Next Article in Journal
Angiotensin II-Induced Signal Transduction Mechanisms for Cardiac Hypertrophy
Next Article in Special Issue
Impact of Chronic Multi-Generational Exposure to an Environmentally Relevant Atrazine Concentration on Testicular Development and Function in Mice
Previous Article in Journal
FIP200 Methylation by SETD2 Prevents Trim21-Induced Degradation and Preserves Autophagy Initiation
Previous Article in Special Issue
Bisphenol Exposure Disrupts Cytoskeletal Organization and Development of Pre-Implantation Embryos
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Endocrine-Disrupting Chemicals, Gut Microbiota, and Human (In)Fertility—It Is Time to Consider the Triad

1
B-Woman, Via dei Monti Parioli 6, 00197 Rome, Italy
2
Clinica Valle Giulia, GeneraLife IVF, Via De Notaris 2B, 00197 Rome, Italy
3
Laboratory of Developmental Biology, Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy
4
Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
5
Department of Medicine and Aging Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
6
Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
7
Centre for Health Technologies (CHT), University of Pavia, Via Ferrata 5, 27100 Pavia, Italy
8
Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, Via Sant’Andrea 34, 61029 Urbino, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 1 September 2022 / Revised: 12 October 2022 / Accepted: 17 October 2022 / Published: 22 October 2022

Abstract

:
The gut microbiota (GM) is a complex and dynamic population of microorganisms living in the human gastrointestinal tract that play an important role in human health and diseases. Recent evidence suggests a strong direct or indirect correlation between GM and both male and female fertility: on the one hand, GM is involved in the regulation of sex hormone levels and in the preservation of the blood–testis barrier integrity; on the other hand, a dysbiotic GM is linked to the onset of pro-inflammatory conditions such as endometriosis or PCOS, which are often associated with infertility. Exposure to endocrine-disrupting chemicals (EDCs) is one of the main causes of GM dysbiosis, with important consequences to the host health and potential transgenerational effects. This perspective article aims to show that the negative effects of EDCs on reproduction are in part due to a dysbiotic GM. We will highlight (i) the link between GM and male and female fertility; (ii) the mechanisms of interaction between EDCs and GM; and (iii) the importance of the maternal–fetal GM axis for offspring growth and development.

1. Introduction

In this perspective, we will describe mounting evidence that suggests a damaging effect of endocrine-disrupting chemicals (EDCs) on the gut microbiota (GM), and we will bring experimental, clinical, and epidemiological evidence that shows how dysregulation of the GM eubiotic condition might be the linking ring between EDCs and male and female infertility. Whilst this dysregulation of EDCs on the GM is evident during adult life, we will suggest that it might occur also during the early post-natal phases of life and fetal development, speculating, for the latter, a possible transgenerational effect.

2. Gut Microbiota: A New Player in Town

The GM is a complex and dynamic population of microorganisms living in the human gastrointestinal tract that exerts biochemical functions otherwise absent in the host. GM is considered a hidden metabolic organ of the human body influencing human health and diseases [1]. Particularly, GM regulates a range of physiological functions [2] mainly involved in (i) preserving the intestinal barrier integrity [3], (ii) protecting against pathogens [4], (iii) regulating host immunity [5,6], (iv) ensuring energy metabolism [7], and (v) modulating immune development [8].
The main GM phyla are Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, Fusobacteria, and Verrucomicrobia, with the former two representing 90% of the whole population [9]. The biodiversity of GM is of utmost importance since it serves as a functional expansion of host genomes. Indeed, the GM’s genome, named gut microbiome, harbors different extra genes encoding enzymatic proteins, non-encoded by the host, that contribute to the regulation of the host physiology [10].

The Key Role of Gut Microbiota in Health and Disease

The GM is a potential controller of wellness and disease. As demonstrated by experimental, clinical, and epidemiological evidence, a dysbiotic microbiota (i.e., a microbiota that deviates from the “eubiotic” status in terms of diversity and functionality [11]) is implicated in a range of diseases in adults, including inflammatory bowel disease [12,13], arthritis [14], cancer [15], neurological and neuropsychiatric disorders [16], cardiometabolic [17] and cardiovascular [18] disease, obesity [19], type 2 diabetes [20,21,22], and, as detailed below, infertility.
Gut eubiosis is pivotal also for the maintenance of the intestinal barrier integrity, essential to prevent the so-called leaky-gut syndrome (LGS), a condition that causes intestinal permeability resulting in the permeation of antigens, endotoxins, and pathogens and the altered production of neurotransmitters and metabolites such as short-chain fatty acids, leading to chronic low-grade inflammation, a state linked to the development of several diseases [23].
Furthermore, increasing evidence suggests that the acquisition and development of a healthy microbiota in the infant are pivotal to exerting long-lasting beneficial effects in disease prevention [24]. The maternal microbial reservoir is crucial in the maternal-to-infant passage. Maternal vaginal, oral, gut, skin, and breast milk microbial communities contribute to establishing the infant’s own gut microbial community [25,26,27,28], thus regulating correct fetal growth, neurodevelopment, and immune programming [29,30] and providing a prophylactic potential of non-communicable diseases (NCDs), such as obesity, immunoinflammatory disorders, and neurocognitive complications [31].
Extensive microbial colonization takes place post-partum [24] through the mode of delivery, contact with the mother (such as skin-to-skin care), maternal diet, and breastfeeding [32]; however, gut colonization might occur already during the prenatal period [33]. Recent, although controversial [30], findings question the dogma that the womb is sterile, suggesting that the fetus incorporates an initial microbial inoculum already before birth (in utero colonization hypothesis) [29,30,34,35,36], followed by postnatally supplemented maternal microbes.

3. The Dangerous Effects of Endocrine-Disrupting Chemicals on the Gut Microbiota

Several factors can affect GM composition and homeostasis, and EDCs are among the most critical [37]. EDCs are highly heterogeneous environmental contaminants that include both natural and synthetic molecules [38]. Phytoestrogens (e.g., genistein and coumestrol) are natural EDCs found in several human and animal food, whereas solvents/lubricants (e.g., polychlorinated biphenyls and dioxins), plastics (e.g., bisphenol A), plasticizers (e.g., phthalates), pesticides and fungicides (e.g., methoxychlor, dichlorodiphenyltrichloroethane, and vinclozolin), and pharmaceutical agents (e.g., diethylstilbestrol) are synthetic EDCs applied to anthropic activities [39]. EDCs enter the organism through the food chain, resulting in adverse interference with hormonally controlled functions [38]. Indeed, they exert their toxicity mimicking estrogen and/or androgen hormone actions, binding to their specific endogenous receptors. They promote impaired activation, synthesis, and secretion of endogenous hormones, thus influencing several hormonal and metabolic processes [38,40]. Moreover, some EDCs show genotoxic effects [41,42] and perturb the epigenetic landscape, inducing alterations of target cells [43,44]. A number of clinical and experimental studies have shown the impact of EDCs on human GM [45]. For instance, increased urinary lead (Pb) was associated with significant changes in the human adult gut microbiota biodiversity [46], affecting both the richness of microbial taxa (α-diversity) and the variability in taxa composition (β-diversity) [47].
Post-natal exposure to di-(2-ethylhexyl) phthalate (DEHP) altered GM composition in new-born babies, with levels of Bifidobacterium longum, a key microbial species for normal gut colonization and development [48], significantly decreased compared to control [49]. Moreover, using an in vitro simulator of the human intestinal microbial ecosystem (SHIME), Wang and colleagues demonstrated that BPA exposure significantly changed the variability of the microbial community and increased the percentage of microbes shared in ascending, transverse, and descending colons, observing an upregulated expression of genes related to estrogenic effect and oxidative stress [50].
Many other experimental studies extensively reviewed by Galvez-Ontiveros and colleagues [45] have been conducted in model animals such as rodents, zebrafish, rabbits, and dogs, showing that exposure to polychlorinated biphenyls, parabens, phytoestrogens, metals, triclosan and triclocarban, phthalates, and BPA and its analogs affects GM composition and functionality and triggers metabolic disease.

