1
|
Echeveste Sanchez M, Guerry H, Grady S, Herman M. Sex-Specific Effects in Acute Nicotine Vapor Exposure on Binge Drinking and Activity in the Central Amygdala and Ventral Tegmental Area. Nicotine Tob Res 2024:ntae268. [PMID: 39749458 DOI: 10.1093/ntr/ntae268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/14/2024] [Accepted: 11/11/2024] [Indexed: 01/04/2025]
Abstract
INTRODUCTION The increasing prevalence of electronic nicotine delivery systems and alcohol drinking has led to increases in nicotine and alcohol co-use. However, the impact of ENDs on brain activity and binge drinking behavior is not fully understood. AIMS AND METHODS We subjected female and male C57BL/6J mice to a voluntary drinking and electronic nicotine vapor exposure paradigm. We first established baseline drinking using a drinking-in-the-dark paradigm. In the third week, mice were exposed to 3 hours sessions of electronic nicotine vapor prior to ethanol access, and ethanol and water drinking levels were measured. Following the last nicotine vapor and drinking session we measured blood alcohol levels (BAL) and collected tissue to measure brain activity by cFos expression. RESULTS Nicotine vapor exposure decreased both alcohol and water intake in males, only reduced ethanol intake in females, and did not alter binge alcohol intake in either sex. Nicotine and alcohol increased activity in the central amygdala in females but had no effect in males. In the ventral tegmental area (VTA) nicotine increased activity in females but alcohol did not produce any additional increases. Nicotine and alcohol-exposed males had significantly lower BAL as compared to females, suggesting sex differences in drinking patterns where males appeared to "front-load" and females drank more throughout the end of the session. CONCLUSIONS These results highlight important sex differences in the impact of nicotine and alcohol co-exposure and underscore the importance of further investigating the neurobiological effects of alcohol and nicotine on brain activity and metabolism in males and females. IMPLICATIONS With increasing nicotine and alcohol use, it is imperative to understand the neurobiological and behavioral consequences of co-use with a focus on sex differences. Using passive nicotine vapor inhalation and a binge drinking paradigm, we investigated the effects of nicotine and alcohol co-use in mice. We found sex differences in alcohol intake and brain activity from this poly-substance use approach. Nicotine decreased alcohol and water intake in males, only reduced alcohol intake in females, and did not change binge alcohol drinking. Alcohol and nicotine produced sex-specific increases in the central amygdala and only nicotine increased VTA activity.
Collapse
Affiliation(s)
- Maria Echeveste Sanchez
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hayley Guerry
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shyenne Grady
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Melissa Herman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Venkatesh SK, Stangl BL, Yan J, Quijano Cardé NA, Stein EA, Diazgranados N, Schwandt ML, Sun H, Momenan R, Goldman D, De Biasi M, Ramchandani VA. Smoking-Related Increases in Alcohol Outcomes and Preliminary Evidence for the Protective Effect of a Functional Nicotine Receptor Gene (CHRNA5) Variant on Alcohol Consumption in Individuals Without Alcohol Use Disorder. Int J Neuropsychopharmacol 2024; 27:pyae035. [PMID: 39208422 PMCID: PMC11450629 DOI: 10.1093/ijnp/pyae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Alcohol and nicotine interact with the nicotinic acetylcholine receptor system to alter reward-related responses, thereby contributing to the co-use and misuse of these drugs. A missense polymorphism rs16969968 (G>A) in the CHRNA5 gene has shown a strong association with nicotine-related phenotypes. However, less is known about the impact of this variant on alcohol-related phenotypes. METHODS We assessed the main and interactive effect of smoking and rs16969968 polymorphism on alcohol consumption using the Alcohol Use Disorders Identification Test (AUDIT), Timeline Follow Back (TLFB), and Lifetime Drinking History (LDH) in 980 healthy adults without alcohol use disorder. We further examined the effect of the rs16969968 polymorphism on acute alcohol consumption using a free-access i.v. alcohol self-administration (IV-ASA) human laboratory paradigm in a subset of 153 nonsmoking participants. Subjective alcohol responses, alcohol sensitivity, and expectancy measures were compared between genotype groups (GG; AA/AG). RESULTS We observed a significant association of smoking with AUDIT, TLFB, and LDH measures across genotype groups, with smokers showing higher scores compared with nonsmokers. Additionally, we found an association between genotype and TLFB-total drinks in the IV-ASA subset, with the GG group showing higher scores than AA/AG group. Relatedly, the alcohol negative expectancy score was significantly lower in the GG group than the AA/AG group. CONCLUSIONS Our findings underscore the association of smoking with alcohol measures. We found preliminary evidence for the protective effect of the functional CHRNA5 polymorphism on alcohol consumption and its association with increased negative alcohol expectancies, which highlights the substantial heterogeneity in alcohol responses.
Collapse
Affiliation(s)
- Shyamala K Venkatesh
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Bethany L Stangl
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Jia Yan
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Natalia A Quijano Cardé
- Pharmacology Graduate Group, Biomedical Graduate Studies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elliot A Stein
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Nancy Diazgranados
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Melanie L Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Hui Sun
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Reza Momenan
- Clinical Neuroimaging Research Core, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Mariella De Biasi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vijay A Ramchandani
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Robinson LE, Didier N, Thomas R, Vena A, King A. Combination Treatment With Varenicline and Nicotine Patch on Smoking Cessation Outcomes in Heavy Drinkers at 26-Week Follow-up. J Clin Psychopharmacol 2024; 44:362-368. [PMID: 38752924 PMCID: PMC11211054 DOI: 10.1097/jcp.0000000000001864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
PURPOSE/BACKGROUND People who smoke cigarettes and drink alcohol heavily are less likely to quit smoking compared with those who do not drink heavily. The current study examined the effects of a 12-week treatment phase of combination varenicline and nicotine patch compared with placebo and nicotine patch on smoking cessation (primary outcome) and alcohol consumption (secondary outcome) in heavy drinking smokers at 26-week follow-up. METHODS/PROCEDURES Participants were daily smokers who met heavy drinking criteria. They were randomly assigned to receive either varenicline and nicotine patch (n = 61) or placebo and nicotine patch (n = 61) for 12 weeks. At week 26, self-reports of point prevalence cigarette abstinence were biochemically confirmed, and past-month alcohol drinking days and heavy drinking days were assessed. FINDINGS/RESULTS At week 26, smoking quit rates did not differ by treatment group (25% varenicline and 26% placebo). Relative to week 12 outcomes, week 26 quit rates significantly dropped off in the varenicline group but not in the placebo group. Alcohol drinking reductions for the whole sample that were previously observed from baseline to week 12 were sustained at week 26, although they did not differ between treatment groups. IMPLICATIONS/CONCLUSIONS In heavy drinking smokers, smoking cessation success was evident in a quarter of the total sample at 3 months postmedication discontinuation. At this time, quit rates were the same between those who received varenicline and nicotine patch and those who received nicotine patch alone. Future research is warranted to examine what may aid in longer-term smoking quit rates in heavy drinking smokers.
Collapse
Affiliation(s)
- Layne E Robinson
- From the Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL
| | - Nathan Didier
- From the Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL
| | - Riya Thomas
- From the Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL
| | | | - Andrea King
- From the Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL
| |
Collapse
|
4
|
Duratkar A, Patel R, Jain NS. Neuronal nicotinic acetylcholine receptor of the central amygdala modulates the ethanol-induced tolerance to anxiolysis and withdrawal-induced anxiety in male rats. Behav Pharmacol 2024; 35:132-146. [PMID: 38451025 DOI: 10.1097/fbp.0000000000000770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
The nicotine acetylcholinergic receptor (nAchR) in the central nucleus of the amygdala (CeA) is known to modulate anxiety traits as well as ethanol-induced behavioral effects. Therefore, the present study investigated the role of CeA nAChR in the tolerance to ethanol anxiolysis and withdrawal-induced anxiety-related effects in rats on elevated plus maze (EPM). To develop ethanol dependence, rats were given free access to an ethanol-containing liquid diet for 10 days. To assess the development of tolerance, separate groups of rats were challenged with ethanol (2 g/kg, i.p.) on days 1, 3, 5, 7 and 10 during the period of ethanol exposure, followed by an EPM assessment. Moreover, expression of ethanol withdrawal was induced after switching ethanol-dependent rats to a liquid diet on day 11, and withdrawal-induced anxiety-like behavior was noted at different post-withdrawal time points using the EPM test. The ethanol-dependent rats were pretreated with intra-CeA (i.CeA) (bilateral) injections of nicotine (0.25 µg/rat) or mecamylamine (MEC) (5 ng/rat) before the challenge dose of ethanol on subthreshold tolerance on the 5th day or on peak tolerance day, that is, 7th or 10th, and before assessment of postwithdrawal anxiety on the 11th day on EPM. Bilateral i.CeA preadministration of nicotine before the challenge dose of ethanol on days 5, 7 and 10 exhibited enhanced tolerance, while injection of MEC, completely mitigated the tolerance to the ethanol-induced antianxiety effect. On the other hand, ethanol-withdrawn rats pretreated i.CeA with nicotine exacerbated while pretreatment with MEC, alleviated the ethanol withdrawal-induced anxiety on all time points. Thus, the present investigation indicates that stimulation of nAChR in CeA negatively modulates the ethanol-induced chronic behavioral effects on anxiety in rats. It is proposed that nAChR antagonists might be useful in the treatment of alcohol use disorder and ethanol withdrawal-related anxiety-like behavior.
Collapse
Affiliation(s)
- Antariksha Duratkar
- Department of Pharmacology, J.L. Chaturvedi College of Pharmacy, Nagpur, Maharashtra
| | - Richa Patel
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| | - Nishant Sudhir Jain
- Department of Pharmacology, J.L. Chaturvedi College of Pharmacy, Nagpur, Maharashtra
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Koni, Bilaspur, Chhattisgarh, India
| |
Collapse
|
5
|
Söderpalm B, Ericson M. Alcohol and the dopamine system. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 175:21-73. [PMID: 38555117 DOI: 10.1016/bs.irn.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
The mesolimbic dopamine pathway plays a major role in drug reinforcement and is likely involved also in the development of drug addiction. Ethanol, like most addictive drugs, acutely activates the mesolimbic dopamine system and releases dopamine, and ethanol-associated stimuli also appear to trigger dopamine release. In addition, chronic exposure to ethanol reduces the baseline function of the mesolimbic dopamine system. The molecular mechanisms underlying ethanol´s interaction with this system remain, however, to be unveiled. Here research on the actions of ethanol in the mesolimbic dopamine system, focusing on the involvement of cystein-loop ligand-gated ion channels, opiate receptors, gastric peptides and acetaldehyde is briefly reviewed. In summary, a great complexity as regards ethanol´s mechanism(s) of action along the mesolimbic dopamine system has been revealed. Consequently, several new targets and possibilities for pharmacotherapies for alcohol use disorder have emerged.
Collapse
Affiliation(s)
- Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
6
|
Palombo P, Maeda R, Riberti Zaniboni C, Antonagi Engi S, Yokoyama T, Bonetti Bertagna N, Anesio A, Cristina Bianchi P, Righi T, Emily Boaventura Tavares G, Souccar C, da Silva FBR, Cardoso Cruz F. Unlocking the role of dorsal hippocampal α4β2 nicotinic acetylcholine receptors in Ethanol-Induced conditioned place preference in mice. Neurosci Lett 2024; 824:137666. [PMID: 38331019 DOI: 10.1016/j.neulet.2024.137666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Alcohol Use Disorder (AUD) presents a significant and challenging public health concern, marked by a dearth of effective pharmacological treatments. Understanding the neurobiological underpinnings of AUD is of paramount importance for the development of efficacious interventions. The process of addiction entails the acquisition of associative behaviors, prominently engaging the dorsal region of the hippocampus for encoding these associative memories. Nicotinic receptor systems have been implicated in mediating the rewarding effects of ethanol, as well as memory and learning processes. In our current investigation, we delved into the role of α4β2 nicotinic acetylcholine receptors (nAChRs) within the dorsal hippocampus in the context of ethanol-induced conditioned place preference (CPP), a robust model for scrutinizing the rewarding properties and drug-associated behaviors. To establish CPP, ethanol (2 g/kg) was administered intraperitoneally during a 8-day conditioning phase. Fos immunohistochemistry was employed to assess the involvement of discrete subregions within the dorsal hippocampus in ethanol-induced CPP. Additionally, we probed the influence of α4β2 nAChRs on CPP via microinjections of a selective nAChR antagonist, dihydro-β-erythroidine (DHBE, at dosages of 6, 12, and 18 µg/0.5 µL per hemisphere) within the hippocampus. Our results unveiled that ethanol-induced CPP was associated with an increase Fos -positive cells in various subregions of the dorsal hippocampus, including CA1, CA2, CA3, and the dentate gyrus. Intrahippocampal administration of DHBE (at doses of 6 and 18 µg/0.50 µL per hemisphere) effectively blocked ethanol-induced CPP, while leaving locomotor activity unaffected. These findings underscore the critical involvement of the dorsal hippocampus and α4β2 nAChRs in the acquisition of ethanol-associated learning and reward.
