101
|
Klee EW, Ebbert JO, Schneider H, Hurt RD, Ekker SC. Zebrafish for the study of the biological effects of nicotine. Nicotine Tob Res 2011; 13:301-12. [PMID: 21385906 DOI: 10.1093/ntr/ntr010] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Zebrafish are emerging as a powerful animal model for studying the molecular and physiological effects of nicotine exposure. The zebrafish have many advantageous physical characteristics, including small size, high fecundity rates, and externally developing transparent embryos. When combined with a battery of molecular-genetic tools and behavioral assays, these attributes enable studies to be conducted that are not practical using traditional animal models. METHODS We reviewed the literature on the application of the zebrafish model as a preclinical model to study the biological effects of nicotine exposure. RESULTS The identified studies used zebrafish to examine the effects of nicotine exposure on early development, addiction, anxiety, and learning. The methods used included green fluorescent protein-labeled proteins to track in vivo nicotine-altered neuron development, nicotine-conditioned place preference, and locomotive sensitization linked with high-throughput molecular and genetic screens and behavioral models of learning and stress response to nicotine. Data are presented on the complete homology of all known human neural nicotinic acetylcholine receptors in zebrafish and on the biological similarity of human and zebrafish dopaminergic signaling. CONCLUSIONS Tobacco dependence remains a major health problem worldwide. Further understanding of the molecular effects of nicotine exposure and genetic contributions to dependence may lead to improvement in patient treatment strategies. While there are limitations to the use of zebrafish as a preclinical model, it should provide a valuable tool to complement existing model systems. The reviewed studies demonstrate the enormous opportunity zebrafish have to advance the science of nicotine and tobacco research.
Collapse
Affiliation(s)
- Eric W Klee
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | |
Collapse
|
102
|
LaPlant Q, Nestler EJ. CRACKing the histone code: cocaine's effects on chromatin structure and function. Horm Behav 2011; 59:321-30. [PMID: 20594965 PMCID: PMC2948759 DOI: 10.1016/j.yhbeh.2010.05.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2010] [Revised: 05/14/2010] [Accepted: 05/22/2010] [Indexed: 12/21/2022]
Abstract
Epigenetics, the nongenetic component of how chromatin structure influences gene expression, is amazingly complex, and linking how environmental stimuli can influence epigenetic 'gene programs' in specific nerve cells to ultimately control behavior is a seemingly insurmountable puzzle. Cocaine is a highly potent stimulus capable of influencing behavior for the lifetime of an organism. Not surprisingly, psychostimulant-induced epigenetic regulation of gene expression has thus been identified as key to understanding the pathology of addiction. In addition to identifying this essential role of epigenetics in addiction, several important concepts have emerged such as the importance of global, temporal, and spatial control of mRNA expression in considering any given histone modification's influence on a given gene. Adding to this complexity, one has to account for the cumulative influence of other epigenetic modifications on a gene's transcription in addition to the interplay between transcription factors and chromatin structure. This review highlights how bioinformatic, molecular, and behavioral studies on addiction provide new insight into these concepts and outlines two distinct psychostimulant-induced patterns of chromatin regulation which are thought to underlie unique programs of gene expression that contribute importantly to the addicted state.
Collapse
Affiliation(s)
- Quincey LaPlant
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1065, New York, NY 10029-6574, USA
| | | |
Collapse
|
103
|
Abstract
Addiction is a debilitating psychiatric disorder, with a complex aetiology involving the interaction of inherited predispositions and environmental factors. Emerging evidence suggests that epigenetic alterations to the genome, including DNA methylation and histone modifications, are important mechanisms underlying addiction and the neurobiological response to addictive substances. In this review, we introduce the reader to epigenetic mechanisms and describe a potential role for dynamic epigenetic changes in mediating addictive behaviours via long-lasting changes in gene expression. We summarize recent findings from both molecular and behavioural experiments elucidating the role of epigenetic changes in mediating the addictive potential of various drugs of abuse, including cocaine, amphetamine and alcohol. The implications of these findings for molecular studies of addiction and the future development of novel therapeutic interventions are also discussed.
