Abstract
The etiology of polycystic ovary syndrome (PCOS) remains unclear, although studies indicate that both genetic and environmental factors contribute to the syndrome. In 2012, Tremellen and Pearce proposed the idea that dysbiosis of the intestinal (gut) microbiome is a causative factor of metabolic and reproductive manifestations of PCOS. In the past 5 years, studies in both humans and rodent models have demonstrated that changes in the taxonomic composition of gut bacteria are associated with PCOS. Studies have also clearly shown that these changes in gut microbiota are associated with PCOS as opposed to obesity, since these changes are observed in women with PCOS that are both of a normal weight or obese, as well as in adolescent girls with PCOS and obesity compared with body mass index- and age-matched females without the disorder. Additionally, studies in both women with PCOS and rodent models of PCOS demonstrated that hyperandrogenism is associated with gut microbial dysbiosis, indicating that androgens may modulate the gut microbial community in females. One study reported that the fecal microbiome transplantation of stool from women with PCOS or exposure to certain bacteria resulted in a PCOS-like phenotype in mice, while other studies showed that exposure to a healthy gut microbiome, pre/probiotics, or specific gut metabolites resulted in protection from developing PCOS-like traits in mice. Altogether, these results suggest that dysbiosis of the gut microbiome may be sufficient to develop PCOS-like symptoms and that modulation of the gut microbiome may be a potential therapeutic target for PCOS.
Keywords: polycystic ovary syndrome, hyperandrogenism, insulin resistance, gut microbiome, bile acids
Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders, affecting 5% to 10% of women of reproductive age worldwide [1, 2]. Diagnosis of PCOS includes 2/3 clinical presentations: (1) clinical or biochemical hyperandrogenism (HA), (2) oligomenorrhea or anovulation, and (3) polycystic ovaries [3]. Women with PCOS are at a higher risk for infertility and pregnancy complications [4–7]. In PCOS, metabolic dysregulation is correlated with HA, occurs independently of body mass index (BMI), and includes obesity, insulin resistance (IR), and dyslipidemia (Fig. 1) [8–10], which leads to an increased risk of developing type 2 diabetes (T2D), hypertension, and nonalcoholic fatty liver disease [6, 11–14]. Due to its prevalence and association with multiple chronic diseases, studying the mechanisms driving the etiology and pathogenesis of PCOS is of utmost importance in facilitating the development of novel therapies for PCOS.
Figure 1.
Correlations between hyperandrogenism (HA), dysbiosis of the gut microbiome, and metabolic dysfunction. A: Link between HA and dysbiosis of the gut microbiome. Recent studies in women and rodent models demonstrated that HA is correlated with dysbiosis of the gut microbiome, including changes in the overall biodiversity of gut bacteria as well as the relative abundance of certain bacteria. In addition, 1 study reported that introducing stool from women with PCOS or a bacterial species (B. vulgatus) in antibiotic-treated mice resulted in HA, suggesting that gut microbial dysbiosis or the overabundance of specific bacteria may be sufficient to induce PCOS-like symptoms. B: Link between HA and metabolic dysfunction. Metabolic dysfunction, including weight gain, insulin resistance (IR), and dyslipidemia, occurs predominantly in women with PCOS diagnosed with HA and ovulatory dysfunction, independent of body mass index. C: Link between dysbiosis of the gut microbiome and metabolic dysfunction. Gut dysbiosis has been associated with obesity, IR, and impaired lipid metabolism in metabolic diseases, including metabolic syndrome, type 2 diabetes, nonalcoholic fatty liver disease, and PCOS. Despite the tripartite set of correlations between HA, gut microbial dysbiosis, and metabolic dysfunction, the mechanisms of how each player affects the other 2 are still largely unknown. Future studies will be required to decipher how the gut microbiome communicates with the host and vice versa in order to alter or respond to varying levels of steroid and metabolic hormones.
The intestinal (gut) microbiome is comprised of a community of diverse bacteria, archaea, fungi, protozoa, and viruses, along with their metabolites. This community plays an important role in host physiology, including immunity, the health of the gut epithelial barrier, production of vitamin B12, and production of short-chain fatty acids (SCFAs) via fermentation of fiber, metabolism, and neurological functions [15]. Alterations in gut microbiota have been associated with metabolic diseases (Fig. 1), autoimmune diseases, neurological disorders, and cardiovascular disease [16]. In 2012, Tremellen and Pearce proposed that a connection might exist between dysbiosis of the gut microbiome and the metabolic and reproductive manifestations of PCOS [17]. Two studies in 2016 and 2017 first reported evidence that changes in the gut microbiome were associated with PCOS in a mouse model of PCOS and in women with the disorder [18, 19]. Since then, multiple subsequent studies in humans [20–28] and rodents [29–32] provided further evidence that dysbiosis of the gut microbiome is associated with PCOS.
In this review, we highlight recent findings on the association between the gut microbiome and PCOS, the relationship between HA and the gut microbiome in PCOS, the relationship between substrates and metabolites of the gut microbiota and PCOS, and potential gut microbiota-altering treatments as therapies of PCOS. To do so, we used the following search terms in NCBI PubMed: “microbiome,” “microbiota,” and “polycystic ovary syndrome.” We confined our search criteria to primary research articles of human and rodent studies on PCOS and the gut microbiome between 2016 and 2020.
Dysbiosis of the Gut Microbiome is Associated with polycystic ovary syndrome (PCOS)
Alpha diversity of the gut microbiota in humans
The overall composition of gut microbiota can be represented by metrics of alpha diversity, which estimate the species richness and/or evenness of a community, sometimes taking phylogenetic relationships into account. Recent studies have shown that alpha diversity of gut microbiota is altered in women with PCOS compared with healthy women. By sampling the fecal microbial content and sequencing 16S ribosomal RNA (rRNA) genes amplified with universal bacterial primers, multiple studies demonstrated that alpha diversity of gut bacteria decreased in premenopausal women with PCOS as compared with age-matched, healthy women [19, 22, 26–28, 33]. In contrast, 3 studies did not observe significant changes in alpha diversity between women with PCOS and healthy women, potentially due to small sample sizes [21, 24, 34]. Interestingly, studies that used shotgun metagenomic sequencing of the gut microbiome also did not report changes in alpha diversity [20, 23, 25]. All of the aforementioned studies included women diagnosed with PCOS using the Rotterdam criteria [35, 36] from limited geographical locations in Asia and Europe, including China, Turkey, Austria, Poland, and Spain. A recent study on gut microbial changes (using 16S rRNA sequencing) in adolescent girls (14–16 years old) with PCOS and obesity and weight- and age-matched healthy controls from an ethnically diverse population in the United States reported that PCOS was also associated with a decrease in alpha diversity [37]. This study indicates that decreased biodiversity of gut microbes, like other features of PCOS [38], manifests by adolescence. High alpha diversity was proposed as an indication of productivity and stability in an ecosystem, implying overall health of the community [39]. Decreases in alpha diversity have been observed in immune diseases such as Chron’s disease, ulcerative colitis, type 1 diabetes mellitus, celiac disease, and allergies [40–42]; in cardiometabolic diseases such as obesity, T2D, and vascular stiffness [42–44]; colorectal cancer [42]; and autism [42]. Thus, loss of microbial diversity in the gut may serve as a biomarker of disease or as an indication of a functional problem, especially when correlated with changes in metabolites of the gut microbiome, as in T2D [44].
Beta diversity of the gut microbiota in humans
In addition to alpha diversity, beta diversity (how similar or different the composition of 1 gut microbial community is compared to another community) can also be estimated using distance metrics that take or do not take phylogenetic relationships or abundance into account. Using 16S rRNA gene sequencing, multiple studies reported that beta diversity was altered in fecal samples obtained from women with PCOS compared with healthy women [19, 22, 28]. However, in other studies, no significant difference in beta diversity was detected in women with PCOS compared with healthy women [21, 26, 33, 34], potentially due to small sample sizes. Comparing adolescent girls with PCOS and obesity to BMI-matched controls, changes in beta diversity were also observed between the 2 groups [37]. Unlike alpha diversity, changes in beta diversity were observed in 2 studies where shotgun metagenomics were used to sequence gut microbiota in women with PCOS [23, 25]. In contrast, 1 study using metagenomic sequencing did not observe differences in gut microbial beta diversity between women with and without PCOS [20]. Overall, these results indicate that differences in beta diversity also appear to be associated with PCOS.
Relative abundance of bacterial taxa in humans
In addition to looking at gut microbial diversity at the community level, various studies assessed differences in the relative abundance (RA) of specific bacterial taxa. In the healthy gut, the phyla Firmicutes, Bacteroidetes, Actinobacteria, and Verrucomicrobia are the most dominant, while Proteobacteria and Tenericutes exist at low abundance [45]. Table 1 summarizes cohort characteristics of women included in studies of PCOS and the gut microbiome, while Table 2 summarizes the different taxa that were significantly altered in women with PCOS from the studies reviewed herein and is organized by bacterial phyla. Of the genera within phylum Bacteroidetes, Bacteroides were positively associated with PCOS in 5/7 studies and Parabacteroides were positively associated with PCOS in 2 studies [20, 23–25, 28, 33], while the family S24-7 was negatively associated with PCOS in 2 studies [19, 33]. Of the genera within phylum Firmicutes, family Clostridiaceae was positively associated with PCOS in 2 studies [22, 25] and family Veillonellaceae in 2 other studies [27, 33]. Of the genera within phylum Proteobacteria, Escherichia, and Shigella were positively associated with PCOS in 2 studies [20, 22].
Table 1.
