Sex, puberty, and the gut microbiome

in Reproduction
Authors:
Laura Sisk-HackworthDepartment of Biology, San Diego State University, San Diego, California, USA

Search for other papers by Laura Sisk-Hackworth in
Current site
Google Scholar
PubMed
Close
,
Scott T KelleyDepartment of Biology, San Diego State University, San Diego, California, USA

Search for other papers by Scott T Kelley in
Current site
Google Scholar
PubMed
Close
, and
Varykina G ThackrayDepartment of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, California, USA

Search for other papers by Varykina G Thackray in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-6672-6117
View More View Less

Correspondence should be addressed to V Thackray; Email: vthackray@ucsd.edu
Free access

In brief

Sex differences in the gut microbiome may impact multiple aspects of human health and disease. In this study, we review the evidence for microbial sex differences in puberty and adulthood and discuss potential mechanisms driving differentiation of the sex-specific gut microbiome.

Abstract

In humans, the gut microbiome is strongly implicated in numerous sex-specific physiological processes and diseases. Given this, it is important to understand how sex differentiation of the gut microbiome occurs and how these differences contribute to host health and disease. While it is commonly believed that the gut microbiome stabilizes after 3 years of age, our review of the literature found considerable evidence that the gut microbiome continues to mature during and after puberty in a sex-dependent manner. We also review the intriguing, though sparse, literature on potential mechanisms by which host sex may influence the gut microbiome, and vice versa, via sex steroids, bile acids, and the immune system. We conclude that the evidence for the existence of a sex-specific gut microbiome is strong but that there is a dearth of research on how host–microbe interactions lead to this differentiation. Finally, we discuss the types of future studies needed to understand the processes driving the maturation of sex-specific microbial communities and the interplay between gut microbiota, host sex, and human health.

Abstract

In brief

Sex differences in the gut microbiome may impact multiple aspects of human health and disease. In this study, we review the evidence for microbial sex differences in puberty and adulthood and discuss potential mechanisms driving differentiation of the sex-specific gut microbiome.

Abstract

In humans, the gut microbiome is strongly implicated in numerous sex-specific physiological processes and diseases. Given this, it is important to understand how sex differentiation of the gut microbiome occurs and how these differences contribute to host health and disease. While it is commonly believed that the gut microbiome stabilizes after 3 years of age, our review of the literature found considerable evidence that the gut microbiome continues to mature during and after puberty in a sex-dependent manner. We also review the intriguing, though sparse, literature on potential mechanisms by which host sex may influence the gut microbiome, and vice versa, via sex steroids, bile acids, and the immune system. We conclude that the evidence for the existence of a sex-specific gut microbiome is strong but that there is a dearth of research on how host–microbe interactions lead to this differentiation. Finally, we discuss the types of future studies needed to understand the processes driving the maturation of sex-specific microbial communities and the interplay between gut microbiota, host sex, and human health.

Introduction

Next-generation sequencing (NGS) has revolutionized our understanding of the human gut microbiome. The combination of NGS, multiplex-library construction, and bioinformatics has revealed gut microbial diversity to be temporally dynamic and highly variable among both populations and individuals. Moreover, the gut microbial community collectively contains two orders of magnitude of more genes than the human genome (Zhu et al. 2010, Huttenhower et al. 2012, Falony et al. 2016, Levy et al. 2020). Variation in abiotic (pH, oxygen levels, nutrition) and biotic factors (immune surveillance, signaling molecules) creates numerous niches along the alimentary tract that select for highly differentiated communities of microorganisms (Barlow et al. 2020). Conversely, the microbes in the mammalian gut influence a broad range of host physiological parameters ranging from metabolism (Baars et al. 2018, Acharya et al. 2019, Colldén et al. 2019) to immunity (Markle et al. 2014, Candon et al. 2015, Vatanen et al. 2016), to the nervous system (Desbonnet et al. 2015, Murray et al. 2020, Heinzel et al. 2021), and to the reproductive system (Acharya et al. 2019, Arroyo et al. 2019).

The human gut microbiome develops rapidly after birth, when the alimentary canal is seeded by the mother and environmental microbiomes (Breitbart et al. 2008, Bäckhed et al. 2015, Dominguez-Bello et al. 2016, Ferretti et al. 2018, Helve et al. 2019, Song et al. 2021). Studies showed that the infant gut microbiome rapidly increases in diversity and begins to resemble adult gut microbiome taxonomic composition and functional pathways by 2–3 years of age (Kurokawa et al. 2007, Koenig et al. 2011, Yatsunenko et al. 2012, Bokulich et al. 2016). Based on these initial studies, the gut microbiome of young children was thought to be fully mature by age 3. However, the gut microbiomes of children can be readily distinguished from adults (Agans et al. 2011, Yatsunenko et al. 2012, Ringel-Kulka et al. 2013, Dominianni et al. 2015, Hollister et al. 2015).

Many important and profound changes in human physiology occur during puberty, which may also be the case with the gut microbiome. In humans, sex-specific processes have been linked to the gut microbiome, including cognition and anxiety (Desbonnet et al. 2015, Murray et al. 2019), liver metabolism (van Keulen et al. 2020, Bhat et al. 2021), immunity (Shepherd et al. 2020, Wilharm et al. 2021), and menstrual regularity (Wang et al. 2020a ). In addition, diseases which emerge during and after puberty have been linked to changes in gut microbial composition, often in a sex-specific manner. These include inflammatory bowel disease (IBD) (Ferguson & Sedgwick 1994, Franzosa et al. 2019), type I diabetes (T1D) (Markle et al. 2013, Yurkovetskiy et al. 2013), lupus (Young et al. 2014, He et al. 2016), obesity (Joriba et al. 2019), polycystic ovary syndrome (PCOS) (Torres et al. 2018, van Hooff et al. 1999, Jobira et al. 2020), and endometriosis (Yuan et al. 2018, Svensson et al. 2021). In addition to playing a role in the etiology and pathophysiology of various diseases, emerging differences in gut microbiota during puberty may lead to variable responses to drug treatments. Understanding the development of sex differences in the gut microbiome may be critical for designing and optimizing therapeutic approaches to treat gut microbiome-related diseases. Given the number of sex-specific diseases related to the gut and the potential for these sex differences to impact drug therapies, it is worth reviewing what is currently known about sex differentiation of the gut microbiome and the processes that drive maturation of the sex-specific gut microbial community.

Sex differences and the gut microbiome

A survey of the literature investigating the relationship between sex and the gut microbiome strongly supports the hypothesis that human and rodent gut microbiomes differentiate during puberty and that this differentiation results in sex-specific communities by adulthood. While early data from the Human Microbiome Project did not detect sex differences in the gut microbiome (Huttenhower et al. 2012), subsequent studies have consistently identified distinct gut microbial communities between males and females. Numerous studies in both mice and humans have found sex differences in gut microbiome beta diversity (between-sample taxonomic composition) (Fig. 1) (Mueller et al. 2006, Li et al. 2008, Markle et al. 2013, Ding & Schloss 2014, Dominianni et al. 2015, Org et al. 2016, Falony et al. 2016, Sheng et al. 2017, Borgo et al. 2018, Gao et al. 2018, Santos-Marcos et al. 2018, Sinha et al. 2019, Takagi et al. 2019, Mayneris-Perxachs et al. 2020, Cui et al. 2021, Peters et al. 2022). Multiple studies have also shown that alpha diversity (within-sample taxonomic diversity) tends to be higher in adult females than adult males, though these differences are less pronounced in older adults (Falony et al. 2016, Borgo et al. 2018, Gao et al. 2018, Cuesta-Zuluaga et al. 2019, Sinha et al. 2019, Takagi et al. 2019, Peters et al. 2022). Sex was also among the ten factors that most explained variability in human gut microbial composition (Falony et al. 2016).

Figure 1
Figure 1

Sex differences in the human gut microbiome arise during puberty. Studies providing evidence of sex differences are categorized by developmental stage, type of data analysis (e.g. 16S rRNA gene sequencing, metagenomics, or untargeted metabolomics) and observed effect on the gut microbiome. Created with BioRender.com.

Citation: Reproduction 165, 2; 10.1530/REP-22-0303

Since multiple studies support the idea that there are sex differences in the adult gut microbiome, it follows that there must be a point during development where the gut microbiome differentiates by sex. While initial studies reported that the microbial community in young children (1–3 years of age) resembled that of adults (Kurokawa et al. 2007, Yatsunenko et al. 2012), follow-up studies showed significant differentiation of beta diversity between child and adult microbiomes (Agans et al. 2011, Ringel-Kulka et al. 2013, Hollister et al. 2015). A study of human dizygotic twins found no differentiation in gut microbial beta diversity between male and female infant twins but did find differentiation between male and female pubertal twins (Yatsunenko et al. 2012). Similarly, another study revealed no sex differentiation in gut microbial beta diversity in pre-pubertal children but did show differentiation by sex in pubertal subjects (Yuan et al. 2020). Moreover, beta diversity of male and female microbiomes in late puberty were more similar to adult microbiomes of the same sex than in early puberty (Korpela et al. 2021). Similar patterns occurred in mouse studies: sex differences in alpha and beta diversity in mouse gut microbiomes were not observed before puberty but became evident after puberty (Markle et al. 2013, Yurkovetskiy et al. 2013). While some taxonomic differences between sexes in puberty were noted (Yuan et al. 2020, Korpela et al. 2021), there have been too few studies with small sample sizes to make strong conclusions about pubertal-specific taxonomic differences. Collectively, these studies support the idea that the mammalian gut microbiome differentiates during puberty, resulting in sex differences in adulthood.

Despite a consistent pattern of community-level sex differentiation in the gut microbiome, the specific taxa differentiated by sex varies among studies. For example, early work suggested that the ratio of the common gut phyla, Bacteroidetes and Firmicutes, differed between male and female gut microbiomes (Gomez et al. 2012, Huttenhower et al. 2012). However, follow-up studies found no difference in the Bacteroidetes to Firmicutes ratio, indicating that it is not a reliable indicator of sex differences (Singh & Manning 2016, Elderman et al. 2018, Santos-Marcos et al. 2018, Takagi et al. 2019). The abundances of specific bacterial genera or species have also not been reliable markers of sex differentiation. While individual studies identified bacterial taxa associated with microbiome sex differentiation, the same taxa did not change consistently in all studies. The reasons for the lack of consistency are unclear but may partly be due to external factors influencing the microbial gut composition in the various study populations (e.g. geography, age, and diet) (Kim et al. 2019, Jaggar et al. 2020, Valeri & Endres 2021). This inconsistency may also be a result of functional redundancy in the gut microbiome. So far, most studies of gut microbiome sex differentiation have employed 16S rRNA bacterial gene sequencing which cannot directly analyze gene function (Fig. 1). Future studies applying techniques such as metagenomics, transcriptomics, or metabolomics to study gut microbiome function could be useful in determining the precise nature of gut microbiome sex differentiation.