Endocrine-Disrupting Chemicals and Gut Microbiota: Mechanisms of Interaction

Gastrointestinal microbiota and EDC interactions are multiple and interdependent. On the one hand, environmental contaminants alter gastrointestinal bacteria composition and/or the metabolic activity that shapes the host’s microbiotype; on the other hand, GM extensively metabolizes environmental chemicals, thus modulating their toxicity in the host [51]. Indeed, the microbiota is pictured as an additional organ involved in xenobiotic biotransformation [52] and influencing the pharmacokinetics of environmental chemicals [53]. Therefore, an altered symbiotic flora may differently modulate chemical toxicity [11,45].
There are three main mechanisms by which EDCs and GM interact [51]:
(1)
Through the direct and indirect metabolism of xenobiotics, i.e., chemical substances that are not produced by the organism. More specifically, xenobiotics enter the human body mainly through the gastrointestinal tract and reach the distal gut where they can be directly metabolized by GM performing diverse chemical transformations such as hydrolysis, removal of the succinate group, dehydroxylation, acetylation, deacetylation, proteolysis, denitration, deconjugation, or thiazole ring opening. In other circumstances, after ingestion, xenobiotics such as the non-polar ones are transported to the liver for detoxification where they are oxidized and subsequently eliminated in urine or secreted into the bile. In the latter case, they move to the small intestine where they can be absorbed, or they progress down to the large intestine where they are metabolized by the GM.
(2)
By altering the microbial diversity and thus inducing dysbiosis. For instance, it has been demonstrated that both BPA and ethinylestradiol exposure can elevate the amount of Bifidobacterium spp. in mice, leading to metabolic disorders [54]; instead, methylparaben, diethyl phthalate, and triclosan (or their mixture) exposure modifies, in rats, the ratio between Bacteroidetes and Firmicutes spp. [55], a relevant marker of gut dysbiosis [56]. Moreover, EDCs reduce the number of microbial species such as Lactobacillus spp., important for xenobiotic biotransformation and involved in the maintenance of a proper intestinal barrier [57], thus resulting in the enhanced absorption of contaminants and toxicity in the host.
(3)
EDCs interfere with the GM enzymatic activity. Indeed, GM significantly contributes to the host metabolism by providing enzymes encoded by the gut microbiome (i.e., the genome of GM) which are involved in both the xenobiotic and endobiotic metabolism. Among the most important of these enzymes, β-glycosidase catalyzes the hydrolysis of plant polyphenol glycosides, and β-glucuronidase (GUSB) catalyzes the removal of glucuronic acid from liver-produced glucuronides [58]. Consequently, EDCs, by perturbating GM, may alter host physiological processes mediated by these enzymes.

4. Gut Microbiota and (In)Fertility

Emerging evidence indicates that GM composition is key also in reproductive health for both women and men.

4.1. Female Fertility

Variations to the GM homeostasis may affect female fertility via different pathways. Primarily, GM is able to influence female fertility by affecting the level of sex hormones [59]. Indeed, as described before, the gut microbiome encodes different enzymes involved in host metabolism, and one of them, the enzyme GUSB, is responsible for the metabolism and modulation of circulating estrogen hormones [60] since it deconjugates estrogens, enabling their binding to estrogen receptors and leading to physiological downstream effects [61,62]. Therefore, changes to the microbial population encoding the enzyme GUSB, known as the estrobolome, affect the endogenous estrogen metabolism by modulating the enterohepatic circulation of these hormones, with a subsequent impact on the woman’s hormonal balance and, therefore, on her fertility [59].
Secondly, GM seems inextricably linked to female infertility due to the important relationship that exists between a healthy GM and the immune system [63]. Indeed, a dysbiotic GM is observed in several infertility-related disorders such as endometriosis [64,65], polycystic ovary syndrome (PCOS), insulin resistance (IR) [66,67,68,69,70,71], and obesity [72,73], characterized by an unbalanced immune profile and pro-inflammatory status, known to negatively affect fertility [74]. All these conditions are characterized by a reduced GM biodiversity and specific microbial imbalances in both the gut and reproductive tract leading to immune dysfunction, compromised immunosurveillance, and altered immune cell profiles. For instance, in women affected by endometriosis, diminished Lactobacillus spp. dominance, an altered Firmicutes:Bacteroidetes ratio, and an abundance of vaginosis-related bacteria and other opportunistic pathogens [64,65] determine upregulated ovarian estrogen secretion through neuro-active metabolites that stimulate GnRH neurons, in turn worsening hormonal homeostasis [64]. A further example is that of PCOS patients, in which the GM shows an abnormal Escherichia:Shigella ratio and an excess of Bacteroides compared to healthy women [71], a condition occurring also in IR and in obese women characterized by an increased Firmicutes:Bacteroidetes ratio [56,72]. Moreover, GM exerts a role in the pathogenesis of thyroid autoimmune disease, a frequent disorder in infertility patients [75,76,77], and a relationship between the gut microbiome and premature ovarian insufficiency (POI) has been suggested [78,79].
Lastly, GM eubiosis plays a key role in female fertility since it has been demonstrated that GM can influence the whole genital tract microbiota through a continuous crosstalk between uterus and vagina ecosystems [80]. Therefore, a condition of dysbiosis in the gut could possibly lead to vaginal and uterine dysbiosis, negatively affecting endometrial receptivity at the time of implantation [81,82].
Noteworthy, oral administration of probiotics influences vaginal microbiota composition and immunity [23], and different microbial species, such as the Gram-positive Lactobacillus spp. that dominates the vaginal microbiota in physiological conditions, originate from the gut [83]. Moreover, GM dysbiosis can induce the leaky-gut syndrome leading to intestinal permeability and leakage of bacteria and bacterial products from the gut into the circulation, thus affecting the female genital tract microbiota [80].

