Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
BMC Microbiol ; 24(1): 169, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760705

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is an endocrinopathy in childbearing-age females which can cause many complications, such as diabetes, obesity, and dyslipidemia. The metabolic disorders in patients with PCOS were linked to gut microbial dysbiosis. However, the correlation between the gut microbial community and dyslipidemia in PCOS remains unillustrated. Our study elucidated the different gut microbiota in patients with PCOS and dyslipidemia (PCOS.D) compared to those with only PCOS and healthy women. RESULTS: In total, 18 patients with PCOS, 16 healthy females, and 18 patients with PCOS.D were enrolled. The 16 S rRNA sequencing in V3-V4 region was utilized for identifying the gut microbiota, which analyzes species annotation, community diversity, and community functions. Our results showed that the ß diversity of gut microbiota did not differ significantly among the three groups. Regarding gut microbiota dysbiosis, patients with PCOS showed a decreased abundance of Proteobacteria, and patients with PCOS.D showed an increased abundance of Bacteroidota compared to other groups. With respect to the gut microbial imbalance at genus level, the PCOS.D group showed a higher abundance of Clostridium_sensu_stricto_1 compared to other two groups. Furthermore, the abundances of Faecalibacterium and Holdemanella were lower in the PCOS.D than those in the PCOS group. Several genera, including Faecalibacterium and Holdemanella, were negatively correlated with the lipid profiles. Pseudomonas was negatively correlated with luteinizing hormone levels. Using PICRUSt analysis, the gut microbiota community functions suggested that certain metabolic pathways (e.g., amino acids, glycolysis, and lipid) were altered in PCOS.D patients as compared to those in PCOS patients. CONCLUSIONS: The gut microbiota characterizations in patients with PCOS.D differ from those in patients with PCOS and controls, and those might also be related to clinical parameters. This may have the potential to become an alternative therapy to regulate the clinical lipid levels of patients with PCOS in the future.


Subject(s)
Bacteria , Dysbiosis , Dyslipidemias , Gastrointestinal Microbiome , Polycystic Ovary Syndrome , RNA, Ribosomal, 16S , Humans , Polycystic Ovary Syndrome/microbiology , Female , Dyslipidemias/microbiology , Adult , Dysbiosis/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , RNA, Ribosomal, 16S/genetics , Young Adult , Feces/microbiology
2.
Curr Opin Clin Nutr Metab Care ; 36(3): 134-147, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38656809

ABSTRACT

PURPOSE OF REVIEW: The analysis of microbiome in association with female health is today a "hot topic" with the main focus on microbes in the female reproductive tract. Nevertheless, recent studies are providing novel information of the possible influence of the gut microbiome on gynecological health outcomes, especially as we start to understand that the gut microbiome is an extended endocrine organ influencing female hormonal levels. This review summarizes the current knowledge of the gut microbes in association with gynecological health. RECENT FINDINGS: The gut microbiome has been associated with endometriosis, polycystic ovary syndrome, gynecological cancers, and infertility, although there is a lack of consistency and consensus among studies due to different study designs and protocols used, and the studies in general are underpowered. SUMMARY: The interconnection between the gut microbiome and reproductive health is complex and further research is warranted. The current knowledge in the field emphasizes the link between the microbiome and gynecological health outcomes, with high potential for novel diagnostic and treatment tools via modulation of the microenvironment.


Subject(s)
Endometriosis , Gastrointestinal Microbiome , Polycystic Ovary Syndrome , Reproductive Health , Humans , Female , Gastrointestinal Microbiome/physiology , Endometriosis/microbiology , Polycystic Ovary Syndrome/microbiology , Genitalia, Female/microbiology , Genital Neoplasms, Female/microbiology , Infertility, Female/microbiology , Genital Diseases, Female/microbiology
3.
Hum Reprod ; 39(6): 1291-1302, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38614956

