Elsevier

NeuroToxicology

Volume 33, Issue 1, January 2012, Pages 23-36
NeuroToxicology

Neonatal Bisphenol A exposure alters sexually dimorphic gene expression in the postnatal rat hypothalamus

https://doi.org/10.1016/j.neuro.2011.11.002Get rights and content

Abstract

Developmental exposure to Bisphenol A (BPA), a component of polycarbonate and epoxy resins, has been purported to adversely impact reproductive function in female rodents. Because neonatal life is a critical window for the sexual dimorphic organization of the hypothalamic–pituitary–gonadal (HPG) axis, interference with this process could underlie compromised adult reproductive physiology. The goal of the present study was to determine if neonatal BPA exposure interferes with sex specific gene expression of estrogen receptor alpha (ERα), ER beta (ERβ) and kisspeptin (Kiss1) in the anterior and mediobasal hypothalamus. Long Evans (LE) neonatal rats were exposed to vehicle, 10 μg estradiol benzoate (EB), 50 mg/kg BPA or 50 μg/kg BPA by subcutaneous injection daily from postnatal day 0 (PND 0) to PND 2. Gene expression was assessed by in situ hybridization on PNDs 4 and 10. Within the anterior hypothalamus ERα expression was augmented by BPA in PND 4 females, then fell to male-typical levels by PND 10. ERβ expression was not altered by BPA on PND 4, but significantly decreased or eliminated in both sexes by PND 10. Kiss1 expression was diminished by BPA in the anterior hypothalamus, especially in females. There were no significant impacts of BPA in the mediobasal hypothalamus. Collectively, BPA effects did not mirror those of EB. The results show that neonatal hypothalamic ER and Kiss1 expression is sensitive to BPA exposure. This disruption may alter sexually dimorphic hypothalamic organization and underlie adult reproductive deficiencies. Additionally, the discordant effects of EB and BPA indicate that BPA likely disrupts hypothalamic organization by a mechanism other than simply acting as an estrogen mimic.

Highlights

► The hypothalamus undergoes sexual dimorphism in neonatal life. ► Hormones are critical for this process therefore it may be vulnerable to endocrine disruption. ► Neonatal exposure to BPA altered the sex specific expression of estrogen receptors. ► Neonatal exposure to BPA altered the sex specific expression of Kiss1. ► These gene expression changes may underlie reproductive deficiencies that emerge later in life.

Introduction

BPA was initially developed as a possible synthetic estrogen (Dodds and Lawson, 1936), and is now a high volume production component of polycarbonate plastics, epoxy resins, dental sealants, thermal receipts and other products (Biedermann et al., 2010, Vandenberg et al., 2007). Human exposure is nearly ubiquitous, with urinary levels higher in children than adults (Calafat et al., 2008). The health impacts of BPA exposure remain controversial but growing evidence suggests that it has the potential to impose adverse outcomes on reproductive (Beronius et al., 2010, Cabaton et al., 2011, Howdeshell et al., 1999, Vandenberg et al., 2009) cardiovascular (Pant et al., 2011) and metabolic (Groff, 2010, Newbold, 2010) health. The hypothalamus is an integral part of each of these systems and thus conceivably a central target through which BPA could induce widespread effects across multiple organ systems. We and others have shown that BPA exposure can alter the sex specific organization of hypothalamic regions in the murine brain known to be important for coordinating gonadotropin release and sexual behavior, most notably the anteroventral periventricular nucleus of the hypothalamus (AVPV) (Patisaul et al., 2006, Patisaul et al., 2007, Rubin et al., 2006) and the arcuate nucleus (ARC) (Patisaul et al., 2009). Collectively these observations support the hypothesis that disruption of hypothalamic organization during critical windows of development could underlie a suite of neuroendocrine BPA effects.

Although long considered weakly estrogenic, the specific mechanisms by which BPA interacts with molecular and cellular targets within the hypothalamus and elsewhere are not clearly established (vom Saal et al., 2007). Classically, BPA is thought to disrupt genomic pathways mediated by the two primary forms of the nuclear estrogen receptor (ER); ER alpha (ERα) and ER beta (ERβ). Compared to estradiol, BPA has a binding affinity approximately 10,000–100,000 fold weaker for both ER isoforms (Andersen et al., 1999, Barkhem et al., 1998, Blair et al., 2000, Gould et al., 1998) but appears to bind each with relatively equal affinity (Kuiper et al., 1998) indicating that it has the potential to interact with either.

