51
|
Gremel CM, Lovinger DM. Associative and sensorimotor cortico-basal ganglia circuit roles in effects of abused drugs. GENES BRAIN AND BEHAVIOR 2016; 16:71-85. [PMID: 27457495 DOI: 10.1111/gbb.12309] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/21/2016] [Accepted: 07/22/2016] [Indexed: 02/06/2023]
Abstract
The mammalian forebrain is characterized by the presence of several parallel cortico-basal ganglia circuits that shape the learning and control of actions. Among these are the associative, limbic and sensorimotor circuits. The function of all of these circuits has now been implicated in responses to drugs of abuse, as well as drug seeking and drug taking. While the limbic circuit has been most widely examined, key roles for the other two circuits in control of goal-directed and habitual instrumental actions related to drugs of abuse have been shown. In this review we describe the three circuits and effects of acute and chronic drug exposure on circuit physiology. Our main emphasis is on drug actions in dorsal striatal components of the associative and sensorimotor circuits. We then review key findings that have implicated these circuits in drug seeking and taking behaviors, as well as drug use disorders. Finally, we consider different models describing how the three cortico-basal ganglia circuits become involved in drug-related behaviors. This topic has implications for drug use disorders and addiction, as treatments that target the balance between the different circuits may be useful for reducing excessive substance use.
Collapse
Affiliation(s)
- C M Gremel
- Neurosciences Graduate Program, Department of Psychology, University of California San Diego, La Jolla, CA
| | - D M Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
52
|
Differential regulation of the phosphorylation of Trimethyl-lysine27 histone H3 at serine 28 in distinct populations of striatal projection neurons. Neuropharmacology 2016; 107:89-99. [DOI: 10.1016/j.neuropharm.2016.02.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 02/23/2016] [Accepted: 02/27/2016] [Indexed: 01/02/2023]
|
53
|
Abstract
Drug addiction can be viewed as a form of neural plasticity—drug—induced neural plasticity. This is because most features of addiction develop gradually and progressively in response to repeated exposure to a drug of abuse and can persist for a long time, perhaps even a lifetime, after discontinuation of drug administration. Within this context of neural plasticity, studies of drug addiction offer a unique opportunity to establish the biological basis of a complex and clinically relevant behavioral abnormality. This derives from the fact that many aspects of addiction can be reproduced with increasing accuracy in laboratory animals. The availability of animal models, in turn, has made it possible to identify specific regions of the brain that mediate behavioral aspects of addiction. Identification of these discrete brain regions has made it possible to begin the process of identifying the molecular and cellular basis of addiction mediated via these regions. This situation for drug addiction contrasts markedly with that for most other model systems of neural plasticity, for which behavioral, let alone clinical, correlates are not readily apparent. It also contrasts with many other neuropsychiatric abnormalities, for example, schizophrenia and depression, for which animal models are much less straight forward and much more difficult to interpret. Advances made in the field of drug addiction have the potential of providing insight into the types of mechanisms underlying other forms of neural plasticity and even, per haps, other neuropsychiatric abnormalities. The goal of this review is to describe recent progress in under standing the molecular basis of addictive states. The Neuroscientist 1:212-220, 1995
Collapse
Affiliation(s)
- Eric J. Nestler
- Laboratory of Molecular Psychiatry Departments of Psychiatry
and Pharmacology Yale University School of Medicine New Haven, Connecticut
| |
Collapse
|
54
|
Gerfen CR. D1 Dopamine Receptor Supersensitivity in the Dopamine-Depleted Striatum Animal Model of Parkinson’s Disease. Neuroscientist 2016; 9:455-62. [PMID: 14678578 DOI: 10.1177/1073858403255839] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dopamine acts in the striatum principally through the D1 and D2 dopamine receptor subtypes, which are segregated to the direct and indirect striatal projection neurons, respectively. As a consequence, degeneration of the dopamine input to the striatum results in opposing affects in these pathways. The resulting functional imbalance is thought to be responsible for the bradykinesia of Parkinson’s disease, which may be temporarily normalized by dopamine replacement therapy. However, direct striatal projection neurons become irreversibly supersensitive to D1 dopamine receptor activation, despite the fact that there is an actual decrease in receptor number. Recent studies show that this D1-supersensitive response results from a switch from the normal D1-mediated activation of protein-kinase A to an aberrant activation of ERK1/2/MAPkinase. This switch in D1-receptor-mediated regulation of protein kinase systems responsible for neuronal plasticity is suggested to underlie dyskinesia produced by L-DOPA treatment of Parkinson’s disease.
Collapse
Affiliation(s)
- Charles R Gerfen
- Section on Neuroanatomy, Laboratory of Systems Neuroscience, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
55
|
Nutsch VL, Will RG, Robison CL, Martz JR, Tobiansky DJ, Dominguez JM. Colocalization of Mating-Induced Fos and D2-Like Dopamine Receptors in the Medial Preoptic Area: Influence of Sexual Experience. Front Behav Neurosci 2016; 10:75. [PMID: 27147996 PMCID: PMC4834303 DOI: 10.3389/fnbeh.2016.00075] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/01/2016] [Indexed: 01/23/2023] Open
Abstract
Dopamine in the medial preoptic area (mPOA) stimulates sexual activity in males. This is evidenced by microdialysis and microinjection experiments revealing that dopamine receptor antagonists in the mPOA inhibit sexual activity, whereas agonists facilitate behavior. Microdialysis experiments similarly show a facilitative role for dopamine, as levels of dopamine in the mPOA increase with mating. While the majority of evidence suggests an important role for dopamine receptors in the mPOA in the regulation of male sexual behaviors, whether sexual activity or sexual experience influence dopamine receptor function in the mPOA has not been previously shown. Here we used immunohistochemical assays to determine whether varying levels of sexual activity or experience influence the number of cells containing Fos or D2 receptor immunoreactivity. Results show that sexual experience facilitated subsequent behavior, namely experience decreased latencies. Moreover, the number of cells with immunoreactivity for Fos or D2 correlated with levels of sexual experience and sexual activity. Sexual activity increased Fos immunoreactivity. Sexually experienced animals also had significantly more D2-positive cells. Sexually inexperienced animals copulating for the first time had a larger percentage of D2-positive cells containing Fos, when compared to sexually experienced animals. Finally, regardless of experience, animals that had sex prior to sacrifice had significantly more D2-positive cells that contained Fos, vs. animals that did not copulate. These findings are noteworthy because sexually experienced animals display increased sexual efficiency. The differences in activation of D2 and changes in receptor density may play a role in this efficiency and other behavioral changes across sexual experience.
Collapse
Affiliation(s)
- Victoria L Nutsch
- Institute for Neuroscience, The University of Texas at Austin Austin, TX, USA
| | - Ryan G Will
- Department of Psychology, The University of Texas at Austin Austin, TX, USA
| | | | - Julia R Martz
- Department of Psychology, The University of Texas at Austin Austin, TX, USA
| | - Daniel J Tobiansky
- Department of Psychology, The University of Texas at Austin Austin, TX, USA
| | - Juan M Dominguez
- Institute for Neuroscience, The University of Texas at AustinAustin, TX, USA; Department of Psychology, The University of Texas at AustinAustin, TX, USA
| |
Collapse
|
56
|
Zuloaga DG, Johnson LA, Weber S, Raber J. Immediate and lasting effects of chronic daily methamphetamine exposure on activation of cells in hypothalamic-pituitary-adrenal axis-associated brain regions. Psychopharmacology (Berl) 2016; 233:381-92. [PMID: 26525566 PMCID: PMC4815259 DOI: 10.1007/s00213-015-4114-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/11/2015] [Indexed: 10/22/2022]
Abstract
RATIONALE Chronic methamphetamine (MA) abuse leads to dependence and symptoms of withdrawal after use has ceased. Negative mood states associated with withdrawal, as well as drug reinstatement, have been linked to drug-induced disruption of the hypothalamic-pituitary-adrenal (HPA) axis. However, effects of chronic MA exposure or acute MA exposure following withdrawal on neural activation patterns within brain regions that regulate the HPA axis are unknown. OBJECTIVES In this study, neural activation patterns were assessed by quantification of c-Fos protein in mice exposed to different regimens of MA administration. METHODS (Experiment 1) Adult male mice were treated with MA (5 mg/kg) or saline once or once daily for 10 days. (Experiment 2) Mice were treated with MA or saline once daily for 10 days and following a 10-day withdrawal period were re-administered a final dose of MA or saline. c-Fos was quantified in brains after the final injection. RESULTS (Experiment 1) Compared to exposure to a single dose of MA (5 mg/kg), chronic MA exposure decreased the number of c-Fos expressing cells in the paraventricular hypothalamus, dorsomedial hypothalamus, central amygdala, basolateral amygdala, bed nucleus of the stria terminalis (BNST), and CA3 hippocampal region. (Experiment 2) Compared to mice receiving their first dose of MA, mice chronically treated with MA, withdrawn, and re-administered MA, showed decreased c-Fos expressing cells within the central and basolateral amygdala, BNST, and CA3. CONCLUSIONS HPA axis-associated amygdala, extended amygdala, and hippocampal regions endure lasting effects following chronic MA exposure and therefore may be linked to stress-related withdrawal symptoms.
