Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Case Series
Commentary
Editorial
Invited Article
Original Article
Review Article
Short Communication
Special Issue on COVID-19 & ART
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Case Series
Commentary
Editorial
Invited Article
Original Article
Review Article
Short Communication
Special Issue on COVID-19 & ART
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Case Series
Commentary
Editorial
Invited Article
Original Article
Review Article
Short Communication
Special Issue on COVID-19 & ART
View/Download PDF

Translate this page into:

Invited Article
2020
:1;
2
doi:
10.25259/JRHM_11_2020

Endocrine disruption and female reproductive health: Implications on cross-talk between endocrine and autocrine/paracrine axes in the ovary

Department of Zoology, Visva-Bharati University, Santiniketan, West Bengal, India

*Corresponding author: Sudipta Maitra, Department of Zoology, Visva-Bharati University, Santiniketan - 731 235, West Bengal, India. s.maitra@visva-bharati.ac.in

Licence
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Biswas S, Mukherjee U, Maitra S. Endocrine disruption and female reproductive health: Implications on cross-talk between endocrine and autocrine/paracrine axes in the ovary. J Reprod Healthc Med 2020;1:2.

Abstract

Female reproduction is a blend of neuroendocrine, endocrine, and autocrine/paracrine factors that maintain the appropriate ovarian micro-environment. The growing urbanization prompted exposure to a myriad of environmental toxins carrying the ability to interfere with reproductive processes governed by endogenous hormones, making reproductive health a major global concern. These environmental anthropogenic contaminants, popularly termed as endocrine-disrupting chemicals (EDCs), can disrupt the ovarian homeostasis leading to serious perturbations, namely, anovulation, infertility, estrogen deficiency, and premature ovarian failure. Although gonadotropin action, biosynthesis of gonadal steroids vis-à-vis growth factors comprise the essential modulators within the ovary, the redox balance along with inflammatory and cell death response can dramatically influence the framework of ovarian dynamics; however, details of which remain relatively less understood. The present overview provides an update on candidates (endocrines and autocrine/paracrine) of oogenesis, and the potential impact of EDCs on diverse intra-ovarian entities including but not limited to gonadotropin action, steroidogenic potential, expression of growth factors, and modulation of maturational competence. Moreover, the relative importance of free radical-induced stress, inflammation, and elevated cell death (follicular atresia), in the regulation of ovarian functions and how these intricate yet conjoined mechanisms may alter the reproductive performance of a female will be an issue of discussion.

Keywords

Endocrine disruption
Gonadal steroids
Insulin-like growth factors and epidermal growth factor
Redox balance and follicular apoptosis
Infertility

INTRODUCTION

Reproduction is the sole process in any species that is responsible to sustain its progeny on earth. A female who starts her journey with 2 million oocytes ends with just 400 follicles capable of ovulating during her reproductive life.[1] This marks the immense importance of every single follicle for the nature that has taken all considerations intelligently in designing and making it of utmost quality for positive successful fertilization. In today’s urbanized globe, females are exposed to a myriad of anthropogenic substances, drugs, and exogenous compounds in the environment, popularly termed as endocrine disrupting chemicals (EDCs), which might lead to an overabundance of infertility related disorders. In addition to altered lifestyle, early and recurrent pregnancy; multitudes of environmental anthropogenic contaminants, poor nutritional status, sanitation facility, and ill-developed infrastructure at rural areas are posing great risks on female reproductive health specially in developing nations. The majority of EDCs interfere with neuroendocrine and endocrine functions thereby distressing reproduction and embryonic development.[2] Moreover, females are constantly undergoing steroidal/nonsteroidal drug regimens during various ovarian disorders, for example, for ovulation induction in women with polycystic ovary syndrome (PCOS), aromatase inhibition therapy for advanced breast cancer or even drugs used as contraceptive pills that might pose side effects on female reproductive health.[3,4] In this context, active and systematic research initiatives may help to better understand the complex dialogs between neuroendocrine, endocrine and juxtacrine modulators, their potential targets, and extent of damage inflicted by various harmful agents [Figure 1]. Based on these major amendments in existing policies, long-term planning and implementation of child and women health-care measures at the grass-root level may help in empowering women of this country to gain access in the globe of sound reproductive health.

Risks of drugs/environmental toxicants on female reproduction.
Figure 1:
Risks of drugs/environmental toxicants on female reproduction.

ENDOCRINE DISRUPTORS: AN UPDATE

A functional endocrine system is a balanced network of chemical messengers in the form of hormones that regulate and coordinate bodily functions. In the present milieu of 21st century with growing urbanization and profuse use of medications, a plethora of chemicals exist that possess the ability to interfere with the natural course of hormone action. They are potent enough to disrupt the normal homeostatic processes of hormone activity and have been termed as endocrine-disrupting chemicals (EDCs). Endocrine disruptors are known to (a) mimic the effects of endogenous hormones; (b) antagonize the effects of endogenous hormones; (c) alter the pattern of synthesis and metabolism of natural hormones; and (d) modify hormone receptor levels.[5] Considering the pervasive nature and risks involved from exposure to EDCs, many of which are estrogenic in nature, United States Environmental Protection Agency developed a two-tier screening and testing program to detect xenobiotics. While tier I screening protocol included battery of assays to identify chemicals that interact with estrogen, androgen, and thyroid systems, tier two screening was executed for those chemicals that are positive in first screening and to further determine whether the chemical substance actually could interfere with endocrine-related processes.[6]

