Elsevier

Neuroscience

Volume 410, 1 July 2019, Pages 274-292
Neuroscience

Research Article
Sex Differences in Neuroplasticity- and Stress-Related Gene Expression and Protein Levels in the Rat Hippocampus Following Oxycodone Conditioned Place Preference

https://doi.org/10.1016/j.neuroscience.2019.04.047Get rights and content

Highlights

  • Hippocampal gene expression and protein levels vary depending on sex after oxycodone (Oxy) conditioned place preference (CPP)

  • After Oxy CPP, Npy mRNA expression decreases but a subset of NPY-labeled cells increases in the dentate gyrus (DG) in females

  • In CA3, plasticity and stress markers differentially increase in Oxy CPP females (ARC, Crhr2) and males (Arc, CRHR1)

  • After Oxy CPP, signaling kinases (Akt1, pAKT, pMAPK) change in opposite directions in females and males in CA1 and DG

  • Differences in signaling, kinase, and stress markers suggest sex-specific hippocampal pathways for opioid-associated learning

Abstract

Prescription opioid abuse is a serious public health issue. Recently, we showed that female and male Sprague–Dawley rats acquire conditioned place preference (CPP) to the mu opioid receptor agonist oxycodone. Anatomical analysis of the hippocampus from these rats unveiled sex differences in the opioid system in a way that would support excitation and opiate associative learning processes especially in females. In this study, we examined the expression and protein densities of opioid, plasticity, stress and related kinase and signaling molecules in the hippocampus of female and male rats following oxycodone CPP. Oxycodone CPP females have: a) increases in ARC (activity regulated cytoskeletal-associated protein)-immunoreactivity (ir) in CA3 pyramidal cells; b) decreases in Npy (neuropeptide Y) gene expression in the medial hippocampus but higher numbers of NPY-containing hilar interneurons compared to males; c) increases in Crhr2 (corticotropin releasing factor receptor 2) expression in CA2/3; d) increases in Akt1 (AKT serine/threonine kinase 1) expression in medial hippocampus; and e) decreases in phosphorylated MAPK (mitogen activated protein kinase)-ir in CA1 and dentate gyrus. Oxycodone CPP males have: a) increases in Bdnf (brain derived-neurotrophic factor) expression, which is known to be produced in granule cells, relative to females; b) elevated Mapk1 expression and pMAPK-ir in the dentate hilus which harbors newly generated granule cells; and c) increases in CRHR1-ir in CA3 pyramidal cell soma. These sex-specific changes in plasticity, stress and kinase markers in hippocampal circuitry parallel previously observed sex differences in the opioid system after oxycodone CPP.

Introduction

Prescription opioid abuse has increased dramatically over the past two decades and has become a serious public health issue (CDC, 2015). Notably, drug overdose rates involving synthetic opioids and heroin have dramatically increased in women (VanHouten et al., 2019). The influence of sex in opioid addiction processes has been challenging to elucidate, as it is difficult to separate the role of genes, environment, and multidrug use in humans. However, the finding that women may have altered sensitivity to morphine over the menstrual cycle (Ribeiro-Dasilva et al., 2011) suggests that ovarian hormones could be involved in addictive processes. Indeed, rodent studies have indicated that the hormonal milieu likely plays a part in opioid addiction (Becker et al., 2017). For example, fluctuations in circulating estrogen levels over the estrous cycle in rats have been reported to alter the patterns of heroin self-administration (Lacy et al., 2016).

Neural circuits involved in associative memory formation and the encoding of motivational incentives are critically involved in the pathway to drug addiction for both sexes (Koob and Volkow, 2010). In rodents, opioid signaling in the CA3 region of the hippocampus is implicated in associative learning and spatial memory (Meilandt et al., 2004, Kesner and Warthen, 2010). Additionally, a type of opioid-mediated long-term potentiation (LTP) has been demonstrated in mossy fiber-CA3 synapses from proestrus (high estrogen) female rats, but not diestrus (low estrogen) female or male rats (Harte-Hargrove et al., 2015), suggesting that opioid associative learning processes could be heightened in females in particular hormonal states.