Collapse
Affiliation(s)
- Damian G. Zuloaga
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, Portland, OR 97239, USA,Department of Psychology, University at Albany, Albany, NY 12222, USA
| | - Lance A. Johnson
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, Portland, OR 97239, USA
| | - Sydney Weber
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, Portland, OR 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, Portland, OR 97239, USA,Department of Neurology, Oregon Health and Science University Portland, Portland, OR 97239, USA,Department of Radiation Medicine, Oregon Health and Science University Portland, Portland, OR 97239, USA,Division of Neuroscience, ONPRC, Oregon Health and Science University Portland, Portland, OR 97239, USA
| |
Collapse
|
57
|
CB1 Cannabinoid Agonist (WIN55,212-2) Within the Basolateral Amygdala Induced Sensitization to Morphine and Increased the Level of μ-Opioid Receptor and c-fos in the Nucleus Accumbens. J Mol Neurosci 2016; 58:446-55. [DOI: 10.1007/s12031-016-0716-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/12/2016] [Indexed: 12/17/2022]
|
58
|
Crittenden J, Graybiel A. Disease-Associated Changes in the Striosome and Matrix Compartments of the Dorsal Striatum. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2016. [DOI: 10.1016/b978-0-12-802206-1.00039-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
59
|
Smith ML, Li J, Cote DM, Ryabinin AE. Effects of isoflurane and ethanol administration on c-Fos immunoreactivity in mice. Neuroscience 2015; 316:337-43. [PMID: 26742790 DOI: 10.1016/j.neuroscience.2015.12.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/17/2015] [Accepted: 12/25/2015] [Indexed: 11/19/2022]
Abstract
Noninvasive functional imaging holds great promise for the future of translational research, due to the ability to directly compare between preclinical and clinical models of psychiatric disorders. Despite this potential, concerns have been raised regarding the necessity to anesthetize rodent and monkey subjects during these procedures, because anesthetics may alter neuronal activity. For example, in studies on drugs of abuse and alcohol, it is not clear to what extent anesthesia can interfere with drug-induced neural activity. Therefore, the current study investigated whole-brain c-Fos activation following isoflurane anesthesia as well as ethanol-induced activation of c-Fos in anesthetized mice. In the first experiment, we examined effects of one or three sessions of gaseous isoflurane on c-Fos activation across the brain in male C57BL/6J mice. Isoflurane administration led to c-Fos activation in several areas, including the piriform cortex and lateral septum. Lower or similar levels of activation in these areas were detected after three sessions of isoflurane, suggesting that multiple exposures may eliminate some of the enhanced neuronal activation caused by acute isoflurane. In the second experiment, we investigated the ability of ethanol injection (1.5 or 2.5g/kgi.p.) to induce c-Fos activation under anesthesia. Following three sessions of isoflurane, 1.5g/kg of ethanol induced c-Fos in the central nucleus of amygdala and the centrally-projecting Edinger-Westphal nucleus (EWcp). This induction was lower after 2.5g/kg of ethanol. These results demonstrate that ethanol-induced neural activation can be detected in the presence of isoflurane anesthesia. They also suggest, that while habituation to isoflurane helps reduce neuronal activation, interaction between effects of anesthesia and alcohol can occur. Studies using fMRI imaging could benefit from using habituated animals and dose-response analyses.
Collapse
Affiliation(s)
- M L Smith
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L470, Portland, OR 97239, USA
| | - J Li
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L470, Portland, OR 97239, USA
| | - D M Cote
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L470, Portland, OR 97239, USA
| | - A E Ryabinin
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code L470, Portland, OR 97239, USA; Portland Alcohol Research Center, Portland, OR, USA.
| |
Collapse
|
60
|
Synergistic Interactions of D1- and D2-Selective Dopamine Agonists in Animal Models for Parkinson’s Disease: Sites of Action and Implications for the Pathogenesis of Dyskinesias. Can J Neurol Sci 2015. [DOI: 10.1017/s0317167100041536] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
ABSTRACT:The addition of a D2 agonist such as bromocriptine to L-Dopa therapy can often improve the response of patients with Parkinson’s disease dramatically. Simultaneous activation of D1 and D2 dopamine receptors can produce a synergistic effect on locomotion in rats and primates. However, despite the importance of this addition of a D2 agonist to the D1/D2 agonist L-Dopa, little is known of the sites of action of these agents. Recent work suggests that, in addition to D1 and D2 dopamine receptor sites in the striatum (caudate-putamen), L-Dopa and D1 agonists have important effects at D1 dopamine receptors in the substantia nigra. Animal experiments suggest that D1 and D2 dopamine receptor agonists probably also affect different outflow pathways from the striatum. An understanding of these pathways and how dopamine agonists affect them gives insight into some of the clinical problems experienced in treating Parkinson’s disease (the “on-off phenomenon, for example). D1/D2 dopamine receptors also differentially affect gene expression and regulation in the striatum. An understanding of the anatomical and biochemical location of the actions of dopamine receptor agonists will be important in maximizing the beneficial effects and minimizing the side-effects of both presently-used drugs and new treatments.
Collapse
|
61
|
Torres OV, McCoy MT, Ladenheim B, Jayanthi S, Brannock C, Tulloch I, Krasnova IN, Cadet JL. CAMKII-conditional deletion of histone deacetylase 2 potentiates acute methamphetamine-induced expression of immediate early genes in the mouse nucleus accumbens. Sci Rep 2015; 5:13396. [PMID: 26300473 PMCID: PMC4547138 DOI: 10.1038/srep13396] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 07/28/2015] [Indexed: 12/03/2022] Open
Abstract
Methamphetamine (METH) produces increases in the expression of immediate early genes (IEGs) and of histone deacetylase 2 (HDAC2) in the rat nucleus accumbens (NAc). Here, we tested whether HDAC2 deletion influenced the effects of METH on IEG expression in the NAc. Microarray analyses showed no baseline differences in IEG expression between wild-type (WT) and HDAC2 knockout (KO) mice. Quantitative-PCR analysis shows that an acute METH injection produced time-dependent increases in mRNA levels of several IEGs in both genotypes. Interestingly, HDAC2KO mice displayed greater METH-induced increases in Egr1 and Egr2 mRNA levels measured at one hour post-injection. The levels of Fosb, Fra2, Egr1, and Egr3 mRNAs stayed elevated in the HDAC2KO mice 2 hours after the METH injection whereas these mRNAs had normalized in the WT mice. In WT mice, METH caused increased HDAC2 recruitment to the promoters some IEGs at 2 hours post injection. METH-induced prolonged increases in Fosb, Fra2, Egr1, and Egr3 mRNA levels in HDAC2KO mice were associated with increased enrichment of phosphorylated CREB (pCREB) on the promoters of these genes. Based on our observations, we hypothesize that HDAC2 may regulate the expression of these genes, in part, by prolonging the actions of pCREB in the mouse NAc.
Collapse
Affiliation(s)
- Oscar V Torres
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224
| | - Michael T McCoy
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224
| | - Christie Brannock
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224
| | - Ingrid Tulloch
- Department of Psychology, Stevenson University, Stevenson, MD 21283
| | - Irina N Krasnova
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224
| |
Collapse
|
62
|
Casarsa BS, Marinzalda MÁ, Marchese NA, Paz MC, Vivas L, Baiardi G, Bregonzio C. A previous history of repeated amphetamine exposure modifies brain angiotensin II AT1 receptor functionality. Neuroscience 2015; 307:1-13. [PMID: 26299338 DOI: 10.1016/j.neuroscience.2015.08.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/29/2015] [Accepted: 08/13/2015] [Indexed: 11/19/2022]
Abstract
UNLABELLED Previous results from our laboratory showed that angiotensin II AT1 receptors (AT1-R) are involved in the neuroadaptative changes induced by amphetamine. The aim of the present work was to study functional and neurochemical responses to angiotensin II (ANG II) mediated by AT1-R activation in animals previously exposed to amphetamine. For this purpose male Wistar rats (250-320 g) were treated with amphetamine (2.5mg/kg/day intraperitoneal) or saline for 5 days and implanted with intracerebroventricular (i.c.v.) cannulae. Seven days after the last amphetamine administration the animals received ANG II (400 pmol) i.c.v. One group was tested in a free choice paradigm for sodium (2% NaCl) and water intake and sacrificed for Fos immunoreactivity (Fos-IR) determinations. In a second group of rats, urine and plasma samples were collected for electrolytes and plasma renin activity determination and then they were sacrificed for Fos-IR determination in Oxytocinergic neurons (Fos-OT-IR). RESULTS Repeated amphetamine exposure (a) prevented the increase in sodium intake and Fos-IR cells in caudate-putamen and accumbens nucleus induced by ANG II i.c.v. (b) potentiated urinary sodium excretion and Fos-OT-IR in hypothalamus and (c) increased the inhibitory response in plasma renin activity, in response to ANG II i.c.v. Our results indicate a possible functional desensitisation of AT1-R in response to ANG II, induced by repeated amphetamine exposure. This functional AT1-R desensitisation allows to unmask the effects of ANG II i.c.v. mediated by oxytocin. We conclude that the long lasting changes in brain AT1-R functionality should be considered among the psychostimulant-induced neuroadaptations.