Collapse
Affiliation(s)
- Chloe C Y Wong
- Institute of Psychiatry, SGDP Research Centre, King's College London, De Crespigny Park, Denmark Hill, UK
| | | | | |
Collapse
|
104
|
Host L, Dietrich JB, Carouge D, Aunis D, Zwiller J. Cocaine self-administration alters the expression of chromatin-remodelling proteins; modulation by histone deacetylase inhibition. J Psychopharmacol 2011; 25:222-9. [PMID: 19939859 DOI: 10.1177/0269881109348173] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Injection of the histone deacetylases inhibitor trichostatin A to rats has been shown to decrease the reinforcing properties of cocaine. In the present study, we investigated alterations in gene expression patterns in the anterior cingulate cortex, caudate-putamen and nucleus accumbens of rats self-administering cocaine and treated with trichostatin A. As recent studies highlighted the importance of chromatin remodelling in the regulation of gene transcription in neurons, we studied the expression of Mecp2 and of several histone deacetylases. Cocaine self-administration was accompanied by an increased synthesis of Mecp2, HDAC2 and HDAC11 and by a decreased nuclear localization of HDAC5 and of the phospho-form of HDAC5, suggesting a nuclear export of this protein in response to the drug. The latter mechanism was further addressed by the demonstration of an enhanced expression of MEF2C transcription factor. Among the genes we examined, treatment with trichostatin A before each cocaine self-administration session was found to mostly affect Mecp2 and HDAC11 expression. A correlation was found between the modification of Mecp2 and MEF2C gene expression and the reinforcing property of cocaine. The two factors known to regulate gene transcription are likely to play a role in the neurobiological mechanism underlying a decrease in the reinforcing properties of cocaine.
Collapse
Affiliation(s)
- Lionel Host
- INSERM, U575, Centre de Neurochimie, Université de Strasbourg, Strasbourg, France
| | | | | | | | | |
Collapse
|
105
|
Pastor V, Host L, Zwiller J, Bernabeu R. Histone deacetylase inhibition decreases preference without affecting aversion for nicotine. J Neurochem 2011; 116:636-45. [PMID: 21166804 DOI: 10.1111/j.1471-4159.2010.07149.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epigenetic mechanisms have recently been shown to be involved in the long-term effects of drugs of abuse. A well described epigenetic mechanism modulating transcriptional activity consists in the binding to DNA of methyl-CpG binding proteins, such as MeCP2, recruiting histone deacetylases (HDACs). Nicotine causes long-term changes in the brain, but little is known concerning the mechanisms involved in nicotine-preference. Using a nicotine-conditioned place preference protocol, we demonstrate here that the histone deacetylase inhibitor phenylbutyrate was able to dramatically reduce the preference for nicotine, without altering the aversive properties of the drug. We measured immunohistochemically the acetylation of lysine-9 of histone H3, and the expression of phosphorylated cAMP-response element-binding protein, HDAC2 and methyl-CpG-binding protein 2 in the striatum and prefrontal cortex of rats displaying nicotine-preference or aversion and treated with phenylbutyrate. We show that, at the dose administered, the inhibitor was effective in inhibiting HDAC activity. The data suggest that phosphorylated cAMP-response element-binding protein participates in the establishment of conditioned place preference, but not in the reduction of nicotine-preference in response to phenylbutyrate. Moreover, striatal expression of HDAC2 in response to phenylbutyrate mirrored the behavioral effects of the inhibitor, suggesting that HDAC2 is involved in promoting synaptic plasticity underlying the preference for nicotine.
Collapse
Affiliation(s)
- Veronica Pastor
- Departamento de Fisiología e Instituto de Biología Celular, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
106
|
Sandner G, Host L, Angst MJ, Guiberteau T, Guignard B, Zwiller J. The HDAC Inhibitor Phenylbutyrate Reverses Effects of Neonatal Ventral Hippocampal Lesion in Rats. Front Psychiatry 2011; 1:153. [PMID: 21423460 PMCID: PMC3059629 DOI: 10.3389/fpsyt.2010.00153] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 12/17/2010] [Indexed: 12/11/2022] Open
Abstract
Recent evidence suggests that epigenetic mechanisms play a role in psychiatric diseases. In this study, we considered rats with neonatal ventral hippocampal lesions (NVHL) that are currently used for modeling neurodevelopmental aspects of schizophrenia. Contribution of epigenetic regulation to the effects of the lesion was investigated, using a histone deacetylase (HDAC) inhibitor. Lesioned or sham-operated rats were treated with the general HDAC inhibitor phenylbutyrate, which was injected daily from the day after surgery until adulthood. Changes in the volume of the lesion were monitored by magnetic resonance imaging (MRI). Anxiety was analyzed in the Plus Maze Test. Hypersensitivity of the dopaminergic system was evaluated by measuring the locomotor response to apomorphine. An associative conditioning test rewarded with food was used to evaluate learning abilities. The volume of the lesions expanded long after surgery, independently of the treatment, as assessed by MRI. Removal of the ventral hippocampus reduced anxiety, and this remained unchanged when animals were treated with phenylbutyrate. In contrast, NVHL rats' hypersensitivity to apomorphine and deterioration of the associative learning were reduced by the treatment. Global HDAC activity, which was increased in the prefrontal cortex of lesioned non-treated rats, was found to be reversed by HDAC inhibition. The study provides evidence that chromatin remodeling may be useful for limiting behavioral consequences due to lesioning of the ventral hippocampus at an early age. This represents a novel approach for treating disorders resulting from insults occurring during brain development.
