1
|
Chellian R, Behnood-Rod A, Bruijnzeel AW. Sex differences in nicotine intake and relapse behavior in nicotine-dependent adult wistar rats. Front Pharmacol 2024; 15:1415219. [PMID: 39391691 PMCID: PMC11464435 DOI: 10.3389/fphar.2024.1415219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Tobacco use is highly addictive and the leading cause of premature mortality in the world. Long-access nicotine self-administration procedures in rats closely model human smoking behavior. However, significant gaps remain in our understanding of sex differences in the development of dependence and relapse in adult rats. Methods In the present study, we investigated operant responding for both nicotine and saline and the development of dependence in adult rats of both sexes. The rats had daily access to nicotine or saline for 6 h per day, 7 days per week. Dependence was assessed by evaluating precipitated and spontaneous somatic withdrawal signs, measuring locomotor activity in the small open field test, and assessing anxiety-like behavior in the large open field and elevated plus maze test. The sucrose preference test was used to determine if cessation of nicotine intake leads to anhedonia. It was also investigated if a period of forced abstinence affects nicotine-seeking behavior. Results This study showed that nicotine intake is higher in females than in males when given daily long access to nicotine. Daily nicotine self-administration led to more precipitated and spontaneous somatic withdrawal signs compared to saline self-administration, with no sex differences observed. In addition, cessation of nicotine intake led to a similar increase in activity in both males and females in the small open field test. However, cessation of nicotine intake did not increase anxiety-like behavior or cause anhedonia in either males or females. A time course analysis revealed that the nicotinic acetylcholine receptor antagonist mecamylamine affected nicotine intake differently in males and females, increasing intake in males and decreasing intake in females. Three weeks of forced abstinence led to an increase in nicotine and saline-seeking behavior. The rats exhibited more nicotine than saline seeking, and the females displayed more nicotine seeking than the males. Discussion The present findings demonstrate that females self-administer more nicotine and display more nicotine-seeking behavior than males. Furthermore, there were no sex differences in somatic withdrawal signs or activity during abstinence from nicotine. This work underscores the importance of considering sex differences across various aspects of addiction, including intake and relapse, when developing novel treatments for tobacco use disorder.
Collapse
|
2
|
Carrica LK, Choi CY, Walter FA, Noonan BL, Shi L, Johnson CT, Bradshaw HB, Liang NC, Gulley JM. Effects of combined use of alcohol and delta-9-tetrahydrocannibinol on working memory in Long Evans rats. Behav Brain Res 2023; 449:114475. [PMID: 37146720 PMCID: PMC10247469 DOI: 10.1016/j.bbr.2023.114475] [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: 02/01/2023] [Revised: 03/31/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
The increase in social acceptance and legalization of cannabis over the last several years is likely to increase the prevalence of its co-use with alcohol. In spite of this, the potential for effects unique to co-use of these drugs, especially in moderate doses, has been studied relatively infrequently. We addressed this in the current study using a laboratory rat model of voluntary drug intake. Periadolescent male and female Long-Evans rats were allowed to orally self-administer ethanol, Δ9-tetrahydrocannibinol (THC), both drugs, or their vehicle controls from postnatal day (P) 30 to P47. They were subsequently trained and tested on an instrumental behavior task that assesses attention, working memory and behavioral flexibility. Similar to previous work, consumption of THC reduced both ethanol and saccharin intake in both sexes. Blood samples taken 14 h following the final self-administration session revealed that females had higher levels of the THC metabolite THC-COOH. There were modest effects of THC on our delayed matching to position (DMTP) task, with females exhibiting reduced performance compared to their control group or male, drug using counterparts. However, there were no significant effects of co-use of ethanol or THC on DMTP performance, and drug effects were also not apparent in the reversal learning phase of the task when non-matching to position was required as the correct response. These findings are consistent with other published studies in rodent models showing that use of these drugs in low to moderate doses does not significantly impact memory or behavioral flexibility following a protracted abstinence period.
Collapse
Affiliation(s)
- Lauren K Carrica
- Department of Psychology, University of Illinois at Urbana-Champaign, USA
| | - Chan Young Choi
- Department of Psychology, University of Illinois at Urbana-Champaign, USA
| | - Francis A Walter
- Neuroscience Program, University of Illinois at Urbana-Champaign, USA
| | - Brynn L Noonan
- Department of Psychology, University of Illinois at Urbana-Champaign, USA
| | - Linyuan Shi
- Department of Psychology, University of Illinois at Urbana-Champaign, USA
| | - Clare T Johnson
- Department of Psychological & Brain Science, Indiana University, Bloomington, IN, USA
| | - Heather B Bradshaw
- Department of Psychological & Brain Science, Indiana University, Bloomington, IN, USA
| | - Nu-Chu Liang
- Department of Psychology, University of Illinois at Urbana-Champaign, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, USA
| | - Joshua M Gulley
- Department of Psychology, University of Illinois at Urbana-Champaign, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, USA.
| |
Collapse
|
3
|
Serra V, Aroni S, Bortolato M, Frau R, Melis M. Endocannabinoid-dependent decrease of GABAergic transmission on dopaminergic neurons is associated with susceptibility to cocaine stimulant effects in pre-adolescent male MAOA hypomorphic mice exposed to early life stress. Neuropharmacology 2023; 233:109548. [PMID: 37080337 DOI: 10.1016/j.neuropharm.2023.109548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Vulnerability to cocaine use disorder depends upon a combination of genetic and environmental risk factors. While early life adversity is a critical environmental vulnerability factor for drug misuse, allelic variants of the monoamine oxidase A (MAOA) gene have been shown to moderate its influence on the risk of drug-related problems. However, data on the interactions between MAOA variants and early life stress (ES) with respect to predisposition to cocaine abuse are limited. Here, we show that a mouse model capturing the interaction of genetic (low-activity alleles of the Maoa gene; MAOANeo) and environmental (i.e., ES) vulnerability factors displays an increased sensitivity to repeated in vivo cocaine psychomotor stimulant actions associated with a reduction of GABAA receptor-mediated inhibition of dopamine neurons of the ventral tegmental area (VTA). Depolarization-induced suppression of inhibition (DSI), a 2-arachidonoylglycerol (2AG)-dependent form of short-term plasticity, also becomes readily expressed by dopamine neurons from male MAOANeo ES mice repeatedly treated with cocaine. The activation of either dopamine D2 or CB1 receptors contributes to cocaine-induced DSI expression, decreased GABA synaptic efficacy, and hyperlocomotion. Next, in vivo pharmacological enhancement of 2AG signaling during repeated cocaine exposure occludes its actions both in vivo and ex vivo. This data extends our knowledge of the multifaceted sequelae imposed by this gene-environment interaction in VTA dopamine neurons of male pre-adolescent mice and contributes to our understanding of neural mechanisms of vulnerability for early onset cocaine use.
Collapse
Affiliation(s)
- Valeria Serra
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy
| | - Sonia Aroni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, 84112, USA
| | - Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042, Monserrato, Italy.
| |
Collapse
|
4
|
Carrica LK, Choi CY, Walter FA, Noonan BL, Shi L, Johnson CT, Bradshaw HB, Liang NC, Gulley JM. Effects of combined use of alcohol and delta-9-tetrahydrocannibinol on working memory in Long Evans rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526698. [PMID: 36778500 PMCID: PMC9915622 DOI: 10.1101/2023.02.02.526698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The increase in social acceptance and legalization of cannabis over the last several years is likely to increase the prevalence of its co-use with alcohol. In spite of this, the potential for effects unique to co-use of these drugs, especially in moderate doses, has been studied relatively infrequently. We addressed this in the current study using a laboratory rat model of voluntary drug intake. Periadolescent male and female Long-Evans rats were allowed to orally self-administer ethanol, Î" 9 -tetrahydrocannibinol (THC), both drugs, or their vehicle controls from postnatal day (P) 30 to P47. They were subsequently trained and tested on an instrumental behavior task that assesses attention, working memory and behavioral flexibility. Similar to previous work, consumption of THC reduced both ethanol and saccharin intake in both sexes. Blood samples taken 14h following the final self-administration session revealed that females had higher levels of the THC metabolite THC-COOH. There were modest effects of THC on our delayed matching to position (DMTP) task, with females exhibiting reduced performance compared to their control group or male, drug using counterparts. However, there were no significant effects of co-use of ethanol or THC on DMTP performance, and drug effects were also not apparent in the reversal learning phase of the task when non-matching to position was required as the correct response. These findings are consistent with other published studies in rodent models showing that use of these drugs in low to moderate doses does not significantly impact memory or behavioral flexibility following a protracted abstinence period.
