1
|
Pirino BE, Kelley AM, Karkhanis AN, Barson JR. A critical review of effects on ethanol intake of the dynorphin/kappa opioid receptor system in the extended amygdala: From inhibition to stimulation. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1027-1038. [PMID: 37042026 PMCID: PMC10289127 DOI: 10.1111/acer.15078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/13/2023]
Abstract
The dynorphin (DYN)/kappa opioid receptor (KOR) system has increasingly been investigated as a possible pharmacotherapeutic target for alcohol use disorder, but findings on the direction of its effects have been mixed. Activation of KORs by DYN has been shown to elicit dysphoric effects, and the DYN/KOR system has canonically been considered particularly important in driving alcohol intake through negative reinforcement in dependent states. However, this review also highlights its activity in opposing the positive reinforcement that drives alcohol intake at earlier stages. Both DYN and KORs are concentrated in the extended amygdala, a set of interconnected regions that includes the bed nucleus of the stria terminalis, central nucleus of the amygdala, and nucleus accumbens shell. This review focuses on the role of the DYN/KOR system in the extended amygdala in ethanol use. It begins by examining the effects of ethanol on the expression of DYN/KOR in the extended amygdala, expression of DYN/KOR in alcohol-preferring and alcohol-avoiding animals, and the effects of knocking out DYN/KOR genes on ethanol intake. Then, it examines the effects on ethanol use in both dependent and nondependent states from systemic pharmacological manipulations of DYN/KOR and from specific manipulation of this system in regions of the extended amygdala. We propose that greater expression and binding of DYN/KOR, by reducing the positive reinforcement that drives early stages of intake, initially acts to prevent the escalation of ethanol drinking. However, prolonged, binge-like, or intermittent ethanol intake enhances levels of DYN/KOR in the extended amygdala such that the system ultimately facilitates the negative reinforcement that drives later stages of ethanol drinking. This review highlights the potential of the DYN/KOR system as a target that can affect different outcomes across different stages of ethanol drinking and the development of alcohol use disorder.
Collapse
Affiliation(s)
- Breanne E. Pirino
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, P.A. 19129
| | - Abigail M. Kelley
- Department of Psychology, Binghamton University – SUNY, Binghamton, N.Y. 13902
| | | | - Jessica R. Barson
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, P.A. 19129
| |
Collapse
|
2
|
Alcohol Withdrawal and the Associated Mood Disorders-A Review. Int J Mol Sci 2022; 23:ijms232314912. [PMID: 36499240 PMCID: PMC9738481 DOI: 10.3390/ijms232314912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2022] Open
Abstract
Recreational use of alcohol is a social norm in many communities worldwide. Alcohol use in moderation brings pleasure and may protect the cardiovascular system. However, excessive alcohol consumption or alcohol abuse are detrimental to one's health. Three million deaths due to excessive alcohol consumption were reported by the World Health Organization. Emerging evidence also revealed the danger of moderate consumption, which includes the increased risk to cancer. Alcohol abuse and periods of withdrawal have been linked to depression and anxiety. Here, we present the effects of alcohol consumption (acute and chronic) on important brain structures-the frontal lobe, the temporal lobe, the limbic system, and the cerebellum. Apart from this, we also present the link between alcohol abuse and withdrawal and mood disorders in this review, thus drawing a link to oxidative stress. In addition, we also discuss the positive impacts of some pharmacotherapies used. Due to the ever-rising demands of life, the cycle between alcohol abuse, withdrawal, and mood disorders may be a never-ending cycle of destruction. Hence, through this review, we hope that we can emphasise the importance and urgency of managing this issue with the appropriate approaches.
Collapse
|
3
|
Demery-Poulos C, Chambers JM. Genetic variation in alcoholism and opioid addiction susceptibility and treatment: a pharmacogenomic approach. AIMS MOLECULAR SCIENCE 2021. [DOI: 10.3934/molsci.2021016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
<abstract>
<p>Alcohol and opioid abuse have pervasive and detrimental consequences from the individual to societal level. The extent of genetic contribution to alcoholism has been studied for decades, yielding speculative and often inconsistent results since the previous discovery of two pharmacokinetic variants strongly protective against alcoholism. The neurobiology of addiction involves innumerate genes with combinatorial and epistatic interactions, creating a difficult landscape for concrete conclusions. In contrast, pharmacogenomic variation in the treatment of alcoholism yields more immediate clinical utility, while also emphasizing pathways crucial to the progression of addiction. An improved understanding of genetic predisposition to alcohol abuse has inherent significance for opioid addiction and treatment, as the two drugs induce the same reward pathway. This review outlines current knowledge, treatments, and research regarding genetic predisposition to alcoholism, focusing on pharmacodynamic variation within the dopaminergic system and shared implications for opioid abuse. Multifaceted and highly polygenic, the phenotype of addiction seems to grow more complex as new research extends the scope of its impact on the brain, body, and progeny.</p>
</abstract>
Collapse
|
4
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
5
|
Karkhanis AN, Al-Hasani R. Dynorphin and its role in alcohol use disorder. Brain Res 2020; 1735:146742. [PMID: 32114059 DOI: 10.1016/j.brainres.2020.146742] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
The dynorphin / kappa opioid receptor (KOR) system has been implicated in many aspects that influence neuropsychiatric disorders. Namely, this system modulates neural circuits that primarily regulate reward seeking, motivation processing, stress responsivity, and pain sensitivity, thus affecting the development of substance and alcohol use disorder (AUD). The effects of this system are often bidirectional and depend on projection targets. To date, a majority of the studies focusing on this system have examined the KOR function using agonists and antagonists. Indeed, there are studies that have examined prodynorphin and dynorphin levels by measuring mRNA and tissue content levels; however, static levels of the neuropeptide and its precursor do not explain complete and online function of the peptide as would be explained by measuring dynorphin transmission in real time. New and exciting methods using optogenetics, chemogenetics, genetic sensors, fast scan cyclic voltammetry are now being developed to detect various neuropeptides with a focus on opioid peptides, including dynorphin. In this review we discuss studies that examine dynorphin projections in areas involved in AUD, its functional involvement in AUD and vulnerability to develop AUD at various ages. Moreover, we discuss dynorphin's role in promoting AUD by dysregulation motivation circuits and how advancements in opioid peptide detection will further our understanding.
Collapse
Affiliation(s)
- Anushree N Karkhanis
- Department of Psychology, Developmental Exposure Alcohol Research Center, Center for Developmental and Behavioral Neuroscience, Binghamton University - SUNY, 4400 Vestal Parkway East, Binghamton, NY 13902, USA.
| | - Ream Al-Hasani
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, Department of Anesthesiology Washington University in St. Louis, Center for Clinical Pharmacology, Washington University School of Medicine & St. Louis College of Pharmacy 660 S.Euclid, Box 8054, St. Louis, MO 63110, USA.
| |
Collapse
|
6
|
Wille-Bille A, Miranda-Morales RS, Pucci M, Bellia F, D'Addario C, Pautassi RM. Prenatal ethanol induces an anxiety phenotype and alters expression of dynorphin & nociceptin/orphanin FQ genes. Prog Neuropsychopharmacol Biol Psychiatry 2018; 85:77-88. [PMID: 29678771 DOI: 10.1016/j.pnpbp.2018.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/11/2018] [Accepted: 04/15/2018] [Indexed: 11/16/2022]
Abstract
Animal models have suggested that prenatal ethanol exposure (PEE) alters the κ opioid receptor system. The present study investigated the brain expression of dynorphin and nociceptin/orphanin FQ related genes and assessed anxiety-like behavior in the light-dark box (LDB), shelter-seeking and risk-taking behaviors in the concentric square field (CSF) test, and ethanol-induced locomotion in the open field (OF), in infant or adolescent Wistar rats that were exposed to PEE (0.0 or 2.0 g/kg, intragastrically, gestational days 17-20). We measured brain mRNA levels of prodynorphin (PDYN), κ opioid receptors (KOR), the nociceptin/orphanin FQ opioid peptide precursor prepronociceptin (ppN/OFQ) and nociceptine/orphanin FQ receptors (NOR). Prenatal ethanol exposure upregulated PDYN and KOR mRNA levels in the ventral tegmental area (VTA) in infant and adolescent rats and KOR mRNA levels in the prefrontal cortex in infant rats. The changes in gene expression in the VTA were accompanied by a reduction of DNA methylation at the PDYN gene promoter, and by a reduction of DNA methylation at the KOR gene promoter. The PEE-induced upregulation of PDYN/KOR in the VTA was accompanied by lower NOR gene expression in the VTA, and lower PDYN gene expression in the nucleus accumbens. PEE rats exhibited hypolocomotion in the OF, greater avoidance of the white and brightly lit areas in the LDB and CSF, and greater preference for the sheltered area in the CSF test. These results suggest that PEE upregulates the dynorphin system, resulting in an anxiety-prone phenotype and triggering compensatory responses in the nociceptin/orphanin FQ system. These findings may help elucidate the mechanisms that underlie the effects of PEE and suggest that the dynorphin and nociceptin/orphanin FQ systems may be possible targets for the prevention and treatment of PEE-induced alterations.
Collapse
Affiliation(s)
- Aranza Wille-Bille
- Instituto de Investigación Médica M. y M. Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba C.P. 5000, Argentina
| | - Roberto Sebastián Miranda-Morales
- Instituto de Investigación Médica M. y M. Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba C.P. 5000, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | | | - Claudio D'Addario
- Università degli Studi di Teramo, Teramo, Italy; Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| | - Ricardo Marcos Pautassi
- Instituto de Investigación Médica M. y M. Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba C.P. 5000, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
7
|
Lundberg S, Abelson KSP, Nylander I, Roman E. Few long-term consequences after prolonged maternal separation in female Wistar rats. PLoS One 2017; 12:e0190042. [PMID: 29267376 PMCID: PMC5739456 DOI: 10.1371/journal.pone.0190042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 12/07/2017] [Indexed: 01/21/2023] Open
Abstract
Environmental factors during the early-life period are known to have long-term consequences for the adult phenotype. An intimate interplay between genes and environment shape the individual and may affect vulnerability for psychopathology in a sex-dependent manner. A rodent maternal separation model was here used to study the long-term effects of different early-life rearing conditions on adult behavior, HPA axis activity and long-term voluntary alcohol intake in female rats. Litters were subjected to 15 min (MS15) or 360 min (MS360) of daily maternal separation during postnatal day 1–21. In adulthood, the behavioral profiles were investigated using the multivariate concentric square field™ (MCSF) test or examined for HPA axis reactivity by cat-odor exposure with subsequent characterization of voluntary alcohol intake and associated changes in HPA axis activity. Adult female MS360 offspring showed mostly no, or only minor, effects on behavior, HPA axis reactivity and long-term alcohol intake relative to MS15. Instead, more pronounced effects were found dependent on changes in the natural hormonal cycle or by the choice of animal supplier. However, changes were revealed in corticosterone load after long-term alcohol access, as females subjected to MS360 had higher concentrations of fecal corticosterone. The present findings are in line with and expand on previous studies on the long-term effects of maternal separation in female rats with regard to behavior, HPA axis activity and voluntary alcohol intake. It can also be a window into further studies detailing how early-life experiences interact with other risk and protective factors to impact the adult phenotype and how possible sex differences play a role.