Collapse
Affiliation(s)
- B S Casarsa
- Laboratorio de Neurofarmacología, Instituto de Investigaciones Biológicas y Tecnológicas (IIByT-CONICET), Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Córdoba, Argentina
| | - M Á Marinzalda
- Laboratorio de Neurofarmacología, Instituto de Investigaciones Biológicas y Tecnológicas (IIByT-CONICET), Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Córdoba, Argentina
| | - N A Marchese
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M C Paz
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - L Vivas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - G Baiardi
- Laboratorio de Neurofarmacología, Instituto de Investigaciones Biológicas y Tecnológicas (IIByT-CONICET), Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Córdoba, Argentina
| | - C Bregonzio
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
63
|
Pisanu A, Lecca D, Valentini V, Bahi A, Dreyer JL, Cacciapaglia F, Scifo A, Piras G, Cadoni C, Di Chiara G. Impairment of acquisition of intravenous cocaine self-administration by RNA-interference of dopamine D1-receptors in the nucleus accumbens shell. Neuropharmacology 2015; 89:398-411. [PMID: 25446574 DOI: 10.1016/j.neuropharm.2014.10.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 01/17/2023]
Abstract
Microdialysis during i.v. drug self-administration (SA) have implicated nucleus accumbens (NAc) shell DA in cocaine and heroin reinforcement. However, this correlative evidence has not been yet substantiated by experimental evidence obtained by studying the effect of selective manipulation of NAc shell DA transmission on cocaine and heroin SA. In order to investigate this issue, DA D1a receptor (D1aR) expression was impaired in the NAc shell and core by locally infusing lentiviral vectors (LV) expressing specific D1aR-siRNAs (LV-siRNAs). Control rats were infused in the same areas with LV expressing GFP. Fifteen days later, rats were trained to acquire i.v. cocaine or heroin self-administration (SA). At the end of behavioral experiments, in order to evaluate the effect of LV-siRNA on D1aR expression, rats were challenged with amphetamine and the brains were processed for immunohistochemical detection of c-Fos and D1aR. Control rats acquired i.v. cocaine and heroin SA. Infusion of LV-siRNAs in the medial NAc shell reduced D1aR density and the number of c-Fos positive nuclei in the NAc shell, while sparing the core, and prevented the acquisition of cocaine, but not heroin SA. In turn, LV-siRNAs infusion in the core reduced D1aR density and the number of c-Fos positive nuclei in the same area, while sparing the shell, and failed to affect acquisition of cocaine. The differential effect of LV impairment of NAc shell D1aR on cocaine and heroin SA indicates that NAc shell DA acting on D1aR specifically mediates cocaine reinforcement.
Collapse
Affiliation(s)
- Augusta Pisanu
- Institute of Neuroscience, National Research Council of Italy, 09124 Cagliari, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Murray RC, Logan MC, Horner KA. Striatal patch compartment lesions reduce stereotypy following repeated cocaine administration. Brain Res 2015; 1618:286-98. [PMID: 26100338 DOI: 10.1016/j.brainres.2015.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/06/2015] [Accepted: 06/12/2015] [Indexed: 12/13/2022]
Abstract
Stereotypy can be characterized as inflexible, repetitive behaviors that occur following repeated exposure to psychostimulants, such as cocaine (COC). Stereotypy may be related to preferential activation of the patch (striosome) compartment of striatum, as enhanced relative activation of the patch compartment has been shown to positively correlate with the emergence of stereotypy following repeated psychostimulant treatment. However, the specific contribution of the patch compartment to COC-induced stereotypy following repeated exposure is unknown. To elucidate the involvement of the patch compartment to the development of stereotypy following repeated COC exposure, we determined if destruction of this sub-region altered COC-induced behaviors. The neurons of the patch compartment were ablated by bilateral infusion of the neurotoxin dermorphin-saporin (DERM-SAP; 17 ng/μl) into the striatum. Animals were allowed to recover for eight days following the infusion, and then were given daily injections of COC (25mg/kg) or saline for one week, followed by a weeklong drug-free period. Animals were then given a challenge dose of saline or COC, observed for 2h in activity chambers and sacrificed. The number of mu-labeled patches in the striatum were reduced by DERM-SAP pretreatment. In COC-treated animals DERM-SAP pretreatment significantly reduced the immobilization and intensity of stereotypy but increased locomotor activity. DERM-SAP pretreatment attenuated COC-induced c-Fos expression in the patch compartment, while enhancing COC-induced c-Fos expression in the matrix compartment. These data indicate that the patch compartment contributes to repetitive behavior and suggests that alterations in activity in the patch vs matrix compartments may underlie to this phenomenon.
Collapse
Affiliation(s)
- Ryan C Murray
- Division of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA 31207, United States
| | - Mary C Logan
- Division of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA 31207, United States
| | - Kristen A Horner
- Division of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA 31207, United States.
| |
Collapse
|
65
|
Wright KN, Hollis F, Duclot F, Dossat AM, Strong CE, Francis TC, Mercer R, Feng J, Dietz DM, Lobo MK, Nestler EJ, Kabbaj M. Methyl supplementation attenuates cocaine-seeking behaviors and cocaine-induced c-Fos activation in a DNA methylation-dependent manner. J Neurosci 2015; 35:8948-58. [PMID: 26063926 PMCID: PMC4461693 DOI: 10.1523/jneurosci.5227-14.2015] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/06/2015] [Accepted: 05/07/2015] [Indexed: 12/14/2022] Open
Abstract
Epigenetic mechanisms, such as histone modifications, regulate responsiveness to drugs of abuse, such as cocaine, but relatively little is known about the regulation of addictive-like behaviors by DNA methylation. To investigate the influence of DNA methylation on the locomotor-activating effects of cocaine and on drug-seeking behavior, rats receiving methyl supplementation via chronic l-methionine (MET) underwent either a sensitization regimen of intermittent cocaine injections or intravenous self-administration of cocaine, followed by cue-induced and drug-primed reinstatement. MET blocked sensitization to the locomotor-activating effects of cocaine and attenuated drug-primed reinstatement, with no effect on cue-induced reinstatement or sucrose self-administration and reinstatement. Furthermore, upregulation of DNA methyltransferase 3a and 3b and global DNA hypomethylation were observed in the nucleus accumbens core (NAc), but not in the medial prefrontal cortex (mPFC), of cocaine-pretreated rats. Glutamatergic projections from the mPFC to the NAc are critically involved in the regulation of cocaine-primed reinstatement, and activation of both brain regions is seen in human addicts when reexposed to the drug. When compared with vehicle-pretreated rats, the immediate early gene c-Fos (a marker of neuronal activation) was upregulated in the NAc and mPFC of cocaine-pretreated rats after cocaine-primed reinstatement, and chronic MET treatment blocked its induction in both regions. Cocaine-induced c-Fos expression in the NAc was associated with reduced methylation at CpG dinucleotides in the c-Fos gene promoter, effects reversed by MET treatment. Overall, these data suggest that drug-seeking behaviors are, in part, attributable to a DNA methylation-dependent process, likely occurring at specific gene loci (e.g., c-Fos) in the reward pathway.
Collapse
Affiliation(s)
- Katherine N Wright
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, Florida 32306
| | - Fiona Hollis
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, Florida 32306, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
| | - Florian Duclot
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, Florida 32306
| | - Amanda M Dossat
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, Florida 32306
| | - Caroline E Strong
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, Florida 32306
| | - T Chase Francis
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Roger Mercer
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, Florida 32306
| | - Jian Feng
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, and
| | - David M Dietz
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York 14214
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Eric J Nestler
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, and
| | - Mohamed Kabbaj
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University, Tallahassee, Florida 32306,
| |
Collapse
|
66
|
Integration of neural networks activated by amphetamine in females with different estrogen levels: a functional imaging study in awake rats. Psychoneuroendocrinology 2015; 56:200-12. [PMID: 25827963 DOI: 10.1016/j.psyneuen.2015.02.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/10/2015] [Accepted: 02/24/2015] [Indexed: 01/15/2023]
Abstract
Previous studies demonstrate that schizophrenia symptomatology in women is dependent upon estrogen levels. Estrogen has beneficial properties when administered in conjunction with antipsychotics, and estrogen also alters, in rats, dopamine neurotransmission, which is a common target of all antipsychotic medications, suggesting a possible interaction between the two. The aim of the current study was to investigate this possible interaction using functional magnetic resonance imaging in awake, female rats. Amphetamine-sensitized, ovariectomized rats receiving no, chronic low, or phasic high levels of estradiol replacement were used, and changes in blood-oxygen-level-dependent (BOLD) signal were recorded over time in response to an acute amphetamine injection. Increasing levels of estradiol enhanced BOLD activation in pathways previously known to be implicated in schizophrenia symptomatology, such as the mesocorticolimbic, habenular and olfactory pathways, as well as more widespread areas. We propose here the first comprehensive "amphetamine activation map" integrating brain regions where amphetamine-related BOLD activity is influenced by estrogen levels in sensitized female rats.
Collapse
|
67
|
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
|
68
|
Kufahl PR, Peartree NA, Heintzelman KL, Chung M, Neisewander JL. Region-specific effects of isoflurane anesthesia on Fos immunoreactivity in response to intravenous cocaine challenge in rats with a history of repeated cocaine administration. Brain Res 2015; 1594:256-66. [PMID: 25451087 PMCID: PMC4805112 DOI: 10.1016/j.brainres.2014.10.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
We have previously shown that acute intravenous (i.v.) administration of cocaine increases Fos immunoreactivity in rats under isoflurane anesthesia. Given that Fos expression is a marker of neural activation, the results suggested that isoflurane is appropriate for imaging cocaine effects under anesthesia. However, most imaging research in this area utilizes subjects with a history of repeated cocaine exposure and this drug history may interact with anesthetic use differently from acute cocaine exposure. Thus, this study further examined Fos expression under isoflurane in rats with a history of repeated i.v. cocaine administration. Rats received daily injections of either saline or cocaine (2mg/kg, i.v.) across 7 consecutive days, followed by 5 days of no drug exposure. On the test day, rats were either nonanesthetized or anesthetized under isoflurane and were given an acute challenge of cocaine (2mg/kg, i.v.). Additional saline-exposed controls received a saline challenge. Ninety min after the drug challenge, the rats were perfused under isoflurane anesthesia and their brains were processed for Fos protein immunohistochemistry. We found that challenge injections of cocaine following a regimen of repeated cocaine exposure resulted in Fos expression in the prefrontal cortex and striatum roughly equivalent to that found in rats who had received the cocaine challenge after a history of vehicle injections. Additionally, isoflurane anesthesia resulted in a heterogeneous attenuation of cocaine-induced Fos expression, with the most robust effect in the orbital cortex but no effect in the nucleus accumbens core (NAcC). These results indicate that cocaine-induced Fos is preserved in the NAcC under isoflurane, suggesting that isoflurane can be used in imaging studies involving cocaine effects in this region.