Collapse
Affiliation(s)
- Lauren K. Carrica
- Department of Psychology, University of Illinois at Urbana-Champaign
| | - Chan Young Choi
- Department of Psychology, University of Illinois at Urbana-Champaign
| | | | - Brynn L. Noonan
- Department of Psychology, University of Illinois at Urbana-Champaign
| | - Linyuan Shi
- Department of Psychology, University of Illinois at Urbana-Champaign
| | - Clare T. Johnson
- Department of Psychological & Brain Science, Indiana University, Bloomington, Indiana
| | - Heather B. Bradshaw
- Department of Psychological & Brain Science, Indiana University, Bloomington, Indiana
| | - Nu-Chu Liang
- Department of Psychology, University of Illinois at Urbana-Champaign
- Neuroscience Program, University of Illinois at Urbana-Champaign
| | - Joshua M. Gulley
- Department of Psychology, University of Illinois at Urbana-Champaign
- Neuroscience Program, University of Illinois at Urbana-Champaign
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign
| |
Collapse
|
5
|
Sauton P, Jeanblanc J, Benzerouk F, Gierski F, Naassila M. Sex-specific decision-making impairments and striatal dopaminergic changes after binge drinking history in rats. Front Pharmacol 2023; 14:1076465. [PMID: 36726581 PMCID: PMC9885167 DOI: 10.3389/fphar.2023.1076465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Binge drinking (BD) is a harmful behavior for health and is a predictive factor for the development of alcohol addiction. Weak decision-making (DM) capacities could play a role in the vulnerability to BD which in turn would lead to DM impairments, thus perpetuating BD. Longitudinal preclinical studies are however lacking and necessary to understand this complex relationship. Both DM and BD are influenced by sex and involve dopamine release in the core of the nucleus accumbens, a central mechanism regulated by dopamine D2/3 autoreceptors. In this context, we used an operant self-administration procedure of BD in male and female rats, and longitudinally assessed DM capacity, memory and anxiety-like behavior. To better understand the mechanisms potentially involved in the relationship between DM and BD, ex vivo dopamine transmission was assessed short term after the end of the binge exposure in the core of the nucleus accumbens (NAc) using the fast-scan cyclic voltammetry (FSCV) technique and the D2/3 agonist quinpirole. We found important basal sex differences in DM, with female rats showing better performances at baseline. Choice processes were impaired exclusively in males after BD history, associated with a decrease in impulse control in both sexes, while memory and anxiety-like behavior were not affected. Our neurobiological results demonstrate that BD did not affect basal dopamine signaling in the NAc core, regardless of the sex, but reveal changes in the sensitivity to the inhibitory effects of quinpirole in females. DM impairments were neither associated with changes in basal dopamine signaling nor pre-synaptic D2 activity. Overall, our findings show that BD affects both DM processes and dopamine transmission in the core of the NAc in a sex-related manner, further suggesting that these effects may play a role in the vicious cycle leading to BD perpetuation and the early onset of AUD. Our results may inform novel strategies for therapeutic and prevention interventions.
Collapse
Affiliation(s)
- Pierre Sauton
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France
| | - Jerome Jeanblanc
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France
| | - Farid Benzerouk
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France,Université de Reims Champagne-Ardenne, Laboratoire Cognition, Santé, Société (C2S, EA6291), Reims, France
| | - Fabien Gierski
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France,Université de Reims Champagne-Ardenne, Laboratoire Cognition, Santé, Société (C2S, EA6291), Reims, France
| | - Mickael Naassila
- INSERM UMR 1247—Research Group on Alcohol & Pharmacodependences (GRAP), Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Amiens, France,*Correspondence: Mickael Naassila,
| |
Collapse
|
6
|
Pareek T, Platt DM, Rüedi-Bettschen D. Daily, limited access to methamphetamine self-administration during pregnancy leads to increased methamphetamine sensitivity in adult offspring. Dev Psychobiol 2023; 65:e22350. [PMID: 36567658 PMCID: PMC10038219 DOI: 10.1002/dev.22350] [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: 02/07/2022] [Revised: 09/20/2022] [Accepted: 11/04/2022] [Indexed: 12/14/2022]
Abstract
Methamphetamine use by women, even throughout pregnancy, is common. But there is limited knowledge about the effects in prenatally methamphetamine-exposed children. This study investigated how prenatal methamphetamine exposure in rats, via maternal i.v. self-administration, affected the sensitivity of adult offspring to methamphetamine in comparison with controls. The offspring were generated from dams either self-administering methamphetamine daily under limited-access conditions prior to and throughout pregnancy, or their respective saline-yoked control dams. Spontaneous and methamphetamine-induced locomotor activity was assessed in male and female offspring of both exposure groups after a range of methamphetamine doses. In a separate group of offspring, acquisition of i.v. methamphetamine self-administration, responding under fixed and progressive ratio schedules of methamphetamine reinforcement, and reinstatement of extinguished drug-seeking behavior were assessed. Methamphetamine dose-dependently increased locomotor activity in both exposure groups. However, methamphetamine-exposed males showed significantly enhanced locomotor activity compared with controls at 1 mg/kg, and methamphetamine-exposed females showed significantly enhanced locomotor activity compared with controls at 3.2 mg/kg. Methamphetamine-exposed offspring of both sexes acquired methamphetamine self-administration faster and showed overall higher levels of methamphetamine-induced reinstatement compared with controls. Taken together, these results indicate that prenatal methamphetamine exposure to relatively low levels alters methamphetamine sensitivity in male and female adult offspring.
Collapse
Affiliation(s)
- Tanya Pareek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
- Graduate Program in Neuroscience, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Donna M. Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Daniela Rüedi-Bettschen
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| |
Collapse
|
7
|
Iyer V, Rangel-Barajas C, Woodward TJ, Kulkarni A, Cantwell L, Crystal JD, Mackie K, Rebec GV, Thakur GA, Hohmann AG. Negative allosteric modulation of CB 1 cannabinoid receptor signaling suppresses opioid-mediated reward. Pharmacol Res 2022; 185:106474. [PMID: 36179954 PMCID: PMC9948526 DOI: 10.1016/j.phrs.2022.106474] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/15/2022] [Accepted: 09/25/2022] [Indexed: 01/18/2023]
Abstract
Blockade of cannabinoid type 1 (CB1)-receptor signaling decreases the rewarding properties of many drugs of abuse and has been proposed as an anti-addiction strategy. However, psychiatric side-effects limit the clinical potential of orthosteric CB1 antagonists. Negative allosteric modulators (NAMs) represent a novel and indirect approach to attenuate CB1 signaling by decreasing affinity and/or efficacy of CB1 ligands. We hypothesized that a CB1-NAM would block opioid reward while avoiding the unwanted effects of orthosteric CB1 antagonists. GAT358, a CB1-NAM, failed to elicit cardinal signs of direct CB1 activation or inactivation when administered by itself. GAT358 decreased catalepsy and hypothermia but not antinociception produced by the orthosteric CB1 agonist CP55,940, suggesting that a CB1-NAM blocked cardinal signs of CB1 activation. Next, GAT358 was evaluated using in vivo assays of opioid-induced dopamine release and reward in male rodents. In the nucleus accumbens shell, a key component of the mesocorticolimbic reward pathway, morphine increased electrically-evoked dopamine efflux and this effect was blocked by a dose of GAT358 that lacked intrinsic effects on evoked dopamine efflux. Moreover, GAT358 blocked morphine-induced reward in a conditioned place preference (CPP) assay without producing reward or aversion alone. GAT358-induced blockade of morphine CPP was also occluded by GAT229, a CB1 positive allosteric modulator (CB1-PAM), and absent in CB1-knockout mice. Finally, GAT358 also reduced oral oxycodone (but not water) consumption in a two-bottle choice paradigm. Our results support the therapeutic potential of CB1-NAMs as novel drug candidates aimed at preventing opioid reward and treating opioid abuse while avoiding unwanted side-effects.
