1
|
Tobore TO. On the Neurobiological Role of Oxidative Stress in Alcohol-Induced Impulsive, Aggressive and Suicidal Behavior. Subst Use Misuse 2019; 54:2290-2303. [PMID: 31369300 DOI: 10.1080/10826084.2019.1645179] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objectives: Alcohol abuse is known to result in behavioral impairments (such as increased impulsivity, aggressive, and suicidal behavior), but the neurobiological basis for these behavioral impairments remains unknown. The objective of this review is to propose a neurobiological basis for alcohol-induced aggression, impulsivity, and suicidal behavior. Methods: Search was done by accessing PubMed/Medline, EBSCO, and PsycINFO databases. The search string used was "(Alcohol OR Alcoholism* OR Alcohol Abuse) AND (Behavior* OR Behavioral Impairment or Disorder) AND (Oxidative Stress OR Reactive Oxygen Species)." The electronic databases were searched for titles or abstracts containing these terms in all published articles between January 1, 1960, and May 31, 2019. The search was limited to studies published in English and other languages involving both animal and human subjects. Articles selected included randomized clinical trials (RCTs), observational studies, meta-analyses, and both systemic and narrative reviews, providing both quantitative and qualitative information with a measure of alcohol abuse or alcoholism as an outcome. Exclusion criteria were unpublished data of any form, including conference proceedings and dissertation. New key terms were identified (new term included: "Antioxidants, Neurotransmitters, Dopamine, Serotonin, GABA, Glutamate. Aggression, Impulsivity, Suicidal Behavior, hippocampus, prefrontal cortex, limbic system, psychiatric disorders, PTSD, Anxiety, Depression. These new terms were searched with Alcohol or Alcoholism or Alcohol Abuse and Oxidative Stress separately resulting in the identification of over 3000 articles. 196 were included in this article. Results: Multiple lines of evidence indicate that oxidative stress (OS) plays a critical underlying role in alcohol toxicity and behavioral impairments. Conclusions/Importance: People diagnosed with PTSD, anxiety disorder, depression, and those with a personality high in psychoticism as measured by the P Scale of the Eysenck Personality Questionnaire, with comorbid alcohol abuse or alcohol use disorder (AUD), may display increased impulsivity, aggression, and suicidal behavior because of the potentiating effect of alcohol-induced OS on their elevated brain oxidative status. Antioxidant therapy should be an integral part of acute alcohol intoxication and AUD treatment. Further research is necessary to fully understand the relationship between OS and alcohol-induced behavioral impairments.
Collapse
|
2
|
Froehlich JC, Fischer SM, Nicholson ER, Dilley JE, Filosa NJ, Smith TN, Rademacher LC. A Combination of Naltrexone + Varenicline Retards the Expression of a Genetic Predisposition Toward High Alcohol Drinking. Alcohol Clin Exp Res 2017; 41:644-652. [PMID: 28055135 PMCID: PMC5332282 DOI: 10.1111/acer.13326] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/03/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND This study examined whether naltrexone (NTX) or varenicline (VAR), alone or in combination, can retard the phenotypic expression of a genetic predisposition toward high alcohol drinking in rats selectively bred for high alcohol intake when drug treatment is initiated prior to, or concomitantly with, the onset of alcohol drinking. METHODS Alcohol-naïve P rats were treated daily with NTX (15.0 mg/kg BW), VAR (1.0 mg/kg BW), a combination of NTX (15.0 mg/kg BW) + VAR (1.0 mg/kg BW), or vehicle (VEH) for 2 weeks prior to, or concomitantly with, their first opportunity to drink alcohol and throughout 21 days of daily 2-hour alcohol access. Drug treatment was then discontinued for 3 weeks followed by reinstatement of drug treatment for an additional 3 weeks. RESULTS When P rats were pretreated with drug for 2 weeks prior to onset of alcohol access, only NTX + VAR in combination blocked the acquisition of alcohol drinking in alcohol-naïve P rats. When drug treatment was initiated concomitantly with the first opportunity to drink alcohol, NTX alone, VAR alone, and NTX + VAR blocked the acquisition of alcohol drinking. Following termination of drug treatment, NTX + VAR and VAR alone continued to reduce alcohol drinking but by the end of 3 weeks without drug treatment, alcohol intake in all groups was comparable to that seen in the vehicle-treated group as the expression of a genetic predisposition toward high alcohol drinking emerged in the drug-free P rats. After 3 weeks without drug treatment, reinstatement of NTX + VAR treatment again reduced alcohol intake. CONCLUSIONS A combination of NTX + VAR, when administered prior to, or concomitantly with, the first opportunity to drink alcohol, blocks the acquisition of alcohol drinking during both initial access to alcohol and during a later period of alcohol access in P rats with a genetic predisposition toward high alcohol intake. The results suggest that NTX + VAR may be effective in curtailing alcohol drinking in individuals at high genetic risk of developing alcoholism.
Collapse
Affiliation(s)
| | | | | | - Julian E. Dilley
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Teal N. Smith
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
3
|
Xie K, Colgan LA, Dao MT, Muntean BS, Sutton LP, Orlandi C, Boye SL, Boye SE, Shih CC, Li Y, Xu B, Smith RG, Yasuda R, Martemyanov KA. NF1 Is a Direct G Protein Effector Essential for Opioid Signaling to Ras in the Striatum. Curr Biol 2016; 26:2992-3003. [PMID: 27773571 DOI: 10.1016/j.cub.2016.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/24/2016] [Accepted: 09/07/2016] [Indexed: 01/19/2023]
Abstract
It is well recognized that G-protein-coupled receptors (GPCRs) can activate Ras-regulated kinase pathways to produce lasting changes in neuronal function. Mechanisms by which GPCRs transduce these signals and their relevance to brain disorders are not well understood. Here, we identify a major Ras regulator, neurofibromin 1 (NF1), as a direct effector of GPCR signaling via Gβγ subunits in the striatum. We find that binding of Gβγ to NF1 inhibits its ability to inactivate Ras. Deletion of NF1 in striatal neurons prevents the opioid-receptor-induced activation of Ras and eliminates its coupling to Akt-mTOR-signaling pathway. By acting in the striatal medium spiny neurons of the direct pathway, NF1 regulates opioid-induced changes in Ras activity, thereby sensitizing mice to psychomotor and rewarding effects of morphine. These results delineate a novel mechanism of GPCR signaling to Ras pathways and establish a critical role of NF1 in opioid addiction.
Collapse
Affiliation(s)
- Keqiang Xie
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Lesley A Colgan
- Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Maria T Dao
- Department of Metabolism and Aging, The Scripps Research Institute, 120 Scripps Way, Jupiter, FL 33458, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Laurie P Sutton
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, 1395 Center Drive, Gainesville, FL 32610, USA
| | - Shannon E Boye
- Department of Ophthalmology, University of Florida, 1395 Center Drive, Gainesville, FL 32610, USA
| | - Chien-Cheng Shih
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Yuqing Li
- Department of Neurology, University of Florida, 1600 SW Archer Road, Gainesville, FL 32610, USA
| | - Baoji Xu
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Roy G Smith
- Department of Metabolism and Aging, The Scripps Research Institute, 120 Scripps Way, Jupiter, FL 33458, USA
| | - Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA.
| |
Collapse
|
4
|
Froehlich JC, Fischer SM, Dilley JE, Nicholson E, Smith T, Filosa N, Rademacher L. Combining Varenicline (Chantix) with Naltrexone Decreases Alcohol Drinking More Effectively Than Does Either Drug Alone in a Rodent Model of Alcoholism. Alcohol Clin Exp Res 2016; 40:1961-70. [PMID: 27469281 PMCID: PMC5297249 DOI: 10.1111/acer.13157] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/08/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND This study examined whether varenicline (VAR), or naltrexone (NTX), alone or in combination, reduces alcohol drinking in alcohol-preferring (P) rats with a genetic predisposition toward high voluntary alcohol intake. METHODS Alcohol-experienced P rats that had been drinking alcohol (15% v/v) for 2 h/d for 4 weeks were fed either vehicle (VEH), VAR alone (0.5, 1.0, or 2.0 mg/kg body weight [BW]), NTX alone (10.0, 15.0, or 20.0 mg/kg BW), or VAR + NTX in 1 of 4 dose combinations (0.5 VAR + 10.0 NTX, 0.5 VAR + 15.0 NTX, 1.0 VAR + 10.0 NTX, or 1.0 VAR + 15.0 NTX) at 1 hour prior to alcohol access for 10 consecutive days, and the effects on alcohol intake were assessed. RESULTS When administered alone, VAR in doses of 0.5 or 1.0 mg/kg BW did not alter alcohol intake but a dose of 2.0 mg/kg BW decreased alcohol intake. This effect disappeared when drug treatment was terminated. NTX in doses of 10.0 and 15.0 mg/kg BW did not alter alcohol intake but a dose of 20.0 mg/kg BW decreased alcohol intake. Combining low doses of VAR and NTX into a single medication reduced alcohol intake as well as did high doses of each drug alone. Reduced alcohol intake occurred immediately after onset of treatment with the combined medication and continued throughout prolonged treatment. CONCLUSIONS Low doses of VAR and NTX, when combined in a single medication, reduce alcohol intake in a rodent model of alcoholism. This approach has the advantage of reducing potential side effects associated with each drug. Lowering the dose of NTX and VAR in a combined treatment approach that maintains efficacy while reducing the incidence of negative side effects may increase patient compliance and improve clinical outcomes for alcoholics and heavy drinkers who want to reduce their alcohol intake.