ABSTRACT

STUDY QUESTION: How does the gut bacteriome differ based on mood disorders (MDs) in women with polycystic ovary syndrome (PCOS), and how can the gut bacteriome contribute to the associations between these two conditions? SUMMARY ANSWER: Women with PCOS who also have MDs exhibited a distinct gut bacteriome with reduced alpha diversity and a significantly lower abundance of Butyricicoccus compared to women with PCOS but without MDs. WHAT IS KNOWN ALREADY: Women with PCOS have a 4- to 5-fold higher risk of having MDs compared to women without PCOS. The gut bacteriome has been suggested to influence the pathophysiology of both PCOS and MDs. STUDY DESIGN, SIZE, DURATION: This population-based cohort study was derived from the Northern Finland Birth Cohort 1966 (NFBC1966), which includes all women born in Northern Finland in 1966. Women with PCOS who donated a stool sample at age 46 years (n = 102) and two BMI-matched controls for each case (n = 205), who also responded properly to the MD criteria scales, were included. PARTICIPANTS/MATERIALS, SETTING, METHODS: A total of 102 women with PCOS and 205 age- and BMI-matched women without PCOS were included. Based on the validated MD criteria, the subjects were categorized into MD or no-MD groups, resulting in the following subgroups: PCOS no-MD (n = 84), PCOS MD (n = 18), control no-MD (n = 180), and control MD (n = 25). Clinical characteristics were assessed at age 31 years and age 46 years, and stool samples were collected from the women at age 46 years, followed by the gut bacteriome analysis using 16 s rRNA sequencing. Alpha diversity was assessed using observed features and Shannon's index, with a focus on genera, and beta diversity was characterized using principal components analysis (PCA) with Bray-Curtis Dissimilarity at the genus level. Associations between the gut bacteriome and PCOS-related clinical features were explored by Spearman's correlation coefficient. A P-value for multiple testing was adjusted with the Benjamini-Hochberg false discovery rate (FDR) method. MAIN RESULTS AND THE ROLE OF CHANCE: We observed changes in the gut bacteriome associated with MDs, irrespective of whether the women also had PCOS. Similarly, PCOS MD cases showed a lower alpha diversity (Observed feature, PCOS no-MD, median 272; PCOS MD, median 208, FDR = 0.01; Shannon, PCOS no-MD, median 5.95; PCOS MD, median 5.57, FDR = 0.01) but also a lower abundance of Butyricicoccus (log-fold changeAnalysis of Compositions of Microbiomes with Bias Correction (ANCOM-BC)=-0.90, FDRANCOM-BC=0.04) compared to PCOS no-MD cases. In contrast, in the controls, the gut bacteriome did not differ based on MDs. Furthermore, in the PCOS group, Sutterella showed positive correlations with PCOS-related clinical parameters linked to obesity (BMI, r2=0.31, FDR = 0.01; waist circumference, r2=0.29, FDR = 0.02), glucose metabolism (fasting glucose, r2=0.46, FDR < 0.001; fasting insulin, r2=0.24, FDR = 0.05), and gut barrier integrity (zonulin, r2=0.25, FDR = 0.03). LIMITATIONS, REASONS FOR CAUTION: Although this was the first study to assess the link between the gut bacteriome and MDs in PCOS and included the largest PCOS dataset for the gut microbiome analysis, the number of subjects stratified by the presence of MDs was limited when contrasted with previous studies that focused on MDs in a non-selected population. WIDER IMPLICATIONS OF THE FINDINGS: The main finding is that gut bacteriome is associated with MDs irrespective of the PCOS status, but PCOS may also modulate further the connection between the gut bacteriome and MDs. STUDY FUNDING/COMPETING INTEREST(S): This research was funded by the European Union's Horizon 2020 Research and Innovation Programme under the Marie Sklodowska-Curie Grant Agreement (MATER, No. 813707), the Academy of Finland (project grants 315921, 321763, 336449), the Sigrid Jusélius Foundation, Novo Nordisk Foundation (NNF21OC0070372), grant numbers PID2021-12728OB-100 (Endo-Map) and CNS2022-135999 (ROSY) funded by MCIN/AEI/10.13039/501100011033 and ERFD A Way of Making Europe. The study was also supported by EU QLG1-CT-2000-01643 (EUROBLCS) (E51560), NorFA (731, 20056, 30167), USA/NIH 2000 G DF682 (50945), the Estonian Research Council (PRG1076, PRG1414), EMBO Installation (3573), and Horizon 2020 Innovation Grant (ERIN, No. EU952516). The funders did not participate in any process of the study. We have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER: N/A.


Subject(s)
Gastrointestinal Microbiome , Mood Disorders , Polycystic Ovary Syndrome , Humans , Female , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/microbiology , Finland/epidemiology , Middle Aged , Mood Disorders/epidemiology , Adult , Cohort Studies , Case-Control Studies , Feces/microbiology
4.
Reprod Sci ; 31(6): 1508-1520, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38228976

ABSTRACT

Polycystic ovary syndrome (PCOS) is a multifaceted disease with an intricate etiology affecting reproductive-aged women. Despite attempts to unravel the pathophysiology, the molecular mechanism of PCOS remains unknown. There are no effective or suitable therapeutic strategies available to ameliorate PCOS; however, the symptoms can be managed. In recent years, a strong association has been found between the gut microbiome and PCOS, leading to the formulation of novel ideas on the genesis and pathological processes of PCOS. Further, gut microbiome dysbiosis involving microbial metabolites may trigger PCOS symptoms via many mechanistic pathways including those associated with carbohydrates, short-chain fatty acids, lipopolysaccharides, bile acids, and gut-brain axis. We present the mechanistic pathways of PCOS-related microbial metabolites and therapeutic opportunities available to treat PCOS, such as prebiotics, probiotics, and fecal microbiota therapy. In addition, the current review highlights the emerging treatment strategies available to alleviate the symptoms of PCOS.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Polycystic Ovary Syndrome , Probiotics , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/microbiology , Polycystic Ovary Syndrome/therapy , Humans , Female , Gastrointestinal Microbiome/physiology , Dysbiosis/therapy , Probiotics/therapeutic use , Probiotics/administration & dosage , Prebiotics/administration & dosage , Brain-Gut Axis/physiology , Fecal Microbiota Transplantation , Animals
5.
Reprod Sci ; 29(1): 69-83, 2022 01.
Article in English | MEDLINE | ID: mdl-33409871