Collapse
Affiliation(s)
- Peter R Kufahl
- Department of Psychology, Arizona State University, Tempe, AZ 85287-1104, United States.
| | - Natalie A Peartree
- Department of Psychology, Arizona State University, Tempe, AZ 85287-1104, United States
| | - Krista L Heintzelman
- Department of Psychology, Arizona State University, Tempe, AZ 85287-1104, United States
| | - Maggie Chung
- Department of Psychology, Arizona State University, Tempe, AZ 85287-1104, United States
| | - Janet L Neisewander
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, United States
| |
Collapse
|
69
|
Effects of homeopathic Anax imperator on behavioural and pain models in mice. HOMEOPATHY 2015; 104:15-23. [DOI: 10.1016/j.homp.2014.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/07/2014] [Accepted: 05/04/2014] [Indexed: 12/25/2022]
|
70
|
Gill MJ, Weiss ML, Cain ME. Effects of differential rearing on amphetamine-induced c-fos expression in rats. Drug Alcohol Depend 2014; 145:231-4. [PMID: 25454411 DOI: 10.1016/j.drugalcdep.2014.09.779] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 09/02/2014] [Accepted: 09/25/2014] [Indexed: 11/20/2022]
Abstract
BACKGROUND Rearing rats in environmental enrichment alters psychostimulant-induced locomotor activity as well as neurotransmitter expression. Exposure to novelty and psychostimulants induces c-fos expression in neurons in the mesolimbic dopamine (DA) pathway. Here we investigated changes in the expression of the immediate early gene c-fos in the mesolimbic DA pathway of enriched, isolated, or socially reared rats due to the neurobiological changes that result from rearing conditions and influence drug taking behavior. METHODS Rats were reared in either enriched (EC), isolated (IC), or social (SC) conditions for 30 days, after which they received an acute amphetamine or saline injection (1.0 mg/kg) and locomotor activity was measured. Following immunohistochemical staining c-fos positive neurons were quantified in the NAcc, mPFC, and amygdala. RESULTS Greater locomotor activity was observed in differentially reared rats treated with amphetamine compared to saline, as well as in SC compared to EC and IC rats. Rats reared in an IC context demonstrated greater c-fos expression than EC rats in the NAcc when treated with amphetamine, and EC saline rats demonstrated greater c-fos expression in the cingulate and prelimbic cortices compared to SC saline rats. Additionally, IC amphetamine rats displayed greater c-fos expression in the NAcc compared to IC saline rats, while EC saline rats displayed greater c-fos expression in the prelimbic cortex compared to EC amphetamine rats. CONCLUSIONS These results suggest regional specificity of psychostimulant-induced c-fos expression in the prelimbic/NAcc pathway that is altered in differential rearing, and influences initial c-fos activation following psychostimulant exposure.
Collapse
Affiliation(s)
- Margaret J Gill
- Kansas State University, Manhattan, KS 66506, United States; North Central College, Department of Psychology, 30 North Brainard St, Naperville, IL 60540, United States.
| | - Mark L Weiss
- Kansas State University, Manhattan, KS 66506, United States
| | - Mary E Cain
- Kansas State University, Manhattan, KS 66506, United States
| |
Collapse
|
71
|
Cruz FC, Javier Rubio F, Hope BT. Using c-fos to study neuronal ensembles in corticostriatal circuitry of addiction. Brain Res 2014; 1628:157-73. [PMID: 25446457 DOI: 10.1016/j.brainres.2014.11.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 10/27/2014] [Accepted: 11/01/2014] [Indexed: 01/02/2023]
Abstract
Learned associations between drugs and environment play an important role in addiction and are thought to be encoded within specific patterns of sparsely distributed neurons called neuronal ensembles. This hypothesis is supported by correlational data from in vivo electrophysiology and cellular imaging studies in relapse models in rodents. In particular, cellular imaging with the immediate early gene c-fos and its protein product Fos has been used to identify sparsely distributed neurons that were strongly activated during conditioned drug behaviors such as drug self-administration and context- and cue-induced reinstatement of drug seeking. Here we review how Fos and the c-fos promoter have been employed to demonstrate causal roles for Fos-expressing neuronal ensembles in prefrontal cortex and nucleus accumbens in conditioned drug behaviors. This work has allowed identification of unique molecular and electrophysiological alterations within Fos-expressing neuronal ensembles that may contribute to the development and expression of learned associations in addiction.
Collapse
Affiliation(s)
- Fabio C Cruz
- Behavioral Neuroscience Branch, IRP/NIDA/NIH/DHHS, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, United States
| | - F Javier Rubio
- Behavioral Neuroscience Branch, IRP/NIDA/NIH/DHHS, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, United States
| | - Bruce T Hope
- Behavioral Neuroscience Branch, IRP/NIDA/NIH/DHHS, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, United States.
| |
Collapse
|
72
|
Lepelletier FX, Tauber C, Nicolas C, Solinas M, Castelnau P, Belzung C, Emond P, Cortese S, Faraone SV, Chalon S, Galineau L. Prenatal exposure to methylphenidate affects the dopamine system and the reactivity to natural reward in adulthood in rats. Int J Neuropsychopharmacol 2014; 18:pyu044. [PMID: 25522388 PMCID: PMC4360227 DOI: 10.1093/ijnp/pyu044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 06/24/2014] [Accepted: 07/08/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Methylphenidate (MPH) is a commonly-used medication for the treatment of children with Attention-Deficit/Hyperactivity Disorders (ADHD). However, its prescription to adults with ADHD and narcolepsy raises the question of how the brain is impacted by MPH exposure during pregnancy. The goal of this study was to elucidate the long-term neurobiological consequences of prenatal exposure to MPH using a rat model. METHODS We focused on the effects of such treatment on the adult dopamine (DA) system and on the reactivity of animals to natural rewards. RESULTS This study shows that adult male rats prenatally exposed to MPH display elevated expression of presynaptic DA markers in the DA cell bodies and the striatum. Our results also suggest that MPH-treated animals could exhibit increased tonic DA activity in the mesolimbic pathway, altered signal-to-noise ratio after a pharmacological stimulation, and decreased reactivity to the locomotor effects of cocaine. Finally, we demonstrated that MPH rats display a decreased preference and motivation for sucrose. CONCLUSIONS This is the first preclinical study reporting long-lasting neurobiological alterations of DA networks as well as alterations in motivational behaviors for natural rewards after a prenatal exposure to MPH. These results raise concerns about the possible neurobiological consequences of MPH treatment during pregnancy.
Collapse
Affiliation(s)
- François-Xavier Lepelletier
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Clovis Tauber
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Céline Nicolas
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Marcello Solinas
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Pierre Castelnau
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Catherine Belzung
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Patrick Emond
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Samuele Cortese
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Stephen V Faraone
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Sylvie Chalon
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Laurent Galineau
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone).
| |
Collapse
|
73
|
A Genetic Mouse Model of Parkinson's Disease Shows Involuntary Movements and Increased Postsynaptic Sensitivity to Apomorphine. Mol Neurobiol 2014; 52:1152-1164. [PMID: 25307288 DOI: 10.1007/s12035-014-8911-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 09/28/2014] [Indexed: 10/24/2022]
Abstract
Alpha-synuclein (SNCA) protein aggregation plays a causal role in Parkinson's disease (PD). The SNCA protein modulates neurotransmission via the SNAP receptor (SNARE) complex assembly and presynaptic vesicle trafficking. The striatal presynaptic dopamine deficit is alleviated by treatment with levodopa (L-DOPA), but postsynaptic plastic changes induced by this treatment lead to a development of involuntary movements (dyskinesia). While this process is currently modeled in rodents harboring neurotoxin-induced lesions of the nigrostriatal pathway, we have here explored the postsynaptic supersensitivity of dopamine receptor-mediated signaling in a genetic mouse model of early PD. To this end, we used mice with prion promoter-driven overexpression of A53T-SNCA in the nigrostriatal and corticostriatal projections. At a symptomatic age (18 months), mice were challenged with apomorphine (5 mg/kg s.c.) and examined using both behavioral and molecular assays. After the administration of apomorphine, A53T-transgenic mice showed more severe stereotypic and dystonic movements in comparison with wild-type controls. Molecular markers of extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation and dephosphorylation, and Fos messenger RNA (mRNA), were examined in striatal tissue at 30 and 100 min after apomorphine injection. At 30 min, wild-type and transgenic mice showed a similar induction of phosphorylated ERK1/2, Dusp1, and Dusp6 mRNA (two MAPK phosphatases). At the same time point, Fos mRNA was induced more strongly in mutant mice than in wild-type controls. At 100 min after apomorphine treatment, the induction of both Fos, Dusp1, and Dusp6 mRNA was significantly larger in mutant mice than wild-type controls. At this time point, apomorphine caused a reduction in phospho-ERK1/2 levels specifically in the transgenic mice. Our results document for the first time a disturbance of ERK1/2 signaling regulation associated with apomorphine-induced involuntary movements in a genetic mouse model of synucleinopathy. This mouse model will be useful to identify novel therapeutic targets that can counteract abnormal dopamine-dependent striatal plasticity during both prodromal and manifest stages of PD.
