1
|
Liu Y, Wang XQ, Zhang P, Haghparast A, He WB, Zhang JJ. Research progress of DNA methylation on the regulation of substance use disorders and the mechanisms. Front Cell Neurosci 2025; 19:1566001. [PMID: 40230379 PMCID: PMC11994631 DOI: 10.3389/fncel.2025.1566001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Drug abuse can damage the central nervous system and lead to substance use disorder (SUD). SUD is influenced by both genetic and environmental factors. Genes determine an individual's susceptibility to drug, while the dysregulation of epigenome drives the abnormal transcription processes, promoting the development of SUD. One of the most widely studied epigenetic mechanisms is DNA methylation, which can be inherited stably. In ontogeny, DNA methylation pattern is dynamic. DNA dysmethylation is prevalent in drug-related psychiatric disorders, resulting in local hypermethylation and transcriptional silencing of related genes. In this review, we summarize the role and regulatory mechanisms of DNA methylation in cocaine, opioids, and methamphetamine in terms of drug exposure, addiction memory, withdrawal relapse, intergenerational inheritance, and focus on cell-specific aspects of the studies with a view to suggesting possible therapeutic regimens for targeting methylation in both human and animal research.
Collapse
Affiliation(s)
- Ya Liu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Xiao-Qian Wang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Peng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Abbas Haghparast
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Wen-Bin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Jian-Jun Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
2
|
Fang T, Liu MN, Liu MQ, Tian XY, Zhang XJ, Liu F, Hao W, Wu N, Li H, Li J. A preliminary study on the association of single nucleotide polymorphisms and methylation of dopamine system-related genes with psychotic symptoms in patients with methamphetamine use disorder. Eur J Neurosci 2024; 59:1428-1440. [PMID: 38151046 DOI: 10.1111/ejn.16238] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/29/2023]
Abstract
Methamphetamine use disorder (MAUD) can substantially jeopardize public security due to its high-risk social psychology and behaviour. Given that the dopamine reward system is intimately correlated with MAUD, we investigated the association of single nucleotide polymorphisms (SNPs), as well as methylation status of dopamine receptor type 4 (DRD4), catechol-O-methyltransferase (COMT) genes, and paranoid and motor-impulsive symptoms in MAUD patients. A total of 189 MAUD patients participated in our study. Peripheral blood samples were used to detect 3 SNPs and 35 CpG units of methylation in the DRD4 gene promoter region and 5 SNPs and 39 CpG units in the COMT gene. MAUD patients with the DRD4 rs1800955 C allele have a lower percentage of paranoid symptoms than those with the rs1800955 TT allele. Individuals with paranoid symptoms exhibited a reduced methylation degree at a particular DRD4 CpG2.3 unit. The interaction of the DRD4 rs1800955 C allele and the reduced DRD4CpG2.3 methylation degree were associated with a lower occurrence of paranoid symptoms. Meanwhile, those with the COMT rs4818 CC allele had lower motor-impulsivity scores in MAUD patients but greater COMT methylation levels in the promoter region and methylation degree at the COMT CpG 51.52 unit. Therefore, based only on the COMT rs4818 CC polymorphism, there was a negative correlation between COMT methylation and motor-impulsive scores. Our preliminary results provide a clue that the combination of SNP genotype and methylation status of the DRD4 and COMT genes serve as biological indicators for the prevalence of relatively high-risk psychotic symptoms in MAUD patients.
Collapse
Affiliation(s)
- Ting Fang
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Meng-Nan Liu
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Meng-Qi Liu
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiao-Yu Tian
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xiao-Jie Zhang
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feng Liu
- Compulsory Detoxification Center of Changsha Public Security Bureau, Changsha, China
| | - Wei Hao
- National Clinical Research Center for Mental Disorders, and Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ning Wu
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Hong Li
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jin Li
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
3
|
De Sa Nogueira D, Bourdy R, Alcala-Vida R, Filliol D, Andry V, Goumon Y, Zwiller J, Romieu P, Merienne K, Olmstead MC, Befort K. Hippocampal Cannabinoid 1 Receptors Are Modulated Following Cocaine Self-administration in Male Rats. Mol Neurobiol 2022; 59:1896-1911. [PMID: 35032317 DOI: 10.1007/s12035-022-02722-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023]
Abstract
Cocaine addiction is a complex pathology inducing long-term neuroplastic changes that, in turn, contribute to maladaptive behaviors. This behavioral dysregulation is associated with transcriptional reprogramming in brain reward circuitry, although the mechanisms underlying this modulation remain poorly understood. The endogenous cannabinoid system may play a role in this process in that cannabinoid mechanisms modulate drug reward and contribute to cocaine-induced neural adaptations. In this study, we investigated whether cocaine self-administration induces long-term adaptations, including transcriptional modifications and associated epigenetic processes. We first examined endocannabinoid gene expression in reward-related brain regions of the rat following self-administered (0.33 mg/kg intravenous, FR1, 10 days) cocaine injections. Interestingly, we found increased Cnr1 expression in several structures, including prefrontal cortex, nucleus accumbens, dorsal striatum, hippocampus, habenula, amygdala, lateral hypothalamus, ventral tegmental area, and rostromedial tegmental nucleus, with most pronounced effects in the hippocampus. Endocannabinoid levels, measured by mass spectrometry, were also altered in this structure. Chromatin immunoprecipitation followed by qPCR in the hippocampus revealed that two activating histone marks, H3K4Me3 and H3K27Ac, were enriched at specific endocannabinoid genes following cocaine intake. Targeting CB1 receptors using chromosome conformation capture, we highlighted spatial chromatin re-organization in the hippocampus, as well as in the nucleus accumbens, suggesting that destabilization of the chromatin may contribute to neuronal responses to cocaine. Overall, our results highlight a key role for the hippocampus in cocaine-induced plasticity and broaden the understanding of neuronal alterations associated with endocannabinoid signaling. The latter suggests that epigenetic modifications contribute to maladaptive behaviors associated with chronic drug use.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France.,Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Romain Bourdy
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Rafael Alcala-Vida
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Dominique Filliol
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Virginie Andry
- Institut Des Neurosciences Cellulaires Et Intégratives (INCI), UPR 3212, CNRS, 8 Allée du Général Rouvillois, 67000, Strasbourg, France
| | - Yannick Goumon
- Institut Des Neurosciences Cellulaires Et Intégratives (INCI), UPR 3212, CNRS, 8 Allée du Général Rouvillois, 67000, Strasbourg, France
| | - Jean Zwiller
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Pascal Romieu
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Karine Merienne
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France
| | - Mary C Olmstead
- Department of Psychology, Center for Neuroscience Studies, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Katia Befort
- Laboratoire de Neurosciences Cognitives Et Adaptatives (LNCA), Centre de La Recherche Nationale Scientifique, Université de Strasbourg, 12 rue Goethe, 67000, Strasbourg, France.