ABSTRACT

Polycystic ovary Syndrome (PCOS) is one of the most popular diseases that cause menstrual dysfunction and infertility in women. Recently, the relationships between the gastrointestinal microbiome and metabolic disorders such as obesity, type 2 diabetes and PCOS have been discovered. However, the association between the gut microbiome and PCOS symptoms has not been well established. We systematically reviewed existing studies comparing gut microbial composition in PCOS and healthy volunteers to explore evidence for this association. A systematic search was carried out in PubMed, Embase, Cochrane Library, and Web of Science from inception to May 26, 2020, for all original cross-sectional, cohort, or case-control studies comparing the fecal microbiomes of patients with PCOS with microbiomes of healthy volunteers (controls). The primary outcomes were differences in specific gut microbes between patients with PCOS and controls. The search identified 256 citations; 10 studies were included. The total population study of these articles consists of 611 participants (including PCOS group and healthy controls group). Among the included 10 studies, nine studies compared α-diversity, and six studies demonstrated that α-diversity has a significant reduction in PCOS patients. Seven of them reported that there was a significant difference of ß-diversity composition between healthy controls groups and PCOS patients. The most common bacterial alterations in PCOS patients included Bacteroidaceae, Coprococcus, Bacteroides, Prevotella, Lactobacillus, Parabacteroides, Escherichia/Shigella, and Faecalibacterium prausnitzii. No consensus has emerged from existing human studies of PCOS and gut microbiome concerning which bacterial taxa are most relevant to it. In this systematic review, we identified specific bacteria associated with microbiomes of patients with PCOS vs controls. Higher level of evidence is needed to determine whether these microbes are a product or cause of PCOS.


Subject(s)
Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Polycystic Ovary Syndrome/microbiology , Female , Humans
6.
Front Endocrinol (Lausanne) ; 12: 747888, 2021.
Article in English | MEDLINE | ID: mdl-34858330

ABSTRACT

Purpose: Sleeve gastrectomy (SG) is a surgical intervention for polycystic ovary syndrome (PCOS), especially for patients with obesity. Here, we explored the effects of SG on the gut microbiota of rats with PCOS and investigated the association between the intestinal flora and efficacy of SG in PCOS. Methods: Dehydroepiandrosterone (DHEA) injection was administered alone and in combination with a high-fat diet to induce PCOS in rats. SG was performed in rats with PCOS, and the effects of SG on the fecal and gut microbiota and the short-chain fatty acid (SCFA) content were observed. Furthermore, the association among gut microbiota, SCFA content and hyperandrogenism or other hallmarks of PCOS was evaluated. Results: The abundance of Firmicutes reduced and that of Bacteroidetes increased in response to SG in the DHEA-induced PCOS rat model. At the genus level, the abundances of Bacteroides and Blautia increased and those of Ruminococcus, Clostridium, and Alistipes reduced distinctly in the PCOS-SG groups. Moreover, the levels of fecal SCFAs, especially butyric acid, reduced after SG. SG significantly ameliorated PCOS-related symptoms such as hyperandrogenism, disrupted ovary function, and impaired glucose tolerance. Bacteroides and Blautia exhibited a negative correlation and Ruminococcus, Clostridium, and Alistipes exhibited a positive correlation with the levels of fecal SCFAs, luteinizing hormone, testosterone, and inflammatory factors. Conclusions: The amelioration of PCOS-related reproductive and metabolic disorders following SG was associated with the regulation of microbial taxa and SCFA content. Our findings provide a novel perspective on the microbial mechanisms in PCOS after SG.


Subject(s)
Fatty Acids, Volatile/metabolism , Feces/microbiology , Gastrectomy , Gastrointestinal Microbiome/physiology , Polycystic Ovary Syndrome/metabolism , Animals , Dehydroepiandrosterone , Diet, High-Fat , Female , Polycystic Ovary Syndrome/microbiology , Rats
7.
Nutrients ; 13(11)2021 Oct 24.
Article in English | MEDLINE | ID: mdl-34836015

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in reproductive-aged women. Recently, various dietary interventions have been used extensively as a novel therapy against PCOS. In the present study, we show that soy isoflavone metabolites and resistant starch, together with gut microbiota modulations, were successful in decreasing the severity of PCOS-like reproductive features while increasing the expression of gut barrier markers and butyric acid in the gut. In the letrozole-induced PCOS model rats, the intake of both 0.05% soy isoflavones and 11% resistant starch, even with letrozole treatment, reduced the severity of menstrual irregularity and polycystic ovaries with a high concentration of soy isoflavones and equol in plasma. Antibiotic cocktail treatment suppressed soy isoflavone metabolism in the gut and showed no considerable effects on reducing the PCOS-like symptoms. The mRNA expression level of occludin significantly increased with soy isoflavone and resistant starch combined treatment. Bacterial genera such as Blautia, Dorea and Clostridium were positively correlated with menstrual irregularity under resistant starch intake. Moreover, the concentration of butyric acid was elevated by resistant starch intake. In conclusion, we propose that both dietary interventions and gut microbiota modulations could be effectively used in reducing the severity of PCOS reproductive features.