IMPACT OF EDC EXPOSURE ON FEMALE REPRODUCTIVE HEALTH: A FOCUS ON XENOESTROGENS

A major subset of EDCs capable of eliciting estrogenic effects is the “xenoestrogens” (XEs) that have received a global concern for putting female reproduction at stake. XEs can exert their influence at both genomic and non-genomic level thereby activating a wide variety of signaling pathways and downstream kinases.[7] Exposure to XEs has been reported to dysregulate the entire hypothalamic-pituitary-gonadal system through increased GnRH production resulting in an increased endogenous estrogen production and premature puberty.[8] Estrogen, the female steroid hormone, regulates a wide array of female reproductive events, including endometrial growth, menstrual cycle regulation, and breast development. Interestingly, ovaries and uteri are most sensitive to XEs leading to a surfeit of female reproductive disorders such as early puberty, premature ovarian failure, impaired fertility as well as breast, ovarian, and uterine cancers.[9] Given the host of endocrine abnormalities associated with multitude of ovarian diseases (obesity, PCOS, endometriosis, ovarian cancer, etc.), EDCs in the environment are worth considering to have plausible association with such pathophysiologic conditions. Over 70% of the women have been diagnosed with sporadic breast cancer and this risk has increased at an exponential rate with the increased exposure to estrogenic chemicals.[10] Endometriosis, another estrogen-dependent gynecological disease, wherein endometrium-like tissue grows outside uterine cavity is also one of the most common causes of infertility in women of reproductive age.[11] Surveys have revealed that women who have used estrogen replacement therapy are more likely to develop endometrial cancer than those who never underwent estrogen replacement therapy.[12] Moreover, estrogen also stimulates the growth of ovarian tumor cell lines expressing the estrogen receptor (ER), making ovarian cancer preponderant among women undergoing various hormone therapy regimens.[13] Collectively, these reports suggest that EDCs can disrupt a wide range of reproductive functions through hormone-driven processes leading to elevated risks for a spectrum of diseases.

Although EDCs might enter into living organisms throughout their life, a particularly vulnerable time for exposure is the prenatal period, covering the entire voyage from gamete production, and fertilization through to intrauterine and post-natal development of progeny.[14] Developmental exposure to EDCs may lead to serious alterations that will persist into adult life, marking the field of endocrine disruption and female reproductive health a major area of concern. Bisphenol-A (BPA), a widely used plasticizer having xenoestrogenic properties, has been reported to be higher in circulation of women with PCOS. BPA reportedly potentially displaces a proportion of bound androgens or interferes with androgen catabolism leading to androgen excess in PCOS.[15] BPA can be transferred from the mother to child both through lactational and transplacental routes. Moreover, women undergoing infertility treatment has been demonstrated to have an inverse correlation between BPA levels in the urine with the number of quality eggs recovered in in vitro fertilization.[16] Recently, BPA per os (p.o.) has been shown to impact uterine morphology and androgen synthesis by interfering with the RNA methylation-related genes.[17] Notably, bisphenol S and fluorine-9-bisphenol, supposedly more environment friendly alternatives of BPA, have been shown to promote aberrant spindle formation in mature mouse and porcine oocytes.[18,19] Moreover, women exposed to di(2-ethylhexyl) phthalate (DEHP), a phthalate ester used in medical bags and tubing, may promote shorter pregnancy duration and increased incidences of miscarriage.[20] Numerous uterine defects that include precocious menstruation, irregular estrous cycle, and even short reproductive lifespan are well documented in mice treated with genistein-an isoflavone phytoestrogen, acting as a potent angiogenesis inhibitor.[21] Surprisingly, diethylstilbestrol, a nonsteroidal synthetic estrogen prescribed to pregnant women earlier, has been implicated in multi-generational effects in addition to reproductive tract dysfunction and poor pregnancy outcomes.[22] These evidences suggest a close association between developmental EDC exposure and adverse reproductive outcomes.