| |
Collapse
|
4
|
Vaillancourt K, Chen GG, Fiori L, Maussion G, Yerko V, Théroux JF, Ernst C, Labonté B, Calipari E, Nestler EJ, Nagy C, Mechawar N, Mash DC, Turecki G. Methylation of the tyrosine hydroxylase gene is dysregulated by cocaine dependence in the human striatum. iScience 2021; 24:103169. [PMID: 34693223 PMCID: PMC8517202 DOI: 10.1016/j.isci.2021.103169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/15/2021] [Accepted: 09/21/2021] [Indexed: 02/01/2023] Open
Abstract
Cocaine dependence is a chronic, relapsing disorder caused by lasting changes in the brain. Animal studies have identified cocaine-related alterations in striatal DNA methylation; however, it is unclear how methylation is related to cocaine dependence in humans. We generated methylomic profiles of the nucleus accumbens using human postmortem brains from a cohort of individuals with cocaine dependence and healthy controls (n = 25 per group). We found hypermethylation in a cluster of CpGs within the gene body of tyrosine hydroxylase (TH), containing a putative binding site for the early growth response 1 (EGR1) transcription factor, which is hypermethylated in the caudate nucleus of cocaine-dependent individuals. We replicated this finding and found it to be specific to striatal neuronal nuclei. Furthermore, this locus demonstrates enhancer activity which is attenuated by methylation and enhanced by EGR1 overexpression. These results suggest that cocaine dependence alters the epigenetic regulation of dopaminergic signaling genes.
Collapse
Affiliation(s)
- Kathryn Vaillancourt
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Gang G. Chen
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Verdun, QC, Canada
| | - Laura Fiori
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Verdun, QC, Canada
| | - Gilles Maussion
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, QC, Canada
| | - Volodymyr Yerko
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Verdun, QC, Canada
| | - Jean-François Théroux
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Verdun, QC, Canada
| | - Carl Ernst
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Benoit Labonté
- Centre de Recherche Cervo, Université Laval, Québec, QC, Canada
| | - Erin Calipari
- Departments of Pharmacology, Molecular Physiology and Biophysics, Psychiatry and Behavioral Sciences; Vanderbilt Center for Addiction Research; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Deborah C. Mash
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Verdun, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
5
|
Saad L, Zwiller J, Kalsbeek A, Anglard P. Epigenetic Regulation of Circadian Clocks and Its Involvement in Drug Addiction. Genes (Basel) 2021; 12:1263. [PMID: 34440437 PMCID: PMC8394526 DOI: 10.3390/genes12081263] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 12/19/2022] Open
Abstract
Based on studies describing an increased prevalence of addictive behaviours in several rare sleep disorders and shift workers, a relationship between circadian rhythms and addiction has been hinted for more than a decade. Although circadian rhythm alterations and molecular mechanisms associated with neuropsychiatric conditions are an area of active investigation, success is limited so far, and further investigations are required. Thus, even though compelling evidence connects the circadian clock to addictive behaviour and vice-versa, yet the functional mechanism behind this interaction remains largely unknown. At the molecular level, multiple mechanisms have been proposed to link the circadian timing system to addiction. The molecular mechanism of the circadian clock consists of a transcriptional/translational feedback system, with several regulatory loops, that are also intricately regulated at the epigenetic level. Interestingly, the epigenetic landscape shows profound changes in the addictive brain, with significant alterations in histone modification, DNA methylation, and small regulatory RNAs. The combination of these two observations raises the possibility that epigenetic regulation is a common plot linking the circadian clocks with addiction, though very little evidence has been reported to date. This review provides an elaborate overview of the circadian system and its involvement in addiction, and we hypothesise a possible connection at the epigenetic level that could further link them. Therefore, we think this review may further improve our understanding of the etiology or/and pathology of psychiatric disorders related to drug addiction.
Collapse
Affiliation(s)
- Lamis Saad
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands;
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jean Zwiller
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - Andries Kalsbeek
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands;
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Patrick Anglard
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- Institut National de la Santé et de la Recherche Médicale (INSERM), 75013 Paris, France
| |
Collapse
|
6
|
Saad L, Kalsbeek A, Zwiller J, Anglard P. Rhythmic Regulation of DNA Methylation Factors and Core-Clock Genes in Brain Structures Activated by Cocaine or Sucrose: Potential Role of Chromatin Remodeling. Genes (Basel) 2021; 12:genes12081195. [PMID: 34440369 PMCID: PMC8392220 DOI: 10.3390/genes12081195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/25/2022] Open
Abstract
The circadian system interacts with the mesocorticolimbic reward system to modulate reward and memory in a time-of-day dependent manner. The circadian discrimination of reward, however, remains difficult to address between natural reinforcers and drugs of abuse. Circadian rhythms control cocaine sensitization and conversely cocaine causes long-term alteration in circadian periodicity in part through the serotonergic neurotransmission. Since neural circuits activated by cocaine and natural reinforcers do not completely overlap, we compared the effect of cocaine with that of sucrose, a strong reinforcer in rodents, by using passive chronic administration. The expression of fifteen genes playing a major role in DNA methylation (Dnmts, Tets), circadian rhythms (Clock, Bmal1, Per1/2, Cry1/2, Rev-Erbβ, Dbp1), appetite, and satiety (Orexin, Npy) was analyzed in dopamine projection areas like the prefrontal cortex, the caudate putamen, and the hypothalamus interconnected with the reward system. The corresponding proteins of two genes (Orexin, Per2) were examined by IHC. For many factors controlling biological and cognitive functions, striking opposite responses were found between the two reinforcers, notably for genes controlling DNA methylation/demethylation processes and in global DNA methylation involved in chromatin remodeling. The data are consistent with a repression of critical core-clock genes by cocaine, suggesting that, consequently, both agents differentially modulate day/night cycles. Whether observed cocaine and sucrose-induced changes in DNA methylation in a time dependent manner are long lasting or contribute to the establishment of addiction requires further neuroepigenetic investigation. Understanding the mechanisms dissociating drugs of abuse from natural reinforcers remains a prerequisite for the design of selective therapeutic tools for compulsive behaviors.
