The gut microbial composition in polycystic ovary syndrome with insulin resistance: Findings from a normal-weight Population

DOI: https://doi.org/10.21203/rs.3.rs-121236/v1

Abstract

Background

A few studies have reported on the relationship between intestinal flora dysbiosis and clinical characteristics in polycystic ovary syndrome (PCOS). But the structure and characteristics of gut microbiota in PCOS have not been fully elucidated.

Objective

To analyze the composition of the Intestinal flora population in normal-weight women with PCOS and insulin resistance(IR) compared to PCOS alone and healthy women.

Methods

The study included 14 PCOS patients with insulin resistant(PCOS-IR) and 12 PCOS alone༈PCOS-NIR༉,and 10 age- and body mass index-matched healthy control women. BMI: 18.5–23.9 kg/m2. The bacterial 16S rDNA V3-V4 fragment was amplified and sequenced. The sequencing data were analyzed for species annotation, community diversity, inter-group differences, to explore the composition characteristics of gut microbial of the subjects and their correlation with clinical parameters.

Results

No significant difference in diversity was observed between PCOA and sample cluster analysis among the three groups (Beta-diversity) and Alpha-diversity. The relative abundance of Rothia, Ruminococcus, and Enterococcus were significantly higher in the PCOS-IR group than in the other two groups (P < 0.05), whereas the abundance of the Prevotella was dramatically decreased (P < 0.05). The abundance of Enterococcus was positively correlated with waist circumference, hip circumference, diastolic blood pressure, and insulin resistance index. Meanwhile, Rothia abundance was positively associated with waist circumference and free fatty acids.

Conclusion

The gut microbial composition of PCOS patients with insulin resistance is different from that of PCOS alone and healthy women. The difference is correlated with the clinical characteristics of PCOS, with regards to insulin resistance, abdominal obesity, free fatty acids, and other indicators. PCOS-IR patients have an increased abundance of Enterococcus. It may affect the intestinal environment of the host by enriching the metabolic pathways related to insulin resistance, leading to the occurrence and development of PCOS.

1. Introduction

Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disease in women, which is related to hirsutism, hyperandrogenism, ovulation dysfunction, menstrual disorders, and infertility[1]. About 50%-70% of cases of anovulatory infertility in patients are linked to PCOS[2], especially those accompanied by low ovulation induction rate, low pregnancy rate, and high abortion rate. PCOS is associated with several metabolic disorders, including insulin resistance (IR), obesity, cardiovascular disease, diabetes, and other long-term metabolic syndromes. At present, the etiology and pathogenesis of PCOS are still unknown, which may involve lifestyle, neuroendocrine, genetic factors, immune and metabolic dysfunction[3]. Insulin resistance (IR) is considered to be the main pathological basis of reproductive dysfunction in polycystic ovary syndrome and has nothing to do with obesity[48]. On the one hand, insulin resistance and hyperinsulinemia affect testosterone synthesis and secretion, while hyper androgen levels can lead to hirsutism, acne, ovulation disorders and menstrual disorders; on the other hand, insulin resistance can have a long-term and serious effect on metabolism in patients with polycystic ovary syndrome[9].

Intestinal flora,the "second genome" obtained by the human body, co-evolves with the host to help promote metabolism and immune response[10]. Evidence indicates that gut microbiome disorders are closely related to the occurrence and development of metabolic diseases, including PCOS[11, 12]. Also, many studies have linked the appearance of IR to gut microbiota dysbiosis[13, 14]. The changes of intestinal flora may affect insulin sensitivity by regulating chronic inflammation mediated by LPS, branched chain amino acid (BCAA), short chain fatty acid (SCFA) and bile acid metabolism, and stimulating the secretion of enterocerebral peptides, resulting in insulin resistance and hyperinsulinemia[1517]. The structure and characteristics of intestinal microbiota in patients with PCOS have not been fully elucidated[1822]. Also, only a few studies have reported on the relationship between intestinal flora dysbiosis and clinical characteristics and metabolism in patients with PCOS.

Intestinal flora imbalance can cause IR, which is closely linked to the occurrence of PCOS. Herein, excluding the influence of obesity, we conducted a pilot study to examine the correlation between gut microbiota, insulin resistance, and clinical characteristics of PCOS patients. These findings may provide new insights into understanding the mechanism of occurrence and development of PCOS, which could accelerate the formulation of new strategies for the prevention and treatment of PCOS.

