacks overt inflammatory symptoms despite microbial overgrowth.

Implications of this Review

This review emphasizes the need to redefine BV beyond a simple microbial imbalance. It emphasizes that the metabolic products of BV-associated bacteria and their impact on host immunity are central to disease progression and recurrence. Clinicians should consider not only microbial community shifts but also metabolite profiles and immune responses when diagnosing and managing BV. The review calls for the integration of multi-omic data, microbiome, metabolome, and immunome, to develop more accurate diagnostics and targeted therapeutic strategies. Understanding these interactions may guide the design of personalized interventions to restore microbial and metabolic homeostasis, reduce BV recurrence, and mitigate associated reproductive health risks.

The right bug in the right place: opportunities for bacterial vaginosis treatment

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review explores the role of vaginal microbiota in bacterial vaginosis and highlights emerging microbiome-informed treatments. It emphasizes microbial signatures of BV, the limitations of antibiotics, and the potential of targeted biotherapeutics to restore microbial balance and reduce recurrence.

What was Reviewed?

This review examines the current understanding of the vaginal microbiome and its relationship to bacterial vaginosis (BV). It discusses how microbial dysbiosis contributes to the onset and persistence of BV and evaluates the potential therapeutic strategies that could leverage microbiome science to treat and prevent the condition. The authors explore the complexity of vaginal microbial communities, particularly focusing on the imbalance between health-associated Lactobacillus species and BV-associated anaerobic bacteria. They review both existing antibiotic treatments and emerging microbiome-informed interventions, including live biotherapeutics and vaginal microbiota transplants (VMT).

Who was Reviewed?

The review focused on published research involving women diagnosed with bacterial vaginosis, as well as healthy women with Lactobacillus-dominated vaginal microbiota. The authors synthesized data from clinical studies, in vitro experiments, and microbiome profiling studies that examined microbial composition, treatment responses, and microbial dynamics in BV-affected and healthy populations. They also reviewed preclinical studies exploring potential microbial therapeutics, including specific bacterial strains and vaginal microbiome restoration strategies.

What were the Most Important Findings?

The review highlighted that bacterial vaginosis is characterized by a distinct microbial signature: a depletion of Lactobacillus species (notably L. crispatus, L. jensenii, and L. gasseri) and an overgrowth of anaerobic bacteria. This microbial imbalance leads to elevated vaginal pH and inflammation, contributing to symptoms and increasing susceptibility to other infections.

The authors emphasized that standard antibiotic treatments, like metronidazole and clindamycin, often result in high recurrence rates and can disrupt both pathogenic and beneficial bacterial populations. They reviewed emerging microbiome-based therapies aimed at correcting vaginal dysbiosis without harming commensal microbes. These include probiotic formulations containing Lactobacillus strains, VMT, and precision antimicrobials targeting specific BV-associated pathogens. Notably, they discussed the importance of strain-specific effects, showing that not all Lactobacillus strains equally promote vaginal health, and that strain selection is critical for therapeutic success.

A key finding was that sustained remission from BV is linked to successful re-establishment of a Lactobacillus-dominant community, specifically L. crispatus. The review also addressed how host factors, sexual activity, and antibiotic exposure influence microbial dynamics, indicating the need for personalized, microbiome-informed approaches to BV treatment.

What are the Implications of this Review?

This review carries significant implications for clinicians managing bacterial vaginosis. It highlights the limitations of antibiotic-centric treatments and underscores the need for microbiome-conscious strategies that restore and maintain vaginal microbial balance. The evidence supports moving toward targeted interventions such as live biotherapeutics and VMT, which can selectively suppress BV-associated pathogens while promoting beneficial lactobacilli. Clinicians should consider that effective, long-term BV management may depend not only on pathogen eradication but also on rebuilding a resilient, health-associated vaginal microbiome. The review points to the potential of precision microbial therapies tailored to individual microbial profiles, marking a shift toward personalized vaginal microbiome medicine. For microbiome signatures research, the paper enriches the understanding of the specific bacterial players involved in BV dysbiosis and recovery.

Tinidazole in the treatment of bacterial vaginosis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This review assesses the use of tinidazole for treating bacterial vaginosis, comparing its efficacy, side effects, and cost to metronidazole. It highlights tinidazole’s favorable side effect profile and its role in treating recurrent BV.

What was studied?

The study evaluated the clinical effectiveness of tinidazole in treating bacterial vaginosis (BV) and compared different dosing regimens. It specifically examined the use of tinidazole in comparison with placebo, focusing on its impact on cure rates and recurrence prevention. Additionally, the pharmacokinetics, safety, and microbial susceptibility of tinidazole in the context of BV treatment were assessed.

Who was studied?

The study involved patients diagnosed with bacterial vaginosis, and it included women treated with tinidazole to evaluate its efficacy. Various groups were compared, including those receiving different doses of tinidazole and a placebo group.

What were the most important findings?

The study found that tinidazole demonstrated significant efficacy in the treatment of BV, with cure rates notably higher in the tinidazole groups compared to placebo. Specifically, the 2 g single-dose regimen was shown to be more effective than placebo, but there was no significant difference in efficacy between tinidazole given in a 2-day regimen versus a single 2 g dose. Moreover, the research revealed that the drug's antimicrobial activity extended beyond typical BV-associated pathogens like Gardnerella vaginalis to other anaerobic species. However, resistance was noted in 54% of G. vaginalis isolates and 96% of Lactobacillus isolates, indicating the complex dynamics of the vaginal microbiome in BV. The study also evaluated the safety profile of tinidazole, showing that it was generally well-tolerated compared to other treatments like metronidazole, with fewer gastrointestinal side effects.

What are the greatest implications of this study?

The study suggests that tinidazole is a viable alternative to metronidazole for BV treatment, especially for recurrent cases where metronidazole may have limited effectiveness. The findings support the use of tinidazole in patients who have not responded well to first-line treatments and indicate that it may be a useful agent for reducing recurrence, particularly when administered with proper dosing regimens. Moreover, tinidazole's action against G. vaginalis and other anaerobes reinforces the need to understand microbial resistance patterns when treating BV, highlighting the complexity of the vaginal microbiome. These results could encourage clinicians to adopt tinidazole more frequently in clinical practice, particularly for cases where standard therapies fail.

Treatment of Bacterial Vaginosis with Topical Clindamycin or Metronidazole

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study compares the microbiological effects of metronidazole and clindamycin in treating bacterial vaginosis, highlighting differences in antimicrobial resistance and efficacy in eliminating BV-associated pathogens.

What was studied?

The study investigated the microbiologic response to treatment for bacterial vaginosis (BV) with topical clindamycin and metronidazole. It focused on the microbiological changes observed in vaginal flora before and after treatment, assessing the impact of these treatments on bacterial populations, including Gardnerella vaginalis, Mycoplasma hominis, and anaerobic gram-negative rods.

Who was studied?

The study included 119 nonpregnant, premenopausal women aged 18 to 45 diagnosed with BV using clinical and Gram stain criteria. They were randomized to receive either clindamycin vaginal ovules or metronidazole vaginal gel. The study also evaluated the microbiologic response over a 3-month follow-up period.

What were the most important findings?

The study revealed that both metronidazole and clindamycin treatments resulted in significant changes in the vaginal microflora. Both treatments led to decreased colonization by Gardnerella vaginalis and Mycoplasma hominis, common BV-associated pathogens. However, metronidazole was more effective in reducing the colonization of Prevotella bivia and black-pigmented Prevotella species. Clindamycin treatment resulted in the emergence of resistant subpopulations of P. bivia and black-pigmented Prevotella species, with resistance to clindamycin increasing significantly 7 to 12 days after treatment. In contrast, metronidazole showed no such increase in resistance. The study found that while both treatments resulted in similar clinical cure rates, the microbiological response differed between the two, with metronidazole proving to be more effective in eradicating anaerobic gram-negative rods. The study further emphasized that the increased clindamycin resistance following treatment with clindamycin could complicate the management of BV, especially with recurrent cases.

What are the implications of this study?

The study highlights the differences in the microbiologic response to clindamycin and metronidazole, suggesting that while both are effective in treating BV, metronidazole may offer a more favorable outcome, particularly in terms of preventing the emergence of antibiotic resistance. The increased clindamycin resistance observed with repeated use suggests that clindamycin may not be the ideal choice for recurrent BV cases. This finding has implications for clinicians in choosing the most appropriate treatment for BV, especially for patients with recurrent infections. The study underscores the importance of antimicrobial stewardship and the potential for developing resistance with the overuse of antibiotics like clindamycin.

Unveiling Resistance and Virulence Mechanisms under Darwinian Positive Selection for Novel Drug Discovery for Gardnerella vaginalis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

The study analyzed the genetic evolution of Gardnerella vaginalis, focusing on its resistance and virulence under Darwinian positive selection. It identifies new drug targets and emphasizes the pathogen's evolving resistance mechanisms.

What was Studied?

The study focused on Gardnerella vaginalis, a significant pathogen responsible for bacterial vaginosis(BV), examining its mechanisms of resistance and virulence under Darwinian positive selection. The researchers utilized comparative genomic analysis to identify resistance and virulence-related genes and their evolutionary patterns. The study also aimed to discover potential new drug targets by analyzing these genomic features in the context of the pathogen's evolutionary adaptations.

Who was Studied?

The study analyzed 97 genomes of Gardnerella vaginalis strains, representing a diverse collection of isolates obtained from the National Center for Biotechnology Information (NCBI) datasets. The strains were carefully selected to reflect the genetic variability and resistance phenotypes of this important pathogen, enabling a comprehensive understanding of its evolution.

What were the most Important Findings?

The study identified several crucial findings that provide new insights into the evolution and pathogenic potential of G. vaginalis. The pathogen exhibits significant genomic diversity, which plays a role in its survival and adaptation to selective pressures, particularly from antibiotics. The analysis revealed some genes, such as Mef(A), associated with resistance to macrolides, and tet(M) and tet(L), linked to resistance against tetracycline. These resistance genes were found to be positively selected in multiple G. vaginalis lineages, reflecting the evolutionary pressures that have shaped the pathogen's resistance capabilities.

Furthermore, the study highlighted the pathogen’s ability to form biofilms, a feature that enhances its survival in the host and increases its resistance to antibiotic treatment. This biofilm formation is also associated with the pathogen's ability to engage in horizontal gene transfer, further complicating the treatment landscape. The pan-resistome analysis indicated that the pathogen has an "open" resistome, suggesting its high capacity to acquire new resistance genes, making it a continuously evolving threat. The researchers also identified two potential drug targets, sigA, a sigma factor involved in transcription initiation, and UDP-N-acetylenolpyruvoylglucosamine reductase, an enzyme crucial for cell wall synthesis. These proteins are vital to the pathogen's survival and represent promising targets for the development of new therapeutic approaches.

What are the Implications of this Study?

The study’s findings highlight the dynamic nature of Gardnerella vaginalis and its ability to rapidly adapt to environmental pressures, particularly through the acquisition of resistance genes. The evolution of resistance mechanisms and the presence of virulence factors underscore the pathogen's significant role in reproductive and sexual health complications. The open pan-resistome suggests that G. vaginalis can continue to evolve and acquire new resistance traits, posing an ongoing challenge to existing treatments. The identification of novel drug targets like sigA and UDP-N-acetylenolpyruvoylglucosamine reductase offers valuable insights into how future therapies could be designed to combat infections caused by this pathogen. This research calls for continued surveillance of G. vaginalis strains to track resistance trends and refine clinical treatment strategies.

Vaginal Microbiome Recovery After Bacterial Vaginosis Treatment

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study examines vaginal microbiome recovery after metronidazole treatment for BV. Findings show delayed microbiota stabilization, L. iners dominance post-treatment, and the need for improved diagnostic tools to predict BV recurrence.

What Was Studied?

This observational study examined how the vaginal microbiota recovers after standard antibiotic treatment for bacterial vaginosis (BV). Researchers assessed changes in microbial composition before and after a five-day metronidazole treatment to determine how long it takes for the vaginal microbiome to return to a healthy state.

Who Was Studied?

The study included 30 women diagnosed with BV and 30 healthy women as controls. Researchers collected vaginal swabs before treatment (Day 1) and at follow-ups on Day 8 and Day 15 to compare microbiota recovery between the two groups. They analyzed microbial composition using 16S rRNA gene sequencing and measured BV status using Nugent scores.

Most Important Findings

Before treatment, BV-positive women had lower levels of Lactobacillus crispatus and Lactobacillus jensenii and a higher presence of Gardnerella vaginalis, Prevotella timonensis/bivia, and Atopobium vaginae. After metronidazole treatment, microbial diversity significantly decreased, and Lactobacillus iners became dominant (67.8% on Day 8). By Day 15, the vaginal microbiota of BV-treated women closely resembled that of healthy women. However, some BV-associated bacteria, including G. vaginalis and P. timonensis/bivia, began to re-emerge in a subset of participants.

The study also highlighted inconsistencies between Nugent scoring and microbiome sequencing results. While Nugent scores normalized in most BV-treated women by Day 8, sequencing data showed that bacterial communities still differed from those of healthy women, only stabilizing around Day 15. These findings suggest that clinical diagnostic methods may not fully capture microbiome recovery dynamics.

Implications of the Study

This study provides valuable insights into BV treatment outcomes and microbiome recovery. While metronidazole effectively reduces BV-associated bacteria, microbiome shifts continue for up to two weeks post-treatment. The dominance of L. iners after treatment raises questions about its role in BV recurrence. Future treatments should focus on restoring a L. crispatus-dominated microbiota, which is more protective against BV. The study also highlights the need for molecular-based diagnostics to better track microbiome recovery and predict BV recurrence.

Vaginal Microbiota Molecular Profiling in Women with Bacterial Vaginosis

May 20, 2025
  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

This study assessed vaginal microbiota in BV-positive women using real-time PCR. It found reduced Lactobacillus crispatus, biofilm-associated pathogens, and high diagnostic accuracy for molecular testing. Results suggest PCR-based diagnostics improve BV detection and highlight the need for microbiome-targeted therapies.

What Was Studied?

This study evaluated the microbial composition of the vaginal microbiota in women with bacterial vaginosis (BV) using molecular profiling techniques. Researchers aimed to determine the dominant bacterial species in BV, assess the role of Lactobacillus species, and evaluate the diagnostic potential of a multiplex real-time PCR test. This approach was considered as an alternative to traditional diagnostic methods like Amsel’s criteria and Nugent scoring, which often lack consistency.

Who Was Studied?

The study included 331 non-pregnant women who reported vaginal discharge. BV was confirmed through clinical examination and Nugent scoring. To gain a more detailed understanding of microbial composition, researchers analyzed vaginal microbiota using a real-time PCR test called Femoflor. This test detects key BV-associated bacteria, sexually transmitted disease (STD) pathogens, and some viruses, providing a broader perspective on vaginal health.

Most Important Findings

The study found that BV-positive women had significantly lower Lactobacillus levels compared to healthy controls. More specifically, Lactobacillus crispatus was severely depleted, while Lactobacillus iners remained dominant in many BV cases. This distinction is important, as L. crispatus is associated with a healthy vaginal microbiome, whereas L. iners is often found in transitional microbiota states and may contribute to recurrence.

Additionally, the study identified a significant presence of anaerobic bacteria in BV cases. Gardnerella vaginalis was the most prevalent, followed closely by Atopobium vaginae (recently renamed Fannyhessea vaginae), Prevotella bivia, Mobiluncus spp., Peptostreptococcus anaerobius, and Megasphaera spp.. These bacteria play a crucial role in biofilm formation, which not only protects BV-associated pathogens from antibiotic treatment but also increases the likelihood of recurrence.

The study emphasized the high diagnostic accuracy of the Femoflor real-time PCR test. Unlike traditional methods, this molecular test demonstrated high sensitivity and specificity in detecting BV-associated microbiota, even in cases where the vaginal microbiota appeared intermediate. The test was also able to detect STD pathogens such as Mycoplasma genitalium, Chlamydia trachomatis, and Neisseria gonorrhoeae, which were present only in BV-positive women. This finding suggests a potential link between BV and increased susceptibility to sexually transmitted infections.

Implications of the Study

These findings reinforce the need for molecular diagnostics in BV management. Traditional methods like Amsel’s criteria and Nugent scoring rely on subjective interpretation and often fail to identify BV in women with intermediate microbiota. The study suggests that real-time PCR testing offers a more reliable alternative, improving diagnostic accuracy and guiding more targeted treatment approaches.

The depletion of L. crispatus and dominance of L. iners in BV cases also raises concerns about the effectiveness of current treatment strategies. L. iners is often present in transitional microbiota states and does not provide the same protective benefits as L. crispatus. Future therapies should focus on restoring L. crispatus-dominated microbiota while addressing biofilm-associated bacterial communities to prevent recurrence. The study supports integrating molecular diagnostics into routine gynecological care and highlights the need for microbiome-targeted interventions to improve BV outcomes.

Clindamycin

Go to Category Page

A Review of the Anti-inflammatory Properties of Clindamycin in the Treatment of Acne Vulgaris

May 20, 2025
  • Clindamycin
    Clindamycin

    Clindamycin is an antibiotic commonly used to treat a variety of bacterial infections, including skin, bone, joint infections, and bacterial vaginosis. Known for its ability to target Gram-positive bacteria and anaerobes, Clindamycin disrupts protein synthesis in bacteria, halting their growth. While highly effective, its impact on the gut microbiome and the potential for Clostridium difficile infections make it essential to use with caution in certain populations.

This review details clindamycin’s anti-inflammatory role in acne, showing its impact on C. acnes, cytokines, and immune responses beyond bacterial suppression.

What Was Reviewed?

This review explores the anti-inflammatory properties of clindamycin in the treatment of acne vulgaris. It compiles data on how clindamycin, traditionally recognized for its antibacterial activity, also exhibits significant immunomodulatory effects. The paper focuses on clindamycin’s ability to inhibit Propionibacterium acnes (now Cutibacterium acnes), a key player in acne pathogenesis, and its impact on the inflammatory cascade triggered by this bacterium. The review outlines how clindamycin affects proinflammatory cytokines, leukocyte chemotaxis, phagocytosis, and various cellular pathways, reinforcing the idea that its therapeutic effects in acne extend beyond mere bacterial suppression​.

Who Was Reviewed?

The subjects of this review are patients with acne vulgaris, with particular emphasis on the microbiological and immunological dynamics within their pilosebaceous units. The review highlights the involvement of C. acnes and its interactions with host immune responses, detailing cytokine production and inflammatory cell recruitment. Human keratinocytes, monocytes, and neutrophils, both in vitro and in vivo, are discussed extensively to illustrate the inflammatory processes and clindamycin’s effects on them​.

What Were the Most Important Findings?

This review underscores that clindamycin’s acne-fighting power lies in both direct antibacterial action and critical anti-inflammatory activities. Clindamycin effectively inhibits C. acnes growth, lipase production, and the resulting free fatty acid buildup, all contributing to acne lesion development. The drug also inhibits the production of key inflammatory mediators, including interleukin-1β (IL-1β), interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), and granulocyte-macrophage colony-stimulating factor (GM-CSF), while suppressing leukocyte chemotaxis and enhancing phagocytosis. The review provides detailed evidence that clindamycin diminishes oxidative bursts from phagocytes and curbs the inflammatory cascade at multiple points, reinforcing that its clinical efficacy in acne likely stems from these combined effects, not solely its antibacterial function​.

What Are the Greatest Implications of This Review?

The findings emphasize the need for clinicians to recognize clindamycin’s dual-action nature. Its anti-inflammatory properties are especially relevant in cases where bacterial resistance is a concern, offering therapeutic benefits even when antibacterial effects are limited. The paper advocates for the continued use of clindamycin, particularly in combination therapies, while highlighting the necessity of antibiotic stewardship. This dual-action insight informs more nuanced acne treatment strategies, acknowledging the complex interplay between microbes and host immunity​.

Current indications for the use of clindamycin: A critical review

May 20, 2025
  • Clindamycin
    Clindamycin

    Clindamycin is an antibiotic commonly used to treat a variety of bacterial infections, including skin, bone, joint infections, and bacterial vaginosis. Known for its ability to target Gram-positive bacteria and anaerobes, Clindamycin disrupts protein synthesis in bacteria, halting their growth. While highly effective, its impact on the gut microbiome and the potential for Clostridium difficile infections make it essential to use with caution in certain populations.

This review analyzes clindamycin’s evidence-based use in infections, its anti-toxin benefits, and microbiome risks, especially C. difficile impact.

What Was Reviewed?

This critical review evaluates the current indications for clindamycin use, consolidating evidence-based guidelines across a range of infectious conditions. It synthesizes data from randomized clinical trials, cohort studies, and expert opinions published between 1966 and 1996, focusing on therapeutic efficacy and safety. The review categorizes evidence by clinical syndrome, discussing clindamycin’s roles in skin and soft tissue infections, necrotizing fasciitis, diabetic foot infections, osteomyelitis, head and neck infections, preoperative prophylaxis, pneumonia, intra-abdominal and pelvic infections, and select protozoal diseases. The review also highlights adverse effects, particularly the significant risk of Clostridium difficile-associated diarrhea​.

Who Was Reviewed?

This review encompasses a broad spectrum of patient populations with bacterial and protozoal infections treated with clindamycin. Key patient groups include individuals with skin and soft tissue infections, diabetic foot ulcers, osteomyelitis, necrotizing fasciitis, and recurrent group A streptococcal pharyngitis. The review also covers surgical patients requiring perioperative prophylaxis and those with anaerobic pulmonary infections or pelvic infections. Special attention is given to vulnerable populations such as diabetics and hospitalized patients at risk of C. difficile colonization, illustrating the intersection between microbial ecology and clinical outcomes​.

What Were the Most Important Findings?

The review confirms that clindamycin remains a vital alternative antibiotic, especially in patients allergic to beta-lactams or for infections involving toxin-producing pathogens like Streptococcus pyogenes. Notably, clindamycin’s efficacy in anaerobic lung infections, diabetic foot infections, and necrotizing fasciitis is well-supported, with its dual action of inhibiting bacterial protein synthesis and suppressing toxin production. However, the review underscores significant concerns over C. difficile-associated diarrhea, with inpatient colonization risks reaching up to 30%. While clindamycin demonstrates broad-spectrum anaerobic activity, its use in abdominal infections is increasingly challenged by cheaper, safer alternatives like metronidazole. From a microbiome standpoint, the paper emphasizes that clindamycin’s prolonged intestinal presence profoundly disrupts gut flora, increasing susceptibility to pathogenic colonization, especially in inpatient settings​.

What Are the Greatest Implications of This Review?

For clinicians, the key takeaway is the nuanced application of clindamycin: it should be reserved for cases with compelling evidence of benefit, such as invasive streptococcal infections or anaerobic lung abscesses. Stewardship is critical to limit microbiome disruption and resistance proliferation, particularly given the high incidence of C. difficile-related complications. The review calls for judicious prescribing practices, recommending metronidazole-based regimens where feasible and advocating for combination therapies in severe infections to optimize outcomes while minimizing adverse microbiome impacts​.

Scientific Rationale and Clinical Basis for Clindamycin Use in the Treatment of Dermatologic Disease

May 20, 2025
  • Clindamycin
    Clindamycin

    Clindamycin is an antibiotic commonly used to treat a variety of bacterial infections, including skin, bone, joint infections, and bacterial vaginosis. Known for its ability to target Gram-positive bacteria and anaerobes, Clindamycin disrupts protein synthesis in bacteria, halting their growth. While highly effective, its impact on the gut microbiome and the potential for Clostridium difficile infections make it essential to use with caution in certain populations.

This review examines clindamycin’s clinical utility in dermatology, spotlighting its antibacterial and anti-inflammatory roles, resistance concerns, and impact on skin microbiota, with a focus on acne vulgaris.

What Was Reviewed?

This review paper focuses on the scientific rationale and clinical basis for using clindamycin in treating dermatologic diseases. It details clindamycin’s antibacterial and anti-inflammatory roles, particularly its established use in acne vulgaris, folliculitis, rosacea, hidradenitis suppurativa (HS), and staphylococcal infections. The authors also explore its mechanism of action, highlighting how clindamycin inhibits bacterial protein synthesis by targeting the 50S subunit of the bacterial ribosome. The review contrasts clindamycin’s dermatological indications with those of tetracyclines. It checked trends in antibiotic resistance, offering a robust understanding of how clindamycin has maintained relevance in dermatology despite rising resistance concerns​.

Who Was Reviewed?

The review primarily covers patients affected by dermatologic conditions where clindamycin is indicated, including acne vulgaris sufferers, individuals with bacterial folliculitis, rosacea, HS, and staphylococcal infections. The paper contextualizes microbial involvement, particularly the role of Cutibacterium acnes (formerly Propionibacterium acnes), and outlines the bacterium's phylotypes and pathogenicity. It also reviews bacterial species relevant to resistance patterns, such as Staphylococcus aureus and Staphylococcus epidermidis, reflecting the broader impact of clindamycin on the skin microbiome​.

What Were the Most Important Findings?

Key findings center on clindamycin’s dual action: its efficacy in suppressing C. acnes growth and its notable anti-inflammatory effects. The review emphasizes that specific ribotypes of C. acnes are more virulent, contributing to inflammation and biofilm formation in acne. Topical clindamycin, often combined with benzoyl peroxide or retinoids, proves highly effective across various acne severities, with additional success noted in folliculitis, rosacea, and HS. Resistance trends are a major concern, with C. acnes resistance to clindamycin and macrolides reported in up to 90% of cases in some regions, driven by gene transfers. The review underscores the need for stewardship to mitigate resistance while recognizing clindamycin’s continued clinical value due to its safety, effectiveness, and versatility​.

What Are the Greatest Implications of This Review?

The review highlights the importance of balancing effective clindamycin use with antibiotic stewardship to limit resistance. For clinicians, the detailed exploration of C. acnes pathogenicity, clindamycin’s mechanism of action, and emerging resistance informs better treatment planning. The paper suggests that, despite high resistance rates, topical clindamycin remains a core treatment, particularly when combined with other agents to reduce monotherapy risks. Additionally, the work calls attention to the delicate balance of the skin microbiome and the need for ongoing surveillance and innovation in dermatologic therapies​.

Premenstrual Syndrome (PMS)

Go to Category Page

A Systematic Review and Meta-Analysis of Premenstrual Syndrome with Special Emphasis on Herbal Medicine and Nutritional Supplements.

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This systematic review and meta-analysis explores the effectiveness of herbal medicine and nutritional supplements in reducing PMS symptoms, highlighting significant reductions in physical, mood, and behavioral symptoms.

What was reviewed?

This article presents a systematic review and meta-analysis focusing on the effectiveness of herbal medicine and nutritional supplements in managing Premenstrual Syndrome (PMS). It reviews randomized controlled trials (RCTs) assessing the impact of various herbal remedies and supplements on both somatic and psycho-behavioral symptoms of PMS. The review highlights the safety, efficacy, and mechanisms of action of these treatments, while also addressing the limitations of current evidence.

Who was reviewed?

The review concentrates on women experiencing PMS, particularly those with both physical and psychological symptoms that significantly affect their daily functioning. It includes trials involving a wide range of herbal and nutritional interventions, aiming to provide clinicians with evidence of alternative therapies for managing PMS symptoms.

What were the most important findings?

The review reveals that herbal medicines and nutritional supplements can significantly reduce the severity of PMS symptoms, including physical, mood, and behavioral issues. It highlights specific interventions such as Vitex agnus castus (chaste tree), Zingiber officinale (ginger), and Crocus sativus (saffron) as particularly effective. The meta-analysis demonstrated a substantial reduction in Premenstrual Symptoms Screening Tool (PSST) scores, indicating notable symptom improvement. Additionally, the review found that certain interventions, such as Ginkgo biloba and vitamin B1, offered improvements in psychological symptoms like anxiety and depression.

Secondary findings also revealed positive effects on mood, physical symptoms, and behavior, with significant reductions in scores for physical symptoms and mood-related symptoms. The study concluded that herbal treatments, particularly when used over multiple cycles, have the potential to alleviate PMS symptoms significantly. However, it emphasized the need for high-quality trials to confirm these results and establish optimal treatment protocols. Importantly, the review also addressed the safety profile of these treatments, with most studies reporting mild adverse effects such as nausea or digestive discomfort.

What are the greatest implications of this review?

The greatest implication of this review is that herbal medicine and nutritional supplements represent a viable, non-pharmacological option for managing PMS symptoms, offering a safer alternative to traditional treatments with fewer side effects. This study provides clinicians with evidence-based recommendations for integrating these alternative therapies into PMS management, especially for patients who prefer natural treatments or experience adverse reactions to conventional medications. The review also underscores the need for more robust, high-quality studies to refine treatment guidelines, improve symptom assessment tools, and explore the mechanisms underlying the therapeutic effects of herbal interventions. Clinicians can consider incorporating these findings into patient care plans while awaiting further evidence from future studies.

Characteristics of the Gut Microbiota in Japanese Patients with Premenstrual Syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This study explores the gut microbiota in Japanese women with Premenstrual Syndrome (PMS), finding significant differences in microbial composition and highlighting the potential role of Collinsella in PMS pathophysiology.

What was studied?

This study investigated the characteristics of the gut microbiota in Japanese women with Premenstrual Syndrome (PMS), aiming to compare the microbial composition between PMS-affected individuals and healthy controls. By analyzing stool samples using 16S rRNA gene sequencing, the study examined the diversity of the microbiome and sought to identify microbial genera potentially associated with the severity of PMS symptoms.

Who was studied?

The study involved a total of 168 women, including 24 diagnosed with PMS and 144 healthy controls. The participants were between the ages of 24 and 49, with the control group selected to match the PMS group in terms of age and other relevant demographic factors. Women with other health conditions, those who had recently used antibiotics, or those with major dietary restrictions were excluded from the study. The aim was to understand how differences in the gut microbiome between the two groups might correlate with PMS symptoms.

What were the most important findings?

The study found that the gut microbiota of women with PMS differed significantly from that of healthy controls. The PMS group exhibited higher α-diversity, which was assessed using the Simpson index, indicating a more varied microbial community. When comparing the overall microbial composition between the groups, there were notable differences in β-diversity and statistical tests. The study also identified several microbial genera that were more abundant in women with PMS. Notably, Collinsella was found to be significantly more abundant in the PMS group, particularly among women aged 30–40 years. This genus was found to be 4.5 times more abundant in the PMS group compared to the controls, and its presence correlated with the severity of PMS symptoms. Bifidobacterium and Blautia, known for their roles in carbohydrate metabolism, were also more prevalent in the PMS group, potentially linking microbiome composition with metabolic processes that could influence PMS symptoms.

What are the greatest implications of this study?

The findings from this study suggest that the gut microbiota may play a significant role in the development and severity of PMS. The identification of Collinsella as a key player in the microbial composition of PMS-affected individuals opens new potential avenues for intervention. The findings imply that microbiome modulation, particularly through dietary adjustments or prebiotics targeting Collinsella, could offer a therapeutic strategy for managing PMS symptoms. The study also emphasizes the importance of further research to confirm these associations and explore the underlying mechanisms connecting microbiome imbalances with PMS.

Characteristics of the gut microbiota in women with premenstrual symptoms: A cross-sectional study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This study explores the association between gut microbiota composition and the severity of premenstrual symptoms, suggesting a potential role for microbiome-based therapies in managing PMS.

What was studied?

The study investigated the characteristics of the gut microbiota in women experiencing premenstrual symptoms, focusing on the potential association between gut microbial composition and the severity of these symptoms. The research aimed to identify microbial differences between women with premenstrual disorders (PMDs) and healthy controls, and to explore how these differences might correlate with the severity of premenstrual syndrome (PMS) symptoms.

Who was studied?

The study involved 56 women, including 27 women with self-reported premenstrual symptoms and 29 women without significant symptoms. Among the 27 women reporting PMS, 21 were further identified as having premenstrual disorders (PMDs) due to the significant interference of their symptoms with their social life. The control group consisted of 22 women with no significant premenstrual symptoms. These participants were matched by age and other demographic factors.

What were the most important findings?

The study found that there were notable differences in the gut microbiota between women experiencing premenstrual disorders (PMDs) and healthy controls. At the phylum level, the abundance of Bacteroidetes was significantly lower in the PMDs group. At the genus level, several bacteria were found to be either more or less prevalent in the PMDs group. Specifically, the PMDs group had lower levels of beneficial bacteria such as Butyricicoccus, Megasphaera, and Parabacteroides, while Anaerotaenia was more prevalent in this group. However, after applying false discovery rate correction, these differences were no longer statistically significant. The abundance of certain microbes, such as Anaerotaenia, correlated positively with the severity of PMS as measured by the Premenstrual Symptoms Questionnaire (PSQ), while Parabacteroides and Megasphaera were negatively associated with symptom severity.

These findings suggest that the gut microbiota may play a role in the pathophysiology of premenstrual disorders, potentially influencing the severity of symptoms. However, the study also noted that there were no significant differences in inflammatory markers (such as CRP, LBP, and sCD14) between the PMDs and control groups, which means that the microbiota’s role might not be directly related to inflammation but may involve other mechanisms such as gut-brain communication.

What are the greatest implications of this study?

This study provides preliminary evidence that the gut microbiota could be linked to the severity of premenstrual symptoms, suggesting that microbiome-based interventions might be a potential therapeutic strategy for managing PMS and PMDs. However, due to the cross-sectional nature of the study, it is not possible to establish causality between microbiota characteristics and the severity of premenstrual symptoms. The study's findings highlight the need for future longitudinal studies to confirm these associations and determine the potential therapeutic role of gut microbiota modulation. Furthermore, the identification of specific microbial imbalances could help develop microbiome-based biomarkers for the diagnosis and treatment of PMS and PMDs, providing a more personalized approach to care.

Epidemiology of Premenstrual Syndrome (PMS)-A Systematic Review and Meta-Analysis Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This systematic review and meta-analysis estimate the global prevalence of PMS, revealing wide variations across countries and highlighting the need for standardized diagnostic approaches.

What was reviewed?

This paper presents a systematic review and meta-analysis of the global prevalence of Premenstrual Syndrome (PMS). It focuses on synthesizing existing studies to determine the overall rate of PMS occurrence among women, analyzing factors influencing its prevalence, and exploring variations in prevalence rates across different countries and regions. The review compiles data from multiple sources to estimate the global burden of PMS and identify trends over time, employing meta-regression to examine factors that might affect PMS prevalence.

Who was reviewed?

The review examines data from studies involving women of reproductive age, specifically those diagnosed with PMS based on various symptom screening tools, such as the Premenstrual Symptoms Screening Tool (PSST), and other diagnostic scales. The included studies span different regions and countries, offering a broad view of PMS prevalence across diverse populations.

What were the most important findings?

The systematic review and meta-analysis found that the pooled prevalence of PMS across 17 studies was 47.8%, with substantial variation between different countries. The lowest prevalence was reported in France (12%), while Iran had the highest (98%). This wide range of prevalence is indicative of various factors, such as different diagnostic criteria, sample populations, and cultural or environmental influences on PMS reporting and diagnosis. The review also highlighted a trend of increasing PMS prevalence between 1996 and 2011, though the correlation with the year of study was not statistically significant.

Meta-regression analysis revealed a significant correlation between the sample size and the reported prevalence of PMS, with larger studies tending to report lower prevalence rates. The review also noted that the differences in measurement tools used to diagnose PMS across studies could contribute to the observed variability in prevalence. The results underscore the need for standardized diagnostic criteria and more comprehensive studies to better understand the factors driving PMS prevalence globally.

What are the greatest implications of this review?

The findings of this review have significant implications for public health and clinical practice. The high global prevalence of PMS, with nearly half of reproductive-aged women affected, underscores the need for effective diagnostic and management strategies. Clinicians should be aware of the significant variation in PMS prevalence, influenced by geographical and methodological factors, which can impact patient care and treatment approaches. The review emphasizes the importance of further research to standardize diagnostic tools and explore the role of environmental, cultural, and genetic factors in PMS. Moreover, the findings suggest that larger, high-quality studies are needed to provide more reliable data on PMS prevalence, which can inform public health policies and interventions aimed at improving women's reproductive health globally.

Premenstrual Syndrome and Exercise: A Narrative Review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This narrative review explores how exercise can help manage the symptoms of Premenstrual Syndrome (PMS), focusing on the physiological and psychological benefits of regular physical activity.

What was reviewed?

This narrative review examines the connection between Premenstrual Syndrome (PMS) and exercise. It explores the potential role of exercise in mitigating PMS symptoms and evaluates existing studies to understand how physical activity might alleviate both the physical and psychological impacts of PMS. The review also critiques the methodology of current research and emphasizes the need for further studies to refine exercise interventions for PMS management.

Who was reviewed?

The review targets women who experience PMS, particularly those suffering from physical, psychological, and behavioral symptoms that impact their quality of life. The study highlights the importance of understanding how exercise may serve as a complementary treatment for PMS. The aim is to improve the health outcomes and daily functioning of affected women.

What were the most important findings?

The review identifies key findings regarding the positive effects of exercise on PMS symptoms. PMS symptoms, which include fatigue, mood swings, bloating, and irritability, significantly impair women’s daily activities. The review suggests that exercise, especially aerobic and resistance exercises, can reduce these symptoms, improving both physical and psychological well-being. Regular physical activity helps decrease fatigue, relieve pain (including breast tenderness), and improve mood by regulating hormonal fluctuations. Studies reviewed show that exercise enhances estrogen and progesterone levels and promotes endorphin release, which further alleviates pain and stress.

Exercise improves overall well-being by stimulating the release of neurochemicals like endorphins, which play a crucial role in mood enhancement and pain reduction. Furthermore, exercise reduces the impact of PMS on daily life, including work performance and social engagement. However, the review highlights the inconsistency across studies regarding the optimal exercise prescription for PMS. It calls for more detailed research to determine the best exercise duration, intensity, and frequency needed to achieve maximum benefit.

What are the greatest implications of this review?

This review suggests that exercise is a cost-effective, accessible, and powerful non-pharmacological approach to managing PMS. It encourages clinicians to incorporate exercise recommendations into treatment plans, considering the physical and psychological benefits it offers. Regular physical activity can serve as a complementary treatment alongside pharmacological options, especially for women seeking a holistic management approach. The review also stresses the importance of personalized exercise prescriptions tailored to individual needs and symptom profiles. As research on exercise and PMS continues to evolve, clearer guidelines will emerge, allowing healthcare providers to better support women with PMS through structured exercise programs.

Premenstrual Syndrome and Premenstrual Dysphoric Disorder as Centrally Based Disorders

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

PMS and PMDD stem from neuroendocrine and neurochemical imbalances, especially altered allopregnanolone and GABA activity. Hormonal and neuroactive therapies improve symptoms, while microbiome and neuroinflammation represent promising research areas.

What was studied?

This review examined the neuroendocrine and neurobiological mechanisms underlying Premenstrual Syndrome (PMS) and Premenstrual Dysphoric Disorder (PMDD), emphasizing their classification as centrally based disorders influenced by hormonal fluctuations. It also discussed current and novel therapeutic strategies targeting neuroactive steroids and neuroinflammation in PMS/PMDD.

Who was studied?

The review synthesized findings from clinical, neuroimaging, pharmacological, and molecular studies involving women diagnosed with PMS and PMDD across various reproductive stages, incorporating prospective symptom tracking and biochemical assessments to explore hormone-neurotransmitter interactions and brain sensitivity.

What were the most important findings?

The review highlighted that PMS and PMDD are neuro-hormonal disorders marked by increased central nervous system sensitivity to normal cyclical fluctuations of estrogens and progesterone, especially its metabolite allopregnanolone. This neurosteroid modulates GABA_A receptor activity, affecting mood regulation, and its altered function correlates with emotional and behavioral symptoms in PMDD. Impairments in opioid and serotonergic systems also contribute. Neuroinflammation via GABAergic pathways and elevated pro-inflammatory markers may play a role. Treatment focuses on stabilizing hormones, mainly with combined hormonal contraception, and modulating neuroactive steroids. SSRIs reduce symptoms by affecting serotonin pathways. Novel therapies targeting neurosteroid pathways, including progesterone receptor modulators, 5α-reductase inhibitors, and GABA_A receptor antagonists, show promise. However, treatment responses vary depending on hormonal regimens and individual profiles. Emerging evidence also suggests the gut-brain axis and microbiome influence symptom severity through neuroimmune interactions, though further study is needed.

What are the greatest implications of this study?

This review consolidates the understanding of PMS/PMDD as disorders rooted in neuroendocrine and neurochemical dysregulation, shifting the clinical perspective from purely gynecological or psychiatric frameworks to integrated neurobiological models. It underscores the necessity for personalized therapeutic approaches that combine hormonal regulation with neuroactive agents. The identification of neuroinflammation and microbiome influences opens novel research pathways and potential non-hormonal interventions. Clinicians should consider both established and emerging treatments to optimize symptom control, and researchers must prioritize elucidating the gut-brain interactions and refining neurosteroid-targeted therapies for improved patient outcomes.

Premenstrual syndrome, a common but underrated entity: review of the clinical literature

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This review explores premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD), focusing on symptoms, prevalence, risk factors, and treatment options.

What was reviewed?

This paper is a review of the clinical literature concerning premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD). It focuses on their symptoms, prevalence, risk factors, etiology, and current diagnostic criteria. The review examines treatment options ranging from lifestyle changes to pharmacological interventions, and it highlights the impact of PMS and PMDD on women's health.

Who was reviewed?

The review focuses on the clinical characteristics and findings related to PMS and PMDD in women of reproductive age. It draws from a wide range of studies to summarize the current understanding of these conditions, examining both epidemiological data and clinical treatments.

What were the most important findings?

The review identifies key characteristics of PMS and PMDD, noting that these disorders manifest during the luteal phase of the menstrual cycle and subside with menstruation. It was found that PMS affects a significant portion of the female population, with the prevalence ranging from 10% to 98%, while PMDD affects 2-8% of women. Symptoms can be physical, such as bloating and breast tenderness, or psychological, including mood swings, irritability, and anxiety. The pathogenesis of PMS and PMDD is linked to hormonal fluctuations, particularly estrogen and progesterone, and the interaction of these hormones with central neurotransmitter systems, notably serotonin, GABA, and beta-endorphins.

The review highlights serotonin’s role in the pathogenesis, with women experiencing PMS showing lower serotonin levels in various bodily fluids. Although the exact cause remains unclear, studies suggest that serotonin may be the key mediator of the mood symptoms seen in these disorders. The review also addresses the controversial role of vitamins and minerals in the treatment of PMS, with limited evidence supporting their efficacy over a placebo. Furthermore, it outlines various therapeutic approaches, such as selective serotonin reuptake inhibitors (SSRIs), combined oral contraceptives (COCs), and lifestyle changes, which have been shown to improve symptoms in many patients.

What are the greatest implications of this review?

This review underscores the importance of recognizing PMS and PMDD as significant health issues that can affect a woman’s quality of life. It suggests that, despite their prevalence, these disorders are often underdiagnosed. The review calls for better recognition and diagnosis, particularly using prospective symptom tracking over multiple cycles, as is recommended by the DSM-5 for diagnosing PMDD. Clinicians should consider both pharmacological and non-pharmacological treatments based on the severity of symptoms, as well as individualized care strategies, including SSRIs and COCs for more severe cases. Furthermore, the review suggests that future research should focus on understanding the complex hormonal and neurotransmitter interactions that underpin PMS and PMDD, potentially offering new avenues for treatment development.

Premenstrual syndrome: New insights into etiology and review of treatment methods

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

This review examines the latest insights into the etiology and treatment of Premenstrual Syndrome (PMS) and Premenstrual Dysphoric Disorder (PMDD), focusing on hormonal and neurosteroid imbalances.

What was reviewed?

This paper is a review of the etiology and treatment options for Premenstrual Syndrome (PMS) and Premenstrual Dysphoric Disorder (PMDD). It examines the underlying causes of these conditions, including hormonal fluctuations, neurotransmitter imbalances, and the role of neurosteroids like allopregnanolone. The review also analyzes a variety of treatment options, including pharmacological methods (SSRIs, hormonal therapies, neurosteroid treatments) and non-pharmacological interventions, to provide a comprehensive overview of how PMS and PMDD can be managed.

Who was reviewed?

The review focuses on women of reproductive age who experience PMS and PMDD, with special attention to those who suffer from severe symptoms that significantly impair their daily functioning. The article explores clinical data and findings from various studies to provide a thorough understanding of the disorder's impact on women's health, as well as the varying responses to treatment.

What were the most important findings?

The review highlights several key findings about the etiology and treatment of PMS and PMDD. One of the most significant insights is the role of hormonal fluctuations, particularly the progesterone metabolite allopregnanolone, in the onset of PMS symptoms. It is noted that this metabolite modulates the GABA-A receptor in the central nervous system (CNS), which may explain some of the mood and anxiety-related symptoms of PMS. The review also underscores the complexity of the disorder, as it involves multiple physiological systems, including the hypothalamic-pituitary-adrenal (HPA) axis and neurotransmitter pathways such as serotonin.

SSRIs, commonly used to manage mood symptoms, provide rapid relief and are considered the first-line pharmacological treatment for severe cases. Hormonal therapies, particularly those that stabilize estrogen and progesterone levels, are also effective but require careful selection to avoid exacerbating symptoms. The review suggests that therapies targeting neurosteroids like allopregnanolone may offer new avenues for treatment, although more research is needed. In terms of treatment strategies, the review emphasizes the importance of personalized care, where treatments are tailored to individual symptoms and underlying mechanisms. For instance, oral contraceptives containing drospirenone and ethinylestradiol are effective for controlling physical symptoms, while SSRIs are more beneficial for psychological symptoms.

What are the greatest implications of this review?

The greatest implication of this review is the need for a more nuanced approach to diagnosing and treating PMS and PMDD. By understanding the complex hormonal and neurochemical interactions involved, clinicians can better tailor treatments to individual patients. The review suggests that effective treatment goes beyond symptom alleviation to address the root causes of the disorders. Additionally, the paper points to the importance of considering non-pharmacological interventions alongside medications, particularly for patients who experience mild to moderate symptoms. Future research into the role of neurosteroids and their modulation in the CNS could lead to more targeted treatments with fewer side effects.

Tobacco consumption and premenstrual syndrome: A case-control study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This study highlights the significant link between tobacco consumption and the increased risk of premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD). Women who smoke are at higher odds of developing these disorders, with a dose-response relationship.

What was studied?

The study aimed to assess the relationship between tobacco smoking and premenstrual syndrome (PMS), including its more severe form, premenstrual dysphoric disorder (PMDD). It focused on understanding how smoking may contribute to the occurrence of these menstrual disorders. The study utilized a case-control design, comparing women with PMS and PMDD to age-matched controls.

Who was studied?

The study population consisted of women diagnosed with PMS, women diagnosed with PMDD, and control groups who did not have PMS or PMDD. Participants were recruited from three major public hospitals and one family counseling center in Santiago de Compostela, Spain, ensuring a sample that included women of various age groups and social backgrounds. All participants completed a self-administered questionnaire that gathered information on their smoking habits, socio-demographic factors, and menstrual health.

What were the most important findings?

The study found that tobacco consumption is significantly associated with both PMS and PMDD. Specifically, current smokers had a higher likelihood of experiencing PMS and PMDD compared to non-smokers. This association was evident even among ex-smokers, although the odds ratio was less precise due to the small sample size of this group. The findings also indicated a dose-response relationship: women who smoked more than 15 cigarettes per day or those with higher pack-years of tobacco consumption had an increased risk of developing PMS and PMDD, suggesting a potential cumulative effect of tobacco exposure. The results were confirmed by a cubic spline model, which further demonstrated a correlation between the amount of tobacco consumed and the likelihood of developing these menstrual disorders.

What are the greatest implications of this study?

The study’s findings suggest that tobacco consumption may be a modifiable risk factor for the development of PMS and PMDD. Given the high prevalence of tobacco use among women globally, particularly in Europe and the United States, these results highlight the need for public health strategies aimed at reducing smoking in women, particularly those in their reproductive years. Health professionals should be aware of the potential link between smoking and these menstrual disorders, as this could inform both prevention and treatment strategies. This study calls for further research to explore the long-term effects of smoking cessation and whether reducing tobacco exposure could lower the risk of developing PMS or PMDD in susceptible women.

Nickel

Go to Category Page

Adverse Events Due to Suspected Nickel Hypersensitivity in Patients with Essure Micro-Inserts

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

The study reviewed adverse events linked to nickel hypersensitivity in Essure implants, noting rare reactions and the role of the menstrual cycle in modulating hypersensitivity.

What was studied?

This study aimed to review adverse events associated with suspected nickel hypersensitivity in patients implanted with Essure micro-inserts, a device used for hysteroscopic sterilization. The researchers specifically sought to determine the correlation between reported symptoms and positive results of nickel patch testing, providing insights into the relevance of nickel sensitivity in clinical outcomes. The role of the menstrual cycle in modulating nickel hypersensitivity was also examined to better understand potential confounding factors.

Who was studied?

The study utilized data from adverse event reports collected between 2001 and July 2010, including 63 patients with suspected nickel hypersensitivity. These data were drawn from the MAUDE database, direct manufacturer reports, and published clinical trials involving 650 patients. Patch testing, performed at the discretion of treating physicians, was reported for 20 patients, with 13 testing positive and 7 testing negative.

What were the most important findings?

The study found that the incidence of adverse events potentially related to nickel hypersensitivity in Essure users was exceptionally low, at 0.01%. Among the 13 patients with positive nickel patch tests, symptom resolution occurred in only 4 cases after device removal, with symptoms such as rash, itching, and asthma attributed to nickel allergy. However, two cases demonstrated unresolved symptoms, and the remaining showed no definitive link to nickel hypersensitivity. For the 7 patients with negative patch tests, none of their symptoms were deemed related to the implants. Notably, nickel ion release from Essure devices was minimal, with leaching rates 2,143 times lower than daily dietary nickel intake.

The findings highlight inconsistencies between patch test results and clinical symptoms, questioning the predictive reliability of these tests for implant-related nickel hypersensitivity. The report underscores that nickel-sensitive individuals did not universally experience symptoms, and adverse reactions were rare compared to the prevalence of nickel allergy in the general population.

What are the greatest implications of this study?

The findings suggest that nickel hypersensitivity, as determined by patch testing, is not a clinically significant contraindication for Essure device placement. Despite the presence of nickel in these implants, adverse reactions were rare, and many suspected symptoms lacked a clear link to nickel sensitivity. The study emphasizes the importance of careful evaluation before attributing symptoms to nickel hypersensitivity and suggests that device removal should be reserved for confirmed cases. The data also support the continued use of nickel-containing implants with appropriate monitoring, providing reassurance to clinicians and patients about their safety.

Nickel Allergy is Found in a Majority of Women with Chronic Fatigue Syndrome and Muscle Pain—and may be Triggered by Cigarette Smoke and Dietary Nickel Intake

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

Nickel allergy, smoking, and dietary nickel intake may worsen chronic fatigue and muscle pain. Managing exposure can improve symptoms.

What was studied?

This study investigated the relationship between nickel allergy, cigarette smoking, and dietary nickel intake in women diagnosed with chronic fatigue syndrome (CFS) and muscle pain. The primary aim was to evaluate the prevalence of nickel allergy in this population and explore how smoking and dietary nickel may trigger or exacerbate symptoms.

Who was studied?

The study involved 204 women aged 21 to 73 years with chronic fatigue and muscle pain, meeting the criteria for fibromyalgia and chronic fatigue syndrome but with no signs of autoimmune disorders. The participants underwent immune stimulation therapy using a Staphylococcus vaccine for six months, and their nickel allergy history, smoking habits, and treatment responses were analyzed.

What were the most important findings?

The study found that 52% of the women had a history suggestive of nickel allergy, and 28% were habitual smokers. Nickel allergy and smoking significantly influenced treatment outcomes, with non-allergic, non-smoking participants showing the highest treatment success rates (39%), compared to only 6% in allergic smokers. Additionally, two case reports highlighted the impact of dietary and smoking changes: one participant improved after quitting smoking and reducing dietary nickel intake, while another experienced sustained symptom relief by following a low-nickel diet. Notably, nickel hypersensitivity was associated with increased fatigue and muscle pain symptoms triggered by dietary nickel or cigarette smoke, both of which contain trace amounts of the metal. The findings indicate that systemic nickel allergy, potentially exacerbated by smoking or high dietary nickel, may contribute to chronic fatigue and muscle pain.

What are the greatest implications of this study?

This research emphasizes the importance of recognizing nickel allergy as a potential factor in chronic fatigue syndrome and muscle pain. The interplay between nickel hypersensitivity, dietary nickel intake, and smoking could have significant clinical implications. Managing nickel exposure through dietary adjustments such as a low-nickel diet and smoking cessation may serve as a non-invasive strategy to alleviate symptoms in affected patients. Furthermore, the study underscores the need for broader awareness and diagnostic consideration of systemic nickel allergy in chronic fatigue-related conditions, particularly in women.

Nickel Sensitivity and Symptom Management in Endometriosis: The Role of a Low-Nickel Diet

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Low‑Nickel Diet (LNiD)
    Low‑Nickel Diet (LNiD)

    A low-nickel diet (LNiD) is a therapeutic dietary intervention that eliminates high-nickel foods, primarily plant-based sources such as legumes, nuts, whole grains, and cocoa, to reduce systemic nickel exposure. It is clinically validated for managing systemic nickel allergy syndrome (SNAS) and nickel-induced eczema. Its relevance is well-established in microbiome modulation, with studies demonstrating clinical benefits in conditions such as endometriosis, fibromyalgia, irritable bowel syndrome, and GERD.

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

This study offers new insights into the potential link between nickel sensitivity and symptom severity in endometriosis, suggesting that a low-nickel diet may be a promising intervention for alleviating associated gastrointestinal and gynecological symptoms.

What Was Studied?

This pilot study investigated the prevalence of nickel (Ni) allergic contact mucositis (ACM) in women with endometriosis who experience gastrointestinal symptoms and evaluated the effects of a low-nickel diet on these symptoms. The study focused on assessing the gastrointestinal, extra-intestinal, and gynecological symptom reductions associated with Ni ACM and dietary interventions.

Who Was Studied?

The study enrolled 84 women of reproductive age diagnosed with endometriosis who reported significant gastrointestinal symptoms. Thirty-one participants completed the study, undergoing a diagnostic nickel oral mucosa patch test (omPT) and a subsequent three-month low-nickel diet intervention. Participants were evaluated using symptom questionnaires both at baseline and after dietary changes.

What Were the Most Important Findings?

The study found that 90.3% of participants tested positive for Ni ACM, suggesting a high prevalence of nickel sensitivity among women with endometriosis. Following three months of adhering to a low-nickel diet, significant reductions in all evaluated symptoms were reported. Gastrointestinal symptoms such as abdominal pain, bloating, and diarrhea showed marked improvement. Extra-intestinal symptoms, including fatigue and headaches, and gynecological symptoms such as pelvic pain and dysmenorrhea, also exhibited statistically significant decreases. These findings indicate that nickel sensitivity may contribute to the symptomatic burden of endometriosis, and dietary interventions targeting nickel can alleviate these issues.

The study suggests a potential mechanistic link between nickel exposure, immune responses, and the exacerbation of endometriosis symptoms. Major microbial associations (MMAs) relevant to this context include those influenced by dietary changes, although specific microbiome alterations were not detailed.

What Are the Greatest Implications of This Study?

This research highlights nickel sensitivity as a significant yet previously under-recognized contributor to gastrointestinal and systemic symptoms in endometriosis patients. The findings suggest that incorporating nickel sensitivity screening and low-nickel dietary recommendations could represent a transformative approach to symptom management in endometriosis. Although the sample size was small, the results offer strong preliminary evidence for revising dietary protocols in clinical practice to include low-nickel guidelines, potentially improving the quality of life for patients.

The influence of nickel on intestinal microbiota disturbances

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

Excess nickel disrupts gut microbiota, promoting dysbiosis and contributing to conditions like obesity and systemic nickel allergy syndrome (SNAS). Probiotics and nickel-restricted diets show promise in mitigating these effects, underscoring the need for further research and clinical intervention.

What was reviewed?

The paper reviewed the influence of nickel on intestinal microbiota disturbances, drawing on 59 scientific publications from the past 20 years. The analysis focused on nickel’s dual role as an essential element for microbial enzymatic reactions and a disruptor of gut microbiota, especially under conditions of excessive exposure or systemic nickel allergy syndrome (SNAS).

Who was reviewed?

The review encompassed research involving humans, animals, and microbial models. Specific attention was given to populations exposed to high levels of nickel, individuals with SNAS, and animal studies demonstrating changes in microbial communities under nickel exposure.

What were the most important findings?

Nickel acts as a cofactor for metalloenzymes like urease, hydrogenase, and [NiFe]-hydrogenase, essential for microbial survival. However, excess nickel promotes dysbiosis, characterized by reductions in beneficial taxa and increases in nickel-resistant bacteria. In humans with SNAS, the microbiota showed decreased levels of beneficial genera such as Bifidobacterium and Lactobacillus, known for their probiotic effects and urease activity, and increases in nickel-tolerant taxa, including Clostridiaceae and Bacillaceae. Similarly, animal studies indicated reduced Verrucomicrobia and Bacteroidetes while promoting Escherichia coli and Enterococcus.

Nickel exposure also leads to an increased abundance of Bacteroides fragilis, Bacteroidales S24-7, and Interstinimonas, with a concurrent decline in Firmicutes, disrupting the Firmicutes-to-Bacteroidetes ratio, a critical marker of gut health. This imbalance contributed to systemic inflammation and altered immune responses. Moreover, nickel-reliant pathogens, such as Helicobacter pylori, which require Ni2+-dependent enzymes like urease for colonization, further highlighted nickel’s role in microbial pathogenicity. Probiotic strains such as Lactobacillus fermentum demonstrated detoxifying effects by metabolizing nickel, suggesting their therapeutic potential.

What are the greatest implications of this review?

The findings reveal that nickel exposure significantly alters gut microbial ecology, driving dysbiosis and systemic inflammation in susceptible populations. The rise of nickel-tolerant taxa, coupled with the decline of protective bacteria, underscores nickel’s role as a disruptor of gut homeostasis, contributing to conditions like obesity and SNAS. Probiotic supplementation, particularly strains capable of nickel detoxification, and dietary restrictions like a low-nickel diet, have shown promise in mitigating these effects. This review highlights the urgent need for dietary nickel regulations and further clinical studies on therapeutic interventions targeting nickel-induced microbial dysbiosis.

Premenstrual Dysphoric Disorder (PMDD)

Go to Category Page

Allopregnanolone in premenstrual dysphoric disorder (PMDD)

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This review identifies impaired sensitivity of GABA-A receptors to the neurosteroid allopregnanolone as central to PMDD, linking receptor plasticity and stress dysregulation to mood symptoms, and highlights promising treatments targeting this pathway.

What was studied?

This review comprehensively examined the role of the neuroactive steroid allopregnanolone (ALLO), a potent positive allosteric modulator of the GABA-A receptor (GABAA-R), in the pathophysiology of premenstrual dysphoric disorder (PMDD). It focused on the evidence supporting altered sensitivity or dysregulation of GABAA-Rs in response to ALLO fluctuations across the menstrual cycle, linking these neurobiological changes to the characteristic mood symptoms and stress sensitivity of PMDD.

Who was studied?

As a review article, this paper synthesized findings from both human clinical studies and animal models, particularly rodents, to elucidate mechanisms underlying PMDD. Human studies included neuroendocrine and neurophysiological investigations of women diagnosed with PMDD compared to controls, focusing on hormonal dynamics, receptor sensitivity, stress response, and symptomatology. Rodent models primarily involved progesterone or ALLO withdrawal paradigms to mimic PMDD symptoms and investigate GABAA-R subunit changes and behavior.

What were the most important findings?

The review highlighted that PMDD is not caused by abnormal circulating hormone levels but rather by impaired CNS sensitivity to normal fluctuations of ALLO. In rodent models, rapid withdrawal from progesterone or ALLO induces anxiety- and depression-like behaviors linked to upregulation of the GABAA-R α4 subunit, implicating receptor plasticity in symptom manifestation. Clinical studies in women with PMDD demonstrated altered GABAA-R function, such as lack of ALLO-induced sedation during the luteal phase and elevated anxiety-potentiated startle responses, indicating dysfunctional receptor adaptation to hormonal changes. The review also detailed how ALLO modulates the hypothalamic-pituitary-adrenal (HPA) axis, with women with PMDD showing altered stress responsivity likely due to impaired ALLO-GABAA-R interaction, leading to heightened stress sensitivity during the luteal phase. Importantly, treatments effective in PMDD, including selective serotonin reuptake inhibitors (SSRIs) and novel GABA-modulating drugs appear to normalize ALLO-GABA signaling, further supporting this pathophysiological model.

What are the greatest implications of this study?

This review consolidates strong evidence that PMDD is fundamentally a disorder of impaired neurosteroid modulation of GABAA-Rs, rather than hormone level abnormalities alone, positioning GABAA-R plasticity and ALLO sensitivity as central to its pathophysiology. Understanding this mechanism clarifies why PMDD symptoms cyclically align with hormonal fluctuations and why patients experience heightened stress sensitivity. Clinically, this suggests that future therapeutic strategies should target the neurosteroid-GABAergic system directly to restore receptor function or stabilize neurosteroid levels, promising more rapid and effective symptom relief than traditional antidepressants. Moreover, this framework encourages the development and testing of novel GABAergic agents tailored to PMDD and related reproductive mood disorders, enhancing personalized medicine for affected women worldwide.

Biological rhythms in premenstrual syndrome and premenstrual dysphoric disorder: a systematic review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Women with PMS/PMDD experience disrupted biological rhythms, notably lower melatonin, higher nighttime temperature, and poor sleep quality. These circadian disturbances contribute to symptom severity, suggesting chronobiological targets for improved diagnosis and treatment.

What was reviewed?

This paper systematically reviewed the existing literature on biological rhythm disruptions in women with premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD). The focus was on circadian and other biological rhythms, including sleep–wake cycles, melatonin secretion, core body temperature, cortisol, prolactin, and thyroid-stimulating hormone levels. The review synthesized findings from 25 studies that compared women diagnosed with PMS/PMDD to healthy controls, assessing both subjective and objective markers of biological rhythms to clarify their association with premenstrual symptoms and the underlying pathophysiology.

Who was reviewed?

The review analyzed studies published between 1989 and 2022 across multiple countries, involving women aged 18 to 45 diagnosed with PMS or PMDD using standardized criteria, mostly DSM-III-R to DSM-5. The total sample sizes varied widely, with some studies including over 600 participants. Healthy control groups consisted of women without PMS/PMDD or psychiatric disorders. Studies included diverse methodologies such as polysomnography, actimetry, hormonal assays, core body temperature measurements, and subjective sleep quality assessments, enabling comprehensive evaluation of biological rhythms in the premenstrual context.

What were the most important findings?

The review found consistent evidence that women with PMS/PMDD exhibit significant disruptions in biological rhythms compared to healthy controls. Notably, they present with lower nocturnal melatonin levels, elevated nighttime core body temperature, and poorer subjective sleep quality, all indicating altered circadian regulation. While objective sleep parameters and activity rhythms showed mixed or nonsignificant differences, hormonal rhythms such as cortisol and prolactin demonstrated inconsistent findings across studies. These rhythm disturbances likely contribute to the psychological and physiological symptoms experienced during the luteal phase. The review highlights melatonin dysregulation as a potential key factor in PMS/PMDD pathophysiology and calls for further research into circadian-based mechanisms and their therapeutic implications.

What are the greatest implications of this review?

This review highlights the importance of biological rhythm disruptions in PMS and PMDD, positioning circadian dysfunction, especially melatonin alterations, as a promising target for understanding symptom development and designing novel interventions. Clinicians should recognize that sleep complaints and temperature regulation abnormalities in these disorders reflect deeper circadian disturbances rather than isolated symptoms. The review advocates for integrating chronobiological assessments into clinical evaluations and exploring circadian-modulating treatments, such as light therapy or melatonin supplementation, to improve patient outcomes. It also calls for future research to clarify inconsistent findings in hormonal rhythms and to investigate the potential of personalized circadian therapies tailored to premenstrual symptom profiles.

Characteristics of the gut microbiota in women with premenstrual symptoms: A cross-sectional study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This study identified specific gut microbiota alterations, including reduced butyrate- and GABA-producing bacteria, associated with premenstrual disorder severity, offering new insights into PMD pathophysiology and potential microbiome-targeted treatments.

What was studied?

This cross-sectional pilot study examined the gut microbiota characteristics in women experiencing premenstrual disorders (PMDs) compared to healthy controls, aiming to uncover microbial associations with the severity of premenstrual symptoms and to evaluate inflammatory markers indicative of bacterial translocation.

Who was studied?

The study involved 43 Japanese women aged 20 to 45 years, with 21 women experiencing PMDs severe enough to disrupt social functioning and 22 controls without significant premenstrual symptoms, all selected to exclude confounding factors like recent medication use, neuropsychiatric disorders, and gastrointestinal diseases.

What were the most important findings?

The study revealed that although overall gut microbial diversity did not differ significantly, women with PMDs exhibited lower levels of the Bacteroidetes phylum and reduced abundance of butyrate-producing genera such as Butyricicoccus and Megasphaera, alongside decreased Parabacteroides, a GABA-related genus, while Anaerotaenia was elevated; these microbial shifts correlated with symptom severity, but inflammatory markers linked to endotoxemia showed no group differences, suggesting unique microbiome alterations in PMDs distinct from major depressive disorder.

What are the greatest implications of this study?

These findings suggest that specific gut microbiota alterations, particularly reductions in beneficial butyrate- and GABA-producing bacteria, may underlie premenstrual symptom severity through the gut-brain axis, highlighting potential microbiome-based biomarkers and therapeutic targets for PMDs, and warranting further longitudinal and intervention research to establish causality and clinical applications.

Determinants of premenstrual dysphoric disorder and associated factors among regular undergraduate students at Hawassa University Southern, Ethiopia

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Among Ethiopian female university students, PMDD affects nearly two-thirds and links strongly to severe menstrual pain, irregular cycles, low social support, and contraceptive use. Early identification and comprehensive care are essential to alleviate its academic and psychological impacts.

What was studied?

This institutional-based cross-sectional study investigated the prevalence and determinants of premenstrual dysphoric disorder (PMDD) among regular undergraduate female students at Hawassa University, Ethiopia. It aimed to quantify PMDD prevalence using a standardized premenstrual symptoms screening tool and to identify socio-demographic, clinical, psychological, gynecological, and behavioral factors associated with PMDD in this population.

Who was studied?

The study included 374 regular female undergraduate students aged 18 and above from the College of Medicine and Health Sciences at Hawassa University. Participants were selected using stratified random sampling and completed a self-administered questionnaire assessing premenstrual symptoms, menstrual characteristics, social support, contraceptive use, and behavioral factors. Students with illnesses or absent during data collection were excluded.

What were the most important findings?

The study found a high PMDD prevalence of approximately 63% among participants, significantly higher than many global estimates. Key factors independently associated with PMDD included severe menstrual pain, irregular menstrual cycles, poor or moderate social support, and contraceptive use. Severe dysmenorrhea exacerbated emotional and behavioral symptoms, while social support appeared protective. These findings indicate that PMDD substantially impairs daily functioning, including academic performance, and is influenced by a complex interplay of physiological, psychological, and social factors. The study emphasizes the importance of early screening and tailored interventions to mitigate PMDD's impact on student well-being and success.

What are the greatest implications of this study?

This study highlights PMDD as a prevalent and underrecognized condition with significant negative effects on young women’s mental health and academic performance in a low-resource setting. It underscores the urgent need for integrating PMDD screening and psychosocial support into university health services, especially focusing on managing menstrual pain, providing social support, and carefully evaluating contraceptive use. These findings advocate for multidisciplinary interventions that address both physical and psychological determinants of PMDD, aiming to improve quality of life and academic outcomes. Moreover, the research supports policymakers and educators in developing targeted health promotion programs and facilitating access to effective treatment for PMDD in similar contexts.