Collapse
Affiliation(s)
- Lamis Saad
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, 1066 EA Amsterdam, The Netherlands
| | - Andries Kalsbeek
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, 1066 EA Amsterdam, The Netherlands
- Correspondence: (A.K.); or (P.A.)
| | - Jean Zwiller
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- CNRS, Centre National de la Recherche Scientifique, 75016 Paris, France
| | - Patrick Anglard
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Université de Strasbourg, Neuropôle de Strasbourg, 67000 Strasbourg, France; (L.S.); (J.Z.)
- INSERM, Institut National de la Santé et de la Recherche Médicale, 75013 Paris, France
- Correspondence: (A.K.); or (P.A.)
| |
Collapse
|
7
|
Faillace MP, Bernabeu RO. Epigenetic Mechanisms Mediate Nicotine-Induced Reward and Behaviour in Zebrafish. Curr Neuropharmacol 2021; 20:510-523. [PMID: 34279203 PMCID: PMC9608226 DOI: 10.2174/1570159x19666210716112351] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/03/2021] [Accepted: 07/11/2021] [Indexed: 11/26/2022] Open
Abstract
Nicotine induces long-term changes in the neural activity of the mesocorticolimbic reward pathway structures. The mechanisms involved in this process have not been fully characterized. The hypothesis discussed here proposed that epigenetic regulation participates in the installation of persistent adaptations and long-lasting synaptic plasticity generated by nicotine action on the mesolimbic dopamine neurons of zebrafish. The epigenetic mechanisms induced by nicotine entail histone and DNA chemical modifications, which have been described to lead to changes in gene expression. Among the enzymes that catalyze epigenetic chemical modifications, histone deacetylases (HDACs) remove acetyl groups from histones, thereby facilitating DNA relaxation and making DNA more accessible to gene transcription. DNA methylation, which is dependent on DNA methyltransferase (DNMTs) activity, inhibits gene expression by recruiting several methyl binding proteins that prevent RNA polymerase binding to DNA. In zebrafish, phenylbutyrate (PhB), an HDAC inhibitor, abolishes nicotine rewarding properties together with a series of typical reward-associated behaviors. Furthermore, PhB and nicotine alter long- and short-term object recognition memory in zebrafish, respectively. Regarding DNA methylation effects, a methyl group donor L-methionine (L-met) was found to dramatically reduce nicotine-induced conditioned place preference (CPP) in zebrafish. Simultaneous treatment with DNMT inhibitor 5-aza-2’-deoxycytidine (AZA) was found to reverse the L-met effect on nicotine-induced CPP as well as nicotine reward-specific effects on genetic expression in zebrafish. Therefore, pharmacological interventions that modulate epigenetic regulation of gene expression should be considered as a potential therapeutic method to treat nicotine addiction.
Collapse
Affiliation(s)
- Maria Paula Faillace
- Departamento de Fisiología, Facultad de Medicina e Instituto de Fisiología y Biofísica Profesor Bernardo Houssay (IFIBIO-Houssay, CONICET-UBA), Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Ramón O Bernabeu
- Departamento de Fisiología, Facultad de Medicina e Instituto de Fisiología y Biofísica Profesor Bernardo Houssay (IFIBIO-Houssay, CONICET-UBA), Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
8
|
De Sa Nogueira D, Bourdy R, Filliol D, Romieu P, Befort K. Hippocampal mu opioid receptors are modulated following cocaine self-administration in rat. Eur J Neurosci 2021; 53:3341-3349. [PMID: 33811699 DOI: 10.1111/ejn.15217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 03/18/2021] [Indexed: 11/30/2022]
Abstract
Cocaine addiction is a complex pathology induced by long-term brain changes. Understanding the neurochemical changes underlying the reinforcing effects of this drug of abuse is critical for reducing the societal burden of drug addiction. The mu opioid receptor plays a major role in drug reward. This receptor is modulated by chronic cocaine treatment in specific brain structures, but few studies investigated neurochemical adaptations induced by voluntary cocaine intake. In this study, we investigated whether intravenous cocaine-self administration (0.33 mg/kg/injection, fixed-ratio 1 [FR1], 10 days) in rats induces transcriptional and functional changes of the mu opioid receptor in reward-related brain regions. Epigenetic processes with histone modifications were examined for two activating marks, H3K4Me3, and H3K27Ac. We found an increase of mu opioid receptor gene expression along with a potentiation of its functionality in hippocampus of cocaine self-administering animals compared to saline controls. Chromatin immunoprecipitation followed by qPCR revealed no modifications of the histone mark H3K4Me3 and H3K27Ac levels at mu opioid receptor promoter. Our study highlights the hippocampus as an important target to further investigate neuroadaptive processes leading to cocaine addiction.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Romain Bourdy
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Dominique Filliol
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Pascal Romieu
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Katia Befort
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
9
|
Hong Q, Xu W, Lin Z, Liu J, Chen W, Zhu H, Lai M, Zhuang D, Xu Z, Fu D, Zhou W, Liu H. Role of GABRD Gene Methylation in the Nucleus Accumbens in Heroin-Seeking Behavior in Rats. Front Pharmacol 2021; 11:612200. [PMID: 33551813 PMCID: PMC7859445 DOI: 10.3389/fphar.2020.612200] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/27/2020] [Indexed: 11/13/2022] Open
Abstract
Epigenetic modifications such as DNA methylation play important roles in regulating gene expression and may mediate neuroplasticity and lead to drug-induced aberrant behaviors. Although several brain regions and neurobiological mechanisms have been suggested to be involved in these processes, there is remarkably little known about the effects of DNA methylation on heroin-seeking behavior. Using a Sprague-Dawley rat model, we show that heroin self-administration resulted in gamma-aminobutyric acid type A receptor subunit delta (GABRD) gene hypomethylation, which was associated with transcriptional upregulation of GABRD in the nucleus accumbens (NAc). Systemic l-methionine (MET) administration significantly strengthened the reinstatement of heroin-seeking behavior induced by heroin priming, whereas intra-NAc injections of the DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine (5-Aza-dC) had the opposite effect on heroin-seeking. Meanwhile, 5-Aza-dC treatment decreased DNA methylation and upregulated the expression of GABRD in the NAc, whereas MET had the opposite effect. Our results also reveal that 5-Aza-dC might alter the methylation landscape of the GABRD gene by directly repressing DNMT1 and DNMT3A expression. Furthermore, reinstatement of heroin-seeking behavior was significantly inhibited by directly overexpressing GABRD and remarkably reinforced by GABRD gene silencing in the NAc. Collectively, these results suggest that targeting the GABRD gene and its methylation might represent a novel pharmacological strategy for treating heroin addiction and relapse.