Collapse
Affiliation(s)
| | | | - Julian E. Dilley
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Emily Nicholson
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Teal Smith
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nick Filosa
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Logan Rademacher
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
5
|
Fritz BM, Boehm SL. Rodent models and mechanisms of voluntary binge-like ethanol consumption: Examples, opportunities, and strategies for preclinical research. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:297-308. [PMID: 26021391 PMCID: PMC4668238 DOI: 10.1016/j.pnpbp.2015.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 05/02/2015] [Accepted: 05/21/2015] [Indexed: 02/03/2023]
Abstract
Binge ethanol consumption has widespread negative consequences for global public health. Rodent models offer exceptional power to explore the neurobiology underlying and affected by binge-like drinking as well as target potential prevention, intervention, and treatment strategies. An important characteristic of these models is their ability to consistently produce pharmacologically-relevant blood ethanol concentration. This review examines the current available rodent models of voluntary, pre-dependent binge-like ethanol consumption and their utility in various research strategies. Studies have demonstrated that a diverse array of neurotransmitters regulate binge-like drinking, resembling some findings from other drinking models. Furthermore, repeated binge-like drinking recruits neuroadaptive mechanisms in mesolimbocortical reward circuitry. New opportunities that these models offer in the current context of mechanistic research are also discussed.
Collapse
Affiliation(s)
| | - Stephen L Boehm
- Indiana Alcohol Research Center, Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States.
| |
Collapse
|
6
|
Pacini M, Maremmani AGI, Ceccanti M, Maremmani I. Former Heroin-Dependent Alcohol Use Disorder Patients. Prevalence, Addiction History and Clinical Features. Alcohol Alcohol 2015; 50:451-7. [PMID: 25827777 DOI: 10.1093/alcalc/agv028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/05/2015] [Indexed: 01/28/2023] Open
Abstract
AIMS To examine the prevalence of former heroin dependence (FHA) in Alcohol Use Disorder (AUD) patients; to compare the clinical characteristics of FHA-AUD patients versus AUD patients without any past use of heroin at alcohol treatment entry; to document the heroin dependence history of FHA-AUD patients, and review treatment strategies for this group. METHODS Retrospective case review of 448 consecutive AUD patients. RESULTS The annual entry of FHA-AUD showed stability over the study period of 3 years overall 60/448 (13.3%). FHA-AUD patients showed higher concomitant use of cocaine, benzodiazepines, cannabis and hallucinogens than other heroin addicts. They consumed higher amounts of alcohol at the beginning of their alcohol dependence history, and reached a high maximum level of alcohol consumption, than other AUD patients, and tended to have more physical disorders. The most important signals of FHA-AUD were polyabuse and older age at the time of presentation. FHA-AUD patients tended to have had a severe pattern of heroin dependence associated with inadequate agonist opiate treatment. CONCLUSIONS The prevalence of FHA-AUD patients is not negligible. This may relate to previous inadequate treatment of heroin addiction contributing to the development of severe AUD. For these patients we propose a reconsideration of 'soft' (low dose) agonist opiate treatment.
Collapse
Affiliation(s)
- Matteo Pacini
- G. De Lisio Institute of Behavioural Sciences, Pisa, Italy
| | - Angelo Giovanni Icro Maremmani
- Vincent P. Dole Dual Diagnosis Unit, Department of Neurosciences, Santa Chiara University Hospital, University of Pisa, Pisa, Italy AU-CNS, Association for the Application of Neuroscientific Knowledge to Social Aims, Pietrasanta, Lucca, Italy
| | - Mauro Ceccanti
- Centre for the Assessment and Treatment of Alcohol-Related Pathology, La Sapienza University, Rome, Italy
| | - Icro Maremmani
- G. De Lisio Institute of Behavioural Sciences, Pisa, Italy Vincent P. Dole Dual Diagnosis Unit, Department of Neurosciences, Santa Chiara University Hospital, University of Pisa, Pisa, Italy AU-CNS, Association for the Application of Neuroscientific Knowledge to Social Aims, Pietrasanta, Lucca, Italy
| |
Collapse
|
7
|
Froehlich JC, Hausauer BJ, Rasmussen DD. Combining naltrexone and prazosin in a single oral medication decreases alcohol drinking more effectively than does either drug alone. Alcohol Clin Exp Res 2013; 37:1763-70. [PMID: 23875623 PMCID: PMC3795831 DOI: 10.1111/acer.12148] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 02/25/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Naltrexone (NTX) is underutilized in clinical treatment settings because its efficacy is modest, and it is not effective for all alcoholics and, when it is effective, a significant number of alcoholics fail to maintain initial treatment gains and subsequently relapse to heavy drinking. This has slowed acceptance of NTX by the treatment community, and there is a clear need for additional treatments for alcoholism and alcohol use disorders. Given that NTX and prazosin can each reduce alcohol drinking in rats selectively bred for alcohol preference and high voluntary alcohol drinking (alcohol-preferring "P" rats), we tested whether a combination of NTX + prazosin is more effective in decreasing alcohol drinking than is either drug alone. METHODS P rats were given access to a 15% (v/v) alcohol solution for 2 hours daily. Rats were fed NTX and prazosin, alone or in combination, prior to onset of the daily 2-hour alcohol access period for 4 weeks and the effect of drug treatment on alcohol and water intake was assessed. RESULTS During the first week of treatment, neither a low dose of NTX, nor prazosin, was effective in decreasing alcohol intake when each drug was administered alone, but combining the 2 drugs in a single medication significantly reduced alcohol intake. The combination was as effective as was a higher dose of NTX. Using a low dose of NTX in combination with prazosin may reduce the potential for undesirable side effects early in treatment which, in turn, may improve patient compliance and result in a more successful outcome when NTX is used for treating alcoholism and alcohol use disorders. CONCLUSIONS Combining low-dose NTX and prazosin in a single medication may be more useful than is either drug alone for treating both inpatient and outpatient alcoholics and heavy drinkers early in the treatment process.
Collapse
|
8
|
Youngentob SL, Kent PF, Youngentob LM. Gestational naltrexone ameliorates fetal ethanol exposures enhancing effect on the postnatal behavioral and neural response to ethanol. Exp Biol Med (Maywood) 2012; 237:1197-208. [PMID: 23045720 DOI: 10.1258/ebm.2012.012132] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The association between gestational exposure to ethanol and adolescent ethanol abuse is well established. Recent animal studies support the role of fetal ethanol experience-induced chemosensory plasticity as contributing to this observation. Previously, we established that fetal ethanol exposure, delivered through a dam's diet throughout gestation, tuned the neural response of the peripheral olfactory system of early postnatal rats to the odor of ethanol. This occurred in conjunction with a loss of responsiveness to other odorants. The instinctive behavioral response to the odor of ethanol was also enhanced. Importantly, there was a significant contributory link between the altered response to the odor of ethanol and increased ethanol avidity when assessed in the same animals. Here, we tested whether the neural and behavioral olfactory plasticity, and their relationship to enhanced ethanol intake, is a result of the mere exposure to ethanol or whether it requires the animal to associate ethanol's reinforcing properties with its odor attributes. In this later respect, the opioid system is important in the mediation (or modulation) of the reinforcing aspects of ethanol. To block endogenous opiates during prenatal life, pregnant rats received daily intraperitoneal administration of the opiate antagonist naltrexone from gestational day 6-21 jointly with ethanol delivered via diet. Relative to control progeny, we found that gestational exposure to naltrexone ameliorated the enhanced postnatal behavioral response to the odor of ethanol and postnatal drug avidity. Our findings support the proposition that in utero ethanol-induced olfactory plasticity (and its relationship to postnatal intake) requires, at least in part, the associative pairing between ethanol's odor quality and its reinforcing aspects. We also found suggestive evidence that fetal naltrexone ameliorated the untoward effects of gestational ethanol exposure on the neural response to non-fetal-exposure odorants. Thus, gestational naltrexone may also have a neuroprotective and/or neuroproliferative impact on olfactory development.