Subject(s)
Gastrointestinal Microbiome , Isoflavones/administration & dosage , Polycystic Ovary Syndrome/microbiology , Polycystic Ovary Syndrome/therapy , Resistant Starch/administration & dosage , Animals , Anti-Bacterial Agents , Biomarkers/analysis , Butyric Acid/metabolism , Disease Models, Animal , Equol/blood , Female , Isoflavones/blood , Letrozole , Polycystic Ovary Syndrome/chemically induced , Rats , Severity of Illness Index , Soy Foods
8.
FASEB J ; 35(11): e21971, 2021 11.
Article in English | MEDLINE | ID: mdl-34653284

ABSTRACT

It has been recently recognized that prenatal androgen exposure is involved in the development of polycystic ovary syndrome (PCOS) in adulthood. In addition, the gut microbiome in adult patients and rodents with PCOS differs from that of healthy individuals. Moreover, recent studies have suggested that the gut microbiome may play a causative role in the pathogenesis of PCOS. We wondered whether prenatal androgen exposure induces gut microbial dysbiosis early in life and is associated with the development of PCOS in later life. To test this hypothesis, we studied the development of PCOS-like phenotypes in prenatally androgenized (PNA) female mice and compared the gut microbiome of PNA and control offspring from 4 to 16 weeks of age. PNA offspring showed a reproductive phenotype from 6 weeks and a metabolic phenotype from 12 weeks of age. The α-diversity of the gut microbiome of the PNA group was higher at 8 weeks and lower at 12 and 16 weeks of age, and the ß-diversity differed from control at 8 weeks. However, a significant difference in the composition of gut microbiome between the PNA and control groups was already apparent at 4 weeks. Allobaculum and Roseburia were less abundant in PNA offspring, and may therefore be targets for future interventional studies. In conclusion, abnormalities in the gut microbiome appear as early as or even before PCOS-like phenotypes develop in PNA mice. Thus, the gut microbiome in early life is a potential target for the prevention of PCOS in later life.


Subject(s)
Androgens/metabolism , Gastrointestinal Microbiome , Polycystic Ovary Syndrome , Prenatal Exposure Delayed Effects/microbiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/microbiology , Pregnancy
9.
Biomed Pharmacother ; 143: 112171, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34536755

ABSTRACT

Several studies reported an increase in cardiovascular risk (CVR) in women with polycystic ovary syndrome (PCOS), considered primarily as the result of the combination of all the clinical features that characterize the syndrome, including hyperandrogenism, insulin resistance, diabetes, obesity chronic low-grade inflammation. Interestingly, in 2012 it has been proposed the so-called DOGMA theory, suggesting the pivotal role played by microbiota alteration in the development of PCOS. Subsequently, several authors evidenced the existence in PCOS women of a marked dysbiosis, which is related to the development of metabolic diseases and cardiovascular complications, mainly due to the production of bacteria-derived metabolites that interfere with various pathways. Among these, trimethylamine-N-oxide (TMAO) is emerging as one of the most important and studied microbiota-derived metabolites related to the increase in CVR, due to its pro-atherosclerotic effect. The purpose of the present review is to summarize the evidence in order to support the hypothesis that, in women with PCOS, dysbiosis might be further involved in enhancement of the CVR via contributing to the increase of circulating TMAO. Although no observational studies on a large number of patients directly investigated the serum levels of TMAO in PCOS women, this manuscript aimed to drive future studies in this field, concurring in providing a novel approach for both comprehension and treatment of the CVR in PCOS.


Subject(s)
Bacteria/metabolism , Cardiovascular Diseases/metabolism , Gastrointestinal Microbiome , Methylamines/blood , Polycystic Ovary Syndrome/metabolism , Animals , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/microbiology , Dysbiosis , Female , Heart Disease Risk Factors , Humans , Hyperandrogenism/epidemiology , Hyperandrogenism/metabolism , Hyperandrogenism/microbiology , Polycystic Ovary Syndrome/epidemiology , Polycystic Ovary Syndrome/microbiology , Prognosis , Risk Assessment , Testosterone/blood
10.
Biomed Pharmacother ; 142: 112094, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34449321

