1
|
Bayoglu M, Ozturk Bintepe M, Kanit L, Balkan B, Gozen O, Koylu EO, Keser A. Decreased anxiety-like behavior in a selectively bred high nicotine-preferring rat line. Int J Neurosci 2024; 134:1403-1413. [PMID: 37929683 DOI: 10.1080/00207454.2023.2279505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Genetic vulnerability contributes significantly to the individual variability observed in nicotine dependence. Selective breeding for sensitivity to a particular effect of abused drugs has produced rodent lines useful for studying genetic vulnerability to drug addiction. Previous research showed that anxiety-related personality traits are associated with nicotine dependence. Therefore, we examined the differences in anxiety-like behavior between a high nicotine-preferring rat line and their controls. At the beginning of the study, all rats, naïve to any drug, were exposed sequentially to open field arena, marble-burying and elevated plus-maze paradigms. In the second step, all rats received nicotine in drinking water for 7 weeks. Behavioral tests were rerun on the final 2 weeks of chronic nicotine treatment. Elevated plus-maze testings under basal condition and during chronic nicotine treatment showed that the time spent on the open arms, preference for being in the open arms, and the latency to enter the closed arms were higher, whereas open arm avoidance index was lower in nicotine-preferring rats compared to the controls. In the open field test, nicotine-preferring rats spent longer time in the central zone and excreted less fecal pellets; they buried less marbles in the marble-burying test. These findings indicate a lower level of anxiety-like behavior in nicotine-preferring rat line under basal conditions and during chronic nicotine treatment. We conclude that lower anxiety level in nicotine-preferring rat line is consistent with novelty-seeking personality type and may increase vulnerability to nicotine dependence in this rat line.
Collapse
Affiliation(s)
- Merve Bayoglu
- Neuroscience Department, Ege University, Institute of Health Sciences, Izmir, Turkey
| | | | - Lutfiye Kanit
- Neuroscience Department, Ege University, Institute of Health Sciences, Izmir, Turkey
- Faculty of Medicine, Physiology Department, Ege University, Izmir, Turkey
- Center for Brain Research, Ege University, Izmir, Turkey
| | - Burcu Balkan
- Neuroscience Department, Ege University, Institute of Health Sciences, Izmir, Turkey
- Faculty of Medicine, Physiology Department, Ege University, Izmir, Turkey
- Center for Brain Research, Ege University, Izmir, Turkey
| | - Oguz Gozen
- Neuroscience Department, Ege University, Institute of Health Sciences, Izmir, Turkey
- Faculty of Medicine, Physiology Department, Ege University, Izmir, Turkey
- Center for Brain Research, Ege University, Izmir, Turkey
| | - Ersin O Koylu
- Neuroscience Department, Ege University, Institute of Health Sciences, Izmir, Turkey
- Faculty of Medicine, Physiology Department, Ege University, Izmir, Turkey
- Center for Brain Research, Ege University, Izmir, Turkey
| | - Aysegul Keser
- Neuroscience Department, Ege University, Institute of Health Sciences, Izmir, Turkey
- Faculty of Medicine, Physiology Department, Ege University, Izmir, Turkey
- Center for Brain Research, Ege University, Izmir, Turkey
| |
Collapse
|
2
|
Roy TA, Bubier JA, Dickson PE, Wilcox TD, Ndukum J, Clark JW, Sukoff Rizzo SJ, Crabbe JC, Denegre JM, Svenson KL, Braun RE, Kumar V, Murray SA, White JK, Philip VM, Chesler EJ. Discovery and validation of genes driving drug-intake and related behavioral traits in mice. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12875. [PMID: 38164795 PMCID: PMC10780947 DOI: 10.1111/gbb.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 01/03/2024]
Abstract
Substance use disorders are heritable disorders characterized by compulsive drug use, the biological mechanisms for which remain largely unknown. Genetic correlations reveal that predisposing drug-naïve phenotypes, including anxiety, depression, novelty preference and sensation seeking, are predictive of drug-use phenotypes, thereby implicating shared genetic mechanisms. High-throughput behavioral screening in knockout (KO) mice allows efficient discovery of the function of genes. We used this strategy in two rounds of candidate prioritization in which we identified 33 drug-use candidate genes based upon predisposing drug-naïve phenotypes and ultimately validated the perturbation of 22 genes as causal drivers of substance intake. We selected 19/221 KO strains (8.5%) that had a difference from control on at least one drug-naïve predictive behavioral phenotype and determined that 15/19 (~80%) affected the consumption or preference for alcohol, methamphetamine or both. No mutant exhibited a difference in nicotine consumption or preference which was possibly confounded with saccharin. In the second round of prioritization, we employed a multivariate approach to identify outliers and performed validation using methamphetamine two-bottle choice and ethanol drinking-in-the-dark protocols. We identified 15/401 KO strains (3.7%, which included one gene from the first cohort) that differed most from controls for the predisposing phenotypes. 8 of 15 gene deletions (53%) affected intake or preference for alcohol, methamphetamine or both. Using multivariate and bioinformatic analyses, we observed multiple relations between predisposing behaviors and drug intake, revealing many distinct biobehavioral processes underlying these relationships. The set of mouse models identified in this study can be used to characterize these addiction-related processes further.
Collapse
Affiliation(s)
- Tyler A. Roy
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - Jason A. Bubier
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - Price E. Dickson
- Joan C Edwards School of MedicineMarshall UniversityHuntingtonWest VirginiaUSA
| | - Troy D. Wilcox
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - Juliet Ndukum
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - James W. Clark
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - Stacey J. Sukoff Rizzo
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
- School of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - John C. Crabbe
- VA Portland Health Care SystemOregon Health & Science UniversityPortlandOregonUSA
| | - James M. Denegre
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - Karen L. Svenson
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - Robert E. Braun
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - Vivek Kumar
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - Stephen A. Murray
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | | | - Vivek M. Philip
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| | - Elissa J. Chesler
- Center for Addiction BiologyThe Jackson LaboratoryBar HarborMaineUSA
| |
Collapse
|
3
|
Roy TA, Bubier JA, Dickson PE, Wilcox TD, Ndukum J, Clark JW, Rizzo SJS, Crabbe JC, Denegre JM, Svenson KL, Braun RE, Kumar V, Murray SA, White JK, Philip VM, Chesler EJ. DISCOVERY AND VALIDATION OF GENES DRIVING DRUG-INTAKE AND RELATED BEHAVIORAL TRAITS IN MICE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.09.548280. [PMID: 37503148 PMCID: PMC10369854 DOI: 10.1101/2023.07.09.548280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Substance use disorders (SUDs) are heritable disorders characterized by compulsive drug use, but the biological mechanisms driving addiction remain largely unknown. Genetic correlations reveal that predisposing drug-naïve phenotypes, including anxiety, depression, novelty preference, and sensation seeking, are predictive of drug-use phenotypes, implicating shared genetic mechanisms. Because of this relationship, high-throughput behavioral screening of predictive phenotypes in knockout (KO) mice allows efficient discovery of genes likely to be involved in drug use. We used this strategy in two rounds of screening in which we identified 33 drug-use candidate genes and ultimately validated the perturbation of 22 of these genes as causal drivers of substance intake. In our initial round of screening, we employed the two-bottle-choice paradigms to assess alcohol, methamphetamine, and nicotine intake. We identified 19 KO strains that were extreme responders on at least one predictive phenotype. Thirteen of the 19 gene deletions (68%) significantly affected alcohol use three methamphetamine use, and two both. In the second round of screening, we employed a multivariate approach to identify outliers and performed validation using methamphetamine two-bottle choice and ethanol drinking-in-the-dark protocols. We identified 15 KO strains that were extreme responders across the predisposing drug-naïve phenotypes. Eight of the 15 gene deletions (53%) significantly affected intake or preference for three alcohol, eight methamphetamine or three both (3). We observed multiple relations between predisposing behaviors and drug intake, revealing many distinct biobehavioral processes underlying these relationships. The set of mouse models identified in this study can be used to characterize these addiction-related processes further.
Collapse
Affiliation(s)
| | | | - Price E. Dickson
- Joan C Edwards School of Medicine, Marshall University Huntington, WV
| | | | | | | | - Stacey J. Sukoff Rizzo
- The Jackson Laboratory, Bar Harbor, ME
- University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - John C. Crabbe
- Oregon Health & Science University and VA Portland Health Care System, Portland, OR
| | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Bahi A, Dreyer JL. Effects of chronic psychosocial stress on 'binge-like' sucrose intake in mice. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110625. [PMID: 36055562 DOI: 10.1016/j.pnpbp.2022.110625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 11/26/2022]
Abstract
Binge eating episodes are persistent and are essential features of numerous eating disorders (EDs). Susceptibility to EDs is largely presumed to be associated with early life stress. In fact, converging evidence from preclinical animal studies have implicated stress as a driver of binge eating. Still, literature examination indicates that vulnerability to EDs may depend on factors such as severity, time, and the type of stressor. Therefore, we aimed at exploring the link between chronic psychosocial stress and 'binge-like' sucrose intake in adolescent mice. To this aim, intruders' experimental mice were exposed to the chronic subordinate colony (CSC) housing, in the presence of a resident aggressive mouse for 2 weeks. At the end of the stress period, mice were tested for anxiety-like behavior then assessed for 'binge-like' intake of sucrose using a long-term drinking in the dark (DID) method that successfully replicates binge eating in humans. As expected, and compared to single housed colony controls (SHC), CSC exposure elicited an anxiogenic-like response in the open field (OF) and elevated-plus maze (EPM) tests and reduced weight gain. Most importantly, we report here for the first time, that mice exposed to chronic psychosocial stress displayed a 'binge-like' consumption of sucrose. However, neither quinine (bitter) nor saccharin (sweet) intakes were affected by CSC exposure. Finally, using Pearson's correlation, results showed a strong correlation between anxiety-like behavior parameters and sucrose intake. Overall these findings support the validity of our chronic psychosocial stress to model binge EDs and establish the long-term consequences of stress on 'binge-like' eating in male mice. These data suggest that chronic psychosocial stress is a risk factor for developing anxiety-associated EDs.
Collapse
Affiliation(s)
- Amine Bahi
- College of Medicine, Ajman University, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Jean-Luc Dreyer
- Division of Biochemistry, Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| |
Collapse
|
5
|
Karam CS, Williams BL, Morozova I, Yuan Q, Panarsky R, Zhang Y, Hodgkinson CA, Goldman D, Kalachikov S, Javitch JA. Functional Genomic Analysis of Amphetamine Sensitivity in Drosophila. Front Psychiatry 2022; 13:831597. [PMID: 35250674 PMCID: PMC8894854 DOI: 10.3389/fpsyt.2022.831597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/24/2022] [Indexed: 11/14/2022] Open
Abstract
Abuse of psychostimulants, including amphetamines (AMPHs), is a major public health problem with profound psychiatric, medical, and psychosocial complications. The actions of these drugs at the dopamine transporter (DAT) play a critical role in their therapeutic efficacy as well as their liability for abuse and dependence. To date, however, the mechanisms that mediate these actions are not well-understood, and therapeutic interventions for AMPH abuse have been limited. Drug exposure can induce broad changes in gene expression that can contribute to neuroplasticity and effect long-lasting changes in neuronal function. Identifying genes and gene pathways perturbed by drug exposure is essential to our understanding of the molecular basis of drug addiction. In this study, we used Drosophila as a model to examine AMPH-induced transcriptional changes that are DAT-dependent, as those would be the most relevant to the stimulatory effects of the drug. Using this approach, we found genes involved in the control of mRNA translation to be significantly upregulated in response to AMPH in a DAT-dependent manner. To further prioritize genes for validation, we explored functional convergence between these genes and genes we identified in a genome-wide association study of AMPH sensitivity using the Drosophila Genetic Reference Panel. We validated a number of these genes by showing that they act specifically in dopamine neurons to mediate the behavioral effects of AMPH. Taken together, our data establish Drosophila as a powerful model that enables the integration of behavioral, genomic and transcriptomic data, followed by rapid gene validation, to investigate the molecular underpinnings of psychostimulant action.
Collapse
Affiliation(s)
- Caline S Karam
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Brenna L Williams
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Irina Morozova
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, United States.,Department of Chemical Engineering, Columbia University, New York, NY, United States
| | - Qiaoping Yuan
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Rony Panarsky
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Yuchao Zhang
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Colin A Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Sergey Kalachikov
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, United States.,Department of Chemical Engineering, Columbia University, New York, NY, United States
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States.,Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| |
Collapse
|
6
|
Stafford AM, Yamamoto BK, Phillips TJ. Combined and sequential effects of alcohol and methamphetamine in animal models. Neurosci Biobehav Rev 2021; 131:248-269. [PMID: 34543650 PMCID: PMC8642292 DOI: 10.1016/j.neubiorev.2021.09.019] [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: 09/25/2020] [Revised: 09/11/2021] [Accepted: 09/12/2021] [Indexed: 11/16/2022]
Abstract
Comorbid drug use, often alcohol with other drugs, poses significant health and societal concerns. Methamphetamine is among the illicit drugs most often co-used with alcohol. The current review examines the animal literature for impacts of comorbid alcohol and methamphetamine exposure. We found evidence for additive or synergistic effects of combined or sequential exposure on behavior and physiology. Dopaminergic, serotonergic, and glutamatergic systems are all impacted by combined exposure to alcohol and methamphetamine and cyclooxygenase-2 activity plays an important role in their combined neurotoxic effects. Adverse consequences of comorbid exposure include altered brain development with prenatal exposure, impaired learning and memory, motor deficits, gastrotoxicity, hepatotoxicity, and augmented intake under some conditions. Given high susceptibility to drug experimentation in adolescence, studies of co-exposure during the adolescent period and of how adolescent exposure to one drug impacts later use or sensitivity to the other drug should be a priority. Further, to gain traction on prevention and treatment, additional research to identify motivational and neurobiological drivers and consequences of comorbid use is needed.