Collapse
Affiliation(s)
- Steven L Youngentob
- Department of Psychiatry and Behavioral Sciences, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA.
| | | | | |
Collapse
|
9
|
Ulmer A, Müller M, Frietsch B. Dihydrocodeine/Agonists for alcohol dependents. Front Psychiatry 2012; 3:21. [PMID: 22470353 PMCID: PMC3311265 DOI: 10.3389/fpsyt.2012.00021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 02/28/2012] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Alcohol addiction too often remains insufficiently treated. It shows the same profile as severe chronic diseases, but no comparable, effective basic treatment has been established up to now. Especially patients with repeated relapses, despite all therapeutic approaches, and patients who are not able to attain an essential abstinence to alcohol, need a basic medication. It seems necessary to acknowledge that parts of them need any agonistic substance, for years, possibly lifelong. For >14 years, we have prescribed such substances with own addictive character for these patients. METHODS We present a documented best possible practice, no designed study. Since 1997, we prescribed Dihydrocodeine (DHC) to 102 heavily alcohol addicted patients, later, also Buprenorphine, Clomethiazole (>6 weeks), Baclofen, and in one case Amphetamine, each on individual indication. This paper focuses on the data with DHC, especially. The Clomethiazole-data has been submitted to a German journal. The number of treatments with the other substances is still low. RESULTS The 102 patients with the DHC treatment had 1367 medically assisted detoxifications and specialized therapies before! The 4 years-retention rate was 26.4%, including 2.8% successfully terminated treatments. In our 12-steps scale on clinical impression, we noticed a significant improvement from mean 3.7 to 8.4 after 2 years. The demand for medically assisted detoxifications in the 2 years remaining patients was reduced by 65.5%. Mean GGT improved from 206.6 U/l at baseline to 66.8 U/l after 2 years. Experiences with the other substances are similar but different in details. CONCLUSION Similar to the Italian studies with GHB and Baclofen, we present a new approach, not only with new substances, but also with a new setting and much more trusting attitude. We observe a huge improvement, reaching an almost optimal, stable, long term status in around 1/4 of the patients already. Many further optimizations are possible.
Collapse
Affiliation(s)
- Albrecht Ulmer
- Gemeinschaftspraxis Dres.med. Ulmer, Frietsch, MuellerStuttgart, Germany
| | - Markus Müller
- Gemeinschaftspraxis Dres.med. Ulmer, Frietsch, MuellerStuttgart, Germany
| | - Bernhard Frietsch
- Gemeinschaftspraxis Dres.med. Ulmer, Frietsch, MuellerStuttgart, Germany
| |
Collapse
|
10
|
Pautassi RM, Nizhnikov ME, Acevedo MB, Spear NE. Naloxone blocks ethanol-mediated appetitive conditioning and locomotor activation in adolescent rats. Behav Brain Res 2010; 216:262-9. [PMID: 20708642 DOI: 10.1016/j.bbr.2010.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 07/30/2010] [Accepted: 08/04/2010] [Indexed: 12/20/2022]
Abstract
Age-related differences in ethanol sensitivity could put adolescents at risk for developing alcohol-related problems. Little information exists, however, about adolescent sensitivity to ethanol's appetitive effects and the neurobiological mechanisms underlying ethanol reinforcement during this developmental stage. The present study assessed the role of the opioid system in adolescent rats in an appetitive second-order schedule of ethanol reinforcement and ethanol-induced locomotor stimulation. On postnatal day 32 (PD32), animals were pretreated with the general opioid antagonist naloxone (0.0, 0.75, 1.50, or 2.5 mg/kg) and then given pairings of ethanol (0.0 or 2.0 g/kg, intragastrically) with intraoral pulses of water (conditioned stimulus 1 [CS₁], first-order conditioning phase). CS₁ delivery occurred 30-45 min after ethanol administration when the effect of ethanol was assumed to be appetitive. On PD33, adolescents were exposed to CS₁ (second-order conditioning phase) while in a chamber featuring distinctive exteroceptive cues (CS₂). Preference for CS₂ was then tested. Adolescents given CS₁-ethanol pairings exhibited greater preference for CS₂ than controls, indicating ethanol-mediated reinforcement, but only when not pretreated with naloxone. Blood alcohol levels during conditioning were not altered by naloxone. Experiment 2 revealed that ethanol-induced locomotor activation soon after administration, and naloxone dose-dependently suppressed this stimulating effect. The present study indicates that adolescent rats are sensitive to ethanol's reinforcing and locomotor-stimulating effects. Both effects of ethanol appear to be mediated by endogenous opioid system activation.
Collapse
Affiliation(s)
- Ricardo Marcos Pautassi
- Instituto de Investigación Médica M. y M. Ferreyra (INIMEC-CONICET), Córdoba, C.P. 5000, Argentina.
| | | | | | | |
Collapse
|
11
|
The delta(1) opioid receptor is a heterodimer that opposes the actions of the delta(2) receptor on alcohol intake. Biol Psychiatry 2009; 66:777-84. [PMID: 19576572 PMCID: PMC2757485 DOI: 10.1016/j.biopsych.2009.05.019] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 05/07/2009] [Accepted: 05/08/2009] [Indexed: 12/22/2022]
Abstract
BACKGROUND Opioid receptors are clinically important targets for both pain and alcohol abuse. Three opioid receptors have been cloned: mu, delta, and kappa, all of which effect alcohol consumption in animal models. Naltrexone is a nonselective opioid antagonist used for alcoholism, the clinical utility of which is limited by poor efficacy and adverse side effects. Here, we demonstrate that the therapeutic limitations of naltrexone may reflect its poor selectivity. Despite decades of research, several mysteries surround the pharmacology of these receptors. For example, two pharmacologically defined subtypes of delta receptors exist in vivo. METHODS Effects of delta subtype-selective ligands (naltrindole, naltriben, tan-67, 7-benzylidene naltrexone) were measured on ethanol consumption in C57BL/6 wildtype and opioid receptor knockout mice using a limited access two-bottle choice paradigm. Affinity and efficacy of naltriben, 7-benzylidenenaltrexone and tan-67 was measured in vitro using radioligand binding and Ca(2+)-mobilizationa assays. RESULTS We show that the subtypes of the delta receptor, delta(1) and delta(2), have opposing effects on ethanol consumption. We find that these effects are synergistic; thereby suggesting that delta(1) and delta(2) receptors are distinct molecular targets. Indeed, we provide both in vitro as well as in vivo evidence that the delta(1) subtype is a micro-delta heterodimer and that the delta(2) subtype is most likely a delta homomer. CONCLUSIONS Together these data provide insight into the limited actions of the clinically important drug naltrexone and identify a novel target with improved specificity and efficacy for the development of new therapeutics for the treatment of alcoholism.
Collapse
|
12
|
Pian JP, Criado JR, Walker BM, Ehlers CL. Milk consumption during adolescence decreases alcohol drinking in adulthood. Pharmacol Biochem Behav 2009; 94:179-85. [PMID: 19698741 DOI: 10.1016/j.pbb.2009.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 08/03/2009] [Accepted: 08/06/2009] [Indexed: 10/20/2022]
Abstract
Early onset of alcohol consumption increases the risk for the development of dependence. Whether adolescent consumption of other highly palatable solutions may also affect alcohol drinking in adulthood is not known. The purpose of this study was to determine the effects of adolescent consumption of four solutions: water, sucrose, sucrose-milk and milk on ethanol drinking in adult rats. Rats had limited access to one of the four solutions from day PND 29 to PND 51 and were subsequently trained to consume ethanol (E) using a sucrose (S) fade-out procedure. Adolescent consumption of sucrose and sucrose-milk solutions increased intake of 2.5% E when it was combined with 10% S but it had no effect on the drinking of 10% E alone. Adolescent consumption of milk and sucrose-milk significantly decreased the intake of 10% E when it was combined with 10% S, and milk significantly reduced 10% E consumption alone and when it was combined with 5% S. Adolescent exposure to the sucrose-milk and sucrose solutions was also found to increase sucrose and sucrose-milk consumption. Our findings suggest adolescent exposure to sucrose increases, whereas, exposure to milk reduces ethanol consumption in adult rats. Our results may provide a new theoretical approach to the early prevention of alcoholism.
