Review
Epigenetic effects of endocrine-disrupting chemicals on female reproduction: An ovarian perspective

https://doi.org/10.1016/j.yfrne.2010.06.003Get rights and content

Abstract

The link between in utero and neonatal exposure to environmental toxicants, such as endocrine-disrupting chemicals (EDCs) and adult female reproductive disorders is well established in both epidemiological and animal studies. Recent studies examining the epigenetic mechanisms involved in mediating the effects of EDCs on female reproduction are gathering momentum. In this review, we describe the developmental processes that are susceptible to EDC exposures in female reproductive system, with a special emphasis on the ovary. We discuss studies with select EDCs that have been shown to have physiological and correlated epigenetic effects in the ovary, neuroendocrine system, and uterus. Importantly, EDCs that can directly target the ovary can alter epigenetic mechanisms in the oocyte, leading to transgenerational epigenetic effects. The potential mechanisms involved in such effects are also discussed.

Introduction

Female fertility disorders are becoming increasingly prevalent and an emerging women’s health concern [13], [63], [74]. These disease outcomes reflect the interplay between individuals’ genetic composition and the environment. Since the genetic composition at the population level is relatively slow to change, the significant role of our dramatically changed environment, in the last century, has to be considered. The impact of the environment, especially during development is highly relevant to adult health and is highlighted by the Barker hypothesis, also known as the developmental origins of health and disease (DoHAD) [11], [12], [105]. Although the Barker hypothesis was originally based on nutritional deprivation in utero and cardiovascular or metabolic disorders in adulthood, its perspective can be applied to developmental exposure to environmental endocrine-disrupting chemicals (EDCs) and fertility disorders in females during adulthood [74].

EDCs are synthetic and natural compounds in the environment that interfere with (i.e., mimic and/or antagonize) the actions of endogenous hormones and disrupt the functions of the endocrine system. The United States Environmental Protection Agency (EPA) has identified hundreds of EDCs that have estrogenic, anti-estrogenic, or anti-androgenic activities [13], [65], [322]. Potentially there are many compounds that either already exist in the environment or are newly introduced, whose effects have not yet been studied in the female reproductive system. EDCs include several groups of chemicals such as insecticides, herbicides and fungicides; plastics and plasticizers; surfactants; industrial chemicals such as polychloro biphenyls (PCBs), polybrominated biphenyls (PBBs), and dioxins; flame retardants; pharmaceuticals; and phytoestrogens such as genistein and coumestrol. There are numerous ways in which exposures to EDCs can occur. While a few compounds are for medicinal use, exposures to the vast majority of these EDCs are through unintended exposure. For example, DDT, which was banned in 1972, was intended for use as a pesticide against disease-bearing insects. It was heavily used for many years and eventually it was found that wildlife, especially bird populations were drastically diminished due to consumption of DDT tainted fish [38].

The overall fertility rate of women aged 15–44 years in the United States dropped 44% between 1960 and 2002 [121]. Lifestyle choices may have been a major contributor of this decline as this study included all women in this age group. However, according to the data from the National Survey for Family Growth, the ‘impaired fecundity rate’ was increased from 11% to 15% between 1982 and 2002 [117]. Furthermore, the incidence of female reproductive disorders such as early puberty, irregular menstrual cycles, endometriosis, premature ovarian failure, and polycystic ovarian disorder is increasing in parallel with the increasing number of EDCs in the environment [63].

Human epidemiological studies suggest that there is a clear association between developmental EDC exposure and adverse health outcomes in females. For example, high maternal serum concentration of pp′-DDT significantly reduces the probability of pregnancy for their daughters [54]. In addition, there is an association between potential exposure to other EDCs, such as bisphenol A (BPA) and genistein and female reproductive problems. Levels of BPA in blood are associated with a variety of conditions in women including endometrial hyperplasia, recurrent miscarriages, sterility, and polycystic ovary syndrome (PCOS) [240], [333], [335]. An example is that of the inverse correlation between the levels of BPA in urine of women undergoing infertility treatment with the number of eggs recovered in in vitro fertilization (IVF) clinics and serum estradiol levels [219]. High BPA exposure is also associated with chromosomal abnormalities [349]. More alarming recent evidence suggests that there is an association between intrauterine and early life exposure to soy formula and uterine fibroids in adulthood [71]. The same study also showed a similar association between in utero diethylstilbestrol (DES) exposure and fibroids.