Collapse
Affiliation(s)
- Stina Lundberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
- * E-mail:
| | - Klas S. P. Abelson
- Department of Experimental Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ingrid Nylander
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Erika Roman
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
8
|
Anderson RI, Becker HC. Role of the Dynorphin/Kappa Opioid Receptor System in the Motivational Effects of Ethanol. Alcohol Clin Exp Res 2017; 41:1402-1418. [PMID: 28425121 DOI: 10.1111/acer.13406] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 04/13/2017] [Indexed: 12/20/2022]
Abstract
Evidence has demonstrated that dynorphin (DYN) and the kappa opioid receptor (KOR) system contribute to various psychiatric disorders, including anxiety, depression, and addiction. More recently, this endogenous opioid system has received increased attention as a potential therapeutic target for treating alcohol use disorders. In this review, we provide an overview and synthesis of preclinical studies examining the influence of alcohol (ethanol [EtOH]) exposure on DYN/KOR expression and function, as well as studies examining the effects of DYN/KOR manipulation on EtOH's rewarding and aversive properties. We then describe work that has characterized effects of KOR activation and blockade on EtOH self-administration and EtOH dependence/withdrawal-related behaviors. Finally, we address how the DYN/KOR system may contribute to stress-EtOH interactions. Despite an apparent role for the DYN/KOR system in motivational effects of EtOH, support comes from relatively few studies. Nevertheless, review of this literature reveals several common themes: (i) rodent strains genetically predisposed to consume more EtOH generally appear to have reduced DYN/KOR tone in brain reward circuitry; (ii) acute and chronic EtOH exposure typically up-regulate the DYN/KOR system; (iii) KOR antagonists reduce behavioral indices of negative affect associated with stress and chronic EtOH exposure/withdrawal; and (iv) KOR antagonists are effective in reducing EtOH consumption, but are often more efficacious under conditions that engender high levels of consumption, such as dependence or stress exposure. These results support the contention that the DYN/KOR system plays a significant role in contributing to dependence- and stress-induced elevation in EtOH consumption. Overall, more comprehensive analyses (on both behavioral and mechanistic levels) are needed to provide additional insight into how the DYN/KOR system is engaged and adapts to influence the motivation effects of EtOH. This information will be critical for the development of new pharmacological agents targeting KORs as promising novel therapeutics for alcohol use disorders and comorbid affective disorders.
Collapse
Affiliation(s)
- Rachel I Anderson
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Howard C Becker
- Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina.,Department of Neuroscience , Medical University of South Carolina, Charleston, South Carolina.,RHJ Department of Veterans Affairs Medical Center , Charleston, South Carolina
| |
Collapse
|
9
|
Uhari-Väänänen J, Raasmaja A, Bäckström P, Oinio V, Airavaara M, Piepponen P, Kiianmaa K. Accumbal μ-Opioid Receptors Modulate Ethanol Intake in Alcohol-Preferring Alko Alcohol Rats. Alcohol Clin Exp Res 2016; 40:2114-2123. [PMID: 27508965 DOI: 10.1111/acer.13176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 07/09/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND The nucleus accumbens shell is a key brain area mediating the reinforcing effects of ethanol (EtOH). Previously, it has been shown that the density of μ-opioid receptors in the nucleus accumbens shell is higher in alcohol-preferring Alko Alcohol (AA) rats than in alcohol-avoiding Alko Non-Alcohol rats. In addition, EtOH releases opioid peptides in the nucleus accumbens and opioid receptor antagonists are able to modify EtOH intake, all suggesting an opioidergic mechanism in the control of EtOH consumption. As the exact mechanisms of opioidergic involvement remains to be elucidated, the aim of this study was to clarify the role of accumbal μ- and κ-opioid receptors in controlling EtOH intake in alcohol-preferring AA rats. METHODS Microinfusions of the μ-opioid receptor antagonist CTOP (0.3 and 1 μg/site), μ-opioid receptor agonist DAMGO (0.03 and 0.1 μg/site), nonselective opioid receptor agonist morphine (30 μg/site), and κ-opioid receptor agonist U50488H (0.3 and 1 μg/site) were administered via bilateral guide cannulas into the nucleus accumbens shell of AA rats that voluntarily consumed 10% EtOH solution in an intermittent, time-restricted (90-minute) 2-bottle choice access paradigm. RESULTS CTOP (1 μg/site) significantly increased EtOH intake. Conversely, DAMGO resulted in a decreasing trend in EtOH intake. Neither morphine nor U50488H had any effect on EtOH intake in the used paradigm. CONCLUSIONS The results provide further evidence for the role of accumbens shell μ-opioid receptors but not κ-opioid receptors in mediating reinforcing effects of EtOH and in regulating EtOH consumption. The results also provide support for views suggesting that the nucleus accumbens shell has a major role in mediating EtOH reward.
Collapse
Affiliation(s)
- Johanna Uhari-Väänänen
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland. .,Department of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| | - Atso Raasmaja
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Pia Bäckström
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Ville Oinio
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland.,Department of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Petteri Piepponen
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Kalervo Kiianmaa
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
10
|
Alongkronrusmee D, Chiang T, van Rijn RM. Delta Opioid Pharmacology in Relation to Alcohol Behaviors. Handb Exp Pharmacol 2016; 247:199-225. [PMID: 27316912 DOI: 10.1007/164_2016_30] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Delta opioid receptors (DORs) are heavily involved in alcohol-mediated processes in the brain. In this chapter we provide an overview of studies investigating how alcohol directly impacts DOR pharmacology and of early studies indicating DOR modulation of alcohol behavior. We will offer a brief summary of the different animal species used in alcohol studies investigating DORs followed by a broader overview of the types of alcohol behaviors modulated by DORs. We will highlight a small set of studies investigating the relationship between alcohol and DORs in analgesia. We will then provide an anatomical overview linking DOR expression in specific brain regions to different alcohol behaviors. In this section, we will provide two models that try to explain how endogenous opioids acting at DORs may influence alcohol behaviors. Next, we will provide an overview of studies investigating certain new aspects of DOR pharmacology, including the formation of heteromers and biased signaling. Finally, we provide a short overview of the genetics of the DORs in relation to alcohol use disorders (AUDs) and a short statement on the potential of using DOR-based therapeutics for treatment of AUDs.
Collapse
Affiliation(s)
- Doungkamol Alongkronrusmee
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Terrance Chiang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
11
|
Sarkisyan D, Hussain MZ, Watanabe H, Kononenko O, Bazov I, Zhou X, Yamskova O, Krishtal O, Karpyak VM, Yakovleva T, Bakalkin G. Downregulation of the endogenous opioid peptides in the dorsal striatum of human alcoholics. Front Cell Neurosci 2015; 9:187. [PMID: 26029055 PMCID: PMC4428131 DOI: 10.3389/fncel.2015.00187] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 04/28/2015] [Indexed: 01/18/2023] Open
Abstract
The endogenous opioid peptides dynorphins and enkephalins may be involved in brain-area specific synaptic adaptations relevant for different stages of an addiction cycle. We compared the levels of prodynorphin (PDYN) and proenkephalin (PENK) mRNAs (by qRT-PCR), and dynorphins and enkephalins (by radioimmunoassay) in the caudate nucleus and putamen between alcoholics and control subjects. We also evaluated whether PDYN promoter variant rs1997794 associated with alcoholism affects PDYN expression. Postmortem specimens obtained from 24 alcoholics and 26 controls were included in final statistical analysis. PDYN mRNA and Met-enkephalin-Arg-Phe, a marker of PENK were downregulated in the caudate of alcoholics, while PDYN mRNA and Leu-enkephalin-Arg, a marker of PDYN were decreased in the putamen of alcoholics carrying high risk rs1997794 C allele. Downregulation of opioid peptides in the dorsal striatum may contribute to development of alcoholism including changes in goal directed behavior and formation of a compulsive habit in alcoholics.
Collapse
Affiliation(s)
- Daniil Sarkisyan
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden
| | | | - Hiroyuki Watanabe
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden
| | - Olga Kononenko
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden ; Department for Cellular Membranology, Bogomoletz Institute of Physiology Kyiv, Ukraine
| | - Igor Bazov
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden
| | - Xingwu Zhou
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden
| | - Olga Yamskova
- Department of Functional Pharmacology, Institute for Neuroscience, Uppsala University Uppsala, Sweden
| | - Oleg Krishtal
- Department for Cellular Membranology, Bogomoletz Institute of Physiology Kyiv, Ukraine
| | - Victor M Karpyak
- Department of Psychiatry and Psychology, Mayo Clinic Rochester, MN, USA
| | - Tatiana Yakovleva
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden
| | - Georgy Bakalkin
- Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University Uppsala, Sweden
| |
Collapse
|
12
|
Barker JM, Taylor JR. Habitual alcohol seeking: modeling the transition from casual drinking to addiction. Neurosci Biobehav Rev 2014; 47:281-94. [PMID: 25193245 PMCID: PMC4258136 DOI: 10.1016/j.neubiorev.2014.08.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 07/17/2014] [Accepted: 08/25/2014] [Indexed: 12/29/2022]
Abstract
The transition from goal-directed actions to habitual ethanol seeking models the development of addictive behavior that characterizes alcohol use disorders. The progression to habitual ethanol-seeking behavior occurs more rapidly than for natural rewards, suggesting that ethanol may act on habit circuit to drive the loss of behavioral flexibility. This review will highlight recent research that has focused on the formation and expression of habitual ethanol seeking, and the commonalities and distinctions between ethanol and natural reward-seeking habits, with the goal of highlighting important, understudied research areas that we believe will lead toward the development of novel treatment and prevention strategies for uncontrolled drinking.
Collapse
Affiliation(s)
- Jacqueline M Barker
- Department of Psychiatry, Yale University School of Medicine, Ribicoff Labs, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Jane R Taylor
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
13
|
Faisal M, Waseem D, Ismatullah H, Taqi MM. A molecular prospective provides new insights into implication of PDYN and OPRK1 genes in alcohol dependence. Comput Biol Med 2014; 53:250-7. [PMID: 25177835 DOI: 10.1016/j.compbiomed.2014.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 07/24/2014] [Accepted: 07/26/2014] [Indexed: 12/15/2022]
Abstract
Single nucleotide polymorphisms (SNPs) both in coding and non-coding regions govern gene functions prompting differential vulnerability to diseases, heterogeneous response to pharmaceutical regimes and environmental anomalies. These genetic variations, SNPs, may alter an individual׳s susceptibility for alcohol dependence by remodeling DNA-protein interaction patterns in prodynorphin (PDYN) and the κ-opioid receptor (OPRK1) genes. In order to elaborate the underlying molecular mechanism behind these susceptibility differences we used bioinformatics tools to retrieve differential DNA-protein interactions at PDYN and OPRK1 SNPs significantly associated with alcohol dependence. Our results show allele-specific DNA-protein interactions depicting allele-specific mechanisms implicated in differential regulation of gene expression. Several transcription factors, for instance, VDR, RXR-alpha, NFYA, CTF family, USF-1, USF2, ER, AR and predominantly SP family show an allele-specific binding affinity with PDYN gene; likewise, GATA, TBP, AP-1, USF-2, C/EBPbeta, Cart-1 and ER interact with OPRK1 SNPs on intron 2 in an allele-specific manner. In a nutshell, transition of a single nucleotide may modify differential DNA-protein interactions at OPRK1 and PDYN׳s SNPs, significantly associated with pathology that may lead to altered individual vulnerability for alcohol dependence.