Collapse
Affiliation(s)
- Vishakh Iyer
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | | | - Taylor J. Woodward
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Abhijit Kulkarni
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Lucas Cantwell
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Jonathon D. Crystal
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Ken Mackie
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA,Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - George V. Rebec
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Ganesh A. Thakur
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Andrea G. Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA,Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA,Corresponding Author: Andrea G. Hohmann, Psychological and Brain Sciences, Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-7007,
| |
Collapse
|
8
|
Melugin PR, Wu F, Munoz C, Phensy A, Pradhan G, Luo Y, Nofal A, Manepalli R, Kroener S. The effects of acamprosate on prefrontal cortical function are mimicked by CaCl2 and they are influenced by the history of alcohol exposure. Neuropharmacology 2022; 212:109062. [PMID: 35430241 PMCID: PMC10804777 DOI: 10.1016/j.neuropharm.2022.109062] [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: 04/11/2021] [Revised: 03/29/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022]
Abstract
Alcohol use disorder is associated with functional changes in the medial prefrontal cortex (mPFC), which include altered glutamatergic transmission and deficits in executive functions that contribute to relapse. Acamprosate (calcium-bis N-acetylhomotaurinate) reduces alcohol craving and relapse, effects that are thought to be mediated by acamprosate's ability to ameliorate alcohol-induced dysregulation of glutamatergic signaling. Treatment with acamprosate and its active moiety calcium (CaCl2) both improve deficits in cognitive flexibility in postdependent mice following chronic intermittent ethanol (CIE) exposure. Here, we show that mice that self-administered alcohol under goal-directed conditions (i.e., operant responding on a fixed-ratio schedule) also display similar deficits in cognitive flexibility and altered glutamatergic signaling in the mPFC, both of which were improved with acamprosate or CaCl2. However, under conditions shown to bias behavior towards habitual responding (operant self-administration after CIE exposure, or on a variable interval schedule), alcohol-induced changes to glutamatergic transmission were unaffected by either acamprosate or CaCl2 treatment. Together, these findings suggest that the variable effects of acamprosate on synaptic signaling may reflect a shift in mPFC networks related to the loss of behavioral control in habitual alcohol-seeking.
Collapse
Affiliation(s)
- Patrick R Melugin
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Fei Wu
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA; Institute of Neurobiology, Jining Medical University, Jining, China
| | - Crystal Munoz
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Aarron Phensy
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Grishma Pradhan
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Yi Luo
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Abraham Nofal
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Rohan Manepalli
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA
| | - Sven Kroener
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, TX, USA.
| |
Collapse
|
9
|
Levis SC, Baram TZ, Mahler SV. Neurodevelopmental origins of substance use disorders: Evidence from animal models of early-life adversity and addiction. Eur J Neurosci 2022; 55:2170-2195. [PMID: 33825217 PMCID: PMC8494863 DOI: 10.1111/ejn.15223] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 01/06/2023]
Abstract
Addiction is a chronic relapsing disorder with devastating personal, societal, and economic consequences. In humans, early-life adversity (ELA) such as trauma, neglect, and resource scarcity are linked with increased risk of later-life addiction, but the brain mechanisms underlying this link are still poorly understood. Here, we focus on data from rodent models of ELA and addiction, in which causal effects of ELA on later-life responses to drugs and the neurodevelopmental mechanisms by which ELA increases vulnerability to addiction can be determined. We first summarize evidence for a link between ELA and addiction in humans, then describe how ELA is commonly modeled in rodents. Since addiction is a heterogeneous disease with many individually varying behavioral aspects that may be impacted by ELA, we next discuss common rodent assays of addiction-like behaviors. We then summarize the specific addiction-relevant behavioral phenotypes caused by ELA in male and female rodents and discuss some of the underlying changes in brain reward and stress circuits that are likely responsible. By better understanding the behavioral and neural mechanisms by which ELA promotes addiction vulnerability, we hope to facilitate development of new approaches for preventing or treating addiction in those with a history of ELA.
Collapse
Affiliation(s)
- Sophia C. Levis
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA
| | - Tallie Z. Baram
- Department of Anatomy & Neurobiology, University of California Irvine, Irvine, CA
- Department of Pediatrics, University of California Irvine, Irvine, CA
| | - Stephen V. Mahler
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA
| |
Collapse
|
10
|
Neuroadaptations and TGF-β signaling: emerging role in models of neuropsychiatric disorders. Mol Psychiatry 2022; 27:296-306. [PMID: 34131268 PMCID: PMC8671568 DOI: 10.1038/s41380-021-01186-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023]
Abstract
Neuropsychiatric diseases are manifested by maladaptive behavioral plasticity. Despite the greater understanding of the neuroplasticity underlying behavioral adaptations, pinpointing precise cellular mediators has remained elusive. This has stymied the development of pharmacological interventions to combat these disorders both at the level of progression and relapse. With increased knowledge on the putative role of the transforming growth factor (TGF- β) family of proteins in mediating diverse neuroadaptations, the influence of TGF-β signaling in regulating maladaptive cellular and behavioral plasticity underlying neuropsychiatric disorders is being increasingly elucidated. The current review is focused on what is currently known about the TGF-β signaling in the central nervous system in mediating cellular and behavioral plasticity related to neuropsychiatric manifestations.
Collapse
|
11
|
Iyer V, Woodward TJ, Pacheco R, Hohmann AG. A limited access oral oxycodone paradigm produces physical dependence and mesocorticolimbic region-dependent increases in DeltaFosB expression without preference. Neuropharmacology 2021; 205:108925. [PMID: 34921830 DOI: 10.1016/j.neuropharm.2021.108925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/02/2021] [Accepted: 12/11/2021] [Indexed: 01/07/2023]
Abstract
The abuse of oral formulations of prescription opioids has precipitated the current opioid epidemic. We developed an oral oxycodone consumption model consisting of a limited access (4 h) two-bottle choice drinking in the dark (TBC-DID) paradigm and quantified dependence with naloxone challenge using mice of both sexes. We also assessed neurobiological correlates of withdrawal and dependence elicited via oral oxycodone consumption using immunohistochemistry for DeltaFosB (ΔFosB), a transcription factor described as a molecular marker for drug addiction. Neither sex developed a preference for the oxycodone bottle, irrespective of oxycodone concentration, bottle position or prior water restriction. Mice that volitionally consumed oxycodone exhibited hyperlocomotion in an open field test and supraspinal but not spinally-mediated antinociception. Both sexes also developed robust, dose-dependent levels of opioid withdrawal that was precipitated by the opioid antagonist naloxone. Oral oxycodone consumption followed by naloxone challenge led to mesocorticolimbic region-dependent increases in the number of ΔFosB expressing cells. Naloxone-precipitated withdrawal jumps, but not the oxycodone bottle % preference, was positively correlated with the number of ΔFosB expressing cells specifically in the nucleus accumbens shell. Thus, limited access oral consumption of oxycodone produced physical dependence and increased ΔFosB expression despite the absence of opioid preference. Our TBC-DID paradigm allows for the study of oral opioid consumption in a simple, high-throughput manner and elucidates the underlying neurobiological substrates that accompany opioid-induced physical dependence.
