1
|
Rezayof A, Ghasemzadeh Z, Sahafi OH. Addictive drugs modify neurogenesis, synaptogenesis and synaptic plasticity to impair memory formation through neurotransmitter imbalances and signaling dysfunction. Neurochem Int 2023; 169:105572. [PMID: 37423274 DOI: 10.1016/j.neuint.2023.105572] [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: 04/19/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023]
Abstract
Drug abuse changes neurophysiological functions at multiple cellular and molecular levels in the addicted brain. Well-supported scientific evidence suggests that drugs negatively affect memory formation, decision-making and inhibition, and emotional and cognitive behaviors. The mesocorticolimbic brain regions are involved in reward-related learning and habitual drug-seeking/taking behaviors to develop physiological and psychological dependence on the drugs. This review highlights the importance of specific drug-induced chemical imbalances resulting in memory impairment through various neurotransmitter receptor-mediated signaling pathways. The mesocorticolimbic modifications in the expression levels of brain-derived neurotrophic factor (BDNF) and the cAMP-response element binding protein (CREB) impair reward-related memory formation following drug abuse. The contributions of protein kinases and microRNAs (miRNAs), along with the transcriptional and epigenetic regulation have also been considered in memory impairment underlying drug addiction. Overall, we integrate the research on various types of drug-induced memory impairment in distinguished brain regions and provide a comprehensive review with clinical implications addressing the upcoming studies.
Collapse
Affiliation(s)
- Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Zahra Ghasemzadeh
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Oveis Hosseinzadeh Sahafi
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
2
|
Martínez-Rivera A, Hao J, Rice R, Inturrisi CE, Rajadhyaksha AM. Ca v1.3 L-type Ca 2+ channel-activated CaMKII/ERK2 pathway in the ventral tegmental area is required for cocaine conditioned place preference. Neuropharmacology 2023; 224:109368. [PMID: 36481277 PMCID: PMC9796157 DOI: 10.1016/j.neuropharm.2022.109368] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
We have previously demonstrated that pharmacological blockade of ventral tegmental area (VTA) Cav1.3 L-type calcium channels (LTCCs) using Cav1.2 dihydropyridine insensitive (Cav1.2DHP-/-) mutant mice attenuates cocaine conditioned place preference (CPP). However, the molecular mechanisms by which Cav1.3 channels mediate the effects of cocaine in the VTA remain largely unknown. In this study using Cav1.2DHP-/- male mice, we find that cocaine place preference increases CaM kinase IIα, ERK2, and CREB phosphorylation in the VTA, proteins strongly linked to cocaine behaviors. To further explore the causal role of these intracellular signaling proteins in cocaine preference, the CaM kinase II inhibitor, KN93 was directly injected into the VTA of male mice before each cocaine conditioning session. We found that KN93 attenuates conditioned preference for cocaine compared to vehicle treated mice and decreased VTA ERK2 and CREB phosphorylation. Additionally, blockade of the ERK pathway with the MEK inhibitor, U0126 or knockdown of ERK2 using siRNA, attenuated cocaine preference and VTA CREB phosphorylation but not CaMKIIα phosphorylation, suggesting that ERK is activated downstream of CaMKIIα. Examination of postsynaptic density (PSD) GluA1 subunit of AMPA receptors in the nucleus accumbens (NAc) that we have previously shown to be upregulated following long withdrawal periods, was blunted by KN93, U0126 and ERK2 siRNA when examined 30 days following cocaine CPP. Taken together, these findings demonstrate that Cav1.3 channels in the VTA are required for cocaine reward behavior and activation of the CaMKIIα/ERK/CREB signaling pathway in the VTA is necessary for long-lasting changes in the NAc. This article is part of the Special Issue on 'L-type calcium channel mechanisms in neuropsychiatric disorders'.
Collapse
Affiliation(s)
- Arlene Martínez-Rivera
- Department of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA; Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Jin Hao
- Department of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Richard Rice
- Department of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
| | | | - Anjali M Rajadhyaksha
- Department of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA; Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
3
|
Morikawa H, Young CC, Smits JA. Usage of L-type calcium channel blockers to suppress drug reward and memory driving addiction: Past, present, and future. Neuropharmacology 2022; 221:109290. [PMID: 36241085 PMCID: PMC10476140 DOI: 10.1016/j.neuropharm.2022.109290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/02/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Over the past three decades, L-type Ca2+ channel (LTCC) blockers have been considered a potential therapeutic drug to alleviate the symptoms of drug addiction. This idea has been supported, in part, by 1) expression of LTCCs in the brain dopaminergic circuits that are thought to play critical roles in the development and expression of addictive behaviors and 2) common usage of LTCC blockers in treating hypertension, which may enable off-label use of these drugs with good brain penetration as therapeutics for brain disorders. Addiction can be viewed as a maladaptive form of learning where powerful memories of drug-associated stimuli and actions drive compulsive drug intake. Largely under this framework, we will focus on the dopaminergic system that is thought be critically involved in drug-associated learning and memory and provide a brief overview of the past and recent studies testing the therapeutic potential of LTCC blockers for addictive disorders in animal models and humans and offer a future perspective on the use of LTCC blockers in drug addiction and, possibly, addiction to other non-drug rewards (e.g., gambling, eating, shopping). Interested readers can refer to other related articles in this issue and a comprehensive review available elsewhere (Little, 2021) to gain further insights into the roles of LTCCs in drug addiction and withdrawal symptoms associated with dependence. This article is part of the Special Issue on 'L-type calcium channel mechanisms in neuropsychiatric disorders'.
Collapse
Affiliation(s)
- Hitoshi Morikawa
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, USA.
| | | | - Jasper A Smits
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
4
|
Little HJ. L-Type Calcium Channel Blockers: A Potential Novel Therapeutic Approach to Drug Dependence. Pharmacol Rev 2021; 73:127-154. [PMID: 34663686 DOI: 10.1124/pharmrev.120.000245] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This review describes interactions between compounds, primarily dihydropyridines, that block L-type calcium channels and drugs that cause dependence, and the potential importance of these interactions. The main dependence-inducing drugs covered are alcohol, psychostimulants, opioids, and nicotine. In preclinical studies, L-type calcium channel blockers prevent or reduce important components of dependence on these drugs, particularly their reinforcing actions and the withdrawal syndromes. The channel blockers also reduce the development of tolerance and/or sensitization, and they have no intrinsic dependence liability. In some instances, their effects include reversal of brain changes established during drug dependence. Prolonged treatment with alcohol, opioids, psychostimulant drugs, or nicotine causes upregulation of dihydropyridine binding sites. Few clinical studies have been carried out so far, and reports are conflicting, although there is some evidence of effectiveness of L-channel blockers in opioid withdrawal. However, the doses of L-type channel blockers used clinically so far have necessarily been limited by potential cardiovascular problems and may not have provided sufficient central levels of the drugs to affect neuronal dihydropyridine binding sites. New L-type calcium channel blocking compounds are being developed with more selective actions on subtypes of L-channel. The preclinical evidence suggests that L-type calcium channels may play a crucial role in the development of dependence to different types of drugs. Mechanisms for this are proposed, including changes in the activity of mesolimbic dopamine neurons, genomic effects, and alterations in synaptic plasticity. Newly developed, more selective L-type calcium channel blockers could be of considerable value in the treatment of drug dependence. SIGNIFICANCE STATEMENT: Dependence on drugs is a very serious health problem with little effective treatment. Preclinical evidence shows drugs that block particular calcium channels, the L-type, reduce dependence-related effects of alcohol, opioids, psychostimulants, and nicotine. Clinical studies have been restricted by potential cardiovascular side effects, but new, more selective L-channel blockers are becoming available. L-channel blockers have no intrinsic dependence liability, and laboratory evidence suggests they reverse previously developed effects of dependence-inducing drugs. They could provide a novel approach to addiction treatment.
Collapse
Affiliation(s)
- Hilary J Little
- Section of Alcohol Research, National Addiction Centre, Institute of Psychiatry, King's College, London, United Kingdom
| |
Collapse
|
5
|
Cocaine- and stress-primed reinstatement of drug-associated memories elicit differential behavioral and frontostriatal circuit activity patterns via recruitment of L-type Ca 2+ channels. Mol Psychiatry 2020; 25:2373-2391. [PMID: 31501511 PMCID: PMC7927165 DOI: 10.1038/s41380-019-0513-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/12/2019] [Accepted: 07/31/2019] [Indexed: 01/07/2023]
Abstract
Cocaine-associated memories are critical drivers of relapse in cocaine-dependent individuals that can be evoked by exposure to cocaine or stress. Whether these environmental stimuli recruit similar molecular and circuit-level mechanisms to promote relapse remains largely unknown. Here, using cocaine- and stress-primed reinstatement of cocaine conditioned place preference to model drug-associated memories, we find that cocaine drives reinstatement by increasing the duration that mice spend in the previously cocaine-paired context whereas stress increases the number of entries into this context. Importantly, both forms of reinstatement require Cav1.2 L-type Ca2+ channels (LTCCs) in cells of the prelimbic cortex that project to the nucleus accumbens core (PrL→NAcC). Utilizing fiber photometry to measure circuit activity in vivo in conjunction with the LTCC blocker, isradipine, we find that LTCCs drive differential recruitment of the PrL→ NAcC pathway during cocaine- and stress-primed reinstatement. While cocaine selectively activates PrL→NAcC cells prior to entry into the cocaine-paired chamber, a measure that is predictive of duration in that chamber, stress increases persistent activity of this projection, which correlates with entries into the cocaine-paired chamber. Using projection-specific chemogenetic manipulations, we show that PrL→NAcC activity is required for both cocaine- and stress-primed reinstatement, and that activation of this projection in Cav1.2-deficient mice restores reinstatement. These data indicate that LTCCs are a common mediator of cocaine- and stress-primed reinstatement. However, they engage different patterns of behavior and PrL→NAcC projection activity depending on the environmental stimuli. These findings establish a framework to further study how different environmental experiences can drive relapse, and supports further exploration of isradipine, an FDA-approved LTCC blocker, as a potential therapeutic for the prevention of relapse in cocaine-dependent individuals.
Collapse
|
6
|
Moon AL, Brydges NM, Wilkinson LS, Hall J, Thomas KL. Cacna1c Hemizygosity Results in Aberrant Fear Conditioning to Neutral Stimuli. Schizophr Bull 2020; 46:1231-1238. [PMID: 31910256 PMCID: PMC7505182 DOI: 10.1093/schbul/sbz127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CACNA1C, a gene that encodes an alpha-1 subunit of L-type voltage-gated calcium channels, has been strongly associated with psychiatric disorders including schizophrenia and bipolar disorder. An important objective is to understand how variation in this gene can lead to an increased risk of psychopathology. Altered associative learning has also been implicated in the pathology of psychiatric disorders, particularly in the manifestation of psychotic symptoms. In this study, we utilize auditory-cued fear memory paradigms in order to investigate whether associative learning is altered in rats hemizygous for the Cacna1c gene. Cacna1c hemizygous (Cacna1c+/-) rats and their wild-type littermates were exposed to either delay, trace, or unpaired auditory fear conditioning. All rats received a Context Recall (24 h post-conditioning) and a Cue Recall (48 h post-conditioning) to test their fear responses. In the delay condition, which results in strong conditioning to the cue in wild-type animals, Cacna1c+/- rats showed increased fear responses to the context. In the trace condition, which results in strong conditioning to the context in wild-type animals, Cacna1c+/- rats showed increased fear responses to the cue. Finally, in the unpaired condition, Cacna1c+/- rats showed increased fear responses to both context and cue. These results indicate that Cacna1c heterozygous rats show aberrantly enhanced fear responses to inappropriate cues, consistent with key models of psychosis.
