1
|
Chen Z, Lu N, Li X, Liu Q, Li Y, Li X, Yu X, Zhao H, Liu C, Tang X, Wang X, Huang W. The Effect of a Caffeine and Nicotine Combination on Nicotine Withdrawal Syndrome in Mice. Nutrients 2024; 16:3048. [PMID: 39339647 PMCID: PMC11435009 DOI: 10.3390/nu16183048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Nicotine dependence is an important cause of excessive exposure to tobacco combustion compounds in most smokers. Nicotine replacement therapy is the main method to treat nicotine dependence, but it still has its shortcomings, such as the inability to mitigate withdrawal effects and limited applicability. It has been hypothesized that a combination of low-dose nicotine and caffeine could achieve the same psychological stimulation effect as a high dose of nicotine without causing nicotine withdrawal effects. To establish a model of nicotine dependence, male C57BL/6J mice were subcutaneously injected four times a day with nicotine (2 mg/kg) for 15 days and fed with water containing nicotine at the same time. They were randomly divided into four groups. After 24 h of withdrawal, different groups were injected with saline, nicotine (0.25 mg/kg or 0.1 mg/kg), or nicotine (0.1 mg/kg) and caffeine (20 mg/kg). Behavioral and physiological changes were evaluated by an assessment of physical signs, open field tests, elevated plus maze experiments, forced swimming tests, hot plate tests, and new-object-recognition tests. The changes in dopamine release in the prefrontal cortex (PFC) and ventral tegmental area (VTA) in the midbrain were analyzed using ELISA. The results showed that a combination of caffeine and nicotine could effectively relieve nicotine withdrawal syndrome, increase movement ability and pain thresholds, reduce anxiety and depression, enhance memory and cognitive ability, and increase the level of dopamine release in the PFC and VTA. Thus, caffeine combined with nicotine has potential as a stable and effective treatment option to help humans with smoking cessation.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Naiyan Lu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Sports and Health Research Institute, Jiangnan University, Wuxi 214122, China
| | - Xu Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qingrun Liu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yujie Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Xiyue Li
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University, Wuxi 214126, China
| | - Ximiao Yu
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University, Wuxi 214126, China
| | - Haotian Zhao
- Sports and Health Research Institute, Jiangnan University, Wuxi 214122, China
| | - Chang Liu
- School of Sport Science, Beijing Sport University, Beijing 100084, China
| | - Xue Tang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xun Wang
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University, Wuxi 214126, China
| | - Weisun Huang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
| |
Collapse
|
2
|
Garcia-Rivas V, Fiancette JF, Tostain J, de Maio G, Ceau M, Wiart JF, Gaulier JM, Deroche-Gamonet V. Individual variations in motives for nicotine self-administration in male rats: evidence in support for a precision psychopharmacology. Transl Psychiatry 2024; 14:85. [PMID: 38336930 PMCID: PMC10858238 DOI: 10.1038/s41398-024-02774-6] [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: 11/30/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 02/12/2024] Open
Abstract
The significant heterogeneity in smoking behavior among smokers, coupled with the inconsistent efficacy of approved smoking cessation therapies, supports the presence of individual variations in the mechanisms underlying smoking. This emphasizes the need to shift from standardized to personalized smoking cessation therapies. However, informed precision medicine demands precision fundamental research. Tobacco smoking is influenced and sustained by diverse psychopharmacological interactions between nicotine and environmental stimuli. In the classical experimental rodent model for studying tobacco dependence, namely intravenous self-administration of nicotine, seeking behavior is reinforced by the combined delivery of nicotine and a discrete cue (nicotine+cue). Whether self-administration behavior is driven by the same psychopharmacological mechanisms across individual rats remains unknown and unexplored. To address this, we employed behavioral pharmacology and unbiased cluster analysis to investigate individual differences in the mechanisms supporting classical intravenous nicotine self-administration (0.04 mg/kg/infusion) in male outbred Sprague-Dawley rats. Our analysis identified two clusters: one subset of rats sought nicotine primarily for its reinforcing effects, while the second subset sought nicotine to enhance the reinforcing effects of the discrete cue. Varenicline (1 mg/kg i.p.) reduced seeking behavior in the former group, whereas it tended to increase in the latter group. Crucially, despite this fundamental qualitative difference revealed by behavioral manipulation, the two clusters exhibited quantitatively identical nicotine+cue self-administration behavior. The traditional application of rodent models to study the reinforcing and addictive effects of nicotine may mask individual variability in the underlying motivational mechanisms. Accounting for this variability could significantly enhance the predictive validity of translational research.
Collapse
Affiliation(s)
- Vernon Garcia-Rivas
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33000, Bordeaux, France.
- INSERM, Magendie, U1215, F-33000, Bordeaux, France.
- Yale University School of Medicine, Department of Psychiatry, New Haven, CT, USA.
| | - Jean-François Fiancette
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33000, Bordeaux, France
- INSERM, Magendie, U1215, F-33000, Bordeaux, France
| | - Jessica Tostain
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33000, Bordeaux, France
- INSERM, Magendie, U1215, F-33000, Bordeaux, France
| | - Giulia de Maio
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33000, Bordeaux, France
- INSERM, Magendie, U1215, F-33000, Bordeaux, France
| | - Matias Ceau
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33000, Bordeaux, France
- INSERM, Magendie, U1215, F-33000, Bordeaux, France
| | | | - Jean-Michel Gaulier
- CHU Lille, Unité Fonctionnelle de Toxicologie, F-59037, Lille, France
- Univ. Lille, ULR 4483, IMPECS - IMPact de l'Environnement Chimique sur la Santé humaine, F-59045, Lille, France
| | - Véronique Deroche-Gamonet
- Univ. Bordeaux, INSERM, Magendie, U1215, F-33000, Bordeaux, France.
- INSERM, Magendie, U1215, F-33000, Bordeaux, France.
| |
Collapse
|
3
|
Vargas-Medrano J, Carcoba LM, Vidal Martinez G, Mulla ZD, Diaz V, Ruiz-Velasco A, Alvarez-Primo F, Colina G, Iñiguez SD, Thompson PM, O’Dell LE, Gadad BS. Sex and diet-dependent gene alterations in human and rat brains with a history of nicotine exposure. Front Psychiatry 2023; 14:1104563. [PMID: 36846236 PMCID: PMC9950561 DOI: 10.3389/fpsyt.2023.1104563] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/19/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Chronic nicotine exposure induces changes in the expression of key regulatory genes associated with metabolic function and neuronal alterations in the brain. Many bioregulatory genes have been associated with exposure to nicotine, but the modulating effects of sex and diet on gene expression in nicotine-exposed brains have been largely unexplored. Both humans and rodents display motivation for nicotine use and the emergence of withdrawal symptoms during abstinence. Research comparing pre-clinical models with human subjects provides an important opportunity to understand common biomarkers of the harmful effects of nicotine as well as information that may help guide the development of more effective interventions for nicotine cessation. Methods Human postmortem dorsolateral prefrontal cortex (dLPFC) tissue BA9 was collected from female and male subjects, smokers and non-smokers (N = 12 per group). Rat frontal lobes were collected from female and male rats that received a regular diet (RD) or a high-fat diet (HFD) (N = 12 per group) for 14 days following implantation of a osmotic mini-pump (Alzet) that delivered nicotine continuously. Controls (control-s) received a sham surgical procedure. RNA was extracted from tissue from human and rat samples and reversed-transcribed to cDNA. Gene expression of CHRNA10 (Cholinergic receptor nicotinic alpha 10), CERKL (Ceramide Kinase-Like), SMYD1 (SET and MYD Domin Containing 1), and FA2H (Fatty Acid 2-Hydrolase) in humans was compared to rats in each subset of groups and quantified by qPCR methods. Additionally, protein expression of FA2H was analyzed by immunohistochemistry (IHC) in human dLPFC. Results Humans with a history of smoking displayed decreased CHRNA10 (p = 0.0005), CERKL (p ≤ 0.0001), and SMYD1 (p = 0.0005) expression and increased FA2H (p = 0.0097) expression compared to non-smokers (p < 0.05). Similar patterns of results were observed in nicotine exposed vs. control rats. Interestingly, sex-related differences in gene expression for CERKL and FA2H were observed. In addition, ANCOVA analysis showed a significant effect of nicotine in a sex-different manner, including an increase in CERKL in male and female rats with RD or HFD. In rats exposed to an HFD, FA2H gene expression was lower in nicotine-treated rats compared to RD rats treated with nicotine. Protein expression of FA2H (p = 0.001) by IHC was significantly higher in smokers compared to non-smokers. Conclusion These results suggest that a history of long-term nicotine exposure in humans alters the expression of sphingolipid metabolism-related (CERKL, SMYD1, and FA2H) and neuronal (CHRNA10) marker genes similarly as compared to rats. Sex- and diet-dependent differences appear in nicotine-exposed rats, critical in regulating sphingolipid metabolism and nicotinic acetylcholine receptors. This research enhances the construct validity of rat models of nicotine usage by showing a similar pattern of changes in gene expression in human subjects with a smoking history.
Collapse
Affiliation(s)
- Javier Vargas-Medrano
- Department of Psychiatry, Texas Tech University Health Sciences Center, El Paso, TX, United States
- Southwest Brain Bank, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Luis M. Carcoba
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Guadalupe Vidal Martinez
- Department of Psychiatry, Texas Tech University Health Sciences Center, El Paso, TX, United States
- Southwest Brain Bank, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Zuber D. Mulla
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Victoria Diaz
- Department of Psychiatry, Texas Tech University Health Sciences Center, El Paso, TX, United States
- Southwest Brain Bank, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Alejandra Ruiz-Velasco
- Department of Psychiatry, Texas Tech University Health Sciences Center, El Paso, TX, United States
- Southwest Brain Bank, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Fabian Alvarez-Primo
- Department of Psychiatry, Texas Tech University Health Sciences Center, El Paso, TX, United States
- Southwest Brain Bank, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Gabriela Colina
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Sergio D. Iñiguez
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States
| | - Peter M. Thompson
- Department of Psychiatry, Texas Tech University Health Sciences Center, El Paso, TX, United States
- Southwest Brain Bank, Texas Tech University Health Sciences Center, El Paso, TX, United States
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Laura E. O’Dell
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States
| | - Bharathi S. Gadad
- Department of Psychiatry, Texas Tech University Health Sciences Center, El Paso, TX, United States
- Southwest Brain Bank, Texas Tech University Health Sciences Center, El Paso, TX, United States
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| |
Collapse
|
4
|
Hamdan JN, Sierra-Fonseca JA, Flores RJ, Saucedo S, Miranda-Arango M, O’Dell LE, Gosselink KL. Early-life adversity increases anxiety-like behavior and modifies synaptic protein expression in a region-specific manner. Front Behav Neurosci 2022; 16:1008556. [PMID: 36338879 PMCID: PMC9626971 DOI: 10.3389/fnbeh.2022.1008556] [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: 07/31/2022] [Accepted: 09/29/2022] [Indexed: 02/11/2024] Open
Abstract
Early-life adversity (ELA) can induce persistent neurological changes and increase the risk for developing affective or substance use disorders. Disruptions to the reward circuitry of the brain and pathways serving motivation and emotion have been implicated in the link between ELA and altered adult behavior. The molecular mechanisms that mediate the long-term effects of ELA, however, are not fully understood. We examined whether ELA in the form of neonatal maternal separation (MatSep) modifies behavior and synaptic protein expression in adults. We hypothesized that MatSep would affect dopaminergic and glutamatergic signaling and enhance sensitivity to methamphetamine (Meth) reward or increase anxiety. Male Wistar rats were subjected to MatSep for 180 min/d on postnatal days (PND) 2-14 and allowed to grow to adulthood (PND 60) with no further manipulation. The hippocampus (Hipp), medial prefrontal cortex (mPFC), nucleus accumbens (NAc), and caudate putamen (CPu) were isolated from one subgroup of animals and subjected to Western blot and protein quantitation for tyrosine hydroxylase (TH), α-synuclein (ALPHA), NMDA receptor (NMDAR), dopamine receptor-1 (D1) and -2 (D2), dopamine transporter (DAT), and postsynaptic density 95 (PSD95). Separate group of animals were tested for anxiety-like behavior and conditioned place preference (CPP) to Meth at 0.0, 0.1, and 1.0 mg/kg doses. MatSep rats displayed an increase in basal levels of anxiety-like behavior compared to control animals. MatSep rats also demonstrated CPP to Meth, but their responses did not differ significantly from controls at any drug dose. Increased NMDAR, D2, and ALPHA expression was observed in the NAc and CPu following MatSep; D2 and ALPHA levels were also elevated in the mPFC, along with DAT. MatSep rats had reduced D1 expression in the mPFC and Hipp, with the Hipp also showing a reduction in D2. Only the CPu showed elevated TH and decreased DAT expression levels. No significant changes were found in PSD95 expression in MatSep rats. In conclusion, ELA is associated with long-lasting and region-specific changes in synaptic protein expression that diminish dopamine neurotransmission and increase anxiety-like behavior in adults. These findings illustrate potential mechanisms through which ELA may increase vulnerability to stress-related illness.
