Elsevier

Drug and Alcohol Dependence

Volume 175, 1 June 2017, Pages 9-23
Drug and Alcohol Dependence

Full length article
Transcriptomic profiling of the ventral tegmental area and nucleus accumbens in rhesus macaques following long-term cocaine self-administration

https://doi.org/10.1016/j.drugalcdep.2017.01.030Get rights and content

Highlights

  • Patterns of cocaine self-administration in rhesus macaques model humans.

  • Gene expression differences are observed in the mesolimbic system after exposure.

  • Changes in the ventral tegmental area are associated with the dopaminergic system.

  • Changes in the nucleus accumbens reflect neuroinflammation and chromatin remodeling.

Abstract

Background

The behavioral consequences associated with addiction are thought to arise from drug-induced neuroadaptation. The mesolimbic system plays an important initial role in this process, and while the dopaminergic system specifically has been strongly interrogated, a complete understanding of the broad transcriptomic effects associated with cocaine use remains elusive.

Methods

Using next generation sequencing approaches, we performed a comprehensive evaluation of gene expression differences in the ventral tegmental area and nucleus accumbens of rhesus macaques that had self-administered cocaine for roughly 100 days and saline-yoked controls. During self-administration, the monkeys increased daily consumption of cocaine until almost the maximum number of injections were taken within the first 15 min of the one hour session for a total intake of 3 mg/kg/day.

Results

We confirm the centrality of dopaminergic differences in the ventral tegmental area, but in the nucleus accumbens we see the strongest evidence for an inflammatory response and large scale chromatin remodeling.

Conclusions

These findings suggest an expanded understanding of the pathology of cocaine addiction with the potential to lead to the development of alternative treatment strategies.

Introduction

Drug abuse is associated with a number of molecular and cellular effects on the brain including changes in neurocircuitry, gene expression, and epigenetic regulation. These changes are believed to be linked with the transition from substance use to abuse: compulsive drug seeking, loss of restraint, and negative affect (Koob and Volkow, 2010, Luscher and Malenka, 2011). The mesolimbic dopamine system, connecting the ventral tegmental area (VTA) and the nucleus accumbens (NAc), drives the salient effects of reward and has been consistently and repeatedly shown to be the primary site of action of drugs of abuse (Nestler, 2005).

There have been a number of studies in rodents and human post-mortem tissues that focus on gene expression differences associated with addiction both in the mesolimbic pathway as well as other areas of the brain (Zhou et al., 2014b). Microarray studies in rodents have identified differences in immediate-early genes (genes activated rapidly following extracellular stimulation) and dopaminergic pathways (Piechota et al., 2010, Yuferov et al., 2003, Zhang et al., 2005) in accordance with previous findings using Northern blots and immunohistochemistry (Hope et al., 1992). More recent studies using next generation sequencing technologies to study the effect of cocaine on the mouse NAc found differences from controls across multiple neurotransmitter systems including dopaminergic, cholinergic, glutamatergic, GABAergic systems as well as cadherin and Wnt signaling pathways (Eipper-Mains et al., 2013). This has been extended to non-coding RNAs with functional effects imputed using informatics approaches (Bu et al., 2012, Chen et al., 2013).

There have also been a number of microarray studies of gene expression on human cocaine abusers in the NAc (Albertson et al., 2004, Bannon et al., 2005) and midbrain dopaminergic regions (Tang et al., 2003). While confirming many of the dopaminergic findings from rodents, more widespread transcriptional changes were also identified. These findings have been suggested to reflect epigenetic reprogramming resulting from chronic drug exposure (Zhou et al., 2011). The differences observed between rodent and human expression studies may result from methodological differences in the duration of exposure to drug, acute compared to chronic usage (Zhou et al., 2014b). Indeed, transgenic mouse work has shown that histone acetylation is important for response to chronic, but not acute, cocaine exposure (Renthal et al., 2007).

