Research Feeds

View All
Characterizing the gut microbiota in females with infertility and preliminary results of a water-soluble dietary fiber intervention study A prebiotic dietary pilot intervention restores faecal metabolites and may be neuroprotective in Parkinson’s Disease Diagnosis of the menopause: NICE guidance and quality standards Causes of Death in End-Stage Kidney Disease: Comparison Between the United States Renal Data System and a Large Integrated Health Care System Factors affecting the absorption and excretion of lead in the rat Factors associated with age at menarche, menstrual knowledge, and hygiene practices among schoolgirls in Sharjah, UAE Cadmium transport in blood serum The non-pathogenic Escherichia coli strain Nissle 1917 – features of a versatile probiotic Structured Exercise Benefits in Euthyroid Graves’ Disease: Improved Capacity, Fatigue, and Relapse Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease A Pilot Microbiota Study in Parkinson’s Disease Patients versus Control Subjects, and Effects of FTY720 and FTY720-Mitoxy Therapies in Parkinsonian and Multiple System Atrophy Mouse Models Dysbiosis of the Saliva Microbiome in Patients With Polycystic Ovary Syndrome Integrated Microbiome and Host Transcriptome Profiles Link Parkinson’s Disease to Blautia Genus: Evidence From Feces, Blood, and Brain Gut microbiota modulation: a narrative review on a novel strategy for prevention and alleviation of ovarian aging Long-term postmenopausal hormone therapy and endometrial cancer

Gut microbiota regulate Alzheimer’s disease pathologies and cognitive disorders via PUFA-associated neuroinflammation. Original paper

Researched by:

  • Dr. Umar ID
    Dr. Umar

    User avatarClinical Pharmacist and Clinical Pharmacy Master’s candidate focused on antibiotic stewardship, AI-driven pharmacy practice, and research that strengthens safe and effective medication use. Experience spans digital health research with Bloomsbury Health (London), pharmacovigilance in patient support programs, and behavioral approaches to mental health care. Published work includes studies on antibiotic use and awareness, AI applications in medicine, postpartum depression management, and patient safety reporting. Developer of an AI-based clinical decision support system designed to enhance antimicrobial stewardship and optimize therapeutic outcomes.

    Read More

November 24, 2025

Researched by:

  • Dr. Umar ID
    Dr. Umar

    User avatarClinical Pharmacist and Clinical Pharmacy Master’s candidate focused on antibiotic stewardship, AI-driven pharmacy practice, and research that strengthens safe and effective medication use. Experience spans digital health research with Bloomsbury Health (London), pharmacovigilance in patient support programs, and behavioral approaches to mental health care. Published work includes studies on antibiotic use and awareness, AI applications in medicine, postpartum depression management, and patient safety reporting. Developer of an AI-based clinical decision support system designed to enhance antimicrobial stewardship and optimize therapeutic outcomes.

    Read More

Last Updated: 2023-01-01

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.

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.

Location
Canada
China
Japan
Taiwan
United States of America
Sample Site
Feces
Species
Homo sapiens

What was studied?

This study investigated how the gut microbiome regulates Alzheimer’s disease (AD) pathologies and cognitive impairment using the 3xTg-AD mouse model. The focus was on microbiome-driven shifts in polyunsaturated fatty acid metabolism, especially arachidonic acid–derived inflammatory lipids, and their impact on the C/EBPβ–AEP (asparagine endopeptidase) pathway, a key driver of amyloid-β and tau cleavage. Analyses included longitudinal microbiome profiling, comparisons between germ-free and specific-pathogen-free mice, fecal microbiota transplantation (FMT) from AD patients and healthy donors, hippocampal RNA-seq, and metabolomics (faeces, serum, and brain). The work emphasises how gut dysbiosis, including Bacteroides enrichment, promotes inflammatory PUFA metabolites that activate microglia and accelerate hallmark AD lesions.

Who was studied?

Experimental models included germ-free, specific-pathogen-free, and human-microbiota-associated 3xTg mice. Human fecal donors were older adults (≥65 years) from nursing facilities, including individuals with clinically diagnosed AD and cognitively healthy controls. All mice were female to control for sex-based microbiome variability. Human-derived microbial communities were successfully engrafted into germ-free mice, allowing evaluation of the causal effects of AD-associated microbiota on neuroinflammation, PUFA metabolism, microglial activation, and cognitive performance.

Most important findings

Loss of gut microbiota dramatically reduced AD pathology, where germ-free mice showed far lower Aβ, Thioflavin-S–positive plaques, AT8-positive tau, and T22-positive tau oligomers. Metabolomics demonstrated substantial reductions in arachidonic-acid–derived metabolites in germ-free mice. Transcriptomics revealed broad downregulation of inflammatory and arachidonic-acid pathways. AD-donor FMT produced powerful pathological effects: ex-germ-free mice colonised with AD microbiota showed increases in Aβ42, tau phosphorylation, microglial activation, and worsening performance on Y-maze cognitive testing. Enrichment of Bacteroides intestinalis, B. fragilis, and B. xylanisolvens aligned with elevated secretion of arachidonic-acid metabolites in vitro. These microbial strains generated PGE2, LTB4, and 12-HHT, directly linking microbiome structure with PUFA-driven neuroinflammation. SCFA supplementation activated the C/EBPβ–AEP axis and worsened AD pathology in germ-free mice, while PGE2-G (a key inflammatory PUFA derivative) amplified microglial maturation.

Key implications

These results show that gut dysbiosis is not just correlated with AD but functionally contributes to its pathogenesis through PUFA-associated neuroinflammation. Specific microbial taxa—especially Bacteroides species with PUFA-metabolising capacity—appear central in producing inflammatory lipids that activate microglia and drive AEP-dependent cleavage of APP and tau. Clinically, this work strengthens the rationale for microbiome-targeted therapies, including dietary modulation of PUFA metabolism, probiotics promoting butyrate-producing species, personalised microbiota restructuring, and potentially carefully selected fecal transplants. Importantly, microbiome signatures involving Bacteroides elevation, reduced Firmicutes, decreased butyrate producers, elevated PGE2/LTB4–pathway activity, and activation of the C/EBPβ–AEP axis emerge as relevant biomarkers for microbiome-based AD risk stratification.

Citation

Chen C, Liao J, Xia Y, et al. Gut microbiota regulate Alzheimer’s disease pathologies and cognitive disorders via PUFA-associated neuroinflammation.Gut. 2022;71(11):2233-2252. doi:10.1136/gutjnl-2021-326269

Fecal Microbiota Transplantation (FMT)

Fecal Microbiota Transplantation (FMT) involves transferring fecal bacteria from a healthy donor to a patient to restore microbiome balance.

Short-chain Fatty Acids (SCFAs)

Short-chain fatty acids are microbially derived metabolites that regulate epithelial integrity, immune signaling, and microbial ecology. Their production patterns and mechanistic roles provide essential functional markers within microbiome signatures and support the interpretation of MBTIs, MMAs, and systems-level microbial shifts across clinical conditions.

Join the Roundtable

Contribute to published consensus reports, connect with top clinicians and researchers, and receive exclusive invitations to roundtable conferences.