1
|
Hrabak M, Ahmed R, Soriano MG, Powell A, Thanos PK, Steiner H. Vilazodone, a Novel SSRI Antidepressant with 5-HT1A Partial Agonist Properties: Diminished Potentiation of Chronic Oral Methylphenidate-Induced Dynorphin Expression in the Striatum in Adolescent Male Rats. Mol Neurobiol 2025; 62:4520-4532. [PMID: 39466575 DOI: 10.1007/s12035-024-04569-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Selective serotonin reuptake inhibitor (SSRI) antidepressants such as fluoxetine are used in combination with the medical psychostimulant methylphenidate (Ritalin) in a variety of treatments in children and adults. Unintended co-exposure to these psychotropic medications also occurs in patients on SSRIs who abuse methylphenidate as a "cognitive enhancer" or recreational drug. Preclinical research shows that SSRIs such as fluoxetine when given in conjunction with methylphenidate potentiate addiction-related gene regulation by methylphenidate in the striatum, consistent with the known facilitatory role for serotonin in psychostimulant-induced neuronal and behavioral changes. Moreover, fluoxetine combined with methylphenidate also facilitated subsequent acquisition of cocaine self-administration in adolescent rats, suggesting an increased addiction liability for methylphenidate. In the present study, we investigated the impact of a novel SSRI, vilazodone, on methylphenidate-induced gene regulation in adolescent male rats. In contrast to prototypical SSRIs such as fluoxetine, vilazodone also acts as a partial 5-HT1A serotonin receptor agonist and is thus proposed to temper serotonin input to the striatum. We compared the effects of chronic treatment (4 weeks) with vilazodone (10 mg/kg, twice daily) with those of fluoxetine (5 mg/kg, twice daily) on striatal dynorphin expression induced by oral methylphenidate treatment (30/60 mg/kg/day in drinking water, 8 h access daily). Our results demonstrate that, in contrast to fluoxetine, vilazodone had minimal or no potentiating effects on methylphenidate-induced dynorphin expression. This diminished impact on gene regulation was seen throughout the striatum, including the nucleus accumbens, where increased dynorphin expression has previously been associated with various aspects of addiction. Our findings suggest that vilazodone may serve as a better adjunct SSRI with reduced addiction-facilitating properties.
Collapse
Affiliation(s)
- Michael Hrabak
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Rania Ahmed
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Michelle G Soriano
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Aidan Powell
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
2
|
Sequeira-Cordero A, Brenes JC. Time-dependent changes in striatal monoamine levels and gene expression following single and repeated amphetamine administration in rats. Eur J Pharmacol 2021; 904:174148. [PMID: 33961872 DOI: 10.1016/j.ejphar.2021.174148] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/23/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
As drug addiction may result from pathological usurpations of learning and memory's neural mechanisms, we focused on the amphetamine-induced time-dependent neurochemical changes associated with neural plasticity. We used juvenile rats as the risk for drug abuse is higher during adolescence. Experiment 1 served to define the appropriate amphetamine dose and the neurochemical effects of a single administration. In experiment 2, rats received seven amphetamine or saline injections in the open-field test throughout a twelve-day period. We measured the mRNA levels of the brain-derived neurotrophic factor (BDNF), its tropomyosin receptor kinase B (TrkB), the cAMP response element-binding protein (CREB), the microRNA-132, the Rho GTPase-activating protein 32 (p250GAP), the corticotropin-releasing factor (CRF), and monoamines and amino-acids contents in the nucleus accumbens and the dorsal striatum 45, 90, and 180 min after the last injection. We found that amphetamine changed gene expression only at certain time points and in a dose and region-dependent manner. Repeated but not single administrations upregulated accumbal and striatal BDNF (180 min) and striatal pri-miR-132 (90 min) expression, while downregulated accumbal CREB levels (90 min). As only some drug users develop addiction, we compared brain parameters between low and high amphetamine responders. Prone subjects characterized by having reduced striatal 5-HT metabolism, higher accumbal BDNF and TrkB expression, and lower levels of CREB in the dorsal striatum and p250GAP in both regions. Thus, individual differences in drug-induced changes in neurotransmission and gene expression in nigrostriatal and mesolimbic dopaminergic pathways may underlie the plasticity adaptations associated with behavioral sensitization to amphetamine.
Collapse
Affiliation(s)
- Andrey Sequeira-Cordero
- Instituto de Investigaciones en Salud, Universidad de Costa Rica, Costa Rica; Centro de Investigación en Neurociencias, Universidad de Costa Rica, Costa Rica.
| | - Juan C Brenes
- Instituto de Investigaciones Psicológicas, Universidad de Costa Rica, Costa Rica; Centro de Investigación en Neurociencias, Universidad de Costa Rica, Costa Rica.
| |
Collapse
|
3
|
Moon C, Marion M, Thanos PK, Steiner H. Fluoxetine Potentiates Oral Methylphenidate-Induced Gene Regulation in the Rat Striatum. Mol Neurobiol 2021; 58:4856-4870. [PMID: 34213723 DOI: 10.1007/s12035-021-02466-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/21/2021] [Indexed: 11/24/2022]
Abstract
Methylphenidate (MP) is combined with selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine (FLX) to treat various disorders. MP, a dopamine reuptake inhibitor, helps manage attention-deficit hyperactivity disorder (ADHD) and is abused as a cognitive enhancer; it has a reduced addiction liability. We showed that combining FLX (serotonin) with MP potentiates MP-induced gene regulation in the striatum. These studies used intraperitoneal drug administration, which is relevant for MP abuse. Clinically, MP and FLX are taken orally (slower bioavailability). Here, we investigated whether chronic oral administration of MP and FLX also altered striatal gene regulation. MP (30/60 mg/kg/day), FLX (20 mg/kg/day), and MP + FLX were administered in rats' drinking water for 8 h/day over 4 weeks. We assessed the expression of dynorphin and substance P (both markers for striatal direct pathway neurons) and enkephalin (indirect pathway) by in situ hybridization histochemistry. Chronic oral MP alone produced a tendency for increased dynorphin and substance P expression and no changes in enkephalin expression. Oral FLX alone did not increase gene expression. In contrast, when given together, FLX greatly enhanced MP-induced expression of dynorphin and substance P and to a lesser degree enkephalin. Thus, FLX potentiated oral MP-induced gene regulation predominantly in direct pathway neurons, mimicking cocaine effects. The three functional domains of the striatum were differentially affected. MP + SSRI concomitant therapies are indicated in ADHD/depression comorbidity and co-exposure occurs with MP misuse as a cognitive enhancer by patients on SSRIs. Our findings indicate that MP + SSRI combinations, even given orally, may enhance addiction-related gene regulation.
Collapse
Affiliation(s)
- Connor Moon
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Matt Marion
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute On Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University At Buffalo, Buffalo, NY, 14203, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute On Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University At Buffalo, Buffalo, NY, 14203, USA
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA. .,Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
4
|
Crittenden JR, Gipson TA, Smith AC, Bowden HA, Yildirim F, Fischer KB, Yim M, Housman DE, Graybiel AM. Striatal transcriptome changes linked to drug-induced repetitive behaviors. Eur J Neurosci 2021; 53:2450-2468. [PMID: 33759265 DOI: 10.1111/ejn.15116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/23/2020] [Accepted: 01/09/2021] [Indexed: 11/30/2022]
Abstract
Disruptive or excessive repetitive motor patterns (stereotypies) are cardinal symptoms in numerous neuropsychiatric disorders. Stereotypies are also evoked by psychomotor stimulants such as amphetamine. The acquisition of motor sequences is paralleled by changes in activity patterns in the striatum, and stereotypies have been linked to abnormal plasticity in these reinforcement-related circuits. Here, we designed experiments in mice to identify transcriptomic changes that underlie striatal plasticity occurring alongside the development of drug-induced stereotypic behavior. We identified three schedules of amphetamine treatment inducing different degrees of stereotypy and used bulk RNAseq to compare striatal gene expression changes among groups of mice treated with the different drug-dose schedules and vehicle-treated, cage-mate controls. Mice were identified as naïve, sensitized, or tolerant to drug-induced stereotypy. All drug-treated groups exhibited expression changes in genes that encode members of the extracellular signal-regulated kinase (ERK) cascades known to regulate psychomotor stimulant responses. In the sensitized group with the most prolonged stereotypy, we found dysregulation of 20 genes that were not changed in other groups. Gene set enrichment analysis indicated highly significant overlap with genes regulated by neuregulin 1 (Nrg1). Nrg1 is known to be a schizophrenia and autism susceptibility gene that encodes a ligand for Erb-B receptors, which are involved in neuronal migration, myelination, and cell survival, including that of dopamine-containing neurons. Stimulant abuse is a risk factor for schizophrenia onset, and these two disorders share behavioral stereotypy phenotypes. Our results raise the possibility that drug-induced sensitization of the Nrg1 signaling pathway might underlie these links.
Collapse
Affiliation(s)
- Jill R Crittenden
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Theresa A Gipson
- Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anne C Smith
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Hilary A Bowden
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ferah Yildirim
- Department of Neuropsychiatry, Department of Psychiatry and Psychotherapy, and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kyle B Fischer
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael Yim
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David E Housman
- Institute for Integrative Cancer Research, The Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ann M Graybiel
- McGovern Institute for Brain Research, The Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, The Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
5
|
Sadakierska-Chudy A, Frankowska M, Jastrzębska J, Wydra K, Miszkiel J, Sanak M, Filip M. Cocaine Administration and Its Withdrawal Enhance the Expression of Genes Encoding Histone-Modifying Enzymes and Histone Acetylation in the Rat Prefrontal Cortex. Neurotox Res 2017; 32:141-150. [PMID: 28393332 PMCID: PMC5487868 DOI: 10.1007/s12640-017-9728-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 12/14/2022]
Abstract
Chronic exposure to cocaine, craving, and relapse are attributed to long-lasting changes in gene expression arising through epigenetic and transcriptional mechanisms. Although several brain regions are involved in these processes, the prefrontal cortex seems to play a crucial role not only in motivation and decision-making but also in extinction and seeking behavior. In this study, we applied cocaine self-administration and extinction training procedures in rats with a yoked triad to determine differentially expressed genes in prefrontal cortex. Microarray analysis showed significant upregulation of several genes encoding histone modification enzymes during early extinction training. Subsequent real-time PCR testing of these genes following cocaine self-administration or early (third day) and late (tenth day) extinction revealed elevated levels of their transcripts. Interestingly, we found the enrichment of Brd1 messenger RNA in rats self-administering cocaine that lasted until extinction training during cocaine withdrawal with concomitant increased acetylation of H3K9 and H4K8. However, despite elevated levels of methyl- and demethyltransferase-encoded transcripts, no changes in global di- and tri-methylation of histone H3 at lysine 4, 9, 27, and 79 were observed. Surprisingly, at the end of extinction training (10 days of cocaine withdrawal), most of the analyzed genes in the rats actively and passively administering cocaine returned to the control level. Together, the alterations identified in the rat prefrontal cortex may suggest enhanced chromatin remodeling and transcriptional activity induced by early cocaine abstinence; however, to know whether they are beneficial or not for the extinction of drug-seeking behavior, further in vivo evaluation is required.
