Did you know?
Women with PCOS produce more than double the amount of anti-Müllerian hormone (AMH) compared to women without PCOS—so much so that some researchers have proposed renaming the condition “AMH syndrome.”
Polycystic ovary syndrome (PCOS)
Polycystic ovary syndrome (COS) 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.
-
Divine Aleru
I am a biochemist with a deep curiosity for the human microbiome and how it shapes human health, and I enjoy making microbiome science more accessible through research and writing. With 2 years experience in microbiome research, I have curated microbiome studies, analyzed microbial signatures, and now focus on interventions as a Microbiome Signatures and Interventions Research Coordinator.
-
Karen Pendergrass
Karen Pendergrass is a microbiome researcher specializing in microbiome-targeted interventions (MBTIs). She systematically analyzes scientific literature to identify microbial patterns, develop hypotheses, and validate interventions. As the founder of the Microbiome Signatures Database, she bridges microbiome research with clinical practice. In 2012, based on her own investigative research, she became the first documented case of FMT for Celiac Disease—four years before the first published case study.
Page Snapshot
- References
- Identified MMAs
Microbiome Signatures identifies and validates condition-specific microbiome shifts and interventions to accelerate clinical translation. Our multidisciplinary team supports clinicians, researchers, and innovators in turning microbiome science into actionable medicine.
I am a biochemist with a deep curiosity for the human microbiome and how it shapes human health, and I enjoy making microbiome science more accessible through research and writing. With 2 years experience in microbiome research, I have curated microbiome studies, analyzed microbial signatures, and now focus on interventions as a Microbiome Signatures and Interventions Research Coordinator.
Overview
Polycystic ovary syndrome (PCOS) is a common and complex endocrine and metabolic disorder affecting women of reproductive age, with a global prevalence ranging from 4% to 20% depending on the diagnostic criteria used.[1][2] It is typically characterized by the presence of hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology,[3] and is strongly associated with various health issues including infertility, insulin resistance (IR), type 2 diabetes mellitus, and cardiovascular disease.[4][5] Although the precise pathogenesis of PCOS is not fully understood, accumulating evidence suggests a complex interplay of genetic predisposition, epigenetic modifications, and environmental factors such as dietary choices and lifestyle,[6] with emerging research highlighting the causal role of gut dysbiosis in its development. [7][8][9]
Diagnosis
PCOS diagnosis typically involves the identification of a combination of clinical and biochemical features, primarily focusing on hyperandrogenism, ovulatory dysfunction including irregular menstrual cycles, infrequent ovulation, or anovulation, and polycystic ovarian morphology (PCOM) on ultrasound.[10] The Rotterdam criteria requires at least two out of these three features for diagnosis, after excluding other causes, as ovarian cysts alone are not enough for a diagnosis, as they can occur in other conditions. [11]
Causal Theories
The pathogenesis of PCOS is multifactorial, involving genetic, environmental, and hormonal factors. Insulin resistance is a central feature, and hyperinsulinemia leads to increased ovarian androgen production.[12] This can disrupt normal follicular development and contribute to the clinical manifestations of PCOS. Environmental factors, including diet and lifestyle, can exacerbate insulin resistance, while genetic predispositions to hyperandrogenism and metabolic dysfunction further complicate the condition.[13] There is emerging evidence supporting the role of gut microbiota in the development and progression of PCOS, with altered microbial compositions being linked to insulin resistance and inflammation.[14]
Associated Conditions
PCOS is a complex condition with significant associations to a variety of metabolic, hormonal, and psychological comorbidities. These include infertility, which is a hallmark feature due to anovulation, metabolic syndrome, and insulin resistance, which elevate the risk of type 2 diabetes.[15][16] Women with PCOS also face a higher risk of cardiovascular diseases, such as hypertension and dyslipidemia, due to hormonal imbalances and metabolic dysfunctions.[17] Endometrial cancer is another concern, as prolonged anovulation leads to unopposed estrogen exposure, increasing the risk of endometrial hyperplasia.[18] Additionally, women with PCOS are more likely to experience depression, anxiety, and eating disorders, potentially exacerbated by physical symptoms like hirsutism, acne, and weight gain.
Primer
To understand PCOS comprehensively, it is essential to explore the intersections of hormonal dysregulation, insulin resistance, and the gut microbiome. The elevated levels of insulin in PCOS not only contribute to metabolic abnormalities but also directly affect ovarian steroidogenesis, exacerbating the symptoms of hyperandrogenism.[19] Dysbiosis in the gut microbiome is now being recognized as a significant contributor to the inflammation and metabolic dysfunction observed in PCOS. Studies suggest that gut microbial imbalances may lead to the development of insulin resistance through mechanisms such as the leakage of lipopolysaccharide (LPS) into the bloodstream, which in turn activate immune pathways that interfere with insulin receptor function.[20][21] This relationship between the gut microbiome and insulin resistance is crucial to understanding the broader pathophysiology of PCOS, particularly regarding the gut-brain axis and hormonal regulation and presents opportunities for microbiome-based diagnostics and microbiome-targeted interventions (MBTIs).[22]
Metallomic Signature
Heavy metals and trace elements play a significant role in the pathophysiology of polycystic ovary syndrome (PCOS). Studies have revealed imbalances in several essential trace elements such as zinc, copper, manganese, and the accumulation of toxic metals like lead (Pb), and cadmium (Cd) in PCOS patients. These elements contribute to oxidative stress (OS), inflammation, and hormonal dysregulation, which are hallmarks of PCOS. Elevated zinc and nickel levels, in particular, are indicative of inflammation and metabolic dysfunction, which are key components of PCOS pathology. By examining the metallomic signature alongside microbial shifts in PCOS, it becomes clear that targeting these imbalances could offer novel therapeutic avenues, including dietary interventions and chelation therapies, to manage oxidative stress and inflammation in PCOS patients.