It is well established that estrogen is masculinizing during perinatal development in the murine brain, and that exposure to even low levels of estrogen or xenoestrogens during this time can permanently alter neuroendocrine pathways critical for mediating steroid negative feedback, gonadotropin release, energy homeostasis and sexual behavior (Amateau et al., 2004, Bader et al., 2011, Faulds et al., 2011, Gore, 2008, Simerly, 2002). We have recently shown that the expression of ERα and ERβ is sexually dimorphic within numerous subregions of the neonatal rat hypothalamus (Cao and Patisaul, 2011). For example, within the AVPV, ERβ expression is higher in males on the day of birth while ERα expression is higher in females, suggesting that the two isoforms may play different functional roles in the sexual differentiation of this region. This sex difference is transient, and eliminated within 48 h, suggesting that if BPA exposure during only the first two days of life alters expression levels, estrogen sensitivity during this critical period of sexual differentiation may be impacted. Here we tested the hypothesis that neonatal BPA exposure can affect the sex specific hypothalamic expression of ERα and ERβ across postnatal development, focusing specifically on the AVPV, ARC and two other subregions (the medial preoptic area (MPOA) and the ventrolateral division of the ventromedial nucleus (VMNvl)) known to be essential for the neuroendocrine control of gonadotropin release and other aspects of female reproductive physiology, metabolic regulation and behavior (Nance, 1976, Pfaff and Keiner, 1973, Pfaff et al., 1994, Simerly, 2002). Although previous studies have explored similar hypotheses in a variety of species, they primarily looked at adult expression in only a single sex and either employed a different exposure window (Ceccarelli et al., 2007) focused on only one hypothalamic area (Mahoney and Padmanabhan, 2010, Monje et al., 2007), or employed techniques with less anatomical resolution such as RT-PCR (Khurana et al., 2000, Monje et al., 2007). Thus, the present studies build on this prior work by (1) characterizing the specific hypothalamic subregions vulnerable to disruption in both sexes (2) identifying this disruption early in postnatal development and (3) more carefully identifing the critical window of exposure for ER disruption by BPA. Collectively, these studies test the hypothesis that perturbation of ER expression during the neonatal period of hypothalamic sexual differentiation could be a mechanism by which BPA induces a wide range of effects, some of which may not emerge until adulthood.

We also sought to determine if BPA exposure can disrupt the expression of Kiss1, a gene which codes for the kisspeptin family of peptides. Kisspeptin is now recognized to be the primary “gatekeeper” of gonadotropin release and essential for initiating pubertal onset (Oakley et al., 2009, Pineda et al., 2010). It also plays a role in energy homeostasis and is a putative effector of leptin actions on gonadotropin releasing (GnRH) neurons (Castellano et al., 2010). There are two populations of Kiss1 neurons in the murine brain, an anterior group which extends through the AVPV into the MPOA (a region now collectively referred to as the rostral periventricular area of the third ventricle (RP3V)) and a mediobasal hypothalamic group confined to the ARC. In adults, the RP3V population is sexually dimorphic, with females having higher expression levels than males (Kauffman et al., 2007), while the ARC population is not (Losa et al., 2010). These observations suggest that the anterior population participates in steroid positive feedback on gonadotropin release, while the posterior population plays a role in steroid negative feedback (Oakley et al., 2009, Pineda et al., 2010). Nearly all Kiss1 neurons in the RP3V and ARC co-express ERα, and a subset of RP3V Kiss1 neurons co-express ERβ (Smith et al., 2005, Smith et al., 2006) suggesting that disruption of ER expression within this neuronal phenotype could affect its sex specific ontogeny and function. Gestational and/or postnatal BPA exposure has been shown to alter Kiss1 mRNA expression within whole hypothalamic micropunches (Navarro et al., 2009, Xi et al., 2010), and RP3V Kiss1-immunoreactivity (Bai et al., 2011, Panzica et al., 2009) during peripuberty and adulthood. Although RP3V Kiss1 expression is not apparent in either sex until approximately the second week of life (Cao and Patisaul, 2011, Clarkson et al., 2009) we have shown that neonatal exposure to estrogen or an ERα selective agonist (Bateman and Patisaul, 2008, Patisaul et al., 2009) can defeminize kisspeptin signaling pathways in the adult RP3V, suggesting that the first few days of life may be a critical window of BPA vulnerability, and that this disruption may be apparent prior to puberty. In contrast to the RP3V, Kiss1 mRNA in the ARC is robustly expressed from PND 0 through PND 19, with higher levels in females than males within the first week of life (Cao and Patisaul, 2011). This indicates that Kiss1 expression may be influenced by neonatal estrogen and vulnerable to endocrine disruption by a chemical like BPA. Here we examined whether neonatal BPA exposure affects the sexual dimorphic ontogeny of Kiss1 mRNA expression in the RP3V and ARC.