Collapse
Affiliation(s)
- Paola Palombo
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.
| | - Roberta Maeda
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Caroline Riberti Zaniboni
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sheila Antonagi Engi
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Thais Yokoyama
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Natalia Bonetti Bertagna
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Augusto Anesio
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Paula Cristina Bianchi
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Thamires Righi
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Caden Souccar
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Fabio Cardoso Cruz
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Hauser SR, Waeiss RA, Deehan GA, Engleman EA, Bell RL, Rodd ZA. Adolescent alcohol and nicotine exposure alters the adult response to alcohol use. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2023; 3:11880. [PMID: 38389816 PMCID: PMC10880795 DOI: 10.3389/adar.2023.11880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/31/2023] [Indexed: 02/24/2024]
Abstract
Adolescence through young adulthood is a unique period of neuronal development and maturation. Numerous agents can alter this process, resulting in long-term neurological and biological consequences. In the clinical literature, it is frequently reported that adolescent alcohol consumption increases the propensity to develop addictions, including alcohol use disorder (AUD), during adulthood. A general limitation of both clinical and human pre-clinical adolescent alcohol research is the high rate of co-using/abusing more than one drug during adolescence, such as co-using/abusing alcohol with nicotine. A primary goal of basic research is elucidating neuroadaptations produced by adolescent alcohol exposure/consumption that promote alcohol and other drug self-administration in adulthood. The long-term goal is to develop pharmacotherapeutics for the prevention or amelioration of these neuroadaptations. This review will focus on studies that have examined the effects of adolescent alcohol and nicotine exposure on adult alcohol consumption, the hypersensitivity of the mesolimbic dopaminergic system, and enhanced responses not only to alcohol but also to nicotine during adulthood. Again, the long-term goal is to identify potential cholinergic agents to prevent or ameliorate the consequences of, peri-adolescent alcohol abuse.
Collapse
Affiliation(s)
- Sheketha R Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Robert A Waeiss
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Gerald A Deehan
- Department of Psychology, East Tennessee State University, Johnson City, TN, United States
| | - Eric A Engleman
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Richard L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zachary A Rodd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
8
|
Quintanilla ME, Rivera-Meza M, Berríos-Cárcamo P, Cassels BK. Reduction of nicotine and ethanol intake in alcohol-preferring (UChB) female rats by the α4β2 nicotinic acetylcholine receptor partial agonists 5-bromocytisine and cytisine. Drug Alcohol Depend 2023; 250:110900. [PMID: 37515828 DOI: 10.1016/j.drugalcdep.2023.110900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/25/2023] [Accepted: 07/11/2023] [Indexed: 07/31/2023]
Abstract
RATIONALE Neuronal nicotinic acetylcholine receptors (nAChRs) are implicated in the reinforcing effects of nicotine and ethanol. Previous studies have shown that cytisine and its 5-bromo derivative are partial agonists at the α4β2 nAChRs and that the parent molecule cytisine is effective in reducing both nicotine- and ethanol-self-administration in rats. However, whether 5-bromocytisine affects nicotine or ethanol self-administration was unknown. OBJECTIVES The present study compared the effects of 5-bromocytisine and cytisine on nicotine self-administration and further assessed the effect of daily drug injection on voluntary ethanol consumption in alcohol-preferring female rats. Animals were administered a 1.5mg/kg i.p. dose of 5-bromocytisine or cytisine every day for 15-16 days. RESULTS The initial efficacy of 5-bromocytisine and cytisine in reducing nicotine intake was similar (-80%) while for voluntary ethanol intake 5-bromocytisine was a superior inhibitor over cytisine (-78% and -40% respectively). The efficacy of cytisine began to diminish after 10 days of daily administration, which was attributed to tolerance development to its inhibitory effects both on nicotine and ethanol self-administration. Tolerance did not develop for 5-bromocytisine. CONCLUSION 5-Bromocytisine, a weaker α4β2 nAChR partial agonist than cytisine, also produces a sustained inhibition of both nicotine and ethanol self-administration, and unlike cytisine, it does not develop tolerance.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Mario Rivera-Meza
- Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile.
| | - Pablo Berríos-Cárcamo
- Center for Regenerative Medicine, Faculty of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago 7710162, Chile.
| | - Bruce K Cassels
- Department of Chemistry, Faculty of Sciences, University of Chile, Santiago 7800003, Chile.
| |
Collapse
|
9
|
Gheidi A, Fitzpatrick CJ, Gregory JD, Morrow JD. Nicotinic and muscarinic acetylcholine receptor antagonism dose-dependently decreases sign- but not goal-tracking behavior in male rats. Psychopharmacology (Berl) 2023; 240:871-880. [PMID: 36795109 PMCID: PMC10599605 DOI: 10.1007/s00213-023-06328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 01/27/2023] [Indexed: 02/17/2023]
Abstract
RATIONALE Acetylcholinergic antagonists have shown some promise in reducing addiction-related behaviors in both preclinical and clinical studies. However, the psychological mechanisms by which these drugs are able to affect addictive behavior remain unclear. A particular key process for the development of addiction is the attribution of incentive salience to reward-related cues, which can be specifically measured in animals using a Pavlovian conditioned approach procedure. When confronted with a lever that predicts food delivery, some rats engage with the lever directly (i.e., they sign track), indicating attribution of incentive-motivational properties to the lever itself. In contrast, others treat the lever as a predictive cue and approach the location of impending food delivery (i.e., they goal track), without treating the lever itself as a reward. OBJECTIVES We tested whether systemic antagonism of the either nicotinic or muscarinic acetylcholine receptors would selectively affect sign- or goal-tracking behavior, indicating a selective effect on incentive salience attribution. METHODS A total of 98 male Sprague Dawley rats were either given the muscarinic antagonist scopolamine (100, 50, or 10 µg/kg i.p.) or the nicotinic antagonist mecamylamine (0.3, 1.0, or 3 mg/kg i.p.) before being trained on a Pavlovian conditioned approach procedure. RESULTS Scopolamine dose-dependently decreased sign tracking behavior and increased goal-tracking behavior. Mecamylamine reduced sign-tracking but did not affect goal-tracking behavior. CONCLUSIONS Antagonism of either muscarinic or nicotinic acetylcholine receptors can reduce incentive sign-tracking behavior in male rats. This effect appears to be specifically due to a reduction in incentive salience attribution since goal-tracking either increased or was not affected by these manipulations.
Collapse
Affiliation(s)
- Ali Gheidi
- Department of Biomedical Sciences, Mercer University, Macon, USA
| | | | - Jordan D Gregory
- Department of Psychiatry, University of Michigan, Ann Arbor, USA
| | - Jonathan D Morrow
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, USA.
- Department of Psychiatry, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
10
|
Kamens HM, Flarend G, Horton WJ. The role of nicotinic receptors in alcohol consumption. Pharmacol Res 2023; 190:106705. [PMID: 36813094 PMCID: PMC10083870 DOI: 10.1016/j.phrs.2023.106705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/14/2023] [Accepted: 02/19/2023] [Indexed: 02/22/2023]
Abstract
The use of alcohol causes significant morbidity and mortality across the globe. Alcohol use disorder (AUD) is defined by the excessive use of this drug despite a negative impact on the individual's life. While there are currently medications available to treat AUD, they have limited efficacy and several side effects. As such, it is essential to continue to look for novel therapeutics. One target for novel therapeutics is nicotinic acetylcholine receptors (nAChRs). Here we systematically review the literature on the involvement of nAChRs in alcohol consumption. Data from both genetic and pharmacology studies provide evidence that nAChRs modulate alcohol intake. Interestingly, pharmacological modulation of all nAChR subtypes examined can decrease alcohol consumption. The reviewed literature demonstrates that nAChRs should continue to be investigated as novel therapeutics for AUD.
Collapse
Affiliation(s)
- Helen M Kamens
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA 16802, United States.
| | - Geneva Flarend
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA 16802, United States
| | - William J Horton
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA 16802, United States
| |
Collapse
|
11
|
Sved AF, Caggiula AR, Donny EC. Elucidating the reinforcing effects of nicotine: a tribute to Nadia Chaudhri. Psychopharmacology (Berl) 2023; 240:417-430. [PMID: 36329195 PMCID: PMC11188050 DOI: 10.1007/s00213-022-06266-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Nadia Chaudhri worked with us as a graduate student in the Center for Neuroscience at the University of Pittsburgh from 1999 until she earned her PhD in 2005, a time that coincided with the discovery in our lab of the dual reinforcing actions of nicotine, a concept that she played an important role in shaping. The research that was described in her doctoral thesis is among the foundational pillars of the now well-accepted notion that nicotine acts as both a primary reinforcer and an amplifier of other reinforcer stimuli. This reinforcement-enhancing action of nicotine is robust and likely to be a powerful driver of nicotine use. Below, we discuss the evidence that these two actions of nicotine - primary reinforcement and reinforcement enhancement - are distinct and dissociable, a finding that Nadia was closely associated with. We go on to address two other topics that greatly interested Nadia during that time, the generalizability of the reinforcement-enhancing action of nicotine to multiple classes of reinforcing stimuli and potential sex differences in the dual reinforcing actions of nicotine. The research has greatly expanded since Nadia's involvement, but the core ideas that she helped to develop remain central to the concept of the dual reinforcing actions of nicotine and its importance for understanding the drivers of nicotine use.
Collapse
Affiliation(s)
- Alan F Sved
- Departments of Neuroscience, Psychiatry and Psychology and the Center for Neuroscience, University of Pittsburgh, 210 Langley Hall, Pittsburgh, PA, 15260, USA.
| | - Anthony R Caggiula
- Departments of Psychology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Eric C Donny
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| |
Collapse
|
12
|
Straub CJ, Rusali LE, Kremiller KM, Riley AP. What We Have Gained from Ibogaine: α3β4 Nicotinic Acetylcholine Receptor Inhibitors as Treatments for Substance Use Disorders. J Med Chem 2023; 66:107-121. [PMID: 36440853 PMCID: PMC10034762 DOI: 10.1021/acs.jmedchem.2c01562] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
For decades, ibogaine─the main psychoactive alkaloid found in Tabernanthe iboga─has been investigated as a possible treatment for substance use disorders (SUDs) due to its purported ability to interrupt the addictive properties of multiple drugs of abuse. Of the numerous pharmacological actions of ibogaine and its derivatives, the inhibition of α3β4 nicotinic acetylcholine receptors (nAChRs), represents a probable mechanism of action for their apparent anti-addictive activity. In this Perspective, we examine several classes of compounds that have been discovered and developed to target α3β4 nAChRs. Specifically, by focusing on compounds that have proven efficacious in pre-clinical models of drug abuse and have been evaluated clinically, we highlight the promising potential of the α3β4 nAChRs as viable targets to treat a wide array of SUDs. Additionally, we discuss the challenges faced by the existing classes of α3β4 nAChR ligands that must be overcome to develop them into therapeutic treatments.