Collapse
Affiliation(s)
- Anna L Moon
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Nichola M Brydges
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Lawrence S Wilkinson
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- School of Psychology, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Kerrie L Thomas
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
7
|
Du Y, Du Y, Zhang Y, Huang Z, Fu M, Li J, Pang Y, Lei P, Wang YT, Song W, He G, Dong Z. MKP-1 reduces Aβ generation and alleviates cognitive impairments in Alzheimer's disease models. Signal Transduct Target Ther 2019; 4:58. [PMID: 31840000 PMCID: PMC6895219 DOI: 10.1038/s41392-019-0091-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 12/23/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) phosphatase 1 (MKP-1) is an essential negative regulator of MAPKs by dephosphorylating MAPKs at both tyrosine and threonine residues. Dysregulation of the MAPK signaling pathway has been associated with Alzheimer's disease (AD). However, the role of MKP-1 in AD pathogenesis remains elusive. Here, we report that MKP-1 levels were decreased in the brain tissues of patients with AD and an AD mouse model. The reduction in MKP-1 gene expression appeared to be a result of transcriptional inhibition via transcription factor specificity protein 1 (Sp1) cis-acting binding elements in the MKP-1 gene promoter. Amyloid-β (Aβ)-induced Sp1 activation decreased MKP-1 expression. However, upregulation of MKP-1 inhibited the expression of both Aβ precursor protein (APP) and β-site APP-cleaving enzyme 1 by inactivating the extracellular signal-regulated kinase 1/2 (ERK)/MAPK signaling pathway. Furthermore, upregulation of MKP-1 reduced Aβ production and plaque formation and improved hippocampal long-term potentiation (LTP) and cognitive deficits in APP/PS1 transgenic mice. Our results demonstrate that MKP-1 impairment facilitates the pathogenesis of AD, whereas upregulation of MKP-1 plays a neuroprotective role to reduce Alzheimer-related phenotypes. Thus, this study suggests that MKP-1 is a novel molecule for AD treatment.
Collapse
Affiliation(s)
- Yehong Du
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Yexiang Du
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, 400016 PR China
| | - Yun Zhang
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Zhilin Huang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Min Fu
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Junjie Li
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Yayan Pang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| | - Peng Lei
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041 Sichuan China
| | - Yu Tian Wang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
- Brain Research Centre, The University of British Columbia, Vancouver, BC V6T 2B5 Canada
| | - Weihong Song
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Guiqiong He
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, 400016 PR China
| | - Zhifang Dong
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 PR China
| |
Collapse
|
8
|
Addy NA, Nunes EJ, Hughley SM, Small KM, Baracz SJ, Haight JL, Rajadhyaksha AM. The L-type calcium channel blocker, isradipine, attenuates cue-induced cocaine-seeking by enhancing dopaminergic activity in the ventral tegmental area to nucleus accumbens pathway. Neuropsychopharmacology 2018; 43:2361-2372. [PMID: 29773910 PMCID: PMC6180103 DOI: 10.1038/s41386-018-0080-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 12/27/2022]
Abstract
Previous preclinical and clinical investigations have focused on the L-type calcium channel (LTCC) as a potential therapeutic target for substance abuse. While some clinical studies have examined the ability of LTCC blockers to alter cocaine's subjective effects, very few LTCC studies have examined cocaine relapse. Here, we examined whether ventral tegmental area (VTA)-specific or systemic administration of the LTCC inhibitor, isradipine, altered cocaine-seeking behavior in a rat model. Male Sprague-Dawley rats first received 10 days of cocaine self-administration training (2 h sessions), where active lever depression resulted in delivery of a ∼0.5 mg/kg cocaine infusion paired with a tone + light cue. Rats then underwent 10 days of forced abstinence, without access to cocaine or cocaine cues. Rats were then returned to the opertant chamber for the cue-induced cocaine-seeking test, where active lever depression in the original training context resulted in tone + light cue presentation. We found VTA specific or systemic isradipine administration robustly attenuated cocaine-seeking, without altering cocaine-taking nor natural reward seeking. Dopamine (DA) signaling in the nucleus accumbens (NAc) core is necessary and sufficient for cue-induced drug-seeking. Surprisingly in our study, isradipine enhanced tonic and phasic DA signaling in cocaine abstinent rats, with no change in sucrose abstinent nor naïve rats. Strikingly, isradipine's behavioral effects were dependent upon NAc core DA receptor activation. Together, our findings reveal a novel mechanism by which the FDA-approved drug, isradipine, could act to decrease cocaine relapse.
Collapse
Affiliation(s)
- Nii A Addy
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA.
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, 06511, USA.
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, 06511, USA.
| | - Eric J Nunes
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Shannon M Hughley
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Keri M Small
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Sarah J Baracz
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Joshua L Haight
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Anjali M Rajadhyaksha
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA
| |
Collapse
|
9
|
Martínez-Rivera A, Hao J, Tropea TF, Giordano TP, Kosovsky M, Rice RC, Lee A, Huganir RL, Striessnig J, Addy NA, Han S, Rajadhyaksha AM. Enhancing VTA Ca v1.3 L-type Ca 2+ channel activity promotes cocaine and mood-related behaviors via overlapping AMPA receptor mechanisms in the nucleus accumbens. Mol Psychiatry 2017; 22:1735-1745. [PMID: 28194001 PMCID: PMC5555837 DOI: 10.1038/mp.2017.9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 11/30/2016] [Accepted: 12/23/2016] [Indexed: 02/07/2023]
Abstract
Genetic factors significantly influence susceptibility for substance abuse and mood disorders. Rodent studies have begun to elucidate a role of Cav1.3 L-type Ca2+ channels in neuropsychiatric-related behaviors, such as addictive and depressive-like behaviors. Human studies have also linked the CACNA1D gene, which codes for the Cav1.3 protein, with bipolar disorder. However, the neurocircuitry and the molecular mechanisms underlying the role of Cav1.3 in neuropsychiatric phenotypes are not well established. In the present study, we directly manipulated Cav1.3 channels in Cav1.2 dihydropyridine insensitive mutant mice and found that ventral tegmental area (VTA) Cav1.3 channels mediate cocaine-related and depressive-like behavior through a common nucleus accumbens (NAc) shell calcium-permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (CP-AMPAR) mechanism that requires GluA1 phosphorylation at S831. Selective activation of VTA Cav1.3 with (±)-BayK-8644 (BayK) enhanced cocaine conditioned place preference and cocaine psychomotor activity while inducing depressive-like behavior, an effect not observed in S831A phospho-mutant mice. Infusion of the CP-AMPAR-specific blocker Naspm into the NAc shell reversed the cocaine and depressive-like phenotypes. In addition, activation of VTA Cav1.3 channels resulted in social behavioral deficits. In contrast to the cocaine- and depression-related phenotypes, GluA1/A2 AMPARs in the NAc core mediated social deficits, independent of S831-GluA1 phosphorylation. Using a candidate gene analysis approach, we also identified single-nucleotide polymorphisms in the CACNA1D gene associated with cocaine dependence in human subjects. Together, our findings reveal novel, overlapping mechanisms through which VTA Cav1.3 mediates cocaine-related, depressive-like and social phenotypes, suggesting that Cav1.3 may serve as a target for the treatment of neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Arlene Martínez-Rivera
- Dept. of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Jin Hao
- Dept. of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Thomas F. Tropea
- Dept. of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Thomas P. Giordano
- Dept. of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Maria Kosovsky
- Dept. of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Richard C. Rice
- Dept. of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Richard L. Huganir
- Department of Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joerg Striessnig
- Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Austria; Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Nii A. Addy
- Department of Psychiatry and Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale Graduate School of Arts and Science, New Haven, CT, USA
| | - Shizhong Han
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Anjali M. Rajadhyaksha
- Dept. of Pediatrics, Division of Pediatric Neurology, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
10
|
Kabir ZD, Martínez-Rivera A, Rajadhyaksha AM. From Gene to Behavior: L-Type Calcium Channel Mechanisms Underlying Neuropsychiatric Symptoms. Neurotherapeutics 2017; 14:588-613. [PMID: 28497380 PMCID: PMC5509628 DOI: 10.1007/s13311-017-0532-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The L-type calcium channels (LTCCs) Cav1.2 and Cav1.3, encoded by the CACNA1C and CACNA1D genes, respectively, are important regulators of calcium influx into cells and are critical for normal brain development and plasticity. In humans, CACNA1C has emerged as one of the most widely reproduced and prominent candidate risk genes for a range of neuropsychiatric disorders, including bipolar disorder (BD), schizophrenia (SCZ), major depressive disorder, autism spectrum disorder, and attention deficit hyperactivity disorder. Separately, CACNA1D has been found to be associated with BD and autism spectrum disorder, as well as cocaine dependence, a comorbid feature associated with psychiatric disorders. Despite growing evidence of a significant link between CACNA1C and CACNA1D and psychiatric disorders, our understanding of the biological mechanisms by which these LTCCs mediate neuropsychiatric-associated endophenotypes, many of which are shared across the different disorders, remains rudimentary. Clinical studies with LTCC blockers testing their efficacy to alleviate symptoms associated with BD, SCZ, and drug dependence have provided mixed results, underscoring the importance of further exploring the neurobiological consequences of dysregulated Cav1.2 and Cav1.3. Here, we provide a review of clinical studies that have evaluated LTCC blockers for BD, SCZ, and drug dependence-associated symptoms, as well as rodent studies that have identified Cav1.2- and Cav1.3-specific molecular and cellular cascades that underlie mood (anxiety, depression), social behavior, cognition, and addiction.
Collapse
Affiliation(s)
- Zeeba D Kabir
- Pediatric Neurology, Pediatrics, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, NY, USA
| | - Arlene Martínez-Rivera
- Pediatric Neurology, Pediatrics, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, NY, USA
| | - Anjali M Rajadhyaksha
- Pediatric Neurology, Pediatrics, Weill Cornell Medicine, New York, NY, USA.
- Weill Cornell Autism Research Program, Weill Cornell Medicine, New York, NY, USA.
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Mohamed RMP, Kumar J, Yap E, Mohamed IN, Sidi H, Adam RL, Das S. Try to Remember: Interplay between Memory and Substance Use Disorder. Curr Drug Targets 2017. [PMID: 28641520 DOI: 10.2174/1389450118666170622092824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Memories associated with substance use disorders, or substance-associated cues increase the likelihood of craving and relapse during abstinence. There is a growing consensus that manipulation of synaptic plasticity may reduce the strength of substance abuse-related memories. On the biological front, there are new insights that suggest memories associated with substance use disorder may follow unique neurobiological pathways that render them more accessible to pharmacological intervention. In parallel to this, research in neurochemistry has identified several potential candidate molecules that could influence the formation and maintenance of long-term memory. Drugs that target these molecules (blebbistatin, isradipine and zeta inhibitory peptide) have shown promise at the preclinical stage. In this review, we shall provide an overview of the evolving understanding on the biochemical mechanisms involved in memory formation and expound on the premise that substance use disorder is a learning disorder.
Collapse
Affiliation(s)
- Rashidi Mohamed Pakri Mohamed
- Department of Psychological Medicine, Faculty of Medicine, University of Malaya, Lembah Pantai, 59100 Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Ernie Yap
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Hatta Sidi
- Department of Psychiatry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Raja Lope Adam
- Department of Psychiatry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Srijit Das
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Terrillion CE, Dao DT, Cachope R, Lobo MK, Puche AC, Cheer JF, Gould TD. Reduced levels of Cacna1c attenuate mesolimbic dopamine system function. GENES, BRAIN, AND BEHAVIOR 2017; 16:495-505. [PMID: 28186690 PMCID: PMC5457318 DOI: 10.1111/gbb.12371] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/22/2017] [Accepted: 02/02/2017] [Indexed: 12/25/2022]
Abstract
Genetic variation in CACNA1C, which codes for the L-type calcium channel (LTCC) Cav 1.2, is associated with clinical diagnoses of bipolar disorder, depression and schizophrenia. Dysregulation of the mesolimbic-dopamine (ML-DA) system is linked to these syndromes and LTCCs are required for normal DAergic neurotransmission between the ventral tegmental area (VTA) and nucleus accumbens (NAc). It is unclear, however, how variations in CACNA1C genotype, and potential subsequent changes in expression levels in these regions, modify risk. Using constitutive and conditional knockout mice, and treatment with the LTCC antagonist nimodipine, we examined the role of Cacna1c in DA-mediated behaviors elicited by psychomotor stimulants. Using fast-scan cyclic voltammetry, DA release and reuptake in the NAc were measured. We find that subsecond DA release in Cacna1c haploinsufficient mice lacks normal sensitivity to inhibition of the DA transporter (DAT). Constitutive haploinsufficiency of Cacna1c led to attenuation of hyperlocomotion following acute administration of stimulants specific to DAT, and locomotor sensitization of these mice to the DAT antagonist GBR12909 did not reach the same level as wild-type mice. The maintenance of sensitization to GBR12909 was attenuated by administration of nimodipine. Sensitization to GBR12909 was attenuated in mice with reduced Cacna1c selectively in the VTA but not in the NAc. Our findings show that Cacna1c is crucial for normal behavioral responses to DA stimulants and that its activity in the VTA is required for behavioral sensitization. Cacna1c likely exerts these effects through modifications to presynaptic ML-DA system function.