What are the metallomic signatures of PCOS?
Copper
Copper is an essential trace element involved in various enzymatic and redox processes, but in the context of polycystic ovary syndrome (PCOS), its role is complex. Studies have shown that copper levels are often elevated in women with PCOS, and this imbalance contributes to oxidative stress and inflammation, two key features of the condition.[23][24] Copper is involved in redox reactions, and when present in excess, it can generate reactive oxygen species (ROS), exacerbating oxidative damage to cells and tissues. This contributes to insulin resistance and impaired ovarian function, both of which are central to the pathophysiology of PCOS. High copper levels have also been linked to increased estrogenic activity, which might aggravate hormonal imbalances seen in PCOS.[25][26]
Zinc
Zinc is critical for many enzymatic functions, including those involved in immune response and reproductive health. Zinc deficiency could lead to biochemical features of PCOS via decreasing the antioxidant capacity, causing insulin resistance and apoptosis.[27] In women with PCOS, zinc supplementation has been associated with improved ovulatory function and reduced androgen levels. Increased zinc levels have also been linked to lower levels of oxidative stress markers such as malondialdehyde (MDA), which is commonly elevated in PCOS.[28][29][x]
Selenium
Selenium is a trace element that plays a vital role in antioxidant defense mechanisms. In women with PCOS, serum selenium levels are significantly lower in hyperandrogenic women, suggesting its potential role in regulating oxidative stress and reproductive dysfunction.[30] Selenium supplementation has been shown to improve glucose homeostasis and reduce free androgen index levels in women with PCOS.[31]
Magnesium
Magnesium is involved in over 300 enzymatic processes, including those related to insulin sensitivity and glucose metabolism. In women with PCOS, magnesium deficiency is associated with higher levels of oxidative stress and insulin resistance.[32] Supplementation with magnesium has been shown to reduce biomarkers of inflammation and improve hormonal balance . Additionally, magnesium plays a role in the regulation of testosterone and estradiol levels in women with PCOS, with a positive correlation observed between magnesium and testosterone in obese patients.[33]
Chromium
Chromium is essential for normal glucose metabolism. It enhances insulin sensitivity, and in women with PCOS, chromium supplementation has demonstrated beneficial effects on glucose homeostasis and insulin resistance, key issues in the condition.[34][35][x] It is also associated with improvements in metabolic parameters, including serum lipids and oxidative stress markers.[36] However, some studies have shown mixed results, with variations in chromium levels observed between different populations of women with PCOS.
Manganese
Manganese is another essential trace element involved in the defense against oxidative stress. Studies have indicated that lower levels of manganese are observed in women with PCOS, particularly in those with insulin resistance.[37] Its deficiency is thought to exacerbate oxidative stress, contributing to the pathophysiology of PCOS.[38]
Nickel
Nickel has been found to accumulate at higher levels in women with PCOS, especially in those with obesity. Nickel exposure is associated with endocrine disruption and mitochondrial dysfunction, which can impair folliculogenesis and ovulation.[39][40] The metal also induces inflammatory responses, further complicating the symptoms of PCOS
Lead
Lead exposure is another significant concern in women with PCOS. Even at low levels, lead has been shown to contribute to oxidative stress, which exacerbates the metabolic and reproductive dysfunctions in PCOS. Studies indicate that elevated blood lead levels (BLLs) are associated with a higher incidence of insulin resistance, a key feature of PCOS. Lead disrupts cellular functions by generating ROS and impairing antioxidant defenses, contributing to the chronic inflammation seen in PCOS.[41] Additionally, lead exposure can alter hormone levels, including those involved in the regulation of ovarian function, thus worsening symptoms like anovulation and infertility.
Cadmium
Cadmium, a heavy metal known for its toxicity, is linked to increased oxidative stress in women with PCOS. Cadmium exposure leads to the generation of reactive oxygen species (ROS), which can damage cellular components and exacerbate insulin resistance.[42][43]
Nutritional Immunity
In PCOS, imbalances in essential micronutrients like zinc, copper, and manganese contribute to immune dysfunction and inflammation. Deficiencies in these nutrients are linked to increased oxidative stress and metabolic issues, such as insulin resistance and hormonal disturbances.[44][45][x][46] In addition to these micronutrients, calprotectin, a protein involved in inflammation and immune response, plays a critical role in modulating the immune system in PCOS. Elevated calprotectin levels have been associated with increased inflammation and oxidative stress in PCOS patients, further highlighting the importance of balancing trace elements and inflammatory markers.[47] Addressing these nutritional imbalances and the inflammatory pathways, including calprotectin regulation, through targeted interventions could help manage inflammation, improve metabolic function, and restore microbial balance, offering potential benefits for PCOS management.