The present study is the first to precisely characterize when, where and how neonatal BPA exposure affects sexual dimorphic ER and Kiss1 gene expression in the postnatal rat brain. Collectively, the results reveal the potential for BPA to disregulate the differential expression of ERα, ERβ and Kiss1 during a critical window of hypothalamic organization, findings which will help elucidate the mechanisms by which BPA can induce persistent effects across neuroendocrine systems.

Section snippets

Animal care, neonatal exposure and tissue collection

Pups were born to timed pregnant Long Evans rats (n = 13; Charles River, Raleigh, NC), housed at the Biological Resource Facility at North Carolina State University (NCSU) under a 14:10 h light:dark cycle (lights on at 07:00 h) at 23 °C and 50% average relative humidity. Animals were housed in thoroughly washed polysulfone caging with woodchip bedding, and fed a semi-purified, phytoestrogen-free diet ad libitum (AIN-93G, Test Diet, Richmond, IN) to minimize exposure to exogenous BPA, phytoestrogens,

Summary of results

A summary of the overall results is provided in Table 1. In the anterior hypothalamus, ER expression was higher in females than males by PND 10 in the MPOA but equivalent to male expression in the AVPV. Neonatal exposure to EB eliminated the sex differences primarily by reducing ER expression in females. BPA effects were dose and age dependent. On PND 4, ERα expression was augmented in both sexes of the HBPA group, but, by PND 10, female expression was lower compared to the unexposed controls.

Discussion

The present study provides comprehensive evidence that neonatal exposure to BPA alters early postnatal, sexually dimorphic expression of ERα, ERβ and Kiss1 mRNA in the rat hypothalamus, particularly in the RP3V region. Disruption of ER expression during this time period may have lifelong consequences because it is the age at which the hypothalamus is undergoing steroid hormone directed sexual differentiation. Perturbation of ER expression by BPA presumably alters sensitivity to endogenous

Conflict of interest statement

None.

Acknowledgements

This work is supported by NIEHS grant RO1 ES016001 to HBP. We thank Karina Todd for assistance with animal care and facilitating ISHH.

References (87)

  • A.S. Kauffman

    Sexual differentiation and the Kiss1 system: hormonal and developmental considerations

    Peptides

    (2009)
  • M.M. Mahoney et al.

    Developmental programming: impact of fetal exposure to endocrine-disrupting chemicals on gonadotropin-releasing hormone and estrogen receptor mRNA in sheep hypothalamus

    Toxicol Appl Pharmacol

    (2010)
  • L. Monje et al.

    Exposure of neonatal female rats to bisphenol A disrupts hypothalamic LHRH pre-mRNA processing and estrogen receptor alpha expression in nuclei controlling estrous cyclicity

    Reprod Toxicol

    (2010)
  • T. Nagao et al.

    Reproductive function in rats exposed neonatally to bisphenol A and estradiol benzoate

    Reprod Toxicol

    (1999)
  • B.M. Nugent et al.

    Hormonally mediated epigenetic changes to steroid receptors in the developing brain: implications for sexual differentiation

    Horm Behav

    (2011)
  • H.B. Patisaul et al.

    Immediate and residual effects of tamoxifen and ethynylestradiol in the female rat hypothalamus

    Brain Res

    (2003)
  • H.B. Patisaul et al.

    Neonatal genistein or bisphenol-A exposure alters sexual differentiation of the AVPV

    Neurotoxicol Teratol

    (2006)
  • H.B. Patisaul et al.

    Differential disruption of nuclear volume and neuronal phenotype in the preoptic area by neonatal exposure to genistein and bisphenol-A

    Neurotoxicology

    (2007)
  • H.B. Patisaul et al.