2. Materials And Methods

Participants

A total of 26 women with Polycystic ovary syndrome (PCOS) aged 18–35 years who visited the Department of Assisted Reproduction(Xiangya Hospital, Central South University)between August 2019 to December 2019 were recruited. Also, we recruited 10 normal women who visited the same department during the same period to have assisted reproduction due to the "male-factor" or "fallopian tube factor" as the control group. This study was approved by the Ethics Committee of the Department of Assisted Reproduction(Xiangya Hospital,Central South University,)as well as the China Registered Clinical Trial Ethics Review Committee (Ethical Review No.: CHiECRT1900028223). All participants provided written informed consent. The participants were non-obese women(BMI 18.5–23.9 kg/m2)[23, 24]. PCOS diagnosis was conducted as per the Rotterdam criteria revised on the 2003 Conference; PCOS was diagnosed if two out of the following three features were present: Oligoovulation or amenorrhea; clinical manifestations of androgen and/or biochemical hyperandrogenism (HA); polycystic ovary: ultrasound reports show that the follicles of unilateral or bilateral ovaries with a diameter of 2-9mm are larger than 12, and/or the volume of the ovary ≥10ml[7]. Individuals excluded from one of the following conditions: Cushing syndrome, congenital adrenocortical hyperplasia, androgen-secreting tumors. None of the subjects were treated with hormone drugs, insulin sensitizers, antibiotics, probiotics and probiotics, traditional Chinese medicines, and immunosuppressants less than three months before the commencement of this study. Women in the control group had no history of menstrual disorder, endocrine diseases, or diagnosed PCOS according to the Rotterdam criteria. The HOMA-IR index was calculated as follows: fasting insulin (FINs, mIU/L)﹡fasting plasma glucose (FPGs, mmol/L)/22.5. Insulin resistance was defined as fasting insulin>10mU/ml, or HOMA-IR>1.66, or abnormal insulin release curve (insulin peak more than ten times the basic value; insulin peak delayed to 1h after taking sugared water; area under the insulin curve increased; insulin level not returning to the normal fasting level 3h after taking sugared water), but with normal fasting blood glucose and glucose tolerance levels [25, 26].

Sampling

Data regarding anthropometry and metabolic parameters were collected for all participants. These included: 1)measurements, including height, weight, waist circumference, hip circumference, and blood pressure, 2)detection of biochemical indicators, such as the sex hormone levels, including progesterone (P), prolactin (PRL), testosterone(T), luteinizing hormone (LH), follicle-stimulating hormone (FSH), oestradiol (E2), sex binding globulin (SHBG), Anti-Mullerian hormone (AMH), and glucose and insulin levels during an oral glucose tolerance test on the third day of the menstrual cycle (In the case of amenorrhea patients, blood samples were collected on any day of the menstrual cycle); meanwhile, biochemical indexes, such as Triglyceride (TG), total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL), total bile acid, free fatty acid; inflammatory markers such as C-reactive protein (CRP), interleukin-6 (IL-6), tumor necrosis factor (TNF-α) were measured. 3) Fecal samples were collected after menstruation, and the samples were immediately frozen and stored at -80℃ until analysis.

DNA extraction and PCR amplification

Microbial DNA was extracted from stool samples using a TIANgen stool DNA kit, following the manufacturer's instructions. The 16S rDNA V3+V4 region of the ribosomal RNA gene was amplified by polymerase chain reaction (PCR). The V3–V4 variable region of the bacterial 16sRNA was amplified via PCR using (5′-CCTACGGRRBGCASCAGKVRVGAAT-3′) and (5′-GGACTACNVGGGTWTCTAATCC-3′) primers under the following conditions: denaturing at 94°C for 3 minutes followed by 24 cycles of 94°C for 5 seconds, 57°C for 90 seconds, and 72°C for 10 seconds, then a final extension at 72°C for 5 minutes. Each PCR reaction mix (25 µL) included 2.5 µL TransStart Buffer, 2 µL dNTPs, 1 µL primer (2 µM), 0.5 µL TransStart Taq DNA and 20 ng of DNA template. The PCR products were assessed using 1.5% agarose gel electrophoresis, quantified by Qubit3.0 Fluorometer (Invitrogen, Carlsbad, CA), then pair-end sequenced on Illumina Miseq PE250 platform (Illumina, San Diego, CA, USA).

Next-generation sequencing

Filtration of raw tags was performed using QIIME (V1.9.1) to dislodge the noisy sequences[27]. The filtered clean tags were searched against the Gold database to identify chimeric tags, which were then removed using the UCHIME algorithm to obtain Effective Tags. Based on the Effective Tags of each sample, the OUT(Operational Taxonomic Units) were clustered using QIIME software (Version 1.9.1) based on the GreenGene database[27-29]. Alpha diversity was reflected by Chao1, Observed OTUs,Simpson, and Shannon indexes. Beta diversity was presented on principal coordinates analysis (PCoA) charts created using PCoA statistical analysis method with R language(version 3.3.3). Based on the species abundance table, the P-value was obtained by Kruskal–Wallis H test analysis and then modified by Benjamini and Hochberg False Discovery Rate method to obtain the Q value. The species with significant differences between groups were obtained with Q value < 0.05 as the threshold[30]. Linear discriminant analysis (LDA) effect size (LEfSe) was used to identify the bacterial taxa and metabolism-associated clinical parameters with significant differences between groups. Logarithmic LDA values > 2.0 and P < 0.05 were set as the threshold for differential flora identification.

Statistical analysis of clinical data

Data were analyzed using SPSS 22.0. Quantitative demographic and clinical data with normal distribution were presented as the mean ± standard deviations (SD). Unpaired t-test was used to determine the difference between two groups. Qualitative demographic and clinical data were expressed as percentages and analyzed using the chi-square test. The Kolmogorov-Smirnov test of normality was applied to all data sets. Data that did not conform to normal distribution were analyzed using Mann-Whitney test. A probability (p) value of < 0.05 was used to determine statistical significance.