OOGENESIS: AN UPDATE ON AUTOCRINE/ PARACRINE ENTITIES

Oogenesis, the mother process of continuation of life, is under the tight surveillance of intra-ovarian factors aiming to develop fertilizable female gametes of utmost quality, a pre-requisite for perpetuation of the species. Ovary, the supreme player of this long run, relies on amalgamation of neuroendocrine, endocrine, and autocrine/paracrine factors.[23] Regulation of ovarian development and function is primarily controlled by the concert of hypothalamus-pituitary-gonadal axis, FSH, and LH being the sole players to stimulate synthesis of major gonadal sex steroids.[24] Binding of FSH to its cognate receptor (FSHR) at follicular cell surface promotes growth, while luteinizing hormone receptor (LHR) drives the oocyte toward final maturation.[24] In addition to gonadotropins and gonadal steroids, an increasing body of evidence in recent times have suggested the participation of a wide spectrum of locally produced non-steroidal peptide factors from the follicular theca-granulosa cell layer and the oocyte itself, thus forming a bi-directional regulatory network within the ovarian follicles. The notion of the oocyte as a passive partner receiving cues from its surrounding counterpart, the somatic follicular cells, has been replaced with the current perception of a complex yet regulated crosstalk between the granulosa- and oocyte-derived factors to orchestrate follicle development.[25-27] Recent few years have seen remarkable parallel contribution of a variety of growth factors, namely, insulin-like growth factors (IGFs),[28-30] epidermal growth factor (EGF) family members,[31-33] TGFβ superfamily members[34-36] in the ovary of mammals as well as fish. While IGFs role in modulation of steroidogenesis, oocyte maturation, follicular growth, and survival of growing oocytes has been reported widely, recent evidences recognized EGF family as yet another essential mediator of LH action on oocyte maturation in mammals. Moreover, oocyte-derived factors belonging to the TGFβ super family, primarily GDF9 and BMP15, are expressed during early stages of follicle development in the oocytes of mammalian species.[27,37,38]

BIDIRECTIONAL COMMUNICATION IN THE OVARY: A TARGET FOR EDC ACTION?

Numerous reports from mammalian models have depicted EDC modulation of follicular dynamics at the level of gonadotropin receptors. HPTE (bis-hydroxy methoxychlor), the active metabolite of the organochlorine pesticide methoxychlor, is a potent suppressor of progesterone and LH receptor in rat granulosa cells.[39] Further, dioxins such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can negatively influence both expression and stability of LH receptor transcripts in the cultured rat granulosa cells.[40] Not limited to mammals, BPA and diisononyl phthalate can modulate follicle-stimulating hormone receptor and LHR expression in zebrafish ovary.[41,42] Besides, adverse impact of EDC exposure on ovarian steroidogenesis and local (juxtacrine) factors has been reported earlier. Previously, serum estradiol and progesterone levels have been shown to undergo significant alteration in rats following DDT exposure.[43] Besides, negative impact of TCDD, a halogenated hydrocarbon, and on aryl hydrocarbon receptors has earlier been implicated in proliferation of granulosa cells, gonadotropin receptor expression, folliculogenesis, and intra-ovarian estrogen signaling.[44] Besides, EDCs such as polycyclic aromatic hydrocarbons, cadmium, and isoflavones impair ovarian steroidogenesis.[45-47] Not limited to mammals, the negative impact of BPA on transcript abundance of ovarian steroidogenic enzyme genes has been elucidated in rare minnow.[48] Recently, the ability of BPA to deregulate epigenetic mechanism leading to negative impact on female reproductive system in zebrafish has also been reported.[41] Besides, the deleterious impact of EDCs on ovarian growth factor expression and function has also been studied in recent past.[49,50] TCDD at environmentally relevant concentration can negatively modulate the interaction between IGF1 and FSH leading to decreased LH receptor mRNA abundance in human granulosa cells.[40] Moreover, studies from human luteinized granulosa cells have shown the up regulation of VEGF and IGF1 due to 1,1-dichloro-2,2-bis(p-chlorophenyl) ethylene, a metabolite of DDT, eventually leading to impaired ovarian steroidogenesis and infertility.[51] Notwithstanding, BPA attenuation of FSH-induced aromatase expression in human granulosa-like tumor cell line has been shown to involve peroxisome proliferator-activated receptor-gamma signaling and reduced expression of IGF1 and IGF receptors (IGFR).[50] Although, a group of researchers has demonstrated the altered mRNA expressions of IGF-1/IGFR on BPA exposure in a piscine model;[49] existing literature and research on fish ovarian growth factors are far from adequate and requires more research initiatives to unveil the molecular mechanism underlying EDC modulation of ovarian functions.

Although EDC-induced ovotoxicity has been demonstrated in several mammalian and non-mammalian models, our understanding of the relative importance of free radicals-induced stress, inflammatory or apoptotic response in promotion of deleterious biological changes in the ovary is limited. For gaining an in-depth knowledge about the extent of damage inflicted by various EDCs and their probable interference with diverse intra-ovarian signaling events, we need thorough research initiatives not only on complex dialogues between neuroendocrine, endocrine, and juxtacrine modulators but also on the redox balance, inflammatory, and apoptotic status of the ovarian follicles.