Collapse
Affiliation(s)
- Alexandra M Stafford
- Department of Behavioral Neuroscience, Portland Alcohol Abuse Research Center and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA.
| | - Bryan K Yamamoto
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tamara J Phillips
- Department of Behavioral Neuroscience, Portland Alcohol Abuse Research Center and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA; Veterans Affairs Portland Health Care System, Portland, OR, USA
| |
Collapse
|
7
|
Phillips TJ, Aldrich SJ. Peri-adolescent exposure to (meth)amphetamine in animal models. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:1-51. [PMID: 34801166 PMCID: PMC9134876 DOI: 10.1016/bs.irn.2021.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Experimentation with psychoactive drugs is often initiated in the peri-adolescent period, but knowledge of differences in the outcomes of peri-adolescent- vs adult-initiated exposure is incomplete. We consider the existing animal research in this area for (meth)amphetamines. Established for a number of phenotypes, is lower sensitivity of peri-adolescents than adults to acute effects of (meth)amphetamines, including neurotoxic effects of binge-level exposure. More variable are data for long-term consequences of peri-adolescent exposure on motivational and cognitive traits. Moreover, investigations often exclude an adult-initiated exposure group critical for answering questions about outcomes unique to peri-adolescent initiation. Despite this, it is clear from the animal research that (meth)amphetamine exposure during the peri-adolescent period, whether self- or other-administered, impacts brain motivational circuitry and cognitive function, and alters adult sensitivity to other drugs and natural rewards. Such consequences occurring in humans have the potential to predispose toward unfortunate and potentially disastrous family, social and livelihood outcomes.
Collapse
Affiliation(s)
- T J Phillips
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States; Veterans Affairs Portland Health Care System, Portland, OR, United States.
| | - S J Aldrich
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
8
|
Kamens HM, Miller CN, Caulfield JI, Zeid D, Horton WJ, Silva CP, Sebastian A, Albert I, Gould TJ, Fishbein D, Grigson PS, Cavigelli SA. Adolescent Stress Reduces Adult Morphine-Induced Behavioral Sensitization in C57BL/6J Mice. Front Behav Neurosci 2021; 15:678102. [PMID: 34149372 PMCID: PMC8209305 DOI: 10.3389/fnbeh.2021.678102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022] Open
Abstract
Deaths related to opioid use have skyrocketed in the United States, leading to a public health epidemic. Research has shown that both biological (genes) and environmental (stress) precursors are linked to opioid use. In particular, stress during adolescence-a critical period of frontal lobe development-influences the likelihood of abusing drugs. However, little is known about the biological mechanisms through which adolescent stress leads to long-term risk of opioid use, or whether genetic background moderates this response. Male and female C57BL/6J and BALB/cJ mice were exposed to chronic variable social stress (CVSS) or control conditions throughout adolescence and then tested for morphine locomotor sensitization or morphine consumption in adulthood. To examine possible mechanisms that underlie stress-induced changes in morphine behaviors, we assessed physiological changes in response to acute stress exposure and prefrontal cortex (PFC) miRNA gene expression. Adolescent stress did not influence morphine sensitization or consumption in BALB/cJ animals, and there was limited evidence of stress effects in female C57BL/6J mice. In contrast, male C57BL/6J mice exposed to adolescent CVSS had blunted morphine sensitization compared to control animals; no differences were observed in the acute locomotor response to morphine administration or morphine consumption. Physiologically, C57BL/6J mice exposed to CVSS had an attenuated corticosterone recovery following an acute stressor and downregulation of twelve miRNA in the PFC compared to control mice. The specificity of the effects for C57BL/6J vs. BALB/cJ mice provides evidence of a gene-environment interaction influencing opioid behaviors. However, this conclusion is dampened by limited locomotor sensitization observed in BALB/cJ mice. It remains possible that results may differ to other doses of morphine or other behavioral responses. Long-term differences in stress reactivity or miRNA expression in C57BL/6J mice suggests two possible biological mechanisms to evaluate in future research.
Collapse
Affiliation(s)
- Helen M. Kamens
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, United States
| | - Carley N. Miller
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, United States
| | - Jasmine I. Caulfield
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Dana Zeid
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, United States
| | - William J. Horton
- Department of Psychology, Bucknell University, Lewisburg, PA, United States
| | - Constanza P. Silva
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, United States
| | - Aswathy Sebastian
- Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States
| | - Istvan Albert
- Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States
| | - Thomas J. Gould
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, United States
| | - Diana Fishbein
- Department of Human Development and Family Studies, The Pennsylvania State University, University Park, PA, United States
- FPG Child Development Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Patricia Sue Grigson
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, United States
| | - Sonia A. Cavigelli
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
9
|
Stocco MR, El-Sherbeni AA, Zhao B, Novalen M, Tyndale RF. The role of CYP2D in rat brain in methamphetamine-induced striatal dopamine and serotonin release and behavioral sensitization. Psychopharmacology (Berl) 2021; 238:1791-1804. [PMID: 33649968 PMCID: PMC8233297 DOI: 10.1007/s00213-021-05808-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/22/2021] [Indexed: 11/28/2022]
Abstract
RATIONALE Cytochrome P450 2D (CYP2D) enzymes metabolize many addictive drugs, including methamphetamine. Variable CYP2D metabolism in the brain may alter CNS drug/metabolite concentrations, consequently affecting addiction liability and neuropsychiatric outcomes; components of these can be modeled by behavioral sensitization in rats. METHODS To investigate the role of CYP2D in the brain in methamphetamine-induced behavioral sensitization, rats were pretreated centrally with a CYP2D irreversible inhibitor (or vehicle) 20 h prior to each of 7 daily methamphetamine (0.5 mg/kg subcutaneous) injections. In vivo brain microdialysis was used to assess brain drug and metabolite concentrations, and neurotransmitter release. RESULTS CYP2D inhibitor (versus vehicle) pretreatment enhanced methamphetamine-induced stereotypy response sensitization. CYP2D inhibitor pretreatment increased brain methamphetamine concentrations and decreased the brain p-hydroxylation metabolic ratio. With microdialysis conducted on days 1 and 7, CYP2D inhibitor pretreatment exacerbated stereotypy sensitization and enhanced dopamine and serotonin release in the dorsal striatum. Day 1 brain methamphetamine and amphetamine concentrations correlated with dopamine and serotonin release, which in turn correlated with the stereotypy response slope across sessions (i.e., day 1 through day 7), used as a measure of sensitization. CONCLUSIONS CYP2D-mediated methamphetamine metabolism in the brain is sufficient to alter behavioral sensitization, brain drug concentrations, and striatal dopamine and serotonin release. Moreover, day 1 methamphetamine-induced neurotransmitter release may be an important predictor of subsequent behavioral sensitization. This suggests the novel contribution of CYP2D in the brain to methamphetamine-induced behavioral sensitization and suggests that the wide variation in human brain CYP2D6 may contribute to differential methamphetamine responses and chronic effects.
Collapse
Affiliation(s)
- Marlaina R. Stocco
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario Canada ,Campbell Family Mental Health Research Institute, CAMH, Toronto, Ontario Canada
| | - Ahmed A. El-Sherbeni
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario Canada ,Department of Clinical Pharmacy, Tanta University, Tanta, Egypt
| | - Bin Zhao
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario Canada ,Campbell Family Mental Health Research Institute, CAMH, Toronto, Ontario Canada
| | - Maria Novalen
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario Canada ,Campbell Family Mental Health Research Institute, CAMH, Toronto, Ontario Canada
| | - Rachel F. Tyndale
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario Canada ,Campbell Family Mental Health Research Institute, CAMH, Toronto, Ontario Canada ,Department of Psychiatry, University of Toronto, Toronto, Ontario Canada
| |
Collapse
|
10
|
Shab G, Fultz EK, Page A, Coelho MA, Brewin LW, Stailey N, Brown CN, Bryant CD, Kippin TE, Szumlinski KK. The motivational valence of methamphetamine relates inversely to subsequent methamphetamine self-administration in female C57BL/6J mice. Behav Brain Res 2020; 398:112959. [PMID: 33053382 DOI: 10.1016/j.bbr.2020.112959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/21/2020] [Accepted: 10/05/2020] [Indexed: 01/21/2023]
Abstract
Understanding the mechanisms underpinning individual variance in addiction vulnerability requires the development of validated, high-throughput screens. In a prior study of a large sample of male isogenic C57BL/6J mice, the direction and magnitude of methamphetamine (MA)-induced place-conditioning predicts the propensity to acquire oral MA self-administration, as well as the efficacy of MA to serve as a reinforcer. The present study examined whether or not such a predictive relationship also exists in females. Adult C57BL/6J females underwent a 4-day MA place-conditioning paradigm (once daily injections of 2 mg/kg) and were then trained to nose-poke for delivery of a 20 mg/L MA solution under increasing schedules of reinforcement, followed by dose-response testing (5-400 mg/L MA). Akin to males, 53 % of the females exhibited a conditioned place-preference, while 32 % of the mice were MA-neutral and 15 % exhibited a conditioned place-aversion. However, unlike males, the place-conditioning phenotype did not transfer to MA-reinforced nose-poking behavior under operant-conditioning procedures, with 400 mg/L MA intake being inversely correlated place-conditioning. While only one MA-conditioning dose has been assayed to date, these data indicate that sex does not significantly shift the proportion of C57BL/6J mice that perceive MA's interoceptive effects as positive, neutral or aversive. However, a sex difference appears to exist regarding the predictive relationship between the motivational valence of MA and subsequent drug-taking behavior; females exhibit MA-taking behavior and reinforcement, despite their initial perception of the stimulant interoceptive effects as positive, neutral or negative.
Collapse
Affiliation(s)
- Gabriella Shab
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, CA, USA
| | - Elissa K Fultz
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, CA, USA
| | - Ariana Page
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, CA, USA
| | - Michal A Coelho
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, CA, USA
| | - Lindsey W Brewin
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, CA, USA
| | - Nicholas Stailey
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, CA, USA
| | - Chelsea N Brown
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, CA, USA
| | - Camron D Bryant
- Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Tod E Kippin
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, CA, USA; Department of Molecular, Cellular and Developmental Biology and the Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, CA, USA; Institute for Collaborative Biology, University of California at Santa Barbara, Santa Barbara, CA, USA
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, CA, USA; Department of Molecular, Cellular and Developmental Biology and the Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, CA, USA.
| |
Collapse
|
11
|
Reed C, Stafford AM, Mootz JRK, Baba H, Erk J, Phillips TJ. A breeding strategy to identify modifiers of high genetic risk for methamphetamine intake. GENES BRAIN AND BEHAVIOR 2020; 20:e12667. [PMID: 32424970 DOI: 10.1111/gbb.12667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/12/2022]
Abstract
Trace amine-associated receptor 1 (Taar1) impacts methamphetamine (MA) intake. A mutant allele (Taar1m1J ) derived from the DBA/2J mouse strain codes for a non-functional receptor, and Taar1m1J/m1J mice consume more MA than mice possessing the reference Taar1+ allele. To study the impact of this mutation in a genetically diverse population, heterogeneous stock-collaborative cross (HS-CC) mice, the product of an eight-way cross of standard and wild-derived strains, were tested for MA intake. HS-CC had low MA intake, so an HS-CC by DBA/2J strain F2 intercross was created to transfer the mutant allele onto the diverse background, and used for selective breeding. To study residual variation in MA intake existing in Taar1m1J/m1J mice, selective breeding for higher (MAH) vs lower (MAL) MA intake was initiated from Taar1m1J/m1J F2 individuals; a control line of Taar1+/+ individuals (MAC) was retained. The lines were also examined for MA-induced locomotor and thermal responses, and fluid and tastant consumption. Taar1m1J/m1J F2 mice consumed significantly more MA than Taar1+/+ F2 mice. Response to selection was significant by generation 2 and there were corresponding differences in fluid consumed. Fluid consumption was not different in non-MA drinking studies. Taar1m1J/m1J genotype (MAL or MAH vs MAC mice) was associated with heighted MA locomotor and reduced hypothermic responses. MAL mice exhibited greater sensitization than MAH mice, but the selected lines did not consistently differ for thermal or tastant phenotypes. Residual variation among high-risk Taar1m1J/m1J mice appears to involve mechanisms associated with neuroadaptation to MA, but not sensitivity to hypothermic effects of MA.
