Elsevier

Neurotoxicology and Teratology

Volume 47, January–February 2015, Pages 66-79
Neurotoxicology and Teratology

Prenatal arsenic exposure alters the programming of the glucocorticoid signaling system during embryonic development

https://doi.org/10.1016/j.ntt.2014.11.006Get rights and content

Highlights

  • The glucocorticoid system was assessed in placenta and fetal brain following prenatal arsenic.

  • Prenatal arsenic reduced GR expression in the fetal brain.

  • GR reduction may be due to a prolonged increase in 11βHSD2 levels.

  • Prenatal arsenic alters glucocorticoid programming.

Abstract

The glucocorticoid system, which plays a critical role in a host of cellular functions including mood disorders and learning and memory, has been reported to be disrupted by arsenic. In previous work we have developed and characterized a prenatal moderate arsenic exposure (50 ppb) model and identified several deficits in learning and memory and mood disorders, as well as alterations within the glucocorticoid receptor signaling system in the adolescent mouse. In these present studies we assessed the effects of arsenic on the glucocorticoid receptor (GR) pathway in both the placenta and the fetal brain in response at two critical periods, embryonic days 14 and 18. The focus of these studies was on the 11β-hydroxysteroid dehydrogenase enzymes (11β-HSD1 and 11β-HSD2) which play a key role in glucorticoid synthesis, as well as the expression and set point of the GR negative feedback regulation. Negative feedback regulation is established early in development. At E14 we found arsenic exposure significantly decreased expression of both protein and message in brain of GR and the 11β-HSD1, while 11β-HSD2 enzyme protein levels were increased but mRNA levels were decreased in the brain. These changes in brain protein continued into the E18 time point, but mRNA levels were no longer significantly altered. Placental HSD11B2 mRNA was not altered by arsenic treatment but protein levels were elevated at E14. GR placental protein levels were decreased at E18 in the arsenic exposed condition. This suggests that arsenic exposure may alter GR expression levels as a consequence of a prolonged developmental imbalance between 11β-HSD1 and 11β-HSD2 protein expression despite decreased 11HSDB2 mRNA. The suppression of GR and the failure to turn down 11β-HSD2 protein expression during fetal development may lead to an altered set point for GR signaling throughout adulthood. To our knowledge, these studies are the first to demonstrate that gestational exposure to moderate levels of arsenic results in altered fetal programming of the glucocorticoid system.

Introduction

Early developmental and adult exposure to arsenic is associated with a multitude of health problems, including cognitive (Rodriguez-Barranco et al., 2013, Rosado et al., 2007, von Ehrenstein et al., 2007, Wasserman et al., 2004), cardiovascular (McClintock et al., 2014, Moon et al., 2012, Wu et al., 2014), metabolic (Chen et al., 2009, Joshi and Shrestha, 2010, Lu et al., 2014) and metastatic (Ferreccio et al., 2013, Mostafa and Cherry, 2013, Wong et al., 1998, Xie et al., 2014) disorders that are manifested across the lifespan. The developmental origins of health and disease (DOHaD) theory (Gluckman et al., 2007, McMullen and Mostyn, 2009, Wadhwa et al., 2009) proposes that susceptibilities to chronic diseases are, in part, determined by physiologic changes initiated in utero in response to an altered prenatal environment. The theory posits that an adverse intra-uterine environment programs the development of the fetus to accommodate for this toxic environment by altering gene expressions. These adaptations are likely through epigenetic modifications. One system, the glucocorticoid signaling pathway has been demonstrated to be programmed in response to prenatal environments (Bolten et al., 2013, Conradt et al., 2013, Khulan and Drake, 2012, Lukaszewski et al., 2013, Reynolds, 2013) and these programming changes have been observed into adulthood (Goldstein et al., 2014, Khalife et al., 2013), including the transmission of the altered programming across generations (Luo et al., 2014). Further, there is support for the suggestion that altered glucocorticoid receptor (GR) programming could impact adult health (Brunton and Russell, 2011, Harris and Seckl, 2011, Merlot et al., 2008, Sarkar et al., 2008).