Collapse
Affiliation(s)
- Vishakh Iyer
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Taylor J Woodward
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Romario Pacheco
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Andrea G Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA; Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
12
|
Rath M, Tawfic J, Abrorkhujaeva A, Sowell S, Wu S, Eans SO, Peris J, McLaughlin JP, Stevens SM, Liu B. Binge ethanol consumption-associated behavioral impairments in male mice using a gelatin-based drinking-in-the dark model. Alcohol 2021; 95:25-36. [PMID: 34029701 PMCID: PMC10629591 DOI: 10.1016/j.alcohol.2021.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/16/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Acute intoxication caused by binge ethanol drinking is linked to widespread impairments in brain functions. Various alcohol administration paradigms have been used in animals to model the heterogeneous clinical manifestation of intoxication in people. It is challenging to model a procedure that produces "visible intoxication" in rodents; however, manipulation of variables such as route of alcohol administration, time of availability, frequency, and duration and amount of ethanol exposure has achieved some success. In the current study, we employed a modified drinking-in-the-dark model to assess the validity of this model in producing "post-ethanol consumption intoxication" impairments following prolonged repeated daily voluntary "binge" ethanol consumption. METHODS Adult male C57BL/6J mice were allowed a daily 3-h access to non-alcoholic plain or ethanol-containing gel during the dark cycle for a total of 83 days. After the initial 2-month daily DID, ethanol intake patterns were intensely characterized during the next 3 weeks. Immediately following the last DID session (day 83), plain and ethanol gel-consuming mice were then subjected to behavioral tests of locomotor ability and/or anxiety (cylinder, wire grip, open field) followed by blood ethanol concentration measurement. RESULT Mice exhibited a relatively consistent ethanol consumption pattern during and across daily access periods. Ethanol intake of individual mice positively correlated with blood ethanol concentration that averaged 61.64 ± 2.84 mg/dL (n = 12). Compared to the plain gel-consuming control mice, ethanol gel mice exhibited significant locomotor impairment as well as anxiety-like behavior, with the magnitude of impairments of key indices well correlated with blood ethanol levels. CONCLUSION The gelatin vehicle-based voluntary ethanol drinking-in-the-dark model reliably produced post consumption acute movement impairments as well as anxiety-like behaviors even after 2 months of daily binge ethanol consumption in male mice. Taken together, this mouse binge ethanol model should facilitate the investigation of mechanisms of binge drinking in subjects chronically abusing ethanol and the search for effective novel treatment strategies.
Collapse
Affiliation(s)
- Meera Rath
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, United States
| | - Jasmin Tawfic
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, United States
| | - Aziza Abrorkhujaeva
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, United States
| | - Sam Sowell
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, United States
| | - Sara Wu
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, United States
| | - Shainnel O Eans
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, United States
| | - Joanna Peris
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, United States
| | - Jay P McLaughlin
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, United States
| | - Stanley M Stevens
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, 33620, United States
| | - Bin Liu
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, 32610, United States.
| |
Collapse
|
13
|
Caffino L, Moro F, Mottarlini F, Targa G, Di Clemente A, Toia M, Orrù A, Giannotti G, Fumagalli F, Cervo L. Repeated exposure to cocaine during adolescence enhances the rewarding threshold for cocaine-conditioned place preference in adulthood. Addict Biol 2021; 26:e13012. [PMID: 33511707 DOI: 10.1111/adb.13012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/22/2020] [Accepted: 01/14/2021] [Indexed: 01/24/2023]
Abstract
Previous studies have shown that adolescent exposure to cocaine increases drug use in adulthood, albeit incubation of cocaine seeking was found to be attenuated in rats trained to self-administer cocaine during adolescence. We here hypothesize that adolescent exposure to cocaine could alter the rewarding properties of the psychostimulant in adulthood. By employing two of the most widely used animal-experimental-preclinical models to investigate drug addiction, we evaluated whether contingent versus non-contingent cocaine self-administration during adolescence modulates its rewarding threshold in adulthood evaluated by conditioned place preference (CPP). Cocaine self-administration during adolescence increases the rewarding threshold in adulthood; CPP for cocaine was observed at the higher (20 mg/kg), but not at the lower (10 mg/kg), dose employed. Rats exposed to either contingent or non-contingent cocaine during adolescence exhibited the same behavior in the CPP paradigm suggesting that, under our experimental conditions, cocaine rewarding properties are shaped by the psychostimulant itself and not by its motivational effects. From a mechanistic standpoint, the preference for the 20 mg/kg cocaine-paired side in a CPP paradigm appears to depend, at least partially, upon the formation of GluA2-lacking Ca2+ -permeable AMPA receptors and the consequent increase of αCaMKII activity in the NAc, both of which are instead reduced when the 10 mg/kg dose was used. In conclusion, contingent or non-contingent cocaine exposure during adolescence desensitizes adult animals to a rewarding dose of cocaine (10 mg/kg) elevating the rewarding threshold necessary (20 mg/kg) to drive conditioned place preference, an effect that may predispose to higher consumption of cocaine during adulthood.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Italy
| | - Federico Moro
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research IRCCS Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Italy
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Italy
| | - Angelo Di Clemente
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research IRCCS Italy
| | - Marianna Toia
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research IRCCS Italy
| | - Alessandro Orrù
- Parco Scientifico e Tecnologico della Sardegna Institute of Translational Pharmacology (C.N.R.) Italy
| | - Giuseppe Giannotti
- Department of Anesthesiology University of Colorado Anschutz Medical Campus Aurora CO USA
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano Italy
| | - Luigi Cervo
- Experimental Psychopharmacology, Department of Neuroscience Mario Negri Institute for Pharmacological Research IRCCS Italy
| |
Collapse
|
14
|
Flores RJ, Alshbool FZ, Giner P, O'Dell LE, Mendez IA. Exposure to nicotine vapor produced by an electronic nicotine delivery system causes short-term increases in impulsive choice in adult male rats. Nicotine Tob Res 2021; 24:358-365. [PMID: 34232312 DOI: 10.1093/ntr/ntab141] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 07/05/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Traditional cigarette use influences cost-benefit decision making by promoting impulsive choice. However, the impact of exposure via electronic nicotine delivery systems on impulsive choice remains unclear. Hence, the present study examined the short- and long-term effects of nicotine vapor on impulsive choice. METHODS Twenty-four adult male rats were trained in the delay discounting task, to choose between small, immediate food rewards or large, delayed food rewards. After 24 days of training in the task, rats were exposed to vapor containing either 0, 12, or 24 mg/mL of nicotine, for ten days. To validate inhalation of nicotine vapor, serum cotinine levels were analyzed on exposure days 1, 5, and 10 using enzyme-linked immunosorbent assay (ELISA). Following vapor exposure, rats were retrained in the discounting task until rats displayed stable responding, and the effects of nicotine vapor on choice preference were assessed. RESULTS Rats exposed to 12 and 24 mg/mL nicotine vapor displayed higher serum cotinine levels than control rats exposed to 0 mg/mL vapor. There were no differences in impulsive choice between any vapor exposure groups when tested 15 days after exposure, across 6 days of stable responding, suggesting that nicotine vapor does not have long lasting effects on impulsive choice. Interestingly, a subsequent nicotine vapor challenge revealed short-term increases in impulsive choice immediately following a single exposure to 24 mg/mL nicotine vapor, relative to choice preference immediately following exposure to 0 mg/mL vapor. CONCLUSIONS These results suggest that exposure to nicotine vapor causes immediate, short-term increases in impulsive choice. IMPLICATIONS E-cigarette use is increasing at an alarming rate, particularly among adolescents and young adults. This is concerning given the lack of research into the effects of nicotine vapor exposure on the brain and behavior. The present study describes a viable rodent model of human e-cigarette use and suggest that exposure to nicotine vapor produces short-term increases in impulsive choice.