Sex steroids and the gut microbiome

Sex steroids are produced by the gonads during puberty in response to activation of the hypothalamic–pituitary–gonadal axis (Fig. 2). Observational studies in humans and studies in rodent models collectively suggest a linkage between sex steroids and the gut microbiome. For example, several studies manipulating sex steroid levels in mice indicated that estrogen and testosterone influenced gut microbial diversity and function. Gonadectomy of male and female mice shifted the gut community composition in both sexes and resulted in lower levels of sex differentiation as measured by beta diversity (Harada et al. 2016, Org et al. 2016, Choi et al. 2017, Kaliannan et al. 2018, Gao et al. 2021). Since gonad removal affects more than just the production of sex steroids, gonadectomy plus steroid replacement studies are necessary to discern whether sex steroids are responsible for a certain phenotype. One such study showed that treatment of ovariectomized female mice with estradiol shifted gut microbiome beta diversity to be more like intact females (Kaliannan et al. 2018). Another study demonstrated that treating castrated male mice with dihydrotestosterone shifted the gut microbiome beta diversity to be more like intact males (Gao et al. 2021).

Figure 2
Figure 2

Activation of the hypothalamic–pituitary–gonadal axis during puberty. (1) Gonadotropin-releasing hormone (GnRH) production is stimulated by kisspeptin secreted from neurons in the hypothalamus. (2) GnRH induces pituitary gonadotrope cells to produce and secrete the gonadotropins, follicle-stimulating hormone (FSH), and luteinizing hormone (LH). (3) In males, LH signaling in testicular Leydig cells results in testosterone production, while in females, LH stimulates testosterone production in ovarian theca cells, which is then converted to estrogen in granulosa cells by aromatase. (4A) Testosterone exerts negative feedback in the hypothalamus and pituitary, while (4B) estrogen exerts both negative and positive feedback. (5) Sex steroids may directly or indirectly modulate the gut microbiome to be sexually dimorphic. Created with BioRender.com.

Citation: Reproduction 165, 2; 10.1530/REP-22-0303

In addition to studies with gonadectomized mice, which results in steroid insufficiency, other studies have employed steroid excess to study the relationship between sex steroids and the gut microbiome. For example, studies have shown that treatment of pubertal female mice or rats with the aromatase inhibitor letrozole, which elevates testosterone and decreases estrogen (Kauffman et al. 2015), results in a shift in gut bacterial alpha and beta diversity (Guo et al. 2016, Kelley et al. 2016). Treatment of female rats with dihydrotestosterone (a non-aromatizable androgen) was also found to alter the gut microbiome (Zheng et al. 2021, Rodriguez Paris et al. 2022). Interestingly, letrozole treatment affected the gut microbiome and metabolic phenotype of pubertal females differently than adult females, indicating that puberty may be a sensitive period for effects of steroids on the gut microbiome as well as the host (Arroyo et al. 2019, Torres, Ho et al. 2019, Torres, Skarra et al. 2019). Moreover, cohousing letrozole- and placebo-treated mice, in which mice exchanged gut microbiota through coprophagy, reduced testosterone levels and ameliorated reproductive and metabolic phenotypes in letrozole-treated mice (Torres, Ho et al. 2019, Ho et al. 2021). This potential bidirectional relationship between host sex steroids and the gut microbiome was also suggested by another study which showed that maternal high-fat diet induced metabolic dysregulation, early puberty, and infertility could be partially reversed in female offspring cohoused with healthy females (Wang et al. 2020a ).

In humans, the evidence for a relationship between sex steroids and the gut microbiome comes from various observational studies with postmenopausal women or women using oral contraceptives. Two gut microbiome studies of women using oral contraceptive pills (estrogen and progesterone) found they had lower alpha diversity, differentiated beta diversity, and distinct gut microbial metabolic pathways compared to women not using oral contraceptives (Mihajlovic et al. 2021, Hua et al. 2022). Other studies showed that urinary estrogens and their metabolites were negatively correlated with gut microbiota alpha diversity in postmenopausal females but not premenopausal females (Flores et al. 2012, Fuhrman et al. 2014). In postmenopausal women, estrone levels and the ratio of parent estrogens to estrogen metabolites were associated with an increased relative abundance of specific bacterial taxa (Flores et al. 2012, Fuhrman et al. 2014). A metagenomics study also found significant differences in the taxonomic and functional pathway beta diversity of postmenopausal women compared to premenopausal women (Zhao et al. 2019). Interestingly, the one study we found that investigated changes in the gut microbiome directly within the intestinal tract, namely the duodenum, also showed a relationship between sex steroids and the microbiome. Specifically, Leite et al. reported that the duodenal microbiome of postmenopausal women treated with hormone replacement therapy was more like premenopausal women than postmenopausal women not taking hormone replacement therapy (Leite et al. 2022).

Another link between sex steroids and the gut microbiome was identified in women with PCOS with elevated testosterone levels (hyperandrogenism). Studies of gut microbial diversity found that women and adolescent girls with PCOS had lower alpha diversity and differences in beta diversity compared to healthy women and adolescent girls (Lindheim et al. 2017, Liu et al. 2017, Torres et al. 2018, Qi et al. 2019, Jobira et al. 2020, Garcia-Beltran et al. 2021). Testosterone levels in women with PCOS also correlated with alpha diversity and the abundance of specific members of the gut microbial community (reviewed in (Rizk & Thackray 2021)). Intriguingly, Qi et al. reported that female mice transplanted with stool from women with PCOS developed metabolic and reproductive dysregulation (Qi et al. 2019), suggesting that changes in the gut microbiome may be sufficient to induce PCOS-like phenotypes.

Microbial modifications of sex steroids

Several key studies support the hypothesis that gut bacteria play a critical role in enterohepatic circulation of sex steroids (Fig. 3). Studies in humans and rats demonstrated that antibiotic treatment dramatically increased the ratio of conjugated to deconjugated estrogens excreted in urine and feces (Adlercreutz et al. 1984, Goldin & Gorbach 1984, Flores et al. 2012). Moreover, Colldén et al. found negligible deconjugation of testosterone glucuronide in the intestinal tract of germ-free male mice (Colldén et al. 2019). This study also showed that the concentration of androgen glucuronide in normal mice was high in the small intestine and low in the cecum and colon, indicating that microbial deconjugation of androgen glucuronide (and presumably other steroids) occurs largely in the small intestine.

Figure 3
Figure 3

Enterohepatic circulation of sex steroids and bile acids. (1) Sex steroids (S) are conjugated with glucuronide from uridine 5'-diphosphoglucuronic acid (UDP-GlcA) by liver enzymes. Bile acids (BAs) are synthesized and conjugated with glycine (Gly) or taurine (Tau) in the liver. (2) Sex steroid glucuronides (S-glucuronide) and primary BAs are transported to the gallbladder and excreted into the small intestine via the bile duct. (3) Gut bacteria deconjugate S-glucuronide and transform BAs into secondary BAs or other BA metabolites. (4) S-glucuronides that are not deconjugated are excreted through the feces. (5) Secondary BAs, other BAs metabolites, and deconjugated sex steroids are primarily reabsorbed through the hepatic portal vein to the liver. Alternatively, they are excreted through feces or returned to circulation, to be reabsorbed by the liver or excreted through urine. Created with BioRender.com.

Citation: Reproduction 165, 2; 10.1530/REP-22-0303

While gut bacteria facilitate enterohepatic cycling of sex steroids, the role of specific bacterial species in this process is not well understood. Studies have shown that a bacterial enzyme, β-glucuronidase, is primarily responsible for the deconjugation of steroid glucuronides (Pellock & Redinbo 2017, Sui et al. 2021) and that β-glucuronidase homologs are present and active in many common gut bacteria in mice and humans (Lombardi et al. 1978, Gadelle et al. 1985, Winter & Bokkenheuser 1987, Dabek et al. 2008, Gloux et al. 2011, McIntosh et al. 2012). β-glucuronidase homologs were found in members of many common gut bacterial families, including Ruminococcaea, Lachnospiraceae, Clostridiaceae, Bacteroidaceae, and Tannerellaceae. Many studies have demonstrated Escherichia coli deconjugation of various estrogen-glucuronides (Buehler et al. 1951, Graef et al. 1977, Lombardi et al. 1978, Legler et al. 2002), though studies on androgen glucuronides are lacking. Recently, Ervin et al. reported deconjugation of estrogen glucuronides by β-glucuronidases isolated from other gut bacteria, including Faecalibacteriumprausnitzii, Roseburia species, and Clostridium species (Ervin et al. 2019). As β-glucuronidase genes are not present in all gut microbes, possession of this gene could provide a competitive advantage during puberty. Phylogenetic and structural diversity analyses of bacterial β-glucuronidase genes indicate that microbial β-glucuronidases are substrate-specific and fall into functional classes (Little et al. 2018, Biernat et al. 2019, Ervin et al. 2019). The fact that many common gut bacteria possess β-glucuronidase activity with different substrate specificity provides a potential mechanism by which sex steroids could differentiate the gut microbiome. In support of this idea, glucuronidase genes in gut metagenomes were reported to be higher in premenopausal women than in postmenopausal women and adult men (Peters et al. 2022).

Another mechanism by which sex steroids may impact bacterial communities is via bacterial metabolism of unconjugated (free) steroids. Metabolism of free androgens and estrogens, including redox reactions and partial cleavage of steroids, has been reported in human fecal cultures (Eriksson & Gustafsson 1971, Lombardi et al. 1978). These studies indicated that free steroids were not broken down completely by bacteria, so this activity may be for detoxifying steroids rather than for energy or anabolism. Indeed, free testosterone was shown to be toxic to gut bacteria such as E. faecalis and E. coli, suggesting that high concentrations of steroids may be toxic to some bacteria but not others (Plotkin et al. 2003). On the other hand, additional studies showed that estrogen and progesterone were completely degraded when added to fecal cultures (Li et al. 2018, Coombes et al. 2020). Collectively, these studies provide evidence that gut microbes metabolize sex steroids and that this metabolism could potentially influence the composition of the gut microbial community in a sex-specific manner. However, the mechanisms by which this might occur are poorly understood and many more studies are needed to determine if and how these processes occur.