Collapse
|
74
|
Varani AP, Moutinho Machado L, Balerio GN. Baclofen prevented the changes in c-Fos and brain-derived neutrophic factor expressions during mecamylamine-precipitated nicotine withdrawal in mice. Synapse 2014; 68:508-17. [DOI: 10.1002/syn.21763] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/12/2014] [Accepted: 06/30/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Andrés P. Varani
- Instituto de Investigaciones Farmacológicas (UBA-CONICET); Junín 956, 5° piso, (C1113AAD) Buenos Aires Argentina
| | - Lirane Moutinho Machado
- Instituto de Investigaciones Farmacológicas (UBA-CONICET); Junín 956, 5° piso, (C1113AAD) Buenos Aires Argentina
| | - Graciela N. Balerio
- Instituto de Investigaciones Farmacológicas (UBA-CONICET); Junín 956, 5° piso, (C1113AAD) Buenos Aires Argentina
- Cátedra de Farmacología; Facultad de Farmacia y Bioquímica; Universidad de Buenos Aires; Junín 956 5° Piso, (C1113AAD) Buenos Aires Argentina
| |
Collapse
|
75
|
Pistollato F, Louisse J, Scelfo B, Mennecozzi M, Accordi B, Basso G, Gaspar JA, Zagoura D, Barilari M, Palosaari T, Sachinidis A, Bremer-Hoffmann S. Development of a pluripotent stem cell derived neuronal model to identify chemically induced pathway perturbations in relation to neurotoxicity: effects of CREB pathway inhibition. Toxicol Appl Pharmacol 2014; 280:378-88. [PMID: 25150140 DOI: 10.1016/j.taap.2014.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 07/01/2014] [Accepted: 08/11/2014] [Indexed: 01/09/2023]
Abstract
According to the advocated paradigm shift in toxicology, acquisition of knowledge on the mechanisms underlying the toxicity of chemicals, such as perturbations of biological pathways, is of primary interest. Pluripotent stem cells (PSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer a unique opportunity to derive physiologically relevant human cell types to measure molecular and cellular effects of such pathway modulations. Here we compared the neuronal differentiation propensity of hESCs and hiPSCs with the aim to develop novel hiPSC-based tools for measuring pathway perturbation in relation to molecular and cellular effects in vitro. Among other fundamental pathways, also, the cAMP responsive element binding protein (CREB) pathway was activated in our neuronal models and gave us the opportunity to study time-dependent effects elicited by chemical perturbations of the CREB pathway in relation to cellular effects. We show that the inhibition of the CREB pathway, using 2-naphthol-AS-E-phosphate (KG-501), induced an inhibition of neurite outgrowth and synaptogenesis, as well as a decrease of MAP2(+) neuronal cells. These data indicate that a CREB pathway inhibition can be related to molecular and cellular effects that may be relevant for neurotoxicity testing, and, thus, qualify the use of our hiPSC-derived neuronal model for studying chemical-induced neurotoxicity resulting from pathway perturbations.
Collapse
Affiliation(s)
| | - Jochem Louisse
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Bibiana Scelfo
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Milena Mennecozzi
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Benedetta Accordi
- Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy
| | - Giuseppe Basso
- Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy
| | - John Antonydas Gaspar
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Dimitra Zagoura
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Manuela Barilari
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Taina Palosaari
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Agapios Sachinidis
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | | |
Collapse
|
76
|
Ruffle JK. Molecular neurobiology of addiction: what's all the (Δ)FosB about? THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2014; 40:428-37. [PMID: 25083822 DOI: 10.3109/00952990.2014.933840] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The transcription factor ΔFosB is upregulated in numerous brain regions following repeated drug exposure. This induction is likely to, at least in part, be responsible for the mechanisms underlying addiction, a disorder in which the regulation of gene expression is thought to be essential. In this review, we describe and discuss the proposed role of ΔFosB as well as the implications of recent findings. The expression of ΔFosB displays variability dependent on the administered substance, showing region-specificity for different drug stimuli. This transcription factor is understood to act via interaction with Jun family proteins and the formation of activator protein-1 (AP-1) complexes. Once AP-1 complexes are formed, a multitude of molecular pathways are initiated, causing genetic, molecular and structural alterations. Many of these molecular changes identified are now directly linked to the physiological and behavioral changes observed following chronic drug exposure. In addition, ΔFosB induction is being considered as a biomarker for the evaluation of potential therapeutic interventions for addiction.
Collapse
Affiliation(s)
- James K Ruffle
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| |
Collapse
|
77
|
Crittenden JR, Lacey CJ, Lee T, Bowden HA, Graybiel AM. Severe drug-induced repetitive behaviors and striatal overexpression of VAChT in ChAT-ChR2-EYFP BAC transgenic mice. Front Neural Circuits 2014; 8:57. [PMID: 24904300 PMCID: PMC4036131 DOI: 10.3389/fncir.2014.00057] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/12/2014] [Indexed: 01/01/2023] Open
Abstract
In drug users, drug-related cues alone can induce dopamine release in the dorsal striatum. Instructive cues activate inputs to the striatum from both dopaminergic and cholinergic neurons, which are thought to work together to support motor learning and motivated behaviors. Imbalances in these neuromodulatory influences can impair normal action selection and might thus contribute to pathologically repetitive and compulsive behaviors such as drug addiction. Dopamine and acetylcholine can have either antagonistic or synergistic effects on behavior, depending on the state of the animal and the receptor signaling systems at play. Semi-synchronized activation of cholinergic interneurons in the dorsal striatum drives dopamine release via presynaptic nicotinic acetylcholine receptors located on dopamine terminals. Nicotinic receptor blockade is known to diminish abnormal repetitive behaviors (stereotypies) induced by psychomotor stimulants. By contrast, blockade of postsynaptic acetylcholine muscarinic receptors in the dorsomedial striatum exacerbates drug-induced stereotypy, exemplifying how different acetylcholine receptors can also have opposing effects. Although acetylcholine release is known to be altered in animal models of drug addiction, predicting whether these changes will augment or diminish drug-induced behaviors thus remains a challenge. Here, we measured amphetamine-induced stereotypy in BAC transgenic mice that have been shown to overexpress the vesicular acetylcholine transporter (VAChT) with consequent increased acetylcholine release. We found that drug-induced stereotypies, consisting of confined sniffing and licking behaviors, were greatly increased in the transgenic mice relative to sibling controls, as was striatal VAChT protein. These findings suggest that VAChT-mediated increases in acetylcholine could be critical in exacerbating drug-induced stereotypic behaviors and promoting exaggerated behavioral fixity.
Collapse
Affiliation(s)
- Jill R Crittenden
- Department of Brain and Cognitive Sciences and McGovern Institute for Brain Research, Massachusetts Institute of Technology Cambridge, MA, USA
| | - Carolyn J Lacey
- Department of Brain and Cognitive Sciences and McGovern Institute for Brain Research, Massachusetts Institute of Technology Cambridge, MA, USA
| | - Tyrone Lee
- Department of Brain and Cognitive Sciences and McGovern Institute for Brain Research, Massachusetts Institute of Technology Cambridge, MA, USA
| | - Hilary A Bowden
- Department of Brain and Cognitive Sciences and McGovern Institute for Brain Research, Massachusetts Institute of Technology Cambridge, MA, USA
| | - Ann M Graybiel
- Department of Brain and Cognitive Sciences and McGovern Institute for Brain Research, Massachusetts Institute of Technology Cambridge, MA, USA
| |
Collapse
|
78
|
Issy A, Del Bel E. 7-Nitroindazole blocks the prepulse inhibition disruption and c-Fos increase induced by methylphenidate. Behav Brain Res 2014; 262:74-83. [DOI: 10.1016/j.bbr.2013.12.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/17/2013] [Accepted: 12/21/2013] [Indexed: 01/03/2023]
|
79
|
Santos L, Opris I, Hampson R, Godwin DW, Gerhardt G, Deadwyler S. Functional dynamics of primate cortico-striatal networks during volitional movements. Front Syst Neurosci 2014; 8:27. [PMID: 24653682 PMCID: PMC3947991 DOI: 10.3389/fnsys.2014.00027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 02/06/2014] [Indexed: 01/08/2023] Open
Abstract
The motor cortex and dorsal striatum (caudate nucleus and putamen) are key regions in motor processing but the interface between the cortex and striatum is not well understood. While dorsal striatum integrates information from multiple brain regions to shape motor learning and habit formation, the disruption of cortico-striatal circuits compromises the functionality of these circuits resulting in a multitude of neurologic disorders, including Parkinson's disease. To better understand the modulation of the cortico-striatal circuits we recorded simultaneously single neuron activity from four brain regions, primary motor, and sensory cortices, together with the rostral and caudal segments of the putamen in rhesus monkeys performing a visual motor task. Results show that spatial and temporal-task related firing relationships between these cortico-striatal circuit regions were modified by the independent administration of the two drugs (cocaine and baclofen). Spatial tuning and correlated firing of neurons from motor cortex and putamen were severely disrupted by cocaine and baclofen on correct trials, while the two drugs have dramatically decreased the functional connectivity of the motor cortical-striatal network. These findings provide insight into the modulation of cortical-striatal firing related to movement with implications for therapeutic approaches to Parkinson's disease and related disorders.