Collapse
Affiliation(s)
- Anna Sadakierska-Chudy
- Institute of Pharmacology Polish Academy of Sciences, Department of Pharmacology, Laboratory of Drug Addiction Pharmacology, Polish Academy of Sciences, ul. Smetna 12, 31-343, Krakow, Poland.
| | - Małgorzata Frankowska
- Institute of Pharmacology Polish Academy of Sciences, Department of Pharmacology, Laboratory of Drug Addiction Pharmacology, Polish Academy of Sciences, ul. Smetna 12, 31-343, Krakow, Poland
| | - Joanna Jastrzębska
- Institute of Pharmacology Polish Academy of Sciences, Department of Pharmacology, Laboratory of Drug Addiction Pharmacology, Polish Academy of Sciences, ul. Smetna 12, 31-343, Krakow, Poland
| | - Karolina Wydra
- Institute of Pharmacology Polish Academy of Sciences, Department of Pharmacology, Laboratory of Drug Addiction Pharmacology, Polish Academy of Sciences, ul. Smetna 12, 31-343, Krakow, Poland
| | - Joanna Miszkiel
- Institute of Pharmacology Polish Academy of Sciences, Department of Pharmacology, Laboratory of Drug Addiction Pharmacology, Polish Academy of Sciences, ul. Smetna 12, 31-343, Krakow, Poland
| | - Marek Sanak
- Laboratory of Molecular Biology and Clinical Genetics, Medical College, Jagiellonian University, ul. Skawinska 8, 31-066, Krakow, Poland
| | - Małgorzata Filip
- Institute of Pharmacology Polish Academy of Sciences, Department of Pharmacology, Laboratory of Drug Addiction Pharmacology, Polish Academy of Sciences, ul. Smetna 12, 31-343, Krakow, Poland
| |
Collapse
|
6
|
Fernàndez-Castillo N, Cabana-Domínguez J, Soriano J, Sànchez-Mora C, Roncero C, Grau-López L, Ros-Cucurull E, Daigre C, van Donkelaar MMJ, Franke B, Casas M, Ribasés M, Cormand B. Transcriptomic and genetic studies identify NFAT5 as a candidate gene for cocaine dependence. Transl Psychiatry 2015; 5:e667. [PMID: 26506053 PMCID: PMC4930134 DOI: 10.1038/tp.2015.158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 08/19/2015] [Indexed: 11/29/2022] Open
Abstract
Cocaine reward and reinforcing effects are mediated mainly by dopaminergic neurotransmission. In this study, we aimed at evaluating gene expression changes induced by acute cocaine exposure on SH-SY5Y-differentiated cells, which have been widely used as a dopaminergic neuronal model. Expression changes and a concomitant increase in neuronal activity were observed after a 5 μM cocaine exposure, whereas no changes in gene expression or in neuronal activity took place at 1 μM cocaine. Changes in gene expression were identified in a total of 756 genes, mainly related to regulation of transcription and gene expression, cell cycle, adhesion and cell projection, as well as mitogen-activeated protein kinase (MAPK), CREB, neurotrophin and neuregulin signaling pathways. Some genes displaying altered expression were subsequently targeted with predicted functional single-nucleotide polymorphisms (SNPs) in a case-control association study in a sample of 806 cocaine-dependent patients and 817 controls. This study highlighted associations between cocaine dependence and five SNPs predicted to alter microRNA binding at the 3'-untranslated region of the NFAT5 gene. The association of SNP rs1437134 with cocaine dependence survived the Bonferroni correction for multiple testing. A functional effect was confirmed for this variant by a luciferase reporter assay, with lower expression observed for the rs1437134G allele, which was more pronounced in the presence of hsa-miR-509. However, brain volumes in regions of relevance to addiction, as assessed with magnetic resonance imaging, did not correlate with NFAT5 variation. These results suggest that the NFAT5 gene, which is upregulated a few hours after cocaine exposure, may be involved in the genetic predisposition to cocaine dependence.
Collapse
Affiliation(s)
- N Fernàndez-Castillo
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - J Cabana-Domínguez
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - J Soriano
- Departament d'Estructura i Constituents de la Matèria, Universitat de Barcelona, Barcelona, Spain
| | - C Sànchez-Mora
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Psychiatric Genetics Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Spain
| | - C Roncero
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Spain
- Addiction and Dual Diagnosis Unit, Psychiatric Service, Hospital Universitari Vall d'Hebron, Agència de Salut Pública, Barcelona, Spain
- Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - L Grau-López
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Spain
- Addiction and Dual Diagnosis Unit, Psychiatric Service, Hospital Universitari Vall d'Hebron, Agència de Salut Pública, Barcelona, Spain
- Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - E Ros-Cucurull
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Addiction and Dual Diagnosis Unit, Psychiatric Service, Hospital Universitari Vall d'Hebron, Agència de Salut Pública, Barcelona, Spain
- Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - C Daigre
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Spain
- Addiction and Dual Diagnosis Unit, Psychiatric Service, Hospital Universitari Vall d'Hebron, Agència de Salut Pública, Barcelona, Spain
- Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M M J van Donkelaar
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Raboud University, Nijmegen, The Netherlands
| | - B Franke
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Raboud University, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Casas
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Spain
- Addiction and Dual Diagnosis Unit, Psychiatric Service, Hospital Universitari Vall d'Hebron, Agència de Salut Pública, Barcelona, Spain
- Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M Ribasés
- Department of Psychiatry, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Psychiatric Genetics Unit, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Spain
| | - B Cormand
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
7
|
Ciudad-Roberts A, Camarasa J, Ciudad CJ, Pubill D, Escubedo E. Alcohol enhances the psychostimulant and conditioning effects of mephedrone in adolescent mice; postulation of unique roles of D3 receptors and BDNF in place preference acquisition. Br J Pharmacol 2015; 172:4970-84. [PMID: 26228024 DOI: 10.1111/bph.13266] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/15/2015] [Accepted: 07/20/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE The psychostimulant mephedrone is often consumed in combination with alcohol (EtOH). This kind of drug consumption during adolescence is a matter of concern. EXPERIMENTAL APPROACH We studied, in adolescent CD-1 mice, whether EtOH could enhance the psychostimulant (locomotor acivity) and rewarding [conditioned place preference (CPP)] effects of mephedrone. We also determined the transcriptional changes associated with a conditioning treatment with these drugs. KEY RESULTS Mephedrone (10 mg·kg(-1)) increased locomotor activity, which was further enhanced by 40% when combined with EtOH (1 g·kg(-1)). This enhancement was blocked by haloperidol. Furthermore, mephedrone (25 mg·kg(-1)) induced CPP, which increased by 70% when administered with a dose of EtOH that was not conditioning by itself (0.75 g·kg(-1)). There was enhanced expression of the D3 dopamine receptor mRNA (Drd3) and Arpc5 in all drug-treated groups. The D3 receptor antagonist SB-277011A and the BDNF receptor antagonist ANA-12 completely prevented CPP as well as the increases in Drd3 in all groups. Accordingly, increased expression of BDNF mRNA in medial prefrontal cortex was detected at 2 and 4 h after mephedrone administration. CONCLUSIONS AND IMPLICATIONS If translated to humans, the enhancement of mephedrone effects by ethanol could result in increased abuse liability. D3 receptors and BDNF play a key role in the establishment of CPP by mephedrone, although an accompanying increase in other synaptic plasticity-related genes may also be necessary.
Collapse
Affiliation(s)
- Andrés Ciudad-Roberts
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section), Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, Barcelona, Spain
| | - Jorge Camarasa
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section), Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, Barcelona, Spain
| | - Carlos J Ciudad
- Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, Barcelona, Spain
| | - David Pubill
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section), Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, Barcelona, Spain
| | - Elena Escubedo
- Department of Pharmacology and Therapeutic Chemistry (Pharmacology Section), Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
8
|
Ozburn AR, Janowsky AJ, Crabbe JC. Commonalities and Distinctions Among Mechanisms of Addiction to Alcohol and Other Drugs. Alcohol Clin Exp Res 2015; 39:1863-77. [PMID: 26431116 PMCID: PMC4594192 DOI: 10.1111/acer.12810] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/10/2015] [Indexed: 01/25/2023]
Abstract
BACKGROUND Alcohol abuse is comorbid with abuse of many other drugs, some with similar pharmacology and others quite different. This leads to the hypothesis of an underlying, unitary dysfunctional neurobiological basis for substance abuse risk and consequences. METHODS In this review, we discuss commonalities and distinctions of addiction to alcohol and other drugs. We focus on recent advances in preclinical studies using rodent models of drug self-administration. RESULTS While there are specific behavioral and molecular manifestations common to alcohol, psychostimulant, opioid, and nicotine dependence, attempts to propose a unifying theory of the addictions inevitably face details where distinctions are found among classes of drugs. CONCLUSIONS For alcohol, versus other drugs of abuse, we discuss and compare advances in: (i) neurocircuitry important for the different stages of drug dependence; (ii) transcriptomics and genetical genomics; and (iii) enduring effects, noting in particular the contributions of behavioral genetics and animal models.
Collapse
Affiliation(s)
- Angela R. Ozburn
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA
- Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Aaron J. Janowsky
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA
- Department of Psychiatry, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - John C. Crabbe
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA
- Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
9
|
Kummari E, Guo-Ross SX, Eells JB. Laser capture microdissection--a demonstration of the isolation of individual dopamine neurons and the entire ventral tegmental area. J Vis Exp 2015:e52336. [PMID: 25742438 PMCID: PMC4354571 DOI: 10.3791/52336] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Laser capture microdissection (LCM) is used to isolate a concentrated population of individual cells or precise anatomical regions of tissue from tissue sections on a microscope slide. When combined with immunohistochemistry, LCM can be used to isolate individual cells types based on a specific protein marker. Here, the LCM technique is described for collecting a specific population of dopamine neurons directly labeled with tyrosine hydroxylase immunohistochemistry and for isolation of the dopamine neuron containing region of the ventral tegmental area using indirect tyrosine hydroxylase immunohistochemistry on a section adjacent to those used for LCM. An infrared (IR) capture laser is used to both dissect individual neurons as well as the ventral tegmental area off glass slides and onto an LCM cap for analysis. Complete dehydration of the tissue with 100% ethanol and xylene is critical. The combination of the IR capture laser and the ultraviolet (UV) cutting laser is used to isolate individual dopamine neurons or the ventral tegmental area when using PEN membrane slides. A PEN membrane slide has significant advantages over a glass slide as it offers better consistency in capturing and collecting cells, is faster collecting large pieces of tissue, is less reliant on dehydration and results in complete removal of the tissue from the slide. Although removal of large areas of tissue from a glass slide is feasible, it is considerably more time consuming and frequently leaves some residual tissue behind. Data shown here demonstrate that RNA of sufficient quantity and quality can be obtained using these procedures for quantitative PCR measurements. Although RNA and DNA are the most commonly isolated molecules from tissue and cells collected with LCM, isolation and measurement of microRNA, protein and epigenetic changes in DNA can also benefit from the enhanced anatomical and cellular resolution obtained using LCM.