Collapse
Affiliation(s)
- Chantelle E. Terrillion
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David T. Dao
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Roger Cachope
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mary Kay Lobo
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Adam C. Puche
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Joseph F. Cheer
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Todd D. Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Liu Y, Harding M, Dore J, Chen X. Ca v1.2, but not Ca v1.3, L-type calcium channel subtype mediates nicotine-induced conditioned place preference in mice. Prog Neuropsychopharmacol Biol Psychiatry 2017; 75:176-182. [PMID: 28185965 DOI: 10.1016/j.pnpbp.2017.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/03/2017] [Indexed: 11/30/2022]
Abstract
Nicotine use is one of the most common forms of drug addiction. Although L-type calcium channels (LTCCs) are involved in nicotine addiction, the contribution of the two primary LTCC subtypes (Cav1.2 and 1.3) is unknown. This study aims to determine the contribution of these two LTCC subtypes to nicotine-induced conditioned place preference (CPP) responses by using transgenic mouse models that do not express Cav1.3 (Cav1.3-/-) or contain a mutation in the dihydropyridine (DHP) site of the Cav1.2 (Cav1.2DHP-/-). We found a hyperbolic dose dependent nicotine (0.1-1mg/kg; 0.5mg/kg optimum) effect on place preference in wild type (WT) mice, that could be prevented by the DHP LTCC blocker nifedipine pretreatment. Similarly, Cav1.3-/- mice showed nicotine-induced place preference which was antagonized by nifedipine. In contrast, nifedipine pretreatment of Cav1.2DHP-/- mice had no effect on nicotine-induced CPP responses, suggesting an involvement of Cav1.2 subtype in the nicotine-induced CPP response. Nifedipine alone failed to produce either conditioned place aversion or CPP in WT mice. These results collectively indicate Cav1.2, but not Cav1.3 LTCC subtype regulates, at least in part, the reinforcing effects of nicotine use.
Collapse
Affiliation(s)
- Yudan Liu
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.
| | - Meghan Harding
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jules Dore
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Xihua Chen
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada.
| |
Collapse
|
14
|
Kabir ZD, Lee AS, Rajadhyaksha AM. L-type Ca 2+ channels in mood, cognition and addiction: integrating human and rodent studies with a focus on behavioural endophenotypes. J Physiol 2016; 594:5823-5837. [PMID: 26913808 PMCID: PMC5063939 DOI: 10.1113/jp270673] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/28/2015] [Indexed: 01/07/2023] Open
Abstract
Brain Cav 1.2 and Cav 1.3 L-type Ca2+ channels play key physiological roles in various neuronal processes that contribute to brain function. Genetic studies have recently identified CACNA1C as a candidate risk gene for bipolar disorder (BD), schizophrenia (SCZ), major depressive disorder (MDD) and autism spectrum disorder (ASD), and CACNA1D for BD and ASD, suggesting a contribution of Cav 1.2 and Cav 1.3 Ca2+ signalling to the pathophysiology of neuropsychiatric disorders. Once considered sole clinical entities, it is now clear that BD, SCZ, MDD and ASD share common phenotypic features, most likely due to overlapping neurocircuitry and common molecular mechanisms. A major future challenge lies in translating the human genetic findings to pathological mechanisms that are translatable back to the patient. One approach for tackling such a daunting scientific endeavour for complex behaviour-based neuropsychiatric disorders is to examine intermediate biological phenotypes in the context of endophenotypes within distinct behavioural domains. This will better allow us to integrate findings from genes to behaviour across species, and improve the chances of translating preclinical findings to clinical practice.
Collapse
Affiliation(s)
- Z D Kabir
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
- Weill Cornell Autism Research Program, Weill Cornell Medical College, New York, NY, USA
| | - A S Lee
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
- Weill Cornell Autism Research Program, Weill Cornell Medical College, New York, NY, USA
| | - A M Rajadhyaksha
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA.
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA.
- Weill Cornell Autism Research Program, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
15
|
Degoulet M, Stelly CE, Ahn KC, Morikawa H. L-type Ca²⁺ channel blockade with antihypertensive medication disrupts VTA synaptic plasticity and drug-associated contextual memory. Mol Psychiatry 2016; 21:394-402. [PMID: 26100537 PMCID: PMC4689680 DOI: 10.1038/mp.2015.84] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 04/14/2015] [Accepted: 04/30/2015] [Indexed: 02/08/2023]
Abstract
Drug addiction is driven, in part, by powerful and enduring memories of sensory cues associated with drug intake. As such, relapse to drug use during abstinence is frequently triggered by an encounter with drug-associated cues, including the drug itself. L-type Ca(2+) channels (LTCCs) are known to regulate different forms of synaptic plasticity, the major neural substrate for learning and memory, in various brain areas. Long-term potentiation (LTP) of NMDA receptor (NMDAR)-mediated glutamatergic transmission in the ventral tegmental area (VTA) may contribute to the increased motivational valence of drug-associated cues triggering relapse. In this study, using rat brain slices, we found that isradipine, a general LTCC antagonist used as antihypertensive medication, not only blocks the induction of NMDAR LTP but also promotes the reversal of previously induced LTP in the VTA. In behaving rats, isradipine injected into the VTA suppressed the acquisition of cocaine-paired contextual cue memory assessed using a conditioned place preference (CPP) paradigm. Furthermore, administration of isradipine or a CaV1.3 subtype-selective LTCC antagonist (systemic or intra-VTA) before a single extinction or reinstatement session, while having no immediate effect at the time of administration, abolished previously acquired cocaine and alcohol (ethanol) CPP on subsequent days. Notably, CPP thus extinguished cannot be reinstated by drug re-exposure, even after 2 weeks of withdrawal. These results suggest that LTCC blockade during exposure to drug-associated cues may cause unlearning of the increased valence of those cues, presumably via reversal of glutamatergic synaptic plasticity in the VTA.
Collapse
Affiliation(s)
- Mickael Degoulet
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Claire E. Stelly
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Kee-Chan Ahn
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA
| | - Hitoshi Morikawa
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, USA,Corresponding author: Hitoshi Morikawa, Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, 2400 Speedway, Austin, TX 78712, USA., Tel: 1-512-232-9299, Fax: 1-512-471-3878,
| |
Collapse
|
16
|
Cheng W, Zhu Y, Wang H. The MAPK pathway is involved in the regulation of rapid pacing-induced ionic channel remodeling in rat atrial myocytes. Mol Med Rep 2016; 13:2677-82. [PMID: 26847818 DOI: 10.3892/mmr.2016.4862] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 01/11/2016] [Indexed: 11/06/2022] Open
Abstract
Alterations to the expression L‑type calcium channels (LTCCs) and Kv4.3 potassium channels form the possible basis of atrial electrical remodeling during rapid pacing. The mitogen‑activated protein kinase (MAPK) pathway is affected by increases in cytoplasmic Ca2+, and therefore represents an attractive candidate for the regulation and mediation of Ca2+‑induced ion channel remodeling. The present study aimed to investigate alterations to the ion channel‑MAPK axis, and to determine its influence on ion channel remodeling during atrial fibrillation. Rat atrial myocytes were isolated, cultured, and in vitro rapid pacing was established. Intracellular Ca2+ signals were monitored using the Fluo‑3/AM Ca2+ indicator. Verapamil, PD98058 and SB203580 were added to the culture medium of various groups at specific time‑points. The mRNA expression levels of LTCC‑α1c and Kv4.3 potassium channels were detected by reverse transcription‑polymerase chain reaction. Western blotting was performed to determine the expression levels of channel and signaling proteins. The results demonstrated that fast pacing significantly increased the intracellular Ca2+ concentration in atrial myocytes, whereas treatment with verapamil markedly inhibited this increase. In addition, verapamil significantly antagonized the rapid pacing‑induced activation of extracellular signal‑regulated kinase (ERK) and p38MAPK. These results indicated that the MAPK pathway may have an important role in the opening of LTCCs, and alterations to MAPK molecule expression could affect the expression and remodeling of ion channels.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yun Zhu
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Haidong Wang
- Department of Cardiothoracic Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
17
|
Cunningham JJ, Orr E, Lothian BC, Morgen J, Brebner K. Effects of fendiline on cocaine-seeking behavior in the rat. Psychopharmacology (Berl) 2015; 232:4401-10. [PMID: 26345344 DOI: 10.1007/s00213-015-4061-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 08/27/2015] [Indexed: 11/26/2022]
Abstract
RATIONALE L-type Ca(2+) channels (LTCC) and GABAB receptors are both possible targets in the development of new pharmacological compounds for cocaine addiction. Drugs that target either receptor attenuate a wide range of cocaine-seeking behaviors in the rat. However, there is no current human-approved pharmacotherapeutic intervention for psychostimulant addiction. OBJECTIVES This study examined the effects of a human-approved LTCC blocker, fendiline, on cocaine-taking and cocaine-seeking behavior in rats. The effects of combining fendiline with the GABAB receptor agonist baclofen on cocaine self-administration were also tested. METHODS Male Wistar rats were trained to self-administer cocaine, and the effects of fendiline pretreatment (vehicle, 1.78, 3.16, 5.62 mg/kg, intraperitoneal (IP)) were tested on progressive ratio responding and cue- and drug-induced reinstatement. The effects of baclofen (vehicle, 0.56, 1.78, 3.16, 5.62 mg/kg, IP) combined with fendiline (5.62 mg/kg, IP) were tested on progressive ratio responding. Control experiments measured locomotor activity and lever pressing for food in rats that received both baclofen and fendiline prior to the test session. RESULTS Acute injections of fendiline prior to cue- or drug-induced reinstatement significantly attenuated lever-pressing behavior (p < 0.05). Fendiline and baclofen, but not fendiline alone, not only significantly attenuated breakpoints, but also impaired general motor behavior and naturalistic reinforcement (p < 0.05). CONCLUSION These data suggest that the LTCC blocker fendiline may represent a novel pharmacotherapeutic intervention to prevent reinstatement to cocaine seeking. Also, co-administration of fendiline and baclofen not only can attenuate the motivation to take cocaine, but also impairs general motor behavior and naturalistic reinforcement.
Collapse
Affiliation(s)
- Jonathan J Cunningham
- Department of Psychiatry, University of British Columbia, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, British Columbia, V6T 2A1, Canada.
| | - Erin Orr
- Department of Psychology, St. Francis Xavier University, 2320 Notre Dame Blvd., Antigonish, Nova Scotia, B2G 2W5, Canada
| | - Barbara C Lothian
- Department of Psychology, St. Francis Xavier University, 2320 Notre Dame Blvd., Antigonish, Nova Scotia, B2G 2W5, Canada
| | - Jennifer Morgen
- Department of Psychology, St. Francis Xavier University, 2320 Notre Dame Blvd., Antigonish, Nova Scotia, B2G 2W5, Canada
| | - Karen Brebner
- Department of Psychology, St. Francis Xavier University, 2320 Notre Dame Blvd., Antigonish, Nova Scotia, B2G 2W5, Canada.
| |
Collapse
|
18
|
Sun WL, Quizon PM, Zhu J. Molecular Mechanism: ERK Signaling, Drug Addiction, and Behavioral Effects. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 137:1-40. [PMID: 26809997 DOI: 10.1016/bs.pmbts.2015.10.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Addiction to psychostimulants has been considered as a chronic psychiatric disorder characterized by craving and compulsive drug seeking and use. Over the past two decades, accumulating evidence has demonstrated that repeated drug exposure causes long-lasting neurochemical and cellular changes that result in enduring neuroadaptation in brain circuitry and underlie compulsive drug consumption and relapse. Through intercellular signaling cascades, drugs of abuse induce remodeling in the rewarding circuitry that contributes to the neuroplasticity of learning and memory associated with addiction. Here, we review the role of the extracellular signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase, and its related intracellular signaling pathways in drug-induced neuroadaptive changes that are associated with drug-mediated psychomotor activity, rewarding properties and relapse of drug seeking behaviors. We also discuss the neurobiological and behavioral effects of pharmacological and genetic interferences with ERK-associated molecular cascades in response to abused substances. Understanding the dynamic modulation of ERK signaling in response to drugs may provide novel molecular targets for therapeutic strategies to drug addiction.