Collapse
Affiliation(s)
- Carolyn J Straub
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Lisa E Rusali
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Kyle M Kremiller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Andrew P Riley
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| |
Collapse
|
13
|
Echeveste Sanchez M, Quadir SG, Whindleton CM, Hoffman JL, Faccidomo SP, Guhr Lee TN, Esther CR, Hodge CW, Herman MA. The effects of electronic nicotine vapor on voluntary alcohol consumption in female and male C57BL/6 J mice. Drug Alcohol Depend 2022; 241:109676. [PMID: 36343590 PMCID: PMC9979979 DOI: 10.1016/j.drugalcdep.2022.109676] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Alcohol drinking and nicotine vaping often co-occur and dependence on both substances is common. However, the impact of nicotine vaping on alcohol consumption is not fully understood. METHODS We examined the effects of nicotine vaping on ethanol drinking in female and male C57BL/6 J mice using an electronic nicotine delivery system and intermittent access two-bottle choice (IA-2BC) drinking. Mice were exposed to electronic nicotine vapor (3%) or propylene glycol/vegetable glycerol (PG/VG) control for 3 h sessions daily for 4 weeks and voluntary alcohol consumption was monitored. Nicotine vapor exposure was stopped and voluntary alcohol drinking was measured for a 2 week abstinence period. We also examined the effects of alcohol and nicotine on locomotion, temperature, and nicotine metabolism. RESULTS Following acute nicotine vapor exposure, alcohol drinking was increased in males but not in females. Thermoregulation was disrupted following nicotine vapor exposure and voluntary drinking. Male and female mice displayed increased locomotor activity immediately following chronic nicotine vapor exposure, and an anxiolytic effect was seen in males. In nicotine vapor abstinence, female mice displayed increased alcohol consumption. Locomotor activity and anxiolytic effects remained elevated in male but not female mice. Female mice displayed higher levels of serum nicotine and hydroxycotinine, suggesting impaired metabolism following chronic drinking and nicotine vapor exposure. CONCLUSION Collectively, these results suggest that while both male and female ethanol-drinking mice experience the stimulatory effects of nicotine vapor, only in males is there a parallel increase in ethanol drinking and only females display impairments in nicotine metabolism after drinking.
Collapse
Affiliation(s)
- M Echeveste Sanchez
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - S G Quadir
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - C M Whindleton
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - J L Hoffman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - S P Faccidomo
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - T N Guhr Lee
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Division of Pediatric Pulmonology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - C R Esther
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Division of Pediatric Pulmonology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - C W Hodge
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - M A Herman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Neurobiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
14
|
UFR2709, an Antagonist of Nicotinic Acetylcholine Receptors, Delays the Acquisition and Reduces Long-Term Ethanol Intake in Alcohol-Preferring UChB Bibulous Rats. Biomedicines 2022; 10:biomedicines10071482. [PMID: 35884787 PMCID: PMC9312520 DOI: 10.3390/biomedicines10071482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022] Open
Abstract
Alcoholism is a worldwide public health problem with high economic cost and which affects health and social behavior. It is estimated that alcoholism kills 3 million people globally, while in Chile it is responsible for around 9 thousand deaths per year. Nicotinic acetylcholine receptors (nAChRs) are ligand-gated ion channels expressed in the central nervous system, and they were suggested to modulate the ethanol mechanism involved in abuse and dependence. Previous work demonstrated a short-term treatment with UFR2709, a nAChRs antagonist, which reduced ethanol intake using a two-bottle free-choice paradigm in University of Chile bibulous (UChB) rats. Here, we present evidence of the UFR2709 efficacy in reducing the acquisition and long-term ethanol consumption. Our results show that UFR2709 (2.5 mg/kg i.p.) reduces the seek behavior and ethanol intake, even when the drug administration was stopped, and induced a reduction in the overall ethanol intake by around 55%. Using naïve UChB bibulous rats, we demonstrate that UFR2709 could delay and reduce the genetically adaptive impulse to seek and drink ethanol and prevent its excessive intake.
Collapse
|
15
|
King CP, Meyer PJ. The incentive amplifying effects of nicotine: Roles in alcohol seeking and consumption. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 93:171-218. [PMID: 35341566 DOI: 10.1016/bs.apha.2021.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Nicotine has a unique profile among drugs of abuse. To the noninitiated user, nicotine has powerful aversive effects and its relatively weak euphorigenic effects undergo rapid tolerance. Despite this, nicotine is commonly abused despite negative heath consequences, and nicotine users have enormous difficulty quitting. Further, nicotine is one of the most commonly co-abused substances, in that it is often taken in combination with other drugs. One explanation of this polydrug use is that nicotine has multiple appetitive and consummatory conditioning effects. For example, nicotine is a reinforcement enhancer in that it can potently increase the incentive value of other stimuli, including those surrounding drugs of abuse such as alcohol. In addition, nicotine also has a unique profile of neurobiological effects that alter regulation of alcohol intake and interoception. This review discusses the psychological and biological mechanisms surrounding nicotine's appetitive conditioning and consummatory effects, particularly its interactions with alcohol.
Collapse
Affiliation(s)
- Christopher P King
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY, United States; Clinical and Research Institute on Addictions, State University of New York at Buffalo, Buffalo, NY, United States
| | - Paul J Meyer
- Department of Psychology, State University of New York at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
16
|
Angelyn H, Loney GC, Meyer PJ. Nicotine Enhances Goal-Tracking in Ethanol and Food Pavlovian Conditioned Approach Paradigms. Front Neurosci 2021; 15:561766. [PMID: 34483813 PMCID: PMC8416423 DOI: 10.3389/fnins.2021.561766] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
Rationale Nicotine promotes alcohol intake through pharmacological and behavioral interactions. As an example of the latter, nicotine can facilitate approach toward food- and alcohol-associated stimuli ("sign-tracking") in lever-Pavlovian conditioned approach (PavCA) paradigms. However, we recently reported that nicotine can also enhance approach toward locations of reward delivery ("goal-tracking") triggered by ethanol-predictive stimuli when the location of ethanol delivery is non-static (i.e., a retractable sipper bottle). Objective To determine whether the non-static nature of the reward location could have biased the development of goal-tracking in our previous study (Loney et al., 2019); we assessed the effect of nicotine in a lever-PavCA paradigm wherein the location of ethanol delivery was static (i.e., a stationary liquid receptacle). Then, to determine whether nicotine's enhancement of goal-tracking is unique to ethanol-predictive stimuli, we assessed the effect of systemic nicotine on approach triggered by food-predictive stimuli in a lever-PavCA paradigm. Methods Long-Evans rats were used in two PavCA experiments wherein a lever predicted the receipt of ethanol (15% vol/vol; experiment 1) or food (experiment 2) into a stationary receptacle. Prior to testing, rats were administered nicotine (0.4 mg/kg subcutaneously) or saline systemically. Results In both experiments, nicotine increased measures of goal-tracking, but not sign-tracking. Conclusion Nicotine can facilitate approach to reward locations without facilitating approach to reward-predictive stimuli. As such, conceptualization of the mechanisms by which nicotine affects behavior must be expanded to explain an enhancement of goal-tracking by nicotine.
Collapse
Affiliation(s)
- Hailley Angelyn
- Behavioral Neuroscience Program, Department of Psychology, State University of New York at Buffalo, Buffalo, NY, United States
| | - Gregory C Loney
- Behavioral Neuroscience Program, Department of Psychology, State University of New York at Buffalo, Buffalo, NY, United States
| | - Paul J Meyer
- Behavioral Neuroscience Program, Department of Psychology, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
17
|
Domi A, Barbier E, Adermark L, Domi E. Targeting the Opioid Receptors: A Promising Therapeutic Avenue for Treatment in “Heavy Drinking Smokers”. Alcohol Alcohol 2021; 56:127-138. [DOI: 10.1093/alcalc/agaa139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 01/12/2023] Open
Abstract
Abstract
Aims
Despite a general decline in tobacco use in the last decades, the prevalence of tobacco smoking in individuals with alcohol use disorder (AUD) remains substantial (45–50%). Importantly, the co-use of both substances potentiates the adverse effects, making it a significant public health problem. Substantial evidence suggests that AUD and Tobacco use disorder (TUD) may share common mechanisms. Targeting these mechanisms may therefore provide more effective therapy. Numerous studies describe a potential role of the endogenous opioid system in both AUD and TUD. Reviewing this literature, we aim to evaluate the efficacy of molecules that target the opioid system as promising therapeutic interventions for treating alcohol and tobacco co-use disorders.
Methods
We provide a synthesis of the current epidemiological knowledge of alcohol and tobacco co-use disorders. We evaluate clinical and preclinical research that focuses on the regulation of the endogenous opioid system in alcohol, nicotine, and their interactions.
Results
The epidemiological data confirm that smoking stimulates heavy drinking and facilitates alcohol craving. Pharmacological findings suggest that treatments that are efficacious in the dual addiction provide a beneficial treatment outcome in comorbid AUD and TUD. In this regard, MOP, DOP and NOP-receptor antagonists show promising results, while the findings prompt caution when considering KOP-receptor antagonists as a treatment option in alcohol and tobacco co-use disorders.
Conclusions
Existing literature suggests a role of the opioid system in sustaining the high comorbidity rates of AUD and TUD. Molecules targeting opioid receptors may therefore represent promising therapeutic interventions in ‘heavy drinking smokers.’
Collapse
Affiliation(s)
- Ana Domi
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy University of Gothenburg, Box 410, Gothenburg 405 30, Sweden
| | - Estelle Barbier
- Center for Social and Affective Neuroscience, Linköping University, Campus US, Entrance 65, Linköping 581 85, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy University of Gothenburg, Box 410, Gothenburg 405 30, Sweden
| | - Esi Domi
- Center for Social and Affective Neuroscience, Linköping University, Campus US, Entrance 65, Linköping 581 85, Sweden
| |
Collapse
|
18
|
Montanari C, Secci ME, Driskell A, McDonald KO, Schratz CL, Gilpin NW. Chronic nicotine increases alcohol self-administration in adult male Wistar rats. Psychopharmacology (Berl) 2021; 238:201-213. [PMID: 33000333 PMCID: PMC7796964 DOI: 10.1007/s00213-020-05669-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/21/2020] [Indexed: 12/25/2022]
Abstract
RATIONALE Alcohol and nicotine co-dependence is common in humans, and nicotine increases alcohol drinking in humans without alcohol use disorder (AUD). Nevertheless, there is little basic research on the interactions between the reinforcing effects of these two drugs. OBJECTIVES The aim of this study was to investigate the effects of chronic nicotine injections on oral alcohol self-administration in alcohol non-dependent rats. METHODS After stable alcohol self-administration was reached (baseline) and a period without alcohol access, adult male rats were treated with chronic nicotine or saline injections for 105 days during which time they were tested intermittently for alcohol self-administration. There were 3 experimental groups: (1) saline, rats treated with saline for 105 days; (2) early nicotine, rats treated with nicotine for 70 days, and then with saline for 35 days; and (3) late nicotine: rats treated with saline for 35 days, and then with nicotine for 70 days. RESULTS Our results indicate that (1) chronic nicotine increases alcohol consumption regardless of whether exposure to alcohol was interrupted (early nicotine) or not (late nicotine) before the start of nicotine treatment, (2) the number of alcohol reinforcements correlates to blood-alcohol levels, and (3) alcohol self-administration rapidly decreases when nicotine is no longer available (early nicotine). CONCLUSIONS These discoveries may have clinical implications in social drinkers that use nicotine products, in that chronic nicotine can escalate alcohol drinking and cessation of nicotine exposure may decrease alcohol use.