Microbiome Signature: Polycystic ovary syndrome (PCOS)
Interventions
Our validation method confirms the effectiveness of microbiome-targeted interventions (MBTIs) by linking their therapeutic impact on microbial imbalances with clinical outcomes, key pathological markers, and major microbial associations (MMAs) associated with PCOS. This dual approach ensures that interventions effectively target both the microbial imbalances and the fundamental biological processes of PCOS, reinforcing the precision of microbiome signatures and the success of MBTIs. Moreover, promising candidates demonstrating preliminary benefits or partial alignment are highlighted for further exploration, contributing to a pipeline of novel treatments for future validation.
Intervention | Classification | MBTI Status |
Fecal Microbiota Transplantation (FMT) | Microbiota-based therapy | Validated |
Metformin | Drug Repurposing | Validated |
Curcumin | Supplement | Validated |
Resistant Dextrin | Supplement | Validated |
Statin Therapy | Pharmaceutical | Validated |
Probiotics | Supplement | Validated |
Synbiotics | Supplement | Validated |
FAQs
What is the role of the microbiome in PCOS?
Emerging research suggests that the gut microbiome may play a significant role in the development and progression of PCOS. Imbalances in gut bacteria, known as dysbiosis, may contribute to the inflammation, insulin resistance, and metabolic dysfunction often seen in PCOS. Certain microbial populations have been linked to increased oxidative stress, which worsens symptoms like acne, hirsutism, and insulin resistance. Additionally, microbial imbalances can affect the body’s ability to process hormones and nutrients, which may contribute to the hormonal irregularities characteristic of PCOS. Research is ongoing, but maintaining a healthy gut microbiome through diet, prebiotics, probiotics, and possibly fecal microbiota transplantation may offer new ways to manage PCOS-related symptoms.
What treatments are available for managing PCOS symptoms?
Treatment for PCOS typically includes hormonal therapies such as birth control pills or anti-androgens to regulate periods and reduce symptoms like acne and hirsutism. For women trying to conceive, ovulation-inducing medications may be prescribed. Metformin is also used to manage insulin resistance, which is common in PCOS.
How is PCOS diagnosed?
PCOS is diagnosed through a combination of clinical symptoms, physical examination, and medical tests. A healthcare provider will evaluate symptoms such as irregular menstrual cycles, hirsutism, and acne. Blood tests are used to measure hormone levels, including androgens and insulin, while an ultrasound may be performed to check for the presence of cysts on the ovaries. The diagnosis is often confirmed if at least two of the following criteria are met: irregular or absent ovulation, elevated levels of androgens (male hormones), and polycystic ovaries visible on ultrasound. It is important for women to consult with a healthcare provider for proper diagnosis and treatment options.
Research Feed
Did you know?
Women with PCOS produce more than double the amount of anti-Müllerian hormone (AMH) compared to women without PCOS—so much so that some researchers have proposed renaming the condition “AMH syndrome.”
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.
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.
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.
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
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.
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.
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.
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.
This review links gut microbiota disruption to the development and symptoms of PCOS, including insulin resistance and hyperandrogenism. It highlights microbiota-targeted therapies—like probiotics, prebiotics, and traditional medicine—as promising options to improve PCOS outcomes and repositions the gut microbiome as a central factor in understanding and treating the disorder.
What Was Reviewed?
This paper reviewed the current scientific understanding of the relationship between gut microbiota and polycystic ovary syndrome (PCOS), positioning the gut microbiome as a potentially central factor in the pathogenesis and management of the condition. It discussed how alterations in gut microbiota composition influence the development and manifestation of PCOS through complex metabolic, inflammatory, and hormonal pathways. The review synthesized a broad range of studies from both clinical research and animal models to clarify how specific microbial shifts contribute to insulin resistance, obesity, hyperandrogenism, inflammation, and even mood disturbances, all characteristic features of PCOS. The study also examined how clinicians can leverage therapeutic interventions, particularly those that modulate the microbiota, such as fecal microbiota transplantation, probiotics, prebiotics, lifestyle changes, and traditional Chinese medicine, to improve clinical outcomes in PCOS patients.
Who Was Reviewed?
The review incorporated findings from a range of human and animal studies focused on subjects diagnosed with PCOS, including those with and without obesity or insulin resistance. It analyzed data from trials where women with PCOS exhibited reduced microbial diversity and distinct microbial profiles compared to healthy controls. The paper also examined results from animal models of PCOS, particularly those induced with letrozole, where researchers demonstrated that fecal transplants from PCOS patients replicated metabolic and reproductive symptoms in recipient mice. In addition, it evaluated interventional studies involving patients treated with probiotics and prebiotics, which demonstrated favorable shifts in microbial composition and clinical markers. These various sources allowed the review to build a robust case for the role of gut microbiota in the etiology and progression of PCOS and to evaluate the therapeutic potential of microbiota modulation across diverse populations.
What Were the Most Important Findings?
The most critical takeaway from this review is the identification of gut microbiota dysbiosis as a key driver of PCOS pathophysiology. Reduced microbial diversity and an imbalance in the relative abundance of bacterial phyla such as Firmicutes and Bacteroidetes are consistently observed in PCOS patients. This microbial disruption correlates with multiple features of the syndrome, including insulin resistance, hyperandrogenism, and chronic inflammation. The review detailed several pathways through which gut microbiota contribute to PCOS. Disrupted energy metabolism allows the host to extract more calories from food, exacerbating obesity and insulin resistance.