Bile acids and the gut microbiome

While direct interaction of sex steroids with gut microbiota may lead to a sexually dimorphic gut microbiome, this is not the only possible mechanism. Sex steroids may also indirectly influence the gut microbial community by regulating host production and secretion of bile acids, which may then modulate the gut microbiota. Bile acids are important for emulsifying fats during digestion and as signaling molecules in glucose and lipid metabolism (Perino et al. 2021). In humans, the liver produces cholic acid (CA) and chenodeoxycholic acid (CDCA) from cholesterol (Russell 2003). Muricholic acid (MCA) is produced instead of CDCA in mice. Bile acids are conjugated with either taurine or glycine, then secreted as primary bile acids through the bile duct into the small intestine. Like sex steroids, bile acids circulate between the liver and the intestine through enterohepatic circulation (Fig. 3). Once secreted into the small intestine, microbial bile salt hydrolase (BSH) deconjugates bile acids with glycine or taurine. 70–90% of bile acids are reabsorbed from the ileum and transported to the liver through the hepatic portal vein (Angelin et al. 1982, Wahlström et al. 2016). The BSH gene is present in most major gut microbiome phyla (Firmicutes, Bacteroidetes, Actinobacteria, Fusobacteria, and Proteobacteria), and BSH enzymes in different phyla have different substrate specificity for glycine- or taurine-conjugated bile acids (Jones et al. 2008, Song et al. 2019). The widespread presence of BSH among members of the gut microbiota suggests that it could confer a competitive advantage by providing resistance to bile acid detergent toxicity (Jones et al. 2008). Another transformation of bile acids by gut microbiota is 7α-dehydroxylation, which produces the secondary bile acids deoxycholic acid (DCA) and lithocholic acid (MDCA in mice) (Lucas et al. 2021). 7α-dehydroxylase activity is less widespread among human gut bacteria than BSH activity, which may lead to niche specialization by species with this activity (Lucas et al. 2021). Less common transformations of bile acids include dehydroxylation, epimerization, isomerization, and oxidation (Eyssen et al. 1983, Degirolamo et al. 2014). As secondary bile acids have varying levels of toxicity to different microbes, producing more secondary bile acids may provide a competitive advantage for bacteria able to tolerate this toxicity and a mechanism for the host to influence the gut microbial community (Tian et al. 2020).

Multiple reviews indicate that microbial bile acid metabolism is widespread within the gut microbiota and that it creates a large diversity of bile acid-derived metabolites (Wahlström et al. 2016, Foley et al. 2019, Winston & Theriot 2020). However, apart from BSH activity, the prevalence of these functions within gut microbiota is unknown (Winston & Theriot 2020). While some effects of secondary bile acids on the host are known, such as anti-inflammatory properties, much remains to be discovered about how microbial transformation of bile acids affects host physiology (Ward et al. 2017, Lajczak-McGinley et al. 2020). A growing body of literature indicate that bile acids have a profound effect on host biology. For instance, both primary and secondary bile acids bind to host receptors such as farnesoid X receptor, G protein-coupled bile acid receptor 1, pregnane X receptor, vitamin D receptor, and retinoid-related orphan receptor and regulate metabolic and immune functions (Chiang 2013, Fiorucci et al. 2021). FXR, specifically, has been widely studied and implicated in glucose metabolism, lipid homeostasis, inflammation, and permeability of the intestinal barrier (Ding et al. 2015).

Sex differences and bile acids

In humans, the bile acid profiles of newborn infants (infants less than 48 h old) were largely composed of primary bile acids and were markedly different from adult bile acid profiles (Wang et al. 2020b ). In mice and rats, the gut microbiota and bile acid pool both diversify rapidly after birth. Interestingly, the diversity of secondary bile acids appears to increase until puberty after which time it stabilizes (Morris et al. 1983, van Best et al. 2020). During and after puberty, studies have shown that primary and secondary bile acid levels are higher in female than male rodents (Morris et al. 1983, Turley et al. 1998, Schwarz et al. 2001, Li-Hawkins et al. 2002, Jahnel et al. 2015, Baars et al. 2018). Ma et al. also identified differences in secondary bile acid levels between young and older adult mice and determined that these differences were reduced by cohousing, indicating that the gut microbiome influences bile acid levels (Ma et al. 2020).

There is also evidence for sex differences in bile acids in humans. Multiple studies have determined that total bile acid concentrations and levels of bile acid synthesis are higher in males than females (Bennion et al. 1978, Gälman et al. 2011, Steiner et al. 2012, Xiang et al. 2012, Frommherz et al. 2016). Since mice show the opposite pattern, this difference is important to consider when using mice as a model for the effects of bile acids on physiology. While there are consistent sex differences in the total bile acid pool in humans, the reported sex differences in terms of the types of primary and secondary acids vary among studies (Fisher & Yousef 1973, Bennion et al. 1978, Wang et al. 2020b ). In addition, little is known about how age interacts with sex differences in primary and secondary bile acid metabolism although one study indicated that sex steroids, which change in concentration throughout the lifespan, may indirectly affect secondary bile acid metabolism (Ridlon et al. 2010).

Interplay between bile acids, the gut microbiome, and PCOS

Ho et al. found a relationship between gut microbiota and secondary bile acids in mice. In this study, letrozole treatment of female mice during puberty resulted in changes to secondary bile acid levels, gut microbiota abundances, metabolism, and the reproductive axis (Ho et al. 2021). In humans, women with PCOS were reported to have different levels of specific secondary bile acids compared to healthy controls and higher levels of BSH genes in their gut microbial metagenomes (Qi et al. 2019, Zhang et al. 2019, Yang et al. 2021a ). Transplantation of stool from women with PCOS into adult female mice resulted in impaired fertility and glucose and insulin sensitivity, potentially modulated by the secondary bile acids tauroursodeoxycholic acid and glycoursodeoxycholic acid (Qi et al. 2019). Another study with fecal microbiome transplantation from women with PCOS to adult female mice showed decreased expression of the FXR receptor in the ileum and impaired insulin sensitivity (Yang et al. 2021b ). These studies indicate a potential influence of the gut/liver axis on metabolic dysregulation and vice versa.

The immune system, sex, and the gut microbiome

The immune system differentiates in a sex-specific manner during puberty and remains sexually dimorphic in adulthood (Hooper et al. 2012, Rizzetto et al. 2018, Fuhler 2020). Since both innate and adaptive immunity shapes many of the interactions in the gut microbiome and vice versa (Hooper et al. 2012, Foster et al. 2017, Shi et al. 2017, Wiertsema et al. 2021), the sexually dimorphic nature of the immune system creates another possible mechanism for generating a sex-specific gut microbiome (Whitacre et al. 1999, Lamason et al. 2006, Klein & Flanagan 2016, Sharma et al. 2018). The immune system is known to influence the gut microbiome through a number of mechanisms, such as preventing infiltration of the gut microbiota into host tissues (Gallo & Hooper 2012), secreting mucins and antimicrobial compounds into the intestine, and propagating microbial signals to other immune cells (Abreu 2010, Peterson & Artis 2014). Adaptive immune cells also surveil the gut microbiota and alter community composition through immunoglobulin secretion (Zhang et al. 2015, Fransen et al. 2015). In turn, the gut microbiome affects differentiation of T and B cells and pro- and anti-inflammatory cytokine levels (Fujimura et al. 2016, Sampson et al. 2016, Luu et al. 2018, Zhao & Elson 2018, Li et al. 2019, Lajczak-McGinley et al. 2020).

Given the importance of the immune system in regulating the gut microbiome, sex-specific differences in the immune system could shape the gut microbiome in a sex-specific manner. This idea is supported by studies showing an association between the gut microbiome and autoimmune disorders. Ulcerative colitis, a form of IBD, is diagnosed more frequently in females, while Chron’s disease, another form of IBD, is diagnosed more often in males (Henderson et al. 2012, Hayter & Cook 2012, Herzog et al. 2014). Studies indicate that the gut microbiome is a major factor in IBD incidence and severity. Microbiome and metabolome composition in patients with IBD was different from healthy patients and associated with gut inflammation levels (Clooney et al. 2019, Dong et al. 2019, Franzosa et al. 2019). Gut microbiota may affect severity of IBD directly by activating immune cells through pattern recognition receptors or immunoglobulin binding or indirectly through bile acid signaling (Ding et al. 2015, Schirmer et al. 2019). Additionally, fecal microbiome transplantation is an effective treatment for many patients, though differences in effectiveness by donor and recipient are not well understood (Mańkowska-Wierzbicka et al. 2020, Cheng et al. 2021, Zhou et al. 2021). While differences in gut microbiome composition between patients with ulcerative colitis or Crohn’s disease were reported, sex differences were not observed (Clooney et al. 2019, Dong et al. 2019). However, since sex differences in IBD patients or their microbiomes are often not analyzed or reported, it is possible that sex differentiation of the gut microbiome may contribute to differences in the development and pathology of the IBD subtypes and warrants future investigation. Given that sex-specific immune responses to bacterial metabolites are a candidate for how the gut microbiome contributes to sex differences in the incidence of IBD (Murray et al. 2019, Murray et al. 2020, Bhattarai et al. 2020, Spichak et al. 2021), it may also be important to determine if the sex of the donor and recipient needs to be considered in fecal microbiome transplantation for IBD treatment.

Another notable example of sex-specific differences in the immune system–gut microbiome relationship occurs in the non-obese diabetic (NOD) mouse model for the autoimmune disorder, T1D. In the NOD mouse model, T1D incidence after 30 weeks of age is ~80% in females and 20% in males, although this ratio varies depending on the mouse colony (Makino et al. 1980, Pozzilli et al. 1993, Yurkovetskiy et al. 2013). Intriguingly, castration of NOD males results in a T1D incidence similar to females, indicating that gonadal sex steroids may be important in protecting male mice from T1D (Makino et al. 1981). Additionally, Markle et al. found no sex differences in T1D in NOD mice raised in a germ-free environment, indicating that the gut microbiome was required for the sex differences in T1D incidence (Markle et al. 2013). This and other studies also showed that the male microbiome was protective against T1D and that delivery of male gut microbiota to female mice via fecal transplantation was protective against T1D (Markle et al. 2013, Yurkovetskiy et al. 2013, Markle et al. 2014). Altogether, this evidence suggests that host–microbe interactions may play a key role in modulating sex differences in autoimmunity.