Collapse
Affiliation(s)
- Wei-Lun Sun
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Pamela M Quizon
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA.
| |
Collapse
|
19
|
Zamponi GW. Targeting voltage-gated calcium channels in neurological and psychiatric diseases. Nat Rev Drug Discov 2015; 15:19-34. [DOI: 10.1038/nrd.2015.5] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
20
|
Yap JJ, Chartoff EH, Holly EN, Potter DN, Carlezon WA, Miczek KA. Social defeat stress-induced sensitization and escalated cocaine self-administration: the role of ERK signaling in the rat ventral tegmental area. Psychopharmacology (Berl) 2015; 232:1555-69. [PMID: 25373870 PMCID: PMC4397167 DOI: 10.1007/s00213-014-3796-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 10/21/2014] [Indexed: 01/20/2023]
Abstract
RATIONALE Intermittent social defeat stress can induce neuroadaptations that promote compulsive drug taking. Within the mesocorticolimbic circuit, repeated cocaine administration activates extracellular signal-regulated kinase (ERK). OBJECTIVE The present experiments examine whether changes in ERK phosphorylation are necessary for the behavioral and neural adaptations that occur as a consequence of intermittent defeat stress. MATERIALS AND METHODS Rats were exposed to four brief intermittent defeats over the course of 10 days. Ten days after the last defeat, rats were challenged with cocaine (10 mg/kg, i.p.) or saline, and ERK activity was examined in mesocorticolimbic regions. To determine the role of ERK in defeat stress-induced behavioral sensitization, we bilaterally microinjected the MAPK/ERK kinase inhibitor U0126 (1 μg/side) or vehicle (20 % DMSO) into the ventral tegmental area (VTA) prior to each of four defeats. Ten days following the last defeat, locomotor activity was assessed for the expression of behavioral cross-sensitization to cocaine (10 mg/kg, i.p.). Thereafter, rats self-administered cocaine under fixed and progressive ratio schedules of reinforcement, including a 24-h continuous access "binge" (0.3 mg/kg/infusion). RESULTS We found that repeated defeat stress increased ERK phosphorylation in the VTA. Inhibition of VTA ERK prior to each social defeat attenuated the development of stress-induced sensitization and prevented stress-induced enhancement of cocaine self-administration during a continuous access binge. CONCLUSIONS These results suggest that enhanced activation of ERK in the VTA due to brief defeats is critical in the induction of sensitization and escalated cocaine taking.
Collapse
Affiliation(s)
- Jasmine J Yap
- Department of Psychology, Tufts University, Medford, MA, 02155, USA,
| | | | | | | | | | | |
Collapse
|
21
|
Andres MA, Cooke IM, Bellinger FP, Berry MJ, Zaporteza MM, Rueli RH, Barayuga SM, Chang L. Methamphetamine acutely inhibits voltage-gated calcium channels but chronically up-regulates L-type channels. J Neurochem 2015; 134:56-65. [PMID: 25807982 DOI: 10.1111/jnc.13104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 03/04/2015] [Accepted: 03/06/2015] [Indexed: 11/30/2022]
Abstract
In neurons, calcium (Ca(2+) ) channels regulate a wide variety of functions ranging from synaptic transmission to gene expression. They also induce neuroplastic changes that alter gene expression following psychostimulant administration. Ca(2+) channel blockers have been considered as potential therapeutic agents for the treatment of methamphetamine (METH) dependence because of their ability to reduce drug craving among METH users. Here, we studied the effects of METH exposure on voltage-gated Ca(2+) channels using SH-SY5Y cells as a model of dopaminergic neurons. We found that METH has different short- and long-term effects. A short-term effect involves immediate (< 5 min) direct inhibition of Ca(2+) ion movements through Ca(2+) channels. Longer exposure to METH (20 min or 48 h) selectively up-regulates the expression of only the CACNA1C gene, thus increasing the number of L-type Ca(2+) channels. This up-regulation of CACNA1C is associated with the expression of the cAMP-responsive element-binding protein (CREB), a known regulator of CACNA1C gene expression, and the MYC gene, which encodes a transcription factor that putatively binds to a site proximal to the CACNA1C gene transcription initiation site. The short-term inhibition of Ca(2+) ion movement and later, the up-regulation of Ca(2+) channel gene expression together suggest the operation of cAMP-responsive element-binding protein- and C-MYC-mediated mechanisms to compensate for Ca(2+) channel inhibition by METH. Increased Ca(2+) current density and subsequent increased intracellular Ca(2+) may contribute to the neurodegeneration accompanying chronic METH abuse. Methamphetamine (METH) exposure has both short- and long-term effects. Acutely, methamphetamine directly inhibits voltage-gated calcium channels. Chronically, neurons compensate by up-regulating the L-type Ca(2+) channel gene, CACNA1C. This compensatory mechanism is mediated by transcription factors C-MYC and CREB, in which CREB is linked to the dopamine D1 receptor signaling pathway. These findings suggest Ca(2+) -mediated neurotoxicity owing to over-expression of calcium channels.
Collapse
Affiliation(s)
- Marilou A Andres
- Bekesy Laboratory of Neurobiology, Pacific Biosciences Research Center, University of Hawaii, Honolulu, Hawaii, USA
| | - Ian M Cooke
- Bekesy Laboratory of Neurobiology, Pacific Biosciences Research Center, University of Hawaii, Honolulu, Hawaii, USA.,Department of Biology, University of Hawaii, Honolulu, Hawaii, USA
| | - Frederick P Bellinger
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Marla J Berry
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Maribel M Zaporteza
- Bekesy Laboratory of Neurobiology, Pacific Biosciences Research Center, University of Hawaii, Honolulu, Hawaii, USA
| | - Rachel H Rueli
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Stephanie M Barayuga
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Linda Chang
- Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
22
|
CACNA1C risk variant affects reward responsiveness in healthy individuals. Transl Psychiatry 2014; 4:e461. [PMID: 25290268 PMCID: PMC4350510 DOI: 10.1038/tp.2014.100] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/01/2014] [Accepted: 08/21/2014] [Indexed: 12/21/2022] Open
Abstract
The variant at rs1006737 in the L-type voltage-gated calcium channel (alpha 1c subunit) CACNA1C gene is reliably associated with both bipolar disorder and schizophrenia. We investigated whether this risk variant affects reward responsiveness because reward processing is one of the central cognitive-motivational domains implicated in both disorders. In a sample of 164 young, healthy individuals, we show a dose-dependent response, where the rs1006737 risk genotype was associated with blunted reward responsiveness, whereas discriminability did not significantly differ between genotype groups. This finding suggests that the CACNA1C risk locus may have a role in neural pathways that facilitate value representation for rewarding stimuli. Impaired reward processing may be a transdiagnostic phenotype of variation in CACNA1C that could contribute to anhedonia and other clinical features common to both affective and psychotic disorders.
Collapse
|
23
|
The role of L-type voltage-gated calcium channels Cav1.2 and Cav1.3 in normal and pathological brain function. Cell Tissue Res 2014; 357:463-76. [DOI: 10.1007/s00441-014-1936-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
|
24
|
Liu Y, Harding M, Pittman A, Dore J, Striessnig J, Rajadhyaksha A, Chen X. Cav1.2 and Cav1.3 L-type calcium channels regulate dopaminergic firing activity in the mouse ventral tegmental area. J Neurophysiol 2014; 112:1119-30. [PMID: 24848473 DOI: 10.1152/jn.00757.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dopaminergic projections from the ventral tegmental area (VTA) constitute the mesolimbocortical system that underlies addiction and psychosis primarily as a result of increased dopaminergic transmission. Dopamine release is spike dependent. L-type calcium channels (LTCCs) play an important role in regulating firing activities, but the contribution of specific subtypes remains unclear. This article describes different functions of Cav1.2 and Cav1.3 subtypes in regulating firing properties with two transgenic mouse strains. For basal firing, Cav1.3-deficient (Cav1.3(-/-)) mice had a lower basal firing frequency. The dihydropyridine (DHP) channel blocker nifedipine reduced single-spike firing in mice expressing DHP-insensitive Cav1.2 channels (Cav1.2DHP(-/-) mice), confirming the significant contribution from the Cav1.3 subtype in basal firing. Moreover, the DHP channel activator (S)-(-)-Bay K8644 and the non-DHP channel activator FPL 64176 converted firing patterns from single spiking to bursting in Cav1.2DHP(-/-) mice. Nifedipine inhibited burst firing induced by both activators, suggesting that Cav1.3 also serves an essential role in burst firing. However, FPL 64176 also induced bursting in Cav1.3(-/-) mice. These results indicate that the Cav1.3 subtype is crucial to regulation of basal single-spike firing, while activation of both Cav1.2 and Cav1.3 can support burst firing of VTA neurons.
Collapse
Affiliation(s)
- Yudan Liu
- Department of Physiology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning, China;
| | - Meghan Harding
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Andrea Pittman
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Jules Dore
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria; and
| | - Anjali Rajadhyaksha
- Division of Pediatric Neurology, Department of Pediatrics and Brain and Mind Research Institute, Weill Cornell Medical College of Cornell University, New York
| | - Xihua Chen
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
25
|
Mitogen-activated protein kinase phosphatase (MKP)-1 as a neuroprotective agent: promotion of the morphological development of midbrain dopaminergic neurons. Neuromolecular Med 2013; 15:435-46. [PMID: 23584919 DOI: 10.1007/s12017-013-8230-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 04/05/2013] [Indexed: 12/24/2022]
Abstract
A greater understanding of the mechanisms that promote the survival and growth of dopaminergic neurons is essential for the advancement of cell replacement therapies for Parkinson's disease (PD). Evidence supports a role for the mitogen-activated protein kinase p38 in the demise of dopaminergic neurons, while mitogen-activated protein kinase phosphatase-1 (MKP-1), which negatively regulates p38 activity, has not yet been investigated in this context. Here, we show that MKP-1 is expressed in dopaminergic neurons cultured from E14 rat ventral mesencephalon (VM). When dopaminergic neurons were transfected to overexpress MKP-1, they displayed a more complex morphology than their control counterparts in vitro. Specifically, MKP-1-transfection induced significant increases in neurite length and branching with a maximum increase observed in primary branches. We demonstrate that inhibition of dopaminergic neurite growth induced by treatment of rat VM neurons with the dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA) in vitro is mediated by p38 and is concomitant with a significant and selective decrease in MKP-1 expression in these neurons. We further show that overexpression of MKP-1 in dopaminergic neurons contributes to neuroprotection against the effects of 6-OHDA. Collectively, we report that MKP-1 can promote the growth and elaboration of dopaminergic neuronal processes and can help protect them from the neurotoxic effects of 6-OHDA. Thus, we propose that strategies aimed at augmenting MKP-1 expression or activity may be beneficial in protecting dopaminergic neurons and may provide potential therapeutic approaches for PD.