4.2. Male Fertility

Growing interest has been devoted also to the role of the microbiome in male reproduction. First, differences in GM between genders have been demonstrated, with a considerably less variety of gut microbiota in men as compared to women, with sex hormones likely responsible for these differences [84,85]. Although the underlying mechanisms by which GM contributes to the regulation of androgens and steroids are still unclear, it is known that some gut microbes can express steroid-processing enzymes and produce steroid hormones with an impact on the metabolism [86,87], and in turn, sex steroids themselves may regulate the structure and function of the GM [87,88].
The GM communicates with the distal organs of the host, including the testis, through various mechanisms, and it may affect male reproduction at several levels.
It has been suggested that a defective gut barrier function can end with the dissemination within the blood flow of microbial-associated molecular patterns (MAMPs) such as lipopolysaccharide, lipoprotein acids, peptidoglycans, and lipoproteins into the testis and epididymis. Indeed, immune system activation, induced by GM translocation, leads to testicular and epididymal inflammation and, also, triggers insulin resistance together with gastrointestinal hormones which, in turn, affect the secretion of sex hormones [luteinizing hormone (LH), follicle-stimulating hormone (FSH), and testosterone (T)] and their role in the regulation of spermatogenesis [89].
This condition determines: (i) the activation of specialized receptors known as pattern recognition receptors which serve as MAMP sensors that recognize the essential microbial components and trigger an immune response; (ii) Langerhans islet inflammation; and (iii) gastrointestinal hormonal changes with insulin resistance and the alteration of LH, FSH, and free T levels [89].
Moreover, a novel role for the GM in the regulation of testicular development and function has been outlined, demonstrating the involvement of the GM in the regulation of the endocrine function of the testis and the integrity of the blood–testis barrier (BTB) [90].
Noteworthy, in addition to the GM, the testicular microenvironment is not completely sterile, containing low amounts of Actinobacteria, Bacteroidetes, Firmicutes, and Proteobacteria. Although the role of testicular bacterial species in the testis remains to be elucidated, it has been suggested that these microbes play an important role in regulating and shaping the immune response in the testis. The gut microbes and testicular microbes together can influence human reproduction, as demonstrated recently in the study of Zhang et al. [91], in which GM altered bile acid levels and affected vitamin A absorption in the gut. Thus, the abnormal vitamin A metabolism can be transferred to the testis through the blood circulation, ultimately resulting in a sharp decline in spermatogonia differentiation in the metabolic syndrome model. Alfano et al. [92] showed a dysbiotic bacterial community in idiopathic nonobstructive azoospermia (iNOA) patients, describing a decrease in Clostridium abundance in those patients with unsuccessful sperm retrieval compared to successful sperm-retrieval patients. This finding has potential clinical relevance since the presence of the class Clostridia has been linked with improved sperm motility and morphology [93,94].
In addition, these authors suggested a link between gut microbes and testicular microbes since testicular bacterial microbiome modifications observed in iNOA men were similar to those previously reported in the gut of elderly individuals [92].
Altered intestinal flora can contribute to increased serum trimethylamine-N-oxide levels, promote vascular inflammation leading to cavernous endothelial and smooth muscle cell damage and to the development of erectile dysfunction [95].
EDCs can cause dysbiosis of the GM with consequent alteration of both the function and the anatomy of the intestinal barrier, activation of the immune system, development of metabolic disorders, and IR which, in turn, affect spermatogenesis and sperm viability [89]. La Merrill et al. [44] identified ten specific key characteristics or main levels where EDCs can interfere with hormone regulation and action, resulting in reproduction disorders. On the other hand, several EDCs compromise BTB integrity and consequently sperm quantity and quality [96], and the GM can protect the germ cells from environmental noxious substances, including EDCs themselves [90]. Indeed, in mice, the GM can modulate the permeability of the blood–testis barrier and influence intratesticular testosterone as well as serum LH and FSH levels. Conversely, the exposure of germ-free mice to commensal bacteria such as Clostridium tyrobutyricum can restore the blood–testis barrier integrity [90].

5. Endocrine-Disrupting Chemicals, Gut Microbiota, and Reproductive Health: An Intricate Triad with Possible Implication for Future Generations

EDC exposure during adulthood has been clearly associated with negative effect for human health; however, EDC exposure in both pre- and post-natal periods may exert even worse consequences: Firstly, because in this phase the human gut is much less resilient and much more responsive to external and environmental factors than the adult gut. Indeed, fetuses are exposed to a greater risk than adults from food contaminants due to their higher absorption rate, poor detoxification and elimination capacity, faster cell proliferation, and the still immature DNA repair mechanism [36]. Secondly, because during the perinatal and neonatal period, the microbial ecosystem inside undergoes an unprecedented process of shaping [25]. Therefore, any disturbance in this timeframe may lead to more detrimental effects than at any other moment in life affecting both the acquisition and constitution of a healthy GM, with subsequent implications for the exposed individual, offspring health, and their reproductive capability. In light of this, it is not surprising that a growing literature is showing the negative consequences of prenatal EDC exposure on offspring reproductive health.
For instance, it has been recently highlighted that gestational EDC exposure causes serious damage to the reproductive system in male offspring along with disturbing the GM. Gestational exposure to dibutyl phthalate determines an increased abundance of Bacteroidetes, Prevotella, and P. copri and causes gut dysbacteriosis in the offspring with also an increase in seminiferous atrophy and spermatogenic cell apoptosis [97]. Moreover, phthalate exposure during the gestational period seems to cause testicular damage in the offspring and abnormal phenotypes (e.g., cryptorchidism, loss of reproductive organs, hypospadias) [97]. Lastly, prenatal EDC exposure and the consequent gut–genital microbiota damage are considered at the origin and pathogenesis of endometriosis [65] and male infertility [57,97] in the offspring. Overall, these studies suggest that environmental chemicals may impair in utero programming and, in some cases (e.g., BPA), may be associated with a trans-generationally increased risk of infertility [98].
A key point now is the understanding of the underlying mechanisms regulating the observed transgenerational effects. In this regard, some authors speculated that specific GM strains can induce epigenetic changes in the host genes relevant to immunological, metabolic, and neurological development and functions [24]. For instance, Lactobacillus and Bifidobacterium can affect DNA methylation by regulating methyl-donor availability through their production of folate [99], or, via butyrate production, they act as histone deacetylase (HDAC) inhibitors and suppressors of nuclear factor-kB (NF-kB) activation, upregulating the expression of PPARγ and decreasing interferon-γ production [100,101]. Moreover, an increased Firmicutes:Bacteroidetes ratio seems to affect DNA methylation [102]. For instance, Kumar and colleagues [103] reported distinct DNA methylation profiles in blood samples from women 6 months after delivery, depending on the predominance of either Firmicutes or Bacteroidetes and Proteobacteria phyla in their fecal microbiota during pregnancy. The differences in methylation patterns affected genes whose function is linked to obesity, metabolism, and inflammation, thus highlighting their link between metabolic disorders and gut microbiota [104].
The exact mechanism by which GM chemically modulate epigenetic marks remains to be clarified. The existence of a “microbiota–nutrient metabolism–host epigenetic” axis has been postulated [105]. According to this hypothesis, the GM acts as a regulator of DNA methylation and histone modifications by altering the levels of nutrients and metabolites: on one hand, directly inhibiting enzymes that catalyze the processes, and on the other, altering the availability of substrates necessary for the enzymatic reactions. In other words, the link between epigenetic marks and GM could be mediated by host-microbial metabolites, acting as substrates and cofactors for epigenetic enzymes. In this scenario, EDCs inducing GM dysbiosis may induce a downstream effect on epigenetic programming and regulation with critical consequences if occurring during the first 1000 days of life of a human individual, a period in which epigenetic DNA imprinting activity is most active and different factors such as nutrition, microbiome, and epigenome play a key role in developmental programming, influencing the individual susceptibility to the development of diseases later in life [24].