Conclusions and future perspectives

Our review of the current literature on the relationship of sex to the gut microbiome in humans and in rodent models showed that a sex-specific gut microbiome develops during puberty and continues into adulthood. The majority of these studies relied on NGS of bacterial 16S rRNA gene amplicon libraries due to its cost-effectiveness and its ability to taxonomically identify both cultured and uncultured bacteria across all phyla. However, while the 16S gene provides useful information about gut bacterial diversity, it reveals little about the function of gut microbes. Metagenomic, metabolomic, and transcriptomic approaches can inform us about microbial gene content and expression and metabolite production and can identify changes in archaea, fungi, and viruses in addition to bacteria. Future studies employing these approaches will be needed to understand comprehensively how sex and sex steroids regulate the composition and function of gut microbial communities along the alimentary canal.

Understanding the role of gut microbiota in sex-specific human health and disease will require better knowledge of how sex steroids affect the gut microbiome, and vice versa. In humans, there is a lack of clinical studies investigating the relationship between sex steroids and the gut microbiome in men. Given that men commonly experience hypogonadism due to factors such as age, disease (e.g. obesity and type 2 diabetes), or drug treatment, for example, GnRH agonist for prostate cancer (Mittan et al. 2002, Zarotsky et al. 2014), future studies are needed to discern the relationship between sex steroids and the gut microbiome in men. Since gender-affirming care for transgender individuals often involves hormone therapy, study of the gut microbiome in individuals receiving cross sex steroid treatment is also important.

While many studies in humans and rodents have established correlations between sex differences and factors such as sex steroids, bile acids, and the immune system, few studies have investigated causal relationships and mechanisms. More studies are needed to investigate how microbiome manipulation affects sex steroid levels (e.g. via antibiotics, cohousing, or fecal transplants) and how sex steroid manipulation affects the gut microbiome (e.g. via gonadectomy and steroid replacement). Additionally, research should be undertaken to measure the levels of both conjugated and unconjugated steroids in males and females, especially when paired with gut microbiome manipulation. The use of germ-free or antibiotic-depleted mice will be key in establishing which sex-specific diseases require the presence of the gut microbiome and which disease phenotypes can be recapitulated by fecal microbiome transplantation.

Since there is growing evidence that there are sex differences in bile acid production in the liver, future research should investigate when sex differences in primary and secondary bile acid levels emerge in humans and rodents and how the gut microbiome contributes to these differences. Gut microbiota manipulation may provide insight into how the gut microbiome affects sex-specific bile acid metabolism and signaling. Altering host bile acid levels through host genetic knock-outs in the bile acid pathway or through diet supplementation could potentially be used to determine how sex-specific bile acid levels affect gut microbiota composition. Deciphering this relationship may be essential to understand how gut microbiota contributes to sex-specific metabolic disorders like PCOS and type 2 diabetes.

Finally, given the number of sex-specific autoimmune diseases, understanding the relationship between the gut microbiome and the immune system is likely to be important for developing microbial-based preventative measures and therapeutics for these disorders. Studies manipulating the immune system are needed to understand how the immune system shapes the sex-specific gut microbiome. Likewise, more studies are required to provide insight into how gut microbiota manipulation affects development of sex-specific autoimmune diseases like IBD and type I diabetes. Understanding how sex steroids, bile acids, the immune system, and the gut microbiome interact to create and maintain sex differences in microbiome-related diseases may have far-reaching implications especially since the gut microbiome is increasingly being considered a therapeutic target.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

Funding

This work was funded by Grant R01 HD095412 (V G T), F31 HD105403 (L S-H) and a SDSU ARCS Foundation Scholar Award (L S-H)

Author contribution statement

L S-H, S T K and V G T conceived of the topic, created the figures, wrote, and edited the manuscript.

References

  • Abreu MT 2010 Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function. Nature Reviews. Immunology 10 131144. (https://doi.org/10.1038/nri2707)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Acharya KD, Gao X, Bless EP, Chen J & Tetel MJ 2019 Estradiol and high fat diet associate with changes in gut microbiota in female ob/ob mice. Scientific Reports 9 20192. (https://doi.org/10.1038/s41598-019-56723-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Adlercreutz H, Pulkkinen MO, Hämäläinen EK & Korpela JT 1984 Studies on the role of intestinal bacteria in metabolism of synthetic and natural steroid hormones. Journal of Steroid Biochemistry 20 217229. (https://doi.org/10.1016/0022-4731(8490208-5)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Agans R, Rigsbee L, Kenche H, Michail S, Khamis HJ & Paliy O 2011 Distal gut microbiota of adolescent children is different from that of adults. FEMS Microbiology Ecology 77 404412. (https://doi.org/10.1111/j.1574-6941.2011.01120.x)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Angelin B, Björkhem I, Einarsson K & Ewerth S 1982 Hepatic uptake of bile acids in man: fasting and postprandial concentrations of individual bile acids in portal venous and systemic blood serum. Journal of Clinical Investigation 70 724731. (https://doi.org/10.1172/jci110668)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arroyo P, Ho BS, Sau L, Kelley ST & Thackray VG 2019 Letrozole treatment of pubertal female mice results in activational effects on reproduction, metabolism and the gut microbiome. PLoS One 14 e0223274. (https://doi.org/10.1371/journal.pone.0223274)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baars A, Oosting A, Lohuis M, Koehorst M, El Aidy S, Hugenholtz F, Smidt Hauke, Mischke M, Boekschoten MV & Verkade HJ et al.2018 Sex differences in lipid metabolism are affected by presence of the gut microbiota. Scientific Reports 8 1342613426. (https://doi.org/10.1038/s41598-018-31695-w)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, Li Y, Xia Y, Xie H & Zhong H et al.2015 Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host and Microbe 17 690703. (https://doi.org/10.1016/j.chom.2015.04.004)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barlow JT, Bogatyrev SR & Ismagilov RF 2020 A quantitative sequencing framework for absolute abundance measurements of mucosal and lumenal microbial communities. Nature Communications 11 2590. (https://doi.org/10.1038/s41467-020-16224-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bennion LJ, Drobny E, Knowler WC, Ginsberg RL, Garnick MB, Adler RD & Duane WC 1978 Sex differences in the size of bile acid pools. Metabolism: Clinical and Experimental 27 961969. (https://doi.org/10.1016/0026-0495(7890140-3)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bhat SF, Pinney SE, Kennedy KM, McCourt CR, Mundy MA, Surette MG, Sloboda DM & Simmons RA 2021 Exposure to high fructose corn syrup during adolescence in the mouse alters hepatic metabolism and the microbiome in a sex-specific manner. Journal of Physiology 599 14871511. (https://doi.org/10.1113/JP280034)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bhattarai Y, Jie S, Linden DR, Ghatak S, Mars RAT, Williams BB, Pu M, Sonnenburg JL, Fischbach MA & Farrugia G et al.2020 Bacterially Derived Tryptamine increases Mucus Release by activating a host receptor in a mouse model of inflammatory bowel disease. iScience 23 101798. (https://doi.org/10.1016/j.isci.2020.101798)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Biernat KA, Pellock SJ, Bhatt AP, Bivins MM, Walton WG, Tran BN T, Wei L, Snider MC, Cesmat AP & Tripathy A et al.2019 Structure, function, and inhibition of drug reactivating human gut microbial β-glucuronidases. Scientific Reports 9 825. (https://doi.org/10.1038/s41598-018-36069-w)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bokulich NA, Chung J, Battaglia T, Henderson N, Jay M, Li H, Lieber AD, Wu F, Perez-Perez GI & Chen Y et al.2016 Antibiotics, birth mode, and diet shape microbiome maturation during early life. Science Translational Medicine 8 343ra82. (https://doi.org/10.1126/scitranslmed.aad7121)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Borgo F, Garbossa S, Riva A, Severgnini M, Luigiano C, Benetti A, Pontiroli AE, Morace G & Borghi E 2018 Body mass index and sex affect diverse microbial niches within the gut. Frontiers in Microbiology 9 213. (https://doi.org/10.3389/fmicb.2018.00213)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Breitbart M, Haynes M, Kelley S, Angly F, Edwards RA, Felts B, Mahaffy JM, Mueller J, Nulton J & Rayhawk S et al.2008 Viral diversity and dynamics in an infant gut. Research in Microbiology 159 367373. (https://doi.org/10.1016/j.resmic.2008.04.006)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buehler HJ, Katzman PA & Doisy EA 1951 Studies on beta-glucuronidase from E. coli. Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine 76 672676. (https://doi.org/10.3181/00379727-76-18591)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Candon S, Perez-Arroyo A, Marquet C, Valette F, Foray A-P, Pelletier B, Milani C, Ventura M, Bach J-F & Chatenoud L 2015 Antibiotics in early life alter the gut microbiome and increase disease incidence in a spontaneous mouse model of autoimmune insulin-dependent diabetes. PLoS One 10 e0125448. (https://doi.org/10.1371/journal.pone.0125448)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheng F, Huang Z, Wei W & Li Z 2021 Fecal microbiota transplantation for Crohn’s disease: a systematic review and meta-analysis. Techniques in Coloproctology 25 495504. (https://doi.org/10.1007/s10151-020-02395-3)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chiang JY 2013 Bile acid metabolism and signaling. Comprehensive Physiology 3 11911212. (https://doi.org/10.1002/cphy.c120023)

  • Choi S, Hwang YJ, Shin MJ & Yi H 2017 Difference in the gut microbiome between ovariectomy-induced obesity and diet-induced obesity. Journal of Microbiology and Biotechnology 27 22282236. (https://doi.org/10.4014/jmb.1710.10001)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clooney AG, Sutton TDS, Shkoporov AN, Holohan RK, Daly KM, O’Regan O, Ryan FJ, Draper LA, Plevy SE & Ross RP et al.2019 Whole-virome analysis sheds light on viral dark matter in inflammatory bowel disease. Cell Host and Microbe 26 764778.e5. (https://doi.org/10.1016/j.chom.2019.10.009)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Colldén H, Landin A, Wallenius V, Elebring E, Fändriks L, Nilsson ME, Ryberg H, Poutanen M, Sjögren K & Vandenput L et al.2019 The gut microbiota is a major regulator of androgen metabolism in intestinal contents. American Journal of Physiology. Endocrinology and Metabolism 317 E1182E1192. (https://doi.org/10.1152/ajpendo.00338.2019)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Coombes Z, Yadav V, McCoubrey L, Freire C, Basit A, Conlan R & Gonzalez D 2020 Progestogens are metabolized by the Gut Microbiota: implications for colonic drug delivery. Pharmaceutics 12. (https://doi.org/10.3390/pharmaceutics12080760)