Collapse
Affiliation(s)
- Cheryl Reed
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Alexandra M Stafford
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - John R K Mootz
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Harue Baba
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Jason Erk
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Tamara J Phillips
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA.,Veterans Affairs Portland Health Care System, Portland, Oregon, USA
| |
Collapse
|
12
|
Xu H, Das S, Sturgill M, Hodgkinson C, Yuan Q, Goldman D, Grasing K. Extracellular dopamine, acetylcholine, and activation of dopamine D1 and D2 receptors after selective breeding for cocaine self-administration in rats. Psychopharmacology (Berl) 2017; 234:2475-2487. [PMID: 28547130 PMCID: PMC5538921 DOI: 10.1007/s00213-017-4640-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 04/30/2017] [Indexed: 12/21/2022]
Abstract
RATIONALE The low self-administration (LS)/Kgras (LS) and high self-administration (HS)/Kgras (HS) rat lines were generated by selective breeding for low- and high-intravenous cocaine self-administration, respectively, from a common outbred Wistar stock (Crl:WI). This trait has remained stable after 13 generations of breeding. OBJECTIVE The objective of the present study is to compare cocaine preference, neurotransmitter release, and dopamine receptor activation in LS and HS rats. METHODS Levels of dopamine, acetylcholine, and cocaine were measured in the nucleus accumbens (NA) shell of HS and LS rats by tandem mass spectrometry of microdialysates. Cocaine-induced locomotor activity and conditioned-place preference were compared between LS and HS rats. RESULTS HS rats displayed greater conditioned-place preference scores compared to LS and reduced basal extracellular concentrations of dopamine and acetylcholine. However, patterns of neurotransmitter release did not differ between strains. Low-dose cocaine increased locomotor activity in LS rats, but not in HS animals, while high-dose cocaine augmented activity only in HS rats. Either dose of cocaine increased immunoreactivity for c-Fos in the NA shell of both strains, with greater elevations observed in HS rats. Activation identified by cells expressing both c-Fos and dopamine receptors was generally greater in the HS strain, with a similar pattern for both D1 and D2 dopamine receptors. CONCLUSIONS Diminished levels of dopamine and acetylcholine in the NA shell, with enhanced cocaine-induced expression of D1 and D2 receptors, are associated with greater rewarding effects of cocaine in HS rats and an altered dose-effect relationship for cocaine-induced locomotor activity.
Collapse
Affiliation(s)
- Haiyang Xu
- Substance Abuse Research Laboratory, Kansas City Veterans Affairs Medical Center, 4801 Linwood Boulevard, Kansas City, MO 64128
| | - Sasmita Das
- Molecular Bio-Nanotechnology, Imaging and Therapeutic Research Unit, Kansas City Veterans Affairs Medical Center, 4801 Linwood Boulevard, Kansas City, MO 64128,Division of Hematology and Oncology, Department of Medicine, University of Kansas School of Medicine, Kansas City, KS 66160
| | - Marc Sturgill
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| | | | - Qiaoping Yuan
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20852
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20852
| | - Kenneth Grasing
- Substance Abuse Research Laboratory, 151, Kansas City Veterans Affairs Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 64128, USA. .,Division of Clinical Pharmacology, Department of Medicine, University of Kansas School of Medicine, Kansas City, KS, 66160, USA.
| |
Collapse
|
13
|
Szumlinski KK, Lominac KD, Campbell RR, Cohen M, Fultz EK, Brown CN, Miller BW, Quadir SG, Martin D, Thompson AB, von Jonquieres G, Klugmann M, Phillips TJ, Kippin TE. Methamphetamine Addiction Vulnerability: The Glutamate, the Bad, and the Ugly. Biol Psychiatry 2017; 81:959-970. [PMID: 27890469 PMCID: PMC5391296 DOI: 10.1016/j.biopsych.2016.10.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/30/2016] [Accepted: 10/05/2016] [Indexed: 01/23/2023]
Abstract
BACKGROUND The high prevalence and severity of methamphetamine (MA) abuse demands greater neurobiological understanding of its etiology. METHODS We conducted immunoblotting and in vivo microdialysis procedures in MA high/low drinking mice, as well as in isogenic C57BL/6J mice that varied in their MA preference/taking, to examine the glutamate underpinnings of MA abuse vulnerability. Neuropharmacological and Homer2 knockdown approaches were also used in C57BL/6J mice to confirm the role for nucleus accumbens (NAC) glutamate/Homer2 expression in MA preference/aversion. RESULTS We identified a hyperglutamatergic state within the NAC as a biochemical trait corresponding with both genetic and idiopathic vulnerability for high MA preference and taking. We also confirmed that subchronic subtoxic MA experience elicits a hyperglutamatergic state within the NAC during protracted withdrawal, characterized by elevated metabotropic glutamate 1/5 receptor function and Homer2 receptor-scaffolding protein expression. A high MA-preferring phenotype was recapitulated by elevating endogenous glutamate within the NAC shell of mice and we reversed MA preference/taking by lowering endogenous glutamate and/or Homer2 expression within this subregion. CONCLUSIONS Our data point to an idiopathic, genetic, or drug-induced hyperglutamatergic state within the NAC as a mediator of MA addiction vulnerability.
Collapse
Affiliation(s)
- Karen K Szumlinski
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California; Molecular, Cellular and Developmental Biology, University of California at Santa Barbara, Santa Barbara, California.
| | - Kevin D Lominac
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| | - Rianne R Campbell
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| | - Matan Cohen
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| | - Elissa K Fultz
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| | - Chelsea N Brown
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| | - Bailey W Miller
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| | - Sema G Quadir
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| | - Douglas Martin
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| | - Andrew B Thompson
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California
| | - Georg von Jonquieres
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, New South Wales, Australia
| | - Matthias Klugmann
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, New South Wales, Australia
| | - Tamara J Phillips
- Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University; VA Portland Health Care System, Portland, Oregon
| | - Tod E Kippin
- Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California; Molecular, Cellular and Developmental Biology, University of California at Santa Barbara, Santa Barbara, California; Neuroscience Research Institute, and Institute for Collaborative Biotechnology, University of California at Santa Barbara, Santa Barbara, California
| |
Collapse
|
14
|
Ohia-Nwoko O, Haile CN, Kosten TA. Sex differences in the acute locomotor response to methamphetamine in BALB/c mice. Behav Brain Res 2017; 327:94-97. [PMID: 28359885 DOI: 10.1016/j.bbr.2017.03.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/15/2017] [Accepted: 03/21/2017] [Indexed: 02/06/2023]
Abstract
Women use methamphetamine more frequently than men and are more vulnerable to its negative psychological effects. Rodent models have been an essential tool for evaluating the sex-dependent effects of psychostimulants; however, evidence of sex differences in the behavioral responses to methamphetamine in mice is lacking. In the present study, we investigated acute methamphetamine-induced (1mg/kg and 4mg/kg) locomotor activation in female and male BALB/c mice. We also evaluated whether basal locomotor activity was associated with the methamphetamine-induced locomotor response. The results indicated that female BALB/c mice displayed enhanced methamphetamine-induced locomotor activity compared to males, while basal locomotor activity was positively correlated with methamphetamine-induced activity in males, but not females. This study is the first to show sex-dependent locomotor effects of methamphetamine in BALB/c mice. Our observations emphasize the importance of considering sex when assessing behavioral responses to methamphetamine.
Collapse
Affiliation(s)
- Odochi Ohia-Nwoko
- University of Houston, Department of Psychology, Houston, TX 77204-6022, United States; Texas Institute for Measurement, Evaluation and Statistics (TIMES), Houston, TX 77204-6022, United States
| | - Colin N Haile
- University of Houston, Department of Psychology, Houston, TX 77204-6022, United States; Texas Institute for Measurement, Evaluation and Statistics (TIMES), Houston, TX 77204-6022, United States
| | - Therese A Kosten
- University of Houston, Department of Psychology, Houston, TX 77204-6022, United States; Texas Institute for Measurement, Evaluation and Statistics (TIMES), Houston, TX 77204-6022, United States.
| |
Collapse
|
15
|
Kaniuga E, Taracha E, Stępień T, Wierzba-Bobrowicz T, Płaźnik A, Chrapusta SJ. Rats showing low and high sensitization of frequency-modulated 50-kHz vocalization response to amphetamine differ in amphetamine-induced brain Fos expression. Brain Res 2016; 1648:356-364. [DOI: 10.1016/j.brainres.2016.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/18/2016] [Accepted: 08/05/2016] [Indexed: 12/27/2022]
|
16
|
Vitamin D3: A Role in Dopamine Circuit Regulation, Diet-Induced Obesity, and Drug Consumption. eNeuro 2016; 3:eN-NWR-0122-15. [PMID: 27257625 PMCID: PMC4875352 DOI: 10.1523/eneuro.0122-15.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 04/27/2016] [Accepted: 05/03/2016] [Indexed: 12/31/2022] Open
Abstract
The influence of micronutrients on dopamine systems is not well defined. Using mice, we show a potential role for reduced dietary vitamin D3 (cholecalciferol) in promoting diet-induced obesity (DIO), food intake, and drug consumption while on a high fat diet. To complement these deficiency studies, treatments with exogenous fully active vitamin D3 (calcitriol, 10 µg/kg, i.p.) were performed. Nondeficient mice that were made leptin resistant with a high fat diet displayed reduced food intake and body weight after an acute treatment with exogenous calcitriol. Dopamine neurons in the midbrain and their target neurons in the striatum were found to express vitamin D3 receptor protein. Acute calcitriol treatment led to transcriptional changes of dopamine-related genes in these regions in naive mice, enhanced amphetamine-induced dopamine release in both naive mice and rats, and increased locomotor activity after acute amphetamine treatment (2.5 mg/kg, i.p.). Alternatively, mice that were chronically fed either the reduced D3 high fat or chow diets displayed less activity after acute amphetamine treatment compared with their respective controls. Finally, high fat deficient mice that were trained to orally consume liquid amphetamine (90 mg/L) displayed increased consumption, while nondeficient mice treated with calcitriol showed reduced consumption. Our findings suggest that reduced dietary D3 may be a contributing environmental factor enhancing DIO as well as drug intake while eating a high fat diet. Moreover, these data demonstrate that dopamine circuits are modulated by D3 signaling, and may serve as direct or indirect targets for exogenous calcitriol.
Collapse
|
17
|
Taracha E, Kaniuga E, Wyszogrodzka E, Płaźnik A, Stefański R, Chrapusta SJ. Poor sensitization of 50-kHz vocalization response to amphetamine predicts rat susceptibility to self-administration of the drug. Psychopharmacology (Berl) 2016; 233:2827-40. [PMID: 27256355 PMCID: PMC4917579 DOI: 10.1007/s00213-016-4328-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 05/09/2016] [Indexed: 11/27/2022]
Abstract
RATIONALE Our previous studies showed promise for using sensitization of the frequency-modulated 50-kHz vocalization response to amphetamine (AMPH) as an index of rat vulnerability to AMPH addiction. OBJECTIVE This study aimed to test the utility of sensitizing frequency-modulated (FM) 50-kHz vocalization in the AMPH self-administration paradigm as well as the ability of N-acetylcysteine to prevent self-administration relapse. METHODS Rats were subjected to the so-called two-injection protocol of sensitization (TIPS) using AMPH and were categorized as low-sensitized callers (LCTIPS) or high-sensitized callers (HCTIPS) based on the individual outcomes. Then, they were given 44 sessions of AMPH self-administration followed by a 17-session N-acetylcysteine-aided extinction course and a single session of AMPH-primed self-administration reinstatement. RESULTS LCTIPS compared to HCTIPS rats showed no considerable difference in the FM 50-kHz vocalization rate during the self-administration training or extinction course, but they were considerably more likely to acquire AMPH self-administration and experience drug-induced reinstatement of this trait. Moreover, the LCTIPS rats were more likely than HCTIPS rats to have a markedly higher FM 50-kHz vocalization rate after AMPH reinstatement. N-acetylcysteine did not affect the course of self-administration extinction or the instrumental or FM 50-kHz vocalization responses to AMPH reinstatement. CONCLUSIONS There is no link between the FM 50-kHz vocalization and key characteristics of AMPH self-administration. Additionally, N-acetylcysteine does not help prevent AMPH self-administration relapse. However, there is a high predictive value for poor sensitization of the FM 50-kHz vocalization response to AMPH with respect to the acquisition and maintenance of self-administration of this psychostimulant.