Premenstrual disorders and PMDD - a review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Premenstrual dysphoric disorder (PMDD) significantly impairs women’s lives due to abnormal sensitivity to hormonal fluctuations. Accurate diagnosis relies on prospective symptom tracking. Evidence supports SSRIs and hormonal treatments as effective management strategies. Emerging therapies and multidisciplinary care models promise improved outcomes for this complex and under-recognized disorder.

What was reviewed?

This paper provides a comprehensive review of premenstrual disorders (PMDs), with a specific focus on premenstrual dysphoric disorder (PMDD). It synthesizes current knowledge regarding the definitions, classification, prevalence, diagnosis, etiology, and treatment of PMDs and PMDD. The review covers consensus guidelines from professional bodies such as the International Society for Premenstrual Disorders (ISPMD), diagnostic criteria from DSM-V and ICD-11, and evaluates various therapeutic approaches, including non-pharmacological interventions, pharmacotherapy (particularly SSRIs), hormonal treatments, novel agents targeting neuroactive steroids, and surgical options. It also highlights challenges in diagnosis, the significant impact on quality of life and suicidality risk, and research gaps.

Who was reviewed?

The review critically assesses a wide body of clinical, epidemiological, and mechanistic studies involving women experiencing PMDs and PMDD worldwide. It references population prevalence data, genetic and neurobiological studies, clinical trials evaluating treatments such as SSRIs and combined oral contraceptive pills (COCPs), and guidelines developed by multidisciplinary expert panels. The authors draw upon systematic reviews, randomized controlled trials, observational studies, and consensus statements to present a balanced perspective. The review specifically incorporates data related to symptom measurement tools like the Daily Record of Severity of Problems (DRSP) and discusses patient management strategies applicable in primary and specialist care settings.

What were the most important findings?

The review clarifies that PMDD represents a severe subset of PMDs, affecting approximately 5% of women, and carries significant physical, psychological, and social burdens, including a markedly increased risk of suicide attempts. It emphasizes that PMDD symptoms arise from abnormal sensitivity to normal menstrual hormonal fluctuations, especially allopregnanolone's paradoxical effect on GABA-A receptors, rather than altered hormone levels per se. Genetic factors, serotonergic dysregulation, inflammation, and stress history also contribute to pathophysiology. Accurate diagnosis depends on prospective symptom tracking over at least two menstrual cycles to confirm symptom cyclicity, severity, and functional impact.

Treatment must be multidisciplinary and individualized. SSRIs are the first-line pharmacological treatment, acting rapidly and effectively even when dosed intermittently in the luteal phase or symptom-onset, mitigating side effects associated with continuous dosing. Hormonal treatments, particularly COCPs containing drospirenone, show efficacy, though hormonal sensitivity varies and progestogen intolerance complicates therapy. Emerging therapies targeting allopregnanolone modulation and 5-alpha reductase inhibitors are under investigation but require further evidence. GnRH analogues and surgical oophorectomy remain options for severe refractory cases but carry significant risks, including bone density loss. Non-pharmacological approaches, including cognitive behavioral therapy (CBT), dietary modifications, and supplements (calcium, magnesium, vitamin B6), provide complementary benefits.

What are the greatest implications of this review?

This review bridges the knowledge gap between evolving scientific insights into PMDD's neuroendocrine mechanisms and practical clinical management strategies. It advocates for a precision medicine approach tailored to individual hormonal sensitivities and symptom profiles. By synthesizing current evidence, it empowers clinicians to improve diagnostic accuracy through prospective symptom monitoring, recognize the disorder’s profound impact on mental health, and adopt evidence-based treatments, minimizing side effects. The emphasis on rapid SSRI efficacy and flexible dosing regimens offers clinicians practical tools to enhance adherence and patient quality of life. Moreover, highlighting the multidisciplinary nature of optimal care and emerging pharmacotherapies signals future directions for research and therapeutic innovation.

Premenstrual Dysphoric Disorder and the Brain

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

PMDD involves altered brain activation in prefrontal regions tied to hormone sensitivity. Symptoms appear cyclically with hormonal fluctuations, distinguishing it from other mood disorders. Understanding this brain-hormone interaction aids targeted treatment and improves clinical recognition of PMDD.

What was reviewed?

This paper reviewed the neurological basis of premenstrual dysphoric disorder (PMDD), emphasizing its recognition as a distinct mood disorder linked to menstrual cycle hormonal fluctuations. It summarized advances in brain imaging and neurophysiological studies demonstrating altered brain function in PMDD patients, particularly in prefrontal cortex regions involved in executive function and emotion regulation. The review highlighted the significance of hormone sensitivity, especially to estradiol and progesterone, and how these hormonal changes affect cerebral blood flow and neural activation patterns in women with PMDD compared to controls.

Who was reviewed?

The review focused on women diagnosed with PMDD according to rigorous DSM criteria, including prospective symptom tracking. It integrated findings from neuroimaging studies (fMRI, PET), hormonal manipulation paradigms (gonadotropin-releasing hormone agonist followed by hormone add-back), and psychophysiological assessments conducted on small to moderate cohorts of women with PMDD and matched healthy controls. The studies collectively evaluated brain activation, cerebral blood flow, neurotransmitter activity, and behavioral correlates of hormone-driven mood symptoms.

What were the most important findings?

The review underscored that women with PMDD show abnormal activation in the dorsolateral prefrontal cortex and medial frontal gyrus during cognitive tasks, regardless of hormonal state, suggesting a trait vulnerability. Brain activation differences correlated with symptom severity, especially irritability, which is a hallmark PMDD symptom. The cerebellum also showed heightened activity in PMDD. The disorder’s symptom manifestation requires the fluctuating hormonal environment of the luteal phase, implicating hormone sensitivity as a key pathophysiological factor. Unlike other mood disorders, PMDD’s brain dysfunction is specifically linked to normal hormonal changes rather than baseline abnormalities, explaining the cyclical nature of symptoms.

What are the greatest implications of this review?

This review clarifies that PMDD arises from an interaction between inherent brain vulnerabilities and normal hormonal fluctuations, particularly estradiol and progesterone. It encourages clinicians to view PMDD as a neurobiologically distinct disorder with predictable symptom timing linked to menstrual phases. These insights justify targeted hormonal and neuropharmacological treatments and support ongoing research into brain-based biomarkers and personalized therapies. The findings also highlight the importance of early diagnosis and symptom monitoring to improve patient care and quality of life for affected women.

Premenstrual dysphoric disorder-an undervalued diagnosis? A cross-sectional study in Hungarian women

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Hungarian women with probable PMDD experience high rates of anxio-depressive symptoms and reduced well-being. Retrospective screening suggests higher-than-expected prevalence, highlighting the need for better diagnostic tools and tailored treatment to address this underrecognized disorder.

What was studied?

This cross-sectional study assessed the prevalence of probable premenstrual dysphoric disorder (PMDD) among Hungarian women and examined the relationship between probable PMDD, anxio-depressive symptom severity, and overall well-being. The researchers aimed to validate a retrospective DSM-5-based PMDD screening tool in this population and explore psychological symptom patterns related to PMDD, using standardized questionnaires for depression, anxiety, and well-being.

Who was studied?

The study included 112 women of reproductive age from Hungary with regular menstrual cycles who were not using hormonal contraceptives and had no significant neurological, psychiatric, endocrine, or gynecological disorders. The participants were divided into two groups based on PMDD screening results: a probable PMDD group (n=67) and a non-PMDD group (n=45). They completed validated questionnaires measuring probable PMDD symptoms, anxio-depressive severity, and subjective well-being.

What were the most important findings?

The study revealed a surprisingly high prevalence of probable PMDD at nearly 60%, exceeding international estimates, which the authors attribute partly to retrospective screening limitations and recruitment bias. Women with probable PMDD reported significantly greater depressive and anxiety symptoms and lower well-being than controls, regardless of menstrual cycle phase. Logistic regression confirmed that higher anxiety and depression scores predicted probable PMDD diagnosis. The findings corroborate prior evidence that PMDD involves substantial psychological distress that impacts life quality. Notably, anxiety symptom severity did not vary significantly across cycle phases, suggesting persistent affective symptoms. These results highlight the challenges of accurate PMDD diagnosis, especially given the burden of prospective symptom tracking, and underscore the need for tailored psychological assessment and treatment strategies.

What are the greatest implications of this study?

This study emphasizes that probable PMDD is a prevalent and underrecognized condition that severely affects women's mental health and well-being, even beyond the premenstrual phase. It highlights the utility and limitations of retrospective screening tools in estimating PMDD prevalence and calls for improved diagnostic protocols that balance accuracy with practicality. The findings advocate for personalized mental health support and further research to refine diagnostic tools and treatment approaches tailored to the needs of women with PMDD, especially in underrepresented populations.

Premenstrual Dysphoric Disorder: Epidemiology and Treatment

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This review delineates PMDD’s unique neurobiology, highlighting hormone sensitivity, GABAergic dysfunction, and stress interaction. SSRIs provide rapid symptom relief, with hormonal and behavioral therapies complementing care. It emphasizes precise diagnosis and individualized, multidisciplinary treatment to improve outcomes in this debilitating disorder.

What was reviewed?

This paper presents a thorough review of the epidemiology, pathophysiology, and treatment options for premenstrual dysphoric disorder (PMDD). It examines PMDD’s diagnostic criteria as established by DSM-5, highlighting the importance of mood symptoms and prospective symptom tracking for accurate diagnosis. The review synthesizes current understanding of PMDD’s biological underpinnings, including the role of neurosteroids like allopregnanolone, estrogen’s influence on serotonergic systems, brain-derived neurotrophic factor (BDNF) polymorphisms, and the impact of stress and inflammation. It further explores neuroimaging and psychophysiological findings that differentiate PMDD from other affective disorders. The review then evaluates therapeutic approaches, emphasizing SSRIs as the first-line treatment and discussing intermittent dosing strategies, hormonal therapies, cognitive-behavioral therapy, and alternative treatments.

Who was reviewed?

The authors critically analyzed studies involving women diagnosed with PMDD across community and clinical samples worldwide. The review includes epidemiological data, genetic and neurobiological research, and clinical trials assessing treatment efficacy. It references consensus guidelines from psychiatric and gynecological professional bodies, neuroimaging studies comparing PMDD patients to healthy controls, and meta-analyses evaluating pharmacologic and psychotherapeutic interventions. The paper also integrates findings from animal models of hormone sensitivity and neurosteroid modulation relevant to PMDD pathophysiology.

What were the most important findings?

Women with PMDD do not differ in peripheral hormone levels but show altered GABA_A receptor function and neurosteroid sensitivity, contributing to affective symptoms. Estrogen’s modulation of serotonin receptors and transporters further implicates serotonergic dysregulation in PMDD. Genetic factors such as polymorphisms in estrogen receptor and serotonin transporter genes, as well as BDNF variants, may increase susceptibility. Stress history correlates with PMDD diagnosis and may influence neurosteroid responses and HPA axis regulation. Neuroimaging reveals structural and functional brain differences in areas regulating emotion and cognition, including the amygdala and prefrontal cortex, with altered GABA and glutamate levels detected in PMDD patients.

Regarding treatment, SSRIs demonstrate moderate to large effect sizes in symptom reduction, with rapid onset of action allowing for intermittent or symptom-onset dosing regimens that minimize side effects and improve adherence. Hormonal treatments, particularly combined oral contraceptives containing drospirenone, show some efficacy but with high placebo responses and variable individual tolerance. Cognitive-behavioral therapy provides sustained symptom improvement and complements pharmacotherapy, though combined approaches do not necessarily enhance outcomes beyond monotherapy. Alternative therapies such as calcium supplementation and omega-3 fatty acids offer limited benefits and require further validation.

What are the greatest implications of this review?

This review consolidates the complex neurobiological, genetic, and psychosocial factors contributing to PMDD, underscoring its distinction from other mood disorders and the importance of precision in diagnosis and treatment. It reinforces SSRIs as the cornerstone of pharmacotherapy and advocates for flexible dosing strategies tailored to symptom patterns, enhancing patient quality of life and medication adherence. The emerging understanding of neurosteroid modulation opens promising avenues for novel therapeutics targeting GABAergic pathways. The findings call for multidisciplinary, individualized treatment plans incorporating pharmacological, psychological, and lifestyle interventions. The review highlights gaps in long-term safety data for hormonal therapies and the need for improved diagnostic tools and biomarkers. Overall, it equips clinicians with an evidence-based framework to optimize PMDD management and encourages ongoing research to address unmet clinical needs.

Premenstrual dysphoric disorder: General overview, treatment strategies, and focus on sertraline for symptom-onset dosing

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Symptom-onset dosing of sertraline effectively reduces PMDD symptoms, especially mood-related ones, with fewer side effects and limited drug exposure. This targeted approach offers a promising, patient-friendly alternative to continuous SSRI treatment for premenstrual dysphoric disorder.

What was studied?

This paper studied the efficacy and tolerability of symptom-onset dosing of sertraline, a selective serotonin reuptake inhibitor (SSRI), for the treatment of premenstrual dysphoric disorder (PMDD). The focus was on assessing whether administering sertraline starting at the onset of PMDD symptoms, rather than continuous or luteal-phase dosing, could effectively reduce symptom severity and improve clinical outcomes over six menstrual cycles. The study also explored the side effect profile and discontinuation symptoms associated with this targeted treatment approach.

Who was studied?

The study population comprised 252 women with prospectively confirmed PMDD, aged approximately 34 years on average, predominantly white (around 70%), and without significant medical or psychiatric comorbidities. These participants were randomized into two groups: 125 women received flexible doses of sertraline (50–100 mg/day) beginning at symptom onset and continuing until menstruation began, while 127 women received placebo treatment following the same schedule.

What were the most important findings?

The study demonstrated that symptom-onset treatment with sertraline significantly reduced the severity of PMDD symptoms compared to placebo. Specifically, women treated with sertraline showed statistically significant improvements in depressive symptoms as measured by clinician-rated scales and a significant reduction in the daily record of problem severity, including the anger/irritability subscale. While the reduction in premenstrual tension ratings narrowly missed statistical significance, sertraline outperformed placebo in global improvement ratings and had higher clinical response rates (67% vs. 52%). Noticeably, emission rates were not significantly different. The average duration of sertraline use was only about seven days per menstrual cycle, minimizing exposure to the drug and related side effects. Adverse effects, primarily nausea and insomnia, were more common in the sertraline group, but abrupt discontinuation did not lead to withdrawal symptoms. These findings indicate that targeted, short-term SSRI treatment timed to symptom onset is effective and well tolerated in managing PMDD.

What are the greatest implications of this study?

This research challenges traditional views that antidepressants require continuous administration to be effective in PMDD treatment by demonstrating that symptom-onset dosing with sertraline is both efficacious and has a favorable side effect profile. This approach minimizes medication exposure and associated adverse effects, potentially improving adherence and reducing treatment costs. It provides a practical strategy to manage PMDD symptoms precisely when needed, aligning with the disorder's cyclical nature. Future research is needed to compare symptom-onset dosing directly with luteal-phase and continuous dosing regimens and to explore treatment strategies for non-responders to symptom-onset sertraline. Clinically, this study supports personalized, flexible pharmacotherapy for PMDD, enhancing therapeutic outcomes while mitigating risks.

Premenstrual Syndrome and Premenstrual Dysphoric Disorder as Centrally Based Disorders

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

PMS and PMDD stem from neuroendocrine and neurochemical imbalances, especially altered allopregnanolone and GABA activity. Hormonal and neuroactive therapies improve symptoms, while microbiome and neuroinflammation represent promising research areas.

What was studied?

This review examined the neuroendocrine and neurobiological mechanisms underlying Premenstrual Syndrome (PMS) and Premenstrual Dysphoric Disorder (PMDD), emphasizing their classification as centrally based disorders influenced by hormonal fluctuations. It also discussed current and novel therapeutic strategies targeting neuroactive steroids and neuroinflammation in PMS/PMDD.

Who was studied?

The review synthesized findings from clinical, neuroimaging, pharmacological, and molecular studies involving women diagnosed with PMS and PMDD across various reproductive stages, incorporating prospective symptom tracking and biochemical assessments to explore hormone-neurotransmitter interactions and brain sensitivity.

What were the most important findings?

The review highlighted that PMS and PMDD are neuro-hormonal disorders marked by increased central nervous system sensitivity to normal cyclical fluctuations of estrogens and progesterone, especially its metabolite allopregnanolone. This neurosteroid modulates GABA_A receptor activity, affecting mood regulation, and its altered function correlates with emotional and behavioral symptoms in PMDD. Impairments in opioid and serotonergic systems also contribute. Neuroinflammation via GABAergic pathways and elevated pro-inflammatory markers may play a role. Treatment focuses on stabilizing hormones, mainly with combined hormonal contraception, and modulating neuroactive steroids. SSRIs reduce symptoms by affecting serotonin pathways. Novel therapies targeting neurosteroid pathways, including progesterone receptor modulators, 5α-reductase inhibitors, and GABA_A receptor antagonists, show promise. However, treatment responses vary depending on hormonal regimens and individual profiles. Emerging evidence also suggests the gut-brain axis and microbiome influence symptom severity through neuroimmune interactions, though further study is needed.

What are the greatest implications of this study?

This review consolidates the understanding of PMS/PMDD as disorders rooted in neuroendocrine and neurochemical dysregulation, shifting the clinical perspective from purely gynecological or psychiatric frameworks to integrated neurobiological models. It underscores the necessity for personalized therapeutic approaches that combine hormonal regulation with neuroactive agents. The identification of neuroinflammation and microbiome influences opens novel research pathways and potential non-hormonal interventions. Clinicians should consider both established and emerging treatments to optimize symptom control, and researchers must prioritize elucidating the gut-brain interactions and refining neurosteroid-targeted therapies for improved patient outcomes.

Premenstrual syndrome, a common but underrated entity: review of the clinical literature

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This review explores premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD), focusing on symptoms, prevalence, risk factors, and treatment options.

What was reviewed?

This paper is a review of the clinical literature concerning premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD). It focuses on their symptoms, prevalence, risk factors, etiology, and current diagnostic criteria. The review examines treatment options ranging from lifestyle changes to pharmacological interventions, and it highlights the impact of PMS and PMDD on women's health.

Who was reviewed?

The review focuses on the clinical characteristics and findings related to PMS and PMDD in women of reproductive age. It draws from a wide range of studies to summarize the current understanding of these conditions, examining both epidemiological data and clinical treatments.

What were the most important findings?

The review identifies key characteristics of PMS and PMDD, noting that these disorders manifest during the luteal phase of the menstrual cycle and subside with menstruation. It was found that PMS affects a significant portion of the female population, with the prevalence ranging from 10% to 98%, while PMDD affects 2-8% of women. Symptoms can be physical, such as bloating and breast tenderness, or psychological, including mood swings, irritability, and anxiety. The pathogenesis of PMS and PMDD is linked to hormonal fluctuations, particularly estrogen and progesterone, and the interaction of these hormones with central neurotransmitter systems, notably serotonin, GABA, and beta-endorphins.

The review highlights serotonin’s role in the pathogenesis, with women experiencing PMS showing lower serotonin levels in various bodily fluids. Although the exact cause remains unclear, studies suggest that serotonin may be the key mediator of the mood symptoms seen in these disorders. The review also addresses the controversial role of vitamins and minerals in the treatment of PMS, with limited evidence supporting their efficacy over a placebo. Furthermore, it outlines various therapeutic approaches, such as selective serotonin reuptake inhibitors (SSRIs), combined oral contraceptives (COCs), and lifestyle changes, which have been shown to improve symptoms in many patients.

What are the greatest implications of this review?

This review underscores the importance of recognizing PMS and PMDD as significant health issues that can affect a woman’s quality of life. It suggests that, despite their prevalence, these disorders are often underdiagnosed. The review calls for better recognition and diagnosis, particularly using prospective symptom tracking over multiple cycles, as is recommended by the DSM-5 for diagnosing PMDD. Clinicians should consider both pharmacological and non-pharmacological treatments based on the severity of symptoms, as well as individualized care strategies, including SSRIs and COCs for more severe cases. Furthermore, the review suggests that future research should focus on understanding the complex hormonal and neurotransmitter interactions that underpin PMS and PMDD, potentially offering new avenues for treatment development.

Prevalence and associated factors of premenstrual dysphoric disorder among high school students in Finote Selam town, northwest Ethiopia

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

Among Ethiopian high school girls, PMDD affects one-third, linked to irregular cycles, depression, prolonged menstruation, and stress. Early screening and mental health support are crucial to improving well-being and academic success.

What was studied?

This cross-sectional study examined the prevalence of premenstrual dysphoric disorder (PMDD) and its associated factors among high school female students in Finote Selam town, northwest Ethiopia. Using DSM-5 criteria and self-administered questionnaires, the study aimed to quantify PMDD prevalence and identify clinical, psychosocial, and menstrual-related predictors affecting this population's mental health and academic performance.

Who was studied?

The research included 548 high school female students aged 15 to 22 years with regular menstrual cycles, excluding those with serious illness or recent school transfers. Participants completed validated questionnaires assessing PMDD symptoms, depression, perceived stress, social support, menstrual characteristics, and behavioral factors such as substance use.

What were the most important findings?

The study found a high PMDD prevalence (33%), with physical symptoms like breast tenderness and fatigue being most common. Key factors significantly associated with PMDD included irregular menstrual cycles, depressive symptoms, longer menstruation duration, and high perceived stress. PMDD significantly impacted academic performance, social functioning, and psychological well-being. The findings aligned with prior Ethiopian and African studies but were higher than reports from developed countries, possibly reflecting sociocultural, infrastructural, and menstrual hygiene differences influencing symptom expression and health-seeking behaviors.

What are the greatest implications of this study?

This study highlights PMDD as a prevalent and underrecognized condition adversely affecting adolescent females' mental health and educational outcomes in low-resource settings. It emphasizes the urgent need for early screening, stress reduction interventions, and targeted mental health support within primary healthcare and school systems. Addressing menstrual health education, improving hygiene management, and mitigating psychosocial stressors could reduce PMDD burden and improve quality of life. These insights guide clinicians and policymakers toward culturally sensitive, accessible strategies for PMDD diagnosis and management in similar populations.

Tobacco consumption and premenstrual syndrome: A case-control study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Premenstrual Syndrome (PMS)
    Premenstrual Syndrome (PMS)

    Premenstrual Syndrome (PMS) involves physical and emotional symptoms linked to hormonal fluctuations. Recent research highlights the role of heavy metals and gut microbiome imbalances in worsening these symptoms. Lifestyle changes, microbiome-targeted therapies, and toxin reduction show promise in effective PMS management.

  • Premenstrual Dysphoric Disorder (PMDD)
    Premenstrual Dysphoric Disorder (PMDD)

    OverviewPremenstrual Dysphoric Disorder (PMDD) affects roughly 3–9% of women of reproductive age and manifests as severe mood, behavioral, and physical symptoms tightly linked to the luteal phase of the menstrual cycle, distinguishing it from milder premenstrual syndrome (PMS).[1][2] Central to PMDD’s pathophysiology is an altered sensitivity of the central nervous system to normal fluctuations of […]

This study highlights the significant link between tobacco consumption and the increased risk of premenstrual syndrome (PMS) and premenstrual dysphoric disorder (PMDD). Women who smoke are at higher odds of developing these disorders, with a dose-response relationship.

What was studied?

The study aimed to assess the relationship between tobacco smoking and premenstrual syndrome (PMS), including its more severe form, premenstrual dysphoric disorder (PMDD). It focused on understanding how smoking may contribute to the occurrence of these menstrual disorders. The study utilized a case-control design, comparing women with PMS and PMDD to age-matched controls.

Who was studied?

The study population consisted of women diagnosed with PMS, women diagnosed with PMDD, and control groups who did not have PMS or PMDD. Participants were recruited from three major public hospitals and one family counseling center in Santiago de Compostela, Spain, ensuring a sample that included women of various age groups and social backgrounds. All participants completed a self-administered questionnaire that gathered information on their smoking habits, socio-demographic factors, and menstrual health.

What were the most important findings?

The study found that tobacco consumption is significantly associated with both PMS and PMDD. Specifically, current smokers had a higher likelihood of experiencing PMS and PMDD compared to non-smokers. This association was evident even among ex-smokers, although the odds ratio was less precise due to the small sample size of this group. The findings also indicated a dose-response relationship: women who smoked more than 15 cigarettes per day or those with higher pack-years of tobacco consumption had an increased risk of developing PMS and PMDD, suggesting a potential cumulative effect of tobacco exposure. The results were confirmed by a cubic spline model, which further demonstrated a correlation between the amount of tobacco consumed and the likelihood of developing these menstrual disorders.

What are the greatest implications of this study?

The study’s findings suggest that tobacco consumption may be a modifiable risk factor for the development of PMS and PMDD. Given the high prevalence of tobacco use among women globally, particularly in Europe and the United States, these results highlight the need for public health strategies aimed at reducing smoking in women, particularly those in their reproductive years. Health professionals should be aware of the potential link between smoking and these menstrual disorders, as this could inform both prevention and treatment strategies. This study calls for further research to explore the long-term effects of smoking cessation and whether reducing tobacco exposure could lower the risk of developing PMS or PMDD in susceptible women.

Irritable Bowel Syndrome (IBS)

Go to Category Page

Alterations in composition and diversity of the intestinal microbiota in patients with diarrhea-predominant irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

The study investigated gut microbiota in diarrhea-predominant irritable bowel syndrome (D-IBS), revealing distinct microbial shifts, including increased Enterobacteriaceae and reduced Faecalibacterium populations, indicating potential inflammatory mechanisms

What was studied?

The study examined the composition and diversity of the gut microbiota in patients with diarrhea-predominant irritable bowel syndrome (D-IBS) compared to healthy controls. Using 16S rRNA gene sequencing, the researchers evaluated microbial populations, community structure, and specific taxonomic shifts associated with D-IBS, aiming to understand the microbial dysbiosis that may underlie the pathophysiology of this condition.

Who was studied?

The study included 23 patients diagnosed with diarrhea-predominant irritable bowel syndrome (D-IBS) and 23 healthy controls (HC). All participants were recruited from the University of North Carolina at Chapel Hill and were screened to exclude other gastrointestinal conditions.

What were the most important findings?

The analysis revealed significant dysbiosis in the gut microbiota of D-IBS patients compared to healthy controls. Key findings included a substantial increase in the family Enterobacteriaceae, particularly unclassified genera, which are known to encompass pathogenic species. Conversely, the beneficial genus Faecalibacterium, particularly F. prausnitzii, was significantly reduced in D-IBS patients. Faecalibacterium is recognized for its anti-inflammatory properties and is generally considered protective for gut health. This reduction may indicate an underlying pro-inflammatory state within the gut microbiota of D-IBS patients. Additionally, D-IBS patients exhibited lower microbial diversity (α-diversity) and greater variability in microbial community composition (β-diversity), suggesting an imbalance in microbial homeostasis. The study also identified specific increases in Enterococcus, Fusobacterium, and unclassified members of Lactobacillaceae and Veillonella, which were largely undetectable in healthy individuals. These shifts point towards a microbial environment that may exacerbate gut inflammation and motility disturbances characteristic of D-IBS.

ParameterFindings in D-IBS Patients
Microbial DiversityReduced α-diversity, indicating lower microbial richness
Microbial Community StructureIncreased β-diversity, suggesting greater community variability
Increased GeneraEnterobacteriaceae, Enterococcus, Fusobacterium, Veillonella
Decreased GeneraFaecalibacterium, specifically F. prausnitzii
Pathogenic AssociationsElevated Enterobacteriaceae includes potentially pathogenic species
Inflammatory IndicatorsLoss of F. prausnitzii, a known anti-inflammatory bacterium
Microbial DysbiosisImbalanced harmful and beneficial bacteria, indicating gut inflammation

What are the greatest implications of this study?

The findings underscore the significant role of gut microbiota in the pathophysiology of D-IBS, marked by a distinct microbial signature that includes elevated Enterobacteriaceae and diminished Faecalibacterium populations. These microbial alterations reflect potential mechanisms driving gut inflammation and motility disorders. Importantly, the study suggests that microbial dysbiosis could serve as both a biomarker for diagnosing D-IBS and a potential target for therapeutic interventions aimed at restoring microbial balance. Future strategies may include microbiome-targeted therapies such as probiotics or prebiotics aimed at re-establishing beneficial bacterial populations and mitigating pro-inflammatory species.

Altered Gut Microbiota in Irritable Bowel Syndrome and Its Association with Food Components

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study explores the altered gut microbiota in IBS and its association with caffeine and dietary fiber. Findings indicate that high caffeine intake is linked to increased microbial diversity and altered gut microbiome composition in IBS patients, offering insights into dietary strategies for symptom modulation.

What Was Studied?

This study investigated the altered gut microbiota in irritable bowel syndrome (IBS) and its association with specific food components, primarily focusing on the impact of dietary patterns and food intake on the microbial diversity and composition in IBS patients compared to healthy controls (HC). The study aimed to understand how specific nutrients, particularly caffeine and dietary fiber, influence gut microbiome patterns in IBS, contributing to symptom modulation and microbiota alterations.

Who Was Studied?

The study was conducted on 80 young adults diagnosed with IBS based on Rome III or IV criteria, alongside 21 healthy controls. Participants were surveyed for food consumption using a food frequency questionnaire, and fecal samples were collected for microbiome analysis through 16S rRNA Illumina sequencing. The participants were primarily students, which may have influenced dietary habits and lifestyle factors.

What Were the Most Important Findings?

The study found significant differences in gut microbiota composition and diversity between IBS patients and healthy controls, despite similar dietary patterns. Notably, caffeine intake was significantly higher in IBS patients (246.42 mg/d) compared to controls (82.93 mg/d), with high caffeine consumption (>400 mg/d) correlating with increased alpha diversity and alterations in microbiome structure. IBS patients showed a higher abundance of Verrucomicrobia, Coriobacteriia, Bacilli, and Erysipelotrichia at the class level, and Coriobacteriaceae, Porphyromonadaceae, Verrucomicrobiaceae, Lachnospiraceae, and Erysipelotrichaceae at the family level. Genera such as Parabacteroides, Blautia, Lachnospiraceae-unclassified, Veillonella, Oscillibacter, Flavonifractor, and Akkermansia were more abundant in IBS patients, whereas Prevotella was higher in controls. Interestingly, higher microbial diversity and abundance were observed in IBS patients with high caffeine intake, suggesting a potential modulatory role of caffeine on gut microbiota. The study also demonstrated significant correlations between dietary fiber intake and microbial diversity in the IBS group, reinforcing the role of dietary components in gut microbiome dynamics.

Microbiota LevelKey Alterations in IBS Patients
PhylumIncreased Verrucomicrobia, Coriobacteriia, Bacilli, Erysipelotrichia
ClassHigher levels of Verrucomicrobia, Coriobacteriia, Bacilli, Erysipelotrichia
OrderVerrucomicrobiales, Coriobacteriales, Lactobacillales, Erysipelotrichales
FamilyCoriobacteriaceae, Porphyromonadaceae, Verrucomicrobiaceae, Lachnospiraceae, Erysipelotrichaceae
GenusParabacteroides, Blautia, Lachnospiraceae-unclassified, Veillonella, Oscillibacter, Flavonifractor, Akkermansia
Diet-Microbiome CorrelationHigh caffeine intake (>400 mg/d) linked to increased microbial diversity; dietary fiber associated with higher alpha diversity
Symptom RelevanceAltered microbial composition correlated with IBS symptom modulation
Therapeutic ImplicationDietary modulation of caffeine and fiber intake could influence gut microbiome and symptoms in IBS

What Are the Greatest Implications of This Study?

The study's findings underscore the influence of specific food components—particularly caffeine and dietary fiber—on gut microbiome diversity and structure in IBS patients. These results suggest that dietary modulation could be a viable strategy for managing IBS symptoms, particularly through the regulation of caffeine and fiber intake. The observation of increased microbial diversity and specific microbial alterations in high-caffeine consumers points to potential therapeutic or dietary adjustments that could improve gut health in IBS patients. Furthermore, the identification of specific microbial taxa enriched in IBS provides a potential avenue for biomarker development for diagnostic and therapeutic purposes.

Altered profiles of intestinal microbiota and organic acids may be the origin of symptoms in irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study reveals that altered microbiota and elevated organic acids in IBS patients correlate with increased symptom severity, poorer quality of life, and heightened negative emotions. The findings suggest that microbial shifts leading to increased acetic and propionic acid production play a critical role in the pathophysiology of IBS.

What was studied?

This study investigated the profiles of intestinal microbiota and organic acids in patients diagnosed with Irritable Bowel Syndrome (IBS) compared to healthy controls. The researchers hypothesized that the gut microbiota of IBS patients is disrupted, contributing to elevated levels of organic acids, which may be linked to the manifestation of gastrointestinal (GI) symptoms. To evaluate this, the study analyzed fecal samples for microbial populations using quantitative real-time PCR and culture methods, as well as organic acid concentrations through high-performance liquid chromatography (HPLC). Abdominal gas was also measured via X-ray imaging to determine if microbial fermentation contributed to bloating and discomfort commonly reported by IBS patients.

Who was studied?

The study involved 26 patients diagnosed with IBS according to the Rome II criteria, further confirmed by the Rome III modular questionnaire. The cohort was composed of 11 patients with constipation-predominant IBS (IBS-C), 8 with diarrhea-predominant IBS (IBS-D), and 7 with mixed IBS (IBS-M). Twenty-six age- and sex-matched healthy controls were also included for comparison. All participants underwent comprehensive medical evaluations, including physical exams, lab tests, and radiological assessments to exclude any organic GI diseases or systemic health issues.

What were the most important findings?

IBS patients demonstrated significant alterations in their gut microbiota and organic acid levels compared to healthy controls. Notably, the study found higher counts of Veillonella (p = 0.046) and Lactobacillus (p = 0.031) in IBS patients, two bacterial groups known for their role in organic acid production. Correspondingly, fecal analysis revealed that IBS patients exhibited significantly elevated levels of acetic acid (p = 0.049), propionic acid (p = 0.025), and total organic acids (p = 0.014). These organic acids are byproducts of microbial fermentation and have been implicated in gut motility and sensory processing.

Importantly, patients with high levels of acetic or propionic acid experienced more severe GI symptoms, poorer quality of life (QOL), and greater levels of negative emotions compared to those with lower acid concentrations. This correlation suggests that microbial shifts favoring Veillonella and Lactobacillus may drive acid production, which in turn exacerbates abdominal discomfort and impacts psychological well-being. Interestingly, despite the elevated organic acid levels, there were no significant differences in abdominal gas volume between IBS patients and controls, indicating that symptom severity may be driven more by microbial metabolites than gas accumulation.

Microbial GroupIBS PatientsHealthy ControlsStatistical SignificanceAssociated Effects
VeillonellaIncreasedNormal levelsp = 0.046Linked to increased acetic and propionic acid production
LactobacillusIncreasedNormal levelsp = 0.031Associated with organic acid elevation
Acetic AcidElevatedNormal levelsp = 0.049Correlates with abdominal pain and bloating
Propionic AcidElevatedNormal levelsp = 0.025Linked to worse GI symptoms and negative emotions
Total Organic AcidsElevatedNormal levelsp = 0.014Higher levels linked to symptom severity
Abdominal Gas VolumeNo significant changeNormal levelsNot significantSymptoms not driven by gas volume but microbial metabolites

What are the greatest implications of this study?

The study's findings provide compelling evidence that microbial imbalances and organic acid overproduction are critical components of IBS symptomatology. Elevated levels of Veillonella and Lactobacillus are linked to increased acetic and propionic acid production, which correlates with symptom severity, reduced QOL, and heightened negative emotional states. These findings support the concept that IBS may not be merely a functional disorder but is mechanistically influenced by microbial fermentation and its metabolic byproducts. Understanding these microbial and metabolic interactions opens the door for targeted probiotic, prebiotic, or dietary interventions aimed at modulating organic acid production, potentially offering novel therapeutic avenues for IBS management.

Alternation of the gut microbiota in irritable bowel syndrome: an integrated analysis based on multicenter amplicon sequencing data

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study explores the alteration of the gut microbiota in IBS patients, revealing distinct microbial shifts linked to IBS subtypes. Findings highlight potential for microbiome-based therapies and personalized intervention strategies to alleviate symptoms and restore gut health in IBS-D, IBS-C, and IBS-M patients.

What Was Studied?

This study focused on the alteration of the gut microbiota in patients with Irritable Bowel Syndrome (IBS) through an integrated analysis of multicenter amplicon sequencing data. Researchers aimed to elucidate specific microbial changes associated with IBS pathophysiology and its subtypes, including IBS-D (diarrhea-predominant), IBS-C (constipation-predominant), and mixed-type IBS (IBS-M). Utilizing 16S rRNA data from the GMrepo database, the study analyzed microbial diversity, composition, and co-occurrence networks to identify key taxa and potential microbial biomarkers linked to IBS symptomatology and gut dysbiosis.

Who Was Studied?

The study analyzed 708 individuals, with 354 diagnosed with IBS (subcategorized into IBS-D, IBS-C, and IBS-M) and 354 matched healthy controls. Data was retrieved from multicenter cohorts to ensure a diverse representation across age, sex, BMI, and geographical regions. The participants were selected using a propensity score matching (PSM) algorithm to reduce confounding variables and ensure balanced comparisons.

What Were the Most Important Findings?

The study revealed significant alterations in the gut microbiota composition of IBS patients compared to healthy controls. At the phylum level, IBS patients showed a marked reduction in Firmicutes, Euryarchaeota, Cyanobacteria, Acidobacteria, and Lentisphaerae, while Proteobacteria and Bacteroidetes were notably enriched. Interestingly, the Firmicutes/Proteobacteria ratio was significantly decreased in IBS patients, indicative of microbial imbalance. At the family level, the study found an enrichment of Enterobacteriaceae, Moraxellaceae, and Sphingobacteriaceae in IBS patients, while Ruminococcaceae and Bifidobacteriaceae were significantly reduced. Genus-level analysis highlighted increases in Streptococcus, Bacillus, Enterocloster, Sphingobacterium, Holdemania, and Acinetobacter among IBS cohorts. Conversely, Faecalibacterium, Bifidobacterium, and Ruminococcus were substantially depleted, suggesting a loss of anti-inflammatory and SCFA-producing microbiota. Network analysis identified Faecalibacterium prausnitzii, Bifidobacterium longum, and Bifidobacterium breve as key hub species with strong positive interactions, indicating their potential role in maintaining gut homeostasis. Subgroup analysis for IBS-D and IBS-C further revealed distinct microbial signatures, such as the enrichment of Streptococcus in both subtypes, while Faecalibacterium and Ruminococcus were consistently depleted. These findings suggest that microbial dysbiosis in IBS is subtype-specific, potentially influencing disease symptoms and progression through microbial interactions and metabolic shifts.

ParameterFindings in IBS-DFindings in IBS-CFindings in IBS-M
Bacterial DiversityReduced diversity with enrichment of Proteobacteria and BacteroidetesAltered diversity with reductions in Firmicutes and EuryarchaeotaMixed microbial shifts with increased Bacteroidetes and Proteobacteria
Phylum-Level ChangesDecreased Firmicutes, Euryarchaeota, Cyanobacteria, Acidobacteria, Lentisphaerae; increased Proteobacteria and BacteroidetesReduced Firmicutes and Euryarchaeota, with moderate increases in ProteobacteriaIncreased Bacteroidetes and Proteobacteria, reduced Cyanobacteria
Key GeneraEnrichment of Streptococcus, Bacillus, Enterocloster, Sphingobacterium, Holdemania, AcinetobacterEnrichment of Streptococcus; depletion of Faecalibacterium and RuminococcusMixed representation of Streptococcus and Bacillus
Metabolic PathwaysElevated pathways linked to hydrogen sulfide production and bile acid metabolismAltered short-chain fatty acid (SCFA) production pathwaysMixed shifts in SCFA production and bile acid metabolism
Inflammatory AssociationsLinked to increased gut permeability and pro-inflammatory markersAssociated with constipation-related dysbiosis and inflammationMixed inflammatory markers reflecting both diarrhea and constipation
Diagnostic PotentialIdentification of Streptococcus and Enterobacteriaceae as microbial markersFaecalibacterium and Ruminococcus as indicators of microbial imbalanceMixed indicators with potential microbial biomarkers for symptom fluctuation

What Are the Greatest Implications of This Study?

This study emphasizes the critical role of gut microbiota alterations in IBS pathogenesis, with distinct microbial imbalances linked to different IBS subtypes. The identification of specific IBS-exclusive genera like Enterobacteriaceae and the depletion of protective species such as Faecalibacterium prausnitzii highlights potential targets for microbiome-based interventions. The findings suggest that personalized therapeutic strategies could be developed based on microbial profiling, potentially leveraging probiotics, dietary modifications, or fecal microbiota transplantation (FMT) to restore microbial balance and alleviate symptoms. The study also underscores the importance of further research into microbial biomarkers for IBS diagnosis and treatment stratification, particularly in distinguishing between IBS-D and IBS-C through targeted microbiome modulation.

Association of aberrant brain network dynamics with gut microbial composition uncovers disrupted brain-gut-microbiome interactions in irritable bowel syndrome: Preliminary findings

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study identifies disrupted brain-gut-microbiome interactions in IBS patients, revealing altered brain network dynamics and microbial shifts linked to symptom severity and cognitive dysfunction. These findings highlight the potential for targeted therapies addressing both brain connectivity and gut microbiota to improve IBS management.

What was studied?

This study explored the association between brain network dynamics and gut microbial composition in patients with Irritable Bowel Syndrome (IBS), highlighting disrupted brain–gut–microbiome (BGM) interactions as a contributing factor to IBS pathophysiology. The research integrated neuroimaging data, specifically resting-state functional magnetic resonance imaging (rs-fMRI), with gut microbiota profiling through 16S rRNA gene sequencing. Dynamic functional connectivity (DFC) analysis was employed to identify temporal changes in brain networks, while microbial diversity and community composition were assessed in fecal samples. This dual analysis aimed to bridge the gap between neural dysfunction and microbial imbalances in IBS patients.

Who was studied?

The study included 33 IBS patients (23 with diarrhea-predominant IBS, 4 with constipation-predominant IBS, and 6 with mixed IBS) and 32 healthy controls, all right-handed and matched for age and gender. Participants were recruited from Lanzhou University Second Hospital, and all met the Rome IV criteria for IBS. Neuroimaging and fecal sampling were performed to analyze both brain network dynamics and gut microbial composition. Exclusion criteria included the use of probiotics or antibiotics within one month of the study, as well as a history of neurological, psychiatric, or significant gastrointestinal disorders.

What were the most important findings?

The study identified significant alterations in both brain network dynamics and gut microbial diversity in IBS patients. In the DFC analysis, four distinct connectivity states were identified, with IBS patients exhibiting prolonged fraction and mean dwell times in State 4—a state associated with weak brain connectivity linked to self-focused thinking and emotional dysregulation. Furthermore, reduced transitions from State 3 to State 1 were noted, indicating impaired cognitive flexibility, a hallmark of IBS symptomatology. Temporal variability of functional connectivity was also disrupted, particularly within the cognitive control network (CCN) and the sensorimotor network (SMN), implicating these brain regions in IBS-related cognitive and sensory processing abnormalities.

In parallel, gut microbiome analysis revealed decreased alpha diversity and altered beta diversity in IBS patients. Notably, IBS patients showed higher abundances of Anaerostipes, Streptococcus, and Ruminococcus, along with elevated levels of Erysipelotrichaceae. These microbial shifts were associated with symptom severity, depression, and anxiety scores. Correlation analysis demonstrated that specific microbial abundances corresponded to changes in brain network connectivity, suggesting a mechanistic link between gut dysbiosis and brain dysfunction in IBS.

FindingIBS PatientsClinical Implications
Brain State 4 (DFC Analysis)Prolonged dwell timeAssociated with emotional dysregulation and self-focused rumination
Transitions from State 3 to State 1ReducedImpaired cognitive flexibility, symptom exacerbation
Temporal Variability in CCN and SMNDecreasedLinked to cognitive and sensory processing deficits
Alpha Diversity (Microbiota)DecreasedIndicative of microbial dysbiosis
Beta Diversity (Microbiota)AlteredReflects community structure changes in IBS
Anaerostipes, Streptococcus, RuminococcusIncreasedLinked to symptom severity, depression, anxiety
ErysipelotrichaceaeIncreasedAssociated with IBS-specific gut dysfunction

What are the greatest implications of this study?

The findings underscore the critical role of brain-gut-microbiome interactions in IBS pathophysiology, highlighting dynamic brain network disruptions alongside distinct microbial imbalances. Prolonged dwell time in weakly connected brain states and altered microbiota composition suggest that IBS symptomatology is driven by both central neural dysregulation and peripheral microbial alterations. These insights open avenues for targeted therapies aimed at modulating both brain connectivity and gut microbiota to alleviate IBS symptoms. The study also establishes a foundational framework for future investigations into BGM axis interventions as a therapeutic strategy for IBS.

Differences in gut microbial composition correlate with regional brain volumes in irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study identified two distinct IBS subgroups based on gut microbiome profiles, revealing correlations with regional brain volumes. The findings highlight the role of gut dysbiosis in structural brain alterations, offering potential biomarkers for subtype classification.

What was studied?

The study explored the correlation between gut microbial composition and regional brain structural changes in patients with Irritable Bowel Syndrome (IBS). Specifically, it aimed to identify distinct subgroups of IBS patients based on gut microbial profiles and examine how these subgroups correlated with structural brain alterations, particularly in sensory integration and salience network regions. This study is the first of its kind to demonstrate a direct association between gut microbiota composition and brain architecture in IBS, offering insights into the gut-brain axis and its potential implications in IBS pathophysiology.

Who was studied?

The study included 29 adult IBS patients and 23 healthy controls (HCs). Among the IBS patients, distinct subgroups were identified: IBS1, which showed a unique gut microbial signature, and HC-like IBS, whose microbial composition resembled that of healthy controls. These participants underwent stool sample analysis for microbial profiling and structural brain imaging to assess regional brain volumes and correlating microbial taxa.

What were the most important findings?

The study identified two primary subgroups within the IBS population (IBS1 and HC-like IBS) based on gut microbial composition. The IBS1 subgroup exhibited significant differences in the relative abundance of certain microbial taxa, including higher levels of Clostridia and reduced levels of Bacteroidia, which were strongly associated with structural brain changes. Notably, IBS1 showed increased volumes in sensory and motor brain regions while experiencing reduced volumes in the insula and prefrontal cortices. These changes correlated with microbial diversity and the relative abundance of Firmicutes and Bacteroidetes, suggesting that distinct microbial clusters may influence sensory processing and brain structure in IBS patients. Furthermore, the findings indicated that early life trauma and long-standing symptoms were more prevalent in the IBS1 subgroup, hinting at the potential role of gut microbial metabolites in altering brain development and sensory integration pathways. The study suggests that IBS subgroups defined by microbial signatures, rather than traditional clinical classifications, could improve the personalization of therapeutic interventions.

ParameterFindings in IBS1 Group
Microbial DiversityHigher alpha diversity and richness compared to HCs
Firmicutes and BacteroidetesIncreased Firmicutes (Clostridia) and decreased Bacteroidetes (Bacteroidia)
Brain Structural ChangesEnlarged sensory and motor regions, reduced volumes in insula and prefrontal cortices
Sensory Integration RegionsCorrelated with Clostridia abundance
Salience Network RegionsAltered structural changes linked to microbial composition
Early Life TraumaMore common in IBS1, potentially linked to gut-brain axis alterations
Therapeutic ImplicationsSuggests targeted microbial-based therapies for distinct IBS subtypes

What are the greatest implications of this study?

The study's findings underscore the potential for redefining IBS subtypes based on gut microbial composition rather than solely clinical characteristics. This microbial stratification could enable more targeted interventions, such as diet modification, prebiotics, probiotics, and antibiotic therapies, specifically tailored to the microbiome of the IBS1 subgroup. Additionally, the observed brain structural changes in sensory and salience-related regions support the hypothesis that gut microbiota play a role in the neurobiological mechanisms underlying IBS symptoms. These insights also point towards the gut-brain axis as a therapeutic target, where modulation of microbial communities could influence not only gastrointestinal symptoms but also associated neurological outcomes.

Fecal Microbiota in Patients with Irritable Bowel Syndrome Compared with Healthy Controls Using Real-Time Polymerase Chain Reaction: An Evidence of Dysbiosis

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

The study identifies significant fecal microbiota dysbiosis in IBS patients, marked by increased Pseudomonas aeruginosa and Bacteroides thetaiotamicron, highlighting their potential as microbial biomarkers for IBS subtypes.

What was studied?

This study investigated fecal microbiota dysbiosis in patients with Irritable Bowel Syndrome (IBS) compared to healthy controls (HC) using quantitative real-time polymerase chain reaction (qPCR). The research aimed to identify specific microbial shifts that characterize different subtypes of IBS, including constipation-predominant (IBS-C), diarrhea-predominant (IBS-D), and unclassified IBS (IBS-U), highlighting the potential role of microbial imbalance in the pathophysiology of IBS.

Who was studied?

The study included 47 patients with IBS, diagnosed using the Rome III criteria, and 30 healthy controls. Among the IBS patients, 20 were classified as IBS-C, 20 as IBS-D, and 7 as IBS-U. Participants were recruited from a gastroenterology outpatient clinic in northern India. Exclusion criteria included recent use of antibiotics, probiotics, or prokinetics, as well as a history of gastrointestinal surgery, inflammatory bowel disease, or celiac disease.

What were the most important findings?

The study found significant microbial shifts in the fecal samples of IBS patients compared to healthy controls. Notably, the abundance of beneficial microbes like Bifidobacterium and Lactobacillus species was reduced, while pathogenic and opportunistic bacteria such as Pseudomonas aeruginosa, Bacteroides thetaiotamicron, Veillonella, Ruminococcus productus, and Gram-negative bacteria were significantly elevated in IBS patients. Among IBS subtypes, P. aeruginosa was found in 97.9% of IBS cases compared to only 33.3% of healthy controls, indicating its potential role as a microbial marker for IBS. Additionally, Bacteroides thetaiotamicron and segmented filamentous bacteria (SFB) were more abundant in IBS-D than in IBS-C, while Veillonella was elevated specifically in IBS-C patients. Abdominal distension and bloating were associated with increased numbers of Bacteroides thetaiotamicron, Clostridium coccoides, and P. aeruginosa, suggesting that microbial dysbiosis might contribute to symptom severity. Principal component analysis further confirmed distinct microbial profiles across IBS subtypes, supporting the hypothesis that gut microbiota composition is intrinsically linked to symptom manifestation in IBS.

ParameterFindings in IBS Patients
Microbial DiversityReduced abundance of Bifidobacterium and Lactobacillus species
Key Pathogenic MicrobesElevated Pseudomonas aeruginosa, Bacteroides thetaiotamicron, Veillonella, Gram-negative bacteria
IBS Subtype DifferencesPseudomonas aeruginosa prevalent in 97.9% of IBS cases; Veillonella elevated in IBS-C, SFB in IBS-D
Symptom AssociationsAbdominal distension and bloating correlated with Bacteroides thetaiotamicron, Clostridium coccoides, P. aeruginosa
Principal Component AnalysisClear microbial separation between IBS subtypes and healthy controls
Diagnostic ImplicationsPseudomonas aeruginosa and Bacteroides thetaiotamicron as potential biomarkers for IBS diagnosis

What are the greatest implications of this study?

This study strongly supports the hypothesis that gut microbiota dysbiosis is associated with IBS and its subtypes. The pronounced presence of Pseudomonas aeruginosa and Bacteroides thetaiotamicron in IBS patients, especially those with diarrhea-predominant symptoms, suggests that these bacteria could serve as microbial biomarkers for IBS. Furthermore, the differential microbial patterns observed between IBS-C and IBS-D imply that targeted microbial therapies could be developed to address specific dysbiotic signatures. Importantly, the study highlights the role of gut dysbiosis in driving clinical symptoms such as bloating and abdominal distension, reinforcing the need for microbiome-targeted interventions as part of a comprehensive therapeutic strategy for IBS.

Functional dysbiosis within the gut microbiota of patients with constipated-irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

The study highlights functional dysbiosis in constipated-irritable bowel syndrome, with increased sulfate-reducing bacteria and disrupted SCFA profiles.

What was studied?

This study investigated the functional dysbiosis in the gut microbiota of women diagnosed with constipated-irritable bowel syndrome (C-IBS) compared to healthy controls. Unlike previous studies that focused predominantly on phylogenetic composition, this research employed a function-based approach to analyze metabolic capabilities and the presence of specific functional microbial groups. The primary objective was to identify shifts in microbial populations linked to fermentation processes, short-chain fatty acid (SCFA) production, and hydrogen metabolism that may contribute to the pathophysiology of C-IBS.

Who was studied?

The study included 14 women diagnosed with C-IBS according to Rome II criteria and 12 sex-matched healthy controls. All participants were assessed for gastrointestinal symptoms and underwent fecal sampling to analyze microbial populations and fermentation capabilities. None of the participants had taken antibiotics, probiotics, or experienced gastrointestinal infections for at least two months prior to sampling to minimize confounding microbial shifts.

What were the most important findings?

The study revealed a distinct microbial dysbiosis in the gut microbiota of C-IBS patients characterized by significant shifts in fermentation pathways and hydrogen metabolism. The abundance of beneficial lactate-producing bacteria such as Bifidobacterium and Lactobacillus was markedly reduced in C-IBS patients. In contrast, sulfate-reducing bacteria (SRB) populations were elevated by 100-fold compared to healthy controls. This shift was associated with enhanced hydrogen sulfide (H2S) production, a gas implicated in gut motility disturbance and visceral hypersensitivity. Additionally, methanogenic archaea and reductive acetogens, crucial for hydrogen consumption, were significantly lower in C-IBS patients. The decrease in butyrate-producing bacteria like the Roseburia–E. rectale group further disrupted SCFA profiles, potentially impairing anti-inflammatory and gut barrier functions. In vitro starch fermentation assays demonstrated that C-IBS microbiota produced significantly more hydrogen and sulfides, but less butyrate compared to controls. This altered fermentative output underscores the role of functional dysbiosis in generating gastrointestinal symptoms characteristic of C-IBS, such as bloating, constipation, and abdominal pain.

ParameterFindings in C-IBS Patients
Microbial DiversityReduced abundance of lactate-producing Bifidobacterium and Lactobacillus
Sulfate-Reducing Bacteria (SRB)100-fold increase compared to healthy controls
Hydrogen MetabolismDecreased methanogens and reductive acetogens, increased hydrogen production
Butyrate ProductionLower production due to reduced Roseburia–E. rectale populations
Short-Chain Fatty Acid (SCFA) ProfileLess butyrate, more hydrogen sulfide (H2S)
In Vitro FermentationC-IBS microbiota produced more hydrogen and sulfides, less butyrate
Pathophysiological ImplicationsLinked to bloating, constipation, and abdominal pain through H2S toxicity

What are the greatest implications of this study?

This study underscores the importance of functional dysbiosis in the pathogenesis of C-IBS. The enhanced presence of sulfate-reducing bacteria and the resulting increase in hydrogen sulfide production point to a mechanistic link between microbial metabolism and the gastrointestinal symptoms of C-IBS. These findings suggest that targeting SRB populations and restoring lactate- and butyrate-producing bacteria may offer therapeutic benefits in mitigating symptom severity. Furthermore, this research supports the need for microbiome-targeted interventions that focus not only on microbial composition but also on metabolic functionality to effectively manage C-IBS.

Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study identifies distinct microbiome signatures in pediatric IBS, revealing unique microbial imbalances linked to disease subtypes.

What was studied?

This study examined the gastrointestinal microbiome signatures in pediatric patients with Irritable Bowel Syndrome (IBS) compared to healthy controls. Using advanced 16S ribosomal RNA (rRNA) gene sequencing and PhyloChip microarray analyses, the researchers aimed to identify specific microbial patterns associated with IBS subtypes in children. The primary goal was to determine how microbial dysbiosis in pediatric IBS differs from that of healthy children and whether specific bacterial taxa are indicative of disease subtypes.

Who was studied?

The study involved 22 pediatric patients diagnosed with IBS based on the Pediatric Rome III criteria and 22 age-matched healthy controls. The participants, aged 7 to 12 years, were recruited from a large healthcare network in the Houston metropolitan area. Stool samples were collected and analyzed using high-resolution sequencing techniques, including 454 pyrosequencing and PhyloChip hybridization, which enabled the identification of thousands of bacterial taxa and their relative abundances.

What were the most important findings?

The analysis revealed significant microbial differences between pediatric IBS patients and healthy controls. Notably, the microbiomes of IBS patients showed a marked increase in the abundance of α-Proteobacteria, particularly Haemophilus parainfluenzae, which was significantly elevated in IBS patients compared to healthy children (0.89% vs. 0.07%, p < 0.05). At the genus level, Dorea and Veillonella were also more prevalent in IBS patients, while Eubacterium and Anaerovorax were reduced. Interestingly, the study identified a novel Ruminococcus-like organism linked specifically to IBS, suggesting potential undiscovered microbial contributors to the condition.

Subgroup analysis further distinguished IBS subtypes, with the microbiomes of IBS-C (constipation-predominant) and IBS-U (unsubtyped IBS) showing distinct bacterial profiles. For example, Bacteroides vulgatus was less abundant in IBS patients, whereas Alistipes, Akkermansia, and Parabacteroides were found to be more abundant in those with higher pain frequency. These findings highlight the possibility of using microbiome signatures to not only diagnose IBS in children but also potentially classify its subtypes with high accuracy.

Microbial GroupIBS PatientsHealthy ControlsStatistical SignificanceSubtype Specificity
α-ProteobacteriaIncreasedLow prevalencep < 0.05General IBS
Haemophilus parainfluenzaeIncreasedAbsent or lowp < 0.05General IBS
DoreaIncreasedNormal levelsp < 0.05General IBS
VeillonellaIncreasedNormal levelsp < 0.05General IBS
EubacteriumDecreasedHigh prevalencep < 0.05General IBS
AnaerovoraxDecreasedHigh prevalencep < 0.05General IBS
Bacteroides vulgatusDecreasedNormal levelsp < 0.05IBS-C and IBS-U
AlistipesIncreasedNormal levelsp < 0.05High pain frequency
AkkermansiaIncreasedNormal levelsp < 0.05High pain frequency
ParabacteroidesIncreasedNormal levelsp < 0.05High pain frequency
Ruminococcus-like organismIncreasedNot presentp < 0.01General IBS

What are the greatest implications of this study?

The study's findings underscore the critical role of gut microbial composition in pediatric IBS, identifying specific bacterial signatures that correlate with disease presence and subtype classification. The enrichment of α-Proteobacteria, Haemophilus parainfluenzae, and Veillonella in IBS patients, along with the depletion of Eubacterium and Anaerovorax, suggests that microbial dysbiosis contributes to the pathophysiology of IBS. These microbial patterns could serve as biomarkers for diagnosing pediatric IBS and differentiating its subtypes, providing a non-invasive diagnostic tool for clinicians. Moreover, the identification of novel microbes associated with IBS points toward new therapeutic targets that could be explored to restore microbial balance and alleviate symptoms in pediatric patients.

Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study identifies global microbiota signatures in IBS patients, revealing distinct microbial imbalances that differentiate them from healthy controls. Key findings include a 2-fold increase in the Firmicutes/Bacteroidetes ratio, reduced Bifidobacterium and Faecalibacterium populations, and significant shifts in methanogens. These alterations suggest potential diagnostic markers and therapeutic targets for IBS.

What was studied?

This study conducted a global and deep molecular analysis of microbiota signatures in fecal samples from patients diagnosed with Irritable Bowel Syndrome (IBS). The primary aim was to comprehensively identify microbial populations that distinguish IBS patients from healthy individuals and correlate these microbial profiles with IBS symptom severity. The study employed high-throughput phylogenetic microarrays and quantitative polymerase chain reaction (qPCR) to achieve high-resolution profiling of the fecal microbiota, allowing for in-depth assessment of species-level differences and community shifts associated with IBS.

Who was studied?

The study included 62 primary care IBS patients and 46 healthy controls, recruited from the Helsinki area in Finland. Participants met the Rome I criteria for IBS diagnosis and were subdivided into three IBS subtypes: diarrhea-predominant (IBS-D), constipation-predominant (IBS-C), and alternating IBS (IBS-A). Healthy controls were age-matched and confirmed to have no history of gastrointestinal disorders. Fecal samples were collected, frozen, and analyzed using the Human Intestinal Tract Chip (HITChip) microarray and qPCR to characterize microbial composition and abundance.

What were the most important findings?

The analysis revealed distinct microbial imbalances in IBS patients compared to healthy controls. Most notably, there was a 2-fold increased Firmicutes to Bacteroidetes ratio in IBS patients, driven by a significant rise in Dorea, Ruminococcus, and Clostridium spp., alongside a marked reduction in Bacteroidetes (p < 0.0001). Additionally, Bifidobacterium and Faecalibacterium species were notably decreased (p < 0.05), reflecting disruptions in populations typically associated with gut health and anti-inflammatory properties. The study also observed a dramatic reduction in methanogens, with a 4-fold lower average number in IBS patients compared to controls (p < 0.003), which was particularly pronounced in IBS-C patients.

Correlation analyses linked these microbial shifts to IBS symptom scores, suggesting that increased levels of Firmicutes and Proteobacteria may exacerbate gut symptoms through mechanisms like mucosal barrier dysfunction and inflammation. Moreover, redundancy analysis showed that IBS patients consistently clustered separately from healthy controls based on their microbial profiles, underscoring the potential for microbiota-based diagnostics.

Microbial GroupIBS PatientsHealthy ControlsStatistical SignificanceSubtype Specificity
Firmicutes/Bacteroidetes RatioIncreased 2-foldNormal levelsp < 0.0002General IBS
Dorea, Ruminococcus, Clostridium spp.ElevatedNormal levelsp < 0.005General IBS
BacteroidetesDecreasedNormal levelsp < 0.0001General IBS
Bifidobacterium spp.DecreasedNormal levelsp < 0.05General IBS
Faecalibacterium spp.DecreasedNormal levelsp < 0.05General IBS
MethanogensReduced 4-foldNormal levelsp < 0.003Most reduced in IBS-C
ProteobacteriaIncreasedNormal levelsp < 0.05Linked to higher symptom severity
Ruminococcus torquesIncreasedLow prevalence in controlsp < 0.01Associated with higher IBS symptom scores

What are the greatest implications of this study?

The findings highlight the potential of global microbiota signatures as diagnostic markers for IBS and suggest that specific microbial imbalances could serve as therapeutic targets. The elevated Firmicutes/Bacteroidetes ratio, alongside reductions in Bifidobacterium and Faecalibacterium, indicates that IBS is characterized by dysbiotic shifts that may drive symptomatology through immune modulation and disrupted gut barrier function. Furthermore, the substantial reduction in methanogens points to altered fermentation pathways in IBS, potentially contributing to gas production and bowel irregularities. These insights suggest that restoring microbial balance through targeted therapies may be an effective strategy for managing IBS symptoms.

Gut microbiome signatures reflect different subtypes of irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study explores gut microbiome signatures across IBS subtypes, highlighting distinct microbial and metabolic shifts. Findings reveal subtype-specific dysbiosis, with implications for personalized dietary interventions and microbiome-targeted therapies for IBS-D, IBS-C, and IBS-U patients.

What Was Studied?

This study investigated the gut microbiome signatures across different subtypes of Irritable Bowel Syndrome (IBS), specifically IBS-Diarrhea (IBS-D), IBS-Constipation (IBS-C), and unclassified IBS (IBS-U). Researchers aimed to determine distinct microbiota compositions and their functional implications, linking these microbial patterns to clinical features such as inflammation, depression, and dietary habits. A cohort of 942 IBS patients was deeply phenotyped and matched with 942 non-IBS controls based on age, gender, BMI, geography, and dietary patterns. The study utilized 16S rRNA sequencing data to analyze microbial compositions, and MetaCyc pathway analysis to evaluate functional metabolic shifts across subtypes.

Who Was Studied?

The study involved 942 patients diagnosed with IBS, categorized into three subtypes: IBS-D (302 subjects), IBS-C (180 subjects), and IBS-U (460 subjects). These patients were matched with 942 non-IBS controls using strict criteria to minimize confounding factors like age, gender, BMI, geography, and dietary habits. Microbiome samples were collected and analyzed to compare taxonomic and functional compositions between IBS subtypes and healthy controls.

What Were the Most Important Findings?

The study identified distinct gut microbiome signatures across the three IBS subtypes, highlighting significant differences in bacterial diversity and composition. Patients with IBS-D and IBS-U exhibited reduced bacterial diversity (Shannon index; p < .01), while IBS-C showed no significant difference in diversity compared to controls. At the phylum level, IBS-D and IBS-U were characterized by a reduction in Firmicutes, Actinobacteriota, Verrucomicrobiota, and Campilobacterota, alongside an enrichment of Proteobacteria. IBS-C, in contrast, displayed an increased abundance of Verrucomicrobiota and Desulfobacterota. Functional pathway analysis revealed that hydrogen sulfide production pathways (SO4ASSIM) were elevated in IBS-D and IBS-U, potentially linking microbial metabolism to symptom severity. IBS-C showed an increase in palmitoleate biosynthesis pathways, which may contribute to stool hardness through the production of calcium palmitate. Notably, Escherichia/Shigella populations were consistently elevated across all IBS subtypes, while beneficial genera such as Bifidobacterium, Sutterella, and Butyricimonas were depleted, particularly in IBS patients with comorbid depression. The presence of these pathogens, coupled with reductions in short-chain fatty acid (SCFA) production pathways, suggests a disruption in anti-inflammatory microbial activity. Moreover, the study found that specific dietary factors, such as lactose consumption, influenced microbial compositions differently across IBS subtypes, indicating the potential for diet-based modulation of gut microbiota in therapeutic strategies.

ParameterFindings in IBS-DFindings in IBS-CFindings in IBS-U
Bacterial DiversityReduced diversity (Shannon index; p < .01)No significant difference compared to controlsReduced diversity (Shannon index; p < .01)
Phylum-Level ChangesDecreased Firmicutes, Actinobacteriota, Verrucomicrobiota, and Campilobacterota; increased ProteobacteriaIncreased Verrucomicrobiota and DesulfobacterotaDecreased Firmicutes, Actinobacteriota, Verrucomicrobiota, and Campilobacterota; increased Proteobacteria
Key GeneraElevated Escherichia/Shigella; reduced Bifidobacterium, Sutterella, and ButyricimonasElevated Verrucomicrobiota, reduced beneficial SCFA producersElevated Escherichia/Shigella; reduced Bifidobacterium, Sutterella, and Butyricimonas
Functional Pathway ShiftsElevated hydrogen sulfide production pathways (SO4ASSIM)Increased palmitoleate biosynthesis, potentially contributing to stool hardnessElevated hydrogen sulfide production pathways (SO4ASSIM)
Inflammatory AssociationsMicrobial dysbiosis linked to inflammation and symptom severityLinked to constipation mechanisms via altered lipid metabolismAssociated with heightened gut inflammation
Dietary InfluencesSensitivity to lactose consumption linked to symptom exacerbationPotential sensitivity to high-fat diets affecting lipid metabolismVariable dietary influences; lactose consumption sensitivity noted

What Are the Greatest Implications of This Study?

This study underscores the subtype-specific microbial signatures in IBS, linking gut dysbiosis to distinct metabolic and inflammatory pathways. The identification of hydrogen sulfide production in IBS-D and IBS-U suggests microbial-driven exacerbation of diarrhea symptoms, while palmitoleate biosynthesis in IBS-C provides insights into constipation mechanisms. Importantly, the findings emphasize that personalized microbiome modulation, possibly through dietary interventions or targeted probiotics, could offer therapeutic benefits tailored to IBS subtype. Additionally, the association of Escherichia/Shigella overgrowth with inflammation and depression highlights a potential microbiome-targeted approach for addressing psychiatric comorbidities in IBS patients. These findings propose a precision medicine approach, leveraging gut microbiome signatures for individualized treatment strategies in IBS.