Collapse
Affiliation(s)
- Guy Sandner
- U666 INSERM, Faculté de Médecine, Université de StrasbourgStrasbourg, France
| | - Lionel Host
- UMR 7237, Centre National de la Recherche Scientifique, Faculté de Psychologie, Université de StrasbourgStrasbourg, France
| | - Marie-Josée Angst
- U666 INSERM, Faculté de Médecine, Université de StrasbourgStrasbourg, France
| | - Thierry Guiberteau
- UMR 7191, Centre National de la Recherche Scientifique/Université Louis Pasteur, Faculté de Médecine, Université de StrasbourgStrasbourg, France
| | - Blandine Guignard
- UMR 7191, Centre National de la Recherche Scientifique/Université Louis Pasteur, Faculté de Médecine, Université de StrasbourgStrasbourg, France
| | - Jean Zwiller
- UMR 7237, Centre National de la Recherche Scientifique, Faculté de Psychologie, Université de StrasbourgStrasbourg, France
| |
Collapse
|
107
|
Alvarenga TA, Ribeiro DA, Araujo P, Hirotsu C, Mazaro-Costa R, Costa JL, Battisti MC, Tufik S, Andersen ML. Sleep loss and acute drug abuse can induce DNA damage in multiple organs of mice. Hum Exp Toxicol 2010; 30:1275-81. [DOI: 10.1177/0960327110388535] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The purpose of the present study was to characterize the genetic damage induced by paradoxical sleep deprivation (PSD) in combination with cocaine or ecstasy (3,4-methylenedioxymethamphetamine; MDMA) in multiple organs of male mice using the single cell gel (comet) assay. C57BL/6J mice were submitted to PSD by the platform technique for 72 hours, followed by drug administration and evaluation of DNA damage in peripheral blood, liver and brain tissues. Cocaine was able to induce genetic damage in the blood, brain and liver cells of sleep-deprived mice at the majority of the doses evaluated. Ecstasy also induced increased DNA migration in peripheral blood cells for all concentrations tested. Analysis of damaged cells by the tail moment data suggests that ecstasy is a genotoxic chemical at the highest concentrations tested, inducing damage in liver or brain cells after sleep deprivation in mice. Taken together, our results suggest that cocaine and ecstasy/MDMA act as potent genotoxins in multiple organs of mice when associated with sleep loss.
Collapse
Affiliation(s)
- TA Alvarenga
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo Brazil
| | - DA Ribeiro
- Departamento de Biociencias, Universidade Federal de São Paulo, Santos, Brazil
| | - P Araujo
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo Brazil
| | - C Hirotsu
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo Brazil
| | - R Mazaro-Costa
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo Brazil
| | - JL Costa
- Instrumental Analysis Laboratory, Criminalistic Institute, São Paulo, Brazil
| | - MC Battisti
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo Brazil
| | - S Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo Brazil
| | - ML Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo Brazil
| |
Collapse
|
108
|
Ben-Ari Y, Spitzer NC. Phenotypic checkpoints regulate neuronal development. Trends Neurosci 2010; 33:485-92. [PMID: 20864191 DOI: 10.1016/j.tins.2010.08.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 08/22/2010] [Accepted: 08/22/2010] [Indexed: 12/22/2022]
Abstract
Nervous system development proceeds by sequential gene expression mediated by cascades of transcription factors in parallel with sequences of patterned network activity driven by receptors and ion channels. These sequences are cell type- and developmental stage-dependent and modulated by paracrine actions of substances released by neurons and glia. How and to what extent these sequences interact to enable neuronal network development is not understood. Recent evidence demonstrates that CNS development requires intermediate stages of differentiation providing functional feedback that influences gene expression. We suggest that embryonic neuronal functions constitute a series of phenotypic checkpoint signatures; neurons failing to express these functions are delayed or developmentally arrested. Such checkpoints are likely to be a general feature of neuronal development and constitute presymptomatic signatures of neurological disorders when they go awry.