Collapse
Affiliation(s)
- Jameel N. Hamdan
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
- Antharis Therapeutics, San Diego, CA, United States
| | - Jorge A. Sierra-Fonseca
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
- Department of Science, Chatham University, Pittsburgh, PA, United States
| | - Rodolfo J. Flores
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States
- National Institutes of Health, National Institute of General Medical Sciences, Bethesda, MD, United States
| | - Sigifredo Saucedo
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
| | - Manuel Miranda-Arango
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
| | - Laura E. O’Dell
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States
| | - Kristin L. Gosselink
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, United States
- Department of Physiology and Pathology, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| |
Collapse
|
5
|
Goldberg LR, Gould TJ. Genetic influences impacting nicotine use and abuse during adolescence: Insights from human and rodent studies. Brain Res Bull 2022; 187:24-38. [PMID: 35738503 DOI: 10.1016/j.brainresbull.2022.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/18/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
Nicotine use continues to be a major public health concern, with an alarming recent rise in electronic cigarette consumption. Heritability estimates of nicotine use and abuse range from 40% to 80%, providing strong evidence that genetic factors impact nicotine addiction-relevant phenotypes. Although nicotine use during adolescence is a key factor in the development of addiction, it remains unclear how genetic factors impact adolescent nicotine use and abuse. This review will discuss studies investigating genetic factors impacting nicotine use during adolescence. Evidence from both rodent and human studies will be summarized and integrated when possible. Human adolescent studies have largely included candidate gene studies for genes identified in adult populations, such as genes involved in nicotine metabolism, nicotinic acetylcholine receptor signaling, dopaminergic signaling, and other neurotransmitter signaling systems. Alternatively, rodent studies have largely taken a discovery-based approach identifying strain differences in adolescent nicotine addiction-relevant behaviors. Here, we aim to answer the following three questions by integrating human and rodent findings: (1) Are there genetic variants that uniquely impact nicotine use during adolescence? (2) Are there genetic variants that impact both adolescent and adult nicotine use? and (3) Do genetic factors in adolescence significantly impact long-term consequences of adolescent nicotine use? Determining answers for these three questions will be critical for the development of preventative measures and treatments for adolescent nicotine use and addiction.
Collapse
Affiliation(s)
- Lisa R Goldberg
- Department of Biobehavioral Heatlh, Pennsylvania State University, University Park, PA, USA
| | - Thomas J Gould
- Department of Biobehavioral Heatlh, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
6
|
Maurer JJ, Wimmer ME, Turner CA, Herman RJ, Zhang Y, Ragnini K, Ferrante J, Kimmey BA, Crist RC, Christopher Pierce R, Schmidt HD. Paternal nicotine taking elicits heritable sex-specific phenotypes that are mediated by hippocampal Satb2. Mol Psychiatry 2022; 27:3864-3874. [PMID: 35595980 PMCID: PMC9675874 DOI: 10.1038/s41380-022-01622-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/04/2022] [Accepted: 05/11/2022] [Indexed: 02/08/2023]
Abstract
Nicotine intake, whether through tobacco smoking or e-cigarettes, remains a global health concern. An emerging preclinical literature indicates that parental nicotine exposure produces behavioral, physiological, and molecular changes in subsequent generations. However, the heritable effects of voluntary parental nicotine taking are unknown. Here, we show increased acquisition of nicotine taking in male and female offspring of sires that self-administered nicotine. In contrast, self-administration of sucrose and cocaine were unaltered in male and female offspring suggesting that the intergenerational effects of paternal nicotine taking may be reinforcer specific. Further characterization revealed memory deficits and increased anxiety-like behaviors in drug-naive male, but not female, offspring of nicotine-experienced sires. Using an unbiased, genome-wide approach, we discovered that these phenotypes were associated with decreased expression of Satb2, a transcription factor known to play important roles in synaptic plasticity and memory formation, in the hippocampus of nicotine-sired male offspring. This effect was sex-specific as no changes in Satb2 expression were found in nicotine-sired female offspring. Finally, increasing Satb2 levels in the hippocampus prevented the escalation of nicotine intake and rescued the memory deficits associated with paternal nicotine taking in male offspring. Collectively, these findings indicate that paternal nicotine taking produces heritable sex-specific molecular changes that promote addiction-like phenotypes and memory impairments in male offspring.
Collapse
Affiliation(s)
- John J Maurer
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mathieu E Wimmer
- Department of Psychology, College of Liberal Arts, Temple University, Philadelphia, PA, 19122, USA
| | - Christopher A Turner
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rae J Herman
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yafang Zhang
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kael Ragnini
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julia Ferrante
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Blake A Kimmey
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Richard C Crist
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - R Christopher Pierce
- Brain Health Institute and Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Heath D Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
Sheardown E, Mech AM, Petrazzini MEM, Leggieri A, Gidziela A, Hosseinian S, Sealy IM, Torres-Perez JV, Busch-Nentwich EM, Malanchini M, Brennan CH. Translational relevance of forward genetic screens in animal models for the study of psychiatric disease. Neurosci Biobehav Rev 2022; 135:104559. [PMID: 35124155 PMCID: PMC9016269 DOI: 10.1016/j.neubiorev.2022.104559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/10/2021] [Accepted: 02/01/2022] [Indexed: 12/16/2022]
Abstract
Psychiatric disorders represent a significant burden in our societies. Despite the convincing evidence pointing at gene and gene-environment interaction contributions, the role of genetics in the etiology of psychiatric disease is still poorly understood. Forward genetic screens in animal models have helped elucidate causal links. Here we discuss the application of mutagenesis-based forward genetic approaches in common animal model species: two invertebrates, nematodes (Caenorhabditis elegans) and fruit flies (Drosophila sp.); and two vertebrates, zebrafish (Danio rerio) and mice (Mus musculus), in relation to psychiatric disease. We also discuss the use of large scale genomic studies in human populations. Despite the advances using data from human populations, animal models coupled with next-generation sequencing strategies are still needed. Although with its own limitations, zebrafish possess characteristics that make them especially well-suited to forward genetic studies exploring the etiology of psychiatric disorders.
Collapse
Affiliation(s)
- Eva Sheardown
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Aleksandra M Mech
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | | | - Adele Leggieri
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Agnieszka Gidziela
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Saeedeh Hosseinian
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Ian M Sealy
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jose V Torres-Perez
- UK Dementia Research Institute at Imperial College London and Department of Brain Sciences, Imperial College London, 86 Wood Lane, London W12 0BZ, UK
| | - Elisabeth M Busch-Nentwich
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Margherita Malanchini
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Caroline H Brennan
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK.
| |
Collapse
|
8
|
Carstens E, Carstens MI. Sensory Effects of Nicotine and Tobacco. Nicotine Tob Res 2022; 24:306-315. [PMID: 33955474 PMCID: PMC8842437 DOI: 10.1093/ntr/ntab086] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/28/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Ingestion of nicotine by smoking, vaping, or other means elicits various effects including reward, antinociception, and aversion due to irritation, bitter taste, and unpleasant side effects such as nausea and dizziness. AIMS AND METHODS Here we review the sensory effects of nicotine and the underlying neurobiological processes. RESULTS AND CONCLUSIONS Nicotine elicits oral irritation and pain via the activation of neuronal nicotinic acetylcholine receptors (nAChRs) expressed by trigeminal nociceptors. These nociceptors excite neurons in the trigeminal subnucleus caudalis (Vc) and other brainstem regions in a manner that is significantly reduced by the nAChR antagonist mecamylamine. Vc neurons are excited by lingual application of nicotine and exhibit a progressive decline in firing to subsequent applications, consistent with desensitization of peripheral sensory neurons and progressively declining ratings of oral irritation in human psychophysical experiments. Nicotine also elicits a nAChR-mediated bitter taste via excitation of gustatory afferents. Nicotine solutions are avoided even when sweeteners are added. Studies employing oral self-administration have yielded mixed results: Some studies show avoidance of nicotine while others report increased nicotine intake over time, particularly in adolescents and females. Nicotine is consistently reported to increase human pain threshold and tolerance levels. In animal studies, nicotine is antinociceptive when delivered by inhalation of tobacco smoke or systemic infusion, intrathecally, and by intracranial microinjection in the pedunculopontine tegmentum, ventrolateral periaqueductal gray, and rostral ventromedial medulla. The antinociception is thought to be mediated by descending inhibition of spinal nociceptive transmission. Menthol cross-desensitizes nicotine-evoked oral irritation, reducing harshness that may account for its popularity as a flavor additive to tobacco products. IMPLICATIONS Nicotine activates brain systems underlying reward and antinociception, but at the same time elicits aversive sensory effects including oral irritation and pain, bitter taste, and other unpleasant side effects mediated largely by nicotinic acetylcholine receptors (nAChRs). This review discusses the competing aversive and antinociceptive effects of nicotine and exposure to tobacco smoke, and the underlying neurobiology. An improved understanding of the interacting effects of nicotine will hopefully inform novel approaches to mitigate nicotine and tobacco use.
Collapse
Affiliation(s)
- Earl Carstens
- Department of Neurobiology, Physiology and Behavior University of California, Davis, CA, USA
| | - M Iodi Carstens
- Department of Neurobiology, Physiology and Behavior University of California, Davis, CA, USA
| |
Collapse
|
9
|
Stojanovic T, Velarde Gamez D, Schuld GJ, Bormann D, Cabatic M, Uhrin P, Lubec G, Monje FJ. Age-Dependent and Pathway-Specific Bimodal Action of Nicotine on Synaptic Plasticity in the Hippocampus of Mice Lacking the miR-132/212 Genes. Cells 2022; 11:261. [PMID: 35053378 PMCID: PMC8774101 DOI: 10.3390/cells11020261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 12/19/2022] Open
Abstract
Nicotine addiction develops predominantly during human adolescence through smoking. Self-administration experiments in rodents verify this biological preponderance to adolescence, suggesting evolutionary-conserved and age-defined mechanisms which influence the susceptibility to nicotine addiction. The hippocampus, a brain region linked to drug-related memory storage, undergoes major morpho-functional restructuring during adolescence and is strongly affected by nicotine stimulation. However, the signaling mechanisms shaping the effects of nicotine in young vs. adult brains remain unclear. MicroRNAs (miRNAs) emerged recently as modulators of brain neuroplasticity, learning and memory, and addiction. Nevertheless, the age-dependent interplay between miRNAs regulation and hippocampal nicotinergic signaling remains poorly explored. We here combined biophysical and pharmacological methods to examine the impact of miRNA-132/212 gene-deletion (miRNA-132/212-/-) and nicotine stimulation on synaptic functions in adolescent and mature adult mice at two hippocampal synaptic circuits: the medial perforant pathway (MPP) to dentate yrus (DG) synapses (MPP-DG) and CA3 Schaffer collaterals to CA1 synapses (CA3-CA1). Basal synaptic transmission and short-term (paired-pulse-induced) synaptic plasticity was unaltered in adolescent and adult miRNA-132/212-/- mice hippocampi, compared with wild-type controls. However, nicotine stimulation promoted CA3-CA1 synaptic potentiation in mature adult (not adolescent) wild-type and suppressed MPP-DG synaptic potentiation in miRNA-132/212-/- mice. Altered levels of CREB, Phospho-CREB, and acetylcholinesterase (AChE) expression were further detected in adult miRNA-132/212-/- mice hippocampi. These observations propose miRNAs as age-sensitive bimodal regulators of hippocampal nicotinergic signaling and, given the relevance of the hippocampus for drug-related memory storage, encourage further research on the influence of miRNAs 132 and 212 in nicotine addiction in the young and the adult brain.
Collapse
Affiliation(s)
- Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - David Velarde Gamez
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - Gabor Jorrid Schuld
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - Daniel Bormann
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
- Laboratory for Cardiac and Thoracic Diagnosis, Department of Surgery, Regeneration and Applied Immunology, Medical University of Vienna, Research Laboratories Vienna General Hospital, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Division of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Maureen Cabatic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, 1090 Vienna, Austria;
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, 1090 Vienna, Austria; (D.V.G.); (G.J.S.); (D.B.); (M.C.)
| |
Collapse
|
10
|
Bagdas D, Kebede N, Zepei AM, Harris L, Minanov K, Picciotto MR, Addy NA. Animal Models to Investigate the Impact of Flavors on Nicotine Addiction and Dependence. Curr Neuropharmacol 2022; 20:2175-2201. [PMID: 35611777 PMCID: PMC9886843 DOI: 10.2174/1570159x20666220524120231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 04/17/2022] [Accepted: 05/22/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Tobacco use in humans is a long-standing public health concern. Flavors are common additives in tobacco and alternative tobacco products, added to mask nicotine's harsh orosensory effects and increase the appeal of these products. Animal models are integral for investigating nicotine use and addiction and are helpful for understanding the effects of flavor additives on the use of nicotine delivery products. OBJECTIVE This review focuses on preclinical models to evaluate the contribution of flavor additives to nicotine addiction. MATERIALS AND METHODS An electronic literature search was conducted by authors up to May 2022. Original articles were selected. RESULTS The behavioral models of rodents described here capture multiple dimensions of human flavored nicotine use behaviors, including advantages and disadvantages. CONCLUSION The consensus of the literature search was that human research on nicotine use behavior has not caught up with fast-changing product innovations, marketing practices, and federal regulations. Animal models are therefore needed to investigate mechanisms underlying nicotine use and addiction. This review provides a comprehensive overvie.
Collapse
Affiliation(s)
- Deniz Bagdas
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Yale Tobacco Center of Regulatory Science, Yale School of Medicine, New Haven, CT, USA
| | - Nardos Kebede
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Andy Ma Zepei
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Lilley Harris
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Karina Minanov
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Marina R. Picciotto
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Yale Tobacco Center of Regulatory Science, Yale School of Medicine, New Haven, CT, USA
| | - Nii A. Addy
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Yale Tobacco Center of Regulatory Science, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| |
Collapse
|
11
|
Wang H, Chen H, Han S, Fu Y, Tian Y, Liu Y, Wang A, Hou H, Hu Q. Decreased mitochondrial DNA copy number in nerve cells and the hippocampus during nicotine exposure is mediated by autophagy. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 226:112831. [PMID: 34592525 DOI: 10.1016/j.ecoenv.2021.112831] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 06/13/2023]
Abstract
Cigarette smoke is a harmful air pollutant and nicotine dependence is the essential cause of the tobacco epidemic. Since mitochondrial abnormalities are associated with substance addiction, in this work we used mitochondrial DNA (mtDNA) copy number as an indicator of mitochondrial function to investigate whether nicotine addicts also exhibit mitochondrial abnormalities. We found significantly lower mtDNA copy number in the peripheral blood of healthy nicotine addicts than in non-smokers, indicating that long-term nicotine exposure through smoking has detrimental effects on mitochondria. We also examined the effects of nicotine on mtDNA levels in a rat conditioned place preference (CPP) model of addiction and in cultured neuron cells, which revealed that the mtDNA copy number was significantly reduced in the hippocampus of CPP rats, in human neuroblastoma SH-SY5Y cells, and in rat pheochromocytoma PC12 cells, suggesting that significantly reduced mtDNA copy number is a potential biomarker of nicotine addiction. In SH-SY5Y cells, nicotine treatment induced several mitochondrial defects, such as increased mtDNA damage, increased reactive oxygen species (ROS) levels, decreased mitochondrial membrane potential (△Ψm), and stimulation of autophagic flux via transcriptional up-regulation of several autophagy-related genes and elevated marker protein accumulation, although genes controlling mtDNA replication were unaffected. In addition, pretreatment with the autophagy inhibitor Bafilomycin A1 led to accumulation of microtubule-associated protein 1 light chain 3b-II (LC3B-II) and counteracted the nicotine-induced decrease in mtDNA copy number. These results were recapitulated in PC12 cells, which also showed significant down-regulation of the marker SQSTM1/P62, suggesting that the decrease in mtDNA copy number is mediated by autophagy. This study shows that prolonged nicotine exposure, such as that in nicotine addicts, leads to a decrease of mtDNA copy number in neurons due to enhanced induction of autophagy. CAPSULE: It was found that smoking or nicotine exposure decreased mtDNA copy number based on population, animal, and cell models, and these effects appear to be mediated by autophagy.
Collapse
Affiliation(s)
- Hongjuan Wang
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China.
| | - Huan Chen
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China.
| | - Shulei Han
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China.
| | - Yaning Fu
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China.
| | - Yushan Tian
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China.
| | - Yong Liu
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.
| | - An Wang
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.
| | - Hongwei Hou
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China.
| | - Qingyuan Hu
- China National Tobacco Quality Supervision and Test Center, Zhengzhou, China.
| |
Collapse
|
12
|
Huggett SB, Johnson EC, Hatoum AS, Lai D, Srijeyanthan J, Bubier JA, Chesler EJ, Agrawal A, Palmer AA, Edenberg HJ, Palmer RHC. Genes identified in rodent studies of alcohol intake are enriched for heritability of human substance use. Alcohol Clin Exp Res 2021; 45:2485-2494. [PMID: 34751961 DOI: 10.1111/acer.14738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/24/2021] [Accepted: 10/29/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Rodent paradigms and human genome-wide association studies (GWAS) on drug use have the potential to provide biological insight into the pathophysiology of addiction. METHODS Using GeneWeaver, we created rodent alcohol and nicotine gene-sets derived from 19 gene expression studies on alcohol and nicotine outcomes. We partitioned the SNP-heritability of these gene-sets using four large human GWAS: 1) alcoholic drinks per week, 2) problematic alcohol use, 3) cigarettes per day and 4) smoking cessation. We benchmarked our findings with curated human alcoholism and nicotine addiction gene-sets and performed specificity analyses using other rodent gene-sets (e.g., locomotor behavior) and other human GWAS (e.g., height). RESULTS The rodent alcohol gene-set was enriched for heritability of drinks per week, cigarettes per day, and smoking cessation, but not problematic alcohol use. However, the rodent nicotine gene-set was not significantly associated with any of these traits. Both rodent gene-sets showed enrichment for several non-substance use GWAS, and the extent of this relationship tended to increase as a function of trait heritability. In general, larger gene-sets demonstrated more significant enrichment. Finally, when evaluating human traits with similar heritabilities, both rodent gene-sets showed greater enrichment for substance use traits. CONCLUSION Our results suggest that rodent gene expression studies can help to identify genes that contribute to heritability of some substance use traits in humans, yet there was less specificity than expected. We outline various limitations, interpretations and considerations for future research.
Collapse
Affiliation(s)
- Spencer B Huggett
- Department of Human Genetics, Emory University, Atlanta, GA, USA.,Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, GA University, Atlanta, USA
| | - Emma C Johnson
- Department of Psychiatry, Washington University, St Louis School of Medicine, St. Louis, MO, USA
| | - Alexander S Hatoum
- Department of Psychiatry, Washington University, St Louis School of Medicine, St. Louis, MO, USA
| | - Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jenani Srijeyanthan
- Department of Human Genetics, Emory University, Atlanta, GA, USA.,Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, GA University, Atlanta, USA
| | | | | | - Arpana Agrawal
- Department of Psychiatry, Washington University, St Louis School of Medicine, St. Louis, MO, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.,Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Howard J Edenberg
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Rohan H C Palmer
- Department of Human Genetics, Emory University, Atlanta, GA, USA.,Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, GA University, Atlanta, USA
| |
Collapse
|
13
|
Rosa MLDP, Machado CA, Oliveira BDS, Toscano ECDB, Asth L, de Barros JLVM, Teixeira AL, Moreira FA, de Miranda AS. Role of cytokine and neurotrophic factors in nicotine addiction in the conditioned place preference paradigm. Neurosci Lett 2021; 764:136235. [PMID: 34508846 DOI: 10.1016/j.neulet.2021.136235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 12/01/2022]
Abstract
The mechanisms involved in the maintenance of cigarette smoking and nicotine reward remain unclear. Immune response might play an important role in this context. Nicotine may induce both central and systemic inflammatory responses as well as changes in the regulation of brain-derived neurotrophic factor (BDNF). The conditioned place preference (CPP) is a method used for the evaluation of nicotine-induced reward, reproducing nicotine-seeking behavior in humans. So far, there are no studies investigating the relationship between neuroinflammation and nicotine-induced CPP. This study aimed to evaluate the levels of inflammatory mediators and neurotrophic factors in key areas of the central nervous system (CNS) of mice subject to nicotine-induced CPP. CPP was induced with an intraperitoneal administration of 0.5 mg/kg of nicotine in male Swiss mice, using an unbiased protocol. Control group received vehicle by the same route. The levels of cytokines, chemokines, and neurotrophic factors were measured using Enzyme-Linked Immunosorbent Assay (ELISA) in the brain after CPP test. As expected, nicotine induced place preference behavior. In parallel, we observed increased peripheral levels of IL-6 and IL-10 alongside increased hippocampal levels of NGF but decreased GDNF in mice treated with nicotine compared to controls. In the striatum, nicotine promoted decrease of IL-1ß, IL-10 and GDNF levels, while the levels of all the mediators were similar between groups in the pre-frontal cortex. Our results provide evidence on the role of cytokines and neurotrophic factors in nicotine-induced CPP in mice.
Collapse
Affiliation(s)
- Magda Luciana de Paula Rosa
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Caroline Amaral Machado
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Bruna da Silva Oliveira
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Eliana Cristina de Brito Toscano
- Laboratório de Patologia Celular e Molecular, Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Laila Asth
- Departmento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - João Luís Vieira Monteiro de Barros
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, TX, USA
| | - Fabrício A Moreira
- Departmento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Aline Silva de Miranda
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil.
| |
Collapse
|
14
|
Havermans A, Zwart EP, Cremers HWJM, van Schijndel MDM, Constant RS, Mešković M, Worutowicz LX, Pennings JLA, Talhout R, van der Ven LTM, Heusinkveld HJ. Exploring Neurobehaviour in Zebrafish Embryos as a Screening Model for Addictiveness of Substances. TOXICS 2021; 9:toxics9100250. [PMID: 34678946 PMCID: PMC8539716 DOI: 10.3390/toxics9100250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/23/2021] [Accepted: 09/29/2021] [Indexed: 12/04/2022]
Abstract
Tobacco use is the leading cause of preventable death worldwide and is highly addictive. Nicotine is the main addictive compound in tobacco, but less is known about other components and additives that may contribute to tobacco addiction. The zebrafish embryo (ZFE) has been shown to be a good model to study the toxic effects of chemicals on the neurological system and thus may be a promising model to study behavioral markers of nicotine effects, which may be predictive for addictiveness. We aimed to develop a testing protocol to study nicotine tolerance in ZFE using a locomotion test with light-dark transitions as behavioral trigger. Behavioral experiments were conducted using three exposure paradigms: (1) Acute exposure to determine nicotine’s effect and potency. (2) Pre-treatment with nicotine dose range followed by a single dose of nicotine, to determine which pre-treatment dose is sufficient to affect the potency of acute nicotine. (3) Pre-treatment with a single dose combined with acute exposure to a dose range to confirm the hypothesized decreased potency of the acute nicotine exposure. These exposure paradigms showed that (1) acute nicotine exposure decreased ZFE activity in response to dark conditions in a dose-dependent fashion; (2) pre-treatment with increasing concentrations dose-dependently reversed the effect of acute nicotine exposure; and (3) a fixed pre-treatment dose of nicotine induced a decreased potency of the acute nicotine exposure. This effect supported the induction of tolerance to nicotine by the pre-treatment, likely through neuroadaptation. The interpretation of these effects, particularly in view of prediction of dependence and addictiveness, and suitability of the ZFE model to test for such effects of other compounds than nicotine, are discussed.
Collapse
|
15
|
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
|
16
|
Rahmadi M, Suasana D, Lailis SR, Ratri DMN, Ardianto C. The effects of quercetin on nicotine-induced reward effects in mice. J Basic Clin Physiol Pharmacol 2021; 32:327-333. [PMID: 34214359 DOI: 10.1515/jbcpp-2020-0418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/21/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Tobacco smoking remains the primary cause of preventable mortality and morbidity in the world. The complexity of the nicotine dependency process included the withdrawal effect that triggers recurrence being the main problem. Quercetin, known as an antioxidant, binds free radicals and modulates endogenous antioxidants through Nrf2 activations is expected as a potential agent to reduce the risk of nicotine dependence. This research aims to evaluate quercetin's effects on reducing the risk of nicotine addiction. METHODS Conditioned Place Preference (CPP) with a biased design was used to evaluate nicotine's reward effects in male Balb/C mice. Preconditioning test was performed on day 1; conditioning test was done twice daily on day 2-4 by administering quercetin (i.p.) 50 mg/kg along with nicotine (s.c.) 0.5 mg/kg or Cigarette Smoke Extract (CSE) (s.c.) contained nicotine 0.5 mg/kg; and postconditioning test was performed on day 5 continue with extinction test on day 6, 8, 10, 12, and reinstatement test on day 13. The duration spent in each compartment was recorded and analyzed. RESULTS Nicotine 0.5 mg/kg and CSE 0.5 mg/kg significantly induced reward effects (p<0.05). There was no decrease of reward effect during the extinction-reinstatement stage of the postconditioning phase (p>0.05), while quercetin 50 mg/kg both induced along with nicotine or CSE was able to inhibit the reward effect of nicotine (p>0.05). CONCLUSIONS Quercetin reduced the risk of nicotine dependence and has a potential effect to use as a therapy for nicotine dependence, especially as a preventive agent.
Collapse
Affiliation(s)
- Mahardian Rahmadi
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Airlangga, Surabaya, Indonesia
| | - Dian Suasana
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Airlangga, Surabaya, Indonesia
| | - Silvy Restuning Lailis
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Airlangga, Surabaya, Indonesia
| | | | - Chrismawan Ardianto
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Airlangga, Surabaya, Indonesia
| |
Collapse
|
17
|
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
|
18
|
Assessing Neutralized Nicotine Distribution Using Mice Vaccinated with the Mucosal Conjugate Nicotine Vaccine. Vaccines (Basel) 2021; 9:vaccines9020118. [PMID: 33546163 PMCID: PMC7913222 DOI: 10.3390/vaccines9020118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 11/16/2022] Open
Abstract
Tobacco smoking continues to be a global epidemic and the leading preventable cause of cancer and cardiovascular disease. Nicotine vaccines have been investigated as an alternative to currently available smoking cessation strategies as a means to increase rates of success and long-term abstinence. Recently, we demonstrated that a mucosal nicotine vaccine was able to induce robust mucosal and systemic antibodies when delivered heterologously using intranasal and intramuscular routes. Herein, we investigated the neutralization ability of the anti-nicotine antibodies using both intranasal and intracardiac nicotine challenges. Combining the extraction of lyophilized organ samples with RP-HPLC methods, we were able to recover between 47% and 56% of the nicotine administered from the blood, brain, heart, and lungs up to 10 min after challenge, suggesting that the interaction of the antibodies with nicotine forms a stable complex independently of the route of vaccination or challenge. Although both challenge routes can be used for assessing systemic antibodies, only the intranasal administration of nicotine, which is more physiologically similar to the inhalation of nicotine, permitted the crucial interaction of nicotine with the mucosal antibodies generated using the heterologous vaccination route. Notably, these results were obtained 6 months after the final vaccination, demonstrating stable mucosal and systemic antibody responses.
Collapse
|
19
|
Zhang L, McCarthy DM, Eskow Jaunarajs KL, Biederman J, Spencer TJ, Bhide PG. Frontal Cortical Monoamine Release, Attention, and Working Memory in a Perinatal Nicotine Exposure Mouse Model Following Kappa Opioid Receptor Antagonism. Cereb Cortex 2021; 31:483-496. [PMID: 32869057 DOI: 10.1093/cercor/bhaa238] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 02/27/2024] Open
Abstract
Perinatal nicotine exposure (PNE) produces frontal cortical hypo-dopaminergic state and attention and working memory deficits consistent with neurodevelopmental disorders such as attention deficit hyperactivity disorder (ADHD). Methylphenidate alleviates ADHD symptoms by increasing extracellular dopamine and noradrenaline. Kappa opioid receptor (KOR) antagonism may be another mechanism to achieve the same results because KOR activation inhibits frontal cortical dopamine release. We administered the selective KOR antagonist norbinaltorphimine (norBNI) (20 mg/kg; intraperitoneal) or methylphenidate (0.75 mg/kg; intraperitoneal) to PNE mouse model and examined frontal cortical monoamine release, attention, and working memory. Both compounds increased dopamine and noradrenaline release but neither influenced serotonin release. Both compounds improved object-based attention and working memory in the PNE group, with norBNI's effects evident at 2.5 h and 5.5 h but absent at 24 h. Methylphenidate's effects were evident at 0.5 h but not at 2.5 h. norBNI's effects temporally coincided with frontal cortical c-Jun N-terminal kinase phosphorylation. norBNI did not alter tissue dopamine content in the nucleus accumbens, offering preliminary support for lack of reinforcement.
Collapse
Affiliation(s)
- Lin Zhang
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Deirdre M McCarthy
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | | | - Joseph Biederman
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Thomas J Spencer
- Pediatric Psychopharmacology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pradeep G Bhide
- Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
20
|
Íbias J, Nazarian A. Sex differences in nicotine-induced impulsivity and its reversal with bupropion in rats. J Psychopharmacol 2020; 34:1382-1392. [PMID: 32684065 PMCID: PMC7708527 DOI: 10.1177/0269881120937543] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Enhancement in cognitive impulsivity and the resulting alterations in decision making serve as a contributing factor for the development and maintenance of substance-use disorders. Nicotine-induced increases in impulsivity has been previously reported in male humans and rodents. Although the potential for sex differences in nicotine-induced impulsivity has not been examined. AIMS AND METHODS In the present study, male and female Sprague Dawley rats were submitted to a delay discounting task, in which several consecutive measures of self-control were taken. Firstly, rats were tested with vehicle, and next with nicotine doses of 0.4 and 0.8 mg/kg. Thereafter, chronic treatment with bupropion started, and the animals were tested again. Half the animals continued to receive 0.8 mg/kg of nicotine, while the rest received nicotine and also a daily dose of 30 mg/kg of bupropion. RESULTS When the animals were first tested with nicotine, female rats showed a significant nicotine dose dependent increase of impulsive behaviour, whereas male rats only showed a decrease on their elections of the larger but delayed reward under the highest dose of 0.8 mg/kg of nicotine. Treatment with bupropion blocked the effect of nicotine on decision making in female rats, as they showed results close to their baseline levels. On the other hand, bupropion did not affect the nicotine-induced delay discounting in male rats. CONCLUSION These findings demonstrate sexually dimorphic effects of nicotine on cognitive impulsivity which may help to shed light on nicotine use vulnerabilities observed in women.
Collapse
Affiliation(s)
| | - Arbi Nazarian
- Correspondence: Arbi Nazarian, Ph.D., Department of Pharmaceutical Sciences, Western University of Health Sciences, 309 E. Second Street, Pomona, CA 91766, USA. , (909) 469-5424
| |
Collapse
|
21
|
Chellian R, Wilson R, Polmann M, Knight P, Behnood-Rod A, Bruijnzeel AW. Evaluation of Sex Differences in the Elasticity of Demand for Nicotine and Food in Rats. Nicotine Tob Res 2020; 22:925-934. [PMID: 31603225 DOI: 10.1093/ntr/ntz171] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 09/04/2019] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Animal studies can inform policy regarding nicotine levels in tobacco products and e-cigarette solutions. Increasing the price of nicotine-containing products decreases their use, but it is unknown how the relationship between price and consumption is affected by both sex and nicotine dose. METHODS A behavioral economics procedure was used to determine the demand elasticity for nicotine in male and female rats. Demand elasticity describes the relationship between price and consumption. A high level of elasticity indicates that consumption is relatively sensitive to increases in price. The rats self-administered a low dose (0.01 mg/kg/inf) or a standard dose (0.03 mg/kg/inf) of nicotine for 9 days under a fixed-ratio (FR) 1 schedule. Then the price (FR schedule) of nicotine was increased, and a demand analysis was conducted. A similar study was conducted with palatable food pellets. RESULTS There were no sex differences in nicotine or food intake under the FR1 schedule. However, demand for 0.03 mg/kg/inf of nicotine was more elastic in females than males. Demand for 0.01 mg/kg/inf of nicotine and food was more elastic in males than females. CONCLUSIONS These findings indicate that there are no differences in nicotine and food intake between males and females when the price is low. When the price of nicotine or food is increased, males maintain their old level of intake longer than females when they have access to a standard dose of nicotine, and females maintain their intake longer when they have access to a low dose of nicotine or food. IMPLICATIONS This behavioral economics analysis indicates that there is no sex difference in nicotine intake when the price of nicotine is low. Increasing the price of nicotine decreases nicotine intake in a dose- and sex-specific manner. Males maintain their old level of intake longer when they have access to a standard dose of nicotine and females when they have access to a low dose. This has implications for tobacco regulatory policy. In a regulatory environment where only low nicotine-containing products are allowed, increasing the price of nicotine products may lead to a greater decrease in nicotine use in males than females.
Collapse
Affiliation(s)
| | - Ryann Wilson
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Michaela Polmann
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Parker Knight
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL.,Department of Neuroscience, University of Florida, Gainesville, FL
| |
Collapse
|
22
|
Flores RJ, Cruz B, Uribe KP, Correa VL, Arreguin MC, Carcoba LM, Mendez IA, O'Dell LE. Estradiol promotes and progesterone reduces anxiety-like behavior produced by nicotine withdrawal in female rats. Psychoneuroendocrinology 2020; 119:104694. [PMID: 32540678 PMCID: PMC7423767 DOI: 10.1016/j.psyneuen.2020.104694] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/17/2020] [Accepted: 04/11/2020] [Indexed: 12/15/2022]
Abstract
This study assessed sex differences and the role of ovarian hormones in nicotine withdrawal. Study 1 compared physical signs, anxiety-like behavior, and corticosterone levels in male, intact female, and ovariectomized (OVX) female rats during nicotine withdrawal. Estradiol (E2) and progesterone levels were also assessed in intact females that were tested during different phases of the 4-day estrous cycle. Study 2 assessed the role of ovarian hormones in withdrawal by comparing the same measures in OVX rats that received vehicle, E2, or E2+progesterone prior to testing. Briefly, rats received a sham surgery or an ovariectomy procedure. Fifteen days later, rats were prepared with a pump that delivered nicotine for 14 days. On the test day, rats received saline or the nicotinic receptor antagonist, mecamylamine to precipitate withdrawal. Physical signs and anxiety-like behavior were assessed on the elevated plus maze (EPM) and light-dark transfer (LDT) tests. During withdrawal, intact females displayed greater anxiety-like behavior and increases in corticosterone levels as compared to male and OVX rats. Females tested in the estrus phase (when E2 is relatively low) displayed less anxiety-like behavior and had lower corticosterone levels versus all other phases. Anxiety-like behavior and corticosterone levels were positively correlated with E2 and negatively correlated with progesterone levels. Intact females displaying high E2/low progesterone showed greater anxiety-like behavior and corticosterone levels as compared to females displaying low E2/high progesterone. Lastly, OVX-E2 rats displayed greater anxiety-like behavior than OVX-E2+progesterone rats. These data suggest that E2 promotes and progesterone reduces anxiety-like behavior produced by nicotine withdrawal.
Collapse
Affiliation(s)
- Rodolfo J Flores
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, USA
| | - Bryan Cruz
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, USA
| | - Kevin P Uribe
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, USA
| | - Victor L Correa
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, USA
| | - Montserrat C Arreguin
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, USA
| | - Luis M Carcoba
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, USA
| | - Ian A Mendez
- School of Pharmacy, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, USA
| | - Laura E O'Dell
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, USA.
| |
Collapse
|
23
|
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
|
24
|
Cepeda-Benito A. Nicotine Effects, Body Weight Concerns and Smoking: A Literature Review. Curr Pharm Des 2020; 26:2316-2326. [PMID: 32233995 DOI: 10.2174/1381612826666200401083040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/12/2020] [Indexed: 12/23/2022]
Abstract
How people become addicted to cigarette smoking and remain addicted despite repeated attempts to quit requires piecing together a rather complex puzzle. The present review contextualizes the role of nicotine and smoking sensory stimulation on maintaining smoking, describes nicotine's effects on feeding behavior and body weight, and explores the impact of smoking outcome expectancies, including the belief that nicotine suppresses appetite and body weight, on the decision to smoke or vape (use of e-cigarettes). The analysis concludes with a review of rat models of human nicotine intake that attempt to isolate the effects of nicotine on appetite and weight gain. Animal research replicates with relative closeness phenomena observed in smokers, but the rat model falls short of replicating the long-term weight gain observed post-smoking cessation.
Collapse
Affiliation(s)
- Antonio Cepeda-Benito
- Department of Psychological Science, Department of Medicine, University of Vermont Cancer Center, University of Vermont, Vermont, United States
| |
Collapse
|
25
|
Nonhuman animal models of substance use disorders: Translational value and utility to basic science. Drug Alcohol Depend 2020; 206:107733. [PMID: 31790978 PMCID: PMC6980671 DOI: 10.1016/j.drugalcdep.2019.107733] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The National Institute on Drug Abuse (NIDA) recently released a Request for Information (RFI) soliciting comments on nonhuman animal models of substance use disorders (SUD). METHODS A literature review was performed to address the four topics outlined in the RFI and one topic inspired by the RFI: (1) animal models that best recapitulate SUD, (2) animal models that best balance the trade-offs between resources and ecological validity, (3) animal models whose translational value are frequently misrepresented or overrepresented by the scientific community, (4) aspects of SUD that are not currently being modeled in animals, and (5) animal models that are optimal for examining the basic mechanisms by which drugs produce their abuse-related effects. RESULTS Models that employ response-contingent drug administration, use complex schedules of reinforcement, measure behaviors that mimic the distinguishing features of SUD, and use animals that are phylogenetically similar to humans have the greatest translational value. Models that produce stable and reproducible baselines of behavior, lessen the number of uncontrolled variables, and minimize the influence of extraneous factors are best at examining basic mechanisms contributing to drug reward and reinforcement. CONCLUSIONS Nonhuman animal models of SUD have undergone significant refinements to increase their utility for basic science and translational value for SUD. The existing literature describes numerous examples of how these models may best be utilized to answer mechanistic questions of drug reward and identify potential therapeutic interventions for SUD. Progress in the field could be accelerated by further collaborations between researchers using animals versus humans.
Collapse
|
26
|
Garcia‐Rivas V, Deroche‐Gamonet V. Not all smokers appear to seek nicotine for the same reasons: implications for preclinical research in nicotine dependence. Addict Biol 2019; 24:317-334. [PMID: 29480575 DOI: 10.1111/adb.12607] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 12/11/2017] [Accepted: 01/15/2018] [Indexed: 01/08/2023]
Abstract
Tobacco use leads to 6 million deaths every year due to severe long-lasting diseases. The main component of tobacco, nicotine, is recognized as one of the most addictive drugs, making smoking cessation difficult, even when 70 percent of smokers wish to do so. Clinical and preclinical studies have demonstrated consistently that nicotine seeking is a complex behavior involving various psychopharmacological mechanisms. Evidence supports that the population of smokers is heterogeneous, particularly as regards the breadth of motives that determine the urge to smoke. Here, we review converging psychological, genetic and neurobiological data from clinical and preclinical studies supporting that the mechanisms controlling nicotine seeking may vary from individual to individual. It appears timely that basic neuroscience integrates this heterogeneity to refine our understanding of the neurobiology of nicotine seeking, as tremendous progress has been made in modeling the various psychopharmacological mechanisms driving nicotine seeking in rodents. For a better understanding of the mechanisms that drive nicotine seeking, we emphasize the need for individual-based research strategies in which nicotine seeking, and eventually treatment efficacy, are determined while taking into account individual variations in the mechanisms of nicotine seeking.
Collapse
Affiliation(s)
- Vernon Garcia‐Rivas
- Université de Bordeaux France
- INSERM U1215, Psychobiology of Drug AddictionNeuroCentre Magendie France
| | - Véronique Deroche‐Gamonet
- Université de Bordeaux France
- INSERM U1215, Psychobiology of Drug AddictionNeuroCentre Magendie France
| |
Collapse
|
27
|
Schmidt HD, Rupprecht LE, Addy NA. Neurobiological and Neurophysiological Mechanisms Underlying Nicotine Seeking and Smoking Relapse. MOLECULAR NEUROPSYCHIATRY 2019; 4:169-189. [PMID: 30815453 PMCID: PMC6388439 DOI: 10.1159/000494799] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/23/2018] [Indexed: 12/19/2022]
Abstract
Tobacco-related morbidity and mortality continue to be a significant public health concern. Unfortunately, current FDA-approved smoking cessation pharmacotherapies have limited efficacy and are associated with high rates of relapse. Therefore, a better understanding of the neurobiological and neurophysiological mechanisms that promote smoking relapse is needed to develop novel smoking cessation medications. Here, we review preclinical studies focused on identifying the neurotransmitter and neuromodulator systems that mediate nicotine relapse, often modeled in laboratory animals using the reinstatement paradigm, as well as the plasticity-dependent neurophysiological mechanisms that facilitate nicotine reinstatement. Particular emphasis is placed on how these neuroadaptations relate to smoking relapse in humans. We also highlight a number of important gaps in our understanding of the neural mechanisms underlying nicotine reinstatement and critical future directions, which may lead toward the development of novel, target pharmacotherapies for smoking cessation.
Collapse
Affiliation(s)
- Heath D. Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laura E. Rupprecht
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nii A. Addy
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
- Interdepartmental Neuroscience Program, Yale Graduate School of Arts and Sciences, New Haven, Connecticut, USA
| |
Collapse
|
28
|
Karelitz JL, Perkins KA. Tobacco smoking may delay habituation of reinforcer effectiveness in humans. Psychopharmacology (Berl) 2018; 235:2315-2321. [PMID: 29777289 PMCID: PMC6045957 DOI: 10.1007/s00213-018-4927-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 05/10/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND The effectiveness of nonconsummatory reinforcers habituate, as their ability to maintain reinforced responding declines over repeated presentations. Preclinical research has shown that nicotine can delay habituation of reinforcer effectiveness, but this effect has not been directly demonstrated in humans. OBJECTIVE In preliminary translational research, we assessed effects of nicotine from tobacco smoking (vs. a no smoking control) on within-session patterns of responding for a brief visual reinforcer. METHODS Using a within-subjects design, 32 adult dependent smokers participated in two experimental sessions, varying by smoking condition: no smoking following overnight abstinence (verified by CO ≤ 10 ppm), or smoking of own cigarette without overnight abstinence. Adapted from preclinical studies, habituation of reinforcer effectiveness was assessed by determining the rate of decline in responding on a simple operant computer task for a visual reinforcer, available on a fixed ratio schedule. RESULTS Reinforced responding and duration of responding were each significantly higher in the smoking vs. no smoking condition. The within-session rate of responding declined significantly more slowly during the smoking vs. no smoking condition, consistent with delayed habituation of reinforcer effectiveness. Follow-up analyses indicated that withdrawal relief did not influence the difference in responding between conditions, suggesting the patterns of responding reflected positive, but not negative, reinforcement. CONCLUSIONS These results are a preliminary demonstration in humans that smoked nicotine may attenuate habituation, thereby maintaining the effectiveness of a reinforcer over a longer period of access. Further research is needed to confirm habituation and rule out alternative causes of declines in within-session responding.
Collapse
Affiliation(s)
- Joshua L Karelitz
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA.
| | - Kenneth A Perkins
- Department of Psychiatry, WPIC, University of Pittsburgh School of Medicine, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| |
Collapse
|
29
|
Kowiański P, Lietzau G, Steliga A, Czuba E, Ludkiewicz B, Waśkow M, Spodnik JH, Moryś J. Nicotine-induced CREB and DeltaFosB activity is modified by caffeine in the brain reward system of the rat. J Chem Neuroanat 2018; 88:1-12. [PMID: 29100904 DOI: 10.1016/j.jchemneu.2017.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 12/21/2022]
Abstract
Coffee and nicotine consumption are frequently combined, indicating possible intensifying effect of caffeine on smoking behavior, although neurobiological background of this phenomenon remains unknown. We aimed at determining the effect of caffeine and nicotine, applied separately or simultaneously, on activation of six structures of the brain reward system: nucleus accumbens (NAc), ventral tegmental area (VTA), amygdala (Amg), hippocampus (Hip), medial prefrontal cortex (mPfr) and dorsal striatum (CdP) in the adult male Wistar rats. Activation of two transcription factors, the phosphorylated form of cyclic AMP-response element binding protein (pCREB) and DeltaFosB (ΔFosB) was assessed by immunohistochemistry after multiple-dose five-days psychostimulants administration followed by 20min and 24h survival, respectively. Nicotine evoked the highest increase of pCREB-immunoreactivity (-ir) in NAc, while caffeine exerted the weakest effect in mPfr and CdP. Nicotine/caffeine co-administration resulted in decrease of pCREB-ir in NAc and increase in Amg, compared with the effect of each psychostimulant used separately. Nicotine was the strongest psychostimulant activating ΔFosB-ir in Amg, whereas caffeine - in Hip. Nicotine/caffeine-exerted effect upon ΔFosB-ir in Amg was weaker, whereas in mPfr stronger, than nicotine-evoked effect in these structures. In summary, pCREB and ΔFosB activation is dependent on the type of stimulus, brain structure and functional context. Activation of both transcription factors is responsible for caffeine's modifying effect upon nicotine-related behaviors and must be taken into account while quitting cigarette smoking.
Collapse
Affiliation(s)
- Przemysław Kowiański
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland; Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200 Slupsk, Poland.
| | - Grażyna Lietzau
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland; Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Aleksandra Steliga
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200 Slupsk, Poland
| | - Ewelina Czuba
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland
| | - Beata Ludkiewicz
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland
| | - Monika Waśkow
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200 Slupsk, Poland
| | - Jan H Spodnik
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland
| | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Str., 80-211 Gdansk, Poland
| |
Collapse
|
30
|
Pogun S, Yararbas G, Nesil T, Kanit L. Sex differences in nicotine preference. J Neurosci Res 2017; 95:148-162. [PMID: 27870459 DOI: 10.1002/jnr.23858] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/29/2016] [Accepted: 07/11/2016] [Indexed: 01/01/2023]
Abstract
Smoking is the major cause of preventable deaths worldwide, and although there is a decline in overall smoking prevalence in developed countries, the decline in women is less pronounced than in men. Women become dependent faster and experience greater difficulties in quitting. Similar trends have been observed in animal models of nicotine/tobacco addiction. Individual differences in vulnerability to drug abuse are also observed in nicotine/tobacco addiction and point to the importance of sex differences. This Review, summarizes findings from three experimental approaches used to depict nicotine preference in animal models, intravenous and oral nicotine self-administration and nicotine-induced conditioned place preference. Nicotine preference is considered to be reflected in the animal's motivation to administer the drug (intravenously or orally) or to prefer an environment paired with the presence of the drug (conditioned place preference). These approaches all point to the importance of sex and age of the subjects; the preference of females and adolescents appear to be more pronounced than that of males and adults, respectively. A closer look at these factors will help us understand the mechanisms that underlie nicotine addiction and develop strategies to cope. Ignoring sex differences and reaching conclusions based only on studies using male subjects has resulted in erroneous generalizations in the past. Sex differences in nicotine preference have been clearly documented, and awareness on this aspect of nicotine dependence will significantly impact our success in translational research. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sakire Pogun
- Center for Brain Research, Ege University, Izmir, Turkey
| | - Gorkem Yararbas
- Institute on Drug Abuse, Toxicology and Pharmaceutical Science, Ege University, Izmir, Turkey
| | - Tanseli Nesil
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, Virginia
| | - Lutfiye Kanit
- Center for Brain Research, Ege University, Izmir, Turkey.,Physiology Department, School of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
31
|
Pipkin JA, Cruz B, Flores RJ, Hinojosa CA, Carcoba LM, Ibarra M, Francis W, Nazarian A, O'Dell LE. Both nicotine reward and withdrawal are enhanced in a rodent model of diabetes. Psychopharmacology (Berl) 2017; 234:1615-1622. [PMID: 28342091 PMCID: PMC5437741 DOI: 10.1007/s00213-017-4592-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 03/07/2017] [Indexed: 01/09/2023]
Abstract
RATIONALE It is presently unclear whether diabetic rats experience greater rewarding effects of nicotine and/or negative affective states produced by nicotine withdrawal. OBJECTIVE The present study utilized a rodent model of diabetes to examine the rewarding effects of nicotine and negative affective states and physical signs produced by withdrawal. METHODS Separate groups of rats received systemic administration of either vehicle or streptozotocin (STZ), which destroys insulin-producing beta cells in the pancreas and elevates glucose levels. Place conditioning procedures were utilized to compare the rewarding effects of nicotine (conditioned place preference; CPP) and negative affective states produced by withdrawal (conditioned place aversion; CPA) in vehicle- and STZ-treated rats. CPA and physical signs of withdrawal were compared after administration of the nicotinic receptor antagonist mecamylamine to precipitate withdrawal in nicotine-dependent rats. A subsequent study utilized elevated plus maze (EPM) procedures to compare anxiety-like behavior produced by nicotine withdrawal in vehicle- and STZ-treated rats. RESULTS STZ-treated rats displayed greater rewarding effects of nicotine and a larger magnitude of aversive effects and physical signs produced by withdrawal as compared to vehicle-treated controls. STZ-treated rats also displayed higher levels of anxiety-like behavior on the EPM during nicotine withdrawal as compared to controls. CONCLUSION The finding that both nicotine reward and withdrawal are enhanced in a rodent model of diabetes implies that the strong behavioral effects of nicotine promote tobacco use in persons with metabolic disorders, such as diabetes.
Collapse
Affiliation(s)
- Joseph A Pipkin
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79902, USA
| | - Bryan Cruz
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79902, USA
| | - Rodolfo J Flores
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79902, USA
| | - Cecilia A Hinojosa
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79902, USA
| | - Luis M Carcoba
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79902, USA
| | - Melissa Ibarra
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79902, USA
| | - Wendy Francis
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79902, USA
| | - Arbi Nazarian
- Department of Pharmaceutical Sciences, Western University of Health Sciences, 309 East Second Street, Pomona, CA, 91766, USA
| | - Laura E O'Dell
- Department of Psychology, The University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79902, USA.
| |
Collapse
|
32
|
Dalaveri F, Nakhaee N, Esmaeilpour K, Mahani SE, Sheibani V. Effects of maternal separation on nicotine-induced conditioned place preference and subsequent learning and memory in adolescent female rats. Neurosci Lett 2016; 639:151-156. [PMID: 27931777 DOI: 10.1016/j.neulet.2016.11.059] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/09/2016] [Accepted: 11/28/2016] [Indexed: 11/17/2022]
Abstract
Adverse early life experiences can potentially increase risk for drug abuse later in life. However, little research has been conducted studying the effects of maternal separation (MS), an experimental model for early life stress, on the rewarding effects of nicotine. Cognitive function may be affected by MS. So, we also investigated whether nicotine administration affect spatial learning and memory in MS adolescent female rats. Rat pups were subjected to daily MS for 15min (MS15) or 180min (MS180) during the first 2 weeks of life or reared under normal animal facility rearing (AFR) conditions. The place preference test was performed with nicotine (0.6mg/kg,s.c.) or vehicle over a period of 6 conditioning trials during adolescence. Spatial learning and memory performance was evaluated by using Morris water maze (MWM). In our study, adolescent female rats exposed to MS180 shown a significantly greater preference for a nicotine-paired compartment during the testing phase than the MS15 group. Nicotine altered the MS-induced spatial learning defects in the MS180 group. These findings suggest that MS may increase sensitivity to the rewarding effects of nicotine and also it is possible to suggest that nicotine administration may influence learning dysfunction induced by MS in adolescent female rats.
Collapse
Affiliation(s)
- Fatemeh Dalaveri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Nouzar Nakhaee
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Esmaeili Mahani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Physiology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
33
|
Perkins KA, Kunkle N, Michael VC, Karelitz JL, Donny EC. Assessing Discrimination of Nicotine in Humans Via Cigarette Smoking. Nicotine Tob Res 2016; 18:1830-6. [PMID: 27013335 PMCID: PMC4978980 DOI: 10.1093/ntr/ntw082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/01/2016] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Nicotine's interoceptive stimulus effects likely help explain smoking's reinforcing efficacy, but human studies have been limited by difficulties controlling dosing via tobacco inhalation. Our objective was to describe a procedure to study nicotine discrimination via smoking. METHODS Dependent smokers abstinent overnight (>12 hours) were first "trained" to discriminate between two cigarettes differing in nicotine content, based on four puffs of exposure, and then tested on whether they successfully acquired that discrimination. After piloting with Quest brand commercial cigarettes, 29 subjects engaged in the main study with cigarettes available through NIDA (Spectrum; 16mg vs. 0.4mg nicotine content). Discrimination training first involved two trials, one with each cigarette, prior to six testing trials. Due to results with the first 20 subjects, the remaining nine received two training trials with each cigarette (four total). Subjective perceptions were also assessed during each testing trial, and puff choice between the two cigarettes available concurrently was assessed after testing, on the last two trials. RESULTS All five pilot subjects successfully discriminated Quest 1 versus Quest 3 (defined by at least five out of six trials correct, ie, >80%). Yet, only 10 of 20 subjects (50%) were able to discriminate the two Spectrum cigarettes based on two training trials. After changing to four training trials, eight of nine subjects were able to discriminate (89%). Subjective perceptions and puff choice differed between cigarettes more in those able versus unable to discriminate them. CONCLUSIONS With sufficient training exposures, smokers can discriminate nicotine between cigarettes differing in nicotine contents. IMPLICATIONS The interoceptive stimulus effects of nicotine are critical to understanding reinforcement from cigarette smoking behavior. Because of the very recent availability of Spectrum research cigarettes from NIDA, with specific known amounts of nicotine content, the study of nicotine discrimination in humans via cigarette smoking may now be feasible. Our results demonstrate that, with sufficient training, smokers can behaviorally discriminate nicotine from four puffs' exposure between cigarettes differing in nicotine contents. Future research should evaluate human discrimination of nicotine from greater amounts of cigarette smoke exposure, as well as in response to other procedural variations.
Collapse
Affiliation(s)
- Kenneth A Perkins
- Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA;
| | - Nicole Kunkle
- Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Valerie C Michael
- Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Joshua L Karelitz
- Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Eric C Donny
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
34
|
A key role for the N/OFQ-NOP receptor system in modulating nicotine taking in a model of nicotine and alcohol co-administration. Sci Rep 2016; 6:26594. [PMID: 27199205 PMCID: PMC4873733 DOI: 10.1038/srep26594] [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: 01/25/2016] [Accepted: 05/05/2016] [Indexed: 11/08/2022] Open
Abstract
Alcohol and nicotine are often co-abused. Although the N/OFQ-NOP receptor system is considered a potential target for development of drug abuse pharmacotherapies, especially for alcoholism, little is known about the role of this system in nicotine dependence. Furthermore, the effect of prior history of nicotine dependence on subsequent nicotine and alcohol taking is understudied. Using an operant co-administration paradigm, in which rats concurrently self-administer nicotine and alcohol, we found that nicotine dependent rats increased nicotine self-administration over time as compared to non-dependent animals, while patterns of alcohol lever pressing did not change between groups. Pretreatment with the potent NOP receptor agonist AT-202 (0.3–3 mg/kg) increased nicotine lever pressing of both dependent and non-dependent groups, whereas the selective antagonist SB612111 (1–10 mg/kg) elicited a clear reduction of nicotine responses, in both dependent and non-dependent rats. In parallel, AT-202 only produced minor changes on alcohol responses and SB612111 reduced alcohol taking at a dose that also reduced locomotor behavior. Results indicate that a history of nicotine dependence affects subsequent nicotine- but not alcohol-maintained responding, and that NOP receptor antagonism, rather than agonism, blocks nicotine self-administration, which strongly suggests a critical role for the endogenous N/OFQ in the modulation of nicotine reinforcement processes.
Collapse
|
35
|
Insight into the Potential Factors That Promote Tobacco Use in Vulnerable Populations. CURRENT ADDICTION REPORTS 2016. [DOI: 10.1007/s40429-016-0091-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
36
|
Schassburger RL, Pitzer EM, Smith TT, Rupprecht LE, Thiels E, Donny EC, Sved AF. Adolescent Rats Self-Administer Less Nicotine Than Adults at Low Doses. Nicotine Tob Res 2016; 18:1861-1868. [PMID: 26764255 DOI: 10.1093/ntr/ntw006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/04/2016] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Although nearly 90% of current smokers initiated tobacco use during adolescence, little is known about reinforcement by nicotine in adolescents. Researchers are currently investigating whether a potential public health policy setting a tobacco product standard with very low nicotine levels would improve public health, and it is essential to understand whether data generated in adults translates to adolescents, particularly as it relates to the threshold dose of nicotine required to support smoking. The present study compared self-administration of low doses of nicotine between adolescent and adult rats. METHODS Adolescent (postnatal day [P] 30) and adult (P90) male and female rats were allowed to nosepoke to receive intravenous infusions of nicotine (3-100 μg/kg/infusion) during 16 daily 1-hour sessions. RESULTS At 10 μg/kg/infusion nicotine, adolescent rats earned significantly fewer infusions than adults. When responding for 30 μg/kg/infusion nicotine, rats of both ages earned a similar number of infusions; however, there were subtle differences in the distribution of infusions across the 1-hour session. No sex differences were apparent in either age group at any dose. CONCLUSIONS These results demonstrate that adolescent rats are less sensitive than adults to the primary reinforcing effects of nicotine. However, at nicotine doses that support self-administration in both age groups, adolescent and adult rats do not differ in acquisition or number of infusions earned. These results suggest that reducing nicotine levels in cigarettes to a level that does not support smoking in adults may be sufficient to reduce the acquisition of smoking in adolescents. IMPLICATIONS The results of the present studies demonstrate that adolescent rats are less sensitive than adults to the primary reinforcing effects of nicotine. These results suggest that reducing nicotine levels in cigarettes to a level that does not support smoking in adults will be sufficient to reduce the acquisition of smoking in adolescents.
Collapse
Affiliation(s)
| | - Emily M Pitzer
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh , Pittsburgh, PA
| | - Tracy T Smith
- Department of Psychology, Dietrich School of Arts and Sciences, University of Pittsburgh , Pittsburgh, PA
| | | | - Edda Thiels
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA.,Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Eric C Donny
- Department of Psychology, Dietrich School of Arts and Sciences, University of Pittsburgh , Pittsburgh, PA
| | - Alan F Sved
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA.,Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA.,Department of Psychology, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
37
|
Caenorhabditis elegans as a Model to Study the Molecular and Genetic Mechanisms of Drug Addiction. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 137:229-52. [PMID: 26810004 DOI: 10.1016/bs.pmbts.2015.10.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Drug addiction takes a massive toll on society. Novel animal models are needed to test new treatments and understand the basic mechanisms underlying addiction. Rodent models have identified the neurocircuitry involved in addictive behavior and indicate that rodents possess some of the same neurobiologic mechanisms that mediate addiction in humans. Recent studies indicate that addiction is mechanistically and phylogenetically ancient and many mechanisms that underlie human addiction are also present in invertebrates. The nematode Caenorhabditis elegans has conserved neurobiologic systems with powerful molecular and genetic tools and a rapid rate of development that enables cost-effective translational discovery. Emerging evidence suggests that C. elegans is an excellent model to identify molecular mechanisms that mediate drug-induced behavior and potential targets for medications development for various addictive compounds. C. elegans emit many behaviors that can be easily quantitated including some that involve interactions with the environment. Ethanol (EtOH) is the best-studied drug-of-abuse in C. elegans and at least 50 different genes/targets have been identified as mediating EtOH's effects and polymorphisms in some orthologs in humans are associated with alcohol use disorders. C. elegans has also been shown to display dopamine and cholinergic system-dependent attraction to nicotine and demonstrate preference for cues previously associated with nicotine. Cocaine and methamphetamine have been found to produce dopamine-dependent reward-like behaviors in C. elegans. These behavioral tests in combination with genetic/molecular manipulations have led to the identification of dozens of target genes/systems in C. elegans that mediate drug effects. The one target/gene identified as essential for drug-induced behavioral responses across all drugs of abuse was the cat-2 gene coding for tyrosine hydroxylase, which is consistent with the role of dopamine neurotransmission in human addiction. Overall, C. elegans can be used to model aspects of drug addiction and identify systems and molecular mechanisms that mediate drug effects. The findings are surprisingly consistent with analogous findings in higher-level organisms. Further, model refinement is warranted to improve model validity and increase utility for medications development.
Collapse
|
38
|
Sanchez V, Lycas MD, Lynch WJ, Brunzell DH. Wheel running exercise attenuates vulnerability to self-administer nicotine in rats. Drug Alcohol Depend 2015; 156:193-198. [PMID: 26433561 PMCID: PMC4633318 DOI: 10.1016/j.drugalcdep.2015.09.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 09/04/2015] [Accepted: 09/11/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Preventing or postponing tobacco use initiation could greatly reduce the number of tobacco-related deaths. While evidence suggests that exercise is a promising treatment for tobacco addiction, it is not clear whether exercise could prevent initial vulnerability to tobacco use. Thus, using an animal model, we examined whether exercise attenuates vulnerability to the use and reinforcing effects of nicotine, the primary addictive chemical in tobacco. METHODS Initial vulnerability was assessed using an acquisition procedure wherein exercising (unlocked running wheel, n=10) and sedentary (locked or no wheel, n=12) male adolescent rats had access to nicotine infusions (0.01-mg/kg) during daily 21.5-h sessions beginning on postnatal day 30. Exercise/sedentary sessions (2-h/day) were conducted prior to each of the acquisition sessions. The effects of exercise on nicotine's reinforcing effects were further assessed in separate groups of exercising (unlocked wheel, n=7) and sedentary (no wheel, n=5) rats responding for nicotine under a progressive-ratio schedule with exercise/sedentary sessions (2-h/day) conducted before the daily progressive-ratio sessions. RESULTS While high rates of acquisition of nicotine self-administration were observed among both groups of sedentary controls, acquisition was robustly attenuated in the exercise group with only 20% of exercising rats meeting the acquisition criterion within the 16-day testing period as compared to 67% of the sedentary controls. Exercise also decreased progressive-ratio responding for nicotine as compared to baseline and to sedentary controls. CONCLUSIONS Exercise may effectively prevent the initiation of nicotine use in adolescents by reducing the reinforcing effects of nicotine.
Collapse
Affiliation(s)
- Victoria Sanchez
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, 450 Ray C. Hunt Drive, Room G167, Charlottesville, VA 22902, USA
| | - Matthew D Lycas
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, 450 Ray C. Hunt Drive, Room G167, Charlottesville, VA 22902, USA
| | - Wendy J Lynch
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, 450 Ray C. Hunt Drive, Room G167, Charlottesville, VA 22902, USA.
| | - Darlene H Brunzell
- Department of Pharmacology and Toxicology, Interdepartmental Neuroscience Program, Virginia Commonwealth University, PO Box 980613, 410 N. 12th St. Richmond, VA 23298, USA
| |
Collapse
|
39
|
Lenoir M, Starosciak AK, Ledon J, Booth C, Zakharova E, Wade D, Vignoli B, Izenwasser S. Sex differences in conditioned nicotine reward are age-specific. Pharmacol Biochem Behav 2015; 132:56-62. [PMID: 25735492 PMCID: PMC4552616 DOI: 10.1016/j.pbb.2015.02.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 02/17/2015] [Accepted: 02/20/2015] [Indexed: 02/05/2023]
Abstract
Women constitute half of all smokers and many studies suggest that adult males and females differ in factors that maintain tobacco smoking, yet there is limited information about sex differences in nicotine reward during adolescence. Limited studies suggest that adolescent male rats self-administer more nicotine than adults, suggesting that drug administration during adolescence leads to different behavioral effects than during adulthood. In the present study, male rats developed a significant conditioned place preference (CPP) to lower doses of nicotine than females, regardless of age. In addition, adolescents were more sensitive than adults. In female rats, adolescents exhibited a CPP of greater magnitude than adult females. In males, the magnitude of the CPP did not differ as a function of age, but adolescents exhibited CPP to lower doses than adults. There also were differences in nicotinic acetylcholinergic receptor binding in nucleus accumbens and caudate putamen in response to nicotine across age and sex. These findings suggest that it is necessary to consider sex- and age-specific effects of drugs such as nicotine when developing strategies for improving smoking cessation treatments.
Collapse
Affiliation(s)
- Magalie Lenoir
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Rm 4113A (D-80), Miami, FL 33136, USA
| | - Amy K Starosciak
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Rm 4113A (D-80), Miami, FL 33136, USA
| | - Jennifer Ledon
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Rm 4113A (D-80), Miami, FL 33136, USA
| | - Caitlin Booth
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Rm 4113A (D-80), Miami, FL 33136, USA
| | - Elena Zakharova
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Rm 4113A (D-80), Miami, FL 33136, USA
| | - Dean Wade
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Rm 4113A (D-80), Miami, FL 33136, USA
| | - Beatrice Vignoli
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Rm 4113A (D-80), Miami, FL 33136, USA
| | - Sari Izenwasser
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, Rm 4113A (D-80), Miami, FL 33136, USA.
| |
Collapse
|
40
|
Qi X, Yamada H, Corrie LW, Ji Y, Bauzo RM, Alexander JC, Bruijnzeel AW. A critical role for the melanocortin 4 receptor in stress-induced relapse to nicotine seeking in rats. Addict Biol 2015; 20:324-35. [PMID: 24612112 DOI: 10.1111/adb.12129] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tobacco addiction is characterized by a lack of control over smoking and relapse after periods of abstinence. Smoking cessation leads to a dysphoric state that contributes to relapse to smoking. After the acute withdrawal phase, exposure to stressors increases the risk for relapse. Blockade of melanocortin 4 (MC4 ) receptors has anxiolytic and antidepressant-like effects in animal models. The aim of these studies was to investigate the role of MC4 receptors in the dysphoria associated with nicotine withdrawal and stress-induced reinstatement of nicotine seeking. To study stress-induced reinstatement, rats self-administered nicotine for 16 days and then nicotine seeking was extinguished by substituting saline for nicotine. Nicotine seeking was reinstated by intermittent footshock stress. The intracranial self-stimulation (ICSS) procedure was used to assess the negative mood state associated with nicotine withdrawal. Elevations in the ICSS thresholds are indicative of a dysphoric state. The selective MC4 receptor antagonists HS014 and HS024 prevented stress-induced reinstatement of extinguished nicotine seeking. Drug doses that prevented stress-induced relapse did not affect responding for food pellets, which indicates that the drugs did not induce sedation or motor impairments. In the ICSS experiments, the nicotinic acetylcholine receptor antagonist mecamylamine elevated the ICSS thresholds of the nicotine-dependent rats. Pre-treatment with HS014 or HS024 did not prevent the elevations in ICSS thresholds. These studies indicate that MC4 receptors play a critical role in stress-induced reinstatement of nicotine seeking, but these receptors may not play a role in the dysphoria associated with acute nicotine withdrawal.
Collapse
Affiliation(s)
- Xiaoli Qi
- Department of Psychiatry; McKnight Brain Institute; University of Florida; Gainesville FL USA
| | - Hidetaka Yamada
- Department of Psychiatry; McKnight Brain Institute; University of Florida; Gainesville FL USA
| | - Lu W. Corrie
- Department of Psychiatry; McKnight Brain Institute; University of Florida; Gainesville FL USA
| | - Yue Ji
- Department of Psychiatry; McKnight Brain Institute; University of Florida; Gainesville FL USA
| | - Rayna M. Bauzo
- Department of Psychiatry; McKnight Brain Institute; University of Florida; Gainesville FL USA
| | - Jon C. Alexander
- Department of Psychiatry; McKnight Brain Institute; University of Florida; Gainesville FL USA
| | - Adrie W. Bruijnzeel
- Department of Psychiatry; McKnight Brain Institute; University of Florida; Gainesville FL USA
| |
Collapse
|
41
|
Goodwin AK, Hiranita T, Paule MG. The Reinforcing Effects of Nicotine in Humans and Nonhuman Primates: A Review of Intravenous Self-Administration Evidence and Future Directions for Research. Nicotine Tob Res 2015; 17:1297-310. [PMID: 25673111 DOI: 10.1093/ntr/ntv002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 01/02/2015] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Cigarette smoking is largely driven by the reinforcing properties of nicotine. Intravenous (IV) self-administration procedures are the gold standard for investigating the reinforcing effects of psychoactive drugs. The goal of this review was to examine the results of published investigations of the reinforcing effects of nicotine measured using IV self-administration procedures in humans and nonhuman primates. RESULTS The body of literature using nonhuman primate subjects indicates nicotine functions as a positive reinforcer when available for self-administration via IV catheters. However, it can also be difficult to establish IV nicotine self-administration in nonhuman primates and sometimes supplemental strategies have been required (e.g., priming injections or food deprivation) before subjects acquire the behavior. Although the body of literature using human subjects is limited, the evidence indicates nicotine functions as a reinforcer via the IV route of administration in adult cigarette smokers. Rates of nicotine self-injection can be variable across subjects and responding is sometimes inconsistent across sessions in both humans and nonhuman primates. CONCLUSIONS The Family Smoking Prevention and Tobacco Control Act, enacted in 2009, gave the Food and Drug Administration regulatory authority over the manufacture, marketing, and distribution of tobacco products. Research examining the threshold reinforcing doses for initiation and maintenance of nicotine self-administration, comparisons of the reinforcing effects of nicotine in adolescent versus adult subjects, investigations of gender differences in the reinforcing effects of nicotine, and studies of the abuse liability of non-nicotine tobacco product constituents and their ability to alter the reinforcing effects of nicotine will inform potential tobacco regulatory actions.
Collapse
Affiliation(s)
- Amy K Goodwin
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR
| | - Takato Hiranita
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR
| | - Merle G Paule
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR
| |
Collapse
|
42
|
Abstract
There is abundant evidence that the dopamine (DA) neurons that project to the nucleus accumbens play a central role in neurobiological mechanisms underpinning drug dependence. This chapter considers the ways in which these projections facilitate the addiction to nicotine and tobacco. It focuses on the complimentary roles of the two principal subdivisions of the nucleus accumbens, the accumbal core and shell, in the acquisition and maintenance of nicotine-seeking behavior. The ways in which tonic and phasic firing of the neurons contributes to the ways in which the accumbens mediate the behavioral responses to nicotine are also considered. Experimental studies suggest that nicotine has relatively weak addictive properties which are insufficient to explain the powerful addictive properties of tobacco smoke. This chapter discusses hypotheses that seek to explain this conundrum. They implicate both discrete sensory stimuli closely paired with the delivery of tobacco smoke and contextual stimuli habitually associated with the delivery of the drug. The mechanisms by which each type of stimulus influence tobacco dependence are hypothesized to depend upon the increased DA release and overflow, respectively, in the two subdivisions of the accumbens. It is suggested that a majority of pharmacotherapies for tobacco dependence are not more successful because they fail to address this important aspect of the dependence.