Collapse
Affiliation(s)
- Christian Montanari
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
| | - Maria E Secci
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ashlyn Driskell
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Katherine O McDonald
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Connor L Schratz
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Nicholas W Gilpin
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.,Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.,Alcohol & Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.,Southeast Louisiana VA Healthcare System (SLVHCS), New Orleans, LA, 70119, USA
| |
Collapse
|
19
|
Miller CN, Kamens HM. The role of nicotinic acetylcholine receptors in alcohol-related behaviors. Brain Res Bull 2020; 163:135-142. [PMID: 32707263 DOI: 10.1016/j.brainresbull.2020.07.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/23/2020] [Accepted: 07/17/2020] [Indexed: 12/29/2022]
Abstract
Alcohol use disorder (AUD) causes an alarming economic and health burden in the United States. Unfortunately, this disease does not exist in isolation; AUD is highly comorbid with nicotine use. Results from both human and animal models demonstrate a genetic correlation between alcohol and nicotine behaviors. These data support the idea of shared genetic and neural mechanisms underlying these behaviors. Nicotine acts directly at nicotinic acetylcholine receptors (nAChR) to have its pharmacological effect. Interestingly, alcohol also acts both directly and indirectly at these receptors. Research utilizing genetically engineered rodents and pharmacological manipulations suggest a role for nAChR in several ethanol behaviors. The current manuscript collates this literature and discusses findings that implicate specific nAChR subunits in ethanol phenotypes. These data suggest future directions for targeting nAChR as novel therapeutics for AUD.
Collapse
Affiliation(s)
- C N Miller
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, 16802, United States
| | - H M Kamens
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, 16802, United States.
| |
Collapse
|
20
|
Ozturk B, Pogun S, Kanit L. Increased alcohol preference and intake in nicotine-preferring rats. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2020; 46:408-420. [PMID: 31860364 DOI: 10.1080/00952990.2019.1695808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Alcohol and tobacco are among the leading substances that are misused together and shared genetic vulnerability is likely. Increased susceptibility to nicotine self-administration has been shown in alcohol-preferring rat-lines. However, a nicotine-preferring (nP) rat-line has not been studied for alcohol preference. OBJECTIVES To evaluate alcohol preference and intake in male and female nP rats. We hypothesized that nP rats and females would drink more ethanol than control rats and males, respectively. METHODS nP rats are being selectively outbred for high oral nicotine intake at Ege University. Seventeen nP (18th generation) and 20 naïve female and male SD rats, not previously exposed to alcohol or nicotine, were used. Twelve-week-old rats were given intermittent access to 20% ethanol in a 2-bottle-choice-procedure for six weeks. After one week withdrawal, six weeks of oral nicotine self-administration was applied. RESULTS nP rats drank significantly more ethanol than controls and their preference for ethanol over water was higher. Female rats' ethanol intake was higher than males'. The nP rats' nicotine preference and intake were higher than controls, and they gained less weight. CONCLUSION We have shown for the first time that nP rats also have high alcohol intake. Our results support the hypothesis that shared genetic factors may underlie concurrent addiction to nicotine and alcohol and have translational value in understanding their misuse. Considering the increased vulnerability for alcohol use disorder in smokers and sex differences observed, early preventive measures in families with a history of tobacco addiction, specifically targeting female members, could have public health benefits.
Collapse
Affiliation(s)
- Baran Ozturk
- Center for Brain Research and School of Medicine Department of Physiology, Ege University , Izmir, Turkey
| | - Sakire Pogun
- Center for Brain Research and School of Medicine Department of Physiology, Ege University , Izmir, Turkey
| | - Lutfiye Kanit
- Center for Brain Research and School of Medicine Department of Physiology, Ege University , Izmir, Turkey
| |
Collapse
|
21
|
Junghanns K, Backhaus J, Tietz U, Lange W, Rink L, Wetterling T, Driessen M. The consumption of cigarettes, coffee and sweets in detoxified alcoholics and its association with relapse and a family history of alcoholism. Eur Psychiatry 2020; 20:451-5. [PMID: 16171658 DOI: 10.1016/j.eurpsy.2004.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2003] [Revised: 06/14/2004] [Accepted: 08/25/2004] [Indexed: 10/26/2022] Open
Abstract
AbstractThirty male alcohol dependent inpatients without concurrent depressive disorder, 13 of them with a positive family history of alcohol dependence in a first degree relative (PFH), were questioned about their desire and consumption habits with respect to cigarettes, coffee, and sweets while on a three-week inpatient treatment after detoxification from alcohol. Six weeks after discharge from hospital, the patients were reassessed for relapse. Eleven patients (36.6%) had relapsed at follow-up. Relapsers were younger than abstainers. The days until relapse correlated negatively with intensity of desire to drink alcohol, desire to smoke cigarettes, and with a higher consumption of cigarettes. PFH patients did not relapse earlier but they had a stronger desire to drink coffee and eat sweets and had a higher coffee consumption.
Collapse
Affiliation(s)
- Klaus Junghanns
- Department of Psychiatry and Psychotherapy, University of Luebeck, Ratzeburger Allee 160, D-23538 Luebeck, Germany.
| | | | | | | | | | | | | |
Collapse
|
22
|
Unequal interactions between alcohol and nicotine co-consumption: suppression and enhancement of concurrent drug intake. Psychopharmacology (Berl) 2020; 237:967-978. [PMID: 31858160 PMCID: PMC7124972 DOI: 10.1007/s00213-019-05426-6] [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] [Received: 07/19/2019] [Accepted: 12/06/2019] [Indexed: 12/26/2022]
Abstract
RATIONALE Alcohol and nicotine addiction are prevalent conditions that co-occur. Despite the prevalence of co-use, factors that influence the suppression and enhancement of concurrent alcohol and nicotine intake are largely unknown. OBJECTIVES Our goals were to assess how nicotine abstinence and availability influenced concurrent alcohol consumption and to determine the impact of quinine adulteration of alcohol on aversion-resistant alcohol consumption and concurrent nicotine consumption. METHODS Male and female C57BL/6J mice voluntarily consumed unsweetened alcohol, nicotine, and water in a chronic 3-bottle choice procedure. In experiment 1, nicotine access was removed for 1 week and re-introduced the following week, while the alcohol and water bottles remained available at all times. In experiment 2, quinine (100-1000 μM) was added to the 20% alcohol bottle, while the nicotine and water bottles remained unaltered. RESULTS In experiment 1, we found that alcohol consumption and preference were unaffected by the presence or absence of nicotine access in both male and female mice. In experiment 2a, we found that quinine temporarily suppressed alcohol intake and enhanced concurrent nicotine, but not water, preference in both male and female mice. In experiment 2b, chronic quinine suppression of alcohol intake increased nicotine consumption and preference in female mice without affecting water preference, whereas it increased water and nicotine preference in male mice. CONCLUSIONS Quinine suppression of alcohol consumption enhanced the preference for concurrent nicotine preference in male and female mice, suggesting that mice compensate for the quinine adulteration of alcohol by increasing their nicotine preference.
Collapse
|
23
|
Barrett ST, Thompson BM, Emory JR, Larsen CE, Pittenger ST, Harris EN, Bevins RA. Sex Differences in the Reward-Enhancing Effects of Nicotine on Ethanol Reinforcement: A Reinforcer Demand Analysis. Nicotine Tob Res 2020; 22:238-247. [PMID: 30982885 PMCID: PMC7297098 DOI: 10.1093/ntr/ntz056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 04/10/2019] [Indexed: 11/13/2022]
Abstract
BACKGROUND Alcohol is often consumed with tobacco, and dependence to alcohol and tobacco are highly comorbid. In addition, there are differences in the prevalence of nicotine- and alcohol-abuse between the sexes. Nicotine produces enhancing effects on the value of other reinforcers, which may extend to alcohol. METHODS Male and female Wistar rats were trained to self-administer 15% ethanol solution in 30-minute sessions. Once ethanol self-administration was established, demand for ethanol was evaluated using an exponential reinforcer demand method, in which the response cost per reinforcer delivery was systematically increased over blocks of several sessions. Within each cost condition, rats were preinjected with nicotine (0.05, 0.1, 0.2, or 0.4 mg/kg base, SC) or saline 5 minutes before self-administration sessions. The effects of nicotine dose and biological sex were evaluated using the estimates generated by the reinforcer demand model. RESULTS Under saline conditions, males showed greater sensitivity to ethanol reinforcement than females. Nicotine enhanced the reinforcement value of alcohol and this varied with sex. In both sexes, 0.4 mg/kg nicotine decreased intensity of ethanol demand. However, 0.05, 0.1, and 0.2 mg/kg nicotine decreased elasticity of ethanol demand in females, but not in males. CONCLUSIONS Nicotine enhances ethanol reinforcement, which may partially drive comorbidity between nicotine-abuse and alcohol-abuse. Males showed signs of greater ethanol reinforcement value than females under saline conditions, and nicotine attenuated this effect by increasing ethanol reinforcement value in the females. These findings highlight that a complete understanding of alcohol-abuse must include a thorough study of alcohol use in the context of other drug use, including nicotine. IMPLICATIONS Nicotine dose dependently enhances the alcohol reinforcement value in a manner that is clearly influenced by biological sex. Under saline baseline conditions, males show lower elasticity of demand for alcohol reinforcement than females, indicative of greater reinforcement value. However, nicotine attenuated this difference by enhancing alcohol reward in the females. Specifically, low-to-moderate doses (0.05-0.2 mg/kg) of nicotine decreased elasticity of alcohol demand in female rats, increasing the perseverance of their alcohol taking behavior. These data indicate that the well-documented reward-enhancing effects of nicotine on sensory reinforcement extend to alcohol reinforcement and that these vary with biological sex.
Collapse
Affiliation(s)
- Scott T Barrett
- Department of Psychology, University of Nebraska–Lincoln, Lincoln, NE
| | - Brady M Thompson
- Department of Psychology, University of Nebraska–Lincoln, Lincoln, NE
| | - Jessica R Emory
- Department of Psychology, University of Nebraska–Lincoln, Lincoln, NE
| | - Chris E Larsen
- Department of Psychology, University of Nebraska–Lincoln, Lincoln, NE
| | | | - Edward N Harris
- Department of Biochemistry, University of Nebraska–Lincoln, Lincoln, NE
| | - Rick A Bevins
- Department of Psychology, University of Nebraska–Lincoln, Lincoln, NE
| |
Collapse
|
24
|
Ren M, Lotfipour S. Nicotine Gateway Effects on Adolescent Substance Use. West J Emerg Med 2019; 20:696-709. [PMID: 31539325 PMCID: PMC6754186 DOI: 10.5811/westjem.2019.7.41661] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 04/15/2019] [Accepted: 07/01/2019] [Indexed: 11/11/2022] Open
Abstract
Given the rise in teenage use of electronic nicotine delivery systems ("vaping") in congruence with the increasing numbers of drug-related emergencies, it is critical to expand the knowledge of the physical and behavioral risks associated with developmental nicotine exposure. A further understanding of the molecular and neurochemical underpinnings of nicotine's gateway effects allows emergency clinicians to advise patients and families and adjust treatment accordingly, which may minimize the use of tobacco, nicotine, and future substances. Currently, the growing use of tobacco products and electronic cigarettes among teenagers represents a major public health concern. Adolescent exposure to tobacco or nicotine can lead to subsequent abuse of nicotine and other substances, which is known as the gateway hypothesis. Adolescence is a developmentally sensitive time period when risk-taking behaviors, such as sensation seeking and drug experimentation, often begin. These hallmark behaviors of adolescence are largely due to maturational changes in the brain. The developing brain is particularly vulnerable to the harmful effects of drugs of abuse, including tobacco and nicotine products, which activate nicotinic acetylcholine receptors (nAChRs). Disruption of nAChR development with early nicotine use may influence the function and pharmacology of the receptor subunits and alter the release of reward-related neurotransmitters, including acetylcholine, dopamine, GABA, serotonin, and glutamate. In this review, we emphasize that the effects of nicotine are highly dependent on timing of exposure, with a dynamic interaction of nAChRs with dopaminergic, endocannabinoid, and opioidergic systems to enhance general drug reward and reinforcement. We analyzed available literature regarding adolescent substance use and nicotine's impact on the developing brain and behavior using the electronic databases of PubMed and Google Scholar for articles published in English between January 1968 and November 2018. We present a large collection of clinical and preclinical evidence that adolescent nicotine exposure influences long-term molecular, biochemical, and functional changes in the brain that encourage subsequent drug abuse.