Collapse
Affiliation(s)
- Jerry P Pian
- Department of Molecular and Integrative Neurosciences, The Scripps Research Institute, 10550 North Torrey Pines Road, SP30-1501, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
13
|
Tampier L, Quintanilla ME. Effect of concurrent saccharin intake on ethanol consumption by high-alcohol-drinking (UChB) rats. Addict Biol 2009; 14:276-82. [PMID: 19413565 DOI: 10.1111/j.1369-1600.2009.00147.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This study examined the effect of concurrent presentation of a highly palatable saccharin solution on ethanol consumption during the acquisition or maintenance of ethanol drinking by high-alcohol-drinking (UChB) rats. Rats were exposed to ethanol (10% v/v) and water under a home cage, two-bottle, free-choice regimen with unlimited access for 24 hours/day. After 7 days (acquisition) of ethanol exposure, a third bottle containing saccharin (0.2% w/v) was concomitantly offered for an additional seven consecutive days, and the same process was repeated after 3 months (maintenance) of ethanol exposure. We found that concurrent saccharin intake significantly reduced ethanol intake by UChB rats after 7 days of ethanol exposure indicating that preference for sweet taste tends to override the preference for ethanol. However, the concurrent saccharin presentation to rats after 3 months of stable ethanol consumption did not reduce ethanol intake, whereas their saccharin consumption reached polydipsic-like values. These results support the notion that in UChB rats, a time-dependent sensitization to the rewarding effects of ethanol is developed that may account for the increases in ethanol volition seen following chronic ethanol intake.
Collapse
Affiliation(s)
- Lutske Tampier
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | | |
Collapse
|
14
|
Pautassi RM, Nizhnikov ME, Spear NE. Assessing appetitive, aversive, and negative ethanol-mediated reinforcement through an immature rat model. Neurosci Biobehav Rev 2009; 33:953-74. [PMID: 19428502 PMCID: PMC2693872 DOI: 10.1016/j.neubiorev.2009.03.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 03/13/2009] [Accepted: 03/13/2009] [Indexed: 11/25/2022]
Abstract
The motivational effects of drugs play a key role during the transition from casual use to abuse and dependence. Ethanol reinforcement has been successfully studied through Pavlovian and operant conditioning in adult rats and mice genetically selected for their ready acceptance of ethanol. Another model for studying ethanol reinforcement is the immature (preweanling) rat, which consumes ethanol and exhibits the capacity to process tactile, odor and taste cues and transfer information between different sensorial modalities. This review describes the motivational effects of ethanol in preweanling, heterogeneous non-selected rats. Preweanlings exhibit ethanol-mediated conditioned taste avoidance and conditioned place aversion. Ethanol's appetitive effects, however, are evident when using first- and second-order conditioning and operant procedures. Ethanol also devalues the motivational representation of aversive stimuli, suggesting early negative reinforcement. It seems that preweanlings are highly sensitive not only to the aversive motivational effects of ethanol but also to its positive and negative (anti-anxiety) reinforcement potential. The review underscores the advantages of using a developing rat to evaluate alcohol's motivational effects.
Collapse
Affiliation(s)
- Ricardo M Pautassi
- Center for Development and Behavioral Neuroscience, State University of New York at Binghamton, Binghamton, NY 13902-6000, USA.
| | | | | |
Collapse
|
15
|
Varlinskaya EI, Spear LP. Ethanol-induced social facilitation in adolescent rats: role of endogenous activity at mu opioid receptors. Alcohol Clin Exp Res 2009; 33:991-1000. [PMID: 19302088 DOI: 10.1111/j.1530-0277.2009.00920.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Ethanol consumption is considerably elevated during adolescence. Attractiveness of alcohol for humans during the adolescent developmental period is based, in part, on its ability to induce social facilitation--a facilitation of social interactions not only evident in human adolescents but also in adolescent rats. Endogenous opioid systems are among the multiple neural systems implicated in the behavioral and reinforcing effects of ethanol and may play a substantial role in modulating stimulatory effects of low doses of ethanol on social behavior during adolescence. This possibility was explored in the present study through the use of an animal model of peer-directed social behavior. METHODS Sprague-Dawley rats were challenged early in adolescence with saline or ethanol intraperitoneally (i.p.), placed into an individual holding cage for 30 minutes, and then tested in a familiar situation with a nonmanipulated partner of the same age and sex. In Experiment 1, each test subject was injected subcutaneously with one of the three doses of a nonselective opioid antagonist naloxone (0, 0.05, and 0.1 mg/kg), 5 minutes prior to the social interaction test and 25 minutes following challenge with saline or ethanol (0.5 g/kg), whereas in Experiment 2 animals were challenged with one of the six doses of ethanol (0, 0.25, 0.5, 0.75, 1.0, and 1.25 g/kg) prior to injection of either saline or naloxone (0.05 mg/kg). In Experiment 3, animals were pretreated i.p. with the selective mu-opioid antagonist CTOP (0, 0.01, 0.025, 0.05, and 0.1 mg/kg) 30 minutes prior to challenge with saline or ethanol (0.5 g/kg). RESULTS Low doses of ethanol (0.5 and 0.75 g/kg) produced social facilitation, as indexed by significant increases in play fighting and social investigation. Both doses of naloxone and the three highest doses of CTOP blocked the stimulatory effects of ethanol on play fighting but not on social investigation. These effects were not associated with alterations in ethanol pharmacokinetic properties or with shifts in the biphasic ethanol dose-response curve. CONCLUSIONS Ethanol-induced facilitation of social play behavior seen in adolescent animals is mediated in part through ethanol-induced release of endogenous ligands for the mu-opioid receptor or an ethanol-associated enhancement of sensitivity of these receptors for their endogenous ligands.
Collapse
Affiliation(s)
- Elena I Varlinskaya
- The Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY 13902-6000, USA.
| | | |
Collapse
|
16
|
Shippenberg TS, LeFevour A, Chefer VI. Targeting endogenous mu- and delta-opioid receptor systems for the treatment of drug addiction. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2008; 7:442-53. [PMID: 19128202 PMCID: PMC3730841 DOI: 10.2174/187152708786927813] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Drug addiction is a chronic, relapsing disorder that is characterized by a compulsion to take drug regardless of the adverse consequences that may ensue. Although the involvement of mesoaccumbal dopamine neurons in the initiation of drug abuse is well-established, neuroadaptations within the limbic cortical- striatopallidal circuit that occur as a consequence of repeated drug use are thought to lead to the behavioral dysregulation that characterizes addiction. Opioid receptors and their endogenous ligands are enriched in brain regions comprising this system and are, thus, strategically located to modulate neurotransmission therein. This article will review data suggesting an important role of mu-opioid receptor (MOPr) and delta opioid receptor (DOPr) systems in mediating the rewarding effects of several classes of abused drugs and that aberrant activity of these opioid systems may not only contribute to the behavioral dysregulation that characterizes addiction but to individual differences in addiction vulnerability.
Collapse
Affiliation(s)
- T S Shippenberg
- Integrative Neuroscience Section, NIH/ NIDA Intramural Research Program, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | | | | |
Collapse
|
17
|
|
18
|
Gilpin NW, Richardson HN, Koob GF. Effects of CRF1-receptor and opioid-receptor antagonists on dependence-induced increases in alcohol drinking by alcohol-preferring (P) rats. Alcohol Clin Exp Res 2008; 32:1535-42. [PMID: 18631323 DOI: 10.1111/j.1530-0277.2008.00745.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Selective breeding of rats over generations and induction of alcohol dependence via chronic vapor inhalation both enhance alcohol consumption in animal models. The purpose of this study was to determine whether dependence-induced increases in alcohol consumption by P rats is sensitive to naltrexone, a general opioid receptor antagonist (but with highest affinity at the mu-opioid receptor at low doses), and the recently characterized small molecule CRF(1)-receptor antagonist MPZP (N,N-bis(2-methoxyethyl)-3-(4-methoxy-2-methylphenyl)-2,5-dimethyl-pyrazolo[1,5-a]pyrimidin-7-amine). METHODS P rats (n = 20) were trained to respond for alcohol and water in a 2-lever operant situation during daily 30-minute sessions. P rats were then matched for alcohol intake and exposed to chronic intermittent alcohol vapor (n = 10) or ambient air (n = 10) for approximately 10 weeks. All rats were then administered MPZP and naltrexone in 2 separate and consecutive Latin-square designs. RESULTS MPZP attenuated dependence-induced increases in alcohol intake by P rats while having no effect on alcohol consumption by nondependent controls. Conversely, operant alcohol responding was reduced similarly in dependent and nondependent P rats by naltrexone. CONCLUSIONS These results confirm a role for brain CRF(1)-receptor systems in dependence-induced changes in the reinforcing properties of alcohol, and CRF(1)-receptor blockade appears to suppress dependence-induced drinking at lower doses in P rats relative to other rat lines. Therefore, brain CRF(1)-receptor systems are important in the regulation of dependence-induced alcohol consumption, whereas brain opioid systems are important in the regulation of basal alcohol consumption by rats.