ABSTRACT

Gut microbiota forms a symbiotic relationship with the host and maintains the ecological balance of the internal and external environment of the human body. However, dysbiosis of the gut microbiota and immune deficiency, as well as environmental changes, can destroy the host-microbial balance, leading to the occurrence of a variety of diseases, such as polycystic ovary syndrome (PCOS), type 2 diabetes mellitus (T2DM), and obesity. Meanwhile, diseases can also affect gut microbiota, forming a vicious cycle. The role of the intestinal microbiota in different diseases have been proven by several studies; however, as a common target of PCOS and T2DM, there are few reports on the treatment of different diseases through the regulation of intestinal microbiota as the critical correlation. This review analyzed the common mechanisms of intestinal microbiota in PCOS and T2DM, including the dysbiosis of gut microbiota, endotoxemia, short-chain fatty acids, biotransformation of bile acids, and synthesis of amino acid in regulating insulin resistance, obesity, chronic inflammation, and mitochondrial dysfunction. The possible therapeutic effects of probiotics and/or prebiotics, fecal microbiota transplantation, bariatric surgery, dietary intervention, drug treatment, and other treatments targeted at regulating intestinal microbiota were also elucidated.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Gastrointestinal Microbiome/physiology , Polycystic Ovary Syndrome/physiopathology , Animals , Diabetes Mellitus, Type 2/microbiology , Dysbiosis/complications , Dysbiosis/therapy , Fecal Microbiota Transplantation/methods , Female , Humans , Polycystic Ovary Syndrome/microbiology , Prebiotics/administration & dosage , Probiotics/administration & dosage
11.
Endocrinology ; 162(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-34145455

ABSTRACT

CONTEXT: The key gut microbial biomarkers for polycystic ovarian syndrome (PCOS) and how dysbiosis causes insulin resistance and PCOS remain unclear. OBJECTIVE: To assess the characteristics of intestinal flora in PCOS and explore whether abnormal intestinal flora can affect insulin resistance and promote PCOS and whether chenodeoxycholic acid (CDCA) can activate intestinal farnesoid X receptor (FXR), improving glucose metabolism in PCOS. SETTING AND DESIGN: The intestinal flora of treatment-naïve PCOS patients and hormonally healthy controls was analyzed. Phenotype analysis, intestinal flora analysis, and global metabolomic profiling of caecal contents were performed on a letrozole-induced PCOS mouse model; similar analyses were conducted after 35 days of antibiotic treatment on the PCOS mouse model, and glucose tolerance testing was performed on the PCOS mouse model after a 35-day CDCA treatment. Mice receiving fecal microbiota transplants from PCOS patients or healthy controls were evaluated after 10 weeks. RESULTS: Bacteroides was significantly enriched in treatment-naïve PCOS patients. The enrichment in Bacteroides was reproduced in the PCOS mouse model. Gut microbiota removal ameliorated the PCOS phenotype and insulin resistance and increased relative FXR mRNA levels in the ileum and serum fibroblast growth factor 15 levels. PCOS stool-transplanted mice exhibited insulin resistance at 10 weeks but not PCOS. Treating the PCOS mouse model with CDCA improved glucose metabolism. CONCLUSIONS: Bacteroides is a key microbial biomarker in PCOS and shows diagnostic value. Gut dysbiosis can cause insulin resistance. FXR activation might play a beneficial rather than detrimental role in glucose metabolism in PCOS.


Subject(s)
Gastrointestinal Microbiome , Insulin Resistance , Polycystic Ovary Syndrome/microbiology , Animals , Bacteroides , Biomarkers/metabolism , Case-Control Studies , Chenodeoxycholic Acid/metabolism , Female , Fibroblast Growth Factors/metabolism , Glucose/metabolism , Glucose Tolerance Test , Humans , Letrozole/pharmacology , Metabolomics , Mice , Mice, Inbred C57BL , Phenotype , RNA, Ribosomal, 16S , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Analysis, DNA
12.
Front Endocrinol (Lausanne) ; 12: 662725, 2021.
Article in English | MEDLINE | ID: mdl-33967963

ABSTRACT

Background: Previous studies suggest that the vaginal microbiome is associated with polycystic ovary syndrome (PCOS). However, the clinical manifestations of PCOS are heterogeneous. Whether the vaginal microbiome is related with different clinical symptoms was unknown. Materials and Methods: In this cross-sectional study, 89 female patients with PCOS admitted to Zhongda Hospital (Nanjing, China) were included. Basic demographic information, health-related behaviors, clinical manifestations and sex hormone levels were comprehensively recorded for all patients. Vaginal swabs were acquired for microbiota sequencing of the V3-V4 region of the 16S rRNA gene. Results: The prevalence of bacterial vaginitis and vulvovaginal candidiasis was 15.7% and 13.5%, respectively, within the PCOS patients, which were the most important factors affecting the vaginal microbiome (permutational multivariate analysis of variance test, R2 = 0.108, P = 0.001). The vaginal microbiome was associated with specific clinical manifestations of PCOS, including acanthosis nigricans, intermenstrual bleeding, pregnancy history, testosterone level and anti-müllerian hormone level, with P values < 0.05. The abundance of Lactobacillus crispatus was higher (P = 0.010) while that of Lactobacillus iners was lower (P = 0.036) among PCOS patients with elevated testosterone levels. Other potential bacterial biomarkers were not statistically significant after adjusting for confounding factors. No evidence of associations of other common manifestations of PCOS, such as obesity and acne, with the vaginal microbiome was obtained. Conclusion: Vaginal bacterial species among PCOS patients with variable clinical manifestations, especially differences in testosterone levels, are distinct. Further studies are essential to investigate the microbiota and molecular mechanisms underpinning this disease.