    • Search Google Scholar
    • Export Citation
  • Cuesta-Zuluaga J, Kelley ST, Chen Y, Escobar JS, Mueller NT, Ley RE, McDonald D, Huang S, Swafford AD & Knight R et al.2019. Age- and sex-dependent patterns of gut microbial diversity in human adults. mSystems 4 e00261-19. (https://doi.org/10.1128/mSystems.00261-19)

    • Search Google Scholar
    • Export Citation
  • Cui M, Trimigno A, Aru V, Rasmussen MA, Khakimov B & Engelsen SB 2021 Influence of age, sex, and diet on the human fecal metabolome investigated by (1)H NMR spectroscopy. Journal of Proteome Research 20 36423653. (https://doi.org/10.1021/acs.jproteome.1c00220)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dabek M, McCrae SI, Stevens VJ, Duncan SH & Louis P 2008 Distribution of beta-glucosidase and beta-glucuronidase activity and of beta-glucuronidase gene gus in human colonic bacteria. FEMS Microbiology Ecology 66 487495. (https://doi.org/10.1111/j.1574-6941.2008.00520.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Degirolamo C, Rainaldi S, Bovenga F, Murzilli S & Moschetta A 2014 Microbiota modification with probiotics induces hepatic bile acid synthesis via downregulation of the Fxr-Fgf15 axis in mice. Cell Reports 7 1218. (https://doi.org/10.1016/j.celrep.2014.02.032)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Desbonnet L, Clarke G, Traplin A, O’Sullivan O, Crispie F, Moloney RD, Cotter PD, Dinan TG & Cryan JF 2015 Gut microbiota depletion from early adolescence in mice: implications for brain and behaviour. Brain, Behavior, and Immunity 48 165173. (https://doi.org/10.1016/j.bbi.2015.04.004)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Ding L, Yang L, Wang Z & Huang W 2015 Bile acid nuclear receptor FXR and digestive system diseases. Acta Pharmaceutica Sinica. B 5 135144. (https://doi.org/10.1016/j.apsb.2015.01.004)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ding T & Schloss PD 2014 Dynamics and associations of microbial community types across the human body. Nature 509 357360. (https://doi.org/10.1038/nature13178)

  • Dominguez-Bello MG, De Jesus-Laboy KM, Shen N, Cox LM, Amir A, Gonzalez A, Bokulich NA, Song SJ, Hoashi M & Rivera-Vinas JI et al.2016 Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer. Nature Medicine 22 250253. (https://doi.org/10.1038/nm.4039)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dominianni C, Sinha R, Goedert JJ, Pei Z, Yang L, Hayes RB & Ahn J 2015 Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLoS One 10 e0124599–e99. (https://doi.org/10.1371/journal.pone.0124599)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dong LN, Wang M, Guo J & Wang JP 2019 Role of intestinal microbiota and metabolites in inflammatory bowel disease. Chinese Medical Journal 132 16101614. (https://doi.org/10.1097/CM9.0000000000000290)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Elderman M, Hugenholtz F, Belzer C, Boekschoten M, van Beek A, de Haan B, Savelkoul H, de Vos P & Faas M 2018 Sex and strain dependent differences in mucosal immunology and microbiota composition in mice. Biology of Sex Differences 9 26. (https://doi.org/10.1186/s13293-018-0186-6)

    • Search Google Scholar
    • Export Citation
  • Eriksson H & Gustafsson JA 1971 Excretion of steroid hormones in adults. Steroids in faeces from adults. European Journal of Biochemistry 18 146150. (https://doi.org/10.1111/j.1432-1033.1971.tb01225.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ervin SM, Li H, Lim L, Roberts LR, Liang X, Mani S & Redinbo MR 2019 Gut microbial β-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. Journal of Biological Chemistry 294 1858618599. (https://doi.org/10.1074/jbc.RA119.010950)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Eyssen H, De Pauw G, Stragier J & Verhulst A 1983 Cooperative formation of omega-muricholic acid by intestinal microorganisms. Applied and Environmental Microbiology 45 141147. (https://doi.org/10.1128/aem.45.1.141-147.1983)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, Kurilshikov A, Bonder MJ, Valles-Colomer M & Vandeputte D et al.2016 Population-level analysis of gut microbiome variation. Science 352 560564. (https://doi.org/10.1126/science.aad3503)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ferguson A & Sedgwick DM 1994 Juvenile onset inflammatory bowel disease: height and body mass index in adult life. BMJ 308 12591263. (https://doi.org/10.1136/bmj.308.6939.1259)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ferretti P, Pasolli E, Tett A, Asnicar F, Gorfer V, Fedi S, Armanini F, Duy Tin T, Manara S & Zolfo M et al.2018 Mother-to-infant microbial transmission from different body sites shapes the developing infant gut microbiome. Cell Host and Microbe 24 133-45.e5. (https://doi.org/10.1016/j.chom.2018.06.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fiorucci S, Carino A, Baldoni M, Santucci L, Costanzi E, Graziosi L, Distrutti E & Biagioli M 2021 Bile acid signaling in inflammatory bowel diseases. Digestive Diseases and Sciences 66 674693. (https://doi.org/10.1007/s10620-020-06715-3)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fisher MM & Yousef IM 1973 Sex differences in the bile acid composition of human bile: studies in patients with and without gallstones. Canadian Medical Association Journal 109 190193.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, Gajer P, Ravel J & Goedert JJ 2012 Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: a cross-sectional study. Journal of Translational Medicine 10 253. (https://doi.org/10.1186/1479-5876-10-253)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Foley MH, O’Flaherty S, Barrangou R & Theriot CM 2019 Bile salt hydrolases: gatekeepers of bile acid metabolism and host-microbiome crosstalk in the gastrointestinal tract. PLoS Pathogens 15 e1007581. (https://doi.org/10.1371/journal.ppat.1007581)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Foster KR, Schluter J, Coyte KZ & Rakoff-Nahoum S 2017 The evolution of the host microbiome as an ecosystem on a leash. Nature 548 4351. (https://doi.org/10.1038/nature23292)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fransen F, Zagato E, Mazzini E, Fosso B, Manzari C, El Aidy S, Chiavelli A, D’Erchia AM, Sethi MK & Pabst O et al.2015 BALB/c and C57BL/6 mice differ in polyreactive IgA abundance, which impacts the generation of antigen-specific IgA and microbiota diversity. Immunity 43 527540. (https://doi.org/10.1016/j.immuni.2015.08.011)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Franzosa EA, Sirota-Madi A, Avila-Pacheco J, Fornelos N, Haiser HJ, Reinker S, Vatanen T, Hall AB, Mallick H & McIver LJ et al.2019 Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nature Microbiology 4 293305. (https://doi.org/10.1038/s41564-018-0306-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Frommherz L, Bub A, Hummel E, Rist MJ, Roth A, Watzl B & Kulling SE 2016 Age-related changes of plasma bile acid concentrations in healthy adults—results from the cross-sectional KarMeN study. PLoS One 11 e0153959. (https://doi.org/10.1371/journal.pone.0153959)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fuhler GM 2020 The immune system and microbiome in pregnancy. Best Practice and Research. Clinical Gastroenterology 44–45 101671. (https://doi.org/10.1016/j.bpg.2020.101671)

    • Search Google Scholar
    • Export Citation
  • Fuhrman BJ, Feigelson HS, Flores R, Gail MH, Xu X, Ravel J & Goedert JJ 2014 Associations of the fecal microbiome with urinary estrogens and estrogen metabolites in postmenopausal women. Journal of Clinical Endocrinology and Metabolism 99 46324640. (https://doi.org/10.1210/jc.2014-2222)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fujimura KE, Sitarik AR, Havstad S, Lin DL, Levan S, Fadrosh D, Panzer AR, LaMere B, Rackaityte Elze & Lukacs NW et al.2016 Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nature Medicine 22 11871191. (https://doi.org/10.1038/nm.4176)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gadelle D, Raibaud P & Sacquet E 1985 Beta-glucuronidase activities of intestinal bacteria determined both in vitro and in vivo in gnotobiotic rats. Applied and Environmental Microbiology 49 682685. (https://doi.org/10.1128/aem.49.3.682-685.1985)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Gallo RL & Hooper LV 2012 Epithelial antimicrobial defence of the skin and intestine. Nature Reviews. Immunology 12 503516. (https://doi.org/10.1038/nri3228)

  • Gälman C, Angelin B & Rudling M 2011 Pronounced variation in bile acid synthesis in humans is related to gender, hypertriglyceridaemia and circulating levels of fibroblast growth factor 19. Journal of Internal Medicine 270 580588. (https://doi.org/10.1111/j.1365-2796.2011.02466.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gao A, Su J, Liu R, Zhao S, Li W, Xu X, Li D, Shi J, Gu B & Zhang J et al.2021 Sexual dimorphism in glucose metabolism is shaped by androgen-driven gut microbiome. Nature Communications 12 7080. (https://doi.org/10.1038/s41467-021-27187-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gao X, Zhang M, Xue J, Huang J, Zhuang R, Zhou X, Zhang H, Fu Q & Hao Y 2018 Body mass index differences in the gut microbiota are gender specific. Frontiers in Microbiology 9 1250. (https://doi.org/10.3389/fmicb.2018.01250)