Collapse
Affiliation(s)
- Nicholas W Gilpin
- Committee on Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | |
Collapse
|
19
|
Time dependent alterations on tyrosine hydroxylase, opioid and cannabinoid CB1 receptor gene expressions after acute ethanol administration in the rat brain. Eur Neuropsychopharmacol 2008; 18:373-82. [PMID: 17964122 DOI: 10.1016/j.euroneuro.2007.09.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2007] [Revised: 08/21/2007] [Accepted: 09/14/2007] [Indexed: 12/20/2022]
Abstract
The aim of this study was to examine the differential regulation after acute ethanol administration on tyrosine hydroxylase, proenkephalin and cannabinoid CB(1) receptor gene expressions in selected areas of the rat brain. Rats received an intragastric administration of 3 g/kg ethanol and were killed by decapitation at 1, 2, 4, 8 and 24 h. The results showed an activation of tyrosine hydroxylase gene expression in the ventral tegmental area and the substantia nigra, increased proenkephalin gene expression in the caudate-putamen, nucleus accumbens core and shell, central and medial amygdala, ventromedial hypothalamic nucleus and the paraventricular hypothalamic nucleus. In contrast, a significant decrease in the cannabinoid CB1 receptor gene expression was found in caudate-putamen, central amygdala and ventromedial hypothalamic nucleus. In conclusion, the results suggest that an acute dose of ethanol induces neuroplastic alterations in proenkephalin, tyrosine hydroxylase and cannabinoid CB1 receptor gene expressions that may contribute to trigger the rewarding effects of ethanol consumption.
Collapse
|
20
|
Kamdar NK, Miller SA, Syed YM, Bhayana R, Gupta T, Rhodes JS. Acute effects of naltrexone and GBR 12909 on ethanol drinking-in-the-dark in C57BL/6J mice. Psychopharmacology (Berl) 2007; 192:207-17. [PMID: 17273875 DOI: 10.1007/s00213-007-0711-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Accepted: 01/13/2007] [Indexed: 10/23/2022]
Abstract
RATIONALE Recently, a simple procedure was described, drinking in the dark (DID), in which C57BL/6J mice self-administer ethanol to the point of intoxication. The test consists of replacing the water with 20% ethanol in the home cage for 2 or 4 h early during the dark phase of the light/dark cycle. OBJECTIVES To determine whether the model displays predictive validity with naltrexone, and whether opioid or dopaminergic mechanisms mediate excessive drinking in the model. MATERIALS AND METHODS Naltrexone or GBR 12909 were administered via intraperitoneal injections immediately before offering ethanol solutions, plain tap water, or 10% sugar water to male C57BL/6J mice, and consumption was monitored over a 2- or 4-h period using the DID procedure. RESULTS Naltrexone (0.5, 1, or 2 mg/kg) dose dependently decreased ethanol drinking but these same doses had no significant effect on the consumption of plain water or 10% sugar water. GBR 12909 (5, 10, and 20 mg/kg) dose dependently reduced the consumption of ethanol and sugar water but had no effect on plain water drinking. CONCLUSIONS The DID model demonstrates predictive validity. Both opioid and dopamine signaling are involved in ethanol drinking to intoxication. Different physiological pathways mediate high ethanol drinking as compared to water or sugar water drinking in DID. DID may be a useful screening tool to find new alcoholism medications and to discover genetic and neurobiological mechanisms relevant to the human disorder.
Collapse
Affiliation(s)
- N K Kamdar
- Department of Psychology, Beckman Institute, Urbana, IL 61801, USA
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
Pharmacological relapse prevention in alcoholism is a rather new clinical field with few drugs being available. Acamprosate, acting predominantly via glutamatergic pathways, and the opioid receptor antagonist naltrexone, were both shown to be efficient in improving rates for continuous abstinence, and not relapsing to heavy drinking in a number of clinical trials and meta-analyses. There are conflicting data on both drugs, especially for acamprosate, according to some recent US studies. However, overall, the evidence is good. Both drugs are approved in most European countries and the US. Efficacy data for disulfiram are mixed; it is a second-line medication compared with other drugs, and is probably most effective when used in a supervised setting. Recently, anticonvulsants including carbamazepine and topiramate have been discussed as possible anti-craving drugs, but there is still limited evidence for their efficacy. Although there is a significant comorbidity for alcoholism with affective disorder, anxiety and schizophrenia, relatively few controlled clinical trials have been performed in this area. Tricyclics have been found to be more effective than serotonin reuptake inhibitors in improving depressive symptoms in these patients.
Collapse
Affiliation(s)
- Michael Soyka
- Private Hospital Meiringen, P.O.Box 612, CH-3860 Meiringen, Switzerland.
| | | |
Collapse
|
22
|
Kampov-Polevoy AB, Eick C, Boland G, Khalitov E, Crews FT. Sweet liking, novelty seeking, and gender predict alcoholic status. Alcohol Clin Exp Res 2004; 28:1291-8. [PMID: 15365298 DOI: 10.1097/01.alc.0000137808.69482.75] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The relationship between a hedonic response to sweet taste and a propensity to excessive alcohol drinking is supported by both animal and human studies. There is evidence indicating that the tendency to rate more concentrated sweet solutions as the most pleasurable (i.e., sweet liking) is associated with the genetic vulnerability to alcoholism. However, sweet liking by itself is insufficient to predict the alcoholic status of the individual. Our previous study indicated that alcoholic status can be predicted by a combination of hedonic response to sweet taste and personality profile as measured by the Tridimensional Personality Questionnaire (TPQ). This study was designed to further test this hypothesis. METHODS Participants were 165 patients admitted to a residential treatment program for the treatment of alcoholism, drug dependence and/or interpersonal problems secondary to substance-abusing family members. In addition to a routine medical examination, on the 24th day after admission, patients completed the TPQ, the standard sweet taste test was conducted, and paternal family history of alcoholism was evaluated. RESULTS Sweet liking was strongly associated with a paternal history of alcoholism. The odds of receiving an alcohol dependence diagnosis were shown to increase, on the average, by 11% for every one-point increase in the TPQ novelty-seeking score in sweet-liking but not in sweet-disliking subjects. Gender contributed independently to the probability of alcohol dependence, with males exhibiting higher rates of alcoholism than females. CONCLUSIONS These findings support the hypothesis that a hedonic response to sweet taste is associated with a genetic risk for alcoholism. Alcoholic status may be predicted by a combination of sweet liking, the TPQ novelty-seeking score, and gender in a mixed group of alcoholic, polysubstance-dependent, and psychiatric patients.
Collapse
Affiliation(s)
- Alexey B Kampov-Polevoy
- Mount Sinai School of Medicine, Division of Addiction Psychiatry, Bronx Veterans Affairs Medical Center, Bronx, New York 10468, USA.
| | | | | | | | | |
Collapse
|
23
|
Chester JA, Blose AM, Zweifel M, Froehlich JC. Effects of stress on alcohol consumption in rats selectively bred for high or low alcohol drinking. Alcohol Clin Exp Res 2004; 28:385-93. [PMID: 15084895 DOI: 10.1097/01.alc.0000117830.54371.7a] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Stress has long been thought to influence the initiation and maintenance of alcohol drinking in humans. However, results of studies in animals suggest that the relationship between stress and alcohol drinking is not well understood. The purpose of this study was to examine the effect of unpredictable and uncontrollable restraint stress on alcohol consumption in two sets of rat lines selectively bred for alcohol preference (P) and high alcohol drinking (HAD1) and for alcohol nonpreference (NP) and low alcohol drinking (LAD1). METHODS Male P (n = 26) and NP (n = 26) and HAD1 (n = 17) and LAD1 (n = 20) rats were counterbalanced on the basis of alcohol intake and assigned, in matched pairs, to either a stress (Stress) or a no-stress (Control) group. All rats were given a free choice between a 10% v/v alcohol solution and water, with food freely available. Unpredictable, uncontrollable stress, which consisted of immobilization in a nylon restraint sleeve for 30 to 120 min/day, was applied for 10 consecutive days. RESULTS Stress moderately reduced alcohol intake in both P and HAD1 rats versus controls and had no effect on alcohol intake in either the NP or the LAD1 rats during the 10 days of stress application. Alcohol intake was increased for the first 5 days after stress termination in P rats but not in HAD1 rats. Alcohol intake remained stable for several weeks in both the NP and LAD1 lines after stress termination and then increased during the last 15 days of the 35-day poststress period in NP rats, but not in LAD1 rats. CONCLUSIONS A reduction in alcohol intake during stress in rats with a genetic predisposition toward high alcohol intake seems to be a moderate but consistent finding, whereas an increase in alcohol intake after stress termination is less consistent and may be influenced by genetic background.