Collapse
Affiliation(s)
- Muhammad Faisal
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Durdana Waseem
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Humaira Ismatullah
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Malik Mumtaz Taqi
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
14
|
Darlington TM, Ehringer MA, Larson C, Phang TL, Radcliffe RA. Transcriptome analysis of Inbred Long Sleep and Inbred Short Sleep mice. GENES BRAIN AND BEHAVIOR 2013; 12:263-74. [PMID: 23433184 DOI: 10.1111/gbb.12018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 11/26/2012] [Accepted: 12/27/2012] [Indexed: 11/28/2022]
Abstract
Many studies have utilized the Inbred Long Sleep and Inbred Short Sleep mouse strains to model the genetic influence on initial sensitivity to ethanol. The mechanisms underlying this divergent phenotype are still not completely understood. In this study, we attempt to identify genes that are differentially expressed between these two strains and to identify baseline networks of co-expressed genes, which may provide insight regarding their phenotypic differences. We examined the whole brain and striatal transcriptomes of both strains, using next generation RNA sequencing techniques. Many genes were differentially expressed between strains, including several in chromosomal regions previously shown to influence initial sensitivity to ethanol. These results are in concordance with a similar sample of striatal transcriptomes measured using microarrays. In addition to the higher dynamic range, RNA-Seq is not hindered by high background noise or polymorphisms in probesets as with microarray technology, and we are able to analyze exome sequence of abundant genes. Furthermore, utilizing Weighted Gene Co-expression Network Analysis, we identified several modules of co-expressed genes corresponding to strain differences. Several candidate genes were identified, including protein phosphatase 1 regulatory unit 1b (Ppp1r1b), prodynorphin (Pdyn), proenkephalin (Penk), ras association (RalGDS/AF-6) domain family member 2 (Rassf2), myosin 1d (Myo1d) and transthyretin (Ttr). In addition, we propose a role for potassium channel activity as well as map kinase signaling in the observed phenotypic differences between the two strains.
Collapse
Affiliation(s)
- T M Darlington
- Institute for Behavioral Genetics, University of Colorado, Boulder, Boulder, CO 80309, USA.
| | | | | | | | | |
Collapse
|
15
|
Morales M, Anderson RI, Spear LP, Varlinskaya EI. Effects of the kappa opioid receptor antagonist, nor-binaltorphimine, on ethanol intake: impact of age and sex. Dev Psychobiol 2013; 56:700-12. [PMID: 23754134 DOI: 10.1002/dev.21137] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 04/16/2013] [Indexed: 12/27/2022]
Abstract
The kappa opioid receptor (KOR) antagonist, nor-binaltorphimine (nor-BNI), was used to investigate the role of the KOR system in mediating ethanol intake. On P25 (adolescent) or P67 (adult) male and female rats were individually housed and given ad libitum access to food and water. The experimental procedure was initiated on P28 or P70: animals were given 30 min/day access to a 10% ethanol/supersaccharin solution every other day (3 baseline exposures). On the day after the final baseline test, rats were injected with nor-BNI (0, 2.5, 5, 10 mg/kg), with testing initiated 24 hr later (30-min access every other day, 3 test exposures). Nor-BNI (10 mg/kg) increased ethanol intake in adult males, whereas the same dose decreased intake in adult females, suggesting pronounced sex differences in KOR-associated mediation of ethanol intake in adulthood. There was no impact of nor-BNI in adolescent animals of either sex, suggesting that the KOR may play less of a role in modulating ethanol intake during adolescence.
Collapse
Affiliation(s)
- Melissa Morales
- Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, P.O. Box 6000, Binghamton, New York, 13902-6000.
| | | | | | | |
Collapse
|
16
|
Bell RL, Sable HJ, Colombo G, Hyytia P, Rodd ZA, Lumeng L. Animal models for medications development targeting alcohol abuse using selectively bred rat lines: neurobiological and pharmacological validity. Pharmacol Biochem Behav 2012; 103:119-55. [PMID: 22841890 PMCID: PMC3595005 DOI: 10.1016/j.pbb.2012.07.007] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 07/07/2012] [Accepted: 07/18/2012] [Indexed: 02/04/2023]
Abstract
The purpose of this review paper is to present evidence that rat animal models of alcoholism provide an ideal platform for developing and screening medications that target alcohol abuse and dependence. The focus is on the 5 oldest international rat lines that have been selectively bred for a high alcohol-consumption phenotype. The behavioral and neurochemical phenotypes of these rat lines are reviewed and placed in the context of the clinical literature. The paper presents behavioral models for assessing the efficacy of pharmaceuticals for the treatment of alcohol abuse and dependence in rodents, with particular emphasis on rats. Drugs that have been tested for their effectiveness in reducing alcohol/ethanol consumption and/or self-administration by these rat lines and their putative site of action are summarized. The paper also presents some current and future directions for developing pharmacological treatments targeting alcohol abuse and dependence.
Collapse
Affiliation(s)
- Richard L. Bell
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Helen J.K. Sable
- Department of Psychology, University of Memphis, Memphis, Tennessee, USA
| | - Giancarlo Colombo
- Neuroscience Institute, National Research Council of Italy, Section of Cagliari, Monserrato, Italy
| | - Petri Hyytia
- Institute of Biomedicine, University of Helsinki, Finland
| | - Zachary A. Rodd
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lawrence Lumeng
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
17
|
Nylander I, Roman E. Neuropeptides as mediators of the early-life impact on the brain; implications for alcohol use disorders. Front Mol Neurosci 2012; 5:77. [PMID: 22783165 PMCID: PMC3389713 DOI: 10.3389/fnmol.2012.00077] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/14/2012] [Indexed: 12/22/2022] Open
Abstract
The brain is constantly exposed to external and internal input and to function in an ever-changing environment we are dependent on processes that enable the brain to adapt to new stimuli. Exposure to postnatal environmental stimuli can interfere with vital adaption processes and cause long-term changes in physiological function and behavior. Early-life alterations in brain function may result in impaired ability to adapt to new situations, in altered sensitivity to challenges later in life and thereby mediate risk or protection for psychopathology such as alcohol use disorders (AUD). In clinical research the studies of mechanisms, mediators, and causal relation between early environmental factors and vulnerability to AUD are restricted and attempts are made to find valid animal models for studies of the early-life influence on the brain. This review focuses on rodent models and the effects of adverse and naturalistic conditions on peptide networks within the brain and pituitary gland. Importantly, the consequences of alcohol addiction are not discussed but rather neurobiological alterations that can cause risk consumption and vulnerability to addiction. The article reviews earlier results and includes new data and multivariate data analysis with emphasis on endogenous opioid peptides but also oxytocin and vasopressin. These peptides are vital for developmental processes and it is hypothesized that early-life changes in peptide networks may interfere with neuronal processes and thereby contribute the individual vulnerability for AUD. The summarized results indicate a link between early-life rearing conditions, opioids, and ethanol consumption and that the ethanol-induced effects and the treatment with opioid antagonists later in life are dependent on early-life experiences. Endogenous opioids are therefore of interest to further study in the early-life impact on individual differences in vulnerability to AUD and treatment outcome.
Collapse
Affiliation(s)
- Ingrid Nylander
- Department of Pharmaceutical Biosciences, Neuropharmacology Addiction and Behaviour, Uppsala UniversityUppsala, Sweden
| | | |
Collapse
|
18
|
Palm S, Roman E, Nylander I. Differences in basal and ethanol-induced levels of opioid peptides in Wistar rats from five different suppliers. Peptides 2012; 36:1-8. [PMID: 22564490 DOI: 10.1016/j.peptides.2012.04.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 04/20/2012] [Accepted: 04/20/2012] [Indexed: 11/23/2022]
Abstract
One major cause for discrepancies in results from animal experimental studies is the use of different animal strains and suppliers. We have previously reported that Wistar rats from five different suppliers display profound differences in ethanol intake and behavior. One of the neurobiological processes that could be underlying these differences is the endogenous opioid system, which has been implicated in the rewarding and reinforcing effects of alcohol. We therefore hypothesized that the differences between the supplier groups would also be evident in the endogenous opioid system. Radioimmunoassay was used to determine the levels of the opioid peptides Met-enkephalin-Arg(6)Phe(7) and dynorphin B in several brain areas of ethanol-drinking and ethanol naïve Wistar rats from five different suppliers. In the ethanol naïve animals, differences between the supplier groups were found in the pituitary gland, hypothalamus, frontal cortex, dorsal striatum and hippocampus. In the ethanol-drinking rats, differences were found in the same structures, with the addition of medial prefrontal cortex and substantia nigra. Correlations between ethanol intake and peptide levels were also found in several of the areas examined. The structures in which differences were found have all been implicated in the transition from drug use to addiction and these differences may lead to different propensities and vulnerability to this transition. Because the endogenous opioids have been suggested to be involved in a number of neurobiological disorders the results do not only have implications for research on alcohol or drug addiction, but many other fields as well.
Collapse
Affiliation(s)
- Sara Palm
- Department of Pharmaceutical Biosciences, Division of Neuropharmacology, Addiction & Behavior, Uppsala University, Uppsala, Sweden.
| | | | | |
Collapse
|
19
|
Acute ethanol administration differentially alters enkephalinase and aminopeptidase N activity and mRNA levels in regions of the nigrostriatal pathway. J Mol Neurosci 2012; 49:289-300. [PMID: 22688357 DOI: 10.1007/s12031-012-9823-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 05/28/2012] [Indexed: 10/28/2022]
Abstract
Opioid peptides play a key role in ethanol reinforcement and may also represent important determinants in brain sensitivity to ethanol through modulation of nigrostriatal dopaminergic activity. Regulation of opioid levels by peptidase-degrading enzymes could be relevant in ethanol's actions. The aim of this work was to study the acute ethanol (2.5 g/kg) effects on the activity and mRNA expression of enkephalinase (NEP) and aminopeptidase N (APN) in the rat substantia nigra (SN) and the anterior-medial (amCP) and medial-posterior (mpCP) regions of the caudate-putamen (CP). Enzymatic activities were measured by fluorometric assays and mRNA expression by reverse transcriptase polymerase chain reaction. Acute ethanol administration differentially altered peptidase activities and mRNA expression with different kinetics. Ethanol increased and decreased NEP mRNA levels in the SN and amCP, respectively, but produced biphasic effects in the mpCP. APN mRNA levels were increased by ethanol in all brain regions. Ethanol induced a transient and long-lasting increase in NEP (mpCP) and APN (amCP) activities, respectively. Peptidase activities were not changed by ethanol in the SN. Our results indicate that striatal NEP and APN are important ethanol targets. Ethanol-induced changes in these neuropeptidases in the CP could contribute to the mechanisms involved in brain sensitivity to ethanol.