Collapse
Affiliation(s)
- Ewa Taracha
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego St., 02-957, Warsaw, Poland.
| | - Ewelina Kaniuga
- />Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego St., 02-957 Warsaw, Poland
| | - Edyta Wyszogrodzka
- />Department of Pharmacology and Physiology of the Nervous System, Institute of Psychiatry and Neurology, 9 Sobieskiego St., 02-957 Warsaw, Poland
| | - Adam Płaźnik
- />Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego St., 02-957 Warsaw, Poland , />Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, 26/28 Krakowskie Przedmieście St., 00-927 Warsaw, Poland
| | - Roman Stefański
- />Department of Pharmacology and Physiology of the Nervous System, Institute of Psychiatry and Neurology, 9 Sobieskiego St., 02-957 Warsaw, Poland
| | - Stanisław J. Chrapusta
- />Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland
| |
Collapse
|
18
|
Phillips TJ, Shabani S. An animal model of differential genetic risk for methamphetamine intake. Front Neurosci 2015; 9:327. [PMID: 26441502 PMCID: PMC4585292 DOI: 10.3389/fnins.2015.00327] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/31/2015] [Indexed: 11/13/2022] Open
Abstract
The question of whether genetic factors contribute to risk for methamphetamine (MA) use and dependence has not been intensively investigated. Compared to human populations, genetic animal models offer the advantages of control over genetic family history and drug exposure. Using selective breeding, we created lines of mice that differ in genetic risk for voluntary MA intake and identified the chromosomal addresses of contributory genes. A quantitative trait locus was identified on chromosome 10 that accounts for more than 50% of the genetic variance in MA intake in the selected mouse lines. In addition, behavioral and physiological screening identified differences corresponding with risk for MA intake that have generated hypotheses that are testable in humans. Heightened sensitivity to aversive and certain physiological effects of MA, such as MA-induced reduction in body temperature, are hallmarks of mice bred for low MA intake. Furthermore, unlike MA-avoiding mice, MA-preferring mice are sensitive to rewarding and reinforcing MA effects, and to MA-induced increases in brain extracellular dopamine levels. Gene expression analyses implicate the importance of a network enriched in transcription factor genes, some of which regulate the mu opioid receptor gene, Oprm1, in risk for MA use. Neuroimmune factors appear to play a role in differential response to MA between the mice bred for high and low intake. In addition, chromosome 10 candidate gene studies provide strong support for a trace amine-associated receptor 1 gene, Taar1, polymorphism in risk for MA intake. MA is a trace amine-associated receptor 1 (TAAR1) agonist, and a non-functional Taar1 allele segregates with high MA consumption. Thus, reduced TAAR1 function has the potential to increase risk for MA use. Overall, existing findings support the MA drinking lines as a powerful model for identifying genetic factors involved in determining risk for harmful MA use. Future directions include the development of a binge model of MA intake, examining the effect of withdrawal from chronic MA on MA intake, and studying potential Taar1 gene × gene and gene × environment interactions. These and other studies are intended to improve our genetic model with regard to its translational value to human addiction.
Collapse
Affiliation(s)
- Tamara J Phillips
- VA Portland Health Care System Portland, OR, USA ; Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University Portland, OR, USA
| | | |
Collapse
|
19
|
Kamens HM, Miyamoto J, Powers MS, Ro K, Soto M, Cox R, Stitzel JA, Ehringer MA. The β3 subunit of the nicotinic acetylcholine receptor: Modulation of gene expression and nicotine consumption. Neuropharmacology 2015; 99:639-49. [PMID: 26318101 DOI: 10.1016/j.neuropharm.2015.08.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/20/2015] [Accepted: 08/21/2015] [Indexed: 02/06/2023]
Abstract
Genetic factors explain approximately half of the variance in smoking behaviors, but the molecular mechanism by which genetic variation influences behavior is poorly understood. SNPs in the putative promoter region of CHRNB3, the gene that encodes the β3 subunit of the nicotinic acetylcholine receptor (nAChR), have been repeatedly associated with nicotine behaviors. In this work we sought to identify putative function of three SNPs in the promoter region of CHRNB3 on in vitro gene expression. Additionally, we used β3 null mutant mice as a model of reduced gene expression to assess the effects on nicotine behaviors. The effect of rs13277254, rs6474413, and rs4950 on reporter gene expression was examined using a luciferase reporter assay. A major and minor parent haplotype served as the background on which alleles at the three SNPs were flipped onto different backgrounds (e.g. minor allele on major haplotype background). Constructs were tested in three human cell lines: BE(2)-C, SH-SY5Y and HEK293T. In all cell types the major haplotype led to greater reporter gene expression compared to the minor haplotype, and results indicate that this effect is driven by rs6474413. Moreover, mice lacking the β3 subunit showed reduced voluntary nicotine consumption compared that of wildtype animals. These data provide evidence that the protective genetic variant at rs6474413 identified in human genetic studies reduces gene expression and that decreased β3 gene expression in mice reduces nicotine intake. This work contributes to our understanding of the molecular mechanisms that contribute to the human genetic associations of tobacco behaviors.
Collapse
Affiliation(s)
- Helen M Kamens
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Jill Miyamoto
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80303, USA
| | - Matthew S Powers
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80303, USA
| | - Kasey Ro
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80303, USA
| | - Marissa Soto
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80303, USA
| | - Ryan Cox
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80303, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80303, USA; Department of Integrative Physiology, University of Colorado, Boulder, CO 80303, USA
| | - Marissa A Ehringer
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80303, USA; Department of Integrative Physiology, University of Colorado, Boulder, CO 80303, USA
| |
Collapse
|
20
|
Harkness JH, Shi X, Janowsky A, Phillips TJ. Trace Amine-Associated Receptor 1 Regulation of Methamphetamine Intake and Related Traits. Neuropsychopharmacology 2015; 40:2175-84. [PMID: 25740289 PMCID: PMC4613607 DOI: 10.1038/npp.2015.61] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/17/2015] [Accepted: 03/03/2015] [Indexed: 12/17/2022]
Abstract
Continued methamphetamine (MA) use is dependent on a positive MA experience and is likely attenuated by sensitivity to the aversive effects of MA. Bidirectional selective breeding of mice for high (MAHDR) or low (MALDR) voluntary consumption of MA demonstrates a genetic influence on MA intake. Quantitative trait locus (QTL) mapping identified a QTL on mouse chromosome 10 that accounts for greater than 50% of the genetically-determined differences in MA intake in the MAHDR and MALDR lines. The trace amine-associated receptor 1 gene (Taar1) is within the confidence interval of the QTL and encodes a receptor (TAAR1) that modulates monoamine neurotransmission and at which MA serves as an agonist. We demonstrate the existence of a non-functional allele of Taar1 in the DBA/2J mouse strain, one of the founder strains of the selected lines, and show that this non-functional allele co-segregates with high MA drinking and with reduced sensitivity to MA-induced conditioned taste aversion (CTA) and hypothermia. The functional Taar1 allele, derived from the other founder strain, C57BL/6J, segregates with low MA drinking and heightened sensitivity to MA-induced CTA and hypothermia. A role for TAAR1 in these phenotypes is corroborated in Taar1 transgenic mice: Taar1 knockout mice consume more MA and exhibit insensitivity to MA-induced CTA and hypothermia, compared with Taar1 wild-type mice. These are the first data to show that voluntary MA consumption is, in part, regulated by TAAR1 function. Behavioral and physiological studies indicate that TAAR1 function increases sensitivity to aversive effects of MA, and may thereby protect against MA use.
Collapse
Affiliation(s)
- John H Harkness
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Xiao Shi
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA,Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA
| | - Aaron Janowsky
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA,Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA,Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Tamara J Phillips
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA,Veterans Affairs Portland Health Care System, Portland, OR, USA,Veterans Affairs Portland Health Care System, R&D 32, 3710 SW US Veterans Hospital Road, Portland, OR 97239, USA, Tel: +1 503 220 8262 ex: 56674, Fax: +1 503 721 1029, E-mail:
| |
Collapse
|
21
|
Horton WJ, Gissel HJ, Saboy JE, Wright KP, Stitzel JA. Melatonin administration alters nicotine preference consumption via signaling through high-affinity melatonin receptors. Psychopharmacology (Berl) 2015; 232:2519-30. [PMID: 25704105 PMCID: PMC4482784 DOI: 10.1007/s00213-015-3886-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 02/08/2015] [Indexed: 12/13/2022]
Abstract
RATIONALE While it is known that tobacco use varies across the 24-h day, the time-of-day effects are poorly understood. Findings from several previous studies indicate a potential role for melatonin in these time-of-day effects; however, the specific underlying mechanisms have not been well characterized. Understanding of these mechanisms may lead to potential novel smoking cessation treatments. OBJECTIVE The objective of this study is examine the role of melatonin and melatonin receptors in nicotine free-choice consumption METHODS A two-bottle oral nicotine choice paradigm was utilized with melatonin supplementation in melatonin-deficient mice (C57BL/6J) or without melatonin supplementation in mice proficient at melatonin synthesis (C3H/Ibg) compared to melatonin-proficient mice lacking both or one of the high-affinity melatonin receptors (MT1 and MT2; double-null mutant DM, or MT1 or MT2). Preference for bitter and sweet tastants also was assessed in wild-type and MT1 and MT2 DM mice. Finally, home cage locomotor monitoring was performed to determine the effect of melatonin administration on activity patterns. RESULTS Supplemental melatonin in drinking water significantly reduced free-choice nicotine consumption in C57BL/6J mice, which do not produce endogenous melatonin, while not altering activity patterns. Independently, genetic deletion of both MT1 and MT2 receptors in a melatonin-proficient mouse strain (C3H) resulted in significantly more nicotine consumption than controls. However, single genetic deletion of either the MT1 or MT2 receptor alone did not result in increased nicotine consumption. Deletion of MT1 and MT2 did not impact taste preference. CONCLUSIONS This study demonstrates that nicotine consumption can be affected by exogenous or endogenous melatonin and requires at least one of the high-affinity melatonin receptors. The fact that expression of either the MT1 or MT2 melatonin receptor is sufficient to maintain lower nicotine consumption suggests functional overlap and potential mechanistic explanations.
Collapse
Affiliation(s)
- William J. Horton
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80303,Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303
| | - Hannah J. Gissel
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80303
| | - Jennifer E. Saboy
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80303
| | - Kenneth P. Wright
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303
| | - Jerry A. Stitzel
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, 80303,Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303
| |
Collapse
|
22
|
Kesby JP, Hubbard DT, Markou A, Semenova S. Expression of HIV gp120 protein increases sensitivity to the rewarding properties of methamphetamine in mice. Addict Biol 2014; 19:593-605. [PMID: 23252824 DOI: 10.1111/adb.12023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Methamphetamine abuse and human immunodeficiency virus (HIV) infection induce neuropathological changes in corticolimbic brain areas involved in reward and cognitive function. Little is known about the combined effects of methamphetamine and HIV infection on cognitive and reward processes. The HIV/gp120 protein induces neurodegeneration in mice, similar to HIV-induced pathology in humans. We investigated the effects of gp120 expression on associative learning, preference for methamphetamine and non-drug reinforcers, and sensitivity to the conditioned rewarding properties of methamphetamine in transgenic (tg) mice expressing HIV/gp120 protein (gp120-tg). gp120-tg mice learned the operant response for food at the same rate as non-tg mice. In the two-bottle choice procedure with restricted access to drugs, gp120-tg mice exhibited greater preference for methamphetamine and saccharin than non-tg mice, whereas preference for quinine was similar between genotypes. Under conditions of unrestricted access to methamphetamine, the mice exhibited a decreased preference for increasing methamphetamine concentrations. However, male gp120-tg mice showed a decreased preference for methamphetamine at lower concentrations than non-tg male mice. gp120-tg mice developed methamphetamine-induced conditioned place preference at lower methamphetamine doses compared with non-tg mice. No differences in methamphetamine pharmacokinetics were found between genotypes. These results indicate that gp120-tg mice exhibit no deficits in associative learning or reward/motivational function for a natural reinforcer. Interestingly, gp120 expression resulted in increased preference for methamphetamine and a highly palatable non-drug reinforcer (saccharin) and increased sensitivity to methamphetamine-induced conditioned reward. These data suggest that HIV-positive individuals may have increased sensitivity to methamphetamine, leading to high methamphetamine abuse potential in this population.
Collapse
Affiliation(s)
- James P. Kesby
- Department of Psychiatry; School of Medicine; University of California San Diego; La Jolla CA USA
| | - David T. Hubbard
- Department of Psychiatry; School of Medicine; University of California San Diego; La Jolla CA USA
| | - Athina Markou
- Department of Psychiatry; School of Medicine; University of California San Diego; La Jolla CA USA
| | - Svetlana Semenova
- Department of Psychiatry; School of Medicine; University of California San Diego; La Jolla CA USA
| |
Collapse
|
23
|
Belknap JK, McWeeney S, Reed C, Burkhart-Kasch S, McKinnon CS, Li N, Baba H, Scibelli AC, Hitzemann R, Phillips TJ. Genetic factors involved in risk for methamphetamine intake and sensitization. Mamm Genome 2013; 24:446-58. [PMID: 24217691 PMCID: PMC3880562 DOI: 10.1007/s00335-013-9484-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/11/2013] [Indexed: 11/26/2022]
Abstract
Lines of mice were created by selective breeding for the purpose of identifying genetic mechanisms that influence the magnitude of the selected trait and to explore genetic correlations for additional traits thought to be influenced by shared mechanisms. DNA samples from high and low methamphetamine-drinking (MADR) and high and low methamphetamine-sensitization lines were used for quantitative trait locus (QTL) mapping. Significant additive genetic correlations between the two traits indicated a common genetic influence, and a QTL on chromosome X was detected for both traits, suggesting one source of this commonality. For MADR mice, a QTL on chromosome 10 accounted for more than 50 % of the genetic variance in that trait. Microarray gene expression analyses were performed for three brain regions for methamphetamine-naïve MADR line mice: nucleus accumbens, prefrontal cortex, and ventral midbrain. Many of the genes that were differentially expressed between the high and low MADR lines were shared in common across the three brain regions. A gene network highly enriched in transcription factor genes was identified as being relevant to genetically determined differences in methamphetamine intake. When the mu opioid receptor gene (Oprm1), located on chromosome 10 in the QTL region, was added to this top-ranked transcription factor network, it became a hub in the network. These data are consistent with previously published findings of opioid response and intake differences between the MADR lines and suggest that Oprm1, or a gene that impacts activity of the opioid system, plays a role in genetically determined differences in methamphetamine intake.