3. Results

Clinical characteristics of the participants

Feces samples were collected from all patients. Table 1 summarizes the clinical, hormonal and metabolic data of the recruited participants. There were no significant differences in age, height, or BMI among the three groups (P > 0.05). The menstrual cycle, waist circumference, and waist-to-hip ratio of the PCOS group were significantly higher than those of the HC group. The waist-to-hip ratio of the PCOS-IR group was higher than that of the other two groups (IR VS NIR: 0.85±0.04 VS 0.82±0.03, IR VS HC:0.85±0.04 VS 0.77±0.03 P<0.05). Regarding sex hormones, patients with PCOS exhibited higher levels of T, LH, LH/FSH, and lower levels of estradiol, relative to the control patients (P<0.001); however, there was no significant difference between the PCOS-IR group and the PCOS-NIR group. Concerning glucose and plasma lipid levels, although fasting glucose was not different among the three groups, women with PCOS had higher fasting insulin, glycosylated hemoglobin, and homeostasis model assessment of insulin resistance (HOMA-IR). Notably, the PCOS-IR group had higher HOMA-IR values(P<0.05). Compared with healthy individuals, the plasma levels of pro-inflammatory cytokines in PCOS patients, including IL-6 and TNF-a, were significantly higher. The PCOS-IR group showed higher levels of C-reactive protein, Il-6, TNF-α, and free Fatty acid (FFA)(P<0.05).

Effect of PCOS on gut microbiota diversity

These sequences comprised 297 OTUs clustered at a 97% similarity level. The dilution curve and Shannon-Wiener curve were used to judge whether the sequencing quantity was sufficient and estimate the species richness (Fig. 1A). The curve tended to flatten, indicating that the sequencing depth was sufficient to reflect the species diversity of the samples. We drew the Venn diagram of OTU number distribution of the three groups of samples to intuitively reflect the common and unique characteristics between the groups (Fig. 1B). Alpha diversity analysis showed that, compared with the HC group, observed OTUs and Chao1 index were decreased in the PCOS-IR group and the PCOS-NIR group, but the differences were not statistically significant (Fig. 1C and 1D). The Beta diversity analysis showed that the principal coordinate analysis (PCoA analysis) and sample clustering analysis were similar among the three groups of samples (Fig. 1E and 1F).

Gut microbiota composition among PCOS patients

At the phylum level, FIG. 2 shows the phylum abundance distribution in the bacterial kingdom of the 36 samples, among which Firmicutes were the most abundant, followed by Proteobacteria, then Actinobacteria. The overall abundance of other bacteria was about 1%. The abundance of Fusobacteria and Verrucomicrobia differed between the PCOS group and the HC group, but the difference was not significant (P > 0.05) (Fig 2). At the family level, significant differences were observed in the abundance of Lactobacillus, Enterococcaceae, Peptostreptococcaceae, and Micrococcaeae among the three groups. In the PCOS-IR group, the abundance of Peptostreptococcaceae, Enterococcaceae, and Micrococcaeae were higher than that of the other two groups. Lactobacillaceae was the highest in the PCOS-NIR group (Fig 3). Compared with the HC group, PCOS patients exhibited a higher abundance of Enterococcus. The relative abundance of Rothia, Ruminococcus, Lachnospira, and Enterococcus was significantly higher in the PCOS-IR patients than in the other two groups (P < 0.05), whereas the abundance of the Prevotella was dramatically decreased (P < 0.05). Lactobacillus and Akkermansia were more abundant in the PCOS-NIR group than in the PCOS-IR and HC groups (P < 0.05) (Fig 4).

Next, to further identify species bacterial taxa with significant differences among the three groups, we used LEfSe multilevel species discrimination and LDA. Rothia and Lactobacillus were identified as the characteristic microbiota in the PCOS-IR and the PCOS-NIR groups, respectively, whereas the dominant bacteria in the HC group was Prevotella(LDA score > 2.0 and P<0.05)(Fig 5).

Correlations between gut microbiota and metabolic parameters or sex hormones

At the genera level, Enterococcus was positively correlated with waist circumference, hip circumference, diastolic blood pressure, and HOMA-IR index. Rothia was positively correlated with waist circumference and free fatty acid (FFA) (P<0.05) (Fig 6).

4. Discussion

Gut microbial plays a vital role in regulating energy storage and human metabolism. As such, substantial focus has been directed to microbiota-targeted agents as a new target for the treatment of polycystic ovary syndrome (PCOS) and related metabolic diseases. Several studies have found variations in gut microbiota composition between PCOS patients and healthy people. Besides, obese PCOS patients have been shown to exhibit more severe gut dysbiosis. However, the precise mechanism underlying the relationship between gut microbiota and the occurrence and development of PCOS has not been fully revealed. Insulin resistance (IR) is considered the important pathological basis of reproductive dysfunction in patients with PCOS [4-7]. 50% of patients with PCOS have IR, independent of obesity[31, 32]. However, there is limited information on the roles played by intestinal flora in the development of IR and its link with PCOS. Herein, our results revealed that gut dysbiosis was more severe in PCOS patients with insulin resistance than in the PCOS-NIR and HC groups. Furthermore, several taxa at the phylum level were related to the clinical characteristics of PCOS and were significantly correlated with metabolic biomarkers, including HOMA-IR, abdominal obesity, free fatty acids, and other indicators.