Collapse
|
26
|
Bhat S, Dao DT, Terrillion CE, Arad M, Smith RJ, Soldatov NM, Gould TD. CACNA1C (Cav1.2) in the pathophysiology of psychiatric disease. Prog Neurobiol 2012; 99:1-14. [PMID: 22705413 PMCID: PMC3459072 DOI: 10.1016/j.pneurobio.2012.06.001] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 05/27/2012] [Accepted: 06/06/2012] [Indexed: 10/28/2022]
Abstract
One of the most consistent genetic findings to have emerged from bipolar disorder genome wide association studies (GWAS) is with CACNA1C, a gene that codes for the α(1C) subunit of the Ca(v)1.2 voltage-dependent L-type calcium channel (LTCC). Genetic variation in CACNA1C have also been associated with depression, schizophrenia, autism spectrum disorders, as well as changes in brain function and structure in control subjects who have no diagnosable psychiatric illness. These data are consistent with a continuum of shared neurobiological vulnerability between diverse-Diagnostic and Statistical Manual (DSM) defined-neuropsychiatric diseases. While involved in numerous cellular functions, Ca(v)1.2 is most frequently implicated in coupling of cell membrane depolarization to transient increase of the membrane permeability for calcium, leading to activation and, potentially, changes in intracellular signaling pathway activity, gene transcription, and synaptic plasticity. Ca(v)1.2 is involved in the proper function of numerous neurological circuits including those involving the hippocampus, amygdala, and mesolimbic reward system, which are strongly implicated in psychiatric disease pathophysiology. A number of behavioral effects of LTCC inhibitors have been described including antidepressant-like behavioral actions in rodent models. Clinical studies suggest possible treatment effects in a subset of patients with mood disorders. We review the genetic structure and variation of CACNA1C, discussing relevant human genetic and clinical findings, as well as the biological actions of Ca(v)1.2 that are most relevant to psychiatric illness.
Collapse
Affiliation(s)
- Shambhu Bhat
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - David T. Dao
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - Michal Arad
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Robert J. Smith
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - Todd D. Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
- Program in Neuroscience, University of Maryland, Baltimore, MD, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
27
|
Horio M, Kohno M, Fujita Y, Ishima T, Inoue R, Mori H, Hashimoto K. Role of serine racemase in behavioral sensitization in mice after repeated administration of methamphetamine. PLoS One 2012; 7:e35494. [PMID: 22530033 PMCID: PMC3329469 DOI: 10.1371/journal.pone.0035494] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 03/20/2012] [Indexed: 02/07/2023] Open
Abstract
Background The N-methyl-D-aspartate (NMDA) receptors play a role in behavioral abnormalities observed after administration of the psychostimulant, methamphetamine (METH). Serine racemase (SRR) is an enzyme which synthesizes D-serine, an endogenous co-agonist of NMDA receptors. Using Srr knock-out (KO) mice, we investigated the role of SRR on METH-induced behavioral abnormalities in mice. Methodology/Principal Findings Evaluations of behavior in acute hyperlocomotion, behavioral sensitization, and conditioned place preference (CPP) were performed. The role of SRR on the release of dopamine (DA) in the nucleus accumbens after administration of METH was examined using in vivo microdialysis technique. Additionally, phosphorylation levels of ERK1/2 proteins in the striatum, frontal cortex and hippocampus were examined using Western blot analysis. Acute hyperlocomotion after a single administration of METH (3 mg/kg) was comparable between wild-type (WT) and Srr-KO mice. However, repeated administration of METH (3 mg/kg/day, once daily for 5 days) resulted in behavioral sensitization in WT, but not Srr-KO mice. Pretreatment with D-serine (900 mg/kg, 30 min prior to each METH treatment) did not affect the development of behavioral sensitization after repeated METH administration. In the CPP paradigm, METH-induced rewarding effects were demonstrable in both WT and Srr-KO mice. In vivo microdialysis study showed that METH (1 mg/kg)-induced DA release in the nucleus accumbens of Srr-KO mice previously treated with METH was significantly lower than that of the WT mice previously treated with METH. Interestingly, a single administration of METH (3 mg/kg) significantly increased the phosphorylation status of ERK1/2 in the striatum of WT, but not Srr-KO mice. Conclusions/Significance These findings suggest first, that SRR plays a role in the development of behavioral sensitization in mice after repeated administration of METH, and second that phosphorylation of ERK1/2 by METH may contribute to the development of this sensitization as seen in WT but not Srr-KO mice.
Collapse
Affiliation(s)
- Mao Horio
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Mami Kohno
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Yuko Fujita
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Tamaki Ishima
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Ran Inoue
- Department of Molecular Neuroscience, Toyama University Graduate School of Medicine, Toyama, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Toyama University Graduate School of Medicine, Toyama, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
- * E-mail:
| |
Collapse
|
28
|
Role of calcineurin in the VTA in rats behaviorally sensitized to methamphetamine. Psychopharmacology (Berl) 2012; 220:117-28. [PMID: 21901318 DOI: 10.1007/s00213-011-2461-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2011] [Accepted: 08/17/2011] [Indexed: 10/17/2022]
Abstract
RATIONALE Chronic psychostimulant administration increases locomotor activity, which is referred to as locomotor sensitization. Calcineurin has been suggested to participate in psychostimulant-induced sensitization, but the underlying neurobiological mechanism is poorly understood. OBJECTIVES This study was designed to examine whether calcineurin activity and its substrates participate in methamphetamine (METH)-induced locomotor sensitization in rats. MATERIALS AND METHODS Two weeks daily METH (1 mg/kg, i.p.) was administrated to rats to induce locomotor sensitization, activity of calcineurin and its substrates Synapsin and glycogen synthase kinase-3β (GSK-3β) were detected. The initiation and expression of locomotor sensitization were tested by inhibition of calcineurin activity systematically or locally in the ventral tegmental area (VTA). RESULTS Expression of the calcineurin A subunit (catalytic subunit) increased in the VTA but not prefrontal cortex, nucleus accumbens, or hippocampus in rats sensitized to METH. The calcineurin inhibitor cyclosporine A, systemically administered or microinfused into the VTA, suppressed the initiation but not expression of METH-induced locomotor sensitization. Chronic METH exposure upregulated the expression of the calcineurin A subunit in the VTA, which was negatively associated with downregulation of the phosphorylation of Synapsin and GSK-3β. Moreover, the related molecular changes were blocked by systemically administered cyclosporine A or microinjections into the VTA. CONCLUSIONS These data elucidate the critical role of calcineurin in the neurobiological mechanism underlying METH-induced locomotor sensitization, suggesting that calcineurin might participate in the initiation of METH-induced locomotor sensitization by negatively regulating the activity of Synapsin and GSK-3β in the VTA.
Collapse
|
29
|
Cav1.2 L-type Ca²⁺ channels mediate cocaine-induced GluA1 trafficking in the nucleus accumbens, a long-term adaptation dependent on ventral tegmental area Ca(v)1.3 channels. J Neurosci 2011; 31:13562-75. [PMID: 21940447 DOI: 10.1523/jneurosci.2315-11.2011] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AMPA receptor (AMPAR) plasticity at glutamatergic synapses in the mesoaccumbal dopaminergic pathway has been implicated in persistent cocaine-induced behavioral responses; however, the precise mechanism underlying these changes remains unknown. Utilizing cocaine psychomotor sensitization, we have examined phosphorylation of GluA1 at key residues serine 845 (S845) and S831, as well as GluA1 cell surface levels in the nucleus accumbens (NAc) of cocaine-preexposed mice and the role of brain-specific Ca(v)1.2 and Ca(v)1.3 L-type Ca²⁺ channels (LTCCs), therein. We found higher basal levels of S845 phospho-GluA1 (P-GluA1) and cell surface GluA1 in the NAc following protracted withdrawal from cocaine exposure, changes that occur independently of LTCCs. In contrast, we found that a cocaine challenge that elicits expression of the cocaine-sensitized response increases S831 P-GluA1 that further increases surface GluA1 beyond the higher basal levels. Intra-NAc pharmacological manipulations indicate that the Ca(v)1.2-activated CaM kinase II (CaMKII) mediates cocaine-induced increase in S831 P-GluA1 and that both Ca(v)1.2-activated CaMKII and extracellular signal-regulated kinase 2 (ERK2) mediate the increase in GluA1 cell surface levels specific to the sensitized response. Experiments using adenoassociated viral vectors expressing Ca(v)1.3 and ERK2 siRNA further indicate that recruitment of the Ca(v)1.2 pathway in the NAc is dependent on ventral tegmental area Ca(v)1.3 LTCCs and ERK2. Together, these results identify candidate pathways that mediate cocaine-induced AMPAR plasticity in the NAc and provide a mechanism linking LTCCs and GluA1 plasticity to cocaine-induced persistent behavioral changes.
Collapse
|
30
|
Previous ethanol experience enhances synaptic plasticity of NMDA receptors in the ventral tegmental area. J Neurosci 2011; 31:5205-12. [PMID: 21471355 DOI: 10.1523/jneurosci.5282-10.2011] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Alcohol addiction (alcoholism) is one of the most prevalent substance abuse disorders worldwide. Addiction is thought to arise, in part, from a maladaptive learning process in which enduring memories of drug experiences are formed. However, alcohol (ethanol) generally interferes with synaptic plasticity mechanisms in the CNS and thus impairs various types of learning and memory. Therefore, it is unclear how powerful memories associated with alcohol experience are formed during the development of alcoholism. Here, using brain slice electrophysiology in mice, we show that repeated in vivo ethanol exposure (2 g/kg, i.p., three times daily for 7 d) causes increased susceptibility to the induction of long-term potentiation (LTP) of NMDA receptor (NMDAR)-mediated transmission in mesolimbic dopamine neurons, a form of synaptic plasticity that may drive the learning of stimuli associated with rewards, including drugs of abuse. Enhancement of NMDAR plasticity results from an increase in the potency of inositol 1,4,5-trisphosphate (IP(3)) in producing facilitation of action potential-evoked Ca(2+) signals, which is critical for LTP induction. This increase in IP(3) effect, which lasts for a week but not a month after ethanol withdrawal, occurs through a protein kinase A (PKA)-dependent mechanism. Corticotropin-releasing factor, a stress-related neuropeptide implicated in alcoholism and other addictions, further amplifies the PKA-mediated increase in IP(3) effect in ethanol-treated mice. Finally, we found that ethanol-treated mice display enhanced place conditioning induced by the psychostimulant cocaine. These data suggest that repeated ethanol experience may promote the formation of drug-associated memories by enhancing synaptic plasticity of NMDARs in dopamine neurons.
Collapse
|
31
|
Molecular switch from L-type Ca v 1.3 to Ca v 1.2 Ca2+ channel signaling underlies long-term psychostimulant-induced behavioral and molecular plasticity. J Neurosci 2011; 30:17051-62. [PMID: 21159975 DOI: 10.1523/jneurosci.2255-10.2010] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
L-type Ca(2+) channel (LTCC)-activated signaling cascades contribute significantly to psychostimulant-induced locomotor sensitization; however, the precise contribution of the two brain-specific subunits Ca(v)1.2 and Ca(v)1.3 remains mostly unknown. In this study, by using amphetamine and cocaine locomotor sensitization in mutant mice expressing dihydropyridine (DHP)-insensitive Ca(v)1.2 LTCCs (Ca(v)1.2DHP(-/-)), we find that, as opposed to a previously identified role of the Ca(v)1.3 subunit of LTCCs in development of sensitization, the Ca(v)1.2 subunit mediates expression of amphetamine and cocaine sensitization when examined after a 14 d drug-free period. Molecular studies to further elucidate the role of Ca(v)1.2 versus Ca(v)1.3 LTCCs in activating signaling pathways in the nucleus accumbens (NAc) of drug-naive versus drug-preexposed mice examined 14 d later revealed that an acute amphetamine and cocaine challenge in drug-naive mice increases Ser133 cAMP response element-binding protein (CREB) phosphorylation in the NAc via Ca(v)1.3 channels and via a dopamine D(1)-dependent mechanism, independent of the extracellular signal-regulated kinase (ERK) pathway, an important mediator of psychostimulant-induced plasticity. In contrast, in amphetamine- and cocaine-preexposed mice, an amphetamine or cocaine challenge no longer activates CREB unless Ca(v)1.2 LTCCs are blocked. This Ca(v)1.2-dependent blunting of CREB activation that underlies expression of locomotor sensitization occurs only after extended drug-free periods and involves recruitment of D(1) receptors and the ERK pathway. Thus, our results demonstrate that specific LTCC subunits are required for the development (Ca(v)1.3) versus expression (Ca(v)1.2) of psychostimulant sensitization and that subunit-specific signaling pathways recruited by psychostimulants underlies long-term drug-induced behavioral responses.