Recently, we have shown that both female and male Sprague–Dawley rats acquire conditioned place preference (CPP) to the mu opioid receptor (MOR) agonist oxycodone (Ryan et al., 2018). Notably, anatomical analysis of the hippocampus from these rats unveiled sex differences in the opioid system in a way that would bolster excitation and opiate associative learning processes to a greater degree in females compared to males (Ryan et al., 2018). In particular, delta opioid receptors (DOR) redistributed within mossy fiber-CA3 synapses in both oxycodone-injected (Oxy) females and males in a manner similar to that demonstrated to be important for opioid-mediated LTP in proestrus females (Harte-Hargrove et al., 2015, Ryan et al., 2018). Additionally, MORs and DORs rearranged in hilar interneurons in Oxy-females in a configuration that could enhance granule cell disinhibition using two different circuits: 1) plasmalemmal associated MORs increased in parvalbumin (PARV)-labeled dendrites (Ryan et al., 2018) which are known to inhibit granule cell soma (Drake et al., 2007); 2) plasmalemmal associated DORs increased on GABAergic interneuron dendrites (Ryan et al., 2018) which are known to contain neuropeptide Y (NPY) and are important for promoting lateral perforant pathway (lpp) LTP (Sperk et al., 2007). However, whether these changes in hippocampal opioid receptor trafficking also are accompanied by changes in gene expression is unknown.

Thus, the goal of this study was to investigate the effect of oxycodone CPP in female and male rats on opioid peptide and receptor gene expression as well as that of synaptic plasticity, stress and related signaling molecules. Genes selected for analysis were the same as those in our recent study in which we demonstrated sex differences in their expression following chronic stress (Randesi et al., 2018). To elucidate these findings, we examined the immunocytochemical distributions of the relevant proteins in the hippocampus from a separate cohort of rats that had been given oxycodone in a CPP paradigm.

Section snippets

Animals

A total of 48 adult Sprague–Dawley female (~ 225–250 g upon arrival) and male (~ 275–325 g upon arrival) rats were used in this study (RGD Cat# 734476, RRID:RGD_734,476; Charles River Laboratories, Wilmington, PA). To maintain rigor and reproducibility and minimize experimental variability due to differences in the vendor and institute environment as well as experimenter handling, molecular experiments were performed on a single cohort of 24 female and male rats obtained from the vendor at the

Female and male rats both acquire oxycodone CPP

Two-way ANOVA showed a significant main effect of condition (saline vs. oxycodone CPP) for the cohort of female and male rats used in the molecular study [F(3,18) = 28.7; p < 0.0001)]. Neither a significant main effect of sex nor a significant interaction between sex and condition was observed. Post-hoc analysis showed that Oxy-females and Oxy-males had a greater percent change in preference for the oxycodone-paired side of the box compared to their saline-injected (Sal) counterparts (females:

Discussion

The present study demonstrates that oxycodone CPP results in regional sex-dependent changes in plasticity (Arc, Bdnf and Npy), stress (Crhr1 and Crhr2), and related kinase (Akt1 and Mapk1) gene expression in the hippocampus. Similarly, there were sex-dependent differences in the related proteins (ARC, NPY, CRHR1, pAKT, pMAPK) following oxycodone CPP (Fig. 9). These changes could be involved in the previously observed sex differences in the opioid system after oxycodone CPP (Ryan et al., 2018).

Acknowledgements

Supported by NIH grants DA08259 (T.A.M., M.J.K., B.S.M.), HL098351 (T.A.M.), HL 136520 (T.A.M.), MH041256 (B.S.M.) and MH102065 (J.D.G.), and Hope for Depression Research grant (B.S.M.). We thank Megan Johnson for assistance with figure preparation.