Collapse
Affiliation(s)
- Evangel Kummari
- Department of Basic Sciences, Mississippi State University College of Veterinary Medicine
| | - Shirley X Guo-Ross
- Department of Basic Sciences, Mississippi State University College of Veterinary Medicine
| | - Jeffrey B Eells
- Department of Basic Sciences, Mississippi State University College of Veterinary Medicine;
| |
Collapse
|
10
|
Herrero-Turrión MJ, Rodríguez-Martín I, López-Bellido R, Rodríguez RE. Whole-genome expression profile in zebrafish embryos after chronic exposure to morphine: identification of new genes associated with neuronal function and mu opioid receptor expression. BMC Genomics 2014; 15:874. [PMID: 25294025 PMCID: PMC4201762 DOI: 10.1186/1471-2164-15-874] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 09/24/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A great number of studies have investigated changes induced by morphine exposure in gene expression using several experimental models. In this study, we examined gene expression changes during chronic exposure to morphine during maturation and differentiation of zebrafish CNS. RESULTS Microarray analysis showed 254 genes whose expression was identified as different by at least 1.3 fold change following chronic morphine exposure as compared to controls. Of these, several novel genes (grb2, copb2, otpb, magi1b, grik-l, bnip4 and sox19b) have been detected for the first time in an experimental animal model treated with morphine. We have also identified a subset of genes (dao.1, wls, bnip4 and camk1γb) differentially expressed by chronic morphine exposure whose expression is related to mu opioid receptor gene expression. Altered expression of copb2, bnip4, sox19b, otpb, dao.1, grik-l and wls is indicative of modified neuronal development, CNS patterning processes, differentiation and dopaminergic neurotransmission, serotonergic signaling pathway, and glutamatergic neurotransmission. The deregulation of camk1γb signaling genes suggests an activation of axonogenesis and dendritogenesis. CONCLUSIONS Our study identified different functional classes of genes and individual candidates involved in the mechanisms underlying susceptibility to morphine actions related to CNS development. These results open new lines to study the treatment of pain and the molecular mechanisms involved in addiction. We also found a set of zebrafish-specific morphine-induced genes, which may be putative targets in human models for addiction and pain processes.
Collapse
Affiliation(s)
| | | | | | - Raquel E Rodríguez
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca 37007, Spain.
| |
Collapse
|
11
|
Abstract
Despite the importance of numerous psychosocial factors, at its core, drug addiction involves a biological process: the ability of repeated exposure to a drug of abuse to induce changes in a vulnerable brain that drive the compulsive seeking and taking of drugs, and loss of control over drug use, that define a state of addiction. Here, we review the types of molecular and cellular adaptations that occur in specific brain regions to mediate addiction-associated behavioral abnormalities. These include alterations in gene expression achieved in part via epigenetic mechanisms, plasticity in the neurophysiological functioning of neurons and synapses, and associated plasticity in neuronal and synaptic morphology mediated in part by altered neurotrophic factor signaling. Each of these types of drug-induced modifications can be viewed as a form of “cellular or molecular memory.” Moreover, it is striking that most addiction-related forms of plasticity are very similar to the types of plasticity that have been associated with more classic forms of “behavioral memory,” perhaps reflecting the finite repertoire of adaptive mechanisms available to neurons when faced with environmental challenges. Finally, addiction-related molecular and cellular adaptations involve most of the same brain regions that mediate more classic forms of memory, consistent with the view that abnormal memories are important drivers of addiction syndromes. The goal of these studies which aim to explicate the molecular and cellular basis of drug addiction is to eventually develop biologically based diagnostic tests, as well as more effective treatments for addiction disorders.
Collapse
Affiliation(s)
- Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
12
|
dela Peña I, de la Peña JB, Kim BN, Han DH, Noh M, Cheong JH. Gene expression profiling in the striatum of amphetamine-treated spontaneously hypertensive rats which showed amphetamine conditioned place preference and self-administration. Arch Pharm Res 2014; 38:865-75. [PMID: 25163681 DOI: 10.1007/s12272-014-0470-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/19/2014] [Indexed: 12/11/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD), the most commonly diagnosed neurobehavioral disorder of childhood, is usually treated with psychostimulants (e.g., amphetamine). Little is known about the neuronal and behavioral consequences of chronic amphetamine use or abuse in individuals with ADHD. Of all ADHD animal models, the spontaneously hypertensive rat (SHR) is the most validated and widely used. Here, we analyzed striatal transcriptomes in amphetamine-pretreated SHRs (5 mg/kg, i.p. for 7 days [twice daily]), which showed a conditioned place preference to and self-administration of amphetamine. Microarray analyses revealed increased mRNA expression of 55 genes (>1.65-fold increase), while 17 genes were downregulated (<0.6-fold) in the striatum of SHRs. The main functional categories overrepresented among the differentially expressed genes in the striatum include those involved in transcription (e.g., Cebpb, Per2), genes associated with angiogenesis (e.g., Kdr, Klf5), cell adhesion (e.g., Col11a1, Ctgf), apoptosis (e.g., Nfkbia, Perp) and neuronal development (e.g., Egr2, Nr4a3). In conclusion, we dissected the striatal transcriptional responses to the reinforcing effects of repeated amphetamine treatment in the SHR model of ADHD. Future studies should determine the influence of these altered transcripts on amphetamine reinforcement in amphetamine-treated SHRs, and the clinical relevance of the present findings with regard to amphetamine use/abuse in ADHD individuals.
Collapse
Affiliation(s)
- Ike dela Peña
- Uimyung Research Institute for Neuroscience, Sahmyook University, 26-21 Kongreung-2-dong, Hwarangro-815, Nowon-gu, Seoul, 139-742, Korea
| | | | | | | | | | | |
Collapse
|
13
|
Recent updates on drug abuse analyzed by neuroproteomics studies: Cocaine, Methamphetamine and MDMA. TRANSLATIONAL PROTEOMICS 2014. [DOI: 10.1016/j.trprot.2014.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
14
|
dela Peña I, Kim HJ, Sohn A, Kim BN, Han DH, Ryu JH, Shin CY, Noh M, Cheong JH. Prefrontal cortical and striatal transcriptional responses to the reinforcing effect of repeated methylphenidate treatment in the spontaneously hypertensive rat, animal model of attention-deficit/hyperactivity disorder (ADHD). Behav Brain Funct 2014; 10:17. [PMID: 24884696 PMCID: PMC4077266 DOI: 10.1186/1744-9081-10-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 04/25/2014] [Indexed: 01/12/2023] Open
Abstract
Background Methylphenidate is the most commonly used stimulant drug for the treatment of attention-deficit/hyperactivity disorder (ADHD). Research has found that methylphenidate is a “reinforcer” and that individuals with ADHD also abuse this medication. Nevertheless, the molecular consequences of long-term recreational methylphenidate use or abuse in individuals with ADHD are not yet fully known. Methods Spontaneously hypertensive rats (SHR), the most validated and widely used ADHD animal model, were pretreated with methylphenidate (5 mg/kg, i.p.) during their adolescence (post-natal day [PND] 42–48) and tested for subsequent methylphenidate-induced conditioned place preference (CPP) and self-administration. Thereafter, the differentially expressed genes in the prefrontal cortex (PFC) and striatum of representative methylphenidate-treated SHRs, which showed CPP to and self-administration of methylphenidate, were analyzed. Results Genome-wide transcriptome profiling analyses revealed 30 differentially expressed genes in the PFC, which include transcripts involved in apoptosis (e.g. S100a9, Angptl4, Nfkbia), transcription (Cebpb, Per3), and neuronal plasticity (Homer1, Jam2, Asap1). In contrast, 306 genes were differentially expressed in the striatum and among them, 252 were downregulated. The main functional categories overrepresented among the downregulated genes include those involved in cell adhesion (e.g. Pcdh10, Ctbbd1, Itgb6), positive regulation of apoptosis (Perp, Taf1, Api5), (Notch3, Nsbp1, Sik1), mitochondrion organization (Prps18c, Letm1, Uqcrc2), and ubiquitin-mediated proteolysis (Nedd4, Usp27x, Ube2d2). Conclusion Together, these changes indicate methylphenidate-induced neurotoxicity, altered synaptic and neuronal plasticity, energy metabolism and ubiquitin-dependent protein degradation in the brains of methylphenidate-treated SHRs, which showed methylphenidate CPP and self-administration. In addition, these findings may also reflect cognitive impairment associated with chronic methylphenidate use as demonstrated in preclinical studies. Future studies are warranted to determine the clinical significance of the present findings with regard to long-term recreational methylphenidate use or abuse in individuals with ADHD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Minsoo Noh
- Uimyung Research Institute for Neuroscience, Sahmyook University, 26-21 Kongreung-2-dong, Hwarangro- 815 Nowon-gu, Seoul 139-742, Korea.
| | | |
Collapse
|
15
|
Matsumoto I, Alexander-Kaufman K, Iwazaki T, Kashem MA, Matsuda-Matsumoto H. CNS proteomes in alcohol and drug abuse and dependence. Expert Rev Proteomics 2014; 4:539-52. [PMID: 17705711 DOI: 10.1586/14789450.4.4.539] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Drugs of abuse, including alcohol, can induce dependency formation and/or brain damage in brain regions important for cognition. 'High-throughput' approaches, such as cDNA microarray and proteomics, allow the analysis of global expression profiles of genes and proteins. These technologies have recently been applied to human brain tissue from patients with psychiatric illnesses, including substance abuse/dependence and appropriate animal models to help understand the causes and secondary effects of these complex disorders. Although these types of studies have been limited in number and by proteomics techniques that are still in their infancy, several interesting hypotheses have been proposed. Focusing on CNS proteomics, we aim to review and update current knowledge in this rapidly advancing area.