Collapse
Affiliation(s)
- Yehezkel Ben-Ari
- Institut de Neurobiologie de la Méditerranée (INMED), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 901, Parc Scientifique de Luminy, Marseille CEDEX 09, France.
| | | |
Collapse
|
109
|
Sadri-Vakili G, Kumaresan V, Schmidt HD, Famous KR, Chawla P, Vassoler FM, Overland RP, Xia E, Bass CE, Terwilliger EF, Pierce RC, Cha JHJ. Cocaine-induced chromatin remodeling increases brain-derived neurotrophic factor transcription in the rat medial prefrontal cortex, which alters the reinforcing efficacy of cocaine. J Neurosci 2010; 30:11735-44. [PMID: 20810894 PMCID: PMC2943400 DOI: 10.1523/jneurosci.2328-10.2010] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 06/28/2010] [Accepted: 07/11/2010] [Indexed: 02/07/2023] Open
Abstract
Cocaine self-administration alters patterns of gene expression in the brain that may underlie cocaine-induced neuronal plasticity. In the present study, male Sprague Dawley rats were allowed to self-administer cocaine (0.25 mg/infusion) 2 h/d for 14 d, followed by 7 d of forced abstinence. Compared with yoked saline control rats, cocaine self-administration resulted in increased brain-derived neurotrophic factor (BDNF) protein levels in the rat medial prefrontal cortex (mPFC). To examine the functional relevance of this finding, cocaine self-administration maintained under a progressive ratio schedule of reinforcement was assessed after short hairpin RNA-induced suppression of BDNF expression in the mPFC. Decreased BDNF expression in the mPFC increased the cocaine self-administration breakpoint. Next, the effect of cocaine self-administration on specific BDNF exons was assessed; results revealed selectively increased BDNF exon IV-containing transcripts in the mPFC. Moreover, there were significant cocaine-induced increases in acetylated histone H3 (AcH3) and phospho-cAMP response element binding protein (pCREB) association with BDNF promoter IV. In contrast, there was decreased methyl-CpG-binding protein 2 (MeCP2) association with BDNF promoter IV in the mPFC of rats that previously self-administered cocaine. Together, these results indicate that cocaine-induced increases in BDNF promoter IV transcript in the mPFC are driven by increased binding of AcH3 and pCREB as well as decreased MeCP2 binding at this BDNF promoter. Collectively, these results indicate that cocaine self-administration remodels chromatin in the mPFC, resulting in increased expression of BDNF, which appears to represent a compensatory neuroadaptation that reduces the reinforcing efficacy of cocaine.
Collapse
Affiliation(s)
- Ghazaleh Sadri-Vakili
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129-4404, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Bredy TW, Sun YE, Kobor MS. How the epigenome contributes to the development of psychiatric disorders. Dev Psychobiol 2010; 52:331-42. [PMID: 20127889 DOI: 10.1002/dev.20424] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Epigenetics commonly refers to the developmental process by which cellular traits are established and inherited without a change in DNA sequence. These mechanisms of cellular memory also orchestrate gene expression in the adult brain and recent evidence suggests that the "epigenome" represents a critical interface between environmental signals, activation, repression and maintenance of genomic responses, and persistent behavior. We here review the current state of knowledge regarding the contribution of the epigenome toward the development of psychiatric disorders.
Collapse
Affiliation(s)
- Timothy W Bredy
- Queensland Brain Institute, University of Queensland, Brisbane, QC 4072, Australia.
| | | | | |
Collapse
|
111
|
Aguilera O, Fernández AF, Muñoz A, Fraga MF. Epigenetics and environment: a complex relationship. J Appl Physiol (1985) 2010; 109:243-51. [PMID: 20378707 DOI: 10.1152/japplphysiol.00068.2010] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The epigenomes of higher organisms constantly change over time. Many of these epigenetic changes are necessary to direct normal cellular development and differentiation in the developing organism. However, developmental abnormalities may occur in response to inappropriate epigenetic signaling that occurs secondarily to still poorly understood causes. In addition to genetic and stochastic influences on epigenetic processes, epigenetic variation can arise as a consequence of environmental factors. Here we review the effects of such environmental factors on the epigenomes of higher organisms. We discuss the possible impact of epigenetic changes on physiological and pathophysiological processes, depending in part on whether these changes occur during embryonic development or adulthood.