Collapse
Affiliation(s)
- Julia A Chester
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | |
Collapse
|
24
|
Kampov-Polevoy AB, Ziedonis D, Steinberg ML, Pinsky I, Krejci J, Eick C, Boland G, Khalitov E, Crews FT. Association Between Sweet Preference and Paternal History of Alcoholism in Psychiatric and Substance Abuse Patients. Alcohol Clin Exp Res 2003; 27:1929-36. [PMID: 14691380 DOI: 10.1097/01.alc.0000099265.60216.23] [Citation(s) in RCA: 45] [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
BACKGROUND The relationship between preference for stronger sweet solutions and propensity to excessive alcohol drinking is supported by both animal and human studies. This study was designed to test the hypothesis that sweet preference is associated with the genetic risk of alcoholism as measured by a paternal history of alcoholism. METHODS Participants were 180 patients admitted to a residential treatment program for the treatment of alcoholism, drug dependence, or psychiatric conditions. In addition to a routine medical examination, patients completed the standard sweet preference test twice (on the 9th and 24th days after admission), and the family history of alcoholism was evaluated. RESULTS Sweet preference was shown to be stable over time. It was strongly associated with a paternal history of alcoholism, with family history-positive patients approximately 5 times more likely to prefer stronger sweet solutions than family history-negative subjects. Such factors as dependence on alcohol, cocaine, opiates, cannabis, other drugs (including prescription drugs), and tobacco smoking, as well as demographics (gender and age), did not significantly interfere with association between sweet preference and paternal history of alcoholism. CONCLUSIONS These findings provide some support for the hypothesis that preference for stronger sweet solutions is associated with a genetic predisposition to alcoholism as measured by a paternal history of alcoholism.
Collapse
Affiliation(s)
- A B Kampov-Polevoy
- Mount Sinai School of Medicine, Division of Psychiatry, Bronx Veterans Affairs Medical Center, New York 10468, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
BACKGROUND The relationship between a hedonic response to sweet tastes and a propensity to excessive alcohol drinking is supported by both animal and human studies. This study was designed to test the hypothesis that the genetic risk for alcoholism as measured by a paternal history of alcoholism in young social drinkers is associated with sweet-liking, defined as rating the strongest offered sucrose solution (i.e., 0.83 M) as the most palatable during the standard sweet test. METHODS Participants were 163 subjects (39% male) without a lifetime history of alcohol or drug abuse or dependence. Eighty-one subjects had a paternal history of alcoholism (FH+), and 82 did not (FH-). Each subject rated a series of sucrose solutions for intensity of sweetness and palatability. Subjects were categorized as sweet-likers if they rated the highest sucrose concentration as the most pleasurable. RESULTS The estimated odds of being a sweet-liker were 2.5 times higher for FH+ than for FH- subjects. FH+ subjects disliked the tastes of the two weakest offered sucrose concentrations (0.05 and 0.10 M), whereas FH- subjects reported these tastes to be neutral. CONCLUSIONS The results of this study support the hypothesis that sweet-liking is associated with a genetic vulnerability to alcoholism.
Collapse
|
26
|
Le Y, Gagneten S, Larson T, Santha E, Dobi A, v Agoston D, Sauer B. Far-upstream elements are dispensable for tissue-specific proenkephalin expression using a Cre-mediated knock-in strategy. J Neurochem 2003; 84:689-97. [PMID: 12562513 DOI: 10.1046/j.1471-4159.2003.01573.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Several cis-regulatory DNA elements are present in the 5' upstream regulatory region of the enkephalin gene (ENK) promoter. To determine their role in conferring organ-specificity of ENK expression in mice and to circumvent the position effects from random gene insertion that are known to often frustrate such analysis in transgenic mice, we used a Cre-mediated gene knock-in strategy to target reporter constructs to a "safe haven" loxP-tagged locus in the hypoxanthine phosphoribosyltransferase (HPRT) gene. Here we report reliable and reproducible reporter gene expression under the control of the 5' upstream regulatory region of the mouse ENK gene in gene-modified mice using this Cre-mediated knock-in strategy. Comparison of two 5'ENK regulatory regions (one with and the other without known cis-regulatory DNA elements) in the resulting adult mice showed that conserved far-upstream cis-regulatory DNA elements are dispensable for correct organ-specific gene expression. Thus the proximal 1.4 kb of the murine ENK promoter region is sufficient for organ-specificity of ENK gene expression when targeted to a safe-haven genomic locus. These results suggest that conservation of the far-upstream DNA elements serves more subtle roles, such as the developmental or cell-specific expression of the ENK gene.
Collapse
Affiliation(s)
- Yunzheng Le
- Laboratory of Biochemistry and Metabolism, National Institute of Diabetes, Digestive and Kidney Diseases/NIH, Bethesda, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Kuzmin A, Sandin J, Terenius L, Ogren SO. Acquisition, expression, and reinstatement of ethanol-induced conditioned place preference in mice: effects of opioid receptor-like 1 receptor agonists and naloxone. J Pharmacol Exp Ther 2003; 304:310-8. [PMID: 12490606 DOI: 10.1124/jpet.102.041350] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The ability of the two opioid receptor-like receptor 1 (ORL1) agonists nociceptin (5 nmol i.c.v.) and synthetic (1S,3aS)-8-(2,3,3a,4,5,6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza-spiro[4.5]decan-4-one hydrochloride (Ro 64-6198; 0.1, 0.3, and 1.0 mg/kg i.p.) and the opioid antagonist naloxone (0.1, 1.0, and 10.0 mg/kg s.c.) to modify ethanol-induced conditioned place preference was examined in NMRI male mice. The ORL1 agonists were found to significantly reduce the acquisition, expression, and ethanol-induced reinstatement of conditioned place preference. Unlike the ORL1 agonists, naloxone at the doses relevant for opioid receptor blockade failed to significantly influence the acquisition of ethanol-induced conditioned place preference. However, naloxone at 1.0 but not 0.1 mg/kg s.c. potently blocked the expression of ethanol-induced conditioned place preference and significantly inhibited ethanol-induced reinstatement of the conditioned place preference after extinction. Separate experiments indicated that nociceptin and Ro 64-6198 are both devoid of reinforcing or aversive properties. Naloxone, however, at 1.0 and 10.0 mg/kg, produced conditioned place aversion, indicating motivational properties of its own. Both nociceptin and Ro 64-6198 reduced locomotor activity after acute administration. However, tolerance developed very quickly to this effect and already after three i.c.v. (or i.p.) injections, there was no significant reduction of locomotor activity. It is concluded that ORL1 agonists can modulate the acquisition, expression, and reinstatement of the conditioned reinforcing effects of ethanol with no reinforcing or aversive properties of their own. This property might be a potential advantage in the treatment of alcoholism compared with nonselective opioid antagonist naltrexone.
Collapse
Affiliation(s)
- A Kuzmin
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | | | | |
Collapse
|
28
|
Shoemaker WJ, Vavrousek-Jakuba E, Arons CD, Kwok FC. The acquisition and maintenance of voluntary ethanol drinking in the rat: effects of dopaminergic lesions and naloxone. Behav Brain Res 2002; 137:139-48. [PMID: 12445720 DOI: 10.1016/s0166-4328(02)00290-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Wistar male rats were microinfused bilaterally with 6-hydroxydopamine or vehicle into the ventral tegmental area. After recovery, ethanol drinking was established using a sucrose-fading paradigm, i.e. rats were given twice a day access to drinks containing increasing amounts of ethanol and decreasing amounts of sucrose. Mean daily intakes at each ethanol/sucrose concentration were similar irrespective of the level of dopamine depletion that, in some animals, reached 80-90%. The percentage of rats testing as ethanol preferers in a two-bottle choice test also appeared similar in both the lesioned and control groups. After completing the sucrose-fading protocol, all rats were switched to one access per day during which they were presented with a drink containing 10% ethanol with 5% sucrose. Naloxone administration (15 min before the daily access period) decreased ethanol beverage consumption by about 50%, irrespective of the level of dopamine depletion. Total daily water intake was not altered by naloxone. In a two-bottle choice situation, naloxone suppressed intake of an ethanol drink (10% ethanol/5% sucrose), but not the intake of 5% sucrose alone. Thus, a lesion of the dopaminergic cell bodies that results in extensive depletion of dopamine in mesolimbic target regions produced no measurable effect on intake of the sweetened ethanol drinks during the acquisition phase of the sucrose-fading paradigm. Furthermore, during the maintenance phase of drinking, the marked effect of naloxone in inhibiting ethanol beverage ingestion (but not water ingestion or sucrose alone solutions) occurred despite extensive loss of dopaminergic innervation to telencephalic target regions. A preliminary account of these experiments appeared in an abstract form and as an Internet publication. (Supported by NIAAA grants P50-03510 and T32-0720).