Cohort characteristics of human studies on PCOS and the gut microbiome
Country | Cohort Groups | N | Diagnosis | Age (years) | T (nmol/L) | BMI (kg/m2) | HOMA-IR | Method | Ref |
---|---|---|---|---|---|---|---|---|---|
Austria | Controls | 19 | Rotterdam Criteria |
32.0 | 1.1 | 22.3 | 0.8 | 16S rRNA | [19] |
PCOS | 24 | 27.0 | 1.3 | 24.9 | 1.7 | (V1–V2) | |||
China | Nonoverweight controls | 7 | Rotterdam Criteria |
30.3 | 1.1 | 20.6 | n/a | Metagenomics | [20] |
Overweight controls | 7 | 28.6 | 0.7 | 27.1 | n/a | ||||
Nonoverweight PCOS | 7 | 27.1 | 1.8 | 21.0 | n/a | ||||
Overweight PCOS | 7 | 29.1 | 1.4 | 27.9 | n/a | ||||
Spain | Nonobese controls | 8 | Rotterdam Criteria |
27.3 | 1.6 | 23.4 | 1.6 | 16S rRNA (V4) | [21] |
Obese controls | 8 | 27.3 | 2.0 | 35.9 | 3.3 | ||||
Nonobese PCOS | 7 | 23.0 | 2.5 | 24.4 | 1.5 | ||||
Obese PCOS | 8 | 29.9 | 2.4 | 37.0 | 2.6 | ||||
China | Nonobese controls | 12 | Rotterdam Criteria |
32.2 | 0.8 | 21.9 | 1.7 | 16S rRNA (V3–V4) | [22] |
Obese controls | 6 | 33.0 | 1.0 | 27.5 | 3.5 | ||||
Nonobese PCOS | 12 | 25.5 | 4.5 | 21.6 | 1.1 | ||||
Obese PCOS | 21 | 29.3 | 5.4 | 30.0 | 3.3 | ||||
China | Controls | 43 | Rotterdam Criteria |
29.6 | 1.56a | 23.7 | 1.6 | Metagenomics | [23] |
PCOS | 50 | 29.9 | 2.11a | 24.7 | 3.1 | ||||
China | Controls | 8 | Rotterdam Criteria |
26.4 | 0.7 | 20.8 | 1.4 | 16S rRNA (V3–V4) | [24] |
NIR-PCOS | 8 | 26.1 | 1.9 | 22.6 | 1.9 | ||||
IR-PCOS | 9 | 25.1 | 2.1 | 22.6 | 4.1 | ||||
China | Controls | 26 | B-ultrasound | 26.7 | 0.8 | n/a | n/a | 16S rRNA (V3–V4) | [25] |
PCOS | 38 | Oligomenorrhea Hormones |
27.6 | 6.0 | n/a | n/a | |||
Metagenomics | |||||||||
China | Nonobese controls | 30 | Rotterdam Criteria |
22.1 | 1.66a | n/a | n/a | 16S rRNA (V3–V4) | [26] |
Obese controls | 11 | 25.3 | 1.8a | n/a | n/a | ||||
Nonobese PCOS | 30 | 25.1 | 2.67a | n/a | 2.4 | ||||
Obese PCOS | 30 | 26.9 | 2.63a | n/a | 6.4 | ||||
China | Controls | 9 | Rotterdam Criteria |
27.9 | 1.3 | 20.9 | 1.7 | 16S rRNA (V3–V4) | [27] |
Nonobese PCOS | 10 | 25.7 | 1.9 | 20.7 | 1.4 | ||||
Obese PCOS | 8 | 27.1 | 2.2 | 29.5 | 3.8 | ||||
Poland | Controls | 48 | Rotterdam Criteria |
29.4 | 1.04a | 23.7 | 1.8 | 16S rRNA (V4) | [28] |
PCOM | 42 | 29.8 | 1.04a | 22.6 | 1.7 | ||||
PCOS | 73 | 27.4 | 0.56a | 25.6 | 2.3 | ||||
Austria | Controls | 20 | Rotterdam Criteria |
32.0 | 1.1 | 22.3 | 0.8 | 16S rRNA (V1–V2) | [33] |
PCOS | 24 | 27.0 | 1.3 | 24.9 | 1.7 | ||||
Turkey | Controls | 15 | Rotterdam Criteria |
22.0 | 0.97a | 31.5 | 2.1 | 16S rRNA (V3–V4) | [34] |
PCOS | 17 | 20.0 | 2.22a | 19.6 | 2.0 | ||||
USA | Obese controls | 21 | NIH Criteria |
14.5 | 0.69a | 35.0 | 4.1 | 16S rRNA (V3–V4) | [37] |
Obese PCOS | 37 | 16.1 | 1.49a | 36.0 | 4.5 |
V1, V2, V3, and V4 are variable regions of the bacterial 16S ribosomal ribonucleic acid (rRNA) gene. Primers are designed to target these regions for 16S rRNA gene sequencing to identify specific bacterial genera.
Abbreviations: BMI, body mass index; B-ultrasound, brightness-mode ultrasound; HOMA-IR, homeostatic model assessment for insulin resistance; IR, insulin-resistant; n/a, not applicable; NIH, National Institutes of Health; NIR, non-insulin-resistant; PCOM, polycystic ovarian morphology; PCOS, polycystic ovary syndrome; Ref, reference number; rRNA, ribosomal ribonucleic acid.
a Converted from ng/mL, ug/L, or ng/dL.
Table 2.
Changes in bacterial taxa associated with PCOS in women
Phylum | Family | Genus | Change | Ref |
---|---|---|---|---|
Actinobacteria | Bifidobacteriaceae | Bifidobacterium | ↓ | [25] |
Actinobacteria | Coriobacteriaceae | Collinsella | ↑ | [25] |
Actinobacteria | ↑ | [37] | ||
Bacteroidetes | Bacteroidaceae | Bacteroides | ↑ | [20] |
Bacteroidetes | Bacteroidaceae | Bacteroides | ↑ | [23] |
Bacteroidetes | Bacteroidaceae | Bacteroides | ↑ | [24] |
Bacteroidetes | Bacteroidaceae | Bacteroides | ↑ | [28] |
Bacteroidetes | Bacteroidaceae | Bacteroides | ↑ | [33] |
Bacteroidetes | Bacteroidaceae | Bacteroides | ↓ | [22] |
Bacteroidetes | Bacteroidaceae | Bacteroides | ↓ | [37] |
Bacteroidetes | Porphyromonadaceae | Odoribacter | ↓ | [28] |
Bacteroidetes | Porphyromonadaceae | Parabacteroides | ↑ | [20] |
Bacteroidetes | Porphyromonadaceae | Parabacteroides | ↑ | [25] |
Bacteroidetes | Porphyromonadaceae | Porphyromonas | ↑ | [28] |
Bacteroidetes | Porphyromonadaceae | ↓ | [37] | |
Bacteroidetes | Prevotellaceae | Alloprevotella | ↓ | [26] |
Bacteroidetes | Prevotellaceae | Prevotella | ↑ | [25] |
Bacteroidetes | Prevotellaceae | Prevotella | ↑ | [37] |
Bacteroidetes | Prevotellaceae | Prevotella | ↓ | [24] |
Bacteroidetes | Prevotellaceae | Prevotella | ↓ | [33] |
Bacteroidetes | S24-7 | ↓ | [19] | |
Bacteroidetes | S24-7 | ↓ | [33] | |
Bacteroidetes | ↑ | [27] | ||
Firmicutes | Clostridiaceae | Clostridium | ↑ | [25] |
Firmicutes | Clostridiaceae | ClostridiumIV | ↑ | [22] |
Firmicutes | Erysipelotrichidae | Catenibacterium | ↑ | [21] |
Firmicutes | Erysipelotrichidae | Kandleria | ↑ | [21] |
Firmicutes | Lachnospiraceae | Blautia | ↑ | [28] |
Firmicutes | Lachnospiraceae | Blautia | ↓ | [20] |
Firmicutes | Lachnospiraceae | Blautia | ↓ | [25] |
Firmicutes | Lachnospiraceae | Coprococcus | ↑ | [26] |
Firmicutes | Lachnospiraceae | Oribacterium | ↑ | [21] |
Firmicutes | Lachnospiraceae | Roseburia | ↓ | [28] |
Firmicutes | Lachnospiraceae | ↓ | [27] | |
Firmicutes | Lactobacillaceae | Lactobacillus | ↓ | [22] |
Firmicutes | Oscillospiraceae | Oscillibacter | ↓ | [22] |
Firmicutes | Ruminococcaceae | Faecalibacterium | ↑ | [28] |
Firmicutes | Ruminococcaceae | Faecalibacterium | ↓ | [20] |
Firmicutes | Ruminococcaceae | Faecalibacterium | ↓ | [25] |
Firmicutes | Ruminococcaceae | Ruminococcus | ↓ | [28] |
Firmicutes | Ruminococcaceae | Subdoligranulum | ↑ | [27] |
Firmicutes | Ruminococcaceae | ↑ | [34] | |
Firmicutes | Ruminococcaceae | ↓ | [22] | |
Firmicutes | Streptococcaceae | Lactococcus | ↓ | [26] |
Firmicutes | Streptococcaceae | Streptococcus | ↓ | [22] |
Firmicutes | Streptococcaceae | ↑ | [37] | |
Firmicutes | Veillonellaceae | Megamonas | ↑ | [27] |
Firmicutes | Veillonellaceae | Megasphaera | ↑ | [33] |
Firmicutes | Anaerococcus | ↓ | [28] | |
Proteobacteria | Comamonadaceae | Comamonas | ↑ | [20] |
Proteobacteria | Enterobacteriaceae | Escherichia | ↑ | [20] |
Proteobacteria | Enterobacteriaceae | Escherichia | ↑ | [22] |
Proteobacteria | Enterobacteriaceae | Shigella | ↑ | [20] |
Proteobacteria | Enterobacteriaceae | Shigella | ↑ | [22] |
Synergistetes | ↓ | [26] | ||
Tenericutes | ↓ | [19] | ||
Tenericutes | ↓ | [26] | ||
Verrucomicrobia | Akkermansiaceae | Akkermansia | ↓ | [22] |
Abbreviations: PCOS, polycystic ovary syndrome; Ref, reference number.