Collapse
Affiliation(s)
- Qingxiao Hong
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| | - Wenjin Xu
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| | - Zi Lin
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
| | - Jing Liu
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
| | - Weisheng Chen
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| | - Huaqiang Zhu
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| | - Miaojun Lai
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| | - Dingding Zhuang
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| | - Zemin Xu
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| | - Dan Fu
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| | - Wenhua Zhou
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| | - Huifen Liu
- Laboratory of Behavioral Neuroscience, Ningbo Kangning Hospital, Ningbo Institute of Microcirculation and Henbane, School of Medicine, Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, China
| |
Collapse
|
10
|
Pisera-Fuster A, Zwiller J, Bernabeu R. Methionine Supplementation Abolishes Nicotine-Induced Place Preference in Zebrafish: a Behavioral and Molecular Analysis. Mol Neurobiol 2021; 58:2590-2607. [PMID: 33475949 DOI: 10.1007/s12035-020-02260-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/10/2020] [Indexed: 12/26/2022]
Abstract
In zebrafish, nicotine is known to regulate sensitivity to psychostimulants via epigenetic mechanisms. Little however is known about the regulation of addictive-like behavior by DNA methylation processes. To evaluate the influence of DNA methylation on nicotine-induced conditioned place preference (CPP), zebrafish were exposed to methyl supplementation through oral L-methionine (Met) administration. Met was found to reduce dramatically nicotine-induced CPP as well as behaviors associated with drug reward. The reduction was associated with the upregulation of DNA methyltransferases (DNMT1 and 3) as well as with the downregulation of methyl-cytosine dioxygenase-1 (TET1) and of nicotinic receptor subunits. Met also increased the expression of histone methyltransferases in nicotine-induced CPP groups. It reversed the nicotine-induced reduction in the methylation at α7 and NMDAR1 gene promoters. Treatment with the DNMT inhibitor 5-aza-2'-deoxycytidine (AZA) was found to reverse the effects of Met in structures of the reward pathway. Interestingly, Met did not modify the amount of the phospho-form of CREB (pCREB), a key factor establishing nicotine conditioning, whereas AZA increased pCREB levels. Our data suggest that nicotine-seeking behavior is partially dependent on DNA methylation occurring probably at specific gene loci, such as α7 and NMDAR1 receptor gene promoters. Overall, they suggest that Met should be considered as a potential therapeutic drug to treat nicotine addiction.
Collapse
Affiliation(s)
- Antonella Pisera-Fuster
- Department of Physiology and Institute of Physiology and Biophysics, School of Medicine, University of Buenos Aires, Paraguay 2155 7thfloor (C1121ABG), Ciudad Autónoma de Buenos Aires, Argentina
| | - Jean Zwiller
- Laboratoire de Neurosciences Cognitives et Adaptatives, Université de Strasbourg, Strasbourg, France
| | - Ramon Bernabeu
- Department of Physiology and Institute of Physiology and Biophysics, School of Medicine, University of Buenos Aires, Paraguay 2155 7thfloor (C1121ABG), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
11
|
Abstract
This review explores how different classes of drugs, including those with therapeutic and abuse potential, alter brain functions and behavior via the epigenome. Epigenetics, in its simplest interpretation, is the study of the regulation of a genes' transcriptional potential. The epigenome is established during development but is malleable throughout life by a wide variety of drugs, with both clinical utility and abuse potential. An epigenetic effect can be central to the drug's therapeutic or abuse potential, or it can be independent from the main effect but nevertheless produce beneficial or adverse side effects. Here, I discuss the various epigenetic effects of main pharmacological drug classes, including antidepressants, antiepileptics, and drugs of abuse.
Collapse
Affiliation(s)
- Miklos Toth
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA;
| |
Collapse
|
12
|
Franco-Enzástiga Ú, García G, Murbartián J, González-Barrios R, Salinas-Abarca AB, Sánchez-Hernández B, Tavares-Ferreira D, Herrera LA, Barragán-Iglesias P, Delgado-Lezama R, Price TJ, Granados-Soto V. Sex-dependent pronociceptive role of spinal α 5 -GABA A receptor and its epigenetic regulation in neuropathic rodents. J Neurochem 2020; 156:897-916. [PMID: 32750173 DOI: 10.1111/jnc.15140] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/26/2020] [Accepted: 07/22/2020] [Indexed: 12/23/2022]
Abstract
Extrasynaptic α5 -subunit containing GABAA (α5 -GABAA ) receptors participate in chronic pain. Previously, we reported a sex difference in the action of α5 -GABAA receptors in dysfunctional pain. However, the underlying mechanisms remain unknown. The aim of this study was to examine this sexual dimorphism in neuropathic rodents and the mechanisms involved. Female and male Wistar rats or ICR mice were subjected to nerve injury followed by α5 -GABAA receptor inverse agonist intrathecal administration, L-655,708. The drug produced an antiallodynic effect in nerve-injured female rats and mice, and a lower effect in males. We hypothesized that changes in α5 -GABAA receptor, probably influenced by hormonal and epigenetic status, might underlie this sex difference. Thus, we performed qPCR and western blot. Nerve injury increased α5 -GABAA mRNA and protein in female dorsal root ganglia (DRG) and decreased them in DRG and spinal cord of males. To investigate the hormonal influence over α5 -GABAA receptor actions, we performed nerve injury to ovariectomized rats and reconstituted them with 17β-estradiol (E2). Ovariectomy abrogated L-655,708 antiallodynic effect and E2 restored it. Ovariectomy decreased α5 -GABAA receptor and estrogen receptor α protein in DRG of neuropathic female rats, while E2 enhanced them. Since DNA methylation might contribute to α5 -GABAA receptor down-regulation in males, we examined CpG island DNA methylation of α5 -GABAA receptor coding gene through pyrosequencing. Nerve injury increased methylation in male, but not female rats. Pharmacological inhibition of DNA methyltransferases increased α5 -GABAA receptor and enabled L-655,708 antinociceptive effect in male rats. These results suggest that α5 -GABAA receptor is a suitable target to treat chronic pain in females.