Author Contributions

T.A.M., M.J.K., B.S.M. designed research; Y.Z., M.R., N.H.C., B.R., J.R.B., F.Y., J.D.G, T.A.M. performed research; N.H.C., M.R., F.Y., T.A.M. analyzed data; M.R., N.H.C., B.R., T.A.M., B.S.M., M.J.K. wrote the paper.

References (111)

  • HR McAlinn et al.

    Sex differences in the subcellular distribution of Corticotropin-releasing factor receptor 1 in the rat Hippocampus following chronic immobilization stress

    Neuroscience

    (2018)
  • LO Murphy et al.

    MAPK signal specificity: the right place at the right time

    Trends Biochem. Sci.

    (2006)
  • M Opendak et al.

    Adult neurogenesis: a substrate for experience-dependent change

    Trends Cogn. Sci.

    (2015)
  • JP Pierce et al.

    Hippocampal mossy fiber leu-enkephalin immunoreactivity in female rats is significantly altered following both acute and chronic stress

    J. Chem. Neuroanat.

    (2014)
  • R Pinaud et al.

    Upregulation of the immediate early gene arc in the brains of rats exposed to environmental enrichment: implications for molecular plasticity

    Brain Res. Mol. Brain Res.

    (2001)
  • N Plath et al.

    Arc/Arg3.1 is essential for the consolidation of synaptic plasticity and memories

    Neuron

    (2006)
  • M Randesi et al.

    Sex differences after chronic stress in the expression of opioid- and neuroplasticity-related genes in the rat hippocampus

    Neurol.Biol.Stress

    (2018)
  • L Regev et al.

    Corticotropin releasing factor in neuroplasticity

    Front. Neuroendocrinol.

    (2014)
  • MC Ribeiro-Dasilva et al.

    Evaluation of menstrual cycle effects on morphine and pentazocine analgesia

    Pain

    (2011)
  • S.A. Rogers et al.

    Enkephalin levels and the number of neuropeptide Y-containing interneurons in the hippocampus are decreased in female cannabinoid-receptor 1 knock-out mice

    Neurotic. Lett.

    (2016)
  • JD Ryan et al.

    Sex differences in the rat hippocampal opioid system after oxycodone conditioned place preference

    Neuroscience

    (2018)
  • M Sauvage et al.

    Detection of corticotropin-releasing hormone receptor 1 immunoreactivity in cholinergic, dopaminergic and noradrenergic neurons of the murine basal forebrain and brainstem nuclei--potential implication for arousal and attention

    Neuroscience

    (2001)
  • HE Scharfman et al.

    Sex differences in the neurobiology of epilepsy: a preclinical perspective

    Neurol. Biol. Dis. 72 Pt. B.

    (2014)
  • F Shen et al.

    Essential role of the NO signaling pathway in the hippocampal CA1 in morphine-associated memory depends on glutaminergic receptors

    Neuropharmacology

    (2016)
  • JL Spencer et al.

    Uncovering the mechanisms of estrogen effects on hippocampal function

    Front. Neuroendocrinol.

    (2008)
  • G Sperk et al.

    Neuropeptide Y in the dentate gyrus

    Prog. Brain Res.

    (2007)
  • N Trapaidze et al.

    Opioid receptor endocytosis and activation of MAP kinase pathway

    Brain Res. Mol. Brain Res.

    (2000)
  • QD Walker et al.

    Sex differences in cocaine-stimulated motor behavior: disparate effects of gonadectomy

    Neuro. psycho. pharmacology

    (2001)
  • QD Walker et al.

    Vaginal lavage attenuates cocaine-stimulated activity and establishes place preference in rats

    Pharmacol. Biochem. Behav.

    (2002)
  • KH Wang et al.

    In vivo two-photon imaging reveals a role of arc in enhancing orientation specificity in visual cortex

    Cell

    (2006)
  • M Waselus et al.