    • Search Google Scholar
    • Export Citation
  • Garcia-Beltran C, Malpique R, Carbonetto B, González-Torres P, Henares D, Brotons P, Muñoz-Almagro C, López-Bermejo A, de Zegher F & Ibáñez L 2021 Gut microbiota in adolescent girls with polycystic ovary syndrome: effects of randomized treatments. Pediatric Obesity 16 e12734. (https://doi.org/10.1111/ijpo.12734)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gloux K, Berteau O, El Oumami H, Béguet F, Leclerc M & Doré J 2011 A metagenomic β-glucuronidase uncovers a core adaptive function of the human intestinal microbiome. Proceedings of the National Academy of Sciences of the United States of America Proceedings of the National Academy of Sciences 108(Supplement 1) 45394546. (https://doi.org/10.1073/pnas.1000066107)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Goldin BR & Gorbach SL 1984 Alterations of the intestinal microflora by diet, oral antibiotics, and Lactobacillus: decreased production of free amines from aromatic nitro compounds, azo dyes, and glucuronides. Journal of the National Cancer Institute 73 689695. (https://doi.org/10.1093/jnci/73.3.689)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gomez A, Luckey D, Yeoman CJ, Marietta EV, Margret E, Miller B, Murray JA, White BA & Taneja V 2012 Loss of sex and age driven differences in the gut microbiome characterize arthritis-susceptible *0401 mice but not arthritis-resistant *0402 mice. PLoS One 7 e36095. (https://doi.org/10.1371/journal.pone.0036095)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Graef V, Furuya E & Nishikaze O 1977 Hydrolysis of steroid glucuronides with beta-glucuronidase preparations from bovine liver, Helix pomatia, and E. coli. Clinical Chemistry 23 532535. (https://doi.org/10.1093/clinchem/23.3.532)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guo Y, Qi Y, Yang X, Zhao L, Wen S, Liu Y & Tang L 2016 Association between polycystic ovary syndrome and gut microbiota. PLoS One 11 e0153196. (https://doi.org/10.1371/journal.pone.0153196)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Harada N, Hanaoka R, Horiuchi H, Kitakaze T, Mitani T, Inui H & Yamaji R 2016 Castration influences intestinal microflora and induces abdominal obesity in high-fat diet-fed mice. Scientific Reports 6 23001. (https://doi.org/10.1038/srep23001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hayter SM & Cook MC 2012 Updated assessment of the prevalence, spectrum and case definition of autoimmune disease. Autoimmunity Reviews 11 754765. (https://doi.org/10.1016/j.autrev.2012.02.001)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • He Z, Shao T, Li H, Xie Z & Wen C 2016 Alterations of the gut microbiome in Chinese patients with systemic lupus erythematosus. Gut Pathogens 8 64. (https://doi.org/10.1186/s13099-016-0146-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Heinzel S, Aho VTE, Suenkel U, von Thaler AK, Schulte C, Deuschle C, Paulin L, Hantunen S, Brockmann K & Eschweiler GW et al.2021 Gut microbiome signatures of risk and prodromal markers of Parkinson's disease. Annals of Neurology 90 E1E12. (https://doi.org/10.1002/ana.26128)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Helve O, Korpela K, Kolho K-L, Saisto T, Skogberg K, Dikareva E, Stefanovic V, Salonen A, de Vos WM & Andersson S 2019 2843. Maternal fecal transplantation to infants born by Cesarean section: safety and feasibility.' Open Forum Infectious Diseases 6 S68S68. (https://doi.org/10.1093/ofid/ofz359.148)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Henderson P, Hansen R, Cameron FL, Gerasimidis K, Rogers P, Bisset WM, Reynish EL, Drummond HE, Anderson NH & Van Limbergen J et al.2012 Rising incidence of pediatric inflammatory bowel disease in Scotland. Inflammatory Bowel Diseases 18 9991005. (https://doi.org/10.1002/ibd.21797)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Herzog D, Buehr P, Koller R, Rueger V, Heyland K, Nydegger A, Spalinger J, Schibli S, Braegger CP & Swiss IBD Cohort Study Group 2014 Gender differences in paediatric patients of the swiss inflammatory bowel disease cohort study. Pediatric Gastroenterology, Hepatology and Nutrition 17 147154. (https://doi.org/10.5223/pghn.2014.17.3.147)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Ho B, Ryback D, Benson B, Mason CN, Torres PJ, Quinn RA, Thackray VG, Kelley ST & Metcalf JL 2021 Gut metabolites are more predictive of disease and cohoused states than gut bacterial features in a polycystic ovary syndrome-like mouse model. mSystems 6 e01149-20. (https://doi.org/10.1128/mSystems.01149-20)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hollister EB, Riehle K, Luna RA, Weidler EM, Rubio-Gonzales M, Mistretta T-A, Raza Sabeen, Doddapaneni HV, Metcalf GA & Muzny DM et al.2015 Structure and function of the healthy pre-adolescent pediatric gut microbiome. Microbiome 3 36. (https://doi.org/10.1186/s40168-015-0101-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hooper LV, Littman DR & Macpherson AJ 2012 Interactions between the microbiota and the immune system. Science 336 12681273. (https://doi.org/10.1126/science.1223490)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Hua X, Cao Y, Morgan DM, Miller K, Chin SM, Bellavance D & Khalili H 2022 Longitudinal analysis of the impact of oral contraceptive use on the gut microbiome. Journal of Medical Microbiology 71. (https://doi.org/10.1099/jmm.0.001512)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, Creasy HH, Earl AM, FitzGerald MG & Fulton RS et al.2012 Structure, function and diversity of the healthy human microbiome. Nature 486 207214. (https://doi.org/10.1038/nature11234)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jaggar M, Rea K, Spichak S, Dinan TG & Cryan JF 2020 You’ve got male: sex and the microbiota-gut-brain axis across the lifespan. Frontiers in Neuroendocrinology 56 100815. (https://doi.org/10.1016/j.yfrne.2019.100815)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jahnel J, Zöhrer E, Scharnagl H, Erwa W, Fauler G & Stojakovic T 2015 Reference ranges of serum bile acids in children and adolescents. Clinical Chemistry and Laboratory Medicine 53 18071813. (https://doi.org/10.1515/cclm-2014-1273)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jobira B, Frank DN, Pyle L, Silveira LJ, Kelsey MM, Garcia-Reyes Y, Robertson CE, DIr, Nadeau KJ & Cree-Green M 2020 Obese adolescents with PCOS have altered biodiversity and relative abundance in gastrointestinal microbiota. Journal of Clinical Endocrinology and Metabolism 105 e2134-44. (https://doi.org/10.1210/clinem/dgz263)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jones BV, Begley M, Hill C, Gahan CG & Marchesi JR 2008 Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proceedings of the National Academy of Sciences of the United States of America Proceedings of the National Academy of Sciences 105 1358013585. (https://doi.org/10.1073/pnas.0804437105)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Joriba, Beza, Frank DN, Pyle L, Susan Gross DIR, Pendelton W, Robertson CE, Naedeau KJ & Kelsey MM 2019 183-lb: the gut microbiome and puberty in girls at risk for type 2 diabetes (T2D). Diabetes 68 183-LB. (https://doi.org/10.2337/db19-183-LB)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaliannan K, Robertson RC, Murphy K, Stanton C, Kang C, Wang B, Hao L, Bhan AK & Kang JX 2018 Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. Microbiome 6 205. (https://doi.org/10.1186/s40168-018-0587-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kauffman AS, Thackray VG, Ryan GE, Tolson KP, Glidewell-Kenney CA, Semaan SJ, Poling MC, Iwata N, Breen KM & Duleba AJ et al.2015 A novel letrozole model recapitulates both the reproductive and metabolic phenotypes of polycystic ovary syndrome in female Mice1. Biology of Reproduction 93. (https://doi.org/10.1095/biolreprod.115.131631)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kelley ST, Skarra DV, Rivera AJ & Thackray VG 2016 The gut microbiome is altered in a letrozole-induced mouse model of polycystic ovary syndrome. PLoS One 11 e0146509. (https://doi.org/10.1371/journal.pone.0146509)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Klein SL & Flanagan KL 2016 Sex differences in immune responses. Nature Reviews. Immunology 16 626638. (https://doi.org/10.1038/nri.2016.90)

  • Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, Angenent LT & Ley RE 2011 Succession of microbial consortia in the developing infant gut microbiome. Proceedings of the National Academy of Sciences of the United States of America 108(Supplement 1) 45784585. (https://doi.org/10.1073/pnas.1000081107)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Korpela K, Kallio S, Salonen A, Hero M, Kukkonen AK, Miettinen PJ, Savilahti E, Kohva E, Kariola L & Suutela M et al.2021 Gut microbiota develop towards an adult profile in a sex-specific manner during puberty. Scientific Reports 11 23297. (https://doi.org/10.1038/s41598-021-02375-z)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kurokawa K, Itoh T, Kuwahara T, Oshima K, Toh H, Toyoda A, Takami H, Morita H, Sharma VK & Srivastava TP et al.2007 Comparative metagenomics revealed commonly enriched gene sets in human gut microbiomes. DNA Research 14 169181. (https://doi.org/10.1093/dnares/dsm018)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lajczak-McGinley NK, Porru E, Fallon CM, Smyth J, Curley C, McCarron PA, Tambuwala MM, Roda A & Keely SJ 2020 The secondary bile acids, ursodeoxycholic acid and lithocholic acid, protect against intestinal inflammation by inhibition of epithelial apoptosis. Physiological Reports 8 e14456. (https://doi.org/10.14814/phy2.14456)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lamason R, Zhao P, Rawat R, Davis A, Hall JC, Chae JJ, Agarwal R, Cohen P, Rosen A & Hoffman EP et al.2006 Sexual dimorphism in immune response genes as a function of puberty. BMC Immunology 7 2. (https://doi.org/10.1186/1471-2172-7-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Legler J, Jonas A, Lahr J, Vethaak AD, Brouwer A & Murk AJ 2002 Biological measurement of estrogenic activity in urine and bile conjugates with the in vitro ER-CALUX reporter gene assay. Environmental Toxicology and Chemistry 21 473479. (https://doi.org/10.1002/etc.5620210301)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Leite G, Barlow GM, Parodi G, Pimentel ML, Chang C, Hosseini A, Wang J, Pimentel M & & Mathur R 2022 Duodenal microbiome changes in postmenopausal women: effects of hormone therapy and implications for cardiovascular risk. Menopause 29 264275. (https://doi.org/10.1097/GME.0000000000001917)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Levy R, Magis AT, Earls JC, Manor O, Wilmanski T, Lovejoy J, Gibbons SM, Omenn GS, Hood L & Price ND 2020 Longitudinal analysis reveals transition barriers between dominant ecological states in the gut microbiome. Proceedings of the National Academy of Sciences of the United States of America 117 1383913845. (https://doi.org/10.1073/pnas.1922498117)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li M, Wang B, Zhang M, Rantalainen M, Wang S, Zhou H, Zhang Y, Shen J, Pang X & Zhang M et al.2008 Symbiotic gut microbes modulate human metabolic phenotypes. Proceedings of the National Academy of Sciences of the United States of America 105 21172122. (https://doi.org/10.1073/pnas.0712038105)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li M, Zhao X, Zhang X, Wu D & Leng S 2018 Biodegradation of 17β-estradiol by bacterial co-culture Isolated from Manure. Scientific Reports 8 37873787. (https://doi.org/10.1038/s41598-018-22169-0)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li N, Wang Q, Wang Y, Sun A, Lin Y, Jin Y & Li X 2019 Fecal microbiota transplantation from chronic unpredictable mild stress mice donors affects anxiety-like and depression-like behavior in recipient mice via the gut microbiota-inflammation-brain axis. Stress 22 592602. (https://doi.org/10.1080/10253890.2019.1617267)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Li-Hawkins J, Gåfvels M, Olin M, Lund EG, Andersson U, Schuster G, Björkhem I, Russell DW & Eggertsen G 2002 Cholic acid mediates negative feedback regulation of bile acid synthesis in mice. Journal of Clinical Investigation 110 11911200. (https://doi.org/10.1172/JCI16309)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lindheim L, Bashir M, Münzker J, Trummer C, Zachhuber V, Leber B, Horvath A, Pieber TR, Gorkiewicz G & Stadlbauer V et al.2017 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 12 e0168390. (https://doi.org/10.1371/journal.pone.0168390)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Little MS, Pellock SJ, Walton WG, Tripathy A & Redinbo MR 2018 Structural basis for the regulation of β-glucuronidase expression by human gut Enterobacteriaceae. Proceedings of the National Academy of Sciences of the United States of America 115 E152E161. (https://doi.org/10.1073/pnas.1716241115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu R, Zhang C, Shi Y, Zhang F, Li L, Wang X, Ling Y, Fu H, Dong W & Shen J et al.2017 Dysbiosis of gut microbiota associated with clinical parameters in polycystic ovary syndrome. Frontiers in Microbiology 8 324. (https://doi.org/10.3389/fmicb.2017.00324)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lombardi P, Goldin B, Boutin E & Gorbach SL 1978 Metabolism of androgens and estrogens by human fecal microorganisms. Journal of Steroid Biochemistry 9 795801. (https://doi.org/10.1016/0022-4731(7890203-0)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Lucas LN, Barrett K, Kerby RL, Zhang Q, Cattaneo LE, Stevenson D, Rey FE & Amador-Noguez D 2021 Dominant bacterial phyla from the human gut show widespread ability to transform and conjugate bile acids. mSystems e0080521. (https://doi.org/10.1128/mSystems.00805-21)