Collapse
Affiliation(s)
- John K. Belknap
- Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239 USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Shannon McWeeney
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR, 97239 USA
- Division of Biostatistics of Public Health & Preventative Medicine, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Cheryl Reed
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Sue Burkhart-Kasch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Carrie S. McKinnon
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Na Li
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Harue Baba
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Angela C. Scibelli
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Robert Hitzemann
- Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239 USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Tamara J. Phillips
- Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239 USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| |
Collapse
|
24
|
Darlington TM, McCarthy RD, Cox RJ, Ehringer MA. Mesolimbic transcriptional response to hedonic substitution of voluntary exercise and voluntary ethanol consumption. Behav Brain Res 2013; 259:313-20. [PMID: 24239693 DOI: 10.1016/j.bbr.2013.11.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 10/17/2013] [Accepted: 11/06/2013] [Indexed: 12/14/2022]
Abstract
The mesolimbic dopaminergic pathway has been implicated in many rewarding behaviors, including the consumption of ethanol and voluntary exercise. It has become apparent that different rewarding stimuli activate this pathway, and therefore it is possible for these behaviors to influence each other, i.e. hedonic substitution. Using adult female C57BL/6J mice, we demonstrate that voluntary access to a running wheel substantially reduces the consumption and preference of ethanol. Furthermore, we examined gene expression of several genes involved in regulating the mesolimbic dopaminergic pathway, which we hypothesized to be the main pathway involved in hedonic substitution. In the striatum, we observed a reduction in mRNA expression of Drd1a due to exercise. Hippocampal Bdnf mRNA increased in response to exercise and decreased in response to ethanol. Furthermore, there was an interaction effect of exercise and ethanol on the expression of Slc18a2 in the midbrain. These data suggest an important role for this pathway, and especially for Bdnf and Slc18a2 in regulating hedonic substitution.
Collapse
Affiliation(s)
- Todd M Darlington
- Institute for Behavioral Genetics, Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO 80303, USA
| | - Riley D McCarthy
- Institute for Behavioral Genetics, Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO 80303, USA
| | - Ryan J Cox
- Institute for Behavioral Genetics, Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO 80303, USA
| | - Marissa A Ehringer
- Institute for Behavioral Genetics, Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO 80303, USA.
| |
Collapse
|
25
|
Methamphetamine increases locomotion and dopamine transporter activity in dopamine d5 receptor-deficient mice. PLoS One 2013; 8:e75975. [PMID: 24155877 PMCID: PMC3796526 DOI: 10.1371/journal.pone.0075975] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 08/19/2013] [Indexed: 11/24/2022] Open
Abstract
Dopamine regulates the psychomotor stimulant activities of amphetamine-like substances in the brain. The effects of dopamine are mediated through five known dopamine receptor subtypes in mammals. The functional relevance of D5 dopamine receptors in the central nervous system is not well understood. To determine the functional relevance of D5 dopamine receptors, we created D5 dopamine receptor-deficient mice and then used these mice to assess the roles of D5 dopamine receptors in the behavioral response to methamphetamine. Interestingly, D5 dopamine receptor-deficient mice displayed increased ambulation in response to methamphetamine. Furthermore, dopamine transporter threonine phosphorylation levels, which regulate amphetamine-induced dopamine release, were elevated in D5 dopamine receptor-deficient mice. The increase in methamphetamine-induced locomotor activity was eliminated by pretreatment with the dopamine transporter blocker GBR12909. Taken together, these results suggest that dopamine transporter activity and threonine phosphorylation levels are regulated by D5 dopamine receptors.
Collapse
|
26
|
Gubner NR, Reed C, McKinnon CS, Phillips TJ. Unique genetic factors influence sensitivity to the rewarding and aversive effects of methamphetamine versus cocaine. Behav Brain Res 2013; 256:420-7. [PMID: 23994231 DOI: 10.1016/j.bbr.2013.08.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 08/13/2013] [Accepted: 08/19/2013] [Indexed: 11/29/2022]
Abstract
Genetic factors significantly influence addiction-related phenotypes. This is supported by the successful bidirectional selective breeding of two replicate sets of mouse lines for amount of methamphetamine consumed. Some of the same genetic factors that influence methamphetamine consumption have been previously found also to influence sensitivity to the conditioned rewarding and aversive effects of methamphetamine. The goal of the current studies was to determine if some of the same genetic factors influence sensitivity to the conditioned rewarding and aversive effects of cocaine. Cocaine conditioned reward was examined in methamphetamine high drinking and low drinking line mice using a conditioned place preference procedure and cocaine conditioned aversion was measured using a conditioned taste aversion procedure. In addition, a general sensitivity measure, locomotor stimulant response to cocaine, was assessed in these lines; previous data indicated no difference between the selected lines in sensitivity to methamphetamine-induced stimulation. In contrast to robust differences for methamphetamine, the methamphetamine high and low drinking lines did not differ in sensitivity to either the rewarding or aversive effects of cocaine. They also exhibited comparable sensitivity to cocaine-induced locomotor stimulation. These data suggest that the genetic factors that influence sensitivity to the conditioned rewarding and aversive effects of methamphetamine in these lines of mice do not influence sensitivity to these effects of cocaine. Thus, different genetic factors may influence risk for methamphetamine versus cocaine use.
Collapse
Affiliation(s)
- Noah R Gubner
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | | | | |
Collapse
|
27
|
Neve KA, Ford CP, Buck DC, Grandy DK, Neve RL, Phillips TJ. Normalizing dopamine D2 receptor-mediated responses in D2 null mutant mice by virus-mediated receptor restoration: comparing D2L and D2S. Neuroscience 2013; 248:479-87. [PMID: 23811070 DOI: 10.1016/j.neuroscience.2013.06.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 05/31/2013] [Accepted: 06/18/2013] [Indexed: 11/29/2022]
Abstract
D2 receptor null mutant (Drd2(-/-)) mice have altered responses to the rewarding and locomotor effects of psychostimulant drugs, which is evidence of a necessary role for D2 receptors in these behaviors. Furthermore, work with mice that constitutively express only the D2 receptor short form (D2S), as a result of genetic deletion of the long form (D2L), provides the basis for a current model in which D2L is thought to be the postsynaptic D2 receptor on medium spiny neurons in the basal forebrain, and D2S the autoreceptor that regulates the activity of dopamine neurons and dopamine synthesis and release. Because constitutive genetic deletion of the D2 or D2L receptor may cause compensatory changes that influence functional outcomes, our approach is to identify aspects of the abnormal phenotype of a Drd2(-/-) mouse that can be normalized by virus-mediated D2 receptor expression. Drd2(-/-) mice are deficient in basal and methamphetamine-induced locomotor activation and lack D2 receptor agonist-induced activation of G protein-regulated inward rectifying potassium channels (GIRKs) in dopaminergic neurons. Here we show that virus-mediated expression of D2L in the nucleus accumbens significantly restored methamphetamine-induced locomotor activation, but not basal locomotor activity, compared to mice receiving the control virus. It also restored the effect of methamphetamine to decrease time spent in the center of the activity chamber in female but not male Drd2(-/-) mice. Furthermore, the effect of expression of D2S was indistinguishable from D2L. Similarly, virus-mediated expression of either D2S or D2L in substantia nigra neurons restored D2 agonist-induced activation of GIRKs. In this acute expression system, the alternatively spliced forms of the D2 receptor appear to be equally capable of acting as postsynaptic receptors and autoreceptors.
Collapse
Affiliation(s)
- K A Neve
- Research Service, Portland VA Medical Center, and Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR 97239, USA.
| | - C P Ford
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - D C Buck
- Research Service, Portland VA Medical Center, and Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR 97239, USA.
| | - D K Grandy
- Department of Physiology & Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA.
| | - R L Neve
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA.
| | - T J Phillips
- Research Service, Portland VA Medical Center, and Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
28
|
Eshleman AJ, Wolfrum KM, Hatfield MG, Johnson RA, Murphy KV, Janowsky A. Substituted methcathinones differ in transporter and receptor interactions. Biochem Pharmacol 2013; 85:1803-15. [PMID: 23583454 DOI: 10.1016/j.bcp.2013.04.004] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/03/2013] [Accepted: 04/03/2013] [Indexed: 12/31/2022]
Abstract
The use of synthetic methcathinones, components of "bath salts," is a world-wide health concern. These compounds, structurally similar to methamphetamine (METH) and 3,4-methylendioxymethamphetamine (MDMA), cause tachycardia, hallucinations and psychosis. We hypothesized that these potentially neurotoxic and abused compounds display differences in their transporter and receptor interactions as compared to amphetamine counterparts. 3,4-Methylenedioxypyrovalerone and naphyrone had high affinity for radioligand binding sites on recombinant human dopamine (hDAT), serotonin (hSERT) and norepinephrine (hNET) transporters, potently inhibited [³H]neurotransmitter uptake, and, like cocaine, did not induce transporter-mediated release. Butylone was a lower affinity uptake inhibitor. In contrast, 4-fluoromethcathinone, mephedrone and methylone had higher inhibitory potency at uptake compared to binding and generally induced release of preloaded [³H]neurotransmitter from hDAT, hSERT and hNET (highest potency at hNET), and thus are transporter substrates, similar to METH and MDMA. At hNET, 4-fluoromethcathinone was a more efficacious releaser than METH. These substituted methcathinones had low uptake inhibitory potency and low efficacy at inducing release via human vesicular monoamine transporters (hVMAT2). These compounds were low potency (1) h5-HT(1A) receptor partial agonists, (2) h5-HT(2A) receptor antagonists, (3) weak h5-HT(2C) receptor antagonists. This is the first report on aspects of substituted methcathinone efficacies at serotonin (5-HT) receptors and in superfusion release assays. Additionally, the drugs had no affinity for dopamine receptors, and high-nanomolar to mid-micromolar affinity for hSigma1 receptors. Thus, direct interactions with hVMAT2 and serotonin, dopamine, and hSigma1 receptors may not explain psychoactive effects. The primary mechanisms of action may be as inhibitors or substrates of DAT, SERT and NET.
Collapse
Affiliation(s)
- Amy J Eshleman
- Research Service, Veterans Affairs Medical Center, Portland, OR, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Hart AB, de Wit H, Palmer AA. Candidate gene studies of a promising intermediate phenotype: failure to replicate. Neuropsychopharmacology 2013; 38:802-16. [PMID: 23303064 PMCID: PMC3671998 DOI: 10.1038/npp.2012.245] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 11/07/2012] [Accepted: 11/26/2012] [Indexed: 11/08/2022]
Abstract
Many candidate gene studies use 'intermediate phenotypes' instead of disease diagnoses. It has been proposed that intermediate phenotypes have simpler genetic architectures such that individual alleles account for a larger percentage of trait variance. This implies that smaller samples can be used to identify genetic associations. Pharmacogenomic drug challenge studies may be an especially promising class of intermediate phenotype. We previously conducted a series of 12 candidate gene analyses of acute subjective and physiological responses to amphetamine in 99-162 healthy human volunteers (ADORA2A, SLC6A3, BDNF, SLC6A4, CSNK1E, SLC6A2, DRD2, FAAH, COMT, OPRM1). Here, we report our attempt to replicate these findings in over 200 additional participants ascertained using identical methodology. We were unable to replicate any of our previous findings. These results raise critical issues related to non-replication of candidate gene studies, such as power, sample size, multiple testing within and between studies, publication bias and the expectation that true allelic effect sizes are similar to those reported in genome-wide association studies. Many of these factors may have contributed to our failure to replicate our previous findings. Our results should instill caution in those considering similarly designed studies.
Collapse
Affiliation(s)
- Amy B Hart
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - Abraham A Palmer
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| |
Collapse
|
30
|
Gubner NR, McKinnon CS, Reed C, Phillips TJ. Accentuating effects of nicotine on ethanol response in mice with high genetic predisposition to ethanol-induced locomotor stimulation. Drug Alcohol Depend 2013; 127:108-14. [PMID: 22795175 PMCID: PMC3505243 DOI: 10.1016/j.drugalcdep.2012.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 06/12/2012] [Accepted: 06/14/2012] [Indexed: 01/12/2023]
Abstract
BACKGROUND Co-morbid use of nicotine-containing tobacco products and alcohol is prevalent in alcohol dependent individuals. Common genetic factors could influence initial sensitivity to the independent or interactive effects of these drugs and play a role in their co-abuse. METHODS Locomotor sensitivity to nicotine and ethanol, alone and in combination, was assessed in mice bred for high (FAST) and low (SLOW) sensitivity to the locomotor stimulant effects of ethanol and in an inbred strain of mouse (DBA/2J) that has been shown to have extreme sensitivity to ethanol-induced stimulation in comparison to other strains. RESULTS The effects of nicotine and ethanol, alone and in combination, were dependent on genotype. In FAST and DBA/2J mice that show high sensitivity to ethanol-induced stimulation, nicotine accentuated the locomotor stimulant response to ethanol. This effect was not found in SLOW mice that are not stimulated by ethanol alone. CONCLUSIONS These data indicate that genes underlying differential sensitivity to the stimulant effects of ethanol alone also influence sensitivity to nicotine in combination with ethanol. Sensitivity to the stimulant effects of nicotine alone does not appear to predict the response to the drug combination, as FAST mice are sensitive to nicotine-induced stimulation, whereas SLOW and DBA/2J mice are not. The combination of nicotine and ethanol may have genotype-dependent effects that could impact co-abuse liability.