Collapse
|
32
|
Mood disorder susceptibility gene CACNA1C modifies mood-related behaviors in mice and interacts with sex to influence behavior in mice and diagnosis in humans. Biol Psychiatry 2010; 68:801-10. [PMID: 20723887 PMCID: PMC2955812 DOI: 10.1016/j.biopsych.2010.06.019] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 05/21/2010] [Accepted: 06/08/2010] [Indexed: 02/04/2023]
Abstract
BACKGROUND Recent genome-wide association studies have associated polymorphisms in the gene CACNA1C, which codes for Ca(v)1.2, with a bipolar disorder and depression diagnosis. METHODS The behaviors of wild-type and Cacna1c heterozygous mice of both sexes were evaluated in a number of tests. Based upon sex differences in our mouse data, we assessed a gene × sex interaction for diagnosis of mood disorders in human subjects. Data from the National Institute of Mental Health Genetics Initiative Bipolar Disorder Consortium and the Genetics of Recurrent Early-Onset Major Depression Consortium were examined using a combined dataset that included 2021 mood disorder cases (1223 female cases) and 1840 control subjects (837 female subjects). RESULTS In both male and female mice, Cacna1c haploinsufficiency was associated with lower exploratory behavior, decreased response to amphetamine, and antidepressant-like behavior in the forced swim and tail suspension tests. Female, but not male, heterozygous mice displayed decreased risk-taking behavior or increased anxiety in multiple tests, greater attenuation of amphetamine-induced hyperlocomotion, decreased development of learned helplessness, and a decreased acoustic startle response, indicating a sex-specific role of Cacna1c. In humans, sex-specific genetic association was seen for two intronic single nucleotide polymorphisms, rs2370419 and rs2470411, in CACNA1C, with effects in female subjects (odds ratio = 1.64, 1.32) but not in male subjects (odds ratio = .82, .86). The interactions by sex were significant after correction for testing 190 single nucleotide polymorphisms (p = 1.4 × 10⁻⁴, 2.1 × 10⁻⁴; p(corrected) = .03, .04) and were consistent across two large datasets. CONCLUSIONS Our preclinical results support a role for CACNA1C in mood disorder pathophysiology, and the combination of human genetic and preclinical data support an interaction between sex and genotype.
Collapse
|
33
|
Mosharov EV, Larsen KE, Kanter E, Phillips KA, Wilson K, Schmitz Y, Krantz DE, Kobayashi K, Edwards RH, Sulzer D. Interplay between cytosolic dopamine, calcium, and alpha-synuclein causes selective death of substantia nigra neurons. Neuron 2009; 62:218-29. [PMID: 19409267 DOI: 10.1016/j.neuron.2009.01.033] [Citation(s) in RCA: 402] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 01/19/2009] [Accepted: 01/24/2009] [Indexed: 11/19/2022]
Abstract
The basis for selective death of specific neuronal populations in neurodegenerative diseases remains unclear. Parkinson's disease (PD) is a synucleinopathy characterized by a preferential loss of dopaminergic neurons in the substantia nigra (SN), whereas neurons of the ventral tegmental area (VTA) are spared. Using intracellular patch electrochemistry to directly measure cytosolic dopamine (DA(cyt)) in cultured midbrain neurons, we confirm that elevated DA(cyt) and its metabolites are neurotoxic and that genetic and pharmacological interventions that decrease DA(cyt) provide neuroprotection. L-DOPA increased DA(cyt) in SN neurons to levels 2- to 3-fold higher than in VTA neurons, a response dependent on dihydropyridine-sensitive Ca2+ channels, resulting in greater susceptibility of SN neurons to L-DOPA-induced neurotoxicity. DA(cyt) was not altered by alpha-synuclein deletion, although dopaminergic neurons lacking alpha-synuclein were resistant to L-DOPA-induced cell death. Thus, an interaction between Ca2+, DA(cyt), and alpha-synuclein may underlie the susceptibility of SN neurons in PD, suggesting multiple therapeutic targets.
Collapse
Affiliation(s)
- Eugene V Mosharov
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
A substrate trapping mutant form of striatal-enriched protein tyrosine phosphatase prevents amphetamine-induced stereotypies and long-term potentiation in the striatum. Biol Psychiatry 2009; 65:637-45. [PMID: 19026408 PMCID: PMC2678172 DOI: 10.1016/j.biopsych.2008.10.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Revised: 10/03/2008] [Accepted: 10/04/2008] [Indexed: 11/23/2022]
Abstract
BACKGROUND Chronic, intermittent exposure to psychostimulant drugs results in striatal neuroadaptations leading to an increase in an array of behavioral responses on subsequent challenge days. A brain-specific striatal-enriched tyrosine phosphatase (STEP) regulates synaptic strengthening by dephosphorylating and inactivating several key synaptic proteins. This study tests the hypothesis that a substrate-trapping form of STEP will prevent the development of amphetamine-induced stereotypies. METHODS A substrate-trapping STEP protein, TAT-STEP (C-S), was infused into the ventrolateral striatum on each of 5 consecutive exposure days and 1 hour before amphetamine injection. Animals were challenged to see whether sensitization to the stereotypy-producing effects of amphetamine developed. The same TAT-STEP (C-S) protein was used on acute striatal slices to determine the impact on long-term potentiation and depression. RESULTS Infusion of TAT-STEP (C-S) blocks the increase of amphetamine-induced stereotypies when given during the 5-day period of sensitization. The TAT-STEP (C-S) has no effect if only infused on the challenge day. Treatment of acute striatal slices with TAT-STEP (C-S) blocks the induction of long-term potentiation and potentates long-term depression. CONCLUSIONS A substrate trapping form of STEP blocks the induction of amphetamine-induced neuroplasticity within the ventrolateral striatum and supports the hypothesis that STEP functions as a tonic break on synaptic strengthening.
Collapse
|
35
|
Liu Y, Chen X. Cholinergic excitation of dopaminergic cells depends on sequential activation of protein kinase C and the L-type calcium channel in ventral tegmental area slices. Brain Res 2008; 1245:41-51. [PMID: 18929546 DOI: 10.1016/j.brainres.2008.09.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Accepted: 09/22/2008] [Indexed: 10/21/2022]
Abstract
Dopaminergic projections from the ventral tegmental area (VTA) constitute the mesolimbocortical system that underlies addiction and psychosis primarily as the result of increased dopaminergic transmission. Dopaminergic neurons in the VTA receive glutamatergic and cholinergic innervations that regulate their firing activities. Both transmitter systems can activate protein kinase C (PKC) by increasing intracellular calcium and lipid second messengers, however, whether PKC mediates increased firing following glutamatergic and cholinergic activation remains unknown. This paper examined the effects of acute PKC inhibition on firing responses to carbachol, NMDA or AMPA using patch clamp recordings from brain slices. The three ligands all induced a reversible increase in firing, however, only carbachol-induced increase in firing was attenuated by the PKC inhibitors chelerythrine or GF 109203X. The L-type calcium channel blocker nifedipine partially blocked carbachol-induced excitation similar to PKC inhibitors. PKC inhibition and L-type channel blockade did not significantly alter NMDA- or AMPA-induced excitation. Concurrent blockade of PKC and L-type channels with chelerythrine and nifedipine did not additively suppress carbachol-induced excitation indicating they were sequential events in the same signaling pathway. Furthermore, preincubation with the PKC inhibitor GF 109203X reduced the carbachol-induced increase in nifedipine-sensitive high-voltage gated calcium currents. These results indicate that cholinergic activation enhances PKC activity, which in turn facilitates L-type channel opening to excite dopaminergic cells, a finding that is in line with reports of increased PKC in the VTA in animals displaying addictive behavior.
Collapse
Affiliation(s)
- Yudan Liu
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada A1B3V6
| | | |
Collapse
|
36
|
Abstract
Although drugs of abuse have different chemical structures and interact with different protein targets, all appear to usurp common neuronal systems that regulate reward and motivation. Addiction is a complex disease that is thought to involve drug-induced changes in synaptic plasticity due to alterations in cell signaling, gene transcription, and protein synthesis. Recent evidence suggests that drugs of abuse interact with and change a common network of signaling pathways that include a subset of specific protein kinases. The best studied of these kinases are reviewed here and include extracellular signal-regulated kinase, cAMP-dependent protein kinase, cyclin-dependent protein kinase 5, protein kinase C, calcium/calmodulin-dependent protein kinase II, and Fyn tyrosine kinase. These kinases have been implicated in various aspects of drug addiction including acute drug effects, drug self-administration, withdrawal, reinforcement, sensitization, and tolerance. Identifying protein kinase substrates and signaling pathways that contribute to the addicted state may provide novel approaches for new pharmacotherapies to treat drug addiction.
Collapse
Affiliation(s)
- Anna M Lee
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California at San Francisco, Emeryville, CA 94608, USA
| | | |
Collapse
|
37
|
Sun WL, Zhou L, Hazim R, Quinones-Jenab V, Jenab S. Effects of dopamine and NMDA receptors on cocaine-induced Fos expression in the striatum of Fischer rats. Brain Res 2008; 1243:1-9. [PMID: 18822274 DOI: 10.1016/j.brainres.2008.09.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 07/28/2008] [Accepted: 09/08/2008] [Indexed: 11/28/2022]
Abstract
Cocaine is an addictive psychostimulant that induces immediate early gene (IEG) expression by activating dopamine (DA) D1 and glutamate NMDA receptors in the striatum. In this study, we show that a single cocaine administration (30 mg/kg) time-dependently increases ERK phosphorylation, c-Fos and FosB protein expression, and MKP-1 phosphorylation (p-MKP-1), in the caudate-putamen (CPu) and nucleus accumbens (NAc) of Fischer rats. In the CPu, 1 h after cocaine injection, the increase in c-Fos and FosB protein expressions is totally abolished by pre-administration of DA-D1 receptor antagonist, SCH23390. In the NAc, SCH23390 also inhibits cocaine-induced c-Fos protein expression. The pre-treatment of NMDA receptor antagonist, MK801, partially reduces cocaine-activated c-Fos protein expression in the CPu. Furthermore, the escalation of p-MKP-1 after acute cocaine administration is dependent on both DA-D1 and NMDA receptor activation in both brain regions examined. Our data suggest that cocaine may modulate ERK pathway signaling through the activation of DA-D1 and NMDA receptors, subsequently influencing the IEG protein expression.
Collapse
Affiliation(s)
- Wei-Lun Sun
- Department of Psychology, Hunter College, CUNY, 695 Park Ave, New York, NY 10065, USA.
| | | | | | | | | |
Collapse
|
38
|
Covington HE, Tropea TF, Rajadhyaksha AM, Kosofsky BE, Miczek KA. NMDA receptors in the rat VTA: a critical site for social stress to intensify cocaine taking. Psychopharmacology (Berl) 2008; 197:203-16. [PMID: 18097654 PMCID: PMC2664317 DOI: 10.1007/s00213-007-1024-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Accepted: 11/13/2007] [Indexed: 01/11/2023]
Abstract
RATIONALE Cocaine strengthens behaviors associated with its administration. The stress response by individuals that are defeated in a brief aggressive confrontation can also promote enduring behavioral consequences similar to those of stimulants. OBJECTIVES The study intends to find whether intermittent episodes of defeat promote cocaine's reinforcing effects by triggering N-methyl-D: -aspartic acid (NMDA)-receptor-mediated plasticity in the ventral tegmental area (VTA). MATERIALS AND METHODS Long-Evans rats were investigated after four social defeats in three experiments. Two experiments examined systemic or intra-VTA antagonism of NMDA receptors during stress on the later expression of behavioral sensitization and cocaine self-administration during fixed and progressive ratio (PR) schedules of reinforcement (0.3 mg/kg/infusion), including a novel 24-h variable-dose continuous access binge (0.2, 0.4, and 0.8 mg/kg/infusion, delivered in an irregular sequence). Third, the expression of receptor proteins NR1 (NMDA) and GluR1 [alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)] were examined in VTA and nucleus accumbens. RESULTS Intermittent defeats augment locomotor responses to cocaine and increase cocaine taking. Rates of responding during binges are increased after defeat stress. These effects are prevented when NMDA or AMPA receptor antagonists are administered before defeats. VTA infusions of the NMDA antagonist AP-5 (5 nmol/side) before stress prevents locomotor sensitization to cocaine and intensified responding for cocaine during a PR schedule or binge. Episodic defeats increase GluR1 AMPA subunit protein expression in the VTA. CONCLUSIONS Social stress stimulates NMDA receptors in the VTA, and this neural action of defeat may be essential for prompting a later increase in cocaine intake during binges.