Collapse
Affiliation(s)
- Michelle Ren
- University of California, Irvine, Department of Pharmaceutical Sciences, Irvine, California
| | - Shahrdad Lotfipour
- University of California, Irvine, Department of Emergency Medicine and Pharmaceutical Sciences, Irvine, California
| |
Collapse
|
25
|
Troisi JR. Ethanol→Nicotine & Nicotine→Ethanol drug-sequence discriminations: Conditional stimulus control with two interoceptive drug elements in rats. Alcohol 2019; 77:125-134. [PMID: 30408489 PMCID: PMC6500766 DOI: 10.1016/j.alcohol.2018.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/25/2018] [Accepted: 10/29/2018] [Indexed: 10/27/2022]
Abstract
Self-administration of ethanol (E) and nicotine (N) occurs frequently in tandem orders (i.e., N→E vs. E→N) and thereby produces differing interoceptive profiles of subjective effects in humans. If the interoceptive stimulus characteristics of N→E differ from E→N, it is possible that such differences contribute to their co-dependence. The rationale for the present investigation was to determine whether ethanol, when preceded or followed by nicotine, produces different discriminative stimulus effects in rats. In two experiments, using a one-manipulandum operant drug discrimination procedure, rats were trained to discriminate temporal sequential administrations of ethanol (1.0 g/kg) that was followed or preceded by nicotine (0.3 mg/kg). Sessions alternated between food-reinforcement sessions on a variable interval 30-sec schedule (i.e., SD) and non-reinforcement sessions (i.e., SΔ). In Experiment 1, administrations of ethanol were followed or preceded by a 10-min interval of nicotine. Training sessions took place 10 min following the second drug injection. Four groups of rats were trained to discriminate only one sequence from sequential administrations of saline, and each drug sequence was counterbalanced across groups for their roles as SD or SΔ. There was robust stimulus control. N→E and E→N functioned equally well as SD or SΔ. Experiment 2 used two groups of rats. For one group, the E→N sequence functioned as the SD and the N→E sequence functioned as the SΔ. The drug sequences were counterbalanced for the other group. Brief non-reinforcement tests revealed significantly greater responding during the SD sequence compared to the SΔ sequence for both groups. These results suggest that different drug sequences of ethanol followed or preceded by nicotine established reliable discriminative stimulus control over operant responding, potentially because of characteristic differences in the overlapping pharmacokinetic profiles of the NE compound. The results are discussed in terms of: 1) conditional stimulus control among two interoceptive drug states; and 2) the clinical modulation of human alcohol consumption and tobacco smoking.
Collapse
Affiliation(s)
- Joseph R Troisi
- Department of Psychology, Saint Anselm College, Manchester, NH, United States.
| |
Collapse
|
26
|
Bold KW, Zweben A, Fucito LM, Piepmeier ME, Muvvala S, Wu R, Gueorguieva R, O'Malley SS. Longitudinal Findings from a Randomized Clinical Trial of Varenicline for Alcohol Use Disorder with Comorbid Cigarette Smoking. Alcohol Clin Exp Res 2019; 43:937-944. [PMID: 30817018 DOI: 10.1111/acer.13994] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/20/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND This study is the first to examine longitudinal posttreatment outcomes of a placebo-controlled trial of varenicline for alcohol use disorder (AUD) with comorbid cigarette smoking. METHODS Participants were 131 adults (n = 39 female) seeking alcohol treatment in a randomized, double-blind, parallel group, placebo-controlled, 16-week multisite trial of varenicline combined with medical management (MM). Timeline follow-back assessments of alcohol and smoking behavior were conducted at the end of treatment (4 months), with follow-ups at 6, 9, and 12 months. Outcomes were percentage of heavy drinking days (PHDD), percent of participants with no heavy drinking days (NHDD), cotinine-confirmed prolonged smoking abstinence (PA), and good clinical outcome on either NHDD or PA. RESULTS Treatment improvements were maintained posttreatment. For the sample overall, PHDD or NHDD did not differ significantly by treatment condition (ps > 0.13), but varenicline produced higher rates of PA versus placebo at 4, 9, and 12 months (p < 0.05). Significant differences were observed by sex: Males had higher rates of NHDD with varenicline (28.9%) versus placebo (6.4%) at the end of treatment (p = 0.004), and these effects were maintained at 12 months (varenicline: 40.0% vs. placebo: 19.2%, p = 0.03). Higher rates of PA were seen for varenicline in both males (8.9%) and females (21.1%) versus placebo (males/females: 0%) at the end of treatment (p = 0.05), and this effect was maintained at 12 months for females (varenicline: 21.1% vs. placebo, 0.0%, p = 0.05). CONCLUSIONS Varenicline treatment combined with MM appears to have enduring benefits for patients with co-occurring AUD and cigarette smoking, and these effects may differ by sex.
Collapse
Affiliation(s)
- Krysten W Bold
- Department of Psychiatry , Yale School of Medicine, New Haven, Connecticut
| | - Allen Zweben
- School of Social Work , Columbia University, New York, New York
| | - Lisa M Fucito
- Department of Psychiatry , Yale School of Medicine, New Haven, Connecticut
| | | | - Srinivas Muvvala
- Department of Psychiatry , Yale School of Medicine, New Haven, Connecticut
| | - Ran Wu
- Department of Psychiatry , Yale School of Medicine, New Haven, Connecticut
| | - Ralitza Gueorguieva
- Department of Psychiatry , Yale School of Medicine, New Haven, Connecticut.,Department of Biostatistics , Yale School of Public Health, New Haven, Connecticut
| | | |
Collapse
|
27
|
Quiroz G, Guerra-Díaz N, Iturriaga-Vásquez P, Rivera-Meza M, Quintanilla ME, Sotomayor-Zárate R. Erysodine, a competitive antagonist at neuronal nicotinic acetylcholine receptors, decreases ethanol consumption in alcohol-preferring UChB rats. Behav Brain Res 2018; 349:169-176. [PMID: 29704599 DOI: 10.1016/j.bbr.2018.04.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 12/20/2022]
Abstract
Alcohol abuse is a worldwide health problem with high economic costs to health systems. Emerging evidence suggests that modulation of brain nicotinic acetylcholine receptors (nAChRs) may be a therapeutic target for alcohol dependence. In this work, we assess the effectiveness of four doses of erysodine (1.5, 2.0, 4.0 or 8.0 mg/kg/day, i.p.), a competitive antagonist of nAChRs, on voluntary ethanol consumption behavior in alcohol-preferring UChB rats, administered during three consecutive days. Results show that erysodine administration produces a dose-dependent reduction in ethanol consumption respect to saline injection (control group). The highest doses of erysodine (4 and 8 mg/kg) reduce (45 and 66%, respectively) the ethanol intake during treatment period and first day of post-treatment compared to control group. While, the lowest doses of erysodine (1.5 and 2 mg/kg) only reduce ethanol intake during one day of treatment period. These effective reductions in ethanol intake were 23 and 29% for 1.5 and 2 mg/kg erysodine, respectively. Locomotor activity induced by a high dose of erysodine (10 mg/kg) was similar to those observed with saline injection in control rats, showing that the reduction in ethanol intake was not produced by hypolocomotor effect induced by erysodine. This is the first report showing that erysodine reduces ethanol intake in UChB rats in a dose-dependent manner. Our results highlight the role of nAChRs in the reward effects of ethanol and its modulation as a potentially effective pharmacological alternative for alcohol dependence treatment.
Collapse
Affiliation(s)
- Gabriel Quiroz
- Programa de Doctorado en Farmacología, Universidad de Chile, Santiago, Chile
| | - Nicolás Guerra-Díaz
- Programa de Doctorado en Química, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Patricio Iturriaga-Vásquez
- Laboratorio de Farmacoquímica, Departamento de Ciencias Químicas y Recursos Naturales, Facultad de Ingeniería y Ciencias, Universidad de la Frontera, Temuco, Chile
| | - Mario Rivera-Meza
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - María Elena Quintanilla
- Programa de Farmacología Molecular y Clínica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Ramón Sotomayor-Zárate
- Laboratorio de Neuroquímica y Neurofarmacología, CENFI, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
28
|
Cippitelli A, Brunori G, Schoch J, Armishaw CJ, Wu J, Zaveri NT, Giulianotti MA, Welmaker GS, Toll L. Differential regulation of alcohol taking and seeking by antagonism at α4β2 and α3β4 nAChRs. Psychopharmacology (Berl) 2018; 235:1745-1757. [PMID: 29572652 PMCID: PMC5949259 DOI: 10.1007/s00213-018-4883-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 03/13/2018] [Indexed: 12/17/2022]
Abstract
RATIONALE Alcoholism is a serious public health problem throughout the world. Current pharmacotherapies for the treatment of this disorder are poorly effective. Preclinical and clinical findings point to nicotinic acetylcholine receptors (nAChRs) as a promising target for the development of novel and effective medications. Assuage Pharmaceuticals, in collaboration with Torrey Pines Institute for Molecular Studies, has discovered a new class of potent and selective α4β2 nAChR antagonists. OBJECTIVE Here, it was hypothesized that α4β2 nAChR antagonism is a viable approach for treatment of alcohol use disorders. RESULTS When tested in rats, one lead compound, AP-202, attenuated both operant alcohol and nicotine self-administration in a paradigm in which the two reinforcers were concurrently available. The conotoxin TP2212-59, a selective α3β4 nAChR antagonist, was only effective in reducing nicotine self-administration. AP-202 also reduced alcohol but not food responding when alcohol was presented as the only reinforcer, whereas the commercially available α4β2 nAChR antagonist dihydro-β-erythroidine failed to alter alcohol self-administration. AP-202 did not block relapse-like behavior induced by previously alcohol-associated stimuli or yohimbine stress. In a reinstatement paradigm, in which alcohol seeking was triggered by a nicotine challenge, a behavior successfully inhibited by the nonselective nAChR antagonist mecamylamine, AP-202 was not effective, while pretreatment with TP2212-59 abolished nicotine-induced reinstatement of alcohol seeking. CONCLUSIONS These findings suggest differential roles for α4β2 and α3β4 nAChR on alcohol taking and seeking with selective blockade of α4β2 nAChR being more implicated in modulating alcohol taking while selective blockade of α3β4 nAChR is involved in nicotine-induced alcohol seeking.
Collapse
Affiliation(s)
- Andrea Cippitelli
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL, 34987, USA.
| | - Gloria Brunori
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Jennifer Schoch
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Christopher J. Armishaw
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Jinhua Wu
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA,Assuage Pharmaceuticals, Inc., 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Nurulain T. Zaveri
- Astraea Therapeutics, LLC, 320 Logue Avenue, Mountain View, CA 94043, USA
| | - Marc A. Giulianotti
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA,Assuage Pharmaceuticals, Inc., 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Gregory S. Welmaker
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA,Assuage Pharmaceuticals, Inc., 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Lawrence Toll
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA,Assuage Pharmaceuticals, Inc., 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| |
Collapse
|
29
|
Stoklosa TM, Morley KC, Volovets A, Haber PS. Pharmacotherapy for Alcohol Use Disorder in the Context of Liver Disease. CURRENT ADDICTION REPORTS 2018. [DOI: 10.1007/s40429-018-0211-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
30
|
Touchette JC, Maertens JJ, Mason MM, O'Rourke KY, Lee AM. The nicotinic receptor drug sazetidine-A reduces alcohol consumption in mice without affecting concurrent nicotine consumption. Neuropharmacology 2018; 133:63-74. [PMID: 29355641 PMCID: PMC5858984 DOI: 10.1016/j.neuropharm.2018.01.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/09/2018] [Accepted: 01/13/2018] [Indexed: 01/01/2023]
Abstract
Alcohol and nicotine addiction are frequently co-morbid. The nicotinic acetylcholine receptors (nAChRs) are critical for both alcohol and nicotine addiction mechanisms, since nAChR drugs that reduce nicotine consumption have been shown to also reduce alcohol consumption. Sazetidine-A, a pre-clinical nAChR drug with agonist and desensitizing effects at α4β2 and α7 nAChRs, has been reported to reduce alcohol consumption and nicotine self-administration in rats when administered at high doses. However, this effect has not been replicated in mice. In this study, we examined the effect of sazetidine-A on alcohol and nicotine consumption in male and female mice utilizing voluntary oral consumption procedures previously developed in our lab. We found that sazetidine-A (1 mg/kg, i.p) reduced overnight alcohol consumption, but did not affect nicotine consumption when presented either alone or concurrently with alcohol. Sazetidine-A did not reduce water or saccharin consumption at any dose tested. In a chronic co-consumption experiment in which either alcohol or nicotine was re-introduced after one week of forced abstinence, sazetidine-A attenuated post-abstinence consumption of alcohol but not nicotine. Sazetidine-A also significantly reduced alcohol consumption in an acute, binge drinking-in-the-dark procedure. Finally, we tested the effect of sazetidine-A on alcohol withdrawal, and found that sazetidine-A significantly reduced handling-induced convulsions during alcohol withdrawal. Collectively, these data suggest a novel role for the nAChR targets of sazetidine-A in specifically mediating alcohol consumption, separate from the involvement of nAChRs in mediating nicotine consumption. Delineation of this pathway may provide insight into novel therapies for the treatment of alcohol use disorders.