Altered short-chain fatty acid production impairs glucose uptake and hormonal regulation. Elevated levels of lipopolysaccharides from gram-negative bacteria promote systemic inflammation and worsen insulin resistance. The metabolism of dietary choline into trimethylamine-N-oxide by gut microbes links PCOS to increased cardiovascular risk. Gut bacteria also regulate bile acid metabolism and influence hormonal balance through receptors such as FXR and TGR5. Furthermore, increased intestinal permeability in PCOS permits microbial endotoxins to leak into circulation, triggering inflammatory responses that disrupt insulin signaling and androgen metabolism. The review also emphasized the brain–gut axis, highlighting how microbiota disturbances contribute to emotional dysregulation in PCOS via serotonin and other neurotransmitter pathways.
What Are the Implications of This Review?
This review redefines PCOS as not merely a hormonal or reproductive disorder, but as a complex condition deeply rooted in metabolic and microbiome-related imbalances. The implications are significant for both diagnosis and treatment. Clinicians can begin to consider the gut microbiota profile as a biomarker for PCOS risk, symptom severity, and treatment response. It also suggests a paradigm shift in therapy, moving beyond symptomatic management toward strategies that address the underlying microbial disruptions. The effectiveness of probiotic and prebiotic interventions, shown to improve insulin sensitivity, reduce androgen levels, and regulate menstrual cycles, opens new therapeutic possibilities. Fecal microbiota transplantation (FMT), though still experimental, has shown early promise in animal models and may become a viable clinical tool. Traditional Chinese medicine, with compounds such as berberine and quercetin, offers additional microbiota-modulating options. Altogether, the review provides strong evidence that targeting the gut microbiome can become a foundational strategy in the long-term management of PCOS.
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.
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 CandidatusSoleaferrea 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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
Did you know?
Women with PCOS produce more than double the amount of anti-Müllerian hormone (AMH) compared to women without PCOS—so much so that some researchers have proposed renaming the condition “AMH syndrome.”
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.
Did you know?
Women with PCOS produce more than double the amount of anti-Müllerian hormone (AMH) compared to women without PCOS—so much so that some researchers have proposed renaming the condition “AMH syndrome.”
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.
Did you know?
Women with PCOS produce more than double the amount of anti-Müllerian hormone (AMH) compared to women without PCOS—so much so that some researchers have proposed renaming the condition “AMH syndrome.”
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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
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.
Lipopolysaccharide (LPS), a potent endotoxin present in the outer membrane of Gram-negative bacteria that causes chronic immune responses associated with inflammation.
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.
A metallomic signature is the condition-specific profile of trace metals and metal-binding molecules that reflects disrupted metal homeostasis.
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.
Major Microbial Associations (MMAs) are fundamental in understanding disease-microbiome interactions and play a crucial role in advancing microbiome-targeted interventions aimed at treating or preventing diseases through microbial modulation.
Fecal Microbiota Transplantation (FMT) involves transferring fecal bacteria from a healthy donor to a patient to restore microbiome balance.
Metformin is a synthetic derivative of guanidine derived from the guanidine alkaloid of the plant Galega officinalis L. with significant hypoglycemic effects. It is a first-line antihyperglycemic agent due to its efficacy, low cost, and favorable safety profile.
Metformin targets both the microbiome and inflammatory mechanisms in PCOS, improving insulin sensitivity, reducing hyperandrogenism, and restoring ovulation. Its microbial effects validate the condition’s microbiome signature, making it a promising microbiome-targeted intervention for managing infertility in PCOS.
Resistant dextrin targets both metabolic and microbial imbalances in PCOS. It reduces fasting blood glucose, lipids, and androgens, while improving microbial composition, supporting its role as a microbiome-targeted intervention (MBTI) for PCOS-related infertility and metabolic health.
Statins target both microbial imbalances and inflammatory mechanisms in PCOS. By improving gut microbial composition and metabolic parameters, statins validate the microbiome signature of PCOS and enhance fertility and metabolic health outcomes.
Probiotics are live microorganisms that offer significant health benefits when administered in adequate amounts. They primarily work by modulating the gut microbiome, supporting a balanced microbial ecosystem. Probiotics have been shown to improve gut health, modulate immune responses, and even influence metabolic and mental health disorders. With growing evidence supporting their therapeutic potential, probiotics are increasingly recognized for their role in treating conditions like irritable bowel syndrome (IBS), antibiotic-associated diarrhea (AAD), and even mental health conditions like depression and anxiety through their impact on the gut-brain axis.
Probiotic supplementation addresses microbial dysbiosis and inflammatory processes in PCOS, improving metabolic and hormonal outcomes, thereby validating its role as an MBTI.
Synbiotics restore gut microbiome balance in PCOS, improving insulin sensitivity, reducing inflammation, and normalizing androgen levels. This validation reinforces its role as a microbiome-targeted intervention.
References
- Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization study.. Sun J, Wang M, Kan Z.. (Front Endocrinol (Lausanne). 2024)
- Gut and Vaginal Microbiomes in PCOS: Implications for Women’s Health.. Gu Y, Zhou G, Zhou F, Li Y, Wu Q, He H, Zhang Y, Ma C, Ding J, Hua K.. (Front Endocrinol (Lausanne). 2022.)
- Gut microbiota alterations reveal potential gut–brain axis changes in polycystic ovary syndrome.. Liang, Z., Di, N., Li, L., & Yang, D.. (Journal of Endocrinological Investigation. 2021.)
- Insulin Resistance and the Polycystic Ovary Syndrome Revisited: An Update on Mechanisms and Implications.. Diamanti-Kandarakis, E., & Dunaif, A.. (Endocrine Reviews, 2012.)