EDCS ON OVARIAN REDOX BALANCE, INFLAMMATORY, AND APOPTOTIC RESPONSE

Reactive oxygen species (ROS) are the by-products of oxidative metabolism and play crucial role in different cell signaling pathways. An imbalance between the production and clearance of ROS concomitant with attenuated antioxidant enzyme activity leads to the induction of elevated oxidative stress. Altered redox balance may lead to increased lipid peroxidation, heightened apoptosis, inflammation, and mitochondrial dysfunction at various target tissues.[52] Our recent data demonstrate disruption of metabolic homeostasis congruent with compromised inflammation, insulin signaling, and steatosis in BPA-exposed model freshwater teleost (Labeo bata)[53] indicating potential threat to aquatic species favored for human consumption. Importantly, EDC-mediated stress response may have malefic impact on biological macromolecules and intracellular machinery causing reproductive dysfunction. Moreover, EDCs may modulate immune cell dysfunction leading to acute or chronic inflammation. Importantly, EDC exposure may induce polycystic ovarian disease (PCOD), an inflammatory ovarian disorder. EDC induced oxidative stress can produce advanced glycation end products (AGEs), pro-inflammatory glycotoxin, that promote activation of pro-inflammatory condition and contributes significantly in ageing and female fertility. While it has been reported earlier that woman with PCOS have high serum AGEs,[54] exposure to persistent organic pollutants (POPs) such as polychlorinated biphenyls (PCBs), hexachlorobenzene, BPA, and phthalates leads to the formation of AGEs and inflammatory disorders including PCOD.[54] Moreover, congruent with altered GnRH pulsatality, high dose of BPA has been shown to elicit PCOD-like critical phenotypes characterized through increased plasma testosterone and 17β estradiol (E2) levels in rodents.[55] Apparently, EDC modulation of sex steroid receptors and steroidogenesis with hyperandrogenism may alter redox balance, inflammatory response, insulin sensitivity leading to PCOS, or inflammatory reproductive disorders. Furthermore, organophosphate and carbamate pesticides have potent role in endocrine disruption and reproductive disorders such as pelvic inflammatory disease that results into infertility, birth defects, and adverse pregnancy outcomes.[56] Another important inflammatory disorder, commonly referred as endometriosis, reveals growth of endometrial tissue and stroma outside the endometrial cavity and has been considered as one of the main reasons behind infertility in females. Certain EDCs such as persistent chemicals (metals, dioxin like compounds, polybrominated diphenyl ethers, and PCBs) and some non-persistent chemicals (BPA and phthalates) can exacerbate endometriosis such as lesions potentially through altered ER functions and elevated inflammatory signaling.[57] Interestingly, low-dose BPA exposure in human trophoblastic cell in vitro induces apoptosis and inflammation that leads to the etiopathology of preeclampsia,[58] an immune system dysfunction related disorder observed at certain stages of gestation due to hypertension where chronic immune activation takes place due to imbalance between CD4+ T cells and regulatory T cells as well as pro-inflammatory and anti-inflammatory cytokines.

While enrolment and survival of primordial follicles, selection or loss of follicles through apoptotic screening, luteogenesis, and atresia are of paramount importance for the regulation of well-orchestrated reproductive fitness, elevated oxidative stress has significant negative influence on apoptotic signaling, death of follicular cells, disrupted steroidogenesis, and premature reproductive aging. Congruent with increase in B-cell lymphoma 2 (BCL2)-associated X protein (Bax), transformation-related protein 53 expression but decreased level of Cyclin D2, BPA exposure may prevent follicular cell proliferation, growth of follicles leading to follicular atresia.[59] BPA-induced apoptosis in granulosa cells involves elevated Bax expression, caspase 3 activation, and attenuated G2-M1 transition promoting follicular atresia and luteal regression.[60,61] Besides, methylmercury may act as a potent EDC by lowering E2 level in circulation and promoting ovarian primary growth arrest as well as follicular apoptosis.[62] Moreover, dioxin and DEHP-induced granulosa cell apoptosis in mammals has been reported earlier.[63,64] Recently, Wang et al.[65] have shown the effect of another potent EDC soy isoflavones on ovarian apoptosis in rats marked by chromatin marginalization, condensation of the nucleus with increased of apoptotic bodies and vacuolation in granulosa cells. Congruent with reduced Bcl2 expression, elevated expression of Fas, caspase 3, caspase 8, and Bax has also been reported. More recently, BPA modulation of gonadotropin receptors, markers associated with steroidogenesis and growth factor expression along with altered ERα/ERβ homeostasis has been reported from our laboratory as a fallout of elevated oxidative/nitrosative stress, elicited inflammatory, and cell death modulators in the zebrafish ovary.[66] Clearly, the tight link between EDC mediated deregulated redox balance, inflammation, induction of apoptosis, and loss of ovarian homeostasis has been less investigated topic and need a steadfast approach in future.