    • Search Google Scholar
    • Export Citation
  • Luu M, Weigand K, Wedi F, Breidenbend C, Leister H, Pautz S, Adhikary T & Visekruna A 2018 Regulation of the effector function of CD8+ T cells by gut microbiota-derived metabolite butyrate. Scientific Reports 8 14430. (https://doi.org/10.1038/s41598-018-32860-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ma J, Hong Y, Zheng N, Xie G, Lyu Y, Gu Y, Xi C, Chen L, Wu G & Li Y et al.2020 Gut microbiota remodeling reverses aging-associated inflammation and dysregulation of systemic bile acid homeostasis in mice sex-specifically. Gut Microbes 11 14501474. (https://doi.org/10.1080/19490976.2020.1763770)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Makino S, Kunimoto K, Muraoka Y & Katagiri K 1981 Effect of castration on the appearance of diabetes in NOD mouse. Jikken Dobutsu. Experimental Animals 30 137140. (https://doi.org/10.1538/expanim1978.30.2_137)

    • Search Google Scholar
    • Export Citation
  • Makino S, Kunimoto K, Muraoka Y, Mizushima Y, Katagiri K & Tochino Y 1980 Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu. Experimental Animals 29 113. (https://doi.org/10.1538/expanim1978.29.1_1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mańkowska-Wierzbicka D, Stelmach-Mardas M, Gabryel M, Tomczak H, Skrzypczak-Zielińska M, Zakerska-Banaszak O, Sowińska A, Mahadea D, Baturo A & Wolko Ł et al.2020 The effectiveness of multi-session FMT treatment in active ulcerative colitis patients: a pilot study. Biomedicines 8 268. (https://doi.org/10.3390/biomedicines8080268)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Markle JGM, Frank DN, Adeli K, von Bergen M & Danska JS 2014 Microbiome manipulation modifies sex-specific risk for autoimmunity. Gut Microbes 5 485493. (https://doi.org/10.4161/gmic.29795)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Markle JGM, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, von Bergen M, McCoy KD, Macpherson AJ & Danska JS 2013 Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science 339 10841088. (https://doi.org/10.1126/science.1233521)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mayneris-Perxachs J, Arnoriaga-Rodríguez M, Luque-Córdoba D, Priego-Capote F, Pérez-Brocal V, Moya A, Burokas A, Maldonado R & Fernández-Real JM 2020 Gut microbiota steroid sexual dimorphism and its impact on gonadal steroids: influences of obesity and menopausal status. Microbiome 8 136. (https://doi.org/10.1186/s40168-020-00913-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McIntosh FM, Maison N, Holtrop G, Young P, Stevens VJ, Ince J, Johnstone AM, Lobley GE, Flint HJ & Louis P 2012 Phylogenetic distribution of genes encoding beta-glucuronidase activity in human colonic bacteria and the impact of diet on faecal glycosidase activities. Environmental Microbiology 14 18761887. (https://doi.org/10.1111/j.1462-2920.2012.02711.x)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mihajlovic J, Leutner M, Hausmann B, Kohl G, Schwarz J, Röver H, Stimakovits N, Wolf P, Maruszczak K & Bastian M et al.2021 Combined hormonal contraceptives are associated with minor changes in composition and diversity in gut microbiota of healthy women. Environmental Microbiology 23 30373047. (https://doi.org/10.1111/1462-2920.15517)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mittan D, Lee S, Miller E, Perez RC, Basler JW & Bruder JM 2002 Bone Loss following hypogonadism in men with prostate cancer treated with GnRH Analogs. Journal of Clinical Endocrinology and Metabolism 87 36563661. (https://doi.org/10.1210/jcem.87.8.8782)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Morris AI, Little JM & Lester R 1983 Development of the bile acid pool in rats from neonatal life through puberty to maturity. Digestion 28 216224. (https://doi.org/10.1159/000198991)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mueller S, Saunier K, Hanisch C, Norin E, Alm L, Midtvedt T, Cresci A, Silvi S, Orpianesi C & Verdenelli MC et al.2006 Differences in fecal microbiota in different European study populations in relation to age, gender, and country: a cross-sectional study. Applied and Environmental Microbiology 72 10271033. (https://doi.org/10.1128/AEM.72.2.1027-1033.2006)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Murray E, Sharma R, Smith KB, Mar KD, Barve R, Lukasik M, Pirwani AF, Malette-Guyon E, Lamba S & Thomas BJ et al.2019 Probiotic consumption during puberty mitigates LPS-induced immune responses and protects against stress-induced depression- and anxiety-like behaviors in adulthood in a sex-specific manner. Brain, Behavior, and Immunity 81 198212. (https://doi.org/10.1016/j.bbi.2019.06.016)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Murray E, Smith KB, Stoby KS, Thomas BJ, Swenson MJ, Arber LA, Frenette E & Ismail N 2020 Pubertal probiotic blocks LPS-induced anxiety and the associated neurochemical and microbial outcomes, in a sex dependent manner. Psychoneuroendocrinology 112 104481. (https://doi.org/10.1016/j.psyneuen.2019.104481)

    • Search Google Scholar
    • Export Citation
  • Org E, Mehrabian M, Parks BW, Shipkova P, Liu X, Drake TA & Lusis AJ 2016 Sex differences and hormonal effects on gut microbiota composition in mice. Gut Microbes 7 313322. (https://doi.org/10.1080/19490976.2016.1203502)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pellock SJ & Redinbo MR 2017 Glucuronides in the gut: sugar-driven symbioses between microbe and host. Journal of Biological Chemistry 292 85698576. (https://doi.org/10.1074/jbc.R116.767434)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Perino A, Demagny H, Velazquez-Villegas L & Schoonjans K 2021 Molecular physiology of bile acid signaling in health, disease, and aging. Physiological Reviews 101 683731. (https://doi.org/10.1152/physrev.00049.2019)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Peters BA, Lin J, Qi Q, Usyk M, Isasi CR, Mossavar-Rahmani Y, Derby CA, Santoro N, Perreira KM & Daviglus ML et al.2022 Menopause is associated with an altered gut microbiome and estrobolome, with implications for adverse cardiometabolic risk in the Hispanic community health study/study of latinos. mSystems 7 e0027322. (https://doi.org/10.1128/msystems.00273-22)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Peterson LW & Artis D 2014 Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nature Reviews. Immunology 14 141153. (https://doi.org/10.1038/nri3608)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Plotkin BJ, Roose RJ, Erikson Q & Viselli SM 2003 Effect of androgens and glucocorticoids on microbial growth and antimicrobial susceptibility. Current Microbiology 47 514520. (https://doi.org/10.1007/s00284-003-4080-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pozzilli P, Signore A, Williams AJ & Beales PE 1993 NOD mouse colonies around the world--recent facts and figures. Immunology Today 14 193196. (https://doi.org/10.1016/0167-5699(9390160-M)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Qi X, Yun C, Sun L, Xia J, Wu Q, Wang Y, Wang L, Zhang Y, Liang X & Wang L et al.2019 Gut microbiota-bile acid-interleukin-22 axis orchestrates polycystic ovary syndrome. Nature Medicine 25 12251233. (https://doi.org/10.1038/s41591-019-0509-0)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ridlon JM, Kang D-J & Hylemon PB 2010 Isolation and characterization of a bile acid inducible 7α-dehydroxylating operon in Clostridium hylemonae TN271. Anaerobe 16 137146. (https://doi.org/10.1016/j.anaerobe.2009.05.004)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Ringel-Kulka T, Cheng J, Ringel Y, Salojärvi J, Carroll I, Palva A, de Vos WM & Satokari R 2013 Intestinal microbiota in healthy U.S. Young children and adults—A high throughput microarray analysis. PLoS One 8 e64315. (https://doi.org/10.1371/journal.pone.0064315)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rizk MG & Thackray VG 2021 Intersection of polycystic ovary syndrome and the gut microbiome. Journal of the Endocrine Society 5 bvaa177. (https://doi.org/10.1210/jendso/bvaa177)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rizzetto L, Fava F, Tuohy KM & Selmi C 2018 Connecting the immune system, systemic chronic inflammation and the gut microbiome: the role of sex. Journal of Autoimmunity 92 1234. (https://doi.org/10.1016/j.jaut.2018.05.008)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rodriguez Paris V, Wong XYD, Solon-Biet SM, Edwards MC, Aflatounian A, Gilchrist RB, Simpson SJ, Handelsman DJ, Kaakoush NO & Walters KA 2022 The interplay between PCOS pathology and diet on gut microbiota in a mouse model. Gut Microbes 14 2085961. (https://doi.org/10.1080/19490976.2022.2085961)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Russell DW 2003 The enzymes, regulation, and genetics of bile acid synthesis. Annual Review of Biochemistry 72 137174. (https://doi.org/10.1146/annurev.biochem.72.121801.161712)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, Challis C, Schretter CE, Rocha S & Gradinaru V et al.2016 Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease. Cell 167 14691480.e12. (https://doi.org/10.1016/j.cell.2016.11.018)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Santos-Marcos JA, Rangel-Zuñiga OA, Jimenez-Lucena R, Quintana-Navarro GM, Garcia-Carpintero S, Malagon MM, Landa BB, Tena-Sempere M, Perez-Martinez P & Lopez-Miranda J et al.2018 Influence of gender and menopausal status on gut microbiota. Maturitas 116 4353. (https://doi.org/10.1016/j.maturitas.2018.07.008)