Collapse
|
20
|
Femenía T, Manzanares J. Increased ethanol intake in prodynorphin knockout mice is associated to changes in opioid receptor function and dopamine transmission. Addict Biol 2012; 17:322-37. [PMID: 21966993 DOI: 10.1111/j.1369-1600.2011.00378.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The purpose of this study was to examine the role of the prodynorphin gene in alcohol sensitivity, preference and vulnerability to alcohol consumption. Handling-induced convulsion (HIC) associated to alcohol, alcohol-induced loss of righting reflex (LORR), hypothermic effects in response to acute ethanol challenge, blood ethanol levels (BELs), conditioned place preference, voluntary ethanol consumption and preference, tyrosine hydroxylase (TH), dopamine transporter (DAT) and proenkephalin (PENK) gene expression, and µ-, δ- and κ-opioid agonist-stimulated [S(35) ]- guanosine 5'-triphosphate-binding autoradiography were studied in prodynorphin knockout (PDYN KO) and wild-type (WT) mice. There were no differences in HIC, LORR or the decrease in body temperature in response to acute ethanol challenge between PDYN KO and WT mice. PDYN KO mice presented higher BEL, higher ethanol-conditioned place preference and more ethanol consumption and preference in a two-bottle choice paradigm than WT mice. These findings were associated with lower TH and higher DAT gene expression in the ventral tegmental area and substantia nigra, and with lower PENK gene expression in the caudate-putamen (CPu), accumbens core (AcbC) and accumbens shell (AcbSh) in PDYN KO. The functional activity of the µ-opioid receptor was lower in the CPu, AcbC, AcbSh and cingulate cortex (Cg) of PDYN KO mice. In contrast, δ- and κ-opioid receptor-binding autoradiographies were increased in the CPu and Cg (δ), and in the CPu, AcbC and Cg (κ) of PDYN KO. These results suggest that deletion of the PDYN gene increased vulnerability for ethanol consumption by altering, at least in part, PENK, TH and DAT gene expression, and µ-, δ- and κ-opioid receptor functional activity in brain areas closely related to ethanol reinforcement.
Collapse
Affiliation(s)
- Teresa Femenía
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | | |
Collapse
|
21
|
Barson JR, Morganstern I, Leibowitz SF. Neurobiology of consummatory behavior: mechanisms underlying overeating and drug use. ILAR J 2012; 53:35-58. [PMID: 23520598 PMCID: PMC3954603 DOI: 10.1093/ilar.53.1.35] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Consummatory behavior is driven by both caloric and emotional need, and a wide variety of animal models have been useful in research on the systems that drive consumption of food and drugs. Models have included selective breeding for a specific trait, manipulation of gene expression, forced or voluntary exposure to a substance, and identification of biomarkers that predict which animals are prone to overconsuming specific substances. This research has elucidated numerous brain areas and neurochemicals that drive consummatory behavior. Although energy homeostasis is primarily mediated by the hypothalamus, reinforcement is more strongly mediated by nuclei outside the hypothalamus, in mesocorticolimbic regions. Orexigenic neurochemicals that control food intake can provide a general signal for promoting caloric intake or a more specific signal for stimulating consumption of a particular macronutrient, fat, carbohydrate, or protein. The neurochemicals involved in controlling fat ingestion--galanin, enkephalin, orexin, melanin-concentrating hormone, and the endocannabinoids--show positive feedback with this macronutrient, as these peptides both increase fat intake and are further stimulated by its intake. This positive association offers some explanation for why foods high in fat are so often overconsumed. Consumption of ethanol, a drug of abuse that also contains calories, is similarly driven by the neurochemical systems involved in fat intake, according to evidence that closely relates fat and ethanol consumption. Further understanding of the systems involved in consummatory behavior will enable the development of effective therapies for the treatment of both overeating and drug abuse.
Collapse
Affiliation(s)
- Jessica R Barson
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, New York 10065, USA
| | | | | |
Collapse
|
22
|
Kemppainen H, Raivio N, Suo-Yrjo V, Kiianmaa K. Opioidergic Modulation of Ethanol Self-Administration in the Ventral Pallidum. Alcohol Clin Exp Res 2011; 36:286-93. [DOI: 10.1111/j.1530-0277.2011.01611.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
23
|
Méndez M, Barbosa-Luna IG, Pérez-Luna JM, Cupo A, Oikawa J. Effects of acute ethanol administration on methionine-enkephalin expression and release in regions of the rat brain. Neuropeptides 2010; 44:413-20. [PMID: 20605629 DOI: 10.1016/j.npep.2010.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 04/15/2010] [Accepted: 05/08/2010] [Indexed: 10/19/2022]
Abstract
The dopaminergic mesolimbic system plays a key role in mediating the reinforcing properties of ethanol and other drugs of abuse. Ethanol reinforcement and high alcohol drinking behaviour have been suggested to involve the ethanol-induced activation of endogenous opioid systems. Ethanol may alter opioidergic transmission at different levels, including opioid peptide biosynthesis and release, as well as binding to opioid receptors. The aim of this work was to investigate the effects of different ethanol doses on methionine-enkephalin (Met-enk) release from the rat nucleus accumbens (NAcc). Ethanol effects were also studied on Met-enk content in the NAcc, prefrontal cortex (PFC) and caudate-putamen (CP). Met-enk release was studied by microdialysis in Wistar anesthetized rats and peptide concentrations were quantitated by radioimmunoassay. Ethanol was administered by intraperitoneal injection after a 2-h basal release period. Ethanol doses of 0.5, 1 and 2.5 g/kg induced a 2.7-, 4.9- and 3.4-fold increase in Met-enk release from the NAcc. However, ethanol responses followed different kinetics, with earliest effects observed with the highest ethanol dose. In comparison, a 2.5-fold increase in peptide release was produced by 100 mM KCl. Ethanol, at a dose of 2.5 g/kg, induced a significant 66.7% decrease in Met-enk content in the NAcc, as well as a 76.4% reduction in peptide levels in the CP. Lower ethanol doses did not alter Met-enk content in these regions. On the other hand, an ethanol dose of 0.5 g/kg produced a non-significant decrease in Met-enk levels in the PFC. Our results suggest that ethanol-induced changes in enkephalin expression and release in regions of the mesocorticolimbic and nigrostriatal pathways could be involved in ethanol central effects. Released enkephalins by ethanol may modulate the dopaminergic activity of mesolimbic neurons and play a critical role in ethanol reinforcement mechanisms.
Collapse
Affiliation(s)
- M Méndez
- Departamento de Neuroquímica, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente, Calzada México Xochimilco 101, Col. San Lorenzo Huipulco, 14370 México D.F., Mexico.
| | | | | | | | | |
Collapse
|
24
|
Panula P, Nuutinen S. Histamine and H3 receptor in alcohol-related behaviors. J Pharmacol Exp Ther 2010; 336:9-16. [PMID: 20864504 DOI: 10.1124/jpet.110.170928] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Data from rat models for alcohol preference and histidine decarboxylase knockout (HDC KO) mice suggest that brain histamine regulates alcohol-related behaviors. Histamine levels are higher in alcohol-preferring than in alcohol-nonpreferring rat brains, and expression of histamine H(3) receptor (H(3)R) is different in key areas for addictive behavior. H(3)R inverse agonists decrease alcohol responding in one alcohol-preferring rat line. Conditioned place preference induced by alcohol is stronger in HDC KO mice than in control mice. The HDC KO mice display a weaker stimulatory response to acute alcohol than the wild-type (WT) mice. In male inbred C57BL/6 mice the H(3)R antagonist ciproxifan inhibits ethanol-evoked stimulation of locomotor activity. Ciproxifan also potentiates the ethanol reward, but does not alone result in the development of place preference. At least in one rat model developed to study alcohol sensitivity high histamine levels are characteristic of the alcohol-insensitive rat line, and lowering brain histamine with a HDC inhibitor increases alcohol sensitivity in the tilting plane test. However, the motor skills of HDC KO mice do not seem to differ from those of the WT mice. Current evidence suggests that the histaminergic system is involved in the regulation of place preference behavior triggered by alcohol, possibly through an interaction with the mesolimbic dopamine system. Histamine may also interact with dopamine in the regulation of the cortico-striato-pallido-thalamo-cortical motor pathway and cerebellar mechanisms, which may be important in different motor behaviors beyond alcohol-induced motor disturbances. H(3)R ligands may have significant effects on alcohol addiction.
Collapse
Affiliation(s)
- Pertti Panula
- Neuroscience Center and Institute of Biomedicine/Anatomy, POB 63, FI-00014, University of Helsinki, Helsinki, Finland.
| | | |
Collapse
|
25
|
Kim DH, Schwartz CE. The genetics of pain: implications for evaluation and treatment of spinal disease. Spine J 2010; 10:827-40. [PMID: 20615760 DOI: 10.1016/j.spinee.2010.05.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 03/07/2010] [Accepted: 05/22/2010] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Variability in human pain experience appears to be at least partially determined by genetic inheritance. To the extent that awareness of individual pain sensitivity and the tendency to develop chronic pain after injury or surgery would be informative for clinical decision making, development and use of genetic testing for specific pain markers could contribute to improved outcomes in management of spinal disease. PURPOSE To review important and illustrative results from both classical and modern pain genetics studies and to introduce readers to critical definitions and concepts necessary to interpret the growing body of genetics literature relevant to spinal disease. STUDY DESIGN/SETTING Literature review and commentary. METHODS A review was performed of published English language studies in which genetic techniques were used to analyze the molecular basis of nociceptive signaling or processing with a particular emphasis on studies addressing genetic determinants of interindividual variability in pain sensitivity or predisposition to chronic pain. RESULTS There is compelling evidence indicating that interindividual differences in pain sensitivity and the risk of developing chronic pain syndromes are genetically determined. Despite a growing list of putative "pain genes," genetic association studies remain plagued with difficulty replicating initial findings in different cohorts. CONCLUSIONS Genome-wide association studies are potentially powerful means of identifying clinically relevant genetic markers predicting disease susceptibility, severity, and treatment response. However, accurate results require rigorous study design with use of large homogeneous populations and precise phenotypes.
