Elsevier

Pharmacological Research

Volume 114, December 2016, Pages 209-218
Pharmacological Research

Original article
Opioid gene expression changes and post-translational histone modifications at promoter regions in the rat nucleus accumbens after acute and repeated 3,4-methylenedioxy-methamphetamine (MDMA) exposure

https://doi.org/10.1016/j.phrs.2016.10.023Get rights and content

Abstract

The recreational drug of abuse 3,4-methylenedioxymethamphetamine (MDMA) has been shown to produce neurotoxic damage and long-lasting changes in several brain areas. In addition to the involvement of serotoninergic and dopaminergic systems, little information exists about the contribution of nociceptin/orphaninFQ (N/OFQ)-NOP and dynorphin (DYN)-KOP systems in neuronal adaptations evoked by MDMA. Here we investigated the behavioral and molecular effects induced by acute (8 mg/kg) or repeated (8 mg/kg twice daily for seven days) MDMA exposure.

MDMA exposure affected body weight gain and induced hyperlocomotion; this latter effect progressively decreased after repeated administration. Gene expression analysis indicated a down-regulation of the N/OFQ system and an up-regulation of the DYN system in the nucleus accumbens (NAc), highlighting an opposite systems regulation in response to MDMA exposure.

Since histone modifications have been strongly associated to the addiction-related maladaptive changes, we examined two permissive (acH3K9 and me3H3K4) and two repressive transcription marks (me3H3K27 and me2H3K9) at the pertinent opioid gene promoter regions. Chromatin immunoprecipitation assays revealed that acute MDMA increased me3H3K4 at the pN/OFQ, pDYN and NOP promoters. Following acute and repeated treatment a significant decrease of acH3K9 at the pN/OFQ promoter was observed, which correlated with gene expression results. Acute treatment caused an acH3K9 increase and a me2H3K9 decrease at the pDYN promoter which matched its mRNA up-regulation.

Our data indicate that the activation of the DYNergic stress system together with the inactivation of the N/OFQergic anti-stress system contribute to the neuroadaptive actions of MDMA and offer novel epigenetic information associated with MDMA abuse.

Introduction

MDMA (3,4-methylenedioxy-methamphetamine), popularly known as “ecstasy”, is a synthetic psychoactive drug which exhibits similarities to amphetamine and to the hallucinogen mescaline [1]. It produces feelings of euphoria, emotional warmth and distortions in sensory and time perceptions [2], [3]. MDMA was initially popular among adolescents and young adults in the nightclub scene or at “raves” (long dance parties), but currently it affects a broader range of users. The powerful effects of MDMA on the serotonergic system seem to be responsible for most actions of this drug of abuse. The massive release of serotonin (5-HT) is mainly due to MDMA action as a substrate-type releaser on the serotonin transporter (SERT) [4]. Furthermore, MDMA rapidly increases dopamine (DA) release [5], [6] and several studies demonstrated the involvement of other neurotransmitter circuits such as the glutamatergic, cholinergic and opioid systems in MDMA-mediated behavioral and neurochemical effects [7], [8], [9]. Notably, the opioid antagonist naloxone is able to block the locomotor stimulation caused by MDMA in mice [10] and it can attenuate MDMA ability to produce conditioned place preference (CPP) in rats [11]. In addition, the δ-opioid antagonist naltrindole blocks the increase of intracranial brain stimulation induced by MDMA [12].

In vivo studies demonstrated that MDMA treatment alters the dynorphinergic system in specific rat brain regions. It has been observed that a single MDMA administration increases dynorphin (DYN) A and prodynorphin (pDYN) mRNA levels in the striatum [13], [14], [15] and markedly up-regulates pDYN levels in the prefrontal cortex and in the brainstem [8], [15]. Conversely, a single MDMA treatment promoted pDYN down-regulation in the ventral tegmental area [15] suggesting a specific response depending on the brain area analyzed.

The nociceptin/orphaninFQ (N/OFQ) peptide shows high structural homology to DYN A and interacts with a G protein-coupled receptor known as the nociceptin (NOP) receptor, which is closely related to the κ-opioid (KOP) receptor [16]. It has been previously suggested that the N/OFQ system is involved in neuronal adaptations evoked by amphetamine since N/OFQ treatment can inhibit amphetamine-induced CPP acquisition in rats [17].

Gene expression alterations contribute to the long-lasting changes induced by MDMA in the brain reward circuitry but the detailed molecular steps through which such alterations are induced are unknown. Several studies identify chromatin remodeling as one of the main gene expression regulators for many drugs of abuse including alcohol, cocaine and amphetamine [18], [19], [20], [21], contributing substantially to the addictive phenotype. Chromatin modifications can activate or repress the transcriptional activity through post-translational histone modifications, which play a crucial role in the maintenance of the steady-state of chromatin [22], [23], [24]. It has been demonstrated that the increase of histone-3 (H3) and histone-4 (H4) acetylation on lysine (K) residues is linked to increased gene expression, whereas histone de-acetylation has been connected with a decrease of several mRNA levels in addiction paradigms [25], [26]. Conversely, H3 or H4 K methylation may act as permissive or repressive marker depending on the methylation site [27].