Alpha and beta diversity in rodent models of PCOS
In addition to human studies, changes in overall gut microbial diversity were also observed in PCOS-like rodent models compared with placebo controls using 16S rRNA gene sequencing. As recently reviewed [46], hyperandrogenic rodent models of PCOS have been created using treatment with dihydrotestosterone (DHT) or the nonsteroidal aromatase inhibitor, letrozole. In a letrozole-induced pubertal PCOS mouse model, alpha diversity of the gut microbiome was lower in letrozole-treated mice than placebo controls, while beta diversity was also changed between the 2 groups [18]. In contrast, letrozole-induced PCOS in adult mice and rats showed no change in alpha diversity [47, 48]. Although adult mice treated with letrozole showed a shift in beta diversity, changes in specific bacterial taxa were distinct between the pubertal and the adult PCOS mouse models [47], and there were no changes in beta diversity observed in adult rats treated with letrozole [48]. In a cohort of 6-week old rats treated with letrozole, changes in beta diversity were observed when compared with placebo-treated rats, while alpha diversity was not different between the 2 groups [49]. These results suggest that the age at which PCOS is induced in rodent models may be critical in order to recapitulate the metabolic dysregulation and gut microbial changes that resemble those found in women with PCOS.
RA of bacterial taxa in rodents
In addition to looking at changes in the overall gut microbial community in rodents, various studies assessed differences in the RA of specific bacterial taxa in PCOS rat and mouse models. Table 3 summarizes cohort characteristics of rodent models of PCOS where gut microbiota was assessed. Table 4 summarizes the different taxa that were altered in the PCOS models and is organized by bacterial phyla. Of the genera within phylum Actinobacteria, Bifidobacterium was negatively associated with PCOS in 2/3 studies [18, 49]. Of the genera within phylum Bacteroidetes, Bacteroides were positively associated with PCOS in 3/4 studies, while Prevotella were positively associated with PCOS in 2 studies [30, 32, 49, 50]. Of the genera within the phylum Firmicutes, Blautia and Roseburia were positively associated with PCOS in 2 studies [18, 29], while Lactobacillus was negatively associated with PCOS in 2/3 studies [29, 30].
Table 3.
Cohort characteristics of rodent studies on PCOS and the gut microbiome
Animal | Treatment | Treatment Format | Start (week) | Duration (week) | N | Method | Ref |
---|---|---|---|---|---|---|---|
C57BL/6N mice | Letrozole | Pellet (50 ug/day) | 4 | 5 | 10/group | 16S rRNA (V4) | [18] |
C57BL/6N mice | Letrozole | Pellet (50 ug/day) | 4 | 5 | 10 placebo; 12 LET | 16S rRNA (V4) | [29] |
Sprague-Dawley rats | Letrozole | Oral gavage (1 mg/kg/day) | 6 | 3 | 8/group | 16S rRNA (V3) | [30] |
C57BL/6N mice | Letrozole | Pellet (50 ug/day) | 4 | 5 | 8/group | 16S rRNA (V4) | [31] |
Sprague-Dawley rats | DHT | Injection (83 ug/day) | 3 | 6 | 5/group | 16S rRNA (V3–V4) | [32] |
Sprague-Dawley rats | Letrozole | Oral gavage (1 mg/kg/day) | 6 | 11 | 8/group | 16S rRNA (V3–V4) | [49] |
Wistar rats | DHT | 15 mg silicone tube | 3 | 10 | 6/group | 16S rRNA (V3–V4) | [50] |
V1, V2, V3, and V4 are variable regions of the bacterial 16S ribosomal ribonucleic acid (rRNA) gene. Primers are designed to target these regions for 16S rRNA gene sequencing to identify specific bacterial genera.
Abbreviations: DHT, dihydrotestosterone; PCOS, polycystic ovary syndrome; Ref, reference number; rRNA, ribosomal ribonucleic acid.
Table 4.
Changes in bacterial taxa associated with rodent models of PCOS
Phylum | Family | Genus | Change | Ref |
---|---|---|---|---|
Actinobacteria | Bifidobacteriaceae | Bifidobacterium | ↑ | [29] |
Actinobacteria | Bifidobacteriaceae | Bifidobacterium | ↓ | [18] |
Actinobacteria | Bifidobacteriaceae | Bifidobacterium | ↓ | [49] |
Actinobacteria | Coriobacteriaceae | Adlercreutzia | ↑ | [31] |
Actinobacteria | ↑ | [32] | ||
Actinobacteria | ↑ | [49] | ||
Bacteroidetes | Bacteroidaceae | Bacteroides | ↑ | [32] |
Bacteroidetes | Bacteroidaceae | Bacteroides | ↑ | [49] |
Bacteroidetes | Bacteroidaceae | Bacteroides | ↑ | [50] |
Bacteroidetes | Bacteroidaceae | Bacteroides | ↓ | [18] |
Bacteroidetes | Odoribacteraceae | Odoribacter | ↓ | [29] |
Bacteroidetes | Porphyromonadaceae | Parabacteroides | ↑ | [29] |
Bacteroidetes | Porphyromonadaceae | Parabacteroides | ↓ | [18] |
Bacteroidetes | Prevotellaceae | Ga6A1 | ↓ | [50] |
Bacteroidetes | Prevotellaceae | Prevotella | ↑ | [30] |
Bacteroidetes | Prevotellaceae | Prevotella_9 | ↑ | [50] |
Bacteroidetes | Prevotellaceae | ↓ | [50] | |
Bacteroidetes | Rikenellaceae | Alistipes | ↑ | [32] |
Bacteroidetes | Rikenellaceae | Alistipes | ↓ | [18] |
Bacteroidetes | S24-7 | ↓ | [18] | |
Bacteroidetes | ↓ | [32] | ||
Cyanobacteria | ↑ | [32] | ||
Firmicutes | Christensenellaceae | Christensenella | ↓ | [31] |
Firmicutes | Clostridiaceae | Anaerotruncus | ↑ | [32] |
Firmicutes | Clostridiaceae | Candidatus Arthromitus | ↓ | [31] |
Firmicutes | Clostridiaceae | Clostridium | ↑ | [32] |
Firmicutes | Clostridiaceae | Clostridium | ↓ | [30] |
Firmicutes | Dehalobacteriaceae | Dehalobacterium | ↓ | [18] |
Firmicutes | Erysipelotrichaceae | Allobaculum | ↑ | [18] |
Firmicutes | Erysipelotrichaceae | Coprobacillus | ↓ | [31] |
Firmicutes | Erysipelotrichaceae | Turicibacter | ↑ | [31] |
Firmicutes | Eubacteriaceae | Eubacterium | ↑ | [32] |
Firmicutes | Hungateiclostridiaceae | Ruminiclostridium | ↓ | [49] |
Firmicutes | Lachnospiraceae | Blautia | ↑ | [18] |
Firmicutes | Lachnospiraceae | Blautia | ↑ | [29] |
Firmicutes | Lachnospiraceae | Butyrivibrio | ↓ | [49] |
Firmicutes | Lachnospiraceae | Coprococcus | ↑ | [18] |
Firmicutes | Lachnospiraceae | Dorea | ↓ | [29] |
Firmicutes | Lachnospiraceae | Dorea | ↓ | [31] |
Firmicutes | Lachnospiraceae | Roseburia | ↑ | [18] |
Firmicutes | Lachnospiraceae | Roseburia | ↑ | [29] |
Firmicutes | Lachnospiraceae | Roseburia | ↓ | [31] |
Firmicutes | Lachnospiraceae | Tyzzerella | ↑ | [32] |
Firmicutes | Lachnospiraceae | ↓ | [29] | |
Firmicutes | Lactobacillaceae | Lactobacillus | ↑ | [31] |
Firmicutes | Lactobacillaceae | Lactobacillus | ↓ | [29] |
Firmicutes | Lactobacillaceae | Lactobacillus | ↓ | [30] |
Firmicutes | Ruminococcaceae | Ruminococcus | ↑ | [18] |
Firmicutes | Ruminococcaceae | Ruminococcus | ↑ | [29] |
Firmicutes | Ruminococcaceae | Ruminococcus | ↓ | [29] |
Firmicutes | Ruminococcaceae | Ruminococcus | ↓ | [30] |
Firmicutes | Ruminococcaceae | Ruminococcus_1 | ↑ | [32] |
Firmicutes | Ruminococcaceae | ↓ | [18] | |
Firmicutes | ↑ | [32] | ||
Proteobacteria | Desulfovibrionaceae | Desulfovibrio | ↓ | [49] |
Proteobacteria | Sutterellaceae | Sutterella | ↑ | [29] |
Saccharibacteria | ↑ | [32] | ||
Spirochaetes | Spirochaetaceae | ↓ | [50] | |
Verrucomicrobia | Akkermansiaceae | Akkermansia | ↑ | [31] |
Abbreviations: PCOS, polycystic ovary syndrome; Ref, reference number.
Caveats and future directions
The diversity in the clinical presentation of PCOS presents a challenge for studies attempting to decipher consistent patterns in the dysbiosis of the gut microbiome in women with PCOS since not all women diagnosed with PCOS have the same pathological phenotypes. Other challenges of studying dysbiosis of the gut microbiome, in general, are geographical and diet-based differences between study populations, which, in turn, can affect the composition of the gut microbiota. Moreover, the use of different methods for the collection and storage of fecal samples, sequencing, and data analysis probably contribute to inconsistent findings in human gut microbiome studies [51]. For instance, 2 factors that may influence the results obtained from the studies outlined herein is the use of different primers for the sequencing of the hypervariable regions (V1-4) of 16S rRNA gene and the use of different bioinformatics programs for data analysis [51] (Tables 1 and 3). In addition, we note that there is considerable variability in the characteristics of the human cohorts with regards to HA, BMI, and IR, which may also explain some of the inconsistencies with regards to alpha diversity, beta diversity, and the RA of specific bacterial taxa.