Direct evidence from unintentional human exposure to EDCs and observations with wildlife species or lab animals shows that developmental exposure to EDCs singly or in mixtures can cause similar types of consequences as described above [115], [116], [143], [155], [176], [229]. Further evidence from lab animal studies for the effects of developmental exposure to EDCs on the female reproductive system is provided in Section 4. The importance of epigenetics (vs. genetics) in disease susceptibility and phenotypic differences are becoming very clear [99], [161], [178], [341]. Epigenetic mechanisms that play a role in the delayed effects of developmental exposure to EDCs are also presented in Section 4 and Table 1. From a public health perspective, the most serious concern emerging from these studies is that developmental exposure to adverse environmental EDCs may lead to effects in subsequent generation(s) via epigenetic mechanisms, broadly termed transgenerational epigenetic effects (see Section 5 for more details).

Section snippets

Epigenetics

The term “epigenetics” was first introduced in the 1940s by Conrad H. Waddington to describe the developmental program where genes determine the individual’s phenotype and internal and external environmental cues are also taken into consideration [152], [133]. Although the word epigenetic was originally derived from an older terminology “epigenesis” – referring to an embryological concept, it literally means “beyond or above genetics” [332]. However, the current use of the term is somewhat

Development of female reproductive system

Successful female reproductive function requires three components that work in close communication with each other: the ovary, the neuroendocrine system or hypothalamus pituitary gonadal (HPG) axis, and the reproductive tract, including the uterus and oviduct. Therefore, EDC exposure that can affect any one these components can impact the others as well. A recent review provides in-depth information on ovarian development and function and on the factors that play a role in these processes [89].

EDCs and their effects on the female reproductive system

In this section, we focus on selected EDCs, methoxychlor (MXC), DES, genistein, and BPA that have been shown in recent animal studies to adversely affect the female reproductive system, especially through epigenetic mechanisms (Table 1). Some of these effects are transgenerational as well (see Section 5). Each EDC section was organized in the following order: brief description of the EDC, human epidemiological studies, experimental animal studies, physiological effects on the ovary, HPG, and

Transgenerational epigenetic effects of EDCs

Transgenerational epigenetic effects, including those that are induced by EDCs have been discussed in recent reviews [297], [356]. Epidemiological studies have suggested that transgenerational epigenetic effects occur in humans. In the Dutch famine of the 1944, not only did the fetuses that were exposed to under-nutrition in utero suffer from cardiovascular or metabolic disorders during their adulthood, but the grand-children also showed a lower birth weight [199]. In an independent study, an

Conclusions and future directions

Data from the reviewed studies collectively suggest that perinatal EDC exposure affects adult ovaries and female reproductive tissues, leading to reproductive dysfunction, supporting the DoHAD concept. In addition, these effects are mediated primarily by steroid hormone receptors, especially the ERs: via ERβ in the ovary and ERα in the uterus and perhaps both in the hypothalamus and pituitary [7], [60], [153], [156], [358]. Furthermore, most of the EDCs discussed in this article lead to MOF (a

Acknowledgments

The authors wish to thank Dr. Kathy Manger for her assistance in the preparation of this manuscript. The research results from our laboratory described in this review were supported in part by the NIEHS Grant ES013854 and the NIEHS sponsored UMDNJ Center for Environmental Exposures and Disease Grant NIEHS P30ES005022.

References (359)

  • F.F. Costa

    Non-coding RNAs: new players in eukaryotic biology

    Gene

    (2005)
  • F.F. Costa

    Non-coding RNAs, epigenetics and complexity

    Gene

    (2008)
  • J.F. Couse et al.

    Estrogen receptor-alpha mediates the detrimental effects of neonatal diethylstilbestrol (DES) exposure in the murine reproductive tract

    Toxicology

    (2004)
  • D.A. Crain et al.

    Female reproductive disorders: the roles of endocrine-disrupting compounds and developmental timing

    Fertil. Steril.

    (2008)
  • A.M. Cummings et al.

    Methoxychlor affects the decidual cell response of the uterus but not other progestational parameters in female rats

    Toxicol. Appl. Pharmacol.

    (1987)
  • A.M. Cummings et al.

    Antifertility effect of methoxychlor in female rats: dose- and time-dependent blockade of pregnancy

    Toxicol. Appl. Pharmacol.

    (1989)
  • A. Dhasarathy et al.

    The MBD protein family-reading an epigenetic mark?

    Mutat. Res.

    (2008)
  • W.J. Dieckmann et al.

    Does the administration of diethylstilbestrol during pregnancy have therapeutic value?

    Am. J. Obstet. Gynecol.

    (1953)
  • V.P. Eroschenko et al.

    Estradiol or methoxychlor stimulates estrogen receptor (ER) expression in uteri

    Reprod. Toxicol.

    (1996)
  • J.G. Falls et al.

    Genomic imprinting: implications for human disease

    Am. J. Pathol.

    (1999)
  • A.P. Feinberg et al.

    DNA methylation and genomic imprinting: insights from cancer into epigenetic mechanisms

    Semin. Cancer Biol.