    Stress-induced redistribution of corticotropin-releasing factor receptor subtypes in the dorsal raphe nucleus

    Biol. Psychiatry

    (2009)
  • MJ Weiser et al.

    Androgen regulation of corticotropin-releasing hormone receptor 2 (CRHR2) mRNA expression and receptor binding in the rat brain

    Exp. Neurol.

    (2008)
  • TJ Williams et al.

    Delta opioid receptors colocalize with corticotropin releasing factor in hippocampal interneurons

    Neuroscience

    (2011)
  • TJ Williams et al.

    Ovarian hormones influence corticotropin releasing factor receptor colocalization with delta opioid receptors in CA1 pyramidal cell dendrites

    Exp. Neurol.

    (2011)
  • TJ Williams et al.

    Hormonal regulation of delta opioid receptor immunoreactivity in interneurons and pyramidal cells in the rat hippocampus

    Neurobiol. Learn. Mem.

    (2011)
  • KT Akama et al.

    Estrogen stimulates postsynaptic density-95 rapid protein synthesis via the Akt/protein kinase B pathway

    J. Neurosci.

    (2003)
  • J Alder et al.

    Transcriptional analysis in the brain: trophin-induced hippocampal synaptic plasticity

    Neurochem. Res.

    (2002)
  • S Arttamangkul et al.

    Differential activation and trafficking of micro-opioid receptors in brain slices

    Mol. Pharmacol.

    (2008)
  • DA Bangasser et al.

    Sex differences in corticotropin-releasing factor receptor signaling and trafficking: potential role in female vulnerability to stress-related psychopathology

    Mol. Psychiatry

    (2010)
  • JB Becker et al.

    Sex differences, gender, and addiction

    J. Neurosci. Res.

    (2017)
  • NC Berchtold et al.

    Estrogen and exercise interact to regulate brain-derived neurotrophic factor mRNA and protein expression in the hippocampus

    Eur. J. Neurosci.

    (2001)
  • R Bi et al.

    The tyrosine kinase and mitogen-activated protein kinase pathways mediate multiple effects of estrogen in hippocampus

    Proc. Natl. Acad. Sci. U. S. A.

    (2000)
  • YN Biala et al.

    Prenatal stress diminishes gender differences in behavior and in expression of hippocampal synaptic genes and proteins in rats

    Hippocampus

    (2011)
  • EA Bolan et al.

    Synergy between mu opioid ligands: evidence for functional interactions among mu opioid receptor subtypes

    J. Pharmacol. Exp. Ther.

    (2002)
  • H Boudin et al.

    Correlative ultrastructural distribution of neurotensin receptor proteins and binding sites in the rat substantia nigra

    J. Neurosci.

    (1998)
  • CR Bramham et al.

    The arc of synaptic memory

    Exp. Brain Res.

    (2010)
  • Centers for Disease Control and Prevention

    Drug overdose deaths hit record numbers in

    (2015)
  • Y Chen et al.

    Immunocytochemical distribution of corticotropin-releasing hormone receptor type-1 (CRF(1))-like immunoreactivity in the mouse brain: light microscopy analysis using an antibody directed against the C-terminus

    J. Comp. Neurol.

    (2000)
  • ZZ Chong et al.

    Activating Akt and the brain's resources to drive cellular survival and prevent inflammatory injury

    Histol. Histopathol.

    (2005)
  • KH Choy et al.