6. Conclusions and Future Perspectives

Figure 1 highlights the main aspects described in this review: emerging concepts provide evidence of the correlation between EDC exposure, GM dysbiosis, and the occurrence of a range of infertility-related diseases in the exposed individual and in the offspring, shedding light on an intriguing triad (EDCs–GM–(in)fertility) and on its possible involvement in the (dis)regulation of reproductive health in both men and women.
In the era of precision medicine, a better understanding of the role of GM in reproduction opens the possibility to develop novel strategies to prevent or treat infertility and the diseases associated with it, such as maternal dietary modification [106], probiotic and prebiotic supplementation, and fecal microbiota transplantation [107]. Moreover, the recent correlation between microbiota composition and host epigenome suggests that the enrichment for certain microbial species could modulate unique gene expression signatures [8]. The epigenome is a dynamic player in host-microbiota crosstalk functional in precision medicine [108]. Should this fascinating hypothesis be confirmed, the control of epigenetic substrate levels produced by microbial species could represent a new avenue for clinical approaches with pre, pro or post-biotic supplementation to regulate epigenetic enzymes in the gut.

Author Contributions

G.F., P.R., D.C., S.G., M.Z., F.M.U. and L.R. conceived the manuscript. G.F., M.A. and M.F. conducted the literature search. G.F., P.R., D.C., M.F., L.S. and M.Z. drafted the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