Subject(s)
Acne Vulgaris/epidemiology , Bacteria/classification , Bacterial Infections/epidemiology , Microbiota , Polycystic Ovary Syndrome/complications , Vagina/microbiology , Acne Vulgaris/microbiology , Adult , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Infections/microbiology , China/epidemiology , Cross-Sectional Studies , Female , Follow-Up Studies , Humans , Polycystic Ovary Syndrome/microbiology
13.
Int J Mol Sci ; 22(4)2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33669557

ABSTRACT

Polycystic ovary syndrome (PCOS) is a complex and heterogeneous endocrine disease. The hypothesis that alterations in the microbiome are involved in the genesis of PCOS has been postulated. Aim of this review is to summarize the available literature data about the relationship between microbiome and PCOS. A search on PubMed and Medline databases was performed from inception to November 20Most of evidence has focused on the connection of intestinal bacteria with sex hormones and insulin-resistance: while in the first case, a relationship with hyperandrogenism has been described, although it is still unclear, in the second one, chronic low-grade inflammation by activating the immune system, with increased production of proinflammatory cytokines which interfere with insulin receptor function, causing IR (Insulin Resistance)/hyperinsulinemia has been described, as well as the role of gastrointestinal hormones like Ghrelin and peptide YY (PYY), bile acids, interleukin-22 and Bacteroides vulgatus have been highlighted. The lower genital tract microbiome would be affected by changes in PCOS patients too. The therapeutic opportunities include probiotic, prebiotics and synbiotics, as well as fecal microbiota transplantation and the use of IL-22, to date only in animal models, as a possible future drug. Current evidence has shown the involvement of the gut microbiome in PCOS, seen how humanized mice receiving a fecal transplant from women with PCOS develop ovarian dysfunction, immune changes and insulin resistance and how it is capable of disrupting the secondary bile acid biosynthesis. A future therapeutic approach for PCOS may involve the human administration of IL-22 and bile acid glycodeoxycholic acid.


Subject(s)
Gastrointestinal Microbiome , Polycystic Ovary Syndrome/microbiology , Animals , Diet , Female , Genitalia, Female/microbiology , Hormones/metabolism , Humans , Insulin Resistance , Polycystic Ovary Syndrome/therapy
14.
Genome Med ; 13(1): 22, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33563315

ABSTRACT

To demonstrate the causative role of gut microbiome in human health and diseases, we first need to identify, via next-generation sequencing, potentially important functional members associated with specific health outcomes and disease phenotypes. However, due to the strain-level genetic complexity of the gut microbiota, microbiome datasets are highly dimensional and highly sparse in nature, making it challenging to identify putative causative agents of a particular disease phenotype. Members of an ecosystem seldomly live independently from each other. Instead, they develop local interactions and form inter-member organizations to influence the ecosystem's higher-level patterns and functions. In the ecological study of macro-organisms, members are defined as belonging to the same "guild" if they exploit the same class of resources in a similar way or work together as a coherent functional group. Translating the concept of "guild" to the study of gut microbiota, we redefine guild as a group of bacteria that show consistent co-abundant behavior and likely to work together to contribute to the same ecological function. In this opinion article, we discuss how to use guilds as the aggregation unit to reduce dimensionality and sparsity in microbiome-wide association studies for identifying candidate gut bacteria that may causatively contribute to human health and diseases.


Subject(s)
Disease , Gastrointestinal Microbiome , Health , Databases, Genetic , Female , Gastrointestinal Microbiome/genetics , Humans , Metagenomics , Obesity/genetics , Phylogeny , Polycystic Ovary Syndrome/microbiology , RNA, Ribosomal, 16S/genetics
16.
Bioprocess Biosyst Eng ; 44(5): 953-964, 2021 May.
Article in English | MEDLINE | ID: mdl-32157446