Collapse
Affiliation(s)
- Rodolfo J Flores
- The University of Texas at El Paso, Department of Psychology, El Paso, Texas, USA
| | - Fatima Z Alshbool
- Texas A&M University, Irma Lerma Rangel College of Pharmacy, Department of Pharmacy Practice, Kingsville, Texas, USA
| | - Priscilla Giner
- The University of Texas at El Paso, School of Pharmacy, Department of Pharmaceutical Sciences, El Paso, Texas, USA
| | - Laura E O'Dell
- The University of Texas at El Paso, Department of Psychology, El Paso, Texas, USA
| | - Ian A Mendez
- The University of Texas at El Paso, School of Pharmacy, Department of Pharmaceutical Sciences, El Paso, Texas, USA
| |
Collapse
|
15
|
De Sa Nogueira D, Bourdy R, Filliol D, Romieu P, Befort K. Hippocampal mu opioid receptors are modulated following cocaine self-administration in rat. Eur J Neurosci 2021; 53:3341-3349. [PMID: 33811699 DOI: 10.1111/ejn.15217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 03/18/2021] [Indexed: 11/30/2022]
Abstract
Cocaine addiction is a complex pathology induced by long-term brain changes. Understanding the neurochemical changes underlying the reinforcing effects of this drug of abuse is critical for reducing the societal burden of drug addiction. The mu opioid receptor plays a major role in drug reward. This receptor is modulated by chronic cocaine treatment in specific brain structures, but few studies investigated neurochemical adaptations induced by voluntary cocaine intake. In this study, we investigated whether intravenous cocaine-self administration (0.33 mg/kg/injection, fixed-ratio 1 [FR1], 10 days) in rats induces transcriptional and functional changes of the mu opioid receptor in reward-related brain regions. Epigenetic processes with histone modifications were examined for two activating marks, H3K4Me3, and H3K27Ac. We found an increase of mu opioid receptor gene expression along with a potentiation of its functionality in hippocampus of cocaine self-administering animals compared to saline controls. Chromatin immunoprecipitation followed by qPCR revealed no modifications of the histone mark H3K4Me3 and H3K27Ac levels at mu opioid receptor promoter. Our study highlights the hippocampus as an important target to further investigate neuroadaptive processes leading to cocaine addiction.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Romain Bourdy
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Dominique Filliol
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Pascal Romieu
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| | - Katia Befort
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA UMR7364), Centre de la Recherche Nationale Scientifique, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
16
|
Zhang H, Lipinski AA, Liktor-Busa E, Smith AF, Moutal A, Khanna R, Langlais PR, Largent-Milnes TM, Vanderah TW. The Effects of Repeated Morphine Treatment on the Endogenous Cannabinoid System in the Ventral Tegmental Area. Front Pharmacol 2021; 12:632757. [PMID: 33953672 PMCID: PMC8090348 DOI: 10.3389/fphar.2021.632757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/26/2021] [Indexed: 12/18/2022] Open
Abstract
The therapeutic utility of opioids is diminished by their ability to induce rewarding behaviors that may lead to opioid use disorder. Recently, the endogenous cannabinoid system has emerged as a hot topic in the study of opioid reward but relatively little is known about how repeated opioid exposure may affect the endogenous cannabinoid system in the mesolimbic reward circuitry. In the present study, we investigated how sustained morphine may modulate the endogenous cannabinoid system in the ventral tegmental area (VTA) of Sprague Dawley rats, a critical region in the mesolimbic reward circuitry. Studies here using proteomic analysis and quantitative real-time PCR (qRT-PCR) found that the VTA expresses 32 different proteins or genes related to the endogenous cannabinoid system; three of these proteins or genes (PLCγ2, ABHD6, and CB2R) were significantly affected after repeated morphine exposure (CB2R was only detected by qRT-PCR but not proteomics). We also identified that repeated morphine treatment does not alter either anandamide (AEA) or 2-arachidonoylglycerol (2-AG) levels in the VTA compared to saline treatment; however, there may be diminished levels of anandamide (AEA) production in the VTA 4 h after a single morphine injection in both chronic saline and morphine pretreated cohorts. Treating the animals with an inhibitor of 2-AG degradation significantly decreased repeated opioid rewarding behavior. Taken together, our studies reveal a potential influence of sustained opioids on the endocannabinoid system in the VTA, suggesting that the endogenous cannabinoid system may participate in the opioid-induced reward.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Austin A. Lipinski
- Department of Medicine, Division of Endocrinology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Erika Liktor-Busa
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Angela F. Smith
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Paul R. Langlais
- Department of Medicine, Division of Endocrinology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Tally M. Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Todd W. Vanderah
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
17
|
Ryu IS, Yoon SS, Choi MJ, Lee YE, Kim JS, Kim WH, Cheong JH, Kim HJ, Jang C, Lee YS, Steffensen SC, Ka M, Woo DH, Jang EY, Seo J. The potent psychomotor, rewarding and reinforcing properties of 3-fluoromethamphetamine in rodents. Addict Biol 2020; 25:e12846. [PMID: 31797481 DOI: 10.1111/adb.12846] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 09/11/2019] [Accepted: 10/02/2019] [Indexed: 11/28/2022]
Abstract
3-fluoromethamphetamine (3-FMA), a derivative of methamphetamine (METH), produces behavioral impairment and deficits in dopaminergic transmission in the striatum of mice. The abuse potential of 3-FMA has not been fully characterized. The aim of this study was to evaluate the effects of 3-FMA on locomotor activity as well as its rewarding and reinforcing properties in the conditioned place preference (CPP) and self-administration procedures. Intravenous (i.v.) administration of 3-FMA (0.5 and 1.0 mg/kg) significantly increased locomotor activity in a dose-dependent manner in rats. In the CPP procedure, intraperitoneal administration of 3-FMA (10 and 30 mg/kg) produced a significant alteration in place preference in mice. In the self-administration paradigms, 3-FMA showed drug-taking behavior at the dose of 0.1 mg/kg/infusion (i.v.) during 2 hr sessions under fixed ratio schedules and high breakpoints at the dose of 0.3 and 1.0 mg/kg/infusion (i.v.) during 6 hr sessions under progressive ratio schedule of reinforcement in rats. A priming injection of 3-FMA (0.4 mg/kg, i.v.), METH (0.2 mg/kg, i.v.), or cocaine (2.0 mg/kg, i.v.) reinstated 3-FMA-seeking behavior after an extinction period in 3-FMA-trained rats during 2 hr session. Taken together, these findings demonstrate robust psychomotor, rewarding and reinforcing properties of 3-FMA, which may underlie its potential for compulsive use in humans.