Collapse
Affiliation(s)
- Izuru Matsumoto
- University of Sydney, Discipline of Pathology, NSW, Australia.
| | | | | | | | | |
Collapse
|
16
|
Steiner H, Warren BL, Van Waes V, Bolaños-Guzmán CA. Life-long consequences of juvenile exposure to psychotropic drugs on brain and behavior. PROGRESS IN BRAIN RESEARCH 2014; 211:13-30. [PMID: 24968775 DOI: 10.1016/b978-0-444-63425-2.00002-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Psychostimulants such as methylphenidate (MPH) and antidepressants such as fluoxetine (FLX) are widely used in the treatment of various mental disorders or as cognitive enhancers. These medications are often combined, for example, to treat comorbid disorders. There is a considerable body of evidence from animal models indicating that individually these psychotropic medications can have detrimental effects on the brain and behavior, especially when given during sensitive periods of brain development. However, almost no studies investigate possible interactions between these drugs. This is surprising given that their combined neurochemical effects (enhanced dopamine and serotonin neurotransmission) mimic some effects of illicit drugs such as cocaine and amphetamine. Here, we summarize recent studies in juvenile rats on the molecular effects in the mid- and forebrain and associated behavioral changes, after such combination treatments. Our findings indicate that these combined MPH+FLX treatments can produce similar molecular changes as seen after cocaine exposure while inducing behavioral changes indicative of dysregulated mood and motivation, effects that often endure into adulthood.
Collapse
Affiliation(s)
- Heinz Steiner
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| | - Brandon L Warren
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Vincent Van Waes
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Carlos A Bolaños-Guzmán
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
17
|
Bough KJ, Amur S, Lao G, Hemby SE, Tannu NS, Kampman KM, Schmitz JM, Martinez D, Merchant KM, Green C, Sharma J, Dougherty AH, Moeller FG. Biomarkers for the development of new medications for cocaine dependence. Neuropsychopharmacology 2014; 39:202-19. [PMID: 23979119 PMCID: PMC3857653 DOI: 10.1038/npp.2013.210] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 06/19/2013] [Accepted: 07/01/2013] [Indexed: 01/20/2023]
Abstract
There has been significant progress in personalized drug development. In large part, this has taken place in the oncology field and been due to the ability of researchers/clinicians to discover and develop novel drug development tools (DDTs), such as biomarkers. In cancer treatment research, biomarkers have permitted a more accurate pathophysiological characterization of an individual patient, and have enabled practitioners to target mechanistically the right drug, to the right patient, at the right time. Similar to cancer, patients with substance use disorders (SUDs) present clinically with heterogeneous symptomatology and respond variably to therapeutic interventions. If comparable biomarkers could be identified and developed for SUDs, significant diagnostic and therapeutic advances could be made. In this review, we highlight current opportunities and difficulties pertaining to the identification and development of biomarkers for SUDs. We focus on cocaine dependence as an example. Putative diagnostic, pharmacodynamic (PD), and predictive biomarkers for cocaine dependence are discussed across a range of methodological approaches. A possible cocaine-dependent clinical outcome assessment (COA)--another type of defined DDT--is also discussed. At present, biomarkers for cocaine dependence are in their infancy. Much additional research will be needed to identify, validate, and qualify these putative tools prior to their potential use for medications development and/or application to clinical practice. However, with a large unmet medical need and an estimated market size of several hundred million dollars per year, if developed, biomarkers for cocaine dependence will hold tremendous value to both industry and public health.
Collapse
Affiliation(s)
- Kristopher J Bough
- Division of Basic Neuroscience and Behavioral Research, National Institute on Drug Abuse, Bethesda, MD, USA
| | - Shashi Amur
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Guifang Lao
- Division of Pharmacotherapies and Medical Consequences, National Institute on Drug Abuse, Bethesda, MD, USA
| | - Scott E Hemby
- Department of Physiology & Pharmacology, Wake Forest University, Winston-Salem, NC, USA
| | - Nilesh S Tannu
- Department of Psychiatry and Behavioral Sciences, University of Texas—Houston Medical School, Houston, TX, USA
| | - Kyle M Kampman
- Department of Psychiatry, University of Pennsylvania—School of Medicine, Philadelphia, PA, USA
| | - Joy M Schmitz
- Department of Psychiatry and Behavioral Sciences, University of Texas—Houston Medical School, Houston, TX, USA
| | - Diana Martinez
- Department of Psychiatry, Columbia University/New York State University, New York, NY, USA
| | | | - Charles Green
- Department of Pediatrics, University of Texas—Houston Medical School, Houston, TX, USA
| | - Jyoti Sharma
- Department of Cardiovascular Medicine, University of Texas—Houston Medical School, Houston, TX, USA
| | - Anne H Dougherty
- Department of Cardiovascular Medicine, University of Texas—Houston Medical School, Houston, TX, USA
| | - F Gerard Moeller
- Department of Psychiatry and Pharmacology and Toxicology, Virginia Commonwealth University Medical School, Richmond, VA, USA
| |
Collapse
|
18
|
Dela Peña I, Jeon SJ, Lee E, Ryu JH, Shin CY, Noh M, Cheong JH. Neuronal development genes are key elements mediating the reinforcing effects of methamphetamine, amphetamine, and methylphenidate. Psychopharmacology (Berl) 2013; 230:399-413. [PMID: 23783774 DOI: 10.1007/s00213-013-3168-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 05/28/2013] [Indexed: 12/24/2022]
Abstract
RATIONALE The molecular mechanisms underlying susceptibility to psychostimulant addiction remain unclear. Searching for commonalities in the effects of addictive drugs on brain gene expression is a prolific approach to determine transcriptional signatures influencing drug abuse. OBJECTIVE We explored the common transcriptional responses to the reinforcing effects of psychostimulants methamphetamine, amphetamine, and methylphenidate. We also aimed to identify transcriptional changes that may subserve abuse of these drugs. METHODS Genome-wide transcriptome profiling analyses were performed to identify common prefrontal cortical (PFC) and striatal gene expression profiles in drug-naïve (cohort 1) and stimulant-pretreated (cohort 2) rats, which showed a conditioned place preference to and self-administration of methamphetamine, amphetamine, and methylphenidate. RESULTS In behavioral studies, stimulant-pretreated rats showed behavioral sensitization characterized by enhanced behavioral response to the rewarding or reinforcing effects of psychostimulants. Inflammation-associated genes (e.g., Alas1, S100a8 and S100a9) were identified as the primary differentially expressed genes (DEGs) in both the PFC and the striatum of cohort 1 rats, while neuronal plasticity (Sgk1)- and brain development (e.g., Bhlhe22, Neurod1, Nr4a2, and Msx1)-associated genes comprised the major upregulated DEGs in the striatum of cohort 2 rats. Furthermore, a meta-analysis of the common striatal DEGs in this study along with morphine-regulated striatal transcriptomes in mice (National Center for Biotechnology Information-Gene Expression Omnibus Database Accession Code GSE7762) suggested similar expression profiles of genes involved in neuronal development (e.g., Bhlhe22, Nr4a2). CONCLUSION This study provides evidence that brain development-associated genes mediate the reinforcing effects of methamphetamine, amphetamine, and methylphenidate and that these transcripts may underlie susceptibility to psychostimulant addiction.
Collapse
Affiliation(s)
- Ike Dela Peña
- Uimyung Research Institute for Neuroscience, Sahmyook University, 26-21 Kongreung-2-dong, Hwarangro-815, Nowon-gu, Seoul, 139-742, South Korea
| | | | | | | | | | | | | |
Collapse
|
19
|
Chambers RA, McClintick JN, Sentir AM, Berg SA, Runyan M, Choi KH, Edenberg HJ. Cortical-striatal gene expression in neonatal hippocampal lesion (NVHL)-amplified cocaine sensitization. GENES BRAIN AND BEHAVIOR 2013; 12:564-75. [PMID: 23682998 DOI: 10.1111/gbb.12051] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 05/01/2013] [Accepted: 05/14/2013] [Indexed: 11/30/2022]
Abstract
Cortical-striatal circuit dysfunction in mental illness may enhance addiction vulnerability. Neonatal ventral hippocampal lesions (NVHL) model this dual diagnosis causality by producing a schizophrenia syndrome with enhanced responsiveness to addictive drugs. Rat genome-wide microarrays containing >24 000 probesets were used to examine separate and co-occurring effects of NVHLs and cocaine sensitization (15 mg/kg/day × 5 days) on gene expression within medial prefrontal cortex (MPFC), nucleus accumbens (NAC), and caudate-putamen (CAPU). Two weeks after NVHLs robustly amplified cocaine behavioral sensitization, brains were harvested for genes of interest defined as those altered at P < 0.001 by NVHL or cocaine effects or interactions. Among 135 genes so impacted, NVHLs altered twofold more than cocaine, with half of all changes in the NAC. Although no genes were changed in the same direction by both NVHL and cocaine history, the anatomy and directionality of significant changes suggested synergy on the neural circuit level generative of compounded behavioral phenotypes: NVHL predominantly downregulated expression in MPFC and NAC while NVHL and cocaine history mostly upregulated CAPU expression. From 75 named genes altered by NVHL or cocaine, 27 had expression levels that correlated significantly with degree of behavioral sensitization, including 11 downregulated by NVHL in MPFC/NAC, and 10 upregulated by NVHL or cocaine in CAPU. These findings suggest that structural and functional impoverishment of prefrontal-cortical-accumbens circuits in mental illness is associated with abnormal striatal plasticity compounding with that in addictive disease. Polygenetic interactions impacting neuronal signaling and morphology within these networks likely contribute to addiction vulnerability in mental illness.
Collapse
Affiliation(s)
- R A Chambers
- Lab for Translational Neuroscience of Dual Diagnosis & Development, Department of Psychiatry, Institute for Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Nielsen DA, Utrankar A, Reyes JA, Simons DD, Kosten TR. Epigenetics of drug abuse: predisposition or response. Pharmacogenomics 2013; 13:1149-60. [PMID: 22909205 DOI: 10.2217/pgs.12.94] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Drug addiction continues to be a serious medical and social problem. Vulnerability to develop an addiction to drugs is dependent on genetic, environmental, social and biological factors. In particular, the interactions of environmental and genetic factors indicate the significance of epigenetic mechanisms, which have been found to occur in response to illicit drug use or as underlying factors in chronic substance abuse and relapse. Epigenetics is defined as the heritable and possibly reversible modifications in gene expression that do not involve alterations in the DNA sequence. This review discusses the various types of epigenetic modifications and their relevance to drug addiction to elucidate whether epigenetics is a predisposing factor, or a response to, developing an addiction to drugs of abuse.