Collapse
Affiliation(s)
| | - Jamie J Maertens
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Margaret M Mason
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Kyu Y O'Rourke
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Anna M Lee
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
31
|
Klenowski PM, Tapper AR. Molecular, Neuronal, and Behavioral Effects of Ethanol and Nicotine Interactions. Handb Exp Pharmacol 2018; 248:187-212. [PMID: 29423839 DOI: 10.1007/164_2017_89] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
32
|
Roberts W, Verplaetse TL, Moore K, Oberleitner L, Picciotto MR, McKee SA. Effects of varenicline on alcohol self-administration and craving in drinkers with depressive symptoms. J Psychopharmacol 2017; 31:906-914. [PMID: 28351203 PMCID: PMC5823265 DOI: 10.1177/0269881117699618] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Varenicline (VAR) is approved to aid in smoking cessation and has been shown to be effective for reducing alcohol consumption in heavy drinkers. Little is known, however, about treatment moderators that may influence efficacy. The current study reanalyzed data from a human laboratory study (Verplaetse et al., 2016) to determine whether VAR was more effective at reducing alcohol use among drinkers reporting symptoms of depression. Participants were 60 adults meeting DSM-IV criteria for alcohol use disorders ( n = 60) who were randomly assigned to receive VAR (1 mg/day, 2 mg/day) or placebo. Following 7 days of medication pretreatment, participants attended a laboratory testing session. They provided self-reported ratings of alcohol craving and performed an ad libitum alcohol consumption task after receiving a priming dose of alcohol (target blood alcohol concentration = 0.030 g/dL). Higher blood VAR plasma levels were associated with less alcohol craving and less drinking among participants with more depressive symptoms. Among participants with fewer depressive symptoms, VAR was associated with more drinking during the ad libitum drinking task. These findings show that depression symptoms may be a moderator of VAR efficacy in alcohol users and provides evidence for the role of nAChRs in depression and alcohol use.
Collapse
Affiliation(s)
- Walter Roberts
- Yale School of Medicine, Department of Psychiatry, New Haven, CT, USA
| | | | - Kelly Moore
- Yale School of Medicine, Department of Psychiatry, New Haven, CT, USA
| | | | | | - Sherry A McKee
- Yale School of Medicine, Department of Psychiatry, New Haven, CT, USA
| |
Collapse
|
33
|
Zipori D, Sadot-Sogrin Y, Goltseker K, Even-Chen O, Rahamim N, Shaham O, Barak S. Re-exposure to nicotine-associated context from adolescence enhances alcohol intake in adulthood. Sci Rep 2017; 7:2479. [PMID: 28559549 PMCID: PMC5449395 DOI: 10.1038/s41598-017-02177-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/04/2017] [Indexed: 11/17/2022] Open
Abstract
Alcohol and nicotine are the two most commonly-abused substances and are often used together. Nicotine enhances alcohol-drinking behaviors in humans and in animals, and was suggested to enhance the reinforcing properties of other reinforcers. Here, we show that nicotine-associated environment, rather than nicotine itself, enhances alcohol intake in rats. Adolescent rats received repeated intermittent injections of nicotine (0.4 mg/kg, i.p., 5 injections, every 3rd day) or saline. The injection was paired with their home cage, or with the subsequent alcohol self-administration context. Rats were then trained to self-administer 20% alcohol. Nicotine given in the home cage did not alter subsequent alcohol intake. However, pairing nicotine with the operant chamber during adolescence led to a long-lasting increased alcohol self-administration in adulthood, compared to nicotine pre-treatment in other contexts. This effect persisted 3 months after nicotine cessation, in a relapse test after abstinence. Furthermore, re-exposure to the nicotine-associated context in adult rats led to a decrease in glial cell line-derived neurotrophic factor (Gdnf) mRNA expression in the ventral tegmental area, an effect that leads to increased alcohol consumption, as we have previously reported. Our findings suggest that retrieval of nicotine-associated contextual memories from adolescence may gate alcohol intake in adulthood, with a possible involvement of GDNF.
Collapse
Affiliation(s)
- Dor Zipori
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Koral Goltseker
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Oren Even-Chen
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nofar Rahamim
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ohad Shaham
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Segev Barak
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
34
|
Yardley MM, Ray LA. Medications development for the treatment of alcohol use disorder: insights into the predictive value of animal and human laboratory models. Addict Biol 2017; 22:581-615. [PMID: 26833803 DOI: 10.1111/adb.12349] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 11/09/2015] [Accepted: 11/13/2015] [Indexed: 12/20/2022]
Abstract
Development of effective treatments for alcohol use disorder (AUD) represents an important public health goal. This review provides a summary of completed preclinical and clinical studies testing pharmacotherapies for the treatment of AUD. We discuss opportunities for improving the translation from preclinical findings to clinical trial outcomes, focusing on the validity and predictive value of animal and human laboratory models of AUD. Specifically, while preclinical studies of medications development have offered important insights into the neurobiology of the disorder and alcohol's molecular targets, limitations include the lack of standardized methods and streamlined processes whereby animal studies can readily inform human studies. Behavioral pharmacology studies provide a less expensive and valuable opportunity to assess the feasibility of a pharmacotherapy prior to initiating larger scale clinical trials by providing insights into the mechanism of the drug, which can then inform recruitment, analyses, and assessments. Summary tables are provided to illustrate the wide range of preclinical, human laboratory, and clinical studies of medications development for alcoholism. Taken together, this review highlights the challenges associated with animal paradigms, human laboratory studies, and clinical trials with the overarching goal of advancing treatment development and highlighting opportunities to bridge the gap between preclinical and clinical research.
Collapse
Affiliation(s)
- Megan M. Yardley
- Department of Psychology; University of California, Los Angeles; Los Angeles CA USA
| | - Lara A. Ray
- Department of Psychology; University of California, Los Angeles; Los Angeles CA USA
- Department of Psychiatry and Biobehavioral Sciences; University of California, Los Angeles; Los Angeles CA USA
| |
Collapse
|
35
|
Tolu S, Marti F, Morel C, Perrier C, Torquet N, Pons S, de Beaurepaire R, Faure P. Nicotine enhances alcohol intake and dopaminergic responses through β2* and β4* nicotinic acetylcholine receptors. Sci Rep 2017; 7:45116. [PMID: 28332590 PMCID: PMC5362818 DOI: 10.1038/srep45116] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 02/17/2017] [Indexed: 12/25/2022] Open
Abstract
Alcohol and nicotine are the most widely co-abused drugs. Both modify the activity of dopaminergic (DA) neurons of the Ventral Tegmental Area (VTA) and lead to an increase in DA release in the Nucleus Accumbens, thereby affecting the reward system. Evidences support the hypothesis that distinct nicotinic acetylcholine receptors (nAChRs), the molecular target of acetylcholine (ACh) and exogenous nicotine, are also in addition implicated in the response to alcohol. The precise molecular and neuronal substrates of this interaction are however not well understood. Here we used in vivo electrophysiology in the VTA to characterise acute and chronic interactions between nicotine and alcohol. Simultaneous injections of the two drugs enhanced their responses on VTA DA neuron firing and chronic exposure to nicotine increased alcohol-induced DA responses and alcohol intake. Then, we assessed the role of β4 * nAChRs, but not β2 * nAChRs, in mediating acute responses to alcohol using nAChR subtypes knockout mice (β2-/- and β4-/- mice). Finally, we showed that nicotine-induced modifications of alcohol responses were absent in β2-/- and β4-/- mice, suggesting that nicotine triggers β2* and β4 * nAChR-dependent neuroadaptations that subsequently modify the responses to alcohol and thus indicating these receptors as key mediators in the complex interactions between these two drugs.
Collapse
Affiliation(s)
- Stefania Tolu
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Fabio Marti
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Carole Morel
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Carole Perrier
- Groupe Hospitalier Paul Guiraud, BP 20065, F-94806, Villejuif, France
| | - Nicolas Torquet
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| | - Stephanie Pons
- Institut Pasteur, Unité Neurobiologie Intégrative des Systèmes Cholinergiques, Département de Neuroscience, F-75724, Paris, France.,CNRS, UMR 3571, F-75724, Paris, France
| | | | - Philippe Faure
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005, Paris, France
| |
Collapse
|
36
|
Abstract
Purpose of the Review Comorbidity of alcohol and tobacco use is highly prevalent and may exacerbate the health effects of either substance alone. However, the mechanisms underlying this comorbidity are not well understood. This review will examine the evidence for shared neurobiological mechanisms of alcohol and nicotine comorbidity and experimental studies of the behavioural consequences of these interactions. Recent Findings Studies examining the shared neurobiology of alcohol and nicotine have identified two main mechanisms of comorbidity: (1) cross-reinforcement via the mesolimbic dopamine pathway and (2) cross-tolerance via shared genetic and nAChR interaction. Animal and human psychopharmacological studies demonstrate support for these two mechanisms of comorbidity. Summary Human behavioural studies indicate that (1) alcohol and tobacco potentiate each other’s rewarding effects and (2) nicotine reduces the sedative and intoxication effects of alcohol. Together, these findings provide a strong evidence base to support the role of the cross-reinforcement and cross-tolerance as mechanisms underlying the comorbidity of alcohol and tobacco use. Methodological concerns in the literature and recommendations for future studies are discussed alongside implications for treatment of comorbid alcohol and tobacco use.
Collapse
Affiliation(s)
- Sally Adams
- Department of Psychology, University of Bath, 10 West, Bath, BA2 7AY UK.,UK Centre for Tobacco and Alcohol Studies, Bath, UK
| |
Collapse
|
37
|
Nicotine-induced enhancement of Pavlovian alcohol-seeking behavior in rats. Psychopharmacology (Berl) 2017; 234:727-738. [PMID: 28011981 DOI: 10.1007/s00213-016-4508-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/09/2016] [Indexed: 01/02/2023]
Abstract
RATIONALE Nicotine enhances responding elicited by Pavlovian cues that predict positive outcomes. OBJECTIVES We tested the hypothesis that nicotine acting at nicotinic acetylcholine receptors (nAChRs) would augment Pavlovian alcohol-seeking. METHODS Male, Long-Evans rats with unrestricted access to food and water were acclimated to drinking 15% ethanol in their home cages and then given Pavlovian conditioning sessions in which each trial of a 15-s conditioned stimulus (CS, 12 trials/session) was paired with 0.2 ml of ethanol (unconditioned stimulus, US, 2.4 ml/session). Entries into a port where ethanol was delivered were used to assess conditioning. Control groups received explicitly unpaired trials of the CS and US. In experiment 1, systemic injections of saline (1 ml/kg) or nicotine (0.4 mg/kg, freebase) were administered before each session. In experiments 2 and 3, an identical regimen of saline or nicotine injections was administered before the start of Pavlovian conditioning sessions. RESULTS All paired groups acquired conditioned port-entry responding to the CS, indicative of Pavlovian alcohol-seeking, whereas unpaired control group did not. Pre-session nicotine injections increased CS port-entries relative to saline, only in the paired group. This nicotine-induced enhancement of Pavlovian alcohol-seeking was blocked by pre-treatment with the nAChR antagonist mecamylamine. Prior exposure to nicotine did not influence the subsequent acquisition of Pavlovian alcohol-seeking. CONCLUSIONS These findings highlight for the first time that nicotine acting at nAChRs augments Pavlovian alcohol-seeking, specifically in non-restricted rats. Individuals who smoke and drink may thus be particularly susceptible to alcohol cues that could trigger further drinking.