Moving forward, consensus methods for studying the gut microbiome in women with PCOS may be required to clearly differentiate differences in the gut microbiome that are due to factors such as geography and diet with those that are related to PCOS. Additionally, since most of the previous studies have relied on 16S rRNA gene sequencing, metagenomic approaches in future studies will be beneficial to investigate whether other gut microbes such as archaea, fungi, and viruses are altered in PCOS and dissect gut microbial gene functions that are associated with PCOS phenotypes. Specifically, metagenomic sequencing of the gut microbiome of adolescent and premenopausal age- and BMI-matched women from diverse ethnic and geographical backgrounds with and without PCOS will be needed to further understand the relationship of PCOS with dysbiosis of the gut microbiome. Mechanistic studies using rodent models of PCOS will also be required to understand how these microbes influence the development and pathogenesis of PCOS.
Interplay Between Gut Dysbiosis and Hyperandrogenism in PCOS
Gut dysbiosis as a driver for HA in PCOS
While the etiology of PCOS remains unknown, there are 2 distinct but related hypotheses that could connect the development of hyperandrogenic PCOS phenotypes to changes in the gut microbiome. As discussed previously, 1 hypothesis proposed by Tremellen and Pearce is that gut dysbiosis, influenced specifically by a high-fat diet (HFD) and a high-carbohydrate diet, leads to inflammation through disruption of the gut barrier, which in turn leads to IR, HA, and ovarian dysfunction [17]. This hypothesis places a strong emphasis on diet and gut dysbiosis as driving factors from which pathogenic features of PCOS, such as HA, emerge. However, it also suggests that obesity and IR are prerequisites for PCOS, although it is well documented that not all women diagnosed with PCOS are obese or insulin resistant [52, 53]. In addition, this hypothesis does not take into account that the incidence of PCOS is relatively similar in countries world wide despite differences in diet [2] and that many animal models of PCOS have been recreated independent of diet [54–58].
In order to begin to mechanistically understand the role of the gut microbiome in the development of PCOS, fecal microbiome transplant (FMT) experiments using stool from women with PCOS are informative. Qi et al performed an FMT from women with PCOS that are normal weight into antibiotic-treated mice and observed reproductive and metabolic changes in the recipient mice [23]. Significant increases in testosterone and luteinizing hormone (LH) levels were observed in mice receiving FMT from women with PCOS (trans-PCOS) compared with mice receiving FMT from healthy women (trans-Control) [23]. In addition to HA, trans-PCOS mice had disrupted estrous cyclicity, decreased ovulation as indicated by a decreased number of corpora lutea in the ovaries, the appearance of ovarian cysts, and a decrease in fertility [23]. Additionally, trans-PCOS mice developed IR as assessed by glucose and insulin tolerance tests and the homeostatic model assessment for IR calculated based on fasting glucose and insulin levels [23]. The researchers also transplanted one of the bacteria, Bacteroides vulgatus, that was positively associated with PCOS into antibiotic-treated mice [23]. Similar reproductive and metabolic phenotypes were observed with B. vulgatus compared with trans-PCOS [23]. This potentially ground-breaking study suggests that transplantation of a dysbiotic gut microbiome from women with PCOS or B. vulgatus is sufficient to induce a PCOS-like phenotype in mice and supports the idea that changes in the gut microbiome may play a causal role in this disorder (Fig. 2).
Figure 2.
Relationship between the gut microbiome and polycystic ovary syndrome (PCOS). Accumulating evidence, in human studies and rodent models, indicates that there is an association between dysbiosis of the gut microbiome and PCOS. A–B: Gut microbes metabolize substrates that enter the gut, from the diet and the host, and produce metabolites that may act directly on the intestines or enter systemic circulation and influence various host tissues whose function is altered in PCOS, such as ovary, skeletal muscle, liver, and adipose tissue. Gut bacterial metabolites reported to be altered in PCOS include secondary bile acids, SCFAs, and TMA. For instance, bile acids bind to receptors, including FXR, in various tissues and activate intracellular signaling. C: Metabolic tissues, including skeletal muscle, liver, and adipocytes, produce metabolites (such as conjugated primary and secondary bile acids, TMAO, lactate, and glucose) that enter the gut and may alter the composition of gut bacteria by serving as substrates, thus providing selective advantages to certain strains of bacteria over others. D: The host reproductive axis regulates sex steroid hormone production. In PCOS, elevated levels of androgens may alter the composition of the gut microbial community. E: Crosstalk between host metabolic tissues and the reproductive axis also occurs independently of the gut microbiome and may be a driver of the pathology and development of PCOS. Further studies are needed to decipher how the interactions outlined in this figure occur mechanistically. Abbreviations: FSH, follicle-stimulating hormone; FXR, farnesoid X receptor; GnRH, gonadotropin-releasing hormone; IR, insulin resistance; LH, luteinizing hormone; SCFAs, short-chain fatty acids; TMA, trimethylamine; TMAO, trimethylamine N-oxide.
Caveats for this study include the use of antibiotics to deplete the gut microbiome instead of the use of germ-free mice. Antibiotics have been shown to affect metabolism in mice [59], and no data were provided to demonstrate that the gut microbiome was actually depleted by antibiotic treatment prior to the FMT or that the FMT resulted in the establishment of gut microbes after the FMT. In addition, no data were provided on whether the mice gained weight or not; thus, it is unclear if a dysbiotic gut microbiome from women with PCOS and normal weight could induce obesity in mice. Future studies where the gut microbiome of recipient mice is sampled and sequenced pre- and post-FMT or bacterial transplantation will provide more comprehensive information about the role of the gut microbiome in the emergence of PCOS-like symptoms in mice. Additionally, using germ-free mice as recipients will clarify the direct effects of gut microbiota on the development of PCOS and whether obesity and IR precede HA [60]. Finally, FMT from women with PCOS that are normal weight and obese as well as non-IR versus IR will help parse out the role of different gut microbiota on the development of the different metabolic phenotypes associated with PCOS.
HA as a driver for gut dysbiosis in PCOS
A second hypothesis is that HA leads to gut dysbiosis in association with the development of PCOS (Fig. 1). Potential mechanisms through which testosterone could alter the gut microbiome include a direct effect as a substrate for gut microbial enzymes and an indirect effect via activation of host androgen receptors or modulation of the immune system (reviewed previously [61, 62]). Although human studies cannot be used to determine causation of gut dysbiosis by HA, it is notable that multiple studies reported correlations between HA and changes in the gut microbiome. Both alpha diversity [22, 28, 37] and beta diversity [28] were associated with HA, indicating that higher testosterone levels are linked with changes in the overall composition of the gut microbial community. Within the phylum Actinobacteria, the genus Collinsella was positively correlated with testosterone levels in 2 studies [25, 26] (Table 5). Within the phylum Bacteroidetes, the genus Bacteroides was positively correlated with testosterone levels in 4 studies [20, 22, 24, 25]. Furthermore, the genus Prevotella was negatively correlated with the levels of testosterone in 1 study [24] and positively correlated with testosterone in 3 studies [25–27]. Within the phylum Proteobacteria, Enterobacteriaceae were positively correlated with testosterone [20–22] (Table 5).
Table 5.