Collapse
Affiliation(s)
- David A Nielsen
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine & the Michael E DeBakey VA Medical Center, 2002 Holcombe Boulevard, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
21
|
Yoo JH, Kitchen I, Bailey A. The endogenous opioid system in cocaine addiction: what lessons have opioid peptide and receptor knockout mice taught us? Br J Pharmacol 2012; 166:1993-2014. [PMID: 22428846 DOI: 10.1111/j.1476-5381.2012.01952.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cocaine addiction has become a major concern in the UK as Britain tops the European 'league table' for cocaine abuse. Despite its devastating health and socio-economic consequences, no effective pharmacotherapy for treating cocaine addiction is available. Identifying neurochemical changes induced by repeated drug exposure is critical not only for understanding the transition from recreational drug use towards compulsive drug abuse but also for the development of novel targets for the treatment of the disease and especially for relapse prevention. This article focuses on the effects of chronic cocaine exposure and withdrawal on each of the endogenous opioid peptides and receptors in rodent models. In addition, we review the studies that utilized opioid peptide or receptor knockout mice in order to identify and/or clarify the role of different components of the opioid system in cocaine-addictive behaviours and in cocaine-induced alterations of brain neurochemistry. The review of these studies indicates a region-specific activation of the µ-opioid receptor system following chronic cocaine exposure, which may contribute towards the rewarding effect of the drug and possibly towards cocaine craving during withdrawal followed by relapse. Cocaine also causes a region-specific activation of the κ-opioid receptor/dynorphin system, which may antagonize the rewarding effect of the drug, and at the same time, contribute to the stress-inducing properties of the drug and the triggering of relapse. These conclusions have important implications for the development of effective pharmacotherapy for the treatment of cocaine addiction and the prevention of relapse.
Collapse
Affiliation(s)
- Ji Hoon Yoo
- Division of Biochemistry, Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | | | | |
Collapse
|
22
|
Addiction-related gene regulation: risks of exposure to cognitive enhancers vs. other psychostimulants. Prog Neurobiol 2012; 100:60-80. [PMID: 23085425 DOI: 10.1016/j.pneurobio.2012.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/02/2012] [Accepted: 10/08/2012] [Indexed: 01/09/2023]
Abstract
The psychostimulants methylphenidate (Ritalin, Concerta), amphetamine (Adderall), and modafinil (Provigil) are widely used in the treatment of medical conditions such as attention-deficit hyperactivity disorder and narcolepsy and, increasingly, as "cognitive enhancers" by healthy people. The long-term neuronal effects of these drugs, however, are poorly understood. A substantial amount of research over the past two decades has investigated the effects of psychostimulants such as cocaine and amphetamines on gene regulation in the brain because these molecular changes are considered critical for psychostimulant addiction. This work has determined in some detail the neurochemical and cellular mechanisms that mediate psychostimulant-induced gene regulation and has also identified the neuronal systems altered by these drugs. Among the most affected brain systems are corticostriatal circuits, which are part of cortico-basal ganglia-cortical loops that mediate motivated behavior. The neurotransmitters critical for such gene regulation are dopamine in interaction with glutamate, while other neurotransmitters (e.g., serotonin) play modulatory roles. This review presents (1) an overview of the main findings on cocaine- and amphetamine-induced gene regulation in corticostriatal circuits in an effort to provide a cellular framework for (2) an assessment of the molecular changes produced by methylphenidate, medical amphetamine (Adderall), and modafinil. The findings lead to the conclusion that protracted exposure to these cognitive enhancers can induce gene regulation effects in corticostriatal circuits that are qualitatively similar to those of cocaine and other amphetamines. These neuronal changes may contribute to the addiction liability of the psychostimulant cognitive enhancers.
Collapse
|
23
|
Matsumoto RR. Targeting sigma receptors: novel medication development for drug abuse and addiction. Expert Rev Clin Pharmacol 2012; 2:351-8. [PMID: 22112179 DOI: 10.1586/ecp.09.18] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Psychostimulant abuse is a serious health and societal problem in industrialized and developing countries. However, the identification of an effective pharmacotherapy to treat it has remained elusive. It has long been known that many psychostimulant drugs, including cocaine and methamphetamine, interact with sigma receptors in the brain and heart, offering a logical target for medication development efforts. However, selective pharmacological agents and molecular biological tools have only recently become available to rigorously evaluate these receptors as viable medication development targets. The current review will summarize provocative preclinical data, demonstrating the ability of sigma receptor antagonists and antisense oligonucleotides to ameliorate cocaine-induced convulsions, lethality, locomotor activity and sensitization, and conditioned place-preference in rodents. Recent studies suggest that the protective effects of sigma receptor antagonists also extend to actions produced by methamphetamine, 3,4-methylenedioxymethamphetamine, ethanol and other abused substances. Together, the data indicate that targeting sigma receptors, particularly the σ(1)-subtype, may offer an innovative approach for combating the effects of cocaine, and perhaps other abused substances.
Collapse
Affiliation(s)
- Rae R Matsumoto
- School of Pharmacy, West Virginia University, PO Box 9500, Morgantown, WV 26506, USA.
| |
Collapse
|
24
|
Schmidt HD, Sangrey GR, Darnell SB, Schassburger RL, Cha JHJ, Pierce RC, Sadri-Vakili G. Increased brain-derived neurotrophic factor (BDNF) expression in the ventral tegmental area during cocaine abstinence is associated with increased histone acetylation at BDNF exon I-containing promoters. J Neurochem 2012; 120:202-9. [PMID: 22043863 PMCID: PMC3243782 DOI: 10.1111/j.1471-4159.2011.07571.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent evidence suggests that the persistence of cocaine seeking during periods of protracted drug abstinence following chronic cocaine exposure is mediated, in part, by neuroadaptations in the mesolimbic dopamine system. Specifically, incubation of cocaine-seeking behavior coincides with increased brain-derived neurotrophic factor (BDNF) protein expression in the ventral tegmental area (VTA). However, the molecular mechanisms that regulate time-dependent changes in VTA BDNF protein expression during cocaine abstinence are unclear. The goal of these experiments was to determine whether VTA BDNF transcript levels are altered following cocaine abstinence and identify the molecular mechanisms regulating cocaine-induced changes in VTA BDNF transcription. Rats were allowed to self-administer cocaine (0.25 mg/infusion, i.v.) for 14 days on a fixed-ratio schedule of reinforcement followed by 7 days of forced drug abstinence. BDNF protein and exon I-containing transcripts were significantly increased in the VTA of cocaine-experienced rats following 7 days of forced drug abstinence compared to yoked saline controls. Cocaine-induced changes in BDNF mRNA were associated with increased acetylation of histone 3 and binding of CREB-binding protein to exon I-containing promoters in the VTA. Taken together, these results suggest that drug abstinence following cocaine self-administration remodels chromatin in the VTA resulting in increased expression of BDNF, which may contribute to neuroadaptations underlying cocaine craving and relapse.
Collapse
Affiliation(s)
- Heath D Schmidt
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania School, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Fernàndez-Castillo N, Orejarena MJ, Ribasés M, Blanco E, Casas M, Robledo P, Maldonado R, Cormand B. Active and passive MDMA ('ecstasy') intake induces differential transcriptional changes in the mouse brain. GENES BRAIN AND BEHAVIOR 2011; 11:38-51. [PMID: 21951708 DOI: 10.1111/j.1601-183x.2011.00735.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, 'ecstasy') is a recreational drug widely used by adolescents and young adults. Although its rewarding effects are well established, there is controversy on its addictive potential. We aimed to compare the consequences of active and passive MDMA administration on gene expression in the mouse brain since all previous studies were based on passive MDMA administration. We used a yoked-control operant intravenous self-administration paradigm combined with microarray technology. Transcriptomic profiles of ventral striatum, frontal cortex, dorsal raphe nucleus and hippocampus were analysed in mice divided in contingent MDMA, yoked MDMA and yoked saline groups, and several changes were validated by quantitative reverse transcription polymerase chain reaction (qRT-PCR). The comparison of contingent MDMA and yoked MDMA vs. yoked saline mice allowed the identification of differential expression in several genes, most of them with immunological and inflammatory functions, but others being involved in neuroadaptation. In the comparison of contingent MDMA vs. yoked MDMA administration, hippocampus and the dorsal raphe nucleus showed statistically significant changes. The altered expression of several genes involved in neuroadaptative changes and synapse function, which may be related to learning self-administration behaviour, could be validated in these two brain structures. In conclusion, our study shows a strong effect of MDMA administration on the expression of immunological and inflammatory genes in all the four brain regions studied. In addition, experiments on MDMA self-administration suggest that the dorsal raphe nucleus and hippocampus may be involved in active MDMA-seeking behaviour, and show specific alterations on gene expression that support the addictive potential of this drug.
Collapse
Affiliation(s)
- N Fernàndez-Castillo
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
26
|
CREB-mediated alterations in the amygdala transcriptome: coordinated regulation of immune response genes following cocaine. Int J Neuropsychopharmacol 2011; 14:1111-26. [PMID: 21138621 PMCID: PMC3970411 DOI: 10.1017/s1461145710001392] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The neuronal circuitry underlying stress- and drug-induced reinstatement of cocaine-seeking has been relatively well characterized; however, less is known regarding the long-term molecular changes following cocaine administration that may promote future reinstatement. The transcription factor cAMP response element-binding protein (CREB) is necessary for stress- but not cocaine-induced reinstatement of conditioned reward, suggesting that different molecular mechanisms may underlie these two types of reinstatement. To explore the relationship between this transcription factor and reinstatement, we utilized the place-conditioning paradigm to examine alterations in gene expression in the amygdala, a neural substrate critically involved in stress-induced reinstatement, following the development of cocaine reward and subsequent extinction. Our findings demonstrate that the amygdala transcriptome was altered by CREB deficiency more than by previous cocaine experience, with an over-representation of genes involved in the immune response. However, a subset of genes involved in stress and immune response demonstrated a drug×genotype interaction, indicating that cocaine produces different long-term alterations in gene expression depending on the presence or absence of CREB. This profile of gene expression in the context of addiction enhances our understanding of the long-term molecular changes that occur throughout the addiction cycle and identifies novel genes and pathways that might lead to the creation of better therapeutic agents.
Collapse
|
27
|
Voigt RM, Mickiewicz AL, Napier TC. Repeated mirtazapine nullifies the maintenance of previously established methamphetamine-induced conditioned place preference in rats. Behav Brain Res 2011; 225:91-6. [PMID: 21771613 DOI: 10.1016/j.bbr.2011.07.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 06/30/2011] [Accepted: 07/05/2011] [Indexed: 11/30/2022]
Abstract
The atypical antidepressant mirtazapine enhances monoaminergic transmission; thus, mirtazapine therapy may counter the hypo-activation of monoamine systems associated with withdrawal from methamphetamine abuse. Human addiction therapy will likely require chronic administration that is given after brain and behavioral maladaptations are established. To emulate this scenario in rats, we ascertained if acute or repeated mirtazapine treatments could antagonize previously established consequences of repeated methamphetamine. Methamphetamine-induced conditioned place preference (CPP) was used, wherein methamphetamine (1mg/kg, i.p.) was administered in a unique environmental context once-daily for three days interposed by saline injections in an alternate context. Subsequently, mirtazapine (5mg/kg, i.p.) was administered in the home cage either as 10 once-daily injections or a single injection. The expression of CPP was determined in drug-free rats three days after the last mirtazapine injection. Expression of methamphetamine-induced CPP was inhibited by 10 home cage administrations of mirtazapine but not by a single injection of mirtazapine. These findings reveal that mirtazapine can inhibit the maintenance of methamphetamine-induced CPP and that treatment duration and/or treatment timing contributes to this effect of mirtazapine.