ABSTRACT

Polycystic ovary syndrome (PCOS) is associated with gut microbiota disturbance. Emerging evidence has shown that gut microbiota plays a major role in the development of PCOS. To better understand how the gut microbiota contributes to the development of PCOS, we investigated the influences of high-fat diet and hyperandrogenism, independently or synergistically, have on the gut microbiota in rats. Furthermore, we explored the associations between gut microbiota and hyperandrogenism or other hallmarks of PCOS. Twenty female SD rats were randomized at aged 3 weeks into 4 groups (n = 5, each); HA: PCOS rats fed with ordinary diet; HF: rats with high-fat diet (HFD); HA-HF: PCOS rats fed with HFD; and C: control rats with ordinary diet. PCOS rat model was induced by 5α-dihydrotestosterone (DHT) injection for 6 weeks. The fasting blood glucose (FBG), plasma insulin, testosterone, free testosterone, TNF-α, MDA, SOD, LPS, TLR4, TG, TC, HDL-C, and LDL-C levels were measured. The molecular ecology of the fecal gut microbiota was analyzed by 16S rDNA high-throughput sequencing. The results showed that rats in the HA and HA-HF group displayed abnormal estrous cycles with increasing androgen level and exhibited multiple large cysts with diminished granulosa layers in ovarian tissues. Compare with the C group, relative abundance of the Bacteroidetes phylum decreased significantly in the other groups (P < 0.05). The Chao1 was the highest in the group C and significantly higher than the HA-HF group (P < 0.05). T, FT, insulin, MDA, LPS, and TNF-α levels had the negative correlation with the richness of community (Chao1 index) in the gut. The rats in the HF and HA-HF groups tended to have lower Shannon and Simpson indices than the C group (P < 0.01, respectively). However, there were no significant differences between C group and the HA group in the Shannon and Simpson values. Beta diversity analysis was then performed based on a weighted UniFrac analysis. The PCoA plots showed a clear separation of the C group from the other groups. ANOSIM analysis of variance confirmed that there were statistically significant separations between the C group and the HA, HA-HF, and HF groups (P < 0.01, respectively). These results showed that DHT with HFD could lower diversity of the gut microbial community. Both HFD and DHT could shift the overall gut microbial composition and change the composition of the microbial community in gut. Furthermore, our analyses demonstrated that the levels of TG, MDA, TNF-α, LPS, TLR4, T, FT, FINS, and HDL-C were correlated with the changes of in the gut microbiome. HFD and DHT were associated with the development and pathology of PCOS by shaping gut microbial communities.


Subject(s)
Diet, High-Fat/adverse effects , Dihydrotestosterone/metabolism , Gastrointestinal Microbiome , Polycystic Ovary Syndrome , Animals , Female , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/microbiology , Rats , Rats, Sprague-Dawley
17.
J Clin Endocrinol Metab ; 106(3): 858-871, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33205157

ABSTRACT

CONTEXT: Despite the gut microbiome being widely studied in metabolic diseases, its role in polycystic ovary syndrome (PCOS) has been scarcely investigated. OBJECTIVE: Compare the gut microbiome in late fertile age women with and without PCOS and investigate whether changes in the gut microbiome correlate with PCOS-related metabolic parameters. DESIGN: Prospective, case-control study using the Northern Finland Birth Cohort 1966. SETTING: General community. PARTICIPANTS: A total of 102 PCOS women and 201 age- and body mass index (BMI)-matched non-PCOS control women. Clinical and biochemical characteristics of the participants were assessed at ages 31 and 46 and analyzed in the context of gut microbiome data at the age of 46. INTERVENTION: (s): None. MAIN OUTCOME MEASURE(S): Bacterial diversity, relative abundance, and correlations with PCOS-related metabolic measures. RESULTS: Bacterial diversity indices did not differ significantly between PCOS and controls (Shannon diversity P = .979, unweighted UniFrac P = .175). Four genera whose balance helps to differentiate between PCOS and non-PCOS were identified. In the whole cohort, the abundance of 2 genera from Clostridiales, Ruminococcaceae UCG-002, and Clostridiales Family XIII AD3011 group, were correlated with several PCOS-related markers. Prediabetic PCOS women had significantly lower alpha diversity (Shannon diversity P = .018) and markedly increased abundance of genus Dorea (false discovery rate = 0.03) compared with women with normal glucose tolerance. CONCLUSION: PCOS and non-PCOS women at late fertile age with similar BMI do not significantly differ in their gut microbial profiles. However, there are significant microbial changes in PCOS individuals depending on their metabolic health.


Subject(s)
Gastrointestinal Microbiome/physiology , Metabolic Diseases/etiology , Polycystic Ovary Syndrome/microbiology , Adult , Cardiometabolic Risk Factors , Case-Control Studies , Cohort Studies , Female , Finland , Humans , Metabolic Diseases/microbiology , Middle Aged , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/metabolism
18.
Eur J Clin Invest ; 51(4): e13417, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32991745

ABSTRACT

BACKGROUND: Limited available animal and human data suggest an association between dysbiosis of gut microbiota and PCOS. We aimed to determine whether gut microbiota in lean women with PCOS shows any alterations compared to healthy women. MATERIALS AND METHODS: Twenty-four lean patients with PCOS phenotype A according to the Rotterdam 2003 diagnostic criteria and 22 BMI-matched healthy women were included in this study. Anthropometric, hormonal and biochemical measurements were carried out in all participants. 16S rRNA gene V3-V4 region amplicon sequencing was performed on stool samples. Preprocessing of the raw data was performed using QIIME, and both QIIME and R packages were used for microbiome analysis. RESULTS: Bacterial richness and diversity did not show a significant difference between patients and controls. Beta diversity was similar between the groups. However, Erysipelotrichaceae, Proteobacteria, Gammaproteobacteria, Enterobacteriaceae, Planococcaceae, Gemmules and Bacillales were significantly abundant in PCOS group according to LEfSe analysis. Clostridium cluster XVII showed increased abundance in patient group, while Clostridium sensustricto and Roseburia were decreased compared to controls. Random forest prediction analysis revealed Clostridium cluster XIVb as the most discriminative feature of patient group and Roseburia for healthy controls. Testosterone and androstenedione were negatively correlated with alpha and phylogenetic diversity. CONCLUSIONS: Our results suggest that gut microbiome of lean PCOS patients with full phenotype shows compositional alterations with similar bacterial richness and diversity compared to controls and that hyperandrogenism is associated with dysbiosis.