Collapse
Affiliation(s)
- David H Kim
- Department of Orthopaedic Surgery, Tufts University Medical School, New England Baptist Hospital, Boston, MA 02120, USA.
| | | |
Collapse
|
26
|
Forebrain PENK and PDYN gene expression levels in three inbred strains of mice and their relationship to genotype-dependent morphine reward sensitivity. Psychopharmacology (Berl) 2010; 208:291-300. [PMID: 19997907 DOI: 10.1007/s00213-009-1730-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 11/10/2009] [Indexed: 12/18/2022]
Abstract
RATIONALE Vulnerability to drug abuse disorders is determined not only by environmental but also by genetic factors. A body of evidence suggests that endogenous opioid peptide systems may influence rewarding effects of addictive substances, and thus, their individual expression levels may contribute to drug abuse liability. OBJECTIVES The aim of our study was to assess whether basal genotype-dependent brain expression of opioid propeptides genes can influence sensitivity to morphine reward. METHODS Experiments were performed on inbred mouse strains C57BL/6J, DBA/2J, and SWR/J, which differ markedly in responses to morphine administration: DBA/2J and SWR/J show low and C57BL/6J high sensitivity to opioid reward. Proenkephalin (PENK) and prodynorphin (PDYN) gene expression was measured by in situ hybridization in brain regions implicated in addiction. The influence of the kappa opioid receptor antagonist nor-binaltorphimine (nor-BNI), which attenuates effects of endogenous PDYN-derived peptides, on rewarding actions of morphine was studied using the conditioned place preference (CPP) paradigm. RESULTS DBA/2J and SWR/J mice showed higher levels of PDYN and lower levels of PENK messenger RNA in the nucleus accumbens than the C57BL/6J strain. Pretreatment with nor-BNI enhanced morphine-induced CPP in the opioid-insensitive DBA/2J and SWR/J strains. CONCLUSIONS Our results demonstrate that inter-strain differences in PENK and PDYN genes expression in the nucleus accumbens parallel sensitivity of the selected mouse strains to rewarding effects of morphine. They suggest that high expression of PDYN may protect against drug abuse by limiting drug-produced reward, which may be due to dynorphin-mediated modulation of dopamine release in the nucleus accumbens.
Collapse
|
27
|
Jarjour S, Bai L, Gianoulakis C. Effect of Acute Ethanol Administration on the Release of Opioid Peptides From the Midbrain Including the Ventral Tegmental Area. Alcohol Clin Exp Res 2009; 33:1033-43. [DOI: 10.1111/j.1530-0277.2009.00924.x] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
28
|
Spanagel R. Alcoholism: A Systems Approach From Molecular Physiology to Addictive Behavior. Physiol Rev 2009; 89:649-705. [DOI: 10.1152/physrev.00013.2008] [Citation(s) in RCA: 491] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Alcohol consumption is an integral part of daily life in many societies. The benefits associated with the production, sale, and use of alcoholic beverages come at an enormous cost to these societies. The World Health Organization ranks alcohol as one of the primary causes of the global burden of disease in industrialized countries. Alcohol-related diseases, especially alcoholism, are the result of cumulative responses to alcohol exposure, the genetic make-up of an individual, and the environmental perturbations over time. This complex gene × environment interaction, which has to be seen in a life-span perspective, leads to a large heterogeneity among alcohol-dependent patients, in terms of both the symptom dimensions and the severity of this disorder. Therefore, a reductionistic approach is not very practical if a better understanding of the pathological processes leading to an addictive behavior is to be achieved. Instead, a systems-oriented perspective in which the interactions and dynamics of all endogenous and environmental factors involved are centrally integrated, will lead to further progress in alcohol research. This review adheres to a systems biology perspective such that the interaction of alcohol with primary and secondary targets within the brain is described in relation to the behavioral consequences. As a result of the interaction of alcohol with these targets, alterations in gene expression and synaptic plasticity take place that lead to long-lasting alteration in neuronal network activity. As a subsequent consequence, alcohol-seeking responses ensue that can finally lead via complex environmental interactions to an addictive behavior.
Collapse
|
29
|
Delta-opioid receptor expression in the ventral tegmental area protects against elevated alcohol consumption. J Neurosci 2009; 28:12672-81. [PMID: 19036960 DOI: 10.1523/jneurosci.4569-08.2008] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Alcoholism is a complex and debilitating syndrome affecting approximately 140 million people worldwide. However, not everyone who consumes ethanol develops abuse, raising the possibility that some individuals have a protective mechanism that inhibits elevated alcohol consumption. We tested the hypothesis that the delta-opioid receptor (DOR) plays such a protective role. Here we show that DOR activity in the ventral tegmental area (VTA) robustly decreases ethanol consumption in rats and that these effects depend on baseline ethanol consumption. Intra-VTA microinjection of the DOR agonist DPDPE decreases drinking, particularly in low-drinking animals. Furthermore, VTA microinjection of the DOR selective antagonist TIPP-Psi increases drinking in low, but not high, drinkers and this increase is blocked by comicroinjection of the GABA(A) antagonist bicuculline. Using electrophysiological techniques we found that in VTA brain slices from drinking rats DPDPE presynaptically inhibits GABA(A) receptor mediated IPSCs in low drinkers, but not in high drinkers or naive animals, most likely through activation of DORs on GABA terminals. This DOR-mediated inhibition of IPSCs also correlates inversely with behavioral correlates of anxiety measured in the elevated plus maze. In contrast, presynaptic inhibition of VTA GABA(A) IPSCs by the mu-opioid receptor agonist DAMGO is significantly reduced in both high- and low-drinking rats (<30%) compared with age-matched nondrinking controls (>70%). Together, our findings demonstrate the protective nature of VTA DORs and identify an important new target for therapeutic intervention for alcoholism.
Collapse
|
30
|
Mitchell JM, Bergren LJ, Chen KS, Rowbotham MC, Fields HL. Naltrexone aversion and treatment efficacy are greatest in humans and rats that actively consume high levels of alcohol. Neurobiol Dis 2009; 33:72-80. [DOI: 10.1016/j.nbd.2008.09.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 09/10/2008] [Accepted: 09/12/2008] [Indexed: 11/26/2022] Open
|
31
|
Teegarden SL, Nestler EJ, Bale TL. Delta FosB-mediated alterations in dopamine signaling are normalized by a palatable high-fat diet. Biol Psychiatry 2008; 64:941-50. [PMID: 18657800 PMCID: PMC2582592 DOI: 10.1016/j.biopsych.2008.06.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 06/06/2008] [Accepted: 06/07/2008] [Indexed: 01/24/2023]
Abstract
BACKGROUND Sensitivity to reward has been implicated as a predisposing factor for behaviors related to drug abuse as well as overeating. However, the underlying mechanisms contributing to reward sensitivity are unknown. We hypothesized that a dysregulation in dopamine signaling might be an underlying cause of heightened reward sensitivity whereby rewarding stimuli could act to normalize the system. METHODS We used a genetic mouse model of increased reward sensitivity, the Delta FosB-overexpressing mouse, to examine reward pathway changes in response to a palatable high-fat diet. Markers of reward signaling in these mice were examined both basally and following 6 weeks of palatable diet exposure. Mice were examined in a behavioral test following high-fat diet withdrawal to assess the vulnerability of this model to removal of rewarding stimuli. RESULTS Our results demonstrate altered reward pathway activation along the nucleus accumbens-hypothalamic-ventral tegmental area circuitry resulting from overexpression of Delta FosB in the nucleus accumbens and striatal regions. Levels of phosphorylated cyclic adenosine monophosphate (cAMP) response element binding protein (pCREB), brain-derived neurotrophic factor (BDNF), and dopamine and cyclic adenosine monophosphate regulated phosphoprotein with a molecular mass of 32 kDa (DARPP-32) in the nucleus accumbens were reduced in Delta FosB mice, suggestive of reduced dopamine signaling. Six weeks of high-fat diet exposure completely ameliorated these differences, revealing the potent rewarding capacity of a palatable diet. Delta FosB mice also showed a significant increase in locomotor activity and anxiety-related responses 24 hours following high-fat withdrawal. CONCLUSIONS These results establish an underlying sensitivity to changes in reward related to dysregulation of Delta FosB and dopamine signaling that can be normalized with palatable diets and may be a predisposing phenotype in some forms of obesity.
Collapse
Affiliation(s)
- Sarah L Teegarden
- Department of Animal Biology, University of Pennsylvania, Philadelphia, PA 19104-6046, USA
| | | | | |
Collapse
|
32
|
Gustafsson L, Oreland S, Hoffmann P, Nylander I. The impact of postnatal environment on opioid peptides in young and adult male Wistar rats. Neuropeptides 2008; 42:177-91. [PMID: 18082882 DOI: 10.1016/j.npep.2007.10.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 09/21/2007] [Accepted: 10/30/2007] [Indexed: 10/22/2022]
Abstract
Early environmental influences can change the neuronal development and thereby affect behavior in adult life. The aim in the present study was to thoroughly examine the impact of early environmental factors on endogenous opioids by using a rodent maternal separation (MS) model. The endogenous opioid peptide system is not fully developed at birth, and short- and/or long-term alterations may occur in these neural networks in animals exposed to manipulation of the postnatal environment. Rat pups were subjected to one of five rearing conditions; 15 min (MS15) litter (l) or individual (i), 360 min (MS360) l or i daily MS, or housed under normal animal facility rearing (AFR) conditions during postnatal days 1-21. Measurements of immunoreactive (ir) Met-enkephalin-Arg6Phe7 (MEAP) and dynorphin B (DYNB) peptide levels in the pituitary gland and in a number of brain areas, were performed at three and 10 weeks of age, respectively. MS-induced changes were more pronounced in ir MEAP levels, especially in individually separated rats at three weeks of age and in litter-separated rats at 10 weeks of age. The enkephalin and dynorphin systems have different developmental patterns, dynorphin appearing earlier, which may point at a more sensitive enkephalin system during the early postnatal weeks. The results provide evidence that opioid peptides are sensitive for early environmental factors and show that the separation conditions are critical and also result in changes manifesting at different time points. MS-induced effects were observed in areas related to stress, drug reward and dependence mechanisms. By describing effects on opioid peptides, the study addresses the possible role of a deranged endogenous opioid system in the previously described behavioral consequences of MS.
Collapse
Affiliation(s)
- Lisa Gustafsson
- Department of Pharmaceutical Biosciences, Division of Pharmacology, Uppsala University, P.O. Box 591, SE-751 24 Uppsala, Sweden
| | | | | | | |
Collapse
|
33
|
Abstract
Despite the generally held view that alcohol is an unspecific pharmacological agent, recent molecular pharmacology studies demonstrated that alcohol has only a few known primary targets. These are the NMDA, GABA(A), glycine, 5-hydroxytryptamine 3 (serotonin) and nicotinic ACh receptors as well as L-type Ca(2+) channels and G-protein-activated inwardly rectifying K(+) channels. Following this first hit of alcohol on specific targets in the brain, a second wave of indirect effects on a variety of neurotransmitter/neuropeptide systems is initiated that leads subsequently to the typical acute behavioural effects of alcohol, ranging from disinhibition to sedation and even hypnosis, with increasing concentrations of alcohol. Besides these acute pharmacodynamic aspects of alcohol, we discuss the neurochemical substrates that are involved in the initiation and maintenance phase of an alcohol drinking behaviour. Finally, addictive behaviour towards alcohol as measured by alcohol-seeking and relapse behaviour is reviewed in the context of specific neurotransmitter/neuropeptide systems and their signalling pathways. The activity of the mesolimbic dopaminergic system plays a crucial role during the initiation phase of alcohol consumption. Following long-term, chronic alcohol consumption virtually all brain neurotransmission seems to be affected, making it difficult to define which of the systems contributes the most to the transition from controlled to compulsive alcohol use. However, compulsive alcohol drinking is characterized by a decrease in the function of the reward neurocircuitry and a recruitment of antireward/stress mechanisms comes into place, with a hypertrophic corticotropin-releasing factor system and a hyperfunctional glutamatergic system being the most important ones.
