Elsevier

Neuropharmacology

Volume 76, Part B, January 2014, Pages 259-268
Neuropharmacology

Invited review
Epigenetic mechanisms of drug addiction

https://doi.org/10.1016/j.neuropharm.2013.04.004Get rights and content

Highlights

  • Epigenetic regulation mediates adaptations to the environment such as abused drugs.

  • Epigenetic regulation includes post-translational modifications of histones.

  • Epigenetic regulation also includes DNA methylation and non-coding RNAs.

  • Each of these epigenetic mechanisms are influenced by drugs of abuse.

  • In turn, these epigenetic mechanisms control behavioral responses to the drugs.

Abstract

Drug addiction involves potentially life-long behavioral abnormalities that are caused in vulnerable individuals by repeated exposure to a drug of abuse. The persistence of these behavioral changes suggests that long-lasting changes in gene expression, within particular regions of the brain, may contribute importantly to the addiction phenotype. Work over the past decade has demonstrated a crucial role for epigenetic mechanisms in driving lasting changes in gene expression in diverse tissues, including brain. This has prompted recent research aimed at characterizing the influence of epigenetic regulatory events in mediating the lasting effects of drugs of abuse on the brain in animal models of drug addiction. This review provides a progress report of this still early work in the field. As will be seen, there is robust evidence that repeated exposure to drugs of abuse induces changes within the brain's reward regions in three major modes of epigenetic regulation—histone modifications such as acetylation and methylation, DNA methylation, and non-coding RNAs. In several instances, it has been possible to demonstrate directly the contribution of such epigenetic changes to addiction-related behavioral abnormalities. Studies of epigenetic mechanisms of addiction are also providing an unprecedented view of the range of genes and non-genic regions that are affected by repeated drug exposure and the precise molecular basis of that regulation. Work is now needed to validate key aspects of this work in human addiction and evaluate the possibility of mining this information to develop new diagnostic tests and more effective treatments for addiction syndromes.

This article is part of a Special Issue entitled ‘NIDA 40th Anniversary Issue’.

Introduction

Drug addiction can be viewed as maladaptive neural plasticity that occurs in vulnerable individuals in response to repeated exposure to a drug of abuse. That vulnerability is determined roughly half by genetic factors (although few specific causative genes have as yet been identified) and half by non-genetic factors which include environmental exposures as well as stochastic events during development. Once formed, in turn, addiction can drive life-long behavioral abnormalities.

These features of addiction suggest an important role for epigenetic mechanisms. The term epigenetics has several definitions; this review utilizes a broad one, which defines epigenetics as a series of biochemical processes through which changes in gene expression are achieved throughout the lifecycle of an organism without a change in DNA sequence (Jaenisch and Bird, 2003). Epigenetics can thus be viewed as the vehicle through which environment interacts with an individual's genome to determine all aspects of function, in health and disease. A subset of epigenetic changes are very stable, which makes them ideal mediators both of addiction vulnerability and of drug-induced brain maladaptations that underlie an addiction syndrome.

Within this context, there are three general roles that epigenetic mechanisms likely play in addiction (Tsankova et al., 2007; Robison and Nestler, 2011). First, repeated exposure to a drug of abuse in adolescence or adulthood causes addiction in vulnerable individuals by inducing stable changes in gene expression through epigenetic regulation of those specific genes. Such epigenetic regulation involves alterations in the steady state expression levels of a set of genes as well as changes in other genes' inducibility—both sensitization (priming) and desensitization—without a change in steady state expression. Regulation of gene inducibility can be seen as “latent” in that it would not be apparent by analysis of mRNA or protein levels. Epigenetic regulation of genes also alters the expression of splice isoforms of a gene, which is usually not apparent from traditional microarray analyses of expressed mRNAs. Second, epigenetic regulation mediates changes in steady state gene expression or inducibility of genes that occur throughout an individual's lifetime in response to a host of environmental exposures, which help determine that individual's vulnerability to drug exposure and addiction later in life (Hiroi and Agatsuma, 2005). Third, there is the possibility that drugs or other environmental exposures induce epigenetic changes in sperm or ova, which are then passed on to offspring and alter their vulnerability to addiction. Such trans-generational epigenetic inheritance of addiction vulnerability remains controversial.