Collapse
Affiliation(s)
- Úrzula Franco-Enzástiga
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Guadalupe García
- Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Janet Murbartián
- Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | | | - Ana B Salinas-Abarca
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Beatriz Sánchez-Hernández
- Departamento de Genética, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana Tavares-Ferreira
- School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Luis A Herrera
- Cancer Biomedical Research Unit, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Paulino Barragán-Iglesias
- School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA.,Department of Physiology and Pharmacology, Center for Basic Sciences, Autonomous University of Aguascalientes, Aguascalientes, Mexico
| | - Rodolfo Delgado-Lezama
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav, Zacatenco, Mexico City, Mexico
| | - Theodore J Price
- School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| |
Collapse
|
13
|
Kaur J, Daoud A, Eblen ST. Targeting Chromatin Remodeling for Cancer Therapy. Curr Mol Pharmacol 2020; 12:215-229. [PMID: 30767757 PMCID: PMC6875867 DOI: 10.2174/1874467212666190215112915] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/25/2019] [Accepted: 01/31/2019] [Indexed: 12/31/2022]
Abstract
Background: Epigenetic alterations comprise key regulatory events that dynamically alter gene expression and their deregulation is commonly linked to the pathogenesis of various diseases, including cancer. Unlike DNA mutations, epigenetic alterations involve modifications to proteins and nucleic acids that regulate chromatin structure without affecting the underlying DNA sequence, altering the accessibility of the transcriptional machinery to the DNA, thus modulating gene expression. In cancer cells, this often involves the silencing of tumor suppressor genes or the increased expression of genes involved in oncogenesis. Advances in laboratory medicine have made it possible to map critical epigenetic events, including histone modifications and DNA methylation, on a genome-wide scale. Like the identification of genetic mutations, mapping of changes to the epigenetic landscape has increased our understanding of cancer progression. However, in contrast to irreversible genetic mutations, epigenetic modifications are flexible and dynamic, thereby making them promising therapeutic targets. Ongoing studies are evaluating the use of epigenetic drugs in chemotherapy sensitization and immune system modulation. With the preclinical success of drugs that modify epigenetics, along with the FDA approval of epigenetic drugs including the DNA methyltransferase 1 (DNMT1) inhibitor 5-azacitidine and the histone deacetylase (HDAC) inhibitor vorinostat, there has been a rise in the number of drugs that target epigenetic modulators over recent years. Conclusion: We provide an overview of epigenetic modulations, particularly those involved in cancer, and discuss the recent advances in drug development that target these chromatin-modifying events, primarily focusing on novel strategies to regulate the epigenome.
Collapse
Affiliation(s)
- Jasmine Kaur
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Abdelkader Daoud
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Scott T Eblen
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, United States
| |
Collapse
|
14
|
Correia C, Romieu P, Olmstead MC, Befort K. Can cocaine-induced neuroinflammation explain maladaptive cocaine-associated memories? Neurosci Biobehav Rev 2020; 111:69-83. [PMID: 31935376 DOI: 10.1016/j.neubiorev.2020.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/20/2019] [Accepted: 01/01/2020] [Indexed: 12/19/2022]
Abstract
Persistent and intrusive memories define a number of psychiatric disorders, including posttraumatic stress disorder and substance use disorder. In the latter, memory for drug-paired cues plays a critical role in sustaining compulsive drug use as these are potent triggers of relapse. As with many drugs, cocaine-cue associated memory is strengthened across presentations as cues become reliable predictors of drug availability. Recently, the targeting of cocaine-associated memory through disruption of the reconsolidation process has emerged as a potential therapeutic strategy; reconsolidation reflects the active process by which memory is re-stabilized after retrieval. In addition, a separate line of work reveals that neuroinflammatory markers, regulated by cocaine intake, play a role in memory processes. Our review brings these two literatures together by summarizing recent findings on cocaine-associated reconsolidation and cocaine-induced neuroinflammation. We discuss the interactions between reconsolidation processes and neuroinflammation following cocaine use, concluding with a new perspective on treatment to decrease risk of relapse to cocaine use.
Collapse
Affiliation(s)
- Caroline Correia
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, UMR 7364, Faculté de Psychologie, 12 rue Goethe, F-67000, Strasbourg, France
| | - Pascal Romieu
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, UMR 7364, Faculté de Psychologie, 12 rue Goethe, F-67000, Strasbourg, France
| | - Mary C Olmstead
- Dept. Psychology, Centre for Neuroscience Studies, Queen's University, Kingston ON, K7L 3N6, Canada
| | - Katia Befort
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Centre de la Recherche Nationale Scientifique, UMR 7364, Faculté de Psychologie, 12 rue Goethe, F-67000, Strasbourg, France.
| |
Collapse
|
15
|
Liu L, Luo T, Dong H, Zhang C, Liu T, Zhang X, Hao W. Genome-Wide DNA Methylation Analysis in Male Methamphetamine Users With Different Addiction Qualities. Front Psychiatry 2020; 11:588229. [PMID: 33192735 PMCID: PMC7645035 DOI: 10.3389/fpsyt.2020.588229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/24/2020] [Indexed: 12/20/2022] Open
Abstract
This paper aimed to explore the genome-wide DNA methylation status of methamphetamine (MA) abusers with different qualities to addiction and to identify differentially methylated candidate genes. A total of 207 male MA abusers with an MA abuse frequency of ≥10 times and an MA abuse duration of ≥1 year were assigned to the high MA addiction quality group (HMAQ group; 168 subjects who met the diagnostic criteria for MA dependence according to the DSM-IV) or to the low MA addictive quality group (LMAQ group; 39 subjects who did not meet the criteria for MA dependence). In addition 105 healthy controls were recruited. Eight HMAQ subjects, eight LMAQ subjects, and eight healthy controls underwent genome-wide DNA methylation scans with an Infinium Human Methylation 450 array (Illumina). The differentially methylated region (DMR) data were entered into pathway analysis, and the differentially methylated position (DMP) data were screened for candidate genes and verified by MethyLight qPCR with all samples. Seven specific pathways with an abnormal methylation status were identified, including the circadian entrainment, cholinergic synapse, glutamatergic synapse, retrograde endocannabinoid signaling, GABAergic synapse, morphine addiction and PI3K-Akt signaling pathways. SLC1A6, BHLHB9, LYNX1, CAV2, and PCSK9 showed differences in their methylation levels in the three groups. Only the number of methylated copies of CAV2 was significantly higher in the LMAQ group than in the HMAQ group. Our findings suggest that the circadian entrainment pathway and the caveolin-2 gene may play key roles in MA addiction quality. Further studies on their functions and mechanisms will help us to better understand the pathogenesis of MA addiction and to explore new targets for drug intervention.