EDC MEDIATED DISRUPTION OF ENERGY SENSING HOMEOSTASIS AND ULTIMATE OVARIAN MISFORTUNE

Oxidative stress mediated compromised mitochondrial functions leads to the disruption of energy sensing homeostasis mechanism. Overproduction of ROS, either through leakage of mitochondrial electron transport system or as the byproduct of metabolic pathways, and elevated oxidative stress led to activation of poly ADP (adenosine diphosphate)-ribose polymerase, a DNA repairing NAD+ consuming enzyme. Besides, ROS inactivation of NAM phosphoribosyltransferase enzyme, the rate limiting component in the mammalian NAD biosynthesis pathway, leads to downregulation of energy sensing marker Sirtuin (Silent mating type information regulation 2 homolog) (SIRT1), an NAD-dependent protein deacetylase that links transcriptional machinery to intracellular energetics and participates in the coordination of cellular functions such as cell cycle, response to DNA damage, metabolism, apoptosis, and autophagy.[67,68] SIRT1 is the regulatory component other than energy sensors such as adenosine monophosphate-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and has crucial role in the regulation of inflammation, apoptosis, and metabolism. Although the molecular mechanism is not yet clearly understood, SIRT modulation of reproductive parameters, for example, spermatogenesis, germ cell apoptosis and premature ovarian failure, or even maintenance of follicle reserve has been reported earlier.[69,70] Downregulation of SIRT1 results into different metabolic disorders such as disruption of lipid and glucose metabolism that results into insulin resistance, type 2 diabetes which also has negative influence on female reproductive fitness. Moreover, SIRT1 has important role in reproductive events such as folliculogenesis, steroidogenesis, spindle assembly, and chromosome alignment. SIRT1 regulation of folliculogenesis in rat ovary and FSH-mediated steroidogenesis in granulosa cells through Forkhead box O3 a and steroidogenic acute regulatory protein (StAR) have been reported earlier.[71,72] Moreover, proliferation and secretory activity of porcine granulosa cells is regulated by SIRT1 modulation of NF-κ and p53 function.[73] Besides, SIRT2 activation of histone H4K16 and α-tubulin may regulate metaphase II spindle assembly, chromosome alignment, and aging process in mouse oocytes.[74] Transcriptional activation of FoxO by AMPK signaling in nutrient-depleted conditions helps in the maintenance of reproductive activities.[75] Folliculogenesis, luteinization, progesterone secretion, and oxidative stress response are controlled by SIRT3 modulation of glutamate dehydrogenase, superoxide dismutase 1, catalase, 17beta-hydroxysteroid dehydrogenase 1, StAR, and human cytochrome P450 aromatase in human granulosa and cumulus cells.[76] Thus, alteration in energy sensors has profound negative impact on female fertility or reproductive fitness.

CONCLUSION

A new life developing within a mother is prone to high risk factors at its various levels of journey starting from preconception, pre-implantation, and fetal period followed by its arrival on earth. Urbanization, pollution, various therapeutic regimens, etc., are proving detrimental for female fertility, requiring utmost concern and attention. To deal and combat with these issues seriously, we need continuous research initiatives to unknot the complex dialogs between neuroendocrine, endocrine and juxtacrine modulators, the basis of reproductive endocrinology [Figure 2]. Accordingly, one can envisage proper assessment of endocrine disruptors with the ultimate goal of making healthy, viable, and superior quality eggs. Restricted production and/or phasing out their use coupled with large scale monitoring are important measures to clean up the environmental burden of EDCs. While, bioaccumulation of EDCs in food chain may pose serious threat to human health and reproductive fitness, more specifically gamete quality, transgenerational adversities, and development of metabolic disorder in both mother and child, microbial degradation of POPs as a component of mitigation strategy may be of significant influence in future.

A schematic representation of endocrine-disrupting chemical-induced oxidative stress and its influence on impaired ovarian homeostasis and female infertility (red and blue arrows signify negative and promoting influence, respectively).
Figure 2:
A schematic representation of endocrine-disrupting chemical-induced oxidative stress and its influence on impaired ovarian homeostasis and female infertility (red and blue arrows signify negative and promoting influence, respectively).

Declaration of patient consent

Patient’s consent not required as there are no patients in this study.

Financial support and sponsorship

SB and UM acknowledge financial support by DST (NO: DST/INSPIRE/03/2015/005022), New Delhi through award of INSPIRE Fellowship and University Grants Commission, India, for research fellowship, respectively. SM gratefully acknowledges ICAR, New Delhi (Grant No. NASF/ ABA- 6018/2016-17).

Conflicts of interest

There are no conflicts of interest.