Although histone modifications induced by cocaine or ethanol in specific brain areas have been characterized using the chromatin immunoprecipitation (ChIP) technique [28], [29], [30], histone changes induced by MDMA exposure have not been sufficiently investigated.

Thus, this study examined the effects of acute and repeated MDMA exposure on the opioid genes expression, as well as the underlying chromatin remodeling mechanisms. The evidence of MDMA-induced mRNA changes prompted us to investigate whether post-translational modifications of histone proteins at the corresponding opioid promoter regions were involved in the regulation of gene expression. Two distinct histone modifications acting as permissive transcription marks were chosen: the acetylation of histone 3 at lysine 9 (acH3K9) and the trimethylation of histone 3 at lysine 4 (me3H3K4). In addition, two histone modification acting as repressive marks were examined: the trimethylation of histone 3 at lysine 27 (me3H3K27) and the dimethylation of histone 3 at lysine 9 (me2H3K9). The brain area designated for the analyses was the nucleus accumbens (NAc) because of its pivotal role in drug addiction. Due to its connectivity with other brain regions, NAc integrates information from cortical and limbic structures to mediate goal-directed behaviors. Accordingly, repeated exposure to several classes of drugs of abuse, including MDMA, may disrupt the plasticity in this region allowing a pathologic motivation for drug seeking.

Section snippets

Drug

MDMA was supplied from the National Institute on Drug Abuse/National Institutes of Health (Research Triangle Institute, Research Triangle Park, NC, USA) and dissolved in saline (0.9% NaCl).

Animals and treatments

Male Sprague-Dawley rats (Harlan, Italy) weighing 200–250 g at the beginning of the experiment were used. Animals were housed two per cage in standard cages (Tecniplast Gazzada, Buguggiate, Italy) and kept in a temperature- and humidity-controlled room with a 12-h light/dark cycle (lights on at 7 a.m.);

MDMA effects on body weight

A significant body weight reduction was observed 2 h after a single MDMA i.p. injection (treatment F(1,12) = 21,85, p < 0.001) (Fig. 2a). Rats repeatedly injected with vehicle showed a physiological body weight increase throughout the treatment (Fig. 2b). Conversely, MDMA-treated animals (twice daily injections of 8.0 mg/kg for seven days) exhibited a significant body weight reduction until the seventh day (treatment F(1,8) = 37.69, p = 0.0003; time F(6,47) = 9.04, p < 0.0001) (Fig. 2b). A significant time x

Discussion

The present study investigated the behavioral and molecular effects evoked by MDMA exposure. So far, a broad range of MDMA doses has been investigated in several experimental paradigms and from 1.5 mg/kg up to 12 or 25 mg/kg specific neuronal alterations [38], [39], [40], [41] and behavioral effects [42], [43], [44], [45] were reported. Previous data obtained in our laboratory showed that the 8.0 mg/kg MDMA, administered once or twice daily, may generate alterations in selected rat brain regions

Conclusions

This study indicates for the first time that MDMA induces specific histone modifications that could be related to the opioid gene expression regulation. In particular, we identified simultaneous changes of N/OFQ-NOP and DYN-KOP systems in the rat NAc after single or repeated MDMA injections. Opposite gene expression changes were recorded suggesting that their dysregulation may contribute to the development of a state of dependence since the activation of the DYNergic brain stress system occurs

Author contributions

FFC and PR conceived and designed the experiments. FFC and MP performed the experiments. FFC, SC and LC analyzed the data. FFC, PR, LC and SC wrote the manuscript. All authors contributed and approved the final manuscript.

Acknowledgements

This work was supported by grants from the University of Bologna (RFO2014 to PR and RFO2013 to SC). We wish to thank theNational Institute on Drug Abuse (NIDA)/National Institute of Health (NIH) (Research Triangle Park) for providing MDMA for experimental use and we wish to thank Dr. Roberto Rimondini for his help in the behavioral experiments.

References (84)

  • T. Kouzarides

    Chromatin modifications and their function

    Cell

    (2007)
  • R.J. Sims et al.

    Histone lysine methylation: a signature for chromatin Function

    Trends Genet.

    (2003)
  • S. Moonat et al.

    Aberrant histone deacetylase2-mediated histone modifications and synaptic plasticity in the amygdala predisposes to anxiety and alcoholism

    Biol. Psychiatry

    (2013)
  • F.F. Caputi et al.

    Dynorphin/KOP and nociceptin/NOP gene expression and epigenetic changes by cocaine in rat striatum and nucleus accumbens

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2014)
  • P. Chomczynski et al.

    Single-step method of RNA isolation by acidguanidinium thiocyanate-phenolchloroform extraction

    Anal. Biochem.

    (1987)
  • K.J. Livak et al.