- Exploration of the Relationship Between Gut Microbiota and Polycystic Ovary Syndrome (PCOS): a Review.. Zhao, X., Jiang, Y., Xi, H., Chen, L., & Feng, X.. (Geburtshilfe Frauenheilkd. 2020)
- Polycystic ovary syndrome: pathophysiology and therapeutic opportunities. Dong J, Rees DA.. (BMJ Med. 2023)
- Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization study.. Sun J, Wang M, Kan Z.. (Front Endocrinol (Lausanne). 2024)
- Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome: a review.. He, F., & Li, Y.. (Journal of Ovarian Research. 2020.)
- Dysbiosis of Gut Microbiota (DOGMA) – A novel theory for the development of Polycystic Ovarian Syndrome. Tremellen, K., & Pearce, K.. (Medical Hypotheses, 2012.)
- Insulin Resistance and the Polycystic Ovary Syndrome Revisited: An Update on Mechanisms and Implications.. Diamanti-Kandarakis, E., & Dunaif, A.. (Endocrine Reviews, 2012.)
- Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization study.. Sun J, Wang M, Kan Z.. (Front Endocrinol (Lausanne). 2024)
- Polycystic ovary syndrome: pathophysiology and therapeutic opportunities. Dong J, Rees DA.. (BMJ Med. 2023 Oct 12;2(1):e000548)
- Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization study. Sun J, Wang M, Kan Z.. (Front Endocrinol (Lausanne). 2024 Feb 1;15:1280983)
- Exploration of the Relationship Between Gut Microbiota and Polycystic Ovary Syndrome (PCOS): a Review. Zhao, X., Jiang, Y., Xi, H., Chen, L., & Feng, X. (2020). (Geburtshilfe Und Frauenheilkunde, 80(02), 161–171.)
- Gut microbiota alterations reveal potential gut–brain axis changes in polycystic ovary syndrome. Liang, Z., Di, N., Li, L., & Yang, D. (2021).. (Journal of Endocrinological Investigation, 44(8), 1727–1737)
- Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization study. Sun J, Wang M, Kan Z.. (Front Endocrinol (Lausanne). 2024 Feb 1;15:1280983)
- Characterization of the gut microbiota in polycystic ovary syndrome with dyslipidemia. Yang, T., Li, G., Xu, Y. et al.. (BMC Microbiol 24, 169 (2024))
- Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome: a review. He, F., & Li, Y. (2020). (Journal of Ovarian Research, 13(1))
- Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary Syndrome. Liu, R., Zhang, C., Shi, Y., Zhang, F., Li, L., Wang, X., Ling, Y., Fu, H., Dong, W., Shen, J., Reeves, A., Greenberg, A. S., Zhao, L., Peng, Y., & Ding, X. (2017). (Frontiers in Microbiology, 8, 234522.)
- Antioxidant status in relation to heavy metals induced oxidative stress in patients with polycystic ovarian syndrome (PCOS). Abudawood, M., Tabassum, H., Alanazi, A.H. et al.. (Sci Rep 11, 22935 (2021))
- Serum Copper Level and Polycystic Ovarian Syndrome: A Meta-Analysis. Qingtao Jiang; Feng Zhang; Lei Han; Baoli Zhu; Xin Liu. (Gynecol Obstet Invest (2021) 86 (3): 239–246.)
- Alteration in gut mycobiota of patients with polycystic ovary syndrome. Chen K, Geng H, Liu J, Ye C.. (Microbiol Spectr11:e02360-23)
- Serum copper assessment in patients with polycystic ovary syndrome and tubal infertility: A retrospective 5-year study. Liu, Y., Zhang, W., Liu, Z., Zheng, A., Liang, B., Li, H., & Meng, Q. (2024).. (Food Science & Nutrition, 12, 5979–5989.)
- Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory disease. Tatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016). (REPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.)
- Association of Trace Elements with Polycystic Ovary Syndrome in Women—A Case-Control Study. Srnovršnik, T., Pinter, B., Horvat, M., Snoj Tratnik, J., Falnoga, I., Mazej, D., Verdenik, I., & Virant-Klun, I. (2025).. (Metabolites, 15(2), 79.)
- Heavy Metals and Essential Elements in Association with Oxidative Stress in Women with Polycystic Ovary Syndrome-A Systematic Review. Srnovršnik T, Virant-Klun I, Pinter B.. (Antioxidants (Basel). 2023 Jul 7;12(7):1398)
- Heavy Metals and Essential Elements in Association with Oxidative Stress in Women with Polycystic Ovary Syndrome-A Systematic Review. Srnovršnik T, Virant-Klun I, Pinter B.. (Antioxidants (Basel). 2023 Jul 7;12(7):1398)
- Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory disease. Tatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016). (REPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.)
- Are Heavy Metal Exposure and Trace Element Levels Related to Metabolic and Endocrine Problems in Polycystic Ovary Syndrome?. Kirmizi, D. A., Baser, E., Turksoy, V. A., Kara, M., Yalvac, E. S., & Gocmen, A. Y. (2020). (Biological Trace Element Research.)
- Serum copper assessment in patients with polycystic ovary syndrome and tubal infertility: A retrospective 5-year study. Liu, Y., Zhang, W., Liu, Z., Zheng, A., Liang, B., Li, H., & Meng, Q. (2024).. (Food Science & Nutrition, 12, 5979–5989.)