References

  1. , . Ovarian follicular atresia In: , ed. Basic Gynecology-Some Related Issues. London: IntechOpen Limited; .
    [Google Scholar]
  2. , , , . Environmental endocrine disruptors: Effects on the human male reproductive system. Rev Endocr Metab Disord. 2015;16:341-57.
    [CrossRef] [PubMed] [Google Scholar]
  3. , , . The importance of steroid uptake and intracrine action in endometrial and ovarian cancers. Front Pharmacol. 2017;8:346.
    [CrossRef] [PubMed] [Google Scholar]
  4. , , . Nonsteroidal anti-inflammatory drugs and risk for ovarian and endometrial cancers in the Iowa Women's Health Study. Cancer Epidemiol Biomarkers Prev. 2010;19:435-42.
    [CrossRef] [Google Scholar]
  5. , , , . Strengths and weaknesses of in vitro assays for estrogenic and androgenic activity. Best Pract Res Clin Endocrinol Metab. 2006;20:15-33.
    [CrossRef] [Google Scholar]
  6. . Assessment of in vivo assays for endocrine disruption. Best Pract Res Clin Endocrinol Metab. 2006;20:35-43.
    [CrossRef] [Google Scholar]
  7. , , , , . Known and emerging factors modulating estrogenic effects of endocrine-disrupting chemicals. Environ Rev. 2014;22:87-98.
    [CrossRef] [Google Scholar]
  8. , . Effects of environmental endocrine disruptors on pubertal development. J Clin Res Pediatr Endocrinol. 2001;3:1-6.
    [CrossRef] [PubMed] [Google Scholar]
  9. . Hazard identification and risk assessment of endocrine disrupting chemicals with regard to developmental effects. Toxicology. 2002;181:367-70.
    [CrossRef] [Google Scholar]
  10. , , , , , , et al. Endocrine-disrupting chemicals: An endocrine society scientific statement. Endocr Rev. 2009;30:293-342.
    [CrossRef] [Google Scholar]
  11. . Endocrine-disrupting compounds and mammary gland development: Early exposure and later life consequences. Endocrinology. 2006;147:S18-24.
    [CrossRef] [PubMed] [Google Scholar]
  12. , . Endometriosis. Lancet. 2004;364:1789-99.
    [CrossRef] [Google Scholar]
  13. , , . P53 in endometrial cancer and its putative precursors: Evidence for diverse pathways of tumorigenesis. Hum Pathol. 1995;26:1268-74.
    [CrossRef] [Google Scholar]
  14. , , . Estrogens and epithelial ovarian cancer. Gynecol Oncol. 2004;94:25-32.
    [CrossRef] [PubMed] [Google Scholar]
  15. , . Polycystic ovary syndrome: Do endocrine-disrupting chemicals play a role? Semin Reprod Med. 2014;32:166-76.
    [CrossRef] [Google Scholar]
  16. , , . Effects of endocrine-disrupting chemicals on female reproductive health. Open Biotechnol J. 2016;10:54-75.
    [CrossRef] [Google Scholar]
  17. , , , , , , et al. Oral bisphenol-a regulates the development and function of reproductive system through differential expression of METTL3. J Ovarian Res. ;2020
    [CrossRef] [Google Scholar]
  18. , , , , , , et al. Acute low-dose bisphenol S exposure affects mouse oocyte quality. Reprod Toxicol. 2020;93:19-27.
    [CrossRef] [PubMed] [Google Scholar]
  19. , , , , , , et al. The toxic effects of fluorene-9-bisphenol on porcine oocyte in vitro maturation. Environ Toxicol. 2020;35:152-8.
    [CrossRef] [PubMed] [Google Scholar]
  20. , , , , , , et al. In utero exposure to di-(2-ethylhexyl) phthalate and duration of human pregnancy. Environ Health Perspect. 2003;111:1783-5.
    [CrossRef] [PubMed] [Google Scholar]
  21. , , . Adverse effects on female development and reproduction in CD-1 mice following neonatal exposure to the phytoestrogen genistein at environmentally relevant doses. Biol Reprod. 2005;73:798-806.
    [CrossRef] [PubMed] [Google Scholar]
  22. , . Effect of diethylstilbestrol on reproductive function. Fertil Steril. 1999;72:1-7.
    [CrossRef] [Google Scholar]
  23. . Insulin-like growth factors and fish reproduction. Biol Reprod. 2010;82:656-61.
    [CrossRef] [PubMed] [Google Scholar]
  24. , . Regulation of oocyte maturation in fish. Dev Growth Differ. 2008;50:S195-219.
    [CrossRef] [PubMed] [Google Scholar]
  25. , , , . Cellular basis for paracrine regulation of ovarian follicle development. Reproduction. 2001;121:647-53.
    [CrossRef] [PubMed] [Google Scholar]
  26. . Intrafollicular paracrine communication in the zebrafish ovary: The state of the art of an emerging model for the study of vertebrate folliculogenesis. Mol Cell Endocrinol. 2005;237:1-10.
    [CrossRef] [PubMed] [Google Scholar]
  27. , . Let's talk and grow together: A bidirectional communication between granulosa-and oocyte-derived factors in the ovary. Glob J Reprod Med. 2017;1:555569.
    [CrossRef] [Google Scholar]
  28. , . The role of the insulin-like growth factor (IGF) system in zebrafish (Danio rerio) ovarian development. Gen Comp Endocrinol. 2010;168:103-10.
    [CrossRef] [PubMed] [Google Scholar]
  29. , , . Expression and functional characterization of intrafollicular GH-IGF system in the zebrafish ovary. Gen Comp Endocrinol. 2016;232:32-42.