Collapse
|
39
|
Zhai H, Li Y, Wang X, Lu L. Drug-induced alterations in the extracellular signal-regulated kinase (ERK) signalling pathway: implications for reinforcement and reinstatement. Cell Mol Neurobiol 2008; 28:157-72. [PMID: 18041576 DOI: 10.1007/s10571-007-9240-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Accepted: 11/06/2007] [Indexed: 12/01/2022]
Abstract
Drug addiction, characterized by high rates of relapse, is recognized as a kind of neuroadaptive disorder. Since the extracellular signal-regulated kinase (ERK) pathway is critical to neuroplasticity in the adult brain, understanding the role this pathway plays is important for understanding the molecular mechanism underlying drug addiction and relapse. Here, we review previous literatures that focus on the effects of exposure to cocaine, amphetamine, Delta(9)-tetrahydrocannabinol (THC), nicotine, morphine, and alcohol on ERK signaling in the mesocorticolimbic dopamine system; these alterations of ERK signaling have been thought to contribute to the drug's rewarding effects and to the long-term maladaptation induced by drug abuse. We then discuss the possible upstreams of the ERK signaling pathway activated by exposure of drugs of abuse and the environmental cues previously paired with drugs. Finally, we argue that since ERK activation is a key molecular process in reinstatement of conditioned place preference and drug self-administration, the pharmacological manipulation of the ERK pathway is a potential treatment strategy for drug addiction.
Collapse
Affiliation(s)
- Haifeng Zhai
- Department of Neuropharmacology, National Institute on Drug Dependence, Peking University, 38, Xue Yuan Road, Hai Dian District, Beijing, 100083, China
| | | | | | | |
Collapse
|
40
|
Biala G, Kruk M. Calcium channel antagonists suppress cross-tolerance to the anxiogenic effects of D-amphetamine and nicotine in the mouse elevated plus maze test. Prog Neuropsychopharmacol Biol Psychiatry 2008; 32:54-61. [PMID: 17761379 DOI: 10.1016/j.pnpbp.2007.07.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 07/06/2007] [Accepted: 07/06/2007] [Indexed: 11/29/2022]
Abstract
The purpose of the current experiments was to examine the anxiety-related effects of repeated amphetamine and nicotine administration using the mouse elevated plus maze (EPM). d-amphetamine was administered daily for 8 days (2 mg/kg, i.p.). On the 9th day, mice were challenged with amphetamine (2 mg/kg, i.p.) or nicotine (0.1 mg/kg, s.c.), and were tested 30 min after this last injection. Additionally, a distinct group of mice was pretreated with nicotine (0.1 mg/kg, s.c., 6 days). These mice were subjected to nicotine (0.1 mg/kg, s.c.) or amphetamine (2 mg/kg, i.p.) challenge on the seventh day to see if full crossover effects developed after the pretreatment of both psychostimulant drugs. Moreover, the L-type voltage-dependent calcium channel antagonists nimodipine (5 and 10 mg/kg, i.p.), flunarizine (5 and 10 mg/kg, i.p.), verapamil (5 and 10 mg/kg, i.p.) and diltiazem (5 and 10 mg/kg, i.p.) were injected prior to each injection of chronic d-amphetamine or nicotine. We observed cross-tolerance to the anxiogenic effects of d-amphetamine and nicotine that was blunted by a pretreatment with calcium channel blockers. Overall our findings imply that similar neural calcium-dependent mechanisms are involved in the anxiety-related responses to chronic amphetamine and nicotine injections. As anxiety seems to be an important factor for the development of psychostimulant dependence, the L-type VDCC antagonists can offer an interesting approach for the pharmacotherapy of addiction, including amphetamine and/or nicotine dependence.
Collapse
Affiliation(s)
- Grazyna Biala
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, 4 Staszica Street, 20-081 Lublin, Poland.
| | | |
Collapse
|
41
|
Katsura M, Shibasaki M, Kurokawa K, Tsujimura A, Ohkuma S. Up-regulation of L-type high voltage-gated calcium channel subunits by sustained exposure to 1,4- and 1,5-benzodiazepines in cerebrocortical neurons. J Neurochem 2007; 103:2518-28. [PMID: 17949410 DOI: 10.1111/j.1471-4159.2007.04984.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The aim of this study is to examine how sustained exposure to two 1,4-benzodiazepines (BZDs) with different action period, diazepam and brotizolam, and a 1,5-BZD, clobazam, affects L-type high voltage-gated calcium channel (HVCC) functions and its mechanisms using primary cultures of mouse cerebral cortical neurons. The sustained exposure to these three BZDs increased [(45)Ca2+] influx, which was due to the enhanced [(45)Ca2+] entry through L-type HVCCs but not through of Cav2.1 and Cav2.2. Increase in [(3)H]diltiazem binding after the exposure to these three BZDs was due to the increase in the binding sites of [(3)H]diltiazem. Western blot analysis showed increase of Cav1.2 and Cav1.3 in association with the increased expression of alpha2/delta1 subunit. Similar changes in [(3)H]diltiazem binding and L-type HVCC subunit expression were found in the cerebral cortex from mouse with BZD physical dependence. These results indicate that BZDs examined here have the potential to increase L-type HVCC functions mediated via the enhanced expression of not only Cav1.2 and Cav1.3 but also alpha2/delta1 subunit after their sustained exposure, which may participate in the development of physical dependence by these BZDs.
Collapse
Affiliation(s)
- Masashi Katsura
- Department of Pharmacology, Kawasaki Medical University, Kurashiki, Japan
| | | | | | | | | |
Collapse
|
42
|
Mills K, Ansah T, Ali S, Mukherjee S, Shockley D. Augmented behavioral response and enhanced synaptosomal calcium transport induced by repeated cocaine administration are decreased by calcium channel blockers. Life Sci 2007; 81:600-8. [PMID: 17689567 PMCID: PMC2765982 DOI: 10.1016/j.lfs.2007.06.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2006] [Revised: 06/19/2007] [Accepted: 06/29/2007] [Indexed: 11/26/2022]
Abstract
Recent studies suggest that calcium influx via L-type calcium channels is necessary for psychostimulant-induced behavioral sensitization. In addition, chronic amphetamine upregulates subtype Cav1.2-containing L-type calcium channels. In the present studies, we assessed the effect of calcium channel blockers (CCBs) on cocaine-induced behavioral sensitization and determined whether the functional activity of L-type calcium channels is altered after repeated cocaine administration. Rats were administered daily intraperitoneal injections of either flunarizine (40 mg/kg), diltiazem (40 mg/kg) or cocaine (20 mg/kg) and the combination of the CCBs and cocaine for 30 days. Motor activities were monitored on Day 1, and every 6th day during the 30-day treatment period. Daily cocaine administration produced increased locomotor activity. Maximal augmentation of behavioral response to repeated cocaine administration was observed on Day 18. Flunarizine pretreatment abolished the augmented behavioral response to repeated cocaine administration while diltiazem was less effective. Measurement of tissue monoamine levels on Day 18 revealed cocaine-induced increases in DA and 5-HT in the nucleus accumbens. By contrast to behavioral response, diltiazem was more effective in attenuating increases in monoamine levels than flunarizine. Cocaine administration for 18 days produced increases in calcium uptake in synaptosomes prepared from the nucleus accumbens and frontal cortex. Increases in calcium uptake were abolished by flunarizine and diltiazem pretreatment. Taken together, the augmented cocaine-induced behavioral response on Day 18 may be due to increased calcium uptake in the nucleus accumbens leading to increased dopamine (DA) and serotonin (5-HT) release. Flunarizine and diltiazem attenuated the behavioral response by decreasing calcium uptake and decreasing neurochemical release.
Collapse
Affiliation(s)
- K. Mills
- Department of Pharmacology, Meharry Medical College, 1005 D.B. Todd Boulevard, Nashville, Tennessee 37208, USA
| | - T.A. Ansah
- Department of Pharmacology, Meharry Medical College, 1005 D.B. Todd Boulevard, Nashville, Tennessee 37208, USA
- Corresponding author: Department of Pharmacology, Meharry Medical College, 1005 D.B. Todd Blvd, Nashville, TN 37208, Tel: 615 327 6295, Fax: 615 327 6632, (T.A. Ansah)
| | - S.F. Ali
- Department of Pharmacology, Meharry Medical College, 1005 D.B. Todd Boulevard, Nashville, Tennessee 37208, USA
- Neurochemistry Laboratory, Division of Neurotoxicology, National Center for Toxicological Research/FDA, 3900 NCTR Drive, Jefferson, Arkansas 72079-9502, USA
| | - S. Mukherjee
- Department of Pharmacology, Meharry Medical College, 1005 D.B. Todd Boulevard, Nashville, Tennessee 37208, USA
| | - D.C. Shockley
- Department of Pharmacology, Meharry Medical College, 1005 D.B. Todd Boulevard, Nashville, Tennessee 37208, USA
| |
Collapse
|
43
|
Giordano TP, Satpute SS, Striessnig J, Kosofsky BE, Rajadhyaksha AM. Up-regulation of dopamine D(2)L mRNA levels in the ventral tegmental area and dorsal striatum of amphetamine-sensitized C57BL/6 mice: role of Ca(v)1.3 L-type Ca(2+) channels. J Neurochem 2006; 99:1197-206. [PMID: 17026527 DOI: 10.1111/j.1471-4159.2006.04186.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Dopamine D(2) long (D(2)L) and D(2) short (D(2)S) isoforms of the D(2) receptor play an important role in psychostimulant-induced neuronal adaptations. In this study, we used quantitative real-time PCR to specifically amplify these two splice variants to examine their mRNA expression in the dorsal striatum (dStr), nucleus accumbens (NAc) and the ventral tegmental area (VTA) of amphetamine-sensitized C57BL/6 mice. We found a significant increase in D(2)L mRNA in the VTA and dStr of amphetamine-treated mice that positively correlated with the sensitized locomotor response. We also found a significant increase in D(2)S mRNA in the VTA. We further examined the role of the Ca(v)1.3 subtype of L-type Ca(2+) channels in up-regulation of D(2)L and D(2)S mRNA in the VTA. Amphetamine-pretreated Ca(v)1.3 wild-type (Ca(v)1.3(+/+)) mice exhibited sensitized behavior and a significant increase in D(2)L and D(2)S mRNA compared with saline-pretreated mice Amphetamine-pretreated homozygous Ca(v)1.3 knockout (Ca(v)1.3(-/-)) mice did not exhibit sensitized behavior. There was a significant increase in D(2)S mRNA, but not D(2)L mRNA. In conclusion, our results find that amphetamine increases D(2)L mRNA expression in the dStr and the VTA, an adaptation that correlates with expression of sensitized behavior and dependence on Ca(v)1.3 Ca(2+) channels.
Collapse
Affiliation(s)
- T P Giordano
- Laboratory of Molecular and Developmental Neuroscience, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | | | | | |
Collapse
|
44
|
Valjent E, Corvol JC, Trzaskos JM, Girault JA, Hervé D. Role of the ERK pathway in psychostimulant-induced locomotor sensitization. BMC Neurosci 2006; 7:20. [PMID: 16512905 PMCID: PMC1420315 DOI: 10.1186/1471-2202-7-20] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 03/02/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Repeated exposure to psychostimulants results in a progressive and long-lasting facilitation of the locomotor response that is thought to have implications for addiction. Psychostimulants and other drugs of abuse activate in specific brain areas extracellular signal-regulated kinase (ERK), an essential component of a signaling pathway involved in synaptic plasticity and long-term effects of drugs of abuse. Here we have investigated the role of ERK activation in the behavioral sensitization induced by repeated administration of psychostimulants in mice, using SL327, a brain-penetrating selective inhibitor of MAP-kinase/ERK kinase (MEK), the enzyme that selectively activates ERK. RESULTS A dose of SL327 (30 mg/kg) that reduced the number of activated ERK-positive neurons by 62 to 89% in various brain areas, had virtually no effect on the spontaneous locomotor activity or the acute hyperlocomotion induced by cocaine or D-amphetamine. Pre-treatment with SL327 (30 mg/kg) prior to each drug administration prevented the locomotor sensitization induced by repeated injections of D-amphetamine or cocaine. The SL327 pre-treatment abolished also conditioned locomotor response of mice placed in the context previously paired with cocaine or D-amphetamine. In contrast, SL327 did not alter the expression of sensitized response to D-amphetamine or cocaine. CONCLUSION Altogether these results show that ERK has a minor contribution to the acute locomotor effects of psychostimulants or to the expression of sensitized responses, whereas it is crucial for the acquisition of locomotor sensitization and psychostimulant-conditioned locomotor response. This study supports the important role of the ERK pathway in long-lasting behavioral alterations induced by drugs of abuse.