While there are many systems affected by arsenic, few have been implicated to play a role in all the various health problems associated with exposure. The GR signaling system, has been linked with cognitive deficits (Rodriguez et al., 2011), cardiovascular (Santos and Joles, 2012), metabolic (Sarr et al., 2012, Spencer, 2012), immune (Bellavance and Rivest, 2014) and metastatic diseases (Kitchin and Wallace, 2008, Schmitz et al., 2009). Several studies have demonstrated that arsenic produces specific disruptions in glucocorticoid transcriptional activity and steroid receptor function (Ahir et al., 2013, Barr et al., 2009, Bodwell et al., 2004, Davey et al., 2007, Gosse et al., 2014, Hamilton et al., 1998, Kaltreider et al., 2001, Shaw et al., 2007). Many of these studies have utilized cell lines or cultured cells, which provide a great deal of mechanistic information regarding the impact of toxins on specific cellular regulators but precludes the ability to assess the impact of arsenic on developmental programming including the interaction between maternal and fetal physiology.

Our own work using an in vivo mouse model exposed prenatally to 50 ppb arsenic in drinking water has found alterations in the levels of glucocorticoid signaling (Goggin et al., 2012, Martinez-Finley et al., 2009, Martinez et al., 2008), in the hippocampus from adolescent offspring. While it is clear that arsenic, both in vivo and in vitro, produces significant changes in the glucocorticoid system, the impact of arsenic on the glucocorticoid system in both the developing fetus and the placenta have not been explored. Fetal levels of glucocorticoid are regulated by both the fetal and the placental tissues through the developmentally regulated isozymes, 11β-hydroxysteroid dehydrogenases (11β-HSD) 1 and 2, which interconvert active and inactive cortisol (corticosterone). In the mouse, placental expression of 11β-HSD2 is elevated until about embryonic day 13 (E13), when it serves to inactivate maternal corticosterone and protects the developing fetus from the high glucocorticoid activation. Expression of 11β-HSD1 begins around E14 and peaks at E18 (Brown et al., 1996, Speirs et al., 2004). 11β-HSD1 is a reductase, which activates corticosterone and regenerates the glucocorticoid increasing fetal corticosterone levels. The developmental expression and levels of these isozymes are seen as being a critical regulation point for hypothalamic–pituitary–adrenal (HPA) axis negative feedback (Brunton and Russell, 2011, Chapman et al., 2013, Reichardt and Schutz, 1996). Maternal exposure to arsenic may alter the timing of 11β-HSD isozyme expression levels in an attempt to adapt to the influence of arsenic on GR signaling. Additionally, arsenic readily crosses the placenta (Caumette et al., 2007, Fei et al., 2013, He et al., 2007, Vahter, 2009) and might directly alter expression and levels of the GR signaling complex within the developing fetal brain. Little is known about the impact of arsenic in utero on the developing fetal brain and placenta.

In the present study, we evaluated the effect of 50 ppb (50 μg/L) arsenic on the glucocorticoid signaling system in both the fetal brain and placenta at two development time-points (E14 and E18) to identify critical shifts in the expression of signaling components which might impact the GR programming and later adult stress responding. The embryonic period from E14 to E18 is critical for the expression shift between 11β-HSD2 and 11β-HSD1 and for development of the negative feedback system for the centrally regulated HPA axis. If prenatal arsenic alters the level and/or the timing of the expression of these proteins which regulate the glucocorticoid response, then it may alter the central regulation of HPA axis programming and affect stress responding throughout life.

Section snippets

Prenatal arsenic exposure paradigm

All procedures were conducted in accordance with protocols approved by the Institutional Animal Care and Use Committee at the University of New Mexico. Animals were maintained in a 22 °C vivarium on a reverse light/dark cycle with lights on at 2000 and ad libitum access to water and food. Exposure to arsenic was performed as previously described during all three trimesters of development (Martinez et al., 2008, Tyler et al., 2014). Briefly, singly housed female C57BL/6J mice (Jackson

Levels of arsenic species (As5, MMA and DMA) were elevated in the brains from arsenic exposed fetuses relative to controls

Both the reduced form of inorganic arsenic, trivalent (As (III)) and the oxidized pentavalent (As (V)) form, accumulate in tissues. Metabolism of arsenic by oxidative methylation leads to the formation of monomethylarsenicals (MMA III and MMA V) and dimethylated arsenicals (DMA III and DMA V). The levels of the trioxide form, arsenite, were not significantly different between the two exposure groups (Table 2).