Collapse
Affiliation(s)
- In Soo Ryu
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| | - Seong Shoon Yoon
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| | - Mee Jung Choi
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| | - Young Eun Lee
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| | - Ji Sun Kim
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| | - Woo Hyun Kim
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| | - Jae Hoon Cheong
- Uimyung Research Institute for Neuroscience, School of Pharmacy Sahmyook University Seoul South Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, School of Pharmacy Sahmyook University Seoul South Korea
| | - Choon‐Gon Jang
- Department of Pharmacology, School of Pharmacy Sungkyunkwan University Suwon South Korea
| | - Yong Sup Lee
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy Kyung Hee University Seoul South Korea
| | - Scott C. Steffensen
- Department of Psychology and Neuroscience Brigham Young University Provo UT USA
| | - Minhan Ka
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| | - Dong Ho Woo
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| | - Eun Young Jang
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| | - Joung‐Wook Seo
- Research Center for Convergence Toxicology Korea Institute of Toxicology Daejeon South Korea
| |
Collapse
|
18
|
Fernandes PR, Almeida FB, da Cunha MMMV, Feddern CF, Freese L, Barros HMT. The effects of caffeine on alcohol oral self-administration behavior in rats. Physiol Behav 2020; 223:112966. [DOI: 10.1016/j.physbeh.2020.112966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/30/2020] [Accepted: 05/10/2020] [Indexed: 02/06/2023]
|
19
|
Geste JR, Levin B, Wilks I, Pompilus M, Zhang X, Esser KA, Febo M, O'Dell L, Bruijnzeel AW. Relationship Between Nicotine Intake and Reward Function in Rats With Intermittent Short Versus Long Access to Nicotine. Nicotine Tob Res 2020; 22:213-223. [PMID: 30958557 DOI: 10.1093/ntr/ntz052] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/01/2019] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Tobacco use improves mood states and smoking cessation leads to anhedonia, which contributes to relapse. Animal studies have shown that noncontingent nicotine administration enhances brain reward function and leads to dependence. However, little is known about the effects of nicotine self-administration on the state of the reward system. METHODS To investigate the relationship between nicotine self-administration and reward function, rats were prepared with intracranial self-stimulation electrodes and intravenous catheters. The rats were trained on the intracranial self-stimulation procedure and allowed to self-administer 0.03 mg/kg/infusion of nicotine. All rats self-administered nicotine daily for 10 days (1 hour/day) and were then switched to an intermittent short access (ShA, 1 hour/day) or long access (LgA, 23 hour/day) schedule (2 days/week, 5 weeks). RESULTS During the first 10 daily, 1-hour sessions, nicotine self-administration decreased the reward thresholds, which indicates that nicotine potentiates reward function. After switching to the intermittent LgA or ShA schedule, nicotine intake was lower in the ShA rats than the LgA rats. The LgA rats increased their nicotine intake over time and they gradually consumed a higher percentage of their nicotine during the light phase. The nicotinic acetylcholine receptor (nAChR) antagonist mecamylamine induced a larger increase in reward thresholds (ie, anhedonia) in the LgA rats than the ShA rats. In the LgA rats, nAChR blockade with mecamylamine decreased nicotine intake for 2 hours and this was followed by a rebound increase in nicotine intake. CONCLUSIONS A brief period of nicotine self-administration enhances reward function and a high level of nicotine intake leads to dependence. IMPLICATIONS These animal studies indicate that there is a strong relationship between the level of nicotine intake and brain reward function. A high level of nicotine intake was more rewarding than a low level of nicotine intake and nicotine dependence was observed after long, but not short, access to nicotine. This powerful combination of nicotine reward and withdrawal makes it difficult to quit smoking. Blockade of nAChRs temporarily decreased nicotine intake, but this was followed by a large rebound increase in nicotine intake. Therefore, nAChR blockade might not decrease the use of combustible cigarettes or electronic cigarettes.
Collapse
Affiliation(s)
- Jean R Geste
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Brandon Levin
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Isaac Wilks
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Marjory Pompilus
- Department of Psychiatry, University of Florida, Gainesville, FL
| | - Xiping Zhang
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL
| | - Karyn A Esser
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL.,Department of Neuroscience, University of Florida, Gainesville, FL
| | - Laura O'Dell
- Department of Psychology, University of Texas at El Paso, El Paso, TX
| | - Adriaan W Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL.,Department of Neuroscience, University of Florida, Gainesville, FL
| |
Collapse
|
20
|
Pickens CL, Cook A, Gaeddert B. Dose-dependent effects of alcohol injections on omission-contingency learning have an inverted-U pattern. Behav Brain Res 2020; 392:112736. [PMID: 32497681 DOI: 10.1016/j.bbr.2020.112736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 05/19/2020] [Accepted: 05/26/2020] [Indexed: 12/31/2022]
Abstract
Previous examinations of the long-term effects of alcohol exposure on omission-contingency learning have produced mixed results across different age or sex groups, with evidence for faster learning or no effect. However, none of these experiments made comparisons using the same exposure-dose across the age/sex groups. Here, we exposed rats to 6 weeks of alcohol injections (3 days/week, 1.75 or 3.5 g/kg/24-h, i.p. broken up into 2 injections/day) in adolescent/early adult males or females (PND27-66) or adult males (PND62-101). We then tested the rats in autoshaping and omission-contingency tasks. In contrast to our hypotheses, the low 1.75-g/kg/24-h dose led to slower omission learning and the higher 3.5-g/kg/24-h dose had no effect. There were no age- or sex-differences in omission learning. Additionally, during autoshaping training, rats exposed in adolescence/early adulthood had a faster shift to sign-tracking in their sign-tracking/goal-tracking ratios than rats exposed in adulthood, with no consistent effect of alcohol exposure or sex-differences. Our results suggest complex effects of alcohol on the neural substrates of omission-contingency learning at different doses, which will require future investigation.
Collapse
Affiliation(s)
- Charles L Pickens
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA.
| | - Anna Cook
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| | - Brooke Gaeddert
- Department of Psychological Sciences, Kansas State University, Manhattan, KS, 66506, USA
| |
Collapse
|
21
|
Pattij T, van Mourik Y, Diergaarde L, de Vries TJ. The role of impulsivity as predisposing behavioural trait in different aspects of alcohol self-administration in rats. Drug Alcohol Depend 2020; 212:107984. [PMID: 32371124 DOI: 10.1016/j.drugalcdep.2020.107984] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 03/03/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Therapeutic interventions to promote abstinence and prevent relapse in alcohol use disorder (AUD) are limitedly available. Therefore, targeting risk factors in the onset and maintenance of AUD could pose an interesting alternative treatment strategy. In this regard, over the last decade trait impulsivity has received considerable attention as such a risk factor predisposing substance dependence both in clinical populations and preclinical rodent studies. This study investigated whether different forms of impulsivity (action versus choice) predict distinct stages of instrumental alcohol self-administration, extinction and cue-induced relapse. METHODS Two cohorts of n = 48 rats each were trained in an operant tasks for either impulsive action or impulsive choice. Subsequently, high and low impulsive rats were then tested in an alcohol self-administration and relapse model and following this retested in the impulsivity tasks to evaluate possible changes in impulsivity levels. RESULTS The current data show that neither impulsive action, nor impulsive choice predict the extent to which rats consume alcohol and the extent to which rats are motivated to self-administer alcohol. Moreover, extinction of responding for alcohol and cue-induced relapse was not predicted by impulsivity. Interestingly, rats and most prominently low impulsive rats became more impulsive after the alcohol self-administration procedure. Although due to employed experimental design it is not clear whether this resulted from alcohol consumption or alcohol abstinence. CONCLUSION Together, these findings lend further support for the notion of a unidirectional relationship between self-administration of the depressant drug alcohol and impulsivity.