Finally, because the large majority of investigations of epigenetic mechanisms have been carried out on cultured cells or peripheral tissues, studies of epigenetic regulation in addiction models will teach the field fundamental principles about epigenetics in the developing and adult nervous system. In this way, such work provides the first ever look at mechanisms of transcriptional regulation in brain, and will likely have enormous impact on the field.

Section snippets

Overview of mechanisms of epigenetic regulation

The 3 billion nucleotides of DNA in a mammalian genome would be ∼2 m long if stretched out linearly, yet fits within a microscopic cell nucleus due to its extraordinary degree of organization and compaction in chromatin—nuclear material composed of DNA, histones, and non-histone proteins (Jaenisch and Bird, 2003). The fundamental unit of chromatin is the nucleosome, which consists of ∼147 base pairs of DNA wrapped around a core histone octamer (∼1.65 turns). Each octamer contains two copies

Role of histone modifications in addiction

Increasing evidence is defining the mechanisms by which chronic exposure to a drug of abuse alters the steady state levels of mRNAs through chromatin regulatory mechanisms or primes or desensitizes additional genes for altered expression after some period of withdrawal (Fig. 3).

Role of DNA methylation in addiction

Given the hypothesized importance of DNA methylation in mediating sustained transcriptional change, it is unfortunate that there have not been more studies of this epigenetic mechanism in drug abuse models. Expression levels of DNMT3a in NAc are differentially altered by acute versus chronic cocaine exposure, and during extended withdrawal (Anier et al., 2010; LaPlant et al., 2010). Local knockout of DNMT3a from the NAc, or local infusion of the DNMT inhibitors RG108 or zebularine, increases

Role of noncoding RNAs in addiction

Multiple miRNAs are reported to be up- or downregulated by drugs of abuse. For instance, cocaine increases levels of miR-181a and decreases miR-124 and let-7d in rat striatum (Chandrasekar and Dreyer, 2009, 2011; Schaefer et al., 2010; Hwang et al., 2012; Sartor et al., 2012), and mimicking the direction of each of these changes enhances cocaine reward. Since miRNAs function via base-pairing with complementary sequences within mRNA molecules, it is possible to infer target mRNAs of drug-altered

Future directions

Although still in relatively early stages, work to date has demonstrated that many forms of epigenetic regulation are altered in brain reward regions by drugs of abuse and in turn serve to regulate drug action. Already these initial studies have raised several key questions that will need to be addressed moving forward.

One surprising finding is that drug exposure alters global levels of several histone modifications, such as increased histone acetylation or decreased H3K9me2 and H3K9me3 in NAc.

Conclusions

The ultimate goal of epigenetic studies of addiction is to understand how repeated exposure to a drug of abuse changes the brain in sustained ways to cause the lasting syndrome of addiction. Such studies are also important to understand how an individual's life experiences establish stable changes at genomic loci, which then help determine that individual's vulnerability to the addiction-causing effects of subsequent drug exposure. It is our expectation that these studies will reveal a host of

Acknowledgments

Preparation of this review was supported by grants from the National Institute on Drug Abuse.

References (85)

  • M.S. Mazei-Robison et al.

    Role for mTOR signaling and neuronal activity in morphine-induced adaptations in ventral tegmental area dopamine neurons

    Neuron

    (2011)
  • M. Pascual et al.

    Changes in histone acetylation in the prefrontal cortex of ethanol-exposed adolescent rats are associated with ethanol-induced place conditioning

    Neuropharmacology

    (2012)
  • A.Z. Pietrzykowski et al.

    Posttranscriptional regulation of BK channel splice variant stability by miR-9 underlies neuroadaptation to alcohol

    Neuron

    (2008)
  • W. Renthal et al.

    Histone deacetylase 5 epigenetically controls behavioral adaptations to chronic emotional stimuli

    Neuron

    (2007)
  • W. Renthal et al.

    Genome wide analysis of chromatin regulation by cocaine reveals a novel role for sirtuins

    Neuron

    (2009)
  • H.Y. Shen et al.

    Additive effects of histone deacetylase inhibitors and amphetamine on histone H4 acetylation, cAMP responsive element binding protein phosphorylation and DeltaFosB expression in the striatum and locomotor sensitization in mice

    Neuroscience

    (2008)
  • J. Sun et al.

    The effects of sodium butyrate, an inhibitor of histone deacetylase, on the cocaine- and sucrose-maintained self administration in rats

    Neurosci. Lett.