Collapse
Affiliation(s)
- N R Gubner
- Department of Behavioral Neuroscience and Portland Alcohol Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | | | | |
Collapse
|
31
|
Harkness JH, Hitzemann RJ, Edmunds S, Phillips TJ. Effects of sodium butyrate on methamphetamine-sensitized locomotor activity. Behav Brain Res 2012; 239:139-47. [PMID: 23137698 DOI: 10.1016/j.bbr.2012.10.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 10/26/2012] [Accepted: 10/29/2012] [Indexed: 10/27/2022]
Abstract
Neuroadaptations associated with behavioral sensitization induced by repeated exposure to methamphetamine (MA) appear to be involved in compulsive drug pursuit and use. Increased histone acetylation, an epigenetic effect resulting in altered gene expression, may promote sensitized responses to psychostimulants. The role of histone acetylation in the expression and acquisition of MA-induced locomotor sensitization was examined by measuring the effect of histone deacetylase inhibition by sodium butyrate (NaB). For the effect on expression, mice were treated repeatedly with MA (10 days of 2mg/kg MA) or saline (10 days), and then vehicle or NaB (630 mg/kg, intraperitoneally) was administered 30 min prior to MA challenge and locomotor response was measured. NaB treatment increased the locomotor response to MA in both acutely MA treated and sensitized animals. For acquisition, NaB was administered 30 min prior to each MA exposure (10 days of 1 or 2mg/kg), but not prior to the MA challenge test. Treatment with NaB during the sensitization acquisition period significantly increased locomotor activation by MA in sensitized mice only. NaB alone did not significantly alter locomotor activity. Acute NaB or MA, but not the combination, increased striatal acetylation at histone H4. Repeated treatment with MA, but not NaB or MA plus NaB, increased striatal acetylation at histone H3. Although increased histone acetylation may alter the expression of genes involved in acute locomotor response to MA and in the acquisition of MA-induced sensitization, results for acetylation at H3 and H4 showed little correspondence with behavior.
Collapse
Affiliation(s)
- John H Harkness
- Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, L470, Portland, OR 97239-3098, USA.
| | | | | | | |
Collapse
|
32
|
de Wit H, Phillips TJ. Do initial responses to drugs predict future use or abuse? Neurosci Biobehav Rev 2012; 36:1565-76. [PMID: 22542906 PMCID: PMC3372699 DOI: 10.1016/j.neubiorev.2012.04.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 03/09/2012] [Accepted: 04/12/2012] [Indexed: 10/28/2022]
Abstract
Individuals vary in their initial reactions to drugs of abuse in ways that may contribute to the likelihood of subsequent drug use. In humans, most drugs of abuse produce positive subjective states such as euphoria and feelings of well-being, which may facilitate repeated use. In nonhumans, many drugs initially increase locomotor activity and produce discriminative stimulus effects, both of which have been considered to be models of human stimulant and subjective states. Both humans and nonhumans vary in their sensitivity to early acute drug effects in ways that may predict future use or self-administration, and some of these variations appear to be genetic in origin. However, it is not known exactly how the initial responses to drugs in either humans or nonhumans relate to subsequent use or abuse. In humans, positive effects of drugs facilitate continued use of a drug while negative effects discourage use, and in nonhumans, greater genetic risk for drug intake is predicted by reduced sensitivity to drug aversive effects; but whether these initial responses affect escalation of drug use, and the development of dependence is currently unknown. Although early use of a drug is a necessary step in the progression to abuse and dependence, other variables may be of greater importance in the transition from use to abuse. Alternatively, the same variables that predict initial acute drug effects and early use may significantly contribute to continued use, escalation and dependence. Here we review the existing evidence for relations between initial direct drug effects, early use, and continued use. Ultimately, these relations can only be determined from systematic longitudinal studies with comprehensive assessments from early drug responses to progression of problem drug use. In parallel, additional investigation of initial responses in animal models as predictors of drug use will shed light on the underlying mechanisms.
Collapse
Affiliation(s)
- Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, 5841 S Maryland Ave, MC3077, Chicago, IL 60637, United States.
| | | |
Collapse
|
33
|
Branch SY, Beckstead MJ. Methamphetamine produces bidirectional, concentration-dependent effects on dopamine neuron excitability and dopamine-mediated synaptic currents. J Neurophysiol 2012; 108:802-9. [PMID: 22592307 DOI: 10.1152/jn.00094.2012] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Amphetamine-like compounds are commonly used to enhance cognition and to treat attention deficit/hyperactivity disorder, but they also function as positive reinforcers and are self-administered at doses far exceeding clinical relevance. Many of these compounds (including methamphetamine) are substrates for dopamine reuptake transporters, elevating extracellular dopamine by inhibiting uptake and promoting reverse transport. This produces an increase in extracellular dopamine that inhibits dopamine neuron firing through autoreceptor activation and consequently blunts phasic dopamine neurotransmission, an important learning signal. However, these mechanisms do not explain the beneficial behavioral effects observed at clinically useful concentrations. In the present study, we have used patch-clamp electrophysiology in slices of mouse midbrain to show that, surprisingly, low concentrations of methamphetamine actually enhance dopamine neurotransmission and increase dopamine neuron firing through a dopamine transporter-mediated excitatory conductance. Both of these effects are reversed by higher concentrations of methamphetamine, which inhibit firing through dopamine D2 autoreceptor activation and decrease the peak amplitude of dopamine-mediated synaptic currents. These competing, concentration-dependent effects of methamphetamine suggest a mechanistic interplay by which lower concentrations of methamphetamine can overcome autoreceptor-mediated inhibition at the soma to increase phasic dopamine transmission.
Collapse
Affiliation(s)
- Sarah Y Branch
- Department of Physiology, University of Texas Health Science Center, San Antonio, Texas 78229, USA
| | | |
Collapse
|
34
|
Giardino WJ, Mark GP, Stenzel-Poore MP, Ryabinin AE. Dissociation of corticotropin-releasing factor receptor subtype involvement in sensitivity to locomotor effects of methamphetamine and cocaine. Psychopharmacology (Berl) 2012; 219:1055-63. [PMID: 21833501 PMCID: PMC3266955 DOI: 10.1007/s00213-011-2433-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 07/29/2011] [Indexed: 11/27/2022]
Abstract
RATIONALE Enhanced sensitivity to the euphoric and locomotor-activating effects of psychostimulants may influence an individual's predisposition to drug abuse and addiction. While drug-induced behaviors are mediated by the actions of several neurotransmitter systems, past research revealed that the corticotropin-releasing factor (CRF) system is important in driving the acute locomotor response to psychostimulants. OBJECTIVES We previously reported that genetic deletion of the CRF type-2 receptor (CRF-R2), but not the CRF type-1 receptor (CRF-R1) dampened the acute locomotor stimulant response to methamphetamine (1 mg/kg). These results contrasted with previous studies implicating CRF-R1 in the locomotor effects of psychostimulants. Since the majority of previous studies focused on cocaine, rather than methamphetamine, we set out to test the hypothesis that these drugs differentially engage CRF-R1 and CRF-R2. METHODS We expanded our earlier findings by first replicating our previous experiments at a higher dose of methamphetamine (2 mg/kg), and by assessing the effects of the CRF-R1-selective antagonist CP-376,395 (10 mg/kg) on methamphetamine-induced locomotor activity. Next, we used both genetic and pharmacological tools to examine the specific components of the CRF system underlying the acute locomotor response to cocaine (5-10 mg/kg). RESULTS While genetic deletion of CRF-R2 dampened the locomotor response to methamphetamine (but not cocaine), genetic deletion and pharmacological blockade of CRF-R1 dampened the locomotor response to cocaine (but not methamphetamine). CONCLUSIONS These findings highlight the differential involvement of CRF receptors in acute sensitivity to two different stimulant drugs of abuse, providing an intriguing basis for the development of more targeted therapeutics for psychostimulant addiction.
Collapse
Affiliation(s)
- William J Giardino
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
35
|
Mandt BH, Johnston NL, Zahniser NR, Allen RM. Acquisition of cocaine self-administration in male Sprague-Dawley rats: effects of cocaine dose but not initial locomotor response to cocaine. Psychopharmacology (Berl) 2012; 219:1089-97. [PMID: 21863236 PMCID: PMC3266438 DOI: 10.1007/s00213-011-2438-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 08/02/2011] [Indexed: 02/06/2023]
Abstract
RATIONALE We have previously described a model in which adult outbred male Sprague-Dawley rats are classified as either low or high cocaine responders (LCRs or HCRs, respectively) based on acute cocaine-induced open-field activation. This model revealed important individual differences in cocaine's effects, including that LCRs exhibited greater responding than HCRs on a progressive ratio schedule of cocaine reinforcement. However, no LCR/HCR differences in acquisition of cocaine self-administration (0.25 mg/kg/12 s infusion) were observed under these conditions. OBJECTIVES To determine if LCRs and HCRs differ in the effectiveness of cocaine to function as a reinforcer under a broader range of conditions, the present study assessed the acquisition of cocaine self-administration (fixed ratio 1 schedule of reinforcement) as a function of i.v. cocaine dose (0.1875, 0.375, 0.5, 1, or 1.5 mg/kg/6 s infusion). RESULTS LCRs and HCRs did not differ significantly on any measure of acquisition examined, including the day to meet acquisition criterion, percent acquired, and cocaine intake. The effect of dose on percent acquired and rate of acquisition peaked at the 1-mg/kg/infusion dose of cocaine. In contrast, the effect of dose on cocaine intake was linear, with the highest rate of intake occurring at the 1.5-mg/kg/infusion dose of cocaine. CONCLUSIONS LCRs and HCRs do not appear to differ in their acquisition of cocaine-reinforced operant responding across a range of cocaine doses, including conditions that lead to high levels of cocaine intake.
Collapse
Affiliation(s)
| | | | - Nancy R. Zahniser
- University of Colorado Denver, Department of Pharmacology and Neuroscience Program
| | | |
Collapse
|
36
|
Parker CC, Cheng R, Sokoloff G, Palmer AA. Genome-wide association for methamphetamine sensitivity in an advanced intercross mouse line. GENES, BRAIN, AND BEHAVIOR 2012; 11:52-61. [PMID: 22032291 PMCID: PMC3368015 DOI: 10.1111/j.1601-183x.2011.00747.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Sensitivity to the locomotor stimulant effects of methamphetamine (MA) is a heritable trait that utilizes neurocircuitry also associated with the rewarding effects of drugs. We used the power of a C57BL/6J × DBA/2J F(2) intercross (n = 676) and the precision of a C57BL/6J × DBA/2J F(8) advanced intercross line (Aap: B6, D2-G8; or F(8) AIL; n = 552) to identify and narrow quantitative trait loci (QTLs) associated with sensitivity to the locomotor stimulant effects of MA. We used the program QTLRel to simultaneously map QTL in the F(2) and F(8) AIL mice. We identified six genome-wide significant QTLs associated with locomotor activity at baseline and seven genome-wide significant QTLs associated with MA-induced locomotor activation. The average per cent decrease in QTL width between the F(2) and the integrated analysis was 65%. Additionally, these QTLs showed a distinct temporal specificity within each session that allowed us to further refine their locations, and identify one QTL with a 1.8-LOD support interval of 1.47 Mb. Next, we utilized publicly available bioinformatics resources to exploit strain-specific sequence data and strain- and region-specific expression data to identify candidate genes. These results illustrate the power of AILs in conjunction with sequence and gene expression data to investigate the genetic underpinnings of behavioral and other traits.