Changes in gut bacteria correlated with testosterone levels in women and PCOS rodent models
Phylum | Family | Genus | Correlation | Host | Ref |
---|---|---|---|---|---|
Actinobacteria | Bifidobacteriaceae | Bifidobacterium | Negative | Human | [25] |
Actinobacteria | Bifidobacteriaceae | Bifidobacterium | Negative | Rodent | [49] |
Actinobacteria | Coriobacteriaceae | Collinsella | Positive | Human | [25] |
Actinobacteria | Coriobacteriaceae | Collinsella | Positive | Human | [26] |
Actinobacteria | Positive | Rodent | [49] | ||
Bacteroidetes | Bacteroidaceae | Bacteroides | Positive | Human | [20] |
Bacteroidetes | Bacteroidaceae | Bacteroides | Positive | Human | [22] |
Bacteroidetes | Bacteroidaceae | Bacteroides | Positive | Human | [24] |
Bacteroidetes | Bacteroidaceae | Bacteroides | Positive | Human | [25] |
Bacteroidetes | Bacteroidaceae | Bacteroides | Positive | Rodent | [49] |
Bacteroidetes | Bacteroidaceae | Positive | Human | [24] | |
Bacteroidetes | Porphyromonadaceae | Parabacteroides | Positive | Human | [20] |
Bacteroidetes | Porphyromonadaceae | Negative | Human | [37] | |
Bacteroidetes | Prevotellaceae | Prevotella | Negative | Human | [24] |
Bacteroidetes | Prevotellaceae | Prevotella | Positive | Human | [25] |
Bacteroidetes | Prevotellaceae | Prevotella | Positive | Human | [26] |
Bacteroidetes | Prevotellaceae | Prevotella | Positive | Human | [27] |
Bacteroidetes | Prevotellaceae | Prevotella | Positive | Rodent | [30] |
Bacteroidetes | Prevotellaceae | Negative | Human | [24] | |
Bacteroidetes | S24-7 | Negative | Human | [19] | |
Firmicutes | Clostridiaceae | Anaerotruncus | Positive | Rodent | [32] |
Firmicutes | Clostridiaceae | Clostridium | Positive | Rodent | [32] |
Firmicutes | Hungateiclostridiaceae | Ruminiclostridium | Negative | Human | [27] |
Firmicutes | Lachnospiraceae | Butyrivibrio | Negative | Rodent | [49] |
Firmicutes | Lachnospiraceae | Coprococcus | Positive | Human | [25] |
Firmicutes | Lachnospiraceae | Fusicatenibacter | Negative | Human | [27] |
Firmicutes | Lachnospiraceae | Tyzzerella | Negative | Human | [27] |
Firmicutes | Lachnospiraceae | Tyzzerella | Positive | Rodent | [32] |
Firmicutes | Lachnospiraceae-ND3007 | Negative | Human | [27] | |
Firmicutes | Lactobacillaceae | Lactobacillus | Negative | Rodent | [49] |
Firmicutes | Rumiacoccaceae-UCG-003 | Negative | Human | [27] | |
Firmicutes | Ruminococcaceae | Faecalibacterium | Negative | Human | [25] |
Firmicutes | Ruminococcaceae | Ruminococcus | Positive | Human | [26] |
Firmicutes | Ruminococcaceae | Ruminococcus | Positive | Rodent | [32] |
Firmicutes | Ruminococcaceae | Subdoligranulum | Negative | Human | [27] |
Firmicutes | Ruminococcaceae | Negative | Human | [22] | |
Firmicutes | Ruminococcaceae | Positive | Human | [34] | |
Firmicutes | Streptococcaceae | Streptococcus | Positive | Human | [22] |
Firmicutes | Streptococcaceae | Streptococcus | Positive | Rodent | [49] |
Firmicutes | Veillonellaceae | Megasphaera | Positive | Human | [26] |
Firmicutes | Negative | Rodent | [49] | ||
Proteobacteria | Desulfovibrionaceae | Desulfovibrio | Negative | Rodent | [32] |
Proteobacteria | Desulfovibrionaceae | Desulfovibrio | Negative | Rodent | [49] |
Proteobacteria | Enterobacteriaceae | Escherichia | Positive | Human | [22] |
Proteobacteria | Enterobacteriaceae | Raoultella | Positive | Human | [21] |
Proteobacteria | Enterobacteriaceae | Shigella | Positive | Human | [22] |
Proteobacteria | Enterobacteriaceae | Positive | Human | [20] | |
Tenericutes | Negative | Human | [19] | ||
Verrucomicrobia | Akkermansiaceae | Akkermansia | Negative | Human | [22] |
Abbreviations: PCOS, polycystic ovary syndrome; Ref, reference number.
Evidence from rodent models also indicates that HA may drive dysbiosis of the gut microbiome (Fig. 1). Two early studies used letrozole, a nonsteroidal inhibitor of aromatase, to induce HA and PCOS-like symptoms in mice and rats. These studies showed that dysbiosis of the gut microbiome occurred as a consequence of treatment with letrozole, most likely due to the resulting HA rather than a direct effect of letrozole [18, 30]. In addition to reproductive and metabolic phenotypes similar to women with PCOS [58, 63], pubertal mice treated with letrozole had diet-independent reductions in alpha diversity, changes in beta diversity of the gut microbiome, and changes in the RA of specific bacteria [18] (Table 4). Bifidobacteriaceae was negatively correlated with testosterone while Bacteroides, Streptococcus [49], and Prevotella were positively correlated with testosterone [30], consistent with the human studies (Table 5). Two rodent studies showed that a proteobacteria called Desulfovibrio was negatively correlated with testosterone [32, 49], while this genus was not observed to be correlated with testosterone in humans (Table 5). Interestingly, these letrozole-induced changes in the gut microbiome appear to be activational rather than organizational changes; removal of letrozole treatment after the establishment of gut dysbiosis restored gut bacterial diversity [29]. Exogenous treatment of rats with DHT, which cannot be converted to estradiol, led to significant reductions in alpha diversity and changes in beta diversity compared with placebo-treated rats [32, 50]. Hyperandrogenism driven by dehydroepiandrosterone (DHEA) treatment altered beta diversity of the gut microbiome in mice when combined with an HFD [64]. Taken together, these results suggest a strong role for testosterone as a modulator of the gut microbiome, although it is unclear whether testosterone exerts an effect on gut microbes directly and/or indirectly through actions in androgen target tissues.
Caveats and future directions
The diagnosis of HA is obtained by measuring hirsutism with the Ferriman-Gallway score and/or biochemically by measuring serum testosterone levels. The “gold standard” method of measuring testosterone is liquid chromatography-mass spectrometry, although methods using radioimmunoassays can provide equivalent results, while other methods such as enzyme-linked immunosorbent assay (ELISA) can overestimate the amount of testosterone in the sample [10, 65]. In the studies reviewed herein, different methods of measuring testosterone were used, including ELISA-based techniques, radioimmunoassays, and liquid chromatography-mass spectrometry. Given that levels of steroid-hormone binding globulin (SHBG) are decreased in women with PCOS [66], it may also be relevant to examine the relationship between changes in the gut microbiome and the free androgen index (ratio of total testosterone to SHBG).
While the 2 hypotheses about the interaction between HA and gut dysbiosis were discussed separately, they are likely interconnected (Fig. 2). In 2017, vom Steeg and Klein reviewed evidence in support of the crosstalk between sex steroid hormones and the microbiota of the host [62]. However, much remains to be discovered about how sex steroid hormones interact with gut bacteria, especially with regards to how testosterone impacts the gut microbiome in females compared with males. To accomplish this, it will be important to employ metagenomic sequencing to identify gut microbial species/strains and microbial genes that are altered in response to increased levels of testosterone in women and PCOS rodent models. Moreover, transplantation of microbiota from women with PCOS or PCOS-like rodent models into germ-free mice will be crucial to comprehend the temporal changes in testosterone levels relative to other symptoms of PCOS as a result of exposure to PCOS-related microbiota. Finally, pharmacological inhibition of the androgen receptor using antiandrogens such as cyproterone or spironolactone [10] will help elucidate the role of androgen signaling in driving gut dysbiosis in women with PCOS or rodent models. Complementary studies using androgen receptor knockdown within specific host tissues will identify which sites of androgen action are required for gut dysbiosis.
Metabolites Associated With Gut Dysbiosis and PCOS
Although association does not equal causation, examining metabolite profiles in women with PCOS compared with healthy controls may provide insight into host/microbe interactions mediated by metabolites that can influence PCOS pathology. An early study in women showed that elevated levels of serum lactate were associated with PCOS [67], and host-produced lactate has been shown to enter the lumen of the gut and serve as a substrate for lactate-utilizing bacteria [68–70], potentially exerting a selective pressure within the microenvironment of the gut (Fig. 2). Moreover, 2 studies reported an association between increased serum levels of trimethylamine N-oxide (TMAO) and PCOS in women [71, 72] (Fig. 2). Trimethylamine N-oxid is a liver metabolite that originates from trimethylamine, which is produced by gut microbes from dietary precursors. Elevated levels of TMAO have been associated with an increased risk of cardiovascular disease [73]. While measuring microbiota-related metabolites in systemic circulation of women with PCOS may help identify mechanisms of regulation of host physiology by gut bacteria, investigating changes in metabolites in the gut or feces (as a proxy) may also highlight regulatory mechanisms. Along these lines, 1 study of the letrozole-induced PCOS rat model showed significant decreases in fecal SCFAs that are produced by gut bacterial fermentation of fiber and serve as signaling molecules in the host [49].
The relationship among bile acids, gut microbiota, and metabolic diseases (extensively reviewed elsewhere [74–77]) highlights an emerging key role for bile acids in regulating metabolic diseases including, potentially, PCOS. Primary bile acids serve as substrates for gut microbial enzymes that result in secondary bile acids that are recycled between the gut and liver via enterohepatic circulation [77]. An examination of glyco- and tauro-conjugated primary bile acids in systemic circulation showed that they were at higher levels in women with PCOS than in healthy women and were positively associated with HA [78]. On the other hand, another study reported that serum glycocholic acid was lower in women with PCOS than in women without the disorder [79]. Additionally, targeted metabolomics showed that the secondary bile acids, glycodeoxycholic acid (GDCA) and tauroursodeoxycholic acid (TUDCA), were lower in the serum and feces of women with PCOS and normal weight compared with healthy women [23]. It is intriguing that TUDCA was reported to be decreased in mice that received an FMT from women with PCOS or transplantation with B. vulgatus [23], suggesting that an altered gut microbiota was sufficient to cause changes in specific bile acid levels. Besides their functions in the absorption of lipids, bile acids act as signaling molecules by binding and activating receptors such as the farnesoid X receptor (FXR) [80–82] (Fig. 2), Takeda G protein receptor 5 [83], vitamin D receptor [84], pregnane X receptor [85], sphingosine-1-phosphate receptor 2 [86], and muscarinic M2 receptor [87]. One study showed that FXR-knockout mice exhibited glucose intolerance and IR when challenged with glucose or insulin, respectively [88]. This suggests a role for bile acid signaling through FXR in regulating metabolism. However, the contributions of specific bile acids and gut bacteria that deconjugate bile acids (such as Bacteroides [89]) in PCOS remain unclear (Fig. 2).
Caveats and future studies
Our understanding of how gut metabolites are altered in PCOS is extremely limited. Most of the studies have investigated metabolomic profiles associated with PCOS within systemic circulation, rather than the feces, and these studies have not been performed in a comprehensive manner due to the difficulty of identifying metabolites using untargeted metabolomics. Both liquid and gas chromatography coupled with mass spectrometry could be utilized to begin to comprehend the full array of gut microbial metabolites that are changed in the feces of women with PCOS, and these analyses should be complemented with quantitative, targeted metabolomics focused on specific metabolites such as bile acids and SCFAs (Fig. 2A and 2B). Moreover, correlations between gut metabolites and microbial species/strains obtained with metagenomic sequencing will help provide a more comprehensive picture of the important interactions occurring in the gut of women with PCOS and potentially shed light on how steroid hormones can influence the gut microbiome.