    (2008)
  • M. Taniguchi et al.

    Histone deacetylase 5 limits cocaine reward through cAMP-induced nuclear import

    Neuron

    (2012)
  • H.C. Tomasiewicz et al.

    Proenkephalin mediates the enduring effects of adolescent cannabis exposure associated with adult opiate vulnerability

    Biol. Psychiatry

    (2012)
  • M. Adli et al.

    Whole-genome chromatin profiling from limited numbers of cells using nano-ChIP-seq

    Nat. Protoc.

    (2011)
  • K. Anier et al.

    DNA methylation regulates cocaine-induced behavioral sensitization in mice

    Neuropsychopharmacology

    (2010)
  • J. Bertran-Gonzalez et al.

    Opposing patterns of signaling activation in dopamine D1 and D2 receptor-expressing striatal neurons in response to cocaine and haloperidol

    J. Neurosci.

    (2008)
  • B. Botia et al.

    Expression of ethanol-induced behavioral sensitization is associated with alteration of chromatin remodeling in mice

    PLoS One

    (2012)
  • K. Brami-Cherrier et al.

    Parsing molecular and behavioral effects of cocaine in mitogen- and stress-activated protein kinase-1-deficient mice

    J. Neurosci.

    (2005)
  • K. Brami-Cherrier et al.

    Role of the ERK/MSK1 signalling pathway in chromatin remodelling and brain responses to drugs of abuse

    J. Neurochem.

    (2009)
  • V. Chandrasekar et al.

    Regulation of MiR-124, Let-7d, and MiR-181a in the accumbens affects the expression, extinction, and reinstatement of cocaine-induced conditioned place preference

    Neuropsychopharmacology

    (2011)
  • C. D'Addario et al.

    Ethanol induces epigenetic modulation of prodynorphin and pronociceptin gene expression in the rat amygdala complex

    J. Mol. Neurosci.

    (2013)
  • D. Damez-Werno et al.

    Drug experience epigenetically primes fosB gene inducibility in rat nucleus accumbens and caudate putamen

    J. Neurosci.

    (2012)
  • D. Damez-Werno et al.

    Histone arginine methylation in the nucleus accumbens in response to chronic cocaine and stress

    Soc. Neurosci. Abs

    (2012)
  • J.V. Deng et al.

    MeCP2 in the nucleus accumbens contributes to neural and behavioral responses to psychostimulants

    Nat. Neurosci.

    (2010)
  • I. Dunham et al.

    An integrated encyclopedia of DNA elements in the human genome

    Nature

    (2012)
  • J.E. Eipper-Mains et al.

    microRNA-Seq reveals cocaine-regulated expression of striatal microRNAs

    RNA

    (2011)
  • D. Ferguson et al.

    Next generation sequencing using ChIP-Seq highlights essential role for SIRT1 in emotional plasticity

    Soc. Neurosci. Abs

    (2012)
  • E. Heller et al.

    Bidirectional regulation of the fosB gene using synthetic zinc-finger transcription factors for the study of addiction and depression

    Soc. Neurosci. Abs

    (2012)
  • N. Hiroi et al.

    Genetic susceptibility to substance dependence

    Mol. Psychiatry

    (2005)
  • J.A. Hollander et al.

    Striatal microRNA controls cocaine intake through CREB signalling

    Nature

    (2010)
  • C.K. Hwang et al.

    Epigenetic programming of mu-opioid receptor gene in mouse brain is regulated by MeCP2 and Brg1 chromatin remodelling factor

    J. Cell. Mol. Med.

    (2009)
  • C.K. Hwang et al.

    MicroRNAs in opioid pharmacology

    J. Neuroimmune Pharmacol.

    (2012)
  • H.I. Im et al.

    MeCP2 controls BDNF expression and cocaine intake through homeostatic interactions with microRNA-212

    Nat. Neurosci.

    (2010)
  • R. Jaenisch et al.

    Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals

    Nat. Genet.

    (2003)
  • T. Jenuwein et al.

    Translating the histone code

    Science

    (2001)
  • Y. Jiang et al.

    Isolation of neuronal chromatin from brain tissue

    BMC Neurosci.

    (2008)
  • Cited by (294)

    • Epigenetic mechanisms of nicotine dependence

      2024, Neuroscience and Biobehavioral Reviews
    View all citing articles on Scopus
    View full text