One of the challenges in developing translational animal models is to recapitulate as closely as possible the most salient features of human behavior while maintaining precise experimental control. In addition to greater genetic and neuroanatomical similarities, nonhuman primate models in particular are valuable when modeling aspects of addiction because their consumption and patterns of drug taking so closely reflect that seen in human drug addiction (Platt and Rowlett, 2012). A rhesus macaque model of self-administration provides a unique opportunity to explore the molecular mechanisms involved in the entirety of the neurochemical systems and circuitry associated with the addictive processes (Weerts et al., 2007).

In the present study, we used a cocaine self-administration procedure in rhesus macaques over three months (approximately 100 consecutive days). After this exposure period, transcriptomic analysis was performed on the NAc and the VTA using next generation RNA-seq. This allows for an unbiased and holistic view of the differences between cocaine- and saline-treated animals in these regions and provides a simultaneous assessment of the two brain regions in long-term cocaine consumption.

Section snippets

Ethics statement

Animals were maintained in accordance with the guidelines of the Committee on Animals of Harvard Medical School and the Guide for the Care and Use of Laboratory Animals (8th edition, 2011). Research protocols were approved by the Harvard Medical School Institutional Animal Care and Use Committee.

Animals

Subjects were 10, experimentally naïve, male young adult (4–7 years) rhesus macaques (Macaca mulatta). All animals were raised in shared conditions with identical diet and husbandry; paired animals

Cocaine self-administration

While there is some variability across animals (Supplemental Fig. S1), when cocaine self-administration was averaged across animals, there was evidence of a gradual increase in self-administration over sessions (Fig. 1A,B). Linear trend analysis revealed a significant fit of these data (84 days (n = 5) [F(1,82) = 18.23, p < 0.001]; 100 days (n = 4) [F(1,98) = 25.06, p < 0.001]) with a positive slope, although the relationship of session and number of injections/hour was relatively weak (r2 = 0.18 or 0.20

Discussion

Under conditions of limited access (1 h/day) and long-term exposure (approximately 100 consecutive days), we observed a general increase over time in cocaine self-administration in monkeys. However, while statistically significant, this escalation was modest in size. While there was a limit on the total amount of cocaine that was available for the animals to consume (so as to not lead to overdose), this did not fully account for the magnitude effect. Rather, what was observed was a relatively

Contributors

W.D.Y. and J.K.R. were responsible for study concept and design. N.M.S. and J.K.R. were responsible for collection of animal data. W.D.Y., S.V.M., D.B.G. were responsible for brain and tissue collection. D.B.G. and E.J.V. were responsible for data analysis. E.J.V. and J.K.R. drafted the manuscript. All authors critically reviewed the manuscript and approved the final version for publication.

Role of funding source

Nothing declared.

Conflict of interest

No conflict declared.

Acknowledgement

This work was supported by grants from the NIH: DA021420 (WDY), OD011103.

References (73)

  • C. Luscher et al.

    Drug-evoked synaptic plasticity in addiction: from molecular changes to circuit remodeling

    Neuron

    (2011)
  • Y.E. Mavromatakis et al.

    Foxa1 and Foxa2 positively and negatively regulate Shh signalling to specify ventral midbrain progenitor identity

    Mech. Dev.

    (2011)
  • T. Nakatani et al.

    Lmx1a and Lmx1b cooperate with Foxa2 to coordinate the specification of dopaminergic neurons and control of floor plate cell differentiation in the developing mesencephalon

    Dev. Biol.

    (2010)
  • E.J. Nestler

    Epigenetic mechanisms of drug addiction

    Neuropharmacology

    (2014)
  • D.M. Platt et al.

    Nonhuman primates in drug and alcohol addiction research

  • W. Renthal et al.

    Histone deacetylase 5 epigenetically controls behavioral adaptations to chronic emotional stimuli

    Neuron

    (2007)
  • X. Zhang et al.

    Limitations of the rhesus macaque draft genome assembly and annotation

    BMC Genomics

    (2012)
  • Z. Zhou et al.

    Gene expression in the addicted brain

    Int. Rev. Neurobiol.

    (2014)
  • D.N. Albertson et al.

    Gene expression profile of the nucleus accumbens of human cocaine abusers: evidence for dysregulation of myelin

    J. Neurochem.

    (2004)
  • M.N. Baliki et al.