    [CrossRef] [PubMed] [Google Scholar]
  30. , , , , . IGFs mediate the action of LH on oocyte maturation in zebrafish. Mol Endocrinol. 2015;29:373-83.
    [CrossRef] [PubMed] [Google Scholar]
  31. , , , , , . EGF-like growth factors as mediators of LH action in the ovulatory follicle. Science. 2004;303:682-4.
    [CrossRef] [PubMed] [Google Scholar]
  32. , . Epidermal growth factor differentially regulates activin subunits in the zebrafish ovarian follicle cells via diverse signaling pathways. Mol Cell Endocrinol. 2012;361:133-42.
    [CrossRef] [PubMed] [Google Scholar]
  33. , . Human chorionic gonadotropin (hCG) induces MAPK3/1 phosphorylation in the zebrafish ovarian follicle cells independent of EGF/EGFR pathway. Gen Comp Endocrinol. 2013;188:251-7.
    [CrossRef] [PubMed] [Google Scholar]
  34. , . Epidermal growth factor and TGFα promote zebrafish oocyte maturation in vitro: Potential role of the ovarian activin regulatory system. Endocrinology. 2002;143:47-54.
    [CrossRef] [PubMed] [Google Scholar]
  35. , . TGF-beta superfamily members and ovarian follicle development. Reproduction. 2006;132:191-206.
    [CrossRef] [PubMed] [Google Scholar]
  36. , , , . Role of activin, transforming growth factor-beta and bone morphogenetic protein 15 in regulating zebrafish oocyte maturation. Comp Biochem Physiol A Mol Integr Physiol. 2009;153:18-23.
    [CrossRef] [PubMed] [Google Scholar]
  37. , , . Oocyte-somatic cell interactions during follicle development in mammals. Anim Reprod Sci 200482-83:431-46
    [CrossRef] [PubMed] [Google Scholar]
  38. , , , , , , et al. Regulation of follicular development and differentiation by intra-ovarian factors and endocrine hormones. Front Biosci (Landmrk Ed). 2019;24:983-93.
    [CrossRef] [Google Scholar]
  39. , , , , , . Effect of the methoxychlor metabolite HPTE on the rat ovarian granulosa cell transcriptome in vitro. Toxicol Sci. 2009;110:95-106.
    [CrossRef] [PubMed] [Google Scholar]
  40. , , , , . Effect of IGF-1 and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the expression of LH receptors during cell differentiation in cultured granulosa cells. Mol Cell Endocrinol. 2003;202:123-31.
    [CrossRef] [Google Scholar]
  41. , , , , , , et al. BPA-induced deregulation of epigenetic patterns: Effects on female zebrafish reproduction. Sci Rep. 2016;6:21982.
    [CrossRef] [PubMed] [Google Scholar]
  42. , , , , , , et al. Effects of diisononyl phthalate on Danio rerio reproduction. Environ Pollut. 2017;231:1051-62.
    [CrossRef] [PubMed] [Google Scholar]
  43. , , , , , , et al. Two-generation reproduction toxicity study in rats with 1,1,1-trichloro-2,2-bis(4-chlorophenyl)ethane (p,p'-DDT) Congenit Anom (Kyoto). 2006;46:105-14.
    [CrossRef] [PubMed] [Google Scholar]
  44. , , , , . Ovarian tumors in rats induced by chronic 2,3,7,8-tetrachlorodibenzo-p-dioxin treatment. Cancer Res. 2000;60:5414-9.
    [Google Scholar]
  45. , , . Endocrine-disrupting chemicals in ovarian function: Effects on steroidogenesis, metabolism and nuclear receptor signaling. Reproduction. 2011;142:633-46.
    [CrossRef] [PubMed] [Google Scholar]
  46. , , , , . Modulation of steroidogenic pathway in rat granulosa cells with subclinical Cd exposure and insulin resistance: An impact on female fertility. Biomed Res Int. 2014;2014:460251.
    [CrossRef] [PubMed] [Google Scholar]
  47. , , , , , , et al. Biochanin A affects steroidogenesis and estrogen receptor-β expression in porcine granulosa cells. Theriogenology. 2013;80:821-8.
    [CrossRef] [PubMed] [Google Scholar]
  48. , , , , , , et al. Effects of 17α-ethinylestradiol and bisphenol A on steroidogenic messenger ribonucleic acid levels in the rare minnow gonads. Aquat Toxicol. 2012;122:19-27.
    [CrossRef] [PubMed] [Google Scholar]
  49. , , . Bisphenol A in oocytes leads to growth suppression and altered stress performance in juvenile rainbow trout. PLoS One. 2010;5:e10741.
    [CrossRef] [PubMed] [Google Scholar]
  50. , , , . Peroxisome proliferator-activated receptor-γ mediates bisphenol A inhibition of FSH-stimulated IGF-1, aromatase, and estradiol in human granulosa cells. Environ Health Perspect. 2010;118:400-6.
    [CrossRef] [PubMed] [Google Scholar]
  51. , , . Influence of dichlorodiphenylchloroethylene on vascular endothelial growth factor and insulin-like growth factor in human and rat ovarian cells. Reprod Toxicol. 2007;24:359-64.
    [CrossRef] [PubMed] [Google Scholar]
  52. , , , , . Oxidative stress and BPA toxicity: An antioxidant approach for male and female reproductive dysfunction. Antioxidants (Basel). 2020;9:405.
    [CrossRef] [PubMed] [Google Scholar]
  53. , , , , , , et al. Bisphenol A-induced oxidative stress, hepatotoxicity and altered estrogen receptor expression in Labeo bata: Impact on metabolic homeostasis and inflammatory response. Ecotoxicol Environ Saf. 2020;202:110944.