Collapse
|
38
|
Tarren JR, Bartlett SE. Alcohol and nicotine interactions: pre-clinical models of dependence. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 43:146-154. [PMID: 27740856 DOI: 10.1080/00952990.2016.1197232] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
While the co-morbidity of alcohol (ethanol) and tobacco (nicotine) dependence is well described, the processes that underpin this strong connection are still under debate. With the increasing popularity of electronic cigarettes (e-cigarettes), it is now becoming more important to look to the neurobiological mechanisms involving alcohol and nicotine interactions to effectively treat a new generation of co-dependent individuals. Researchers have already recognized that the neuropathology produced by the combination of nicotine and ethanol is likely to produce an addictive nature very different to that of either one alone, and are employing a mixture of pre-clinical techniques to establish and investigate every stage in the development of both nicotine and ethanol-seeking behaviors. While it is agreed that multiple pathways orchestrate the complex reward profile of alcohol and nicotine co-addiction, several lines of evidence suggest the convergent site of action is within the mesolimbic dopaminergic system, at neuronal nicotinic acetylcholine receptors (nAChRs). A whole host of strategies are currently being employed to discover and unravel previously unknown or ill understood neurobiological processes in the brain, contributing greatly toward the development of novel pharmacotherapies with the aim of improving patient outcomes. This review intends to shed some light on the most influential and most recent pre-clinical work that is leading the charge in modeling this complicated relationship.
Collapse
Affiliation(s)
- Josephine R Tarren
- a Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology , Woolloongabba , QLD , Australia
| | - Selena E Bartlett
- a Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology , Woolloongabba , QLD , Australia
| |
Collapse
|
39
|
Kohut SJ. Interactions between nicotine and drugs of abuse: a review of preclinical findings. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 43:155-170. [PMID: 27589579 DOI: 10.1080/00952990.2016.1209513] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Polysubstance abuse is common among substance-use disorder patients, and nicotine is one of the most commonly co-used substances. Epidemiological and clinical laboratory studies suggest that nicotine, when combined with other drugs of abuse, increases intake of one or both substances. This review focuses on the preclinical literature regarding nicotine's interaction with alcohol, stimulants (i.e., cocaine, amphetamines), opioids (i.e., morphine, heroin), and Δ9-tetrahydrocannabinol (THC). The current understanding of how these various classes of abused drugs may interact with nicotine on behavioral, physiological, and pharmacological indices that may be important in maintaining co-use of one or both substances in human populations are highlighted. Suggestions as to future areas of research and gaps in knowledge are offered.
Collapse
Affiliation(s)
- Stephen J Kohut
- a McLean Hospital and Department of Psychiatry, Harvard Medical School , Belmont , MA , USA
| |
Collapse
|
40
|
Cross SJ, Lotfipour S, Leslie FM. Mechanisms and genetic factors underlying co-use of nicotine and alcohol or other drugs of abuse. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 43:171-185. [PMID: 27532746 DOI: 10.1080/00952990.2016.1209512] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Concurrent use of tobacco and alcohol or psychostimulants represents a major public health concern, with use of one substance influencing consumption of the other. Co-abuse of these drugs leads to substantial negative health outcomes, reduced cessation, and high economic costs, but the underlying mechanisms are poorly understood. Epidemiological data suggest that tobacco use during adolescence plays a particularly significant role. Adolescence is a sensitive period of development marked by major neurobiological maturation of brain regions critical for reward processing, learning and memory, and executive function. Nicotine exposure during this time produces a unique and long-lasting vulnerability to subsequent substance use, likely via actions at cholinergic, dopaminergic, and serotonergic systems. In this review, we discuss recent clinical and preclinical data examining the genetic factors and mechanisms underlying co-use of nicotine and alcohol or cocaine and amphetamines. We evaluate the critical role of nicotinic acetylcholine receptors throughout, and emphasize the dearth of preclinical studies assessing concurrent drug exposure. We stress important age and sex differences in drug responses, and highlight a brief, low-dose nicotine exposure paradigm that may better model early use of tobacco products. The escalating use of e-cigarettes among youth necessitates a closer look at the consequences of early adolescent nicotine exposure on subsequent alcohol and drug abuse.
Collapse
Affiliation(s)
- Sarah J Cross
- a Department of Anatomy & Neurobiology , School of Medicine, University of California , Irvine , CA , USA
| | - Shahrdad Lotfipour
- b Department of Emergency Medicine , School of Medicine, University of California , Irvine , CA , USA.,c Department of Pharmacology , School of Medicine, University of California , Irvine , CA , USA
| | - Frances M Leslie
- a Department of Anatomy & Neurobiology , School of Medicine, University of California , Irvine , CA , USA.,c Department of Pharmacology , School of Medicine, University of California , Irvine , CA , USA
| |
Collapse
|
41
|
Gawel K, Labuz K, Gibula-Bruzda E, Jenda M, Marszalek-Grabska M, Silberring J, Kotlinska JH. Acquisition and reinstatement of ethanol-induced conditioned place preference in rats: Effects of the cholinesterase inhibitors donepezil and rivastigmine. J Psychopharmacol 2016; 30:676-87. [PMID: 27097732 DOI: 10.1177/0269881116642539] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The present study examined the influence of the cholinesterase inhibitors donepezil (a selective inhibitor of acetylcholinesterase) and rivastigmine (also an inhibitor of butyrylcholinesterase) on the acquisition and reinstatement of ethanol-induced conditioned place preference (CPP) in rats. Before the CPP procedure, animals received a single injection of ethanol (0.5 g/kg, 10% w/v, intraperitoneally [i.p.]) for 15 days. The ethanol-induced CPP (biased method) was developed by four injections of ethanol (0.5 g/kg, 10% w/v, i.p.) every second day. Control rats received saline instead of ethanol. Donepezil (0.5, 1 or 3 mg/kg, i.p.) or rivastigmine (0.03, 0.5 or 1 mg/kg, i.p.) were administered before ethanol during conditioning or before the reinstatement of ethanol-induced CPP. The cholinesterase inhibitors were equally effective in increasing (dose dependently) the acquisition of ethanol-induced CPP. Furthermore, priming injections of both inhibitors reinstated (cross-reinstatement) the ethanol-induced CPP with similar efficacy. These effects of both cholinesterase inhibitors were reversed by mecamylamine (3 mg/kg, i.p.), a nicotinic acetylcholine receptor antagonist, but not by scopolamine (0.5 mg/kg, i.p.), a muscarinic acetylcholine receptor antagonist. Thus, our results show that the cholinergic system is involved in the reinforcing properties of ethanol, and nicotinic acetylcholine receptors play an important role in the relapse to ethanol-seeking behaviour.
Collapse
Affiliation(s)
- Kinga Gawel
- Department of Pharmacology and Pharmacodynamics, Medical University, Lublin, Poland
| | | | - Ewa Gibula-Bruzda
- Department of Pharmacology and Pharmacodynamics, Medical University, Lublin, Poland
| | - Malgorzata Jenda
- Department of Pharmacology and Pharmacodynamics, Medical University, Lublin, Poland
| | | | - Jerzy Silberring
- Department of Biochemistry and Neurobiology, AGH University of Science and Technology, Krakow, Poland
| | - Jolanta H Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University, Lublin, Poland
| |
Collapse
|
42
|
O'Rourke KY, Touchette JC, Hartell EC, Bade EJ, Lee AM. Voluntary co-consumption of alcohol and nicotine: Effects of abstinence, intermittency, and withdrawal in mice. Neuropharmacology 2016; 109:236-246. [PMID: 27342124 DOI: 10.1016/j.neuropharm.2016.06.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 11/17/2022]
Abstract
Alcohol and nicotine are often used together, and there is a high rate of co-occurrence between alcohol and nicotine addiction. Most animal models studying alcohol and nicotine interactions have utilized passive drug administration, which may not be relevant to human co-addiction. In addition, the interactions between alcohol and nicotine in female animals have been understudied, as most studies have used male animals. To address these issues, we developed models of alcohol and nicotine co-consumption in male and female mice that utilized voluntary, oral consumption of unsweetened alcohol, nicotine and water. We first examined drug consumption and preference in single-drug, sequential alcohol and nicotine consumption tests in male and female C57BL/6 and DBA/2J mice. We then tested chronic continuous and intermittent access alcohol and nicotine co-consumption procedures. We found that male and female C57BL/6 mice readily co-consumed unsweetened alcohol and nicotine. In our continuous co-consumption procedures, we found that varying the available nicotine concentration during an alcohol abstinence period affected compensatory nicotine consumption during alcohol abstinence, and affected rebound alcohol consumption when alcohol was re-introduced. Consumption of alcohol and nicotine in an intermittent co-consumption procedure produced higher alcohol consumption levels, but not nicotine consumption levels, compared with the continuous co-consumption procedures. Finally, we found that intermittent alcohol and nicotine co-consumption resulted in physical dependence. Our data show that these voluntary co-consumption procedures can be easily performed in mice and can be used to study behavioral interactions between alcohol and nicotine consumption, which may better model human alcohol and nicotine co-addiction.
Collapse
Affiliation(s)
- Kyu Y O'Rourke
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | | | - Elizabeth C Hartell
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Elizabeth J Bade
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Anna M Lee
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
43
|
Bell RL, Hauser S, Rodd ZA, Liang T, Sari Y, McClintick J, Rahman S, Engleman EA. A Genetic Animal Model of Alcoholism for Screening Medications to Treat Addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:179-261. [PMID: 27055615 PMCID: PMC4851471 DOI: 10.1016/bs.irn.2016.02.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The purpose of this review is to present up-to-date pharmacological, genetic, and behavioral findings from the alcohol-preferring P rat and summarize similar past work. Behaviorally, the focus will be on how the P rat meets criteria put forth for a valid animal model of alcoholism with a highlight on its use as an animal model of polysubstance abuse, including alcohol, nicotine, and psychostimulants. Pharmacologically and genetically, the focus will be on the neurotransmitter and neuropeptide systems that have received the most attention: cholinergic, dopaminergic, GABAergic, glutamatergic, serotonergic, noradrenergic, corticotrophin releasing hormone, opioid, and neuropeptide Y. Herein, we sought to place the P rat's behavioral and neurochemical phenotypes, and to some extent its genotype, in the context of the clinical literature. After reviewing the findings thus far, this chapter discusses future directions for expanding the use of this genetic animal model of alcoholism to identify molecular targets for treating drug addiction in general.
Collapse
Affiliation(s)
- R L Bell
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States.
| | - S Hauser
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Z A Rodd
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - T Liang
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - Y Sari
- University of Toledo, Toledo, OH, United States
| | - J McClintick
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States
| | - E A Engleman
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
44
|
Roche DJO, Ray LA, Yardley MM, King AC. Current insights into the mechanisms and development of treatments for heavy drinking cigarette smokers. CURRENT ADDICTION REPORTS 2016; 3:125-137. [PMID: 27162709 PMCID: PMC4859339 DOI: 10.1007/s40429-016-0081-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is a strong association between cigarette smoking and alcohol use at the epidemiological, behavioral, and molecular levels, and this co-use creates substantial impediments to smoking cessation among smokers who are also heavy drinkers. Compared with individuals who only smoke, those who both drink and smoke heavily experience more severe health consequences and have greater difficulty in quitting smoking. During smoking abstinence, greater alcohol use is associated with decreased odds of smoking cessation, and smokers are substantially more likely to experience a smoking lapse during drinking episodes. As heavy drinking smokers are less responsive to the currently available pharmacological treatments, this subgroup of high-risk substance users possesses a unique clinical profile and treatment needs. Thus, treatment development for heavy drinking smokers represents a significant and understudied research area within the field of smoking cessation. This review will briefly describe findings from epidemiological, behavioral, and molecular studies illustrating alcohol and tobacco co-use and identify how the behavioral and neurobiological mechanisms underlying the interaction of alcohol and nicotine may inform the development of targeted treatments for this unique population of smokers.