Collapse
Affiliation(s)
- Tommy Pattij
- Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam UMC, location VU University Medical Center, De Boelelaan 1105, 1081 HZ, Amsterdam, the Netherlands.
| | - Yvar van Mourik
- Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam UMC, location VU University Medical Center, De Boelelaan 1105, 1081 HZ, Amsterdam, the Netherlands
| | - Leontien Diergaarde
- Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam UMC, location VU University Medical Center, De Boelelaan 1105, 1081 HZ, Amsterdam, the Netherlands
| | - Taco J de Vries
- Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam UMC, location VU University Medical Center, De Boelelaan 1105, 1081 HZ, Amsterdam, the Netherlands
| |
Collapse
|
22
|
Schweppe CA, Burzynski C, Jayanthi S, Ladenheim B, Cadet JL, Gardner EL, Xi ZX, van Praag H, Newman AH, Keck TM. Neurochemical and behavioral comparisons of contingent and non-contingent methamphetamine exposure following binge or yoked long-access self-administration paradigms. Psychopharmacology (Berl) 2020; 237:1989-2005. [PMID: 32388619 PMCID: PMC7974824 DOI: 10.1007/s00213-020-05513-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 03/26/2020] [Indexed: 12/22/2022]
Abstract
RATIONALE Abuse of the psychostimulant methamphetamine (METH) can cause long-lasting damage to brain monoaminergic systems and is associated with profound mental health problems for users, including lasting cognitive impairments. Animal models of METH exposure have been useful in dissecting the molecular effects of the drug on cognition, but many studies use acute, non-contingent "binge" administrations of METH which do not adequately approximate human METH use. Long-term METH exposure via long-access (LgA) self-administration paradigms has been proposed to more closely reflect human use and induce cognitive impairments. OBJECTIVE To better understand the role of contingency and patterns of exposure in METH-induced cognitive impairments, we analyzed behavioral and neurochemical outcomes in adult male rats, comparing non-contingent "binge" METH administration with contingent (LgA) METH self-administration and non-contingent yoked partners. RESULTS Binge METH (40 mg/kg, i.p., over 1 day) dramatically altered striatal and hippocampal dopamine, DOPAC, 5-HT, 5-HIAA, BDNF, and TrkB 75 days after drug exposure. In contrast, 6-h LgA METH self-administration (cumulative 24.8-48.9 mg METH, i.v., over 16 days) altered hippocampal BDNF in both contingent and yoked animals but reduced striatal 5-HIAA in only contingent animals. Neurochemical alterations following binge METH administration were not accompanied by cognitive deficits in Morris water maze, novel object recognition, or Y-maze tests. However, contingent LgA METH self-administration resulted in impaired spatial memory in the water maze. CONCLUSIONS Overall, substantial differences in neurochemical markers between METH exposure and self-administration paradigms did not consistently translate to deficits in cognitive tasks, highlighting the complexity of correlating METH-induced neurochemical changes with cognitive outcomes.
Collapse
Affiliation(s)
- Catherine A. Schweppe
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, 333 Cassell Drive, Baltimore, MD 21224, USA,Present address: Department of Neurology, University of California Los Angeles, 635 Charles E Young Drive South, Los Angeles, CA 90095, USA
| | - Caitlin Burzynski
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Eliot L. Gardner
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Henriette van Praag
- Neuroplasticity and Behavior Unit, Laboratory of Neurosciences, National Institute on Aging – Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA,Department of Biomedical Science, Charles E. Schmidt College of Medicine, and Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Thomas M. Keck
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, 333 Cassell Drive, Baltimore, MD 21224, USA,Department of Chemistry & Biochemistry, Department of Molecular & Cellular Biosciences, College of Science and Mathematics, Rowan University, 201 Mullica Hill Road, Glassboro, NJ 08028, USA
| |
Collapse
|
23
|
Groefsema MM, Engels RC, Voon V, Schellekens AF, Luijten M, Sescousse G. Brain responses to anticipating and receiving beer: Comparing light, at-risk, and dependent alcohol users. Addict Biol 2020; 25:e12766. [PMID: 31066137 PMCID: PMC7187239 DOI: 10.1111/adb.12766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/26/2019] [Accepted: 03/30/2019] [Indexed: 12/15/2022]
Abstract
Impaired brain processing of alcohol‐related rewards has been suggested to play a central role in alcohol use disorder. Yet, evidence remains inconsistent and mainly originates from studies in which participants passively observe alcohol cues or taste alcohol. Here, we designed a protocol in which beer consumption was predicted by incentive cues and contingent on instrumental action closer to real life situations. We predicted that anticipating and receiving beer (compared with water) would elicit activity in the brain reward network and that this activity would correlate with drinking level across participants. The sample consisted of 150 beer‐drinking males, aged 18 to 25 years. Three groups were defined based on alcohol use disorders identification test (AUDIT) scores: light drinkers (n = 39), at‐risk drinkers (n = 64), and dependent drinkers (n = 47). fMRI measures were obtained while participants engaged in the beer incentive delay task involving beer‐ and water‐predicting cues followed by real sips of beer or water. During anticipation, outcome notification and delivery of beer compared with water, higher activity was found in a reward‐related brain network including the dorsal medial prefrontal cortex, orbitofrontal cortex, and amygdala. Yet, no activity was observed in the striatum, and no differences were found between the groups. Our results reveal that anticipating, obtaining, and tasting beer activates parts of the brain reward network, but that these brain responses do not differentiate between different drinking levels.
Collapse
Affiliation(s)
- Martine M. Groefsema
- Executive BoardRadboud University, Behavioural Science Institute Nijmegen The Netherlands
| | | | - Valerie Voon
- Cambridge University, Behavioural and Clinical Neuroscience Institute Cambridge United Kingdom
| | | | - Maartje Luijten
- Executive BoardRadboud University, Behavioural Science Institute Nijmegen The Netherlands
| | - Guillaume Sescousse
- Radboud University, Donders Institute for Brain, Cognition and Behaviour Nijmegen The Netherlands
- Centre de Recherche en Neurosciences de Lyon, INSERM U1028, CNRS UMR5292, PSYR2 Team Lyon France
- CH Le Vinatier, Service Universitaire d'Addictologie Bron France
| |
Collapse
|
24
|
Karel P, Van der Toorn A, Vanderschuren L, Guo C, Sadighi Alvandi M, Reneman L, Dijkhuizen R, Verheij MMM, Homberg JR. Ultrahigh-resolution MRI reveals structural brain differences in serotonin transporter knockout rats after sucrose and cocaine self-administration. Addict Biol 2020; 25:e12722. [PMID: 30748070 PMCID: PMC6916608 DOI: 10.1111/adb.12722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 11/21/2018] [Accepted: 01/08/2019] [Indexed: 12/16/2022]
Abstract
Excessive use of cocaine is known to induce changes in brain white and gray matter. It is unknown whether the extent of these changes is related to individual differences in vulnerability to cocaine addiction. One factor increasing vulnerability involves reduced expression of the serotonin transporter (5-HTT). Human studies have shown that inherited 5-HTT downregulation is associated with structural changes in the brain. These genotype-related structural changes may contribute to risk for cocaine addiction. Here, we tested this idea by using ultrahigh-resolution structural magnetic resonance imaging (MRI) on postmortem tissue of 5-HTT-/- and wild-type (5-HTT+/+ ) rats with a history of long access to cocaine or sucrose (control) self-administration. We found that 5-HTT-/- rats, compared with wild-type control animals, self-administered more cocaine, but not sucrose, under long-access conditions. Ultrahigh-resolution structural MRI subsequently revealed that, independent of sucrose or cocaine self-administration, 5-HTT-/- rats had a smaller amygdala. Moreover, we found an interaction between genotype and type of reward for dorsal raphe nucleus volume. The data point to an important but differential role of the amygdala and dorsal raphe nucleus in 5-HTT genotype-dependent vulnerability to cocaine addiction.