Collapse
Affiliation(s)
| | - Riyan Cheng
- Department of Human Genetics, the University of Chicago, IL 60637
| | - Greta Sokoloff
- Department of Human Genetics, the University of Chicago, IL 60637
| | - Abraham A. Palmer
- Department of Human Genetics, the University of Chicago, IL 60637
- Department of Psychiatry and Behavioral Neuroscience, the University of Chicago, IL 60637
| |
Collapse
|
37
|
Meal schedule influences food restriction-induced locomotor sensitization to methamphetamine. Psychopharmacology (Berl) 2012; 219:795-803. [PMID: 21750897 PMCID: PMC4416415 DOI: 10.1007/s00213-011-2401-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 06/26/2011] [Indexed: 10/18/2022]
Abstract
RATIONALE Traditional protocols for inducing sensitization to psychostimulants use an intermittent or "binge"-like drug administration, and binge eating behavior is comorbid with drug abuse in humans. Food restriction increases the reinforcing properties and self-administration of many drugs of abuse. OBJECTIVE The present study tested the hypotheses that (1) food restriction induces sensitization to the locomotor stimulation observed in response to methamphetamine and (2) a binge-like feeding schedule during food restriction produces increased sensitization compared to equally restricted mice fed in three daily meals. METHODS Male DBA/2J mice were fed ad libitum or were food restricted to either an 8% or 16% loss of body weight. Additionally, the food-restricted mice were divided into two groups that were fed in either one meal (binge) or three equal-sized meals (meal). After the reduced body weight was stable, mice were tested for locomotor activity following saline and methamphetamine (1 mg/kg) injections. RESULTS Both 16% body weight loss groups exhibited sensitization to methamphetamine. Opposite to our hypothesis, the 8% meal but not the 8% binge food-restricted group demonstrated locomotor sensitization. Serum corticosterone levels were significantly higher in the meal-fed groups when compared to the binge- and ad libitum-fed groups. CONCLUSIONS These results support a role for feeding schedule and plasma corticosterone levels in food restriction-induced enhancement of the effects of methamphetamine.
Collapse
|
38
|
Li D, Herrera S, Bubula N, Nikitina E, Palmer AA, Hanck DA, Loweth JA, Vezina P. Casein kinase 1 enables nucleus accumbens amphetamine-induced locomotion by regulating AMPA receptor phosphorylation. J Neurochem 2011; 118:237-47. [PMID: 21564097 PMCID: PMC3129449 DOI: 10.1111/j.1471-4159.2011.07308.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The closely related δ and ε isoforms of the serine/threonine protein kinase casein kinase 1 (Csnk1) have been implicated in the generation of psychostimulant-induced behaviors. In this study, we show that Csnk1δ/ε produces its effects on behavior by acting on the Darpp-32-PP1 signaling pathway to regulate AMPA receptor phosphorylation in the nucleus accumbens (NAcc). Inhibiting Csnk1δ/ε in the NAcc with the selective inhibitor PF-670462 blocks amphetamine induced locomotion and its ability to increase phosphorylation of Darpp-32 at S137 and T34, decrease PP1 activity and increase phosphorylation of the AMPA receptor subunit at S845. Consistent with these findings, preventing GluR1 phosphorylation with the alanine mutant GluR1(S845A) reduces glutamate-evoked currents in cultured medium spiny neurons and blocks the locomotor activity produced by NAcc amphetamine. Thus, Csnk1 enables the locomotor and likely the incentive motivational effects of amphetamine by regulating Darrp-32-PP1-GlurR1(S845) signaling in the NAcc. As such, Csnk1 may be a critical target for intervention in the treatment of drug use disorders.
Collapse
Affiliation(s)
- Dongdong Li
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Stacy Herrera
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Nancy Bubula
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Elena Nikitina
- Department of Medicine, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Abraham A Palmer
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
- Department of Human Genetics, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
- Committee on Neurobiology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Dorothy A Hanck
- Department of Medicine, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
- Committee on Neurobiology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Jessica A Loweth
- Committee on Neurobiology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Paul Vezina
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
- Committee on Neurobiology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| |
Collapse
|
39
|
Shabani S, McKinnon CS, Reed C, Cunningham CL, Phillips TJ. Sensitivity to rewarding or aversive effects of methamphetamine determines methamphetamine intake. GENES BRAIN AND BEHAVIOR 2011; 10:625-36. [PMID: 21554535 DOI: 10.1111/j.1601-183x.2011.00700.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Amphetamines have rewarding and aversive effects. Relative sensitivity to these effects may be a better predictor of vulnerability to addiction than sensitivity to one of these effects alone. We tested this hypothesis in a dose-response study in a second replicate set of mouse lines selectively bred for high vs. low methamphetamine (MA) drinking (MADR). Replicate 2 high (MAHDR-2) and low (MALDR-2) MA drinking mice were bred based on MA consumption in a two-bottle choice procedure and examined for novel tastant drinking. Sensitivities to the rewarding and aversive effects of several doses of MA (0.5, 2 and 4 mg/kg) were measured using a place conditioning procedure. After conditioning, mice were tested in a drug-free and then drug-present state for time spent in the saline- and MA-paired contexts. Similar to the first set of MADR lines, by the end of selection, MAHDR-2 mice consumed about 6 mg MA/kg/18 h, compared to nearly no MA in MALDR-2 mice, but had similar taste preference ratios. MAHDR-2 mice exhibited place preference in both the drug-free and drug-present tests, and no significant place aversion. In contrast, MALDR-2 mice exhibited no place preference or aversion during the drug-free test, but robust place aversion in the drug-present test. These data extend our preliminary findings from the first set of MADR lines and support the hypothesis that the combination of greater sensitivity to the rewarding effects of MA and insensitivity to the aversive effects of MA is genetically associated with heightened risk for MA consumption.
Collapse
Affiliation(s)
- S Shabani
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
40
|
Scibelli AC, McKinnon CS, Reed C, Burkhart-Kasch S, Li N, Baba H, Wheeler JM, Phillips TJ. Selective breeding for magnitude of methamphetamine-induced sensitization alters methamphetamine consumption. Psychopharmacology (Berl) 2011; 214:791-804. [PMID: 21088960 PMCID: PMC3320759 DOI: 10.1007/s00213-010-2086-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 10/30/2010] [Indexed: 02/06/2023]
Abstract
RATIONALE Genetically determined differences in susceptibility to drug-induced sensitization could be related to risk for drug consumption. OBJECTIVES Studies were performed to determine whether selective breeding could be used to create lines of mice with different magnitudes of locomotor sensitization to methamphetamine (MA). MA sensitization (MASENS) lines were also examined for genetically correlated responses to MA. METHODS Beginning with the F2 cross of C57BL/6J and DBA/2J strains, mice were tested for locomotor sensitization to repeated injections of 1 mg/kg MA and bred based on magnitude of sensitization. Five selected offspring generations were tested. All generations were also tested for MA consumption, and some were tested for dose-dependent locomotor-stimulant responses to MA, consumption of saccharin, quinine, and potassium chloride as a measure of taste sensitivity, and MA clearance after acute and repeated MA. RESULTS Selective breeding resulted in creation of two lines [MA high sensitization (MAHSENS) and MA low sensitization (MALSENS)] that differed in magnitude of MA-induced sensitization. Initially, greater MA consumption in MAHSENS mice reversed over the course of selection so that MALSENS mice consumed more MA. MAHSENS mice exhibited greater sensitivity to the acute stimulant effects of MA, but there were no significant differences between the lines in MA clearance from blood. CONCLUSIONS Genetic factors influence magnitude of MA-induced locomotor sensitization and some of the genes involved in magnitude of this response also influence MA sensitivity and consumption. Genetic factors leading to greater MA-induced sensitization may serve a protective role against high levels of MA consumption.
Collapse
Affiliation(s)
- Angela C. Scibelli
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, and Veterans Affairs Medical Center, Portland, OR, USA
| | - Carrie S. McKinnon
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, and Veterans Affairs Medical Center, Portland, OR, USA
| | - Cheryl Reed
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, and Veterans Affairs Medical Center, Portland, OR, USA
| | - Sue Burkhart-Kasch
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, and Veterans Affairs Medical Center, Portland, OR, USA
| | - Na Li
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, and Veterans Affairs Medical Center, Portland, OR, USA
| | - Harue Baba
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, and Veterans Affairs Medical Center, Portland, OR, USA
| | - Jeanna M. Wheeler
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, and Veterans Affairs Medical Center, Portland, OR, USA
| | - Tamara J. Phillips
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, and Veterans Affairs Medical Center, Portland, OR, USA. Portland VA Medical Center, 3710 SW US Veterans Hospital Rd., R&D-32, Portland, OR 97239, USA
| |
Collapse
|
41
|
|
42
|
Methamphetamine causes sustained depression in cerebral blood flow. Brain Res 2010; 1373:91-100. [PMID: 21156163 DOI: 10.1016/j.brainres.2010.12.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 12/02/2010] [Accepted: 12/07/2010] [Indexed: 11/20/2022]
Abstract
The use prevalence of the highly addictive psychostimulant methamphetamine (MA) has been steadily increasing over the past decade. MA abuse has been associated with both transient and permanent alterations in cerebral blood flow (CBF), hemorrhage, cerebrovascular accidents and death. To understand MA-induced changes in CBF, we exposed C56BL/6 mice to an acute bolus of MA (5mg/kg MA, delivered IP). This elicited a biphasic CBF response, characterized by an initial transient increase (~ 5 minutes) followed by a prolonged decrease (~ 30 minutes) of approximately 25% relative to baseline CBF--as measured by laser Doppler flowmetry over the somatosensory cortex. To assess if this was due to catecholamine derived vasoconstriction, phentolamine, an α-adrenergic antagonist was administered prior to MA treatment. This reduced the initial increase in CBF but failed to prevent the subsequent, sustained decrease in CBF. Consistent with prior reports, MA caused a transient increase in mean arterial blood pressure, body temperature and respiratory rate. Elevated respiratory rate resulted in hypocapnia. When respiratory rate was controlled by artificially ventilating mice, blood PaCO(2) levels after MA exposure remained unchanged from physiologic levels, and the MA-induced decrease in CBF was abolished. In vivo two-photon imaging of cerebral blood vessels revealed sustained MA-induced vasoconstriction of pial arterioles, consistent with laser Doppler flowmetry data. These findings show that even a single, acute exposure to MA can result in profound changes in CBF, with potentially deleterious consequences for brain function.
Collapse
|
43
|
Giardino WJ, Pastor R, Anacker AMJ, Spangler E, Cote DM, Li J, Stenzel-Poore MP, Phillips TJ, Ryabinin AE. Dissection of corticotropin-releasing factor system involvement in locomotor sensitivity to methamphetamine. GENES BRAIN AND BEHAVIOR 2010; 10:78-89. [PMID: 20731720 DOI: 10.1111/j.1601-183x.2010.00641.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sensitivity to the euphoric and locomotor-activating effects of drugs of abuse may contribute to risk for excessive use and addiction. Repeated administration of psychostimulants such as methamphetamine (MA) can result in neuroadaptive consequences that manifest behaviorally as a progressive escalation of locomotor activation, termed psychomotor sensitization. The present studies addressed the involvement of specific components of the corticotropin-releasing factor (CRF) system in locomotor activation and psychomotor sensitization induced by MA (1, 2 mg/kg) by utilizing pharmacological approaches, as well as a series of genetic knockout (KO) mice, each deficient for a single component of the CRF system: CRF-R1, CRF-R2, CRF, or the CRF-related peptide Urocortin 1 (Ucn1). CRF-R1 KO mice did not differ from wild-type mice in sensitization to MA, and pharmacological blockade of CRF-R1 with CP-154,526 (15, 30 mg/kg) in DBA/2J mice did not selectively attenuate either the acquisition or expression of MA-induced sensitization. Deletion of either of the endogenous ligands of CRF-R1 (CRF, Ucn1) either enhanced or had no effect on MA-induced sensitization, providing further evidence against a role for CRF-R1 signaling. Interestingly, deletion of CRF-R2 attenuated MA-induced locomotor activation, elucidating a novel contribution of the CRF system to MA sensitivity, and suggesting the participation of the endogenous urocortin peptides Ucn2 and Ucn3. Immunohistochemistry for Fos was used to visualize neural activation underlying CRF-R2-dependent sensitivity to MA, identifying the basolateral and central nuclei of the amygdala as neural substrates involved in this response. Our results support further examination of CRF-R2 involvement in neural processes associated with MA addiction.
Collapse
Affiliation(s)
- W J Giardino
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kamens HM, Andersen J, Picciotto MR. Modulation of ethanol consumption by genetic and pharmacological manipulation of nicotinic acetylcholine receptors in mice. Psychopharmacology (Berl) 2010; 208:613-26. [PMID: 20072781 PMCID: PMC2901400 DOI: 10.1007/s00213-009-1759-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Accepted: 12/09/2009] [Indexed: 12/12/2022]
Abstract
RATIONALE Alcohol and nicotine are commonly co-abused. Genetic correlations between responses to these drugs have been reported, providing evidence that common genes underlie the response to alcohol and nicotine. Nicotinic acetylcholine receptors (nAChRs) in the mesolimbic dopamine system are important in mediating nicotine response, and several studies suggest that alcohol may also interact with these nAChRs. OBJECTIVE The aim of this study was to examine the role of nAChRs containing α7 or β2 subunits in ethanol consumption. METHODS A two-bottle choice paradigm was used to determine ethanol consumption in wild-type and nAChR subunit knockout mice. Challenge studies were performed using the α4β2 nAChR partial agonist varenicline. RESULTS Mice lacking the β2 subunit consumed a similar amount of ethanol compared to their wild-type siblings in an ethanol-drinking paradigm. In contrast, mice lacking the α7 nAChR receptor subunit consumed significantly less ethanol than wild-type mice but consumed comparable amounts of water, saccharin, and quinine. In C57BL/6J mice, varenicline dose-dependently decreased ethanol consumption with a significant effect of 2 mg/kg, without affecting water or saccharin consumption. This effect of varenicline was not reversed in mice lacking either the α7 or β2 subunit, providing evidence that nAChRs containing one of these subunits are not required for this effect of varenicline. CONCLUSIONS This study provides evidence that α7 nAChRs are involved in ethanol consumption and supports the idea that pharmacological manipulation of nAChRs reduces ethanol intake. Additional nAChRs may also be involved in ethanol intake, and there may be functional redundancy in the nicotinic control of alcohol drinking.