Moreover, the results of the study showed that the composition of gut microbiota of PCOS patients with normal BMI was changed, but there was no significant difference in α-diversity among the three groups. It is noteworthy that several studies have reported conflicting results regarding the composition and function of the intestinal flora in PCOS patients. According to a recent meta-analysis, decreased intestinal microbiome diversity and changes in diversity are closely associated with obesity in humans [33, 34]. Previous studies have reported a significant decrease in gut microbiota diversity in PCOS patients or letrozole-induced mouse models[19, 20, 35]. Liu et al. found that obese PCOS patients had the lowest diversity of gut microbiota [19]. Meanwhile, studies have shown that sex hormones are related to changes in gut microbiom[36, 37]. Torres et al. found that PCOS patients exhibited a lower diversity of gut microbiota than healthy controls, and that total testosterone levels were associated with reduced diversity[20]. However, Insener et al. did not find a decrease in the diversity of gut microbiota in all PCOS patients with hyperandrogenemia, whether obese or not. and diversity may be different from PCOS diagnostic criteria[38]. Similarly, in this study, β- diversity of gut microbiota did not differ significantly among the three groups of samples according to weighted and unweighted clustering analysis. In a recent study, weighted UniFrac range-based hierarchical clustering and PCoA analysis indicated a clear distinction between the HC group and the IR group, whereas the NIR group could not be distinguished from the HC and IR groups[39]. Although there is no agreed standard for choosing sample size in microbiome studies, a study estimated that a sample size of 10 subjects per unweighted group (total sample size of 30) and 20 subjects per weighted group (total sample size of 60) may provide accurate statistical results for weighted analysis[40]. The α and β diversity of intestinal flora may be influenced by sex, sex hormones, and obesity. In this study, the BMI of all the subjects was within the normal range, and thus the effect of obesity itself on the gut microbiota composition of the PCOS patients could not be considered. Maybe due to the small sample size, a more definite answer should be given after increasing the sample size in the future.

According to the analysis of the structural composition of gut microbiota conducted in this study, the three groups were mainly composed of Bacteroidetes and Firmicutes at the phylum level. At the genus level, the relative abundance of Enterococcus in the PCOS group increased significantly (P<0.05) and was highest in the IR group. Enterococcus is a common gram-positive bacteria and can be divided into five categories according to phylogenetic similarity. Among them, Enterococcus faecalis and Enterococcus faecium are the major pathogenic bacteria to humans[41, 42]. Although the causal role of genus Enterococcus in the occurrence and development of metabolic diseases has not been fully revealed, previous studies have found that Enterococcus is more abundant in the gut microbiota of obese children and adolescents [43], as well as in mice under a high-fat/high-sugar "Western" diet[44]. A recent study found that Enterococcus can regulate the level of incretin hormone glucagon-like peptide-1 (GLP-1). GeIE secreted by E. fasecalis can degrade GLP-1 (GLP-1), leading to abnormal insulin secretion[45]. At the same time, GeIE can degrade intestinal gastric inhibitory peptides (just like leptin), and thereby interfere with the metabolism of the host. Similarly, in our study, Enterococcus was the most abundant genera in the PCOS-IR group, and its abundance was positively correlated with insulin resistance index. GLP-1 plays a role in regulating glucose homeostasis and reducing appetite in the body. Considering the vital role GLP-1 plays in the development of type 2 diabetes and other metabolic diseases, we speculated that Enterococcus could influence the occurrence and development of PCOS by regulating the GLP-1 signaling pathway, especially in patients with IR. Studies have shown that after oral glucose tolerance test, GLP-1 activity in lean PCOS patients is usually lower than in healthy women [46]. The use of GLP-1 receptor agonists in the treatment of PCOS patients can improve symptoms and reduce metabolic complications by reducing weight and insulin resistance[47, 48]. Insulin secretion and gastric emptying are affected by intestinal flora environment and intestinal flora imbalance. Estelle et al. proved the important role of intestinal flora in controlling GLP-1-induced insulin secretion and gastric emptying in mice[49]. Therefore, the role of Enterococcus in the regulation of GLP-1 level in PCOS patients should be investigated further.