Collapse
|
34
|
Weerts EM, Kim YK, Wand GS, Dannals RF, Lee JS, Frost JJ, McCaul ME. Differences in delta- and mu-opioid receptor blockade measured by positron emission tomography in naltrexone-treated recently abstinent alcohol-dependent subjects. Neuropsychopharmacology 2008; 33:653-65. [PMID: 17487229 DOI: 10.1038/sj.npp.1301440] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Blockade of brain mu-opioid receptor (mu-OR) and delta-opioid receptor (delta-OR) was investigated in recently abstinent alcohol-dependent subjects (N=21) maintained on naltrexone. Subjects completed a 19-day inpatient protocol, which included alcohol abstinence followed by naltrexone treatment (50 mg) on days 15-19. Blood samples were collected after the first administration of naltrexone to evaluate serum levels of naltrexone and 6-beta-naltrexol. Regional brain mu-OR binding potential (BP) and delta-OR Ki was measured using [11C]carfentanil (CAR) positron emission tomography (PET) and [11C]methyl naltrindole ([11C]MeNTI) PET, respectively, before (day 5) and during naltrexone treatment (day 18). Naltrexone inhibition of [11C]CAR BP was near maximal across all brain regions of interest with little variability across subjects (mean+SD% inhibition=94.9+4.9%). Naltrexone only partially inhibited the [11C]MeNTI Ki and there was more variability across subjects (mean+SD% inhibition=21.1+14.49%). Peak serum levels of naltrexone were positively correlated with % inhibition of delta-OR Ki in neocortex and basal ganglia. Peak serum levels of naltrexone were not correlated with % inhibition of mu-OR BP. Peak levels of 6-beta-naltrexol were not significantly correlated with % inhibition of mu-OR BP or delta-OR Ki. Thus, the FDA recommended therapeutic dose of naltrexone was sufficient to produce near complete inhibition of the mu-OR in recently abstinent alcohol dependent subjects. The lower percent inhibition of delta-OR and greater variability in delta-OR blockade by naltrexone across subjects may contribute to individual differences in treatment outcomes to naltrexone. Further investigations on the relationship between individual differences in delta-OR blockade by naltrexone and clinical outcomes should be explored.
Collapse
Affiliation(s)
- Elise M Weerts
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Méndez M, Morales-Mulia M, Pérez-Luna JM. Ethanol-induced changes in proenkephalin mRNA expression in the rat nigrostriatal pathway. J Mol Neurosci 2008; 34:225-34. [PMID: 18227978 DOI: 10.1007/s12031-008-9039-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2007] [Accepted: 01/02/2008] [Indexed: 10/22/2022]
Abstract
Endogenous opioid systems have been suggested to play a key role in ethanol reinforcement mechanisms and alcohol-drinking behavior. Ethanol induces differential alterations in opioid peptide expression in brain areas of the reward circuits, which may be linked to the reinforcing effects of ethanol. In addition, ethanol-induced alterations in opioidergic nigrostriatal transmission could be involved in brain sensitivity to ethanol and play a role in addictive processes. The aim of this work was to study the effects of acute ethanol administration on proenkephalin (proenk) mRNA expression in the rat substantia nigra and caudate-putamen (CP) for up to 24 h post treatment. Male Wistar rats received ethanol (2.5 g/kg) or distilled water by intragastric administration, and proenk mRNA expression was studied by in situ hybridization and densitometry. Ethanol transiently increased proenk mRNA expression in the CP 1 h after drug administration. Proenk mRNA levels remained elevated 2 h post treatment in the anterior-medial and medial-posterior regions of the CP. In contrast, ethanol decreased proenk mRNA expression in the substantia nigra pars compacta and pars reticulata 2 h after drug exposure. Alterations in enkephalin expression in the substantia nigra and CP in response to ethanol exposure could be involved in the mechanisms underlying brain sensitivity to the drug.
Collapse
Affiliation(s)
- Milagros Méndez
- Departamento de Neuroquímica, Calzada México Xochimilco 101, Col. San Lorenzo Huipulco, 14370, México D.F., México.
| | | | | |
Collapse
|
36
|
Shippenberg TS, Zapata A, Chefer VI. Dynorphin and the pathophysiology of drug addiction. Pharmacol Ther 2007; 116:306-21. [PMID: 17868902 PMCID: PMC2939016 DOI: 10.1016/j.pharmthera.2007.06.011] [Citation(s) in RCA: 282] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Accepted: 06/27/2007] [Indexed: 12/30/2022]
Abstract
Drug addiction is a chronic relapsing disease in which drug administration becomes the primary stimulus that drives behavior regardless of the adverse consequence that may ensue. As drug use becomes more compulsive, motivation for natural rewards that normally drive behavior decreases. The discontinuation of drug use is associated with somatic signs of withdrawal, dysphoria, anxiety, and anhedonia. These consequences of drug use are thought to contribute to the maintenance of drug use and to the reinstatement of compulsive drug use that occurs during the early phase of abstinence. Even, however, after prolonged periods of abstinence, 80-90% of human addicts relapse to addiction, suggesting that repeated drug use produces enduring changes in brain circuits that subserve incentive motivation and stimulus-response (habit) learning. A major goal of addiction research is the identification of the neural mechanisms by which drugs of abuse produce these effects. This article will review data showing that the dynorphin/kappa-opioid receptor (KOPr) system serves an essential function in opposing alterations in behavior and brain neurochemistry that occur as a consequence of repeated drug use and that aberrant activity of this system may not only contribute to the dysregulation of behavior that characterizes addiction but to individual differences in vulnerability to the pharmacological actions of cocaine and alcohol. We will provide evidence that the repeated administration of cocaine and alcohol up-regulates the dynorphin/KOPr system and that pharmacological treatments that target this system may prove effective in the treatment of drug addiction.
Collapse
Affiliation(s)
- T S Shippenberg
- Integrative Neuroscience Section, NIH/NIDA Intramural Research Program, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | | | | |
Collapse
|
37
|
D'Addario C, Ming Y, Ogren SO, Terenius L. The role of acetaldehyde in mediating effects of alcohol on expression of endogenous opioid system genes in a neuroblastoma cell line. FASEB J 2007; 22:662-70. [PMID: 17934066 DOI: 10.1096/fj.07-8346com] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Ethanol (EtOH) alters neural activity through interaction with various neurotransmitters and neuromodulators. The endogenous opioid system seems to play a key role in the activities of EtOH, since the opioid antagonist naltrexone (ReVia) attenuates craving. We have investigated the transcriptional regulation of opioid system genes in response to EtOH exposure for up to 96 h in human neuroblastoma SH-SY5Y cells using quantitative real-time polymerase chain reaction. We observed a significant decrease in the expression of opioid peptide precursors (proopiomelanocortin, proenkephalin, and prodynorphin) and of the kappa opioid receptor after 48 and 72 h of EtOH exposure (10 and 40 mM). These alterations were not present when the EtOH metabolism was blocked by 4-methylpyrazole. To evaluate whether the effects evoked by EtOH were possibly due to the first product of EtOH metabolism, cells were exposed to 0.4 mM acetaldehyde. We observed the same pattern of changes for prodynorphin, proenkephalin, and the kappa opioid receptor as after 72 h exposure to EtOH. These results contribute to our understanding of EtOH action at a cellular level and provide evidence of the role of acetaldehyde in mediating some of the EtOH-induced effects.
Collapse
Affiliation(s)
- Claudio D'Addario
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, 17176 Sweden.
| | | | | | | |
Collapse
|
38
|
Lindholm S, Rosin A, Dahlin I, Georgieva J, Franck J. Ethanol alters the effect of kappa receptor ligands on dopamine release in the nucleus accumbens. Physiol Behav 2007; 92:167-71. [PMID: 17610914 DOI: 10.1016/j.physbeh.2007.05.039] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Repeated exposure to ethanol has previously been shown to induce alterations in both midbrain dopamine and dynorphin systems. The aim of this study was to investigate functional changes in the sensitivity of dynorphin/kappa-receptor systems following repeated ethanol administration, using dopamine as an indirect marker. The effects of kappa-opioid receptor ligands on dopamine release in the rat nucleus accumbens were investigated following repeated ethanol administration (2 g/kg body weight, twice daily for 7 days). The selective kappa-receptor agonist U50, 488H reduced dopamine levels in both ethanol- and saline-treated animals, although the decline had a later onset and lasted shorter in the ethanol-treated group. Nor-binaltorphimine, a kappa-antagonist, produced a significant increase of dopamine in ethanol-treated rats, but lacked effect in the saline-treated group. This change in responsiveness of dopamine neurons following repeated ethanol administration could be related to changes in the sensitivity of kappa-receptor systems and/or an increase in dynorphin tone in the nucleus accumbens.
Collapse
Affiliation(s)
- Sara Lindholm
- Karolinska Institutet, Department of Clinical Neuroscience, CMM L8:01, Karolinska, Hospital, S-171 76 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
39
|
Gustafsson L, Zhou Q, Nylander I. Ethanol-induced effects on opioid peptides in adult male Wistar rats are dependent on early environmental factors. Neuroscience 2007; 146:1137-49. [PMID: 17391858 DOI: 10.1016/j.neuroscience.2007.02.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 02/05/2007] [Accepted: 02/20/2007] [Indexed: 11/21/2022]
Abstract
The vulnerability to develop alcoholism is dependent on both genetic and environmental factors. The neurobiological mechanisms underlying these factors are not fully understood but individual divergence in the endogenous opioid peptide system may contribute. We have previously reported that early-life experiences can affect endogenous opioids and also adult voluntary ethanol intake. In the present study, this line of research was continued and the effects of long-term voluntary ethanol drinking on the opioid system are described in animals reared in different environmental settings. Rat pups were subjected to 15 min (MS15) or 360 min (MS360) of daily maternal separation during postnatal days 1-21. At 10 weeks of age, male rats were exposed to voluntary ethanol drinking in a four-bottle paradigm with 5%, 10% and 20% ethanol solution in addition to water for 2 months. Age-matched controls received water during the same period. Immunoreactive (ir) Met-enkephalin-Arg6Phe7 (MEAP) and dynorphin B (DYNB) peptide levels were thereafter measured in the pituitary gland and several brain areas. In water-drinking animals, lower ir MEAP levels were observed in the MS360 rats in the hypothalamus, medial prefrontal cortex, striatum and the periaqueductal gray, whereas no differences were seen in ir DYNB levels. Long-term ethanol drinking induced lower ir MEAP levels in MS15 rats in the medial prefrontal cortex and the periaqueductal gray, whereas higher levels were detected in MS360 rats in the hypothalamus, striatum and the substantia nigra. Chronic voluntary drinking affected ir DYNB levels in the pituitary gland, hypothalamus and the substantia nigra, with minor differences between MS15 and MS360. In conclusion, manipulation of the early environment caused changes in the opioid system and a subsequent altered response to ethanol. The altered sensitivity of the opioid peptides to ethanol may contribute to the previously reported differences in ethanol intake between MS15 and MS360 rats.