Collapse
Affiliation(s)
- Helen M. Kamens
- Department of Psychiatry, School of Medicine, Yale University, 34 Park Street—3rd floor research, New Haven, CT 06508, USA
| | - Jimena Andersen
- Department of Psychiatry, School of Medicine, Yale University, 34 Park Street—3rd floor research, New Haven, CT 06508, USA
| | - Marina R. Picciotto
- Department of Psychiatry, School of Medicine, Yale University, 34 Park Street—3rd floor research, New Haven, CT 06508, USA
| |
Collapse
|
45
|
Harro J. Inter-individual differences in neurobiology as vulnerability factors for affective disorders: implications for psychopharmacology. Pharmacol Ther 2009; 125:402-22. [PMID: 20005252 DOI: 10.1016/j.pharmthera.2009.11.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 11/25/2009] [Indexed: 10/20/2022]
Abstract
Susceptibility to affective disorders is individually different, and determined both by genetic variance and life events that cause significant differences in the CNS structure and function between individual subjects. Therefore it is plausible that search for the inter-individual differences in endophenotypes that mediate the effects of causal factors, both genetic and environmental, will reveal the substrates for vulnerability, help to clarify pathogenetic mechanisms, and possibly aid in developing strategies to discover better, more personalized treatments. This review first examines comparatively a number of animal models of human affect and affect-related disorders that rely on persistent inter-individual differences, and then highlights some of the neurobiological findings in these models that are compatible with much of research in human behavioural and personality traits. Many behaviours occur in specific combinations in several models, but often remarkable dissociations are observed, providing a variety of constellations of traits. It is concluded that more systematic comparative experimentation on behaviour and neurobiology in different models is warranted to reveal possible "building blocks" of affect-related personality common in animals and humans. Looking into the perspectives in psychopharmacology the focus is placed on probable associations of inter-individual differences with brain structure and function, personality and coping strategies, and psychiatric vulnerability, highlighting some unexpected interactions between vulnerability endophenotypes, adverse life events, and behavioural traits. It is argued that further studies on inter-individual differences in affect and underlying neurobiology should include formal modeling of their epistatic, hierarchical and dynamic nature.
Collapse
Affiliation(s)
- Jaanus Harro
- Department of Psychology, University of Tartu, Estonian Centre of Behavioural and Health Sciences, Tiigi 78, 50410 Tartu, Estonia.
| |
Collapse
|
46
|
Wheeler JM, Reed C, Burkhart-Kasch S, Li N, Cunningham CL, Janowsky A, Franken FH, Wiren KM, Hashimoto JG, Scibelli AC, Phillips TJ. Genetically correlated effects of selective breeding for high and low methamphetamine consumption. GENES BRAIN AND BEHAVIOR 2009; 8:758-71. [PMID: 19689456 DOI: 10.1111/j.1601-183x.2009.00522.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Improved prevention and treatment of drug addiction will require deeper understanding of genetic factors contributing to susceptibility to excessive drug use. Intravenous operant self-administration methods have greatly advanced understanding of behavioral traits related to addiction. However, these methods are not suitable for large-scale genetic experiments in mice. Selective breeding of mice can aggregate 'addiction alleles' in a model that has the potential to identify coordinated effects of multiple genes. We produced mouse lines that orally self-administer high (MAHDR) or low (MALDR) amounts of methamphetamine, representing the first demonstration of selective breeding for self-administration of any psychostimulant drug. Conditioned place preference and taste aversion results indicate that MAHDR mice are relatively more sensitive to the rewarding effects and less sensitive to the aversive effects of methamphetamine, compared to MALDR mice. These results validate the oral route of self-administration for investigation of the motivational effects of methamphetamine and provide a viable alternative to intravenous self-administration procedures. Gene expression results for a subset of genes relevant to addiction-related processes suggest differential regulation by methamphetamine of apoptosis and immune pathways in the nucleus accumbens of MAHDR and MALDR mice. In each line, methamphetamine reduced an allostatic state by bringing gene expression back toward 'normal' levels. Genes differentially expressed in the drug-naï ve state, including Slc6a4 (serotonin transporter), Htr3a (serotonin receptor 3A), Rela [nuclear factor kappaB (NFkappaB)] and Fos (cFos), represent candidates whose expression levels may predict methamphetamine consumption and susceptibility to methamphetamine reward and aversion.
Collapse
Affiliation(s)
- J M Wheeler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Bryant CD, Graham ME, Distler MG, Munoz MB, Li D, Vezina P, Sokoloff G, Palmer AA. A role for casein kinase 1 epsilon in the locomotor stimulant response to methamphetamine. Psychopharmacology (Berl) 2009; 203:703-11. [PMID: 19050854 PMCID: PMC2729782 DOI: 10.1007/s00213-008-1417-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Accepted: 11/08/2008] [Indexed: 11/30/2022]
Abstract
RATIONALE We previously colocalized a quantitative trait locus (QTL) for sensitivity to the locomotor stimulant effects of methamphetamine (MA) with a QTL for expression of casein kinase 1 epsilon (Csnk1-epsilon) in the nucleus accumbens (NAc). Subsequently, we identified a single nucleotide polymorphism in CSNK1E (rs135745) that was associated with increased sensitivity to the subjective effects of d-amphetamine in healthy human subjects. Based on these results, we hypothesized that differential expression of Csnk1-epsilon causes differential sensitivity to MA-induced locomotor activity in mice. OBJECTIVE In the present study, we used PF-670462 (PF), which is a selective inhibitor of Csnk1-epsilon, to directly evaluate the role of Csnk1-epsilon in the locomotor stimulant response to MA in male C57BL/6J mice. METHODS We administered vehicle, PF, MA, or MA + PF, either via intraperitoneal injections or bilateral intra-NAc microinjections. We also examined Darpp-32 phosphorylation in mice receiving intraperitoneal injections. RESULTS Intraperitoneal PF (20-40 mg/kg) attenuated the locomotor stimulant response to MA (2 mg/kg) without affecting baseline activity. The high dose of PF also significantly inhibited MA-induced phosphorylation of Darpp-32, providing a potential mechanism by which Csnk1-epsilon contributes to MA-induced locomotor activity. Furthermore, microinjection of PF (5 microg/side) into the NAc completely blocked the locomotor stimulant response to MA (2.5 microg/side) without affecting baseline activity. CONCLUSIONS These results provide direct evidence that Csnk1-epsilon is crucial for the locomotor stimulant response to a moderate dose of MA and suggest that genetic polymorphisms affecting Csnk1-epsilon expression or function could influence sensitivity to amphetamines in both mice and humans.
Collapse
Affiliation(s)
- Camron D. Bryant
- University of Chicago; Department of Human Genetics; 920 E. 58 St. CLSC 507D; Chicago, IL 60637 USA
| | - Melissa E. Graham
- University of Chicago; Department of Human Genetics; 920 E. 58 St. CLSC 507D; Chicago, IL 60637 USA
| | - Margaret G. Distler
- University of Chicago; Department of Human Genetics; 920 E. 58 St. CLSC 507D; Chicago, IL 60637 USA
| | - Michaelanne B. Munoz
- University of Chicago; Department of Human Genetics; 920 E. 58 St. CLSC 507D; Chicago, IL 60637 USA
| | - Dongdong Li
- University of Chicago; Department of Psychiatry and Behavioral Neuroscience; 5841 S. Maryland Av MC 3077; Chicago, IL 60637 USA
| | - Paul Vezina
- University of Chicago; Department of Psychiatry and Behavioral Neuroscience; 5841 S. Maryland Av MC 3077; Chicago, IL 60637 USA
| | - Greta Sokoloff
- University of Chicago; Department of Human Genetics; 920 E. 58 St. CLSC 507D; Chicago, IL 60637 USA
| | - Abraham A. Palmer
- University of Chicago; Department of Human Genetics; 920 E. 58 St. CLSC 507D; Chicago, IL 60637 USA, University of Chicago; Department of Psychiatry and Behavioral Neuroscience; 5841 S. Maryland Av MC 3077; Chicago, IL 60637 USA,Corresponding author: Abraham A. Palmer, Ph.D.; University of Chicago; Department of Human Genetics; 920 E. 58 St. CLSC 507D; Chicago, IL 60637, USA; voice: (773) 834-2897; fax: (773) 834-0505
| |
Collapse
|
48
|
Abstract
Although the role of genetic factors in the response to drugs of abuse has been emphasized, no earlier studies have applied selective breeding to intravenous drug self-administration. Here we report the effects of six generations of selective breeding for rat lines with low or high levels of intravenous drug self-administration (LS and HS lines, respectively). Rats from the outbred founder population and the first selected generation were evaluated for intravenous self-administration of either morphine or cocaine. All subsequent generations were assessed for self-administration of cocaine, using a multifactorial score based on how rapidly self-administration behavior was acquired, levels of self-administration during acquisition, and the response to different doses of cocaine. All changes in cocaine self-administration that occurred in generations three through six were consistent with effects of selection, with most measures differing in sixth-generation LS and HS animals. Sixth-generation HS rats self-administered approximately five times more injections of low-dose cocaine than LS animals under fixed-ratio-5 (a schedule in which an injection is delivered after five lever presses). These findings support a role of genetic factors in influencing cocaine-reinforced behavior. Establishment of the LS and HS lines will allow future studies to evaluate the role of specific genetic factors that underlie these differences and may contribute to substance abuse disorders in humans.
Collapse
|
49
|
Reduced sensitivity to the locomotor-stimulant effects of cocaine is associated with increased sensitivity to its discriminative stimulus properties. Behav Pharmacol 2009; 20:67-77. [PMID: 19125118 DOI: 10.1097/fbp.0b013e3283242fdd] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Outbred Long-Evans rats exhibit wide variation in their locomotor response to cocaine. Here, we investigated the relationship between these individual differences and interoceptive effects of cocaine in low cocaine responder (LCR) and high cocaine responder (HCR) phenotypes. Rats were trained to discriminate cocaine (10.0 mg/kg, intraperitoneally) from saline by repeated pairings of injections with one of two response levers. In subsequent tests for stimulus generalization to other cocaine doses (1.25-15.0 mg/kg), LCRs exhibited partial-to-full generalization at 1.85 and 2.5 mg/kg cocaine, respectively, whereas HCRs did not. When the selective 5-HT reuptake inhibitor fluoxetine (5.0 mg/kg) was coadministered with saline or different cocaine doses, we observed similar upward shifts in dose-response in both phenotypes. In contrast, coadministration of the 5-HT2A/2C agonist (+/-)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI; 0.3 mg/kg) led to partial substitution of DOI for cocaine and enhancement of the stimulus properties of 1.25 mg/kg cocaine in LCRs only. Finally, a retest of cocaine-induced locomotion after discrimination testing revealed marked behavioral sensitization in LCRs and modest changes in behavior in HCRs. Taken together, these results suggest that initial sensitivity to the locomotor-stimulant effects of cocaine is inversely related to its interoceptive properties and that differences in 5-HT systems may contribute to the phenotypic differences observed.
Collapse
|
50
|
Kelly MA, Low MJ, Rubinstein M, Phillips TJ. Role of dopamine D1-like receptors in methamphetamine locomotor responses of D2 receptor knockout mice. GENES BRAIN AND BEHAVIOR 2008; 7:568-77. [PMID: 18363855 DOI: 10.1111/j.1601-183x.2008.00392.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Behavioral sensitization to psychostimulants manifests as an increased locomotor response with repeated administration. Dopamine systems are accepted to play a fundamental role in sensitization, but the role of specific dopamine receptor subtypes has not been completely defined. This study used the combination of dopamine D2 receptor-deficient mice and a D1-like antagonist to examine dopamine D1 and D2 receptor involvement in acute and sensitized locomotor responses to methamphetamine. Absence of the dopamine D2 receptor resulted in attenuation of the acute stimulant effects of methamphetamine. Mutant and wild-type mice exhibited sensitization that lasted longer within the time period of the challenge test in the mutant animals. Pretreatment with the D1-like receptor antagonist SCH 23390 produced more potent reductions in the acute and sensitized locomotor responses to methamphetamine in D2 receptor-deficient mice than in wild-type mice; however, the expression of locomotor sensitization when challenged with methamphetamine alone was equivalently attenuated by previous treatment with SCH 23390. These data suggest that dopamine D2 receptors play a key role in the acute stimulant and sensitizing effects of methamphetamine and act in concert with D1-like receptors to influence the acquisition of methamphetamine-induced behavioral sensitization, traits that may influence continued methamphetamine use.
Collapse
Affiliation(s)
- M A Kelly
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | | | | | | |
Collapse
|