M.F. was supported by Fondazione Umberto Veronesi. P.R., M.Z. and S.G were supported by the Italian Ministry of Education, University and Research (MUR): Dipartimenti di Eccellenza Program (2018–2022)—Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef] [PubMed]
  2. Kho, Z.Y.; Lal, S.K. The Human Gut Microbiome—A Potential Controller of Wellness and Disease. Front. Microbiol. 2018, 9, 1835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Takiishi, T.; Fenero, C.I.M.; Camara, N.O.S. Intestinal barrier and gut microbiota: Shaping our immune responses throughout life. Tissue Barriers 2017, 5, e1373208. [Google Scholar] [CrossRef] [PubMed]
  4. Pickard, J.M.; Zeng, M.Y.; Caruso, R.; Nunez, G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol. Rev. 2017, 279, 70–89. [Google Scholar] [CrossRef]
  5. Wu, H.J.; Wu, E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef] [Green Version]
  6. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
  7. Heiss, C.N.; Olofsson, L.E. Gut Microbiota-Dependent Modulation of Energy Metabolism. J. Innate Immun. 2018, 10, 163–171. [Google Scholar] [CrossRef]
  8. Woo, V.; Alenghat, T. Epigenetic regulation by gut microbiota. Gut Microbes 2022, 14, 2022407. [Google Scholar] [CrossRef]
  9. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [Green Version]
  10. Hooper, L.V.; Gordon, J.I. Commensal host-bacterial relationships in the gut. Science 2001, 292, 1115–1118. [Google Scholar] [CrossRef]
  11. Velmurugan, G.; Ramprasath, T.; Gilles, M.; Swaminathan, K.; Ramasamy, S. Gut Microbiota, Endocrine-Disrupting Chemicals, and the Diabetes Epidemic. Trends Endocrinol. Metab. 2017, 28, 612–625. [Google Scholar] [CrossRef]
  12. Pu, D.; Zhang, Z.; Feng, B. Alterations and Potential Applications of Gut Microbiota in Biological Therapy for Inflammatory Bowel Diseases. Front. Pharmacol. 2022, 13, 906419. [Google Scholar] [CrossRef]
  13. Gill, P.A.; Inniss, S.; Kumagai, T.; Rahman, F.Z.; Smith, A.M. The Role of Diet and Gut Microbiota in Regulating Gastrointestinal and Inflammatory Disease. Front. Immunol. 2022, 13, 866059. [Google Scholar] [CrossRef]
  14. Guan, Z.; Luo, L.; Liu, S.; Guan, Z.; Zhang, Q.; Li, X.; Tao, K. The Role of Depletion of Gut Microbiota in Osteoporosis and Osteoarthritis: A Narrative Review. Front. Endocrinol. 2022, 13, 847401. [Google Scholar] [CrossRef]
  15. Sadrekarimi, H.; Gardanova, Z.R.; Bakhshesh, M.; Ebrahimzadeh, F.; Yaseri, A.F.; Thangavelu, L.; Hasanpoor, Z.; Zadeh, F.A.; Kahrizi, M.S. Emerging role of human microbiome in cancer development and response to therapy: Special focus on intestinal microflora. J. Transl. Med. 2022, 20, 301. [Google Scholar] [CrossRef]
  16. Mitrea, L.; Nemes, S.A.; Szabo, K.; Teleky, B.E.; Vodnar, D.C. Guts Imbalance Imbalances the Brain: A Review of Gut Microbiota Association With Neurological and Psychiatric Disorders. Front. Med. 2022, 9, 813204. [Google Scholar] [CrossRef]
  17. Callender, C.; Attaye, I.; Nieuwdorp, M. The Interaction between the Gut Microbiome and Bile Acids in Cardiometabolic Diseases. Metabolites 2022, 12, 65. [Google Scholar] [CrossRef]
  18. Rajendiran, E.; Ramadass, B.; Ramprasath, V. Understanding connections and roles of gut microbiome in cardiovascular diseases. Can. J. Microbiol. 2021, 67, 101–111. [Google Scholar] [CrossRef]
  19. Asadi, A.; Shadab Mehr, N.; Mohamadi, M.H.; Shokri, F.; Heidary, M.; Sadeghifard, N.; Khoshnood, S. Obesity and gut-microbiota-brain axis: A narrative review. J. Clin. Lab. Anal. 2022, 36, e24420. [Google Scholar] [CrossRef]
  20. Bouter, K.E.; van Raalte, D.H.; Groen, A.K.; Nieuwdorp, M. Role of the Gut Microbiome in the Pathogenesis of Obesity and Obesity-Related Metabolic Dysfunction. Gastroenterology 2017, 152, 1671–1678. [Google Scholar] [CrossRef]
  21. Ogunrinola, G.A.; Oyewale, J.O.; Oshamika, O.O.; Olasehinde, G.I. The Human Microbiome and Its Impacts on Health. Int. J. Microbiol. 2020, 2020, 8045646. [Google Scholar] [CrossRef]
  22. Cho, I.; Blaser, M.J. The human microbiome: At the interface of health and disease. Nat. Rev. Genet. 2012, 13, 260–270. [Google Scholar] [CrossRef] [Green Version]
  23. Kinashi, Y.; Hase, K. Partners in Leaky Gut Syndrome: Intestinal Dysbiosis and Autoimmunity. Front. Immunol. 2021, 12, 673708. [Google Scholar] [CrossRef]
  24. Indrio, F.; Martini, S.; Francavilla, R.; Corvaglia, L.; Cristofori, F.; Mastrolia, S.A.; Neu, J.; Rautava, S.; Russo Spena, G.; Raimondi, F.; et al. Epigenetic Matters: The Link between Early Nutrition, Microbiome, and Long-term Health Development. Front. Pediatr. 2017, 5, 178. [Google Scholar] [CrossRef] [Green Version]
  25. Laursen, M.F.; Bahl, M.I.; Licht, T.R. Settlers of our inner surface—Factors shaping the gut microbiota from birth to toddlerhood. FEMS Microbiol. Rev. 2021, 45, fuab001. [Google Scholar] [CrossRef]
  26. Chu, D.M.; Ma, J.; Prince, A.L.; Antony, K.M.; Seferovic, M.D.; Aagaard, K.M. Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat. Med. 2017, 23, 314–326. [Google Scholar] [CrossRef] [Green Version]
  27. Pannaraj, P.S.; Li, F.; Cerini, C.; Bender, J.M.; Yang, S.; Rollie, A.; Adisetiyo, H.; Zabih, S.; Lincez, P.J.; Bittinger, K.; et al. Association Between Breast Milk Bacterial Communities and Establishment and Development of the Infant Gut Microbiome. JAMA Pediatr. 2017, 171, 647–654. [Google Scholar] [CrossRef] [Green Version]
  28. Ferretti, P.; Pasolli, E.; Tett, A.; Asnicar, F.; Gorfer, V.; Fedi, S.; Armanini, F.; Truong, D.T.; Manara, S.; Zolfo, M.; et al. Mother-to-Infant Microbial Transmission from Different Body Sites Shapes the Developing Infant Gut Microbiome. Cell Host Microbe 2018, 24, 133–145.e5. [Google Scholar] [CrossRef] [PubMed]
  29. Gao, Y.; Nanan, R.; Macia, L.; Tan, J.; Sominsky, L.; Quinn, T.P.; O’Hely, M.; Ponsonby, A.L.; Tang, M.L.K.; Collier, F.; et al. The maternal gut microbiome during pregnancy and offspring allergy and asthma. J. Allergy Clin. Immunol. 2021, 148, 669–678. [Google Scholar] [CrossRef] [PubMed]
  30. Koleva, P.T.; Kim, J.S.; Scott, J.A.; Kozyrskyj, A.L. Microbial programming of health and disease starts during fetal life. Birth Defects Res. C Embryo Today 2015, 105, 265–277. [Google Scholar] [CrossRef] [PubMed]
  31. Hosseinkhani, F.; Heinken, A.; Thiele, I.; Lindenburg, P.W.; Harms, A.C.; Hankemeier, T. The contribution of gut bacterial metabolites in the human immune signaling pathway of non-communicable diseases. Gut Microbes 2021, 13, 1–22. [Google Scholar] [CrossRef]
  32. Neu, J. Developmental aspects of maternal-fetal, and infant gut microbiota and implications for long-term health. Matern. Health Neonatol. Perinatol. 2015, 1, 6. [Google Scholar] [CrossRef] [Green Version]