Collapse
Affiliation(s)
- L Gustafsson
- Department of Pharmaceutical Biosciences, Division of Pharmacology, Uppsala University, P.O. Box 591, SE-751 24 Uppsala, Sweden
| | | | | |
Collapse
|
40
|
Ojanen SP, Palmén M, Hyytiä P, Kiianmaa K. Extracellular glutamate and GABA in the ventral tegmental area of alcohol-preferring AA and alcohol-avoiding ANA rats treated repeatedly with morphine. Eur J Pharmacol 2007; 559:38-45. [PMID: 17198699 DOI: 10.1016/j.ejphar.2006.11.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 10/30/2006] [Accepted: 11/20/2006] [Indexed: 10/23/2022]
Abstract
Glutamate and gamma-amino-butyric acid (GABA) have been implicated in neuronal plasticity related to behavioral sensitization. In the present study, we examined morphine-induced changes in the extracellular concentrations of glutamate and GABA in the ventral tegmental area in alcohol-preferring Alko Alcohol (AA) and alcohol-avoiding Alko Non-Alcohol (ANA) rats that have previously been shown to differ in morphine-induced sensitization. The rats were given escalating doses (5-20 mg/kg) of morphine every other day for five days. This treatment produced behavioral sensitization to locomotor effects of morphine in AA, but not in ANA rats, when challenged with an additional injection of morphine (10 mg/kg) 10 days later. Morphine also increased the levels of glutamate in the ventral tegmental area only in AA rats, while no significant changes were found in the extracellular concentrations of GABA between the lines. Challenging the morphine-treated AA rats with ethanol (1.5 g/kg) did not modify the levels of glutamate or GABA. No changes in the concentrations of glutamate or GABA were seen in saline-treated AA and ANA rats after morphine challenge. These results render increased glutamate transmission in the ventral tegmental area a potential contributor to the higher susceptibility of AA rats to morphine-induced behavioral and neurochemical effects relative to ANA rats.
Collapse
Affiliation(s)
- Sami P Ojanen
- Department of Mental Health and Alcohol Research, National Public Health Institute, POB 33, 00251 Helsinki, Finland
| | | | | | | |
Collapse
|
41
|
Méndez M, Morales-Mulia M. Ethanol exposure differentially alters pro-enkephalin mRNA expression in regions of the mesocorticolimbic system. Psychopharmacology (Berl) 2006; 189:117-24. [PMID: 17047937 DOI: 10.1007/s00213-006-0503-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Accepted: 06/28/2006] [Indexed: 10/24/2022]
Abstract
RATIONALE Opioid peptides have been suggested to play a major role in ethanol reinforcement mechanisms and alcohol drinking behaviour. However, in non-selected strains of rodents, it is not known whether opioid biosynthesis is a critical event in these processes. OBJECTIVE The aim of this work was to study the effects of a high dose of ethanol (2.5 g/kg body weight) on pro-enkephalin (pro-enk) mRNA expression in brain regions of the mesocorticolimbic system for up to 24 h after drug administration. MATERIALS AND METHODS Male Wistar rats were administered with ethanol (2.5 g/kg body weight) or distilled water and were killed 30 min, 1, 2, 4, 8 or 24 h after treatment. Coronal brain sections (20 mu) were obtained and pro-enk mRNA expression was studied by in situ hybridization and densitometry. RESULTS Acute ethanol administration induced a transient decrease and increase in pro-enk mRNA expression in the ventral tegmental area (33.2%) and prefrontal cortex (26.5%) 2 and 4 h after treatment, respectively. In contrast, ethanol induced prolonged increases in pro-enk mRNA expression in the core and shell regions of the nucleus accumbens, with different kinetics. Maximal effects were observed 2 h after ethanol exposure (core, 70.0%; shell, 60.0%). CONCLUSIONS Our results indicate that enkephalin expression in regions of the rat mesocorticolimbic system is differentially altered by acute ethanol treatment and suggest that enkephalins may play a key role in ethanol reinforcement mechanisms.
Collapse
Affiliation(s)
- Milagros Méndez
- Departamento de Neuroquímica, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente, Calzada México Xochimilco 101, Col. San Lorenzo Huipulco, 14370, México DF, México.
| | | |
Collapse
|
42
|
Sommer W, Hyytiä P, Kiianmaa K. The alcohol-preferring AA and alcohol-avoiding ANA rats: neurobiology of the regulation of alcohol drinking. Addict Biol 2006; 11:289-309. [PMID: 16961760 DOI: 10.1111/j.1369-1600.2006.00037.x] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The AA (alko, alcohol) and ANA (alko, non-alcohol) rat lines were among the earliest rodent lines produced by bidirectional selection for ethanol preference. The purpose of this review is to highlight the strategies for understanding the neurobiological factors underlying differential alcohol-drinking behavior in these lines. Most early work evaluated functioning of the major neurotransmitter systems implicated in drug reward in the lines. No consistent line differences were found in the dopaminergic system either under baseline conditions or after ethanol challenges. However, increased opioidergic tone in the ventral striatum and a deficiency in endocannabinoid signaling in the prefrontal cortex of AA rats may comprise mechanisms leading to increased ethanol consumption. Because complex behaviors, such as ethanol drinking, are not likely to be controlled by single factors, system-oriented molecular-profiling strategies have been used recently. Microarray based expression analysis of AA and ANA brains and novel data-mining strategies provide a system biological view that allows us to formulate a hypothesis on the mechanism underlying selection for ethanol preference. Two main factors appear active in the selection: a recruitment of signal transduction networks, including mitogen-activated protein kinases and calcium pathways and involving transcription factors such as Creb, Myc and Max, to mediate ethanol reinforcement and plasticity. The second factor acts on the mitochondrion and most likely provides metabolic flexibility for alternative substrate utilization in the presence of low amounts of ethanol.
Collapse
Affiliation(s)
- Wolfgang Sommer
- Laboratory of Clinical and Translational Studies, NIAAA, National Institutes of Health, USA
| | | | | |
Collapse
|
43
|
Marinelli PW, Lam M, Bai L, Quirion R, Gianoulakis C. A Microdialysis Profile of Dynorphin A1-8 Release in the Rat Nucleus Accumbens Following Alcohol Administration. Alcohol Clin Exp Res 2006; 30:982-90. [PMID: 16737456 DOI: 10.1111/j.1530-0277.2006.00112.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Pharmacological studies have implicated the endogenous opioid system in mediating alcohol intake. Other evidence has shown that alcohol administration can influence endorphinergic and enkephalinergic activity, while very few studies have examined its effect on dynorphinergic systems. The aim of the present study was to investigate the effect of alcohol administration or a mechanical stressor on extracellular levels of dynorphin A(1-8) in the rat nucleus accumbens-a brain region that plays a significant role in the processes underlying reinforcement and stress. METHODS Male Sprague-Dawley rats were implanted with a microdialysis probe aimed at the shell region of the nucleus accumbens. Artificial cerebrospinal fluid was pumped at a rate of 1.5 microL/min in awake and freely moving animals and the dialysate was collected at 30-minute intervals. In one experiment, following a baseline period, rats were injected intraperitoneally with either physiological saline or 1 of 3 doses of alcohol, 0.8, 1.6, or 3.2 g ethanol/kg body weight. In a second experiment, following a baseline period, rats were applied a clothespin to the base of their tail for 20 minutes. The levels of dynorphin A(1-8) in the dialysate were analyzed with solid-phase radioimmunoassay. RESULTS Relative to saline-treated controls, an alcohol dose of 1.6 and 3.2 g/kg caused a transient increase in the extracellular levels of dynorphin A(1-8) in the first 30 minutes of alcohol administration. However, the effect resulting from the high 3.2 g/kg dose was far more pronounced and more significant than with the moderate dose. There was no effect of tail pinch on dynorphin A(1-8) levels in the nucleus accumbens. CONCLUSIONS In this experiment, a very high dose of alcohol was especially capable of stimulating dynorphin A(1-8) release in the nucleus accumbens. Dynorphin release in the accumbens has been previously associated with aversive stimuli and may thus reflect a system underlying the aversive properties of high-dose alcohol administration. However, the lack of effect of tail-pinch stress in the present study suggests that dynorphin A(1-8) is not released in response to all forms of stressful/aversive stimuli.
Collapse
Affiliation(s)
- Peter W Marinelli
- Department of Neurology and Neurosurgery, Douglas Hospital Research Centre and McGill University, Montréal, Québec, Canada
| | | | | | | | | |
Collapse
|
44
|
Zapata A, Shippenberg TS. Endogenous kappa opioid receptor systems modulate the responsiveness of mesoaccumbal dopamine neurons to ethanol. Alcohol Clin Exp Res 2006; 30:592-7. [PMID: 16573576 DOI: 10.1111/j.1530-0277.2006.00069.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Endogenous kappa-opioid receptor (KOPR) systems modulate the actions of several drugs of abuse. Their role in modulating the effects of ethanol is unknown. An increase in nucleus accumbens extracellular dopamine (DA) has been implicated in mediating the rewarding and locomotor-activating effects of ethanol and virtually all drugs of abuse. The present microdialysis studies were conducted to determine whether the lack of KOPR alters ethanol-evoked DA levels in the nucleus accumbens of naïve mice and whether a similar effect is observed in mice repeatedly exposed to ethanol. METHODS Gene deletion techniques were used in conjunction with in vivo microdialysis to examine the influence of lack of KOPR on ethanol-evoked DA in the nucleus accumbens. To determine whether pharmacological inactivation of KOPR produces similar effects in naïve mice and those repeatedly exposed to ethanol, the KOPR antagonist norbinaltorphimine (n-BNI) was administered in wild-type mice before repeated air or ethanol vapor inhalation. Microdialysis was conducted 24 hours later. RESULTS Acute ethanol administration increased DA levels in the nucleus accumbens of wild-type mice. In littermates lacking the KOPR gene, ethanol-evoked DA levels were enhanced. Prior ethanol exposure reduced ethanol-evoked DA levels in vehicle-treated and n-BNI-treated mice. Statistical analysis, however, revealed a significant main effect of n-BNI, indicating that KOPR blockade increased ethanol-evoked DA levels in naïve mice and repeated ethanol exposure attenuated, but did not abolish, this effect. CONCLUSIONS These findings demonstrate that inhibition of KOPR leads to increased sensitivity to the DA-releasing effects of ethanol in the nucleus accumbens.