Gut microbiota composition and functional prediction in diarrhea-predominant irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study reveals that gut microbiota in IBS-D patients is characterized by reduced Firmicutes, Fusobacteria, and Actinobacteria, alongside elevated Proteobacteria. Functional predictions indicate disrupted metabolism and increased inflammatory pathways, suggesting microbiome-targeted interventions may benefit IBS-D symptom management.

What was studied?

This study investigated the gut microbiota composition and functional prediction in patients with diarrhea-predominant irritable bowel syndrome (IBS-D) compared to healthy controls in Nanchang, China. It aimed to identify differences in microbial diversity, composition, and functional metabolic pathways using 16S rRNA sequencing and PICRUSt analysis. The study included 30 IBS-D patients and 30 healthy controls and examined the relative abundance of various microbial taxa, alpha and beta diversity measures, and predicted functional capabilities of the microbiome.

Who was studied?

The study focused on 30 patients diagnosed with diarrhea-predominant IBS (IBS-D) based on Rome IV criteria, alongside 30 healthy controls. Participants ranged in age from 20 to 64 years and were recruited from Nanchang, China. All participants were screened to exclude conditions like inflammatory bowel disease, peptic ulcer, and recent antibiotic or probiotic use to avoid confounding factors.

What were the most important findings?

The study found that gut microbiota richness, but not diversity, was decreased in IBS-D patients compared to healthy controls. At the phylum level, there was a significant decrease in Firmicutes, Fusobacteria, and Actinobacteria, alongside an increase in Proteobacteria in IBS-D patients. At the genus level, Enterobacteriaceae significantly increased, while Alloprevotella and Fusobacterium significantly decreased. Functional predictions using PICRUSt analysis showed up-regulation in pathways associated with cofactor and vitamin metabolism, xenobiotics biodegradation, and metabolism, while environmental adaptation, cell growth, and death pathways were down-regulated. These shifts suggest that microbial imbalances in IBS-D patients may contribute to inflammation, altered metabolism, and disrupted gut barrier function. Additionally, Proteobacteria, identified as a potential microbial signature of disease, was notably elevated, indicating a possible role in driving inflammation in the gut of IBS-D patients.

ParameterIBS-D Patients
Alpha DiversityDecreased richness; no change in diversity
Phylum-Level Changes↓ Firmicutes, Fusobacteria, Actinobacteria; ↑ Proteobacteria
Genus-Level Changes↑ Enterobacteriaceae; ↓ Alloprevotella, Fusobacterium
Functional Pathways (PICRUSt)↑ Metabolism of cofactors, vitamins, and xenobiotics; ↓ Environmental adaptation, cell growth, and death
Microbial SignatureElevated Proteobacteria linked to inflammation
Inflammatory MarkersAssociated with increased Proteobacteria
Therapeutic ImplicationsPotential for microbiome-targeted therapy and dietary interventions

What are the greatest implications of this study?

The findings underscore the significant role of microbiome alterations in the pathogenesis of IBS-D, highlighting Proteobacteria as a potential microbial marker of disease activity. The functional predictions suggest that dysbiosis in IBS-D is not limited to microbial composition but extends to metabolic and detoxification pathways, which may exacerbate symptoms and chronic inflammation. These insights open avenues for microbiome-targeted therapies, such as probiotics, prebiotics, and dietary modifications, aimed at restoring microbial balance and improving patient outcomes. Furthermore, the study emphasizes the need for region-specific microbiome analyses due to geographical variations in gut flora.

IBS-associated phylogenetic unbalances of the intestinal microbiota are not reverted by probiotic supplementation

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study found that while probiotics may relieve IBS symptoms, they do not revert the gut microbiota imbalances associated with the disorder.

What was studied?

This study investigated the phylogenetic unbalances of the intestinal microbiota in individuals diagnosed with Irritable Bowel Syndrome (IBS) and evaluated whether these imbalances could be reverted through daily probiotic supplementation with a novel yogurt containing Bifidobacterium animalis subsp. lactis Bb12 and Kluyveromyces marxianus B0399. The primary goal was to determine if the probiotic intervention would modulate the gut microbiota composition in a way that counteracts the dysbiosis observed in IBS patients.

Who was studied?

Nineteen individuals with clinically diagnosed IBS (10 with diarrhea-predominant IBS, 5 with mixed bowel habits, and 4 with constipation-predominant IBS) participated in a monocentric trial. Their gut microbiota profiles were compared with a control cohort of 24 healthy subjects, matched for age and sex, and previously characterized using the same methodologies.

What were the most important findings?

The study demonstrated that the gut microbiota of IBS patients is significantly different from that of healthy individuals, showing distinct phylogenetic imbalances. The IBS-associated microbiota was characterized by an increased abundance of Lactobacilli, Bacillus cereus, Bacillus clausii, Bifidobacteria, Clostridium cluster IX, and Eubacterium rectale, alongside a marked depletion of the Bacteroides/Prevotella group and the Veillonella genus. Notably, several bacterial groups previously defined as pathobionts, such as members of the Enterobacteriaceae family, Enterococcus faecium, Clostridium difficile, and Campylobacter spp., were enriched in IBS patients.

Despite these findings, the probiotic intervention with B. animalis subsp. lactis Bb12 and K. marxianus B0399 did not significantly alter the microbiota composition in IBS patients. After four weeks of daily consumption, the microbial profiles of the participants showed no major shifts, with high inter-individual diversity persisting. Hierarchical clustering of microarray fingerprints before and after supplementation indicated that most samples remained largely unchanged. Additionally, measures of microbial diversity, such as Shannon and richness indices, were not impacted by the probiotic treatment. This indicates that while the probiotics may provide symptomatic relief, they do not appear to correct the underlying microbial imbalances associated with IBS.

Microbial GroupIBS PatientsHealthy ControlsStatistical SignificanceEffect of Probiotics
LactobacilliIncreasedNormal levelsp < 0.05No significant change observed
Bacillus cereusIncreasedLow prevalencep < 0.001No significant change observed
Bacillus clausiiIncreasedLow prevalencep < 0.001No significant change observed
BifidobacteriaIncreasedNormal levelsp < 0.001No significant change observed
Clostridium cluster IXIncreasedNormal levelsp < 0.001No significant change observed
Eubacterium rectaleIncreasedNormal levelsp < 0.01No significant change observed
Bacteroides/PrevotellaDecreasedHigh prevalencep < 0.01No significant change observed
Veillonella genusDecreasedNormal levelsp < 0.05No significant change observed
EnterobacteriaceaeIncreasedLow prevalencep < 0.001No significant change observed
Enterococcus faeciumIncreasedLow prevalencep < 0.001No significant change observed
Clostridium difficileIncreasedLow prevalencep < 0.01No significant change observed
Campylobacter spp.IncreasedLow prevalencep < 0.05No significant change observed

What are the greatest implications of this study?

The findings suggest that the therapeutic benefits of probiotic supplementation in IBS may not be attributed to substantial shifts in the gut microbiota composition. The persistence of phylogenetic unbalances despite intervention implies that the mechanisms of probiotic efficacy in IBS might be independent of direct compositional changes, possibly involving modulation of immune responses or gut barrier function instead. This challenges the traditional view that microbiota normalization is a key pathway for probiotic effectiveness in IBS treatment and underscores the necessity for mechanistic studies focused on functional, rather than purely compositional, microbiota changes.

Increase in fecal primary bile acids and dysbiosis in patients with diarrhea-predominant irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

The study identified significant shifts in fecal bile acid composition and gut microbiota in IBS-D patients. Elevated primary bile acids and dysbiosis were linked to stool frequency and consistency, suggesting bile acid modulation as a potential therapeutic target for symptom management.

What was studied?

This study examined the alterations in fecal bile acid (BA) composition and gut microbiota dysbiosis in patients with diarrhea-predominant irritable bowel syndrome (IBS-D) compared to healthy subjects. The research aimed to identify how shifts in primary and secondary bile acid levels correlate with symptoms of diarrhea, stool frequency, and microbial changes that could influence bile acid metabolism

Who was studied?

The study included 14 patients diagnosed with diarrhea-predominant IBS (IBS-D) and 18 healthy subjects (HS) as controls. All participants were evaluated for clinical symptoms, and stool samples were collected for analysis of bile acid composition and gut microbiota profiling using quantitative PCR (qPCR) and high-performance liquid chromatography–tandem mass spectrometry (HPLC-MS/MS).

What were the most important findings?

The study uncovered a significant increase in primary bile acids (BA) in the feces of IBS-D patients compared to healthy subjects, with a corresponding decrease in secondary bile acids. Primary bile acids, particularly cholic acid (CA) and chenodeoxycholic acid (CDCA), were markedly elevated, contributing to looser stool consistency and higher stool frequency. These elevations in primary BA were positively correlated with the Bristol Stool Scale score and the frequency of bowel movements. In contrast, secondary bile acids such as deoxycholic acid (DCA) were significantly reduced in IBS-D patients, suggesting impaired microbial transformation. Dysbiosis within the gut microbiota was also evident, characterized by a significant increase in Escherichia coli and a marked decrease in Bifidobacterium and members of the Leptum group. These microbial shifts indicate a reduction in the bacteria responsible for bile acid dehydroxylation, further exacerbating the accumulation of primary BA. The study postulates that this microbial imbalance not only alters bile acid metabolism but may also promote mucosal permeability and colonic motility, contributing to the pathophysiology of diarrhea in IBS-D patients.

ParameterIBS-D Patients
Primary Bile AcidsSignificantly elevated (CA, CDCA)
Secondary Bile AcidsMarkedly reduced (DCA)
Gut Microbiota ImbalanceIncrease in Escherichia coli
Microbial ReductionsBifidobacterium and Leptum groups
Stool FrequencyIncreased; correlated with primary BA levels
Stool ConsistencyLooser; linked to higher primary BA levels
Microbial TransformationImpaired conversion from primary to secondary BA
Inflammatory ImplicationsSuggestive of increased mucosal permeability and motility changes

What are the greatest implications of this study?

This study underscores the critical role of bile acid dysbiosis in the pathogenesis of diarrhea-predominant IBS. The findings suggest that the elevated primary bile acids, linked to microbial dysbiosis, could serve as both biomarkers and therapeutic targets for IBS-D. The observed reduction in secondary bile acids, typically resulting from microbial transformation, highlights the importance of gut microbiota in maintaining bile acid homeostasis. This disruption in bile acid metabolism not only influences stool consistency and motility but may also contribute to chronic gut inflammation. Therapeutic interventions that aim to modulate bile acid levels or restore microbial balance may hold promise for symptom relief in IBS-D patients.

Irritable Bowel Syndrome and Nickel Allergy:What Is the Role of the Low Nickel Diet?

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

A low-nickel diet significantly improved gastrointestinal symptoms and physical well-being in IBS patients with nickel allergy, despite persistent intestinal permeability. Findings suggest nickel-induced immune activation as a therapeutic target in IBS.

What was studied?

This pilot study evaluated the prevalence of nickel (Ni) allergy in individuals diagnosed with irritable bowel syndrome (IBS) and investigated the clinical efficacy of a low-nickel diet (LNiD) in this population. Specifically, the authors assessed the impact of the dietary intervention on gastrointestinal symptoms, intestinal permeability, quality of life, and psychological status in Ni-sensitized IBS patients. It also analyzed how the severity of allergic skin reactions and IBS subtypes might influence changes in gut permeability during LNiD intervention.

Who was studied?

Twenty patients fulfilling Rome III criteria for IBS and exhibiting Ni sensitization (confirmed by positive patch test) were enrolled. These individuals had undergone a thorough exclusion process to rule out other organic gastrointestinal disorders, infections, or metabolic dysfunctions. The sample predominantly consisted of females, with most presenting the diarrhea-predominant (IBS-D) subtype. After baseline evaluation of intestinal permeability, psychological wellbeing, and gastrointestinal symptoms, participants followed a low-Ni diet for three months. Compliance was monitored via dietary diaries. Post-intervention assessments included repeated clinical questionnaires and permeability testing using ⁵¹Cr-EDTA for patients with initially elevated intestinal permeability. A control group of healthy subjects matched by age, sex, and socioeconomic background was included for comparison in permeability analysis.

What were the most important findings?

A strikingly high prevalence of nickel allergy was found among IBS patients, particularly in females. The LNiD significantly alleviated gastrointestinal symptoms (except vomiting) and improved general well-being scores (notably bodily pain and physical functioning). Despite improvements in symptoms, all participants continued to show elevated intestinal permeability post-intervention, suggesting persistent barrier dysfunction possibly due to chronic mucosal immune activation. Interestingly, intestinal permeability changes (Δ⁵¹Cr-EDTA) varied by IBS subtype and allergic response severity. Patients with stronger Ni skin reactivity (++/+++) were more likely to experience worsened barrier function, while those with IBS-M or IBS-U subtypes exhibited improvement. Psychometric testing revealed widespread psychological distress, especially anxiety, supporting the established association between IBS and affective dysregulation. These findings reinforce the concept of systemic nickel allergy syndrome (SNAS) as a relevant comorbidity in IBS and point toward immune-mediated mechanisms involving CD4+ T cell infiltration and Th2 cytokines as underlying contributors to both gastrointestinal and systemic symptoms.

From a microbiome perspective, although the study did not directly assess microbial composition, its findings implicate Ni-induced barrier dysfunction as a potential facilitator of microbial dysbiosis. The interplay between metal exposure, gut immune activation, and barrier integrity represents a critical axis in microbiome-host interaction and warrants further microbial profiling in future studies.

What are the greatest implications of this study?

This study identifies a potentially overlooked driver of IBS symptoms—nickel hypersensitivity—and provides preliminary but compelling evidence for dietary nickel restriction as a therapeutic intervention in select IBS patients. Its implications extend to redefining subgroups within IBS, informing personalized nutrition strategies, and advancing the study of metal-induced gut dysfunction as a contributor to microbial dysbiosis. Moreover, it highlights the need for integrative care models that address metal exposure, gut permeability, immune status, and psychological health concurrently. Given that low-Ni diets do not restrict FODMAPs, this study also challenges prevailing dietary paradigms in IBS management by introducing a non-FODMAP, immune-targeted approach with mechanistic plausibility.

Irritable Bowel Syndrome-Like Disorders in Endometriosis: Prevalence of Nickel Sensitivity and Effects of a Low-Nickel Diet. An Open-Label Pilot Study.

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

Nickel allergic contact mucositis was identified in over 90% of endometriosis patients with IBS-like symptoms. A low-nickel diet significantly reduced gastrointestinal, extra-intestinal, and gynecological symptoms, revealing nickel sensitivity as a key driver of endometriosis symptomatology.

What was studied?

This open-label pilot study investigated the prevalence of nickel allergic contact mucositis (Ni ACM) in women with endometriosis who presented with irritable bowel syndrome (IBS)-like symptoms and assessed the efficacy of a low-nickel diet (LNiD) in ameliorating gastrointestinal, extra-intestinal, and gynecological symptoms. The study aimed to determine whether nickel (Ni) sensitivity contributes to the symptom burden in endometriosis and whether dietary nickel restriction can serve as a therapeutic intervention.

Who was studied?

The study initially screened 83 women of reproductive age diagnosed with endometriosis via imaging or laparoscopy who also experienced at least three gastrointestinal symptoms scoring ≥5 on the Gastrointestinal Symptom Rating Scale (GSRS). After applying exclusion criteria (e.g., celiac disease, IgE-mediated food allergies), 47 patients remained eligible. Sixteen dropped out due to the dietary restrictions, leaving 31 who completed the study. Each participant underwent a nickel oral mucosa patch test (omPT) to identify Ni ACM. Of the 31 patients, 28 (90.3%) tested positive. All participants followed a low-Ni diet for three months, with symptoms reassessed using the GSRS at baseline (T0) and after the intervention (T1).

What were the most important findings?

Nickel ACM was highly prevalent among women with endometriosis and IBS-like symptoms, with 90.3% of study completers testing positive via omPT. All patients who adhered to a three-month low-nickel diet experienced statistically significant reductions in all 15 gastrointestinal symptoms, including bloating, abdominal pain, diarrhea, and constipation. Additionally, the LNiD led to meaningful improvements in seven extra-intestinal symptoms such as headache, fatigue, and joint pain, as well as in hallmark gynecological symptoms of endometriosis: chronic pelvic pain, dysmenorrhea, and dyspareunia. These improvements underscore a systemic role of nickel sensitivity in the symptomatology of endometriosis beyond localized pelvic pathology.

From a microbiome perspective, Ni ACM reflects a low-grade inflammatory response that disrupts mucosal immune regulation and intestinal barrier integrity—two key mechanisms implicated in microbial dysbiosis. Though microbiome composition was not directly assessed, the systemic inflammatory profile induced by dietary nickel may favor enrichment of nickel-tolerant pathobionts, including certain Gammaproteobacteria and urease-producing bacteria, while impairing barrier-supporting commensals. This aligns with broader hypotheses on the metallomic drivers of endometriosis-associated dysbiosis.

What are the greatest implications of this study?

This study provides compelling evidence that nickel hypersensitivity may be a clinically significant and previously underrecognized contributor to the gastrointestinal and systemic symptom burden in endometriosis. It positions Ni ACM not only as a comorbidity but as a potential driver of symptom exacerbation, offering a new lens through which to understand IBS-like manifestations in endometriosis. Importantly, the successful use of a targeted dietary intervention based on objective testing (omPT) introduces a personalized medicine framework that could improve quality of life while minimizing unnecessary dietary restrictions, such as those imposed by low-FODMAP diets. If validated in larger, randomized cohorts, the incorporation of nickel testing and dietary counseling into standard endometriosis management could represent a low-risk, high-reward clinical advance.

Lower Bifidobacteria counts in both duodenal mucosa-associated and fecal microbiota in irritable bowel syndrome patients

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study reveals that IBS patients have significantly lower bifidobacteria counts in both fecal and duodenal samples compared to healthy controls. The marked reduction in Bifidobacterium catenulatum levels suggests a critical link to gut dysbiosis, impaired mucosal integrity, and potential contributions to symptom severity in IBS subtypes.

What was studied?

This study investigated the levels of bifidobacteria in both duodenal mucosa-associated and fecal microbiota of patients with Irritable Bowel Syndrome (IBS) compared to healthy controls. The primary goal was to identify whether IBS patients exhibited significant reductions in bifidobacteria populations, which could play a role in gut dysbiosis and symptom expression. To achieve this, the researchers employed molecular-based techniques including Fluorescent In Situ Hybridization (FISH) and real-time PCR analysis to quantify bifidobacterial species in both fecal and duodenal samples.

Who was studied?

The study cohort consisted of 41 IBS patients meeting the Rome II criteria and 26 healthy subjects without GI symptoms or major abdominal surgery. The IBS group included patients with diarrhea-predominant (IBS-D), constipation-predominant (IBS-C), and alternating subtypes (IBS-A). Samples of fecal matter and duodenal mucosa were collected for microbial analysis, with careful exclusion of participants who had used probiotics, antimicrobials, or other medications known to influence gut flora composition.

What were the most important findings?

The analysis revealed that IBS patients had significantly lower levels of bifidobacteria in both fecal and duodenal mucosa-associated samples compared to healthy controls. FISH analysis demonstrated a 2-fold decrease in bifidobacteria levels in fecal samples of IBS patients (4.2 ± 1.3%) versus healthy subjects (8.3 ± 1.9%, p < 0.01). In particular, Bifidobacterium catenulatum levels were markedly reduced in IBS patients, with concentrations of 6 ± 0.6% compared to 19 ± 2.5% in healthy controls (p < 0.001). The disparity was consistent across all IBS subtypes, indicating a broad-spectrum deficiency rather than subtype-specific microbial shifts.

The study also highlighted that while overall bifidobacteria counts were lower, other major bacterial groups did not show significant differences between IBS patients and healthy subjects, suggesting a targeted disruption rather than widespread microbial imbalance. This finding underscores the potential role of bifidobacteria depletion in IBS pathophysiology, possibly through mechanisms involving mucosal integrity, anti-inflammatory activity, and short-chain fatty acid production.

Microbial GroupIBS PatientsHealthy ControlsStatistical SignificanceAssociated Effects
Total Bifidobacteria (Fecal)4.2 ± 1.3%8.3 ± 1.9%p < 0.01Reduced short-chain fatty acid production, disrupted gut barrier
Bifidobacterium catenulatum6 ± 0.6%19 ± 2.5%p < 0.001Potential role in symptom expression through gut dysbiosis
Total Bifidobacteria (Duodenal)ReducedNormal levelsp < 0.01Suggests mucosal disruption in IBS patients
Other Major Bacterial GroupsNo significant changeNormal levelsNot significantIndicates targeted depletion rather than widespread dysbiosis
FISH Detection Coverage32%44%p < 0.05Lower overall detection in IBS samples

What are the greatest implications of this study?

The results strongly suggest that reduced bifidobacteria populations, particularly Bifidobacterium catenulatum, may contribute to IBS symptomatology through impaired mucosal protection and altered gut homeostasis. This finding is crucial as bifidobacteria are known to produce lactic and acetic acids that lower gut pH, inhibit pathogenic bacteria, and support barrier function. Therapeutic strategies that restore bifidobacteria levels—such as targeted probiotics or prebiotic interventions—may offer novel approaches for symptom management in IBS patients. These insights pave the way for more microbiome-focused therapeutic strategies aimed at recalibrating microbial populations in the gut.

Molecular analysis of the luminal- and mucosal-associated intestinal microbiota in diarrhea-predominant irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study reveals that diarrhea-predominant IBS (D-IBS) is linked to reduced microbial diversity and niche-specific dysbiosis in the gut.

What was studied?

This study investigated the molecular composition of the luminal- and mucosal-associated intestinal microbiota in individuals with diarrhea-predominant irritable bowel syndrome (D-IBS) compared to healthy controls. Using terminal-restriction fragment length polymorphism (T-RFLP) fingerprinting of the bacterial 16S rRNA gene, researchers aimed to uncover compositional differences and biodiversity shifts in both fecal and colonic mucosal niches. The study's objective was to determine whether D-IBS is associated with distinct microbial communities that could explain its pathogenesis and symptomatology.

Who was studied?

The study population consisted of 16 patients diagnosed with D-IBS according to the Rome III criteria and 21 healthy controls. The participants were recruited from the general population of Chapel Hill, NC, and the University of North Carolina Hospitals outpatient clinics. All subjects underwent fecal and colonic mucosal sampling for microbial analysis. Inclusion criteria ensured that participants had no history of GI tract surgery (except appendectomy or cholecystectomy), no other GI disorders, and had not taken antibiotics, anti-inflammatory agents, or probiotics for at least two months prior to sampling.

What were the most important findings?

The analysis revealed significant differences in the composition and biodiversity of the intestinal microbiota between D-IBS patients and healthy controls. The fecal samples of D-IBS patients demonstrated a 1.2-fold reduction in microbial biodiversity compared to healthy subjects (p = 0.008). Interestingly, while fecal samples showed decreased diversity, mucosal samples did not exhibit the same trend, suggesting niche-specific microbial alterations in D-IBS. The study identified a distinct reduction in Clostridiales and Planctomycetaceae in D-IBS fecal samples, two groups previously associated with gut health and immune modulation. Conversely, certain terminal-restriction fragments (T-RFs) were detected in D-IBS patients but not in controls, indicating shifts toward a more pathogenic or dysbiotic microbial environment.

Multivariate analysis of T-RFLP profiles highlighted clear distinctions between luminal and mucosal communities in both groups, with D-IBS patients showing a higher degree of microbial dissimilarity in fecal samples compared to mucosal ones. This niche-specific differentiation underscores the complexity of D-IBS-associated dysbiosis, where luminal disturbances are more pronounced than mucosal alterations.

Microbial GroupD-IBS PatientsNiche-Specific Effect
ClostridialesDecreased (p < 0.05)Significant reduction in fecal samples
PlanctomycetaceaeDecreased (p < 0.01)Reduced in fecal, not mucosal samples
T-RFs unique to D-IBSIncreased (p < 0.01)Luminal-specific dysbiosis
Microbial BiodiversityReduced by 1.2-fold (p < 0.008)Significant only in fecal samples
Mucosal Microbial DiversityNo significant changeBiodiversity unchanged in mucosal samples

What are the greatest implications of this study?

The findings support the hypothesis that D-IBS is linked to significant dysbiosis, particularly within the fecal microbiota. The reduced biodiversity and shifts in specific microbial populations such as Clostridiales suggest that microbial depletion and overrepresentation of dysbiotic species could contribute to gut barrier dysfunction, immune activation, and symptomatic expression in D-IBS. Moreover, the difference in microbial composition between luminal and mucosal niches indicates that therapeutic strategies may need to be niche-specific to effectively target the underlying microbiota imbalances in D-IBS patients. These insights could pave the way for targeted microbiome-based interventions that are specifically designed to restore biodiversity and recalibrate pathogenic imbalances in D-IBS patients.

More than constipation - bowel symptoms in Parkinson's disease and their connection to gut microbiota

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

IBS-like symptoms in Parkinson's disease are linked to distinct gut microbiota changes, particularly Prevotella depletion, implicating the microbiota-gut-brain axis in non-motor symptoms.

What was studied?

The study explored the correlation between bowel symptoms, particularly IBS-like symptoms, and gut microbiota composition in patients with Parkinson's disease (PD). The primary objective was to assess whether IBS-like symptoms in PD patients were linked to distinct microbial signatures, specifically focusing on alterations in gut bacteria such as Prevotella. The study also aimed to evaluate the broader gastrointestinal dysfunction in PD beyond mere constipation, investigating the microbiota-gut-brain axis's role in non-motor symptoms.

Who was studied?

The study included 74 patients with Parkinson's disease (PD) and 75 healthy controls. Participants were assessed for bowel symptoms using the Rome III criteria, and microbiota analysis was performed on fecal samples using 16S rRNA sequencing. The study specifically focused on identifying differences in microbial communities between PD patients with IBS-like symptoms and those without.

What were the most important findings?

The study found that IBS-like symptoms were significantly more prevalent in PD patients (24.3%) compared to controls (5.3%). Importantly, PD patients with IBS-like symptoms exhibited distinct gut microbiota compositions, characterized by a marked reduction in Prevotella and related taxa, which are typically involved in the maintenance of gut barrier integrity and mucosal immunity. In contrast, the genus Bacteroides remained relatively stable. The microbial dysbiosis observed in IBS+ PD patients correlated with more severe non-motor symptoms, such as pain and gastrointestinal distress, which are commonly associated with dysregulation of the gut-brain axis. The results suggest that the gut microbial environment may exacerbate gastrointestinal and non-motor symptoms in PD, providing potential biomarkers for stratification and targeted therapy.

The study also revealed that the presence of IBS-like symptoms in PD patients correlated with a broader spectrum of non-motor symptoms, including increased pain and dysautonomia. These symptoms may reflect a more complex gut-brain axis disruption in PD, where microbial shifts contribute to both local gut dysfunction and central nervous system alterations.

ParameterFindings in PD Patients with IBS-like Symptoms
IBS Prevalence24.3% in PD vs. 5.3% in controls
Key Microbial ChangesMarked reduction in Prevotella
Gut-Brain Axis DisruptionLinked to more severe non-motor symptoms
Non-Motor SymptomsIncreased pain, gastrointestinal distress, dysautonomia
Diagnostic ImplicationsPotential biomarker for identifying at-risk patients
Therapeutic ImplicationsSuggests microbiome-targeted therapies for symptom relief

What are the greatest implications of this study?

The findings suggest that microbial profiling of PD patients could serve as a diagnostic tool for identifying those at risk of severe gastrointestinal dysfunction and non-motor symptoms. Furthermore, the association between Prevotella depletion and IBS-like symptoms highlights the potential for microbiome-targeted interventions to alleviate both bowel symptoms and broader non-motor manifestations in PD. This supports a growing body of evidence that the microbiota-gut-brain axis plays a significant role in the pathophysiology of Parkinson's disease, extending beyond motor symptoms to include gut dysbiosis-driven complications. The study advocates for integrating microbiome analysis into PD management to tailor dietary, probiotic, and therapeutic interventions that restore microbial balance and potentially improve patient outcomes.

Multi-omics profiles of the intestinal microbiome in irritable bowel syndrome and its bowel habit subtypes

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study explores multi-omics profiles of the intestinal microbiome in IBS subtypes, revealing distinct microbial and metabolic signatures. Findings highlight potential for microbiome-based diagnostics and targeted dietary interventions in IBS-D, IBS-C, and IBS-M, enhancing personalized therapeutic strategies.

What Was Studied?

This study investigated the multi-omics profiles of the intestinal microbiome in Irritable Bowel Syndrome (IBS) and its bowel habit subtypes. The researchers aimed to uncover distinct microbial compositions and functional differences in patients with IBS-D (diarrhea-predominant), IBS-C (constipation-predominant), and IBS-M (mixed), compared to healthy controls. This study utilized 16S rRNA sequencing, metatranscriptomics, and untargeted metabolomics to capture both compositional and functional microbial changes. A cohort of 318 IBS patients and 177 healthy controls provided fecal samples, which were analyzed for microbial taxa, gene expression, and metabolic products.

Who Was Studied?

The study included 318 IBS patients, categorized into IBS-D, IBS-C, and IBS-M, alongside 177 healthy controls. These participants were recruited from diverse backgrounds and matched by age, gender, BMI, diet, and anxiety levels to minimize confounding factors. The study used comprehensive multi-omics approaches to investigate microbiome signatures, functional gene expression, and metabolic profiles in these individuals.

What Were the Most Important Findings?

The study identified distinct multi-omics microbial signatures in IBS patients compared to healthy controls. IBS-D patients exhibited increased levels of Bacteroides dorei, alterations in succinate and mannose metabolism, and elevated polyamine synthesis, which are associated with diarrhea severity. Meanwhile, IBS-C patients showed distinct upregulation of butyrate-producing pathways and enrichment in Verrucomicrobiota. The metatranscriptomics analysis revealed heightened expression of genes involved in fructose and polyol metabolism across all IBS subtypes, suggesting a microbiome-driven enhancement of fermentable carbohydrate utilization. Additionally, metabolomic profiling showed increased tyramine, gentisate, and hydrocinnamate in IBS patients, suggesting disruptions in aromatic amino acid metabolism. The multi-omics classifier developed from these findings demonstrated high accuracy (AUC 0.82) in distinguishing IBS patients from healthy controls. Further subclassification models effectively differentiated IBS-D from IBS-C with 86% accuracy, highlighting the role of bile acids, polyamines, and SCFA pathways in bowel habit variability.

ParameterFindings in IBS-DFindings in IBS-CFindings in IBS-M
Bacterial DiversityAltered diversity with increased Bacteroides doreiElevated Verrucomicrobiota and butyrate-producing pathwaysMixed microbial shifts with no clear dominant phylum
Key GeneraEnrichment of Bacteroides dorei, reduction in anti-inflammatory taxaIncreased Verrucomicrobiota and SCFA producersVariable populations of Firmicutes and Bacteroidetes
Metabolic PathwaysEnhanced succinate and mannose metabolism; elevated polyamine synthesisUpregulation of butyrate-producing pathwaysMixed shifts in carbohydrate fermentation and SCFA production
Metatranscriptomics AnalysisHigher expression of genes involved in fructose and polyol metabolismButyrate synthesis pathways more prominentElevated expression of pathways linked to bile acid metabolism
Metabolomic ShiftsIncreased levels of tyramine, gentisate, and hydrocinnamateElevated levels of butyrate and palmitoleateMixed aromatic amino acid metabolism alterations
Inflammatory AssociationsLinked to diarrhea severity through succinate and bile acid dysregulationLinked to constipation through lipid metabolismMixed inflammation markers reflective of both diarrhea and constipation
Diagnostic PotentialMulti-omics classifier with 86% accuracy in distinguishing IBS-DEffective biomarker profiles for IBS-CSubtype differentiation through SCFA and bile acid pathways

What Are the Greatest Implications of This Study?

This study's findings underscore the role of gut microbiome dysbiosis in IBS pathophysiology, driven by specific metabolic and transcriptional shifts. The identification of subtype-specific microbial signatures highlights the potential for personalized microbiome-based diagnostics and targeted dietary interventions. Notably, the association between fermentable carbohydrate metabolism and symptom severity suggests that dietary modifications—such as low-FODMAP or specific carbohydrate restriction—could be effective therapeutic strategies. Additionally, the development of a multi-omics classifier with high diagnostic accuracy presents a promising non-invasive approach for IBS diagnosis and subtype differentiation.

Overlap Between Irritable Bowel Syndrome Diagnosis and Endometriosis in Adolescents

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

The study links endometriosis with a fivefold increased risk of IBS in adolescents, emphasizing the role of acyclic pelvic pain severity and inflammation as shared mechanisms. Early screening for IBS and microbiome-targeted interventions could improve outcomes in this group.

What Was Studied?

The study investigated the association between endometriosis and irritable bowel syndrome (IBS) in adolescent females. Specifically, it aimed to determine the prevalence of IBS in those with and without surgically confirmed endometriosis and to explore how pelvic pain severity and other comorbidities influenced this relationship.

Who Was Studied?

The study analyzed data from 323 adolescent females under 21 years of age who participated in the "Women’s Health Study: Adolescence to Adulthood," a longitudinal cohort. Participants were grouped based on the presence or absence of surgically confirmed endometriosis and whether they met the diagnostic criteria for IBS, as defined by Rome IV guidelines or self-reported clinician diagnoses.

What Were the Most Important Findings?

The study found that adolescents with endometriosis were five times more likely to have IBS than those without endometriosis (adjusted odds ratio [aOR], 5.26). Among participants with endometriosis, the odds of IBS increased with the severity of acyclic pelvic pain, with each 1-point increase in pain severity raising the odds by 31% (aOR, 1.31). A significant overlap in pain-related comorbidities, including migraines, sleep disturbances, and urinary symptoms, was observed in individuals with both conditions. Moreover, central pain sensitization, driven by visceral hypersensitivity and alterations in the peripheral and central nervous systems, was identified as a potential shared mechanism between endometriosis and IBS. This study's microbiome associations highlight the role of inflammatory processes in both conditions. Low-grade mucosal inflammation and mast cell activation, often linked to microbiome dysbiosis, were implicated as contributing factors to the development of IBS in the context of endometriosis.

What Are the Greatest Implications of This Study?

This research underscores the need for integrated screening and management strategies for adolescents presenting with endometriosis and/or IBS. Identifying overlapping symptoms early could reduce diagnostic delays and improve patient outcomes. Furthermore, the findings suggest potential pathways for therapeutic interventions targeting the microbiome and immune regulation, such as central sensitization and inflammation. Clinicians should consider IBS in the differential diagnosis for adolescents with severe acyclic pelvic pain, even in the absence of endometriosis.

The microbiome of the oral mucosa in irritable bowel syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

The study identified distinct oral microbiome profiles in IBS patients, particularly in those overweight, highlighting its potential as a non-invasive diagnostic tool for visceral pain severity and symptom progression.

What was studied?

The study examined the microbiome composition of the oral mucosa in patients with irritable bowel syndrome (IBS) compared to healthy controls. Specifically, it aimed to determine if distinct microbial shifts in the oral cavity could serve as non-invasive biomarkers for IBS diagnosis and symptom severity, particularly visceral pain. The researchers analyzed the buccal mucosal microbiome using PhyloChip microarrays to profile microbial richness, diversity, and composition.

Who was studied?

The study included 38 participants, comprising individuals diagnosed with IBS and healthy controls. Within the IBS group, participants were further classified based on body weight to explore associations between microbial diversity and symptom severity. Overweight IBS participants exhibited the most pronounced microbial shifts, highlighting the impact of both IBS and obesity on the oral microbiome.

What were the most important findings?

The study identified significant alterations in the oral microbiome of IBS patients, with particular emphasis on those who were overweight. Overweight IBS participants demonstrated decreased richness in the phylum Bacteroidetes and the genus Bacillus, while microbial diversity analyses revealed significant shifts in community structure. Analysis of β-diversity indicated a clear separation in microbial composition between overweight IBS patients and other groups. The oral microbiome of IBS participants showed marked increases in Enterobacteriaceae, Streptococcus, Corynebacterium, Pseudomonas, and Flavobacterium, with a strong correlation between these microbial changes and visceral pain severity. Notably, visceral pain in IBS patients was robustly associated with 60 operational taxonomic units (OTUs), 4 genera, 5 families, and 4 orders of bacteria. These correlations suggest that microbial perturbations in the oral cavity reflect systemic dysbiosis linked to symptom severity. Overweight IBS participants, in particular, exhibited a distinct oral microbial profile resembling dysbiosis patterns seen in both gastrointestinal and obesity-related conditions. The findings propose that the oral mucosa could serve as a practical, non-invasive substrate for diagnosing IBS and assessing symptom severity. Moreover, the stability of the oral microbiome compared to the gut highlights its potential as a reliable source for microbial information in IBS diagnostics.

ParameterFindings in IBS Patients
Microbial DiversityReduced richness in Bacteroidetes and Bacillus
Key Microbial ShiftsIncreased Enterobacteriaceae, Streptococcus, Corynebacterium, Pseudomonas, Flavobacterium
Beta DiversityMarked separation in overweight IBS participants
Pain CorrelationIncreased pain severity correlated with 60 OTUs, 4 genera, 5 families, and 4 orders
Diagnostic ImplicationsOral microbiome as a potential non-invasive biomarker for IBS severity and visceral pain

What are the greatest implications of this study?

This study's findings underscore the diagnostic potential of the oral microbiome in IBS, particularly in overweight patients. By identifying distinct microbial signatures linked to visceral pain, the research suggests that oral mucosal sampling could serve as a non-invasive method for diagnosing IBS and monitoring symptom progression. Unlike the gut microbiome, which can be influenced by various transient factors, the oral microbiome remains relatively stable, offering a consistent reflection of systemic microbial changes. This makes it an ideal candidate for longitudinal studies and patient monitoring. Furthermore, the study opens pathways for personalized therapeutic interventions targeting microbial imbalances in IBS patients, particularly those with weight-related symptom exacerbation. Future research could expand on these findings by exploring targeted microbial therapies and correlating oral dysbiosis with specific clinical outcomes

Understanding of the Site-Specific Microbial Patterns towards Accurate Identification for Patients with Diarrhea-Predominant Irritable Bowel Syndrome

May 20, 2025
  • Irritable Bowel Syndrome (IBS)
    Irritable Bowel Syndrome (IBS)

    Irritable Bowel Syndrome (IBS) is a common gastrointestinal disorder characterized by symptoms such as abdominal pain, bloating, and altered bowel habits. Recent research has focused on the gut microbiota's role in IBS, aiming to identify specific microbial signatures associated with the condition.

This study identifies site-specific microbial patterns in IBS-D patients, highlighting Bacteroides, Prevotella, and Oscillospira in the rectal mucosa as biomarkers. Findings suggest that rectal mucosa sampling may enhance diagnostic accuracy, paving the way for personalized microbiome-based treatments for IBS-D.

What Was Studied?

This study focused on understanding the site-specific microbial patterns within the intestinal tract of patients with diarrhea-predominant irritable bowel syndrome (IBS-D). Unlike traditional studies that primarily use fecal samples, this research employed a multi-site sampling strategy to analyze microbial communities at the duodenal mucosa (DM), duodenal lumen (DL), rectal mucosa (RM), and rectal lumen (RL) of IBS-D patients and healthy controls. The primary objective was to identify microbial biomarkers and site-specific microbial signatures that could enhance diagnostic accuracy for IBS-D.

Who Was Studied?

The study included 74 IBS-D patients and 20 healthy controls. A total of 283 samples were collected from four distinct intestinal sites (DM, DL, RM, RL), allowing for a comprehensive evaluation of microbial composition and diversity along the gastrointestinal tract. This site-specific sampling enabled the detection of unique microbial biomarkers that are potentially indicative of IBS-D pathology.

What Were the Most Important Findings?

The study revealed substantial microbial dysbiosis in IBS-D patients, characterized by site-specific microbial alterations that were not evident in healthy controls. In the duodenum, IBS-D patients exhibited higher abundances of Pseudomonas, Streptococcus, and Acinetobacter, whereas Burkholderia and Bacillus were dominant in healthy subjects. Rectal mucosa (RM) samples from IBS-D patients were particularly enriched with Bacteroides, Prevotella, and Oscillospira, which served as potential biomarkers for IBS-D. Notably, these site-specific microbial shifts were associated with symptom severity, including abdominal pain and bloating. The rectal mucosa community (RM) demonstrated a high predictive power for distinguishing IBS-D from healthy controls, with a Random Forest model achieving an AUC of 97.36%. Additionally, the co-abundance network analysis revealed decreased microbial connectivity in IBS-D patients, suggesting a loss of beneficial microbial interactions. The study concluded that rectal mucosa sampling is more diagnostically valuable than fecal samples for identifying IBS-D-specific microbial dysbiosis. This indicates that traditional fecal sampling may not adequately capture the microbial changes occurring within specific gut niches of IBS-D patients.

Intestinal SiteMicrobial Findings in IBS-D Patients
Duodenal Mucosa (DM)Increased abundances of Pseudomonas, Streptococcus, and Acinetobacter. Reduced levels of Burkholderia and Bacillus compared to healthy controls.
Duodenal Lumen (DL)Elevated levels of Enterococcus and Streptococcus, indicating localized dysbiosis in the small intestine.
Rectal Mucosa (RM)Significant enrichment of Bacteroides, Prevotella, and Oscillospira, identified as potential biomarkers for IBS-D.
Rectal Lumen (RL)Altered community structure with increases in pathogenic genera like Fusobacterium and reductions in Faecalibacterium.
Microbial ConnectivityDecreased co-abundance network connections, suggesting disrupted microbial interactions in IBS-D patients.
Diagnostic ValueRectal mucosa samples showed high predictive power for IBS-D diagnosis with an AUC of 97.36% in Random Forest models.

What Are the Greatest Implications of This Study?

The findings of this study underscore the critical importance of site-specific microbial sampling for the accurate diagnosis and characterization of IBS-D. The discovery of RM-specific biomarkers (Bacteroides, Prevotella, Oscillospira) suggests that diagnostic approaches could be significantly enhanced by targeting microbial signatures from the rectal mucosa rather than relying solely on fecal samples. This site-specific understanding opens the door for more precise microbiome-based diagnostics and targeted therapeutic strategies. The study also emphasizes the need for personalized medicine approaches that consider regional microbiota variability along the gastrointestinal tract, which could lead to better symptom management and improved patient outcomes.

Autoimmune Diseases

Go to Category Page

Alterations of the Gut Microbiota in Hashimoto's Thyroiditis Patients

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Hashimoto’s Thyroiditis
    Hashimoto’s Thyroiditis

    OverviewHashimoto’s Thyroiditis (HT) is an autoimmune disorder characterized by the progressive destruction of thyroid follicles due to chronic inflammation, often leading to hypothyroidism. It affects 10-12% of the global population, with a significantly higher prevalence among women​​. While its etiology involves genetic, environmental, and epigenetic factors, increasing evidence highlights the role of gut microbiota in […]

Gut microbiota analysis in Hashimoto’s thyroiditis patients identified significant dysbiosis, with increased pro-inflammatory taxa and reduced beneficial microbes, correlating with thyroid autoantibodies.

What Was Studied?

This study systematically investigated alterations in the gut microbiota composition in patients with Hashimoto’s thyroiditis (HT), an organ-specific autoimmune disease, compared to healthy controls. The researchers used 16S rRNA sequencing to profile and compare the gut microbiota of 50 HT patients and 27 matched healthy controls. The study aimed to identify microbial biomarkers associated with HT and their correlations with clinical parameters, such as thyroid peroxidase antibody (TPO-Ab) and thyroglobulin antibody (TG-Ab) levels.

Who Was Studied?

The study involved two cohorts: an exploration cohort of 28 HT patients and 16 healthy controls, and a validation cohort of 22 HT patients and 11 healthy controls. All participants were of Han Chinese ethnicity, aged between 18 and 65 years, and matched for age, sex, and BMI. Patients included were euthyroid and free from confounding conditions or recent medications that could affect the gut microbiota.

Key Findings

The study revealed significant differences in the gut microbiota composition between HT patients and healthy controls, though overall bacterial diversity and richness were similar. HT patients exhibited a marked increase in Firmicutes and a reduction in Bacteroidetes, with a significantly higher Firmicutes-to-Bacteroidetes (F/B) ratio. At the genus level, the abundances of Blautia, Roseburia, Ruminococcus_torques_group, and Eubacterium_hallii_group were significantly increased in HT patients. In contrast, beneficial genera like Bacteroides, Fecalibacterium, and Prevotella_9 were significantly decreased.

The researchers identified 27 genera with significant differences between HT patients and controls using linear discriminant analysis effect size (LEfSe). Ten genera, including Bacteroides and Fecalibacterium, were highlighted as potential biomarkers, achieving high diagnostic accuracy with AUC values of 0.91 and 0.88 in the exploration and validation cohorts, respectively.

Microbiota changes were correlated with clinical parameters. For instance, increased levels of Blautia and Dorea were positively associated with TPO-Ab and TG-Ab, while reduced levels of Fecalibacterium and Bacteroides correlated inversely with these antibodies.

Greatest Implications

The findings highlight the potential role of gut dysbiosis in the pathogenesis of HT. The observed microbial shifts suggest a loss of anti-inflammatory and barrier-supporting taxa, such as Fecalibacterium, and an increase in pro-inflammatory or mucin-degrading taxa, such as Ruminococcus_torques_group. This dysbiosis may contribute to immune activation and thyroid autoimmunity through mechanisms like increased intestinal permeability and molecular mimicry. Additionally, the identified microbial biomarkers could serve as non-invasive tools for HT diagnosis and disease monitoring. However, longitudinal studies and experimental validation are needed to confirm causality and explore therapeutic interventions targeting the gut microbiota.

Assessment of Thyroid Function and Oxidative Stress State in Foundry Workers Exposed to Lead

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

The study found that foundry workers exposed to lead had higher blood lead levels, increased thyroid hormones, and markers of oxidative stress compared to controls. These results indicate a significant oxidative-antioxidant imbalance due to lead exposure, stressing the need for better occupational health measures to prevent

What was studied?
The study assessed thyroid function and oxidative stress in foundry workers occupationally exposed to lead (Pb) dust and fumes. It investigated the correlation between blood lead levels (BLL) and thyroid hormones, as well as markers of oxidative stress.

Who was studied?
The study involved 59 adult male foundry workers exposed to lead and a control group of 28 male subjects with no history of lead exposure or thyroid abnormalities.

 

What were the most important findings?

Foundry workers had significantly higher blood lead levels (16.5±1.74 µg/dl) compared to the control group (12.8±1.16 µg/dl).

The exposed group exhibited significantly increased levels of free triiodothyronine (FT3) and free thyroxine (FT4), and decreased levels of thyroid stimulating hormone (TSH).

Markers of oxidative stress showed a significant increase in malondialdehyde (MDA) and a significant decrease in glutathione (GSH) among exposed workers.

A significant positive correlation was found between BLL and duration of employment, while a negative correlation existed between BLL and both TSH and GSH levels.

Elevated thyroid hormones were observed in 32.76% of the occupationally exposed workers.

There was a significant positive relationship between GSH and TSH, and between MDA and FT3 and FT4 among exposed workers.

 

What are the greatest implications of this study?
The study suggests that occupational exposure to lead dust and fumes can stimulate thyroid function, resulting in increased thyroid hormone levels, which may contribute to an oxidative-antioxidant imbalance. This imbalance, indicated by increased MDA and decreased GSH levels, underscores the potential health risks associated with prolonged exposure to lead, highlighting the need for improved protective measures and monitoring in industrial settings.

Beneficial Effects of a Low-Nickel Diet on Relapsing IBS-Like and Extraintestinal Symptoms of Celiac Patients during a Proper Gluten-Free Diet: Nickel Allergic Contact Mucositis in Suspected Non-Responsive Celiac Disease

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study provides strong evidence that nickel-rich foods in a gluten-free diet can trigger or exacerbate IBS-like gastrointestinal and extraintestinal symptoms in a subset of celiac disease patients, even when the disease is in serological and histological remission. The findings highlight the importance of nickel sensitivity in the pathogenesis of these relapsing symptoms, suggesting that a low-nickel diet can be a valuable intervention to improve patient outcomes. The use of the nickel oral mucosa patch test (Ni omPT) was also validated as a reliable diagnostic tool for identifying nickel-sensitive patients.

What was studied?

The study investigated the prevalence and effects of nickel allergic contact mucositis (Ni ACM) in celiac disease (CD) patients who were on a proper gluten-free diet (GFD) but experienced a relapse of Irritable Bowel Syndrome (IBS)-like gastrointestinal and extraintestinal symptoms. The main goal was to determine whether nickel-rich foods in a gluten-free diet could trigger these symptoms and to evaluate the impact of a low-nickel diet (LNiD) on symptom reduction in these patients. This pilot study also explored the use of the nickel oral mucosa patch test (Ni omPT) to diagnose nickel sensitivity in these patients.

Who was studied?

The study involved 102 consecutive adult celiac disease patients (74 females, 28 males, mean age 42.3 ± 7.4 years) who had been on a gluten-free diet for at least 12 months and were in serological and histological remission of their disease. These patients were selected because they reported relapsing or persisting gastrointestinal and extraintestinal symptoms despite proper adherence to a GFD. After exclusions based on comorbid conditions like lactose intolerance and Helicobacter pylori infection, 20 patients (all female, age 23–65 years) were included in the final analysis.

What were the most important findings?

In a cohort of 20 patients with persistent symptoms despite adherence to a gluten-free diet (GFD), all tested positive for nickel sensitivity via the nickel oral mucosa patch test (Ni omPT), confirming a diagnosis of nickel allergic contact mucositis (Ni ACM). Following prolonged GFD, 83.3% of patients experienced a relapse of symptoms, including abdominal pain, bloating, loose stools, and fatigue, coinciding with high dietary nickel intake from nickel-rich gluten-free foods such as corn. Implementing a low-nickel diet (LNiD) for three months improved 83.4% of total symptoms, with 41.7% reaching statistical significance. Notably, 80% of gastrointestinal and 88.9% of extraintestinal symptoms improved, including significant relief from abdominal pain, swelling, fatigue, and dermatitis. Combining LNiD with GFD restored patients' well-being to levels comparable to those previously achieved by GFD alone, strongly implicating nickel sensitivity as the primary driver of symptom relapse.

What are the greatest implications of this study?

This study provides strong evidence that nickel-rich foods in a gluten-free diet can trigger or exacerbate IBS-like gastrointestinal and extraintestinal symptoms in a subset of celiac disease patients, even when the disease is in serological and histological remission. The findings highlight the importance of nickel sensitivity (Ni ACM) in the pathogenesis of these relapsing symptoms, suggesting that a low-nickel diet (LNiD) can be a valuable intervention to improve patient outcomes. The use of the nickel oral mucosa patch test (Ni omPT) was also validated as a reliable diagnostic tool for identifying nickel-sensitive patients.

Clinically, these results imply that gastroenterologists should consider nickel sensitivity as a differential diagnosis in celiac patients who are non-responsive to a GFD and continue to experience symptoms. The integration of a low-nickel dietary approach alongside the GFD may become an essential part of managing non-responsive celiac disease with overlapping IBS-like symptoms. Further large-scale studies are needed to confirm these findings and refine dietary guidelines for managing nickel sensitivity in this population.

Comparative Analysis of Taxonomic and Functional Gut Microbiota Profiles in Relation to Seroconversion of Thyroid Peroxidase Antibodies in Euthyroid Participants.

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Hashimoto’s Thyroiditis
    Hashimoto’s Thyroiditis

    OverviewHashimoto’s Thyroiditis (HT) is an autoimmune disorder characterized by the progressive destruction of thyroid follicles due to chronic inflammation, often leading to hypothyroidism. It affects 10-12% of the global population, with a significantly higher prevalence among women​​. While its etiology involves genetic, environmental, and epigenetic factors, increasing evidence highlights the role of gut microbiota in […]

This study explored gut microbiota profiles in TPOAb-positive and TPOAb-negative euthyroid individuals. While no significant diversity differences were found, specific taxa like Desulfovibrionaceae were associated with TPOAb presence. Further research is needed to determine their role in autoimmune thyroid disease progression.

What was studied?

This study investigated the taxonomic and functional gut microbiota profiles of euthyroid individuals with and without thyroid peroxidase antibodies (TPOAb), a marker for autoimmune thyroid diseases such as Hashimoto’s thyroiditis. The goal was to assess whether gut microbiota composition differs in individuals with TPOAb before the clinical onset of autoimmune thyroid disease and to evaluate ethnic variations in thyroid biomarkers.

Who was studied?

The study examined 1,468 euthyroid participants aged 35 years and older from the multiethnic HELIUS cohort, including European Dutch, Moroccan, and Turkish individuals. Of these, 159 participants were TPOAb-positive, and 1,309 were TPOAb-negative. Fecal microbiota composition was analyzed using 16S rRNA sequencing.

What were the most important findings?

The study revealed no significant differences in global gut microbiota diversity (alpha or beta diversity) between TPOAb-positive and TPOAb-negative individuals. However, 138 microbial taxa were nominally associated with TPOAb presence, with 13 taxa consistently significant across multiple statistical methods. Among the most notable taxa, members of the Desulfovibrionaceae family were positively associated with TPOAb presence, while certain taxa from the Clostridiales vadin BB60 group were negatively associated. Functional pathway analysis indicated reduced abundance of pathways related to D-glucarate degradation, glycolysis, and adenosylcobalamin biosynthesis in TPOAb-positive participants, although none of these associations were statistically significant after correction for multiple testing. Ethnicity emerged as a more significant factor in microbiota variation than TPOAb status, with no ethnic differences in thyroid biomarker levels found.

What are the greatest implications of this study?

This study underscores the role of gut microbiota in the early stages of autoimmune thyroid disease, suggesting that microbial alterations may not be the primary driver of TPOAb seroconversion. However, the associations between specific taxa and TPOAb presence warrant further investigation to elucidate their potential involvement in disease progression. The lack of robust differences in microbiota composition between groups highlights the need for longitudinal studies to determine causal relationships between gut dysbiosis and autoimmune thyroiditis. Moreover, the findings emphasize the importance of considering ethnic diversity in microbiome research to ensure accurate interpretation of results.

Elevated Lactoferrin and Anti-Lactoferrin Antibodies in Endometriosis: Autoimmune and Microbiome Insights

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This study confirms elevated lactoferrin and anti-lactoferrin antibody levels in endometriosis, suggesting autoimmune involvement. Anti-lactoferrin drops post-surgery, hinting at a biomarker role, while lactoferrin ties to inflammation and potential microbiome links.

What Was Studied?

This study, conducted by Mori-Yamanaka et al. and published in Tohoku J. Exp. Med. in 2023, definitively explored serum lactoferrin (LTF) and anti-lactoferrin antibody (aLF) levels in patients with endometriosis. Endometriosis, a chronic inflammatory condition marked by ectopic endometrial-like tissue, remains poorly understood in terms of its underlying mechanisms. The researchers aimed to determine whether LTF, an iron-binding glycoprotein with antimicrobial and anti-inflammatory properties, and aLF, an autoantibody tied to immune dysregulation, play roles in the disease’s pathology. By measuring these markers in the blood of endometriosis patients compared to controls and assessing changes after surgical intervention, the study sought to uncover potential links to inflammation and autoimmunity. Although the study did not directly investigate microbiome signatures, LTF’s known role in modulating microbial environments suggests a possible indirect connection to gut or pelvic microbiome alterations in endometriosis.

Who Was Studied?

The research focused on 68 Japanese women undergoing surgery at Shiga University of Medical Science Hospital between November 2020 and May 2022. Of these, 51 had surgically and histopathologically confirmed endometriosis, spanning all stages (I-IV) per the revised American Society for Reproductive Medicine classification. The remaining 17 women, who underwent surgery for other gynecological issues like uterine myomas or benign ovarian tumors, served as controls without endometriosis. This cohort provided a robust sample to compare LTF and aLF levels across disease states and post-treatment outcomes, offering clinicians a clear demographic context for interpreting the findings.

What Were the Most Important Findings?

The study conclusively demonstrated that serum LTF and aLF levels are significantly elevated in endometriosis patients compared to controls, with p-values of 0.016 and 0.028, respectively. These elevations were particularly striking in advanced stages (III and IV), showing stronger statistical significance (LTF: p = 0.024; aLF: p = 0.016) compared to controls. Following surgery in 21 patients, aLF levels dropped markedly (p < 0.001), while LTF levels showed no significant change (p = 0.102). Notably, 43% of endometriosis patients exhibited aLF levels above the reference range, a prevalence akin to autoimmune conditions. Although microbiome data wasn’t directly assessed, LTF’s antimicrobial properties hint at potential microbial associations, possibly involving dysbiosis in the pelvic or gut microbiome, which could exacerbate inflammation in endometriosis. These findings position LTF and aLF as key players in the disease’s inflammatory and possibly autoimmune landscape.

What Are the Greatest Implications of This Study?

This study’s implications are profound for clinicians managing endometriosis. The elevated aLF levels, mirroring patterns in autoimmune diseases, strongly suggest that endometriosis involves an autoimmune component, potentially driven by immune responses to microbial or endogenous triggers. This insight could shift treatment paradigms toward immune-modulating therapies. Moreover, the significant post-surgical decline in aLF levels establishes it as a promising biomarker for monitoring disease activity and treatment success, offering a practical tool for clinical decision-making. While LTF’s role remains less clear, its persistence post-surgery and antimicrobial function imply a complex interplay with inflammation and possibly the microbiome, warranting further investigation into microbial signatures like those of Lactobacillus or Prevotella, known to influence pelvic health. Despite the study’s limitations—its small sample and surgical focus—these findings pave the way for innovative diagnostics and therapies, urging clinicians to consider immune and microbial factors in endometriosis care.

Exploring the Bidirectional Link Between Graves’ Disease and Gut Microbiome: New Insights Into the Thyroid–Gut Axis

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This study confirms a bidirectional causal relationship between Graves’ Disease and the gut microbiome. Key taxa like Deltaproteobacteria elevate GD risk, while others, such as Anaerostipes, are protective. These findings advance our understanding of the thyroid-gut axis and suggest microbiome-targeted interventions for GD.

What was studied?

This study investigated the bidirectional causal relationship between Graves’ Disease (GD) and the gut microbiome. Utilizing Mendelian randomization (MR), it examined how alterations in the gut microbiome might influence GD and vice versa, supporting the thyroid–gut axis (TGA) concept. Genome-wide association study (GWAS) summary datasets, which analyze millions of genetic variants across diverse populations to identify associations between genetic markers and specific traits, were sourced from international consortiums to evaluate these interactions.

Who was studied?

The study involved two large datasets. Gut microbiome data included 18,340 samples spanning diverse ethnic groups (European, Middle Eastern, East Asian, Hispanic/Latin American, and African American), while GD data included 212,453 samples of Asian ethnicity, sourced from Biobank Japan. These comprehensive datasets were analyzed to identify instrumental variables linking genetic variants to gut microbiome composition and GD susceptibility.

What were the most important findings?

The study established a bidirectional causal relationship between Graves’ disease (GD) and the gut microbiome, identifying key microbial associations that act as either risk or protective factors. Risk factors for GD included the classes Deltaproteobacteria (odds ratio [OR] = 3.603) and Mollicutes, as well as the genera Ruminococcus torques group, Oxalobacter, and Ruminococcaceae UCG 011. Protective associations were observed for the family Peptococcaceae and the genus Anaerostipes (OR = 0.489). Furthermore, GD was found to alter gut microbiome composition, increasing the abundance of genera like Anaerofilum (OR = 1.584) and reducing taxa such as the Clostridium innocuum group (OR = 0.918) and Sutterella (OR = 0.953). These findings highlight the regulatory activity of the thyroid–gut axis (TGA) and provide strong evidence for its involvement in GD pathogenesis.

What are the greatest implications of this study?

The findings underscore the critical role of the gut microbiome in GD pathogenesis and its reciprocal interaction with thyroid health. Identifying specific microbial taxa as risk or protective factors offers actionable insights for microbiome-targeted interventions (MBTIs), such as probiotics or dietary modifications, tailored to mitigate GD risk or progression. The bidirectional relationship between GD and the gut microbiome highlights the need for integrated approaches addressing both thyroid and gut health. These results could guide the development of precision medicine strategies, leveraging the gut microbiome to modulate immune responses and improve clinical outcomes for patients with GD. This research also establishes a foundational understanding of major microbial associations (MMAs) within the TGA, paving the way for future therapeutic innovations. Further, this study establishes a methodological precedent for using Mendelian Randomization to discern causal effects in microbiome-related research.

Gut microbiota in early pediatric multiple sclerosis: a case−control study

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This study identified significant gut microbiota dysbiosis in pediatric MS, with increased pro-inflammatory taxa and metabolic shifts. Findings suggest early microbial perturbations may contribute to disease pathogenesis.

What was studied?

This study explored the gut microbiota of children diagnosed with early-onset pediatric multiple sclerosis (MS) and compared it to controls of similar age and sex. The researchers aimed to identify gut microbial community differences, including taxonomic and functional perturbations, and examined the influence of immunomodulatory drug (IMD) exposure. This study also predicted functional metabolic pathways based on microbial profiles.

Who was studied?

The study involved 18 children with relapsing-remitting multiple sclerosis (RRMS) and 17 healthy controls. The participants, aged 4 to 18 years, were enrolled from a University of California, San Francisco pediatric clinic. MS cases were within two years of symptom onset, with half being IMD-naïve. Both groups were matched by age and sex, with controls lacking autoimmune conditions or recent antibiotic exposure.

What were the most important findings?

The study revealed significant microbial differences between pediatric MS cases and controls. MS cases exhibited an enrichment in pro-inflammatory taxa, including Desulfovibrionaceae (e.g., Bilophila, Desulfovibrio) and Christensenellaceae, and a depletion of anti-inflammatory taxa such as Lachnospiraceae and Ruminococcaceae. Additionally, metabolic pathways related to glutathione metabolism were enriched in MS cases, regardless of IMD exposure. Notably, IMD exposure correlated with reduced beta diversity variations, suggesting partial modulation of the microbiome toward a more control-like composition. Furthermore, the study observed shifts in microbial genes involved in lipopolysaccharide biosynthesis and immune modulation, linking gut dysbiosis with potential mechanisms of neuroinflammation and neurodegeneration.

What are the greatest implications of this study?

This study highlights the potential role of gut microbiota in the early pathogenesis of pediatric MS. The observed microbial dysbiosis aligns with a pro-inflammatory milieu that may contribute to immune dysregulation in MS. The findings underscore the importance of gut-targeted interventions, such as dietary modifications or probiotics, as potential therapeutic strategies. The results also emphasize the need for longitudinal studies to elucidate causative versus consequential relationships between gut dysbiosis and MS development.

Integrative analysis of gut microbiome and host transcriptome reveal novel molecular signatures in Hashimoto's thyroiditis

May 20, 2025
  • Hashimoto’s Thyroiditis
    Hashimoto’s Thyroiditis

    OverviewHashimoto’s Thyroiditis (HT) is an autoimmune disorder characterized by the progressive destruction of thyroid follicles due to chronic inflammation, often leading to hypothyroidism. It affects 10-12% of the global population, with a significantly higher prevalence among women​​. While its etiology involves genetic, environmental, and epigenetic factors, increasing evidence highlights the role of gut microbiota in […]

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This study revealed novel molecular signatures linking gut microbiota and transcriptome in Hashimoto's thyroiditis, advancing diagnostic and therapeutic approaches.

What Was Studied?

Integrative analysis reveals novel gut microbiota-transcriptome signatures for Hashimoto's thyroiditis, aiding early diagnosis and treatment.This study explored the molecular signatures of Hashimoto’s thyroiditis (HT) through an integrative analysis of gut microbiome and host transcriptome (miRNA/mRNA). It aimed to identify novel molecular markers and elucidate the gut-thyroid axis, using data from 31 early HT patients and 30 healthy controls across discovery and validation cohorts. The study sought to uncover interactions between the gut microbiota and host gene expression, providing insights into HT pathogenesis.

Who Was Studied?

Participants included 31 early HT patients and 30 healthy individuals aged 18–65. HT cases were defined by elevated thyroid antibodies (TPOAb/TGAb) and morphological abnormalities while maintaining normal thyroid function. Exclusions included antibiotic or probiotic use, significant dietary changes, or comorbid conditions. Blood and fecal samples were collected for transcriptomic and metagenomic sequencing.

What Were the Most Important Findings?

The study identified subtle but significant gut microbiota alterations in early HT patients. While alpha diversity was unchanged, beta diversity analysis revealed compositional shifts, including increased Bacillota_A and Spirochaetota at the phylum level and significant differences in 24 genera and 67 species. Beneficial microbes like Barnesiella intestinihominis were reduced, while opportunistic pathogens like Peptostreptococcus were enriched. Host transcriptome analysis identified 1975 downregulated and 1821 upregulated mRNAs, alongside 27 miRNAs. Immune and inflammation-related pathways were enriched, with hsa-miR-548aq-3p and hsa-miR-374a-5p playing key roles. Key molecular signatures included three bacterial species (Salaquimonas_sp002400845, Clostridium_AI_sp002297865, Enterocloster_citroniae) and six RNAs (e.g., GADD45A, IRS2, SMAD6). These integrated signatures demonstrated strong diagnostic potential (AUC=0.95) in distinguishing HT patients from healthy controls.

What Are the Greatest Implications?

This research advances understanding of the gut-thyroid axis and provides a robust framework for early HT diagnosis and treatment. Molecular signatures identified offer potential for targeted therapies, including microbiome modulation. For example, restoring beneficial microbes such as Barnesiella intestinihominis or targeting specific pathogenic species may offer therapeutic benefits. Integration of gut microbiota and transcriptome data sets a precedent for multidimensional biomarker development in autoimmune conditions.

Mercury and nickel allergy/ Risk factors in fatigue and autoimmunity

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

Hypersensitivity to mercury and nickel was significantly more common in fatigued and autoimmune patients than in healthy controls. Removal of dental metals reversed symptoms and immune activation, suggesting that metal-driven immune dysregulation may underlie fatigue and autoimmunity.

What was studied?

This observational study examined the relationship between hypersensitivity to metals—specifically mercury and nickel—and the prevalence of chronic fatigue and autoimmune disorders. The research utilized the MELISA® (Memory Lymphocyte ImmunoStimulation Assay) to assess in vitro lymphocyte reactivity to various metals among patients with autoimmune thyroiditis, fatigue without endocrinopathy, and occupational exposure to dental metals. The central goal was to evaluate whether metal hypersensitivity constitutes a risk factor for fatigue and autoimmunity and whether removal of metal exposures (e.g., dental amalgam) could reverse symptoms.

Who was studied?

The study analyzed 72 fatigued patients divided into three primary groups: (1) 22 patients with autoimmune thyroiditis, including some with autoimmune polyglandular syndrome (APS); (2) 28 fatigued individuals without endocrinopathies, many of whom experienced local or systemic symptoms linked to dental alloys; and (3) 22 fatigued professionals with long-term occupational metal exposure, including dentists and technicians. A control group of 13 healthy, fatigue-free individuals without autoimmunity was also included. Lymphocyte reactivity to 17 metals was measured using the MELISA® test. Additionally, two patients underwent dental amalgam replacement and were followed for symptom changes and immune reactivity post-intervention.

What were the most important findings?

Lymphocyte stimulation indices revealed that reactivity to inorganic mercury and nickel was significantly elevated in all fatigued patient groups compared to healthy controls. Specifically, 72.7% of autoimmune thyroiditis patients and 61.1% reacted to mercury and nickel, respectively, with similar trends in the other fatigued cohorts. This pattern was not seen with most other metals, suggesting a specific immunologic sensitivity to mercury and nickel in this population. Notably, healthy controls exhibited no reactivity to nickel and minimal reactivity to mercury. Two representative case studies illustrated that removal of metal-containing dental restorations significantly reduced both symptom burden and lymphocyte reactivity in follow-up MELISA® tests. One patient, initially disabled due to fatigue and autoimmune comorbidities, returned to work and experienced sustained health improvements after amalgam replacement.