  33. Miller, W.B., Jr. The Eukaryotic Microbiome: Origins and Implications for Fetal and Neonatal Life. Front. Pediatr. 2016, 4, 96. [Google Scholar] [CrossRef] [Green Version]
  34. Funkhouser, L.J.; Bordenstein, S.R. Mom knows best: The universality of maternal microbial transmission. PLoS Biol. 2013, 11, e1001631. [Google Scholar] [CrossRef] [Green Version]
  35. Perez-Cano, F.J. What Does Influence the Neonatal Microbiome? Nutrients 2020, 12, 2472. [Google Scholar] [CrossRef]
  36. Sarron, E.; Perot, M.; Barbezier, N.; Delayre-Orthez, C.; Gay-Queheillard, J.; Anton, P.M. Early exposure to food contaminants reshapes maturation of the human brain-gut-microbiota axis. World J. Gastroenterol. 2020, 26, 3145–3169. [Google Scholar] [CrossRef]
  37. Hasan, N.; Yang, H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 2019, 7, e7502. [Google Scholar] [CrossRef] [Green Version]
  38. Gore, A.C.; Chappell, V.A.; Fenton, S.E.; Flaws, J.A.; Nadal, A.; Prins, G.S.; Toppari, J.; Zoeller, R.T. Executive Summary to EDC-2: The Endocrine Society’s Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr. Rev. 2015, 36, 593–602. [Google Scholar] [CrossRef] [Green Version]
  39. Diamanti-Kandarakis, E.; Bourguignon, J.P.; Giudice, L.C.; Hauser, R.; Prins, G.S.; Soto, A.M.; Zoeller, R.T.; Gore, A.C. Endocrine-disrupting chemicals: An Endocrine Society scientific statement. Endocr. Rev. 2009, 30, 293–342. [Google Scholar] [CrossRef]
  40. Kabir, E.R.; Rahman, M.S.; Rahman, I. A review on endocrine disruptors and their possible impacts on human health. Environ. Toxicol. Pharmacol. 2015, 40, 241–258. [Google Scholar] [CrossRef]
  41. Claxton, L.D.; Houk, V.S.; Hughes, T.J. Genotoxicity of industrial wastes and effluents. Mutat. Res. 1998, 410, 237–243. [Google Scholar] [CrossRef]
  42. Choudhuri, S.; Kaur, T.; Jain, S.; Sharma, C.; Asthana, S. A review on genotoxicity in connection to infertility and cancer. Chem. Biol. Interact. 2021, 345, 109531. [Google Scholar] [CrossRef]
  43. Rattan, S.; Flaws, J.A. The epigenetic impacts of endocrine disruptors on female reproduction across generationsdagger. Biol. Reprod. 2019, 101, 635–644. [Google Scholar] [CrossRef]
  44. La Merrill, M.A.; Vandenberg, L.N.; Smith, M.T.; Goodson, W.; Browne, P.; Patisaul, H.B.; Guyton, K.Z.; Kortenkamp, A.; Cogliano, V.J.; Woodruff, T.J.; et al. Consensus on the key characteristics of endocrine-disrupting chemicals as a basis for hazard identification. Nat. Rev. Endocrinol. 2020, 16, 45–57. [Google Scholar] [CrossRef] [Green Version]
  45. Galvez-Ontiveros, Y.; Paez, S.; Monteagudo, C.; Rivas, A. Endocrine Disruptors in Food: Impact on Gut Microbiota and Metabolic Diseases. Nutrients 2020, 12, 1158. [Google Scholar] [CrossRef]
  46. Eggers, S.; Safdar, N.; Sethi, A.K.; Suen, G.; Peppard, P.E.; Kates, A.E.; Skarlupka, J.H.; Kanarek, M.; Malecki, K.M.C. Urinary lead concentration and composition of the adult gut microbiota in a cross-sectional population-based sample. Environ. Int. 2019, 133, 105122. [Google Scholar] [CrossRef] [PubMed]
  47. Walters, K.E.; Martiny, J.B.H. Alpha-, beta-, and gamma-diversity of bacteria varies across habitats. PLoS ONE 2020, 15, e0233872. [Google Scholar] [CrossRef]
  48. Matamoros, S.; Gras-Leguen, C.; Le Vacon, F.; Potel, G.; de La Cochetiere, M.F. Development of intestinal microbiota in infants and its impact on health. Trends Microbiol. 2013, 21, 167–173. [Google Scholar] [CrossRef]
  49. Yang, Y.N.; Yang, Y.S.H.; Lin, I.H.; Chen, Y.Y.; Lin, H.Y.; Wu, C.Y.; Su, Y.T.; Yang, Y.J.; Yang, S.N.; Suen, J.L. Phthalate exposure alters gut microbiota composition and IgM vaccine response in human newborns. Food Chem. Toxicol. 2019, 132, 110700. [Google Scholar] [CrossRef] [PubMed]
  50. Wang, Y.; Rui, M.; Nie, Y.; Lu, G. Influence of gastrointestinal tract on metabolism of bisphenol A as determined by in vitro simulated system. J. Hazard. Mater. 2018, 355, 111–118. [Google Scholar] [CrossRef] [PubMed]
  51. Claus, S.P.; Guillou, H.; Ellero-Simatos, S. The gut microbiota: A major player in the toxicity of environmental pollutants? NPJ Biofilms Microbiomes 2016, 2, 16003. [Google Scholar] [CrossRef] [Green Version]
  52. Li, N.; Li, J.; Zhang, Q.; Gao, S.; Quan, X.; Liu, P.; Xu, C. Effects of endocrine disrupting chemicals in host health: Three-way interactions between environmental exposure, host phenotypic responses, and gut microbiota. Environ. Pollut. 2021, 271, 116387. [Google Scholar] [CrossRef]
  53. Rosenfeld, C.S. Gut Dysbiosis in Animals Due to Environmental Chemical Exposures. Front. Cell Infect. Microbiol. 2017, 7, 396. [Google Scholar] [CrossRef] [Green Version]
  54. Javurek, A.B.; Spollen, W.G.; Johnson, S.A.; Bivens, N.J.; Bromert, K.H.; Givan, S.A.; Rosenfeld, C.S. Effects of exposure to bisphenol A and ethinyl estradiol on the gut microbiota of parents and their offspring in a rodent model. Gut Microbes 2016, 7, 471–485. [Google Scholar] [CrossRef] [Green Version]
  55. Hu, J.; Raikhel, V.; Gopalakrishnan, K.; Fernandez-Hernandez, H.; Lambertini, L.; Manservisi, F.; Falcioni, L.; Bua, L.; Belpoggi, F.; Teitelbaum, L.S.; et al. Effect of postnatal low-dose exposure to environmental chemicals on the gut microbiome in a rodent model. Microbiome 2016, 4, 26. [Google Scholar] [CrossRef] [Green Version]
  56. Magne, F.; Gotteland, M.; Gauthier, L.; Zazueta, A.; Pesoa, S.; Navarrete, P.; Balamurugan, R. The Firmicutes/Bacteroidetes Ratio: A Relevant Marker of Gut Dysbiosis in Obese Patients? Nutrients 2020, 12, 1474. [Google Scholar] [CrossRef]
  57. Hampl, R.; Starka, L. Endocrine disruptors and gut microbiome interactions. Physiol. Res. 2020, 69, S211–S223. [Google Scholar] [CrossRef]
  58. McIntosh, F.M.; Maison, N.; Holtrop, G.; Young, P.; Stevens, V.J.; Ince, J.; Johnstone, A.M.; Lobley, G.E.; Flint, H.J.; Louis, P. Phylogenetic distribution of genes encoding beta-glucuronidase activity in human colonic bacteria and the impact of diet on faecal glycosidase activities. Environ. Microbiol. 2012, 14, 1876–1887. [Google Scholar] [CrossRef]
  59. He, S.; Li, H.; Yu, Z.; Zhang, F.; Liang, S.; Liu, H.; Chen, H.; Lu, M. The Gut Microbiome and Sex Hormone-Related Diseases. Front. Microbiol. 2021, 12, 711137. [Google Scholar] [CrossRef]
  60. Baker, J.M.; Al-Nakkash, L.; Herbst-Kralovetz, M.M. Estrogen-gut microbiome axis: Physiological and clinical implications. Maturitas 2017, 103, 45–53. [Google Scholar] [CrossRef]
  61. Ervin, S.M.; Li, H.; Lim, L.; Roberts, L.R.; Liang, X.; Mani, S.; Redinbo, M.R. Gut microbial beta-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. J. Biol. Chem. 2019, 294, 18586–18599. [Google Scholar] [CrossRef]
  62. Plottel, C.S.; Blaser, M.J. Microbiome and malignancy. Cell Host Microbe 2011, 10, 324–335. [Google Scholar] [CrossRef] [Green Version]