- Association of Trace Elements with Polycystic Ovary Syndrome in Women—A Case-Control Study. Srnovršnik, T., Pinter, B., Horvat, M., Snoj Tratnik, J., Falnoga, I., Mazej, D., Verdenik, I., & Virant-Klun, I. (2025).. (Metabolites, 15(2), 79.)
- A Comparative Study of Blood Levels of Manganese, Some Macroelements and Heavy Metals in Obese and Non-Obese Polycystic Ovary Syndrome Patients. H. Mhaibes S, A. Taher M, H. Badr A, H. Badr A.. (Iraqi Journal of Pharmaceutical Sciences [Internet]. 2017 Dec. 26 [cited 2025 Mar. 12];26(2):85-94)
- Levels of Trace Elements in Erythrocytes as Endocrine Disruptors in Obese and Nonobese Women with Polycystic Ovary Syndrome. Pokorska-Niewiada, K., Brodowska, A., Brodowski, J., & Szczuko, M. (2022). (International Journal of Environmental Research and Public Health, 19(2), 976.)
- Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory disease. Tatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016). (REPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.)
- Are Heavy Metal Exposure and Trace Element Levels Related to Metabolic and Endocrine Problems in Polycystic Ovary Syndrome?. Kirmizi, D. A., Baser, E., Turksoy, V. A., Kara, M., Yalvac, E. S., & Gocmen, A. Y. (2020). (Biological Trace Element Research.)
- Heavy Metals and Essential Elements in Association with Oxidative Stress in Women with Polycystic Ovary Syndrome-A Systematic Review. Srnovršnik T, Virant-Klun I, Pinter B.. (Antioxidants (Basel). 2023 Jul 7;12(7):1398)
- Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory disease. Tatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016). (REPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.)
- Levels of Trace Elements in Erythrocytes as Endocrine Disruptors in Obese and Nonobese Women with Polycystic Ovary Syndrome. Pokorska-Niewiada, K., Brodowska, A., Brodowski, J., & Szczuko, M. (2022). (International Journal of Environmental Research and Public Health, 19(2), 976.)
- Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory disease. Tatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016). (REPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.)
- Levels of Trace Elements in Erythrocytes as Endocrine Disruptors in Obese and Nonobese Women with Polycystic Ovary Syndrome. Pokorska-Niewiada, K., Brodowska, A., Brodowski, J., & Szczuko, M. (2022). (International Journal of Environmental Research and Public Health, 19(2), 976.)
- A Comparative Study of Blood Levels of Manganese, Some Macroelements and Heavy Metals in Obese and Non-Obese Polycystic Ovary Syndrome Patients. H. Mhaibes S, A. Taher M, H. Badr A, H. Badr A.. (Iraqi Journal of Pharmaceutical Sciences [Internet]. 2017 Dec. 26 [cited 2025 Mar. 12];26(2):85-94)
- A Comparative Study of Blood Levels of Manganese, Some Macroelements and Heavy Metals in Obese and Non-Obese Polycystic Ovary Syndrome Patients. H. Mhaibes S, A. Taher M, H. Badr A, H. Badr A.. (Iraqi Journal of Pharmaceutical Sciences [Internet]. 2017 Dec. 26 [cited 2025 Mar. 12];26(2):85-94)
- Serum trace elements and heavy metals in polycystic ovary syndrome. Kurdoglu Z, Kurdoglu M, Demir H, Sahin HG. (Hum Exp Toxicol. 2012 May;31(5):452-6.)
- Serum copper assessment in patients with polycystic ovary syndrome and tubal infertility: A retrospective 5-year study. Liu, Y., Zhang, W., Liu, Z., Zheng, A., Liang, B., Li, H., & Meng, Q. (2024).. (Food Science & Nutrition, 12, 5979–5989.)
- Are Heavy Metal Exposure and Trace Element Levels Related to Metabolic and Endocrine Problems in Polycystic Ovary Syndrome?. Kirmizi, D. A., Baser, E., Turksoy, V. A., Kara, M., Yalvac, E. S., & Gocmen, A. Y. (2020). (Biological Trace Element Research.)
- Levels of Trace Elements in Erythrocytes as Endocrine Disruptors in Obese and Nonobese Women with Polycystic Ovary Syndrome. Pokorska-Niewiada, K., Brodowska, A., Brodowski, J., & Szczuko, M. (2022). (International Journal of Environmental Research and Public Health, 19(2), 976.)
- Evaluation of the relationship between polycystic ovary syndrome and intestinal inflammation as measured by fecal calprotectin levels. Bihter Senem Feyzioğlu, Zerrin Avul. (Zeynep Kamil Med J. 2024; 55(4): 207-212)
Sun J, Wang M, Kan Z.
Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization study.Front Endocrinol (Lausanne). 2024
Read ReviewGu Y, Zhou G, Zhou F, Li Y, Wu Q, He H, Zhang Y, Ma C, Ding J, Hua K.
Gut and Vaginal Microbiomes in PCOS: Implications for Women’s Health.Front Endocrinol (Lausanne). 2022.
Read ReviewLiang, Z., Di, N., Li, L., & Yang, D.
Gut microbiota alterations reveal potential gut–brain axis changes in polycystic ovary syndrome.Journal of Endocrinological Investigation. 2021.
Read ReviewDiamanti-Kandarakis, E., & Dunaif, A.