    Parceling human accumbens into putative core and shell dissociates encoding of values for reward and pain

    J. Neurosci.

    (2013)
  • M. Bannon et al.

    Gene expression profiling in the brains of human cocaine abusers

    Addict. Biol.

    (2005)
  • M.J. Bannon et al.

    A molecular profile of cocaine abuse includes the differential expression of genes that regulate transcription, chromatin, and dopamine cell phenotype

    Neuropsychopharmacology

    (2014)
  • M.J. Bannon et al.

    Investigating the potential influence of cause of death and cocaine levels on the differential expression of genes associated with cocaine abuse

    PLoS One

    (2015)
  • O. Bell et al.

    Determinants and dynamics of genome accessibility

    Nat. Rev. Genet.

    (2011)
  • M. Bsibsi et al.

    Toll-like receptor 3 on adult human astrocytes triggers production of neuroprotective mediators

    Glia

    (2006)
  • Q. Bu et al.

    Transcriptome analysis of long non-coding RNAs of the nucleus accumbens in cocaine-conditioned mice

    J. Neurochem.

    (2012)
  • P.W. Czoty et al.

    Long-term cocaine self-administration under fixed-ratio and second-order schedules in monkeys

    Psychopharmacology

    (2007)
  • S. Edwards et al.

    Escalation of drug self-administration as a hallmark of persistent addiction liability

    Behav. Pharmacol.

    (2013)
  • J.E. Eipper-Mains et al.

    Effects of cocaine and withdrawal on the mouse nucleus accumbens transcriptome

    Genes Brain Behav.

    (2013)
  • J. Feng et al.

    Chronic cocaine-regulated epigenomic changes in mouse nucleus accumbens

    Genome Biol.

    (2014)
  • A.L. Ferri et al.

    Foxa1 and Foxa2 regulate multiple phases of midbrain dopaminergic neuron development in a dosage-dependent manner

    Development

    (2007)
  • C.K. Glass et al.

    Molecular control of activation and priming in macrophages

    Nat. Immunol.

    (2016)
  • I. Glezer et al.

    Innate immunity triggers oligodendrocyte progenitor reactivity and confines damages to brain injuries

    FASEB J.

    (2006)
  • L. Heimer et al.

    The accumbens: beyond the core-shell dichotomy

    J. Neuropsychiatry Clin. Neurosci.

    (1997)
  • P.K. Henry et al.

    Cocaine-induced reinstatement during limited and extended drug access conditions in rhesus monkeys

    Psychopharmacology

    (2009)
  • B. Hope et al.

    Regulation of immediate early gene expression and AP-1 binding in the rat nucleus accumbens by chronic cocaine

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

    (1992)
  • Cited by (23)

    • Cocaine self-administration differentially activates microglia in the mouse brain

      2020, Neuroscience Letters
      Citation Excerpt :

      In addition to rodent models, non-human primate rhesus macaques with cocaine exposure also showed increased neuroinflammation. In the nucleus accumbens (NAc) brain region of rhesus monkeys with cocaine self-administration, a cluster of upregulated immune response and inflammation related genes was identified and associated predominately with microglial activation [11]. Consistently, elevations in microglial activation were found in the frontal white matter of these monkeys following prolonged cocaine self-administration [12].

    • Can cocaine-induced neuroinflammation explain maladaptive cocaine-associated memories?

      2020, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      For example, analysis of genomic data (Clark et al., 2013) obtained from whole RNA sequencing studies (Ahmed et al., 2005; Renthal et al., 2007 and Piechota et al., 2010) highlight gene expression increases in inflammatory modulators in several brain reward structures (e.g., dorsal striatum, NAc, PFC, amygdala, septum, lateral hypothalamus, and VTA), following acute or chronic cocaine intake in rats. A global increase in neuroinflammatory marker expression was confirmed in an RNAseq study of both NAc and VTA samples from cocaine self-administering nonhuman primates (Vallender et al., 2017). Pro-inflammatory cytokines, cytokine receptors, chemokines, TLRs, MMPs, ADAMs, PDGFs, and MCSF are among the genes up-regulated by cocaine.

    View all citing articles on Scopus
    1

    Co-first authors.

    2

    Co-senior authors.

    View full text