These findings imply a mechanistic link between metal-driven immune activation and chronic fatigue, potentially via inflammatory disruption of the hypothalamic-pituitary-adrenal (HPA) axis. Nickel exposure, often underestimated, appeared especially important, with sensitization possibly enhanced by environmental and occupational exposures (e.g., stainless steel, dental alloys, cigarette smoke). The results further underscore the inadequacy of patch testing alone in detecting systemic hypersensitivity and support the utility of MELISA® as a diagnostic adjunct.

What are the greatest implications of this study?

This paper provides early, compelling evidence that hypersensitivity to mercury and nickel may play a causative role in the symptomatology of fatigue, autoimmune disorders, and related syndromes like chronic fatigue syndrome (CFS). It introduces the concept that these metals can induce systemic immune activation, not merely local contact dermatitis, thereby contributing to dysregulation of the HPA axis and the development or exacerbation of autoimmunity. Clinically, the study supports consideration of metal sensitization in patients with unexplained fatigue or autoimmune disease, and it opens the door to therapeutic strategies involving metal detoxification or elimination—particularly amalgam removal. Furthermore, the study highlights MELISA® testing as a superior method for detecting metal sensitization compared to standard patch tests, particularly in systemic presentations. This work is foundational in drawing attention to metallomic contributors to chronic inflammatory and autoimmune conditions.

Meta-analysis of gut microbiome studies identifies disease-specific and shared responses

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This meta-analysis standardized and re-analyzed data from 28 gut microbiome studies across ten diseases, identifying consistent microbiome signatures associated with specific diseases and a non-specific response common to multiple conditions. Key findings suggest both potential microbial diagnostics and treatments, emphasizing the importance of understanding shared versus disease-specific microbial responses in future research and clinical applications.

What was studied?

The meta-analysis focused on the human gut microbiome’s association with various diseases by analyzing 28 published case-control gut microbiome studies covering ten diseases. The researchers aimed to standardize the processing and analysis of these datasets to identify consistent patterns and shifts in the gut microbiome associated with specific diseases or a generalized health-disease spectrum.

Who was studied?

The participants of the original case-control studies comprised individuals with different diseases, including colorectal cancer, inflammatory bowel disease (IBD), and others, alongside control groups of healthy individuals. The meta-analysis integrated data only from studies with publicly available 16S amplicon sequencing data of stool samples from at least 15 case patients, excluding studies focused solely on children under 5 years old.

 

What were the most important findings?

Consistent Microbial Patterns: The meta-analysis revealed consistent and specific microbiome changes associated with various diseases. For instance, diseases like colorectal cancer showed an enrichment of pathogenic bacteria, while a depletion of health-associated bacteria marked conditions like IBD.

Non-Specific Microbial Responses: A significant finding was that many microbial associations are not disease-specific but rather indicate a non-specific response shared across multiple disease states. Approximately half of the genera identified were common to more than one disease, suggesting a generalized microbial response to disease states rather than unique disease-specific signatures.

Diagnostic and Therapeutic Implications: The study identified distinct categories of dysbiosis (microbial imbalance) that could guide the development of microbiome-based diagnostics and therapeutics. For example, enriching for depleted beneficial microbes could be a strategy for diseases characterized by such depletions.

 

What are the greatest implications of this meta-analysis?

Improved Disease Understanding: By providing a clearer picture of the microbiome’s role in various diseases, the study helps refine our understanding of disease mechanisms and potential microbial contributions to disease processes.

Guidance for Future Research: The findings suggest that future microbiome research in disease contexts should consider the non-specificity of many microbial changes. This realization could influence how researchers design studies and interpret results, potentially focusing on truly disease-specific microbial signatures.

Clinical Applications: The identification of consistent microbial patterns and signatures across diseases opens pathways to developing novel diagnostics and therapies, such as probiotics or fecal microbiota transplants, targeted at restoring healthy microbial communities or addressing specific dysbioses.

Data Sharing and Standardization: The study underscores the value of making raw data and metadata from microbiome studies publicly available and highlights the benefits of using standardized methods for data processing and analysis to compare and integrate results across studies.

Overall, this meta-analysis clarifies the microbiome’s role in disease and sets a framework for future research and clinical applications by demonstrating the importance of understanding both disease-specific and non-specific microbial responses.

Molecular Alteration Analysis of Human Gut Microbial Composition in Graves' disease Patients

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This study shows significant alterations in gut microbiota diversity in Graves' disease (GD) patients, with increased Prevotellaceae and Pasteurellaceae and decreased Enterobacteriaceae. Findings support gut microbial dysbiosis in GD, potentially contributing to its pathogenesis and informing new treatments.

What was studied?
The study investigated the gut microbial composition in patients with Graves’ disease (GD) compared to healthy controls.

 

Who was studied?
The study involved 27 GD patients and 11 healthy controls, with fecal samples collected for analysis.

 

What were the most important findings?

The association between gut microbiota and host homeostasis is pivotal for understanding various diseases, including autoimmune disorders like Graves’ disease (GD), characterized by hyperthyroidism and ophthalmopathy. This study hypothesized that gut bacteria play a significant role in GD pathogenicity. To investigate this, the intestinal bacterial composition of 27 GD patients and 11 healthy controls was analyzed using PCR-DGGE of the 16S rRNA gene targeting the V3 region and Real-time PCR for specific bacterial groups. High-throughput sequencing of the 16S rRNA gene (V3+V4 regions) was performed on randomly selected samples using the Hiseq2500 platform.

The results revealed a lower diversity of intestinal bacteria in GD patients compared to controls. Statistical analyses indicated significant alterations in bacterial phyla, with a higher relative abundance of Prevotellaceae and Pasteurellaceae, and a lower abundance of Enterobacteriaceae, Veillonellaceae, and Rikenellaceae in GD patients. At the genus level, Prevotella_9 and Haemophilus were significantly increased, whereas Alistipes and Faecalibacterium were decreased in GD patients. Notably, the species Haemophilus parainfluenza was more abundant in GD patients.

 

What are the greatest implications of this study?
The findings support the hypothesis of gut microbial dysbiosis in GD, suggesting that changes in the gut microbiota may contribute to the disease’s pathogenesis. These insights could pave the way for novel therapeutic approaches targeting gut microbiota in GD treatment.

 

 

Molecular estimation of alteration in intestinal microbial composition in Hashimoto's thyroiditis patients

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Hashimoto’s Thyroiditis
    Hashimoto’s Thyroiditis

    OverviewHashimoto’s Thyroiditis (HT) is an autoimmune disorder characterized by the progressive destruction of thyroid follicles due to chronic inflammation, often leading to hypothyroidism. It affects 10-12% of the global population, with a significantly higher prevalence among women​​. While its etiology involves genetic, environmental, and epigenetic factors, increasing evidence highlights the role of gut microbiota in […]

This study revealed novel molecular signatures linking gut microbiota and transcriptome in Hashimoto's thyroiditis, advancing diagnostic and therapeutic approaches.

What Was Studied?

This study examined alterations in the gut microbiota composition of patients with Hashimoto's thyroiditis (HT). It aimed to investigate the relationship between intestinal dysbiosis and HT through quantitative and qualitative analysis of gut microbial diversity and composition using techniques such as PCR-DGGE, real-time PCR, and pyrosequencing of 16S rRNA genes.

Who Was Studied?

The study analyzed fecal samples from 29 HT patients and 12 healthy individuals aged 40–60 years. Patients were diagnosed based on elevated thyroid antibodies (TPOAb and TGAb) and other clinical markers, including TSH and T4 levels. Healthy controls had normal thyroid function and no history of antibiotic or probiotic use in the 60 days preceding the study.

What Were the Most Important Findings?

The study revealed significant gut microbiota dysbiosis in Hashimoto’s thyroiditis (HT) patients compared to healthy controls. HT patients exhibited an increased abundance of inflammatory phyla like Proteobacteria and decreased beneficial phyla such as Firmicutes and Bacteroidetes. At the genus level, Escherichia-Shigella and Parasutterella were elevated, while anti-inflammatory genera such as Prevotella_9 and Dialister were significantly reduced. Escherichia coli was particularly overrepresented, potentially contributing to intestinal barrier disruption and inflammation linked to thyroid autoimmunity.

Real-time PCR showed significant reductions in Bifidobacterium and Lactobacillus, essential for producing immune-regulating SCFAs, while alpha diversity indicated bacterial overgrowth in HT patients. Functional diversity measures showed no significant changes, pointing to microbial imbalance rather than increased functional diversity. Pyrosequencing confirmed these findings, demonstrating a distinct microbial profile in HT patients. These results highlight the role of gut dysbiosis in HT pathogenesis and suggest potential therapeutic strategies targeting microbiome restoration.

What Are the Greatest Implications?

This study highlights gut microbiota dysbiosis as a potential contributor to the pathogenesis of HT. The findings suggest that the overrepresentation of inflammatory and opportunistic pathogens, such as Escherichia coli and Escherichia-Shigella, coupled with the reduction of beneficial microbes like Bifidobacterium and Lactobacillus, may influence immune regulation and thyroid autoimmunity. Restoring microbial balance through probiotics, dietary interventions, or targeted microbiome therapies could serve as novel strategies for managing HT. These results underscore the critical role of gut health in autoimmune diseases and provide a foundation for developing microbiome-targeted interventions.

Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This case-control study investigates the gut microbiota's role in multiple sclerosis (MS) pathogenesis by comparing the fecal microbiota of relapsing-remitting MS patients to healthy controls. Findings reveal significant microbial dysbiosis in MS patients, highlighting differences in the abundance of specific bacterial genera, supporting the gut microbiota's involvement in MS etiology.

What was studied?

The research focused on investigating the potential role of gut microbiota in the pathogenesis of Multiple Sclerosis (MS), particularly relapsing-remitting MS (RRMS). It aimed to compare the fecal microbiota composition between RRMS patients and healthy controls, analyze the microbial diversity, and assess the predictive power of microbiota profiles in distinguishing disease status.

Who was studied?

The study included 31 RRMS patients, categorized based on their disease phase (active or in remission), and 36 age- and sex-matched healthy controls. The RRMS patients were between 18 and 80 years of age, met the McDonald diagnostic criteria for MS, and had an Expanded Disability Status Scale (EDSS) score between 1 and 6. The selection criteria excluded individuals with prior significant surgeries, current antibiotic or probiotic use, or a history of autoimmune diseases other than MS.

What were the most important findings?

Distinct Microbial Community Profiles: RRMS patients had significantly different gut microbiota compositions compared to healthy controls, with specific genera such as Pseudomonas, Pedobacter, Blautia, and Dorea showing higher abundance in RRMS patients, while genera like Adlercreutzia, Parabacteroides, and Lactobacillus were more abundant in controls.

Species Richness and Diversity: Active disease phase was associated with a trend towards lower species richness compared to healthy controls, while remission phase microbiota exhibited similar species richness to controls.

Predictive Power of Gut Microbiota: Using Random Forests (RF) and operational taxonomic unit (OTU) profiles, the study achieved significant classification accuracy in distinguishing RRMS patients from healthy controls based on gut microbiota composition.

Functional Implications: The functional analysis suggested alterations in pathways related to fatty acid metabolism, defense mechanisms, and glycolysis, indicating a broader impact of gut microbiota dysbiosis on metabolic functions.

What are the greatest implications of this study?

The findings underscore the importance of gut microbiota in the etiology and pathogenesis of RRMS, suggesting that dysbiosis may not only be a marker of the disease but also potentially contribute to its development and progression. These results open avenues for future research to explore gut microbiota as a therapeutic target or biomarker for MS. Understanding the specific roles of altered microbiota and their metabolic pathways could lead to new interventions to modulate the gut microbiome to manage or prevent MS. Moreover, the predictive model based on gut microbiota composition presents a novel approach for identifying individuals at risk of RRMS, offering the potential for early intervention and personalized treatment strategies.

Role of Cholestyramine in Refractory Hyperthyroidism: A Case Report and Literature Review

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

A 52-year-old woman with refractory iodine-induced hyperthyroidism showed significant improvement with cholestyramine, reducing FT4 by 30% in 5 days. Despite conventional treatments failing, cholestyramine proved effective, leading to euthyroidism. This highlights cholestyramine's potential as an adjunct therapy.

What was studied?

The study investigated the role of cholestyramine as an additional treatment for refractory iodine-induced hyperthyroidism in a patient who did not respond to conventional therapies.

Who was studied?

A 52-year-old female patient with a history of goiter who developed iodine-induced hyperthyroidism following a CT scan with contrast. The patient had obstructive symptoms and was unresponsive to standard treatments, including dexamethasone, carbimazole, and propranolol.

What were the most important findings?

After adding cholestyramine, the patient’s FT4 levels decreased by 30% within 5 days and normalized by 12 days.

What are the greatest implications of this study?

Cholestyramine can be an effective adjunct therapy for managing refractory iodine-induced hyperthyroidism, suggesting a potential new treatment avenue for similar cases, such as Grave's Disease (GD). This case highlights the need for alternative treatments when conventional therapies fail and emphasizes the utility of cholestyramine in rapid thyroid hormone reduction.

Role of Cholestyramine in Refractory Hyperthyroidism A Case Report and Literature Review

Serendipity in Refractory Celiac Disease: Full Recovery of Duodenal Villi and Clinical Symptoms after Fecal Microbiota Transfer

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

A patient with refractory celiac disease type II achieved complete duodenal villi recovery and symptom resolution after fecal microbiota transfer. This unexpected outcome suggests that microbiome manipulation may offer a novel treatment for RCD II, providing an alternative to immunosuppressive therapies for this challenging condition.

What was studied?

This study examined the impact of fecal microbiota transfer (FMT) on a patient with refractory celiac disease type II (RCD II). The patient initially received FMT as treatment for recurrent Clostridium difficile infection (CDI), but the intervention unexpectedly resulted in full recovery of duodenal villi and resolution of celiac symptoms, suggesting a potential therapeutic role for microbiome manipulation in RCD II.

Who was studied?

A 68-year-old woman with a 10-year history of RCD II was the subject of this study. Despite adherence to a strict gluten-free diet, she experienced persistent villous atrophy and malabsorption. She had been receiving budesonide therapy and later underwent cladribine treatment, neither of which alleviated her condition. The patient was repeatedly hospitalized due to severe diarrhea, dehydration, and infections, and was ultimately treated with FMT for recurrent CDI.

What were the most important findings?

FMT not only resolved the patient’s CDI but also led to complete histological recovery of the duodenal mucosa. Before FMT, duodenal biopsies confirmed villous atrophy (Marsh IIIA) and an abnormal intraepithelial lymphocyte population (>80%). However, post-FMT, the patient experienced significant clinical improvement, gaining weight and becoming symptom-free. Follow-up biopsies at six months showed full villous recovery (Marsh 0), although 71% of intraepithelial lymphocytes remained aberrant.

Microbiome analysis of the FMT donor revealed a high Shannon diversity index (3.81), suggesting a diverse and resilient microbial community. Unfortunately, due to the lack of a pre-FMT stool sample from the patient, a direct comparison of microbiome shifts could not be conducted. However, the resolution of symptoms and histological improvement strongly indicate that microbiome alterations played a role in disease modulation. Given previous studies implicating gut dysbiosis in celiac disease pathogenesis, this case highlights a possible causal role of microbiota in maintaining the chronic inflammatory state of RCD II.

What are the greatest implications of this study?

This study provides compelling evidence that microbiome manipulation may be a viable therapeutic strategy for RCD II, a condition with limited treatment options and high mortality risk. The findings suggest that gut dysbiosis could be a key driver of persistent villous atrophy in RCD II and that FMT may help restore intestinal homeostasis. If confirmed in larger studies, this could shift the treatment paradigm for RCD II, potentially offering an alternative to immunosuppressive therapies or autologous stem cell transplantation. Given the poor prognosis associated with RCD II, the ability to restore mucosal integrity through microbiome-targeted interventions represents a significant advancement. Further research should explore optimal donor selection, microbial composition, and long-term effects of FMT in RCD II patients.

Shared Gut Microbiota Dysbiosis in Immune-Mediated Diseases: Insights and Biomarkers

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Multiple Sclerosis (MS)
    Multiple Sclerosis (MS)

    OverviewIn the past decade, research has shown that the enormous community of microbes that live in the gut, known as the gut microbiota, are closely linked to human health and disease. This relationship is primarily due to the gut microbiota’s impact on systemic immune responses. There is growing evidence that these impacts on immune function are […]

The study identifies gut microbial dysbiosis and specific taxa associated with Crohn's disease, ulcerative colitis, multiple sclerosis, and rheumatoid arthritis. Findings support common microbial markers across IMIDs and their diagnostic potential.

What was studied?

This study explored the gut microbiota composition in patients with immune-mediated inflammatory diseases (IMIDs) such as Crohn’s disease (CD), ulcerative colitis (UC), multiple sclerosis (MS), and rheumatoid arthritis (RA) compared to healthy controls (HC). Using 16S rRNA gene sequencing, researchers assessed microbial diversity, richness, and specific taxonomic biomarkers to identify common and unique microbial features across these IMIDs. Machine learning techniques were applied to differentiate microbial patterns between diseases and controls further.

Who was studied?

The cohort included 79 patients across the four IMIDs (20 with CD, 19 with UC, 19 with MS, 21 with RA) and 23 healthy controls. Participants were adults, not on antibiotics for at least eight weeks, and recruited from clinical centers in Canada. Stool samples were collected twice within a two-month interval for analysis.

What were the most important findings?

The study revealed significant microbial dysbiosis in all IMIDs compared to healthy controls. Richness and diversity were lowest in CD and highest in HCs. Taxa such as Actinomyces, Eggerthella, and Streptococcus were enriched in disease cohorts, while beneficial taxa like Roseburia and Gemmiger were depleted. Disease-specific patterns were also identified: Intestinibacter in CD, Bifidobacterium in UC, and unclassified Erysipelotrichaceae in MS. Machine learning highlighted microbial signatures capable of differentiating diseases from HC, with the best classification accuracy observed in CD versus HC (AUC = 0.95).

Key Findings

Microbial Diversity and Richness: Patients with IMIDs exhibited reduced gut microbial richness and diversity compared to HCs, with CD showing the lowest diversity.

Common Dysbiosis Across IMIDs: Certain taxa, such as Actinomyces, Eggerthella, Faecalicoccus, and Streptococcus, were consistently enriched in IMID patients, while Gemmiger, Lachnospira, and Sporobacter were depleted across all disease cohorts.

Disease-Specific Microbiota Signatures: Intestinibacter was elevated in CD. Bifidobacterium was enriched in UC. Erysipelotrichaceae was more abundant in MS. Roseburia was significantly reduced in RA.

Machine Learning Classification: Machine learning models effectively distinguished between IMID and HC cohorts, with the highest classification accuracy for CD (AUC ~0.95). Features like Gemmiger (elevated in HCs) and Faecalicoccus (elevated in IMIDs) were identified as significant markers.

Gram-Positive Focus: The study highlighted an unusually low abundance of Gram-negative bacteria, focusing analysis on Gram-positive taxa, which still yielded meaningful insights into IMID-specific dysbiosis.

What are the greatest implications of this study?

The findings underscore the potential of gut microbiota as diagnostic biomarkers for IMIDs. Shared microbial patterns suggest a common dysbiotic component in IMID etiology, while distinct taxa provide insight into disease-specific mechanisms. This research highlights the importance of the gut microbiome in IMID pathogenesis and opens avenues for microbiome-targeted interventions (MBTIs).

The Comorbidity of Endometriosis and Systemic Lupus Erythematosus: A Systematic Review

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This review explores the significant comorbidity between systemic lupus erythematosus (SLE) and endometriosis, emphasizing shared pathological pathways.

DOI: 10.7759/cureus.42362

What Was Reviewed?

This systematic review examined the comorbidity between endometriosis and systemic lupus erythematosus (SLE), two chronic conditions with significant implications for women's health. The review aimed to elucidate the prevalence, shared pathophysiological mechanisms, and risk factors linking these diseases, emphasizing immune dysregulation, genetic predispositions, and hormonal influences. The review synthesized findings from nine studies conducted between 2011 and 2021, including case-control, cohort, and systematic review methodologies.

Who Was Reviewed?

The review focused on studies of females aged 12-60, representing the pubertal to postmenopausal age range. The population comprised patients with diagnosed endometriosis and SLE. The studies predominantly included participants from diverse ethnicities and geographies, screened based on standardized inclusion criteria to establish the prevalence and interaction of these conditions.

What Were the Most Important Findings?

The review confirmed a statistically significant correlation between endometriosis and SLE, with women diagnosed with either condition at a heightened risk of developing the other. The findings implicated immune dysregulation, characterized by diminished cytotoxic T-cell activity and elevated humoral immune responses, as a central mechanism. Notable microbial associations include increased systemic inflammation mediated by cytokines such as interleukin-1, interleukin-6, and tumor necrosis factor (TNF-α). Genetic factors also played a role, with gene loci such as PTPN22 associated with increased susceptibility to both conditions. Surgical interventions like hysterectomy were linked to increased inflammation and subsequent autoimmune activation, while modified surgical techniques showed promise in mitigating risk.

What Are the Greatest Implications of This Review?

This review highlights the necessity for clinicians to adopt an interdisciplinary approach when managing patients with either endometriosis or SLE, as their comorbidity exacerbates disease burden and complicates treatment. It emphasizes the importance of targeted therapies to modulate immune response alongside careful evaluation of surgical and hormonal treatment strategies to minimize adverse outcomes. The findings suggest a potential for incorporating microbial and genetic markers into diagnostic and therapeutic protocols to improve outcomes.

The gut microbiota and endometriosis: From pathogenesis to diagnosis and treatment

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

The review explored the connection between gut microbiota and endometriosis, highlighting potential influences on disease mechanisms through hormonal, immune, and inflammatory pathways. It suggests that modulating gut microbiota could lead to innovative diagnostic and therapeutic approaches for endometriosis. This review further suggests that via Fecal Microbiota Transplantation (FMT) may provide a novel therapeutic approach for the clinical treatment of endometriosis.

What was reviewed?

The study reviewed the relationship between the gut microbiota and endometriosis, focusing on how the gut microbiota may influence the pathogenesis, diagnosis, and potential treatment of endometriosis through various mechanisms such as estrogen modulation, immune response, and inflammation.

 

Who was reviewed?

The review did not involve specific individuals as subjects of study but instead synthesized findings from various studies that investigate the gut microbiota’s involvement in patients with endometriosis. It includes analysis of microbial profiles and their correlations with the disease.

 

What were the most important findings?

Important findings highlighted that alterations in the gut microbiota are associated with endometriosis and could potentially influence the disease’s pathogenesis through mechanisms linked to hormonal balance, immune modulation, and inflammatory responses. Specifically, changes in bacterial diversity and specific bacterial groups (such as an increase in the Firmicutes/Bacteroidetes ratio) were noted in patients with endometriosis.

 

What are the greatest implications of this review?

The review suggests that targeting the gut microbiota might offer new strategies for the diagnosis and treatment of endometriosis. Understanding the role of the gut microbiota in endometriosis could lead to non-invasive diagnostic biomarkers and novel therapeutic approaches that involve modulating the gut microbiota through diet, probiotics, or even fecal microbiota transplantation.

Hashimoto’s Thyroiditis

Go to Category Page

Alterations of the Gut Microbiota in Hashimoto's Thyroiditis Patients

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Hashimoto’s Thyroiditis
    Hashimoto’s Thyroiditis

    OverviewHashimoto’s Thyroiditis (HT) is an autoimmune disorder characterized by the progressive destruction of thyroid follicles due to chronic inflammation, often leading to hypothyroidism. It affects 10-12% of the global population, with a significantly higher prevalence among women​​. While its etiology involves genetic, environmental, and epigenetic factors, increasing evidence highlights the role of gut microbiota in […]

Gut microbiota analysis in Hashimoto’s thyroiditis patients identified significant dysbiosis, with increased pro-inflammatory taxa and reduced beneficial microbes, correlating with thyroid autoantibodies.

What Was Studied?

This study systematically investigated alterations in the gut microbiota composition in patients with Hashimoto’s thyroiditis (HT), an organ-specific autoimmune disease, compared to healthy controls. The researchers used 16S rRNA sequencing to profile and compare the gut microbiota of 50 HT patients and 27 matched healthy controls. The study aimed to identify microbial biomarkers associated with HT and their correlations with clinical parameters, such as thyroid peroxidase antibody (TPO-Ab) and thyroglobulin antibody (TG-Ab) levels.

Who Was Studied?

The study involved two cohorts: an exploration cohort of 28 HT patients and 16 healthy controls, and a validation cohort of 22 HT patients and 11 healthy controls. All participants were of Han Chinese ethnicity, aged between 18 and 65 years, and matched for age, sex, and BMI. Patients included were euthyroid and free from confounding conditions or recent medications that could affect the gut microbiota.

Key Findings

The study revealed significant differences in the gut microbiota composition between HT patients and healthy controls, though overall bacterial diversity and richness were similar. HT patients exhibited a marked increase in Firmicutes and a reduction in Bacteroidetes, with a significantly higher Firmicutes-to-Bacteroidetes (F/B) ratio. At the genus level, the abundances of Blautia, Roseburia, Ruminococcus_torques_group, and Eubacterium_hallii_group were significantly increased in HT patients. In contrast, beneficial genera like Bacteroides, Fecalibacterium, and Prevotella_9 were significantly decreased.

The researchers identified 27 genera with significant differences between HT patients and controls using linear discriminant analysis effect size (LEfSe). Ten genera, including Bacteroides and Fecalibacterium, were highlighted as potential biomarkers, achieving high diagnostic accuracy with AUC values of 0.91 and 0.88 in the exploration and validation cohorts, respectively.

Microbiota changes were correlated with clinical parameters. For instance, increased levels of Blautia and Dorea were positively associated with TPO-Ab and TG-Ab, while reduced levels of Fecalibacterium and Bacteroides correlated inversely with these antibodies.

Greatest Implications

The findings highlight the potential role of gut dysbiosis in the pathogenesis of HT. The observed microbial shifts suggest a loss of anti-inflammatory and barrier-supporting taxa, such as Fecalibacterium, and an increase in pro-inflammatory or mucin-degrading taxa, such as Ruminococcus_torques_group. This dysbiosis may contribute to immune activation and thyroid autoimmunity through mechanisms like increased intestinal permeability and molecular mimicry. Additionally, the identified microbial biomarkers could serve as non-invasive tools for HT diagnosis and disease monitoring. However, longitudinal studies and experimental validation are needed to confirm causality and explore therapeutic interventions targeting the gut microbiota.

Comparative Analysis of Taxonomic and Functional Gut Microbiota Profiles in Relation to Seroconversion of Thyroid Peroxidase Antibodies in Euthyroid Participants.

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Hashimoto’s Thyroiditis
    Hashimoto’s Thyroiditis

    OverviewHashimoto’s Thyroiditis (HT) is an autoimmune disorder characterized by the progressive destruction of thyroid follicles due to chronic inflammation, often leading to hypothyroidism. It affects 10-12% of the global population, with a significantly higher prevalence among women​​. While its etiology involves genetic, environmental, and epigenetic factors, increasing evidence highlights the role of gut microbiota in […]

This study explored gut microbiota profiles in TPOAb-positive and TPOAb-negative euthyroid individuals. While no significant diversity differences were found, specific taxa like Desulfovibrionaceae were associated with TPOAb presence. Further research is needed to determine their role in autoimmune thyroid disease progression.

What was studied?

This study investigated the taxonomic and functional gut microbiota profiles of euthyroid individuals with and without thyroid peroxidase antibodies (TPOAb), a marker for autoimmune thyroid diseases such as Hashimoto’s thyroiditis. The goal was to assess whether gut microbiota composition differs in individuals with TPOAb before the clinical onset of autoimmune thyroid disease and to evaluate ethnic variations in thyroid biomarkers.

Who was studied?

The study examined 1,468 euthyroid participants aged 35 years and older from the multiethnic HELIUS cohort, including European Dutch, Moroccan, and Turkish individuals. Of these, 159 participants were TPOAb-positive, and 1,309 were TPOAb-negative. Fecal microbiota composition was analyzed using 16S rRNA sequencing.

What were the most important findings?

The study revealed no significant differences in global gut microbiota diversity (alpha or beta diversity) between TPOAb-positive and TPOAb-negative individuals. However, 138 microbial taxa were nominally associated with TPOAb presence, with 13 taxa consistently significant across multiple statistical methods. Among the most notable taxa, members of the Desulfovibrionaceae family were positively associated with TPOAb presence, while certain taxa from the Clostridiales vadin BB60 group were negatively associated. Functional pathway analysis indicated reduced abundance of pathways related to D-glucarate degradation, glycolysis, and adenosylcobalamin biosynthesis in TPOAb-positive participants, although none of these associations were statistically significant after correction for multiple testing. Ethnicity emerged as a more significant factor in microbiota variation than TPOAb status, with no ethnic differences in thyroid biomarker levels found.

What are the greatest implications of this study?

This study underscores the role of gut microbiota in the early stages of autoimmune thyroid disease, suggesting that microbial alterations may not be the primary driver of TPOAb seroconversion. However, the associations between specific taxa and TPOAb presence warrant further investigation to elucidate their potential involvement in disease progression. The lack of robust differences in microbiota composition between groups highlights the need for longitudinal studies to determine causal relationships between gut dysbiosis and autoimmune thyroiditis. Moreover, the findings emphasize the importance of considering ethnic diversity in microbiome research to ensure accurate interpretation of results.

Integrative analysis of gut microbiome and host transcriptome reveal novel molecular signatures in Hashimoto's thyroiditis

May 20, 2025
  • Hashimoto’s Thyroiditis
    Hashimoto’s Thyroiditis

    OverviewHashimoto’s Thyroiditis (HT) is an autoimmune disorder characterized by the progressive destruction of thyroid follicles due to chronic inflammation, often leading to hypothyroidism. It affects 10-12% of the global population, with a significantly higher prevalence among women​​. While its etiology involves genetic, environmental, and epigenetic factors, increasing evidence highlights the role of gut microbiota in […]

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

This study revealed novel molecular signatures linking gut microbiota and transcriptome in Hashimoto's thyroiditis, advancing diagnostic and therapeutic approaches.

What Was Studied?

Integrative analysis reveals novel gut microbiota-transcriptome signatures for Hashimoto's thyroiditis, aiding early diagnosis and treatment.This study explored the molecular signatures of Hashimoto’s thyroiditis (HT) through an integrative analysis of gut microbiome and host transcriptome (miRNA/mRNA). It aimed to identify novel molecular markers and elucidate the gut-thyroid axis, using data from 31 early HT patients and 30 healthy controls across discovery and validation cohorts. The study sought to uncover interactions between the gut microbiota and host gene expression, providing insights into HT pathogenesis.

Who Was Studied?

Participants included 31 early HT patients and 30 healthy individuals aged 18–65. HT cases were defined by elevated thyroid antibodies (TPOAb/TGAb) and morphological abnormalities while maintaining normal thyroid function. Exclusions included antibiotic or probiotic use, significant dietary changes, or comorbid conditions. Blood and fecal samples were collected for transcriptomic and metagenomic sequencing.

What Were the Most Important Findings?

The study identified subtle but significant gut microbiota alterations in early HT patients. While alpha diversity was unchanged, beta diversity analysis revealed compositional shifts, including increased Bacillota_A and Spirochaetota at the phylum level and significant differences in 24 genera and 67 species. Beneficial microbes like Barnesiella intestinihominis were reduced, while opportunistic pathogens like Peptostreptococcus were enriched. Host transcriptome analysis identified 1975 downregulated and 1821 upregulated mRNAs, alongside 27 miRNAs. Immune and inflammation-related pathways were enriched, with hsa-miR-548aq-3p and hsa-miR-374a-5p playing key roles. Key molecular signatures included three bacterial species (Salaquimonas_sp002400845, Clostridium_AI_sp002297865, Enterocloster_citroniae) and six RNAs (e.g., GADD45A, IRS2, SMAD6). These integrated signatures demonstrated strong diagnostic potential (AUC=0.95) in distinguishing HT patients from healthy controls.

What Are the Greatest Implications?

This research advances understanding of the gut-thyroid axis and provides a robust framework for early HT diagnosis and treatment. Molecular signatures identified offer potential for targeted therapies, including microbiome modulation. For example, restoring beneficial microbes such as Barnesiella intestinihominis or targeting specific pathogenic species may offer therapeutic benefits. Integration of gut microbiota and transcriptome data sets a precedent for multidimensional biomarker development in autoimmune conditions.

Molecular estimation of alteration in intestinal microbial composition in Hashimoto's thyroiditis patients

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Hashimoto’s Thyroiditis
    Hashimoto’s Thyroiditis

    OverviewHashimoto’s Thyroiditis (HT) is an autoimmune disorder characterized by the progressive destruction of thyroid follicles due to chronic inflammation, often leading to hypothyroidism. It affects 10-12% of the global population, with a significantly higher prevalence among women​​. While its etiology involves genetic, environmental, and epigenetic factors, increasing evidence highlights the role of gut microbiota in […]

This study revealed novel molecular signatures linking gut microbiota and transcriptome in Hashimoto's thyroiditis, advancing diagnostic and therapeutic approaches.

What Was Studied?

This study examined alterations in the gut microbiota composition of patients with Hashimoto's thyroiditis (HT). It aimed to investigate the relationship between intestinal dysbiosis and HT through quantitative and qualitative analysis of gut microbial diversity and composition using techniques such as PCR-DGGE, real-time PCR, and pyrosequencing of 16S rRNA genes.

Who Was Studied?

The study analyzed fecal samples from 29 HT patients and 12 healthy individuals aged 40–60 years. Patients were diagnosed based on elevated thyroid antibodies (TPOAb and TGAb) and other clinical markers, including TSH and T4 levels. Healthy controls had normal thyroid function and no history of antibiotic or probiotic use in the 60 days preceding the study.

What Were the Most Important Findings?

The study revealed significant gut microbiota dysbiosis in Hashimoto’s thyroiditis (HT) patients compared to healthy controls. HT patients exhibited an increased abundance of inflammatory phyla like Proteobacteria and decreased beneficial phyla such as Firmicutes and Bacteroidetes. At the genus level, Escherichia-Shigella and Parasutterella were elevated, while anti-inflammatory genera such as Prevotella_9 and Dialister were significantly reduced. Escherichia coli was particularly overrepresented, potentially contributing to intestinal barrier disruption and inflammation linked to thyroid autoimmunity.

Real-time PCR showed significant reductions in Bifidobacterium and Lactobacillus, essential for producing immune-regulating SCFAs, while alpha diversity indicated bacterial overgrowth in HT patients. Functional diversity measures showed no significant changes, pointing to microbial imbalance rather than increased functional diversity. Pyrosequencing confirmed these findings, demonstrating a distinct microbial profile in HT patients. These results highlight the role of gut dysbiosis in HT pathogenesis and suggest potential therapeutic strategies targeting microbiome restoration.

What Are the Greatest Implications?

This study highlights gut microbiota dysbiosis as a potential contributor to the pathogenesis of HT. The findings suggest that the overrepresentation of inflammatory and opportunistic pathogens, such as Escherichia coli and Escherichia-Shigella, coupled with the reduction of beneficial microbes like Bifidobacterium and Lactobacillus, may influence immune regulation and thyroid autoimmunity. Restoring microbial balance through probiotics, dietary interventions, or targeted microbiome therapies could serve as novel strategies for managing HT. These results underscore the critical role of gut health in autoimmune diseases and provide a foundation for developing microbiome-targeted interventions.

Cardiovascular Health

Go to Category Page

Altered Gut Microbiota in Chronic Heart Failure: A Pathway to New Therapies

May 20, 2025
  • Cardiovascular Health
    Cardiovascular Health

    Recent research has revealed that specific gut microbiota-derived metabolites are strongly linked to cardiovascular disease risk—potentially influencing atherosclerosis development more than traditional risk factors like cholesterol levels. This highlights the gut microbiome as a novel therapeutic target for cardiovascular interventions.

  • Heart Failure
    Heart Failure

    Recent research reveals that the gut microbiome significantly influences heart failure progression, contributing to inflammation and other complications.

This review emphasizes the significant alterations in gut microbiota in severe chronic heart failure (CHF) patients and suggests that gut microbiota modulation could be a promising avenue for therapeutic intervention. The study provides a foundation for future research aimed at leveraging gut microbiota to improve CHF management and patient health.

What Was Studied?

This original research focused on alterations in the gut microbiota composition of patients with severe chronic heart failure (CHF) using bacterial 16S rRNA gene sequencing. The study aimed to uncover microbial dysbiosis patterns and their potential functional implications in CHF.

Who Was Studied?

The study examined 29 CHF patients classified under New York Heart Association (NYHA) Class III-IV and compared them to 30 healthy controls. These individuals were recruited from Harbin Medical University hospitals in China. Inclusion criteria ensured the absence of confounding variables like recent antibiotic use or gastrointestinal surgery.

What Were the Most Important Findings?

The study found significant differences in microbial composition and diversity between CHF patients and healthy controls:

Phylum-Level Changes: CHF patients showed a significant decrease in Firmicutes (59.5% vs. 72.4%) and a marked increase in Proteobacteria (21.3% vs. 6.9%), suggesting dysbiosis.

Genus-Level Alterations: Notable reductions in SCFA-producing genera like Ruminococcaceae (UCG-004 and UCG-002), Lachnospiraceae FCS020 group, and Dialister were observed. Conversely, pathogenic genera such as Enterococcus and Klebsiella were elevated.

Diversity Metrics: Alpha diversity (Chao1, PD-whole-tree, Shannon indices) and beta diversity (weighted UniFrac distances) were significantly lower in CHF patients, reflecting reduced microbial richness and altered community structure.

Functional Implications: Predicted microbial functions (using PICRUSt) linked to CHF involved disruptions in pathways like cell cycle control, carbohydrate metabolism, and amino acid metabolism. Dysbiosis is also correlated with reduced SCFA production, potentially exacerbating inflammation and metabolic dysregulation.

What Are the Greatest Implications of This Study?

This research highlights a potential gut-heart axis, where microbial dysbiosis in CHF may contribute to systemic inflammation and metabolic disturbances via SCFA deficiencies and increased endotoxins. The findings suggest that targeting gut microbiota through therapeutic interventions could represent a novel strategy for managing severe CHF. Moreover, the identified microbial signatures could guide biomarker development for CHF diagnosis and progression monitoring.

Evidence of a causal and modifiable relationship between kidney function and circulating trimethylamine N-oxide

May 20, 2025
  • Cardiovascular Health
    Cardiovascular Health

    Recent research has revealed that specific gut microbiota-derived metabolites are strongly linked to cardiovascular disease risk—potentially influencing atherosclerosis development more than traditional risk factors like cholesterol levels. This highlights the gut microbiome as a novel therapeutic target for cardiovascular interventions.

This study identifies kidney function as the primary modifiable factor influencing circulating TMAO levels, which are linked to cardiovascular risk. The bidirectional relationship between TMAO and kidney health suggests that reno-protective therapies, particularly GLP-1 receptor agonists, could lower TMAO levels and mitigate associated risks, providing new insights for cardiovascular and renal disease management.

What was studied?

The study investigated the relationship between kidney function and circulating levels of trimethylamine N-oxide (TMAO), a gut microbiota-derived metabolite that has been implicated in increased cardiovascular risk. The researchers aimed to identify the primary factors influencing serum TMAO levels, with a particular focus on the modifiable role of kidney function. They employed a combination of machine learning, epidemiological analysis, and preclinical experiments to explore this relationship and assess whether kidney function not only regulates TMAO levels but is also affected by them, particularly in the context of kidney fibrosis.

Who was studied?

The study involved a cohort of 1,741 adult Europeans from the MetaCardis study, which included participants across a spectrum of cardiometabolic disease severity, ranging from metabolically healthy individuals to those with metabolic syndrome, type-2 diabetes (T2D), and ischemic heart disease (IHD). The cohort was representative of a European population, with individuals recruited from Denmark, France, and Germany. The study also included specific sub-cohorts such as the MetaCardis Body Mass Index Spectrum subset (BMIS), which focused on overweight or obese individuals presenting with features of metabolic syndrome but without overt T2D or ischemic heart disease.

What were the most important findings?

Kidney Function as the Primary Modifiable Factor Influencing TMAO Levels: The study identified kidney function, measured by estimated glomerular filtration rate (eGFR), as the most significant modifiable factor regulating fasting serum TMAO levels. Lower eGFR, indicative of reduced kidney function, was strongly associated with higher circulating TMAO levels.

Modest Impact of Diet and Gut Microbiota: While diet and gut microbiota composition were found to contribute to circulating TMAO levels, their impact was relatively modest compared to kidney function. The habitual intake of TMAO precursors like red meat and eggs did not significantly correlate with TMAO levels in the study's non-interventional settings.

Bidirectional Relationship Between TMAO and Kidney Function: The study suggested a bidirectional relationship where impaired kidney function leads to higher TMAO levels, and elevated TMAO levels, in turn, contribute to kidney damage, particularly fibrosis. This was corroborated by preclinical models showing that TMAO exposure increases kidney scarring.

Therapeutic Implications of Reno-Protective Drugs: Patients with T2D who were receiving glucose-lowering drugs with reno-protective properties, specifically GLP-1 receptor agonists (GLP-1RAs), had significantly lower circulating TMAO levels compared to matched controls. This finding suggests that reno-protective medications could potentially be used to lower TMAO levels and mitigate associated cardiovascular risks.

What are the greatest implications of this study?

Clinical Management of Cardiovascular Risk: The study highlights the critical role of kidney function in managing cardiovascular risk associated with elevated TMAO levels. It suggests that preserving or improving kidney function could be a key strategy in reducing circulating TMAO levels and, by extension, cardiovascular risk.

Potential for Therapeutic Interventions: The findings imply that reno-protective therapies, particularly those involving GLP-1 receptor agonists, could have a dual benefit in patients with T2D or other cardiometabolic conditions: improving kidney function and lowering TMAO levels. This could lead to novel therapeutic strategies aimed at reducing TMAO-related cardiovascular risk.

Reevaluation of Dietary and Microbiota Interventions: While diet and gut microbiota composition have been previously considered major contributors to TMAO levels, this study suggests that in the context of non-interventional settings, their impact may be secondary to that of kidney function. This could shift the focus of future research and clinical practice towards targeting kidney health as a more effective means of controlling TMAO levels.

Mechanistic Insights into TMAO and Kidney Health: The Evidence of a causal and modifiable relationship between kidney function and circulating trimethylamine N-oxide study provides mechanistic insights into how TMAO contributes to kidney damage, particularly through the promotion of renal fibrosis. This understanding could inform future research into the development of targeted therapies that specifically address the pro-fibrotic effects of TMAO in kidney disease.

Gut microbiota in heart failure and related interventions

May 20, 2025
  • Cardiovascular Health
    Cardiovascular Health

    Recent research has revealed that specific gut microbiota-derived metabolites are strongly linked to cardiovascular disease risk—potentially influencing atherosclerosis development more than traditional risk factors like cholesterol levels. This highlights the gut microbiome as a novel therapeutic target for cardiovascular interventions.

This review explores the gut-heart axis, highlighting how gut microbiota alterations and metabolites like TMAO and SCFAs contribute to heart failure (HF). It evaluates the gut hypothesis, emphasizing bacterial translocation and inflammation in HF, and discusses potential interventions.

What was reviewed?

The Gut Microbiota in Heart Failure and Related Interventions Review article examines the relationship between heart failure (HF) and the gut microbiota, exploring the gut hypothesis of HF, the role of gut microbiota metabolites, and potential microbiome-targeted interventions (MBTIs). The review provides a comprehensive overview of the current understanding of how changes in gut microbiota composition and its metabolites contribute to HF progression and discusses various interventions, including dietary changes, probiotic therapy, fecal microbiota transplantation (FMT), antibiotics, and other novel approaches.

Who was reviewed?

The review synthesizes findings from various studies involving HF patients and animal models to understand the connection between gut microbiota and HF. It also evaluates research on different interventions and their effects on gut microbiota and HF. Specific studies cited include investigations of bacterial species present in HF patients compared to healthy controls, the impact of gut microbiota metabolites like trimethylamine N-oxide (TMAO) and short-chain fatty acids (SCFAs) on HF, and the efficacy of interventions like the DASH diet, Mediterranean diet, probiotics, FMT, and antibiotics.

What were the most important findings of this review?

The review highlights the gut hypothesis of heart failure (HF), where reduced cardiac output and systemic congestion lead to diminished intestinal perfusion, ischemia, and barrier dysfunction. This allows bacterial translocation and endotoxin release, worsening inflammation and HF. HF patients show increased pathogenic bacteria (e.g., Bacteroides, Eubacterium rectale) and decreased beneficial bacteria (e.g., Lachnospiraceae, Ruminococcaceae). Key gut microbiota metabolites, such as TMAO, SCFAs, TMAVA, and PAGln, significantly impact HF. TMAO promotes cardiac fibrosis, hypertrophy, and inflammation, while SCFAs have protective effects, preventing cardiac hypertrophy and fibrosis, reducing inflammation, and providing energy to the failing heart.

What are the greatest implications of this review?

The greatest implications of the Gut Microbiota in Heart Failure and Related Interventions review are manifold. It highlights the therapeutic potential of targeting gut microbiota as a promising avenue for heart failure (HF) treatment, suggesting that a deeper understanding of the interactions between gut microbiota and HF could lead to novel strategies that complement existing therapies. Personalized medicine approaches, including dietary changes, probiotics, and potentially fecal microbiota transplantation (FMT), could be tailored to individual patients to address specific microbial imbalances contributing to HF. The review also underscores the importance of preventive strategies, such as adopting diets that support a healthy gut microbiota, in reducing the risk and progression of HF, which could have significant public health implications. Additionally, the review calls for further research to elucidate the mechanisms linking gut microbiota and HF, assess the long-term efficacy and safety of various interventions, and explore the roles of other metabolites and bacterial species in HF. Such research could pave the way for new diagnostic and therapeutic tools in HF management. Overall, the review emphasizes the critical role of gut microbiota in HF and suggests that targeting it could revolutionize HF treatment and prevention.

The Association between Zinc and Copper Circulating Levels and Cardiometabolic Risk Factors in Adults: A Study of Qatar Biobank Data

May 20, 2025
  • Cardiovascular Health
    Cardiovascular Health

    Recent research has revealed that specific gut microbiota-derived metabolites are strongly linked to cardiovascular disease risk—potentially influencing atherosclerosis development more than traditional risk factors like cholesterol levels. This highlights the gut microbiome as a novel therapeutic target for cardiovascular interventions.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study evaluated the associations of zinc, copper, and Zn/Cu ratio with cardiometabolic risk factors in Qatari adults, revealing copper’s protective role and Zn/Cu ratio’s adverse implications for metabolic health.

What was studied?

This study analyzed the relationship between zinc (Zn), copper (Cu), and the zinc-to-copper (Zn/Cu) ratio with cardiometabolic risk (CMR) factors and metabolic syndrome (MetS) using data from the Qatar Biobank. It sought to determine whether circulating levels of these trace minerals and their ratios were associated with various markers of cardiometabolic health, including lipid profiles, blood pressure, glucose levels, and body composition.

Who was studied?

The study included 437 Qatari adults aged 18 and older, representing both sexes. Participants had detailed cardiometabolic and mineral status profiles measured. Individuals with non-communicable diseases, those taking mineral supplements, and pregnant or lactating women were excluded to ensure a clear analysis of trace mineral associations with CMR factors.

Key Findings

This study revealed several associations between trace mineral levels and cardiometabolic markers. High Cu levels were associated with a reduced risk of MetS, lower diastolic blood pressure (DBP), and decreased prevalence of low HDL cholesterol, suggesting a protective role of copper in cardiometabolic health. Conversely, a higher Zn/Cu ratio was linked to an increased risk of MetS and low HDL, indicating that imbalances in these trace elements could worsen metabolic health.

While Zn alone was not strongly correlated with MetS or most CMR factors, it showed weak positive correlations with waist circumference (WC) and triglycerides (TG), which are notable for metabolic processes. Cu, on the other hand, positively correlated with HDL and total cholesterol (TC) while negatively correlating with DBP. These findings emphasize the differential and sometimes opposing roles of these minerals in cardiometabolic regulation.

In terms of microbiome relevance, trace elements like Zn and Cu influence microbial composition and metabolic functions. For example, Zn deficiency can affect glucose metabolism and inflammation, while Cu is a cofactor for antioxidative enzymes like superoxide dismutase, influencing oxidative stress pathways. Dysregulation of these pathways is often linked to microbial dysbiosis, potentially contributing to MetS and other cardiometabolic conditions.

Greatest Implications

The results underscore the need to consider trace element levels, particularly Cu and the Zn/Cu ratio, in cardiometabolic health assessments. The findings suggest that higher Cu levels confer protective effects against MetS and DBP, whereas an elevated Zn/Cu ratio increases the risk of adverse outcomes, including low HDL and MetS. These insights could inform clinical interventions, such as dietary adjustments or supplementation, to balance trace mineral levels and support cardiometabolic health. Additionally, these results highlight the potential role of trace mineral modulation as part of microbiome-targeted therapies, given their influence on systemic inflammation and metabolism.

TMAO: how gut microbiota contributes to heart failure

May 20, 2025
  • Cardiovascular Health
    Cardiovascular Health

    Recent research has revealed that specific gut microbiota-derived metabolites are strongly linked to cardiovascular disease risk—potentially influencing atherosclerosis development more than traditional risk factors like cholesterol levels. This highlights the gut microbiome as a novel therapeutic target for cardiovascular interventions.

This review underscores the critical role of gut microbiota and TMAO in heart failure pathophysiology and opens up new avenues for therapeutic interventions targeting the gut–TMAO–HF axis. The findings suggest promising directions for future research and clinical applications aimed at improving HF patient care and outcomes.

What Was Reviewed?

This review focused on the involvement of gut microbiota in the pathogenesis and progression of cardiovascular diseases, particularly heart failure (HF). It emphasized the role of gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) in heart failure and explored the potential of the gut–TMAO–HF axis as a therapeutic target for HF treatment.

Who Was Reviewed?

The review considered patients with various forms of heart failure, including acute heart failure (AHF), chronic heart failure (CHF), heart failure with preserved ejection fraction (HFpEF), and heart failure with reduced ejection fraction (HFrEF). It also encompassed studies involving animal models and in vitro experiments that investigated the pathophysiological mechanisms of TMAO in HF.

What Were the Most Important Findings of This Review?

This review highlights the critical role of gut microbiota in heart failure (HF). Gut dysbiosis contributes to HF pathogenesis through mechanisms like splanchnic hypoperfusion and intestinal barrier dysfunction. Trimethylamine N-oxide (TMAO), a gut-derived metabolite, significantly impacts cardiovascular pathology by promoting myocardial hypertrophy and fibrosis, inducing inflammatory responses, and causing endothelial dysfunction. Elevated TMAO levels correlate with poorer prognosis and higher mortality in HF patients, serving as an independent predictor for HF outcomes. Potential therapeutic targets include dietary interventions, probiotics, prebiotics, and inhibitors of TMA synthesis, such as 3,3-dimethyl-1-butanol (DMB). Fecal microbial transplantation (FMT) and certain antibiotics also show promise in modulating gut microbiota and reducing TMAO production. These findings support a multifaceted approach to HF management by targeting gut microbiota and its metabolites.

What Are the Greatest Implications of This Review?

The "TMAO: how gut microbiota contributes to heart failure" review highlights the importance of novel therapeutic strategies, the prognostic value of TMAO, and future research directions.

Novel Therapeutic Strategies: The review suggests that targeting the gut–TMAO–HF axis could be a revolutionary approach in treating HF. By modulating gut microbiota composition and reducing TMAO levels, it may be possible to improve HF prognosis and patient outcomes. Personalized dietary interventions and the use of probiotics, prebiotics, and phytochemicals hold significant potential for HF management.

Prognostic Value of TMAO: TMAO can serve as a valuable prognostic marker for HF, aiding clinicians in identifying high-risk patients and tailoring more effective treatment strategies. Further research is needed to validate TMAO's role across diverse populations and to explore its utility in clinical practice.

Future Research Directions: Prospective studies are needed to establish a causal relationship between gut microbiota changes and HF. Investigating the detailed mechanisms of how TMAO influences HF progression will be crucial for developing targeted therapies.

Heart Failure

Go to Category Page

Altered Gut Microbiota in Chronic Heart Failure: A Pathway to New Therapies

May 20, 2025
  • Cardiovascular Health
    Cardiovascular Health

    Recent research has revealed that specific gut microbiota-derived metabolites are strongly linked to cardiovascular disease risk—potentially influencing atherosclerosis development more than traditional risk factors like cholesterol levels. This highlights the gut microbiome as a novel therapeutic target for cardiovascular interventions.

  • Heart Failure
    Heart Failure

    Recent research reveals that the gut microbiome significantly influences heart failure progression, contributing to inflammation and other complications.

This review emphasizes the significant alterations in gut microbiota in severe chronic heart failure (CHF) patients and suggests that gut microbiota modulation could be a promising avenue for therapeutic intervention. The study provides a foundation for future research aimed at leveraging gut microbiota to improve CHF management and patient health.

What Was Studied?

This original research focused on alterations in the gut microbiota composition of patients with severe chronic heart failure (CHF) using bacterial 16S rRNA gene sequencing. The study aimed to uncover microbial dysbiosis patterns and their potential functional implications in CHF.

Who Was Studied?

The study examined 29 CHF patients classified under New York Heart Association (NYHA) Class III-IV and compared them to 30 healthy controls. These individuals were recruited from Harbin Medical University hospitals in China. Inclusion criteria ensured the absence of confounding variables like recent antibiotic use or gastrointestinal surgery.

What Were the Most Important Findings?

The study found significant differences in microbial composition and diversity between CHF patients and healthy controls:

Phylum-Level Changes: CHF patients showed a significant decrease in Firmicutes (59.5% vs. 72.4%) and a marked increase in Proteobacteria (21.3% vs. 6.9%), suggesting dysbiosis.

Genus-Level Alterations: Notable reductions in SCFA-producing genera like Ruminococcaceae (UCG-004 and UCG-002), Lachnospiraceae FCS020 group, and Dialister were observed. Conversely, pathogenic genera such as Enterococcus and Klebsiella were elevated.

Diversity Metrics: Alpha diversity (Chao1, PD-whole-tree, Shannon indices) and beta diversity (weighted UniFrac distances) were significantly lower in CHF patients, reflecting reduced microbial richness and altered community structure.

Functional Implications: Predicted microbial functions (using PICRUSt) linked to CHF involved disruptions in pathways like cell cycle control, carbohydrate metabolism, and amino acid metabolism. Dysbiosis is also correlated with reduced SCFA production, potentially exacerbating inflammation and metabolic dysregulation.

What Are the Greatest Implications of This Study?

This research highlights a potential gut-heart axis, where microbial dysbiosis in CHF may contribute to systemic inflammation and metabolic disturbances via SCFA deficiencies and increased endotoxins. The findings suggest that targeting gut microbiota through therapeutic interventions could represent a novel strategy for managing severe CHF. Moreover, the identified microbial signatures could guide biomarker development for CHF diagnosis and progression monitoring.

Rheumatoid Arthritis

Go to Category Page

Analyzing the impact of heavy metal exposure on osteoarthritis and rheumatoid arthritis: an approach based on interpretable machine learning

May 20, 2025
  • Rheumatoid Arthritis
    Rheumatoid Arthritis

    OverviewRheumatoid arthritis (RA) is a systemic autoimmune disease marked by chronic joint inflammation, synovitis, and bone erosion, driven by Treg/Th17 imbalance, excessive IL-17, TNF-α, and IL-1 production, and macrophage activation. Emerging evidence links microbial dysbiosis and heavy metal exposure to RA, [1][2] with gut microbiota influencing autoimmune activation via Toll-like receptor (TLR) signaling, inflammasome activation, […]

Machine learning analysis of NHANES data (2003–2020) identifies tungsten, cadmium, arsenic, and antimony as major risk factors for arthritis. SHAP-based models suggest these metals contribute to inflammation and cartilage degradation, reinforcing environmental exposure as a key component of arthritis development.

What Was Studied?

This study examined the impact of heavy metal exposure on the prevalence and differentiation of osteoarthritis (OA) and rheumatoid arthritis (RA) using interpretable machine learning models. Researchers analyzed data from the National Health and Nutrition Examination Survey (NHANES) (2003–2020) to assess how various heavy metals contribute to arthritis risk.

Who Was Studied?

The study population consisted of 14,319 participants from NHANES who met specific inclusion criteria, including age ≥ 20 years and confirmed arthritis status via blood and urine heavy metal testing.

Key Findings

Using machine learning techniques such as LASSO regression and SHapley Additive exPlanations (SHAP), the study identified tungsten, cobalt, cadmium, antimony, arsenic, and blood cadmium as significant risk factors for arthritis, while molybdenum, thallium, lead, and mercury appeared to have a protective or neutral association. Cadmium exposure showed a strong correlation with rheumatoid arthritis (RA), likely due to its role in oxidative stress and inflammation, while arsenic exposure was linked to both osteoarthritis (OA) and RA, with previous studies indicating its contribution to cartilage degradation. Tungsten and antimony emerged as newly recognized risk factors, though their mechanisms remain unclear. In contrast, molybdenum exhibited a potential protective effect, possibly by counteracting inflammation. The study’s machine learning models demonstrated high predictive accuracy, with XGBoost achieving 81% accuracy in identifying arthritis and LightGBM distinguishing between OA and RA with 76% accuracy.

Greatest Implications

This study reinforces the environmental component of arthritis development, suggesting that heavy metal exposure contributes to arthritis risk and progression. Machine learning models, particularly SHAP-based interpretations, provide valuable predictive tools for early detection. The findings highlight tungsten, cobalt, cadmium, and arsenic as potential modifiable risk factors, paving the way for targeted interventions to reduce arthritis prevalence.

Association of Bitter Taste Receptor T2R38 Polymorphisms, Oral Microbiota, and Rheumatoid Arthritis

May 20, 2025
  • Rheumatoid Arthritis
    Rheumatoid Arthritis

    OverviewRheumatoid arthritis (RA) is a systemic autoimmune disease marked by chronic joint inflammation, synovitis, and bone erosion, driven by Treg/Th17 imbalance, excessive IL-17, TNF-α, and IL-1 production, and macrophage activation. Emerging evidence links microbial dysbiosis and heavy metal exposure to RA, [1][2] with gut microbiota influencing autoimmune activation via Toll-like receptor (TLR) signaling, inflammasome activation, […]

This study links T2R38 genotype to oral microbiota composition in rheumatoid arthritis, revealing microbial shifts modulated by taste receptor polymorphisms. Findings suggest buccal dysbiosis in RA is partially shaped by host genetics, offering new insight into mucosal immunity and potential biomarkers for disease risk and microbial-targeted therapy.

What was studied?

This study examined the relationship between T2R38 bitter taste receptor gene polymorphisms, the composition of the oral microbiota, and rheumatoid arthritis (RA) status. Specifically, it assessed whether the T2R38 genotypes (PAV/PAV, PAV/AVI, AVI/AVI) influenced the buccal microbial community and whether this interaction was associated with RA pathogenesis or oral microbial dysbiosis.

Who was studied?

The study involved 99 adults, including 35 individuals with clinically diagnosed RA and 64 non-RA controls. Most of the non-RA participants were first-degree relatives of the RA patients, considered at-risk individuals. Buccal swabs were collected from all participants for both genotyping of the TAS2R38 gene and 16S rRNA sequencing of the oral microbiome. Clinical features, inflammatory markers, and periodontal health symptoms were also recorded.

What were the most important findings?

The study revealed significant shifts in the buccal microbiome between RA and non-RA individuals, independent of diversity indices. RA patients exhibited higher relative abundances of Streptococcus salivarius, Rothia mucilaginosa, and Leptotrichia, whereas non-RA individuals showed enrichment in Porphyromonas, Fusobacterium periodonticum, Aggregatibacter, and Capnocytophaga. These genera are known to modulate inflammation and mucosal immunity, aligning with recognized dysbiosis in RA.

Crucially, the TAS2R38 genotype modified the oral microbial profile. Supertasters (PAV/PAV) with RA had higher levels of Streptococcus salivarius—a known immunomodulatory commensal—compared to non-tasters (AVI/AVI). Conversely, Porphyromonas and Aggregatibacter, associated with periodontal disease and implicated in ACPA production, were depleted in RA. These findings suggest that T2R38 polymorphisms, which influence oral mucosal immunity through differential antimicrobial peptide expression, may shape oral microbial ecology in RA.

FeatureFindingImplication
RA-associated taxa↑ Streptococcus salivarius, Rothia mucilaginosa, LeptotrichiaIndicators of RA-specific dysbiosis and mucosal immune adaptation
Non-RA-enriched taxa↑ Porphyromonas, Fusobacterium, AggregatibacterSuggest immune exclusion of periodontal pathogens in RA
Genotype-microbiome interactionPAV/PAV RA patients had ↑ Streptococcus salivariusTAS2R38-mediated modulation of oral immunity
T2R38 polymorphism distributionOverrepresentation of PAV/PAV in RA vs. published population controlsSuggests genetic predisposition influencing oral-immune axis in RA
Implications for ACPA developmentDepletion of P. gingivalis and Aggregatibacter in established RAPotential early immune targeting of citrullinating pathogens prior to disease onset
Alpha diversityNo significant differences by RA status or T2R38 genotypeDysbiosis not reflected in richness but in compositional shifts
Beta diversitySignificant difference between RA and non-RA (p = 0.02)Confirms distinct microbial communities between groups
Clinical translationSupports microbiome-genotype risk models for RA; possible diagnostic and therapeutic valueMay inform MBTI development targeting host-microbiome interactions mediated by taste receptors

What are the greatest implications of this study?

This study provides novel evidence linking taste receptor polymorphisms, specifically TAS2R38, to oral microbiome composition and RA susceptibility. The differential microbial signatures observed—particularly the RA-associated depletion of periodontal pathogens in PAV/PAV individuals—support the idea that heightened T2R38-mediated immune surveillance may selectively modulate oral dysbiosis. This could have implications for early immune priming and autoantibody production via citrullination pathways involving P. gingivalis and A. actinomycetemcomitans.

For the Microbiome Signatures Database, this study contributes to defining RA-specific Major Microbial Associations (MMAs) in the oral cavity. Clinically, it highlights the potential of integrating genotypic screening (e.g., TAS2R38) with microbiome profiling as a stratification tool in RA risk assessment and precision therapeutics. It also supports future development of microbiome-targeted interventions (MBTIs) aimed at restoring oral eubiosis in genetically susceptible individuals.

Oral Microbiota Identifies Patients in Early Onset Rheumatoid Arthritis

May 20, 2025
  • Rheumatoid Arthritis
    Rheumatoid Arthritis

    OverviewRheumatoid arthritis (RA) is a systemic autoimmune disease marked by chronic joint inflammation, synovitis, and bone erosion, driven by Treg/Th17 imbalance, excessive IL-17, TNF-α, and IL-1 production, and macrophage activation. Emerging evidence links microbial dysbiosis and heavy metal exposure to RA, [1][2] with gut microbiota influencing autoimmune activation via Toll-like receptor (TLR) signaling, inflammasome activation, […]

This study found that oral microbiota in early rheumatoid arthritis displays distinct bacterial enrichments and metabolic shifts. Key taxa, including Filifactor alocis and Porphyromonas endodontalis, were elevated in RA regardless of periodontal status, highlighting their potential as biomarkers and microbial contributors to RA pathogenesis.

What was studied?

This study evaluated the composition of the oral microbiota in untreated patients with early-onset rheumatoid arthritis (eRA), comparing it to age- and sex-matched healthy controls. The investigation employed multiplex 16S rRNA gene sequencing on saliva samples to identify microbial shifts associated with eRA, both with and without clinically defined periodontal disease. The analysis also included functional profiling via PICRUSt to determine whether predicted metabolic functions differed between the groups.

Who was studied?

The study included 61 patients diagnosed with early-onset rheumatoid arthritis (symptom duration ≤12 months) and 59 healthy controls. None of the patients had begun disease-modifying anti-rheumatic drug therapy. Participants were classified by periodontal status to assess whether microbial differences were independent of underlying periodontal disease. Smoking, corticosteroid use, and other demographic factors were also recorded to assess confounders.

What were the most important findings?

The study revealed significant compositional and functional differences in the oral microbiota between eRA patients and controls. Saliva samples from eRA patients showed higher species richness and a more dispersed microbial profile. Notably, species enrichment included Prevotella pleuritidis, Porphyromonas endodontalis, Filifactor alocis, and Treponema denticola, all of which are strongly associated with periodontitis and inflammation. Importantly, these differences persisted regardless of the presence of periodontal disease. Functional predictions indicated increased pathways related to ornithine metabolism, glucosylceramidase activity, and beta-lactamase resistance, suggesting a pro-inflammatory and immunomodulatory shift in the microbial community. From a microbiome signatures perspective, Filifactor alocis emerged as a Major Microbial Association (MMA) due to its consistent elevation and known mechanistic involvement in arginine metabolism and ornithine production, both linked to citrullination—a process implicated in RA pathogenesis.

Microbial Feature or FunctioneRA Patients (Enriched or Altered)Clinical or Mechanistic Relevance
Filifactor alocisEnrichedArginine metabolism → ornithine; inflammation and RA induction
Porphyromonas endodontalisEnrichedPeriodontal pathogen; linked to Sjögren’s and systemic inflammation
Prevotella pleuritidisEnrichedPeriodontitis-associated; pro-inflammatory
Treponema denticolaEnrichedKnown periodontal pathogen; immune-modulatory
Functional PathwaysOrnithine metabolism, beta-lactamase resistance, glucosylceramidaseAssociated with inflammation, immune activation, and metabolic stress in RA
Microbial DiversityHigher in eRASuggests unstable and dysbiotic oral ecosystem
Predicted KO functionsElevated in eRAFatty acid metabolism, sphingolipid and steroid metabolism linked to immune dysregulation

What are the greatest implications of this study?

These findings offer early evidence that oral microbiota dysbiosis precedes and potentially contributes to the pathogenesis of rheumatoid arthritis. The enrichment of species such as Filifactor alocis and Porphyromonas endodontalis, coupled with shifts in microbial metabolic pathways, suggests a mechanistic link between oral microbial communities and systemic autoimmunity via processes like protein citrullination and immune activation. This supports the idea that the oral cavity may act as a trigger site in genetically susceptible individuals. Clinically, these bacterial taxa could serve as non-invasive biomarkers for eRA risk assessment, especially valuable in periodontally healthy patients, and may inform future microbiome-targeted interventions for early disease modulation.

Role of Some Heavy Metals in Rheumatoid Arthritis

May 20, 2025
  • Rheumatoid Arthritis
    Rheumatoid Arthritis

    OverviewRheumatoid arthritis (RA) is a systemic autoimmune disease marked by chronic joint inflammation, synovitis, and bone erosion, driven by Treg/Th17 imbalance, excessive IL-17, TNF-α, and IL-1 production, and macrophage activation. Emerging evidence links microbial dysbiosis and heavy metal exposure to RA, [1][2] with gut microbiota influencing autoimmune activation via Toll-like receptor (TLR) signaling, inflammasome activation, […]

This study explored the association between rheumatoid arthritis and heavy metal exposure, identifying a significant deficiency of copper in RA patients. No significant correlations were found for zinc, cobalt, lead, or nickel. The findings suggest that copper deficiency may play a role in RA, warranting further research on trace metals in autoimmune diseases.

What was studied?

This study investigated the association between heavy metal concentrations and rheumatoid arthritis (RA). Specifically, it examined the serum levels of zinc (Zn), cobalt (Co), lead (Pb), nickel (Ni), and copper (Cu) in RA-positive and RA-negative individuals. The study also analyzed variations in metal levels based on sex and two age groups (below 35 years and 35 years and above) to determine if these factors influenced the relationship between heavy metal exposure and RA.

Who was studied?

The study included blood samples from patients suspected of having RA, collected from diagnostic laboratories in Sargodha, Pakistan. The participants were grouped based on their RA status (RA-positive and RA-negative), sex, and age category. Serum metal concentrations were analyzed using atomic absorption spectrophotometry (AAS), and statistical comparisons were made using a two-sample t-test to determine differences between groups.