Prenatal arsenic did not alter the number of fetuses, crown to rump length or placental weight at the specific gestational time points tested

While our earlier work did not observe differences in the number of live pups or in

Discussion

Several researchers have identified the GR as a key target for arsenic effects (Ahir et al., 2013, Barr et al., 2009, Bodwell et al., 2004, Gosse et al., 2014, Simons et al., 1990, Stancato et al., 1993). The GR pathway sits at a critical interaction and integration point to play a role in all of the key arsenic associated effects from: inflammation to cognition, and from immunity to neoplasia. Using a comparative toxicogemonics data base, Ahir and colleagues (Ahir et al., 2013) identified the

Conclusions

In our previous work assessing the impact of perinatal arsenic exposures on the function of the central and peripheral HPA system, we found significant support to conclude that arsenic exposure in utero resulted in a suppression of the glucocorticoid negative feedback system due to a decrease in GR and 11 HSD1 in the hippocampus. It was unclear if this deficit in HPA function was due to an indirect effect of arsenic resulting in a compensatory response to altered fetal HPA development or if the

Funding and disclosure

This work was supported by a grant from the National Institutes of Health: NIEHS RO1ES019583-01 (AMA). All authors declare no conflicts of interest.

The following are the Supplementary data related to this article.

Transparency document

Transparency document.

Acknowledgments

The authors thank Kevin Caldwell for helpful discussions and criticisms of the work. The authors received computer support from Michael Riblett. Dr. Abdulmehdi Ali performed the arsenic speciation analysis. The work was supported with funding 1RO1ES019583-01 (AMA) from the National Institute of Environmental Health Sciences.

Conceived and designed experiments: AMA and KEC. Performed experiments: all authors. Analyzed the data: all authors. Wrote the paper: all authors.

References (117)

  • S.L. Goggin et al.

    Perinatal exposure to 50 ppb sodium arsenate induces hypothalamic–pituitary–adrenal axis dysregulation in male C57BL/6 mice

    Neurotoxicology

    (2012)
  • J.M. Goldstein et al.

    Disruption of fetal hormonal programming (prenatal stress) implicates shared risk for sex differences in depression and cardiovascular disease

    Front Neuroendocrinol

    (2014)
  • M.V. Govindan et al.

    Human glucocorticoid receptor gene promotor-homologous down regulation

    J Steroid Biochem Mol Biol

    (1991)
  • A. Harris et al.

    Glucocorticoids, prenatal stress and the programming of disease

    Horm Behav

    (2011)
  • A.P. Harris et al.

    Mineralocorticoid and glucocorticoid receptor balance in control of HPA axis and behaviour

    Psychoneuroendocrinology

    (2013)
  • M.C. Holmes et al.

    The role of 11beta-hydroxysteroid dehydrogenases in the brain

    Mol Cell Endocrinol

    (2006)
  • M.F. Hughes et al.

    Accumulation and metabolism of arsenic in mice after repeated oral administration of arsenate

    Toxicol Appl Pharmacol

    (2003)
  • Y. Jin et al.

    Distribution of speciated arsenicals in mice exposed to arsenite at the early life

    Ecotoxicol Environ Saf

    (2010)
  • J.E. Kalinyak et al.

    Tissue-specific regulation of glucocorticoid receptor mRNA by dexamethasone

    J Biol Chem

    (1987)
  • C.E. Keegan et al.

    Expression of corticotropin-releasing hormone transgenes in neurons of adult and developing mice

    Mol Cell Neurosci

    (1994)
  • B. Khulan et al.

    Glucocorticoids as mediators of developmental programming effects

    Best Pract Res Clin Endocrinol Metab

    (2012)
  • K.T. Kitchin et al.

    The role of protein binding of trivalent arsenicals in arsenic carcinogenesis and toxicity

    J Inorg Biochem

    (2008)
  • M.A. Lukaszewski et al.

    The hypothalamic POMC mRNA expression is upregulated in prenatally undernourished male rat offspring under high-fat diet

    Peptides

    (2013)
  • H. Luo et al.

    Prenatal caffeine ingestion induces transgenerational neuroendocrine metabolic programming alteration in second generation rats

    Toxicol Appl Pharmacol

    (2014)
  • E.J. Martinez et al.

    Moderate perinatal arsenic exposure alters neuroendocrine markers associated with depression and increases depressive-like behaviors in adult mouse offspring

    Neurotoxicology

    (2008)
  • E.J. Martinez-Finley et al.

    Learning deficits in C57BL/6 J mice following perinatal arsenic exposure: consequence of lower corticosterone receptor levels?

    Pharmacol Biochem Behav

    (2009)
  • E.J. Martinez-Finley et al.

    Reduced expression of MAPK/ERK genes in perinatal arsenic-exposed offspring induced by glucocorticoid receptor deficits

    Neurotoxicol Teratol

    (2011)
  • T.R. McClintock et al.