  63. Rooks, M.G.; Garrett, W.S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016, 16, 341–352. [Google Scholar] [CrossRef]
  64. Jiang, I.; Yong, P.J.; Allaire, C.; Bedaiwy, M.A. Intricate Connections between the Microbiota and Endometriosis. Int. J. Mol. Sci. 2021, 22, 5644. [Google Scholar] [CrossRef] [PubMed]
  65. Garcia-Penarrubia, P.; Ruiz-Alcaraz, A.J.; Martinez-Esparza, M.; Marin, P.; Machado-Linde, F. Hypothetical roadmap towards endometriosis: Prenatal endocrine-disrupting chemical pollutant exposure, anogenital distance, gut-genital microbiota and subclinical infections. Hum. Reprod. Update 2020, 26, 214–246. [Google Scholar] [CrossRef] [PubMed]
  66. Qi, X.; Yun, C.; Sun, L.; Xia, J.; Wu, Q.; Wang, Y.; Wang, L.; Zhang, Y.; Liang, X.; Wang, L.; et al. Publisher Correction: Gut microbiota-bile acid-interleukin-22 axis orchestrates polycystic ovary syndrome. Nat. Med. 2019, 25, 1459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Giampaolino, P.; Foreste, V.; Di Filippo, C.; Gallo, A.; Mercorio, A.; Serafino, P.; Improda, F.P.; Verrazzo, P.; Zara, G.; Buonfantino, C.; et al. Microbiome and PCOS: State-of-Art and Future Aspects. Int. J. Mol. Sci. 2021, 22, 2048. [Google Scholar] [CrossRef] [PubMed]
  68. He, F.F.; Li, Y.M. Role of gut microbiota in the development of insulin resistance and the mechanism underlying polycystic ovary syndrome: A review. J. Ovarian Res. 2020, 13, 73. [Google Scholar] [CrossRef]
  69. Kriebs, A. IL-22 links gut microbiota to PCOS. Nat. Rev. Endocrinol. 2019, 15, 565. [Google Scholar] [CrossRef]
  70. Yurtdas, G.; Akdevelioglu, Y. A New Approach to Polycystic Ovary Syndrome: The Gut Microbiota. J. Am. Coll. Nutr. 2020, 39, 371–382. [Google Scholar] [CrossRef]
  71. Guo, J.; Shao, J.; Yang, Y.; Niu, X.; Liao, J.; Zhao, Q.; Wang, D.; Li, S.; Hu, J. Gut Microbiota in Patients with Polycystic Ovary Syndrome: A Systematic Review. Reprod. Sci. 2022, 29, 69–83. [Google Scholar] [CrossRef]
  72. Saad, M.J.; Santos, A.; Prada, P.O. Linking Gut Microbiota and Inflammation to Obesity and Insulin Resistance. Physiology 2016, 31, 283–293. [Google Scholar] [CrossRef] [Green Version]
  73. Scheithauer, T.P.M.; Rampanelli, E.; Nieuwdorp, M.; Vallance, B.A.; Verchere, C.B.; van Raalte, D.H.; Herrema, H. Gut Microbiota as a Trigger for Metabolic Inflammation in Obesity and Type 2 Diabetes. Front. Immunol. 2020, 11, 571731. [Google Scholar] [CrossRef]
  74. de Rivero Vaccari, J.P. The Inflammasome in Reproductive Biology: A Promising Target for Novel Therapies. Front. Endocrinol. 2020, 11, 8. [Google Scholar] [CrossRef] [Green Version]
  75. Wang, J.W.; Liao, X.X.; Li, T. Thyroid Autoimmunity in Adverse Fertility and Pregnancy Outcomes: Timing of Assisted Reproductive Technology in AITD Women. J. Transl. Int. Med. 2021, 9, 76–83. [Google Scholar] [CrossRef]
  76. Poppe, K. Management of Endocrine Disease: Thyroid and female infertility: More questions than answers?! Eur. J. Endocrinol. 2021, 184, R123–R135. [Google Scholar] [CrossRef]
  77. Twig, G.; Shina, A.; Amital, H.; Shoenfeld, Y. Pathogenesis of infertility and recurrent pregnancy loss in thyroid autoimmunity. J. Autoimmun. 2012, 38, J275–J281. [Google Scholar] [CrossRef]
  78. Wu, J.; Zhuo, Y.; Liu, Y.; Chen, Y.; Ning, Y.; Yao, J. Association between premature ovarian insufficiency and gut microbiota. BMC Pregnancy Childbirth 2021, 21, 418. [Google Scholar] [CrossRef]
  79. Jiang, L.; Fei, H.; Tong, J.; Zhou, J.; Zhu, J.; Jin, X.; Shi, Z.; Zhou, Y.; Ma, X.; Yu, H.; et al. Hormone Replacement Therapy Reverses Gut Microbiome and Serum Metabolome Alterations in Premature Ovarian Insufficiency. Front. Endocrinol. 2021, 12, 794496. [Google Scholar] [CrossRef]
  80. Amabebe, E.; Anumba, D.O.C. Female Gut and Genital Tract Microbiota-Induced Crosstalk and Differential Effects of Short-Chain Fatty Acids on Immune Sequelae. Front. Immunol. 2020, 11, 2184. [Google Scholar] [CrossRef]
  81. Benner, M.; Ferwerda, G.; Joosten, I.; van der Molen, R.G. How uterine microbiota might be responsible for a receptive, fertile endometrium. Hum. Reprod. Update 2018, 24, 393–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Wang, J.; Li, Z.; Ma, X.; Du, L.; Jia, Z.; Cui, X.; Yu, L.; Yang, J.; Xiao, L.; Zhang, B.; et al. Translocation of vaginal microbiota is involved in impairment and protection of uterine health. Nat. Commun. 2021, 12, 4191. [Google Scholar] [CrossRef] [PubMed]
  83. Amabebe, E.; Anumba, D.O.C. The Vaginal Microenvironment: The Physiologic Role of Lactobacilli. Front. Med. 2018, 5, 181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Dominianni, C.; Sinha, R.; Goedert, J.J.; Pei, Z.; Yang, L.; Hayes, R.B.; Ahn, J. Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLoS ONE 2015, 10, e0124599. [Google Scholar] [CrossRef] [Green Version]
  85. Haro, C.; Rangel-Zuniga, O.A.; Alcala-Diaz, J.F.; Gomez-Delgado, F.; Perez-Martinez, P.; Delgado-Lista, J.; Quintana-Navarro, G.M.; Landa, B.B.; Navas-Cortes, J.A.; Tena-Sempere, M.; et al. Intestinal Microbiota Is Influenced by Gender and Body Mass Index. PLoS ONE 2016, 11, e0154090. [Google Scholar] [CrossRef] [Green Version]
  86. Diviccaro, S.; Giatti, S.; Borgo, F.; Falvo, E.; Caruso, D.; Garcia-Segura, L.M.; Melcangi, R.C. Steroidogenic machinery in the adult rat colon. J. Steroid. Biochem. Mol. Biol. 2020, 203, 105732. [Google Scholar] [CrossRef]
  87. Ly, L.K.; Rowles, J.L., 3rd; Paul, H.M.; Alves, J.M.P.; Yemm, C.; Wolf, P.M.; Devendran, S.; Hudson, M.E.; Morris, D.J.; Erdman, J.W., Jr.; et al. Bacterial steroid-17,20-desmolase is a taxonomically rare enzymatic pathway that converts prednisone to 1,4-androstanediene-3,11,17-trione, a metabolite that causes proliferation of prostate cancer cells. J. Steroid. Biochem. Mol. Biol. 2020, 199, 105567. [Google Scholar] [CrossRef]
  88. Arroyo, P.; Ho, B.S.; Sau, L.; Kelley, S.T.; Thackray, V.G. Letrozole treatment of pubertal female mice results in activational effects on reproduction, metabolism and the gut microbiome. PLoS ONE 2019, 14, e0223274. [Google Scholar] [CrossRef] [Green Version]
  89. Wang, Y.; Xie, Z. Exploring the role of gut microbiome in male reproduction. Andrology 2022, 10, 441–450. [Google Scholar] [CrossRef]
  90. Al-Asmakh, M.; Stukenborg, J.B.; Reda, A.; Anuar, F.; Strand, M.L.; Hedin, L.; Pettersson, S.; Soder, O. The gut microbiota and developmental programming of the testis in mice. PLoS ONE 2014, 9, e103809. [Google Scholar] [CrossRef]
  91. Zhang, T.; Sun, P.; Geng, Q.; Fan, H.; Gong, Y.; Hu, Y.; Shan, L.; Sun, Y.; Shen, W.; Zhou, Y. Disrupted spermatogenesis in a metabolic syndrome model: The role of vitamin A metabolism in the gut-testis axis. Gut 2022, 71, 78–87. [Google Scholar] [CrossRef]