Most Important Findings

The study found a significant decrease in serum copper (Cu) levels in RA-positive patients compared to RA-negative individuals (p=0.04). However, there was no significant relationship between RA and the concentrations of zinc (Zn), cobalt (Co), lead (Pb), and nickel (Ni). Additionally, no significant correlation was observed between heavy metal concentrations and sex or age group within the RA-positive cohort. While some previous studies reported lower zinc levels in RA patients, this study did not find any significant association between RA and Zn deficiency. The findings also contradicted prior research that suggested elevated Pb levels in RA patients, as no significant difference was observed in this study. The observed variations between this study and previous research may be due to genetic, environmental, or dietary differences in the study populations.

Greatest Implications

The study provides evidence that copper deficiency may be associated with RA, potentially implicating copper in the disease's pathophysiology. This aligns with existing research suggesting that copper plays a role in inflammatory and immune system regulation. However, the lack of significant findings for other metals highlights the need for further research to determine their exact impact on RA development and progression. Given that previous studies have reported conflicting results regarding heavy metals and RA, it is crucial to consider factors such as geographic variation, dietary intake, and environmental exposures in future research. Understanding the role of trace metals in RA could contribute to novel therapeutic strategies, including dietary interventions or supplementation for patients with confirmed deficiencies.

Subgingival Microbiome in Rheumatoid Arthritis Patients with Periodontitis

May 20, 2025
  • Rheumatoid Arthritis
    Rheumatoid Arthritis

    OverviewRheumatoid arthritis (RA) is a systemic autoimmune disease marked by chronic joint inflammation, synovitis, and bone erosion, driven by Treg/Th17 imbalance, excessive IL-17, TNF-α, and IL-1 production, and macrophage activation. Emerging evidence links microbial dysbiosis and heavy metal exposure to RA, [1][2] with gut microbiota influencing autoimmune activation via Toll-like receptor (TLR) signaling, inflammasome activation, […]

This study links the subgingival microbiome in rheumatoid arthritis to elevated ACPAs via enrichment of A. butyrica, P. simiae, and P. stomatis. These bacteria contribute to citrullination and may be key microbial biomarkers or MBTI targets in RA-associated periodontitis.

What was studied?

This study investigated the subgingival microbiome in rheumatoid arthritis (RA) patients with periodontitis in a Taiwanese population. It aimed to characterize the compositional and functional differences in subgingival microbial communities between RA patients and matched healthy controls using 16S rRNA gene sequencing. The primary goal was to evaluate correlations between specific microbial taxa and levels of anti-citrullinated protein antibodies (ACPAs), which are hallmark autoantibodies in RA.

Who was studied?

Subjects included RA patients with periodontitis and matched control groups. Subgingival plaque samples were collected and categorized into three clinical groups: individuals with active periodontitis (PD), those with active RA and periodontitis (AM), and periodontally healthy individuals. The final analysis focused on the AM and PD groups for microbial profiling and immunologic correlations with ACPAs.

What were the most important findings?

The study revealed significant differences in subgingival microbiota between RA patients and controls. In both AM and PD groups, Aminipila butyrica and Peptococcus simiae were enriched in RA patients and showed positive correlations with serum ACPA levels. A. butyrica, a strictly anaerobic, arginine-decomposing bacterium, encodes arginine deiminase, facilitating the production of citrulline—potentially leading to the formation of citrullinated antigens that induce ACPA production. This mechanism parallels that of P. gingivalis and supports its role as a potential Major Microbial Association (MMA) in RA.

Peptostreptococcus stomatis also demonstrated a strong positive correlation with ACPA levels and has been implicated in autoimmune processes, likely due to its ability to initiate NETosis and hypercitrullination via pore-forming toxins. Additionally, Streptococcus sanguinis was associated with increased ACPA levels in the PD group, while Pseudomonas batumici showed a negative correlation. PICRUSt2 functional prediction highlighted enrichment in arginine, ornithine, and proline interconversion pathways in RA patients, particularly those involving L-citrulline metabolism.

Microbial TaxaCorrelation with ACPAProposed Mechanism
Aminipila butyricaPositive (AM & PD groups)Arginine deiminase → citrulline → ACPA induction
Peptococcus simiaePositive (AM & PD groups)Potential PAD activation via NETosis
Peptostreptococcus stomatisStrong positive (AM)Immune-inflammatory trigger; linked to autoimmunity
Streptococcus sanguinisPositive (PD group)Possible secondary contributor to ACPA production
Pseudomonas batumiciNegative (PD group)Enriched in controls; potential protective role
Pathways (PICRUSt2)Enriched in RA (AM & PD)Arginine/proline metabolism → L-citrulline biosynthesis

What are the greatest implications of this study?

This study highlights a mechanistic link between subgingival dysbiosis and systemic autoimmunity in RA, particularly through bacteria capable of driving citrullination. The enrichment of A. butyrica and P. simiae, coupled with elevated ACPAs, supports the hypothesis that periodontal microbiota contribute to RA pathogenesis through microbial or host protein citrullination. These findings underscore the diagnostic and therapeutic potential of targeting oral microbial communities—particularly those expressing arginine deiminase or capable of inducing NETosis—in RA patients. From a microbiome signatures perspective, A. butyrica, P. simiae, and P. stomatis are high-confidence MMAs that may serve as predictive biomarkers or targets for MBTIs, especially those modulating arginine metabolism or immunogenic bacterial citrullination.

Microbes

Go to Category Page

Antimicrobial, Probiotic, and Immunomodulatory Potential of Cannabis sativa Extract and Delivery Systems

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

Overall, this study highlights the multifaceted bioactivities of cannabis-derived compounds and underscores their potential in pharmaceutical and probiotic applications.

What was studied?

The study investigated the antimicrobial, probiotic, and immunobiological effects of the Henola Cannabis sativae extract and its combinations with specific carriers: polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer, magnesium aluminometasilicate, and hydroxypropyl-β-cyclodextrin.  The study demonstrated that Henola Cannabis sativae extract, particularly when combined with carriers like hydroxypropyl-β-cyclodextrin, exhibits strong antimicrobial activity against various pathogens and boosts probiotic bacteria growth. Additionally, it affects immune cytokine levels, suggesting its potential as a therapeutic agent for both microbial infections and immune modulation in clinical settings.

 

Who was studied?

The subjects of the study were various microorganisms, including pathogenic bacteria like Clostridium difficile, Listeria monocytogenes, Enterococcus faecalis, Staphylococcus aureus, Staphylococcus pyrogenes, Escherichia coli, Klebsiella pneumoniae, Salmonella typhimurium, Pseudomonas aeruginosa, and the fungus Candida albicans. Additionally, the study examined probiotic bacterial strains such as Lactobacillus acidophilus, Lactobacillus casei, and others.

What were the most important findings?

The research revealed several key findings: Firstly, the Henola Cannabis sativae extract showed significant antimicrobial activity, effectively reducing the counts of various pathogenic bacteria and Candida albicans. Secondly, when combined with hydroxypropyl-β-cyclodextrin, the extract substantially enhanced the growth of beneficial probiotic bacteria. Additionally, the extract and its systems notably affected the levels of important immune cytokines such as IL-6, IL-8, and TNF-α, indicating potent immunomodulatory effects.

 

What are the greatest implications of this study?

The findings from the study on Henola Cannabis sativae extract have considerable implications. The extract’s demonstrated antimicrobial and probiotic-enhancing effects suggest its potential for development into therapeutic agents that could inhibit pathogenic microorganisms or support beneficial microbial communities. Additionally, its impact on immune system cytokines showcases its possible use in managing or treating inflammation-related diseases, indicating that it could beneficially influence immune responses. Moreover, the effectiveness of the extract when combined with specific carriers highlights the potential for creating targeted delivery systems that could enhance the bioavailability and efficacy of cannabinoids, terpenes, and flavonoids in clinical settings.

 

Associations Between Endometriosis and Gut Microbiota

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This case control study explores the gut microbiota's association with endometriosis in women, comparing 66 patients to 198 controls. Using 16S rRNA sequencing, it was found that patients have lower diversity in their gut bacteria and significant differences in the abundance of 12 bacterial types, suggesting that endometriosis may influence gut microbiota composition.

What was studied?

The study examined the gut microbiota in women with endometriosis compared to healthy controls. It aimed to explore differences based on disease localization, symptoms, or treatment and assess the gut microbiota’s potential role in the pathogenesis of endometriosis.

 

Who was studied?

66 women diagnosed with endometriosis at Skåne University Hospital were studied alongside 198 matched controls from the Malmö Offspring Study, assessing their gut microbiota through 16S rRNA sequencing.

 

What were the most important findings?

Significant findings include higher overall microbial diversity in controls compared to endometriosis patients, with specific differences in the abundance of 12 bacteria types between the two groups. After adjusting for false discovery rates, no significant microbiota differences were found within the endometriosis cohort.

 

What are the greatest implications of this study?

The study implies that gut microbiota may be altered in individuals with endometriosis, suggesting a possible link between gut microbiota and the pathogenesis or symptomatology of endometriosis. These findings highlight the need for further research on the gut microbiota’s role in endometriosis, potentially leading to new diagnostic and treatment strategies.

Comparison of Vaginal, Cervical and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

In this prospective cohort, women with stage 3/4 endometriosis exhibited a unique microbial profile, characterized by the absence of Atopobium and increased levels of potentially pathogenic genera like Gardnerella, Streptococcus, Escherichia, Shigella, and Ureoplasma in their vaginal and cervical microbiota.

What was studied?

The study focused on exploring the associations between the microbiota of the vaginal, cervical, and gut regions and stage 3–4 endometriosis in women. This research aimed to uncover potential differences in the microbiome composition between women diagnosed with advanced endometriosis and healthy controls, particularly at the genus level of microbial classification.

 

Who was studied?

The participants included 14 women with histologically proven stage 3–4 endometriosis and 14 healthy controls. These women were carefully selected based on strict criteria to ensure the stability of the microbiota, excluding those who had ever been pregnant or had conditions/medications that could affect the microbiome. All participants belonged to the same ethnicity, and the study managed to maintain a balance between samples collected during different phases of the menstrual cycle in both groups.

 

What were the most important findings?

The study revealed that while the overall composition of the vaginal, cervical, and gut microbiota was similar between women with and without endometriosis, there were notable genus-level differences. Specifically, Atopobium was absent in the endometriosis group’s vaginal and cervical microbiota. Increases in Gardnerella in the cervical microbiota and Escherichia/Shigella in the gut were more common among those with endometriosis. Sensitivity analyses excluding Lactobacillus showed significant increases in Sneathia, Gardnerella, Streptococcus, Escherichia/Shigella, and Ureaplasma, and a decrease in Alloprevotella in the cervical microbiota of the endometriosis group.

 

What are the greatest implications of this study?

The findings suggest a potentially significant association between the composition of the female microbiota and the presence of stage 3–4 endometriosis, particularly regarding the absence and presence of specific microbial genera. These differences might offer insights into the pathophysiology of endometriosis and indicate potential diagnostic markers or therapeutic targets. Specifically, the study raises intriguing questions about the direction of causation between altered microbiota and endometriosis. It suggests the microbiome’s potential as both a screening tool for endometriosis and a therapeutic target, depending on whether changes in the microbiome are a cause or a consequence of the disease. The study also underscores the potential utility of gut microbiome analysis as a predictive tool for surgical decisions, such as the need for bowel resection to treat deep infiltrating endometriosis.

Overall, this research lays the groundwork for further studies to clarify the causal relationships between dysbiosis and endometriosis and to explore the microbiome’s role in the disease’s pathogenesis, diagnosis, and treatment.

Copper in microbial pathogenesis: meddling with the metal

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study explores the dual role of copper in microbial pathogenesis, highlighting how it serves both as a vital nutrient and a potent antimicrobial agent. The research delves into the sophisticated mechanisms developed by pathogens, like Mycobacterium tuberculosis and Pseudomonas aeruginosa, to evade copper's toxicity, including specialized copper pumps and regulatory proteins. It also investigates copper's critical role in the host's immune defense, influencing infection outcomes. Findings suggest that copper's antimicrobial properties could be leveraged in healthcare to develop new treatment strategies, and its application in environmental settings could help control pathogen growth.

What was studied?

The study investigated the role of copper in microbial pathogenesis. Specifically, it examined how copper serves as both a necessary nutrient for microbial organisms and a microbial weapon used by hosts against pathogens. The research explored copper’s dual roles, its involvement in various microbial resistance mechanisms, and its interaction with the host’s immune responses.

 

Who was studied?

The study focused on various microbial species, including bacteria and fungi. It delved into the copper homeostasis mechanisms of pathogens like Mycobacterium tuberculosis and Pseudomonas aeruginosa, and also examined model organisms such as Saccharomyces cerevisiae to understand copper’s role in microbial pathogenesis and resistance.

 

What were the most important findings?

Significant findings from the study demonstrate that copper is utilized by hosts as an antimicrobial agent, significantly impacting pathogen growth and survival. Additionally, pathogens have evolved sophisticated mechanisms to counteract copper toxicity. These adaptations include the development of specific copper pumps and regulatory proteins that meticulously manage copper uptake and expulsion. Moreover, copper is found to play a critical role in the immune defense strategy of hosts, substantially influencing the outcomes of infections. These insights underscore the complex interplay between copper, pathogens, and host defenses.

 

What are the greatest implications of this study?

The implications of this research are broad and significant for both healthcare and environmental management. Understanding copper’s role in microbial pathogenesis could lead to the development of new antimicrobial strategies and treatments that leverage copper’s toxic effects on pathogens. Additionally, this knowledge could inform the use of copper in medical and environmental applications to control pathogen growth, thereby reducing infection rates and enhancing public health safety.

Correlation of fecal metabolomics and gut microbiota in mice with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study explores the relationship between fecal metabolomics and gut microbiota in endometriosis (EMS) mice, finding key metabolic changes and decreased microbiota diversity. Significant pathways, like bile acid biosynthesis and ALA metabolism, were identified, suggesting fecal metabolites affected by dysbacteriosis as potential EMS markers.

What was studied?

The study investigated the interaction between fecal metabolomics and gut microbiota in mice with endometriosis (EMS), aiming to identify metabolic changes and microbiota diversity associated with the disease.

 

Who was studied?

Female C57BL/6J mice, utilized to construct an EMS model, were the subjects of this research, allowing for the examination of fecal metabolites and gut microbiota composition.

 

What were the most important findings?

Significant findings included the identification of 156 differential metabolites, decreasing the diversity and abundance of gut microbiota in EMS mice, and involving key metabolic pathways such as bile acid biosynthesis and alpha-linolenic acid (ALA) metabolism. Notably, increased levels of chenodeoxycholic and ursodeoxycholic acids and decreased levels of ALA and 12,13-EOTrE were found in EMS mice feces.

 

What are the greatest implications of this study?

The study suggests that the identified abnormal fecal metabolites, influenced by gut dysbiosis, may be potential markers for diagnosing EMS. This finding opens new avenues for understanding EMS pathogenesis and developing non-invasive diagnostic tools based on fecal metabolite profiles.

Effect of endometriosis on the fecal bacteriota composition of mice during the acute phase of lesion formation

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study investigated the impact of induced endometriotic lesions on gut microbiota in mice, using GFP+ uterine tissue transplantation. Despite successful lesion formation, no significant changes in gut microbiota composition were observed in the acute phase, suggesting endometriosis may not cause pronounced dysbiosis during early lesion development.

What was studied?

The study investigated whether the induction of endometriosis in mice affects the composition of their gut microbiota. It tested this by transplanting uterine tissue fragments into mice and analyzing changes in the gut microbiota before and after endometriosis induction.

 

Who was studied?

Female C57BL/6 wild-type mice and GFP+ transgenic donor mice were used. Uterine tissue from the donor mice was transplanted into the peritoneal cavity of the wild-type mice to induce endometriosis, with sham-transplanted mice serving as controls.

 

What were the most important findings?

Endometriotic lesions successfully developed in the mice, but the study found no significant alterations in the gut microbiota composition within the 21-day observation period. The bacterial community remained stable, indicating no early-phase intestinal dysbiosis due to endometriosis induction.

 

What are the greatest implications of this study?

The study hypothesizes that there is a bi-directional relationship between gut dysbiosis and endometriosis, where alterations in the gut microbiota may influence the development and progression of endometriosis and vice versa. Although this particular study did not find significant changes in the gut microbiota composition within the early phase of endometriosis induction in mice, it suggests the possibility that the gut microbiota could be involved in hormone-related, inflammatory, angiogenic, and vasculogenic processes associated with endometriosis.

Other studies’ findings, which reported dysbiosis following endometriosis induction, further support the idea of a complex interaction between endometriosis and the gut microbiota. This interaction could potentially impact estrogen metabolism, systemic inflammation, and stem cell homeostasis, all of which are implicated in the pathogenesis of endometriosis. However, the study calls for more research to clarify this relationship, including studies on microbial activity and metabolic function, to understand how gut microbiota might affect endometriosis fully.

Endometriosis induces gut microbiota alterations in mice

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study used high-throughput DNA sequencing to show that murine endometriosis alters gut microbiota, notably increasing the Firmicutes/Bacteroidetes ratio and Bifidobacterium levels by day 42, indicating disease-specific dysbiosis. It underscores the need for further research on the long-term effects of endometriosis on gut microbiota and its bidirectional interaction with the host.

What was studied?

The research focused on investigating the impact of murine endometriosis on gut microbiota composition using high-throughput DNA sequencing to explore how the disease affects intestinal microbial communities.

 

Who was studied?

The study subjects were mice. These animals were divided into two groups: one group with induced endometriosis through the intraperitoneal injection of endometrial tissues and a mock group that served as a control.

 

What were the most important findings?

The study’s key findings include the emergence of a distinct gut microbiota composition in mice with endometriosis by day 42 post-modeling, highlighted by an increased Firmicutes/Bacteroidetes ratio and elevated levels of Bifidobacterium. These changes suggest a specific dysbiosis associated with endometriosis.

 

What are the greatest implications of this study?

The study’s most significant implications lie in its pioneering use of high-throughput DNA sequencing to link endometriosis with specific changes in gut microbiota, highlighting the disease’s potential to induce dysbiosis. It suggests the importance of further research to understand the long-term effects of endometriosis on gut microbiota and the bidirectional interactions between the host and its microbiota. This could lead to novel insights into the pathophysiology of endometriosis and inform new therapeutic strategies targeting the gut microbiome.

Gallium Antimicrobial Agents: Innovations in Combating Drug-Resistant Pathogens

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This review highlights gallium's role as an iron mimic disrupting bacterial iron metabolism, effective against multidrug-resistant pathogens. Innovations in gallium delivery, including nanomaterials and synergistic therapies with antibiotics, address bioavailability challenges and enhance its antimicrobial potency.

What was reviewed?

The study reviewed the advancements in the application of gallium and gallium-based compounds as antimicrobial agents. The review aimed to address the challenges of antimicrobial resistance (AMR) by highlighting gallium's unique properties, mechanisms of action, and its potential as a non-antibiotic antibacterial strategy. The review summarized optimization strategies for gallium compounds, such as improving bioavailability and achieving sustained release, alongside synergistic effects with other antimicrobial agents.

Who was reviewed?

The review focused on multidrug-resistant (MDR) pathogens, including Pseudomonas aeruginosa, Mycobacterium tuberculosis, Acinetobacter baumannii, and methicillin-resistant Staphylococcus aureus (MRSA). It also discussed broader applications against Gram-positive and Gram-negative bacteria.

What were the most important findings?

Gallium mimics iron, a vital nutrient for bacterial growth, disrupting iron metabolism by substituting in iron-dependent processes. This "Trojan horse" mechanism inhibits bacterial proliferation by inactivating critical enzymes like ribonucleotide reductase and preventing biofilm formation, a significant defense strategy for bacteria. Key findings include:

Sustained-release systems, such as gallium-doped bioglasses and alloys, provide long-term antibacterial effects suitable for implant-related infections.

Gallium is redox-inert and disrupts bacterial metabolism by replacing iron, thereby impairing DNA synthesis, electron transport, and oxidative stress responses.

Gallium-based compounds have enhanced their solubility and antibacterial efficacy through chelation and nanomaterial delivery systems.

Synergistic strategies, combining gallium with antibiotics or antimicrobial agents, restore the efficacy of resistant antibiotics, reduce required dosages, and mitigate resistance development.

Gallium acts as a redox-inert iron mimic, disrupting bacterial iron-dependent metabolic pathways and enzyme functions, such as ribonucleotide reductase, critical for DNA synthesis. This mechanism renders it effective against MDR pathogens while minimizing bacterial resistance development. Key findings include:

Bioavailability Challenges: Gallium compounds face hydrolysis in physiological conditions, limiting their effectiveness. Strategies like coordination with ligands, incorporation into nanomaterials, and use of gallium-doped bioglasses have significantly enhanced bioavailability and sustained release.

Synergistic Antibacterial Effects: Gallium combined with antibiotics (e.g., ciprofloxacin, vancomycin) restores antibiotic efficacy against resistant strains and enhances their potency. Additionally, gallium complexes combined with metal ions or photodynamic therapies show amplified antibacterial effects.

Biofilm Disruption: Gallium can inhibit biofilm formation, a major resistance mechanism, particularly in Pseudomonas aeruginosa, by reducing iron availability needed for biofilm maintenance.

Nanotechnology Advances: Gallium-loaded nanomaterials, such as liposomes and Janus micromotors, improve targeting and sustained release, offering a promising avenue for clinical applications.

What are the greatest implications of this review?

Gallium represents a promising alternative to traditional antibiotics, especially against MDR pathogens. The review highlights potential clinical applications in treating implant-related infections, respiratory conditions, and other systemic infections. However, limitations like low bioavailability and the need for targeted delivery require further research. Nonetheless, its role as a complement to existing antibiotics could significantly delay resistance development and enhance antimicrobial strategies.

Gut microbiota imbalance and its correlations with hormone and inflammatory factors in patients with stage 3/4 endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This case control study explored the gut microbiota in stage 3/4 endometriosis (EM) by comparing fecal and blood samples from 12 EM patients and 12 controls using 16S rRNA sequencing. Results showed reduced α diversity and an increased Firmicutes/Bacteroidetes ratio in EM patients, with notable taxonomic differences and elevated estradiol and IL-8 levels. The study suggests microbiota-related pathways may influence EM, indicating directions for further research.

What was studied?

The study investigated the role of gut microbiota in endometriosis (EM), focusing on its differences between individuals with stage 3/4 EM and healthy controls and how these differences correlate with serum hormone levels and inflammatory cytokines.

 

Who was studied?

The research involved 12 patients diagnosed with stage 3/4 endometriosis and 12 healthy control subjects. The researchers compared their gut microbiota compositions and measured serum levels of hormones and inflammatory cytokines.

 

What were the most important findings?

Key findings included a lower α diversity of gut microbiota and a higher Firmicutes/Bacteroidetes ratio in the EM group compared to controls. Significant differences in the abundances of various taxa were observed, along with higher serum levels of estradiol (E2) and interleukin-8 (IL-8) in the EM group. The study also identified correlations between specific microbial abundances and levels of estradiol and IL-8.

 

What are the greatest implications of this study?

The study’s implications suggest that the gut microbiota may play a significant role in the pathophysiology of endometriosis through its influence on hormonal and inflammatory pathways. These findings open potential avenues for novel therapeutic strategies targeting the gut microbiota in endometriosis management and highlight the need for further research to verify and expand upon these preliminary observations.

In vitro antibiofilm, antibacterial, antioxidant, and antitumor activities of the brown alga Padina pavonica biomass extract

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

P. pavonica methanolic extract exhibited strong antibacterial activity against Staphylococcus aureus, Enterococcus faecalis, Pseudomonas aeruginosa, Klebsiella pneumonia, Bacillus subtilis, and moderate antibacterial activity against Escherichia coli, Pseudomonas fluorescens and Streptococcus agalactiae. The study concludes that P. pavonica methanolic extract exhibited effective antibiofilm, antibacterial, antioxidant, and anticancer activities.

What was studied?
The study focused on evaluating the antibiofilm, antibacterial, antioxidant, and anticancer activities of the methanolic extract of the marine algae Padina pavonica L.

 

Who was studied?
The subjects of the study were various microorganisms including Staphylococcus aureus, Enterococcus faecalis, Pseudomonas aeruginosa, Klebsiella pneumoniae, Bacillus subtilis, Escherichia coli, Pseudomonas fluorescens, and Streptococcus agalactiae. Additionally, the extract’s effect on lung carcinoma cells was tested.

 

What were the most important findings?

The extract showed high inhibitory action against biofilm formation (88-99% effectiveness). It displayed strong antibacterial activity against several identified bacterial species and moderate activity against others. The extract demonstrated significant antioxidant properties with 84.59% DPPH radical scavenging activity. And the extract exhibited potent anticancer activity against lung carcinoma with a high range of inhibitory percent (1.79-98.25%) and a low IC50 value (15.14 µg/ml).

 

What are the greatest implications of this study?

This study highlights the potential of Padina pavonica as a source of natural compounds with multiple therapeutic applications, including treating infections, combating cancer, and preventing oxidative stress. The broad spectrum of activities suggests that P. pavonica could be developed into various pharmacological or nutraceutical products, thereby supporting biodiversity conservation and offering new avenues for drug development from marine resources.

Inflammatory cytokines IL-6, IL-10, IL-13, TNF-α and peritoneal fluid flora were associated with infertility in patients with endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This cross-sectional observational study examined the link between inflammatory markers (IL-6, IL-10, IL-13, TNF-α), peritoneal fluid bacterial flora, and infertility in endometriosis patients. Results showed significantly higher white cell counts and elevated levels of inflammatory markers in endometriosis patients compared to controls. Logistic regression confirmed significant associations between these inflammatory markers and infertility.

What was studied?

The study investigated the relationship between inflammatory markers (IL-6, IL-10, IL-13, and TNF-α), the composition of bacterial flora in peritoneal fluid, and infertility in patients with endometriosis.

 

Who was studied?

The participants included 55 patients diagnosed with endometriosis and infertility (observation group) attending a Gynecology Clinic from June 2014 to July 2017 and 30 individuals without endometriosis or infertility issues (control group).

 

What were the most important findings?

The study found elevated white cell counts (monocytes, neutrophils, eosinophils, and basophils) and higher levels of inflammatory cytokines (IL-6, IL-10, IL-13, and TNF-α) in the peritoneal fluid of endometriosis patients with infertility compared to controls. A significant correlation between these inflammatory markers and endometriosis associated with infertility was also established.

 

What are the greatest implications of this study?

The study implies that inflammatory factors in peritoneal fluid play a crucial role in the pathophysiology of infertility associated with endometriosis. These findings suggest that inflammatory cytokines (IL-6, IL-10, IL-13, and TNF-α) could serve as significant biomarkers for diagnosing and understanding the mechanisms of endometriosis-related infertility.

Lippia origanoides derivatives in vitro evaluation on polymicrobial biofilms: Streptococcus mutans, Lactobacillus rhamnosus and Candida albicans

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study assessed the impact of Lippia origanoides derivatives on biofilms formed by Streptococcus mutans, Lactobacillus rhamnosus, and Candida albicans, and their cytotoxic effects on human skin and periodontal cells. Results showed that the essential oils and terpenes, particularly carvacrol and thymol, effectively inhibited biofilm formation and had cytotoxic effects comparable to chlorhexidine. These findings suggest potential clinical applications for L. origanoides compounds.

What was studied?

The study investigated the antimicrobial effects of essential oils derived from the Colombian plant Lippia origanoides, specifically focusing on two terpenes, thymol and carvacrol, against polymicrobial biofilms of Streptococcus mutans, Lactobacillus rhamnosus, and Candida albicans. The study also assessed the cytotoxic effects of these compounds on human skin keratinocytes (HaCaT) and periodontal ligament fibroblasts (FLP).

 

Who was studied?

The microorganisms studied were Streptococcus mutans, Lactobacillus rhamnosus, and Candida albicans. Additionally, the effects of the compounds on human cell lines, specifically HaCaT keratinocytes and FLP fibroblasts, were evaluated to assess cytotoxicity.

 

What were the most important findings?

The study revealed several key findings regarding the effects of Lippia origanoides essential oils, especially those rich in thymol and carvacrol. These oils demonstrated significant antimicrobial activity, inhibiting and eradicating biofilms of the studied microorganisms, both in their formation and preformed states. Additionally, the essential oils were found to have a cytotoxic impact on HaCaT and FLP cell lines, comparable to that of chlorhexidine, indicating their effectiveness without increased toxicity. Furthermore, the oils effectively disrupted biofilm integrity, leading to a reduction in the number of microbial cells and the extracellular matrix.

 

What are the greatest implications of this study?

This study highlights the potential of natural compounds, specifically thymol and carvacrol from Lippia origanoides, as effective alternatives to traditional antimicrobials like chlorhexidine for treating oral biofilms without increasing cytotoxicity. These findings support further research into natural remedies that could combat microbial resistance and provide safer, cost-effective treatments for biofilm-associated infections. Additionally, the study underscores the importance of exploring the biodiversity of plants like Lippia origanoides for developing new therapeutic agents.

Meta-analysis of gut microbiome studies identifies disease-specific and shared responses

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This meta-analysis standardized and re-analyzed data from 28 gut microbiome studies across ten diseases, identifying consistent microbiome signatures associated with specific diseases and a non-specific response common to multiple conditions. Key findings suggest both potential microbial diagnostics and treatments, emphasizing the importance of understanding shared versus disease-specific microbial responses in future research and clinical applications.

What was studied?

The meta-analysis focused on the human gut microbiome’s association with various diseases by analyzing 28 published case-control gut microbiome studies covering ten diseases. The researchers aimed to standardize the processing and analysis of these datasets to identify consistent patterns and shifts in the gut microbiome associated with specific diseases or a generalized health-disease spectrum.

Who was studied?

The participants of the original case-control studies comprised individuals with different diseases, including colorectal cancer, inflammatory bowel disease (IBD), and others, alongside control groups of healthy individuals. The meta-analysis integrated data only from studies with publicly available 16S amplicon sequencing data of stool samples from at least 15 case patients, excluding studies focused solely on children under 5 years old.

 

What were the most important findings?

Consistent Microbial Patterns: The meta-analysis revealed consistent and specific microbiome changes associated with various diseases. For instance, diseases like colorectal cancer showed an enrichment of pathogenic bacteria, while a depletion of health-associated bacteria marked conditions like IBD.

Non-Specific Microbial Responses: A significant finding was that many microbial associations are not disease-specific but rather indicate a non-specific response shared across multiple disease states. Approximately half of the genera identified were common to more than one disease, suggesting a generalized microbial response to disease states rather than unique disease-specific signatures.

Diagnostic and Therapeutic Implications: The study identified distinct categories of dysbiosis (microbial imbalance) that could guide the development of microbiome-based diagnostics and therapeutics. For example, enriching for depleted beneficial microbes could be a strategy for diseases characterized by such depletions.

 

What are the greatest implications of this meta-analysis?

Improved Disease Understanding: By providing a clearer picture of the microbiome’s role in various diseases, the study helps refine our understanding of disease mechanisms and potential microbial contributions to disease processes.

Guidance for Future Research: The findings suggest that future microbiome research in disease contexts should consider the non-specificity of many microbial changes. This realization could influence how researchers design studies and interpret results, potentially focusing on truly disease-specific microbial signatures.

Clinical Applications: The identification of consistent microbial patterns and signatures across diseases opens pathways to developing novel diagnostics and therapies, such as probiotics or fecal microbiota transplants, targeted at restoring healthy microbial communities or addressing specific dysbioses.

Data Sharing and Standardization: The study underscores the value of making raw data and metadata from microbiome studies publicly available and highlights the benefits of using standardized methods for data processing and analysis to compare and integrate results across studies.

Overall, this meta-analysis clarifies the microbiome’s role in disease and sets a framework for future research and clinical applications by demonstrating the importance of understanding both disease-specific and non-specific microbial responses.

Microbiome of the lower genital tract in Chinese women with endometriosis by 16s-rRNA sequencing technique: a pilot study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This case-control study investigates the relationship between genital tract microbiota and endometriosis using 16s-rRNA sequencing. Findings show Atopobium prevalence in endometriosis with adenomyosis cases, highlighting microbiota's distinct functions.

What was studied?

The study investigated the relationship between the genital tract microbiota and endometriosis, particularly focusing on how microbiota diversity and specific bacteria like Atopobium might be associated with the disease.

 

Who was studied?

68 participants, from whom 134 samples were collected from the cervical canal, posterior fornix, and uterine cavity for 16s-rRNA sequencing, were included in the study.

 

What were the most important findings?

Key findings included no significant differences in alpha diversity between the cervical canal and posterior fornix. However, the microbiota profile of patients with adenomyosis and endometriosis differed markedly from the control group, with Atopobium showing significant prevalence in these patients. While no specific biomarkers were identified, PICRUSt analysis revealed several characteristic microbiota functions.

 

What are the greatest implications of this study?

The study suggests a potentially significant role of microbiota, particularly Atopobium, in the pathogenesis of endometriosis combined with adenomyosis. This finding could lead to new insights into the microbiota-immune-endometriosis system interaction, offering new avenues for understanding and possibly treating endometriosis and adenomyosis. Further research is needed to verify the functions of the microbiota identified and their direct association with the diseases.

Microbiome Profile of Deep Endometriosis Patients: Comparison of Vaginal Fluid, Endometrium and Lesion

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This case study found that a distinct bacterial composition was observed in deep endometriotic lesions, characterized by a reduced prevalence of Lactobacillus and an increased abundance of Alishewanella, Enterococcus, and Pseudomonas.

What was studied?

The study focused on identifying and comparing the bacterial patterns present in the vaginal fluid, eutopic endometrium, and endometriotic lesions of patients with endometriosis to those found in the vaginal fluid and eutopic endometrium of control patients without the disease. High-throughput DNA sequencing of the 16S rRNA marker gene was utilized to analyze the microbiome profile in these different biological samples from both groups.

 

Who was studied?

Twenty-one patients participated in this study, divided into two groups: eleven in the control group and ten in the endometriotic group. The control group consisted of women who underwent laparoscopic surgery for benign gynecological diseases or elective tubal ligation, where the absence of endometriosis was confirmed during peritoneal cavity inspection. The endometriotic group included only women with deep endometriosis, confirmed by laparoscopic surgery and histopathology analysis.

 

What were the most important findings?

The study found that microbiome sequencing of vaginal fluid, eutopic endometrium, and endometriotic lesions typically showed similar profiles, dominated by Lactobacillus, Gardnerella, Streptococcus, and Prevotella. Despite no significant overall differences in microbiome diversity between control and endometriotic patients, deep endometriotic lesions exhibited a distinct bacterial composition with less Lactobacillus and a higher abundance of Alishewanella, Enterococcus, and Pseudomonas.

 

What are the greatest implications of this study?

The study highlights several implications for endometriosis management: It provides insights into the pathogenesis by showing distinct bacterial compositions in deep lesions, suggesting microbial involvement in lesion development. This leads to the potential for non-invasive diagnostics by identifying specific microbial patterns, opening avenues for biomarker-based detection. Therapeutically, interventions like antibiotics or probiotics could be new treatment strategies if certain bacteria contribute to pathogenesis. The findings emphasize the need for more research to establish causal links between microbiome composition and endometriosis, understand the bacterial influence on the disease, and explore microbiome-based treatments.

Microbiota composition and distribution along the female reproductive tract of women with endometriosis

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This cross-sectional observational study compared microbiota in various reproductive tract locations finding significant microbiota changes in endometriosis patients, especially a decrease in Lactobacillus and increase in specific bacteria in the cervical area.

What was studied?

The study investigated the microbiota distribution across the entire female reproductive tract of endometriosis (EMS) patients and non-EMS women, aiming to identify EMS-specific bacterial species and examine the relationship between flora and disease development.

 

Who was studied?

Fifty women undergoing laparoscopic surgery for benign gynecological diseases or pelvic endometriosis at Peking University Shenzhen Hospital were studied. They were divided into two groups: 36 with pelvic endometriosis (stages I-IV) and 14 controls without endometriosis symptoms.

 

What were the most important findings?

Significant differences in the microbiota distribution were observed, especially a decrease in Lactobacillus in the upper reproductive tract of EMS patients. Specific Operational Taxonomic Units (OTUs), particularly Sphingobium sp. and Pseudomonas viridiflava, were identified as significantly enriched in the endometrium and peritoneal fluid of EMS patients, suggesting their potential role in EMS pathogenesis.

 

What are the greatest implications of this study?

The study offers a new perspective on the pathogenesis of endometriosis, emphasizing the role of specific bacteria in its development. Identifying microbiota changes associated with EMS could lead to novel diagnostic markers and therapeutic targets, enhancing our understanding of the disease and potentially leading to more effective management strategies for endometriosis and other female reproductive tract diseases.

Molecular detection of microbial colonization in cervical mucus of women with and without endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This cross-sectional observational study investigates cervical mucus in women with and without endometriosis, finding similar microbial distributions overall. However, bacteria such as Corynebacterium, Enterobacteriaceae, Flavobacterium, Pseudomonas, and Streptococcus were more common in women with endometriosis, with Enterobacteriaceae and Streptococcus showing significant associations

What was studied?

The study investigated the microbiota in the cervical mucus of women with and without endometriosis using next-generation sequencing (NGS) technologies. It aimed to clarify whether cervical mucus in women with endometriosis is contaminated with bacteria, which could potentially transmigrate into the intrauterine cavity, influencing the pathogenesis of endometriosis. The research explored the bacterial population’s diversity, its correlation with the disease, and how these findings align with previous studies on intrauterine microbial colonization and its role in endometriosis through LPS/TLR4 engagement of the innate immune system.

 

Who was studied?

The study’s participants consisted of 30 women diagnosed with endometriosis (confirmed by laparoscopy and classified according to the revised American Society for Reproductive Medicine scoring system for stages III-IV) and 39 women without the condition (control group), all of reproductive age (20-44 years). The control group included women with fibroids or benign ovarian tumors other than endometriosis. All subjects had a normal-appearing cervix, were negative for vaginal culturing tests, and had not received endocrine therapy or antibiotics for at least six months before sample collection.

 

What were the most important findings?

The study highlights several findings about the cervical mucus microbiota in women with and without endometriosis. It found a diverse array of microbiota in both groups, with variations not tied to menstrual cycle phases. The endometriosis group exhibited significantly higher alpha diversity, indicating a more diverse bacterial community. Specific bacteria such as Enterobacteriaceae, Corynebacterium, Pseudomonas, Flavobacterium, and Streptococcus were more prevalent in the endometriosis group, with Enterobacteriaceae and Streptococcus showing significantly higher prevalence.

What are the greatest implications of this study?

The findings suggest that the cervical mucus of women with endometriosis harbors a distinct and more diverse bacterial population than women without the condition. The significant presence of specific bacteria, particularly Enterobacteriaceae and Streptococcus, in women with endometriosis may play a role in the disease’s pathogenesis through mechanisms involving bacterial contamination, immune system engagement, and inflammatory responses. These insights open new avenues for understanding endometriosis’s pathophysiology, potentially leading to novel diagnostic and therapeutic strategies. Further, the study implies a possible link between cervical mucus microbiota and adverse pregnancy outcomes, highlighting the need for additional research to explore the mechanistic connections between microbial colonization and endometriosis and its implications for fertility and pregnancy.

Reproductive Microbiomes: Using the Microbiome as a Novel Diagnostic Tool for Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This cross-sectional observational study examines how endometriosis affects the bacterial communities of the uterus and cervix, considering the condition's role in inflammation, pain management, and infertility in women.

What was studied?

The study aimed to investigate how endometriosis affects the uterine and cervical bacterial communities. Utilizing next-generation amplicon sequencing of the bacterial 16S rRNA gene, the research sought to identify alterations in these microbiomes associated with endometriosis and to determine if specific bacterial taxa within the cervix could help diagnose active endometriosis, potentially avoiding the need for invasive diagnostic procedures like laparoscopic surgery.

 

Who was studied?

Nineteen pre-menopausal women undergoing laparoscopic surgery for pelvic pain with suspicion or known endometriosis constituted the experimental group (n=10, with endometriosis stages I-IV), while women undergoing surgery for benign uterine or ovarian conditions served as controls (n=9). The control group was examined during surgery to confirm the absence of endometriotic lesions. The staging of endometriosis for patients in the experimental group was performed using the revised American Society for Reproductive Medicine (rASRM) classification scale.

 

Key findings of the study include significant differences in bacterial communities between uterine and cervical samples, both in species diversity and abundance, with the uterus displaying a diverse profile of Bacteroidetes and Firmicutes, and the cervix dominated by Lactobacillus. No significant differences in bacterial communities were noted across different endometriosis stages on the day of surgery. However, a distinct cervical bacterial community in a stage III endometriosis patient suggests a link between disease severity and microbiome alterations. Additionally, notable fluctuations in the cervical microbiome were observed over time in this patient, indicating dynamic microbiome changes associated with disease progression and treatment.

 

What are the greatest implications of this study?

The findings underscore the potential of bacterial community profiling as a diagnostic tool for endometriosis, offering a non-invasive method to identify the disease in asymptomatic, infertile women. This approach could facilitate earlier diagnosis and treatment, potentially improving fertility outcomes and reducing the need for invasive diagnostic surgeries. The study also highlights the dynamic nature of the uterine and cervical microbiomes in relation to endometriosis, suggesting that microbiome alterations could be linked to disease severity and progression. Understanding these microbial community changes opens new avenues for researching endometriosis pathogenesis and developing novel therapeutic strategies that target microbiome modulation. Moreover, the fluctuations observed in the microbiome over time, especially in patients with advanced disease, may offer insights into predicting disease progression and treatment outcomes, including fertility potential post-treatment.

Role of Cholestyramine in Refractory Hyperthyroidism: A Case Report and Literature Review

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

A 52-year-old woman with refractory iodine-induced hyperthyroidism showed significant improvement with cholestyramine, reducing FT4 by 30% in 5 days. Despite conventional treatments failing, cholestyramine proved effective, leading to euthyroidism. This highlights cholestyramine's potential as an adjunct therapy.

What was studied?

The study investigated the role of cholestyramine as an additional treatment for refractory iodine-induced hyperthyroidism in a patient who did not respond to conventional therapies.

Who was studied?

A 52-year-old female patient with a history of goiter who developed iodine-induced hyperthyroidism following a CT scan with contrast. The patient had obstructive symptoms and was unresponsive to standard treatments, including dexamethasone, carbimazole, and propranolol.

What were the most important findings?

After adding cholestyramine, the patient’s FT4 levels decreased by 30% within 5 days and normalized by 12 days.

What are the greatest implications of this study?

Cholestyramine can be an effective adjunct therapy for managing refractory iodine-induced hyperthyroidism, suggesting a potential new treatment avenue for similar cases, such as Grave's Disease (GD). This case highlights the need for alternative treatments when conventional therapies fail and emphasizes the utility of cholestyramine in rapid thyroid hormone reduction.

Role of Cholestyramine in Refractory Hyperthyroidism A Case Report and Literature Review

The impact of photobiomodulation of major salivary glands on caries risk

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study evaluated the effects of photobiomodulation on salivary glands in high caries-risk patients, finding that light therapies significantly reduced cariogenic bacteria and improved salivary parameters, suggesting a promising non-invasive approach to dental caries prevention.

What was studied? 

The study explored the impact of photobiomodulation therapy using different types of light on the major salivary glands and its subsequent effects on caries risk factors in individuals with high risk for dental caries. The therapy modalities tested included polarized polychromatic light, continuous LED light, and pulsed LED light, each aiming to modify salivary parameters known to influence caries development, such as bacterial counts of Streptococcus mutans and Lactobacillus, salivary flow, and buffering capacity.

 

Who was studied? 

The subjects of this study were thirty-six patients identified as having a high risk for dental caries. These individuals were randomly assigned to one of four groups, with three groups receiving distinct light treatments and one group serving as a control that was exposed to non-therapeutic visible light. The study’s interventions were applied extra-orally and intra-orally over multiple sessions spanning a four-week period.

 

What were the most important findings?  

The study’s findings revealed significant improvements in caries risk factors for the groups treated with light therapies. Notably, there was a reduction in the bacterial counts of Streptococcus mutans and Lactobacillus, and enhancements in both stimulated and unstimulated salivary flow and buffering capacity in the groups exposed to light therapy, with statistically significant changes observed. Conversely, no significant changes were observed in the control group.

 

What are the greatest implications of this study?

The greatest implications of this study are its contributions to the field of non-invasive dental treatments and the potential for reducing caries risk through such innovative approaches. By demonstrating that photobiomodulation can effectively decrease the levels of cariogenic bacteria and enhance salivary function, the study supports the broader application of light therapy in preventive dental care, particularly for patients at high risk of caries. This could lead to more targeted, conservative, and effective management strategies for dental caries, aligning with broader preventive healthcare goals.

The influence of nickel on intestinal microbiota disturbances

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

Excess nickel disrupts gut microbiota, promoting dysbiosis and contributing to conditions like obesity and systemic nickel allergy syndrome (SNAS). Probiotics and nickel-restricted diets show promise in mitigating these effects, underscoring the need for further research and clinical intervention.

What was reviewed?

The paper reviewed the influence of nickel on intestinal microbiota disturbances, drawing on 59 scientific publications from the past 20 years. The analysis focused on nickel’s dual role as an essential element for microbial enzymatic reactions and a disruptor of gut microbiota, especially under conditions of excessive exposure or systemic nickel allergy syndrome (SNAS).

Who was reviewed?

The review encompassed research involving humans, animals, and microbial models. Specific attention was given to populations exposed to high levels of nickel, individuals with SNAS, and animal studies demonstrating changes in microbial communities under nickel exposure.

What were the most important findings?

Nickel acts as a cofactor for metalloenzymes like urease, hydrogenase, and [NiFe]-hydrogenase, essential for microbial survival. However, excess nickel promotes dysbiosis, characterized by reductions in beneficial taxa and increases in nickel-resistant bacteria. In humans with SNAS, the microbiota showed decreased levels of beneficial genera such as Bifidobacterium and Lactobacillus, known for their probiotic effects and urease activity, and increases in nickel-tolerant taxa, including Clostridiaceae and Bacillaceae. Similarly, animal studies indicated reduced Verrucomicrobia and Bacteroidetes while promoting Escherichia coli and Enterococcus.

Nickel exposure also leads to an increased abundance of Bacteroides fragilis, Bacteroidales S24-7, and Interstinimonas, with a concurrent decline in Firmicutes, disrupting the Firmicutes-to-Bacteroidetes ratio, a critical marker of gut health. This imbalance contributed to systemic inflammation and altered immune responses. Moreover, nickel-reliant pathogens, such as Helicobacter pylori, which require Ni2+-dependent enzymes like urease for colonization, further highlighted nickel’s role in microbial pathogenicity. Probiotic strains such as Lactobacillus fermentum demonstrated detoxifying effects by metabolizing nickel, suggesting their therapeutic potential.

What are the greatest implications of this review?

The findings reveal that nickel exposure significantly alters gut microbial ecology, driving dysbiosis and systemic inflammation in susceptible populations. The rise of nickel-tolerant taxa, coupled with the decline of protective bacteria, underscores nickel’s role as a disruptor of gut homeostasis, contributing to conditions like obesity and SNAS. Probiotic supplementation, particularly strains capable of nickel detoxification, and dietary restrictions like a low-nickel diet, have shown promise in mitigating these effects. This review highlights the urgent need for dietary nickel regulations and further clinical studies on therapeutic interventions targeting nickel-induced microbial dysbiosis.

The Revaluation of Plant-Derived Terpenes to Fight Antibiotic-Resistant Infections

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

The study investigated the antimicrobial effects of plant-derived terpenes against antibiotic-resistant pathogens. Key findings revealed that terpenes disrupt microbial membranes, inhibit biofilm formation, and block efflux pumps. These properties suggest terpenes as potential alternatives to traditional antibiotics, offering new strategies to combat the increasing challenge of antibiotic resistance.

What was studied?
The study focused on terpenes derived from plants as potential antimicrobial agents, particularly their effectiveness against antibiotic-resistant infections. This included an exploration of the diverse mechanisms by which terpenes combat microbial resistance, their bioavailability, and the synergistic effects when combined with other antimicrobials.

 

Who was studied?
The research mainly involved microbial organisms that are known to exhibit resistance to conventional antibiotics. This included a variety of pathogens such as Gram-positive and Gram-negative bacteria, with specific attention to multi-drug resistant strains.

 

What were the most important findings?
The study found that plant-derived terpenes have significant antimicrobial activity against resistant strains. It highlighted the mechanisms of action of terpenes, such as disruption of microbial membranes, inhibition of biofilm formation, and efflux pump inhibition. These findings suggest terpenes’ potential as effective agents in combating antibiotic-resistant infections.

 

What are the greatest implications of this study?
The implications are substantial in the context of global health challenges posed by antibiotic resistance. The findings suggest that terpenes could be developed into new antimicrobial agents that offer a viable alternative to traditional antibiotics. This could lead to the development of novel therapeutic strategies and formulations, potentially reducing the prevalence of resistant infections and the healthcare burdens associated with them.

The Vaginal Microbiome as a Tool to Predict rASRM Stage of Disease in Endometriosis: a Pilot Study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This pilot study analyzed gut and vaginal microbiomes in 59 women (35 with endometriosis, 24 controls) using 16S rRNA sequencing and machine learning to explore their diagnostic potential for endometriosis. Findings indicate microbiome variations with the menstrual cycle and disease severity, suggesting that vaginal microbiome profiles could predict endometriosis stages, offering a novel, less-invasive diagnostic method.

What was studied?

The study focused on characterizing the gut and vaginal microbiome profiles of women with endometriosis compared to controls without the disease, exploring the potential of these profiles as less-invasive diagnostic tools for assessing the severity of endometriosis.

 

Who was studied?

Fifty-nine women participated in the study, including 35 with endometriosis and 24 control subjects. Rectal and vaginal samples were collected from all participants at two different periods of their menstrual cycle.

 

What were the most important findings?

Significant findings included variations in the distribution of vaginal community state types (CSTs) across different phases of the menstrual cycle and differences in gut and vaginal microbiome profiles between patients with varying stages of endometriosis as classified by the revised American Society for Reproductive Medicine (rASRM) stages. Machine-learning models could predict the severity of endometriosis (stages 1-2 vs. 3-4) based on these microbiome profiles, with Anaerococcus genus showing the highest predictive value.

 

What are the greatest implications of this study?

The study suggests that analysis of the vaginal microbiome could serve as a novel, less-invasive method to diagnose and predict the stage of endometriosis. This approach could potentially lead to earlier and more accurate diagnoses of endometriosis, improving treatment planning and outcomes for affected women.

Urinary lead concentration and composition of the adult gut microbiota in a cross-sectional population-based sample

May 20, 2025
  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This study explores the link between urinary lead concentration and adult gut microbiota composition. Conducted on 696 participants, it reveals associations between lead levels and increased microbial diversity and richness. Specific bacterial groups, like Proteobacteria, correlate with elevated lead.

What was studied?

The study examined the association between urinary lead concentrations and changes in the gut microbiota composition in a population-based sample of adults. The research explored how varying levels of lead exposure could impact microbial diversity, richness, and specific bacterial taxa within the gut.

 

Who was studied?

The study population comprised adults from Wisconsin, USA, who participated in the Survey of the Health of Wisconsin (SHOW) and its ancillary microbiome study. Participants were mainly over 18 years of age, with a diverse distribution in terms of age, gender, income, and education levels.

 

What were the most important findings?

Key findings from the study revealed that urinary lead concentrations were associated with significant alterations in the gut microbiota. Notably, there was an increase in microbial α-diversity and richness. Furthermore, higher urinary lead levels were correlated with changes in microbial β-diversity, and these changes were linked to the presence of specific bacterial taxa such as Proteobacteria, including members of the Burkholderiales. Intriguingly, these alterations in the microbiota were observed even in adults who had relatively low levels of lead exposure compared to national averages. This suggests that even minimal lead exposure can have noticeable impacts on gut microbiota composition, underscoring the sensitivity of human microbiota to environmental contaminants.

 

What are the greatest implications of this study?

The implications of this study suggest a significant environmental health concern where even low levels of lead exposure can influence gut microbiota composition, potentially affecting overall human health. The findings underscore the need for further research to understand the long-term health outcomes associated with microbiota changes due to environmental contaminants like lead. Additionally, this study contributes to the broader understanding of how environmental factors can impact microbial communities in the human body, which is crucial for developing strategies to mitigate such effects.  These insights highlight the complex interactions between environmental pollutants and human health, emphasizing the importance of monitoring and regulating environmental lead exposures.

Metals

Go to Category Page

Bacterial Iron Detoxification Mechanisms: Insights into Iron Homeostasis and Oxidative Stress Mitigation

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This review explores bacterial iron homeostasis, focusing on detoxification pathways, oxidative stress mitigation, and iron-storage mechanisms. It highlights regulatory proteins like Fur and DtxR and storage proteins like ferritins. These insights provide potential therapeutic targets for limiting bacterial growth and addressing iron-related dysbiosis in host-pathogen interactions.

What was reviewed?

This review examines the mechanisms bacteria employ to manage iron homeostasis, detoxify excess iron, and mitigate oxidative stress caused by reactive oxygen and nitrogen species. The review surveys molecular insights into various iron-regulating proteins and pathways, highlighting their structures, functions, and evolutionary significance. These mechanisms are critical for bacterial survival, especially under oxidative stress and during host-pathogen interactions.

Who was reviewed?

The review focuses on bacteria from diverse ecological niches, including both Gram-positive and Gram-negative species. It examines key bacterial proteins and regulatory systems such as Fur (ferric uptake regulator), DtxR/IdeR (iron-dependent regulators), and novel regulators like RirA and Irr. The study also investigates iron storage proteins like ferritins and mini-ferritins (Dps), alongside mechanisms for iron acquisition, transport, and detoxification.

What were the most important findings?

The review highlights iron's dual role as an essential nutrient and potential toxin due to ROS generation via the Fenton reaction. Proteins like Fur, DtxR, and RirA regulate iron homeostasis by balancing uptake, storage, and efflux to prevent oxidative stress. Ferritins and Dps detoxify excess iron by sequestering it, while siderophores facilitate iron acquisition. The interplay between iron homeostasis and ROS/RNS detoxification is critical during immune responses, with bacteria exhibiting diverse adaptations to manage iron under varying environmental stresses.

What are the greatest implications of this review?

The insights provided by this review have significant implications for understanding bacterial survival strategies under nutrient limitation and oxidative stress. These findings offer potential therapeutic targets for managing bacterial infections. Iron acquisition and storage systems are particularly attractive targets, as their disruption could limit bacterial growth and virulence. Additionally, understanding bacterial iron detoxification pathways can inform microbiome-targeted interventions (MBTIs) such as iron chelation, especially in conditions where dysbiosis may alter iron homeostasis or oxidative stress.

Beneficial Effects of a Low-Nickel Diet on Relapsing IBS-Like and Extraintestinal Symptoms of Celiac Patients during a Proper Gluten-Free Diet: Nickel Allergic Contact Mucositis in Suspected Non-Responsive Celiac Disease

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study provides strong evidence that nickel-rich foods in a gluten-free diet can trigger or exacerbate IBS-like gastrointestinal and extraintestinal symptoms in a subset of celiac disease patients, even when the disease is in serological and histological remission. The findings highlight the importance of nickel sensitivity in the pathogenesis of these relapsing symptoms, suggesting that a low-nickel diet can be a valuable intervention to improve patient outcomes. The use of the nickel oral mucosa patch test (Ni omPT) was also validated as a reliable diagnostic tool for identifying nickel-sensitive patients.

What was studied?

The study investigated the prevalence and effects of nickel allergic contact mucositis (Ni ACM) in celiac disease (CD) patients who were on a proper gluten-free diet (GFD) but experienced a relapse of Irritable Bowel Syndrome (IBS)-like gastrointestinal and extraintestinal symptoms. The main goal was to determine whether nickel-rich foods in a gluten-free diet could trigger these symptoms and to evaluate the impact of a low-nickel diet (LNiD) on symptom reduction in these patients. This pilot study also explored the use of the nickel oral mucosa patch test (Ni omPT) to diagnose nickel sensitivity in these patients.

Who was studied?

The study involved 102 consecutive adult celiac disease patients (74 females, 28 males, mean age 42.3 ± 7.4 years) who had been on a gluten-free diet for at least 12 months and were in serological and histological remission of their disease. These patients were selected because they reported relapsing or persisting gastrointestinal and extraintestinal symptoms despite proper adherence to a GFD. After exclusions based on comorbid conditions like lactose intolerance and Helicobacter pylori infection, 20 patients (all female, age 23–65 years) were included in the final analysis.

What were the most important findings?

In a cohort of 20 patients with persistent symptoms despite adherence to a gluten-free diet (GFD), all tested positive for nickel sensitivity via the nickel oral mucosa patch test (Ni omPT), confirming a diagnosis of nickel allergic contact mucositis (Ni ACM). Following prolonged GFD, 83.3% of patients experienced a relapse of symptoms, including abdominal pain, bloating, loose stools, and fatigue, coinciding with high dietary nickel intake from nickel-rich gluten-free foods such as corn. Implementing a low-nickel diet (LNiD) for three months improved 83.4% of total symptoms, with 41.7% reaching statistical significance. Notably, 80% of gastrointestinal and 88.9% of extraintestinal symptoms improved, including significant relief from abdominal pain, swelling, fatigue, and dermatitis. Combining LNiD with GFD restored patients' well-being to levels comparable to those previously achieved by GFD alone, strongly implicating nickel sensitivity as the primary driver of symptom relapse.

What are the greatest implications of this study?

This study provides strong evidence that nickel-rich foods in a gluten-free diet can trigger or exacerbate IBS-like gastrointestinal and extraintestinal symptoms in a subset of celiac disease patients, even when the disease is in serological and histological remission. The findings highlight the importance of nickel sensitivity (Ni ACM) in the pathogenesis of these relapsing symptoms, suggesting that a low-nickel diet (LNiD) can be a valuable intervention to improve patient outcomes. The use of the nickel oral mucosa patch test (Ni omPT) was also validated as a reliable diagnostic tool for identifying nickel-sensitive patients.

Clinically, these results imply that gastroenterologists should consider nickel sensitivity as a differential diagnosis in celiac patients who are non-responsive to a GFD and continue to experience symptoms. The integration of a low-nickel dietary approach alongside the GFD may become an essential part of managing non-responsive celiac disease with overlapping IBS-like symptoms. Further large-scale studies are needed to confirm these findings and refine dietary guidelines for managing nickel sensitivity in this population.

Critical Review on Zeolite Clinoptilolite Safety and Medical Applications in vivo

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

The review highlights clinoptilolite's detoxifying, immunomodulatory, and antioxidant properties, showing its safety and potential for in vivo use in humans and animals. It can adsorb toxic metals like nickel, support gut health, and modulate immune responses, making it valuable for conditions like systemic nickel allergy syndrome and microbiome imbalances, with promising applications in both human supplements and veterinary care.

What was reviewed?

The review focused on the safety and medical applications of clinoptilolite, a naturally occurring zeolite, for in vivo use. The analysis encompassed the physical and chemical properties of clinoptilolite, its detoxifying capabilities, and its potential therapeutic effects in both human and veterinary medicine. The review also considered various studies on clinoptilolite's role in detoxification, immune modulation, oxidative stress, and its impact on body homeostasis. The safety of clinoptilolite for internal consumption, including concerns about the potential release of heavy metals like lead during use, was also addressed.

Who was reviewed?

The review examined the available scientific literature concerning the effects of clinoptilolite in various contexts, specifically in animal studies and human trials. It included research from both clinical studies and preclinical evaluations that utilized clinoptilolite as a dietary supplement or therapeutic agent. The literature encompassed studies on humans, including clinical trials and case studies, as well as extensive research on animals such as dairy cows, rats, mice, and poultry, assessing both the therapeutic benefits and safety profiles of clinoptilolite-based products.

What were the most important findings of this review?

  1. Detoxification Properties: Clinoptilolite exhibits high cation-exchange capacity, making it effective in adsorbing toxic metals such as lead, cadmium, and nickel from the body. This property makes it valuable in reducing the load of toxic elements in animals and humans.
  2. Safety Profile: Clinoptilolite, particularly in its micronized and purified forms, has been shown to be generally safe for in vivo use at varying dosages, with no significant toxic effects observed in animals or humans. The European Food Safety Authority (EFSA) has classified it as safe for consumption in animal feeds at specified doses.
  3. Immunomodulatory and Antioxidant Effects: Clinoptilolite can modulate immune responses, as evidenced by its effects on immune cell counts and antioxidant enzyme activity in various studies. This includes improved responses in immunodeficient conditions and a reduction in markers of oxidative stress.
  4. Impact on Microbiota and Gastrointestinal Health: Clinoptilolite has shown positive effects on gut health, potentially through its detoxifying role, the maintenance of intestinal wall integrity, and its ability to modulate the microbiota composition. These effects suggest potential therapeutic roles in conditions like gastrointestinal disorders, dysbiosis, and enhanced resistance to pathogens.

What are the greatest implications of this review?

  1. Potential for Human Therapeutics: The review highlights the potential use of clinoptilolite as a nutraceutical for detoxifying heavy metals and improving immune responses in humans. Its safety profile supports further exploration into its use in supplements aimed at mitigating conditions related to heavy metal exposure, such as systemic nickel allergy syndrome (SNAS) and other metal-induced toxicities.
  2. Veterinary Applications: Clinoptilolite's effectiveness in improving animal health—particularly its ability to enhance reproductive health, detoxify nitrates, and manage mycotoxins—suggests that it could serve as a valuable supplement in livestock production. Its role in reducing ammonia and supporting gut health is particularly important for animal productivity and welfare.
  3. Integration in Microbiome-Targeted Therapies: Given its impact on gastrointestinal health and the microbiome, clinoptilolite holds promise as a component of microbiome-targeted interventions (MBTIs). It could serve as an adjunctive therapy in managing conditions like irritable bowel syndrome (IBS) and other dysbiosis-associated disorders.
  4. Safety Considerations and Future Research: The review underlines the importance of rigorous quality control and characterization of clinoptilolite materials for in vivo applications. Variability in clinoptilolite's physical and chemical properties across different sources necessitates careful evaluation before therapeutic use. Future studies are needed to delineate the specific mechanisms by which clinoptilolite exerts its effects on immune modulation and detoxification, as well as to confirm its efficacy and safety in long-term human applications.

Does Exposure of Lead and Cadmium Affect the Endometriosis?

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

The study links lead and cadmium exposure to increased endometriosis risk, emphasizing lead's role at low blood levels and synergistic effects with cadmium. It advocates for strict monitoring and preventive measures to minimize exposure.

What Was Studied?

This study investigated the association between occupational exposure to lead and cadmium and the risk of developing endometriosis (EM) among South Korean female workers. Utilizing medical and biological data from over 26,000 individuals who underwent lead-associated medical examinations between 2000 and 2004, the study examined blood lead levels (BLLs), co-exposure to cadmium, and their relationship with hospital admissions for EM.

Who Was Studied?

The study focused on South Korean female workers exposed to lead as part of their occupation. These individuals underwent specialized medical examinations. A total of 26,542 workers were included, with the study comparing EM admissions in lead-exposed workers against the general population and noise-exposed workers as control groups.

What Were the Most Important Findings?

The study found that lead exposure, even at relatively low levels (BLLs < 5 µg/dL), was significantly associated with an increased risk of EM. The standard admission rate (SAR) for EM in lead-exposed workers was 1.24 times higher than the general population, and for workers with BLLs < 5 µg/dL, it was 1.44 times higher. Co-exposure to lead and cadmium demonstrated a synergistic effect, amplifying the risk of EM beyond what could be expected from exposure to either metal alone. While cadmium exposure alone did not show a statistically significant association with EM, the relative excess risk due to interaction (RERI) was 0.33, indicating a notable combined impact of these metals. The study also highlighted that oxidative stress induced by heavy metal exposure likely underpins these effects, with mechanisms involving the disruption of antioxidant defenses and cellular damage.

What Are the Greatest Implications of This Study?

This research underscores the need to minimize exposure to heavy metals, particularly lead and cadmium, among female workers. It also calls for rigorous monitoring of blood lead and cadmium levels in workplaces to mitigate their combined effects. The findings are critical for understanding the environmental and occupational contributors to EM and suggest that policies limiting heavy metal exposure could have a substantial public health impact, especially for at-risk populations.

Elevated Lead, Nickel, and Bismuth Levels in the Peritoneal Fluid of a Peritoneal Endometriosis Patient without Toxic Habits or Occupational Exposure following a Vegetarian Diet

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

A case study links elevated lead, nickel, and bismuth in peritoneal fluid with endometriosis, highlighting potential dietary and environmental exposures as contributors. Further research may identify these potentially toxic elements (PTEs) as diagnostic biomarkers and therapeutic targets.

What Was Studied?

This case report investigated the multielemental profile of peritoneal fluid (PF) in a 22-year-old woman diagnosed with peritoneal endometriosis. The study aimed to evaluate the concentrations of potentially toxic elements (PTEs), including lead (Pb), nickel (Ni), bismuth (Bi), and cobalt (Co), and to compare them with levels in an age-matched control without endometriosis. The patient had no toxic habits, occupational exposure, or documented environmental exposure, and adhered to a vegetarian diet, raising questions about the dietary and environmental sources of PTEs and their role in the pathogenesis of endometriosis.

Who Was Studied?

The primary subject was a 22-year-old woman diagnosed with peritoneal endometriosis during laparoscopic surgery, where her PF was analyzed. The comparison group included an age-matched control and a reference cohort of ten women diagnosed with non-hormonally dependent benign ovarian cysts.

Most Important Findings

The study revealed significantly elevated levels of Pb (75 µg/L, 90:1 ratio), Ni (40.4 µg/L, 4:1 ratio), Bi (33.3 µg/L, 1.5:1 ratio), and Co (1.39 µg/L, 5:1 ratio) in the PF of the endometriosis patient compared to the control. These findings suggest potential contributions of dietary and environmental exposures to PTEs. Nickel, a cofactor for metalloenzymes, was noted to be higher potentially due to the patient’s vegetarian diet, which is associated with increased nickel intake from plant-based foods such as nuts and legumes. Elevated Pb levels were striking, with concentrations much higher than typical dietary or environmental exposures in industrialized settings. While cobalt and bismuth also showed elevated levels, their specific roles in endometriosis remain unclear. The findings support the hypothesis that environmental and dietary PTE exposure may contribute to the pathogenesis of endometriosis by inducing oxidative stress or endocrine disruption.

Greatest Implications

This study highlights the need to explore PTEs as potential biomarkers for endometriosis diagnosis and as contributors to its etiology. Elevated PTE levels in PF may result from dietary habits, such as a vegetarian diet, or unidentified environmental exposures. This study emphasizes the importance of further investigations into environmental toxicology and dietary patterns in endometriosis patients. Understanding these associations could inform preventative strategies, dietary guidelines, and therapeutic interventions for endometriosis management.

Elevated levels of whole blood nickel in a group of Sri Lankan women with endometriosis: a case control study

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study identified elevated blood nickel levels in women with endometriosis, suggesting a potential role of nickel as a metalloestrogen in its pathogenesis.

What Was Studied?

This study investigated the association between whole blood levels of nickel, cadmium, and lead in women with and without endometriosis. Specifically, it aimed to determine whether these heavy metals, known to have estrogenic properties, could be linked to the pathogenesis of endometriosis. The research involved analyzing the whole blood levels of these metals in 50 women with endometriosis and 50 age-matched controls who were confirmed to be free of the condition via laparoscopy or laparotomy.

Who Was Studied?

The study focused on a group of Sri Lankan women of reproductive age who underwent laparotomy or laparoscopy. The participants were divided into two groups: cases (women diagnosed with endometriosis, n=50) and controls (women without endometriosis, n=50). None of the participants were current smokers, and the groups were matched for age and body mass index.

Most Important Findings

The study revealed significantly elevated levels of nickel in the whole blood of women with endometriosis compared to controls (2.6 μg/L vs. 0.8 μg/L, p=0.016). This finding aligns with previous evidence that nickel, a potent metalloestrogen, can activate estrogen receptors and may contribute to the persistence of ectopic endometrial tissue. In contrast, the blood levels of cadmium and lead did not show statistically significant differences between the two groups. The presence of nickel in ectopic endometrial tissue, previously demonstrated by the researchers, supports the hypothesis that hematogenous routes could transport nickel to ectopic sites. Despite these findings, the study's small sample size limits the ability to conclude definitively that nickel is an etiological factor for endometriosis.

Greatest Implications

The discovery of higher nickel levels in women with endometriosis introduces a novel avenue for understanding the role of environmental pollutants, particularly metalloestrogens, in the condition's pathogenesis. It emphasizes the need for larger-scale studies to explore nickel's potential as a biomarker or contributor to endometriosis. Furthermore, this research underscores the importance of addressing environmental and occupational exposures to nickel, especially for women of reproductive age, as part of preventive strategies for endometriosis.

Gallium Antimicrobial Agents: Innovations in Combating Drug-Resistant Pathogens

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

This review highlights gallium's role as an iron mimic disrupting bacterial iron metabolism, effective against multidrug-resistant pathogens. Innovations in gallium delivery, including nanomaterials and synergistic therapies with antibiotics, address bioavailability challenges and enhance its antimicrobial potency.

What was reviewed?

The study reviewed the advancements in the application of gallium and gallium-based compounds as antimicrobial agents. The review aimed to address the challenges of antimicrobial resistance (AMR) by highlighting gallium's unique properties, mechanisms of action, and its potential as a non-antibiotic antibacterial strategy. The review summarized optimization strategies for gallium compounds, such as improving bioavailability and achieving sustained release, alongside synergistic effects with other antimicrobial agents.

Who was reviewed?

The review focused on multidrug-resistant (MDR) pathogens, including Pseudomonas aeruginosa, Mycobacterium tuberculosis, Acinetobacter baumannii, and methicillin-resistant Staphylococcus aureus (MRSA). It also discussed broader applications against Gram-positive and Gram-negative bacteria.

What were the most important findings?

Gallium mimics iron, a vital nutrient for bacterial growth, disrupting iron metabolism by substituting in iron-dependent processes. This "Trojan horse" mechanism inhibits bacterial proliferation by inactivating critical enzymes like ribonucleotide reductase and preventing biofilm formation, a significant defense strategy for bacteria. Key findings include:

Sustained-release systems, such as gallium-doped bioglasses and alloys, provide long-term antibacterial effects suitable for implant-related infections.

Gallium is redox-inert and disrupts bacterial metabolism by replacing iron, thereby impairing DNA synthesis, electron transport, and oxidative stress responses.

Gallium-based compounds have enhanced their solubility and antibacterial efficacy through chelation and nanomaterial delivery systems.

Synergistic strategies, combining gallium with antibiotics or antimicrobial agents, restore the efficacy of resistant antibiotics, reduce required dosages, and mitigate resistance development.

Gallium acts as a redox-inert iron mimic, disrupting bacterial iron-dependent metabolic pathways and enzyme functions, such as ribonucleotide reductase, critical for DNA synthesis. This mechanism renders it effective against MDR pathogens while minimizing bacterial resistance development. Key findings include:

Bioavailability Challenges: Gallium compounds face hydrolysis in physiological conditions, limiting their effectiveness. Strategies like coordination with ligands, incorporation into nanomaterials, and use of gallium-doped bioglasses have significantly enhanced bioavailability and sustained release.

Synergistic Antibacterial Effects: Gallium combined with antibiotics (e.g., ciprofloxacin, vancomycin) restores antibiotic efficacy against resistant strains and enhances their potency. Additionally, gallium complexes combined with metal ions or photodynamic therapies show amplified antibacterial effects.

Biofilm Disruption: Gallium can inhibit biofilm formation, a major resistance mechanism, particularly in Pseudomonas aeruginosa, by reducing iron availability needed for biofilm maintenance.

Nanotechnology Advances: Gallium-loaded nanomaterials, such as liposomes and Janus micromotors, improve targeting and sustained release, offering a promising avenue for clinical applications.

What are the greatest implications of this review?

Gallium represents a promising alternative to traditional antibiotics, especially against MDR pathogens. The review highlights potential clinical applications in treating implant-related infections, respiratory conditions, and other systemic infections. However, limitations like low bioavailability and the need for targeted delivery require further research. Nonetheless, its role as a complement to existing antibiotics could significantly delay resistance development and enhance antimicrobial strategies.

GS2 Gallium Complex: A Novel Inhibitor of MMP-14 for Anti-Metastatic Cancer Therapy

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Research on GS2, a new gallium complex, showed potent inhibition of cell invasion and MMP activity in cancer cells, particularly MMP-14. These findings indicate GS2's strong potential as a therapeutic agent against metastatic cancers by interfering with key processes of cancer cell invasion and matrix degradation.

What Was Studied?

This study examined the effects of [N-(5-chloro-2-hydroxyphenyl)-L-aspartato] chlorogallate (GS2), a water-soluble gallium complex, on tumor cell invasion and the activity and expression of matrix metalloproteinases (MMPs). Specifically, it evaluated GS2's anti-invasive properties and its regulatory effects on MMP-2, MMP-9, and MMP-14 in two human cancer cell lines: metastatic HT-1080 fibrosarcoma and MDA-MB231 breast carcinoma cells.

Who Was Studied?

The research utilized human cell lines HT-1080 (fibrosarcoma) and MDA-MB231 (breast carcinoma). Additionally, MCF7 cells transfected with MMP-14 and non-transfected fibroblast cells (F40) were used for supplemental experiments.

Most Important Findings

GS2 demonstrated significant anti-invasive and anti-MMP activities at non-cytotoxic concentrations. The compound inhibited MMP-2, MMP-9, and MMP-14 activities in a dose-dependent manner, with IC50 values of 168 µM, 82 µM, and 20 µM, respectively. GS2 reduced the mRNA expression of MMP-14 in both cell lines and inhibited MMP-2 and MMP-9 expression exclusively in MDA-MB231 cells. Western blotting confirmed decreased MMP-14 protein expression in response to GS2. Importantly, GS2 significantly inhibited cell invasion through a type-I collagen-coated matrix, correlating with the downregulation of MMP-14, a critical regulator of the extracellular matrix and tumor invasion. Notably, GS2's inhibition of MMP-14 showed specificity for cells expressing higher MMP-14 levels, a hallmark of invasive cancer phenotypes.

Greatest Implications

The findings suggest GS2 is a promising candidate for anti-metastatic therapy targeting MMP-14. This is particularly relevant for cancers characterized by elevated MMP-14 expression, such as type II endometrial adenocarcinoma and invasive pituitary adenomas. GS2’s ability to selectively inhibit MMP-14 and reduce cancer cell invasion positions it as a potential therapeutic for limiting tumor metastasis. Moreover, its low cytotoxicity at effective concentrations highlights its clinical applicability.

Heavy Metals and Pesticides Toxicity in Agricultural Soil and Plants: Ecological Risks and Human Health Implications

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This review explores the toxicity of heavy metals and pesticides in agricultural ecosystems, detailing their ecological and health impacts. It highlights pathways, bioaccumulation, and synergistic interactions, emphasizing the need for improved agricultural practices and bioremediation to mitigate these toxicants' harmful effects on soil, plants, and human health.

What Was Reviewed?

The paper is a comprehensive review focusing on the toxic effects of heavy metals (cadmium, lead, copper, and zinc) and pesticides on agricultural soil, plants, and human health. It provides an integrated synthesis of the pathways of these toxicants, from their sources to their accumulation in the environment and their implications for ecosystems and humans. The review also highlights the synergistic and antagonistic interactions between these toxicants and their combined effects on soil, plants, and humans.

Who Was Reviewed?

The review synthesizes previous studies involving agricultural soils, plants, and human populations exposed to heavy metals and pesticides. It encompasses research on various sources of these toxicants, including natural (e.g., geological) and anthropogenic sources (e.g., agricultural practices and industrial emissions), along with their bioaccumulation in ecosystems and transmission through food chains.

Most Important Findings

The review identifies key pathways through which heavy metals and pesticides impact agricultural ecosystems and human health. Heavy metals, such as cadmium and lead, were found to disrupt soil properties (e.g., pH, microbial diversity) and plant physiology (e.g., nutrient uptake, photosynthesis). Similarly, pesticides alter soil enzymatic activity, microbial communities, and plant metabolic processes. Both toxicants contribute to human health risks through bioaccumulation and persistence in food chains, leading to diseases like cancer, neurological disorders, and metabolic syndromes.

The synergistic and antagonistic interactions between heavy metals and pesticides exacerbate these effects, creating complex toxicity profiles that are challenging to predict. For example, combined exposure to cadmium and certain pesticides magnifies soil microbial damage and plant stress responses.

Greatest Implications

The review underscores the urgent need for multidisciplinary strategies to mitigate the combined toxicity of heavy metals and pesticides. It calls for improved agricultural practices, stringent regulations on pesticide use, and enhanced research on bioremediation techniques. Furthermore, understanding these toxicants' interactions is crucial for developing effective interventions to protect ecosystems and human health.

Higher incidence of zinc and nickel hypersensitivity in patients with irritable bowel syndrome

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

The study highlights a potential immune-mediated link between hypersensitivity to dental metals, particularly zinc and nickel, and the pathogenesis of IBS in a subset of patients. The findings suggest that metal hypersensitivity may contribute to the mucosal inflammation observed in IBS patients.

What was studied?

This study investigated the incidence of hypersensitivity to dental metals—specifically zinc, nickel, gold, and palladium—in patients with Irritable Bowel Syndrome (IBS). The study aimed to assess whether hypersensitivity to these metals, which are commonly used in dental prostheses, could be linked to immune activation in IBS patients. The focus was on understanding how metal-induced hypersensitivity may contribute to the low-grade mucosal inflammation observed in a subset of IBS patients.

Who was studied?

The study recruited 147 Japanese patients diagnosed with IBS according to the Rome IV diagnostic criteria and 22 healthy controls (HC). The IBS patients were classified into four subtypes:

IBS-D (diarrhea-predominant IBS): 59 patients (40.1%)
IBS-C (constipation-predominant IBS): 9 patients (6.1%)
IBS-M (IBS with mixed bowel habits): 66 patients (44.9%)
IBS-U (unspecified IBS): 13 patients (8.8%)

The subjects underwent the drug-induced lymphocyte stimulation test (DLST) to detect hypersensitivity to the metals examined quantitatively.

What were the most important findings?

Increased hypersensitivity in IBS patients: 56.5% of IBS patients demonstrated hypersensitivity to at least one metal species, compared to only 31.8% of healthy controls.

Higher sensitivity to zinc and nickel: A significant portion of IBS patients was hypersensitive to zinc (36.4%) and nickel (39.6%), whereas none of the healthy controls exhibited hypersensitivity to zinc, and only 21.1% showed sensitivity to nickel.

Severe sensitivity: IBS patients had a significantly higher stimulation index (SI) for both zinc and nickel compared to healthy controls.

No significant difference across IBS subtypes: There was no significant difference in metal hypersensitivity rates or SI values across different IBS subtypes (IBS-D, IBS-C, IBS-M, IBS-U).

What are the greatest implications of this study?

The study highlights a potential immune-mediated link between hypersensitivity to dental metals, particularly zinc and nickel, and the pathogenesis of IBS in a subset of patients. The findings suggest that metal hypersensitivity may contribute to the mucosal inflammation observed in IBS patients. This pilot study underscores the importance of considering hypersensitivity reactions as part of IBS management, potentially leading to personalized treatment approaches, such as dietary modifications like a low-nickel diet, or avoiding exposure to specific metals in dental materials.

 
 
 
 

Metalloestrogens exposure and risk of gestational diabetes mellitus: Evidence emerging from the systematic review and meta-analysis

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This systematic review and meta-analysis identified arsenic, antimony, and copper as metalloestrogens associated with an increased risk of gestational diabetes mellitus (GDM). The findings highlight the significance of environmental metal exposure in pregnancy, underscoring the need for public health policies to mitigate these risks and further research into the mechanisms linking metalloestrogens to GDM.

What Was Reviewed?

The systematic review and meta-analysis examined the relationship between exposure to metalloestrogens—specifically arsenic (As), antimony (Sb), chromium (Cr), cadmium (Cd), copper (Cu), selenium (Se), and mercury (Hg)—and the risk of developing gestational diabetes mellitus (GDM). This review aimed to aggregate findings from observational studies to better understand how these metals, which have estrogenic properties, impact the development of GDM. A comprehensive search of PubMed, Web of Science, and Embase databases was conducted to collect relevant studies up until December 2023.  

Who Was Reviewed?

The meta-analysis included a total of 33 observational studies, which involved 141,175 subjects, comprising 9,450 cases of GDM and 131,725 controls. The review covered a wide geographic distribution, with studies conducted in multiple countries and varying methodologies for exposure assessment. Biological specimens such as blood, urine, and other tissues were analyzed to measure levels of metalloestrogens in pregnant women and correlate them with the occurrence of GDM.  

What Were the Most Important Findings of This Review?

The meta-analysis revealed that exposure to certain metalloestrogens, specifically arsenic (As), antimony (Sb), and copper (Cu), is associated with an increased risk of GDM. Arsenic exposure exhibited a risk ratio (OR = 1.28, 95% CI [1.08, 1.52]), antimony a higher risk (OR = 1.73, 95% CI [1.13, 2.65]), and copper was also linked with a modest increase in GDM risk (OR = 1.29, 95% CI [1.02, 1.63]). However, high heterogeneity in these studies was noted, with substantial variability in the results for arsenic (I2 = 64.1%), antimony (I2 = 80.9%), and copper (I2 = 71.6%). Other metalloestrogens, such as selenium, cadmium, chromium, and mercury, did not show a statistically significant association with GDM in this analysis.  

What Are the Greatest Implications of This Review?

The greatest implication of this review is the emerging recognition that environmental exposure to metalloestrogens could be a significant contributor to the risk of GDM. This finding underscores the importance of considering environmental and occupational exposure to harmful metals in pregnancy, which could lead to metabolic disturbances that increase the likelihood of GDM. The results suggest that public health policies should place greater emphasis on reducing environmental contamination by metalloestrogens, especially in populations at higher risk of GDM. Additionally, it points to the need for more targeted research on how different metals disrupt estrogenic pathways and influence insulin signaling and pancreatic function, which may open new avenues for preventive strategies, early diagnosis, and novel treatments for GDM. The high heterogeneity in the studies also highlights the complexity of the relationship between metalloestrogens and GDM. Variations in exposure levels, methods of measurement, and geographical factors call for standardized approaches in future research to better clarify these associations. Furthermore, the study suggests the necessity for increased awareness and education among healthcare providers regarding environmental factors in pregnancy, encouraging efforts to mitigate exposure in vulnerable populations.

Metalloestrogens: an emerging class of inorganic xenoestrogens with potential to add to the oestrogenic burden of the human breast

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

This review opens new avenues in toxicology and endocrine research, identifying metalloestrogens as a critical factor in hormone disruption and breast cancer risk. Further studies are necessary to confirm these findings and develop effective mitigation strategies for human health protection.

What was reviewed?

This study, published in the Journal of Applied Toxicology, reviewed the concept and emerging evidence of metalloestrogens mimicking estrogenic activity. The review focused on how these metals interact with estrogen receptors (ERs) like organic xenoestrogens, potentially contributing to estrogenic activity in human breast tissue and increasing the risk of hormone-related cancers such as breast cancer. The review primarily covered in vitro and in vivo studies of various metal ions, including aluminum, antimony, arsenite, barium, cadmium, chromium (Cr(II)), cobalt, copper, lead, mercury, nickel, selenite, tin, and vanadate. The review also highlights significant research contributions from multiple studies and scholars focusing on the effects of metalloestrogens on human breast cancer cell lines, such as MCF-7 and T47D, as well as their impact on gene expression and cellular proliferation.

Most Important Findings:

Estrogenic Activity of Metals: The review found that various metal ions can act as estrogen agonists by binding to estrogen receptors, particularly ERα, and mimicking the actions of physiological estrogens. This was demonstrated in studies showing that metals such as cadmium, nickel, and aluminum could displace estradiol from the ligand-binding domain of ERα, leading to altered gene expression and increased cell proliferation in breast cancer cells.

Molecular Mechanisms: Metals such as cadmium were shown to bind directly to the ligand-binding domain (LBD) of the estrogen receptor, interfering with the receptor's normal function. This binding alters the receptor’s ability to interact with estrogen response elements (EREs) on DNA, thereby affecting the transcription of estrogen-regulated genes. For instance, cadmium was found to downregulate ER levels and upregulate estrogen-regulated gene expression, driving cell proliferation.

Cooperative Action with Estrogens: The metals did not antagonize estradiol’s action; instead, they often enhanced the agonist actions of estradiol. In some cases, metals like copper and cobalt increased breast cancer cell proliferation when combined with estradiol, indicating a synergistic effect that may exacerbate estrogenic signaling in hormone-dependent cancers.

In Vivo Evidence: The review highlighted evidence of in vivo estrogenic activity in animal models, particularly for cadmium, which was shown to increase uterine weight, induce mammary gland development, and alter gene expression. The estrogenic effects of cadmium were noted at doses relevant to human exposure, raising significant concerns about environmental exposure to these metals.

Environmental and Occupational Exposure: The presence of metalloestrogens such as cadmium and aluminum in everyday consumer products (e.g., antiperspirants) and the environment (e.g., tobacco smoke, and industrial pollutants) implies widespread human exposure. These metals can accumulate in the body, especially in breast tissue, and may contribute to the burden of aberrant estrogen signaling involved in breast cancer development.

Greatest Implications:

Breast Cancer Risk: The review underscores the potential for metalloestrogens to increase the risk of breast cancer by contributing to estrogenic signaling within breast tissue. Given that breast cancer is often driven by estrogen receptor activation, the cumulative burden of environmental estrogens and metalloestrogens could enhance the likelihood of cancer development and progression.

Environmental Health and Toxicology: The widespread presence of these metals in the environment, their ability to accumulate in the body, and their newly recognized estrogenic activity suggest a need for revised regulatory guidelines and risk assessments for human exposure to metalloestrogens. This includes re-evaluating safe exposure levels, especially for metals like cadmium, which is already classified as a human carcinogen.

Endocrine Disruption: The concept of metalloestrogens extends the traditional understanding of endocrine-disrupting chemicals (EDCs) beyond organic compounds, emphasizing the need for further investigation into how inorganic metals may impact hormone-related diseases. This review calls for more research on the long-term effects of chronic exposure to metalloestrogens in both wildlife and humans.

Public Health Awareness: There is a strong implication for public health education regarding the sources of metalloestrogen exposure, such as antiperspirants, diet, cigarette smoke, and industrial pollutants. Raising awareness could lead to better personal care practices and lifestyle choices to reduce individual exposure to these potentially harmful metal ions.

Metals at the Host–Fungal Pathogen Battleground

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

Fungal pathogens rely on metal acquisition for virulence. This review reveals how host tissues manipulate iron, copper, zinc, and manganese to starve or intoxicate fungi and how pathogens adapt, with implications for antifungal therapy and microbiome-targeted interventions.

What was reviewed?

This review, titled “Metals at the Host–Fungal Pathogen Battleground” by Garg et al. (2024), provides a comprehensive analysis of how fungal pathogens and their mammalian hosts engage in a dynamic, metal-centric battle during infection. The paper focuses on four key transition metals—iron (Fe), copper (Cu), zinc (Zn), and manganese (Mn)—and discusses their dual roles as essential micronutrients and toxic agents. The review examines how the host immune system manipulates metal availability to control fungal infections through nutritional immunity and metal toxicity, and how fungi adapt through sophisticated metal uptake, detoxification, and cofactor-utilization systems.

Who was reviewed?

The review explores fungal pathogens of major clinical concern, primarily Candida albicans, Cryptococcus neoformans, Aspergillus fumigatus, Histoplasma capsulatum, and other pathogenic molds and yeasts. It includes insights drawn from model organisms such as Saccharomyces cerevisiae while highlighting evolutionary expansions in metal acquisition strategies among fungal pathogens. These fungi were examined in the context of their interaction with host immune responses and fluctuating tissue-specific metal availabilities during infection.

What were the most important findings?

One of the central insights is the host's use of “nutritional immunity” to manipulate metal availability—either sequestering metals to starve the fungus or flooding them with toxic concentrations. Calprotectin, a neutrophil-derived metal-binding protein, plays a critical role by sequestering Zn and Mn at infection sites. During C. albicans infection, Zn and Cu levels in kidney tissue follow a biphasic pattern: an initial deprivation phase followed by toxic overload, compelling the fungus to recalibrate metal acquisition and detoxification systems. The review details distinct fungal responses to each metal: siderophore-mediated Fe acquisition, Cu detoxification through P-type ATPases and metallothioneins, Zn scavenging via zincophores like PRA1, and Mn uptake through NRAMP transporters (e.g., SMF12, SMF13). These responses are tightly regulated by metal-specific transcription factors—SEF1 and HAP for Fe, MAC1 and ACE for Cu, ZAP1 for Zn—but notably, no Mn-specific regulon has been identified, suggesting post-transcriptional regulation may dominate Mn homeostasis.

From a microbiome perspective, these findings highlight that major fungal pathogens exhibit clear dependencies on metal-acquisition genes and metalloregulation systems, suggesting that transitions in microbial metal use may be a distinguishing trait in dysbiotic or pathogenic microbiomes. The expression of zincophores and siderophore transporters, as well as evidence of calprotectin-induced nutrient metal depletion, could serve as microbial signature markers (MMAs) for fungal overgrowth and metal imbalance in tissue-specific microbiomes. Additionally, the review underscores that host micronutrient modulation—especially Fe and Mn dynamics—has significant implications for microbiome resilience and therapeutic outcomes, especially in patients receiving iron supplements or experiencing metal dysregulation from comorbidities.

What are the greatest implications of this review?

This review fundamentally shifts the framing of antifungal strategies by revealing that metals are both nutrient targets and weapons in host-pathogen interactions. For clinicians, this offers novel therapeutic opportunities: metal chelation, metallophore inhibition, and interference with fungal metal transporters could be developed into antifungal interventions. Importantly, these approaches must be designed to minimize collateral damage to commensal microbial taxa, many of which share metal acquisition strategies. The review also emphasizes that rising antifungal resistance demands novel targets, and metal-based pathways offer promising, underexploited avenues for drug development. For microbiome researchers, this paper underscores the need to integrate metallomic profiling into microbiome signatures, especially for fungal overgrowth conditions like candidiasis, aspergillosis, and histoplasmosis. Incorporating fungal metallophore gene presence, metal uptake transporters, and responses to host nutritional immunity into microbiome databases will enhance precision in diagnostics and intervention selection.

Nickel Allergic Contact Mucositis in Suspected Non-Responsive Celiac Disease

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

The study points to nickel sensitivity as a novel contributor to persistent symptoms in Celiac Disease (CD) patients and supports the use of a low-nickel diet as an effective intervention, potentially altering future approaches to treating the disease.

What was studied?

This study examined the prevalence of nickel (Ni) allergic contact mucositis (ACM) in celiac disease (CD) patients who were in serological and histological remission from their condition but continued to experience relapsing gastrointestinal and extraintestinal symptoms despite following a strict gluten-free diet (GFD). Additionally, the study investigated the therapeutic effects of a low-nickel diet on these persistent symptoms in the CD patients. The study hypothesized that Ni-rich foods, which are commonly consumed in a GFD, might lead to intestinal sensitization to Ni and induce symptoms similar to irritable bowel syndrome (IBS).

Who was studied?

The study recruited 102 consecutive CD patients (74 female, 28 male, age range 18–65 years) who had been on a GFD for at least 12 months and were in serological and histological remission (Marsh–Oberhuber type 0–I). These patients continued to report relapsing gastrointestinal and/or extraintestinal symptoms despite adherence to the GFD. After applying exclusion criteria (e.g., lactose intolerance, Helicobacter pylori infection, inflammatory bowel diseases), 20 female patients completed the study. All these patients underwent nickel oral mucosa patch testing (omPT) and were subsequently placed on a low-nickel diet for three months.

What were the most important findings?

Prevalence of Ni ACM in CD patients: The study found that 100% of the CD patients who completed the study tested positive for Ni ACM using the omPT, suggesting a high prevalence of nickel sensitivity in CD patients experiencing recurrent symptoms despite a GFD.

Symptom trends and the impact of a low-nickel diet:

Following an initial gluten-free diet (T0 vs. T1), 79.2% of the total symptoms improved, with 58.3% showing statistically significant improvement. However, after prolonged adherence to a GFD (T1 vs. T2), 83.3% of symptoms worsened, with 41.7% of symptoms showing statistically significant exacerbation. This relapse of symptoms was attributed to increased dietary intake of Ni-rich foods commonly consumed in a GFD (e.g., corn). After three months of a low-nickel diet (T2 vs. T3), 83.4% of symptoms improved, with 41.7% showing statistically significant improvement. Both gastrointestinal and extraintestinal symptoms showed substantial relief, including key symptoms such as abdominal pain, bloating, fatigue, and dermatitis.

What are the greatest implications of this study?

Nickel sensitivity as a cause of persistent symptoms in celiac patients: This study suggests that nickel sensitivity (Ni ACM) may be a significant underlying cause of persistent gastrointestinal and extraintestinal symptoms in CD patients who are in remission but continue to suffer despite adherence to a GFD. The findings imply that the ingestion of Ni-rich foods commonly included in a GFD might trigger or exacerbate these symptoms in Ni-sensitive individuals.

Clinical management of celiac disease: The results highlight the potential clinical importance of considering nickel sensitivity in CD patients with unresolved symptoms. Nickel oral mucosa patch testing (omPT) should be considered as part of the diagnostic workup for such patients, and a low-nickel diet (LNiD) could be an effective therapeutic intervention to alleviate these symptoms. This could lead to an improved quality of life for CD patients and more targeted dietary interventions.

Potential changes in dietary guidelines: If larger studies confirm these findings, dietary guidelines for celiac patients may need to be expanded to include advice on limiting Ni-rich foods, particularly for patients who are unresponsive to a GFD alone. This could represent a significant shift in how refractory or non-responsive celiac disease is treated.

Overlap with other dietary interventions: The study also draws attention to the overlap between nickel-rich foods and foods commonly eliminated in low-FODMAP diets, which are often prescribed for IBS-like symptoms. This suggests that some benefits attributed to low-FODMAP diets may, in fact, be due to the concurrent reduction of nickel intake, further emphasizing the need for specific diagnostic testing in dietary management.

Nickel Allergy as a Risk Factor for Endometriosis

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study identifies nickel allergy as an independent risk factor for endometriosis, highlighting shared immune dysregulation and estrogenic pathways. Using a population-based cohort, researchers found a 2.5-fold increased odds of nickel allergy in women with endometriosis, emphasizing the role of environmental exposures in its pathogenesis.

What Was Studied?

This study examined the association between nickel allergy and endometriosis using a population-based nested case-control design. The research aimed to determine whether nickel allergy is an independent risk factor for endometriosis by analyzing a cohort dataset provided by the South Korean National Health Insurance Service (NHIS), which included approximately 1 million individuals. The study was designed to assess causality and improve upon previous correlational studies.

Who Was Studied?

The study included 4,985 women divided into two groups: 997 women with endometriosis and 3,988 controls matched by age and socioeconomic status. The endometriosis group was identified using diagnostic codes, surgery records, and drug prescriptions between 2009 and 2013, while nickel allergy cases were identified between 2002 and 2008 using specific diagnostic and patch test codes.

What Were the Most Important Findings?

The findings revealed that women with endometriosis had a significantly higher prevalence of nickel allergy (0.8%) compared to the control group (0.3%), with an adjusted odds ratio of 2.474 (95% CI: 1.023–5.988; p = 0.044). The study highlights the estrogenic and immune-modulating properties of nickel, suggesting that elevated blood nickel levels associated with nickel allergy may contribute to endometriosis pathogenesis. Both conditions share immunological mechanisms, including cell-mediated hypersensitivity and immune dysregulation. The study also emphasizes the activity of nickel as a metalloestrogen, which may influence endometriosis through its interaction with estrogen receptors. Despite its low clinical prevalence, the findings support nickel allergy as a potential contributing factor to endometriosis, particularly in the context of autoimmune and estrogenic pathways.

What Are the Greatest Implications of This Study?

This study underscores the link between environmental factors, such as nickel exposure, and the development of endometriosis. Identifying nickel allergy as a risk factor paves the way for further research into environmental triggers and immune-mediated mechanisms in endometriosis. These findings could lead to targeted prevention strategies, such as reducing nickel exposure in at-risk populations, and inform therapeutic approaches that address immune and estrogenic pathways in endometriosis management.

Nickel Allergy is Found in a Majority of Women with Chronic Fatigue Syndrome and Muscle Pain—and may be Triggered by Cigarette Smoke and Dietary Nickel Intake

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

Nickel allergy, smoking, and dietary nickel intake may worsen chronic fatigue and muscle pain. Managing exposure can improve symptoms.

What was studied?

This study investigated the relationship between nickel allergy, cigarette smoking, and dietary nickel intake in women diagnosed with chronic fatigue syndrome (CFS) and muscle pain. The primary aim was to evaluate the prevalence of nickel allergy in this population and explore how smoking and dietary nickel may trigger or exacerbate symptoms.

Who was studied?

The study involved 204 women aged 21 to 73 years with chronic fatigue and muscle pain, meeting the criteria for fibromyalgia and chronic fatigue syndrome but with no signs of autoimmune disorders. The participants underwent immune stimulation therapy using a Staphylococcus vaccine for six months, and their nickel allergy history, smoking habits, and treatment responses were analyzed.

What were the most important findings?

The study found that 52% of the women had a history suggestive of nickel allergy, and 28% were habitual smokers. Nickel allergy and smoking significantly influenced treatment outcomes, with non-allergic, non-smoking participants showing the highest treatment success rates (39%), compared to only 6% in allergic smokers. Additionally, two case reports highlighted the impact of dietary and smoking changes: one participant improved after quitting smoking and reducing dietary nickel intake, while another experienced sustained symptom relief by following a low-nickel diet. Notably, nickel hypersensitivity was associated with increased fatigue and muscle pain symptoms triggered by dietary nickel or cigarette smoke, both of which contain trace amounts of the metal. The findings indicate that systemic nickel allergy, potentially exacerbated by smoking or high dietary nickel, may contribute to chronic fatigue and muscle pain.

What are the greatest implications of this study?

This research emphasizes the importance of recognizing nickel allergy as a potential factor in chronic fatigue syndrome and muscle pain. The interplay between nickel hypersensitivity, dietary nickel intake, and smoking could have significant clinical implications. Managing nickel exposure through dietary adjustments such as a low-nickel diet and smoking cessation may serve as a non-invasive strategy to alleviate symptoms in affected patients. Furthermore, the study underscores the need for broader awareness and diagnostic consideration of systemic nickel allergy in chronic fatigue-related conditions, particularly in women.

Nickel chelation therapy as an approach to combat multi-drug resistant enteric pathogens

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study evaluates nickel chelation therapy using DMG against multidrug-resistant Salmonella and Klebsiella. DMG impaired virulence by inhibiting Ni-dependent enzymes, reduced bacterial load in organs, and improved survival in animal models, offering a promising metallomic intervention.

What was studied?

This experimental study investigated the therapeutic potential of the nickel-specific chelator dimethylglyoxime (DMG) as an antimicrobial intervention against multidrug-resistant (MDR) enteric pathogens, specifically Salmonella enterica serovar Typhimurium and Klebsiella pneumoniae. The research assessed whether nickel chelation by DMG could inhibit the growth and virulence of these pathogens in vitro and in vivo through interference with essential Ni-dependent bacterial enzymes.

Who was studied?

The study utilized bacterial strains of MDR Klebsiella pneumoniae (ATCC BAA-2472) and MDR Salmonella Typhimurium (ATCC 700408 and ATCC 14028), in addition to two animal models: Mus musculus (mice) for assessing DMG efficacy and safety in systemic infection, and Galleria mellonella (wax moth larvae) for testing virulence attenuation in an invertebrate model.

What were the most important findings?

This study provides compelling evidence that dimethylglyoxime (DMG), a nickel-specific chelator, exerts a potent bacteriostatic effect against multidrug-resistant Salmonella Typhimurium and Klebsiella pneumoniae by inhibiting key Ni-dependent enzymes—hydrogenase and urease, respectively. At concentrations between 1 and 5 mM, DMG impaired enzyme activity without exhibiting toxicity in murine or invertebrate models. In vivo, DMG administration resulted in 50% survival among infected mice, compared to complete lethality in untreated controls, and led to a 10-fold reduction in bacterial colonization of the liver and spleen. In Galleria mellonella, pre-injection with DMG improved survival by 40–60% after challenge with lethal doses of MDR pathogens. NMR analysis confirmed DMG’s systemic absorption by detecting it in liver tissue. The absence of adverse effects in either model underscores the compound's therapeutic safety. The targeted suppression of nickel-requiring pathogens supports the utility of metallome-directed interventions in combating MDR infections, particularly for pathogens that rely on nickel-dependent enzymes for virulence.

FindingDetails
Bacteriostatic ActivityDMG showed concentration-dependent inhibition (1–5 mM) of MDR S. Typhimurium and K. pneumoniae.
Target EnzymesInhibited hydrogenase activity in Salmonella and urease activity in Klebsiella, both Ni-dependent enzymes.
Animal Survival50% survival in DMG-treated mice vs. 0% in untreated controls.
Organ Burden Reduction10-fold reduction in bacterial colonization in livers and spleens of treated mice.
Larvae Protection40–60% survival in DMG-treated G. mellonella larvae following lethal bacterial challenge.
Systemic BioavailabilityNMR confirmed presence of DMG in liver tissue post-oral administration.
Safety ProfileNo observed toxicity in mice or larvae at therapeutic doses.
Mechanistic RelevanceAligns with a metallomic intervention strategy by targeting Ni-dependent MMAs (Salmonella, Klebsiella).

What are the greatest implications of this study?

This study underscores the translational potential of metallome-targeted interventions, specifically through nickel chelation, as a viable therapeutic approach against MDR pathogens. By inhibiting bacterial nickel-dependent enzymatic machinery critical for virulence and survival, such as hydrogenases and ureases, DMG offers a mechanism-based strategy that bypasses conventional antibiotic resistance pathways. The fact that many high-priority MDR pathogens identified by the WHO possess Ni-dependent enzymes positions nickel chelation as a broadly applicable antimicrobial modality. Moreover, the non-toxic profile of DMG in two distinct animal models supports its development for clinical use, particularly as an adjunct or alternative to antibiotics in cases of resistant infections.

Nickel Sensitivity and Symptom Management in Endometriosis: The Role of a Low-Nickel Diet

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Low‑Nickel Diet (LNiD)
    Low‑Nickel Diet (LNiD)

    A low-nickel diet (LNiD) is a therapeutic dietary intervention that eliminates high-nickel foods, primarily plant-based sources such as legumes, nuts, whole grains, and cocoa, to reduce systemic nickel exposure. It is clinically validated for managing systemic nickel allergy syndrome (SNAS) and nickel-induced eczema. Its relevance is well-established in microbiome modulation, with studies demonstrating clinical benefits in conditions such as endometriosis, fibromyalgia, irritable bowel syndrome, and GERD.

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

This study offers new insights into the potential link between nickel sensitivity and symptom severity in endometriosis, suggesting that a low-nickel diet may be a promising intervention for alleviating associated gastrointestinal and gynecological symptoms.

What Was Studied?

This pilot study investigated the prevalence of nickel (Ni) allergic contact mucositis (ACM) in women with endometriosis who experience gastrointestinal symptoms and evaluated the effects of a low-nickel diet on these symptoms. The study focused on assessing the gastrointestinal, extra-intestinal, and gynecological symptom reductions associated with Ni ACM and dietary interventions.

Who Was Studied?

The study enrolled 84 women of reproductive age diagnosed with endometriosis who reported significant gastrointestinal symptoms. Thirty-one participants completed the study, undergoing a diagnostic nickel oral mucosa patch test (omPT) and a subsequent three-month low-nickel diet intervention. Participants were evaluated using symptom questionnaires both at baseline and after dietary changes.

What Were the Most Important Findings?

The study found that 90.3% of participants tested positive for Ni ACM, suggesting a high prevalence of nickel sensitivity among women with endometriosis. Following three months of adhering to a low-nickel diet, significant reductions in all evaluated symptoms were reported. Gastrointestinal symptoms such as abdominal pain, bloating, and diarrhea showed marked improvement. Extra-intestinal symptoms, including fatigue and headaches, and gynecological symptoms such as pelvic pain and dysmenorrhea, also exhibited statistically significant decreases. These findings indicate that nickel sensitivity may contribute to the symptomatic burden of endometriosis, and dietary interventions targeting nickel can alleviate these issues.

The study suggests a potential mechanistic link between nickel exposure, immune responses, and the exacerbation of endometriosis symptoms. Major microbial associations (MMAs) relevant to this context include those influenced by dietary changes, although specific microbiome alterations were not detailed.

What Are the Greatest Implications of This Study?

This research highlights nickel sensitivity as a significant yet previously under-recognized contributor to gastrointestinal and systemic symptoms in endometriosis patients. The findings suggest that incorporating nickel sensitivity screening and low-nickel dietary recommendations could represent a transformative approach to symptom management in endometriosis. Although the sample size was small, the results offer strong preliminary evidence for revising dietary protocols in clinical practice to include low-nickel guidelines, potentially improving the quality of life for patients.

Nickel Sensitivity in Patients With Irritable Bowel Syndrome

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study suggests that nickel sensitivity may play a significant role in the etiology of IBS, warranting further exploration and potentially influencing future diagnostic and therapeutic strategies for IBS management.

What was studied?

This study investigated the relationship between nickel (Ni) sensitivity and irritable bowel syndrome (IBS). Specifically, the researchers aimed to determine whether patients with IBS are more prone to nickel sensitivity than healthy individuals. The study utilized patch testing, a standard method for diagnosing contact allergies, to evaluate Ni sensitivity in both IBS patients and a healthy control group.

Who was studied?

The study included 50 patients diagnosed with IBS based on the Rome IV criteria and 40 healthy volunteers who served as the control group. Both groups were similar in terms of age, gender distribution, and dietary habits. The mean ages of the patient and control groups were 42.82 and 39.77 years, respectively, with no significant difference in age or gender between the groups. Exclusion criteria were applied to ensure that systemic medication, dietary restrictions, or recent use of nickel-containing products did not confound the results.

What were the most important findings?

Nickel sensitivity prevalence in IBS: The study found that 40% of the IBS patient group exhibited nickel sensitivity, compared to 17.5% of the control group. This difference was statistically significant (p=0.03), suggesting a strong association between Ni sensitivity and IBS.

Gender distribution: While nickel sensitivity was more common in women, with 45.8% of female IBS patients testing positive compared to 34.6% of male patients, the difference in sensitivity between genders was not statistically significant. However, among men, Ni sensitivity was significantly higher in the IBS group compared to the control group (p=0.03).

Nickel sensitivity’s role in IBS: The study supports the hypothesis that nickel sensitivity might play a role in the pathogenesis of IBS, corroborating earlier studies that suggested a link between nickel intake and gastrointestinal symptoms mimicking IBS.

What are the greatest implications of this study?

Clinical relevance of nickel sensitivity in IBS: This study highlights the potential role of dietary nickel in triggering or exacerbating IBS symptoms. If nickel sensitivity is confirmed as a contributor to IBS pathogenesis, dietary modifications such as low-nickel diets may be a viable treatment approach for some IBS patients, possibly improving symptoms and reducing treatment costs.

Future research directions: The findings suggest a need for more comprehensive studies to further explore the relationship between nickel sensitivity and IBS, as well as to investigate the pathophysiological mechanisms underlying this association. Additionally, larger studies could focus on whether low-nickel diets provide significant clinical improvement in IBS patients with nickel sensitivity.

Gender considerations in treatment: While nickel sensitivity is traditionally more associated with women, this study reveals that nickel sensitivity is significantly higher in men with IBS compared to healthy controls. This finding may influence future clinical approaches, encouraging practitioners to consider nickel sensitivity in both male and female IBS patients.

Nutritional immunity: the battle for nutrient metals at the host–pathogen interface

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This review outlines how vertebrate hosts restrict or overload metals like Fe, Zn, Mn, and Cu to combat bacterial pathogens, and how bacteria adapt to survive. These dynamics shape infection outcomes and microbial ecology, offering new targets for antimicrobial therapy and microbiome-based interventions.

What was reviewed?

The review titled “Nutritional Immunity: The Battle for Nutrient Metals at the Host–Pathogen Interface” by Murdoch and Skaar (2022) provides a comprehensive update on how vertebrate hosts manipulate metal bioavailability to control bacterial infections and how bacterial pathogens counteract these strategies. The authors explore the essential roles of trace metals—iron (Fe), zinc (Zn), manganese (Mn), and copper (Cu)—in bacterial and host physiology and examine how their regulated distribution, sequestration, or overload can determine infection outcomes. The review focuses on molecular mechanisms of host-mediated metal withholding (nutritional immunity), bacterial acquisition strategies, and the evolving arms race over these metals at specific host-pathogen interfaces.

Who was reviewed

This review synthesizes findings across multiple Gram-positive and Gram-negative bacterial pathogens, including Staphylococcus aureus, Salmonella Typhimurium, Escherichia coli, Acinetobacter baumannii, Pseudomonas aeruginosa, Mycobacterium tuberculosis, and Neisseria meningitidis, among others. Host systems, including neutrophils, macrophages, epithelial cells, and various tissues like the intestinal tract and respiratory mucosa, are reviewed in terms of their specific roles in metal sequestration, intoxication, and immune modulation.

What were the most important findings?

A central conclusion of this review is that nutritional immunity involves both limitation and intoxication of nutrient metals. Hosts utilize proteins like calprotectin, lipocalin-2, transferrin, and metallothioneins to sequester Fe, Zn, Mn, and Cu, thereby starving pathogens of essential cofactors. Simultaneously, immune cells like macrophages and neutrophils deploy excess Zn and Cu to phagosomes, leveraging metal toxicity to kill internalized bacteria.

Bacterial pathogens have evolved diverse counterstrategies: siderophore production for Fe acquisition, zincophores and metallophores for Zn uptake, NRAMP and ABC transporter systems for Mn and Zn, and metallochaperones (e.g., ZigA, ZagA) to distribute metals to target enzymes under stress. Siderophores such as yersiniabactin not only scavenge Fe but also bind Zn and Cu, expanding their functional utility. Some pathogens even pirate host metal-binding proteins like calprotectin, underscoring the evolutionary sophistication of metal acquisition systems.

Pertinent to the microbiome signatures database, the review identifies multiple trace metal acquisition systems and regulators as conditionally expressed virulence determinants. For example, S. aureus relies on the Isd system for heme-Fe acquisition in calprotectin-rich environments, while E. coli expresses stealth siderophores like salmochelin to evade host lipocalin-2. The regulation of metal transporters by Zur, Fur, and MntR points to coordinated responses within microbial communities to host-induced metal stress. These systems may serve as key microbial markers (MMAs) in tissue-specific dysbiosis and inflammation, particularly in gut, respiratory, and skin-associated microbial niches.

What are the greatest implications of this review?

This review reframes trace metals not just as nutrients, but as strategic tools of host defense and bacterial offense. The dual use of metals as essential cofactors and toxic agents presents a unique therapeutic opportunity. Interventions targeting bacterial metal acquisition—such as siderophore-drug conjugates, gallium-based Fe mimetics, or vaccines targeting outer membrane metal receptors—are already in clinical development, as exemplified by the FDA-approved antibiotic cefiderocol. Importantly, these strategies must be balanced to avoid unintended disruption of commensal microbes that rely on similar metalloproteins and uptake systems.

From a microbiome perspective, this review strongly supports the inclusion of metal utilization traits in microbial signature profiling. Shifts in metal acquisition gene expression or the presence of stealth siderophores, metallophores, and specific metalloregulators could distinguish pathogenic from commensal states in host-associated microbial communities. It also emphasizes that dietary metal intake and host genetic variants in metal regulation significantly impact microbiome composition and infection risk. This opens new avenues for personalized dietary or metallomic interventions in microbiome modulation and pathogen control.

Plants that Hyperaccumulate Heavy Metals

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This chapter reviews hyperaccumulator plants, especially in the Brassicaceae family, and their mechanisms of metal uptake. It highlights implications for human health, including how high-metal diets may shift the microbiome toward dysbiosis—an insight relevant to conditions like endometriosis where metallomic imbalances and microbial disruptions intersect.

What was reviewed?

This chapter, “Plants that Hyperaccumulate Heavy Metals” by Elisa Fasani, provides a detailed review of the physiological, genetic, and ecological mechanisms underlying metal hyperaccumulation in plants, with a particular emphasis on angiosperm species. It defines hyperaccumulators as plants that uptake, translocate, and sequester heavy metals in their aerial tissues at concentrations vastly exceeding those of typical plants, and systematically reviews the elements for which hyperaccumulation occurs (e.g., nickel, zinc, cadmium, arsenic). It also discusses the evolutionary distribution of this trait, highlighting its prevalence in specific plant families, notably the Brassicaceae and Euphorbiaceae, and examines the molecular determinants of hyperaccumulation such as transporters, chelators, and stress-response proteins.

Who was reviewed?

The review encompasses approximately 500 hyperaccumulator taxa, primarily angiosperms, focusing on model species like Arabidopsis halleri and Noccaea caerulescens. These species are genetically similar to A. thaliana and have been extensively studied for their abilities to hyperaccumulate Zn, Cd, and Ni. The genus Alyssum, particularly Alyssum bertolonii and Alyssum lesbiacum, also features prominently, especially in relation to Ni accumulation. Although most hyperaccumulator species are herbaceous perennials adapted to metalliferous soils, the review also includes ferns (e.g., Pteris vittata) for arsenic hyperaccumulation and other non-Brassicaceae families that contribute to the small number of Cd and Se hyperaccumulators.

What were the most important findings?

A central finding of the review is the disproportionate representation of Brassicaceae among metal hyperaccumulator plants, with about 25% of known hyperaccumulator species belonging to this family. This family dominates zinc hyperaccumulation, and includes the only two known angiosperm arsenic hyperaccumulators. The genus Alyssum within Brassicaceae is especially rich in nickel hyperaccumulators, highlighting this family’s strong metallomic signature. Importantly, hyperaccumulation of one metal often correlates with the uptake of others—particularly Zn with Cd and Pb—due to shared ionic characteristics. The physiological mechanisms facilitating hyperaccumulation include enhanced uptake, symplastic mobility, xylem loading, and vacuolar sequestration, while detoxification involves metal ligands like histidine and nicotianamine. A significant portion of the review is dedicated to the molecular biology of hyperaccumulation, emphasizing gene families such as ZIP, CDF, NRAMP, HMA, and YSL, and the roles they play in metal homeostasis. Notably, the review supports the “elemental defense” hypothesis, wherein metal accumulation deters herbivores and pathogens. This is especially relevant for microbiome research: hyperaccumulator plants harbor metal-tolerant microbial communities, a parallel to dysbiotic profiles seen in metal-rich environments or metal-exposed tissues in humans. These plant-microbiome-metal interactions may mirror microbial shifts seen in chronic inflammatory diseases with elevated heavy metal burden, such as endometriosis.

What are the greatest implications of this review?

The implications for clinical microbiome research are substantial. The review reinforces the concept that chronic exposure to dietary heavy metals—even from otherwise health-associated vegetables like those in the Brassicaceae family—could influence host microbiome composition by selecting for metal-resistant, pro-inflammatory taxa. In the context of endometriosis, this is particularly compelling given the elevated presence of these metals in the metallomic signature of the condition. The well-documented accumulation of arsenic, cadmium, lead, nickel, and zinc in Brassicaceae suggests that excessive consumption of these vegetables—especially when sourced from contaminated soils—could contribute to microbial dysbiosis and worsen disease pathology. Furthermore, the elemental defense mechanism in plants that favors metal accumulation over chemical defenses may have unintended consequences when translated to human exposure, especially when these plants become regular components of the diet. The review thus supports reevaluating blanket dietary recommendations for high Brassicaceae intake in individuals with microbiome-mediated conditions or impaired metal detoxification.

Potential Role of Copper in Diabetes and Diabetic Kidney Disease

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This review explores copper's role in diabetes and diabetic kidney disease, emphasizing its impact on oxidative stress, glycemic control, and renal function. Findings highlight the potential of copper homeostasis as a therapeutic target and the Zn/Cu ratio as a biomarker for diabetes management and DKD progression.

What Was Reviewed?

The paper reviewed the role of copper in diabetes and its complications, with a specific focus on diabetic kidney disease (DKD). Copper, as an essential trace element, is pivotal in various biological processes, but its imbalance—either deficiency or excess—can lead to pathophysiological outcomes. This review summarized findings from animal studies, human observational research, and clinical investigations to explore the interplay between copper metabolism and diabetes-related complications.

Who Was Reviewed?

The review incorporated findings from diabetic and non-diabetic human subjects, diabetic animal models (primarily rodents), and observational studies spanning a range of diabetes subtypes, including type 1 diabetes mellitus (T1DM), type 2 diabetes mellitus (T2DM), and gestational diabetes mellitus (GDM). It also examined patients with varying stages of DKD, from normoalbuminuria to microalbuminuria.

Most Important Findings

The review highlighted the dual role of copper in diabetes and DKD. Elevated copper levels in serum or tissue are often linked with poor glycemic control and oxidative stress, which exacerbates diabetes-related complications. In T1DM and T2DM patients, higher copper levels and altered copper-to-zinc (Cu/Zn) ratios were correlated with increased glycated hemoglobin (HbA1c) levels, indicating poor glucose regulation. In DKD, urinary copper excretion increases in advanced stages, suggesting that impaired renal function may disrupt copper homeostasis, leading to nephrotoxicity and disease progression.

Animal studies demonstrated that copper-chelating agents like tetrathiomolybdate and triethylenetetramine (TETA) reduced oxidative stress, improved glucose tolerance, and mitigated renal and cardiac damage in diabetic models. These findings emphasize the potential of copper regulation as a therapeutic avenue. The Zn/Cu ratio emerged as a potential biomarker for assessing metabolic and renal health in diabetes, with a higher ratio associated with better glycemic control and reduced DKD risk.

Greatest Implications

The findings underscore the significance of maintaining copper homeostasis in diabetes management. Dysregulated copper metabolism contributes to oxidative stress, inflammation, and tissue damage, which accelerates the progression of diabetes and its complications. Interventions targeting copper balance, such as dietary adjustments, copper chelators, or enhancing the Zn/Cu ratio, could offer novel therapeutic strategies for preventing or managing DKD. Moreover, the Zn/Cu ratio could serve as a diagnostic and prognostic biomarker in diabetic populations.

Presence of metalloestrogens in ectopic endometrial tissue

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study examined the presence of metalloestrogens in ectopic endometrial tissue from fifty women diagnosed with endometriosis. Cadmium, nickel, and lead were found in all tissue samples, with nickel and lead showing particularly high concentrations. These findings suggest that metalloestrogens play a role in the etiology of endometriosis by interacting with estrogen receptors, emphasizing environmental pollutants' role in endometriosis progression.

What was studied?


This study investigated the presence of metalloestrogens in ectopic endometrial tissue from women with endometriosis. Metalloestrogens, heavy metals that can mimic estrogen and may contribute to estrogen-dependent diseases, were the focus, particularly regarding their potential role in the persistence and pathology of endometriosis. The researchers specifically analyzed levels of cadmium, nickel, and lead in ectopic endometrial samples using advanced metal detection techniques, Total Reflection X-ray Fluorescence (TXRF) and Graphite Furnace Atomic Absorption Spectroscopy (GFASS).

Who was studied?


The study included fifty women of reproductive age diagnosed with endometriosis via laparoscopy or laparotomy at the Professorial Gynecology Unit of the National Hospital, Colombo, Sri Lanka, during 2009-2010. The participants underwent these procedures for diagnosis or treatment, and endometriotic tissue samples were collected during surgery. The participants presented with varied symptoms like infertility, dysmenorrhea, chronic pelvic pain, and endometriomas.

What were the most important findings?


The study found significant levels of cadmium, nickel, and lead in all ectopic endometrial tissue samples. Specifically, geometric mean concentrations were reported as follows: cadmium (2.861 μg/Kg), nickel (17.547 μg/Kg), and lead (25.785 μg/Kg). The concentrations varied by tissue site, with the ovarian endometrioma wall showing higher, though not statistically significant, metal levels than pelvic endometrial patches or nodules in the pouch of Douglas.

Implications


This study is one of the first to identify and quantify metalloestrogens in ectopic endometrial tissue, shedding light on a possible environmental and molecular link to endometriosis. It underscores the mechanism by which these metals could perpetuate endometriosis, given their ability to interact with estrogen receptors in ectopic tissue. The implications are substantial for public health, especially given the widespread environmental exposure to metals such as cadmium, nickel, and lead. These findings suggest that environmental pollution may play a significant role in the etiology and progression of endometriosis, calling for further investigation into the estrogen-mimicking properties of environmental metals and their regulation. Additionally, the study highlights the need for preventive measures to reduce heavy metal exposure to nickel and lead, particularly among women susceptible to estrogen-related diseases.

Presence of metalloestrogens in ectopic endometrial tissue

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study quantified metalloestrogens—cadmium, nickel, and lead—in ectopic endometrial tissue, suggesting their role in endometriosis persistence.

What Was Studied?

This study investigated the presence of metalloestrogens—heavy metals with estrogenic effects—in ectopic endometrial tissue. Metalloestrogens, such as cadmium, nickel, and lead, have been implicated in estrogen-dependent diseases like endometriosis. The study aimed to quantify these metals in ectopic endometrial tissues from women diagnosed with endometriosis, using advanced analytical techniques.

Who Was Studied?

The study included 50 women of reproductive age who had endometriosis confirmed through laparotomy or laparoscopy. The participants were patients from a gynecology unit at a tertiary care hospital in Sri Lanka. Samples of ectopic endometrial tissue were collected from these women during surgical procedures, and the disease severity was classified based on the Revised American Society for Reproductive Medicine classification system.

What Were the Most Important Findings?

The study found significant levels of three metalloestrogens—cadmium (2.861 µg/kg), nickel (17.547 µg/kg), and lead (25.785 µg/kg)—in all ectopic endometrial tissue samples analyzed. Among these, lead exhibited the highest concentration. The study is notable for being the first to report the quantitative detection of metalloestrogens in ectopic endometrial tissue. Notably, the presence of these metals varied slightly depending on the tissue site, such as the wall of an endometrioma or nodules in the pelvic region, though these differences were not statistically significant. The findings suggest a potential role for environmental metalloestrogens in the persistence and progression of endometriosis.

What Are the Greatest Implications of This Study?

The detection of metalloestrogens in ectopic endometrial tissue underscores their role in the etiology and maintenance of endometriosis. These metals may act as endocrine disruptors, binding to estrogen receptors in ectopic tissue and mimicking estrogenic effects, thereby contributing to the persistence of the disease. The findings highlight the need for further research to elucidate the mechanistic pathways by which metalloestrogens influence endometriosis. Clinicians should consider environmental exposures and diet as a factor in managing and preventing this condition.

Relaxed fibronectin: a potential novel target for imaging endometriotic lesions

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

Research on relaxed fibronectin as a target for imaging endometriotic lesions showed that a novel radiotracer binds preferentially to this protein in disease areas. This finding could lead to improved diagnostic techniques for endometriosis, offering a non-invasive method to detect lesions accurately, thereby enhancing treatment planning and patient outcomes.

What was studied?

The study investigated relaxed fibronectin as a novel target for imaging endometriotic lesions. Researchers explored using a preclinical radiotracer, [111In]In-FnBPA5, which binds specifically to relaxed fibronectin, an extracellular matrix protein involved in the pathogenesis of diseases like cancer and fibrosis.

Who was studied?

The study involved preclinical experiments using mice and immunohistochemical analysis on tissue samples from mice and patients diagnosed with endometriosis.

What were the most important findings?

The radiotracer [111In]In-FnBPA5 accumulated in the mouse uterus, with uptake varying according to the estrous cycle, suggesting an increased abundance of relaxed fibronectin during estrogen-dependent phases. Immunohistochemical analysis on patient-derived tissues showed that relaxed fibronectin is preferentially located near the endometriotic stroma, supporting its potential as a target for imaging endometriosis.

What are the greatest implications of this study?

The findings that [111In]In-FnBPA5 uptake varies in the mouse uterus with the estrous cycle, indicating increased relaxed fibronectin during estrogen-dependent phases, hold significant implications for future research on endometriosis.

Biomarker Identification: Understanding the fluctuation of relaxed fibronectin could help identify biomarkers for endometriosis, enabling earlier and more accurate diagnosis.

Pathogenesis Insights: These results suggest that estrogen-driven changes in fibronectin might play a role in the development or exacerbation of endometriosis. This could lead to a better understanding of the disease’s underlying mechanisms.

Targeted Therapies: By highlighting the relationship between estrogen, fibronectin, and endometrial tissue changes, new therapeutic targets may be identified, paving the way for treatments that modulate fibronectin or its pathways.

Diagnostic Imaging: The study suggests that targeting relaxed fibronectin could significantly improve the diagnostic imaging of endometriosis. This approach may lead to developing a specific radiotracer for noninvasive detection of endometriotic lesions, potentially enhancing diagnosis accuracy and aiding in better disease management.

Clinical Application: The researchers also suggest using gallium-68 for potential clinical application, which could further refine imaging techniques and improve patient outcomes.

Role of Cholestyramine in Refractory Hyperthyroidism: A Case Report and Literature Review

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

A 52-year-old woman with refractory iodine-induced hyperthyroidism showed significant improvement with cholestyramine, reducing FT4 by 30% in 5 days. Despite conventional treatments failing, cholestyramine proved effective, leading to euthyroidism. This highlights cholestyramine's potential as an adjunct therapy.

What was studied?

The study investigated the role of cholestyramine as an additional treatment for refractory iodine-induced hyperthyroidism in a patient who did not respond to conventional therapies.

Who was studied?

A 52-year-old female patient with a history of goiter who developed iodine-induced hyperthyroidism following a CT scan with contrast. The patient had obstructive symptoms and was unresponsive to standard treatments, including dexamethasone, carbimazole, and propranolol.

What were the most important findings?

After adding cholestyramine, the patient’s FT4 levels decreased by 30% within 5 days and normalized by 12 days.

What are the greatest implications of this study?

Cholestyramine can be an effective adjunct therapy for managing refractory iodine-induced hyperthyroidism, suggesting a potential new treatment avenue for similar cases, such as Grave's Disease (GD). This case highlights the need for alternative treatments when conventional therapies fail and emphasizes the utility of cholestyramine in rapid thyroid hormone reduction.

Role of Cholestyramine in Refractory Hyperthyroidism A Case Report and Literature Review

Role of Nickel in Microbial Pathogenesis

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

Nickel-dependent enzymes like urease and hydrogenase are essential for pathogen virulence. This review outlines the mechanisms by which pathogens acquire and utilize nickel and explores implications for therapy and microbiome balance.

What was reviewed?

This comprehensive review article by Maier and Benoit (2019) explores the role of nickel as a critical cofactor in microbial pathogenesis, detailing its involvement in virulence-related enzymes and systems across a broad range of prokaryotic and eukaryotic pathogens. The review synthesizes decades of experimental findings regarding nickel-dependent enzymes—primarily urease and [NiFe]-hydrogenase—and their roles in microbial survival, colonization, biofilm formation, and host damage. Additionally, the authors examine nickel uptake systems, transporters, metallochaperones, storage proteins, and host immune mechanisms that attempt to limit nickel availability through nutritional immunity. The paper also emphasizes the dual nature of nickel as both a microbial nutrient and a potential antimicrobial target, especially in pathogens that rely on Ni-enzymes for virulence.

Who was reviewed?

The review includes a detailed catalog of more than 40 prokaryotic and nine eukaryotic pathogens harboring nickel-requiring enzymes. These include Helicobacter pylori, Staphylococcus aureus, Salmonella Typhimurium, Proteus mirabilis, Campylobacter jejuni, and Cryptococcus neoformans, among others. The review discusses these organisms in the context of their nickel metabolic strategies, virulence mechanisms, and interactions with host environments.

What were the most important findings?

The most significant finding is that nickel acts as a vital cofactor for several enzymes directly implicated in pathogenesis. Urease and [NiFe]-hydrogenase, the two primary nickel-dependent enzymes, enable microbial survival in hostile (especially acidic) environments and provide essential metabolic advantages during host colonization. For instance, in H. pylori, urease is essential for stomach colonization and contributes to carcinogenesis by promoting angiogenesis and chronic inflammation. Similarly, S. aureus upregulates urease genes in biofilms and uses the enzyme for kidney persistence. P. mirabilis utilizes urease to form crystalline catheter biofilms that enhance infection. Additionally, [NiFe]-hydrogenases in S. Typhimurium and C. jejuni are shown to fuel ATP production via hydrogen oxidation, aiding in host colonization and immune evasion.

The review also highlights that nickel availability is highly restricted in host tissues, leading pathogens to develop sophisticated acquisition systems such as NikABCDE and NixA transporters, metallophores (e.g., staphylopine, pseudopaline), and histidine-rich chaperones like HypB. Host defense mechanisms—including calprotectin, lactoferrin, and potentially hepcidin—attempt to sequester nickel and inhibit Ni-enzyme activation, with successful outcomes in some models.

From a microbiome perspective, commensal microbes such as urease-positive Bifidobacterium, Lactobacillus, and methanogenic archaea also utilize nickel enzymes, suggesting that systemic nickel depletion or chelation strategies could disrupt microbial homeostasis. This raises an important consideration: while nickel-targeted therapies might inhibit pathogens, they could also cause dysbiosis if not carefully balanced.

What are the greatest implications of this review?

The key implication is that nickel metabolism represents both an Achilles’ heel and a strategic fulcrum for pathogenic microbes. The clear reliance of multiple pathogens on nickel-dependent enzymes opens avenues for antimicrobial development, particularly through chelation therapies, targeted inhibition of Ni-enzyme maturation, or interference with nickel import systems. However, this also necessitates caution: beneficial microbes within the host microbiota depend on similar enzymes, and indiscriminate nickel disruption could lead to dysbiosis, undermining long-term host resilience.

For the microbiome signatures database, this review reinforces the value of including nickel-metabolism genes or Ni-enzyme prevalence in microbial trait profiling. For instance, the consistent enrichment of urease-positive pathogens in urinary tract infections and their association with stone formation could serve as microbiome-level markers. Moreover, the presence of nickel-dependent hydrogenases in enteric pathogens suggests a link between dietary nickel intake, microbial virulence, and gut ecosystem dynamics. Clinicians using microbiome data should be aware that modulating dietary nickel or applying nickel-targeted interventions could shift the microbial landscape significantly, both positively and negatively.

The Association between Zinc and Copper Circulating Levels and Cardiometabolic Risk Factors in Adults: A Study of Qatar Biobank Data

May 20, 2025
  • Cardiovascular Health
    Cardiovascular Health

    Recent research has revealed that specific gut microbiota-derived metabolites are strongly linked to cardiovascular disease risk—potentially influencing atherosclerosis development more than traditional risk factors like cholesterol levels. This highlights the gut microbiome as a novel therapeutic target for cardiovascular interventions.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study evaluated the associations of zinc, copper, and Zn/Cu ratio with cardiometabolic risk factors in Qatari adults, revealing copper’s protective role and Zn/Cu ratio’s adverse implications for metabolic health.

What was studied?

This study analyzed the relationship between zinc (Zn), copper (Cu), and the zinc-to-copper (Zn/Cu) ratio with cardiometabolic risk (CMR) factors and metabolic syndrome (MetS) using data from the Qatar Biobank. It sought to determine whether circulating levels of these trace minerals and their ratios were associated with various markers of cardiometabolic health, including lipid profiles, blood pressure, glucose levels, and body composition.

Who was studied?

The study included 437 Qatari adults aged 18 and older, representing both sexes. Participants had detailed cardiometabolic and mineral status profiles measured. Individuals with non-communicable diseases, those taking mineral supplements, and pregnant or lactating women were excluded to ensure a clear analysis of trace mineral associations with CMR factors.

Key Findings

This study revealed several associations between trace mineral levels and cardiometabolic markers. High Cu levels were associated with a reduced risk of MetS, lower diastolic blood pressure (DBP), and decreased prevalence of low HDL cholesterol, suggesting a protective role of copper in cardiometabolic health. Conversely, a higher Zn/Cu ratio was linked to an increased risk of MetS and low HDL, indicating that imbalances in these trace elements could worsen metabolic health.

While Zn alone was not strongly correlated with MetS or most CMR factors, it showed weak positive correlations with waist circumference (WC) and triglycerides (TG), which are notable for metabolic processes. Cu, on the other hand, positively correlated with HDL and total cholesterol (TC) while negatively correlating with DBP. These findings emphasize the differential and sometimes opposing roles of these minerals in cardiometabolic regulation.

In terms of microbiome relevance, trace elements like Zn and Cu influence microbial composition and metabolic functions. For example, Zn deficiency can affect glucose metabolism and inflammation, while Cu is a cofactor for antioxidative enzymes like superoxide dismutase, influencing oxidative stress pathways. Dysregulation of these pathways is often linked to microbial dysbiosis, potentially contributing to MetS and other cardiometabolic conditions.

Greatest Implications

The results underscore the need to consider trace element levels, particularly Cu and the Zn/Cu ratio, in cardiometabolic health assessments. The findings suggest that higher Cu levels confer protective effects against MetS and DBP, whereas an elevated Zn/Cu ratio increases the risk of adverse outcomes, including low HDL and MetS. These insights could inform clinical interventions, such as dietary adjustments or supplementation, to balance trace mineral levels and support cardiometabolic health. Additionally, these results highlight the potential role of trace mineral modulation as part of microbiome-targeted therapies, given their influence on systemic inflammation and metabolism.

The copper chelator ammonium tetrathiomolybdate inhibits the progression of experimental endometriosis in TNFR1-deficient mice

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

Ammonium tetrathiomolybdate (TM) inhibits endometriosis progression in TNFR1-deficient mice by reducing copper and estradiol levels, lesion growth, angiogenesis, and oxidative stress.

What Was Studied?

This study evaluated the therapeutic potential of ammonium tetrathiomolybdate (TM), a copper chelator, in inhibiting the progression of experimental endometriosis (EDT) in TNFR1-deficient mice. It explored TM's effects on copper and estradiol concentrations, lesion development, angiogenesis, oxidative stress, and inflammatory pathways. The research aimed to address how TM mitigates EDT in a worsened state caused by TNFR1 deficiency, a condition characterized by reduced cell death and increased lesion proliferation.

Who Was Studied?

The subjects were TNFR1-deficient female C57BL/6 mice divided into three groups: sham-operated (KO Sham), EDT-induced (KO EDT), and EDT-induced with TM treatment (KO EDT+TM). EDT was induced via autologous uterine tissue transplantation into the intestinal mesentery, and TM was administered orally postoperatively. Experimental outcomes were evaluated one month after EDT induction.

Most Important Findings

The study revealed several critical findings. First, EDT induction significantly elevated copper and estradiol levels in the peritoneal fluid, both of which were restored to physiological levels with TM treatment. TM also reduced lesion volume and weight, decreased cell proliferation, and suppressed angiogenesis, as evidenced by lower blood vessel counts and reduced expression of Vegfa, Fgf2, and Pdgfb. Furthermore, TM altered oxidative stress markers, decreasing the activity of superoxide dismutase (SOD) and catalase (CAT) while increasing lipid peroxidation, suggesting a pro-oxidative environment conducive to apoptotic signaling.

From a microbiome perspective, copper's role as a metalloestrogen and its involvement in estradiol synthesis underline the relevance of copper chelation in addressing estrogen-dependent diseases like endometriosis. By reducing copper levels, TM may disrupt microbial contributions to oxidative stress and inflammation, though direct microbiome-specific findings were not explored.

Greatest Implications

The study's findings suggest TM's dual role in reducing pro-inflammatory and pro-angiogenic pathways while restoring copper and estradiol homeostasis. These mechanisms are vital for mitigating EDT progression, particularly in the context of TNFR1 deficiency, where pathological signaling is exacerbated. Clinically, TM represents a potential adjunct therapy for managing endometriosis, particularly in cases resistant to conventional hormone treatments. The findings also reinforce the broader therapeutic relevance of targeting trace metals like copper in inflammatory and estrogen-dependent conditions.

The effect of a low-nickel diet and nickel sensitization ongastroesophageal reflux disease: A pilot study

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

A low-nickel diet significantly reduced GERD symptoms in 95% of participants, regardless of patch test status. The results suggest dietary nickel may contribute to GERD pathophysiology independently of classical allergy mechanisms.

What was studied?

This prospective pilot study investigated the therapeutic potential of a Low‑Nickel diet (LNiD) in patients with refractory gastroesophageal reflux disease (GERD) and explored whether epicutaneous patch testing for nickel (Ni) could predict dietary responsiveness. The researchers sought to determine whether dietary nickel contributes to GERD pathophysiology and whether identifying nickel sensitivity via patch testing can inform clinical decision-making.

Who was studied?

Twenty adult patients with a confirmed diagnosis of GERD were enrolled at a single site in West Virginia. All had persistent GERD symptoms despite at least three months of proton pump inhibitor (PPI) therapy and a GERD-HRQL score ≥30 at baseline. Patch testing was conducted using nickel sulfate and three additional common allergens (cobalt chloride, balsam of Peru, and cinnamic aldehyde), but patients were blinded to their patch test results until the end of the study. All participants adhered to a standardized low-nickel diet for eight weeks. GERD symptom severity was evaluated pre- and post-intervention using the validated GERD Health-Related Quality of Life (GERD-HRQL) questionnaire, which assesses heartburn, regurgitation, and overall symptom burden.

What were the most important findings?

Nineteen out of twenty participants (95%) reported substantial improvement in GERD symptoms after following the low-nickel diet. Mean total GERD-HRQL scores dropped by 27.05 points, while mean heartburn and regurgitation scores declined by 11.45 and 10.85 points, respectively—all statistically significant reductions (p < 0.001). Improvements were consistent across participants, regardless of nickel patch test results. Although 3 participants (15%) tested positive for nickel sensitivity, their symptom improvement was only modestly different from those who tested negative. Importantly, the magnitude of symptom reduction was not significantly correlated with patch test status. Nearly half of participants (45%) reported being satisfied with their GERD symptoms post-intervention, a notable shift from the 95% dissatisfaction rate at baseline.

While the study did not measure microbiome parameters directly, the data suggest nickel may act as a dietary irritant that exacerbates esophageal inflammation independently of overt allergic sensitization. This opens the possibility that dietary nickel contributes to barrier disruption or mucosal immune activation along the upper gastrointestinal tract—mechanisms well-documented in nickel-induced contact mucositis and consistent with microbial shifts observed in conditions like irritable bowel syndrome IBS and H. pylori infection, both of which have been responsive to nickel restriction.

What are the greatest implications of this study?

This study establishes that a low-nickel diet may be a powerful non-pharmacologic intervention for GERD, even in patients unresponsive to standard therapy. The data challenge the assumption that only patients with demonstrable nickel allergy benefit from nickel restriction, suggesting that dietary nickel may provoke inflammation through mechanisms not captured by skin-based patch testing. This has important clinical implications: patch test–guided exclusion diets may miss a substantial subset of patients who stand to benefit from nickel reduction. The findings justify broader consideration of dietary nickel as a contributing factor in GERD and potentially other inflammatory gastrointestinal conditions. Importantly, the intervention had excellent adherence and tolerability, supporting its viability in routine care. Future randomized, controlled trials with larger sample sizes and microbial/metallomic profiling are warranted to validate these findings and explore underlying mechanisms.

The Influence of Lactoferrin in Plasma and Peritoneal Fluid on Iron Metabolism in Women with Endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study demonstrates that peritoneal fluid-to-plasma ferritin and lactoferrin ratios distinguish endometriosis stage and severity. Iron overload and shifting iron-binding protein profiles reveal a localized dysregulation that may influence disease progression and potentially pathogenic microbiome selection.

What was studied?

This study investigated the role of lactoferrin (LF) in relation to iron metabolism in women with and without endometriosis by measuring levels of LF, ferritin (FT), transferrin (TF), and iron (Fe) simultaneously in plasma and peritoneal fluid. The authors specifically explored whether the concentrations and ratios of these iron-related proteins in the two biological compartments could distinguish the presence and progression of endometriosis. The goal was to identify noninvasive or minimally invasive biomarkers that may aid in diagnosing or staging the disease based on iron metabolism, especially given endometriosis’ pro-inflammatory, iron-rich microenvironment.

Who was studied?

The study cohort included 90 women of reproductive age undergoing diagnostic laparoscopy, of whom 57 had histologically confirmed endometriosis (stages I–IV) and 33 did not. Plasma and peritoneal fluid samples were collected pre- and intra-operatively. Subjects were classified based on endometriosis diagnosis and stage, and specimens were evaluated for levels of LF, FT, TF, and Fe using ELISA, immunoturbidimetric assay, and colorimetric methods.

What were the most important findings?

Key findings highlight that ferritin and iron concentrations were significantly elevated in peritoneal fluid compared to plasma, especially in patients with advanced-stage endometriosis. In contrast, transferrin was consistently lower in peritoneal fluid. Notably, lactoferrin levels did not significantly differ between women with and without endometriosis when evaluated independently in plasma or peritoneal fluid, but the peritoneal fluid/plasma lactoferrin ratio decreased progressively with increasing disease severity, significantly distinguishing stage I from stage IV. The ferritin ratio was markedly higher in the endometriosis group, underscoring its potential as a disease marker. Correlation analyses revealed that in severe endometriosis, lactoferrin was significantly associated with ferritin and iron in the peritoneal fluid, suggesting a disrupted iron regulation mechanism localized to the disease microenvironment. Importantly, the elevated ferritin concentrations in peritoneal fluid may serve a compensatory, protective role to sequester iron and mitigate oxidative stress, while lactoferrin may lose this protective function as disease progresses.

From a microbiome perspective, this study underscores the iron-dependent ecological shifts that may select for siderophilic pathobionts. The iron overload and pro-oxidative milieu likely fosters the expansion of iron-requiring microbial taxa, potentially including Escherichia, Enterobacter, and Fusobacterium, known to be enriched in some endometriosis microbiome signatures. While microbial profiling was not performed, the metallomic dysregulation described supports the hypothesis that iron availability is a crucial factor in shaping pathogenic microbial communities in endometriosis.

What are the greatest implications of this study?

This study provides compelling evidence that iron-binding proteins—particularly ferritin and lactoferrin—play a localized and differential role in the progression of endometriosis. The findings suggest that peritoneal fluid iron metabolism, and especially the ferritin-to-lactoferrin balance, may be a critical axis of disease progression and potentially a therapeutic target. The study introduces the peritoneal fluid/plasma concentration ratio as a novel diagnostic parameter, offering a more granular assessment than conventional plasma markers. The declining lactoferrin ratio and increasing ferritin ratio with disease severity may signal a transition from iron sequestration and immune modulation toward iron-driven oxidative stress and tissue damage. This may serve as a foundation for the development of metallome-targeted diagnostics and therapies, including exogenous lactoferrin supplementation, which the authors suggest could restore iron balance in advanced disease stages. These findings also have implications for understanding how iron dysregulation may foster microbial dysbiosis, providing a mechanistic link between host iron metabolism and the pathophysiological selection of microbial communities in endometriosis.

The influence of nickel on intestinal microbiota disturbances

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

  • Microbes
    Microbes

    Microbes, short for microorganisms, are tiny living organisms that are ubiquitous in the environment, including on and inside the human body. They play a crucial role in human health and disease, functioning within complex ecosystems in various parts of the body, such as the skin, mouth, gut, and respiratory tract. The human microbiome, which is […]

Excess nickel disrupts gut microbiota, promoting dysbiosis and contributing to conditions like obesity and systemic nickel allergy syndrome (SNAS). Probiotics and nickel-restricted diets show promise in mitigating these effects, underscoring the need for further research and clinical intervention.

What was reviewed?

The paper reviewed the influence of nickel on intestinal microbiota disturbances, drawing on 59 scientific publications from the past 20 years. The analysis focused on nickel’s dual role as an essential element for microbial enzymatic reactions and a disruptor of gut microbiota, especially under conditions of excessive exposure or systemic nickel allergy syndrome (SNAS).

Who was reviewed?

The review encompassed research involving humans, animals, and microbial models. Specific attention was given to populations exposed to high levels of nickel, individuals with SNAS, and animal studies demonstrating changes in microbial communities under nickel exposure.

What were the most important findings?

Nickel acts as a cofactor for metalloenzymes like urease, hydrogenase, and [NiFe]-hydrogenase, essential for microbial survival. However, excess nickel promotes dysbiosis, characterized by reductions in beneficial taxa and increases in nickel-resistant bacteria. In humans with SNAS, the microbiota showed decreased levels of beneficial genera such as Bifidobacterium and Lactobacillus, known for their probiotic effects and urease activity, and increases in nickel-tolerant taxa, including Clostridiaceae and Bacillaceae. Similarly, animal studies indicated reduced Verrucomicrobia and Bacteroidetes while promoting Escherichia coli and Enterococcus.

Nickel exposure also leads to an increased abundance of Bacteroides fragilis, Bacteroidales S24-7, and Interstinimonas, with a concurrent decline in Firmicutes, disrupting the Firmicutes-to-Bacteroidetes ratio, a critical marker of gut health. This imbalance contributed to systemic inflammation and altered immune responses. Moreover, nickel-reliant pathogens, such as Helicobacter pylori, which require Ni2+-dependent enzymes like urease for colonization, further highlighted nickel’s role in microbial pathogenicity. Probiotic strains such as Lactobacillus fermentum demonstrated detoxifying effects by metabolizing nickel, suggesting their therapeutic potential.

What are the greatest implications of this review?

The findings reveal that nickel exposure significantly alters gut microbial ecology, driving dysbiosis and systemic inflammation in susceptible populations. The rise of nickel-tolerant taxa, coupled with the decline of protective bacteria, underscores nickel’s role as a disruptor of gut homeostasis, contributing to conditions like obesity and SNAS. Probiotic supplementation, particularly strains capable of nickel detoxification, and dietary restrictions like a low-nickel diet, have shown promise in mitigating these effects. This review highlights the urgent need for dietary nickel regulations and further clinical studies on therapeutic interventions targeting nickel-induced microbial dysbiosis.

Trace Elements and Endometriosis: Insights into Oxidative Stress and Novel Therapies

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This review explores the role of trace elements and oxidative stress in endometriosis, highlighting their potential as therapeutic targets. It underscores the need for further research into the trace elements’ roles in endometriotic lesions.

What was reviewed?

The article reviews the role of trace elements in the pathogenesis and management of endometriosis, a chronic, estrogen-dependent inflammatory disease. It synthesizes existing research on the impact of oxidative stress and environmental exposure to trace elements like zinc, nickel, cadmium, and copper, linking these factors to the formation and proliferation of endometrial-like lesions outside the uterus.

Who was reviewed?

The review focuses on studies involving women with confirmed endometriosis, highlighting environmental and biological factors such as trace element concentrations in blood, urine, and peritoneal fluid. Additionally, it incorporates experimental findings, including animal models, to explore the mechanistic roles of trace elements.

What were the most important findings?

The review emphasizes the link between oxidative stress and endometriosis, with trace elements acting as potential modulators of this process. Zinc, for instance, is identified for its antioxidant and anti-inflammatory roles, with lower levels in endometriosis patients potentially contributing to lesion formation. Nickel, on the other hand, has been implicated in the condition as a metalloestrogen, as further evidenced by improved symptoms following a low-nickel diet. Cadmium and lead, known for inducing oxidative stress, show conflicting associations with endometriosis, though some evidence suggests their presence synergistically exacerbates disease severity. Copper's involvement in angiogenesis and its elevated levels in endometriosis patients suggest a role in lesion proliferation. The review also highlights discrepancies in study findings, emphasizing the need for further research on trace elements within endometriotic implants rather than just systemic fluids.

What are the greatest implications of this review?

The review underscores the potential of targeting trace elements and oxidative stress as therapeutic strategies for endometriosis. It calls for more comprehensive research into the specific roles of trace elements within endometriotic tissue, as these could pave the way for novel diagnostic markers and treatments. Additionally, the environmental and dietary implications of trace element exposure warrant further exploration, particularly in the context of prevention and symptom management.

Urine metallomics signature as an indicator of pancreatic cancer†

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

This study identifies a distinct urinary metallomic signature in pancreatic cancer patients, marked by altered calcium, magnesium, copper, and zinc levels, along with lighter zinc isotopic composition. These findings suggest that non-invasive urine tests could enable early PDAC detection by leveraging trace metal imbalances and stable isotope shifts.

What was studied?

This study explored the utility of urinary metallomic profiling—specifically concentrations and isotopic composition of essential metals—as a non-invasive diagnostic tool for pancreatic ductal adenocarcinoma (PDAC). The researchers examined urine samples from PDAC patients and healthy controls to identify specific metal dyshomeostasis and isotopic shifts that could serve as biomarkers for PDAC detection.

Who was studied?

Urine samples from 21 patients diagnosed with PDAC and 46 healthy control subjects were analyzed. All samples were collected under ethical approval through the Barts Pancreas Tissue Bank.

Most important findings:

A distinct urinary metallomic signature was identified in pancreatic ductal adenocarcinoma (PDAC) patients, characterized by decreased calcium and magnesium and increased zinc and copper levels. The multivariate model integrating these four elements exhibited outstanding diagnostic accuracy, achieving an area under the curve (AUC) of 0.995 with 99.5% sensitivity. Moreover, stable zinc isotope analysis revealed a shift toward isotopically lighter zinc in PDAC patients (median δ⁶⁶Zn = −0.15‰) compared to healthy controls (median δ⁶⁶Zn = +0.02‰), likely due to oxidative stress-induced oxidation of cysteine-rich metallothioneins, which preferentially bind lighter isotopes. From a microbiome-metallomic perspective, such trace metal imbalances—particularly involving zinc and copper—may influence microbial community structure by selectively enriching pathogenic taxa and diminishing beneficial ones. Although the microbiome was not directly assessed in this study, the metallomic disturbances observed may serve as indirect indicators of host-microbe dysregulation, especially relevant in gastrointestinal malignancies such as PDAC.

ElementChange in PDAC vs. Control
Calcium (Ca)Decreased (***p < 0.0001)
Magnesium (Mg)Decreased (**p = 0.0002)
Zinc (Zn)Increased (*p = 0.015)
Copper (Cu)Increased (*p = 0.02)

Greatest implications of the study:

This work provides strong preliminary evidence that urinary metallomic profiles—specifically Ca, Mg, Cu, Zn concentrations and zinc isotopic signatures—can serve as non-invasive biomarkers for PDAC detection. It is the first study to report isotopic zinc alterations in urine associated with PDAC and proposes a compelling mechanistic link to oxidative stress and metalloprotein dysregulation. If validated in larger cohorts, this approach could represent a breakthrough in early detection of pancreatic cancer, a malignancy notorious for its asymptomatic progression and poor prognosis. The authors propose that isotopic measurements, which offer significantly greater resolution than standard clinical assays, could even function as prognostic tools if longitudinally correlated with disease progression.

Essential Oils

Go to Category Page

Dietary essential oils improves the growth performance, antioxidant properties and intestinal permeability by inhibiting bacterial proliferation, and altering the gut microbiota of yellow-feather broilers

May 20, 2025
  • Essential Oils
    Essential Oils

    Essential oils are powerful natural substances that can modulate the gut microbiome, offering therapeutic benefits such as enhanced digestion, reduced inflammation, and improved immune function. With antimicrobial properties that selectively target harmful bacteria while promoting beneficial gut microbes, EOs like oregano, tea tree, and peppermint are emerging as effective interventions for optimizing health. However, safe application is crucial to avoid potential risks, including skin irritation and interactions with medications. Understanding the proper use of EOs ensures their effectiveness in supporting a balanced microbiome and overall well-being.

This study shows how essential oils improve broiler gut health, boost growth, and reshape microbiota, offering a natural antibiotic alternative.

What was studied?

This experimental study investigated the antibacterial properties of essential oils (EOs) and their effects on growth performance, intestinal morphology, antioxidant capacity, and gut microbiota of yellow-feathered broilers. A total of 720 male chicks were divided into four groups receiving different EO doses over 48 days. The study aimed to determine whether EOs could inhibit bacterial proliferation, improve gut health, and enhance overall poultry production performance. The researchers combined in vitro antimicrobial testing with in vivo assessments of intestinal structure, biochemical markers, and gut microbial shifts.

Who was studied?

The study focused on yellow-feathered broiler chickens, which are commonly used in poultry farming. These birds were fed either a control diet or diets supplemented with varying concentrations of EOs, which contained thymol and carvacrol as active ingredients. Their growth, intestinal morphology, antioxidant responses, serum biochemistry, and cecal microbiota were closely monitored. The microbial focus included pathogenic species like E. coli and Salmonella, along with beneficial genera such as Lactobacillus and Faecalibacterium, enabling a clear view of how the EO interventions reshaped gut microbial communities.

What were the most important findings?

The study demonstrated that EOs significantly inhibited the proliferation and biofilm formation of E. coli O78 and Salmonella pullorum, with minimum inhibitory concentrations (MICs. In vivo, EO supplementation enhanced average daily gain (ADG) and feed efficiency across multiple growth phases. Antioxidant markers such as superoxide dismutase (SOD) and catalase (CAT) activity improved, while intestinal permeability markers (villus height and tight junction protein expression) were positively affected. Gut microbiota analysis revealed that EO supplementation increased beneficial microbes, particularly Lactobacillus and Faecalibacterium, while reducing harmful genera such as Negativibacillus and Flavonifractor. Notably, the EO400 group showed the highest microbial diversity and upregulated pathways for chemoheterotrophy and fermentation. Correlation analysis linked improved gut microbiota profiles to better growth performance, suggesting that EO-driven microbial shifts were central to the observed production benefits.

What are the greatest implications of this study?

This study provides robust evidence that essential oils can serve as effective natural alternatives to antibiotic growth promoters (AGPs) in poultry farming. By reducing pathogenic bacteria and fostering beneficial microbiota, EOs improve gut health and production performance while mitigating risks associated with antibiotic resistance. The findings are particularly relevant as global regulations increasingly restrict AGP use. However, the dose-dependent effects and variable long-term impacts highlight the need for precise formulation and further research to optimize EO-based interventions in commercial settings.

Essential Oils, Chemical Compounds, and Their Effects on the Gut Microorganisms and Broiler Chicken Production

May 20, 2025
  • Essential Oils
    Essential Oils

    Essential oils are powerful natural substances that can modulate the gut microbiome, offering therapeutic benefits such as enhanced digestion, reduced inflammation, and improved immune function. With antimicrobial properties that selectively target harmful bacteria while promoting beneficial gut microbes, EOs like oregano, tea tree, and peppermint are emerging as effective interventions for optimizing health. However, safe application is crucial to avoid potential risks, including skin irritation and interactions with medications. Understanding the proper use of EOs ensures their effectiveness in supporting a balanced microbiome and overall well-being.

This review explores how essential oils enhance broiler chicken health by reshaping gut microbiota, improving growth, and acting as sustainable antibiotic alternatives.

What was reviewed?

This review systematically evaluated the role of essential oils (EOs) in improving broiler chickens' health, gut microbiota, and production outcomes. By analyzing 158 publications, it explored the antimicrobial, antioxidant, and growth-promoting properties of EOs sourced from a wide range of plants such as Origanum, Cymbopogon, Citrus, and Eucalyptus. The focus was on how these plant-derived compounds influence gut microbiota composition, inhibit pathogenic bacteria, and serve as sustainable alternatives to antibiotics in poultry farming. The review also detailed various modes of action, including nutrient absorption enhancement, immune system fortification, and oxidative stress reduction, positioning EOs as a promising tool for sustainable broiler production.

Who was reviewed?

The review focused on studies investigating broiler chickens, specifically those examining the effects of dietary or water supplementation with essential oils. It addressed different microbial communities within the chicken gut, noting common bacterial families such as Lachnospiraceae, Ruminococcaceae, and Veillonellaceae, and pathogens like Escherichia coli, Salmonella spp., and Clostridium perfringens. The review highlighted how EOs influenced these microbiota and the overall health and productivity of broilers.

What were the most important findings?

The review found that essential oils exert significant antimicrobial activity, especially against pathogens like E. coli, Klebsiella pneumoniae, Salmonella spp., and Staphylococcus spp. For example, cinnamaldehyde from cinnamon bark and compounds from Cymbopogon citratus were highly effective in reducing pathogenic bacterial loads, improving intestinal morphology, and promoting a healthier gut environment. Origanum EO not only enhanced gut health but also demonstrated strong antioxidant effects, while Citrus oils showed dual benefits of microbial control and reduction of Eimeria oocysts. Importantly, the use of EOs led to improvements in nutrient absorption through enhanced gut morphology, specifically increasing villi height and enzyme activity, thereby boosting growth performance. These findings emphasize the potential of EOs to rebalance gut microbial populations toward beneficial species like Lactobacillus, reduce pathogenic colonization, and serve as natural growth promoters.

What are the greatest implications of this review?

The review underscores that essential oils can act as sustainable and effective alternatives to antibiotics in poultry production. Their ability to reduce pathogenic bacteria while supporting beneficial gut microbiota is critical in addressing antibiotic resistance issues. The implications extend to safer poultry products for consumers and better environmental outcomes. However, the review also highlighted the need for further research to standardize EO formulations, determine optimal dosages, and assess long-term safety for both animals and humans. The findings encourage poultry producers to consider EOs as part of integrated gut health management strategies that align with global moves toward antibiotic-free animal farming.

Brain Health

Go to Category Page

Effectiveness of Partially Hydrolyzed Guar Gum on Cognitive Function and Sleep Efficiency in Healthy Elderly Subjects in a Randomized, Double-Blind, Placebo-Controlled, and Parallel-Group Study

May 20, 2025
  • Brain Health
    Brain Health

    Brain health encompasses the overall functioning and well-being of the brain, including cognitive function, emotional and psychological well-being, neurological integrity, behavioral health, neurodevelopmental health, age-related brain health, and brain resilience and plasticity.

The study explores the potential of partially hydrolyzed guar gum (PHGG), a water-soluble prebiotic dietary fiber, to improve cognitive function, sleep efficiency, and overall mental health in healthy elderly individuals. Given the growing global concern over cognitive decline and dementia among aging populations, the study investigates PHGG as a functional food component that may positively influence brain health through the gut-brain axis.

What Was Reviewed?

The study reviewed the effectiveness of partially hydrolyzed guar gum (PHGG), a water-soluble prebiotic dietary fiber, on cognitive function, sleep efficiency, and overall mental health in elderly individuals. The research specifically focused on assessing the impact of PHGG supplementation on cognitive domains such as visual memory and simple attention, sleep quality parameters like sleepiness on rising, and mood states including vigor and confusion. The study also considered the safety and tolerability of PHGG in the target population. The review encompasses the potential mechanisms through which PHGG may exert its effects, particularly its role in modulating the gut microbiome and the production of short-chain fatty acids (SCFAs), which are implicated in the gut-brain axis and neuroprotection.

Who Was Reviewed?

The subjects of the review were 66 healthy elderly Japanese individuals aged 60 years or older. These participants were free from cognitive impairment (as indicated by a Mini Mental State Examination score of 24 or higher) and were not undergoing treatment for chronic diseases that could influence the outcomes. The participants were randomly assigned to receive either PHGG supplementation (5 g/day) or a placebo for a duration of 12 weeks. The study specifically targeted an elderly population to investigate whether PHGG could mitigate age-related cognitive decline and improve sleep quality, given that these issues are particularly prevalent in this demographic.

What Were the Most Important Findings of This Review?

Cognitive Function:

The most significant finding was the improvement in visual memory observed in the PHGG group after 12 weeks of supplementation. Visual memory scores were significantly higher in the PHGG group compared to the placebo group, suggesting that PHGG has a positive effect on this critical cognitive domain. Improvements in simple attention were also noted at 8 weeks, although this was less emphasized.


Sleep Quality:

The PHGG group demonstrated significant improvements in sleep quality, particularly in the domain of "sleepiness on rising," after 8 weeks of supplementation. This improvement indicates better sleep efficiency and mental clarity upon waking, which are essential for maintaining daily function in the elderly.


Mood and Mental Health:

Although no significant intergroup differences were observed, within-group analyses revealed that PHGG supplementation led to increased vigor and reduced confusion, suggesting a potential benefit of PHGG on mood states, although these findings were more exploratory.


Safety:

The study confirmed the safety of PHGG, as no adverse events were reported, making it a viable supplement for elderly populations.

What Are the Greatest Implications of This Review?

Potential Role of PHGG in Cognitive Health:

The study suggests that PHGG supplementation could serve as a functional food intervention to enhance cognitive function, particularly visual memory, in elderly individuals. This finding is significant as visual memory is crucial for daily activities and maintaining independence in aging populations. The positive effects observed may indicate that PHGG could be a valuable tool in preventing or delaying cognitive decline.


Enhancement of Sleep Quality:

Improved sleep quality, as evidenced by reduced sleepiness on rising, has broad implications for overall health and well-being in the elderly. Sleep disturbances are common in aging, and interventions like PHGG that can improve sleep efficiency are likely to contribute to better cognitive function, mood, and quality of life.


Implications for the Gut-Brain Axis:

The study reinforces the concept that the gut microbiome, modulated by prebiotic interventions like PHGG, plays a crucial role in brain health. By promoting the production of SCFAs and improving gut health, PHGG may influence brain function through the gut-brain axis, offering a non-pharmacological approach to support cognitive and mental health in the elderly.


Foundation for Future Research:

While the study provides promising data, it also highlights the need for further research with larger sample sizes, longer durations, and objective assessments. The findings lay the groundwork for more comprehensive studies that could explore the long-term effects of PHGG on cognitive decline, its mechanisms of action, and its potential to prevent dementia.

Microbiota–Gut–Brain Axis: Barrier Function and Lymphatic System in Neurological Health

May 20, 2025
  • Brain Health
    Brain Health

    Brain health encompasses the overall functioning and well-being of the brain, including cognitive function, emotional and psychological well-being, neurological integrity, behavioral health, neurodevelopmental health, age-related brain health, and brain resilience and plasticity.

This review explores the microbiota–gut–brain axis, highlighting the role of gut microbiota in barrier integrity and lymphatic transport. It discusses microbial metabolites, vagus nerve signaling, and meningeal lymphatics as critical communication pathways, emphasizing their implications for gastrointestinal and neurological disorders.

What Was Reviewed?

This review explores the microbiota–gut–brain axis (MGBA), with a focus on the interplay between the gut microbiota, intestinal and blood-brain barrier integrity, and the lymphatic system. The authors examine how gut microbes influence barrier function through neural transmission, metabolite production, immune modulation, and gut hormone signaling. A significant aspect of the review is the role of lymphatic vessels as a previously underappreciated conduit between the gut and brain. The review also discusses the impact of microbiota dysbiosis on barrier dysfunction and its implications for both gastrointestinal and neurological diseases.

Who Was Reviewed?

The review synthesizes findings from various microbiome studies, including those investigating the microbiota's role in intestinal permeability, neuroinflammation, and neurological conditions. It integrates evidence from experimental models and human studies to highlight key mechanisms underlying MGBA communication.

Key Findings and Microbiome Associations

The review underscores that the gut microbiota exerts a profound influence on both the intestinal and blood-brain barriers, modulating permeability and contributing to systemic homeostasis. Several key points emerge:

Microbiota and Barrier Function: Gut microbes regulate intestinal and blood-brain barrier integrity through microbial metabolites such as short-chain fatty acids (SCFAs), neurotransmitter production, and immune modulation. Butyrate, for example, strengthens the blood-brain barrier by enhancing tight junction protein expression.

Lymphatic System as a Communication Pathway: The lymphatic network, particularly intestinal lacteals, serves as a conduit for microbiota-derived molecules and immune cells, linking gut health with central nervous system (CNS) function. Dysregulation of lymphatic transport mechanisms is implicated in neurological disorders.

Gut Microbiota Dysbiosis and Neurological Conditions: Altered microbiota composition contributes to neuroinflammatory and neurodegenerative diseases. Increased gut permeability and translocation of microbial products, such as lipopolysaccharides (LPS), trigger systemic inflammation, which can exacerbate conditions like Alzheimer's and Parkinson’s disease.

Vagus Nerve and Microbial Metabolites: The vagus nerve is a major conduit for gut-brain signaling. Microbial-derived neurotransmitters, including serotonin and dopamine precursors, influence neurological health. In animal models, vagotomy disrupts gut microbiota–mediated neurological effects, further supporting the role of direct neural communication.

Meningeal Lymphatics and CNS Immunity: The meningeal lymphatic system is increasingly recognized as an essential pathway for brain waste clearance and immune regulation. Dysfunction in these lymphatic vessels is linked to neuroinflammatory conditions such as multiple sclerosis and Alzheimer's disease.

Implications of This Review

The findings emphasize the importance of maintaining gut microbiota balance to preserve barrier integrity and prevent systemic inflammation that may contribute to neurological diseases. This review suggests that therapeutic interventions targeting the microbiota—such as prebiotics, probiotics, fecal microbiota transplantation (FMT), and microbiota-modulating diets—could play a role in managing both gastrointestinal and neurodegenerative conditions. Additionally, interventions that enhance lymphatic function, such as VEGF-C-mediated lymphangiogenesis, have shown promise in mitigating neuroinflammatory disorders by regulating microbiota-host interactions.

Neuromicrobiology, an Emerging Neurometabolic Facet of the Gut Microbiome?

May 20, 2025
  • Brain Health
    Brain Health

    Brain health encompasses the overall functioning and well-being of the brain, including cognitive function, emotional and psychological well-being, neurological integrity, behavioral health, neurodevelopmental health, age-related brain health, and brain resilience and plasticity.

The paper reviews neuromicrobiology, examining how the gut microbiome influences brain health and cognitive function through neuroactive metabolites like GABA, serotonin, and dopamine, focusing on their biosynthesis, transport, and impact on the gut-brain axis and mental health.

What Was Reviewed?

The paper reviews the emerging field of neuromicrobiology, which explores the interactions between the gut microbiome and the brain, particularly focusing on how gut microbiota produce neuroactive metabolites that influence cognitive function and brain health. It addresses the biosynthesis, absorption, and transport of these neuroactive metabolites, including neurotransmitters such as γ-aminobutyric acid (GABA), serotonin, dopamine, and others. The review also discusses how these compounds interact with the gut-brain axis and their implications for mental health and neurological disorders.

Who Was Reviewed?

The review synthesizes research across multiple studies involving both human and animal models. It examines the gut microbiota's role in producing neuroactive compounds and their potential effects on the central nervous system (CNS). The paper does not focus on a specific population but rather on a broad range of studies that include both healthy and diseased subjects to understand the underlying mechanisms of gut-brain communication via microbial metabolites.

What Were the Most Important Findings of This Review?

Diversity of Neuroactive Metabolites:

The review highlights the diversity of neuroactive metabolites produced by the gut microbiome, including neurotransmitters like GABA, serotonin, dopamine, and their precursors. These metabolites are synthesized by a variety of gut bacteria, and their production is influenced by factors such as diet, genetics, and environmental conditions.


Mechanisms of Interaction with the Brain:

The paper details the pathways through which these neuroactive metabolites interact with the brain, emphasizing the "bottom-up" pathway of the gut-brain axis. This includes the direct signaling of neurotransmitters via the vagus nerve, modulation of the immune system, and the transport of metabolites across the blood-brain barrier (BBB) via transport proteins or secreted microbial extracellular vesicles (MEVs).


Impact on Mental Health and Neurological Disorders:

The review discusses how dysbiosis (an imbalance in gut microbiota) is linked to various mental health disorders, including depression, anxiety, and neurodegenerative diseases like Alzheimer's and Parkinson's. It suggests that microbial metabolites could play a significant role in the pathophysiology of these conditions, offering potential targets for therapeutic interventions.


Microbiota-Targeted Interventions (MBTIs):

The paper underscores the potential of microbiome-targeted interventions (MBTIs), such as prebiotics, probiotics, synbiotics, and postbiotics, to modulate gut-brain interactions. However, it also notes that the precise mechanisms underlying these interventions are not fully understood, which limits their current therapeutic application.


Challenges and Future Directions:

A major theme is the complexity and challenges of translating current findings into clinical practice. The review identifies gaps in understanding how microbial neuroactive metabolites specifically influence brain function and calls for more mechanistic studies to establish causality and therapeutic potential.

What Are the Greatest Implications of This Review?

Advancement of Neuromicrobiology:

The review positions neuromicrobiology as a crucial field for understanding the gut-brain axis and its impact on brain health. It suggests that advances in this area could lead to novel approaches for preventing and treating neurological and psychiatric disorders by targeting the gut microbiome.


Potential for Novel Therapeutics:

The insights into microbial production of neuroactive compounds open up possibilities for developing new microbiota-targeted therapies. These could include specific probiotics engineered to produce neurotransmitters, or prebiotic diets designed to enhance the production of beneficial metabolites, which could be tailored to individual patient needs based on their gut microbiome composition.


Integration of Multi-Omics Approaches:

The paper calls for the integration of metagenomic, metabolomic, and transcriptomic data to better understand the microbiome-gut-brain axis. This could enable a more comprehensive understanding of how gut microbes influence brain health and lead to the identification of biomarkers for disease or targets for intervention.


Need for Mechanistic Research:

The "Neuromicrobiology, an Emerging Neurometabolic Facet of the Gut Microbiome?" review emphasizes the need to move beyond correlation studies and towards mechanistic research that clarifies how specific gut microbes and their metabolites influence brain function. This will be critical for developing evidence-based therapeutic applications and understanding individual variability in response to microbiome-targeted interventions.


Implications for Public Health:

By highlighting the role of the gut microbiome in brain health, the review suggests that dietary and lifestyle interventions targeting the gut microbiome could become a key component of public health strategies for preventing cognitive decline and mental health disorders.

Image modified from the original "Neuromicrobiology, an emerging neurometabolic facet of the gut microbiome?" review paper. Figure 4: The pathways through which gut microbiota-derived neuroactive compounds reach the brain—indirect transportation via modulation of host neurotransmitter biosynthesis, microbial extracellular vesicle (MEV) transportation, and direct transport—highlight the complex interactions between the gut and the brain. These mechanisms provide multiple routes through which the gut microbiome can impact brain function and behavior, emphasizing the importance of the gut-brain axis in health and disease.

Review test

May 20, 2025
  • Brain Health
    Brain Health

    Brain health encompasses the overall functioning and well-being of the brain, including cognitive function, emotional and psychological well-being, neurological integrity, behavioral health, neurodevelopmental health, age-related brain health, and brain resilience and plasticity.

test

The gut microbiome in neurological disorders

May 20, 2025
  • Brain Health
    Brain Health

    Brain health encompasses the overall functioning and well-being of the brain, including cognitive function, emotional and psychological well-being, neurological integrity, behavioral health, neurodevelopmental health, age-related brain health, and brain resilience and plasticity.

This review underscores the pivotal role of the gut microbiome in neurological health and disease, while also highlighting the potential for developing microbiome-based therapies. However, it calls for caution in interpreting the current evidence, advocating for more rigorous research to translate these findings into clinical practice.

What was reviewed?

This review comprehensively examined the emerging role of the gut microbiome in neurological disorders, focusing on the microbiota-gut-brain axis—a bidirectional communication network that links the gut microbiome to central nervous system (CNS) functions. The authors reviewed existing literature and studies that explore how gut microbiota influence neurodevelopment, aging, and the pathophysiology of various neurological disorders, including Alzheimer’s disease, autism spectrum disorder (ASD), multiple sclerosis, Parkinson’s disease, stroke, and traumatic brain injury. The review also examines the potential for microbiome-targeted interventions (MBTIs) as therapeutic strategies in these conditions.

Who was reviewed?

The review primarily focused on:

Animal Models: A significant portion of the evidence comes from studies involving germ-free mice and other animal models, which have been used to demonstrate the impact of the gut microbiota on neurodevelopment, neuroinflammation, and behavior.

Human Studies: The review also included cross-sectional, observational, and interventional studies in human subjects, particularly in populations affected by neurological disorders. These human studies often explored correlations between microbiota composition and disease states, cognitive functions, and responses to microbiome-targeted interventions such as probiotics and dietary changes.

What were the most important findings of this review?

The most important findings of the review include:

Microbiota-Gut-Brain Axis: The gut microbiome plays a crucial role in brain health through multiple pathways, including immune modulation, neurotransmitter production, and regulation of neuroinflammation.

Neurodevelopment: Early-life microbiota composition significantly influences neurodevelopmental processes, with evidence from both animal and human studies suggesting that disruptions in the microbiota-gut-brain axis can affect cognitive and social behaviors.

Aging and Neurological Disorders: A diverse gut microbiome is associated with healthier aging and may protect against cognitive decline. In neurological disorders such as MS, PD, AD, and ASD, altered gut microbiota compositions have been observed, with certain bacterial taxa linked to disease pathophysiology.

Therapeutic Potential: There is growing evidence that microbiome-targeted interventions (e.g., probiotics, prebiotics, dietary changes) may modulate disease outcomes, though the field is nascent, and robust clinical trials are needed to confirm these therapeutic effects.

What are the greatest implications of this review?

The greatest implications of this review are:

Potential for New Therapeutics: Understanding the microbiota–gut–brain axis could lead to novel therapeutic strategies targeting the gut microbiome to treat or prevent neurological disorders. This could involve probiotics, prebiotics, dietary interventions, or fecal microbiota transplantation.

Need for Longitudinal Studies: Many of the findings are based on cross-sectional studies, which provide a snapshot in time but do not establish causality. There is a need for longitudinal cohort studies and randomized controlled trials to better understand how microbiota changes over time in relation to disease progression and treatment response.

Precision Medicine: Integrating microbiome data with other omics (genomics, metabolomics) could help tailor treatments to individual patients based on their microbiota composition, potentially enhancing the effectiveness of therapies for neurological disorders.

Holistic Understanding of Neurological Diseases: The review highlights the importance of considering the gut microbiome as an integral part of understanding and managing neurological diseases, potentially shifting paradigms in neurology towards a more comprehensive systems biology approach.

Integration of Systems Biology: The authors underscore the importance of integrating microbiome research with genomic, metabolomic, and other multiomic data to understand the mechanisms underlying the microbiota-gut-brain axis fully. This approach could lead to the identification of biomarkers and the development of more precise interventions.

Potential for Preventive Measures: The review suggests that modulating the microbiome early in life or during the aging process could serve as a preventive strategy against cognitive decline and other neurological disorders. This could shift the focus from treating diseases after they manifest to preventing them through microbiome management.

Conclusion from the Authors

Recent advances have highlighted the critical role that the gut microbiota plays in both the development and maintenance of brain function. Evidence from a growing body of clinical and animal research strongly supports the involvement of the microbiota in neurological disorders such as Parkinson’s disease, multiple sclerosis, and autism spectrum disorder, with emerging insights into its role in Alzheimer’s disease and stroke. However, the field remains in its early stages, and researchers must exercise caution in interpreting these findings. Small sample sizes, methodological inconsistencies, and potential biases often limit the current body of work. To move forward, there is a pressing need for well-designed, large-scale studies that can accurately elucidate the complex relationships within the microbiota-gut-brain axis.

Future research must shift from observational studies to those that can establish causality and explore functional outcomes. This necessitates a greater emphasis on interventional approaches, such as the use of probiotics, prebiotics, and fecal microbiota transplantation, in longitudinal studies. Such approaches should aim to identify the microbiota as a biomarker of disease and test the efficacy of microbiota-targeted therapies in clinical populations.

Given the considerable interindividual variability in microbiota composition, a significant challenge lies in defining what constitutes a "healthy" microbiome. This variability complicates efforts to develop standardized therapeutic approaches. Nevertheless, it also opens the door to personalized medicine, where treatments are tailored to the individual’s unique microbiome profile. To advance this goal, researchers must delve deeper into the microbial ecosystem, beyond just bacterial genera, employing metagenomic and multi-omic techniques to understand the full spectrum of microbial influence on brain health.

Additionally, expanding research to include other components of the microbiome, such as viruses and bacteriophages, will be crucial to gaining a comprehensive understanding of their role in brain function. Investigating the interaction between host genetics and the microbiome is another underexplored area that holds promise for uncovering the biological mechanisms underlying neurological disorders. Systems biology approaches will be vital for integrating these diverse data streams and providing a holistic view of microbiota-gut-brain interactions.

Diet remains a major factor influencing microbiota composition, especially in the context of neurological disorders that affect nutritional intake. Understanding the relationship between diet, microbiota, and brain health will be key to developing dietary interventions that support neurological health throughout life. As research progresses, the influence of dietary components and microbial metabolites on health will likely become a central focus in the quest to develop microbiota-based therapies.

The interaction between medications and the microbiota is an emerging area of interest, as recent studies indicate that a substantial number of drugs can alter the gut microbiome. This interaction has significant implications for drug efficacy and safety, underscoring the need for further investigation. As we continue to explore these complex relationships, the next five years of research will be pivotal in determining how the microbiota can be harnessed to develop effective therapies for neurological disorders.

Image modified from the original "The gut microbiome in neurological disorders"
Figure 2: Pathways of communication between the microbiota and the brain

The gut microbiota–brain axis in neurological disorder

May 20, 2025
  • Brain Health
    Brain Health

    Brain health encompasses the overall functioning and well-being of the brain, including cognitive function, emotional and psychological well-being, neurological integrity, behavioral health, neurodevelopmental health, age-related brain health, and brain resilience and plasticity.

This review highlights the critical role of gut microbiota in brain health and neurological disorders, suggesting that microbiome-targeted interventions (MBTIs) could revolutionize the treatment and management of these conditions. However, further research is needed to fully understand the mechanisms involved and to develop safe and effective microbiome-targeted therapies.

What was reviewed?

The review paper provides a comprehensive overview of the gut microbiota-brain axis (GBA) and its role in various neurological disorders. Specifically, the paper examined the intricate bidirectional communication between the gut microbiota (GM) and the central nervous system (CNS), exploring how dysbiosis (an imbalance in the gut microbiota) contributes to the pathogenesis of neurological conditions such as Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), autism spectrum disorder (ASD), anxiety, depression, and stroke. The review also delved into the mechanisms by which gut microbiota influence brain health, including neurotransmitter production, endocrine signaling, immune modulation, and neuronal pathways.

Who was reviewed?

The review synthesized findings from a broad range of preclinical and clinical studies, drawing upon research involving animal models, human subjects, and in vitro experiments. The paper reviewed studies that have investigated the composition of gut microbiota in individuals with neurological disorders compared to healthy controls, as well as studies that explored the mechanistic pathways connecting gut microbiota with brain function. Additionally, the review considered research on potential microbiome-targeted interventions (MBTIs) such as probiotics, prebiotics, and dietary modifications that could influence neurological health.

What were the most important findings of this review?

The review highlighted several key findings:

Bidirectional Communication of the Gut-Brain Axis: The gut-brain axis (GBA) operates through complex bidirectional communication involving multiple pathways, including neural, endocrine, immune, and metabolic routes. These pathways allow gut microbiota to influence brain function and, conversely, enable the brain to affect gastrointestinal processes.

Microbiota Dysbiosis and Neurological Disorders: Dysbiosis, or alterations in the gut microbiota composition, is consistently associated with several neurological disorders. For instance, individuals with AD, PD, MS, ASD, and mood disorders exhibit distinct microbiota profiles compared to healthy controls, including reduced diversity and imbalances in specific microbial taxa.

Mechanistic Pathways: The review detailed how gut microbiota impact brain health through various mechanisms:

Neurotransmitters: Gut microbes produce and modulate key neurotransmitters such as serotonin, dopamine, and GABA, which are crucial for CNS function.

Endocrine Signaling: Short-chain fatty acids (SCFAs) produced by gut bacteria influence the release of gut hormones like GLP-1 and PYY, which affect mood, memory, and learning.

Immune Modulation: Gut microbiota influence the immune system, affecting neuroinflammation and playing a role in the pathogenesis of psychiatric and neurodegenerative diseases.

Neuronal Pathways: The vagus nerve serves as a direct communication route between the gut and the brain, with microbial metabolites potentially activating neurons.

What are the greatest implications of this review?

The review has several significant implications:

Personalized Medicine: The variability in gut microbiota among individuals suggests that personalized approaches to treating neurological disorders could be more effective. Tailoring interventions based on an individual's microbiota profile could optimize therapeutic outcomes.

Microbiome as a Therapeutic Target: The findings underscore the potential of microbiome-targeted interventions (MBTIs) in treating neurological disorders. Probiotics, prebiotics, dietary changes, and possibly even fecal microbiota transplants (FMT) could be developed as therapeutic strategies to restore gut microbiota balance and improve neurological outcomes.

Development of Biomarkers: The distinct microbial profiles observed in various neurological disorders suggest that gut microbiota composition could serve as a biomarker for early diagnosis, prognosis, and monitoring of treatment responses in these conditions.

Mechanistic Insights into Neurological Disorders: By elucidating the mechanisms through which gut microbiota influence brain function, the review opens new avenues for understanding the pathophysiology of neurological disorders. This knowledge could lead to more targeted and effective treatments that address the underlying causes of these diseases.

Interdisciplinary Research and Clinical Translation: The review highlights the need for continued collaboration between microbiology, neuroscience, immunology, and clinical research to translate these findings into practical applications. Developing effective MBTIs requires a deep understanding of the gut-brain axis, which can only be achieved through interdisciplinary research.

The therapeutic role of minocycline in Parkinson’s disease

May 20, 2025
  • Parkinson’s Disease
    Parkinson’s Disease

    OverviewParkinson’s disease (PD) is a neurodegenerative disorder primarily characterized by the degeneration of dopaminergic neurons in the nigrostriatal pathway, leading to progressive hypokinetic movements [1], and a range of non-motor symptoms including gastrointestinal (GI) dysfunction [2]. Emerging evidence suggests that the gut microbiome may influence PD through the gut–brain axis. PD is one of the […]

  • Brain Health
    Brain Health

    Brain health encompasses the overall functioning and well-being of the brain, including cognitive function, emotional and psychological well-being, neurological integrity, behavioral health, neurodevelopmental health, age-related brain health, and brain resilience and plasticity.

The review by Cankaya et al. explores minocycline’s neuroprotective potential in Parkinson's disease (PD), highlighting its anti-inflammatory, antioxidant, and anti-apoptotic effects. While preclinical studies show promising neuroprotective results, clinical trials have yet to confirm its efficacy in slowing PD progression. The review emphasizes the need for further research to validate minocycline as a therapeutic agent for PD and other neurodegenerative disorders.

What Was Reviewed?

The review focused on the potential therapeutic role of minocycline, a semisynthetic tetracycline-derived antibiotic, in Parkinson’s disease (PD). It encompassed both preclinical and clinical studies to evaluate minocycline’s neuroprotective effects and its mechanisms of action in various experimental models of neurodegenerative diseases, including cerebral ischemia, traumatic brain injury, amyotrophic lateral sclerosis (ALS), Huntington’s disease (HD), multiple sclerosis (MS), and specifically Parkinson’s disease (PD).

Who Was Reviewed?

The review analyzed data from multiple sources, including:

Animal Models: Various in vivo and in vitro studies were conducted on rodents and other animals to observe the neuroprotective effects of minocycline.

Clinical Trials: Human studies, including randomized, double-blind clinical trials, that assessed the efficacy of minocycline in treating PD and other neurodegenerative disorders.

Literature Reviews: Retrospectively recorded results from studies available on databases such as PubMed, Scopus, and ISI Web of Science, focusing on keywords like “minocycline and Parkinson’s disease,” “minocycline and neuroprotection,” “minocycline,” and “neurodegeneration.”

Most Important Findings

Anti-Inflammatory Effects: Minocycline modulates microglia activation, reduces the release of proinflammatory cytokines, and inhibits pathways leading to neuroinflammation. This helps in attenuating neuroinflammation, a critical aspect of PD pathogenesis.

Antioxidant Effects: The drug reduces oxidative stress by inhibiting the production of reactive oxygen species (ROS) and stabilizing mitochondrial function.

Anti-Apoptotic Effects: Minocycline inhibits apoptotic pathways by stabilizing mitochondrial membranes, reducing the release of cytochrome c, and modulating the expression of B-cell lymphoma 2 (Bcl-2) proteins, thereby preventing neuronal cell death.

Efficacy in Experimental Models:

Parkinson’s Disease Models: In MPTP and 6-OHDA-induced PD models, minocycline reduced dopaminergic neuron degeneration and improved behavioral deficits.

Other Neurodegenerative Models: Minocycline demonstrated neuroprotective effects in models of ALS, HD, and MS by inhibiting microglial activation and reducing neuronal death.

Greatest Implications

The therapeutic role of minocycline in Parkinson’s disease as a Potential Neuroprotective Agent: Despite mixed results in clinical trials, the extensive preclinical data support the potential of minocycline as a neuroprotective agent that could modify disease progression in PD and possibly other neurodegenerative diseases. This highlights the need for further research and well-designed clinical trials to determine its efficacy conclusively.

Mechanistic Insights: The review provides a comprehensive understanding of the mechanisms through which minocycline exerts its effects, such as anti-inflammatory, antioxidant, and anti-apoptotic pathways. This knowledge could be pivotal in developing targeted therapies for neurodegenerative diseases.

Future Research Directions: The findings underline the importance of exploring combination therapies that might enhance the efficacy of minocycline. Additionally, investigating different dosing regimens, treatment durations, and patient populations could yield more definitive results regarding its therapeutic potential.

Broader Implications for Neurodegenerative Diseases: The review suggests that minocycline's therapeutic benefits might extend beyond PD to other conditions like ALS, HD, and MS, making it a promising candidate for broader neuroprotective applications.

Polycystic ovary syndrome (PCOS)

Go to Category Page

Evaluation of the relationship between polycystic ovary syndrome and intestinal inflammation as measured by fecal calprotectin levels

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

PCOS is linked with higher gut inflammation, as shown by elevated fecal calprotectin. This may help guide diagnosis and treatment.

What was studied?

The authors investigated whether intestinal inflammation plays a role in polycystic ovary syndrome (PCOS) by analyzing fecal calprotectin levels, a noninvasive biomarker that reflects neutrophil-driven gut inflammation. Given the increasing evidence that inflammation and gut dysbiosis contribute to PCOS, the researchers aimed to determine if elevated calprotectin could serve as an additional indicator of disease presence or severity.

Who was studied?

The study included 54 adult women: 27 with PCOS and 27 healthy controls. All participants were of reproductive age and had a body mass index within the normal range. The authors excluded individuals with gastrointestinal disorders, systemic illness, or recent antibiotic use to isolate the relationship between PCOS and gut inflammation.

What were the most important findings?

Women with PCOS had significantly higher fecal calprotectin levels compared to healthy controls, suggesting greater intestinal inflammation. Interestingly, systemic inflammation, measured by standard markers like hs-CRP, was similar across groups, indicating that the inflammation in PCOS may be localized to the gut. Although calprotectin wasn’t an independent predictor of PCOS in statistical models, it showed excellent specificity. This means it could help differentiate between PCOS and non-PCOS individuals in clinical settings. The findings support that microbial shifts and increased intestinal permeability—hallmarks of gut dysbiosis—may underlie some aspects of PCOS. Elevated calprotectin levels point toward neutrophil activity in the intestinal lining, often triggered by changes in gut microbiota.

What are the greatest implications of this study?

The study underscores the potential role of intestinal inflammation in PCOS and highlights fecal calprotectin as a promising, low-cost marker that could aid diagnosis or monitoring. These findings open the door to new interventions, such as microbiome-targeted therapies, to manage PCOS symptoms. If confirmed in future studies, strategies that reduce gut inflammation might improve hormonal balance and fertility outcomes in PCOS patients. It also reinforces the value of including microbiome-related biomarkers in gynecological evaluations.

Evidence for statin therapy in polycystic ovary syndrome. 

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

This review highlights the potential benefits of statin therapy in women with polycystic ovary syndrome (PCOS), showing improvements in metabolic dysfunction, hyperandrogenism, and cardiovascular risk factors. Statins may offer a dual benefit for PCOS management, though further research is needed for long-term clinical outcomes.

What was reviewed?

This paper provides a review of the evidence supporting statin therapy for managing polycystic ovary syndrome (PCOS), focusing on its potential to reduce cardiovascular risks and address some of the metabolic complications associated with PCOS. The review discusses both the lipid-lowering effects of statins and their pleiotropic effects, including improvements in insulin resistance, hyperandrogenemia, and systemic inflammation. These secondary benefits may offer additional therapeutic value for women with PCOS, a condition commonly linked with metabolic and cardiovascular disturbances.

Who was reviewed?

The review examined existing studies and clinical trials investigating the use of statins in PCOS patients. The studies reviewed explored the effectiveness of statins like atorvastatin and simvastatin in reducing various metabolic and biochemical markers in women with PCOS, such as testosterone levels, insulin resistance, and inflammation. The review focused on understanding how statins could be beneficial in managing the hormonal and metabolic dysfunctions seen in PCOS.

What were the most important findings?

The review revealed promising evidence supporting statin therapy for women with PCOS, particularly due to its pleiotropic effects. Statins were shown to improve the lipid profile in women with PCOS, reducing LDL cholesterol levels, which is crucial given the elevated cardiovascular risks associated with the condition. Beyond lipid-lowering, statins also contributed to significant reductions in hyperandrogenemia, insulin resistance, and markers of systemic inflammation such as C-reactive protein (CRP). These findings suggest that statins could offer a dual benefit by improving both metabolic and reproductive parameters in women with PCOS.

Moreover, the review highlighted that statins like atorvastatin and simvastatin have comparable effects on testosterone reduction, an important aspect of managing hyperandrogenism in PCOS. The use of statins led to a decrease in testosterone levels that was similar to the effects of established antiandrogens. This effect was observed independently of the improvement in lipid profiles, which underscores the potential of statins to address some of the hormonal imbalances seen in PCOS.

What are the greatest implications of this review?

The findings from this review suggest that statins may be a valuable addition to the treatment options for PCOS, especially for women who are at high risk of cardiovascular disease due to the metabolic disturbances commonly seen in the condition. The reduction in hyperandrogenemia, improvement in insulin sensitivity, and decrease in inflammation could provide significant therapeutic benefits, particularly for those who have not responded well to other treatments like insulin sensitizers or antiandrogens. However, the review also emphasized the need for further large-scale studies to validate the long-term efficacy of statins in improving fertility outcomes and reducing cardiovascular events in women with PCOS. The potential teratogenic risks of statins, particularly during pregnancy, warrant caution and a careful approach to their use in reproductive-age women.

Metformin use in women with polycystic ovary syndrome

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

This review discusses the role of metformin in treating PCOS-related infertility. It highlights its effectiveness in improving ovulation, reducing insulin resistance, and managing hyperandrogenism. Metformin is recommended as a first-line treatment for non-obese women with PCOS-related infertility, with benefits also seen in IVF and metabolic health.

What was reviewed?

This review examines the role of metformin in managing polycystic ovary syndrome (PCOS), specifically focusing on its effectiveness in treating PCOS-related infertility. The paper reviews evidence from randomized controlled trials (RCTs) and other studies, discussing the use of metformin as an insulin-sensitizing agent for women with PCOS who experience anovulatory infertility. It also explores metformin’s impact on metabolic dysfunctions, hyperandrogenism, and its potential use alongside other treatments like clomiphene for improving fertility outcomes in women with PCOS.

Who was reviewed?

The review considers various studies and clinical trials on the use of metformin in women with PCOS. These studies involve women with varying degrees of obesity and insulin resistance, who are experiencing anovulatory infertility, hyperandrogenism, or both. The review synthesizes results from RCTs that examined the effectiveness of metformin alone or in combination with other treatments like clomiphene citrate and aromatase inhibitors in improving ovulation, fertility, and reducing the metabolic disturbances associated with PCOS.

What were the most important findings?

The review highlights several key findings regarding the use of metformin in treating PCOS-related infertility. Metformin has shown efficacy in improving ovulation rates in women with anovulatory infertility, particularly in non-obese women. A Cochrane review of seven RCTs revealed that metformin significantly increased clinical pregnancy rates compared to placebo. However, while metformin showed promise, it did not outperform clomiphene citrate as a first-line treatment for ovulation induction in women with PCOS, particularly in obese patients. The review also found that metformin, when used in combination with clomiphene, can be effective for women who are resistant to clomiphene alone.

Additionally, the review emphasized that metformin has benefits beyond fertility induction. It helps reduce hyperinsulinemia and insulin resistance, which are common in women with PCOS, and can improve associated metabolic conditions such as dyslipidemia and obesity. Furthermore, metformin was found to reduce the risk of ovarian hyperstimulation syndrome (OHSS) in women undergoing in vitro fertilization (IVF). Although metformin’s role in improving long-term health outcomes, such as the prevention of type 2 diabetes, cardiovascular disease, and endometrial cancer, remains inconclusive, it offers significant short-term reproductive benefits.

What are the greatest implications of this review?

The review suggests that metformin should be considered a suitable first-line treatment for non-obese women with anovulatory infertility due to PCOS. For women who are resistant to clomiphene or prefer an alternative to the oral contraceptive pill (OCP) for managing hyperandrogenic symptoms, metformin can be an effective option. Additionally, metformin’s role in reducing the risk of OHSS during IVF procedures underscores its importance in assisted reproductive treatments. The review also raises the need for further research to better define metformin’s long-term benefits in preventing the metabolic and reproductive complications associated with PCOS, as well as its potential to improve long-term health outcomes like diabetes prevention.

Role of Metformin in Polycystic Ovary Syndrome (PCOS)-Related Infertility

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

This review examines the role of metformin in treating PCOS-related infertility, highlighting its effectiveness in improving insulin sensitivity, reducing hyperandrogenism, and restoring ovulation. Metformin serves as a first-line therapy, offering significant benefits for women with anovulatory infertility due to PCOS.

What was reviewed?

This review explores the role of metformin in treating polycystic ovary syndrome (PCOS)-related infertility. PCOS is a common endocrinological disorder that can lead to infertility, characterized by insulin resistance, hyperandrogenism, and anovulation. The review discusses metformin’s mechanisms, its impact on insulin sensitivity, its role in improving ovulation, and its effectiveness in managing metabolic and hormonal imbalances in women with PCOS. The review also emphasizes the drug's benefits in improving menstrual cyclicity and reducing hyperandrogenism, ultimately aiding in fertility restoration.

Who was reviewed?

The review synthesizes findings from various clinical studies and trials examining the effects of metformin on women with PCOS. It draws on observational studies and randomized controlled trials to evaluate the efficacy of metformin in addressing infertility associated with PCOS. The women studied in these trials typically had anovulatory infertility, hyperandrogenism, and varying degrees of insulin resistance, and they were treated with metformin to assess its impact on ovulation and fertility.

What were the most important findings?

The review found that metformin has significant therapeutic benefits for women with PCOS, particularly in restoring menstrual regularity and improving ovulation rates. Metformin works primarily by improving insulin sensitivity, which reduces hyperinsulinemia—a key factor in the pathogenesis of PCOS. This insulin-sensitizing effect contributes to lower circulating androgen levels, which is crucial in managing symptoms like hirsutism and acne. In several studies, metformin, either alone or in combination with other treatments like clomifene citrate, successfully induced ovulation in women who were resistant to standard treatments.

Furthermore, metformin appears to improve metabolic dysfunctions common in PCOS, including insulin resistance, dyslipidemia, and obesity, all of which contribute to the infertility and long-term health risks associated with the condition. However, the review also noted that while metformin improves metabolic and reproductive outcomes, its efficacy in women with significant obesity is less pronounced. The review also highlights that metformin is generally well-tolerated, although some women may experience gastrointestinal side effects.

What are the greatest implications of this review?

The review underscores metformin’s potential as a first-line treatment for women with PCOS-related infertility, especially for those who are insulin-resistant and non-obese. The findings suggest that metformin could be a safer and more accessible alternative to more invasive fertility treatments like in vitro fertilization (IVF). Moreover, metformin’s role in reducing the risk of ovarian hyperstimulation syndrome during assisted reproductive technology procedures makes it particularly valuable in IVF protocols. The review also emphasizes the need for further studies to determine the optimal dose and long-term benefits of metformin, particularly for women with more severe obesity or metabolic complications.

Usefulness of intermittent clomiphene citrate treatment for women with polycystic ovarian syndrome that is resistant to standard clomiphene citrate treatment

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Polycystic ovary syndrome (PCOS)
    Polycystic ovary syndrome (PCOS)

    Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age, characterized by irregular menstrual cycles, hyperandrogenism, and insulin resistance. It is often associated with metabolic dysfunctions and inflammation, leading to fertility issues and increased risk of type 2 diabetes and cardiovascular disease.

The study explored intermittent clomiphene citrate treatment (ICT) for women with PCOS who were resistant to standard CC treatment. ICT showed an 80.8% response rate, offering an effective and safer alternative to gonadotropin therapy with no risk of OHSS or multiple pregnancies.

What was studied?

The study investigated the efficacy of intermittent clomiphene citrate treatment (ICT) in women with polycystic ovary syndrome (PCOS) who were resistant to standard clomiphene citrate (CC) treatment. Clomiphene citrate is commonly used to induce ovulation in women with PCOS; however, some women do not respond to the standard treatment. This research aimed to assess whether a modified dosing schedule, using intermittent CC treatment, could help induce follicular growth in these resistant patients.

Who was studied?

The study focused on 42 infertile women diagnosed with PCOS who were resistant to standard CC treatment. Of these, 26 women underwent the intermittent CC treatment protocol, and their response to the treatment was monitored and analyzed. The patient cohort was selected based on their non-response to the standard 50 mg/day, 5-day CC regimen, which is typically used as a first-line treatment for anovulatory PCOS.

What were the most important findings?

The study found that intermittent CC treatment was highly effective for women who were resistant to standard CC treatment. The protocol involved administering 100 mg of CC for 5-day periods across different phases of the menstrual cycle. The results showed that 80.8% of the patients who had previously been resistant to standard CC treatment responded positively to ICT, with follicular growth observed in many of the cases. Notably, the majority of those who responded had only a single mature follicle, which is a safer outcome in terms of reducing the risk of multiple pregnancies or ovarian hyperstimulation syndrome (OHSS). Additionally, no cases of OHSS or multiple pregnancies were reported, making ICT a promising alternative to gonadotropin therapy, which carries higher risks and requires more intensive treatment.

The efficacy of ICT was particularly notable in patients resistant to the 100 mg dose of CC, where up to 78.2% responded positively after subsequent doses. This suggests that ICT may be more effective than simply increasing the dose of standard CC, providing a less invasive and lower-risk alternative for patients who fail the initial treatment.

What are the greatest implications of this study?

The findings suggest that ICT could become a preferred treatment option for women with CC-resistant PCOS before advancing to gonadotropin therapy. ICT offers several advantages: it is less invasive, more cost-effective, and poses a lower risk of complications like OHSS and multiple pregnancies. By maintaining high serum FSH levels through repeated administration of CC, ICT seems to trigger follicular growth effectively without the need for prolonged or more invasive treatments. The study's outcomes could help reshape the treatment protocols for PCOS, offering a safer, more accessible alternative to gonadotropin therapy. However, further research with larger patient populations is necessary to validate these findings and assess the long-term efficacy of ICT in achieving successful pregnancy outcomes.

STOPs

Go to Category Page

Exploring the link between dietary zinc intake and endometriosis risk: insights from a cross-sectional analysis of American women

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • STOPs
    STOPs

    A STOP (Suggested Termination Of Practices) is a recommendation that advocates for the discontinuation of certain medical interventions, treatments, or practices based on emerging evidence indicating that these may be ineffective, harmful, or counterproductive in the management of specific conditions.

This study links higher dietary zinc intake with increased endometriosis risk among American women, highlighting zinc’s complex role in immune modulation and estrogen-related pathways. Findings emphasize the importance of balanced intake for managing endometriosis risk.

What was studied?

This study investigated the association between dietary zinc intake and the risk of endometriosis among American women. Using cross-sectional data from the National Health and Nutrition Examination Survey (NHANES) collected between 1999 and 2006, the researchers aimed to evaluate whether zinc intake, as a key nutritional factor, was linked to the prevalence of endometriosis. Zinc is known for its essential roles in immune modulation, antioxidative defense, and regulation of matrix metalloproteinases (MMPs), all of which are implicated in endometriosis progression.

Who was studied?

The study included 4,315 American women aged 20–54 years, of whom 331 were diagnosed with endometriosis based on self-reported doctor diagnoses. Participants’ dietary zinc intake was assessed using 24-hour dietary recall interviews, with additional data on demographics, lifestyle, and health covariates collected. Women with extreme caloric intakes or incomplete data were excluded to ensure robustness of results.

What were the most important findings?

The study revealed a positive correlation between higher dietary zinc intake and the risk of endometriosis. Women consuming over 14 mg/day of zinc had a significantly higher adjusted odds ratio (1.60, 95% CI: 1.12–2.27, p = 0.009) compared to those with intake ≤8 mg/day. Zinc’s dual role in immune modulation and antioxidative defense was emphasized, particularly its regulation of matrix metalloproteinases (MMPs) like MMP-2 and MMP-9, which are key enzymes in tissue remodeling and endometriotic lesion invasion. Interestingly, despite zinc’s known antioxidative and anti-inflammatory roles, excessive intake appeared to have a counterproductive effect. These nuanced findings highlight zinc’s complex role in endometriosis pathophysiology.

What are the greatest implications of this study?

This research underscores the potential for dietary zinc as both a marker and modifiable factor in endometriosis risk. It raises questions about zinc’s dualistic effects, where optimal levels may support immune health, but excess intake could exacerbate estrogen-related pathways in endometriosis. Clinicians should be cautious when recommending zinc supplementation for reproductive health, particularly in populations at risk for endometriosis. Furthermore, this study strengthens the biological plausibility of microbiome involvement in endometriosis, as zinc is a crucial cofactor for microbial activity, and its imbalance may alter the gut and pelvic microbiota implicated in the disease.

Fruit and vegetable consumption and risk of endometriosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • STOPs
    STOPs

    A STOP (Suggested Termination Of Practices) is a recommendation that advocates for the discontinuation of certain medical interventions, treatments, or practices based on emerging evidence indicating that these may be ineffective, harmful, or counterproductive in the management of specific conditions.

Higher fruit intake, especially citrus fruits, was inversely associated with laparoscopically confirmed endometriosis, suggesting a protective effect potentially linked to beta-cryptoxanthin. In contrast, cruciferous vegetables were linked to increased risk, highlighting the complex interplay between diet and endometriosis risk factors.

What Was Studied

This study explored the potential link between the consumption of fruits and vegetables and the risk of laparoscopically confirmed endometriosis. Using data collected from the Nurses' Health Study II, the researchers analyzed dietary habits over a 22-year period, investigating whether certain food groups and nutrients influenced the likelihood of developing endometriosis.

Who Was Studied

Participants included premenopausal women aged 25–42 years who were enrolled in the Nurses' Health Study II cohort. These women completed biennial surveys assessing health status, lifestyle factors, and dietary intake. Those with a history of endometriosis, cancer, infertility, or hysterectomy were excluded from the analysis, ensuring a focused evaluation of diet and disease development.

Most Important Findings

The study found an inverse relationship between fruit consumption, particularly citrus fruits, and the risk of endometriosis. Women who consumed citrus fruits frequently were less likely to develop endometriosis. Conversely, no significant association was found between total vegetable intake and the disease. Cruciferous vegetables, however, were unexpectedly linked to an increased risk. Beta-cryptoxanthin, a nutrient found in citrus fruits, appeared to play a protective role, and the beneficial effects of fruit consumption were especially notable among participants who had a history of smoking. These findings suggest a potential role for specific dietary components in either mitigating or exacerbating the risk of endometriosis.

Implications

The findings highlight the importance of dietary considerations in understanding endometriosis risk. The protective association of citrus fruits underscores the potential of targeted nutritional interventions to reduce risk. The increased risk observed with cruciferous vegetables raises questions about the role of gastrointestinal symptoms, as these vegetables are high in fermentable oligosaccharides, which could exacerbate symptoms and lead to increased diagnosis rates. Future studies exploring these dietary patterns in greater depth are warranted to clarify the underlying mechanisms and to guide dietary recommendations for those at risk.