Collapse
Affiliation(s)
- Agustin Zapata
- Integrative Neuroscience Section, Behavioral Neuroscience Branch, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland 21224, USA.
| | | |
Collapse
|
45
|
Ojanen SP, Hyytiä P, Kiianmaa K. Enhanced Morphine-Induced Ethanol Drinking in Alcohol-Preferring Alko Alcohol Rats Sensitized to Morphine. Alcohol Clin Exp Res 2006; 30:621-9. [PMID: 16573579 DOI: 10.1111/j.1530-0277.2006.00072.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Alcohol-preferring alko alcohol (AA) rats are more susceptible to morphine-induced behavioral and neurochemical sensitization than alcohol nonpreferring alko nonalcohol (ANA) rats. Alko alcohol rats sensitized to morphine, however, do not show enhanced acquisition of ethanol drinking. The purpose of the present study was to clarify further interactions between morphine-induced behavioral sensitization and voluntary ethanol drinking in the AA rats. METHODS Alko alcohol rats drinking ethanol in a limited 6-hour access paradigm were sensitized to morphine with repeated injections of morphine (5-15 mg/kg). Injection days alternated with days of ethanol access. Controls had access only to water and/or were given injections of saline. After a 5-day washout period from ethanol and morphine, the rats were challenged with morphine or saline and subsequent ethanol drinking or locomotor activity was recorded. RESULTS Ethanol intake was suppressed during the repeated treatment with morphine, and the morphine-treated rats did not differ in ethanol intake from the controls when given access to ethanol after the washout. Intake of ethanol was, however, increased when the rats were challenged with morphine [1 or 10 mg/kg, subcutaneously (s.c.)], while in the controls an increase in ethanol intake was seen only after 1 mg/kg morphine. Sensitization to the locomotor stimulating effects of morphine was revealed in the morphine-treated rats after a challenge with morphine (3 or 10 mg/kg, s.c.). The controls that had been drinking ethanol also showed a sensitized response after morphine (3 mg/kg). CONCLUSIONS Ethanol did not interfere with the development of sensitization to morphine. Furthermore, the neuroadaptations induced by repeated exposure to ethanol were sufficient to cause behavioral cross-sensitization to morphine. Sensitization to the behavioral effects of morphine alone, however, neither enhances the reinforcing properties of voluntarily consumed ethanol nor contributes to increase in its intake. The increase in ethanol intake found after an acute dose of morphine was augmented in rats withdrawn from repeated treatment with morphine. The data suggest that the neuronal mechanisms underlying behavioral sensitization to morphine probably are distinct from those mediating reinforcement from ethanol and that the morphine-induced neuroadaptations contribute to the enhancement of increase in ethanol intake by morphine.
Collapse
Affiliation(s)
- Sami P Ojanen
- Department of Mental Health and Alcohol Research, National Public Health Institute, Helsinki, Finland
| | | | | |
Collapse
|
46
|
Guitart-Masip M, Giménez-Llort L, Fernández-Teruel A, Cañete T, Tobeña A, Ogren SO, Terenius L, Johansson B. Reduced ethanol response in the alcohol-preferring RHA rats and neuropeptide mRNAs in relevant structures. Eur J Neurosci 2006; 23:531-40. [PMID: 16420460 DOI: 10.1111/j.1460-9568.2005.04556.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Roman rat strains, genetically selected for high (RHA) or low (RLA) active avoidance acquisition in the two-way shuttle box, differ in dopaminergic activity. These two strains appear to be a valid laboratory model of divergent sensation/novelty and substance-seeking profiles. RHA rats show higher ethanol intake and preference than do RLA rats, and it was suggested that RHA rats are more tolerant than RLA to the effects of alcohol. In the hole-board test, we found that the non-alcohol-preferring RLA rats showed enhanced responsiveness to the stimulatory effects of intraperitoneal administration of 0.25 g/kg ethanol when compared with RHA rats. In situ hybridization analysis showed higher levels of preprodynorphin in the accumbens shell and higher levels of preproenkephalin in the cingulate cortex in RHA rats. RLA rats showed higher levels of enkephalin gene transcripts in restricted areas of the dorsal striatum. Finally, differences in cholecystokinin gene transcript, suggestive of a different arrangement of certain interneurons, were found in different cortical areas. The differences in peptide gene expression found between the two strains might reflect the differences in alcohol preference and sensitivity. RHA rats may have more predictive value than other rodent alcoholism models, as high initial tolerance to ethanol is a risk factor for alcoholism in humans.
Collapse
Affiliation(s)
- Marc Guitart-Masip
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Neuroscience Institute, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Drug addiction is a chronic disease characterized by compulsive drug use despite the severe negative consequences associated with it. Repeated exposure to drugs of abuse results in molecular adaptations in neuronal signaling pathways, which eventually manifest in the complex behavioral alterations that characterize addiction. These include tolerance, sensitization, dependence, drug craving, and relapse. In this Review, we focus on recent studies highlighting signaling cascades initiated by cocaine, as a representative of a drug of abuse with a defined site of action, and alcohol, as a drug with an undefined primary site of action. Specifically, we describe recent studies that emphasize the role of protein-protein interactions, phosphorylation, and compartmentalization in the molecular mechanisms that result in the cellular and behavioral adaptations that underlie addiction. Signaling cascades that contribute to addiction, as well as those that protect or delay the development of addiction, are presented.
Collapse
Affiliation(s)
- Dorit Ron
- Ernest Gallo Clinic and Research Center, University of California San Francisco, Emeryville, CA 94608, USA.
| | | |
Collapse
|
48
|
Mitchell JM, Liang MT, Fields HL. A single injection of the kappa opioid antagonist norbinaltorphimine increases ethanol consumption in rats. Psychopharmacology (Berl) 2005; 182:384-92. [PMID: 16001119 DOI: 10.1007/s00213-005-0067-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Accepted: 04/27/2005] [Indexed: 10/25/2022]
Abstract
RATIONALE Kappa opioid receptor (KOR) agonists interfere with the reinforcing effects of drugs of abuse. KOR agonists decrease heroin, cocaine, and ethanol self-administration, and block heroin and cocaine conditioned place preference (CPP) in rats. However, KOR agonists also produce emesis and dysphoria, making it difficult to determine if their effects on self-administration are due to an action on reward mechanisms or are secondary to the drug's direct aversive effects. Assuming that endogenous KOR ligands modulate circuits involved in drug and alcohol reward, selective KOR antagonists can be used to clarify these issues. If KOR antagonists increase drug self-administration then it is likely that endogenous KOR agonists directly modulate drug intake. OBJECTIVES To determine the effects of nor-BNI, the highly selective KOR antagonist, on ethanol consumption and CPP. METHODS Thirty-eight male Lewis rats were given free access to ethanol until stable self-administration was achieved. Animals were then administered a single injection of nor-BNI (10 mg kg(-1)) while ethanol intake was monitored. RESULTS A single injection of nor-BNI induces a long-lasting increase in ethanol consumption, but does not induce a CPP. A high/low split revealed that this effect was primarily due to an increase in drinking in nor-BNI-treated high drinkers, which drank significantly more than saline-treated high drinkers and also drank significantly more when compared to their own pretreatment baseline. CONCLUSIONS Blocking the KOR system increases ethanol self-administration, suggesting that the decrease in self-administration seen with KOR agonists is due to a direct modulation of reward circuitry.
Collapse
Affiliation(s)
- Jennifer M Mitchell
- Department of Neurology, University of California at San Francisco, P. O. Box 0114, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
49
|
Marinelli PW, Bai L, Quirion R, Gianoulakis C. A Microdialysis Profile of Met-Enkephalin Release in the Rat Nucleus Accumbens Following Alcohol Administration. Alcohol Clin Exp Res 2005; 29:1821-8. [PMID: 16269911 DOI: 10.1097/01.alc.0000183008.62955.2e] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Pharmacological studies have implicated the endogenous opioid system in mediating alcohol intake. Other evidence has shown that alcohol administration can influence opioid activity. In this regard, the majority of studies have concentrated on endorphinergic systems, whereas other opioid systems have been granted comparably less attention. This is the case despite some compelling evidence that has implicated enkephalinergic peptide systems, particularly Met-enkephalin, in mediating alcohol preference. The aim of the present study was to investigate the effect of alcohol administration on extracellular levels of Met-enkephalin in the rat nucleus accumbens--a brain region that plays a significant role in the processes underlying reinforcement and stress. METHODS Male Sprague-Dawley rats were implanted with a microdialysis probe aimed at the shell region of the nucleus accumbens. Artificial cerebrospinal fluid was pumped at a rate of 1.75 mul/min in awake and freely moving rats and dialysates were collected at 30-minute intervals. After several baseline collections, rats were injected intraperitoneally with either physiological saline or one of four doses of alcohol: 0.8, 1.6, 2.4, or 3.2 g/kg ethanol body weight. The levels of Met-enkephalin in the dialysates were analyzed with solid-phase radioimmunoassay. RESULTS Within the first 30 minutes of administration, an alcohol dose of 1.6 g/kg caused a significant and prolonged elevation in the extracellular levels of Met-enkephalin. Alcohol did not have a major effect on the release of Met-enkephalin at any other dose. CONCLUSIONS In this experiment, only a moderate dose of alcohol was capable of stimulating Met-enkephalin release in the nucleus accumbens. Enkephalins may modulate local neurotransmitter release by binding to presynaptic Delta-opioid receptors, or, they may inhibit effector cells by binding to postsynaptic Delta- or mu-opioid receptors. This may be one of multiple neurological mechanisms that modulate alcohol-drinking behavior.
Collapse
Affiliation(s)
- Peter W Marinelli
- Biobehavioural Pharmacology Section, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
50
|
Abstract
Preclinical and clinical data implicate the endogenous opioid system in alcohol dependence. In vitro studies show that rodent pituitary and hypothalamic tissue responds to acute exposure to alcohol by releasing beta-endorphins. In vivo studies suggest differential activity of endogenous opioid receptors in rodents with high and low alcohol preference. Similarly, humans with a family history of alcohol dependence also show a heightened endorphin response to an acute challenge of alcohol compared with those with no family history of alcohol dependence.The effects of opioid agonists and antagonists on rodent and human alcohol consumption further support the opioid-alcohol link. In rodents and humans, small doses of opioid agonists increase alcohol consumption, while pretreatment with large doses decreases consumption. The opioid antagonist naltrexone decreases rodent alcohol consumption, particularly in low doses under acute and intermittent schedules. Most clinical trials in patients with alcohol dependence support modest therapeutic effects of naltrexone in decreasing alcohol consumption. Efforts to identify subgroups of alcohol-dependent patients responsive to naltrexone, as well as psychosocial and pharmacological augmentation strategies, may further improve the clinical usefulness of the drug.
Collapse
|