  92. Alfano, M.; Ferrarese, R.; Locatelli, I.; Ventimiglia, E.; Ippolito, S.; Gallina, P.; Cesana, D.; Canducci, F.; Pagliardini, L.; Vigano, P.; et al. Testicular microbiome in azoospermic men-first evidence of the impact of an altered microenvironment. Hum. Reprod. 2018, 33, 1212–1217. [Google Scholar] [CrossRef]
  93. Hou, D.; Zhou, X.; Zhong, X.; Settles, M.L.; Herring, J.; Wang, L.; Abdo, Z.; Forney, L.J.; Xu, C. Microbiota of the seminal fluid from healthy and infertile men. Fertil. Steril. 2013, 100, 1261–1269. [Google Scholar] [CrossRef] [Green Version]
  94. Weng, S.L.; Chiu, C.M.; Lin, F.M.; Huang, W.C.; Liang, C.; Yang, T.; Yang, T.L.; Liu, C.Y.; Wu, W.Y.; Chang, Y.A.; et al. Bacterial communities in semen from men of infertile couples: Metagenomic sequencing reveals relationships of seminal microbiota to semen quality. PLoS ONE 2014, 9, e110152. [Google Scholar] [CrossRef] [Green Version]
  95. Li, H.; Qi, T.; Huang, Z.S.; Ying, Y.; Zhang, Y.; Wang, B.; Ye, L.; Zhang, B.; Chen, D.L.; Chen, J. Relationship between gut microbiota and type 2 diabetic erectile dysfunction in Sprague-Dawley rats. J. Huazhong Univ. Sci. Technol. Med. Sci. 2017, 37, 523–530. [Google Scholar] [CrossRef]
  96. Wu, D.; Huang, C.J.; Jiao, X.F.; Ding, Z.M.; Zhang, S.X.; Miao, Y.L.; Huo, L.J. Bisphenol AF compromises blood-testis barrier integrity and sperm quality in mice. Chemosphere 2019, 237, 124410. [Google Scholar] [CrossRef]
  97. Zhang, T.; Zhou, X.; Zhang, X.; Ren, X.; Wu, J.; Wang, Z.; Wang, S.; Wang, Z. Gut microbiota may contribute to the postnatal male reproductive abnormalities induced by prenatal dibutyl phthalate exposure. Chemosphere 2022, 287, 132046. [Google Scholar] [CrossRef]
  98. Rodriguez-Caro, H.; Williams, S.A. Strategies to reduce non-communicable diseases in the offspring: Negative and positive in utero programming. J. Dev. Orig. Health Dis. 2018, 9, 642–652. [Google Scholar] [CrossRef]
  99. Rossi, M.; Amaretti, A.; Raimondi, S. Folate production by probiotic bacteria. Nutrients 2011, 3, 118–134. [Google Scholar] [CrossRef] [Green Version]
  100. Berni Canani, R.; Di Costanzo, M.; Leone, L. The epigenetic effects of butyrate: Potential therapeutic implications for clinical practice. Clin. Epigenetics 2012, 4, 4. [Google Scholar] [CrossRef]
  101. Fofanova, T.Y.; Petrosino, J.F.; Kellermayer, R. Microbiome-Epigenome Interactions and the Environmental Origins of Inflammatory Bowel Diseases. J. Pediatr. Gastroenterol. Nutr. 2016, 62, 208–219. [Google Scholar] [CrossRef] [Green Version]
  102. Ramos-Molina, B.; Sanchez-Alcoholado, L.; Cabrera-Mulero, A.; Lopez-Dominguez, R.; Carmona-Saez, P.; Garcia-Fuentes, E.; Moreno-Indias, I.; Tinahones, F.J. Gut Microbiota Composition Is Associated With the Global DNA Methylation Pattern in Obesity. Front. Genet. 2019, 10, 613. [Google Scholar] [CrossRef] [Green Version]
  103. Kumar, H.; Lund, R.; Laiho, A.; Lundelin, K.; Ley, R.E.; Isolauri, E.; Salminen, S. Gut microbiota as an epigenetic regulator: Pilot study based on whole-genome methylation analysis. mBio 2014, 5, e02113–e02114. [Google Scholar] [CrossRef] [Green Version]
  104. Yang, J.Y.; Kweon, M.N. The gut microbiota: A key regulator of metabolic diseases. BMB Rep. 2016, 49, 536–541. [Google Scholar] [CrossRef] [Green Version]
  105. Miro-Blanch, J.; Yanes, O. Epigenetic Regulation at the Interplay Between Gut Microbiota and Host Metabolism. Front. Genet. 2019, 10, 638. [Google Scholar] [CrossRef]
  106. Druart, C.; Alligier, M.; Salazar, N.; Neyrinck, A.M.; Delzenne, N.M. Modulation of the gut microbiota by nutrients with prebiotic and probiotic properties. Adv. Nutr. 2014, 5, 624S–633S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Gulliver, E.L.; Young, R.B.; Chonwerawong, M.; D’Adamo, G.L.; Thomason, T.; Widdop, J.T.; Rutten, E.L.; Rossetto Marcelino, V.; Bryant, R.V.; Costello, S.P.; et al. Review article: The future of microbiome-based therapeutics. Aliment. Pharmacol. Ther. 2022, 56, 192–208. [Google Scholar] [CrossRef] [PubMed]
  108. Peery, R.C.; Pammi, M.; Claud, E.; Shen, L. Epigenome—A mediator for host-microbiome crosstalk. Semin. Perinatol. 2021, 45, 151455. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the intricate relationship among endocrine-disrupting chemicals (EDCs), gut microbiota (GM), and reproductive health. EDCs induce GM damage through several mechanisms of action, leading to GM dysbiosis, intestinal permeability, and chronic low-grade inflammation, conditions linked to several diseases, including those related to infertility. When a dysbiotic condition occurs during pregnancy or in the early post-natal period, it may lead to even worse detrimental effects, affecting both the acquisition and constitution of a healthy GM, with subsequent implications on the exposed individual, the offspring’s health, and reproductive capability.
Figure 1. Overview of the intricate relationship among endocrine-disrupting chemicals (EDCs), gut microbiota (GM), and reproductive health. EDCs induce GM damage through several mechanisms of action, leading to GM dysbiosis, intestinal permeability, and chronic low-grade inflammation, conditions linked to several diseases, including those related to infertility. When a dysbiotic condition occurs during pregnancy or in the early post-natal period, it may lead to even worse detrimental effects, affecting both the acquisition and constitution of a healthy GM, with subsequent implications on the exposed individual, the offspring’s health, and reproductive capability.
Cells 11 03335 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fabozzi, G.; Rebuzzini, P.; Cimadomo, D.; Allori, M.; Franzago, M.; Stuppia, L.; Garagna, S.; Ubaldi, F.M.; Zuccotti, M.; Rienzi, L. Endocrine-Disrupting Chemicals, Gut Microbiota, and Human (In)Fertility—It Is Time to Consider the Triad. Cells 2022, 11, 3335. https://0-doi-org.brum.beds.ac.uk/10.3390/cells11213335

AMA Style

Fabozzi G, Rebuzzini P, Cimadomo D, Allori M, Franzago M, Stuppia L, Garagna S, Ubaldi FM, Zuccotti M, Rienzi L. Endocrine-Disrupting Chemicals, Gut Microbiota, and Human (In)Fertility—It Is Time to Consider the Triad. Cells. 2022; 11(21):3335. https://0-doi-org.brum.beds.ac.uk/10.3390/cells11213335

Chicago/Turabian Style

Fabozzi, Gemma, Paola Rebuzzini, Danilo Cimadomo, Mariachiara Allori, Marica Franzago, Liborio Stuppia, Silvia Garagna, Filippo Maria Ubaldi, Maurizio Zuccotti, and Laura Rienzi. 2022. "Endocrine-Disrupting Chemicals, Gut Microbiota, and Human (In)Fertility—It Is Time to Consider the Triad" Cells 11, no. 21: 3335. https://0-doi-org.brum.beds.ac.uk/10.3390/cells11213335

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop