1
|
Corley C, Craig A, Sadek S, Marusich JA, Chehimi SN, White AM, Holdiness LJ, Reiner BC, Gipson CD. Enhancing translation: A need to leverage complex preclinical models of addictive drugs to accelerate substance use treatment options. Pharmacol Biochem Behav 2024; 243:173836. [PMID: 39067531 PMCID: PMC11344688 DOI: 10.1016/j.pbb.2024.173836] [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: 02/12/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Preclinical models of addictive drugs have been developed for decades to model aspects of the clinical experience in substance use disorders (SUDs). These include passive exposure as well as volitional intake models across addictive drugs and have been utilized to also measure withdrawal symptomatology and potential neurobehavioral mechanisms underlying relapse to drug seeking or taking. There are a number of Food and Drug Administration (FDA)-approved medications for SUDs, however, many demonstrate low clinical efficacy as well as potential sex differences, and we also note gaps in the continuum of care for certain aspects of clinical experiences in individuals who use drugs. In this review, we provide a comprehensive update on both frequently utilized and novel behavioral models of addiction with a focus on translational value to the clinical experience and highlight the need for preclinical research to follow epidemiological trends in drug use patterns to stay abreast of clinical treatment needs. We then note areas in which models could be improved to enhance the medications development pipeline through efforts to enhance translation of preclinical models. Next, we describe neuroscience efforts that can be leveraged to identify novel biological mechanisms to enhance medications development efforts for SUDs, focusing specifically on advances in brain transcriptomics approaches that can provide comprehensive screening and identification of novel targets. Together, the confluence of this review demonstrates the need for careful selection of behavioral models and methodological parameters that better approximate the clinical experience combined with cutting edge neuroscience techniques to advance the medications development pipeline for SUDs.
Collapse
Affiliation(s)
- Christa Corley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ashley Craig
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Safiyah Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Samar N Chehimi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley M White
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lexi J Holdiness
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
2
|
Chellian R, Behnood-Rod A, Bruijnzeel AW. Sex differences in nicotine intake and relapse behavior in nicotine-dependent adult wistar rats. Front Pharmacol 2024; 15:1415219. [PMID: 39391691 PMCID: PMC11464435 DOI: 10.3389/fphar.2024.1415219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Tobacco use is highly addictive and the leading cause of premature mortality in the world. Long-access nicotine self-administration procedures in rats closely model human smoking behavior. However, significant gaps remain in our understanding of sex differences in the development of dependence and relapse in adult rats. Methods In the present study, we investigated operant responding for both nicotine and saline and the development of dependence in adult rats of both sexes. The rats had daily access to nicotine or saline for 6 h per day, 7 days per week. Dependence was assessed by evaluating precipitated and spontaneous somatic withdrawal signs, measuring locomotor activity in the small open field test, and assessing anxiety-like behavior in the large open field and elevated plus maze test. The sucrose preference test was used to determine if cessation of nicotine intake leads to anhedonia. It was also investigated if a period of forced abstinence affects nicotine-seeking behavior. Results This study showed that nicotine intake is higher in females than in males when given daily long access to nicotine. Daily nicotine self-administration led to more precipitated and spontaneous somatic withdrawal signs compared to saline self-administration, with no sex differences observed. In addition, cessation of nicotine intake led to a similar increase in activity in both males and females in the small open field test. However, cessation of nicotine intake did not increase anxiety-like behavior or cause anhedonia in either males or females. A time course analysis revealed that the nicotinic acetylcholine receptor antagonist mecamylamine affected nicotine intake differently in males and females, increasing intake in males and decreasing intake in females. Three weeks of forced abstinence led to an increase in nicotine and saline-seeking behavior. The rats exhibited more nicotine than saline seeking, and the females displayed more nicotine seeking than the males. Discussion The present findings demonstrate that females self-administer more nicotine and display more nicotine-seeking behavior than males. Furthermore, there were no sex differences in somatic withdrawal signs or activity during abstinence from nicotine. This work underscores the importance of considering sex differences across various aspects of addiction, including intake and relapse, when developing novel treatments for tobacco use disorder.
Collapse
|
3
|
Liu C, Filbey FM. Unlocking the age-old secrets of reward and substance use. Pharmacol Biochem Behav 2024; 239:173766. [PMID: 38604456 DOI: 10.1016/j.pbb.2024.173766] [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: 01/31/2024] [Revised: 03/25/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Although substance use is widespread across the lifespan from early adolescence to older adulthood, the prevalence of substance use disorder (SUD) differs between age groups. These age differences in SUD rates necessitate an investigation into how age moderates reward sensitivity, and consequently influences the risks and consequences related to substance use. This theoretical review integrates evidence from the literature to address the dynamic interplay between age and reward in the context of substance use. Overall, increasing evidence demonstrates that age moderates reward sensitivity and underlying reward system neurobiology. Reward sensitivity undergoes a non-linear trajectory across the lifespan. Low levels of reward sensitivity are associated with childhood and late adulthood. In contrast, high levels are associated with early to late adolescence, followed by a decline in the twenties. These fluctuations in reward sensitivity across the lifespan contribute to complex associations with substance use. This lends support to adolescence and young adulthood as vulnerable periods for the risk of subsequent SUD. More empirical research is needed to investigate reward sensitivity during SUD maintenance and recovery. Future research should also involve larger sample sizes and encompass a broader range of age groups, including older adults.
Collapse
Affiliation(s)
- Che Liu
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, United States of America.
| | - Francesca M Filbey
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, United States of America
| |
Collapse
|
4
|
Shaykhutdinova ER, Severyukhina MS, Kholoshenko IV, Gondarenko EA, Shelukhina IV, Kryukova EV, Ismailova AM, Sadovnikova ES, Dyachenko IA, Murashev AN, Tsetlin VI, Utkin YN. Anti-smoking drugs cytisine and varenicline reduce cardiac reperfusion injury in rat model of myocardial ischemia. Biochimie 2024; 216:108-119. [PMID: 37871826 DOI: 10.1016/j.biochi.2023.10.011] [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: 03/13/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Evidence to date indicates that activation of nicotinic acetylcholine receptors (nAChRs) can reduce cardiac injury from ischemia and subsequent reperfusion. The use of nAChR agonists in various animal models leads to a reduction in reperfusion injury. Earlier this effect was shown for the agonists of α7 nAChR subtype. In this work, we demonstrated the expression of mRNA encoding α4, α6 and β2 nAChR subunits in the left ventricle of rat heart. In a rat model of myocardial ischemia, we studied the effect of α4β2 nAChR agonists cytisine and varenicline, medicines used for the treatment of nicotine addiction, and found them to significantly reduce myocardium ischemia-reperfusion injury, varenicline manifesting a higher protection. Dihydro-β-erythroidine, antagonist of α4β2 nAChR, as well as methyllycaconitine, antagonist of α7 and α6β2-containing nAChR, prevented protective effect of varenicline. This together with the presence of α4, α6 and β2 subunit mRNA in the left ventricule of rat heart raises the possibility that the varenicline effect is mediated by α4β2 as well as by α7 and/or α6β2-containing receptors. Our results point to a new way for the use of cytisine and varenicline as cardioprotective agents.
Collapse
Affiliation(s)
- Elvira R Shaykhutdinova
- Biological Testing Laboratory, Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (BIBCh RAS), 6 Prospekt Nauki, 142290, Pushchino, Russia.
| | - Maria S Severyukhina
- Pushchino Branch of the Federal State Budgetary Educational Institution of Higher Education "Russian Biotechnological University (BIOTECH University)", 3 Prospekt Nauki, 142290, Pushchino, Russia.
| | - Inna V Kholoshenko
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (IBCh RAS), 16/10 Miklukho-Maklay Str., 117997, Moscow, Russia; Mendeleev University of Chemical Technology of Russia, 9 Miusskaya square, 125047, Moscow, Russia.
| | - Elena A Gondarenko
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (IBCh RAS), 16/10 Miklukho-Maklay Str., 117997, Moscow, Russia.
| | - Irina V Shelukhina
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (IBCh RAS), 16/10 Miklukho-Maklay Str., 117997, Moscow, Russia.
| | - Elena V Kryukova
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (IBCh RAS), 16/10 Miklukho-Maklay Str., 117997, Moscow, Russia.
| | - Alina M Ismailova
- Biological Testing Laboratory, Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (BIBCh RAS), 6 Prospekt Nauki, 142290, Pushchino, Russia.
| | - Elena S Sadovnikova
- Biological Testing Laboratory, Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (BIBCh RAS), 6 Prospekt Nauki, 142290, Pushchino, Russia.
| | - Igor A Dyachenko
- Biological Testing Laboratory, Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (BIBCh RAS), 6 Prospekt Nauki, 142290, Pushchino, Russia.
| | - Arkady N Murashev
- Biological Testing Laboratory, Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (BIBCh RAS), 6 Prospekt Nauki, 142290, Pushchino, Russia.
| | - Victor I Tsetlin
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (IBCh RAS), 16/10 Miklukho-Maklay Str., 117997, Moscow, Russia.
| | - Yuri N Utkin
- Department of Molecular Neuroimmune Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (IBCh RAS), 16/10 Miklukho-Maklay Str., 117997, Moscow, Russia.
| |
Collapse
|
5
|
Foulds J, Allen SI, Yingst J. Cytisinicline to Speed Smoking Cessation in the United States. JAMA 2023; 330:129-130. [PMID: 37432440 PMCID: PMC11500792 DOI: 10.1001/jama.2023.5939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Affiliation(s)
- Jonathan Foulds
- Penn State Center for Research on Tobacco and Health, Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Sophia I Allen
- Penn State Center for Research on Tobacco and Health, Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Jessica Yingst
- Penn State Center for Research on Tobacco and Health, Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
6
|
Chellian R, Behnood-Rod A, Bruijnzeel AW. Development of Dependence in Smokers and Rodents With Voluntary Nicotine Intake: Similarities and Differences. Nicotine Tob Res 2023; 25:1229-1240. [PMID: 36482774 PMCID: PMC10256892 DOI: 10.1093/ntr/ntac280] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 07/29/2023]
Abstract
INTRODUCTION Smoking and vaping throughout adolescence and early adulthood lead to nicotine dependence. Nicotine withdrawal is associated with somatic and affective withdrawal symptoms that contribute to smoking and relapse. Affective nicotine withdrawal symptoms in humans include craving for cigarettes, depression, anxiety, trouble sleeping, and cognitive deficits. METHODS Herein, we review clinical studies that investigated nicotine dependence in people who smoke or vape. We also discuss studies that investigated the development of dependence in animals with oral nicotine intake, nicotine aerosol self-administration, and intravenous nicotine self-administration. RESULTS Clinical studies report that adolescents who smoke daily develop nicotine dependence before those who smoke infrequently, but ultimately all smokers become dependent in adulthood. Preclinical studies indicate that rats that self-administer nicotine also become dependent. Rats that self-administer nicotine display somatic withdrawal signs and affective withdrawal signs, including increased anxiety and depressive-like behavior, cognitive deficits, and allodynia. Most nicotine withdrawal signs were observed in rodents with daily (7 days/week) or intermittent long access (23-hour) to nicotine. Clinical smoking studies report symptoms of nicotine dependence in adolescents of both sexes, but virtually all preclinical nicotine self-administration studies have been done with adult male rats. CONCLUSIONS The role of sex and age in the development of dependence in nicotine self-administration studies remains under-investigated. However, the role of sex and age in nicotine withdrawal has been thoroughly evaluated in studies in which nicotine was administered noncontingently. We discuss the need for volitional nicotine self-administration studies that explore the gradual development of dependence during adolescence and adulthood in rodents of both sexes. IMPLICATIONS The reviewed clinical studies investigated the development of nicotine dependence in male and female adolescent and young adult smokers and vapers. These studies indicate that most adolescent smokers and vapers gradually become nicotine dependent. Preclinical studies with rodents show that nicotine intake in widely used self-administration models also leads to dependence. However, almost all animal studies that investigated the development of nicotine dependence have been conducted with adult male rats. To better model smoking and vaping, it is important that nicotine intake in rats or mice starts during adolescence and that both sexes are included.
Collapse
Affiliation(s)
| | - Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Adriaan W Bruijnzeel
- Corresponding Author: Adriaan Bruijnzeel, PhD, University of Florida, Department of Psychiatry, 1149 Newell Dr., Gainesville, Florida 32611, USA. Telephone: 352-294-4931; Fax: 352-392-9887; E-mail:
| |
Collapse
|
7
|
Colyer-Patel K, Kuhns L, Weidema A, Lesscher H, Cousijn J. Age-dependent effects of tobacco smoke and nicotine on cognition and the brain: A systematic review of the human and animal literature comparing adolescents and adults. Neurosci Biobehav Rev 2023; 146:105038. [PMID: 36627063 DOI: 10.1016/j.neubiorev.2023.105038] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/21/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
Cigarette smoking is often initiated during adolescence and an earlier age of onset is associated with worse health outcomes later in life. Paradoxically, the transition towards adulthood also marks the potential for recovery, as the majority of adolescents are able to quit smoking when adulthood emerges. This systematic review aimed to evaluate the evidence from both human and animal studies for the differential impact of adolescent versus adult repeated and long-term tobacco and nicotine exposure on cognitive and brain outcomes. The limited human studies and more extensive yet heterogeneous animal studies, provide preliminary evidence of heightened fear learning, anxiety-related behaviour, reward processing, nicotinic acetylcholinergic receptors expression, dopamine expression and serotonin functioning after adolescent compared to adult exposure. Effects of nicotine or tobacco use on impulsivity were comparable across age groups. These findings provide novel insights into the mechanisms underlying adolescents' vulnerability to tobacco and nicotine. Future research is needed to translate animal to human findings, with a focus on directly linking a broader spectrum of brain and behavioural outcomes.
Collapse
Affiliation(s)
- Karis Colyer-Patel
- Neuroscience of Addiction (NofA) Lab, Department of Psychology, Education & Child Studies, Erasmus University Rotterdam, the Netherlands.
| | - Lauren Kuhns
- Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands
| | - Alix Weidema
- Neuroscience of Addiction (NofA) Lab, Department of Psychology, Education & Child Studies, Erasmus University Rotterdam, the Netherlands
| | - Heidi Lesscher
- Department Population Health Sciences, Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Janna Cousijn
- Neuroscience of Addiction (NofA) Lab, Department of Psychology, Education & Child Studies, Erasmus University Rotterdam, the Netherlands; Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
8
|
Hiranita T, Obeng S, Sharma A, Wilkerson JL, McCurdy CR, McMahon LR. In vitro and in vivo pharmacology of kratom. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:35-76. [PMID: 35341571 DOI: 10.1016/bs.apha.2021.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Kratom products have been historically and anecdotally used in south Asian countries for centuries to manage pain and opioid withdrawal. The use of kratom products has dramatically increased in the United States. More than 45 kratom alkaloids have been isolated, yet the overall pharmacology of the individual alkaloids is still not well characterized. The purpose of this chapter is to summarize in vitro and in vivo opioid activities of the primary kratom alkaloid mitragynine and its more potent metabolite 7-hydroxymitragynine. Following are experimental procedures described to characterize opioid receptor activity; receptor binding and functional assays, antinociceptive assays, operant conditioning assays, and respiratory plethysmography. The capacity of kratom alkaloids to confer tolerance and physical dependence as well as their pharmacokinetic properties are also summarized. The data reviewed here suggest that kratom products and mitragynine possess low efficacy agonist activity at the mu-opioid receptor in vivo. In addition, kratom products and mitragynine have been demonstrated to antagonize the effects of high efficacy mu-opioid agonists. The data further suggest that 7-hydroxymitragynine formed in vivo by metabolism of mitragynine may be minimally involved in the overall behavioral profile of mitragynine and kratom, whereas 7-hydroxymitragynine itself, at sufficiently high doses administered exogenously, shares many of the same abuse- and dependence-related behavioral effects associated with traditional opioid agonists. The apparent low efficacy of kratom products and mitragynine at mu-opioid receptors supports the development of these ligands as effective and potentially safe medications for opioid use disorder.
Collapse
Affiliation(s)
- Takato Hiranita
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Samuel Obeng
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Abhisheak Sharma
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Translational Drug Development Core, Clinical and Translational Sciences Institute, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Jenny L Wilkerson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, United States; Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Translational Drug Development Core, Clinical and Translational Sciences Institute, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Lance R McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
9
|
Chellian R, Behnood-Rod A, Wilson R, Bruijnzeel AW. Rewarding Effects of Nicotine Self-administration Increase Over Time in Male and Female Rats. Nicotine Tob Res 2021; 23:2117-2126. [PMID: 33987656 DOI: 10.1093/ntr/ntab097] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/11/2021] [Indexed: 11/14/2022]
Abstract
INTRODUCTION Smoking and the use of other nicotine-containing products is rewarding in humans. The self-administration of nicotine is also rewarding in male rats. However, it is unknown if there are sex differences in the reward-enhancing effects of nicotine self-administration and if the rewarding effects of nicotine change over time. METHODS Rats were prepared with catheters and intracranial self-stimulation (ICSS) electrodes to investigate the effects of nicotine and saline self-administration on reward function. A decrease in thresholds in the ICSS procedure reflects an enhancement of reward function. The ICSS parameters were determined before and after the self-administration sessions from days 1 to 10, and after the self-administration sessions from days 11 to 15. RESULTS During the first 10 days, there was no sex difference in nicotine intake, but during the last 5 days, the females took more nicotine than the males. During the first 10 days, nicotine self-administration did not lower the brain reward thresholds but decreased the response latencies. During the last 5 days, nicotine lowered the reward thresholds and decreased the response latencies. An analysis with the 5-day averages (days 1-5, 6-10, and 11-15) showed that the reward enhancing and stimulatory effects of nicotine increased over time. There were no sex differences in the reward-enhancing and stimulatory effects of nicotine. The nicotinic receptor antagonist mecamylamine diminished the reward-enhancing and stimulatory effects of nicotine. CONCLUSION These findings indicate that the rewarding effects of nicotine self-administration increase over time, and there are no sex differences in the reward-enhancing effects of nicotine self-administration in rats. IMPLICATIONS This study investigated the rewarding effect of nicotine and saline self-administration in male and female rats. The self-administration of nicotine, but not saline, enhanced brain reward function and had stimulatory effects. The rewarding effects of nicotine increased over time in the males and the females. Despite that the females had a higher level of nicotine intake than the males, the reward-enhancing effects of nicotine self-administration were the same. These findings suggest that in new tobacco and e-cigarette users, nicotine's rewarding effects might increase quickly, and a higher level of nicotine use in females might not translate into greater rewarding effects.
Collapse
Affiliation(s)
| | - Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Ryann Wilson
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Abstract
BACKGROUND Animal models are critical to improve our understanding of the neuronal mechanisms underlying nicotine withdrawal. Nicotine dependence in rodents can be established by repeated nicotine injections, chronic nicotine infusion via osmotic minipumps, oral nicotine intake, tobacco smoke exposure, nicotine vapor exposure, and e-cigarette aerosol exposure. The time course of nicotine withdrawal symptoms associated with these methods has not been reviewed in the literature. AIM The goal of this review is to discuss nicotine withdrawal symptoms associated with the cessation of nicotine, tobacco smoke, nicotine vapor, and e-cigarette aerosol exposure in rats and mice. Furthermore, age and sex differences in nicotine withdrawal symptoms are reviewed. RESULTS Cessation of nicotine, tobacco smoke, nicotine vapor, and e-cigarette aerosol exposure leads to nicotine withdrawal symptoms such as somatic withdrawal signs, changes in locomotor activity, anxiety- and depressive-like behavior, learning and memory deficits, attention deficits, hyperalgesia, and dysphoria. These withdrawal symptoms are most pronounced within the first week after cessation of nicotine exposure. Anxiety- and depressive-like behavior, and deficits in learning and memory may persist for several months. Adolescent (4-6 weeks old) rats and mice display fewer nicotine withdrawal symptoms than adults (>8 weeks old). In adult rats and mice, females show fewer nicotine withdrawal symptoms than males. The smoking cessation drugs bupropion and varenicline reduce nicotine withdrawal symptoms in rodents. CONCLUSION The nicotine withdrawal symptoms that are observed in rodents are similar to those observed in humans. Tobacco smoke and e-cigarette aerosol contain chemicals and added flavors that enhance the reinforcing properties of nicotine. Therefore, more valid animal models of tobacco and e-cigarette use need to be developed by using tobacco smoke and e-cigarette aerosol exposure methods to induce dependence.
Collapse
Affiliation(s)
| | - Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, USA
| | | | - Ryann Wilson
- Department of Psychiatry, University of Florida, Gainesville, USA
| | - Vijayapandi Pandy
- Department of Pharmacology, Chalapathi Institute of Pharmaceutical Sciences, Guntur, India
| | | |
Collapse
|
11
|
Smith LC, Tieu L, Suhandynata RT, Boomhower B, Hoffman M, Sepulveda Y, Carrette LLG, Momper JD, Fitzgerald RL, Hanham K, Dowling J, Kallupi M, George O. Cannabidiol reduces withdrawal symptoms in nicotine-dependent rats. Psychopharmacology (Berl) 2021; 238:2201-2211. [PMID: 33909102 PMCID: PMC8295227 DOI: 10.1007/s00213-021-05845-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
RATIONALE Cannabidiol (CBD) reduces craving in animal models of alcohol and cocaine use and is known to modulate nicotinic receptor function, suggesting that it may alleviate symptoms of nicotine withdrawal. However, preclinical evaluation of its efficacy is still lacking. OBJECTIVES The goal of this study was to test the preclinical efficacy of a chronic CBD treatment in reducing nicotine dependence using measures of withdrawal symptoms including somatic signs, hyperalgesia, and weight gain during acute and protracted abstinence. METHODS Male and female Wistar rats were made dependent on nicotine using osmotic minipumps (3.15 mg/kg/day) for 2 weeks, after which minipumps were removed to induce spontaneous withdrawal. Three groups received CBD injections at doses of 7.5, 15, and 30 mg/kg/day for 2 weeks, starting 1 week into chronic nicotine infusion. The control groups included rats with nicotine minipumps that received vehicle injections of sesame oil instead of CBD; rats implanted with saline minipumps received sesame oil injections (double vehicle) or the highest dose of CBD 30 mg/kg/day. Throughout the experiment, serum was collected for determination of CBD and nicotine concentrations, mechanical sensitivity threshold and withdrawal scores were measured, and body weight was recorded. RESULTS CBD prevented rats from exhibiting somatic signs of withdrawal and hyperalgesia during acute and protracted abstinence. There was no dose-response observed for CBD, suggesting a ceiling effect at the doses used and the potential for lower effective doses of CBD. The saline minipump group did not show either somatic signs of withdrawal or hyperalgesia during acute and protracted abstinence, and the highest dose of CBD used (30 mg/kg/day) did not alter these results. CONCLUSIONS This preclinical study suggests that using CBD as a strategy to alleviate the withdrawal symptoms upon nicotine cessation may be beneficial.
Collapse
Affiliation(s)
- Lauren C Smith
- Department of Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, CA, 92037, USA
- Department of Psychiatry, University of California, San Diego School of Medicine, San Diego, CA,, 92093, USA
| | - Lani Tieu
- Department of Psychiatry, University of California, San Diego School of Medicine, San Diego, CA,, 92093, USA
| | - Raymond T Suhandynata
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, 92093, USA
| | - Brent Boomhower
- Department of Psychiatry, University of California, San Diego School of Medicine, San Diego, CA,, 92093, USA
| | - Melissa Hoffman
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, 92093, USA
| | - Yadira Sepulveda
- Division of Pharmaceutical Sciences, University of California, San Diego Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, 92093, USA
| | - Lieselot L G Carrette
- Department of Psychiatry, University of California, San Diego School of Medicine, San Diego, CA,, 92093, USA
| | - Jeremiah D Momper
- Division of Pharmaceutical Sciences, University of California, San Diego Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, 92093, USA
| | - Robert L Fitzgerald
- Department of Pathology, University of California, San Diego School of Medicine, San Diego, CA, 92093, USA
| | - Kate Hanham
- CV Sciences, Inc., 10070 Barnes Canyon Road, San Diego, CA, 92121, USA
| | - Joseph Dowling
- CV Sciences, Inc., 10070 Barnes Canyon Road, San Diego, CA, 92121, USA
| | - Marsida Kallupi
- Department of Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, CA, 92037, USA.
- Department of Psychiatry, University of California, San Diego School of Medicine, San Diego, CA,, 92093, USA.
| | - Olivier George
- Department of Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, CA, 92037, USA.
- Department of Psychiatry, University of California, San Diego School of Medicine, San Diego, CA,, 92093, USA.
| |
Collapse
|
12
|
Cytisine and cytisine derivatives. More than smoking cessation aids. Pharmacol Res 2021; 170:105700. [PMID: 34087351 DOI: 10.1016/j.phrs.2021.105700] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/29/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022]
Abstract
Cytisine, a natural bioactive compound that is mainly isolated from plants of the Leguminosae family (especially the seeds of Laburnum anagyroides), has been marketed in central and eastern Europe as an aid in the clinical management of smoking cessation for more than 50 years. Its main targets are neuronal nicotinic acetylcholine receptors (nAChRs), and pre-clinical studies have shown that its interactions with various nAChR subtypes located in different areas of the central and peripheral nervous systems are neuroprotective, have a wide range of biological effects on nicotine and alcohol addiction, regulate mood, food intake and motor activity, and influence the autonomic and cardiovascular systems. Its relatively rigid conformation makes it an attractive template for research of new derivatives. Recent studies of structurally modified cytisine have led to the development of new compounds and for some of them the biological activities are mediated by still unidentified targets other than nAChRs, whose mechanisms of action are still being investigated. The aim of this review is to describe and discuss: 1) the most recent pre-clinical results obtained with cytisine in the fields of neurological and non-neurological diseases; 2) the effects and possible mechanisms of action of the most recent cytisine derivatives; and 3) the main areas warranting further research.
Collapse
|
13
|
Chellian R, Wilks I, Levin B, Xue S, Behnood-Rod A, Wilson R, McCarthy M, Ravula A, Chandasana H, Derendorf H, Bruijnzeel AW. Tobacco smoke exposure enhances reward sensitivity in male and female rats. Psychopharmacology (Berl) 2021; 238:845-855. [PMID: 33410984 PMCID: PMC7914215 DOI: 10.1007/s00213-020-05736-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
RATIONALE Systemic administration of the tobacco smoke constituent nicotine stimulates brain reward function in rats. However, it is unknown if the inhalation of tobacco smoke affects brain reward function. OBJECTIVES These experiments investigated if exposure to smoke from high-nicotine SPECTRUM research cigarettes increases reward function and affects the rewarding effects of nicotine in adult male and female Wistar rats. METHODS Reward function after smoke or nicotine exposure was investigated using the intracranial self-stimulation (ICSS) procedure. A decrease in reward thresholds reflects an increase in reward function. In the first experiment, the rats were exposed to tobacco smoke for 40 min/day for 9 days, and the rewarding effects of nicotine (0.03-0.6 mg/kg) were investigated 3 weeks later. In the second experiment, the dose effects of tobacco smoke exposure (40-min sessions, 1-4 cigarettes burnt simultaneously) on reward function were investigated. RESULTS Tobacco smoke exposure did not affect the nicotine-induced decrease in reward thresholds or response latencies in male and female rats. Smoke exposure lowered the brain reward thresholds to a similar degree in males and females and caused a greater decrease in latencies in females. There was a positive relationship between plasma nicotine and cotinine levels and the nicotine content of the SPECTRUM research cigarettes. Similar smoke exposure conditions led to higher plasma nicotine and cotinine levels in female than male rats. CONCLUSION These findings indicate that tobacco smoke exposure enhances brain reward function but does not potentiate the rewarding effects of nicotine in male and female rats.
Collapse
Affiliation(s)
- Ranjithkumar Chellian
- Department of Psychiatry, University of Florida, 1149 Newell Dr., Gainesville, FL, 32611, USA
| | - Isaac Wilks
- Department of Psychiatry, University of Florida, 1149 Newell Dr., Gainesville, FL, 32611, USA
| | - Brandon Levin
- Department of Psychiatry, University of Florida, 1149 Newell Dr., Gainesville, FL, 32611, USA
| | - Song Xue
- Department of Psychiatry, University of Florida, 1149 Newell Dr., Gainesville, FL, 32611, USA
| | - Azin Behnood-Rod
- Department of Psychiatry, University of Florida, 1149 Newell Dr., Gainesville, FL, 32611, USA
| | - Ryann Wilson
- Department of Psychiatry, University of Florida, 1149 Newell Dr., Gainesville, FL, 32611, USA
| | - Megan McCarthy
- Department of Psychiatry, University of Florida, 1149 Newell Dr., Gainesville, FL, 32611, USA
| | - Abhigyan Ravula
- Department of Pharmaceutics, University of Florida, Gainesville, USA
| | - Hardik Chandasana
- Department of Pharmaceutics, University of Florida, Gainesville, USA
| | - Hartmut Derendorf
- Department of Pharmaceutics, University of Florida, Gainesville, USA
| | - Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, 1149 Newell Dr., Gainesville, FL, 32611, USA.
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
14
|
Higgins GA, Sellers EM. 5-HT 2A and 5-HT 2C receptors as potential targets for the treatment of nicotine use and dependence. PROGRESS IN BRAIN RESEARCH 2021; 259:229-263. [PMID: 33541678 DOI: 10.1016/bs.pbr.2021.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nicotine use and dependence, typically achieved through cigarette smoking, but increasingly through vape products, is the leading cause of preventable death today. Despite a recognition that many current smokers would like to quit, the success rate at doing so is low and indicative of the persistent nature of nicotine dependence and the high urge to relapse. There are currently three main forms of pharmacotherapy approved as aids to treat nicotine dependence: a variety of nicotine replacement products (NRT's), the mixed NA/DA reuptake inhibitor bupropion (Zyban®), and the preferential nicotinic α4β2 receptor agonist drug, varenicline (Chantix®); the latter being generally recognized to be the most effective. However, each of these approaches afford only limited efficacy, and various other pharmacological approaches are being explored. This chapter focusses on approaches targeted to the serotonin (5-HT) system, namely, selective serotonin reuptake inhibitors (SSRI's) which served a pioneer role in the investigation of serotoninergic modulators in human smoking cessation trials; and secondly drugs selectively interacting with the 5-HT2A and 5-HT2C receptor systems. From an efficacy perspective, measured as smoking abstinence, the 5-HT2A agonist psychedelics, namely psilocybin, seem to show the most promise; although as the article highlights, these findings are both preliminary and there are significant challenges to the route to approval, and therapeutic use of this class should they reach approval status. Additional avenues include 5-HT2C receptor agonists, which until recently was pioneered by lorcaserin, and 5-HT2A receptor antagonists represented by pimavanserin. Each of these approaches has distinct profiles across preclinical tests of nicotine dependence, and may have therapeutic potential. It is anticipated as diagnostic and predictive biomarkers emerge, they may provide opportunities for subject stratification and opportunities for personalizing smoking cessation treatment. The clinical assessment of SSRI, 5-HT2A and/or 5-HT2C receptor-based treatments may be best served by this process.
Collapse
Affiliation(s)
- Guy A Higgins
- Intervivo Solutions Inc, Fergus, ON, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada.
| | - Edward M Sellers
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada; Department of Medicine and Psychiatry, University of Toronto, Toronto, ON, Canada; DL Global Partners Inc., Toronto, ON, Canada
| |
Collapse
|
15
|
Behnood-Rod A, Chellian R, Wilson R, Hiranita T, Sharma A, Leon F, McCurdy CR, McMahon LR, Bruijnzeel AW. Evaluation of the rewarding effects of mitragynine and 7-hydroxymitragynine in an intracranial self-stimulation procedure in male and female rats. Drug Alcohol Depend 2020; 215:108235. [PMID: 32889450 PMCID: PMC7542979 DOI: 10.1016/j.drugalcdep.2020.108235] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/28/2020] [Accepted: 08/11/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Kratom (Mitragyna speciosa Korth.) has been used in Southeast Asia for hundreds of years to increase energy, for relaxation, and to diminish opioid withdrawal. Kratom use has recently spread to Western countries. Kratom could potentially be used for the treatment of opioid withdrawal and pain, but more insight is needed into its abuse potential. Therefore, we investigated the rewarding properties of the primary kratom alkaloid mitragynine and its active metabolite 7-hydroxymitragynine, and morphine as a reference drug in male and female rats. These compounds have agonist activity at mu-opioid receptors. METHODS The compounds were tested in an intracranial self-stimulation (ICSS) procedure, which allows for the evaluation of the rewarding/aversive and sedative effects of drugs. Rewarding doses of drugs decrease the brain reward thresholds, and aversive drug doses have the opposite effect. RESULTS Mitragynine, 7-hydroxymitragynine, and morphine affected the brain reward thresholds. A high dose of 7-hydroxymitragynine (3.2 mg/kg) increased the brain reward thresholds, whereas an intermediate dose of morphine (10 mg/kg) decreased the reward thresholds. 7-Hydroxymitragynine and morphine affected the response latencies. Five mg/kg of morphine increased response latencies. 7-Hydroxymitragynine tended to increase the response latencies, but the post hoc analyses did not reveal a significant effect. There were no sex differences in the effects of mitragynine, 7-hydroxymitragynine, and morphine on the reward thresholds and the response latencies. CONCLUSIONS These initial findings indicate that mitragynine and 7-hydroxymitragynine are not rewarding in the ICSS procedure. The present results suggest that these kratom alkaloids do not have abuse potential.
Collapse
Affiliation(s)
- Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | | | - Ryann Wilson
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Takato Hiranita
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Abhisheak Sharma
- Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA,Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Francisco Leon
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Christopher R. McCurdy
- Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA,Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Lance R. McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | | |
Collapse
|
16
|
Geste JR, Levin B, Wilks I, Pompilus M, Zhang X, Esser KA, Febo M, O'Dell L, Bruijnzeel AW. Relationship Between Nicotine Intake and Reward Function in Rats With Intermittent Short Versus Long Access to Nicotine. Nicotine Tob Res 2020; 22:213-223. [PMID: 30958557 DOI: 10.1093/ntr/ntz052] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/01/2019] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Tobacco use improves mood states and smoking cessation leads to anhedonia, which contributes to relapse. Animal studies have shown that noncontingent nicotine administration enhances brain reward function and leads to dependence. However, little is known about the effects of nicotine self-administration on the state of the reward system. METHODS To investigate the relationship between nicotine self-administration and reward function, rats were prepared with intracranial self-stimulation electrodes and intravenous catheters. The rats were trained on the intracranial self-stimulation procedure and allowed to self-administer 0.03 mg/kg/infusion of nicotine. All rats self-administered nicotine daily for 10 days (1 hour/day) and were then switched to an intermittent short access (ShA, 1 hour/day) or long access (LgA, 23 hour/day) schedule (2 days/week, 5 weeks). RESULTS During the first 10 daily, 1-hour sessions, nicotine self-administration decreased the reward thresholds, which indicates that nicotine potentiates reward function. After switching to the intermittent LgA or ShA schedule, nicotine intake was lower in the ShA rats than the LgA rats. The LgA rats increased their nicotine intake over time and they gradually consumed a higher percentage of their nicotine during the light phase. The nicotinic acetylcholine receptor (nAChR) antagonist mecamylamine induced a larger increase in reward thresholds (ie, anhedonia) in the LgA rats than the ShA rats. In the LgA rats, nAChR blockade with mecamylamine decreased nicotine intake for 2 hours and this was followed by a rebound increase in nicotine intake. CONCLUSIONS A brief period of nicotine self-administration enhances reward function and a high level of nicotine intake leads to dependence. IMPLICATIONS These animal studies indicate that there is a strong relationship between the level of nicotine intake and brain reward function. A high level of nicotine intake was more rewarding than a low level of nicotine intake and nicotine dependence was observed after long, but not short, access to nicotine. This powerful combination of nicotine reward and withdrawal makes it difficult to quit smoking. Blockade of nAChRs temporarily decreased nicotine intake, but this was followed by a large rebound increase in nicotine intake. Therefore, nAChR blockade might not decrease the use of combustible cigarettes or electronic cigarettes.
Collapse
Affiliation(s)
- Jean R Geste
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Brandon Levin
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Isaac Wilks
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Marjory Pompilus
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Xiping Zhang
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL
| | - Karyn A Esser
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL.,Department of Neuroscience, University of Florida, Gainesville, FL
| | - Laura O'Dell
- Department of Psychology, University of Texas at El Paso, El Paso, TX
| | - Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL.,Department of Neuroscience, University of Florida, Gainesville, FL
| |
Collapse
|
17
|
Xue S, Behnood-Rod A, Wilson R, Wilks I, Tan S, Bruijnzeel AW. Rewarding Effects of Nicotine in Adolescent and Adult Male and Female Rats as Measured Using Intracranial Self-stimulation. Nicotine Tob Res 2020; 22:172-179. [PMID: 30452710 DOI: 10.1093/ntr/nty249] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/14/2018] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Tobacco is highly addictive, and after the development of dependence, it is difficult to quit smoking. Therefore, it is important to understand the factors that play a role in the initiation of smoking. The rewarding effects of nicotine play a role in the initiation of smoking and the goal of the present study was to determine the rewarding effects of nicotine in adolescent and adult male and female rats. METHODS Male and female Wistar rats were prepared with intracranial self-stimulation (ICSS) electrodes between postnatal day (P) 23 and 33. They were then trained on the ICSS procedure and the effect of nicotine (0, 0.03, 0.1, 0.3 mg/kg) on the reward thresholds and response latencies was investigated during adolescence (P40-59) or adulthood (>P75). RESULTS Nicotine lowered the brain reward thresholds of the adult and adolescent male and female rats. The nicotine-induced decrease in the reward thresholds was the same in the adult male and adult female rats. However, nicotine induced a greater decrease in the reward thresholds of the adolescent female rats than the adolescent male rats. Nicotine decreased the response latencies of all groups and there was no effect of age or sex. CONCLUSIONS Nicotine enhances reward function and psychomotor performance in adolescent and adult male and female rats. Adolescent female rats are more sensitive to the acute rewarding effects of nicotine than adolescent male rats. Therefore, the rewarding effects of nicotine might play a greater role in the initiation of smoking in adolescent females than in adolescent males. IMPLICATIONS The great majority of people start smoking during adolescence. The present studies suggest that during this period female rats are more sensitive to the acute rewarding effects of low and intermediate doses of nicotine than male rats. The rewarding properties of nicotine play a role in the initiation of smoking and establishing habitual smoking. Therefore, the present findings might explain why adolescent females are at a higher risk for becoming nicotine dependent than adolescent males.
Collapse
Affiliation(s)
- Song Xue
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Ryann Wilson
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Isaac Wilks
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Sijie Tan
- Department of Psychiatry, University of Florida, Gainesville, FL.,Department of Histology and Embryology, University of South China, Hengyang, Hunan, China
| | - Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL.,Department of Neuroscience, University of Florida, Gainesville, FL
| |
Collapse
|
18
|
Moerke MJ, McMahon LR, Wilkerson JL. More than Smoke and Patches: The Quest for Pharmacotherapies to Treat Tobacco Use Disorder. Pharmacol Rev 2020; 72:527-557. [PMID: 32205338 DOI: 10.1124/pr.119.018028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tobacco use is a persistent public health issue. It kills up to half its users and is the cause of nearly 90% of all lung cancers. The main psychoactive component of tobacco is nicotine, primarily responsible for its abuse-related effects. Accordingly, most pharmacotherapies for smoking cessation target nicotinic acetylcholine receptors (nAChRs), nicotine's major site of action in the brain. The goal of the current review is twofold: first, to provide a brief overview of the most commonly used behavioral procedures for evaluating smoking cessation pharmacotherapies and an introduction to pharmacokinetic and pharmacodynamic properties of nicotine important for consideration in the development of new pharmacotherapies; and second, to discuss current and potential future pharmacological interventions aimed at decreasing tobacco use. Attention will focus on the potential for allosteric modulators of nAChRs to offer an improvement over currently approved pharmacotherapies. Additionally, given increasing public concern for the potential health consequences of using electronic nicotine delivery systems, which allow users to inhale aerosolized solutions as an alternative to smoking tobacco, an effort will be made throughout this review to address the implications of this relatively new form of nicotine delivery, specifically as it relates to smoking cessation. SIGNIFICANCE STATEMENT: Despite decades of research that have vastly improved our understanding of nicotine and its effects on the body, only a handful of pharmacotherapies have been successfully developed for use in smoking cessation. Thus, investigation of alternative pharmacological strategies for treating tobacco use disorder remains active; allosteric modulators of nicotinic acetylcholine receptors represent one class of compounds currently under development for this purpose.
Collapse
Affiliation(s)
- M J Moerke
- Division of Preclinical Pharmacology, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (M.J.M.) and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (L.R.M., J.L.W.)
| | - L R McMahon
- Division of Preclinical Pharmacology, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (M.J.M.) and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (L.R.M., J.L.W.)
| | - J L Wilkerson
- Division of Preclinical Pharmacology, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland (M.J.M.) and Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida (L.R.M., J.L.W.)
| |
Collapse
|
19
|
Swain Y, Muelken P, Skansberg A, Lanzdorf D, Haave Z, LeSage MG, Gewirtz JC, Harris AC. Higher anhedonia during withdrawal from initial opioid exposure is protective against subsequent opioid self-administration in rats. Psychopharmacology (Berl) 2020; 237:2279-2291. [PMID: 32388620 PMCID: PMC7354901 DOI: 10.1007/s00213-020-05532-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/17/2020] [Indexed: 12/15/2022]
Abstract
RATIONALE Understanding factors contributing to individual differences in vulnerability to opioid addiction is essential for developing more effective preventions and treatments, yet few reliable behavioral predictors of subsequent opioid self-administration have been identified in rodents. Sensitivity to the acute effects of initial drug exposure predicts later addiction vulnerability in both humans and animals, but the relationship between sensitivity to withdrawal from initial drug exposure and later drug use vulnerability is unclear. OBJECTIVE The goal of the current study was to evaluate whether the degree of anhedonia experienced during withdrawal from early opioid exposure predicts subsequent vulnerability to opioid self-administration. METHODS Rats were first tested for withdrawal sensitivity following acute injections of morphine (i.e., "acute dependence"), measured as elevations in intracranial self-stimulation (ICSS) thresholds (anhedonia-like behavior) during naloxone-precipitated and spontaneous withdrawal. Rats were then tested for addiction-like behavior using various measures of i.v. morphine self-administration (MSA) including acquisition, demand, extinction, and reinstatement induced by morphine, stress, and/or drug-associated cues. RESULTS Greater naloxone-precipitated withdrawal across repeated morphine injections and greater peak spontaneous withdrawal severity following a single morphine injection were associated with lower addiction-like behavior on multiple MSA measures. Withdrawal-induced anhedonia predicted a wider range of MSA measures than did any individual measure of MSA itself. CONCLUSIONS Our data establish WIA as one of the first behavioral measures to predict individual differences in opioid SA in rodents. This model promises to be useful for furthering our understanding of behavioral and neurobiological mechanisms underlying vulnerability to opioid addiction.
Collapse
Affiliation(s)
- Yayi Swain
- Hennepin Healthcare Research Institute,,University of Minnesota Department of Psychology
| | | | - Annika Skansberg
- Hennepin Healthcare Research Institute,,University of Minnesota Department of Psychology
| | - Danielle Lanzdorf
- Hennepin Healthcare Research Institute,,University of Minnesota Department of Psychology
| | - Zachary Haave
- Hennepin Healthcare Research Institute,,University of Minnesota Department of Neuroscience
| | - Mark G. LeSage
- Hennepin Healthcare Research Institute,,University of Minnesota Department of Psychology,,University of Minnesota Department of Medicine
| | - Jonathan C. Gewirtz
- University of Minnesota Department of Psychology,,University of Minnesota Department of Neuroscience
| | - Andrew C. Harris
- Hennepin Healthcare Research Institute,,University of Minnesota Department of Psychology,,University of Minnesota Department of Medicine
| |
Collapse
|
20
|
Chellian R, Behnood-Rod A, Wilson R, Kamble SH, Sharma A, McCurdy CR, Bruijnzeel AW. Adolescent nicotine and tobacco smoke exposure enhances nicotine self-administration in female rats. Neuropharmacology 2020; 176:108243. [PMID: 32702403 DOI: 10.1016/j.neuropharm.2020.108243] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/24/2020] [Accepted: 07/13/2020] [Indexed: 12/30/2022]
Abstract
Most people start experimenting with tobacco products or e-cigarettes in early adolescence and become habitual smokers in late adolescence or adulthood. These studies investigated if exposure to tobacco smoke or nicotine during early and mid-adolescence affects nicotine intake in late adolescence and early adulthood. Male and female rats were exposed to tobacco smoke from low- and high-nicotine SPECTRUM cigarettes or nicotine (0.3 mg/kg, twice a day) from postnatal day (P) 24-42. The self-administration sessions started at P55. The rats self-administered nicotine for 14-15 days under a fixed-ratio 1 schedule, and on the first day, the maximum number of infusions was twenty. Exposure to smoke from high, but not low, nicotine cigarettes during adolescence increased nicotine self-administration in female but not male rats. Adolescent treatment with nicotine facilitated nicotine self-administration. On the first day of nicotine self-administration, nicotine-treated rats reached the maximum number of infusions before the saline-treated control rats. Nicotine intake was also higher in the nicotine-treated female rats than in the saline-treated females. There was no sex difference in nicotine intake in controls when the data from the studies were combined. Smoke exposure led to a dose-dependent increase in plasma nicotine and cotinine levels in adolescent rats. Exposure to smoke from high-nicotine cigarettes and 0.3 mg/kg of nicotine led to plasma nicotine and cotinine levels that are similar to those in tobacco users. These findings indicate that in females, but not males, exposure to nicotine during adolescence may facilitate smoking and e-cigarette use later in life.
Collapse
Affiliation(s)
| | - Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Ryann Wilson
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Shyam H Kamble
- Department of Pharmaceutics, University of Florida, Gainesville, FL, USA; Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA
| | - Abhisheak Sharma
- Department of Pharmaceutics, University of Florida, Gainesville, FL, USA; Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA
| | - Christopher R McCurdy
- Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA; Department of Medicinal Chemistry, University of Florida, Gainesville, FL, USA
| | | |
Collapse
|
21
|
Huang X, Xu H. Advances on the Bioactivities, Total Synthesis, Structural Modification, and Structure-Activity Relationships of Cytisine Derivatives. Mini Rev Med Chem 2020; 20:369-395. [DOI: 10.2174/1389557519666191104121821] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/23/2019] [Accepted: 09/18/2019] [Indexed: 02/05/2023]
Abstract
Cytisine is a quinolizidine alkaloid isolated from various Leguminosae plants. Cytisine and
its derivatives exhibit a broad range of biological properties, such as smoking cessation aid, antidepressant,
neuroprotective, nootropic, anticancer, antiviral, antiparasitic, antidiabetic, insecticidal, and nematicidal
activities. In this review, the progress of cytisine and its derivatives in regard to bioactivities,
total synthesis, structural modifications focusing on their N-12 position and lactam ring is reported.
Additionally, the structure-activity relationships of cytisine and its derivatives are also discussed.
Collapse
Affiliation(s)
- Xiaobo Huang
- Research Institute of Pesticidal Design & Synthesis, College of Plant Protection/College of Chemistry and Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| | - Hui Xu
- Research Institute of Pesticidal Design & Synthesis, College of Plant Protection/College of Chemistry and Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi Province, China
| |
Collapse
|
22
|
Wilkerson JL, Deba F, Crowley ML, Hamouda AK, McMahon LR. Advances in the In vitro and In vivo pharmacology of Alpha4beta2 nicotinic receptor positive allosteric modulators. Neuropharmacology 2020; 168:108008. [PMID: 32113032 DOI: 10.1016/j.neuropharm.2020.108008] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 02/08/2020] [Accepted: 02/11/2020] [Indexed: 11/29/2022]
Abstract
Receptors containing α4 and β2 subunits are a major neuronal nicotinic acetylcholine receptor (nAChR) subtype in the brain. This receptor plays a critical role in nicotine addiction, with potential smoking cessation therapeutics producing modulation of α4β2 nAChR. In addition, compounds that act as agonists at α4β2 nAChR may be useful for the treatment of pathological pain. Further, as the α4β2 nAChR has been implicated in cognition, therapeutics that act as α4β2 nAChR agonists are also being examined as treatments for cognitive disorders and neurological diseases that impact cognitive function, such as Alzheimer's disease and schizophrenia. This review will cover the molecular in vitro evidence that allosteric modulators of the α4β2 neuronal nAChR provide several advantages over traditional α4β2 nAChR orthosteric ligands. Specifically, we explore the concept that nAChR allosteric modulators allow for greater pharmacological selectivity, while minimizing potential deleterious off-target effects. Further, here we discuss the development and preclinical in vivo behavioral assessment of allosteric modulators at the α4β2 neuronal nAChR as therapeutics for smoking cessation, pathological pain, as well as cognitive disorders and neurological diseases that impact cognitive function. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology'.
Collapse
Affiliation(s)
- Jenny L Wilkerson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States.
| | - Farah Deba
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Texas at Tyler, Tyler, TX, 75799, United States
| | - Morgan L Crowley
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States
| | - Ayman K Hamouda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Texas at Tyler, Tyler, TX, 75799, United States.
| | - Lance R McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States.
| |
Collapse
|
23
|
Ravula A, Chandasana H, Jagnarine D, Wall SC, Setlow B, Febo M, Bruijnzeel AW, Derendorf H. Pharmacokinetic and Pharmacodynamic Characterization of Tetrahydrocannabinol-Induced Cannabinoid Dependence After Chronic Passive Cannabis Smoke Exposure in Rats. Cannabis Cannabinoid Res 2019; 4:240-254. [PMID: 32042924 DOI: 10.1089/can.2019.0049] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Introduction: Cannabis is the most widely used illicit drug in the US, and cannabis use among young adults continues to rise. Previous studies have shown that chronic administration of delta 9-tetrahydrocannabinol (THC), the main psychoactive component of cannabis, induces dependence in animal models. Because smoking is the most frequent route of THC self-administration, it is critical to investigate the effects of cannabis smoke inhalation. The goal of the current study was to develop a rat model to characterize the pharmacokinetics (PKs) of THC after cannabis smoke inhalation, and to determine if chronic cannabis smoke inhalation leads to the development of cannabis dependence. Materials and Methods: For the PK study, male Wistar rats were administered THC intravenously (1 mg/kg) or exposed to smoke from 5 or 10 sequentially smoked cannabis cigarettes (5.3% THC) in an automated smoking machine. Plasma samples were collected from 10 min to 10 hours post smoke exposure (or intravenous administration) and analyzed using liquid chromatography-mass spectrometry to characterize the PK of THC. A three-compartment PK model was used to characterize the PKs. In a separate study, three groups of male Wistar rats were trained in an intracranial self-stimulation (ICSS) procedure, and exposed to smoke from burning 5 or 10 cannabis cigarettes (or clean air control conditions), 5 days/week for 4 weeks. Discussion and Conclusions: Across exposure days, the change from baseline in ICSS thresholds for cannabis smoke-exposed groups was significantly lower and response latencies were significantly faster in the cannabis smoke-exposed groups compared to controls, suggesting that chronic cannabis smoke exposure has rewarding properties. Acute administration of the CB1 receptor antagonist rimonabant (0.3, 1.0, 3.0 mg/kg) induced a dose-dependent increase in ICSS thresholds in the smoke-exposed rats, suggestive of dependence and withdrawal. Finally, an effect compartment PK-pharmacodynamic model was used to describe the relationship between THC concentrations and changes in ICSS thresholds after cannabis smoke exposure.
Collapse
Affiliation(s)
- Abhigyan Ravula
- Department of Pharmaceutics, University of Florida, Gainesville, Florida
| | - Hardik Chandasana
- Department of Pharmaceutics, University of Florida, Gainesville, Florida
| | - Darin Jagnarine
- Department of Psychiatry, University of Florida, Gainesville, Florida
| | - Shannon C Wall
- Department of Psychiatry, University of Florida, Gainesville, Florida
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, Florida
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, Florida
| | | | - Hartmut Derendorf
- Department of Pharmaceutics, University of Florida, Gainesville, Florida
| |
Collapse
|
24
|
Tutka P, Vinnikov D, Courtney RJ, Benowitz NL. Cytisine for nicotine addiction treatment: a review of pharmacology, therapeutics and an update of clinical trial evidence for smoking cessation. Addiction 2019; 114:1951-1969. [PMID: 31240783 DOI: 10.1111/add.14721] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/08/2019] [Accepted: 06/18/2019] [Indexed: 01/30/2023]
Abstract
AIMS To review cytisine's history of use, pre-clinical evidence, clinical pharmacokinetics, efficacy, adverse reactions (ARs) and safety for smoking cessation. METHODS A synoptic review of the use of cytisine as a smoking cessation medication, mechanism of action, pharmacokinetics and safety. Relevant literature on data included in these sections were identified through a search of 11 databases with additional literature obtained from reports and monographs. Three databases (PubMed, EMBASE and www.elibrary.ru) were systematically searched for studies published from 2012 to August 2018 in any language to provide an updated meta-analysis of cytisine's efficacy and ARs for smoking cessation compared with placebo. We pooled the relative risks (RR) of abstinence in the efficacy analysis and RR of ARs, either reported by the authors or calculated from the reports. RESULTS Cytisine has been in use since 1964 and is currently marketed in 18 countries. Systemic bioavailability from oral ingestion is high and clearance is primarily renal, with minimal or no metabolism. Brain uptake in animal models is moderate. The plasma half-life averages 4.8 hours. Eight studies were included for meta-analysis of efficacy. With heterogeneous results, the overall RR versus placebo of successful continuous abstinence at the longest follow-up was 1.74 [95% confidence interval (CI) = 1.38-2.19]. Nausea, vomiting, dyspepsia, upper abdominal pain and dry mouth that were mild or moderate were the most common ARs, with RR versus placebo 1.10 (95% CI = 0.95-1.28). The cost of cytisine in eastern and central Europe is several-fold less than that of other smoking cessation medications. CONCLUSIONS Cytisine is a low-cost medication found to increase the likelihood of smoking cessation. The most frequently reported ARs of cytisine involve gastrointestinal symptoms that are mostly reported as either mild or moderate in severity.
Collapse
Affiliation(s)
- Piotr Tutka
- Department of Experimental and Clinical Pharmacology, University of Rzeszów, Rzeszów, Poland.,Laboratory for Innovative Research in Pharmacology, University of Rzeszów, Rzeszów, Poland.,National Drug and Alcohol Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Denis Vinnikov
- School of Public Health, Al-Farabi Kazakh National University, Almaty, Kazakhstan.,Biological Institute, National Research Tomsk State University, Tomsk, Russia
| | - Ryan J Courtney
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Neal L Benowitz
- Division of Clinical Pharmacology and Experimental Therapeutics, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
25
|
Laikowski MM, Reisdorfer F, Moura S. NAChR α4β2 Subtype and their Relation with Nicotine Addiction, Cognition, Depression and Hyperactivity Disorder. Curr Med Chem 2019; 26:3792-3811. [PMID: 29637850 DOI: 10.2174/0929867325666180410105135] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/27/2017] [Accepted: 04/05/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND Neuronal α4β2 nAChRs are receptors involved in the role of neurotransmitters regulation and release, and this ionic channel participates in biological process of memory, learning and attention. This work aims to review the structure and functioning of the α4β2 nAChR emphasizing its role in the treatment of associated diseases like nicotine addiction and underlying pathologies such as cognition, depression and attention-deficit hyperactivity disorder. METHODS The authors realized extensive bibliographic research using the descriptors "Nicotine Receptor α4β2" and "cognition", "depression", "attention-deficit hyperactivity disorder", besides cross-references of the selected articles and after analysis of references in the specific literature. RESULTS As results, it was that found 179 relevant articles presenting the main molecules with affinity to nAChR α4β2 related to the cited diseases. The α4β2 nAChR subtype is a remarkable therapeutic target since this is the most abundant receptor in the central nervous system. CONCLUSION In summary, this review presents perspectives on the pharmacology and therapeutic targeting of α4β2 nAChRs for the treatment of cognition and diseases like nicotine dependence, depression and attention-deficit hyperactivity disorder.
Collapse
Affiliation(s)
- Manuela M Laikowski
- Laboratory of Natural and Synthetics Products, University of Caxias do Sul, Caxias do Sul, Brazil
| | - Fávero Reisdorfer
- Laboratory of Drug Development and Quality Control, University Federal of Pampa, Brazil
| | - Sidnei Moura
- Laboratory of Natural and Synthetics Products, University of Caxias do Sul, Caxias do Sul, Brazil
| |
Collapse
|
26
|
Hamouda AK, Jackson A, Bagdas D, Imad Damaj M. Reversal of Nicotine Withdrawal Signs Through Positive Allosteric Modulation of α4β2 Nicotinic Acetylcholine Receptors in Male Mice. Nicotine Tob Res 2019; 20:903-907. [PMID: 29059422 DOI: 10.1093/ntr/ntx183] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/15/2017] [Indexed: 01/03/2023]
Abstract
Introduction Nicotine withdrawal symptoms are important factors in determining the relapse rate to tobacco smoking and drugs that diminish these symptoms would potentially have a higher success rate as smoking cessation aids. Unlike US Food and Drug administration approved smoke cessation aids (nicotine and varenicline) which act as nicotinic acetylcholine receptors (nAChRs) agonists, desformylflustrabromine (dFBr) acts as a nAChR positive allosteric modulator with higher selectivity to the α4β2 nAChR. In animal studies, dFBr was well tolerated and reduced intravenous nicotine self-administration. In this study, we use behavioral test in mouse model of spontaneous nicotine withdrawal to assess the effect of dFBr on nicotine withdrawal symptoms. Methods Spontaneous nicotine withdrawal in nicotine-dependent ICR male mice was established 18-24 h after termination (minipump removal) of 14 days infusion of nicotine. After that (day 15), spontaneous signs of nicotine withdrawal were examined in the following order: anxiety-like behaviors, somatic signs, and then hyperalgesia using previously published behavioral protocols. Fifteen minutes before withdrawal signs testing, mice received a subcutaneous acute injection of vehicle or dFBr at the doses of 0.02, 0.1, and 1 mg/kg to determine the effect of dFBr on nicotine withdrawal symptoms. Results dFBr produced dose-dependent reversal of nicotine withdrawal signs in mouse model of spontaneous nicotine withdrawal. Implications Positive allosteric modulators of nAChR such as dFBr reduce nicotine withdrawal symptoms supporting the potential clinical use of this novel class of nAChR-based therapeutics as smoking cessation aid.
Collapse
Affiliation(s)
- Ayman K Hamouda
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M Health Sciences Center MS, Kingsville, TX
| | - Asti Jackson
- Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA
| | - Deniz Bagdas
- Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA.,Experimental Animals Breeding and Research Center, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - M Imad Damaj
- Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
27
|
Tan S, Xue S, Behnood-Rod A, Chellian R, Wilson R, Knight P, Panunzio S, Lyons H, Febo M, Bruijnzeel AW. Sex differences in the reward deficit and somatic signs associated with precipitated nicotine withdrawal in rats. Neuropharmacology 2019; 160:107756. [PMID: 31487496 DOI: 10.1016/j.neuropharm.2019.107756] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/27/2019] [Accepted: 08/31/2019] [Indexed: 01/27/2023]
Abstract
Female smokers are more likely to relapse than male smokers, but little is known about sex differences in nicotine withdrawal. Therefore, male and female rats were prepared with minipumps that contained nicotine or saline and sex differences in precipitated and spontaneous nicotine withdrawal were investigated. The intracranial self-stimulation (ICSS) procedure was used to assess mood states. Elevations in brain reward thresholds reflect a deficit in reward function. Anxiety-like behavior was investigated after the acute nicotine withdrawal phase in a large open field and the elevated plus maze test. The nicotinic receptor antagonist mecamylamine elevated the brain reward thresholds of the nicotine-treated rats but did not affect those of the saline-treated control rats. A low dose of mecamylamine elevated the brain reward thresholds of the nicotine-treated male rats but not those of the females. Mecamylamine also precipitated more somatic withdrawal signs in the nicotine-treated male than female rats. Minipump removal elevated the brain reward thresholds of the nicotine-treated rats for about 36 h but did not affect those of the saline-treated rats. There was no sex difference in the reward deficit during spontaneous nicotine withdrawal. In addition, the nicotine-treated male and female rats did not display increased anxiety-like behavior three to four days after minipump removal. In conclusion, these studies suggest that relatively low doses of a nicotinic receptor antagonist induce a greater reward deficit and more somatic withdrawal signs in male than female rats, but there is no sex difference in the reward deficit during spontaneous withdrawal.
Collapse
Affiliation(s)
- Sijie Tan
- Department of Psychiatry, University of Florida, Gainesville, FL, USA; Department of Histology and Embryology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Song Xue
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | | | - Ryann Wilson
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Parker Knight
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Stefany Panunzio
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Hannah Lyons
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL, USA; Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL, USA; Department of Neuroscience, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
28
|
Addicott MA, Sweitzer MM, McClernon FJ. The Effects of Nicotine and Tobacco Use on Brain Reward Function: Interaction With Nicotine Dependence Severity. Nicotine Tob Res 2019; 21:764-771. [PMID: 29584917 PMCID: PMC6784410 DOI: 10.1093/ntr/nty059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 03/22/2018] [Indexed: 11/13/2022]
Abstract
INTRODUCTION This study investigated the effects of nicotine/tobacco on neural activation during performance of a monetary incentive delay task. AIMS AND METHODS Prior to each scan, nonsmokers received nicotine or placebo nasal spray, and smokers were smoking satiated or 24-hour withdrawn. During the scan, participants made timed responses to reward-related cues and received feedback. Parameter estimates from cue- and feedback-related activation in medial prefrontal regions and the nucleus accumbens were extracted and underwent within- and between-group analyses. Smokers' nicotine dependence severity was included as a continuous predictor variable for neural activation. RESULTS Among smokers (n = 21), withdrawal decreased cue-related activation in the supplementary motor area and ventromedial prefrontal cortex, and the difference in activation (satiety > withdrawal) in these regions negatively correlated with nicotine dependence severity (Fagerström Test for Nicotine Dependence). Among nonsmokers (n = 22), nicotine increased the difference in nucleus accumbens activation between rewarded and nonrewarded feedback phases. Tobacco withdrawal and acute nicotine also had widespread effects on activation throughout the brain during the feedback phase. CONCLUSIONS Acute nicotine in nonsmokers may have increased the salience of feedback information, but produced few effects on reward-related activation overall, perhaps reflecting nicotine's modest, indirect effects on reward processing. Conversely, tobacco withdrawal decreased activation compared with satiety, and this difference between conditions correlated with nicotine dependence severity. This suggests that as smokers become more dependent on nicotine, tobacco withdrawal has a more pronounced effect on reward processing. IMPLICATIONS Relative to the acute effects of nicotine in nonsmokers, withdrawal from daily tobacco use had more significant effects on reward-related brain activation. This study suggests that the effects of tobacco withdrawal on reward-related brain function interact with subjects' level of nicotine dependence severity. These are potentially important sources of variability that could contribute to smoking cessation outcomes.
Collapse
Affiliation(s)
- Merideth A Addicott
- Department of Psychiatry, University of Arkansas for Medical Science, Little Rock, AR
| | - Maggie M Sweitzer
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, University of Arkansas for Medical Sciences, Durham, NC
| | - F Joseph McClernon
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, University of Arkansas for Medical Sciences, Durham, NC
| |
Collapse
|
29
|
Doran N, Dubrava S, Anthenelli RM. Effects of Varenicline, Depressive Symptoms, and Region of Enrollment on Smoking Cessation in Depressed Smokers. Nicotine Tob Res 2019; 21:156-162. [PMID: 29471329 PMCID: PMC6329403 DOI: 10.1093/ntr/nty033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/14/2018] [Indexed: 11/12/2022]
Abstract
Introduction Despite effective treatments, relapse to smoking remains a vexing global health problem. One predictor of relapse is depressive symptoms. Medications such as varenicline reduce withdrawal-related symptoms of depression, reducing relapse. This study examined whether varenicline moderated the effect of depressive symptoms on relapse, and whether this varied by region of enrollment. Methods Adult smokers (n = 525; 37% male) with past or current, stable major depressive disorder recruited from United States (n = 255), and European (n = 270) sites participated in a randomized, double-blind cessation treatment trial including 12 weeks of varenicline or placebo, with 40-week nontreatment follow-up. Results Longitudinal and binary logistic regressions were used to model the probability of sustained abstinence by end of treatment and point-prevalence abstinence in follow-up. The association between depression symptoms and abstinence was moderated by intervention group at end of treatment, and by region during follow-up: more severe symptoms were associated with end-of-treatment relapse for placebo (odds ratio [OR] = 0.91, p = .003), but not varenicline (OR = 0.99, p = .568). During follow-up, increased symptoms of depression predicted greater likelihood of smoking for European (p = .009) but not US participants. Europeans were more likely to be abstinent for both outcomes (p < .01). Conclusions These results extend studies demonstrating varenicline is associated with less withdrawal-related depression, and suggest it aids cessation even in smokers with depressive symptoms. Findings also suggest regional differences in the relationship between depressive symptoms and cessation that may be related to differences in prevalence. Implications This study indicates varenicline may aid cessation partially by reducing withdrawal-related symptoms of depression. It also suggests that the impact of depressive symptoms on cessation varies regionally, and that this variation may be related to differences in smoking prevalence.
Collapse
Affiliation(s)
- Neal Doran
- Department of Psychiatry, Health Sciences, University of California, San Diego
- Mental Health Care Line, Veterans Affairs San Diego Healthcare System
| | | | - Robert M Anthenelli
- Department of Psychiatry, Health Sciences, University of California, San Diego
| |
Collapse
|
30
|
Trigo JM, Le Foll B. Nicotine Self-Administration as Paradigm for Medication Discovery for Smoking Cessation: Recent Findings in Medications Targeting the Cholinergic System. Methods Mol Biol 2019; 2011:165-193. [PMID: 31273700 DOI: 10.1007/978-1-4939-9554-7_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Tobacco kills every year approximately six million people as a direct result of direct use, and it is still considered one of the most excruciating threats for human health worldwide. The low successful rates of the currently available pharmacotherapies to assist in quitting tobacco use suggest there is a need for more effective treatments.The intravenous self-administration (IVSA) paradigm is considered the gold standard to study voluntary drug intake in animal models, including nicotine. The IVSA paradigm has been used to identify key mechanisms involved in the addictive properties of nicotine in both rodents and nonhuman primates. In this chapter we describe how the IVSA paradigm has served to further investigate the role of nicotinic acetylcholine receptors (nAChRs) in the reinforcing properties of nicotine. Notably, this review will cover recent advances (i.e., research carried out during the past decade) using the IVSA paradigm, with a focus on the status of research on current smoking cessation medications (such as varenicline and bupropion) and of other nAChR ligands.The combination of the IVSA paradigm with pharmacological and genetic tools (e.g., knockout animals) has greatly contributed to our understanding of the role of specific subtype nAChRs in nicotine reinforcement processes. We also discuss some of the limitations of the IVSA paradigm so these can be taken into consideration when interpreting and designing new studies.
Collapse
Affiliation(s)
- Jose M Trigo
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada.
- Addictions Division, CAMH, Toronto, ON, Canada.
| |
Collapse
|
31
|
Abstract
INTRODUCTION Tobacco use causes one premature death every six seconds. Current smoking cessation aids include nicotine replacement therapies, bupropion, and varenicline. Although more than 70% of smokers express a desire to quit, fewer than 3% remain abstinent for more than one year, highlighting a critical need for more efficacious smoking cessation treatments. Areas covered: The authors discuss the rationale, preclinical and clinical development of varenicline for smoking cessation. They cover the development of varenicline as a partial agonist at α4β2 receptors, the primary neural substrate for nicotine reward. Then, they discuss evidence from preclinical studies indicating varenicline's efficacy in blocking nicotine reward, followed by clinical trials demonstrating safety and efficacy in sustaining abstinence in smokers. Finally, they cover post-market surveillance, including caution in heavy machine operators, putative cardiovascular risk, and the repealed warning for adverse neuropsychiatric events. Expert opinion: Varenicline development was based on strong theoretical rationale and preclinical evidence. Clinical studies indicate that varenicline is safe and more effective in sustaining abstinence than placebo, bupropion or nicotine replacement therapies. However, given that continuous abstinence rates across studies remain low (18 ~ 30% with varenicline; 4 ~ 10% with placebo), novel and more effective medications targeting other nicotinic or glutamate receptors for smoking cessation are required.
Collapse
Affiliation(s)
- Chloe J. Jordan
- Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
32
|
Neuroprotection of Cytisine Against Cerebral Ischemia–Reperfusion Injury in Mice by Regulating NR2B-ERK/CREB Signal Pathway. Neurochem Res 2018; 43:1575-1586. [DOI: 10.1007/s11064-018-2572-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/09/2018] [Accepted: 06/08/2018] [Indexed: 10/28/2022]
|
33
|
Bagdas D, Alkhlaif Y, Jackson A, Carroll FI, Ditre JW, Damaj MI. New insights on the effects of varenicline on nicotine reward, withdrawal and hyperalgesia in mice. Neuropharmacology 2018; 138:72-79. [PMID: 29860196 DOI: 10.1016/j.neuropharm.2018.05.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/16/2018] [Accepted: 05/19/2018] [Indexed: 12/16/2022]
Abstract
Varenicline, a partial agonist for α4β2* nicotinic acetylcholine receptors (nAChRs) and a full agonist for α3β4 and α7 nAChRs, is approved for smoking cessation treatment. Although, partial agonism at α4β2* nAChRs is believed to be the mechanism underlying the effects of varenicline on nicotine reward, the contribution of other nicotinic subtypes to varenicline's effects on nicotine reward is currently unknown. Therefore, we examined the role of α5 and α7 nAChR subunits in the effects of varenicline on nicotine reward using the conditioned place preference (CPP) test in mice. Moreover, the effects of varenicline on nicotine withdrawal-induced hyperalgesia and aversion are unknown. We also examined the reversal of nicotine withdrawal in mouse models of dependence by varenicline. Varenicline dose-dependently blocked the development and expression of nicotine reward in the CPP test. The blockade of nicotine reward by varenicline (0.1 mg/kg) was preserved in α7 knockout mice but reduced in α5 knockout mice. Administration of varenicline at high dose of 2.5 mg/kg resulted in a place aversion that was dependent on α5 nAChRs but not β2 nAChRs. Furthermore, varenicline (0.1 and 0.5 mg/kg) reversed nicotine withdrawal signs such as hyperalgesia and somatic signs and withdrawal-induced aversion in a dose-related manner. Our results indicate that the α5 nAChR subunit plays a role in the effects of varenicline on nicotine reward in mice. Moreover, the mediation of α5 nAChRs, but not β2 nAChRs are probably needed for aversive properties of varenicline at high dose. Varenicline was also shown to reduce several nicotine withdrawal signs.
Collapse
Affiliation(s)
- Deniz Bagdas
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States; The Center for the Study for Tobacco Products, Virginia Commonwealth University, Richmond, VA, United States.
| | - Yasmin Alkhlaif
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Asti Jackson
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - F Ivy Carroll
- Center for Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, NC, United States
| | - Joseph W Ditre
- Department of Psychology, Syracuse University, Syracuse, NY, United States
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States; The Center for the Study for Tobacco Products, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
34
|
Self-administration of the synthetic cathinone MDPV enhances reward function via a nicotinic receptor dependent mechanism. Neuropharmacology 2018; 137:286-296. [PMID: 29778945 DOI: 10.1016/j.neuropharm.2018.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/30/2018] [Accepted: 05/06/2018] [Indexed: 01/28/2023]
Abstract
Methylenedioxypyrovalerone (MDPV) is an addictive synthetic drug with severe side effects. Previous studies have shown that MDPV has positive reinforcing properties. However, little is known about the effect of MDPV self-administration on the state of the brain reward system and the neuronal mechanisms by which MDPV mediates its effects. The goal of the present studies was to determine the effect of MDPV self-administration on reward function and the role of cholinergic neurotransmission in the reinforcing effects of MDPV. To study the effect of MDPV self-administration on the brain reward system, rats were prepared with intravenous catheters and intracranial self-stimulation electrodes (ICSS). For 10 days, the reward thresholds were assessed immediately before (23 h post prior session) and after 1 h of MDPV self-administration. The reward thresholds were decreased immediately after MDPV self-administration, which is indicative of a potentiation of brain reward function. The reward thresholds 23 h after MDPV intake gradually increased over time, which is indicative of anhedonia. Pretreatment with the nicotinic acetylcholine receptor (nAChR) antagonist mecamylamine decreased the self-administration of MDPV and completely prevented the decrease in reward thresholds. A control study with palatable chocolate pellets showed that responding for a natural reinforcer does not affect the state of the brain reward system. Furthermore, mecamylamine did not affect responding for food pellets. In conclusion, the self-administration of MDPV potentiates reward function and nAChR blockade prevents the reward enhancing effects of MDPV self-administration. Preventing the MDPV-induced increase in cholinergic neurotransmission might be a safe approach to diminish MDPV abuse.
Collapse
|
35
|
Colon-Perez LM, Pino JA, Saha K, Pompilus M, Kaplitz S, Choudhury N, Jagnarine DA, Geste JR, Levin BA, Wilks I, Setlow B, Bruijnzeel AW, Khoshbouei H, Torres GE, Febo M. Functional connectivity, behavioral and dopaminergic alterations 24 hours following acute exposure to synthetic bath salt drug methylenedioxypyrovalerone. Neuropharmacology 2018; 137:178-193. [PMID: 29729891 DOI: 10.1016/j.neuropharm.2018.04.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/26/2018] [Accepted: 04/29/2018] [Indexed: 12/22/2022]
Abstract
Among cathinone drugs known as bath salts, methylenedioxypyrovalerone (MDPV) exerts its potent actions via the dopamine (DA) system, and at intoxicating doses may produce adverse behavioral effects. Previous work by our group suggests that prolonged alterations in correlated neural activity between cortical and striatal areas could underlie, at least in part, the adverse reactions to this bath salt drug. In the present study, we assessed the effect of acute MDPV administration on brain functional connectivity at 1 and 24 h in rats. Using graph theory metrics to assess in vivo brain functional network organization we observed that 24 h after MDPV administration there was an increased clustering coefficient, rich club index, and average path length. Increases in these metrics suggests that MDPV produces a prolonged pattern of correlated activity characterized by greater interactions between subsets of high degree nodes but a reduced interaction with regions outside this core subset. Further analysis revealed that the core set of nodes include prefrontal cortical, amygdala, hypothalamic, somatosensory and striatal areas. At the molecular level, MDPV downregulated the dopamine transporter (DAT) in striatum and produced a shift in its subcellular distribution, an effect likely to involve rapid internalization at the membrane. These new findings suggest that potent binding of MDPV to DAT may trigger internalization and a prolonged alteration in homeostatic regulation of DA and functional brain network reorganization. We propose that the observed MDPV-induced network reorganization and DAergic changes may contribute to previously reported adverse behavioral responses to MDPV.
Collapse
Affiliation(s)
- Luis M Colon-Perez
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Addiction Research and Education (CARE), College of Medicine, University of Florida, Gainesville, FL 32610, USA; Advanced Magnetic Resonance Imaging and Spectroscopy (AMRIS) Facility, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jose A Pino
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kaustuv Saha
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Marjory Pompilus
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Sherman Kaplitz
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Nafisa Choudhury
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Darin A Jagnarine
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jean R Geste
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brandon A Levin
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Isaac Wilks
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Barry Setlow
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; The McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Addiction Research and Education (CARE), College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Adriaan W Bruijnzeel
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; The McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Addiction Research and Education (CARE), College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; The McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Addiction Research and Education (CARE), College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Gonzalo E Torres
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Addiction Research and Education (CARE), College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Marcelo Febo
- Department of Psychiatry, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; The McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Addiction Research and Education (CARE), College of Medicine, University of Florida, Gainesville, FL 32610, USA; Advanced Magnetic Resonance Imaging and Spectroscopy (AMRIS) Facility, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
36
|
Rattray NJW, Deziel NC, Wallach JD, Khan SA, Vasiliou V, Ioannidis JPA, Johnson CH. Beyond genomics: understanding exposotypes through metabolomics. Hum Genomics 2018; 12:4. [PMID: 29373992 PMCID: PMC5787293 DOI: 10.1186/s40246-018-0134-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/11/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Over the past 20 years, advances in genomic technology have enabled unparalleled access to the information contained within the human genome. However, the multiple genetic variants associated with various diseases typically account for only a small fraction of the disease risk. This may be due to the multifactorial nature of disease mechanisms, the strong impact of the environment, and the complexity of gene-environment interactions. Metabolomics is the quantification of small molecules produced by metabolic processes within a biological sample. Metabolomics datasets contain a wealth of information that reflect the disease state and are consequent to both genetic variation and environment. Thus, metabolomics is being widely adopted for epidemiologic research to identify disease risk traits. In this review, we discuss the evolution and challenges of metabolomics in epidemiologic research, particularly for assessing environmental exposures and providing insights into gene-environment interactions, and mechanism of biological impact. MAIN TEXT Metabolomics can be used to measure the complex global modulating effect that an exposure event has on an individual phenotype. Combining information derived from all levels of protein synthesis and subsequent enzymatic action on metabolite production can reveal the individual exposotype. We discuss some of the methodological and statistical challenges in dealing with this type of high-dimensional data, such as the impact of study design, analytical biases, and biological variance. We show examples of disease risk inference from metabolic traits using metabolome-wide association studies. We also evaluate how these studies may drive precision medicine approaches, and pharmacogenomics, which have up to now been inefficient. Finally, we discuss how to promote transparency and open science to improve reproducibility and credibility in metabolomics. CONCLUSIONS Comparison of exposotypes at the human population level may help understanding how environmental exposures affect biology at the systems level to determine cause, effect, and susceptibilities. Juxtaposition and integration of genomics and metabolomics information may offer additional insights. Clinical utility of this information for single individuals and populations has yet to be routinely demonstrated, but hopefully, recent advances to improve the robustness of large-scale metabolomics will facilitate clinical translation.
Collapse
Affiliation(s)
- Nicholas J. W. Rattray
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT USA
| | - Nicole C. Deziel
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT USA
| | - Joshua D. Wallach
- Collaboration for Research Integrity and Transparency (CRIT), Yale Law School, New Haven, CT USA
- Center for Outcomes Research and Evaluation (CORE), Yale-New Haven Health System, New Haven, CT USA
| | - Sajid A. Khan
- Department of Surgery, Section of Surgical Oncology, Yale University School of Medicine, New Haven, CT USA
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT USA
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT USA
| | - John P. A. Ioannidis
- Stanford Prevention Research Center, Department of Medicine, Stanford University, Stanford, CA USA
- Department of Health Research and Policy, Stanford University, Stanford, CA USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA USA
- Department of Statistics, Stanford University, Stanford, CA USA
- Meta-Research Innovation Center at Stanford, Stanford University, Stanford, CA USA
| | - Caroline H. Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT USA
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
37
|
Sambo DO, Lin M, Owens A, Lebowitz JJ, Richardson B, Jagnarine DA, Shetty M, Rodriquez M, Alonge T, Ali M, Katz J, Yan L, Febo M, Henry LK, Bruijnzeel AW, Daws L, Khoshbouei H. The sigma-1 receptor modulates methamphetamine dysregulation of dopamine neurotransmission. Nat Commun 2017; 8:2228. [PMID: 29263318 PMCID: PMC5738444 DOI: 10.1038/s41467-017-02087-x] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 11/05/2017] [Indexed: 12/16/2022] Open
Abstract
Dopamine neurotransmission is highly dysregulated by the psychostimulant methamphetamine, a substrate for the dopamine transporter (DAT). Through interactions with DAT, methamphetamine increases extracellular dopamine levels in the brain, leading to its rewarding and addictive properties. Methamphetamine also interacts with the sigma-1 receptor (σ1R), an inter-organelle signaling modulator. Using complementary strategies, we identified a novel mechanism for σ1R regulation of dopamine neurotransmission in response to methamphetamine. We found that σ1R activation prevents methamphetamine-induced, DAT-mediated increases in firing activity of dopamine neurons. In vitro and in vivo amperometric measurements revealed that σ1R activation decreases methamphetamine-stimulated dopamine efflux without affecting basal dopamine neurotransmission. Consistent with these findings, σ1R activation decreases methamphetamine-induced locomotion, motivated behavior, and enhancement of brain reward function. Notably, we revealed that the σ1R interacts with DAT at or near the plasma membrane and decreases methamphetamine-induced Ca2+ signaling, providing potential mechanisms. Broadly, these data provide evidence for σ1R regulation of dopamine neurotransmission and support the σ1R as a putative target for the treatment of methamphetamine addiction.
Collapse
Affiliation(s)
- Danielle O Sambo
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Min Lin
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Anthony Owens
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Joseph J Lebowitz
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Ben Richardson
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Darin A Jagnarine
- Department of Psychiatry, University of Florida, Gainesville, FL, 32611, USA
| | - Madhur Shetty
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Meghan Rodriquez
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Taiwo Alonge
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Mishaal Ali
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Jonathan Katz
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Long Yan
- Max Plank Institute for Neuroscience Jupiter, Jupiter, FL, 33458, USA
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL, 32611, USA
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | | | - Lynette Daws
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
38
|
Bruijnzeel AW. Reducing the Prevalence of Smoking: Policy Measures and Focusing on Specific Populations. Nicotine Tob Res 2017; 19:1003-1004. [PMID: 29054127 PMCID: PMC5896478 DOI: 10.1093/ntr/ntx154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 06/30/2017] [Indexed: 03/15/2024]
Affiliation(s)
- Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL
- Department of Neuroscience, University of Florida, Gainesville, FL
| |
Collapse
|
39
|
Mason BJ. Emerging pharmacotherapies for alcohol use disorder. Neuropharmacology 2017; 122:244-253. [PMID: 28454983 PMCID: PMC5643030 DOI: 10.1016/j.neuropharm.2017.04.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 01/29/2023]
Abstract
The identification of different stages within the alcohol use disorder (AUD) cycle that are linked to neurocircuitry changes in pathophysiology associated with the negative emotional states of abstinence has provided a view of medication development for AUD that emphasizes changes in the brain reward and stress systems. Alcohol use disorder can be defined as a chronic relapsing disorder that involves compulsive alcohol seeking and taking, loss of control over alcohol intake, and emergence of a negative emotional state during abstinence. The focus of early medications development was to block the motivation to seek alcohol in the binge/intoxication stage. More recent work has focused on reversing the motivational dysregulations associated with the withdrawal/negative affect and preoccupation/anticipation stages during protracted abstinence. Advances in our understanding of the neurocircuitry and neuropharmacological mechanisms that are involved in the development and maintenance of the withdrawal/negative affect stage using validated animal models have provided viable targets for future medications. Another major advance has been proof-of-concept testing of potential therapeutics and clinical validation of relevant pharmacological targets using human laboratory models of protracted abstinence. This review focuses on future targets for medication development associated with reversal of the loss of reward function and gain in brain stress function that drive negative reinforcement in the withdrawal/negative affect stage of addiction. Basic research has identified novel neurobiological targets associated with the withdrawal/negative affect stage and preoccupation/anticipation stage, with a focus on neuroadaptive changes within the extended amygdala that account for the transition to dependence and vulnerability to relapse. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Barbara J Mason
- The Pearson Center on Alcoholism and Addiction Research, Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, TPC-5 La Jolla, CA 92037 USA.
| |
Collapse
|
40
|
Sotiriou I, Chalkiadaki K, Nikolaidis C, Sidiropoulou K, Chatzaki E. Pharmacotherapy in smoking cessation: Corticotropin Releasing Factor receptors as emerging intervention targets. Neuropeptides 2017; 63:49-57. [PMID: 28222901 DOI: 10.1016/j.npep.2017.02.082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 02/13/2017] [Accepted: 02/13/2017] [Indexed: 11/29/2022]
Abstract
Smoking represents perhaps the single most important health risk factor and a global contributor to mortality that can unquestionably be prevented. Smoking is responsible for many diseases, including various types of cancer, chronic obstructive pulmonary disease, coronary heart disease, peripheral vascular disease and peptic ulcer, while it adversely affects fetal formation and development. Since smoking habit duration is a critical factor for mortality, the goal of treatment should be its timely cessation and relapse prevention. Drug intervention therapy is an important ally in smoking cessation. Significant positive steps have been achieved in the last few years in the development of supportive compounds. In the present review, we analyze reports studying the role of Corticotropin Releasing Factor (CRF), the principle neuroendocrine mediator of the stress response and its two receptors (CRF1 and CRF2) in the withdrawal phase as well as in the abstinence from nicotine use. Although still in pre-clinical evaluation, therapeutic implications of these data were investigated in order to highlight potential pharmaceutical interventions.
Collapse
Affiliation(s)
- Ioannis Sotiriou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | | | - Christos Nikolaidis
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | | | - Ekaterini Chatzaki
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece.
| |
Collapse
|
41
|
Bruijnzeel AW. Reward Processing and Smoking. Nicotine Tob Res 2017; 19:661-662. [PMID: 28486714 DOI: 10.1093/ntr/ntw303] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 11/07/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL.,Department of Neuroscience, University of Florida, Gainesville, FL
| |
Collapse
|
42
|
Koob GF. Antireward, compulsivity, and addiction: seminal contributions of Dr. Athina Markou to motivational dysregulation in addiction. Psychopharmacology (Berl) 2017; 234:1315-1332. [PMID: 28050629 DOI: 10.1007/s00213-016-4484-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/08/2016] [Indexed: 02/02/2023]
Abstract
RATIONALE AND OBJECTIVES Addiction is defined as a chronically relapsing disorder characterized by compulsive drug seeking that is hypothesized to derive from multiple sources of motivational dysregulation. METHODS AND RESULTS Dr. Athina Markou made seminal contributions to our understanding of the neurobiology of addiction with her studies on the dysregulation of reward function using animal models with construct validity. Repeated overstimulation of the reward systems with drugs of abuse decreases reward function, characterized by brain stimulation reward and presumbably reflecting dysphoria-like states. The construct of negative reinforcement, defined as drug taking that alleviates a negative emotional state that is created by drug abstinence, is particularly relevant as a driving force in both the withdrawal/negative affect and preoccupation/anticipation stages of the addiction cycle. CONCLUSIONS The negative emotional state that drives such negative reinforcement is hypothesized to derive from the dysregulation of key neurochemical circuits that drive incentive-salience/reward systems (dopamine, opioid peptides) in the ventral striatum and from the recruitment of brain stress systems (corticotropin-releasing factor, dynorphin) within the extended amygdala. As drug taking becomes compulsive-like, the factors that motivate behavior are hypothesized to shift to drug-seeking behavior that is driven not only by positive reinforcement but also by negative reinforcement. This shift in motivation is hypothesized to reflect the allostatic misregulation of hedonic tone such that drug taking makes the hedonic negative emotional state worse during the process of seeking temporary relief with compulsive drug taking.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism, 5635 Fishers Lane, Room 2001, Suite 2000, Rockville, MD, 20852, USA.
| |
Collapse
|
43
|
Bruijnzeel AW. Neuropeptide systems and new treatments for nicotine addiction. Psychopharmacology (Berl) 2017; 234:1419-1437. [PMID: 28028605 PMCID: PMC5420481 DOI: 10.1007/s00213-016-4513-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/12/2016] [Indexed: 01/08/2023]
Abstract
RATIONALE The mildly euphoric and cognitive enhancing effects of nicotine play a role in the initiation of smoking, while dysphoria and anxiety associated with smoking cessation contribute to relapse. After the acute withdrawal phase, smoking cues, a few cigarettes (i.e., lapse), and stressors can cause relapse. Human and animal studies have shown that neuropeptides play a critical role in nicotine addiction. OBJECTIVES The goal of this paper is to describe the role of neuropeptide systems in the initiation of nicotine intake, nicotine withdrawal, and the reinstatement of extinguished nicotine seeking. RESULTS The reviewed studies indicate that several drugs that target neuropeptide systems diminish the rewarding effects of nicotine by preventing the activation of dopaminergic systems. Other peptide-based drugs diminish the hyperactivity of brain stress systems and diminish withdrawal-associated symptom severity. Blockade of hypocretin-1 and nociceptin receptors and stimulation of galanin and neurotensin receptors diminishes the rewarding effects of nicotine. Both corticotropin-releasing factor type 1 and kappa-opioid receptor antagonists diminish dysphoria and anxiety-like behavior associated with nicotine withdrawal and inhibit stress-induced reinstatement of nicotine seeking. Furthermore, blockade of vasopressin 1b receptors diminishes dysphoria during nicotine withdrawal, and melanocortin 4 receptor blockade prevents stress-induced reinstatement of nicotine seeking. The role of neuropeptide systems in nicotine-primed and cue-induced reinstatement is largely unexplored, but there is evidence for a role of hypocretin-1 receptors in cue-induced reinstatement of nicotine seeking. CONCLUSION Drugs that target neuropeptide systems might decrease the euphoric effects of smoking and improve relapse rates by diminishing withdrawal symptoms and improving stress resilience.
Collapse
Affiliation(s)
- Adriaan W. Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, Florida, USA,Department of Neuroscience, University of Florida, Gainesville, Florida, USA,Center for Addiction Research and Education, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
44
|
Qi X, Guzhva L, Yang Z, Febo M, Shan Z, Wang KKW, Bruijnzeel AW. Overexpression of CRF in the BNST diminishes dysphoria but not anxiety-like behavior in nicotine withdrawing rats. Eur Neuropsychopharmacol 2016; 26:1378-1389. [PMID: 27461514 PMCID: PMC5067082 DOI: 10.1016/j.euroneuro.2016.07.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 06/06/2016] [Accepted: 07/02/2016] [Indexed: 01/08/2023]
Abstract
Smoking cessation leads to dysphoria and anxiety, which both increase the risk for relapse. This negative affective state is partly mediated by an increase in activity in brain stress systems. Recent studies indicate that prolonged viral vector-mediated overexpression of stress peptides diminishes stress sensitivity. Here we investigated whether the overexpression of corticotropin-releasing factor (CRF) in the bed nucleus of the stria terminalis (BNST) diminishes nicotine withdrawal symptoms in rats. The effect of nicotine withdrawal on brain reward function was investigated with an intracranial self-stimulation (ICSS) procedure. Anxiety-like behavior was investigated in the elevated plus maze test and a large open field. An adeno-associated virus (AAV) pseudotype 2/5 vector was used to overexpress CRF in the lateral BNST and nicotine dependence was induced using minipumps. Administration of the nicotinic receptor antagonist mecamylamine and cessation of nicotine administration led to a dysphoria-like state, which was prevented by the overexpression of CRF in the BNST. Nicotine withdrawal also increased anxiety-like behavior in the elevated plus maze test and large open field test and slightly decreased locomotor activity in the open field. The overexpression of CRF in the BNST did not prevent the increase in anxiety-like behavior or decrease in locomotor activity. The overexpression of CRF increased CRF1 and CRF2 receptor gene expression and increased the CRF2/CRF1 receptor ratio. In conclusion, the overexpression of CRF in the BNST prevents the dysphoria-like state associated with nicotine withdrawal and increases the CRF2/CRF1 receptor ratio, which may diminish the negative effects of CRF on mood.
Collapse
Affiliation(s)
- Xiaoli Qi
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA
| | - Lidia Guzhva
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA
| | - Zhihui Yang
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Zhiying Shan
- Department of Physiology and Functional Genomics, Gainesville, FL 32611, USA
| | - Kevin K W Wang
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL 32611, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
45
|
Varenicline impairs extinction and enhances reinstatement across repeated cycles of nicotine self-administration in rats. Neuropharmacology 2016; 105:463-470. [DOI: 10.1016/j.neuropharm.2016.02.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/11/2016] [Accepted: 02/15/2016] [Indexed: 01/10/2023]
|
46
|
Neuronal Nicotinic Acetylcholine Receptor Modulators Reduce Sugar Intake. PLoS One 2016; 11:e0150270. [PMID: 27028298 PMCID: PMC4814119 DOI: 10.1371/journal.pone.0150270] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/11/2016] [Indexed: 12/21/2022] Open
Abstract
Excess sugar consumption has been shown to contribute directly to weight gain, thus contributing to the growing worldwide obesity epidemic. Interestingly, increased sugar consumption has been shown to repeatedly elevate dopamine levels in the nucleus accumbens (NAc), in the mesolimbic reward pathway of the brain similar to many drugs of abuse. We report that varenicline, an FDA-approved nicotinic acetylcholine receptor (nAChR) partial agonist that modulates dopamine in the mesolimbic reward pathway of the brain, significantly reduces sucrose consumption, especially in a long-term consumption paradigm. Similar results were observed with other nAChR drugs, namely mecamylamine and cytisine. Furthermore, we show that long-term sucrose consumption increases α4β2 * and decreases α6β2* nAChRs in the nucleus accumbens, a key brain region associated with reward. Taken together, our results suggest that nAChR drugs such as varenicline may represent a novel treatment strategy for reducing sugar consumption.
Collapse
|
47
|
A Two-Day Continuous Nicotine Infusion Is Sufficient to Demonstrate Nicotine Withdrawal in Rats as Measured Using Intracranial Self-Stimulation. PLoS One 2015; 10:e0144553. [PMID: 26658557 PMCID: PMC4684239 DOI: 10.1371/journal.pone.0144553] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/19/2015] [Indexed: 01/23/2023] Open
Abstract
Avoidance of the negative affective (emotional) symptoms of nicotine withdrawal (e.g., anhedonia, anxiety) contributes to tobacco addiction. Establishing the minimal nicotine exposure conditions required to demonstrate negative affective withdrawal signs in animals, as well as understanding moderators of these conditions, could inform tobacco addiction-related research, treatment, and policy. The goal of this study was to determine the minimal duration of continuous nicotine infusion required to demonstrate nicotine withdrawal in rats as measured by elevations in intracranial self-stimulation (ICSS) thresholds (anhedonia-like behavior). Administration of the nicotinic acetylcholine receptor antagonist mecamylamine (3.0 mg/kg, s.c.) on alternate test days throughout the course of a 2-week continuous nicotine infusion (3.2 mg/kg/day via osmotic minipump) elicited elevations in ICSS thresholds beginning on the second day of infusion. Magnitude of antagonist-precipitated withdrawal did not change with further nicotine exposure and mecamylamine injections, and was similar to that observed in a positive control group receiving mecamylamine following a 14-day nicotine infusion. Expression of a significant withdrawal effect was delayed in nicotine-infused rats receiving mecamylamine on all test days rather than on alternate test days. In a separate study, rats exhibited a transient increase in ICSS thresholds following cessation of a 2-day continuous nicotine infusion (3.2 mg/kg/day). Magnitude of this spontaneous withdrawal effect was similar to that observed in rats receiving a 9-day nicotine infusion. Our findings demonstrate that rats exhibit antagonist-precipitated and spontaneous nicotine withdrawal following a 2-day continuous nicotine infusion, at least under the experimental conditions studied here. Magnitude of these effects were similar to those observed in traditional models involving more prolonged nicotine exposure. Further development of these models, including evaluation of more clinically relevant nicotine dosing regimens and other measures of nicotine withdrawal (e.g., anxiety-like behavior, somatic signs), may be useful for understanding the development of the nicotine withdrawal syndrome.
Collapse
|
48
|
Mohamed TS, Jayakar SS, Hamouda AK. Orthosteric and Allosteric Ligands of Nicotinic Acetylcholine Receptors for Smoking Cessation. Front Mol Neurosci 2015; 8:71. [PMID: 26635524 PMCID: PMC4658446 DOI: 10.3389/fnmol.2015.00071] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/06/2015] [Indexed: 12/23/2022] Open
Abstract
Nicotine addiction, the result of tobacco use, leads to over six million premature deaths world-wide per year, a number that is expected to increase by a third within the next two decades. While more than half of smokers want and attempt to quit, only a small percentage of smokers are able to quit without pharmacological interventions. Therefore, over the past decades, researchers in academia and the pharmaceutical industry have focused their attention on the development of more effective smoking cessation therapies, which is now a growing 1.9 billion dollar market. Because the role of neuronal nicotinic acetylcholine receptors (nAChR) in nicotine addiction is well established, nAChR based therapeutics remain the leading strategy for smoking cessation. However, the development of neuronal nAChR drugs that are selective for a nAChR subpopulation is challenging, and only few neuronal nAChR drugs are clinically available. Among the many neuronal nAChR subtypes that have been identified in the brain, the α4β2 subtype is the most abundant and plays a critical role in nicotine addiction. Here, we review the role of neuronal nAChRs, especially the α4β2 subtype, in the development and treatment of nicotine addiction. We also compare available smoking cessation medications and other nAChR orthosteric and allosteric ligands that have been developed with emphasis on the difficulties faced in the development of clinically useful compounds with high nAChR subtype selectivity.
Collapse
Affiliation(s)
- Tasnim S Mohamed
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M Health Sciences Center Kingsville, TX, USA
| | - Selwyn S Jayakar
- Department of Neurobiology, Harvard Medical School Boston, MA, USA
| | - Ayman K Hamouda
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M Health Sciences Center Kingsville, TX, USA ; Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Sciences Center Bryan, TX, USA
| |
Collapse
|
49
|
Koob GF, Mason BJ. Existing and Future Drugs for the Treatment of the Dark Side of Addiction. Annu Rev Pharmacol Toxicol 2015; 56:299-322. [PMID: 26514207 DOI: 10.1146/annurev-pharmtox-010715-103143] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The identification of a heuristic framework for the stages of the addiction cycle that are linked to neurocircuitry changes in pathophysiology includes the binge/intoxication stage, the withdrawal/negative affect stage, and the preoccupation/anticipation (craving) stage, which represent neuroadaptations in three neurocircuits (basal ganglia, extended amygdala, and frontal cortex, respectively). The identification of excellent and validated animal models, the development of human laboratory models, and an enormous surge in our understanding of neurocircuitry and neuropharmacological mechanisms have provided a revisionist view of addiction that emphasizes the loss of brain reward function and gain of stress function that drive negative reinforcement (the dark side of addiction) as a key to compulsive drug seeking. Reversing the dark side of addiction not only explains much of the existing successful pharmacotherapies for addiction but also points to vast new opportunities for future medications to alleviate this major source of human suffering.
Collapse
Affiliation(s)
- George F Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037; ,
| | - Barbara J Mason
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California 92037; ,
| |
Collapse
|
50
|
Wiebelhaus JM, Walentiny DM, Beardsley PM. Effects of Acute and Repeated Administration of Oxycodone and Naloxone-Precipitated Withdrawal on Intracranial Self-Stimulation in Rats. J Pharmacol Exp Ther 2015; 356:43-52. [PMID: 26491062 DOI: 10.1124/jpet.115.228940] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/20/2015] [Indexed: 01/24/2023] Open
Abstract
Incidence of prescription opioid abuse and overdose, often led by oxycodone, continues to increase, producing twice as many overdose deaths as heroin. Surprisingly, preclinical reports relevant to oxycodone's abuse-related effects are relatively sparse considering its history and patient usage. The goal of this study was to characterize dose- and time-dependent effects of acute and repeated oxycodone administration in a frequency-rate intracranial self-stimulation (ICSS) procedure, an assay often predictive of drug-related reinforcing effects, in male Sprague-Dawley rats. We hypothesized that oxycodone would produce a biphasic profile of rate-increasing and rate-decreasing effects maintained by ICSS similar to μ-opioid receptor agonists. Oxycodone (0.03, 0.3, 1, and 3 mg/kg, s.c.) produced dose- and time-dependent alterations on ICSS, with the predicted biphasic profile of rate-increasing effects at lower stimulation frequencies followed by rate-decreasing effects at higher frequencies. Peak effects were observed between 30 and 60 minutes, which were reversed by naloxone pretreatment (30 minutes). Tolerance to rate-decreasing effects was observed over a 5-day period when rats were treated with 1 mg/kg oxycodone twice a day. Subsequently, the dosing regimen was increased to 3 mg/kg twice a day over 10 days, although further marked tolerance did not develop. When then challenged with 10 mg/kg naloxone, a significant suppression below baseline levels of ICSS-maintained responding occurred indicative of dependence that recovered to baseline within 5 hours. The results of this study provide the first report of acute and chronic effects of oxycodone on responding maintained by ICSS presentation and the use of ICSS-maintained responding to characterize its tolerance and dependence effects.
Collapse
Affiliation(s)
- Jason M Wiebelhaus
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - D Matthew Walentiny
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Patrick M Beardsley
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|