Collapse
Affiliation(s)
- Oscar Aguilera
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | |
Collapse
|
112
|
Host L, Anglard P, Romieu P, Thibault C, Dembele D, Aunis D, Zwiller J. Inhibition of histone deacetylases in rats self-administering cocaine regulates lissencephaly gene-1 and reelin gene expression, as revealed by microarray technique. J Neurochem 2010; 113:236-47. [DOI: 10.1111/j.1471-4159.2010.06591.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
113
|
Abstract
Substance use disorder is a chronic condition of compulsive drug seeking and use that is mediated by stable changes in central reward pathways. Repeated use of abused drugs causes persistent alterations in gene expression responsible for the long-term behavioral and structural changes. Recently, it has been suggested that epigenetic mechanisms are responsible in part for these drug-induced changes in gene expression. One of the alluring aspects of epigenetic regulation of gene expression is that epigenetic mechanisms may provide transient and potentially stable conditions that in turn may ultimately participate in the molecular mechanisms required for neuronal changes subserving long-lasting changes in behavior. This review describes epigenetic mechanisms of gene regulation and then discusses the emerging role of epigenetics in drug-induced plasticity and behavior. Understanding these mechanisms that establish and maintain drug-dependent plasticity changes may lead to deeper understanding of substance use disorders as well as novel approaches to treatment.
Collapse
Affiliation(s)
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, 106 Bonney Research Lab, Irvine, CA 92697-3800 USA
| |
Collapse
|
114
|
Carouge D, Host L, Aunis D, Zwiller J, Anglard P. CDKL5 is a brain MeCP2 target gene regulated by DNA methylation. Neurobiol Dis 2010; 38:414-24. [PMID: 20211261 DOI: 10.1016/j.nbd.2010.02.014] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 02/21/2010] [Accepted: 02/27/2010] [Indexed: 02/08/2023] Open
Abstract
Rett syndrome and its "early-onset seizure" variant are severe neurodevelopmental disorders associated with mutations within the MECP2 and the CDKL5 genes. Antidepressants and drugs of abuse induce the expression of the epigenetic factor MeCP2, thereby influencing chromatin remodeling. We show that increased MeCP2 levels resulted in the repression of Cdkl5 in rat brain structures in response to cocaine, as well as in cells exposed to serotonin, or overexpressing MeCP2. In contrast, Cdkl5 was induced by siRNA-mediated knockdown of Mecp2 and by DNA-methyltransferase inhibitors, demonstrating its regulation by MeCP2 and by DNA methylation. Cdkl5 gene methylation and its methylation-dependent binding to MeCP2 were increased in the striatum of cocaine-treated rats. Our data demonstrate that Cdkl5 is a MeCP2-repressed target gene providing a link between genes the mutation of which generates overlapping symptoms. They highlight DNA methylation changes as a potential mechanism participating in the long-term plasticity triggered by pharmacological agents.