Collapse
Affiliation(s)
- Emmanuel Valjent
- INSERM, U536, F-75005, Paris; Université Pierre et Marie Curie (UPMC-Paris 6), F-75005, Paris; Institut du Fer a Moulin, F-75005, Paris
| | - Jean-Christophe Corvol
- INSERM, U536, F-75005, Paris; Université Pierre et Marie Curie (UPMC-Paris 6), F-75005, Paris; Institut du Fer a Moulin, F-75005, Paris
| | - James M Trzaskos
- Bristol-Myers Squibb Co., Pharmaceutical Research Institute, Princeton, NJ 08540, USA
| | - Jean-Antoine Girault
- INSERM, U536, F-75005, Paris; Université Pierre et Marie Curie (UPMC-Paris 6), F-75005, Paris; Institut du Fer a Moulin, F-75005, Paris
| | - Denis Hervé
- INSERM, U536, F-75005, Paris; Université Pierre et Marie Curie (UPMC-Paris 6), F-75005, Paris; Institut du Fer a Moulin, F-75005, Paris
| |
Collapse
|
45
|
Valjent E, Corbillé AG, Bertran-Gonzalez J, Hervé D, Girault JA. Inhibition of ERK pathway or protein synthesis during reexposure to drugs of abuse erases previously learned place preference. Proc Natl Acad Sci U S A 2006; 103:2932-7. [PMID: 16473939 PMCID: PMC1413817 DOI: 10.1073/pnas.0511030103] [Citation(s) in RCA: 240] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2005] [Indexed: 11/18/2022] Open
Abstract
Repeated association of drugs of abuse with context leads to long-lasting behavioral responses that reflect reward-controlled learning and participate in the establishment of addiction. Reactivation of consolidated memories is known to produce a reconsolidation process during which memories undergo a labile state. We investigated whether reexposure to drugs had similar effects. Cocaine administration activates extracellular signal-regulated kinase (ERK) in the striatum, and ERK activation is required for the acquisition of cocaine-induced conditioned place preference (CPP). When mice previously conditioned for cocaine-place preference were reexposed to cocaine in the drug-paired compartment after systemic administration of SL327, an inhibitor of ERK activation, CPP response was abolished 24 h later. This procedure also abolished the phosphorylation of ERK and glutamate receptor-1 observed in the ventral and dorsal striatum, 24 h later, during CPP test. Erasure of CPP by SL327 required the combination of cocaine administration and drug-paired context and did not result from enhanced extinction. Similarly, reexposure to morphine in the presence of SL327 long-lastingly abolished response of previously learned morphine-CPP. The effects of SL327 on cocaine- or morphine-CPP were reproduced by protein synthesis inhibition. In contrast, protein synthesis inhibition did not alter previously acquired locomotor sensitization to cocaine. Our findings show that an established CPP can be disrupted when reactivation associates both the conditioned context and drug administration. This process involves ERK, and systemic treatment preventing ERK activation during reexposure erases the previously learned behavioral response. These results suggest potential therapeutic strategies to explore in the context of addiction.
Collapse
Affiliation(s)
- Emmanuel Valjent
- Institut National de la Santé et de la Recherche Médicale (INSERM), U536; Université Pierre et Marie Curie (UPMC-Paris 6); and Institut du Fer à Moulin, F-75005 Paris, France
| | - Anne-Gaëlle Corbillé
- Institut National de la Santé et de la Recherche Médicale (INSERM), U536; Université Pierre et Marie Curie (UPMC-Paris 6); and Institut du Fer à Moulin, F-75005 Paris, France
| | - Jesus Bertran-Gonzalez
- Institut National de la Santé et de la Recherche Médicale (INSERM), U536; Université Pierre et Marie Curie (UPMC-Paris 6); and Institut du Fer à Moulin, F-75005 Paris, France
| | - Denis Hervé
- Institut National de la Santé et de la Recherche Médicale (INSERM), U536; Université Pierre et Marie Curie (UPMC-Paris 6); and Institut du Fer à Moulin, F-75005 Paris, France
| | - Jean-Antoine Girault
- Institut National de la Santé et de la Recherche Médicale (INSERM), U536; Université Pierre et Marie Curie (UPMC-Paris 6); and Institut du Fer à Moulin, F-75005 Paris, France
| |
Collapse
|
46
|
Bonci A, Carlezon WA. Ion channels and intracellular signaling proteins as potential targets for novel therapeutics for addictive and depressive disorders. Pharmacol Ther 2006; 108:65-75. [PMID: 16095714 DOI: 10.1016/j.pharmthera.2005.06.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2005] [Indexed: 11/19/2022]
Abstract
Modern neuroscience is placing increased emphasis on understanding how the activity of ion channels and intracellular molecules in the central nervous system affect behavior. An improved understanding of the brain and the biological bases of conditions such as addictive and depressive disorders is important because it should ultimately enable the design of innovative treatments for these conditions. The development of rational therapies that are based on knowledge of what is different about the addicted or depressed brain would be an important advance. Here, we describe how multidisciplinary studies that combine numerous approaches (behavioral analysis, physiology, molecular biology, and genetic engineering) have begun to provide important advances that have helped to establish causal relationships between the pathophysiology of these conditions and behavior. This type of work has identified classes of molecules on the outside of cells (receptors and ion channels) that receive signals from other cells and initiate cellular events that have short-term effects on the neurons. It has also identified other classes of molecules that are inside of cells (signal transduction molecules) that can have immediate effects on cell function (e.g., ion channel phosphorylation), as well longer term effects (alterations in protein expression) that affect the ways in which neurons function within circuits. Innovative treatments that block, negate, or even reverse the extracellular or intracellular neuroadaptations resulting from exposure to drugs of abuse or stress might be more effective than current therapies because they directly target the molecular processes that cause maladaptive behaviors.
Collapse
Affiliation(s)
- Antonello Bonci
- Ernest Gallo Clinic and Research Center, Department of Neurology, University of California, San Francisco, CA 94110, USA.
| | | |
Collapse
|
47
|
Rajadhyaksha AM, Kosofsky BE. Psychostimulants, L-type calcium channels, kinases, and phosphatases. Neuroscientist 2005; 11:494-502. [PMID: 16151049 PMCID: PMC2706980 DOI: 10.1177/1073858405278236] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
There is growing evidence for the role of voltage-gated L-type Ca2+ channels in mediating aspects of the addictive properties of psychostimulants. L-type Ca2+ channels activate Ca2+ second-messenger pathways that regulate protein phosphorylation and thereby activation of target gene expression. Here the authors will review recent progress in our understanding of L-type Ca2+ channel-activated signal transduction pathways that contribute to molecular neuroadaptations evident following acute and chronic exposures to psychostimulants.
Collapse
Affiliation(s)
- Anjali M Rajadhyaksha
- Labortory of Molecular and Developmental Neuroscience, and Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.
| | | |
Collapse
|
48
|
Rajadhyaksha AM, Kosofsky BE. Psychostimulants, Protein phosphorylation and Gene expression: a growing role of L-type calcium channels. CELLSCIENCE 2005; 2:127-144. [PMID: 16724157 PMCID: PMC1466347 DOI: 10.1901/jaba.2005.2-127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
|
49
|
Guerriero RM, Rajadhyaksha A, Crozatier C, Giros B, Nosten-Bertrand M, Kosofsky BE. Augmented Constitutive CREB Expression in the Nucleus accumbens and Striatum May Contribute to the Altered Behavioral Response to Cocaine of Adult Mice Exposed to Cocaine in utero. Dev Neurosci 2005; 27:235-48. [PMID: 16046859 DOI: 10.1159/000085997] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2004] [Accepted: 10/10/2004] [Indexed: 11/19/2022] Open
Abstract
Neuroadaptations occurring in the mesolimbic dopamine pathway following recurrent exposure to drugs of abuse have been correlated with a behavioral phenomenon known as behavioral sensitization. We have developed an animal model of prenatal cocaine exposure and, using a postnatal sensitization protocol, have examined the subsequent sensitivity of offspring to cocaine. Pregnant Swiss Webster dams were injected twice daily from embryonic day 8 to 17, inclusive, with cocaine (COC40: administered cocaine HCl at a dose of 40 mg/kg/day, and COC20: administered cocaine HCl at a dose of 20 mg/kg/day), or saline (SAL). The SPF40 group (saline pair-fed), a nutritional control group, was 'pair-fed' with COC40 dams. Activity was recorded for 30 min during a 3-day saline habituation, a 14-day 'initiation' phase, when animals received cocaine (15 mg/kg) or saline every other day, and following a 21-day 'withdrawal' period when all mice were challenged with cocaine. COC40 offspring, as compared with SAL controls, did not habituate to a novel environment, demonstrated increased cocaine-induced stereotypy on Coc 1 (first cocaine injection), and blunted locomotor sensitization on challenge as measured by the percentage of each animal's baseline locomotion. Tissue samples of the nucleus accumbens (NAc) and striatum (Str) of all four prenatal treatment groups were examined to determine whether alterations in the transcription factor CREB or glutamate receptor subunit, GluR1, induced by prenatal cocaine treatment may have contributed to the altered behavioral responses. Immunoblot quantitation revealed significantly increased constitutive CREB expression in the NAc and Str of COC40 mice as compared with SAL controls. Such alterations in constitutive CREB levels may contribute to some of the behavioral differences reported in adult mice exposed to cocaine in utero.
Collapse
Affiliation(s)
- Rejean M Guerriero
- Laboratory of Molecular and Developmental Neuroscience, Massachusetts General Hospital-East, Charlestown, MA, USA
| | | | | | | | | | | |
Collapse
|
50
|
Rushlow WJ, Seah YH, Belliveau DJ, Rajakumar N. Changes in calcineurin expression induced in the rat brain by the administration of antipsychotics. J Neurochem 2005; 94:587-96. [PMID: 16033416 DOI: 10.1111/j.1471-4159.2005.03092.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Calcineurin (CN) was recently identified as a susceptibility gene for schizophrenia as well as showing altered RNA expression levels in the post-mortem brains of individuals with schizophrenia. CN knockout mice show a number of behaviours associated with schizophrenia, including deficits in sensorimotor gating, suggesting a link between CN and psychosis. Concurrently, we found, using genome screening techniques, that antipsychotics alter CN expression levels. Therefore, western blotting, in situ hybridization, immunocytochemistry and phosphatase assays were employed to determine what effect antipsychotics have on CN. The results indicate that clozapine, risperidone and haloperidol cause substantial reductions in the A subunit of CN but not CN B at both the RNA and protein levels in the striatum and prefrontal cortex. The changes could only be observed after repeated treatment with antipsychotics but not after acute administration. The alterations in CN protein levels were specific to antipsychotics and mediated by D2 dopamine receptor antagonism. However, despite reductions in CN protein levels, the phosphatase activity of CN was significantly elevated after treatment with antipsychotics. Collectively the results suggest that CN may be a common target for antipsychotics and that antipsychotic-induced alterations in CN may represent one of the mechanisms by which antipsychotics alleviate psychosis.
Collapse
Affiliation(s)
- W J Rushlow
- Department of Psychiatry, the University of Western Ontario and the London Health Sciences Centres, London, Ontario, Canada.
| | | | | | | |
Collapse
|