Collapse
Affiliation(s)
- William J Shoemaker
- Department of Psychiatry, School of Medicine, University of Connecticut Health Center, Farmington Avenue, , Farmington, CT 06030, USA.
| | | | | | | |
Collapse
|
29
|
Abstract
Is it more than a linguistic accident that the same term, craving, is used to describe intense desires for both foods and for a variety of drugs of abuse? There is strong evidence for common pathways that are affected by most addictive drugs. As the other contributors to this volume will indicate, a strong case can also be made for some shared substrates for food and drug rewards in animals. There has been less explicit work on this topic in humans but many lines of evidence support the common mechanism view: Opioid peptides seem to influence food palatability for humans. There is mounting evidence for comorbidity between drug/alcohol abuse and excessive craving or liking for sweets. Anecdotally, elderly individuals tend to 'age-out' of drug abuse, and the elderly also experience markedly fewer food cravings with age. If we focus on the compulsive aspects of food and drug cravings, there is also evidence for overlap: for example, activity in the orbitofrontal cortex is associated with cocaine and alcohol craving. This area is also implicated in the pathology of obsessive-compulsive disorder. Although there is no direct evidence of orbitofrontal involvement in food cravings, there is indirect evidence such as higher than expected co-occurrence of obsessive-compulsive behavior and eating disorders. As a result of bringing together evidence for common substrates for food and for drug rewards, we hope to be able to advance fundamental knowledge of motivational processes and to promote the development of better treatments for drug addiction and for eating disorders.
Collapse
Affiliation(s)
- Marcia L Pelchat
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
30
|
De A, Boyadjieva N, Sarkar DK. Role of protein kinase C in control of ethanol-modulated beta-endorphin release from hypothalamic neurons in primary cultures. J Pharmacol Exp Ther 2002; 301:119-28. [PMID: 11907165 DOI: 10.1124/jpet.301.1.119] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have previously shown that short-term exposure to ethanol stimulates immunoreactive beta-endorphin (IR-beta-EP) release from hypothalamic neurons and that chronic ethanol exposure decreases the IR-beta-EP release from these neurons. The role of protein kinase C (PKC) in the ethanol-regulated beta-EP release from hypothalamic neurons has not been established. In this study, by using the primary cultures of hypothalamic neurons, we tested the effects of PKC stimulator phorbol ester 4 beta-phorbol 12-myristate-13-acetate (PMA) and PKC inhibitor chelerythrine chloride on ethanol-induced IR-beta-EP release. Additionally, the effects of ethanol with or without PMA on expression and translocation of various PKC isoenzymes from cytosolic to membrane fraction were determined. PMA treatment increased IR-beta-EP release in a time- and dose-dependent manner. Acute ethanol treatment (3 h) increased, while chronic ethanol treatment (24 h) reduced, the magnitude of PMA-induced IR-beta-EP release. The stimulatory effect of acute ethanol on IR-beta-EP release was reduced by chelerythrine chloride. Determination of the effects of ethanol with or without PMA on seven different PKC isoenzymes (PKC-alpha, -beta I, -beta II, -gamma, -delta, -epsilon, and -zeta) revealed that the expression and translocation of only two PKC isoenzymes, PKC-delta and PKC-epsilon, were stimulated by acute treatment with ethanol. Acute ethanol also increased PMA-stimulated expression of these two isoenzymes. Chronic ethanol treatment reduced both basal and PMA-induced increase of PKC-delta and PKC-epsilon expression and translocation. These data provide evidence for the first time that ethanol-regulated IR-beta-EP secretion is controlled by the PKC system, possibly involving PKC-delta and PKC-epsilon isoenzymes.
Collapse
Affiliation(s)
- Alok De
- Department of Animal Sciences, Rutgers, The State University of New Jersey, Cook College, New Brunswick, New Jersey 08901-8525, USA
| | | | | |
Collapse
|
31
|
Rasmussen DD, Boldt BM, Wilkinson CW, Mitton DR. Chronic Daily Ethanol and Withdrawal: 3. Forebrain Pro-Opiomelanocortin Gene Expression and Implications for Dependence, Relapse, and Deprivation Effect. Alcohol Clin Exp Res 2002. [DOI: 10.1111/j.1530-0277.2002.tb02572.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
Chester JA, Price CS, Froehlich JC. Inverse Genetic Association Between Alcohol Preference and Severity of Alcohol Withdrawal in Two Sets of Rat Lines Selected for the Same Phenotype. Alcohol Clin Exp Res 2002. [DOI: 10.1111/j.1530-0277.2002.tb02427.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Kim SG, Stromberg MF, Kim MJ, Volpicelli JR, Park JM. The effect of antagonists selective for mu- and delta-opioid receptor subtypes on alcohol consumption in C57BL/6 mice. Alcohol 2000; 22:85-90. [PMID: 11113622 DOI: 10.1016/s0741-8329(00)00109-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Several studies have demonstrated that non-selective opioid receptor antagonists effectively reduce alcohol consumption in both animal models and at the clinical level. However, research examining the contribution of specific opioid receptor subtypes to this effect has yielded conflicting results. Some of these studies have shown that the effect is contingent upon the action of mu receptors while others have suggested that delta receptors are primarily responsible. The data reported here re-examine this question using the alcohol-preferring C57BL/6 mice. The results of this experiment demonstrate that D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2) (CTOP), a mu-selective antagonist, and naltrindole, a delta-selective antagonist, are equally effective at reducing alcohol consumption in a limited access model compared to a saline control group. While there was no specific comparison of the effects of these drugs on alternative appetitive behavior, neither of these drugs had effects on measured off-session food or water consumption. The results of this experiment suggest that alcohol consumption is mediated by both mu- and delta-opioid receptor subtypes.
Collapse
Affiliation(s)
- S G Kim
- Department of Psychiatry, School of Medicine, Pusan National University, 1-ga 10, Ami-dong, Seo-gu, 602-739, Pusan, South Korea.
| | | | | | | | | |
Collapse
|
34
|
Martin-Fardon R, Ciccocioppo R, Massi M, Weiss F. Nociceptin prevents stress-induced ethanol- but not cocaine-seeking behavior in rats. Neuroreport 2000; 11:1939-43. [PMID: 10884047 DOI: 10.1097/00001756-200006260-00026] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study examined whether nociceptin/orphanin FQ (NC), the endogenous ligand of the opioid receptor-like1 (ORL1) receptor, can block drug-seeking behavior induced by foot-shock stress. Male Wistar rats were trained to operantly self-administer ethanol or cocaine, and then subjected to daily extinction training until responding ceased. Subsequent exposure to 15 min of intermittent footshock elicited robust reinstatement of responding at the previously drug-paired lever. NC (0.1-2.0 microg; i.c.v.) significantly inhibited the effects of footshock stress on ethanol- but not cocaine-seeking behavior. The results support the hypothesis that the NC system participates in the regulation of behavioral responses to stress, and that drugs interacting with NC receptors may have therapeutic potential for the treatment of stress-induced alcohol-seeking behavior and relapse.
Collapse
Affiliation(s)
- R Martin-Fardon
- The Scripps Research Institute CVN-15, Department of Neuropharmacology, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
35
|
Petrakis IL, Trevisan L, D'Souza C, Gil R, Krasnicki S, Webb E, Heninger G, Cooney N, Krystal JH. CSF Monoamine Metabolite and Beta Endorphin Levels in Recently Detoxified Alcoholics and Healthy Controls: Prediction of Alcohol Cue-Induced Craving? Alcohol Clin Exp Res 1999. [DOI: 10.1111/j.1530-0277.1999.tb04355.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Eckardt MJ, File SE, Gessa GL, Grant KA, Guerri C, Hoffman PL, Kalant H, Koob GF, Li TK, Tabakoff B. Effects of moderate alcohol consumption on the central nervous system. Alcohol Clin Exp Res 1998; 22:998-1040. [PMID: 9726269 DOI: 10.1111/j.1530-0277.1998.tb03695.x] [Citation(s) in RCA: 464] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The concept of moderate consumption of ethanol (beverage alcohol) has evolved over time from considering this level of intake to be nonintoxicating and noninjurious, to encompassing levels defined as "statistically" normal in particular populations, and the public health-driven concepts that define moderate drinking as the level corresponding to the lowest overall rate of morbidity or mortality in a population. The various approaches to defining moderate consumption of ethanol provide for a range of intakes that can result in blood ethanol concentrations ranging from 5 to 6 mg/dl, to levels of over 90 mg/dl (i.e., approximately 20 mM). This review summarizes available information regarding the effects of moderate consumption of ethanol on the adult and the developing nervous systems. The metabolism of ethanol in the human is reviewed to allow for proper appreciation of the important variables that interact to influence the level of exposure of the brain to ethanol once ethanol is orally consumed. At the neurochemical level, the moderate consumption of ethanol selectively affects the function of GABA, glutamatergic, serotonergic, dopaminergic, cholinergic, and opioid neuronal systems. Ethanol can affect these systems directly, and/or the interactions between and among these systems become important in the expression of ethanol's actions. The behavioral consequences of ethanol's actions on brain neurochemistry, and the neurochemical effects themselves, are very much dose- and time-related, and the collage of ethanol's actions can change significantly even on the rising and falling phases of the blood ethanol curve. The behavioral effects of moderate ethanol intake can encompass events that the human or other animal can perceive as reinforcing through either positive (e.g., pleasurable, activating) or negative (e.g., anxiolysis, stress reduction) reinforcement mechanisms. Genetic factors and gender play an important role in the metabolism and behavioral actions of ethanol, and doses of ethanol producing pleasurable feelings, activation, and reduction of anxiety in some humans/animals can have aversive, sedative, or no effect in others. Research on the cognitive effects of acute and chronic moderate intake of ethanol is reviewed, and although a number of studies have noted a measurable diminution in neuropsychologic parameters in habitual consumers of moderate amounts of ethanol, others have not found such changes. Recent studies have also noted some positive effects of moderate ethanol consumption on cognitive performance in the aging human. The moderate consumption of ethanol by pregnant women can have significant consequences on the developing nervous system of the fetus. Consumption of ethanol during pregnancy at levels considered to be in the moderate range can generate fetal alcohol effects (behavioral, cognitive anomalies) in the offspring. A number of factors--including gestational period, the periodicity of the mother's drinking, genetic factors, etc.--play important roles in determining the effect of ethanol on the developing central nervous system. A series of recommendations for future research endeavors, at all levels, is included with this review as part of the assessment of the effects of moderate ethanol consumption on the central nervous system.