    [CrossRef] [PubMed] [Google Scholar]
  54. , . Polycystic ovary syndrome and environmental toxins. Fertil Steril. 2016;106:948-58.
    [CrossRef] [PubMed] [Google Scholar]
  55. , , , . Neonatal exposure to bisphenol A and reproductive and endocrine alterations resembling the polycystic ovarian syndrome in adult rats. Environ Health Perspect. 2010;118:1217-22.
    [CrossRef] [PubMed] [Google Scholar]
  56. , , , . Evaluation of pelvic inflammatory disease potential in cholinesterase inhibitor pesticide-exposed females. Environ Sci Pollut Res Int. 2018;25:30818-25.
    [CrossRef] [PubMed] [Google Scholar]
  57. , , . Endocrine disrupting chemicals and endometriosis. Fertil Steril. 2016;106:959-66.
    [CrossRef] [PubMed] [Google Scholar]
  58. , , , , , . Urinary concentrations of bisphenol A and phthalate metabolites measured during pregnancy and risk of preeclampsia. Environ Health Perspect. 2016;124:1651-5.
    [CrossRef] [PubMed] [Google Scholar]
  59. , , . Bisphenol A inhibits follicle growth and induces atresia in cultured mouse antral follicles independently of the genomic estrogenic pathway. Biol Reprod. 2012;87:63.
    [CrossRef] [PubMed] [Google Scholar]
  60. , , , , , , et al. Bisphenol A induces apoptosis and G2-to-M arrest of ovarian granulosa cells. Biochem Biophs Res Commun. 2002;292:456-62.
    [CrossRef] [PubMed] [Google Scholar]
  61. , , , , , , et al. Bisphenol A exposure during adulthood causes augmentation of follicular atresia and luteal regression by decreasing 17β-estradiol synthesis via downregulation of aromatase in rat ovary. Environ Health Perspect. 2013;121:663-9.
    [CrossRef] [PubMed] [Google Scholar]
  62. , , . Increased ovarian follicular apoptosis in fathead minnows (Pimephales promelas) exposed to dietary methylmercury. Aquat Toxicol. 2006;79:49-54.
    [CrossRef] [PubMed] [Google Scholar]
  63. , , , . Dioxin perturbs, in a dose-and time-dependent fashion, steroid secretion, and induces apoptosis of human luteinized granulosa cells. Endocrinology. 1998;139:4373-9.
    [CrossRef] [PubMed] [Google Scholar]
  64. , , , , . Di-(2-ethylhcxyl) phthalate reduces progesterone levels and induces apoptosis of ovarian granulosa cell in adult female ICR mice. Environ Toxicol Pharmacol. 2012;34:869-75.
    [CrossRef] [Google Scholar]
  65. , , , , , , et al. Soy isoflavones administered to rats from weaning until sexual maturity affect ovarian follicle development by inducing apoptosis. Food Chem Toxicol. 2014;72:51-60.
    [CrossRef] [PubMed] [Google Scholar]
  66. , , , , , . Bisphenol A impairs reproductive fitness in zebrafish ovary: Potential involvement of oxidative/nitrosative stress, inflammatory and apoptotic mediators. Environ Pollut. 2020;19:115692.
    [CrossRef] [Google Scholar]
  67. , , . Crosstalk between poly(ADPribose) polymerase and sirtuin enzymes. Mol Aspects Med. 2013;34:1168-201.
    [CrossRef] [PubMed] [Google Scholar]
  68. , , , , , , et al. SIRT1 is a regulator of autophagy: Implications in gastric cancer progression and treatment. FEBS Lett. 2015;589:2034-42.
    [CrossRef] [PubMed] [Google Scholar]
  69. , , , , , , et al. Chronic exposure to low dose of bisphenol A impacts on the first round of spermatogenesis via SIRT1 modulation. Sci Rep. 2018;8:1-2.
    [CrossRef] [PubMed] [Google Scholar]
  70. , , , , , , et al. SIRT1 activator (SRT1720) improves the follicle reserve and prolongs the ovarian lifespan of diet-induced obesity in female mice via activating SIRT1 and suppressing mTOR signaling. J Ovarian Res. 2014;7:97.
    [CrossRef] [PubMed] [Google Scholar]
  71. , , , , , , et al. Resveratrol promotes expression of SIRT1 and StAR in rat ovarian granulosa cells: An implicative role of SIRT1 in the ovary. Reprod Biol Endocrinol. 2012;10:14.
    [CrossRef] [PubMed] [Google Scholar]
  72. , , , , , , et al. Obesity accelerates ovarian follicle development and follicle loss in rats. Metabolism. 2014;63:94-103.
    [CrossRef] [PubMed] [Google Scholar]
  73. , , , , . Interrelationships between sirtuin 1 and transcription factors p53 and NF-κB (p50/p65) in the control of ovarian cell apoptosis and proliferation. Cell Tissue Res. 2014;358:627-32.
    [CrossRef] [PubMed] [Google Scholar]
  74. , , , , , . Sirt2 functions in spindle organization and chromosome alignment in mouse oocyte meiosis. Faseb J. 2014;28:1435-45.
    [CrossRef] [PubMed] [Google Scholar]
  75. , . Regulation of reproduction and longevity by nutrient-sensing pathways. J Cell Biol. 2018;217:93-106.
    [CrossRef] [PubMed] [Google Scholar]
  76. , , , , , , et al. Sirtuin functions in female fertility: Possible role in oxidative stress and aging. Oxid Med Cell Longev. 2015;2015:659687.
    [CrossRef] [PubMed] [Google Scholar]
Show Sections