The critical bacterial genus in the intestinal tract of patients with PCOS was identified using LEfSe multilevel species discrimination and LDA. According to the results, Rothia played vital roles in the PCOS-IR group, whereas Prevotella was the dominant bacterial group in the HC group. Rats fed a high-fat diet have been shown to exhibit significantly increased fat, reduced insulin sensitivity, increased abundance of esophageal Rothia, and Rothia were associated with fasting blood glucose and insulin. These observations were associated with the expression of inflammatory genes and fatty acid transport and metabolism in the esophagus[50]. Changes in Rothia flora in the adolescent oral cavity are associated with obesity[51]. Women with gestational diabetes have also shown an increase in Rothia abundance, relative to women with normal blood sugar[52]. In this study, Rothia had significant advantages in the PCOS-IR group, and its abundance was positively correlated with waist circumference and free fatty acid (FFA). Previous studies have shown that non-obese PCOS patients with insulin resistance have a more obvious centrality distribution of fat [37]. In our study, PCOS patients with normal BMI had abdominal fat accumulation. Besides, Rothia abundance in the PCOS-IR group was related to abdominal obesity. Intestinal flora may participate in the process of visceral fat metabolism, releasing too much free fatty acids, increasing lipotoxicity, and reducing insulin sensitivity. Prevotella is a bacterium that produces short-chain fatty acids (SCFA), regulates the uptake of nutrients and hormone levels in the gut. Also, it participates in energy metabolism, and its decreased abundance is significantly associated with increased testosterone and pro-inflammatory cytokines[39]. In this study, the HC group had the highest abundance of Prevotella, maintaining the balance of intestinal flora, whereas the NIR group and IR group had a reduced abundance of Prevotella. However, we did not observe a correlation between the decreased abundance of the Prevotella and the sex hormones and biochemical indicators, which could be due to a small sample size used herein. The relationship between PCOS and intestinal flora is complex and could be related to genetic, lifestyle, and environmental factors [53, 54]. In this study, we excluded the influence of smoking, taking antibiotics and probiotics on intestinal flora in the included subjects. However, different backgrounds (such as region, race, lifestyle, and diet habit) of the subjects could be responsible for the differences in the PCOS-related intestinal flora[55].

5. Conclusion

In conclusion, we analyzed the composition of the intestinal microbiota in PCOS patients with normal BMI combined with insulin resistance and its relationship with clinical characteristics. Notably, no significant difference was found in intestinal microbial diversity among the three groups of patients. However, the abundance of Enterococcus and Rothia increased significantly in the PCOS-IR group and were correlated with insulin resistance, suggesting that the bacterium may play a key role in the pathogenesis of PCOS by regulating the glucagon-like peptidin-1 (GLP-1) level and other mechanisms. The altered intestinal flora may affect the intestinal environment of the host by enriching different metabolic functions and promoting the development of PCOS insulin resistance and disease in women. The present study was an experimental cross-sectional study involving participants from different regions, RACES, and eating habits, and this may have brought heterogeneity. Considering the variations in human intestinal microbiota, many clinical population and animal experiments may be needed to verify the findings of this study. Also, further studies are required to elucidate the precise function of various flora and the mechanism underlying their roles in the pathogenesis of multiple diseases. Our next study will involve performing a metabolomic analysis to explore further the relationship between insulin resistance and intestinal flora in PCOS patients. These findings will provide a theoretical basis for drug design and subsequent clinical treatment of PCOS.

Abbreviations

PCOS:polycystic ovary syndrome; IR:insulin resistance; BCAA:branched chain amino acid; SCFA:short chain fatty acid; HA:hyperandrogenism; P:progesterone; PRL:prolactin; T:testosterone; LH:luteinizing hormone; FSH:follicle-stimulating hormone; E2: oestradiol; SHBG:sex binding globulin; AMH:Anti-Mullerian hormone; TG:Triglyceride; TC:total cholesterol; LDL:low-density lipoprotein; HDL:high-density lipoprotein; CRP:C-reactive protein; IL-6:interleukin-6; TNF-α:tumor necrosis factor; OUT:Operational Taxonomic Units; LDA:Linear discriminant analysis; FFA: free fatty acid; GLP-1: glucagon-like peptide-1.

Declarations

Acknowledgements

We are grateful to the women who participated in the survey.

Author contributions

All authors contributed to the concepts of the manuscript; Fangfang He: Data collection, analysis, and management, manuscript writing. Yumei Li: Project development, manuscript writing and editing. All authors read and approved the final manuscript.

Funding

Not applicable

Availability of date and materials

The datasets are available from the corresponding author on reasonable request.

Ethics approval and consent to participate

This study was approved by the Ethics Committee of the Department of Assisted Reproduction(Xiangya Hospital,Central South University,)as well as the China Registered Clinical Trial Ethics Review Committee (Ethical Review No.: CHiECRT1900028223).

Consent for publication

This manuscript was an original research that has not been published previously, and not under consideration for publication elsewhere, in whole or in part.

Competing interests

The authors have no conflicts of interest to disclose.