    Impact of neonatal exposure to the ERalpha agonist PPT, bisphenol-A or phytoestrogens on hypothalamic kisspeptin fiber density in male and female rats

    Neurotoxicology

    (2009)
  • H.B. Patisaul et al.

    Regulation of estrogen receptor beta mRNA in the brain: opposite effects of 17β-estradiol and the phytoestrogen, coumestrol

    Brain Res Mol Brain Res

    (1999)
  • D.W. Pfaff et al.

    GnRH neurons and other cellular and molecular mechanisms for simple mammalian reproductive behaviors

    Recent Prog Horm Res

    (1994)
  • J.A. Taylor et al.

    No effect of route of exposure (oral; subcutaneous injection) on plasma bisphenol A throughout 24 h after administration in neonatal female mice

    Reprod Toxicol

    (2008)
  • L.N. Vandenberg et al.

    Human exposure to bisphenol A (BPA)

    Reprod Toxicol

    (2007)
  • F.S. vom Saal et al.

    Chapel Hill bisphenol A expert panel consensus statement: integration of mechanisms, effects in animals and potential to impact human health at current levels of exposure

    Reprod Toxicol

    (2007)
  • 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)
  • M. Aihara et al.

    Induction of persistent diestrus followed by persistent estrus is indicative of delayed maturation of tonic gonadotropin-releasing systems in rats

    Biol Reprod

    (1989)
  • S.K. Amateau et al.

    Brain estradiol content in newborn rats: sex differences, regional heterogeneity, and possible de novo synthesis by the female telencephalon

    Endocrinology

    (2004)
  • H.R. Andersen et al.

    Comparison of short-term estrogenicity tests for identification of hormone-disrupting chemicals

    Environ Health Perspect

    (1999)
  • M.I. Bader et al.

    Comparative assessment of estrogenic responses with relevance to the metabolic syndrome and to menopausal symptoms in wild-type and aromatase-knockout mice

    J Steroid Biochem Mol Biol

    (2011)
  • Y. Bai et al.

    Increase of anteroventral periventricular kisspeptin neurons and generation of E2-induced LH-surge system in male rats exposed perinatally to environmental dose of bisphenol-A

    Endocrinology

    (2011)
  • T. Barkhem et al.

    Differential response of estrogen receptor alpha and estrogen receptor beta to partial estrogen agonists/antagonists

    Mol Pharmacol

    (1998)
  • H.L. Bateman et al.

    Disrupted female reproductive physiology following neonatal exposure to phytoestrogens or estrogen specific ligands is associated with decreased GnRH activation and kisspeptin fiber density in the hypothalamus

    Neurotoxicology

    (2008)
  • S. Biedermann et al.

    Transfer of bisphenol A from thermal printer paper to the skin

    Anal Bioanal Chem

    (2010)
  • R.M. Blair et al.

    The estrogen receptor relative binding affinities of 188 natural and xenochemicals: structural diversity of ligands

    Toxicol Sci

    (2000)
  • N.J. Cabaton et al.

    Perinatal exposure to environmentally relevant levels of bisphenol A decreases fertility and fecundity in CD-1 mice

    Environ Health Perspect

    (2011)
  • A.M. Calafat et al.

    Exposure of the U.S. population to bisphenol A and 4-tertiary-octylphenol: 2003–2004

    Environ Health Perspect

    (2008)
  • J. Cao et al.

    Sexually dimorphic expression of hypothalamic estrogen receptors alpha and beta and kiss1 in neonatal male and female rats

    J Comp Neurol

    (2011)
  • F.A. Champagne et al.

    Maternal care associated with methylation of the estrogen receptor-alpha1b promoter and estrogen receptor-alpha expression in the medial preoptic area of female offspring

    Endocrinology

    (2006)
  • J. Clarkson et al.

    Postnatal development of an estradiol-kisspeptin positive feedback mechanism implicated in puberty onset

    Endocrinology

    (2009)
  • J. Clarkson et al.

    Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons

    Endocrinology

    (2006)
  • E.C. Dodds et al.

    Synthetic estrogenic agents without the phenanthrene nucleus

    Nature

    (1936)
  • D.C. Dolinoy et al.

    Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development

    Proc Natl Acad Sci USA

    (2007)
  • F. Farabollini et al.

    Effects of perinatal exposure to bisphenol A on sociosexual behavior of female and male rats

    Environ Health Perspect

    (2002)
  • Cited by (82)

    View all citing articles on Scopus
    View full text