Collapse
Affiliation(s)
- Lucas Santos
- Department of Physiology and Pharmacology, Wake Forest University Medical School Winston-Salem, NC, USA
| | - Ioan Opris
- Department of Physiology and Pharmacology, Wake Forest University Medical School Winston-Salem, NC, USA
| | - Robert Hampson
- Department of Physiology and Pharmacology, Wake Forest University Medical School Winston-Salem, NC, USA
| | - Dwayne W Godwin
- Department of Physiology and Pharmacology, Wake Forest University Medical School Winston-Salem, NC, USA ; Department of Neurobiology and Anatomy, Wake Forest University Medical School Winston-Salem, NC, USA
| | - Greg Gerhardt
- Department of Neurobiology and Neurology, University of Kentucky Lexington, KY, USA
| | - Samuel Deadwyler
- Department of Physiology and Pharmacology, Wake Forest University Medical School Winston-Salem, NC, USA
| |
Collapse
|
80
|
Adult hemiparkinsonian rats do not benefit from tactile stimulation. Behav Brain Res 2014; 261:97-105. [DOI: 10.1016/j.bbr.2013.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/03/2013] [Accepted: 12/07/2013] [Indexed: 10/25/2022]
|
81
|
Bobzean SAM, DeNobrega AK, Perrotti LI. Sex differences in the neurobiology of drug addiction. Exp Neurol 2014; 259:64-74. [PMID: 24508560 DOI: 10.1016/j.expneurol.2014.01.022] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 01/21/2014] [Accepted: 01/27/2014] [Indexed: 01/09/2023]
Abstract
Epidemiological data demonstrate that while women report lower rates of drug use than men, the number of current drug users and abusers who are women continues to increase. In addition women progress through the phases of addiction differently than men; women transition from casual drug use to addiction faster, are more reactive to stimuli that trigger relapse, and have higher rates of relapse then men. Sex differences in physiological and psychological responses to drugs of abuse are well documented and it is well established that estrogen effects on dopamine (DA) systems are largely responsible for these sex differences. However, the downstream mechanisms that result from interactions between estrogen and the effects of drugs of abuse on the DA system are just beginning to be explored. Here we review the basic neurocircuitry which underlies reward and addiction; highlighting the neuroadaptive changes that occur in the mesolimbic dopamine reward and anti-reward/stress pathways. We propose that sex differences in addiction are due to sex differences in the neural systems which mediate positive and negative reinforcement and that these differences are modulated by ovarian hormones. This forms a neurobehavioral basis for the search for the molecular and cellular underpinnings that uniquely guide motivational behaviors and make women more vulnerable to developing and sustaining addiction than men.
Collapse
Affiliation(s)
- Samara A M Bobzean
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Aliza K DeNobrega
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Linda I Perrotti
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA.
| |
Collapse
|
82
|
Matsumoto I, Alexander-Kaufman K, Iwazaki T, Kashem MA, Matsuda-Matsumoto H. CNS proteomes in alcohol and drug abuse and dependence. Expert Rev Proteomics 2014; 4:539-52. [PMID: 17705711 DOI: 10.1586/14789450.4.4.539] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Drugs of abuse, including alcohol, can induce dependency formation and/or brain damage in brain regions important for cognition. 'High-throughput' approaches, such as cDNA microarray and proteomics, allow the analysis of global expression profiles of genes and proteins. These technologies have recently been applied to human brain tissue from patients with psychiatric illnesses, including substance abuse/dependence and appropriate animal models to help understand the causes and secondary effects of these complex disorders. Although these types of studies have been limited in number and by proteomics techniques that are still in their infancy, several interesting hypotheses have been proposed. Focusing on CNS proteomics, we aim to review and update current knowledge in this rapidly advancing area.
Collapse
Affiliation(s)
- Izuru Matsumoto
- University of Sydney, Discipline of Pathology, NSW, Australia.
| | | | | | | | | |
Collapse
|
83
|
Luo J, Wang T, Liang S, Hu X, Li W, Jin F. Experimental gastritis leads to anxiety- and depression-like behaviors in female but not male rats. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2013; 9:46. [PMID: 24345032 PMCID: PMC3878489 DOI: 10.1186/1744-9081-9-46] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 12/10/2013] [Indexed: 02/08/2023]
Abstract
Human and animals studies support the idea that there is a gender-related co-morbidity of pain-related and inflammatory gastrointestinal (GI) diseases with psychological disorders. This co-morbidity is the evidence for the existence of GI-brain axis which consists of immune (cytokines), neural (vagus nerve) and neuroendocrine (HPA axis) pathways. Psychological stress causes disturbances in GI physiology, such as altered GI barrier function, changes in motility and secretion, development of visceral hypersensitivity, and dysfunction of inflammatory responses. Whether GI inflammation would exert impact on psychological behavior is not well established. We examined the effect of experimental gastritis on anxiety- and depression-like behaviors in male and female Sprague-Dawley rats, and evaluated potential mechanisms of action. Gastritis was induced by adding 0.1% (w/v) iodoacetamide (IAA) to the sterile drinking water for 7 days. Sucrose preference test assessed the depression-like behavior, open field test and elevated plus maze evaluated the anxiety-like behavior. IAA treatment induced gastric inflammation in rats of either gender. No behavioral abnormality or dysfunction of GI-brain axis was observed in male rats with IAA-induced gastritis. Anxiety- and depression-like behaviors were apparent and the HPA axis was hyperactive in female rats with IAA-induced gastritis. Our results show that gastric inflammation leads to anxiety- and depression-like behaviors in female but not male rats via the neuroendocrine (HPA axis) pathway, suggesting that the GI inflammation can impair normal brain function and induce changes in psychological behavior in a gender-related manner through the GI-to-brain signaling.
Collapse
Affiliation(s)
- Jia Luo
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tao Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Shan Liang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xu Hu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Feng Jin
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
84
|
Liu W, Peng QX, Lin XL, Luo CH, Jiang MJ, Mo ZX, Yung KKL. Effect of rhynchophylline on the expression of p-CREB and sc-Fos in triatum and hippocampal CA1 area of methamphetamine-induced conditioned place preference rats. Fitoterapia 2013; 92:16-22. [PMID: 24140441 DOI: 10.1016/j.fitote.2013.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 09/30/2013] [Accepted: 10/05/2013] [Indexed: 02/04/2023]
Abstract
To explore the effect of rhynchophylline (Rhy) on the expression of p-CREB and c-Fos in the striatum and hippocampal CA1 area of methamphetamine-induced conditioned place preference (CPP) rat, methamphetamine (2 mg/kg) was injected to rats and the conditioned place preference was observed in these rats treated with or without Rhy. An immunohistochemistry assay was used to determine the expression of p-CREB and c-Fos in the striatum and hippocampal CA1 area. Methamphetamine induced significant behavior alteration in CPP, while after pretreatment with rhynchophylline or ketamine, the time of staying in methamphetamine-paired compartment of rats was significantly reduced. Methamphetamine also increased the number of p-CREB positive cells in the striatum and hippocampal CA1 zone, as well as p-Fos positive cells. However, the compound Rhy could attenuate the effect. These findings show that Rhy can suppress the acquisition of CPP in rats induced by methamphetamine and the action may be related with the reduced expression of p-CREB and p-Fos in the striatum and hippocampus.
Collapse
Affiliation(s)
- Wei Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qiu-Xian Peng
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Xiao-Liang Lin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Chao-Hua Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ming-Jin Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Zhi-Xian Mo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Ken Kin-Lam Yung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| |
Collapse
|
85
|
Tobiansky DJ, Roma PG, Hattori T, Will RG, Nutsch VL, Dominguez JM. The medial preoptic area modulates cocaine-induced activity in female rats. Behav Neurosci 2013; 127:293-302. [PMID: 23565937 DOI: 10.1037/a0031949] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Drugs of abuse exert their effects by exploiting natural neurobiological reward mechanisms, especially the mesolimbic dopamine (DA) system. However, the mesolimbic system does not operate in isolation, and input from other reward-relevant structures may play a role in cocaine's rewarding effects. The medial preoptic area (mPOA) of the hypothalamus is involved in the regulation of two essential and naturally rewarding behaviors: sexual and maternal behaviors. It also makes strong neuroanatomical connections with areas of the mesolimbic system, particularly the ventral tegmental area (VTA). As such, the mPOA is a logical candidate for a neuroanatomical locus modulating activity in the mesolimbic system and emergent behavioral expressions of drug reward, yet the role of this structure is largely unexplored. Here, using a female rat model, we show that the mPOA innervates the VTA in a region-specific manner, that lesions of the mPOA augment cocaine-induced Fos expression in the nucleus accumbens (NAc) and cocaine-induced conditioned place preference. We also show that approximately 68% of mPOA-VTA efferents release γ-aminobutyric acid (GABA), over 75% are sensitive to DA as evidenced by colocalization with DA receptors, and nearly 60% of these contain both DA receptors and GABA, which suggests a novel key role for the mPOA in the inhibition of the mesolimbic DA circuit. Combined, these results reveal the mPOA as a critical modulating structure in cocaine-induced mesolimbic activity and behavioral manifestation of reward, at least in part, via GABAergic output that is sensitive to DA input.
Collapse
Affiliation(s)
- Daniel J Tobiansky
- Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA
| | | | | | | | | | | |
Collapse
|
86
|
Cadet JL, Jayanthi S, McCoy MT, Ladenheim B, Saint-Preux F, Lehrmann E, De S, Becker KG, Brannock C. Genome-wide profiling identifies a subset of methamphetamine (METH)-induced genes associated with METH-induced increased H4K5Ac binding in the rat striatum. BMC Genomics 2013; 14:545. [PMID: 23937714 PMCID: PMC3751638 DOI: 10.1186/1471-2164-14-545] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/20/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND METH is an illicit drug of abuse that influences gene expression in the rat striatum. Histone modifications regulate gene transcription. METHODS We therefore used microarray analysis and genome-scale approaches to examine potential relationships between the effects of METH on gene expression and on DNA binding of histone H4 acetylated at lysine 4 (H4K5Ac) in the rat dorsal striatum of METH-naïve and METH-pretreated rats. RESULTS Acute and chronic METH administration caused differential changes in striatal gene expression. METH also increased H4K5Ac binding around the transcriptional start sites (TSSs) of genes in the rat striatum. In order to relate gene expression to histone acetylation, we binned genes of similar expression into groups of 100 genes and proceeded to relate gene expression to H4K5Ac binding. We found a positive correlation between gene expression and H4K5Ac binding in the striatum of control rats. Similar correlations were observed in METH-treated rats. Genes that showed acute METH-induced increased expression in saline-pretreated rats also showed METH-induced increased H4K5Ac binding. The acute METH injection caused similar increases in H4K5Ac binding in METH-pretreated rats, without affecting gene expression to the same degree. Finally, genes that showed METH-induced decreased expression exhibited either decreases or no changes in H4K5Ac binding. CONCLUSION Acute METH injections caused increased gene expression of genes that showed increased H4K5Ac binding near their transcription start sites.