    Association between arsenic exposure from drinking water and hematuria: results from the Health Effects of Arsenic Longitudinal Study

    Toxicol Appl Pharmacol

    (2014)
  • E. Merlot et al.

    Prenatal stress, fetal imprinting and immunity

    Brain Behav Immun

    (2008)
  • C. Pavlides et al.

    Effects of mineralocorticoid and glucocorticoid receptors on long-term potentiation in the CA3 hippocampal field

    Brain Res

    (1999)
  • R.M. Reynolds

    Glucocorticoid excess and the developmental origins of disease: two decades of testing the hypothesis—2012 Curt Richter Award Winner

    Psychoneuroendocrinology

    (2013)
  • J.S. Rodriguez et al.

    Prenatal betamethasone exposure has sex specific effects in reversal learning and attention in juvenile baboons

    Am J Obstet Gynecol

    (2011)
  • M. Rodriguez-Barranco et al.

    Association of arsenic, cadmium and manganese exposure with neurodevelopment and behavioural disorders in children: a systematic review and meta-analysis

    Sci Total Environ

    (2013)
  • S. Rosewicz et al.

    Mechanism of glucocorticoid receptor down-regulation by glucocorticoids

    J Biol Chem

    (1988)
  • M.S. Santos et al.

    Early determinants of cardiovascular disease

    Best Pract Res Clin Endocrinol Metab

    (2012)
  • M.V. Schmidt et al.

    Regulation of the developing hypothalamic–pituitary–adrenal axis in corticotropin releasing hormone receptor 1-deficient mice

    Neuroscience

    (2003)
  • S.S. Simons et al.

    Arsenite and cadmium(II) as probes of glucocorticoid receptor structure and function

    J Biol Chem

    (1990)
  • M. Styblo et al.

    Identification of methylated metabolites of inorganic arsenic by thin-layer chromatography

    J Chromatogr B Biomed Appl

    (1995)
  • C.R. Tyler et al.

    Fluoxetine treatment ameliorates depression induced by perinatal arsenic exposure via a neurogenic mechanism

    Neurotoxicology

    (2014)
  • C. Abou-Seif et al.

    Tissue specific epigenetic differences in CRH gene expression

    Front Biosci

    (2012)
  • B.K. Ahir et al.

    Systems biology and birth defects prevention: blockade of the glucocorticoid receptor prevents arsenic-induced birth defects

    Environ Health Perspect

    (2013)
  • S.M. Barlow et al.

    The relation between maternal restraint and food deprivation, plasma corticosterone, and induction of cleft palate in the offspring of mice

    Teratology

    (1975)
  • F.D. Barr et al.

    Disruption of histone modification and CARM1 recruitment by arsenic represses transcription at glucocorticoid receptor-regulated promoters

    PLoS ONE

    (2009)
  • M.A. Bellavance et al.

    The HPA — immune axis and the immunomodulatory actions of glucocorticoids in the brain

    Front Immunol

    (2014)
  • J.E. Bodwell et al.

    Arsenic at very low concentrations alters glucocorticoid receptor (GR)-mediated gene activation but not GR-mediated gene repression: complex dose–response effects are closely correlated with levels of activated GR and require a functional GR DNA binding domain

    Chem Res Toxicol

    (2004)
  • R.W. Brown et al.

    The ontogeny of 11 beta-hydroxysteroid dehydrogenase type 2 and mineralocorticoid receptor gene expression reveal intricate control of glucocorticoid action in development

    Endocrinology

    (1996)
  • K.K. Caldwell et al.

    Prenatal alcohol exposure is associated with altered subcellular distribution of glucocorticoid and mineralocorticoid receptors in the adolescent mouse hippocampal formation

    Alcohol Clin Exp Res

    (2014)
  • R.N. Carter et al.

    Hypothalamic–pituitary–adrenal axis abnormalities in response to deletion of 11beta-HSD1 is strain-dependent

    J Neuroendocrinol

    (2009)
  • G. Caumette et al.

    Monitoring the arsenic and iodine exposure of seaweed-eating North Ronaldsay sheep from the gestational and suckling periods to adulthood by using horns as a dietary archive

    Environ Sci Technol

    (2007)
  • K. Chapman et al.

    11Beta-hydroxysteroid dehydrogenases: intracellular gate-keepers of tissue glucocorticoid action

    Physiol Rev

    (2013)
  • Cited by (0)

    View full text