Insulin Resistance and the Polycystic Ovary Syndrome Revisited: An Update on Mechanisms and Implications.Endocrine Reviews, 2012.
Read ReviewZhao, X., Jiang, Y., Xi, H., Chen, L., & Feng, X.
Exploration of the Relationship Between Gut Microbiota and Polycystic Ovary Syndrome (PCOS): a Review.Geburtshilfe Frauenheilkd. 2020
Read ReviewDong J, Rees DA.
Polycystic ovary syndrome: pathophysiology and therapeutic opportunitiesBMJ Med. 2023
Read ReviewSun J, Wang M, Kan Z.
Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization study.Front Endocrinol (Lausanne). 2024
Read ReviewHe, F., & Li, Y.
Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome: a review.Journal of Ovarian Research. 2020.
Read ReviewTremellen, K., & Pearce, K.
Dysbiosis of Gut Microbiota (DOGMA) – A novel theory for the development of Polycystic Ovarian SyndromeMedical Hypotheses, 2012.
Read ReviewDiamanti-Kandarakis, E., & Dunaif, A.
Insulin Resistance and the Polycystic Ovary Syndrome Revisited: An Update on Mechanisms and Implications.Endocrine Reviews, 2012.
Read ReviewSun J, Wang M, Kan Z.
Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization study.Front Endocrinol (Lausanne). 2024
Read ReviewDong J, Rees DA.
Polycystic ovary syndrome: pathophysiology and therapeutic opportunitiesBMJ Med. 2023 Oct 12;2(1):e000548
Read ReviewSun J, Wang M, Kan Z.
Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization studyFront Endocrinol (Lausanne). 2024 Feb 1;15:1280983
Read ReviewZhao, X., Jiang, Y., Xi, H., Chen, L., & Feng, X. (2020)
Exploration of the Relationship Between Gut Microbiota and Polycystic Ovary Syndrome (PCOS): a ReviewGeburtshilfe Und Frauenheilkunde, 80(02), 161–171.
Read ReviewLiang, Z., Di, N., Li, L., & Yang, D. (2021).
Gut microbiota alterations reveal potential gut–brain axis changes in polycystic ovary syndromeJournal of Endocrinological Investigation, 44(8), 1727–1737
Read ReviewSun J, Wang M, Kan Z.
Causal relationship between gut microbiota and polycystic ovary syndrome: a literature review and Mendelian randomization studyFront Endocrinol (Lausanne). 2024 Feb 1;15:1280983
Read ReviewYang, T., Li, G., Xu, Y. et al.
Characterization of the gut microbiota in polycystic ovary syndrome with dyslipidemiaBMC Microbiol 24, 169 (2024)
Read ReviewHe, F., & Li, Y. (2020)
Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome: a reviewJournal of Ovarian Research, 13(1)
Read ReviewLiu, R., Zhang, C., Shi, Y., Zhang, F., Li, L., Wang, X., Ling, Y., Fu, H., Dong, W., Shen, J., Reeves, A., Greenberg, A. S., Zhao, L., Peng, Y., & Ding, X. (2017)
Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary SyndromeFrontiers in Microbiology, 8, 234522.
Read ReviewAbudawood, M., Tabassum, H., Alanazi, A.H. et al.
Antioxidant status in relation to heavy metals induced oxidative stress in patients with polycystic ovarian syndrome (PCOS)Sci Rep 11, 22935 (2021)
Read ReviewQingtao Jiang; Feng Zhang; Lei Han; Baoli Zhu; Xin Liu
Serum Copper Level and Polycystic Ovarian Syndrome: A Meta-AnalysisGynecol Obstet Invest (2021) 86 (3): 239–246.
Read ReviewChen K, Geng H, Liu J, Ye C.
Alteration in gut mycobiota of patients with polycystic ovary syndromeMicrobiol Spectr11:e02360-23
Read ReviewLiu, Y., Zhang, W., Liu, Z., Zheng, A., Liang, B., Li, H., & Meng, Q. (2024).
Serum copper assessment in patients with polycystic ovary syndrome and tubal infertility: A retrospective 5-year studyFood Science & Nutrition, 12, 5979–5989.
Read ReviewTatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016)
Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory diseaseREPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.
Read ReviewSrnovršnik, T., Pinter, B., Horvat, M., Snoj Tratnik, J., Falnoga, I., Mazej, D., Verdenik, I., & Virant-Klun, I. (2025).
Association of Trace Elements with Polycystic Ovary Syndrome in Women—A Case-Control StudyMetabolites, 15(2), 79.
Read ReviewSrnovršnik T, Virant-Klun I, Pinter B.
Heavy Metals and Essential Elements in Association with Oxidative Stress in Women with Polycystic Ovary Syndrome-A Systematic ReviewAntioxidants (Basel). 2023 Jul 7;12(7):1398
Read ReviewSrnovršnik T, Virant-Klun I, Pinter B.
Heavy Metals and Essential Elements in Association with Oxidative Stress in Women with Polycystic Ovary Syndrome-A Systematic ReviewAntioxidants (Basel). 2023 Jul 7;12(7):1398
Read ReviewTatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016)
Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory diseaseREPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.
Read ReviewKirmizi, D. A., Baser, E., Turksoy, V. A., Kara, M., Yalvac, E. S., & Gocmen, A. Y. (2020)
Are Heavy Metal Exposure and Trace Element Levels Related to Metabolic and Endocrine Problems in Polycystic Ovary Syndrome?Biological Trace Element Research.