    • Crossref
    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schirmer M, Garner A, Vlamakis H & Xavier RJ 2019 Microbial genes and pathways in inflammatory bowel disease. Nature Reviews. Microbiology 17 497511. (https://doi.org/10.1038/s41579-019-0213-6)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Schwarz M, Russell DW, Dietschy JM & Turley SD 2001 Alternate pathways of bile acid synthesis in the cholesterol 7alpha-hydroxylase knockout mouse are not upregulated by either cholesterol or cholestyramine feeding. Journal of Lipid Research 42 15941603. (https://doi.org/10.1016/S0022-2275(2032213-6)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Sharma R, Rooke J, Kolmogorova D, Melanson B, Mallet J-F, Matar C, Schwarz J & Ismail N 2018 Sex differences in the peripheral and central immune responses following lipopolysaccharide treatment in pubertal and adult CD-1 mice. International Journal of Developmental Neuroscience 71 94104. (https://doi.org/10.1016/j.ijdevneu.2018.07.012)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Sheng L, Jena PK, Liu H-X, Kalanetra KM, Gonzalez FJ, French SW, Krishnan VV, Mills DA & Wan YY 2017 Gender differences in bile acids and microbiota in relationship with gender dissimilarity in steatosis induced by diet and FXR inactivation. Scientific Reports 7 17481748. (https://doi.org/10.1038/s41598-017-01576-9)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Shepherd R, Cheung AS, Pang K, Saffery R & Novakovic B 2020 Sexual dimorphism in innate immunity: the role of sex hormones and epigenetics. Frontiers in Immunology 11 604000. (https://doi.org/10.3389/fimmu.2020.604000)

    • Search Google Scholar
    • Export Citation
  • Shi N, Li N, Duan X & Niu H 2017 Interaction between the gut microbiome and mucosal immune system. Military Medical Research 4 14. (https://doi.org/10.1186/s40779-017-0122-9)

    • Search Google Scholar
    • Export Citation
  • Singh P & Manning SD 2016 Impact of age and sex on the composition and abundance of the intestinal microbiota in individuals with and without enteric infections. Annals of Epidemiology 26 380385. (https://doi.org/10.1016/j.annepidem.2016.03.007)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Sinha T, Vila AV, Garmaeva S, Jankipersadsing SA, Imhann F, Collij V, Bonder MJ, Jiang X, Gurry T & Alm EJ et al.2019 Analysis of 1135 gut metagenomes identifies sex-specific resistome profiles. Gut Microbes 10 358366. (https://doi.org/10.1080/19490976.2018.1528822)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Song SJ, Wang J, Martino C, Jiang Lingjing, Thompson WK, Shenhav L, McDonald D, Marotz C, Harris PR & Hernandez CD et al.2021 Naturalization of the microbiota developmental trajectory of Cesarean-born neonates after vaginal seeding. Med 2 951964.e5. (https://doi.org/10.1016/j.medj.2021.05.003)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Song Z, Cai Y, Lao Xingzhen, Wang X, Lin X, Cui Y, Kalavagunta PK, Liao J, Jin L & Shang J et al.2019 Taxonomic profiling and populational patterns of bacterial bile salt hydrolase (BSH) genes based on worldwide human gut microbiome. Microbiome 7 9. (https://doi.org/10.1186/s40168-019-0628-3)

    • Search Google Scholar
    • Export Citation
  • Spichak S, Donoso F, Moloney GM, Gunnigle E, Brown JM, Codagnone M, Dinan TG & Cryan JF 2021 Microbially-derived short-chain fatty acids impact astrocyte gene expression in a sex-specific manner', Brain, Behavior, & Immunity. Health 16 100318. (https://doi.org/10.1016/j.bbih.2021.100318)

    • Search Google Scholar
    • Export Citation
  • Steiner C, Holleboom AG, Karuna R, Motazacker MM, Kuivenhoven JA, Frikke-Schmidt R, Tybjaerg-Hansen A, Rohrer L, Rentsch KM & Eckardstein Av 2012 Lipoprotein distribution and serum concentrations of 7α-hydroxy-4-cholesten-3-one and bile acids: effects of monogenic disturbances in high-density lipoprotein metabolism. Clinical Science 122 385396. (https://doi.org/10.1042/CS20110482)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Sui Y, Wu J & Chen J 2021 The role of gut microbial β-glucuronidase in estrogen reactivation and breast cancer. Frontiers in Cell and Developmental Biology 9. (https://doi.org/10.3389/fcell.2021.631552)

    • Search Google Scholar
    • Export Citation
  • Svensson A, Brunkwall L, Roth B, Orho-Melander M & Ohlsson B 2021 Associations between endometriosis and gut microbiota. Reproductive Sciences 28 23672377. (https://doi.org/10.1007/s43032-021-00506-5)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Takagi T, Naito Y, Inoue R, Kashiwagi S, Uchiyama K, Mizushima K, Tsuchiya S, Dohi O, Yoshida N & Kamada K et al.2019 Differences in gut microbiota associated with age, sex, and stool consistency in healthy Japanese subjects. Journal of Gastroenterology 54 5363. (https://doi.org/10.1007/s00535-018-1488-5)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Kim YS, Unno T, Kim B-Y & Park M-S 2019 Sex differences in gut microbiota. Wjmh 38 4860.

  • Tian Y, Gui W, Koo Imhoi, Smith PB, Allman EL, Nichols RG, Rimal B, Cai J, Liu Q & Patterson AD 2020 The microbiome modulating activity of bile acids. Gut Microbes 11 979996. (https://doi.org/10.1080/19490976.2020.1732268)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Torres PJ, Ho BS, Arroyo P, Sau L, Chen A, Kelley ST & Thackray VG 2019 Exposure to a healthy gut microbiome protects against reproductive and metabolic dysregulation in a PCOS mouse model. Endocrinology 160 11931204. (https://doi.org/10.1210/en.2019-00050)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Torres PJ, Siakowska M, Banaszewska B, Pawelczyk L, Duleba AJ, Kelley ST & Thackray VG 2018 Gut microbial diversity in women with polycystic ovary syndrome correlates with hyperandrogenism. Journal of Clinical Endocrinology and Metabolism 103 15021511. (https://doi.org/10.1210/jc.2017-02153)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Torres PJ, Skarra DV, Ho BS, Sau L, Anvar AR, Kelley ST & Thackray VG 2019 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 Microbiology 19 5757. (https://doi.org/10.1186/s12866-019-1425-7)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Turley SD, Schwarz M, Spady DK & Dietschy JM 1998 Gender-related differences in bile acid and sterol metabolism in outbred CD-1 mice fed low- and high-cholesterol diets. Hepatology 28 10881094. (https://doi.org/10.1002/hep.510280425)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Valeri F & Endres K 2021 How biological sex of the host shapes its gut microbiota. Frontiers in Neuroendocrinology 61 100912. (https://doi.org/10.1016/j.yfrne.2021.100912)

    • Search Google Scholar
    • Export Citation
  • van Best N, Rolle-Kampczyk U, Schaap FG, Basic M, Olde Damink SWM, Bleich A, Savelkoul PHM, von Bergen M, Penders J & Hornef MW 2020 Bile acids drive the newborn's gut microbiota maturation. Nature Communications 11 3692. (https://doi.org/10.1038/s41467-020-17183-8)

    • Search Google Scholar
    • Export Citation
  • van Hooff MH, Voorhorst FJ, Kaptein MB, Hirasing RA, Koppenaal C & Schoemaker J 1999 Endocrine features of polycystic ovary syndrome in a random population sample of 14–16 year old adolescents. Human Reproduction 14 22232229. (https://doi.org/10.1093/humrep/14.9.2223)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • van Keulen BJ, Dolan CV, van der Voorn B, Andrew R, Walker BR, Hulshoff Pol H, Boomsma DI, Rotteveel J & Finken MJJ 2020 Sexual dimorphism in cortisol metabolism throughout pubertal development: a longitudinal study. Endocrine Connections 9 542551. (https://doi.org/10.1530/EC-20-0123)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Vatanen T, Kostic AD, d’Hennezel E, Siljander H, Franzosa EA, Yassour M, Kolde R, Vlamakis H, Arthur TD & Hämäläinen A-M et al.2016 Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell 165 842853. (https://doi.org/10.1016/j.cell.2016.04.007)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Wahlström A, Sayin SI, Marschall HU & Bäckhed F 2016 Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metabolism 24 4150. (https://doi.org/10.1016/j.cmet.2016.05.005)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Wang M, Zhang Youjie, Miller D, Rehman NO, Cheng X, Yeo J-Y, Joe B & Hill JW 2020a Microbial reconstitution reverses early female puberty induced by maternal high-fat diet during lactation. Endocrinology 161. (https://doi.org/10.1210/endocr/bqz041)

    • Search Google Scholar
    • Export Citation
  • Wang WX, Wen-Xia LC, Wang GY, Zhang J-L, Tan XW, Lin QH, Chen Y-J, Zhang J, Zhu P-P & Miao J et al.2020b Urinary bile acid profile of newborns born by Cesarean section is characterized by oxidative metabolism of primary bile acids: limited roles of fetal-specific CYP3A7 in cholate oxidations. Drug Metabolism and Disposition: The Biological Fate of Chemicals 48 662672. (https://doi.org/10.1124/dmd.120.000011)

    • Crossref
    • Search Google Scholar
    • Export Citation
  • Ward JBJ, Lajczak NK, Kelly OB, O’Dwyer AM, Giddam AK, Ní Gabhann J, Franco P, Tambuwala MM, Jefferies CA & Keely S et al.2017 Ursodeoxycholic acid and lithocholic acid exert anti-inflammatory actions in the colon. American Journal of Physiology-Gastrointestinal and Liver Physiology 312 G550G558. (https://doi.org/10.1152/ajpgi.00256.2016)

    • Crossref