Collapse
Affiliation(s)
- Delphine Carouge
- INSERM, U575, Université de Strasbourg, Centre de Neurochimie, 67084 Strasbourg, France
| | | | | | | | | |
Collapse
|
115
|
Chronic cocaine-induced H3 acetylation and transcriptional activation of CaMKIIalpha in the nucleus accumbens is critical for motivation for drug reinforcement. Neuropsychopharmacology 2010; 35:913-28. [PMID: 20010550 PMCID: PMC3055366 DOI: 10.1038/npp.2009.193] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The regulation of gene expression in the brain reward regions is known to contribute to the pathogenesis and persistence of drug addiction. Increasing evidence suggests that the regulation of gene transcription is mediated by epigenetic mechanisms that alter the chromatin structure at specific gene promoters. To better understand the involvement of epigenetic regulation in drug reinforcement properties, rats were subjected to cocaine self-administration paradigm. Daily histone deacetylase (HDAC) inhibitor infusions in the shell of the nucleus accumbens (NAc) caused an upward shift in the dose-response curve under fixed-ratio schedule and increased the break point under progressive-ratio schedule, indicating enhanced motivation for self-administered drug. The effect of the HDAC inhibitor is attributed to the increased elevation of histone acetylation induced by chronic, but not acute, cocaine experience. In contrast, neutralizing the chronic cocaine-induced increase in histone modification by the bilateral overexpression of HDAC4 in the NAc shell reduced drug motivation. The association between the motivation for cocaine and the transcriptional activation of addiction-related genes by H3 acetylation in the NAc shell was analyzed. Among the genes activated by chronic cocaine experiences, the expression of CaMKIIalpha, but not CaMKIIbeta, correlated positively with motivation for the drug. Lentivirus-mediated shRNA knockdown experiments showed that CaMKIIalpha, but not CaMKIIbeta, in the NAc shell is essential for the maintenance of motivation to self-administered cocaine. These findings suggest that chronic drug-use-induced transcriptional activation of genes, such as CaMKIIalpha, modulated by H3 acetylation in the NAc is a critical regulatory mechanism underlying motivation for drug reinforcement.
Collapse
|
116
|
Sleiman SF, Basso M, Mahishi L, Kozikowski AP, Donohoe ME, Langley B, Ratan RR. Putting the 'HAT' back on survival signalling: the promises and challenges of HDAC inhibition in the treatment of neurological conditions. Expert Opin Investig Drugs 2010; 18:573-84. [PMID: 19388875 DOI: 10.1517/13543780902810345] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Decreased histone acetyltransferase activity and transcriptional dysfunction have been implicated in almost all neurodegenerative conditions. Increasing net histone acetyltransferase activity through inhibition of the histone deacetylases (HDACs) has been shown to be an effective strategy to delay or halt progression of neurological disease in cellular and rodent models. These findings have provided firm rationale for Phase I and Phase II clinical trials of HDAC inhibitors in Huntington's disease, spinal muscular atrophy, and Freidreich's ataxia. In this review, we discuss the current findings and promise of HDAC inhibition as a strategy for treating neurological disorders. Despite the fact that HDAC inhibitors are in an advanced stage of development, we suggest other approaches to modulating HDAC function that may be less toxic and more efficacious than the canonical agents developed so far.
Collapse
Affiliation(s)
- Sama F Sleiman
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, 10605 NY, USA.
| | | | | | | | | | | | | |
Collapse
|
117
|
Alvarenga TA, Andersen ML, Ribeiro DA, Araujo P, Hirotsu C, Costa JL, Battisti MC, Tufik S. Single exposure to cocaine or ecstasy induces DNA damage in brain and other organs of mice. Addict Biol 2010; 15:96-9. [PMID: 19878142 DOI: 10.1111/j.1369-1600.2009.00179.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We evaluated the overall genetic damage induced by different doses of cocaine and MDMA (3,4-Methylenedioxymethamphetamine) in several organs. One hour after intraperitoneal drug administration, mice were euthanized; peripheral blood, liver and brain were collected, and the cellular suspensions were used for the single cell gel (comet) assay. We determined that all doses of cocaine and MDMA tested were able to induce DNA damage in blood cells. Extensive genotoxic damage was induced by cocaine or MDMA at the highest doses used in liver cells. Brain cells were affected by all doses administrated. These findings demonstrate that cocaine and MDMA are potent genotoxins.
Collapse
|
118
|
|
119
|
Abstract
Alterations in gene expression are implicated in the pathogenesis of several neuropsychiatrie disorders, including drug addiction and depression, increasing evidence indicates that changes in gene expression in neurons, in the context of animal models of addiction and depression, are mediated in part by epigenetic mechanisms that alter chromatin structure on specific gene promoters. This review discusses recent findings from behavioral, molecular, and bioinformatic approaches that are being used to understand the complex epigenetic regulation of gene expression in brain by drugs of abuse and by stress. These advances promise to open up new avenues for improved treatments of these disorders.
