Elsevier

NeuroToxicology

Volume 54, May 2016, Pages 65-71
NeuroToxicology

Full length article
Sex-dependent effects of lead and prenatal stress on post-translational histone modifications in frontal cortex and hippocampus in the early postnatal brain

https://doi.org/10.1016/j.neuro.2016.03.016Get rights and content

Highlights

  • H3K9/14Ac and H3K9Me3 are suggested to be sexually dimorphic histone modifications.

  • H3K9/14Ac and H3K9Me3 were examined for modification by lead and prenatal stress.

  • Sex-dependent differences were seen in both histone modifications.

  • Effects of lead and stress were sex and brain-region-dependent.

Abstract

Environmental lead (Pb) exposure and prenatal stress (PS) are co-occurring risk factors for impaired cognition and other disorders/diseases in adulthood and target common biological substrates in the brain. Sex-dependent differences characterize the neurochemical and behavioral responses of the brain to Pb and PS and sexually dimorphic histone modifications have been reported to occur in at-risk brain regions (cortex and hippocampus) during development. The present study sought to examine levels and developmental timing of sexually dimorphic histone modifications (i.e., H3K9/14Ac and H3K9Me3) and the extent to which they may be altered by Pb ± PS. Female C57/Bl6 mice were randomly assigned to receive distilled deionized drinking water containing 0 or 100 ppm Pb acetate for 2 months prior to breeding and throughout lactation. Half of the dams in each group were exposed to restraint stress (PS, three restraint sessions in plastic cylindrical devices 3×/day at for 30 min/day (1000, 1300, and 1600 h)) from gestational day 11–19 or no stress (NS). At delivery (PND0) and postnatal day 6 (PND6), pups were euthanized and frontal cortex and hippocampus were removed, homogenized, and assayed for levels of H3K9/14Ac and H3K9Me3. Sex-dependent differences in both levels of histone modifications as well as the developmental trajectory of changes in these levels were observed in both structures and these parameters were differentially affected by Pb ± PS in a sex and brain-region-dependent manner. Disruptions of these epigenetic processes by developmental Pb ± PS may underlie some of the sex-dependent neurobehavioral differences previously observed in these animals.

Introduction

Many aspects of both normal and pathological brain functioning exhibit important yet poorly understood sex differences (Wizemann and Pardue, 2001, McCarthy et al., 2012). Sex differences have been described for various cognitive functions, as well as for anxiety and stress responses, outcomes from traumatic brain injury, risk and outcomes from various neurodegenerative diseases, and the risk for developing various emotional and affective disorders (see McCarthy et al., 2012 for review). Sex-dependent differences also characterize the response of the brain to the neurotoxicant lead (Pb) and to prenatal stress (PS). Developmental Pb exposure and PS are important co-occurring risk factors for a variety of adverse health outcomes in adulthood as well as for various cognitive/behavioral problems during childhood, with effects expressed differently in males and females (Virgolini et al., 2008a, Virgolini et al., 2008b, Cory-Slechta et al., 2010, Cory-Slechta et al., 2012, Giesbrecht et al., 2015, Wainstock et al., 2015, Zohar et al., 2015).

The precise molecular mechanisms underlying the effects of developmental Pb exposure and of PS on the brain are not yet completely known, nor are the molecular underpinnings for the sex-related differences in the responses of the brain to such risk factors. However, it seems likely that epigenetic mechanisms are involved (Auger and Auger, 2011). Epigenetic outcomes resulting from environmental perturbations are frequently found to be sex-dependent, particularly in relation to neuroendocrine-acting systems. These sex differences likely reflect reported sex differences in such aspects of epigenetic programming as levels of the DNA methyltransferases (DNMTs) (McCarthy et al., 2009, Kolodkin and Auger, 2011) or DNA methylation patterns, and expression of methyl-binding proteins and co-repressor proteins (McCarthy et al., 2009). The existence of such sex differences early in development has been suggested to be critical to brain differentiation (Auger and Auger, 2011). These differences also establish a different biological substrate for each sex on which environmental stressors, including chemical exposures, then act.

In the mammalian brain, chromatin-level modifications play pivotal roles in coordinating developmental programs. Gene regulation during development is regulated by the binding of a variety of regulatory proteins to gene promoter regions and through epigenetic modification of chromatin by post-translational histone modifications and DNA methylation. The role of epigenetics in determining sex differences in the brain (McCarthy et al., 2009) can also be significantly influenced by the period of development during which these environmental factors act. For example, higher levels of DNMT3a mRNA expression in female amygdala were reported at postnatal day 1, but had disappeared by postnatal day 10, with these changes related to sex differences in steroid exposure levels (Kolodkin and Auger, 2011). These dynamics in the developmental trajectory could influence the extent to which environmental risk factors affect the epigenome in a sex-dependent manner.

Epigenetic alterations may occur in the germline as well as in somatic cells (Skinner et al., 2008), both of which are modifiable by environmental stimuli such as steroid hormones and endocrine-disrupting chemicals (McCarthy et al., 2009, Gore, 2008) and potentially, by environmental factors such as developmental Pb exposure and PS. It is possible that an interaction between genes and sex hormones during development may make a variety of brain regions, including the frontal cortex and hippocampus, which are particularly sensitive to the effects of Pb and PS (Cory-Slechta et al., 1998, Cory-Slechta et al., 1999, Berger et al., 2002, Barros et al., 2004, Virgolini et al., 2008a, Martinez-Tellez et al., 2009, Rossi-George et al., 2011, Neal et al., 2012, Stansfield et al., 2012, Baranowska-Bosiacka et al., 2013, Hu et al., 2014, Li et al., 2014, Li et al., 2015) differentially susceptible to epigenetic alterations, such as post-translational histone modifications, that are associated with modulation of transcriptional regulation.