References

  1. Sahmay S, Aydogan Mathyk B, Sofiyeva N, Atakul N, Azemi A, Erel T. Serum AMH levels and insulin resistance in women with PCOS. Eur J Obstet Gynecol Reprod Biol. 2018;224:159-64.
  2. Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. The Lancet. 2007;370(9588):685-97.
  3. Rosenfield RL, Ehrmann DA. The Pathogenesis of Polycystic Ovary Syndrome (PCOS): The Hypothesis of PCOS as Functional Ovarian Hyperandrogenism Revisited. Endocr Rev. 2016;37(5):467-520.
  4. Macut D, Bjekic-Macut J, Rahelic D, Doknic M. Insulin and the polycystic ovary syndrome. Diabetes Res Clin Pract. 2017;130:163-70.
  5. Diamanti-Kandarakis E, Dunaif A. Insulin resistance and the polycystic ovary syndrome revisited: an update on mechanisms and implications. Endocr Rev. 2012;33(6):981-1030.
  6. Legro RS, Arslanian SA, Ehrmann DA, Hoeger KM, Murad MH, Pasquali R, et al. Diagnosis and treatment of polycystic ovary syndrome: an Endocrine Society clinical practice guideline. The Journal of clinical endocrinology and metabolism. 2013;98(12):4565-92.
  7. Rotterdam EA-SPcwg. Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome (PCOS). Hum Reprod. 2004;19(1):41-7.
  8. Garg D, Tal R. Inositol Treatment and ART Outcomes in Women with PCOS. Int J Endocrinol. 2016;2016:1979654.
  9. Goodman NF, Cobin RH, Futterweit W, Glueck JS, Legro RS, Carmina E, et al. American Association of Clinical Endocrinologists, American College of Endocrinology, and Androgen Excess and Pcos Society Disease State Clinical Review: Guide to the Best Practices in the Evaluation and Treatment of Polycystic Ovary Syndrome - Part 2. Endocr Pract. 2015;21(12):1415-26.
  10. Coyte KZ, Schluter J, Foster KR. The ecology of the microbiome: Networks, competition, and stability. Science. 2015;350(6261):663-6.
  11. Hartstra AV, Bouter KE, Backhed F, Nieuwdorp M. Insights into the role of the microbiome in obesity and type 2 diabetes. Diabetes Care. 2015;38(1):159-65.
  12. Boulange CL, Neves AL, Chilloux J, Nicholson JK, Dumas ME. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 2016;8(1):42.
  13. Scheithauer TP, Dallinga-Thie GM, de Vos WM, Nieuwdorp M, van Raalte DH. Causality of small and large intestinal microbiota in weight regulation and insulin resistance. Mol Metab. 2016;5(9):759-70.
  14. Jiao N, Baker SS, Nugent CA, Tsompana M, Cai L, Wang Y, et al. Gut microbiome may contribute to insulin resistance and systemic inflammation in obese rodents: a meta-analysis. Physiol Genomics. 2018;50(4):244-54.
  15. Tremellen K, Pearce K. Dysbiosis of Gut Microbiota (DOGMA)--a novel theory for the development of Polycystic Ovarian Syndrome. Med Hypotheses. 2012;79(1):104-12.
  16. Thackray VG. Sex, Microbes, and Polycystic Ovary Syndrome. Trends Endocrinol Metab. 2019;30(1):54-65.
  17. Zhao X, Jiang Y, Xi H, Chen L, Feng X. Exploration of the Relationship Between Gut Microbiota and Polycystic Ovary Syndrome (PCOS): a Review. Geburtshilfe und Frauenheilkunde. 2020;80(2):161-71.
  18. Lindheim L, Bashir M, Münzker J, Trummer C, Zachhuber V, Leber B, et al. Alterations in Gut Microbiome Composition and Barrier Function Are Associated with Reproductive and Metabolic Defects in Women with Polycystic Ovary Syndrome (PCOS): A Pilot Study. PLoS One. 2017;12(1):e0168390.
  19. Liu R, Zhang C, Shi Y, Zhang F, Li L, Wang X, et al. Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary Syndrome. Front Microbiol. 2017;8:324.
  20. Torres PJ, Siakowska M, Banaszewska B, Pawelczyk L, Duleba AJ, Kelley ST, et al. Gut Microbial Diversity in Women With Polycystic Ovary Syndrome Correlates With Hyperandrogenism. J Clin Endocrinol Metab. 2018;103(4):1502-11.
  21. Insenser M, Murri M, Del Campo R, Martínez-García M, Fernández-Durán E, Escobar-Morreale HF. Gut Microbiota and the Polycystic Ovary Syndrome: Influence of Sex, Sex Hormones, and Obesity. J Clin Endocrinol Metab. 2018;103(7):2552-62.
  22. Zeng B, Lai Z, Sun L, Zhang Z, Yang J, Li Z, et al. Structural and functional profiles of the gut microbial community in polycystic ovary syndrome with insulin resistance (IR-PCOS): a pilot study. Res Microbiol. 2019;170(1):43-52.
  23. He W, Li Q, Yang M, Jiao J, Ma X, Zhou Y, et al. Lower BMI cutoffs to define overweight and obesity in China. Obesity (Silver Spring). 2015;23(3):684-91.
  24. Oakkar EE, Stevens J, Truesdale KP, Cai J. BMI and all-cause mortality among Chinese and Caucasians: the People's Republic of China and the Atherosclerosis Risk in Communities Studies. Asia Pac J Clin Nutr. 2015;24(3):472-9.
  25. We LXzCYjX. Analysis of insulin release pattern in patients with polycystic ovary syndrome and normal glucose tolerance. Chin J Endocrinol Meta. 2012;28(8).
  26. -Wei, SYYLPSY. Clinical research on insulin releasing curve rating method in evaluation of insulin resistance in patients with PCOS. China Academic Jounal Electronic Publishing. 2015;30(20).
  27. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7(5):335-6.
  28. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics. 2011;27(16):2194-200.
  29. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Applied and environmental microbiology. 2006;72(7):5069-72.
  30. Feng Q, Liang S, Jia H, Stadlmayr A, Tang L, Lan Z, et al. Gut microbiome development along the colorectal adenoma-carcinoma sequence. Nature communications. 2015;6:6528.
  31. Widecka J, Ozegowska K, Banaszewska B, Kazienko A, Safranow K, Branecka-Wozniak D, et al. Is copeptin a new potential biomarker of insulin resistance in polycystic ovary syndrome? Ginekol Pol. 2019;90(3):115-21.
  32. Barber TM. Divergences in insulin resistance between the different phenotypes of the polycystic ovary syndrome. Expert Rev Endocrinol. 2013;8(5):427-9.
  33. Sze MA, Schloss PD. Looking for a Signal in the Noise: Revisiting Obesity and the Microbiome. MBio. 2016;7(4).
  34. Walters WA, Xu Z, Knight R. Meta-analyses of human gut microbes associated with obesity and IBD. 2014;588(22):4223-33.
  35. Kelley ST, Skarra DV, Rivera AJ, Thackray VG. The Gut Microbiome Is Altered in a Letrozole-Induced Mouse Model of Polycystic Ovary Syndrome. PLoS One. 2016;11(1):e0146509.
  36. Thackray VG. Sex, Microbes, and Polycystic Ovary Syndrome. Trends Endocrinol Metab. 2018.
  37. Sherman SB, Sarsour N, Salehi M, Schroering A, Mell B, Joe B, et al. Prenatal androgen exposure causes hypertension and gut microbiota dysbiosis. Gut Microbes. 2018:1-22.
  38. Insenser M, Murri M, Del Campo R, Martinez-Garcia MA, Fernandez-Duran E, Escobar-Morreale HF. Gut Microbiota and the Polycystic Ovary Syndrome: Influence of Sex, Sex Hormones, and Obesity. J Clin Endocrinol Metab. 2018;103(7):2552-62.
  39. Zeng B, Lai Z, Sun L, Zhang Z, Yang J, Li Z, et al. Structural and functional profiles of the gut microbial community in polycystic ovary syndrome with insulin resistance (IR-PCOS): a pilot study. Res Microbiol. 2018.
  40. Kelly BJ, Gross R, Bittinger K, Sherrill-Mix S, Lewis JD, Collman RG, et al. Power and sample-size estimation for microbiome studies using pairwise distances and PERMANOVA. Bioinformatics. 2015;31(15):2461-8.
  41. Byappanahalli MN, Nevers MB, Korajkic A, Staley ZR, Harwood VJ. Enterococci in the Environment. Microbiology and Molecular Biology Reviews. 2012;76(4):685-706.
  42. Ruzickova M, Vitezova M, Kushkevych I. The Characterization of Enterococcus Genus: Resistance Mechanisms and Inflammatory Bowel Disease. Open Med (Wars). 2020;15:211-24.
  43. Hou YP, He QQ, Ouyang HM, Peng HS, Wang Q, Li J, et al. Human Gut Microbiota Associated with Obesity in Chinese Children and Adolescents. Biomed Res Int. 2017;2017:7585989.
  44. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 2009;1(6):6ra14.
  45. LeValley SL, Tomaro-Duchesneau C, Britton RA. Degradation of the Incretin Hormone Glucagon-Like Peptide-1 (GLP-1) by Enterococcus faecalis Metalloprotease GelE. mSphere. 2020;5(1).
  46. Vrbikova J, Hill M, Bendlova B, Grimmichova T, Dvorakova K, Vondra K, et al. Incretin levels in polycystic ovary syndrome. European journal of endocrinology. 2008;159(2):121-7.
  47. Lamos EM, Malek R, Davis SN. GLP-1 receptor agonists in the treatment of polycystic ovary syndrome. Expert Rev Clin Pharmacol. 2017;10(4):401-8.
  48. Niafar M, Pourafkari L, Porhomayon J, Nader N. A systematic review of GLP-1 agonists on the metabolic syndrome in women with polycystic ovaries. Arch Gynecol Obstet. 2016;293(3):509-15.
  49. Grasset E, Puel A, Charpentier J, Collet X, Christensen JE, Terce F, et al. A Specific Gut Microbiota Dysbiosis of Type 2 Diabetic Mice Induces GLP-1 Resistance through an Enteric NO-Dependent and Gut-Brain Axis Mechanism. Cell Metab. 2017;25(5):1075-90 e5.
  50. Kaakoush NO, Lecomte V, Maloney CA, Morris MJ. Cross-talk among metabolic parameters, esophageal microbiota, and host gene expression following chronic exposure to an obesogenic diet. Sci Rep. 2017;7:45753.
  51. de Andrade PAM, Giovani PA, Araujo DS, de Souza AJ, Pedroni-Pereira A, Kantovitz KR, et al. Shifts in the bacterial community of saliva give insights on the relationship between obesity and oral microbiota in adolescents. Arch Microbiol. 2020.
  52. Crusell MKW, Hansen TH, Nielsen T, Allin KH, Rühlemann MC, Damm P, et al. Gestational diabetes is associated with change in the gut microbiota composition in third trimester of pregnancy and postpartum. Microbiome. 2018;6(1).
  53. Vink JM, Sadrzadeh S, Lambalk CB, Boomsma DI. Heritability of polycystic ovary syndrome in a Dutch twin-family study. J Clin Endocrinol Metab. 2006;91(6):2100-4.
  54. Domecq JP, Prutsky G, Mullan RJ, Hazem A, Sundaresh V, Elamin MB, et al. Lifestyle Modification Programs in Polycystic Ovary Syndrome: Systematic Review and Meta-Analysis. The Journal of Clinical Endocrinology & Metabolism. 2013;98(12):4655-63.
  55. Knight R, Vrbanac A, Taylor BC, Aksenov A, Callewaert C, Debelius J, et al. Best practices for analysing microbiomes. Nat Rev Microbiol. 2018;16(7):410-22.

Tables

Table I. Clinical, biochemical and hormonal features of participants

 

 

PCOS

Parametes

HC(n=12)

NIR(n=10)

IR(n=14)

Age(years)

28.25±1.22

26.4±3.41

26.71±2.43

Menstrual cycle(days)

29.67±2.02AB

82.7±26.61B

75.43±27.91A

BMI(Kg/m2)

21.28±1.34

21.32±1.22

21.91±1.4

Waist

67.42±4.4AB

71.8±3.29BC

77.79±5.73AC

Hip

88±5.31

87.8±3.33

91.54±4.43

Waist to hip ratio

0.77±0.03AB

0.82±0.03BC

0.85±0.04AC

FPG (mmol/L)

5±0.27A

5.09±0.47

5.42±0.59A

FINs(uU/mL)

6.91±1.82A

6.95±2.52C

14.81±3.88AC

HOMA-IR

1.54±0.45A

1.61±0.64C

3.57±1.07AC

HbA1-C

5.04±0AB

5.37±0.21B

5.26±0.28A

FSH (IU/L)

6.8±2.18

5.92±2.16

6.43±1.42

LH(IU/L)

6.25±2.88AB

15.55±8.03B

15.37±7.31A

LH/FSH

0.96±0.4AB

2.53±0.89B

2.42±1.2A

T (nmol/L)

0.75±0.21AB

1.93±0.57B

1.81±0.82A

P(nmol/l)

0.54±0.16B

0.93±0.52B

0.74±0.43

E2(pmol/L)

183.51±100.9B

281.47±164.67B

194.25±55.9

AMH

4.99±3.74A

7.99±2.79

9.28±5.12A

TBA (umol/l)

4.7±2.22

5.51±4.77

5.26±2.4

ALT (U/L)

13.18±2.7

21.02±15.13

20.81±11.77

AST(U/L)

18.22±5.91

22.03±6.72

21.64±6.4

Lithic (umol/L)

243.43±37.04B

320.21±56.66B

283.65±105.62

TG( mmol/L)

0.88±0.25A

1.14±0.84

1.29±0.41A

TC( mmol/L)

4.29±1.11

4.39±0.68

4.77±0.88

HDL(mmol/L)

1.47±0.31

1.36±0.26

1.33±0.23

LDL(mmol/L)

2.67±0.59

2.6±0.53

2.9±0.6

APO-a

93.13±71.68

120.14±126.97

194.49±220.43

ApoA/ApoB

2.47±0.48

2.31±0.56

2.22±0.62

FFA(mmol/L)

0.47±0.11A

0.48±0.31

0.66±0.24A

hsCRP(mg/L)

0.69±0.56A

0.8±0.75C

1.63±1.27AC

IL-6(pg/ml)

1.13±0.46AB

2.12±0.26B

2.56±1.28A

TNF

3.76±1.43AB

4.86±1.07B

4.54±0.51A

A: P < 0.05 for HC vs. IR-PCOS group.

B: P < 0.05 for HC vs. NIR-PCOS group.

C: P < 0.05 for IR vs. NIR-PCOS group.

Abbreviation: PCOS, polycystic ovary syndrome; PCOS-IR, PCOS with insulin resistance; PCOS-NIR, PCOS without insulin resistance; BMI, body mass index; WHR: the ratio of waist to hip; FSH, follicular stimulating hormone; LH, luteinizing hormone; TNF-a: tumor necrosis factor-a; FPG, fasting plasma glucose; FINS, fasting plasma insulin; HOMAIR, homeostasis model assessment of insulin resistance; PPG, 3h postprandial plasma glucose; TC, total cholesterol; HDL-C, high density lipoprotein-cholesterol; LDL-C, low density lipoprotein-cholesterol; ALT: alanine aminotransferase; AST: aspartate aminotransferase.