Molecular detection of intrauterine microbial colonization in women with endometriosis

May 20, 2025
  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • STOPs
    STOPs

    A STOP (Suggested Termination Of Practices) is a recommendation that advocates for the discontinuation of certain medical interventions, treatments, or practices based on emerging evidence indicating that these may be ineffective, harmful, or counterproductive in the management of specific conditions.

A case-controlled molecular study on 32 women, half with endometriosis and half without, investigated microbial colonization in the intrauterine environment and ovarian cystic fluid. It found significant bacterial variations, with certain bacteria types increasing or decreasing, particularly after treatment with gonadotropin-releasing hormone agonist (GnRHa). The study suggests GnRHa treatment might promote sub-clinical infections in the intrauterine and ovarian environments.

What was studied?

The research focused on investigating microbial colonization in women’s intrauterine environment and cystic fluid, utilizing a molecular approach to detect bacterial presence. This involved examining variations in bacterial types and their implications in the context of endometriosis and treatment effects.

Who was studied?

The study included 32 women, evenly divided between those diagnosed with endometriosis and those without the condition. Each group was further split, with half receiving gonadotropin-releasing hormone agonist (GnRHa) treatment, to explore the treatment’s impact on microbial colonization.

What were the most important findings?

Key findings revealed a broad range of bacterial presence in both endometrial swabs and cystic fluids, with significant changes in bacterial families (decrease in Lactobacillacae and increase in Streptococcaceae, Staphylococaceae, and Enterobacteriaceae) observed in GnRHa-treated women with endometriosis. The 16S metagenome assay was more effective than traditional culture methods, particularly in identifying bacteria in ovarian endometrioma cystic fluid.

What are the greatest implications of this study?

The study’s findings suggest the presence of sub-clinical infections in the intrauterine environment and ovarian endometrioma cystic fluid, particularly following GnRHa treatment. This raises concerns about the potential for GnRHa therapy to promote silent infections, indicating a need for careful consideration and monitoring of such treatments in women with endometriosis.

Anemia

Go to Category Page

How Severe Anemia Might Influence the Risk of Invasive Bacterial Infections in African Children

May 20, 2025
  • Anemia
    Anemia

    Anemia is a reduction in red blood cells or hemoglobin, often influenced by the gut microbiome's impact on nutrient absorption.

The hypothesis presented in the study is that severe anemia may contribute to the risk of invasive bacterial infections in African children through dysregulation of iron homeostasis and/or iron-regulatory proteins, particularly by affecting the regulation of the hepatic hormone hepcidin and subsequent iron availability for bacterial growth. This hypothesis, therefore, not only opens new avenues for research into the pathophysiology of anemia and bacterial infections but also for developing better therapeutic interventions that could reduce morbidity and mortality in those affected by these conditions.

What Was Reviewed?

This review investigates the hypothesis that severe anemia contributes to the elevated risk of invasive bacterial infections in African children through dysregulation of iron homeostasis, including the disruption of hepcidin—a hepatic hormone that governs iron availability in the body. The authors synthesize evidence from epidemiological, mechanistic, and experimental studies, proposing that the interplay between anemia, haemolysis, immune dysfunction, and gut permeability collectively facilitates systemic infections. The paper places particular emphasis on how bacterial pathogens exploit iron and haem to thrive under conditions of anemia-induced iron dysregulation, especially in regions with high burdens of infectious disease and malnutrition.

Who Was Reviewed?

The review draws on research concerning sub-Saharan African children who commonly experience severe anemia due to malaria, nutritional deficiencies, sickle cell disease, and HIV. It includes human observational data, animal model findings, and in vitro studies related to bacterial infections, immune response, and iron regulation pathways.

What Were the Most Important Findings?

Severe anemia in African children correlates strongly with an increased risk of invasive bacterial infections, particularly with Gram-negative organisms such as non-typhoidal Salmonella (NTS), E. coli, and Haemophilus influenzae, as well as Gram-positive organisms including Staphylococcus aureus and Streptococcus pneumoniae. This elevated susceptibility is mechanistically linked to increased erythropoietic drive and haemolysis, both of which suppress hepcidin via the erythroid hormone erythroferrone (ERFE). Reduced hepcidin enhances plasma iron levels and promotes the release of iron from macrophage stores, thereby removing the “nutritional immunity” that would otherwise limit iron availability to pathogens. Simultaneously, haemolysis releases non-transferrin-bound iron (NTBI) and free haem, which are more readily exploited by pathogens through siderophore-mediated and haem-binding iron acquisition systems.

Of particular relevance to microbiome researchers, the review notes that severe anemia can disrupt gut barrier integrity and provoke dysbiosis. The increased gut permeability facilitates the translocation of enteric pathogens, notably NTS and E. coli, into systemic circulation. This breach is exacerbated by inflammation, destabilization of tight junction proteins like ZO-1, and macrophage-mediated changes to mucosal immunity. The review references mouse models where anemia-induced epithelial dysfunction was dependent on macrophage-driven cytokine signaling, especially IFN-γ, as well as bacterial studies showing enhanced virulence and iron uptake capacity in haem-rich or iron-rich conditions. Importantly, these microbial strategies overlap with the exact niches disrupted in severe anemia, such as haem overload, NTBI availability, and compromised mucosal defenses.

Major microbial associations (MMAs) include:

PathogenKey Iron Acquisition Strategies
Non-typhoidal SalmonellaSiderophores (salmochelin, enterobactin), Fe2+ uptake via ferroportin Strong association with anemia.
E. coliEnterobactin, salmochelin, aerobactin, haem receptors. Strong association with anemia.
Haemophilus influenzaeHaem- and haemoglobin-binding proteins (HgpA/B/C, HxuA). Moderate association with anemia.
Staphylococcus aureusIsd system, staphyloferrin siderophores, transferrin binding. Moderate association with anemia.
Streptococcus pneumoniaeABC transporters (piu, pia, pit), haemoglobin-binding proteins. Moderate association with anemia.

What Are the Greatest Implications of This Review?

The review suggests that treating severe anemia in high-infection-burden areas like sub-Saharan Africa should involve caution, particularly regarding iron supplementation. While iron repletion is essential, excessive or unregulated iron can exacerbate infection risk, especially in the presence of low hepcidin levels. This carries profound public health implications, as many iron supplementation programs do not account for concurrent infectious burdens or the child's hepcidin status. Clinically, these insights demand a reevaluation of iron therapy protocols, particularly in settings where malaria, HIV, or bacterial sepsis are endemic. The review also encourages further exploration of therapies that modulate iron availability (e.g., hepcidin agonists or iron chelators) and highlights the need for comprehensive microbiome assessments in anaemic populations. The tight interconnection between gut microbiota, intestinal permeability, and systemic iron overload represents a mechanistic intersection worth pursuing in microbiome-targeted interventions.

Tinidazole

Go to Category Page

Improved cure of bacterial vaginosis with single dose of tinidazole (2g), Lactobacillus rhamnosus GR-1, and Lactobacillus reuteri RC-14

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Bacterial Vaginosis
    Bacterial Vaginosis

    Bacterial vaginosis (BV) is caused by an imbalance in the vaginal microbiota, where the typically dominant Lactobacillus species are significantly reduced, leading to an overgrowth of anaerobic and facultative bacteria.

  • Tinidazole
    Tinidazole

    Tinidazole is a nitroimidazole antimicrobial that selectively targets anaerobic bacteria and protozoa, reshaping the gut ecosystem by depleting pathogenic anaerobes while preserving commensals. Clinically validated for giardiasis, bacterial vaginosis, and colorectal surgery prophylaxis. Its DNA-disrupting and biofilm-penetrating actions reduce inflammatory triggers and create niches for healthy microbiota to rebound.

This study shows that oral probiotics significantly boost cure rates of bacterial vaginosis when used with tinidazole, restoring a healthy vaginal microbiome.

What Was Studied?

This randomized, double-blind, placebo-controlled clinical trial investigated whether the combination of a single 2g dose of tinidazole and daily oral probiotics (Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14) improved bacterial vaginosis (BV) cure rates in women, compared to tinidazole alone. BV, a condition characterized by dysbiosis in the vaginal microbiome and reduction of protective Lactobacillus species, has shown poor long-term response to antibiotic treatment alone. The study aimed to determine if probiotic supplementation could enhance the therapeutic efficacy of tinidazole by restoring a more favorable microbial balance.

Who Was Studied?

Sixty-four Brazilian women diagnosed with BV based on Amsel’s criteria and Nugent scoring participated in the study. The participants were randomly assigned to either a placebo group or a probiotic group, both of which received the same tinidazole dose. The probiotic group also received daily capsules containing L. rhamnosus GR-1 and L. reuteri RC-14 for four weeks. Women with other vaginal infections, recent antibiotic use, or immunosuppression were excluded. The trial assessed both subjective symptoms and microbiological cure at the end of the treatment period.

What Were the Most Important Findings?

At the end of the four-week treatment, the probiotic group exhibited a significantly higher cure rate compared to the placebo group as measured by Amsel’s criteria and Nugent score. Women in the probiotic group were assessed with “normal” vaginal microbiota based on Gram stain, compared to the placebo group. Importantly, the study also noted reduced BV-associated microbial morphotypes (Gram-variable rods, curved anaerobes) and a statistically significant improvement in key clinical indicators, including pH, discharge, and odor in the probiotic group. While both groups used tinidazole, the probiotics played a key role in enhancing microbiota restoration. Notably, the probiotic strains used in the study are known for producing biosurfactants, bacteriocins, and signaling molecules that can disrupt pathogenic biofilms, particularly those formed by Gardnerella vaginalis. This mechanism may explain their strong microbiome-modulating effect.

What Are the Greatest Implications of This Study?

This study provides robust clinical evidence supporting the adjunctive use of probiotics with antibiotics to treat BV and improve microbiota restoration. By demonstrating that oral administration of specific Lactobacillus strains significantly improves cure rates and promotes a return to healthy vaginal flora, the study bridges microbiome science with practical gynecological care. Clinicians managing recurrent or treatment-resistant BV can consider integrating targeted probiotic strains to reduce recurrence and enhance long-term remission. Additionally, the study underscores the need for strain-specific probiotic selection, given the inconsistent outcomes with nonspecific lactobacilli. The use of probiotics also holds promise in preserving drug efficacy and reducing the need for prolonged antibiotic exposure, which aligns with antimicrobial stewardship principles and microbiome health preservation.

Tinidazole in Anaerobic Infections

May 20, 2025
  • Tinidazole
    Tinidazole

    Tinidazole is a nitroimidazole antimicrobial that selectively targets anaerobic bacteria and protozoa, reshaping the gut ecosystem by depleting pathogenic anaerobes while preserving commensals. Clinically validated for giardiasis, bacterial vaginosis, and colorectal surgery prophylaxis. Its DNA-disrupting and biofilm-penetrating actions reduce inflammatory triggers and create niches for healthy microbiota to rebound.

This review evaluates tinidazole's antibacterial efficacy, pharmacokinetics, and clinical utility in anaerobic infections, highlighting its broad-spectrum activity and potential microbiome implications.

What Was Reviewed?

The article offers a comprehensive evaluation of tinidazole, a nitroimidazole antimicrobial agent. The review delves into its antibacterial spectrum, pharmacokinetics, clinical efficacy, and tolerability, particularly emphasizing its role in managing anaerobic infections. The authors systematically assess tinidazole's in vitro activity against obligate anaerobes, its pharmacological behavior in humans, and its therapeutic outcomes in both prophylactic and treatment settings.

Who Was Reviewed?

This review synthesizes data from various clinical studies and trials involving diverse patient populations subjected to tinidazole treatment. The populations include individuals undergoing elective colorectal and gynecological surgeries, patients with established anaerobic infections, and those receiving tinidazole for prophylactic purposes. The review encompasses findings from both controlled and observational studies, providing a broad perspective on tinidazole's clinical applications across different demographics and clinical scenarios.

What were the most important findings?

Tinidazole exhibits potent in vitro activity against a broad range of obligate anaerobic bacteria, including Bacteroides fragilis, Fusobacterium species, and Clostridium species, with minimum inhibitory concentrations (MICs) typically around 1 µg/ml. The drug demonstrates bactericidal properties, with its efficacy being consistent across various culture media and relatively unaffected by inoculum size, although some variability exists with certain strains like Bacteroides melaninogenicus.

Pharmacokinetically, tinidazole is well-absorbed orally, achieving peak plasma concentrations within 0.5 to 2 hours post-administration and maintaining therapeutic levels for extended periods, which supports its suitability for both single-dose prophylaxis and sustained treatment regimens. Clinically, tinidazole has shown effectiveness in reducing postoperative anaerobic infections when administered prophylactically before surgeries, such as colorectal and gynecological procedures. However, the evidence from double-blind, placebo-controlled studies, particularly in gynecological surgery, remains equivocal, indicating a need for further research to establish definitive conclusions.

What are the greatest implications of this review?

The review underscores tinidazole's efficacy as a therapeutic agent against anaerobic infections, highlighting its favorable pharmacokinetic profile and broad-spectrum activity. For clinicians, tinidazole presents as a viable option for both prophylactic and therapeutic interventions in anaerobic infections. However, the variability in clinical trial outcomes, especially concerning prophylactic use in gynecological surgeries, calls for cautious application and further investigation. Additionally, the potential implications on the microbiome warrant consideration, advocating for strategies that mitigate dysbiosis, such as adjunctive probiotic therapy or targeted antimicrobial stewardship, to preserve microbial homeostasis during treatment.

Endometriomas

Go to Category Page

Iron Overload and Endometriosis: Mechanisms, Implications, and Therapeutic Targets

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Endometriomas
    Endometriomas

    An endometrioma is a type of ovarian cyst filled with old blood, arising from endometrial tissue outside the uterus, typically causing pain and potentially impacting fertility.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

Iron overload in endometriosis contributes to oxidative stress, inflammation, and tissue damage, driving lesion persistence and subfertility. Ferroptosis resistance and dysregulated iron metabolism highlight therapeutic opportunities using iron chelators and modulators.

What Was Reviewed?

This systematic review evaluated the role of iron in the pathophysiology of endometriosis. The review synthesized findings from 53 studies, including both human and animal research, to provide a comprehensive understanding of how excess iron contributes to oxidative stress, inflammation, and tissue damage in endometriosis. It also explored iron-related mechanisms such as ferroptosis and the implications for subfertility, symptom severity, and potential malignant transformation.

Who Was Reviewed?

The review included a total of 53 studies: 47 human studies involving 3,556 participants and 6 animal studies. The human studies primarily examined women diagnosed with endometriosis, and the included research utilized various bio-samples such as ovarian endometriomas, peritoneal fluid, and ectopic endometrial lesions. Animal studies focused on endometriosis models to explore systemic and local iron mechanics.

Key Findings

Iron overload is consistently found in endometriotic tissues and peritoneal fluid but not in systemic circulation. This localized iron accumulation stems from repeated bleeding within lesions, leading to oxidative stress and inflammation that perpetuates the ectopic growth of endometrial tissue. Dysregulated iron transport and the failure of homeostatic mechanisms contribute to this pathology, with increased expression of proteins such as divalent metal transporter-1 (DMT1) and decreased ferroportin expression in affected tissues.

Markers of oxidative stress, including lipid peroxidation and DNA damage, were significantly elevated in endometriotic lesions. Aberrant resistance to ferroptosis, an iron-dependent form of cell death, was identified as a key mechanism supporting lesion persistence. Additionally, iron-induced ferroptosis was linked to the production of pro-inflammatory and angiogenic factors like IL-8 and VEGFA, exacerbating inflammation and lesion vascularization.

Iron overload was implicated in subfertility, as higher iron concentrations in ovarian follicles and endometriomas were associated with impaired oocyte quality and development. These findings suggest that iron mechanics might influence folliculogenesis and embryo viability. Importantly, the review highlighted the therapeutic potential of iron chelators and ferroptosis modulators for managing endometriosis.

Implications of the Review

This review underscores the central role of aberrant iron metabolism in the pathogenesis of endometriosis, providing a mechanistic basis for its persistence, progression, and associated complications such as subfertility and chronic pain. Iron-related oxidative stress emerges as a critical driver of inflammation and tissue damage, making it a promising target for therapeutic intervention. Future research should explore the efficacy of iron-targeted treatments, such as chelators, and further elucidate the role of ferroptosis in endometriosis. These insights could lead to novel strategies for mitigating symptom severity and improving fertility outcomes in affected women.

Lactoferrin

Go to Category Page

Lactoferrin as a Natural Immune Modulator

May 20, 2025
  • Lactoferrin
    Lactoferrin

    OverviewLactoferrin is a multifunctional glycoprotein of significant interest due to its broad spectrum of biological activities. To date, 20 various physiological roles of lactoferrin have been confirmed [x].  The following summary highlights its sources and biological properties:SourcesHuman and Animal Sources: Lactoferrin is naturally present in colostrum, nasal, intestinal, and genital secretions, as well as in […]

Lactoferrin is a key immunomodulatory glycoprotein that bridges innate and adaptive immunity. It regulates oxidative stress, curbs inflammation, and supports mucosal immune integrity, showing promise for microbiome-focused interventions in infections, SIRS, and immune dysfunction.

What was reviewed?

This review comprehensively examined lactoferrin, a multifunctional iron-binding glycoprotein, highlighting its capacity as a natural immunomodulator that bridges innate and adaptive immunity. The paper assessed lactoferrin’s roles in infection, inflammation, oxidative stress, and immune system regulation, including its therapeutic potential in systemic inflammatory response syndrome (SIRS), sepsis, and bacterial infections, including methicillin-resistant Staphylococcus aureus (MRSA).

Who was reviewed?

The review synthesized findings from preclinical in vivo and in vitro studies, particularly in murine models, along with limited human data, to explore the immune mechanisms regulated by lactoferrin. It included evidence across diverse immune cell types, including macrophages, neutrophils, dendritic cells, and T and B lymphocytes.

What were the most important findings?

Lactoferrin significantly modulates immune function through both direct and indirect pathways. It acts as an antimicrobial by binding iron, limiting pathogen proliferation, and neutralizing lipopolysaccharides (LPS). In the microbiome context, this review emphasizes that lactoferrin plays a regulatory role by reducing oxidative stress through iron sequestration and decreasing reactive oxygen species (ROS), which often rise during inflammatory and infectious states.

Lactoferrin dampens excessive immune responses during sepsis and endotoxemia by suppressing mitochondrial ROS and pro-inflammatory cytokines such as IL-6 and TNF-α. It also protects mucosal integrity by reducing bacterial translocation, especially in gut-associated lymphoid tissue. Notably, in both Gram-negative (E. coli) and Gram-positive (MRSA) infection models, lactoferrin improved survival and reduced inflammatory biomarkers. Regarding adaptive immunity, lactoferrin promotes Th1 responses, enhances antigen presentation via dendritic cells and macrophages, and drives T-cell maturation and B-cell isotype switching—thereby reinforcing host microbial surveillance and immunological memory.

Microbiome relevance lies in lactoferrin’s ability to preserve mucosal immunity, reduce gut inflammation, and prevent dysbiosis-linked bacterial dissemination, especially under systemic infectious stress. These actions suggest lactoferrin supports a microbiome-resilient host immune state.

What are the implications of this review?

This review highlights lactoferrin’s potential as a natural immunomodulatory intervention. Its ability to simultaneously enhance protective immunity while dampening harmful inflammation makes it a promising candidate for clinical use in sepsis, autoimmune diseases, infections, and potentially microbiome-targeted therapies. Its role in bridging innate and adaptive immunity also supports its use as a vaccine adjuvant, especially for pathogens requiring strong Th1-type responses. For microbiome-focused clinicians, lactoferrin’s action on mucosal immunity and bacterial translocation pathways suggests a powerful tool for managing dysbiosis-linked systemic inflammation.

Parkinson’s Disease

Go to Category Page

Metallomic analysis of brain tissues distinguishes between cases of dementia with Lewy bodies, Alzheimer’s disease, and Parkinson’s disease dementia

May 20, 2025
  • Parkinson’s Disease
    Parkinson’s Disease

    OverviewParkinson’s disease (PD) is a neurodegenerative disorder primarily characterized by the degeneration of dopaminergic neurons in the nigrostriatal pathway, leading to progressive hypokinetic movements [1], and a range of non-motor symptoms including gastrointestinal (GI) dysfunction [2]. Emerging evidence suggests that the gut microbiome may influence PD through the gut–brain axis. PD is one of the […]

  • Alzheimer’s Dementia
    Alzheimer’s Dementia

    OverviewAlzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by amyloid-beta (Aβ) plaques, neurofibrillary tangles, neuroinflammation, and metabolic dysfunction, ultimately leading to cognitive decline and dementia. Emerging research highlights the microbiota-gut-brain axis as a crucial factor in AD pathogenesis, with gut dysbiosis contributing to neuroinflammation, immune dysregulation, and blood-brain barrier permeability. Microbial metabolites, such as […]

  • Metallomic Signatures
    Metallomic Signatures

    A metallomic signature is the condition-specific profile of trace metals and metal-binding molecules that reflects disrupted metal homeostasis.

Dementia with Lewy bodies (DLB) brains show widespread copper depletion and region-specific sodium, manganese, iron, and selenium alterations. While copper loss is common to AD and PDD, DLB presents a distinct metallomic fingerprint, enabling disease differentiation via PCA. Metallomic profiling may aid in diagnosing overlapping dementias and reveals unique pathophysiological signatures.

What was studied?

This original research study investigated whether the metallomic profile of dementia with Lewy bodies (DLB) differs from that of Alzheimer’s disease (AD) and Parkinson’s disease dementia (PDD). The study sought to determine if post-mortem changes in elemental concentrations—particularly in essential metals—could help differentiate these often-overlapping neurodegenerative conditions. Using ICP-MS (Inductively Coupled Plasma–Mass Spectrometry), the authors quantified concentrations of nine elements (Na, Mg, K, Ca, Mn, Fe, Cu, Zn, and Se) across 10 brain regions from DLB patients and age-/sex-matched controls. These findings were directly compared to previously published metallomic profiles for AD and PDD, produced using identical methodologies. Multivariate analyses (PCA and PLS-DA) were employed to assess the potential for disease discrimination based on metal signatures.

Who was studied?

The study analyzed post-mortem brain tissue from 23 DLB patients and 20 controls, collected across ten distinct brain regions. Comparative analyses included prior datasets from similarly matched AD and PDD patient cohorts.

What were the most important findings?

n this study, region-specific metallomic profiling revealed distinct trace element alterations in Dementia with Lewy Bodies (DLB). Copper (Cu) levels were consistently decreased in five of ten DLB brain regions, including the cingulate gyrus (CG), middle temporal gyrus (MTG), primary visual cortex (PVC), substantia nigra (SN), and putamen (PUT), suggesting a widespread Cu deficiency. Sodium (Na) was elevated in four regions—medulla (MED), cerebellum (CB), MTG, and CG—while more localized changes were observed for other metals. Iron (Fe) levels were increased in the motor cortex (MCX) and CG, whereas manganese (Mn) was decreased in both the PVC and MED. Calcium (Ca) was specifically reduced in the hippocampus, and selenium (Se) was also decreased in the PVC. No significant differences in magnesium, potassium, or zinc levels were observed between DLB and control brains. Multivariate analyses, including Principal Component Analysis (PCA) and Partial Least Squares-Discriminant Analysis (PLS-DA), demonstrated that DLB could be distinctly separated from Alzheimer’s disease (AD) and Parkinson’s disease dementia (PDD) based on metallomic signatures. Specifically, CG, MTG, and PVC profiles enabled discrimination between DLB and AD, while the PVC alone differentiated DLB from PDD. Notably, copper depletion emerged as the only common alteration across DLB, AD, and PDD, underscoring its potential central role in the pathogenesis of neurodegenerative diseases. The authors propose that these metallomic fingerprints may reflect disease-specific mechanisms, including variations in oxidative stress, protein aggregation, and mitochondrial dysfunction.

What are the greatest implications of this study?

This study provides compelling evidence that distinct metallomic signatures exist across DLB, AD, and PDD, despite shared pathology such as copper depletion. It strengthens the emerging concept that trace metal dysregulation is disease-specific, rather than a general byproduct of neurodegeneration. The findings support the idea that metallomic profiling—potentially via cerebrospinal fluid or advanced imaging in living patients—could improve differential diagnosis of dementias with overlapping clinical features. Furthermore, the study reinforces the hypothesis that metal dyshomeostasis, particularly copper depletion, may be a contributing pathogenic mechanism, impairing antioxidant defenses and mitochondrial function. These findings could inform new diagnostic tools and therapeutic targets.

The therapeutic role of minocycline in Parkinson’s disease

May 20, 2025
  • Parkinson’s Disease
    Parkinson’s Disease

    OverviewParkinson’s disease (PD) is a neurodegenerative disorder primarily characterized by the degeneration of dopaminergic neurons in the nigrostriatal pathway, leading to progressive hypokinetic movements [1], and a range of non-motor symptoms including gastrointestinal (GI) dysfunction [2]. Emerging evidence suggests that the gut microbiome may influence PD through the gut–brain axis. PD is one of the […]

  • Brain Health
    Brain Health

    Brain health encompasses the overall functioning and well-being of the brain, including cognitive function, emotional and psychological well-being, neurological integrity, behavioral health, neurodevelopmental health, age-related brain health, and brain resilience and plasticity.

The review by Cankaya et al. explores minocycline’s neuroprotective potential in Parkinson's disease (PD), highlighting its anti-inflammatory, antioxidant, and anti-apoptotic effects. While preclinical studies show promising neuroprotective results, clinical trials have yet to confirm its efficacy in slowing PD progression. The review emphasizes the need for further research to validate minocycline as a therapeutic agent for PD and other neurodegenerative disorders.

What Was Reviewed?

The review focused on the potential therapeutic role of minocycline, a semisynthetic tetracycline-derived antibiotic, in Parkinson’s disease (PD). It encompassed both preclinical and clinical studies to evaluate minocycline’s neuroprotective effects and its mechanisms of action in various experimental models of neurodegenerative diseases, including cerebral ischemia, traumatic brain injury, amyotrophic lateral sclerosis (ALS), Huntington’s disease (HD), multiple sclerosis (MS), and specifically Parkinson’s disease (PD).

Who Was Reviewed?

The review analyzed data from multiple sources, including:

Animal Models: Various in vivo and in vitro studies were conducted on rodents and other animals to observe the neuroprotective effects of minocycline.

Clinical Trials: Human studies, including randomized, double-blind clinical trials, that assessed the efficacy of minocycline in treating PD and other neurodegenerative disorders.

Literature Reviews: Retrospectively recorded results from studies available on databases such as PubMed, Scopus, and ISI Web of Science, focusing on keywords like “minocycline and Parkinson’s disease,” “minocycline and neuroprotection,” “minocycline,” and “neurodegeneration.”

Most Important Findings

Anti-Inflammatory Effects: Minocycline modulates microglia activation, reduces the release of proinflammatory cytokines, and inhibits pathways leading to neuroinflammation. This helps in attenuating neuroinflammation, a critical aspect of PD pathogenesis.

Antioxidant Effects: The drug reduces oxidative stress by inhibiting the production of reactive oxygen species (ROS) and stabilizing mitochondrial function.

Anti-Apoptotic Effects: Minocycline inhibits apoptotic pathways by stabilizing mitochondrial membranes, reducing the release of cytochrome c, and modulating the expression of B-cell lymphoma 2 (Bcl-2) proteins, thereby preventing neuronal cell death.

Efficacy in Experimental Models:

Parkinson’s Disease Models: In MPTP and 6-OHDA-induced PD models, minocycline reduced dopaminergic neuron degeneration and improved behavioral deficits.

Other Neurodegenerative Models: Minocycline demonstrated neuroprotective effects in models of ALS, HD, and MS by inhibiting microglial activation and reducing neuronal death.

Greatest Implications

The therapeutic role of minocycline in Parkinson’s disease as a Potential Neuroprotective Agent: Despite mixed results in clinical trials, the extensive preclinical data support the potential of minocycline as a neuroprotective agent that could modify disease progression in PD and possibly other neurodegenerative diseases. This highlights the need for further research and well-designed clinical trials to determine its efficacy conclusively.

Mechanistic Insights: The review provides a comprehensive understanding of the mechanisms through which minocycline exerts its effects, such as anti-inflammatory, antioxidant, and anti-apoptotic pathways. This knowledge could be pivotal in developing targeted therapies for neurodegenerative diseases.

Future Research Directions: The findings underline the importance of exploring combination therapies that might enhance the efficacy of minocycline. Additionally, investigating different dosing regimens, treatment durations, and patient populations could yield more definitive results regarding its therapeutic potential.

Broader Implications for Neurodegenerative Diseases: The review suggests that minocycline's therapeutic benefits might extend beyond PD to other conditions like ALS, HD, and MS, making it a promising candidate for broader neuroprotective applications.

Alzheimer’s Dementia

Go to Category Page

Metallomic analysis of brain tissues distinguishes between cases of dementia with Lewy bodies, Alzheimer’s disease, and Parkinson’s disease dementia

May 20, 2025
  • Parkinson’s Disease
    Parkinson’s Disease

    OverviewParkinson’s disease (PD) is a neurodegenerative disorder primarily characterized by the degeneration of dopaminergic neurons in the nigrostriatal pathway, leading to progressive hypokinetic movements [1], and a range of non-motor symptoms including gastrointestinal (GI) dysfunction [2]. Emerging evidence suggests that the gut microbiome may influence PD through the gut–brain axis. PD is one of the […]

  • Alzheimer’s Dementia
    Alzheimer’s Dementia

    OverviewAlzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by amyloid-beta (Aβ) plaques, neurofibrillary tangles, neuroinflammation, and metabolic dysfunction, ultimately leading to cognitive decline and dementia. Emerging research highlights the microbiota-gut-brain axis as a crucial factor in AD pathogenesis, with gut dysbiosis contributing to neuroinflammation, immune dysregulation, and blood-brain barrier permeability. Microbial metabolites, such as […]

  • Metallomic Signatures
    Metallomic Signatures

    A metallomic signature is the condition-specific profile of trace metals and metal-binding molecules that reflects disrupted metal homeostasis.

Dementia with Lewy bodies (DLB) brains show widespread copper depletion and region-specific sodium, manganese, iron, and selenium alterations. While copper loss is common to AD and PDD, DLB presents a distinct metallomic fingerprint, enabling disease differentiation via PCA. Metallomic profiling may aid in diagnosing overlapping dementias and reveals unique pathophysiological signatures.

What was studied?

This original research study investigated whether the metallomic profile of dementia with Lewy bodies (DLB) differs from that of Alzheimer’s disease (AD) and Parkinson’s disease dementia (PDD). The study sought to determine if post-mortem changes in elemental concentrations—particularly in essential metals—could help differentiate these often-overlapping neurodegenerative conditions. Using ICP-MS (Inductively Coupled Plasma–Mass Spectrometry), the authors quantified concentrations of nine elements (Na, Mg, K, Ca, Mn, Fe, Cu, Zn, and Se) across 10 brain regions from DLB patients and age-/sex-matched controls. These findings were directly compared to previously published metallomic profiles for AD and PDD, produced using identical methodologies. Multivariate analyses (PCA and PLS-DA) were employed to assess the potential for disease discrimination based on metal signatures.

Who was studied?

The study analyzed post-mortem brain tissue from 23 DLB patients and 20 controls, collected across ten distinct brain regions. Comparative analyses included prior datasets from similarly matched AD and PDD patient cohorts.

What were the most important findings?

n this study, region-specific metallomic profiling revealed distinct trace element alterations in Dementia with Lewy Bodies (DLB). Copper (Cu) levels were consistently decreased in five of ten DLB brain regions, including the cingulate gyrus (CG), middle temporal gyrus (MTG), primary visual cortex (PVC), substantia nigra (SN), and putamen (PUT), suggesting a widespread Cu deficiency. Sodium (Na) was elevated in four regions—medulla (MED), cerebellum (CB), MTG, and CG—while more localized changes were observed for other metals. Iron (Fe) levels were increased in the motor cortex (MCX) and CG, whereas manganese (Mn) was decreased in both the PVC and MED. Calcium (Ca) was specifically reduced in the hippocampus, and selenium (Se) was also decreased in the PVC. No significant differences in magnesium, potassium, or zinc levels were observed between DLB and control brains. Multivariate analyses, including Principal Component Analysis (PCA) and Partial Least Squares-Discriminant Analysis (PLS-DA), demonstrated that DLB could be distinctly separated from Alzheimer’s disease (AD) and Parkinson’s disease dementia (PDD) based on metallomic signatures. Specifically, CG, MTG, and PVC profiles enabled discrimination between DLB and AD, while the PVC alone differentiated DLB from PDD. Notably, copper depletion emerged as the only common alteration across DLB, AD, and PDD, underscoring its potential central role in the pathogenesis of neurodegenerative diseases. The authors propose that these metallomic fingerprints may reflect disease-specific mechanisms, including variations in oxidative stress, protein aggregation, and mitochondrial dysfunction.

What are the greatest implications of this study?

This study provides compelling evidence that distinct metallomic signatures exist across DLB, AD, and PDD, despite shared pathology such as copper depletion. It strengthens the emerging concept that trace metal dysregulation is disease-specific, rather than a general byproduct of neurodegeneration. The findings support the idea that metallomic profiling—potentially via cerebrospinal fluid or advanced imaging in living patients—could improve differential diagnosis of dementias with overlapping clinical features. Furthermore, the study reinforces the hypothesis that metal dyshomeostasis, particularly copper depletion, may be a contributing pathogenic mechanism, impairing antioxidant defenses and mitochondrial function. These findings could inform new diagnostic tools and therapeutic targets.

Metallomic Signatures

Go to Category Page

Metallomic analysis of brain tissues distinguishes between cases of dementia with Lewy bodies, Alzheimer’s disease, and Parkinson’s disease dementia

May 20, 2025
  • Parkinson’s Disease
    Parkinson’s Disease

    OverviewParkinson’s disease (PD) is a neurodegenerative disorder primarily characterized by the degeneration of dopaminergic neurons in the nigrostriatal pathway, leading to progressive hypokinetic movements [1], and a range of non-motor symptoms including gastrointestinal (GI) dysfunction [2]. Emerging evidence suggests that the gut microbiome may influence PD through the gut–brain axis. PD is one of the […]

  • Alzheimer’s Dementia
    Alzheimer’s Dementia

    OverviewAlzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by amyloid-beta (Aβ) plaques, neurofibrillary tangles, neuroinflammation, and metabolic dysfunction, ultimately leading to cognitive decline and dementia. Emerging research highlights the microbiota-gut-brain axis as a crucial factor in AD pathogenesis, with gut dysbiosis contributing to neuroinflammation, immune dysregulation, and blood-brain barrier permeability. Microbial metabolites, such as […]

  • Metallomic Signatures
    Metallomic Signatures

    A metallomic signature is the condition-specific profile of trace metals and metal-binding molecules that reflects disrupted metal homeostasis.

Dementia with Lewy bodies (DLB) brains show widespread copper depletion and region-specific sodium, manganese, iron, and selenium alterations. While copper loss is common to AD and PDD, DLB presents a distinct metallomic fingerprint, enabling disease differentiation via PCA. Metallomic profiling may aid in diagnosing overlapping dementias and reveals unique pathophysiological signatures.

What was studied?

This original research study investigated whether the metallomic profile of dementia with Lewy bodies (DLB) differs from that of Alzheimer’s disease (AD) and Parkinson’s disease dementia (PDD). The study sought to determine if post-mortem changes in elemental concentrations—particularly in essential metals—could help differentiate these often-overlapping neurodegenerative conditions. Using ICP-MS (Inductively Coupled Plasma–Mass Spectrometry), the authors quantified concentrations of nine elements (Na, Mg, K, Ca, Mn, Fe, Cu, Zn, and Se) across 10 brain regions from DLB patients and age-/sex-matched controls. These findings were directly compared to previously published metallomic profiles for AD and PDD, produced using identical methodologies. Multivariate analyses (PCA and PLS-DA) were employed to assess the potential for disease discrimination based on metal signatures.

Who was studied?

The study analyzed post-mortem brain tissue from 23 DLB patients and 20 controls, collected across ten distinct brain regions. Comparative analyses included prior datasets from similarly matched AD and PDD patient cohorts.

What were the most important findings?

n this study, region-specific metallomic profiling revealed distinct trace element alterations in Dementia with Lewy Bodies (DLB). Copper (Cu) levels were consistently decreased in five of ten DLB brain regions, including the cingulate gyrus (CG), middle temporal gyrus (MTG), primary visual cortex (PVC), substantia nigra (SN), and putamen (PUT), suggesting a widespread Cu deficiency. Sodium (Na) was elevated in four regions—medulla (MED), cerebellum (CB), MTG, and CG—while more localized changes were observed for other metals. Iron (Fe) levels were increased in the motor cortex (MCX) and CG, whereas manganese (Mn) was decreased in both the PVC and MED. Calcium (Ca) was specifically reduced in the hippocampus, and selenium (Se) was also decreased in the PVC. No significant differences in magnesium, potassium, or zinc levels were observed between DLB and control brains. Multivariate analyses, including Principal Component Analysis (PCA) and Partial Least Squares-Discriminant Analysis (PLS-DA), demonstrated that DLB could be distinctly separated from Alzheimer’s disease (AD) and Parkinson’s disease dementia (PDD) based on metallomic signatures. Specifically, CG, MTG, and PVC profiles enabled discrimination between DLB and AD, while the PVC alone differentiated DLB from PDD. Notably, copper depletion emerged as the only common alteration across DLB, AD, and PDD, underscoring its potential central role in the pathogenesis of neurodegenerative diseases. The authors propose that these metallomic fingerprints may reflect disease-specific mechanisms, including variations in oxidative stress, protein aggregation, and mitochondrial dysfunction.

What are the greatest implications of this study?

This study provides compelling evidence that distinct metallomic signatures exist across DLB, AD, and PDD, despite shared pathology such as copper depletion. It strengthens the emerging concept that trace metal dysregulation is disease-specific, rather than a general byproduct of neurodegeneration. The findings support the idea that metallomic profiling—potentially via cerebrospinal fluid or advanced imaging in living patients—could improve differential diagnosis of dementias with overlapping clinical features. Furthermore, the study reinforces the hypothesis that metal dyshomeostasis, particularly copper depletion, may be a contributing pathogenic mechanism, impairing antioxidant defenses and mitochondrial function. These findings could inform new diagnostic tools and therapeutic targets.

Low‑Nickel Diet (LNiD)

Go to Category Page

Nickel Sensitivity and Symptom Management in Endometriosis: The Role of a Low-Nickel Diet

May 20, 2025
  • Metals
    Metals

    OverviewHeavy metals play a significant and multifaceted role in the pathogenicity of microbial species. Their involvement can be viewed from two primary perspectives: the toxicity of heavy metals to microbes and the exploitation of heavy metals by microbial pathogens to establish infections and evade the host immune response. Understanding these aspects is critical for both […]

  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Low‑Nickel Diet (LNiD)
    Low‑Nickel Diet (LNiD)

    A low-nickel diet (LNiD) is a therapeutic dietary intervention that eliminates high-nickel foods, primarily plant-based sources such as legumes, nuts, whole grains, and cocoa, to reduce systemic nickel exposure. It is clinically validated for managing systemic nickel allergy syndrome (SNAS) and nickel-induced eczema. Its relevance is well-established in microbiome modulation, with studies demonstrating clinical benefits in conditions such as endometriosis, fibromyalgia, irritable bowel syndrome, and GERD.

  • Nickel
    Nickel

    Bacteria regulate transition metal levels through complex mechanisms to ensure survival and adaptability, influencing both their physiology and the development of antimicrobial strategies.

This study offers new insights into the potential link between nickel sensitivity and symptom severity in endometriosis, suggesting that a low-nickel diet may be a promising intervention for alleviating associated gastrointestinal and gynecological symptoms.

What Was Studied?

This pilot study investigated the prevalence of nickel (Ni) allergic contact mucositis (ACM) in women with endometriosis who experience gastrointestinal symptoms and evaluated the effects of a low-nickel diet on these symptoms. The study focused on assessing the gastrointestinal, extra-intestinal, and gynecological symptom reductions associated with Ni ACM and dietary interventions.

Who Was Studied?

The study enrolled 84 women of reproductive age diagnosed with endometriosis who reported significant gastrointestinal symptoms. Thirty-one participants completed the study, undergoing a diagnostic nickel oral mucosa patch test (omPT) and a subsequent three-month low-nickel diet intervention. Participants were evaluated using symptom questionnaires both at baseline and after dietary changes.

What Were the Most Important Findings?

The study found that 90.3% of participants tested positive for Ni ACM, suggesting a high prevalence of nickel sensitivity among women with endometriosis. Following three months of adhering to a low-nickel diet, significant reductions in all evaluated symptoms were reported. Gastrointestinal symptoms such as abdominal pain, bloating, and diarrhea showed marked improvement. Extra-intestinal symptoms, including fatigue and headaches, and gynecological symptoms such as pelvic pain and dysmenorrhea, also exhibited statistically significant decreases. These findings indicate that nickel sensitivity may contribute to the symptomatic burden of endometriosis, and dietary interventions targeting nickel can alleviate these issues.

The study suggests a potential mechanistic link between nickel exposure, immune responses, and the exacerbation of endometriosis symptoms. Major microbial associations (MMAs) relevant to this context include those influenced by dietary changes, although specific microbiome alterations were not detailed.

What Are the Greatest Implications of This Study?

This research highlights nickel sensitivity as a significant yet previously under-recognized contributor to gastrointestinal and systemic symptoms in endometriosis patients. The findings suggest that incorporating nickel sensitivity screening and low-nickel dietary recommendations could represent a transformative approach to symptom management in endometriosis. Although the sample size was small, the results offer strong preliminary evidence for revising dietary protocols in clinical practice to include low-nickel guidelines, potentially improving the quality of life for patients.

Lipopolysaccharides (LPS)

Go to Category Page

Organophosphorus pesticide chlorpyrifos intake promotes obesity and insulin resistance through impacting gut and gut microbiota

May 20, 2025
  • Lipopolysaccharides (LPS)
    Lipopolysaccharides (LPS)

    Lipopolysaccharide (LPS), a potent endotoxin present in the outer membrane of Gram-negative bacteria that causes chronic immune responses associated with inflammation.

Chronic chlorpyrifos exposure induces obesity and insulin resistance by altering gut microbiota, increasing gut permeability, and driving inflammation.

What was studied?

This study investigated how chronic exposure to the organophosphorus pesticide chlorpyrifos contributes to obesity and insulin resistance (IR) through its impact on gut microbiota and gut barrier integrity. Using C57Bl/6 and CD-1 (ICR) mice fed either normal-fat or high-fat diets, researchers evaluated the effects of chlorpyrifos on gut permeability, microbiota composition, inflammatory responses, and metabolic outcomes such as insulin sensitivity and glucose homeostasis.

Who was studied?

The study utilized male C57Bl/6 and CD-1 (ICR) mice, chosen to represent genetic and dietary variability. Mice were divided into groups based on diet (normal-fat or high-fat) and exposure to chlorpyrifos. Antibiotic treatment and microbiota transplantation were performed to further explore the role of chlorpyrifos-induced microbiota changes in metabolic dysregulation.

What were the most important findings?

Chronic chlorpyrifos exposure disrupted the gut barrier by reducing the expression of tight junction proteins (e.g., occludin, ZO-1), leading to increased intestinal permeability. This facilitated the entry of lipopolysaccharides (LPS) into circulation, promoting low-grade inflammation. Chlorpyrifos exposure altered the gut microbiota composition, with increased Proteobacteria (a source of LPS) and decreased Bacteroidetes—microbial changes commonly associated with obesity. Antibiotic treatment reversed chlorpyrifos-induced obesity and insulin resistance, confirming that microbiota changes played a key role in these outcomes. Mice transplanted with chlorpyrifos-altered microbiota exhibited increased fat accumulation, impaired glucose tolerance, and insulin resistance, highlighting the causal role of microbiota alterations.

What are the greatest implications of this study?

This study underscores the potential role of environmental pollutants like chlorpyrifos in driving the global obesity epidemic by altering gut microbiota and promoting systemic inflammation. These findings suggest that pesticide exposure should be critically evaluated in public health policies and safety assessments. Furthermore, the study provides insights into the gut microbiota as a therapeutic target for metabolic disorders induced by environmental factors.

Hyperbaric Oxygen Therapy (HBOT)

Go to Category Page

Remission of Endometriosis by Hyperbaric Oxygen Treatment in Rats

May 20, 2025
  • Hyperbaric Oxygen Therapy (HBOT)
    Hyperbaric Oxygen Therapy (HBOT)

    Hyperbaric Oxygen Therapy (HBOT) involves breathing pure oxygen in a pressurized chamber, which increases the amount of oxygen dissolved in the blood and delivered to tissues.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

This study demonstrates hyperbaric oxygen therapy (HBOT) achieves complete remission of endometriotic lesions in a rat model by reducing hypoxia, inflammation, and TNF-α levels. While HBOT shows potential as a non-invasive therapy for endometriosis, further studies are needed to validate its impact on microbiome modulation.

What Was Studied?

This study, conducted by Aydin et al., investigated the effects of long-term hyperbaric oxygen therapy (HBOT) on experimentally induced endometriosis in a rat model. The primary objective was to assess whether HBOT could lead to remission of endometriotic lesions and alleviate inflammation by modulating peritoneal cytokine levels, particularly tumor necrosis factor-alpha (TNF-α). The study evaluated the volume, histopathological changes, and proliferation markers (Ki-67) of endometriotic implants after six weeks of HBOT.

Who Was Studied?

The study was performed on 40 non-pregnant, female Wistar-Albino rats. After surgical induction of endometriosis using an autotransplantation technique, the rats were divided into two groups: one receiving HBOT (20 rats) and a control group (19 rats) without treatment. HBOT was administered for 2 hours daily at 2.5 atm for six weeks. Both groups underwent multiple laparotomies to evaluate lesion volume, histopathological scores, and cytokine levels before and after treatment.

What Were the Most Important Findings?

The study demonstrated that HBOT resulted in complete remission of endometriotic lesions in a rat model. Significant reductions were observed in lesion volume, histopathological scores, Ki-67 proliferation markers, and TNF-α levels in the peritoneal fluid of the HBOT-treated group compared to controls. Specifically, the mean lesion volume decreased by 29.5% (57.4 ± 12.5 mm³ in the HBOT group vs. 94.6 ± 17.2 mm³ in controls). TNF-α levels were significantly lower in the HBOT group (5.33 ± 1.02 pg/mL vs. 8.16 ± 1.76 pg/mL in controls). Reduced Ki-67 staining indicated diminished cellular proliferation within endometriotic lesions. The findings suggest that HBOT alleviates endometriosis-associated inflammation by suppressing NFκB-mediated pro-inflammatory pathways and reducing TNF-α levels, key drivers of inflammation and angiogenesis in endometriosis.

From a microbiome perspective, while the study did not directly assess microbial changes, the reduction in hypoxia and inflammation could indirectly modulate microbial communities. Hypoxia-driven dysbiosis, favoring facultative anaerobes like E. coli and GBS, is a known contributor to endometriosis pathogenesis. By restoring oxygen levels and dampening inflammation, HBOT may reduce the selective advantage for these pathogens, potentially rebalancing the peritoneal microbiome.

What Are the Greatest Implications of This Study?

The study positions HBOT as a potential non-invasive therapeutic strategy for endometriosis, with demonstrated efficacy in reducing lesion size and inflammation. By targeting hypoxia and pro-inflammatory cytokines, HBOT addresses two critical drivers of endometriosis pathophysiology. This has implications for both clinical management and microbiome research, suggesting that HBOT could indirectly modulate microbial dysbiosis in endometriosis. However, the absence of direct microbial analyses leaves a critical gap in validating HBOT as a microbiome-targeted intervention (MBTI). Further studies incorporating microbiome sequencing and metabolomics are essential to establish a direct link between HBOT and microbiome modulation.

Autism spectrum disorder (ASD)

Go to Category Page

Reversal of Autism Symptoms among Dizygotic Twins through a Personalized Lifestyle and Environmental Modification Approach: A Case Report and Review of the Literature

May 20, 2025
  • Autism spectrum disorder (ASD)
    Autism spectrum disorder (ASD)

    Autism Spectrum Disorder (ASD) is a neurodevelopmental condition characterized by social, communication, and behavioral challenges. It involves genetic and environmental factors, including microbiome imbalances which influence symptom severity and overall health.

The study showed dramatic, sustained improvements in ASD symptoms in dizygotic female twins through a personalized, multidisciplinary approach targeting modifiable lifestyle and environmental factors, including microbiome-targeted interventions. These included dietary changes, gluten-free and casein-free diets, and supplements like omega-3 fatty acids and probiotics to support gut health and overall wellbeing.

What was studied?

The study focused on the reversal of autism spectrum disorder (ASD) symptoms among dizygotic female twins through a personalized, multidisciplinary therapeutic approach, including microbiome-targeted interventions (MBTIs). The approach primarily targeted modifiable lifestyle and environmental factors believed to contribute to the condition. Following the reversal of autism symptoms in twins, the case report aimed to document the twins' improvements and review the related literature on environmental and lifestyle influences on ASD.  

Who was studied?

The subjects of the study were dizygotic (fraternal) female twin toddlers who were diagnosed with Level 3 severity ASD, which requires very substantial support. The diagnosis was made when the twins were approximately 20 months old. The case report included detailed documentation of the twins' medical history, diagnostic evaluations, and therapeutic interventions over a two-year period.  

What were the most important findings of this case study?

Reversal of Autism Symptoms: Both twins exhibited dramatic improvements in their ASD symptoms, as evidenced by significant reductions in their Autism Treatment Evaluation Checklist (ATEC) scores. One twin's ATEC score decreased from 76 to 32, while the other's decreased from 43 to 4.

Sustained Improvement: The improvements in the twins' symptoms remained relatively stable for six months following the last assessment.

Multidisciplinary Approach: The therapeutic interventions involved a variety of licensed clinicians and focused on environmental and lifestyle modifications tailored to each twin's symptoms, lab results, and other outcome measures. Interventions included dietary changes, nutritional supplements, physical therapies, and environmental modifications.

Parental Involvement: The parents played a crucial role in implementing and achieving the interventions, demonstrating exceptional motivation, compliance, and communication with practitioners.

What are the greatest implications of this case study?

Potential for ASD Reversal: The case report provides encouraging evidence that ASD symptoms can be significantly improved and potentially reversed through a comprehensive, personalized approach that targets modifiable environmental and lifestyle factors.

Role of Environmental and Lifestyle Factors: The findings highlight the significant impact that environmental and lifestyle factors can have on ASD, suggesting that these factors may play a more substantial role than genetic factors in some cases.

Need for Personalized Medicine: The success of the personalized, multidisciplinary approach underscores the importance of individualized treatment plans that consider the unique needs and risk factors of each patient.

Challenges: While the results are promising, the comprehensive and resource-intensive nature of the interventions may not be easily generalizable to all families due to financial and accessibility constraints. This highlights the need for more accessible and cost-effective treatment options.

Future Research: The study calls for prospective studies to further investigate the effectiveness of personalized, multi-modality treatment approaches in reversing ASD symptoms and to establish more precise estimates of the contributions of genetic versus environmental factors in ASD etiology.

Multiple Sclerosis (MS)

Go to Category Page

Shared Gut Microbiota Dysbiosis in Immune-Mediated Diseases: Insights and Biomarkers

May 20, 2025
  • Autoimmune Diseases
    Autoimmune Diseases

    Autoimmune disease is when the immune system mistakenly attacks the body's tissues, often linked to imbalances in the microbiome, which can disrupt immune regulation and contribute to disease development.

  • Multiple Sclerosis (MS)
    Multiple Sclerosis (MS)

    OverviewIn the past decade, research has shown that the enormous community of microbes that live in the gut, known as the gut microbiota, are closely linked to human health and disease. This relationship is primarily due to the gut microbiota’s impact on systemic immune responses. There is growing evidence that these impacts on immune function are […]

The study identifies gut microbial dysbiosis and specific taxa associated with Crohn's disease, ulcerative colitis, multiple sclerosis, and rheumatoid arthritis. Findings support common microbial markers across IMIDs and their diagnostic potential.

What was studied?

This study explored the gut microbiota composition in patients with immune-mediated inflammatory diseases (IMIDs) such as Crohn’s disease (CD), ulcerative colitis (UC), multiple sclerosis (MS), and rheumatoid arthritis (RA) compared to healthy controls (HC). Using 16S rRNA gene sequencing, researchers assessed microbial diversity, richness, and specific taxonomic biomarkers to identify common and unique microbial features across these IMIDs. Machine learning techniques were applied to differentiate microbial patterns between diseases and controls further.

Who was studied?

The cohort included 79 patients across the four IMIDs (20 with CD, 19 with UC, 19 with MS, 21 with RA) and 23 healthy controls. Participants were adults, not on antibiotics for at least eight weeks, and recruited from clinical centers in Canada. Stool samples were collected twice within a two-month interval for analysis.

What were the most important findings?

The study revealed significant microbial dysbiosis in all IMIDs compared to healthy controls. Richness and diversity were lowest in CD and highest in HCs. Taxa such as Actinomyces, Eggerthella, and Streptococcus were enriched in disease cohorts, while beneficial taxa like Roseburia and Gemmiger were depleted. Disease-specific patterns were also identified: Intestinibacter in CD, Bifidobacterium in UC, and unclassified Erysipelotrichaceae in MS. Machine learning highlighted microbial signatures capable of differentiating diseases from HC, with the best classification accuracy observed in CD versus HC (AUC = 0.95).

Key Findings

Microbial Diversity and Richness: Patients with IMIDs exhibited reduced gut microbial richness and diversity compared to HCs, with CD showing the lowest diversity.

Common Dysbiosis Across IMIDs: Certain taxa, such as Actinomyces, Eggerthella, Faecalicoccus, and Streptococcus, were consistently enriched in IMID patients, while Gemmiger, Lachnospira, and Sporobacter were depleted across all disease cohorts.

Disease-Specific Microbiota Signatures: Intestinibacter was elevated in CD. Bifidobacterium was enriched in UC. Erysipelotrichaceae was more abundant in MS. Roseburia was significantly reduced in RA.

Machine Learning Classification: Machine learning models effectively distinguished between IMID and HC cohorts, with the highest classification accuracy for CD (AUC ~0.95). Features like Gemmiger (elevated in HCs) and Faecalicoccus (elevated in IMIDs) were identified as significant markers.

Gram-Positive Focus: The study highlighted an unusually low abundance of Gram-negative bacteria, focusing analysis on Gram-positive taxa, which still yielded meaningful insights into IMID-specific dysbiosis.

What are the greatest implications of this study?

The findings underscore the potential of gut microbiota as diagnostic biomarkers for IMIDs. Shared microbial patterns suggest a common dysbiotic component in IMID etiology, while distinct taxa provide insight into disease-specific mechanisms. This research highlights the importance of the gut microbiome in IMID pathogenesis and opens avenues for microbiome-targeted interventions (MBTIs).

Microbiome Signatures

Go to Category Page

The Microbiome in Precision Medicine: The Way Forward

May 20, 2025
  • Microbiome Signatures
    Microbiome Signatures

    Microbiome signatures define the unique compositions and functions of microbial communities in environments like the human gut, skin, or oral cavity. Characterized by the types and abundances of microbes such as bacteria, viruses, fungi, and archaea, as well as their genetic profiles and metabolites, these signatures are pivotal for diagnosing, understanding, and managing health and disease.

  • Microbiome-Targeted Interventions (MBTIs)
    Microbiome-Targeted Interventions (MBTIs)

    Microbiome Targeted Interventions (MBTIs) are cutting-edge treatments that utilize information from Microbiome Signatures to modulate the microbiome, revolutionizing medicine with unparalleled precision and impact.

The editorial emphasizes how dynamic host–microbiome interactions, combined with genomics and other 'omics' datasets, can inform personalized diagnostics and microbiome-targeted interventions. Despite challenges like microbiome variability and establishing causality, the editorial underscores the microbiome's critical role in shaping the future of individualized medicine.

Overview

Joseph F. Petrosino’s editorial highlights the microbiome's pivotal role in precision medicine, emphasizing its potential to advance personalized diagnostics and therapeutic strategies. The integration of microbiome signatures with other ‘omics’ data, such as genomics and metabolomics, offers a comprehensive understanding of disease mechanisms and tailored treatments. Petrosino draws parallels between microbiome signature research and genomics, with both fields leveraging next-generation sequencing technologies to uncover associations that can be exploited for clinical interventions.

Precision Microbiome Interventions

A notable advancement discussed is the precision editing of the microbiome to treat inflammatory diseases. For instance, Zhu et al. demonstrated how targeting molybdenum-cofactor-dependent pathways with tungstate suppressed Enterobacteriaceae overgrowth in colitis models, restoring microbial diversity and reducing inflammation.[1] This targeted approach exemplifies the potential of microbiome-targeted interventions (MBTIs) to effectively manage dysbiosis without disrupting beneficial microbial communities.

Future Directions

Petrosino advocates for the routine integration of microbiome data with clinical and multi-omics datasets to enhance diagnostic precision, treatment customization, and early disease detection. He envisions a future where microbiome profiling becomes standard practice in clinical trials and patient care, facilitating the stratification of patients for personalized interventions. Furthermore, the editorial calls for banking samples from large cohort studies to enable future integrative analyses, maximizing the clinical potential of microbiome research.

Conclusion

In conclusion, Petrosino establishes the microbiome as a cornerstone of precision medicine, emphasizing the need for continued innovation and translational efforts to unlock its full potential in clinical practice.

FAQs

 

 

 [research-feed]

Microbiome-Targeted Interventions (MBTIs)

Go to Category Page

The Microbiome in Precision Medicine: The Way Forward

May 20, 2025
  • Microbiome Signatures
    Microbiome Signatures

    Microbiome signatures define the unique compositions and functions of microbial communities in environments like the human gut, skin, or oral cavity. Characterized by the types and abundances of microbes such as bacteria, viruses, fungi, and archaea, as well as their genetic profiles and metabolites, these signatures are pivotal for diagnosing, understanding, and managing health and disease.

  • Microbiome-Targeted Interventions (MBTIs)
    Microbiome-Targeted Interventions (MBTIs)

    Microbiome Targeted Interventions (MBTIs) are cutting-edge treatments that utilize information from Microbiome Signatures to modulate the microbiome, revolutionizing medicine with unparalleled precision and impact.

The editorial emphasizes how dynamic host–microbiome interactions, combined with genomics and other 'omics' datasets, can inform personalized diagnostics and microbiome-targeted interventions. Despite challenges like microbiome variability and establishing causality, the editorial underscores the microbiome's critical role in shaping the future of individualized medicine.

Overview

Joseph F. Petrosino’s editorial highlights the microbiome's pivotal role in precision medicine, emphasizing its potential to advance personalized diagnostics and therapeutic strategies. The integration of microbiome signatures with other ‘omics’ data, such as genomics and metabolomics, offers a comprehensive understanding of disease mechanisms and tailored treatments. Petrosino draws parallels between microbiome signature research and genomics, with both fields leveraging next-generation sequencing technologies to uncover associations that can be exploited for clinical interventions.

Precision Microbiome Interventions

A notable advancement discussed is the precision editing of the microbiome to treat inflammatory diseases. For instance, Zhu et al. demonstrated how targeting molybdenum-cofactor-dependent pathways with tungstate suppressed Enterobacteriaceae overgrowth in colitis models, restoring microbial diversity and reducing inflammation.[1] This targeted approach exemplifies the potential of microbiome-targeted interventions (MBTIs) to effectively manage dysbiosis without disrupting beneficial microbial communities.

Future Directions

Petrosino advocates for the routine integration of microbiome data with clinical and multi-omics datasets to enhance diagnostic precision, treatment customization, and early disease detection. He envisions a future where microbiome profiling becomes standard practice in clinical trials and patient care, facilitating the stratification of patients for personalized interventions. Furthermore, the editorial calls for banking samples from large cohort studies to enable future integrative analyses, maximizing the clinical potential of microbiome research.

Conclusion

In conclusion, Petrosino establishes the microbiome as a cornerstone of precision medicine, emphasizing the need for continued innovation and translational efforts to unlock its full potential in clinical practice.

FAQs

 

 

 [research-feed]

Chronic Pelvic Pain (CPP)

Go to Category Page

The role of gut and genital microbiota and the estrobolome in endometriosis, infertility and chronic pelvic pain

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Chronic Pelvic Pain (CPP)
    Chronic Pelvic Pain (CPP)

    Chronic Pelvic Pain (CPP) is persistent pain in the pelvic region lasting six months or longer, often multifactorial, impacting physical and emotional well-being, and associated with various medical conditions.

  • Infertility
    Infertility

    Infertility is the inability to conceive after 12 months of regular, unprotected sex. It affects both men and women and can be due to various physical, hormonal, or genetic factors. Treatments include medication, surgery, assisted reproductive technologies, and lifestyle changes.

This review highlights the gut and genital microbiome's roles in estrogen-driven conditions like endometriosis, infertility, and CPP, emphasizing dysbiosis' impact on inflammation and estrogen metabolism.

What was reviewed?

The reviewed manuscript explored the intricate relationship between the gut and genital microbiomes, the estrobolome, and their roles in the pathophysiology of endometriosis, infertility, and chronic pelvic pain (CPP). The authors critically examined 28 clinical and six preclinical studies to understand microbial dysbiosis's contributions to estrogen metabolism, inflammation, and symptomatology in these conditions. This review also identified methodological gaps in microbiome studies and proposed strategies to improve future research.

Who was reviewed?

The review included human and animal studies, examining women diagnosed with endometriosis, infertility, and CPP, alongside healthy controls. Specific focus was placed on microbial associations in the gut, cervicovaginal, and endometrial microbiomes, with emphasis on bacterial vaginosis-associated bacteria, Lactobacillus depletion, and microbial influences on estrogen-driven mechanisms.

What were the most important findings?

Key findings highlighted that dysbiosis in the gut microbiome disrupts the estrobolome, an essential modulator of estrogen metabolism. This disruption contributes to heightened systemic and local inflammation, potentially exacerbating endometriosis symptoms and infertility. Many studies noted an association between bacterial vaginosis-related bacteria and a reduction in Lactobacillus dominance in the cervicovaginal microbiome with the prevalence of endometriosis and infertility. Additionally, the review underscored a bidirectional relationship between gut microbiota and endometriosis progression in animal models, emphasizing the role of gut dysbiosis in increasing b-glucuronidase activity, leading to elevated circulating estrogen levels.

What are the greatest implications of this review?

This review underscores the need for rigorous, standardized methodologies to better delineate causal relationships between microbiota and gynecological conditions like endometriosis and CPP. The findings of this review suggest that targeting the microbiome could lead to novel diagnostics and therapeutics for estrogen-driven diseases. The review also highlights the potential of leveraging microbiome-based biomarkers for non-invasive diagnostics and monitoring of endometriosis progression, bridging a critical translational gap in gynecological health.

The role of the vaginal microbiome in distinguishing female chronic pelvic pain caused by endometriosis/adenomyosis

May 20, 2025
  • Women’s Health
    Women’s Health

    Women’s health, a vital aspect of medical science, encompasses various conditions unique to women’s physiological makeup. Historically, women were often excluded from clinical research, leading to a gap in understanding the intricacies of women’s health needs. However, recent advancements have highlighted the significant role that the microbiome plays in these conditions, offering new insights and potential therapies. MicrobiomeSignatures.com is at the forefront of exploring the microbiome signature of each of these conditions to unravel the etiology of these diseases and develop targeted microbiome therapies.

  • Endometriosis
    Endometriosis

    Endometriosis involves ectopic endometrial tissue causing pain and infertility. Validated and Promising Interventions include Hyperbaric Oxygen Therapy (HBOT), Low Nickel Diet, and Metronidazole therapy.

  • Chronic Pelvic Pain (CPP)
    Chronic Pelvic Pain (CPP)

    Chronic Pelvic Pain (CPP) is persistent pain in the pelvic region lasting six months or longer, often multifactorial, impacting physical and emotional well-being, and associated with various medical conditions.

This study examines the role of the vaginal microbiome in distinguishing chronic pelvic pain caused by endometriosis and adenomyosis. Findings highlight specific microbial signatures associated with pain severity, offering potential non-invasive biomarkers for differential diagnosis and targeted therapeutic strategies.

What was studied?

This study investigated whether the composition of the vaginal microbiome could serve as a diagnostic biomarker to differentiate chronic pelvic pain (CPP) caused by endometriosis or adenomyosis (EM/AM) from other causes of chronic pelvic pain syndrome (CPPS) in women. Using 16S rRNA sequencing (V4 region), the researchers profiled the vaginal microbiota of 37 women with EM/AM-associated CPP, 25 with CPPS from other causes, and 66 healthy controls without CPPS. Additionally, the study explored whether combining vaginal microbial markers with serum CA125 could improve differential diagnostic accuracy.

Who was studied?

The study included 128 premenopausal women attending the gynecology department of Peking Union Medical College Hospital. These were stratified into three groups: 37 women with surgically confirmed EM/AM-associated CPP, 25 women with non-EM/AM CPPS (adhesions, hydrosalpinx, infertility), and 66 women without any chronic pelvic pain. All participants were HPV-negative, had not recently used antibiotics or vaginal products, and were matched for age, gravidity, parity, and contraceptive method to control for confounding variables.

What were the most important findings?

The vaginal microbiome of women with EM/AM-associated CPP exhibited significantly higher alpha diversity than those in the CPPS and healthy control groups. Taxonomic analyses revealed distinct microbial signatures: increased abundance of Clostridium butyricum, Clostridium disporicum, Alloscardovia omnicolens, and Veillonella montpellierensis, alongside a marked depletion of Lactobacillus jensenii, Lactobacillus reuteri, and Lactobacillus iners. These differentially abundant taxa serve as potential microbiome biomarkers.

Diagnostic performance analysis demonstrated that a combination of microbial biomarkers (specifically, a relative abundance of Clostridium disporicum >0.001105% and Lactobacillus reuteri <0.1911349%) yielded 81.08% sensitivity and 52% specificity for identifying EM/AM-associated CPP. When combined with serum CA125 levels, sensitivity increased to 89.19%, although specificity remained unchanged. Functional predictions via PICRUSt revealed enrichment of metabolic pathways such as amino acid metabolism, energy metabolism, and metabolism of cofactors and vitamins in EM/AM patients, along with downregulation of membrane transport and nucleotide metabolism compared to controls. These shifts may reflect microbial contributions to inflammation and pain signaling pathways implicated in EM/AM-associated CPP.

From a microbiome signature standpoint, the enriched taxa—particularly Clostridium disporicum and Alloscardovia omnicolens—emerge as Major Microbial Associations (MMAs) due to their consistent elevation in EM/AM patients. Conversely, Lactobacillus jensenii and L. reuteri, known for their protective, anti-inflammatory properties, are depleted, suggesting their role in maintaining vaginal eubiosis and preventing EM/AM-associated pathogenesis.

What are the greatest implications of this study?

This research provides compelling evidence that the vaginal microbiome harbors discriminative microbial signatures capable of differentiating EM/AM-associated CPP from other forms of chronic pelvic pain. The incorporation of specific microbial biomarkers, particularly when paired with serum CA125, may improve non-invasive diagnostic accuracy, enabling earlier and more targeted therapeutic intervention. Clinically, these findings underscore the potential of microbiome-informed diagnostics for gynecological conditions where conventional markers fall short. More broadly, this study suggests that vaginal dysbiosis, characterized by Lactobacillus depletion and enrichment of saccharolytic and anaerobic species, could be causally linked to EM/AM pathogenesis, possibly via inflammatory or metabolic pathways. Future studies incorporating metagenomic or metabolomic analyses are warranted to functionally validate these microbial associations and to explore the feasibility of microbial modulation as a therapeutic strategy.

Join the Roundtable

Contribute to published consensus reports, connect with top clinicians and researchers, and receive exclusive invitations to roundtable conferences.