Collapse
Affiliation(s)
- Robin M Voigt
- Department of Pharmacology & Experimental Therapeutics, Loyola University Chicago Medical Center, Maywood, IL, United States.
| | | | | |
Collapse
|
28
|
Hippocampal CA1 region shows differential regulation of gene expression in mice displaying extremes in behavioral sensitization to amphetamine: relevance for psychosis susceptibility? Psychopharmacology (Berl) 2011; 217:525-38. [PMID: 21537941 PMCID: PMC3180555 DOI: 10.1007/s00213-011-2313-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 04/09/2011] [Indexed: 11/27/2022]
Abstract
RATIONALE Psychosis susceptibility is mediated in part by the dopaminergic neurotransmitter system. In humans, individual differences in vulnerability for psychosis are reflected in differential sensitivity for psychostimulants such as amphetamine. We hypothesize that the same genes and pathways underlying behavioral sensitization in mice are also involved in the vulnerability to psychosis. OBJECTIVES The aim of the current study was to investigate which genes and pathways may contribute to behavioral sensitization in different dopaminergic output areas in the mouse brain. METHODS We took advantage of the naturally occurring difference in psychostimulant sensitivity in DBA/2 mice and selected animals displaying extremes in behavioral sensitization to amphetamine. Subsequently, the dopamine output areas, prefrontal cortex, nucleus accumbens, and cornu ammonis 1 (CA1) area of the hippocampus, were isolated by laser microdissection and subjected to DNA microarray analysis 1 h after a challenge dose of amphetamine. RESULTS A large number of genes with differential expression between high and low responders were identified, with no overlap between brain regions. Validation of these gene expression changes with real-time quantitative polymerase chain reaction demonstrated that the most robust and reproducible effects on gene expression were in the CA1 region of the hippocampus. Interestingly, many of the validated genes in CA1 are members of the cAMP response element (CRE) family and targets of the glucocorticoid receptor (GR) and myocyte enhancer factor 2 (Mef2) transcription factors. CONCLUSION We hypothesize that CRE, Mef2, and GR signaling form a transcription regulating network, which underlies differential amphetamine sensitivity, and therefore, may play an important role in susceptibility to psychosis.
Collapse
|
29
|
Ho MK, Goldman D, Heinz A, Kaprio J, Kreek MJ, Li MD, Munafò MR, Tyndale RF. Breaking barriers in the genomics and pharmacogenetics of drug addiction. Clin Pharmacol Ther 2010; 88:779-91. [PMID: 20981002 PMCID: PMC3738009 DOI: 10.1038/clpt.2010.175] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Drug addiction remains a substantial health issue with limited treatment options currently available. Despite considerable advances in the understanding of human genetic architecture, the genetic underpinning of complex disorders remains elusive. On the basis of our current understanding of neurobiology, numerous candidate genes have been implicated in the etiology and response to treatment for different addictions. Genome-wide association (GWA) studies have also identified novel targets. However, replication of these studies is often lacking, and this complicates interpretation. The situation is expected to improve as issues such as phenotypic characterization, the apparent "missing heritability," the identification of functional variants, and possible gene-environment (G × E) interactions are addressed. In addition, there is growing evidence that genetic information can be useful in refining the choice of addiction treatment. As genetic testing becomes more common in the practice of medicine, a variety of ethical and practical challenges, some of which are unique to drug addiction, will also need to be considered.
Collapse
Affiliation(s)
- M K Ho
- Centre for Addiction & Mental Health, Department of Psychiatry, Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
30
|
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: 8.5] [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
|
31
|
Huang MC, Ho CW, Chen CH, Liu SC, Chen CC, Leu SJ. Reduced expression of circadian clock genes in male alcoholic patients. Alcohol Clin Exp Res 2010; 34:1899-904. [PMID: 20735373 DOI: 10.1111/j.1530-0277.2010.01278.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND There are clear interactions between chronic alcohol consumption and circadian rhythmicity that is regulated by several circadian clock genes. The altered expressions of these genes have been mainly described in animals. The mammalian master clock in the suprachiasmatic nuclei orchestrates the biological rhythms in peripheral tissues. As peripheral blood mononuclear cells (PBMCs) are the most accessible tissue clinically, we assessed the mRNA levels of these genes in patients with alcohol dependence (AD) undergoing alcohol-withdrawal (AW) treatment. METHODS Twenty-two male patients fulfilled the DSM-IV diagnostic criteria of AD, and 12 comparison healthy control subjects were recruited. The patients with AD were further divided by the presence of delirium tremens (DTs), the most severe form of AW syndrome, into DT group and non-DT group. All the participants received blood withdrawal at 9 am, while the patients with AD had blood collection twice: on the next morning of admission (baseline) and on the seventh day. PBMCs were isolated from whole blood, and the mRNA expression profiles of hClock1, hBmal1, hPer1, hPer2, hCry1, and hCry2 were determined by quantitative real-time PCR. RESULTS The baseline mRNA levels of the target circadian clock genes were markedly lower in patients with AD than in control subjects. After 1 week of alcohol detoxification, there were very limited restorations of discrete circadian gene expressions. DT group did not differ in the expression patterns of circadian clock genes from non-DT group. CONCLUSIONS This is the first study demonstrating the overall lowering of circadian clock genes among patients with AD. The expression pattern is comparable between patients with and without DTs. Although preliminary with data at only one single time point, the observation of strikingly reduced mRNA levels supports the association between circadian clock gene dysregulation and chronic alcohol intake.
Collapse
Affiliation(s)
- Ming-Chyi Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
32
|
Suman A, Mehta B, Guo ML, Chu XP, Fibuch EE, Mao LM, Wang JQ. Alterations in subcellular expression of acid-sensing ion channels in the rat forebrain following chronic amphetamine administration. Neurosci Res 2010; 68:1-8. [PMID: 20566346 DOI: 10.1016/j.neures.2010.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 05/27/2010] [Accepted: 06/01/2010] [Indexed: 12/20/2022]
Abstract
Acid-sensing ion channels (ASICs) are densely expressed in broad areas of mammalian brains and actively modulate synaptic transmission and a variety of neuronal activities. To explore whether ASICs are linked to addictive properties of drugs of abuse, we investigated the effect of the psychostimulant amphetamine on subcellular ASIC expression in the rat forebrain in vivo. Repeated administration of amphetamine (once daily for 7 days, 1.25 mg/kg for days 1/7, 4 mg/kg for days 2-6) induced typical behavioral sensitization. At a 14-day withdrawal period, ASIC1 protein levels were increased in the defined surface and intracellular compartments in the striatum (both caudate putamen and nucleus accumbens) in amphetamine-treated rats relative to saline-treated rats as detected by a surface protein cross-linking assay. ASIC2 proteins, however, remained stable in the striatum. In the medial prefrontal cortex, repeated amphetamine administration had no effect on ASIC1 expression in either the surface or the intracellular pool. However, amphetamine selectively reduced the surface expression of ASIC2 in this region. These data identify ASICs as a sensitive target to repeated stimulant exposure. The region- and compartment-specific regulation of ASIC1 and ASIC2 expression may constitute a key synaptic adaptation in reward circuits critical for psychomotor plasticity.
Collapse
Affiliation(s)
- Ajay Suman
- Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Hemby SE. Cocainomics: new insights into the molecular basis of cocaine addiction. J Neuroimmune Pharmacol 2010; 5:70-82. [PMID: 20084466 PMCID: PMC3255087 DOI: 10.1007/s11481-009-9189-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 12/21/2009] [Indexed: 02/06/2023]
Abstract
Until recently, knowledge of the impact of abused drugs on gene and protein expression in the brain was limited to less than 100 targets. With the advent of high-throughput genomic and proteomic techniques, investigators are now able to evaluate changes across the entire genome and across thousands of proteins in defined brain regions and generate expression profiles of vulnerable neuroanatomical substrates in rodent and nonhuman primate drug abuse models and in human post-mortem brain tissue from drug abuse victims. The availability of gene and protein expression profiles will continue to expand our understanding of the short- and long-term consequences of drug addiction and other addictive disorders and may provide new approaches or new targets for pharmacotherapeutic intervention. This review summarizes several important genomic and proteomic studies of cocaine abuse/addiction from rodent, nonhuman primate, and human postmortem studies of cocaine abuse and explores how these studies have advanced our understanding of addiction.
Collapse
Affiliation(s)
- Scott E Hemby
- Department of Physiology and Pharmacology, Center for the Neurobiology of Addiction and Treatment, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
34
|
Tannu NS, Howell LL, Hemby SE. Integrative proteomic analysis of the nucleus accumbens in rhesus monkeys following cocaine self-administration. Mol Psychiatry 2010; 15:185-203. [PMID: 18504425 PMCID: PMC3272768 DOI: 10.1038/mp.2008.53] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Revised: 03/12/2008] [Accepted: 04/10/2008] [Indexed: 11/09/2022]
Abstract
The reinforcing effects and long-term consequences of cocaine self-administration have been associated with brain regions of the mesolimbic dopamine pathway, namely the nucleus accumbens (NAc). Studies of cocaine-induced biochemical adaptations in rodent models have advanced our knowledge; however, unbiased detailed assessments of intracellular alterations in the primate brain are scarce, yet essential, to develop a comprehensive understanding of cocaine addiction. To this end, two-dimensional difference in gel electrophoresis (2D-DIGE) was used to compare changes in cytosolic protein abundance in the NAc between rhesus monkeys self-administering cocaine and controls. Following image normalization, spots with significantly differential image intensities (P<0.05) were identified, excised, trypsin digested and analyzed by matrix-assisted laser-desorption ionization time-of-flight time-of-flight (MALDI-TOF-TOF). In total, 1098 spots were subjected to statistical analysis with 22 spots found to be differentially abundant of which 18 proteins were positively identified by mass spectrometry. In addition, approximately 1000 protein spots were constitutively expressed of which 21 proteins were positively identified by mass spectrometry. Increased levels of proteins in the cocaine-exposed monkeys include glial fibrillary acidic protein, syntaxin-binding protein 3, protein kinase C isoform, adenylate kinase isoenzyme 5 and mitochondrial-related proteins, whereas decreased levels of proteins included beta-soluble N-ethylmaleimide-sensitive factor attachment protein and neural and non-neural enolase. Using a complimentary proteomics approach, the differential expression of phosphorylated proteins in the cytosolic fraction of these subjects was examined. Two-dimensional gel electrophoresis (2DGE) was followed by gel staining with Pro-Q Diamond phosphoprotein gel stain, enabling differentiation of approximately 150 phosphoprotein spots between the groups. Following excision and trypsin digestions, MALDI-TOF-TOF was used to confirm the identity of 15 cocaine-altered phosphoproteins. Significant increased levels were detected for gamma-aminobutyric acid type A receptor-associated protein 1, 14-3-3 gamma-protein, glutathione S-transferase and brain-type aldolase, whereas significant decreases were observed for beta-actin, Rab GDP-dissociation inhibitor, guanine deaminase, peroxiredoxin 2 isoform b and several mitochondrial proteins. Results from these studies indicate coordinated dysregulation of proteins related to cell structure, signaling, metabolism and mitochondrial function. These data extend and compliment previous studies of cocaine-induced biochemical alterations in human postmortem brain tissue, using an animal model that closely recapitulates the human condition and provide new insight into the molecular basis of the disease and potential targets for pharmacotherapeutic intervention.