Collapse
Affiliation(s)
- David J K Balfour
- Medical Research Institute, Division of Neuroscience, Ninewells Hospital and Medical School, Dundee, DD1 9SY, Scotland,
| |
Collapse
|
43
|
Abstract
An aversive abstinence syndrome manifests 4-24 h following cessation of chronic use of nicotine-containing products. Symptoms peak on approximately the 3rd day and taper off over the course of the following 3-4 weeks. While the severity of withdrawal symptoms is largely determined by how nicotine is consumed, certain short nucleotide polymorphisms (SNPs) have been shown to predispose individuals to consume larger amounts of nicotine more frequently--as well as to more severe symptoms of withdrawal when trying to quit. Additionally, rodent behavioral models and transgenic mouse models have revealed that specific nicotinic acetylcholine receptor (nAChR) subunits, cellular components, and neuronal circuits are critical to the expression of withdrawal symptoms. Consequently, by continuing to map neuronal circuits and nAChR subpopulations that underlie the nicotine withdrawal syndrome--and by continuing to enumerate genes that predispose carriers to nicotine addiction and exacerbated withdrawal symptoms--it will be possible to pursue personalized therapeutics that more effectively treat nicotine addiction.
Collapse
Affiliation(s)
- Ian McLaughlin
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | | |
Collapse
|
44
|
Rosenthal DG, Weitzman M, Benowitz NL. Nicotine Addiction: Mechanisms and Consequences. INTERNATIONAL JOURNAL OF MENTAL HEALTH 2014. [DOI: 10.2753/imh0020-7411400102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
| | | | - Neal L. Benowitz
- b Departments of Medicine, Bioengineering, and Therapeutic Sciences, University of California San Francisco
| |
Collapse
|
45
|
Ahsan HM, de la Peña JBI, Botanas CJ, Kim HJ, Yu GY, Cheong JH. Conditioned place preference and self-administration induced by nicotine in adolescent and adult rats. Biomol Ther (Seoul) 2014; 22:460-6. [PMID: 25414778 PMCID: PMC4201227 DOI: 10.4062/biomolther.2014.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 06/13/2014] [Accepted: 06/24/2014] [Indexed: 11/05/2022] Open
Abstract
Nicotine addiction is a worldwide problem. However, previous studies characterizing the rewarding and reinforcing effects of nicotine in animal models have reported inconsistent findings. It was observed that the addictive effects are variable on different factors (e.g. route, dose, and age). Here, we evaluated the rewarding and reinforcing effects of nicotine in different routes of administration, across a wide dose range, and in different age groups. Two of the most widely used animal models of drug addiction were employed: the conditioned place preference (CPP) and self-administration (SA) tests. Nicotine CPP was evaluated in different routes [intraperitoneal (i.p.) and subcutaneous (s.c.)], doses (0.05 to 1.0 mg/kg) and age [adolescent and adult rats]. Similarly, intravenous nicotine SA was assessed in different doses (0.01 to 0.06 mg/kg/infusion) and age (adolescent and adult rats). In the CPP test, s.c. nicotine produced greater response than i.p. The 0.2 mg/kg dose produced highest CPP response in adolescent, while 0.6 mg/kg in adult rats; which were also confirmed in 7 days pretreated rats. In the SA test, adolescent rats readily self-administer 0.03 mg/kg/infusion of nicotine. Doses that produced nicotine CPP and SA induced blood nicotine levels that corresponded well with human smokers. In conclusion, we have demonstrated that nicotine produces reliable CPP [0.2 mg/kg dose (s.c.)] in adolescents and [0.6 mg/kg dose (s.c.)] in adults, and SA [0.03 mg/kg/infusion] in adolescent rats. Both tests indicate that adolescent rats are more sensitive to the rewarding and reinforcing effects of nicotine.
Collapse
Affiliation(s)
- Hafiz Muhammad Ahsan
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 139-742
| | - June Bryan I de la Peña
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 139-742
| | - Chrislean Jun Botanas
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 139-742
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 139-742
| | - Gu Yong Yu
- Department of Chemistry, Sahmyook University, Seoul 139-742, Republic of Korea
| | - Jae Hoon Cheong
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 139-742
| |
Collapse
|
46
|
Grassi MC, Ferketich AK, Enea D, Culasso F, Nencini P. Validity of the Italian version of the Severity of Dependence Scale (SDS) for nicotine dependence in smokers intending to quit. Psychol Rep 2014; 114:1-13. [PMID: 24765705 DOI: 10.2466/18.15.pr0.114k16w7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The objective was to test the psychometric properties of an Italian version of the Severity of Dependence Scale, a five-item measure designed to assess the compulsive dimension of drug dependence. 635 smokers enrolled in a tobacco dependence treatment program served as the participants. The Fagerström Test for Nicotine Dependence was used as a comparative measure. Dimensionality of the Severity of Dependence Scale and the Fagerström Test for Nicotine Dependence was assessed by factor analysis. Prediction of smoking at one year was evaluated by logistic regression. Factor analysis yielded a two-factor solution; however, the second factor explained very little variance. Factor 1 had a Cronbach's alpha of .66 (overall Scale coefficient = .44). The total Severity of Dependence Score predicted smoking at one year (OR = 1.10).
Collapse
|
47
|
Ashare RL, Schmidt HD. Optimizing treatments for nicotine dependence by increasing cognitive performance during withdrawal. Expert Opin Drug Discov 2014; 9:579-94. [PMID: 24707983 DOI: 10.1517/17460441.2014.908180] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Current FDA-approved smoking cessation pharmacotherapies have limited efficacy and are associated with high rates of relapse. Therefore, there is a clear need to develop novel antismoking medications. Nicotine withdrawal is associated with cognitive impairments that predict smoking relapse. It has been proposed that these cognitive deficits are a hallmark of nicotine withdrawal that could be targeted in order to prevent smoking relapse. Thus, pharmacotherapies that increase cognitive performance during nicotine withdrawal may represent potential smoking cessation agents. AREAS COVERED The authors review the clinical literature demonstrating that nicotine withdrawal is associated with deficits in working memory, attention and response inhibition. They then briefly summarize different classes of compounds and strategies to increase cognitive performance during nicotine withdrawal. Particular emphasis has been placed on translational research in order to highlight areas for which there is strong rationale for pilot clinical trials of potential smoking cessation medications. EXPERT OPINION There is emerging evidence that supports deficits in cognitive function as a plausible nicotine withdrawal phenotype. The authors furthermore believe that the translational paradigms presented here may represent efficient and valid means for the evaluation of cognitive-enhancing medications as possible treatments for nicotine dependence.
Collapse
Affiliation(s)
- Rebecca L Ashare
- University of Pennsylvania, Perelman School of Medicine, Center for Interdisciplinary Research on Nicotine Addiction, Department of Psychiatry , 3535 Market St, Suite 4100, Philadelphia, PA 19104 , USA +1 215 746 5789 ;
| | | |
Collapse
|
48
|
Dickson PE, Miller MM, Rogers TD, Blaha CD, Mittleman G. Effects of adolescent nicotine exposure and withdrawal on intravenous cocaine self-administration during adulthood in male C57BL/6J mice. Addict Biol 2014; 19:37-48. [PMID: 22978678 DOI: 10.1111/j.1369-1600.2012.00496.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Studies of adolescent drug use show (1) a pattern in which the use of tobacco precedes the use of other drugs and (2) a positive relationship between adolescent tobacco use and later drug use. These observations have led to the hypothesis that a causal relationship exists between early exposure to nicotine and the later use of hard drugs such as cocaine. Using male C57BL/6J mice, we tested the hypothesis that nicotine exposure in adolescence leads to increased intravenous self-administration (IVSA) of cocaine in adulthood. Using miniature osmotic pumps, we exposed mice and their littermate controls to nicotine (24 mg/kg/day) or vehicle, respectively, over the entire course of adolescence [postnatal days (P) 28-56]. Nicotine exposure was terminated on P56 and mice were not exposed to nicotine again during the experiment. On P73, mice were allowed to acquire cocaine IVSA (1.0 mg/kg/infusion) and a dose-response curve was generated (0.18, 0.32, 0.56, 1.0, 1.8 mg/kg/infusion). Lever pressing during extinction conditions was also evaluated. All mice rapidly learned to lever press for the combination of cocaine infusions and non-drug stimuli. Analysis of the dose-response curve revealed that adolescent nicotine-exposed mice self-administered significantly more (P < 0.05) cocaine than controls at all but the highest dose. No significant differences were observed between adolescent nicotine-exposed and control mice during the acquisition or extinction stages. These results indicate that adolescent nicotine exposure can increase cocaine IVSA in mice, which suggests the possibility of a causal link between adolescent tobacco use and later cocaine use in humans.
Collapse
Affiliation(s)
| | | | | | | | - Guy Mittleman
- Department of Psychology; University of Memphis; Memphis TN USA
| |
Collapse
|
49
|
Wang T, Han W, Wang B, Jiang Q, Solberg-Woods LC, Palmer AA, Chen H. Propensity for social interaction predicts nicotine-reinforced behaviors in outbred rats. GENES BRAIN AND BEHAVIOR 2013; 13:202-12. [PMID: 24289793 DOI: 10.1111/gbb.12112] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 10/14/2013] [Accepted: 11/26/2013] [Indexed: 12/30/2022]
Abstract
Social and genetic factors can influence smoking behavior. Using olfactogustatory stimuli as the sensory cue for intravenous nicotine self-administration (SA), we previously showed that social learning of nicotine contingent odor cue prevented rats from developing conditioned taste aversion and allowed them to instead establish stable nicotine SA. We hypothesized that genetic factors influenced socially acquired nicotine SA. A heterogeneous stock (HS; N/NIH) of outbred rats was trained to self-administer nicotine using the social learning protocol. Both male and female HS rats acquired nicotine SA, but females self-administered more nicotine than males. After extinction, the context previously paired with nicotine SA, in conjunction with socially transmitted drug cues, was sufficient to cause reinstatement of drug-seeking behavior. Wide variation in both nicotine intake and reinstatement was observed. Using multiple regression analysis, we found that measures of social interaction were significant predictors of nicotine intake and reinstatement of drug seeking in both males and females. Furthermore, measures of depression were predictors of nicotine intake in both males and females, anxiety was a predictor only in males and response to novelty was a predictor only in females. In males, measures of both depression and anxiety predicted nicotine reinstatement. Together, these data supported the ideas that genetically determined propensities for emotional and social phenotypes are significant determinants for nicotine-reinforced behavior, and that the HS rat is a suitable tool for dissecting genetic mechanisms that may underlie the interaction between social behavior, anxiety, depression and smoking.
Collapse
Affiliation(s)
- T Wang
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Perkins KA, Karelitz JL. Reinforcement enhancing effects of nicotine via smoking. Psychopharmacology (Berl) 2013; 228:479-86. [PMID: 23494236 PMCID: PMC3707934 DOI: 10.1007/s00213-013-3054-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/25/2013] [Indexed: 02/06/2023]
Abstract
RATIONALE In animals, nicotine enhances reinforcement from stimuli unrelated to nicotine intake. Human research is suggestive but has not clearly shown a similar influence of nicotine. OBJECTIVES We assessed acute effects of nicotine via smoking on enhancement of positive (money, music) or negative (termination of noise) reinforcers, or no "reward" (control). These different rewards determined the generalizability of nicotine effects. MATERIALS AND METHODS Dependent (n = 25) and nondependent (n = 27) smokers participated in three sessions, each after overnight abstinence. Using a within-subjects design, sessions involved no smoking or smoking denicotinized (0.05 mg) or nicotine (0.6 mg) Quest(R) brand cigarettes. For comparison, a fourth session involved no abstinence prior to smoking one's own brand to gauge responses under typical nicotine satiation. Reinforcement was assessed by responses on a simple operant computer task for the rewards, each available singly on a progressive ratio schedule during separate trials. RESULTS The reinforcing effect of music, but not other rewards, was greater due to the nicotine cigarette, compared to the denicotinized cigarette or no smoking. Reinforcement enhancing effects of nicotine did not differ between dependent and nondependent groups, indicating no influence of withdrawal relief. Responding due to acute nicotine after abstinence was very similar to responding to one's own brand after no abstinence. CONCLUSIONS Acute nicotine intake per se from smoking after abstinence enhances the reinforcing value of rewards unassociated with smoking, perhaps in a manner comparable to ad lib smoking after no abstinence. Nicotine's reinforcement enhancing effects may be specific to certain rewards, perhaps those sensory in nature.
Collapse
Affiliation(s)
- Kenneth A Perkins
- Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, 3811 O'Hara Street, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|