Treatments of Gut Dysbiosis in PCOS
Fecal microbial transplant
Since gut dysbiosis has been proposed as a driver of PCOS symptoms, treatment with an FMT from healthy donors or representative microbes from a healthy gut might serve as a useful therapy to re-diversify the gut microbiome, although there are limited studies to support this idea. One study showed that performing an FMT from healthy rats into a letrozole-induced PCOS rat model resulted in a reduction of androgen levels, improved estrous cycles, normalized ovarian morphology, and increased levels of Lactobacillus and Clostridium species and decreased Prevotella [30]. Additionally, a co-housing experiment showed improved reproductive and metabolic symptoms of PCOS in letrozole-treated mice that were co-housed with healthy, placebo-treated mice in addition to changing the RA of Coprobacillus and Lactobacillus [31]. Overall, these studies show promise that adjusting the gut microbial community in women with PCOS may alleviate some of the diet-independent, hyperandrogenic-induced symptoms.
Prebiotics: wheat dextrin and inulin
Resistant dextrin, a glucose polysaccharide that is fermented in the colon by microbes rather than absorbed in the small intestine, was given to women with PCOS and women without the disorder for 3 months [90]. Resistant dextrin lowered levels of free testosterone, hirsutism, the interval between menstrual cycles, fasting blood glucose, and lipid profile [90], but changes in alpha diversity or the RA of specific microbes was not assessed in this study. Additionally, the probiotic inulin was shown to improve gut dysbiosis, lower testosterone, and increase estradiol levels while improving ovarian morphology and weight gain in DHEA-treated mice fed an HFD [64]. However, alpha diversity was not assessed in this study, and thus, no conclusions can be specifically made about the role of inulin on the richness or evenness of the gut microbial community. In addition, it is unclear in mice whether inulin is targeting the effects of HFD or DHEA alone or in combination, so further studies investigating the effects of fermentable, dietary fiber on PCOS are warranted.
Probiotics: Bifidobacterium lactis, Lactobacillus, and lactic acid bacteria
Consuming probiotic (or beneficial) strains of bacteria has the potential to improve gut dysbiosis either directly through repopulation of the gut with healthy microbes or indirectly through the production of gut metabolites. Bifidobacterium lactis V9 given as a 10-week treatment for PCOS in 14 women with the disorder decreased LH and increased intestinal SCFAs [25]. Lactobacillus given to letrozole-treated rats reduced androgen levels, improved estrous cyclicity, normalized ovarian morphology, as well as increased Lactobacillus and Clostridium species and decreased Prevotella [30].
Several studies also investigated the effects of probiotic combinations on PCOS phenotypes. A combination of Bifidobacterium, Lactobacillus acidophilus, and Enterococcus faecalis in a DHT-induced PCOS rat model improved reproductive and metabolic functions as well as alpha diversity of the gut microbiome [50]. In addition, certain strains of Lactobacillus and Bifidobacterium (HL2 and HB3) given to letrozole-treated rats protected against pathological changes in the ovaries and restored testosterone levels while upregulating levels of SCFAs [48].
Small molecules: metformin and IL22
Previous studies have shown that metformin decreases total testosterone, hirsutism, acne, LH, BMI, waist-to-hip ratio, and fasting insulin, and increases SHBG, follicle-stimulating hormone, and progesterone, while it also improves menstrual cycles [91, 92]. In addition to its role in the inhibition of androgen biosynthesis [93, 94], metformin treatment of individuals with T2D decreased the RA of Bacteroides fragilis and FXR signaling, while it increased levels of glycoursodeoxycholic acid in the gut in 1 study [95], as well as the RA of Akkermansia muciniphila and SCFA-producing microbes in another study [96]. Although no studies have investigated the effect of metformin on the gut microbiome of women with PCOS, 1 study in DHEA-treated mice showed that it improved dysbiosis of the gut, including increased levels of Bacteroidetes and decreased levels of Helicobacter and Proteobacteria [64]. In this mouse model, metformin also decreased testosterone levels while also improving ovarian function, weight gain, and IR [64].
IL22 is an anti-inflammatory cytokine produced by cells of the lymphoid lineage, including cells in the lamina propria of the intestinal wall called group 3 innate lymphoid cells (ILC3) [97]. IL22 was used to treat symptoms of PCOS in mice that received an FMT from women with PCOS, a transplantation of B. vulgatus, or treated with DHEA [23, 98]. In all of these models, IL22 improved reproductive and metabolic symptoms including a decrease in testosterone levels, increased insulin tolerance, and enhanced browning of adipose tissue [23, 98]. However, it is unknown whether IL22 altered the composition or function of gut microbiome or if other mechanisms were responsible for the beneficial effect. Qi et al hypothesized that an increase in the abundance of B. vulgatus, and potentially other bacteria in the intestine of women with PCOS, results in increased deconjugation of secondary bile acids and lower levels of GDCA and TUDCA, which normally bind to receptors involved in the production of IL22 [23]. Since there is no current evidence demonstrating that GDCA and TUDCA bind to receptors on ILC3 cells in vivo and that this binding leads to the production of IL22, future studies in PCOS rodent models will be useful to identify specific mechanisms by which changes in gut bacteria regulate secondary bile acids and IL22 production, as well as how IL22 signaling influences reproductive and metabolic phenotypes of PCOS.
Summary
In summary, this review highlights recent progress made in understanding the relationship between PCOS and dysbiosis of the gut microbiome in both humans and rodent models. Although there is considerable variability in the results obtained from 16S rRNA and metagenomic gene sequencing, many studies support the idea that changes in gut microbiota are associated with PCOS, including a decrease in biodiversity as well as changes in specific bacterial taxa. Notably, these changes occur both in adolescent girls and women with this disorder that are normal weight or obese, suggesting that a gut dysbiosis manifests along with other features of PCOS during puberty independently of BMI, although it is unclear whether obesity influences changes in the gut microbiome observed in PCOS. In addition, changes in gut microbiota are correlated with HA, indicating that elevated levels of testosterone may regulate the composition of the gut microbiome in females. We explored hypotheses that HA as well as dysbiosis of the gut microbiome could act as drivers of PCOS through their interaction with each other, although future studies are needed to understand the mechanisms involved. Moreover, we reviewed the limited studies that investigated changes in specific microbial metabolites associated with PCOS. While preliminary, these studies justify further exploration of this understudied area, with the exciting potential to uncover novel targets for small molecule therapeutics focused on PCOS. Finally, we reviewed studies investigating the efficacy of prebiotic, probiotic, and postbiotic treatments that modulate the gut microbiome and, in turn, improve symptoms of PCOS in both human and rodent studies. Although most of these studies need to be reproduced, the positive results on reproductive and metabolic features of PCOS from treatment with dietary fibers, probiotics such as Bifidobacterium and Lactobacillus, and small molecules including bile acids and anti-inflammatory cytokines indicate that this is an area deserving of future study.
Acknowledgments
Financial Support: This work was funded by the National Institute of Child Health and Human Development through a cooperative agreement as part of the National Centers for Translational Research in Reproduction and Infertility (Grant P50 HD012303) to V.G.T.. V.G.T. was funded by Grant R01 HD095412 and M.G.R. was funded by T32 HD007203.
Glossary
Abbreviations
- BMI
body mass index
- DHEA
dehydroepiandrosterone
- DHT
dihydrotestosterone
- ELISA
enzyme-linked immunosorbent assay
- FMT
fecal microbiome transplant
- FXR
farnesoid X receptor
- GDCA
glycodeoxycholic acid
- HA
hyperandrogenism
- HFD
high-fat diet
- ILC3
group 3 innate lymphoid cells
- IR
insulin resistance
- LH
luteinizing hormone
- PCOS
polycystic ovary syndrome
- RA
relative abundance
- rRNA
ribosomal ribonucleic acid
- SCFAs
short-chain fatty acids
- SHBG
steroid-hormone binding globulin
- TMAO
trimethylamine N-oxide
- T2D
type 2 diabetes
- TUDCA
tauroursodeoxycholic acid
Additional Information
Disclosure Summary: The authors have nothing to disclose.
Data Availability
Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.