Collapse
Affiliation(s)
- NS Tannu
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - LL Howell
- Neuroscience Division, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - SE Hemby
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Psychiatry and Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
35
|
Winrow CJ, Tanis KQ, Reiss DR, Rigby AM, Uslaner JM, Uebele VN, Doran SM, Fox SV, Garson SL, Gotter AL, Levine DM, Roecker AJ, Coleman PJ, Koblan KS, Renger JJ. Orexin receptor antagonism prevents transcriptional and behavioral plasticity resulting from stimulant exposure. Neuropharmacology 2010; 58:185-94. [DOI: 10.1016/j.neuropharm.2009.07.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 07/02/2009] [Accepted: 07/03/2009] [Indexed: 12/20/2022]
|
36
|
Meaburn EL, Fernandes C, Craig IW, Plomin R, Schalkwyk LC. Assessing individual differences in genome-wide gene expression in human whole blood: reliability over four hours and stability over 10 months. Twin Res Hum Genet 2009; 12:372-80. [PMID: 19653838 DOI: 10.1375/twin.12.4.372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Studying the causes and correlates of natural variation in gene expression in healthy populations assumes that individual differences in gene expression can be reliably and stably assessed across time. However, this is yet to be established. We examined 4-hour test-retest reliability and 10 month test-retest stability of individual differences in gene expression in ten 12-year-old children. Blood was collected on four occasions: 10 a.m. and 2 p.m. on Day 1 and 10 months later at 10 a.m. and 2 p.m. Total RNA was hybridized to Affymetrix-U133 plus 2.0 arrays. For each probeset, the correlation across individuals between 10 a.m. and 2 p.m. on Day 1 estimates test-retest reliability. We identified 3,414 variable and abundantly expressed probesets whose 4-hour test-retest reliability exceeded .70, a conventionally accepted level of reliability, which we had 80% power to detect. Of the 3,414 reliable probesets, 1,752 were also significantly reliable 10 months later. We assessed the long-term stability of individual differences in gene expression by correlating the average expression level for each probe-set across the two 4-hour assessments on Day 1 with the average level of each probe-set across the two 4-hour assessments 10 months later. 1,291 (73.7%) of the 1,752 probe-sets that reliably detected individual differences across 4 hours on two occasions, 10 months apart, also stably detected individual differences across 10 months. Heritability, as estimated from the MZ twin intraclass correlations, is twice as high for the 1,752 reliable probesets versus all present probesets on the array (0.68 vs 0.34), and is even higher (0.76) for the 1,291 reliable probesets that are also stable across 10 months. The 1,291 probesets that reliably detect individual differences from a single peripheral blood collection and stably detect individual differences over 10 months are promising targets for research on the causes (e.g., eQTLs) and correlates (e.g., psychopathology) of individual differences in gene expression.
Collapse
Affiliation(s)
- Emma L Meaburn
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom.
| | | | | | | | | |
Collapse
|
37
|
Le Merrer J, Becker JAJ, Befort K, Kieffer BL. Reward processing by the opioid system in the brain. Physiol Rev 2009; 89:1379-412. [PMID: 19789384 DOI: 10.1152/physrev.00005.2009] [Citation(s) in RCA: 702] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The opioid system consists of three receptors, mu, delta, and kappa, which are activated by endogenous opioid peptides processed from three protein precursors, proopiomelanocortin, proenkephalin, and prodynorphin. Opioid receptors are recruited in response to natural rewarding stimuli and drugs of abuse, and both endogenous opioids and their receptors are modified as addiction develops. Mechanisms whereby aberrant activation and modifications of the opioid system contribute to drug craving and relapse remain to be clarified. This review summarizes our present knowledge on brain sites where the endogenous opioid system controls hedonic responses and is modified in response to drugs of abuse in the rodent brain. We review 1) the latest data on the anatomy of the opioid system, 2) the consequences of local intracerebral pharmacological manipulation of the opioid system on reinforced behaviors, 3) the consequences of gene knockout on reinforced behaviors and drug dependence, and 4) the consequences of chronic exposure to drugs of abuse on expression levels of opioid system genes. Future studies will establish key molecular actors of the system and neural sites where opioid peptides and receptors contribute to the onset of addictive disorders. Combined with data from human and nonhuman primate (not reviewed here), research in this extremely active field has implications both for our understanding of the biology of addiction and for therapeutic interventions to treat the disorder.
Collapse
Affiliation(s)
- Julie Le Merrer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Département Neurobiologie et Génétique, Illkirch, France
| | | | | | | |
Collapse
|
38
|
Schramm-Sapyta NL, Walker QD, Caster JM, Levin ED, Kuhn CM. Are adolescents more vulnerable to drug addiction than adults? Evidence from animal models. Psychopharmacology (Berl) 2009; 206:1-21. [PMID: 19547960 PMCID: PMC3025448 DOI: 10.1007/s00213-009-1585-5] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2008] [Accepted: 05/26/2009] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND RATIONALE Epidemiological evidence suggests that people who begin experimenting with drugs of abuse during early adolescence are more likely to develop substance use disorders (SUDs), but this correlation does not guarantee causation. Animal models, in which age of onset can be tightly controlled, offer a platform for testing causality. Many animal models address drug effects that might promote or discourage drug intake and drug-induced neuroplasticity. METHODS We have reviewed the preclinical literature to investigate whether adolescent rodents are differentially sensitive to rewarding, reinforcing, aversive, locomotor, and withdrawal-induced effects of drugs of abuse. RESULTS AND CONCLUSIONS The rodent model literature consistently suggests that the balance of rewarding and aversive effects of drugs of abuse is tipped toward reward in adolescence. However, increased reward does not consistently lead to increased voluntary intake: age effects on voluntary intake are drug and method specific. On the other hand, adolescents are consistently less sensitive to withdrawal effects, which could protect against compulsive drug seeking. Studies examining neuronal function have revealed several age-related effects but have yet to link these effects to vulnerability to SUDs. Taken together, the findings suggest factors which may promote recreational drug use in adolescents, but evidence relating to pathological drug-seeking behavior is lacking. A call is made for future studies to address this gap using behavioral models of pathological drug seeking and for neurobiologic studies to more directly link age effects to SUD vulnerability.
Collapse
|
39
|
Webb KJ, Norton WHJ, Trümbach D, Meijer AH, Ninkovic J, Topp S, Heck D, Marr C, Wurst W, Theis FJ, Spaink HP, Bally-Cuif L. Zebrafish reward mutants reveal novel transcripts mediating the behavioral effects of amphetamine. Genome Biol 2009; 10:R81. [PMID: 19646228 PMCID: PMC2728535 DOI: 10.1186/gb-2009-10-7-r81] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/16/2009] [Accepted: 07/31/2009] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Addiction is a pathological dysregulation of the brain's reward systems, determined by several complex genetic pathways. The conditioned place preference test provides an evaluation of the effects of drugs in animal models, allowing the investigation of substances at a biologically relevant level with respect to reward. Our lab has previously reported the development of a reliable conditioned place preference paradigm for zebrafish. Here, this test was used to isolate a dominant N-ethyl-N-nitrosourea (ENU)-induced mutant, no addiction (nad(dne3256)), which fails to respond to amphetamine, and which we used as an entry point towards identifying the behaviorally relevant transcriptional response to amphetamine. RESULTS Through the combination of microarray experiments comparing the adult brain transcriptome of mutant and wild-type siblings under normal conditions, as well as their response to amphetamine, we identified genes that correlate with the mutants' altered conditioned place preference behavior. In addition to pathways classically involved in reward, this gene set shows a striking enrichment in transcription factor-encoding genes classically involved in brain development, which later appear to be re-used within the adult brain. We selected a subset of them for validation by quantitative PCR and in situ hybridization, revealing that specific brain areas responding to the drug through these transcription factors include domains of ongoing adult neurogenesis. Finally, network construction revealed functional connections between several of these genes. CONCLUSIONS Together, our results identify a new network of coordinated gene regulation that influences or accompanies amphetamine-triggered conditioned place preference behavior and that may underlie the susceptibility to addiction.
Collapse
Affiliation(s)
- Katharine J Webb
- Department Zebrafish Neurogenetics, Institute of Developmental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University - Munich, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| | - William HJ Norton
- Department Zebrafish Neurogenetics, Institute of Developmental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University - Munich, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| | - Annemarie H Meijer
- Institute of Biology, University of Leiden, Leiden, 2300 RA The Netherlands
| | - Jovica Ninkovic
- Department Zebrafish Neurogenetics, Institute of Developmental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University - Munich, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
- Current address: Institute of Stem Cell Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| | - Stefanie Topp
- Department Zebrafish Neurogenetics, Institute of Developmental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University - Munich, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| | - Daniel Heck
- Institute for Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| | - Carsten Marr
- Institute for Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| | - Fabian J Theis
- Institute for Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| | - Herman P Spaink
- Institute of Biology, University of Leiden, Leiden, 2300 RA The Netherlands
| | - Laure Bally-Cuif
- Department Zebrafish Neurogenetics, Institute of Developmental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
- Center for Integrated Protein Science (Munich), Institute of Developmental Genetics, Technical University - Munich, Ingolstaedter Landstrasse, Neuherberg, 85764 Germany
| |
Collapse
|
40
|
Abstract
Addictive drugs hijack the human brain's 'reward' systems. A zebrafish model of addiction has recently been used to query changes in gene expression during this process.