    (2002)
  • H.B. Adewale et al.

    Neonatal bisphenol-A exposure alters rat reproductive development and ovarian morphology without impairing activation of gonadotropin-releasing hormone neurons

    Biol. Reprod.

    (2009)
  • E.D. Albrecht, G.J. Pepe, Estrogen regulation of placental angiogenesis and fetal ovarian development during primate...
  • C. Allegrucci et al.

    Epigenetics and the germline

    Reproduction

    (2005)
  • M.D. Anway et al.

    Epigenetic transgenerational actions of endocrine disruptors and male fertility

    Science

    (2005)
  • M.D. Anway et al.

    Endocrine disruptor vinclozolin induced epigenetic transgenerational adult-onset disease

    Endocrinology

    (2006)
  • P. Avner et al.

    X-chromosome inactivation: counting, choice and initiation

    Nat. Rev. Genet.

    (2001)
  • J. Bakker et al.

    Alpha-fetoprotein protects the developing female mouse brain from masculinization and defeminization by estrogens

    Nat. Neurosci.

    (2006)
  • H.S. Bal

    Effect of methoxychlor on reproductive systems of the rat

    Proc. Soc. Exp. Biol. Med.

    (1984)
  • D.J. Barker

    Fetal origins of coronary heart disease

    BMJ

    (1995)
  • D.J. Barker

    Developmental origins of adult health and disease

    J. Epidemiol. Commun. Health

    (2004)
  • J. Barrett, S. Gonzalez, H. Sarantis, J. Varshavsky, Girl, Disrupted: Hormone Disruptors and Women’s Reproductive...
  • M.S. Bartolomei

    Genomic imprinting: employing and avoiding epigenetic processes

    Genes Dev.

    (2009)
  • M.A. Bedell et al.

    DNA rearrangements located over 100 kb 5′ of the steel (Sl)-coding region in steel-panda and steel-contrasted mice deregulate Sl expression and cause female sterility by disrupting ovarian follicle development

    Genes Dev.

    (1995)
  • H.W. Bennetts et al.

    The oestrogenic effects of subterranean clover (trifolium subterraneum); uterine maintenance in the ovariectomised ewe on clover grazing

    Aust. J. Exp. Biol. Med. Sci.

    (1951)
  • P. Berta et al.

    Genetic evidence equating SRY and the testis-determining factor

    Nature

    (1990)
  • R.B. Billiar et al.

    Up-regulation of alpha-inhibin expression in the fetal ovary of estrogen-suppressed baboons is associated with impaired fetal ovarian folliculogenesis

    Biol. Reprod.

    (2003)
  • A. Bird

    DNA methylation patterns and epigenetic memory

    Genes Dev.

    (2002)
  • J. Blatt et al.

    Ovarian carcinoma in an adolescent with transgenerational exposure to diethylstilbestrol

    J. Pediatr. Hematol. Oncol.

    (2003)
  • K. Block et al.

    In utero diethylstilbestrol (DES) exposure alters Hox gene expression in the developing mullerian system

    Faseb J.

    (2000)
  • C. Borgeest et al.

    Methoxychlor may cause ovarian follicular atresia and proliferation of the ovarian epithelium in the mouse

    Toxicol. Sci.

    (2002)
  • C. Brede et al.

    Increased migration levels of bisphenol A from polycarbonate baby bottles after dishwashing, boiling and brushing

    Food Addit. Contam.

    (2003)
  • T.G. Bredfeldt et al.

    Xenoestrogen-induced regulation of EZH2 and histone methylation via estrogen receptor signaling to PI3K/AKT

    Mol. Endocrinol.

    (2010)
  • R.M. Brena et al.

    Toward a human epigenome

    Nat. Genet.

    (2006)
  • J. Brennan et al.

    One tissue, two fates: molecular genetic events that underlie testis versus ovary development

    Nat. Rev. Genet.

    (2004)
  • R. Bretveld et al.

    Time to pregnancy among female greenhouse workers

    Scand. J. Work Environ. Health

    (2006)
  • K.L. Britt et al.

    Estrogen actions on follicle formation and early follicle development

    Biol. Reprod.

    (2004)
  • J.R. Brody et al.

    Histologic, morphometric, and immunocytochemical analysis of myometrial development in rats and mice: I. Normal development

    Am. J. Anat.

    (1989)
  • J.G. Bromer et al.

    Hypermethylation of homeobox A10 by in utero diethylstilbestrol exposure: an epigenetic mechanism for altered developmental programming

    Endocrinology

    (2009)
  • A.M. Calafat et al.

    Urinary concentrations of bisphenol A and 4-nonylphenol in a human reference population

    Environ. Health Perspect.

    (2005)
  • Cited by (131)

    View all citing articles on Scopus
    View full text