Collapse
Affiliation(s)
- William Renthal
- Medical Scientist Training Program, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
120
|
Histone modifications, DNA methylation, and schizophrenia. Neurosci Biobehav Rev 2009; 34:882-8. [PMID: 19879893 DOI: 10.1016/j.neubiorev.2009.10.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 09/22/2009] [Accepted: 10/21/2009] [Indexed: 02/08/2023]
Abstract
Studies have demonstrated that several schizophrenia candidate genes are especially susceptible to changes in transcriptional activity as a result of histone modifications and DNA methylation. Increased expression of epigenetic enzymes which generally reduce transcription have been reported in schizophrenia postmortem brain samples. An abnormal chromatin state leading to reduced candidate gene expression can be explained by aberrant coordination of epigenetic mechanisms in schizophrenia. Dynamic epigenetic processes are difficult to study using static measures such as postmortem brain samples. Therefore, we have developed a model using cultured peripheral blood mononuclear cells (PBMCs) capable of pharmacologically probing these processes in human subjects. This approach has revealed several promising findings indicating that schizophrenia subject PBMC chromatin may be less capable of responding to agents which normally 'open' chromatin. We suggest that the ability to appropriately modify chromatin structure may be a factor in treatment response. Several pharmacological approaches for targeting epigenetic processes are reviewed.
Collapse
|
121
|
Macedo CE, Angst MJ, Guiberteau T, Brasse D, O'Brien TJ, Sandner G. Acoustic hypersensitivity in adult rats after neonatal ventral hippocampus lesions. Behav Brain Res 2009; 207:161-8. [PMID: 19818810 DOI: 10.1016/j.bbr.2009.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 09/25/2009] [Accepted: 10/02/2009] [Indexed: 10/20/2022]
Abstract
Rats with a bilateral neonatal ventral hippocampus lesion (NVHL) are used as models of neurobiological aspects of schizophrenia. In view of their decreased number of GABAergic interneurons, we hypothesized that they would show increased reactivity to acoustic stimuli. We systematically characterized the acoustic reactivity of NVHL rats and sham operated controls. They were behaviourally observed during a loud white noise. A first cohort of 7 months' old rats was studied. Then the observations were reproduced in a second cohort of the same age after characterizing the reactivity of the same rats to dopaminergic drugs. A third cohort of rats was studied at 2, 3, 4, 5 and 6 months. In subsets of lesioned and control rats, inferior colliculus auditory evoked potentials were recorded. A significant proportion of rats (50-62%) showed aberrant audiogenic responses with explosive wild running resembling the initial phase of audiogenic seizures. This was not correlated with their well-known enhanced reactivity to dopaminergic drugs. The proportion of rats showing this strong reaction increased with rats' age. After the cessation of the noise, NVHL rats showed a long freezing period that did neither depend on the size of the lesion nor on the rats' age. The initial negative deflection of the auditory evoked potential was enhanced in the inferior colliculus of only NVHL rats that displayed wild running. Complementary anatomical investigations using X-ray scans in the living animal, and alizarin red staining of brain slices, revealed a thin layer of calcium deposit close to the medial geniculate nuclei in post-NVHL rats, raising the possibility that this may contribute to the hyper-reactivity to sounds seen in these animals. The findings of this study provide complementary information with potential relevance for the hyper-reactivity noted in patients with schizophrenia, and therefore a tool to investigate the underlying biology of this endophenotype.
Collapse
Affiliation(s)
- Carlos Eduardo Macedo
- Laboratorio de Psicobiologia, Universidade de São Paulo (USP), Ribeirão Preto, Brazil
| | | | | | | | | | | |
Collapse
|
122
|
Malvaez M, Barrett RM, Wood MA, Sanchis-Segura C. Epigenetic mechanisms underlying extinction of memory and drug-seeking behavior. Mamm Genome 2009; 20:612-23. [PMID: 19789849 PMCID: PMC3157916 DOI: 10.1007/s00335-009-9224-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 08/25/2009] [Indexed: 12/28/2022]
Abstract
An increasing body of evidence shows that structural modifications of chromatin, the DNA-protein complex that packages genomic DNA, do not only participate in maintaining cellular memory (e.g., cell fate), but they may also underlie the strengthening and maintenance of synaptic connections required for long-term changes in behavior. Accordingly, epigenetics has become a central topic in several neurobiology fields such as memory, drug addiction, and several psychiatric and mental disorders. This interest is justified as dynamic chromatin modifications may provide not only transient but also stable (or even potentially permanent) epigenetic marks to facilitate, maintain, or block transcriptional processes, which in turn may participate in the molecular neural adaptations underlying behavioral changes. Through epigenetic mechanisms the genome may be indexed in response to environmental signals, resulting in specific neural modifications that largely determine the future behavior of an organism. In this review we discuss recent advances in our understanding of how epigenetic mechanisms contribute to the formation of long-term memory and drug-seeking behavior and potentially how to apply that knowledge to the extinction of memory and drug-seeking behavior.