Collapse
Affiliation(s)
- Jean Lud Cadet
- Molecular Neuropsychiatry Branch, National Institute on Drug Abuse/IRP, NIH Biomedical Research Center, 251 Bayview Blvd, Baltimore, MD 21224, USA.
| |
Collapse
|
41
|
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: 2.8] [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
|
42
|
Histone acetylation in drug addiction. Semin Cell Dev Biol 2009; 20:387-94. [PMID: 19560043 DOI: 10.1016/j.semcdb.2009.01.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Revised: 01/09/2009] [Accepted: 01/13/2009] [Indexed: 11/22/2022]
Abstract
Regulation of chromatin structure through post-translational modifications of histones (e.g., acetylation) has emerged as an important mechanism to translate a variety of environmental stimuli, including drugs of abuse, into specific changes in gene expression. Since alterations in gene expression are thought to contribute to the development and maintenance of the addicted state, recent efforts are aimed at identifying how drugs of abuse alter chromatin structure and the enzymes which regulate it. This review discusses how drugs of abuse alter histone acetylation in brain reward regions, through which enzymes this occurs, and ultimately what role histone acetylation plays in addiction-related behaviors.
Collapse
|
43
|
Lull ME, Freeman WM, Vrana KE, Mash DC. Correlating human and animal studies of cocaine abuse and gene expression. Ann N Y Acad Sci 2008; 1141:58-75. [PMID: 18991951 DOI: 10.1196/annals.1441.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Gene expression changes resulting from cocaine abuse in both humans and animal models have been studied for several decades. Although human studies have been very useful at illuminating cocaine-related expression changes, there are many factors complicating these studies, including the difficulty of obtaining high-quality postmortem brain tissue and patient comorbidities. Animal models of cocaine abuse have served as valuable additions to human data and allow examination of specific aspects of cocaine abuse, including immediate early gene expression and the molecular effects of abstinence and relapse. In total, human and animal studies of cocaine abuse have uncovered gene expression changes in the brain related to a number of molecular functions, including the extracellular matrix, synaptic communication and neuroplasticity, receptors, ion channels and transporters, oligodendrocytes and myelin, apoptosis and cell death, mitochondrial function, signal transduction, and transcription factors. In addition, the mitogen-activated protein kinase and synaptic long-term potentiation signal transduction pathways are highlighted as pathways in which multiple components are altered by cocaine. Pathways and processes affected by changes in gene expression that overlap among multiple species may be promising pharmacotherapeutic targets for reducing the behavioral effects of cocaine abuse and the relapse potential observed in humans.
Collapse
Affiliation(s)
- Melinda E Lull
- Department of Pharmacology, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
44
|
Kily LJM, Cowe YCM, Hussain O, Patel S, McElwaine S, Cotter FE, Brennan CH. Gene expression changes in a zebrafish model of drug dependency suggest conservation of neuro-adaptation pathways. ACTA ACUST UNITED AC 2008; 211:1623-34. [PMID: 18456890 DOI: 10.1242/jeb.014399] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Addiction is a complex psychiatric disorder considered to be a disease of the brain's natural reward reinforcement system. Repeated stimulation of the 'reward' pathway leads to adaptive changes in gene expression and synaptic organization that reinforce drug taking and underlie long-term changes in behaviour. The primitive nature of reward reinforcement pathways and the near universal ability of abused drugs to target the same system allow drug-associated reward and reinforcement to be studied in non-mammalian species. Zebrafish have proved to be a valuable model system for the study of vertebrate development and disease. Here we demonstrate that adult zebrafish show a dose-dependent acute conditioned place preference (CPP) reinforcement response to ethanol or nicotine. Repeated exposure of adult zebrafish to either nicotine or ethanol leads to a robust CPP response that persists following 3 weeks of abstinence and in the face of adverse stimuli, a behavioural indicator of the establishment of dependence. Microarray analysis using whole brain samples from drug-treated and control zebrafish identified 1362 genes that show a significant change in expression between control and treated individuals. Of these genes, 153 are common to both ethanol- and nicotine-treated animals. These genes include members of pathways and processes implicated in drug dependence in mammalian models, revealing conservation of neuro-adaptation pathways between zebrafish and mammals.
Collapse
Affiliation(s)
- Layla J M Kily
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End, London E1 4NS, UK
| | | | | | | | | | | | | |
Collapse
|
45
|
Characterizing intercellular signaling peptides in drug addiction. Neuropharmacology 2008; 56 Suppl 1:196-204. [PMID: 18722391 DOI: 10.1016/j.neuropharm.2008.07.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Revised: 07/23/2008] [Accepted: 07/28/2008] [Indexed: 11/23/2022]
Abstract
Intercellular signaling peptides (SPs) coordinate the activity of cells and influence organism behavior. SPs, a chemically and structurally diverse group of compounds responsible for transferring information between neurons, are broadly involved in neural plasticity, learning and memory, as well as in drug addiction phenomena. Historically, SP discovery and characterization has tracked advances in measurement capabilities. Today, a suite of analytical technologies is available to investigate individual SPs, as well as entire intercellular signaling complements, in samples ranging from individual cells to entire organisms. Immunochemistry and in situ hybridization are commonly used for following preselected SPs. Discovery-type investigations targeting the transcriptome and proteome are accomplished using high-throughput characterization technologies such as microarrays and mass spectrometry. By integrating directed approaches with discovery approaches, multiplatform studies fill critical gaps in our knowledge of drug-induced alterations in intercellular signaling. Throughout the past 35 years, the National Institute on Drug Abuse has made significant resources available to scientists that study the mechanisms of drug addiction. The roles of SPs in the addiction process are highlighted, as are the analytical approaches used to detect and characterize them.
Collapse
|
46
|
Lynch WJ, Girgenti MJ, Breslin FJ, Newton SS, Taylor JR. Gene profiling the response to repeated cocaine self-administration in dorsal striatum: a focus on circadian genes. Brain Res 2008; 1213:166-77. [PMID: 18452895 PMCID: PMC2494701 DOI: 10.1016/j.brainres.2008.02.106] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Revised: 02/27/2008] [Accepted: 02/29/2008] [Indexed: 01/09/2023]
Abstract
Alterations in gene expression in the dorsal striatum caused by chronic cocaine exposure have been implicated in the long-term behavioral changes associated with cocaine addiction. To gain further insight into the molecular alterations that occur as a result of cocaine self-administration, we conducted a microarray analysis of gene expression followed by bioinformatic gene network analysis that allowed us to identify adaptations at the level of gene expression as well as into interconnected networks. Changes in gene expression were examined in the dorsal striatum of rats 1 day after they had self-administered cocaine for 7 days under a 24-h access, discrete trial paradigm (averaging 98 mg/kg/day). Here we report the regulation of the circadian genes Clock, Bmal1, Cryptochrome1, Period2, as well as several genes that are regulated by/associated with the circadian system (i.e., early growth response 1, dynorphin). We also observed regulation of other relevant genes (i.e., Nur77, beta catenin). These changes were then linked to curated pathways and formulated networks which identified circadian rhythm processes as affected by cocaine self-administration. These data strongly suggest involvement of circadian-associated genes in the brain's response to cocaine and may contribute to an understanding of addictive behavior including disruptions in sleep and circadian rhythmicity.
Collapse
Affiliation(s)
- Wendy J Lynch
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, USA.
| | | | | | | | | |
Collapse
|
47
|
Lee KW, Tian YH, You IJ, Kwon SH, Ha RR, Lee SY, Kim HC, Jang CG. Blockade of M1 muscarinic acetylcholine receptors modulates the methamphetamine-induced psychomotor stimulant effect. Neuroscience 2008; 153:1235-44. [DOI: 10.1016/j.neuroscience.2008.02.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Revised: 02/12/2008] [Accepted: 02/16/2008] [Indexed: 11/27/2022]
|
48
|
Abstract
The articulated goals of Dialogues in Clinical Neuroscience are to serve as "an interface between clinical neuropsychiatry and the neurosciences by providing state-of-the-art information and original insights into relevant clinical, biological, and therapeutic aspects." My laboratory the Laboratory of the Biology of Addictive Diseases at The Rockefeller University, has for years been focused on "bidirectional translational research," that is, learning by careful observations and study in patient populations with the disorders under study, in this case primarily specific addictive diseases, and then using that knowledge to create improved animal models or other laboratory-based research paradigms, while, at the same time, taking research findings made at the bench into the clinic as promptly as that is appropriate and feasible. In this invited review, therefore, the focus will be on perspectives of our Laboratory of the Biology of Addictive Diseases and related National Institutes of Health/National Institute on Drug Abuse research Center, including laboratory-based molecular neurobiological research, research using several animal models designed to mimic human patterns of drug abuse and addiction, as well as basic clinical research, intertwined with treatment-related research.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- Laboratory of Biology of Addictive Diseases, Rockefeller University, New York, NY 10021, USA.
| |
Collapse
|
49
|
Li MD, Wang J. Neuroproteomics and its applications in research on nicotine and other drugs of abuse. Proteomics Clin Appl 2007; 1:1406-27. [PMID: 21136639 DOI: 10.1002/prca.200700321] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Indexed: 12/24/2022]
Abstract
The rapidly growing field of neuroproteomics is able to track changes in protein expression and protein modifications underlying various physiological conditions, including the neural diseases related to drug addiction. Thus, it presents great promise in characterizing protein function, biochemical pathways, and networks to understand the mechanisms underlying drug dependence. In this article, we first provide an overview of proteomics technologies and bioinformatics tools available to analyze proteomics data. Then we summarize the recent applications of proteomics to profile the protein expression pattern in animal or human brain tissues after the administration of nicotine, alcohol, amphetamine, butorphanol, cocaine, and morphine. By comparing the protein expression profiles in response to chronic nicotine exposure with those appearing in response to treatment with other drugs of abuse, we identified three biological processes that appears to be regulated by multiple drugs of abuse: energy metabolism, oxidative stress response, and protein degradation and modification. Such similarity indicates that despite the obvious differences among their chemical properties and the receptors with which they interact, different substances of abuse may cause some similar changes in cellular activities and biological processes in neurons.
Collapse
Affiliation(s)
- Ming D Li
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA, USA.
| | | |
Collapse
|
50
|
Perreau-Lenz S, Zghoul T, Spanagel R. Clock genes running amok. Clock genes and their role in drug addiction and depression. EMBO Rep 2007; 8 Spec No:S20-3. [PMID: 17726437 PMCID: PMC3327520 DOI: 10.1038/sj.embor.7401016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Stéphanie Perreau-Lenz
- Department of Psychopharmacology at the Central Institute of Mental Health in Mannheim, Germany.E-mail:
| | - Tarek Zghoul
- Department of Psychopharmacology at the Central Institute of Mental Health in Mannheim, Germany.E-mail:
| | - Rainer Spanagel
- Department of Psychopharmacology at the Central Institute of Mental Health in Mannheim, Germany.E-mail:
| |
Collapse
|