Collapse
Affiliation(s)
- Liang Liu
- Department of Geriatric Psychiatry, Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| | - Tao Luo
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China.,Department of Clinic Psychiatry, Jiangxi Mental Hospital, Nanchang University, Nanchang, China
| | - Huixi Dong
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| | - Chenxi Zhang
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| | - Tieqiao Liu
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| | - Xiangyang Zhang
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Wei Hao
- Hunan Key Laboratory of Psychiatry and Mental Health, Department of Psychiatry and Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Central South University, Changsha, China
| |
Collapse
|
16
|
Urb M, Niinep K, Matsalu T, Kipper K, Herodes K, Zharkovsky A, Timmusk T, Anier K, Kalda A. The role of DNA methyltransferase activity in cocaine treatment and withdrawal in the nucleus accumbens of mice. Addict Biol 2020; 25:e12720. [PMID: 30730091 DOI: 10.1111/adb.12720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/07/2018] [Accepted: 01/08/2019] [Indexed: 01/02/2023]
Abstract
An increasing number of reports have provided crucial evidence that epigenetic modifications, such as DNA methylation, may be involved in initiating and establishing psychostimulant-induced stable changes at the cellular level by coordinating the expression of gene networks, which then manifests as long-term behavioral changes. In this study, we evaluated the enzyme activity of DNA methyltransferases (DNMTs) after cocaine treatment and during withdrawal. Furthermore, we studied how genetic or pharmacological inhibition of DNMTs in mouse nucleus accumbens (NAc) affects the induction and expression of cocaine-induced behavioral sensitization. Our results showed that after silencing Dnmt3a in the NAc during the induction phase of cocaine-induced sensitization, overall DNMT activity decreases, correlating negatively with behavioral sensitization. Reduced Dnmt3a mRNA during this phase was the largest contributing factor for decreased DNMT activity. Cocaine withdrawal and a challenge dose increased DNMT activity in the NAc, which was associated with the expression of behavioral sensitization. Long-term selective Dnmt3a transcription silencing in the NAc did not alter DNMT activity or the expression of cocaine-induced behavioral sensitization. However, bilateral intra-NAc injection of a non-specific inhibitor of DNMT (RG108) during withdrawal from cocaine decreased DNMT activity in the NAc and had a small effect on the expression of cocaine-induced behavioral sensitization. Thus, cocaine treatment and withdrawal is associated with biphasic changes in DNMT activity in the NAc, and the expression of behavioral sensitization decreases with non-selective inhibition of DNMT but not with selective silencing of Dnmt3a.
Collapse
Affiliation(s)
- Mari Urb
- Department of PharmacologyInstitute of Biomedicine and Translational Medicine, University of Tartu Estonia
| | - Kerly Niinep
- Department of PharmacologyInstitute of Biomedicine and Translational Medicine, University of Tartu Estonia
| | - Terje Matsalu
- Department of PharmacologyInstitute of Biomedicine and Translational Medicine, University of Tartu Estonia
| | - Karin Kipper
- Institute of Chemistry, University of Tartu Estonia
| | - Koit Herodes
- Institute of Chemistry, University of Tartu Estonia
| | - Alexander Zharkovsky
- Department of PharmacologyInstitute of Biomedicine and Translational Medicine, University of Tartu Estonia
| | - Tõnis Timmusk
- Institute of Chemistry and Biotechnology, Tallinn University of Technology Estonia
| | - Kaili Anier
- Department of PharmacologyInstitute of Biomedicine and Translational Medicine, University of Tartu Estonia
| | - Anti Kalda
- Department of PharmacologyInstitute of Biomedicine and Translational Medicine, University of Tartu Estonia
| |
Collapse
|
17
|
Sartor GC. Epigenetic pharmacotherapy for substance use disorder. Biochem Pharmacol 2019; 168:269-274. [PMID: 31306644 PMCID: PMC6733674 DOI: 10.1016/j.bcp.2019.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/10/2019] [Indexed: 12/12/2022]
Abstract
Identifying novel therapeutics for the treatment of substance use disorder (SUD) is an area of intensive investigation. Prior strategies that have attempted to modify one or a few neurotransmitter receptors have had limited success, and currently there are no FDA-approved medications for the treatment of cocaine, methamphetamine, and marijuana use disorders. Because drugs of abuse are known to alter the expression of numerous genes in reward-related brain regions, epigenetic-based therapies have emerged as intriguing targets for therapeutic innovation. Here, I evaluate potential therapeutic approaches and challenges in targeting epigenetic factors for the treatment of SUD and highlight examples of promising strategies and future directions.
Collapse
Affiliation(s)
- Gregory C Sartor
- University of Connecticut, Department of Pharmaceutical Sciences, 69 N. Eagleville Road, Storrs, CT 06269, United States.
| |
Collapse
|
18
|
Saad L, Sartori M, Pol Bodetto S, Romieu P, Kalsbeek A, Zwiller J, Anglard P. Regulation of Brain DNA Methylation Factors and of the Orexinergic System by Cocaine and Food Self-Administration. Mol Neurobiol 2019; 56:5315-5331. [PMID: 30603957 DOI: 10.1007/s12035-018-1453-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/07/2018] [Indexed: 12/22/2022]
Abstract
Inhibitors of DNA methylation and orexin type-1 receptor antagonists modulate the neurobiological effects driving drugs of abuse and natural reinforcers by activating common brain structures of the mesolimbic reward system. In this study, we applied a self-administration paradigm to assess the involvement of factors regulating DNA methylation processes and satiety or appetite signals. These factors include Dnmts and Tets, miR-212/132, orexins, and orx-R1 genes. The study focused on dopamine projection areas such as the prefrontal cortex (PFCx) and caudate putamen (CPu) and in the hypothalamus (HP) that is interconnected with the reward system. Striking changes were observed in response to both reinforcers, but differed depending on contingent and non-contingent delivery. Expression also differed in the PFCx and the CPu. Cocaine and food induced opposite effects on Dnmt3a expression in both brain structures, whereas they repressed both miRs to a different extent, without affecting their primary transcript in the CPu. Unexpectedly, orexin mRNAs were found in the CPu, suggesting a transport from their transcription site in the HP. The orexin receptor1 gene was found to be induced by cocaine in the PFCx, consistent with a regulation by DNA methylation. Global levels of 5-methylcytosines in the PFCx were not significantly altered by cocaine, suggesting that it is rather their distribution that contributes to long-lasting behaviors. Together, our data demonstrate that DNA methylation regulating factors are differentially altered by cocaine and food. At the molecular level, they support the idea that neural circuits activated by both reinforcers do not completely overlap.