    Combined neonatal stress and young-adult glucocorticoid stimulation in rats reduce BDNF expression in hippocampus: effects on learning and memory

    Hippocampus

    (2008)
  • Cited by (18)

    • Sex and chronic stress alter the distribution of glutamate receptors within rat hippocampal CA3 pyramidal cells following oxycodone conditioned place preference

      2022, Neurobiology of Stress
      Citation Excerpt :

      Our previous studies have shown that Oxy CPP primes rats, particularly females, to have enhanced opioid learning and susceptibility to addictive learning behaviors (Ryan et al., 2018). Our other past work demonstrated that Oxy CPP leads to an increase in opioid, stress, and plasticity signaling molecules (such as ARC) in the hippocampus of female rats, contributing to LTP learning processes, synaptic plasticity, and memory (Randesi et al., 2019). While the current data shows a decrease in baseline GluN1 receptor subunit density in the CA3 hippocampus of female rats following Oxy CPP, this does not discount the previous evidence of Oxy CPP enhancing addictive learning behaviors in females, but may suggest a mechanism at play other than glutamatergic signaling in the CA3 pyramidal cell dendrites.

    • Sex differences in the rodent hippocampal opioid system following stress and oxycodone associated learning processes

      2022, Pharmacology Biochemistry and Behavior
      Citation Excerpt :

      However, Oxy CPP males had: a) increases in brain derived-neurotrophic factor (Bdnf) expression, which is known to be produced in granule cells; b) elevated Mapk1 expression and pMAPK-ir in the DG hilus which harbors newly generated granule cells; and c) increases in CRF receptor-ir in CA3 pyramidal cell soma. As previously discussed (Randesi et al., 2019), these findings suggest additional mechanisms by which Oxy CPP could interact with the hippocampal opioid system to differentially enhance plasticity and learning processes in females and males. As discussed in Sections 6 and 7, CIS differentially influences the opioid system as well as plasticity and stress markers in hippocampal neurons in female and male rats in a manner that would “prime” females for opioid mediated learning processes but negatively impact learning processes in males.

    • Chronic oral nicotine administration and withdrawal regulate the expression of neuropeptide Y and its receptors in the mesocorticolimbic system

      2021, Neuropeptides
      Citation Excerpt :

      Rewarding effects of nicotine exposure and aversive effects of nicotine withdrawal are enhanced in females [reviewed in Pogun et al., 2017 and O'Dell and Torres, 2014]. Also, studies report the presence of sex differences in NPY expression in HIP, striatum and hypothalamus during both basal condition and drug exposure (Randesi et al., 2019; Rugarn et al., 1999). Additionally, this study measured only the mRNA expression of NPY and YRs in specific brain regions.

    • Sex and chronic stress alter delta opioid receptor distribution within rat hippocampal CA1 pyramidal cells following behavioral challenges

      2020, Neurobiology of Stress
      Citation Excerpt :

      Moreover, phosphorylated AKT is elevated in the cytoplasm of CA1 dendrites and spines in proestrus females compared to males (Znamensky et al., 2003). Oxy-males have elevated expression of activity regulated cytoskeletal-associated protein (Arc) (Randesi et al., 2019), an immediate early gene targeted to synapses [reviewed in (Bramham et al., 2010)] and brain-derived neurotrophic factor (Bdnf) which is involved in synaptogenesis (Cunha et al., 2010). Arc and Bdnf are especially important in LTP processes (Cunha et al., 2010; Plath et al., 2006).

    • Sex and chronic stress differentially alter phosphorylated mu and delta opioid receptor levels in the rat hippocampus following oxycodone conditioned place preference

      2019, Neuroscience Letters
      Citation Excerpt :

      The increase in pDOR-ir in CA3 SR in the naïve Oxy-males parallels the upregulation of the immediate early gene activity-regulated cytoskeleton-associated protein (Arc) mRNA in CA3 in naïve Oxy-males compared to Sal-males [15]. Additionally, our prior study [15] demonstrated that naïve Oxy-males compared to naïve Oxy-females have elevated levels of brain derived neurotrophic factor (BDNF) expression in the dentate gyrus, which harbors the granule cells known to produce BDNF in the mossy fibers [31]. Both Arc and BDNF are important regulators of synaptic plasticity processes including synaptogenesis and LTP [32,33].

    View all citing articles on Scopus

    Contributed equally.

    Co-Senior authors.

    View full text