Collapse
Affiliation(s)
- M J Eckardt
- Office of Scientific Affairs, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Rasmussen DD, Bryant CA, Boldt BM, Colasurdo EA, Levin N, Wilkinson CW. Acute Alcohol Effects on Opiomelanocortinergic Regulation. Alcohol Clin Exp Res 1998. [DOI: 10.1111/j.1530-0277.1998.tb03870.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
38
|
Wang DS, Sternbach G, Varon J. Nalmefene: a long-acting opioid antagonist. Clinical applications in emergency medicine. J Emerg Med 1998; 16:471-5. [PMID: 9610980 DOI: 10.1016/s0736-4679(98)00019-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The use of the opioid antagonist naloxone is well known to the experienced health care provider. The availability of the longer acting opioid antagonist nalmefene has several potential benefits in clinical practice. Nalmefene has a plasma half-life of almost 11 h, compared to 60-90 min for naloxone. Nalmefene has been shown to reverse opioid intoxication for as long as 8 h, reducing the need for continuous monitoring of intoxicated patients and repeated dosing of naloxone. Single dose administration has also been used effectively in the reversal of opiate-assisted conscious sedation. In addition, this agent has been used in the treatment of diseases as diverse as interstitial cystitis and chronic alcohol dependence. However, the long duration of action enables extended withdrawal reactions in the chronically opioid-dependent patient. The prolonged opioid antagonism of nalmefene has several applications in the clinical practice of emergency medicine, and is a useful addition in certain situations to the pharmacologic armamentarium of the practicing emergency physician.
Collapse
Affiliation(s)
- D S Wang
- Division of Emergency Medicine, Stanford University Medical Center, California 94305, USA
| | | | | |
Collapse
|
39
|
Alvarez C, Prunell M, Boada J. Effect of naloxone on behavioral changes induced by subchronic administration of ethanol in rats. Pharmacol Biochem Behav 1998; 59:961-5. [PMID: 9586856 DOI: 10.1016/s0091-3057(97)00505-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endogenous opioid peptides appear to be involved in acute behavioral effects induced by single doses of ethanol. However, its role in repeated ethanol exposure has not been studied. In the present study ethanol was given to rats at the doses of 2 and 4 g/kg by a stomach gauge for 15 days, and its effects on spontaneous motility, open-field activity, and active avoidance behavior recorded on the 3rd, the 6th and the 15th days. Then the effect of naloxone (0.5 and 2 mg/kg by intraperitoneal route) was tested against a challenge ethanol dose, administrated by oral route, on the 16th day. Control animals received tap water and saline instead of ethanol or naloxone, respectively. Both doses of ethanol induced a decrease in spontaneous motility that was antagonized by naloxone. Open-field ambulations were increased by the high dose of ethanol, low-dose lacking effect; naloxone did not modify these ethanol effects. The low dose of ethanol shortened latency time in shuttlebox, the high dose causing escape and freezing responses; none of these effects were modified by naloxone. Therefore, endogenous opioid peptides appear to play a limited role in the chronic effects of ethanol in rats; particularly its effects in tests inducing an increase in the level of anxiety were resistant to naloxone.
Collapse
Affiliation(s)
- C Alvarez
- Department of Physical Medicine and Pharmacology, University of La Laguna, Tenerife (The Canary Islands), Spain
| | | | | |
Collapse
|
40
|
Bormann NM, Cunningham CL. The effects of naloxone on expression and acquisition of ethanol place conditioning in rats. Pharmacol Biochem Behav 1997; 58:975-82. [PMID: 9408203 DOI: 10.1016/s0091-3057(97)00304-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Naloxone has been shown to facilitate extinction of ethanol-induced conditioned place preference (CPP) in mice. The present-study extended these findings by examining naloxone's effect on the expression (Experiment 1) and acquisition (Experiment 2) of place conditioning with ethanol in rats. In Experiment 1, after place conditioning with ethanol (1.8 g/kg, I.P.), groups N0, N1.5, and N10 received 0, 1.5, or 10 mg/kg naloxone before testing. As expected, ethanol produced a robust conditioned place aversion (CPA). However, naloxone had no effect on expression of CPA. In contrast to studies with mice, the endogenous opioid system does not appear to be involved in the conditioned motivational effects of ethanol in rats. In Experiment 2, groups SE1 and SE2, NS(1.5), NE(1.5), and NE(10), received ethanol alone (1.2 g/kg), naloxone alone (1.5 mg/kg), naloxone 1.5 mg/kg plus ethanol, and naloxone 10 mg/kg plus ethanol during acquisition, respectively. All naloxone-treated groups exhibited CPA. Moreover, group NE(1.5) showed a stronger CPA than group NS(1.5). The CPA produced by coadministration of naloxone and ethanol was attributed to naloxone's effects on the neural processes underlying ethanol's unconditioned aversive effects, or to other nonspecific effects on ethanol's motivational properties.
Collapse
Affiliation(s)
- N M Bormann
- Department of Behavioral Neuroscience, Oregon Health Sciences University, Portland 97201-3098, USA
| | | |
Collapse
|
41
|
Overstreet DH, Halikas JA, Seredenin SB, Kampov-Polevoy AB, Viglinskaya KV, Kashevskaya O, Badishtov BA, Knapp DJ, Mormede P, Kiianmaa K, Li TK, Rezvani AH. Behavioral Similarities and Differences among Alcohol-Preferring and -Nonpreferring Rats: Confirmation by Factor Analysis and Extension to Additional Groups. Alcohol Clin Exp Res 1997. [DOI: 10.1111/j.1530-0277.1997.tb03847.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
42
|
Litten RZ, Allen J, Fertig J. Pharmacotherapies for alcohol problems: a review of research with focus on developments since 1991. Alcohol Clin Exp Res 1996; 20:859-76. [PMID: 8865961 DOI: 10.1111/j.1530-0277.1996.tb05264.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Research on medications to treat alcohol problems has flourished in the last 5 years. Whereas before this time most projects focused on withdrawal agents, at least equal interest has now extended to drugs that may directly reduce urge to drink. The most promising medications in this regard are the opiate antagonists and acamprosate. Considerable attention has also been devoted to serotonergic agents. As aids to detoxification, pharmacologic agents that affect the multiple neural systems disrupted by acute alcohol withdrawal remain under active investigation. Significant progress is also being made in identifying medications to assist alcoholics suffering collateral psychopathology, especially depression and anxiety based disorders. Unfortunately, fewer gains have been realized in the development of medications to assist patients simultaneously dependent on both alcohol and illicit drugs. Also, research to develop amethystic agents remains in its very early stages.
Collapse
|
43
|
Affiliation(s)
- K Tsueda
- Department of Anesthesiology, School of Medicine, University of Louisville, Kentucky 40292, USA
| | | | | | | | | | | |
Collapse
|
44
|
Tsueda K, Loyd GE, Heine MF, Duque F, Haas JE, Stocking JE. Opiates in Ethanol Withdrawal. Anesth Analg 1995. [DOI: 10.1213/00000539-199510000-00039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|