As sexually dimorphic histone modifications have been reported to occur in cortex and hippocampus during development (Tsai et al., 2009), the present study examined the extent to which these histone modifications may be influenced by Pb and/or PS and thus contribute to the sex-dependent differences in the patterns of effects observed in response to these environmental risk factors alone and in combination (Cory-Slechta et al., 2004b, Cory-Slechta et al., 2008, Cory-Slechta et al., 2010, Virgolini et al., 2006, Virgolini et al., 2008a, Virgolini et al., 2008b, Rossi-George et al., 2011). The goal of the present study was to examine levels and developmental timing of sexually dimorphic histone modifications, H3K9/14Ac (associated with gene activation) and H3K9Me3 (associated with gene silencing) (Tsai et al., 2009) and the extent to which they may be altered by Pb and/or PS (Pb ± PS). Modifications of these histone marks by Pb ± PS could have wide-ranging effects on gene transcription as well as later behavioral/cognitive functions.

Section snippets

Animals and lead exposure and prenatal stress

The use of animals was in compliance with NIH Guidelines for the Care and Use of Laboratory Animals and the study was approved by the institutional animal care and use committee at the University of Rochester School of Medicine. Two-week-old female C57/Bl6 mice (Jackson Laboratories, Bar Harbor ME) were randomly assigned to receive distilled deionized water drinking containing 0 or 100 ppm Pb acetate for 2 months prior to breeding and throughout lactation. Standard rat chow diet (Lab Diet,

Dam and offspring weights and litter size

Body weights of dams were collected at 3 different time points over the course of gestation (gestational days 4, 10 and 17). No significant differences in body weight were observed until day 17, when a main effect of stress was found (F = 4.53, p < 0.0001). This reflected a small decrement in body weight of stressed dams (approximately 3 gm), with mean ± SE values of 32.73 ± 0.62, 29.75 ± 0.69, 33.00 ± 0.61 and 30.19 ± 0.64 g for the 0-NS, 0-PS, 100-NS and 100-PS groups, respectively.

Litter weight at birth did

Discussion

Epigenetic changes in response to a stressor can persist long after the stressor has ended and can underlie persisting functional changes in the brain. In this study, we examined the effects of Pb and PS on H3K9/14Ac and H3K9Me3 levels in two brain regions known to be sensitive to the effects of Pb and PS, frontal cortex and hippocampus, in male and female offspring at PND0 and PND6, two time points previously suggested to show different sexually dimorphic effects on these two histone marks (

Conclusions

In conclusion, sex differences in histone modifications that manifest early in development may be involved in the expression of sex-dependent differences in cognitive functions observed later in life. Disruptions of these processes by developmental Pb exposure, PS, and the combination of these factors may underlie some of the sex-dependent neurobehavioral differences observed in these animals in adulthood (Virgolini et al., 2008a, Virgolini et al., 2008b, Virgolini et al., 2006, Cory-Slechta et

Conflicts of interest

The authors have no conflicts of interest to declare.

Acknowledgement

This work was supported by NIH grant R01 ES021534 (Cory-Slechta, D.A.; Schneider, J.S. multi-PIs).

References (41)

  • A.P. Neal et al.

    Enhanced nitric oxide production during lead (Pb(2)(+)) exposure recovers protein expression but not presynaptic localization of synaptic proteins in developing hippocampal neurons

    Brain Res.

    (2012)
  • A. Rossi-George et al.

    Interactions of lifetime lead exposure and stress: behavioral; neurochemical and HPA axis effects

    Neurotoxicology

    (2011)
  • G. Tabibnia et al.

    Sex difference and laterality in the volume of mouse dentate gyrus granule cell layer

    Brain Res.

    (1999)
  • M.B. Virgolini et al.

    Permanent alterations in stress responsivity in female offspring subjected to combined maternal lead exposure and/or stress

    Neurotoxicology

    (2006)
  • M.B. Virgolini et al.

    CNS effects of developmental Pb exposure are enhanced by combined maternal and offspring stress

    Neurotoxicology

    (2008)
  • M.B. Virgolini et al.

    Influence of low level maternal Pb exposure and prenatal stress on offspring stress challenge responsivity

    Neurotoxicology

    (2008)
  • A.P. Auger et al.

    Epigenetic turn ons and turn offs: chromatin reorganization and brain differentiation

    Endocrinology

    (2011)
  • V.G. Barros et al.

    Early adoption modifies the effects of prenatal stress on dopamine and glutamate receptors in adult rat brain

    J. Neurosci. Res.

    (2004)
  • M.A. Berger et al.

    Long-term effects of prenatal stress on dopamine and glutamate receptors in adult rat brain

    Neurochem. Res.

    (2002)
  • S.J. Clapcote et al.

    Simplex PCR assay for sex determination in mice

    Biotechniques

    (2005)
  • Cited by (0)

    View full text