Subject(s)
Gastrointestinal Microbiome/genetics , Polycystic Ovary Syndrome/microbiology , Androstenedione/blood , Bacillales , Body Mass Index , Case-Control Studies , Clostridium , Enterobacteriaceae , Female , Firmicutes , Gammaproteobacteria , Humans , Planococcaceae , Polycystic Ovary Syndrome/blood , Proteobacteria , RNA, Ribosomal, 16S/genetics , Testosterone/blood , Young Adult
19.
Biomed Pharmacother ; 133: 110958, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33171400

ABSTRACT

Polycystic ovarian syndrome (PCOS), characterized by chronic anovulation and hyperandrogenaemia, is a complex endocrine and metabolic disorder commonly seen in women of reproductive age. Multiple factors, including the intestinal microbiome, affect the pathogenesis and development of PCOS. However, the specific mechanisms by which gut microbes play a role in PCOS remain elusive. This review summarizes recent research about the transformational changes in gut microbes revealed in PCOS patients and the possible mechanisms and pathways by which the intestinal microbiome exerts influence on PCOS progression and phenotypes. In addition to the intestinal microbiome, evidence from animal studies suggests changes in the vaginal microbiome under PCOS conditions. The alteration of microbiome could affect oestrus cycle and PCOS phenotypes. Microbiome is closely associated with medicine and therapeutic approaches. Microbiome influences drug and therapy response and itself is a new source of therapy. Accurate modulation of the intestinal and vaginal microbiome is a potential therapy for PCOS patients. Future studies are required to elucidate the specific role of each particular genera of microbiota and the mechanism by which microbiome impacts the pathogenesis, progression and phenotypes of PCOS.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Polycystic Ovary Syndrome/microbiology , Animals , Bacteria/growth & development , Dysbiosis , Female , Host-Pathogen Interactions , Humans , Phenotype , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Polycystic Ovary Syndrome/therapy , Vagina/microbiology
20.
Femina ; 49(10): 631-635, 2021. ilus
Article in Portuguese | LILACS | ID: biblio-1358197

ABSTRACT

Objetivo: Revisar a implicação e a relação existente entre a microbiota intestinal e a síndrome do ovário policístico (SOP). Métodos: Trata-se de uma revisão sistemática de artigos das bases de dados PubMed, Cochrane e Science Direct dos últimos cinco anos, nos idiomas inglês, português e espanhol. Resultados: A disbiose da microbiota intestinal ativa o sistema imunológico do hospedeiro. Tal ativação interfere na função do receptor de insulina, causando hiperinsulinemia, o que aumenta a produção de androgênio ovariano e dificulta o desenvolvimento de um folículo saudável. Além disso, pacientes com SOP apresentam o perfil taxonômico alterado, o qual se associou inversamente com excesso de andrógenos e inflamação da SOP. Foi evidenciado que o uso de probióticos pode regular a resposta inflamatória, diminuir os níveis totais de testosterona e contribuir para que a SOP não prejudique uma possível gravidez. Conclusão: Essa revisão sugere que há íntima associação entre a disbiose microbiana e as alterações patológicas que ocorrem na SOP. Assim, a suplementação de probióticos em tais pacientes pode ter grandes benefícios, como melhora dos sintomas e redução das repercussões da doença.(AU)


Objective: To review the implication and the relationship between the intestinal microbiota and polycystic ovary syndrome. Methods: This is a systematic review of articles from the PubMed, Cochrane and Science Direct databases, from the last five years, in English, Portuguese and Spanish. Results: Dysbiosis of the intestinal microbiota activates the host's immune system. Such activation interferes with the function of the insulin receptor, causing hyperinsulinemia, which increases the production of ovarian androgens and hinders the development of a healthy follicle. In addition, patients with PCOS have an altered taxonomic profile, which is inversely associated with excess androgens and PCOS inflammation. It was evidenced that the use of probiotics can regulate the inflammatory response, decrease the total testosterone levels and contribute so that PCOS does not harm a possible pregnancy. Conclusion: This review suggests that there is a close association between microbial dysbiosis and pathological changes that occur in PCOS. Thus, supplementation of probiotics in such patients can have great benefits, such as improving symptoms and reducing the repercussions of the disease.(AU)


Subject(s)
Humans , Female , Polycystic Ovary Syndrome/microbiology , Gastrointestinal Microbiome , Gonadal Steroid Hormones , Insulin Resistance , Databases, Bibliographic , Dysbiosis
SELECTION OF CITATIONS
SEARCH DETAIL
...