Collapse
Affiliation(s)
- Melissa Malvaez
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697-3800, USA
| | - Ruth M. Barrett
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697-3800, USA
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697-3800, USA
| | | |
Collapse
|
123
|
Abstract
Studies of environmental challenges, such as hazardous air pollutants, nonmutagenic toxins, diet choice, and maternal behavioral patterns, reveal changes in gene expression patterns, DNA methylation, and histone modifications that are in causal association with exogenous exposures. In this article we summarize some of the recent advances in the field of environmental epigenetics and highlight seminal studies that implicate in utero exposures as causative agents in altering not only the epigenome of the exposed gestation, but that of subsequent generations. Current studies of the effects of maternal behavior, exposure to environmental toxins, and exposure to maternal diet and an altered gestational milieu are summarized.
Collapse
Affiliation(s)
- Melissa A Suter
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|
124
|
Thomas EA. Focal nature of neurological disorders necessitates isotype-selective histone deacetylase (HDAC) inhibitors. Mol Neurobiol 2009; 40:33-45. [PMID: 19396637 DOI: 10.1007/s12035-009-8067-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Accepted: 03/31/2009] [Indexed: 11/25/2022]
Abstract
Histone deacetylase (HDAC) inhibitors represent a promising new avenue of therapeutic options for a range of neurological disorders. Within any particular neurological disorder, neuronal damage or death is not widespread; rather, particular brain regions are preferentially affected. Different disorders exhibit distinct focal pathologies. Hence, understanding the region-specific effects of HDAC inhibitors is essential for targeting appropriate brain areas and reducing toxicity in unaffected areas. The outcome of HDAC inhibition depends on several factors, including the diversity in the central nervous system expression of HDAC enzymes, selectivity of a given HDAC inhibitor for different HDAC enzymes, and the presence or absence of cofactors necessary for enzyme function. This review will summarize brain regions associated with various neurological disorders and factors affecting the consequences of HDAC inhibition.
Collapse
Affiliation(s)
- Elizabeth A Thomas
- Department of Molecular Biology, The Scripps Research Institute, MB-10, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| |
Collapse
|
125
|
Gundersen BB, Blendy JA. Effects of the histone deacetylase inhibitor sodium butyrate in models of depression and anxiety. Neuropharmacology 2009; 57:67-74. [PMID: 19393671 DOI: 10.1016/j.neuropharm.2009.04.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 04/02/2009] [Accepted: 04/15/2009] [Indexed: 11/27/2022]
Abstract
Histone modification, which affects the rate of transcription without altering DNA sequence, occurs in response to various psychiatric drugs and in several models of psychiatric disease. As increases in histone acetylation have been seen after treatment with antidepressants, we investigated whether directly increasing histone acetylation using a histone deacetylase inhibitor would have antidepressant effects. We administered sodium butyrate (NaB, 100 mg/kg, i.p.) to mice acutely (3 injections over 24 h) or chronically (twice daily for 21 days) and subjected them to a number of behavioral tests of antidepressant response. This dose of NaB had no effect on overall locomotor activity after either acute or chronic treatment. Acutely treated mice showed an increase in immobility in the forced-swim test (FST) and an increase in latency to consume in the novel environment of the novelty-induced hypophagia (NIH) paradigm, an anxiogenic effect. The effect of NaB on anxiety did not generalize to another test, the elevated zero maze, where it had no effect. Chronic treatment with NaB had no effect on latency to consume in the NIH or immobility in the FST. However, this dose did alter histone acetylation in the hippocampus. While H4 acetylation increased in the hippocampus 30 min following acute NaB, chronic treatment caused a decrease in AcH4. There were no changes in AcH3 following either treatment. While changes in chromatin structure may be involved in the mechanism of action of antidepressant drugs, these data suggest that increasing histone acetylation pharmacologically is not sufficient to produce antidepressant effects.
Collapse
Affiliation(s)
- Brigitta B Gundersen
- Department of Pharmacology, University of Pennysylvania, School of Medicine, Translational Research Labs, 125 S. 31st St, Philadelphia, PA 19104, USA
| | | |
Collapse
|