References and Notes
- 1. Rao P, Bhide P. Controversies in the diagnosis of polycystic ovary syndrome. Ther Adv Reprod Health. 2020;14:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Wolf WM, Wattick RA, Kinkade ON, Olfert MD. Geographical prevalence of polycystic ovary syndrome as determined by region and race/ethnicity. Int J Environ Res Public Health. 2018;15(11):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Teede HJ, Misso ML, Costello MF, et al. ; International PCOS Network . Recommendations from the international evidence-based guideline for the assessment and management of polycystic ovary syndrome. Hum Reprod. 2018;33(9):1602–1618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Hull MG. Epidemiology of infertility and polycystic ovarian disease: endocrinological and demographic studies. Gynecol Endocrinol. 1987;1(3):235–245. [DOI] [PubMed] [Google Scholar]
- 5. Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES, Yildiz BO. The prevalence and features of the polycystic ovary syndrome in an unselected population. J Clin Endocrinol Metab. 2004;89(6):2745–2749. [DOI] [PubMed] [Google Scholar]
- 6. Goodman NF, Cobin RH, Futterweit W, Glueck JS, Legro RS, Carmina E; American Association of Clinical Endocrinologists (AACE) ; American College of Endocrinology (ACE); Androgen Excess and PCOS Society. 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):1415–1426. [DOI] [PubMed] [Google Scholar]
- 7. Palomba S, de Wilde MA, Falbo A, Koster MP, La Sala GB, Fauser BC. Pregnancy complications in women with polycystic ovary syndrome. Hum Reprod Update. 2015;21(5):575–592. [DOI] [PubMed] [Google Scholar]
- 8. Barber TM, Wass JA, McCarthy MI, Franks S. Metabolic characteristics of women with polycystic ovaries and oligo-amenorrhoea but normal androgen levels: implications for the management of polycystic ovary syndrome. Clin Endocrinol (Oxf). 2007;66(4):513–517. [DOI] [PubMed] [Google Scholar]
- 9. Moghetti P, Tosi F, Bonin C, et al. Divergences in insulin resistance between the different phenotypes of the polycystic ovary syndrome. J Clin Endocrinol Metab. 2013;98(4):E628–E637. [DOI] [PubMed] [Google Scholar]
- 10. Goodman NF, Cobin RH, Futterweit W, Glueck JS, Legro RS, Carmina E; American Association of Clinical Endocrinologists (AACE) ; American College of Endocrinology (ACE); Androgen Excess and PCOS Society (AES). 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 1. Endocr Pract. 2015;21(11):1291–1300. [DOI] [PubMed] [Google Scholar]
- 11. Azziz R, Carmina E, Chen Z, et al. Polycystic ovary syndrome. Nat Rev Dis Primers. 2016;2):1–18. [DOI] [PubMed] [Google Scholar]
- 12. Churchill SJ, Wang ET, Pisarska MD. Metabolic consequences of polycystic ovary syndrome. Minerva Ginecol. 2015;67(6):545–555. [PubMed] [Google Scholar]
- 13. Vassilatou E. Nonalcoholic fatty liver disease and polycystic ovary syndrome. World J Gastroenterol. 2014;20(26):8351–8363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Yang R, Yang S, Li R, Liu P, Qiao J, Zhang Y. Effects of hyperandrogenism on metabolic abnormalities in patients with polycystic ovary syndrome: a meta-analysis. Reprod Biol Endocrinol. 2016;14(1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Barko PC, McMichael MA, Swanson KS, Williams DA. The gastrointestinal microbiome: a review. J Vet Intern Med. 2018;32(1):9–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Durack J, Lynch SV. The gut microbiome: relationships with disease and opportunities for therapy. J Exp Med. 2019;216(1): 20–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Tremellen K, Pearce K. Dysbiosis of gut microbiota (DOGMA)–a novel theory for the development of polycystic ovarian syndrome. Med Hypotheses. 2012;79(1):104–112. [DOI] [PubMed] [Google Scholar]
- 18. 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):1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Lindheim L, Bashir M, Münzker J, 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):1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Chu W, Han Q, Xu J, et al. Metagenomic analysis identified microbiome alterations and pathological association between intestinal microbiota and polycystic ovary syndrome. Fertil Steril. 2020;113(6):1286–1298.e4. [DOI] [PubMed] [Google Scholar]
- 21. Insenser M, Murri M, Del Campo R, Martínez-García MÁ, 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):2552–2562. [DOI] [PubMed] [Google Scholar]
- 22. Liu R, Zhang C, Shi Y, et al. Dysbiosis of gut microbiota associated with clinical parameters in polycystic ovary syndrome. Front Microbiol. 2017;8:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Qi X, Yun C, Sun L, et al. Gut microbiota-bile acid-interleukin-22 axis orchestrates polycystic ovary syndrome. Nat Med. 2019;25(8):1225–1233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Zeng B, Lai Z, Sun L, 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. [DOI] [PubMed] [Google Scholar]
- 25. Zhang J, Sun Z, Jiang S, et al. Probiotic Bifidobacterium lactis V9 regulates the secretion of sex hormones in polycystic ovary syndrome patients through the gut-brain Axis. mSystems. 2019;4(2):1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Zhou L, Ni Z, Cheng W, et al. Characteristic gut microbiota and predicted metabolic functions in women with PCOS. Endocr Connect. 2020;9(1):63–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Liang Y, Ming Q, Liang J, Zhang Y, Zhang H, Shen T. Gut microbiota dysbiosis in polycystic ovary syndrome: association with obesity – a preliminary report. Can J Physiol Pharmacol. 2020;98(11):803–809. [DOI] [PubMed] [Google Scholar]
- 28. Torres PJ, Siakowska M, Banaszewska B, et al. Gut microbial diversity in women with polycystic ovary syndrome correlates with hyperandrogenism. J Clin Endocrinol Metab. 2018;103(4):1502–1511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Arroyo P, Ho BS, Sau L, Kelley ST, Thackray VG. Letrozole treatment of pubertal female mice results in activational effects on reproduction, metabolism and the gut microbiome. Plos One. 2019;14(9):1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Guo Y, Qi Y, Yang X, et al. Association between polycystic ovary syndrome and gut microbiota. Plos One. 2016;11(4):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Torres PJ, Ho BS, Arroyo P, et al. Exposure to a healthy gut microbiome protects against reproductive and metabolic dysregulation in a PCOS mouse model. Endocrinology. 2019;160(5):1193–1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Zheng Y, Yu J, Liang C, Li S, Wen X, Li Y. Characterization on gut microbiome of PCOS rats and its further design by shifts in high-fat diet and dihydrotestosterone induction in PCOS rats. Bioprocess Biosyst Eng. 2020. 10.1007/s00449-020-02320-w [DOI] [PubMed] [Google Scholar]
- 33. Haudum C, Lindheim L, Ascani A, et al. Impact of short-term isoflavone intervention in polycystic ovary syndrome (PCOS) patients on microbiota composition and metagenomics. Nutrients. 2020;12(6):1–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Eyupoglu ND, Ergunay K, Acikgoz A, Akyon Y, Yilmaz E, Yildiz BO. Gut microbiota and oral contraceptive use in overweight and obese patients with polycystic ovary syndrome. J Clin Endocrinol Metab. 2020:1–9. [DOI] [PubMed] [Google Scholar]
- 35. Rotterdam EA-SPCWG. Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome. Fertil Steril. 2004;81(1):19–25. [DOI] [PubMed] [Google Scholar]
- 36. 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):41–47. [DOI] [PubMed] [Google Scholar]
- 37. Jobira B, Frank DN, Pyle L, et al. Obese adolescents with PCOS have altered biodiversity and relative abundance in gastrointestinal microbiota. J Clin Endocrinol Metab. 2020;105(6):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Peña AS, Witchel SF, Hoeger KM, et al. Adolescent polycystic ovary syndrome according to the international evidence-based guideline. BMC Med. 2020;18(1):1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Tilman D, Reich PB, Knops J, Wedin D, Mielke T, Lehman C. Diversity and productivity in a long-term grassland experiment. Science. 2001;294(5543):843–845. [DOI] [PubMed] [Google Scholar]
- 40. Kowalska-Duplaga K, Gosiewski T, Kapusta P, et al. Differences in the intestinal microbiome of healthy children and patients with newly diagnosed Crohn’s disease. Sci Rep. 2019;9(1):18880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Duvallet C, Gibbons SM, Gurry T, Irizarry RA, Alm EJ. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat Commun. 2017;8(1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Mosca A, Leclerc M, Hugot JP. Gut microbiota diversity and human diseases: should we reintroduce key predators in our ecosystem? Front Microbiol. 2016;7:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Menni C, Lin C, Cecelja M, et al. Gut microbial diversity is associated with lower arterial stiffness in women. Eur Heart J. 2018;39(25):2390–2397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Menni C, Zhu J, Le Roy CI, et al. Serum metabolites reflecting gut microbiome alpha diversity predict type 2 diabetes. Gut Microbes. 2020;11(6):1632–1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Nageshwar Reddy D. Role of the normal gut microbiota. World J Gastroenterol. 2015;21(29):8787–8803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Stener-Victorin E, Padmanabhan V, Walters KA, et al. Animal models to understand the etiology and pathophysiology of polycystic ovary syndrome. Endocr Rev. 2020;41(4):538–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Torres PJ, Skarra DV, Ho BS, et al. Letrozole treatment of adult female mice results in a similar reproductive phenotype but distinct changes in metabolism and the gut microbiome compared to pubertal mice. BMC Microbiol. 2019;19(1):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. He Y, Wang Q, Li X, et al. Lactic acid bacteria alleviate polycystic ovarian syndrome by regulating sex hormone related gut microbiota. Food Funct. 2020;11(6):5192–5204. [DOI] [PubMed] [Google Scholar]