Collapse
Affiliation(s)
- Lamis Saad
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Neuropôle de Strasbourg, Université de Strasbourg, Strasbourg, France
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
| | - Maxime Sartori
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Neuropôle de Strasbourg, Université de Strasbourg, Strasbourg, France
- IGBMC, Inserm U 964, CNRS UMR 7104, University of Strasbourg, Illkirch, France
| | - Sarah Pol Bodetto
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Neuropôle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Pascal Romieu
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Neuropôle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Andries Kalsbeek
- The Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jean Zwiller
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Neuropôle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Patrick Anglard
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364 CNRS, Neuropôle de Strasbourg, Université de Strasbourg, Strasbourg, France.
- INSERM, Institut National de la Santé et de la Recherche Médicale, Paris, France.
| |
Collapse
|
19
|
Caputi FF, Romualdi P, Candeletti S. Regulation of the Genes Encoding the ppN/OFQ and NOP Receptor. Handb Exp Pharmacol 2019; 254:141-162. [PMID: 30689088 DOI: 10.1007/164_2018_196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Over the years, the ability of N/OFQ-NOP receptor system in modulating several physiological functions, including the release of neurotransmitters, anxiety-like behavior responses, modulation of the reward circuitry, inflammatory signaling, nociception, and motor function, has been examined in several brain regions and at spinal level. This chapter collects information related to the genes encoding the ppN/OFQ and NOP receptor, their regulation, and relative transcriptional control mechanisms. Furthermore, genetic manipulations, polymorphisms, and epigenetic alterations associated with different pathological conditions are discussed. The evidence here collected indicates that the study of ppN/OFQ and NOP receptor gene expression may offer novel opportunities in the field of personalized therapies and highlights this system as a good "druggable target" for different pathological conditions.
Collapse
Affiliation(s)
- Francesca Felicia Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
20
|
Neuroendocrine aging precedes perimenopause and is regulated by DNA methylation. Neurobiol Aging 2018; 74:213-224. [PMID: 30497015 DOI: 10.1016/j.neurobiolaging.2018.09.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 09/21/2018] [Accepted: 09/21/2018] [Indexed: 11/22/2022]
Abstract
Perimenopause marks initiation of female reproductive senescence. Age of onset is only 47% heritable suggesting that additional factors other than inheritance regulate this endocrine aging transition. To elucidate these factors, we characterized transcriptional and epigenomic changes across endocrine aging using a rat model that recapitulates characteristics of the human perimenopause. RNA-seq analysis revealed that hypothalamic aging precedes onset of perimenopause. In the hypothalamus, global DNA methylation declined with both age and reproductive senescence. Genome-wide epigentic analysis revealed changes in DNA methylation in genes required for hormone signaling, glutamate signaling, and melatonin and circadian pathways. Specific epignetic changes in these signaling pathways provide insight into the origin of perimenopause-associated neurological symptoms such as insomnia. Treatment with 5-aza-2'-deoxycytidine, a DNA-methyltransferase-1 inhibitor, accelerated transition to reproductive senescence/ whereas supplementation with methionine, a S-adenosylmethionine precursor, delayed onset of perimenopause and endocrine aging. Collectively, these data provide evidence for a critical period of female neuroendocrine aging in brain that precedes ovarian failure and that DNA methylation regulates the transition duration of perimenopause to menopause.
Collapse
|
21
|
De Sa Nogueira D, Merienne K, Befort K. Neuroepigenetics and addictive behaviors: Where do we stand? Neurosci Biobehav Rev 2018; 106:58-72. [PMID: 30205119 DOI: 10.1016/j.neubiorev.2018.08.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/28/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022]
Abstract
Substance use disorders involve long-term changes in the brain that lead to compulsive drug seeking, craving, and a high probability of relapse. Recent findings have highlighted the role of epigenetic regulations in controlling chromatin access and regulation of gene expression following exposure to drugs of abuse. In the present review, we focus on data investigating genome-wide epigenetic modifications in the brain of addicted patients or in rodent models exposed to drugs of abuse, with a particular focus on DNA methylation and histone modifications associated with transcriptional studies. We highlight critical factors for epigenomic studies in addiction. We discuss new findings related to psychostimulants, alcohol, opiate, nicotine and cannabinoids. We examine the possible transmission of these changes across generations. We highlight developing tools, specifically those that allow investigation of structural reorganization of the chromatin. These have the potential to increase our understanding of alteration of chromatin architecture at gene regulatory regions. Neuroepigenetic mechanisms involved in addictive behaviors could explain persistent phenotypic effects of drugs and, in particular, vulnerability to relapse.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, CNRS, Université de Strasbourg, Team 3 « Abuse of Drugs and Neuroadaptations », Faculté de Psychologie, 12 rue Goethe, F-67000, France
| | - Karine Merienne
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, CNRS, Université de Strasbourg, Team 1 « Dynamics of Memory and Epigenetics », Faculté de Psychologie, 12 rue Goethe, F-67000, France
| | - Katia Befort
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, CNRS, Université de Strasbourg, Team 3 « Abuse of Drugs and Neuroadaptations », Faculté de Psychologie, 12 rue Goethe, F-67000, France.