Collapse
Affiliation(s)
- Daniel J O Roche
- University of California, Los Angeles, Department of Psychology, Los Angeles, CA, 90095, USA
| | - Lara A Ray
- University of California, Los Angeles, Department of Psychology, Los Angeles, CA, 90095, USA; University of California, Los Angeles, Department of Psychiatry and Biobehavioral Sciences, Los Angeles, CA, 90095, USA
| | - Megan M Yardley
- University of California, Los Angeles, Department of Psychology, Los Angeles, CA, 90095, USA
| | - Andrea C King
- University of Chicago, Department of Psychiatry and Behavioral Neuroscience, Chicago, IL 60637, USA
| |
Collapse
|
45
|
Nuutinen S, Panula P, Salminen O. Different Hypothalamic Nicotinic α7 Receptor Expression and Response to Low Nicotine Dose in Alcohol-Preferring and Alcohol-Avoiding Rats. Alcohol Clin Exp Res 2016; 40:329-34. [PMID: 26842251 DOI: 10.1111/acer.12960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/08/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND The aim of this study was to examine possible differences in nicotinic acetylcholine receptors and responses in rats with genetic preference or avoidance for alcohol. This was done by using 2 rat lines with high alcohol preference (Alko Alcohol [AA]) or alcohol avoidance (Alko Non-Alcohol [ANA]). METHODS Locomotor activity was measured following nicotine and histamine H3 receptor (H3R) antagonist treatment. In situ hybridization and receptor ligand binding experiments were used in drug-naïve animals to examine the expression of different α nicotinic receptor subunits. RESULTS The AA rats were found to be more sensitive to the stimulatory effect of a low dose of nicotine than ANA rats, which were not significantly activated. Combination of histamine H3R antagonist, JNJ-39220675, and nicotine resulted to similar locomotor activation as nicotine alone. To further understand the mechanism underlying the difference in nicotine response in AA and ANA rats, we studied the expression of α5, α6, and α7 nicotinic receptor subunits in specific brain areas of AA and ANA rats. We found no differences in the expression of α5 nicotinic receptor subunits in the medial habenula and hippocampus or in α6 subunit in the ventral tegmental area and substantia nigra. However, the level of α7 nicotinic receptor subunit mRNA was significantly lower in the tuberomamillary nucleus of posterior hypothalamus of alcohol-preferring AA rats than in alcohol-avoiding ANA rats. Also the hypothalamic [125I-α-bungarotoxin binding was lower in AA rats indicating lower levels of α7 nicotinic receptors. CONCLUSIONS The lower expression and receptor binding of α7 nicotinic receptors in the tuberomamillary nucleus of AA rats suggest a difference in the regulation of brain histamine neurons between the rat lines since the α7 nicotinic receptors are located in histaminergic neurons. Stronger nicotine-induced locomotor response, mediated partially via α7 receptors, and previously described high alcohol consumption in AA rats could be explained by the found difference in tuberomamillary α7 receptor levels.
Collapse
Affiliation(s)
- Saara Nuutinen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Pertti Panula
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Outi Salminen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
46
|
Van Skike CE, Maggio SE, Reynolds AR, Casey EM, Bardo MT, Dwoskin LP, Prendergast MA, Nixon K. Critical needs in drug discovery for cessation of alcohol and nicotine polysubstance abuse. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:269-87. [PMID: 26582145 PMCID: PMC4679525 DOI: 10.1016/j.pnpbp.2015.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 11/10/2015] [Accepted: 11/11/2015] [Indexed: 01/29/2023]
Abstract
Polysubstance abuse of alcohol and nicotine has been overlooked in our understanding of the neurobiology of addiction and especially in the development of novel therapeutics for its treatment. Estimates show that as many as 92% of people with alcohol use disorders also smoke tobacco. The health risks associated with both excessive alcohol consumption and tobacco smoking create an urgent biomedical need for the discovery of effective cessation treatments, as opposed to current approaches that attempt to independently treat each abused agent. The lack of treatment approaches for alcohol and nicotine abuse/dependence mirrors a similar lack of research in the neurobiology of polysubstance abuse. This review discusses three critical needs in medications development for alcohol and nicotine co-abuse: (1) the need for a better understanding of the clinical condition (i.e. alcohol and nicotine polysubstance abuse), (2) the need to better understand how these drugs interact in order to identify new targets for therapeutic development and (3) the need for animal models that better mimic this human condition. Current and emerging treatments available for the cessation of each drug and their mechanisms of action are discussed within this context followed by what is known about the pharmacological interactions of alcohol and nicotine. Much has been and will continue to be gained from studying comorbid alcohol and nicotine exposure.
Collapse
Affiliation(s)
- C E Van Skike
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States
| | - S E Maggio
- Department of Psychology, University of Kentucky, Lexington, KY 40536, United States
| | - A R Reynolds
- Department of Psychology, University of Kentucky, Lexington, KY 40536, United States
| | - E M Casey
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States
| | - M T Bardo
- Department of Psychology, University of Kentucky, Lexington, KY 40536, United States; Center for Drug Abuse and Research Translation, University of Kentucky, Lexington, KY 40536, United States; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, United States
| | - L P Dwoskin
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States; Center for Drug Abuse and Research Translation, University of Kentucky, Lexington, KY 40536, United States
| | - M A Prendergast
- Department of Psychology, University of Kentucky, Lexington, KY 40536, United States; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, United States
| | - K Nixon
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, United States.
| |
Collapse
|
47
|
Rahman S, Engleman EA, Bell RL. Recent Advances in Nicotinic Receptor Signaling in Alcohol Abuse and Alcoholism. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 137:183-201. [PMID: 26810002 PMCID: PMC4754113 DOI: 10.1016/bs.pmbts.2015.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alcohol is the most commonly abused legal substance and alcoholism is a serious public health problem. It is a leading cause of preventable death in the world. The cellular and molecular mechanisms of alcohol reward and addiction are still not well understood. Emerging evidence indicates that unlike other drugs of abuse, such as nicotine, cocaine, or opioids, alcohol targets numerous channel proteins, receptor molecules, and signaling pathways in the brain. Previously, research has identified brain nicotinic acetylcholine receptors (nAChRs), a heterogeneous family of pentameric ligand-gated cation channels expressed in the mammalian brain, as critical molecular targets for alcohol abuse and dependence. Genetic variations encoding nAChR subunits have been shown to increase the vulnerability to develop alcohol dependence. Here, we review recent insights into the rewarding effects of alcohol, as they pertain to different nAChR subtypes, associated signaling molecules, and pathways that contribute to the molecular mechanisms of alcoholism and/or comorbid brain disorders. Understanding these cellular changes and molecular underpinnings may be useful for the advancement of brain nicotinic-cholinergic mechanisms, and will lead to a better translational and therapeutic outcome for alcoholism and/or comorbid conditions.
Collapse
Affiliation(s)
- Shafiqur Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, South Dakota, USA.
| | - Eric A Engleman
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Richard L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
48
|
Cigarettes and alcohol: The influence of nicotine on operant alcohol self-administration and the mesolimbic dopamine system. Biochem Pharmacol 2015; 97:550-557. [PMID: 26253689 DOI: 10.1016/j.bcp.2015.07.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 07/30/2015] [Indexed: 12/11/2022]
Abstract
Studies in human populations consistently demonstrate an interaction between nicotine and ethanol use, each drug influencing the use of the other. Here we present data and review evidence from animal studies that nicotine influences operant self-administration of ethanol. The operant reinforcement paradigm has proven to be a behaviorally relevant and quantitative model for studying ethanol-seeking behavior. Exposure to nicotine can modify the reinforcing properties of ethanol during different phases of ethanol self-administration, including acquisition, maintenance, and reinstatement. Our data suggest that non-daily intermittent nicotine exposure can trigger a long-lasting increase in ethanol self-administration. The biological basis for interactions between nicotine and ethanol is not well understood but may involve the stress hormone systems and adaptations in the mesolimbic dopamine system. Future studies that combine operant self-administration with techniques for monitoring or manipulating in vivo neurophysiology may provide new insights into the neuronal mechanisms that link nicotine and alcohol use.
Collapse
|
49
|
Karatayev O, Lukatskaya O, Moon SH, Guo WR, Chen D, Algava D, Abedi S, Leibowitz SF. Nicotine and ethanol co-use in Long-Evans rats: Stimulatory effects of perinatal exposure to a fat-rich diet. Alcohol 2015; 49:479-89. [PMID: 25979531 DOI: 10.1016/j.alcohol.2015.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 12/14/2022]
Abstract
Clinical studies demonstrate frequent co-existence of nicotine and alcohol abuse and suggest that this may result, in part, from the ready access to and intake of fat-rich diets. Whereas animal studies show that high-fat diet intake in adults can enhance the consumption of either nicotine or ethanol and that maternal consumption of a fat-rich diet during pregnancy increases operant responding for nicotine in offspring, little is known about the impact of dietary fat on the co-abuse of these two drugs. The goal of this study was to test in Long-Evans rats the effects of perinatal exposure to fat on the co-use of nicotine and ethanol, using a novel paradigm that involves simultaneous intravenous (IV) self-administration of these two drugs. Fat- vs. chow-exposed offspring were characterized and compared, first in terms of their nicotine self-administration behavior, then in terms of their nicotine/ethanol self-administration behavior, and lastly in terms of their self-administration of ethanol in the absence of nicotine. The results demonstrate that maternal consumption of fat compared to low-fat chow during gestation and lactation significantly stimulates nicotine self-administration during fixed-ratio testing. It also increases nicotine/ethanol self-administration during fixed-ratio and dose-response testing, with BEC elevated to 120 mg/dL, and causes an increase in breakpoint during progressive ratio testing. Of particular note is the finding that rats perinatally exposed to fat self-administer significantly more of the nicotine/ethanol mixture as compared to nicotine alone, an effect not evident in the chow-control rats. After removal of nicotine from the nicotine/ethanol mixture, this difference between the fat- and chow-exposed rats was lost, with both groups failing to acquire the self-administration of ethanol alone. Together, these findings suggest that perinatal exposure to a fat-rich diet, in addition to stimulating self-administration of nicotine, causes an even greater vulnerability to the excessive co-use of nicotine and ethanol.
Collapse
|
50
|
Fucito LM, Hanrahan TH. Heavy-Drinking Smokers' Treatment Needs and Preferences: A Mixed-Methods Study. J Subst Abuse Treat 2015; 59:38-44. [PMID: 26297324 DOI: 10.1016/j.jsat.2015.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/12/2015] [Accepted: 07/06/2015] [Indexed: 02/07/2023]
Abstract
The purpose of this mixed methods study was to describe the smoking and psychological characteristics of heavy-drinking smokers, their perceptions of smoking and drinking, and their smoking and alcohol treatment preferences to inform an integrated smoking and alcohol intervention. Heavy-drinking smokers (N=26) completed standardized surveys and participated in semi-structured focus group interviews. Participants reported a strong association between their smoking and drinking. Participants were more motivated to quit smoking than to reduce their drinking but perceived greater barriers to smoking cessation. Stress/negative affect was closely linked with both behaviors. They expressed overall enthusiasm for a smoking and alcohol intervention but had specific format and content preferences. Half preferred an integrated treatment format whereas others preferred a sequential treatment model. The most preferred content included personalized health feedback and a way to monitor health gains after behavior changes.
Collapse
Affiliation(s)
- Lisa M Fucito
- Yale School of Medicine, Department of Psychiatry, 1 Long Wharf Drive, Box 18, New Haven, CT 06511, USA; Smilow Cancer Hospital at Yale-New Haven, 20 York Street, New Haven, CT 06519, USA; Yale Cancer Center, 333 Cedar Street, New Haven, CT, 06510, USA.
| | - Tess H Hanrahan
- Yale School of Medicine, Department of Psychiatry, 1 Long Wharf Drive, Box 18, New Haven, CT 06511, USA
| |
Collapse
|