Collapse
Affiliation(s)
- Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Ziółkowska B, Korostyński M, Piechota M, Kubik J, Przewłocki R. Effects of morphine on immediate-early gene expression in the striatum of C57BL/6J and DBA/2J mice. Pharmacol Rep 2013; 64:1091-104. [PMID: 23238466 DOI: 10.1016/s1734-1140(12)70906-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 05/22/2012] [Indexed: 12/25/2022]
Abstract
BACKGROUND Immediate early gene (IEG) induction elicited by drugs of abuse may contribute to development of plastic changes in the brain responsible for drug-induced behavioral changes leading to addiction. The aim of the present study was to characterize the changes in IEG expression in the striatum and nucleus accumbens produced by an acute or chronic administration of morphine. METHODS In order to search for a possible relationship between morphine-induced IEG expression and behavior, the experiment was performed on two inbred strains of mice, C57BL/6J and DBA/2J, which differ markedly in their sensitivity to the rewarding and locomotor stimulatory actions of opiates. Gene expression was assessed using RT-PCR and DNA microarrays. RESULTS The experiments demonstrated a prolonged or a delayed up-regulation of 14 IEG in the striatum at 4 h after morphine administration. Among them, a cluster of 8 genes, including 6 inducible transcription factors (c-fos, fra-2, junB, zif268 (egr1), egr2, NGFI-B) and 2 effector IEG (arc and mkp1) seemed to be regulated in concert in response to morphine. This group of genes was induced to a greater degree after chronic than acute morphine administration selectively in C57BL/6J mice and the difference bore apparently no relationship to opiate-produced locomotor activation. The strain-selective regulation was also demonstrated for cyclin L2 and tPA after an acute morphine injection. CONCLUSIONS Our data indicate that morphine up-regulates many IEG in the mouse striatum at a strikingly delayed time-point and that these changes are genotype-dependent. They also suggest inter-strain differences in the development of striatal neuroadaptations to chronic morphine treatment.
Collapse
Affiliation(s)
- Barbara Ziółkowska
- Department of Molecular Neuropharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343 Kraków, Poland.
| | | | | | | | | |
Collapse
|
88
|
Niessen NA, Balthazart J, Ball GF, Charlier TD. C-fos down-regulation inhibits testosterone-dependent male sexual behavior and the associated learning. Eur J Neurosci 2013; 38:3325-37. [PMID: 23895306 DOI: 10.1111/ejn.12321] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 06/24/2013] [Indexed: 01/20/2023]
Abstract
Environmental stimulation results in an increased expression of transcription factors called immediate early genes (IEGs) in specific neuronal populations. In male Japanese quail, copulation with a female increases the expression of the IEGs zenk and c-fos in the medial pre-optic nucleus (POM), a key nucleus controlling male sexual behavior. The functional significance of this increased IEG expression that follows performance of copulatory behavior is unknown. We addressed this question by repeatedly quantifying the performance of appetitive (learned social proximity response) and consummatory (actual copulation) sexual behavior in castrated, testosterone-treated males that received daily intra-cerebroventricular injection of an antisense oligodeoxynucleotide targeting c-fos or control vehicle. Daily antisense injections significantly inhibited the expression of copulatory behavior as well as the acquisition of the learned social proximity response. A strong reduction of the proximity response was still observed in antisense-treated birds that copulated with a female, ruling out the indirect effect of the absence of interactions with females on the learning process. After a 2-day interruption of behavioral testing but not of antisense injections, birds were submitted to a final copulatory test that confirmed the behavioral inhibition in antisense-injected birds. Brains were collected at 90 min after the behavioral testing for quantification of c-fos-immunoreactive cells. A significant reduction of the number of c-fos-positive cells in the POM but not in other brain regions was observed following antisense injection. Taken together, the data suggest that c-fos expression in the POM modulates copulatory behavior and sexual learning in male quail.
Collapse
Affiliation(s)
- Neville-Andrew Niessen
- Research Group in Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liège, Avenue de l'Hôpital 1 (Bat. B36), B-4000, Liège 1, Belgium
| | | | | | | |
Collapse
|
89
|
Krasnova IN, Chiflikyan M, Justinova Z, McCoy MT, Ladenheim B, Jayanthi S, Quintero C, Brannock C, Barnes C, Adair JE, Lehrmann E, Kobeissy FH, Gold MS, Becker KG, Goldberg SR, Cadet JL. CREB phosphorylation regulates striatal transcriptional responses in the self-administration model of methamphetamine addiction in the rat. Neurobiol Dis 2013; 58:132-43. [PMID: 23726845 DOI: 10.1016/j.nbd.2013.05.009] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/08/2013] [Accepted: 05/20/2013] [Indexed: 10/26/2022] Open
Abstract
Neuroplastic changes in the dorsal striatum participate in the transition from casual to habitual drug use and might play a critical role in the development of methamphetamine (METH) addiction. We examined the influence of METH self-administration on gene and protein expression that may form substrates for METH-induced neuronal plasticity in the dorsal striatum. Male Sprague-Dawley rats self-administered METH (0.1mg/kg/injection, i.v.) or received yoked saline infusions during eight 15-h sessions and were euthanized 2h, 24h, or 1month after cessation of METH exposure. Changes in gene and protein expression were assessed using microarray analysis, RT-PCR and Western blots. Chromatin immunoprecipitation (ChIP) followed by PCR was used to examine epigenetic regulation of METH-induced transcription. METH self-administration caused increases in mRNA expression of the transcription factors, c-fos and fosb, the neurotrophic factor, Bdnf, and the synaptic protein, synaptophysin (Syp) in the dorsal striatum. METH also caused changes in ΔFosB, BDNF and TrkB protein levels, with increases after 2 and 24h, but decreases after 1month of drug abstinence. Importantly, ChIP-PCR showed that METH self-administration caused enrichment of phosphorylated CREB (pCREB), but not of histone H3 trimethylated at lysine 4 (H3K4me3), on promoters of c-fos, fosb, Bdnf and Syp at 2h after cessation of drug intake. These findings show that METH-induced changes in gene expression are mediated, in part, by pCREB-dependent epigenetic phenomena. Thus, METH self-administration might trigger epigenetic changes that mediate alterations in expression of genes and proteins serving as substrates for addiction-related synaptic plasticity.
Collapse
Affiliation(s)
- Irina N Krasnova
- Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Striatal patch compartment lesions alter methamphetamine-induced behavior and immediate early gene expression in the striatum, substantia nigra and frontal cortex. Brain Struct Funct 2013; 219:1213-29. [PMID: 23625147 DOI: 10.1007/s00429-013-0559-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 04/18/2013] [Indexed: 10/26/2022]
Abstract
Methamphetamine (METH) induces stereotypy, which is characterized as inflexible, repetitive behavior. Enhanced activation of the patch compartment of the striatum has been correlated with stereotypy, suggesting that stereotypy may be related to preferential activation of this region. However, the specific contribution of the patch compartment to METH-induced stereotypy is not clear. To elucidate the involvement of the patch compartment to the development of METH-induced stereotypy, we determined if destruction of this sub-region altered METH-induced behaviors. Animals were bilaterally infused in the striatum with the neurotoxin dermorphin-saporin (DERM-SAP; 17 ng/μl) to specifically ablate the neurons of the patch compartment. Eight days later, animals were treated with METH (7.5 mg/kg), placed in activity chambers, observed for 2 h and killed. DERM-SAP pretreatment significantly reduced the number and total area of mu-labeled patches in the striatum. DERM-SAP pretreatment significantly reduced the intensity of METH-induced stereotypy and the spatial immobility typically observed with METH-induced stereotypy. In support of this observation, DERM-SAP pretreatment also significantly increased locomotor activity in METH-treated animals. In the striatum, DERM-SAP pretreatment attenuated METH-induced c-Fos expression in the patch compartment, while enhancing METH-induced c-Fos expression in the matrix compartment. DERM-SAP pretreatment followed by METH administration augmented c-Fos expression in the SNpc and reduced METH-induced c-Fos expression in the SNpr. In the medial prefrontal, but not sensorimotor cortex, c-Fos and zif/268 expression was increased following METH treatment in animals pre-treated with DERM-SAP. These data indicate that the patch compartment is necessary for the expression of repetitive behaviors and suggests that alterations in activity in the basal ganglia may contribute to this phenomenon.
Collapse
|
91
|
Electroacupuncture reduces cocaine-induced seizures and mortality in mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:134610. [PMID: 23690833 PMCID: PMC3652148 DOI: 10.1155/2013/134610] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 02/17/2013] [Indexed: 12/20/2022]
Abstract
The aims of this study were to characterize the protective profile of electroacupuncture (EA) on cocaine-induced seizures and mortality in mice. Mice were treated with EA (2 Hz, 50 Hz, and 100 Hz), or they underwent needle insertion without anesthesia at the Dazhui (GV14) and Baihui (GV20) acupoints before cocaine administration. EA at 50 Hz applied to GV14 and GV20 significantly reduced the seizure severity induced by a single dose of cocaine (75 mg/kg; i.p.). Furthermore, needle insertion into GV14 and GV20 and EA at 2 Hz and 50 Hz at both acupoints significantly reduced the mortality rate induced by a single lethal dose of cocaine (125 mg/kg; i.p.). In the sham control group, EA at 50 Hz applied to bilateral Tianzong (SI11) acupoints had no protective effects against cocaine. In addition, EA at 50 Hz applied to GV14 and GV20 failed to reduce the incidence of seizures and mortality induced by the local anesthetic procaine. In an immunohistochemistry study, EA (50 Hz) pretreatment at GV14 and GV20 decreased cocaine (75 mg/kg; i.p.)-induced c-Fos expression in the paraventricular thalamus. While the dopamine D3 receptor antagonist, SB-277011-A (30 mg/kg; s.c), did not by itself affect cocaine-induced seizure severity, it prevented the effects of EA on cocaine-induced seizures. These results suggest that EA alleviates cocaine-induced seizures and mortality and that the dopamine D3 receptor is involved, at least in part, in the anticonvulsant effects of EA in mice.