| |
Collapse
|
22
|
Li X, Carreria MB, Witonsky KR, Zeric T, Lofaro OM, Bossert JM, Zhang J, Surjono F, Richie CT, Harvey BK, Son H, Cowan CW, Nestler EJ, Shaham Y. Role of Dorsal Striatum Histone Deacetylase 5 in Incubation of Methamphetamine Craving. Biol Psychiatry 2018; 84:213-222. [PMID: 29397902 PMCID: PMC6026084 DOI: 10.1016/j.biopsych.2017.12.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/28/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Methamphetamine (meth) seeking progressively increases after withdrawal (incubation of meth craving). We previously demonstrated an association between histone deacetylase 5 (HDAC5) gene expression in the rat dorsal striatum and incubation of meth craving. Here we used viral constructs to study the causal role of dorsal striatum HDAC5 in this incubation. METHODS In experiment 1 (overexpression), we injected an adeno-associated virus bilaterally into dorsal striatum to express either green fluorescent protein (control) or a mutant form of HDAC5, which strongly localized to the nucleus. After training rats to self-administer meth (10 days, 9 hours/day), we tested the rats for relapse to meth seeking on withdrawal days 2 and 30. In experiment 2 (knockdown), we injected an adeno-associated virus bilaterally into the dorsal striatum to express a short hairpin RNA either against luciferase (control) or against HDAC5. After training rats to self-administer meth, we tested the rats for relapse on withdrawal days 2 and 30. We also measured gene expression of other HDACs and potential HDAC5 downstream targets. RESULTS We found that HDAC5 overexpression in dorsal striatum increased meth seeking on withdrawal day 30 but not day 2. In contrast, HDAC5 knockdown in the dorsal striatum decreased meth seeking on withdrawal day 30 but not on day 2; this manipulation also altered other HDACs (Hdac1 and Hdac4) and potential HDAC5 targets (Gnb4 and Suv39h1). CONCLUSIONS Results demonstrate a novel role of dorsal striatum HDAC5 in incubation of meth craving. These findings also set up future work to identify HDAC5 targets that mediate this incubation.
Collapse
Affiliation(s)
- Xuan Li
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland.
| | - Maria B Carreria
- Neuroscience Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kailyn R Witonsky
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland
| | - Tamara Zeric
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland
| | - Olivia M Lofaro
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland
| | - Jennifer M Bossert
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland
| | - Jianjun Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Felicia Surjono
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland
| | - Christopher T Richie
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland
| | - Hyeon Son
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, South Korea
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Eric J Nestler
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yavin Shaham
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland
| |
Collapse
|
23
|
Epigenetic mechanisms associated with addiction-related behavioural effects of nicotine and/or cocaine: implication of the endocannabinoid system. Behav Pharmacol 2018; 28:493-511. [PMID: 28704272 DOI: 10.1097/fbp.0000000000000326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The addictive use of nicotine (NC) and cocaine (COC) continues to be a major public health problem, and their combined use has been reported, particularly during adolescence. In neural plasticity, commonly induced by NC and COC, as well as behavioural plasticity related to the use of these two drugs, the involvement of epigenetic mechanisms, in which the reversible regulation of gene expression occurs independently of the DNA sequence, has recently been reported. Furthermore, on the basis of intense interactions with the target neurotransmitter systems, the endocannabinoid (ECB) system has been considered pivotal for eliciting the effects of NC or COC. The combined use of marijuana with NC and/or COC has also been reported. This article presents the addiction-related behavioural effects of NC and/or COC, based on the common behavioural/neural plasticity and combined use of NC/COC, and reviews the interacting role of the ECB system. The epigenetic processes inseparable from the effects of NC and/or COC (i.e. DNA methylation, histone modifications and alterations in microRNAs) and the putative therapeutic involvement of the ECB system at the epigenetic level are also discussed.
Collapse
|
24
|
Qiao X, Yin F, Ji Y, Li Y, Yan P, Lai J. 5-Aza-2'-deoxycytidine in the medial prefrontal cortex regulates alcohol-related behavior and Ntf3-TrkC expression in rats. PLoS One 2017; 12:e0179469. [PMID: 28614398 PMCID: PMC5470731 DOI: 10.1371/journal.pone.0179469] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/31/2017] [Indexed: 12/16/2022] Open
Abstract
Recent studies have indicated that DNA methylation plays an important role in the development of alcohol abuse. 5-Aza-2'-deoxycytidine (5-Aza-dc), an inhibitor of DNA methyltransferases, was FDA approved for myelodysplastic syndrome treatment. However, it is unclear whether 5-Aza-dc is involved in alcohol abuse. In this study, using a chronic alcohol exposure model in rats, 5-Aza-dc was injected into the medial prefrontal cortex (mPFC). Alcohol-drinking behavior and the anxiety related behavior were evaluated by two-bottle choice and open field test. We found that 5-Aza-dc injection into the mPFC significantly decreased alcohol consumption and alcohol preference in alcohol-exposure rats, corresponding to the reduced blood alcohol levels. Although 5-Aza-dc potentiated the anxiety-like behavior of alcohol-exposure rats, it had no effect on the locomotor activity. Moreover, both of the mRNA and protein levels of DNA Methyltransferase 3A (DNMT3A) and DNMT3B in the mPFC were upregulated after 35 days of alcohol exposure and this upregulation could be reversed by 5-Aza-dc treatment. Additionally, 5-Aza-dc reversed the alcohol-induced downregulation of neurotrophin-3 (Ntf3), correspondingly the expression of its receptor-TrkC was reduced. These findings identified a functional role of 5-Aza-dc in alcohol-related behavioral phenotypes and one of the potential target genes, Ntf3. We also provide novel evidence for DNA methyltransferases as potential therapeutic targets in alcohol abuse.
Collapse
Affiliation(s)
- Xiaomeng Qiao
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Fangyuan Yin
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yuanyuan Ji
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yunxiao Li
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Peng Yan
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jianghua Lai
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, Shaanxi, China
| |
Collapse
|
25
|
Vaillancourt K, Ernst C, Mash D, Turecki G. DNA Methylation Dynamics and Cocaine in the Brain: Progress and Prospects. Genes (Basel) 2017; 8:genes8050138. [PMID: 28498318 PMCID: PMC5448012 DOI: 10.3390/genes8050138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/04/2017] [Indexed: 12/27/2022] Open
Abstract
Cytosine modifications, including DNA methylation, are stable epigenetic marks that may translate environmental change into transcriptional regulation. Research has begun to investigate DNA methylation dynamics in relation to cocaine use disorders. Specifically, DNA methylation machinery, including methyltransferases and binding proteins, are dysregulated in brain reward pathways after chronic cocaine exposure. In addition, numerous methylome-wide and candidate promoter studies have identified differential methylation, at the nucleotide level, in rodent models of cocaine abuse and drug seeking behavior. This review highlights the current progress in the field of cocaine-related methylation, and offers considerations for future research.
Collapse
Affiliation(s)
- Kathryn Vaillancourt
- Department of Psychiatry, McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Verdun, QC H4H 1R3, Canada.
| | - Carl Ernst
- Department of Psychiatry, McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Verdun, QC H4H 1R3, Canada.
| | - Deborah Mash
- Department of Neurology, University of Miami Miller School of Medicine, University of Miami, Coral Gables, FL 33146, USA.
| | - Gustavo Turecki
- Department of Psychiatry, McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Verdun, QC H4H 1R3, Canada.
| |
Collapse
|