- 49. Wang T, Sha L, Li Y, et al. Dietary α-linolenic acid-rich flaxseed oil exerts beneficial effects on polycystic ovary syndrome through sex steroid hormones-microbiota-inflammation axis in rats. Front Endocrinol (Lausanne). 2020;11:1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Zhang F, Ma T, Cui P, et al. Diversity of the gut microbiota in dihydrotestosterone-induced PCOS rats and the pharmacologic effects of diane-35, probiotics, and berberine. Front Microbiol. 2019;10:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Allaband C, McDonald D, Vázquez-Baeza Y, et al. Microbiome 101: studying, analyzing, and interpreting gut microbiome data for clinicians. Clin Gastroenterol Hepatol. 2019;17(2):218–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Zhu S, Zhang B, Jiang X, et al. Metabolic disturbances in non-obese women with polycystic ovary syndrome: a systematic review and meta-analysis. Fertil Steril. 2019;111(1):168–177. [DOI] [PubMed] [Google Scholar]
- 53. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Kafali H, Iriadam M, Ozardali I, Demir N. Letrozole-induced polycystic ovaries in the rat: a new model for cystic ovarian disease. Arch Med Res. 2004;35(2):103–108. [DOI] [PubMed] [Google Scholar]
- 55. Mannerås L, Cajander S, Holmäng A, et al. A new rat model exhibiting both ovarian and metabolic characteristics of polycystic ovary syndrome. Endocrinology. 2007;148(8):3781–3791. [DOI] [PubMed] [Google Scholar]
- 56. van Houten EL, Kramer P, McLuskey A, Karels B, Themmen AP, Visser JA. Reproductive and metabolic phenotype of a mouse model of PCOS. Endocrinology. 2012;153(6):2861–2869. [DOI] [PubMed] [Google Scholar]
- 57. Park JH, Choi TS. Polycystic ovary syndrome (PCOS)-like phenotypes in the d-galactose-induced aging mouse model. Biochem Biophys Res Commun. 2012;427(4):701–704. [DOI] [PubMed] [Google Scholar]
- 58. Kauffman AS, Thackray VG, Ryan GE, et al. A novel letrozole model recapitulates both the reproductive and metabolic phenotypes of polycystic ovary syndrome in female mice. Biol Reprod. 2015;93(3):1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Yang JH, Bhargava P, McCloskey D, Mao N, Palsson BO, Collins JJ. Antibiotic-induced changes to the host metabolic environment inhibit drug efficacy and alter immune function. Cell Host Microbe. 2017;22(6):757–765.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Park JC, Im SH. Of men in mice: the development and application of a humanized gnotobiotic mouse model for microbiome therapeutics. Exp Mol Med. 2020;52(9):1383–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Thackray VG. Sex, microbes, and polycystic ovary syndrome. Trends Endocrinol Metab. 2019;30(1):54–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Vom Steeg LG, Klein SL. Sex steroids mediate bidirectional interactions between hosts and microbes. Horm Behav. 2017;88:45–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Skarra DV, Hernández-Carretero A, Rivera AJ, Anvar AR, Thackray VG. Hyperandrogenemia induced by letrozole treatment of pubertal female mice results in hyperinsulinemia prior to weight gain and insulin resistance. Endocrinology. 2017;158(9):2988–3003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Xue J, Li X, Liu P, et al. Inulin and metformin ameliorate polycystic ovary syndrome via anti-inflammation and modulating gut microbiota in mice. Endocr J. 2019;66(10):859–870. [DOI] [PubMed] [Google Scholar]
- 65. Janse F, Eijkemans MJ, Goverde AJ, et al. Assessment of androgen concentration in women: liquid chromatography-tandem mass spectrometry and extraction RIA show comparable results. Eur J Endocrinol. 2011;165(6):925–933. [DOI] [PubMed] [Google Scholar]
- 66. Deswal R, Yadav A, Dang AS. Sex hormone binding globulin – an important biomarker for predicting PCOS risk: a systematic review and meta-analysis. Syst Biol Reprod Med. 2018;64(1):12–24. [DOI] [PubMed] [Google Scholar]
- 67. Zhao Y, Fu L, Li R, et al. Metabolic profiles characterizing different phenotypes of polycystic ovary syndrome: plasma metabolomics analysis. BMC Med. 2012;10:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Scheiman J, Luber JM, Chavkin TA, et al. Meta-omics analysis of elite athletes identifies a performance-enhancing microbe that functions via lactate metabolism. Nat Med. 2019;25(7):1104–1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Hove H, Nordgaard-Andersen I, Mortensen PB. Faecal DL-lactate concentration in 100 gastrointestinal patients. Scand J Gastroenterol. 1994;29(3):255–259. [DOI] [PubMed] [Google Scholar]
- 70. Gillis CC, Hughes ER, Spiga L, et al. Dysbiosis-associated change in host metabolism generates lactate to support salmonella growth. Cell Host Microbe. 2018;23(1):54–64 e56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Eyupoglu ND, Caliskan Guzelce E, Acikgoz A, et al. Circulating gut microbiota metabolite trimethylamine N-oxide and oral contraceptive use in polycystic ovary syndrome. Clin Endocrinol (Oxf). 2019;91(6):810–815. [DOI] [PubMed] [Google Scholar]
- 72. Huang J, Liu L, Chen C, Gao Y. PCOS without hyperandrogenism is associated with higher plasma trimethylamine N-oxide levels. BMC Endocr Disord. 2020;20(1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Velasquez MT, Ramezani A, Manal A, Raj DS. Trimethylamine N-oxide: the good, the bad and the unknown. Toxins (Basel). 2016;8(11):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. McGlone ER, Bloom SR. Bile acids and the metabolic syndrome. Ann Clin Biochem. 2019;56(3):326–337. [DOI] [PubMed] [Google Scholar]
- 75. Li C, Li Y, Gai Z. Bile acids and farnesoid X receptor: novel target for the treatment of diabetic cardiomyopathy. Curr Protein Pept Sci. 2019;20(10):976–983. [DOI] [PubMed] [Google Scholar]
- 76. Jia ET, Liu ZY, Pan M, Lu JF, Ge QY. Regulation of bile acid metabolism-related signaling pathways by gut microbiota in diseases. J Zhejiang Univ Sci B. 2019;20(10):781–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Wahlström A, Sayin SI, Marschall HU, Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016;24(1):41–50. [DOI] [PubMed] [Google Scholar]
- 78. Zhang B, Shen S, Gu T, et al. Increased circulating conjugated primary bile acids are associated with hyperandrogenism in women with polycystic ovary syndrome. J Steroid Biochem Mol Biol. 2019;189:171–175. [DOI] [PubMed] [Google Scholar]
- 79. Jia C, Xu H, Xu Y, Xu Y, Shi Q. Serum metabolomics analysis of patients with polycystic ovary syndrome by mass spectrometry. Mol Reprod Dev. 2019;86(3):292–297. [DOI] [PubMed] [Google Scholar]
- 80. Wang H, Chen J, Hollister K, Sowers LC, Forman BM. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol Cell. 1999;3(5):543–553. [DOI] [PubMed] [Google Scholar]
- 81. Makishima M, Okamoto AY, Repa JJ, et al. Identification of a nuclear receptor for bile acids. Science. 1999;284(5418):1362–1365. [DOI] [PubMed] [Google Scholar]
- 82. Parks DJ, Blanchard SG, Bledsoe RK, et al. Bile acids: natural ligands for an orphan nuclear receptor. Science. 1999;284(5418):1365–1368. [DOI] [PubMed] [Google Scholar]
- 83. Maruyama T, Miyamoto Y, Nakamura T, et al. Identification of membrane-type receptor for bile acids (M-BAR). Biochem Biophys Res Commun. 2002;298(5):714–719. [DOI] [PubMed] [Google Scholar]
- 84. Makishima M, Lu TT, Xie W, et al. Vitamin D receptor as an intestinal bile acid sensor. Science. 2002;296(5571):1313–1316. [DOI] [PubMed] [Google Scholar]
- 85. Goodwin B, Gauthier KC, Umetani M, et al. Identification of bile acid precursors as endogenous ligands for the nuclear xenobiotic pregnane X receptor. Proc Natl Acad Sci U S A. 2003;100(1):223–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Studer E, Zhou X, Zhao R, et al. Conjugated bile acids activate the sphingosine-1-phosphate receptor 2 in primary rodent hepatocytes. Hepatology. 2012;55(1):267–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Raufman JP, Chen Y, Cheng K, Compadre C, Compadre L, Zimniak P. Selective interaction of bile acids with muscarinic receptors: a case of molecular mimicry. Eur J Pharmacol. 2002;457(2–3):77–84. [DOI] [PubMed] [Google Scholar]
- 88. Cariou B, van Harmelen K, Duran-Sandoval D, et al. The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. J Biol Chem. 2006;281(16):11039–11049. [DOI] [PubMed] [Google Scholar]
- 89. .Yao L, Seaton SC, Ndousse-Fetter S, et al. A selective gut bacterial bile salt hydrolase alters host metabolism. Elife. 2018;7:1–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Shamasbi GS, Dehgan P, Charandabi SM-A, Aliasgarzadeh A, Mirghafourvand M. The effect of resistant dextrin as a prebiotic on metabolic parameters and androgen level in women with polycystic ovarian syndrome: a randomized, triple-blind, controlled, clinical trial. Eur J Nutr. 2019;58(2):629–640. [DOI] [PubMed] [Google Scholar]
- 91. Kolodziejczyk B, Duleba AJ, Spaczynski RZ, Pawelczyk L. Metformin therapy decreases hyperandrogenism and hyperinsulinemia in women with polycystic ovary syndrome. Fertil Steril. 2000;73(6):1149–1154. [DOI] [PubMed] [Google Scholar]
- 92. Ciçek MN, Bala A, Celik C, Akyürek C. The comparison of clinical and hormonal parameters in PCOS patients treated with metformin and GnRH analogue. Arch Gynecol Obstet. 2003;268(2):107–112. [DOI] [PubMed] [Google Scholar]
- 93. Lee BH, Indran IR, Tan HM, et al. A dietary medium-chain fatty acid, decanoic acid, inhibits recruitment of Nur77 to the HSD3B2 promoter in vitro and reverses endocrine and metabolic abnormalities in a rat model of polycystic ovary syndrome. Endocrinology. 2016;157(1):382–394. [DOI] [PubMed] [Google Scholar]
- 94. Hirsch A, Hahn D, Kempná P, et al. Metformin inhibits human androgen production by regulating steroidogenic enzymes HSD3B2 and CYP17A1 and complex I activity of the respiratory chain. Endocrinology. 2012;153(9):4354–4366. [DOI] [PubMed] [Google Scholar]
- 95. Sun L, Xie C, Wang G, et al. Gut microbiota and intestinal FXR mediate the clinical benefits of metformin. Nat Med. 2018;24(12):1919–1929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. de la Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, et al. Metformin is associated with higher relative abundance of mucin-degrading akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the gut. Diabetes Care. 2017;40(1):54–62. [DOI] [PubMed] [Google Scholar]
- 97. Dudakov JA, Hanash AM, van den Brink MR. Interleukin-22: immunobiology and pathology. Annu Rev Immunol. 2015;33:747–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Qi X, Yun C, Liao B, Qiao J, Pang Y. The therapeutic effect of interleukin-22 in high androgen-induced polycystic ovary syndrome. J Endocrinol. 2020;245(2):281–289. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.