Read ReviewLiu, Y., Zhang, W., Liu, Z., Zheng, A., Liang, B., Li, H., & Meng, Q. (2024).
Serum copper assessment in patients with polycystic ovary syndrome and tubal infertility: A retrospective 5-year studyFood Science & Nutrition, 12, 5979–5989.
Read ReviewSrnovršnik, T., Pinter, B., Horvat, M., Snoj Tratnik, J., Falnoga, I., Mazej, D., Verdenik, I., & Virant-Klun, I. (2025).
Association of Trace Elements with Polycystic Ovary Syndrome in Women—A Case-Control StudyMetabolites, 15(2), 79.
Read ReviewH. Mhaibes S, A. Taher M, H. Badr A, H. Badr A.
A Comparative Study of Blood Levels of Manganese, Some Macroelements and Heavy Metals in Obese and Non-Obese Polycystic Ovary Syndrome PatientsIraqi Journal of Pharmaceutical Sciences [Internet]. 2017 Dec. 26 [cited 2025 Mar. 12];26(2):85-94
Read ReviewPokorska-Niewiada, K., Brodowska, A., Brodowski, J., & Szczuko, M. (2022)
Levels of Trace Elements in Erythrocytes as Endocrine Disruptors in Obese and Nonobese Women with Polycystic Ovary SyndromeInternational Journal of Environmental Research and Public Health, 19(2), 976.
Read ReviewTatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016)
Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory diseaseREPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.
Read ReviewKirmizi, D. A., Baser, E., Turksoy, V. A., Kara, M., Yalvac, E. S., & Gocmen, A. Y. (2020)
Are Heavy Metal Exposure and Trace Element Levels Related to Metabolic and Endocrine Problems in Polycystic Ovary Syndrome?Biological Trace Element Research.
Read ReviewSrnovršnik T, Virant-Klun I, Pinter B.
Heavy Metals and Essential Elements in Association with Oxidative Stress in Women with Polycystic Ovary Syndrome-A Systematic ReviewAntioxidants (Basel). 2023 Jul 7;12(7):1398
Read ReviewTatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016)
Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory diseaseREPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.
Read ReviewPokorska-Niewiada, K., Brodowska, A., Brodowski, J., & Szczuko, M. (2022)
Levels of Trace Elements in Erythrocytes as Endocrine Disruptors in Obese and Nonobese Women with Polycystic Ovary SyndromeInternational Journal of Environmental Research and Public Health, 19(2), 976.
Read ReviewTatarchuk, T. F., Kosei, N. V., Vetokh, H. V., & Gunkov, S. V. (2016)
Serum micro- and macroelements levels in women with polycystic ovary syndrome associated with pelvic inflammatory diseaseREPRODUCTIVE ENDOCRINOLOGY, (27), 26–29.
Read ReviewPokorska-Niewiada, K., Brodowska, A., Brodowski, J., & Szczuko, M. (2022)
Levels of Trace Elements in Erythrocytes as Endocrine Disruptors in Obese and Nonobese Women with Polycystic Ovary SyndromeInternational Journal of Environmental Research and Public Health, 19(2), 976.
Read ReviewH. Mhaibes S, A. Taher M, H. Badr A, H. Badr A.
A Comparative Study of Blood Levels of Manganese, Some Macroelements and Heavy Metals in Obese and Non-Obese Polycystic Ovary Syndrome PatientsIraqi Journal of Pharmaceutical Sciences [Internet]. 2017 Dec. 26 [cited 2025 Mar. 12];26(2):85-94
Read ReviewH. Mhaibes S, A. Taher M, H. Badr A, H. Badr A.
A Comparative Study of Blood Levels of Manganese, Some Macroelements and Heavy Metals in Obese and Non-Obese Polycystic Ovary Syndrome PatientsIraqi Journal of Pharmaceutical Sciences [Internet]. 2017 Dec. 26 [cited 2025 Mar. 12];26(2):85-94
Read ReviewKurdoglu Z, Kurdoglu M, Demir H, Sahin HG
Serum trace elements and heavy metals in polycystic ovary syndromeHum Exp Toxicol. 2012 May;31(5):452-6.
Read ReviewLiu, Y., Zhang, W., Liu, Z., Zheng, A., Liang, B., Li, H., & Meng, Q. (2024).
Serum copper assessment in patients with polycystic ovary syndrome and tubal infertility: A retrospective 5-year studyFood Science & Nutrition, 12, 5979–5989.
Read ReviewKirmizi, D. A., Baser, E., Turksoy, V. A., Kara, M., Yalvac, E. S., & Gocmen, A. Y. (2020)
Are Heavy Metal Exposure and Trace Element Levels Related to Metabolic and Endocrine Problems in Polycystic Ovary Syndrome?Biological Trace Element Research.
Read ReviewPokorska-Niewiada, K., Brodowska, A., Brodowski, J., & Szczuko, M. (2022)
Levels of Trace Elements in Erythrocytes as Endocrine Disruptors in Obese and Nonobese Women with Polycystic Ovary SyndromeInternational Journal of Environmental Research and Public Health, 19(2), 976.
Read ReviewBihter Senem Feyzioğlu, Zerrin Avul
Evaluation of the relationship between polycystic ovary syndrome and intestinal inflammation as measured by fecal calprotectin levelsZeynep Kamil Med J. 2024; 55(4): 207-212
Read Review