Collapse
|
92
|
Nielsen DA, Utrankar A, Reyes JA, Simons DD, Kosten TR. Epigenetics of drug abuse: predisposition or response. Pharmacogenomics 2013; 13:1149-60. [PMID: 22909205 DOI: 10.2217/pgs.12.94] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Drug addiction continues to be a serious medical and social problem. Vulnerability to develop an addiction to drugs is dependent on genetic, environmental, social and biological factors. In particular, the interactions of environmental and genetic factors indicate the significance of epigenetic mechanisms, which have been found to occur in response to illicit drug use or as underlying factors in chronic substance abuse and relapse. Epigenetics is defined as the heritable and possibly reversible modifications in gene expression that do not involve alterations in the DNA sequence. This review discusses the various types of epigenetic modifications and their relevance to drug addiction to elucidate whether epigenetics is a predisposing factor, or a response to, developing an addiction to drugs of abuse.
Collapse
Affiliation(s)
- David A Nielsen
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine & the Michael E DeBakey VA Medical Center, 2002 Holcombe Boulevard, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
93
|
Burton CL, Rizos Z, Diwan M, Nobrega JN, Fletcher PJ. Antagonizing 5-HT2A receptors with M100907 and stimulating 5-HT2C receptors with Ro60-0175 blocks cocaine-induced locomotion and zif268 mRNA expression in Sprague-Dawley rats. Behav Brain Res 2013. [DOI: 10.1016/j.bbr.2012.11.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
94
|
White SL, Schmidt HD, Vassoler FM, Pierce RC. Acute cocaine increases phosphorylation of CaMKII and GluA1 in the dorsolateral striatum of drug naïve rats, but not cocaine-experienced rats. Neurosci Lett 2013; 537:71-6. [PMID: 23352852 DOI: 10.1016/j.neulet.2013.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 01/09/2013] [Accepted: 01/10/2013] [Indexed: 12/13/2022]
Abstract
Transport of GluA1-containing AMPA glutamate receptors to synapses in the nucleus accumbens, a process that involves phosphorylation of key serine residues by CaMKII, is associated with the reinstatement of cocaine-seeking behavior. A growing body of evidence indicates that the dorsal striatum contributes to aspects of cocaine addiction. However, the potential role of CaMKII-mediated phosphorylation of GluA1 subunits in the dorsolateral (DL) striatum during cocaine reinstatement has not been examined. In this study, rats were trained to self-administer cocaine and were partnered with saline-yoked rats that received injections of saline. Following extinction, each pair of rats received either a systemic priming injection of cocaine (10mg/kg, i.p.) or saline. As expected, cocaine-experienced rats displayed robust reinstatement of cocaine seeking in response to a challenge injection, whereas yoked saline controls did not. The DL striatum was dissected immediately following the reinstatement test session. Results from Western blotting experiments showed increased pGluA1-ser831 and pCaMKII-thr286 in the DL striatum of saline-yoked rats given an acute injection of cocaine. This effect was absent in cocaine-experienced rats that received a saline injection, and no changes were observed following a priming injection of cocaine in cocaine-experienced rats. These results indicate that acute exposure to cocaine in drug naïve rats increased CaMKII-mediated phosphorylation of GluA1-containing AMPA receptors in the DL striatum, an effect that was not observed during cocaine priming-induced reinstatement of drug seeking. It is possible; therefore, that increased phosphorylation of CaMKII and GluA1 following acute cocaine is a compensatory mechanism in the DL striatum.
Collapse
Affiliation(s)
- Samantha L White
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 125 South 31st Street, Room 1102a, Philadelphia, PA 19104, USA.
| | | | | | | |
Collapse
|
95
|
|
96
|
|
97
|
Bepari AK, Sano H, Tamamaki N, Nambu A, Tanaka KF, Takebayashi H. Identification of optogenetically activated striatal medium spiny neurons by Npas4 expression. PLoS One 2012; 7:e52783. [PMID: 23300775 PMCID: PMC3530472 DOI: 10.1371/journal.pone.0052783] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 11/20/2012] [Indexed: 11/19/2022] Open
Abstract
Optogenetics is a powerful neuromodulatory tool with many unique advantages to explore functions of neuronal circuits in physiology and diseases. Yet, interpretation of cellular and behavioral responses following in vivo optogenetic manipulation of brain activities in experimental animals often necessitates identification of photoactivated neurons with high spatial resolution. Although tracing expression of immediate early genes (IEGs) provides a convenient approach, neuronal activation is not always followed by specific induction of widely used neuronal activity markers like c-fos, Egr1 and Arc. In this study we performed unilateral optogenetic stimulation of the striatum in freely moving transgenic mice that expressed a channelrhodopsin-2 (ChR2) variant ChR2(C128S) in striatal medium spiny neurons (MSNs). We found that in vivo blue light stimulation significantly altered electrophysiological activity of striatal neurons and animal behaviors. To identify photoactivated neurons we then analyzed IEG expression patterns using in situ hybridization. Upon light illumination an induction of c-fos was not apparent whereas another neuronal IEG Npas4 was robustly induced in MSNs ipsilaterally. Our results demonstrate that tracing Npas4 mRNA expression following in vivo optogenetic modulation can be an effective tool for reliable and sensitive identification of activated MSNs in the mouse striatum.
Collapse
Affiliation(s)
- Asim K. Bepari
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiromi Sano
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
| | - Nobuaki Tamamaki
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
- Department of Physiological Sciences, Graduate University for Advanced Studies (Sokendai), Okazaki, Japan
| | - Kenji F. Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
- * E-mail: (KT); (HT)
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- PRESTO, Japan Science and Technology Agency (JST), Saitama, Japan
- * E-mail: (KT); (HT)
| |
Collapse
|
98
|
Madsen HB, Brown RM, Lawrence AJ. Neuroplasticity in addiction: cellular and transcriptional perspectives. Front Mol Neurosci 2012; 5:99. [PMID: 23162427 PMCID: PMC3495339 DOI: 10.3389/fnmol.2012.00099] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 10/20/2012] [Indexed: 12/17/2022] Open
Abstract
Drug addiction is a chronic, relapsing brain disorder which consists of compulsive patterns of drug-seeking and taking that occurs at the expense of other activities. The transition from casual to compulsive drug use and the enduring propensity to relapse is thought to be underpinned by long-lasting neuroadaptations in specific brain circuitry, analogous to those that underlie long-term memory formation. Research spanning the last two decades has made great progress in identifying cellular and molecular mechanisms that contribute to drug-induced changes in plasticity and behavior. Alterations in synaptic transmission within the mesocorticolimbic and corticostriatal pathways, and changes in the transcriptional potential of cells by epigenetic mechanisms are two important means by which drugs of abuse can induce lasting changes in behavior. In this review we provide a summary of more recent research that has furthered our understanding of drug-induced neuroplastic changes both at the level of the synapse, and on a transcriptional level, and how these changes may relate to the human disease of addiction.
Collapse
Affiliation(s)
- Heather B Madsen
- Addiction Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health Parkville, VIC, Australia
| | | | | |
Collapse
|
99
|
Addiction-related gene regulation: risks of exposure to cognitive enhancers vs. other psychostimulants. Prog Neurobiol 2012; 100:60-80. [PMID: 23085425 DOI: 10.1016/j.pneurobio.2012.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/02/2012] [Accepted: 10/08/2012] [Indexed: 01/09/2023]
Abstract
The psychostimulants methylphenidate (Ritalin, Concerta), amphetamine (Adderall), and modafinil (Provigil) are widely used in the treatment of medical conditions such as attention-deficit hyperactivity disorder and narcolepsy and, increasingly, as "cognitive enhancers" by healthy people. The long-term neuronal effects of these drugs, however, are poorly understood. A substantial amount of research over the past two decades has investigated the effects of psychostimulants such as cocaine and amphetamines on gene regulation in the brain because these molecular changes are considered critical for psychostimulant addiction. This work has determined in some detail the neurochemical and cellular mechanisms that mediate psychostimulant-induced gene regulation and has also identified the neuronal systems altered by these drugs. Among the most affected brain systems are corticostriatal circuits, which are part of cortico-basal ganglia-cortical loops that mediate motivated behavior. The neurotransmitters critical for such gene regulation are dopamine in interaction with glutamate, while other neurotransmitters (e.g., serotonin) play modulatory roles. This review presents (1) an overview of the main findings on cocaine- and amphetamine-induced gene regulation in corticostriatal circuits in an effort to provide a cellular framework for (2) an assessment of the molecular changes produced by methylphenidate, medical amphetamine (Adderall), and modafinil. The findings lead to the conclusion that protracted exposure to these cognitive enhancers can induce gene regulation effects in corticostriatal circuits that are qualitatively similar to those of cocaine and other amphetamines. These neuronal changes may contribute to the addiction liability of the psychostimulant cognitive enhancers.
Collapse
|
100
|
Chronic hyperdopaminergic activity of schizophrenia is associated with increased ΔFosB levels and cdk-5 signaling in the nucleus accumbens. Neuroscience 2012; 222:124-35. [DOI: 10.1016/j.neuroscience.2012.07.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 07/10/2012] [Accepted: 07/12/2012] [Indexed: 11/18/2022]
|