    Analysis of relative gene expression data usingreal-time quantitative PCR and the 2(-Delta Delta C(T))

    Methods

    (2001)
  • K.D. Johnson et al.

    Dissecting long-range transcriptional mechanisms by chromatin immunoprecipitation

    Methods

    (2002)
  • F. Yuki et al.

    Warning against co-administration of 3,4-methylenedioxymethamphetamine (MDMA) with methamphetamine from the perspective of pharmacokinetic and pharmacodynamic evaluations in rat brain

    Eur. J. Pharm. Sci.

    (2013)
  • S.A. Collins et al.

    MDMA-induced loss of parvalbumin interneurons within the dentate gyrus is mediated by 5HT2A and NMDA receptors

    Eur. J. Pharmacol.

    (2015)
  • M.E. Tancer et al.

    The subjective effects of MDMA and mCPP in moderate MDMA users

    Drug Alcohol Depend.

    (2001)
  • M.R. Thompson et al.

    A role for oxytocin and 5-HT(1A) receptors in the prosocial effects of 3,4 methylenedioxymethamphetamine (ecstasy)

    Neuroscience

    (2007)
  • P.J. Wellman et al.

    Changes in feeding and locomotion induced by amphetamine analogs in rats

    Drug Alcohol Depend.

    (2009)
  • W. White et al.

    The effects of dose and repeated administration on the longer-term hypophagia produced by amphetamine in rats

    Pharmacol. Biochem. Behav.

    (2010)
  • W. White et al.

    Acute withdrawal-related hypophagia elicited by amphetamine is attenuated by pretreatment with selective dopamine D1 or D2 receptor antagonists in rats

    Physiol. Behav.

    (2015)
  • D.Y. Kuo et al.

    Amphetamine-evoked changes of oxidative stress and neuropeptide Y gene expression in hypothalamus: regulation by the protein kinase C-delta signaling

    Chem. Biol. Interact.

    (2009)
  • M.G. McNamara et al.

    Some behavioural and neurochemical aspects of subacute (6)3,4-methylenedioxymethamphetamine administration in rats

    Pharmacol. Biochem. Behav.

    (1995)
  • A. Di Giannuario et al.

    Orphanin FQ reduces morphine-induced dopamine release in the nucleus accumbens: a microdialysis study in rats

    Neurosci. Lett.

    (1999)
  • R. Ciccocioppo et al.

    Effect of nociceptin/orphanin FQ on the rewarding properties of morphine

    Eur. J. Pharmacol.

    (2000)
  • J. Turchan et al.

    Effects of repeated psychostimulant administration on the prodynorphin system activity and kappa opioid receptor density in the rat brain

    Neuroscience

    (1998)
  • A.J. Smith et al.

    Acute amphetamine or methamphetamine alters opioid peptide mRNA expression in rat striatum

    Brain Res. Mol. Brain Res.

    (1994)
  • G.F. Koob

    A role for brain stress systems in addiction

    Neuron

    (2008)
  • J.M. Witkin et al.

    The biology of Nociceptin/Orphanin FQ (N/OFQ) related to obesity stress, anxiety, mood, and drug dependence

    Pharmacol. Ther.

    (2014)
  • A. Gómez-Román et al.

    The neuroendocrine response to stress under the effect of drugs: negative synergy between amphetamine and stressors

    Psychoneuroendocrinology

    (2016)
  • E.J. Nestler

    Epigenetic mechanisms of drug addiction

    Neuropharmacology

    (2014)
  • A. Kalda et al.

    Epigenetic mechanisms of psychostimulant-induced addiction

    Int. Rev. Neurobiol.

    (2015)
  • S. Jayanthi et al.

    Methamphetamine downregulates striatal glutamate receptors via diverse epigenetic mechanisms

    Biol. Psychiatry

    (2014)
  • J.C. Rice et al.

    Histone methylation versus histone acetylation: new insights into epigenetic regulation

    Curr. Opin. Cell Biol.

    (2001)
  • J.P. Capela et al.

    Molecular and cellular mechanisms of ecstasy-induced neurotoxicity: an overview

    Mol. Neurobiol.

    (2009)
  • J. Cami et al.

    Human pharmacology of 3,4-methylenedioxymethamphetamine (ecstasy): psychomotor performance and subjective effects

    J. Clin. Psychopharmacol.

    (2000)
  • C. Michael White

    How MDMA’s pharmacology and pharmacokinetics drive desired effects and harms

    J. Clin. Pharmacol.

    (2014)
  • A.R. Green et al.

    The pharmacology and clinical pharmacology of 3,4-methylenedioxymethamphetamine (MDMA, Ecstasy)

    Pharmacol. Rev.

    (2003)
  • M.I. Colado et al.

    Acute and long-term effects of MDMA on cerebral dopamine biochemistry and function

    Psychopharmacology (Berl.)

    (2004)
  • Cited by (0)

    1

    Equally senior authors.

    View full text