Collapse
Affiliation(s)
- Peter Karel
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| | - Annette Van der Toorn
- Biomedical MR Imaging and Spectroscopy Group, Center for Image SciencesUniversity Medical Center Utrecht and Utrecht UniversityUtrechtThe Netherlands
| | - Louk Vanderschuren
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Chao Guo
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| | - Mina Sadighi Alvandi
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| | - Liesbeth Reneman
- Department of Radiology and Nuclear Medicine, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Brain and CognitionUniversity of AmsterdamAmsterdamThe Netherlands
| | - Rick Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image SciencesUniversity Medical Center Utrecht and Utrecht UniversityUtrechtThe Netherlands
| | - Michel M. M. Verheij
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| |
Collapse
|
25
|
Lack of effect of the combination of metyrapone and oxazepam on brain dopamine. Brain Res 2019; 1724:146435. [DOI: 10.1016/j.brainres.2019.146435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/20/2019] [Accepted: 09/02/2019] [Indexed: 01/04/2023]
|
26
|
Marie N, Canestrelli C, Noble F. Role of pharmacokinetic and pharmacodynamic parameters in neuroadaptations induced by drugs of abuse, with a focus on opioids and psychostimulants. Neurosci Biobehav Rev 2019; 106:217-226. [DOI: 10.1016/j.neubiorev.2018.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/29/2018] [Accepted: 06/06/2018] [Indexed: 01/16/2023]
|
27
|
Mavrikaki M, Anastasiadou E, Ozdemir RA, Potter D, Helmholz C, Slack FJ, Chartoff EH. Overexpression of miR-9 in the Nucleus Accumbens Increases Oxycodone Self-Administration. Int J Neuropsychopharmacol 2019; 22:383-393. [PMID: 30989210 PMCID: PMC6545539 DOI: 10.1093/ijnp/pyz015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/11/2019] [Accepted: 04/12/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND There is an urgent need to identify factors that increase vulnerability to opioid addiction to help stem the opioid epidemic and develop more efficient pharmacotherapeutics. MicroRNAs are small non-coding RNAs that regulate gene expression at a posttranscriptional level and have been implicated in chronic drug-taking in humans and in rodent models. Recent evidence has shown that chronic opioid treatment regulates the microRNA miR-9. The present study was designed to test the hypothesis that miR-9 in the nucleus accumbens potentiates oxycodone addictive-like behavior. METHODS We utilized adeno-associated virus (AAV) to overexpress miR-9 in the nucleus accumbens of male rats and tested the effects on intravenous self-administration of the highly abused prescription opioid, oxycodone, in 1-hour short-access followed by 6-h long-access sessions, the latter of which leads to escalation of drug intake. In separate rats, we assessed the effects of nucleus accumbens miR-9 overexpression on mRNA targets including RE1-silencing transcription factor (REST) and dopamine D2 receptor (DRD2), which have been shown to be regulated by drugs of abuse. RESULTS Overexpression of miR-9 in the nucleus accumbens significantly increased oxycodone self-administration compared with rats expressing a control, scrambled microRNA. Analysis of the pattern of oxycodone intake revealed that miR-9 overexpression increased "burst" episodes of intake and decreased the inter-infusion interval. Furthermore, miR-9 overexpression decreased the expression of REST and increased DRD2 in the nucleus accumbens at time points that coincided with behavioral effects. CONCLUSIONS These results suggest that nucleus accumbens miR-9 regulates oxycodone addictive-like behavior as well as the expression of genes that are involved in drug addiction.
Collapse
Affiliation(s)
- Maria Mavrikaki
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts
| | - Eleni Anastasiadou
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Recep A Ozdemir
- Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - David Potter
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts
| | - Carolin Helmholz
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts
| | - Frank J Slack
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Elena H Chartoff
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts,Correspondence: Elena H. Chartoff, PhD, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont 02478, MA ()
| |
Collapse
|
28
|
Leyrer-Jackson JM, Nagy EK, Olive MF. Cognitive deficits and neurotoxicity induced by synthetic cathinones: is there a role for neuroinflammation? Psychopharmacology (Berl) 2019; 236:1079-1095. [PMID: 30368582 PMCID: PMC6486871 DOI: 10.1007/s00213-018-5067-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023]
Abstract
RATIONALE The number of synthetic derivatives of cathinone, the primary psychoactive alkaloid found in Catha edulis (khat), has risen exponentially in the past decade. Synthetic cathinones (frequently referred to as "bath salts") produce adverse cognitive and behavioral sequelae, share similar pharmacological mechanisms of action with traditional psychostimulants, and may therefore trigger similar cellular events that give rise to neuroinflammation and neurotoxicity. OBJECTIVES In this review, we provide a brief overview of synthetic cathinones, followed by a summary of cognitive deficits in animals and humans that have been documented following acute or repeated exposure. We also summarize growing evidence from in vitro and in vivo studies for synthetic cathinone-induced neurotoxicity, and provide a working hypothetic model of potential cellular mechanisms. RESULTS Synthetic cathinones produce varying effects on markers of monoaminergic terminal function and can increase the formation of reactive oxygen and nitrogen species, induce apoptotic signaling, and cause neurodegeneration and cytotoxicity. We hypothesize that these effects result from biochemical events similar to those induced by traditional psychostimulants. However, empirical evidence for the ability of synthetic cathinones to induce neuroinflammatory processes is currently lacking. CONCLUSIONS Like their traditional psychostimulant counterparts, synthetic cathinones appear to induce neurocognitive dysfunction and cytotoxicity, which are dependent on drug type, dose, frequency, and time following exposure. However, additional studies on synthetic cathinone-induced neuroinflammation are clearly needed, as are investigations into the neurochemical and neuroimmune mechanisms underlying their neurotoxic effects. Such endeavors may lead to novel therapeutic avenues to promote recovery in habitual synthetic cathinone users.
Collapse
Affiliation(s)
| | | | - M. Foster Olive
- Correspondence to: M. Foster Olive, Ph.D. Department of Psychology, Arizona State University, 950 S. McAllister Ave. Tempe, AZ 85287 USA, Phone 1-480-727-9557, Fax 1-480-965-8544,
| |
Collapse
|
29
|
Moazen P, Azizi H, Salmanzadeh H, Semnanian S. Adolescent morphine exposure induces immediate and long-term increases in impulsive behavior. Psychopharmacology (Berl) 2018; 235:3423-3434. [PMID: 30350222 DOI: 10.1007/s00213-018-5051-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 09/24/2018] [Indexed: 02/04/2023]
Abstract
RATIONALE Adolescence in humans represents a unique and critical developmental time point associated with increased risk-taking behavior. Converging clinical and epidemiological studies report a peak of drug use during adolescence, leading to the hypothesis that the developing adolescents brain is at risk to lose control over drug intake. Both adolescence and drug abuse are associated with significant cognitive and psychological changes such as lack of impulse control. A simple definition for impulsive behavior is the tendency to act prematurely without foresight. Increase in impulsivity is evident in acute morphine consumption, but to date, little is known with respect to subchronic morphine administration in impulsive behavior, particularly comparing time-dependent effects in adults, young adults, and adolescents. METHODS To evaluate this, adult, young adult, and adolescent rats were treated with a subchronic regimen of morphine or saline during 5 days (s.c.). Thereafter, we examined impulsive behavioral effects of morphine administration, 24 h and 25 days after administration in rats, while responding under a five-choice serial reaction time task (5-CSRTT). RESULTS Subchronic morphine administration increased premature responding 24 h after the last injection of morphine in adult, young adult, and adolescent rats without increasing motor activity but a significant change in motivation in adult and young adult rats only. After 25 days of abstinence, premature responses were significantly increased in comparison with baseline in adolescent rats but not in adults and young adults. CONCLUSION The main conclusion of this study is that morphine exposure in adolescents has a long-term profound effect on motor impulsive behavior later in adulthood. An implication of our findings might be that we should be especially careful about consuming and prescribing opioid drugs in adolescents.
Collapse
Affiliation(s)
- Parisa Moazen
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamed Salmanzadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
30
|
Vassoler FM, Oranges ML, Toorie AM, Byrnes EM. Oxycodone self-administration during pregnancy disrupts the maternal-infant dyad and decreases midbrain OPRM1 expression during early postnatal development in rats. Pharmacol Biochem Behav 2018; 173:74-83. [PMID: 30055180 DOI: 10.1016/j.pbb.2018.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 12/30/2022]
Abstract
Opioid use and abuse has reached epidemic levels in the United States. As these drugs are frequently used by women of reproductive age, there has been a significant increase in the number of infants born to opioid dependent women. Few preclinical studies have examined voluntary opioid intake during pregnancy, and none have used intravenous self-administration. Thus, the purpose of the current set of studies was to utilize a translational model of oxycodone self-administration in rats to determine the effects of oxycodone intake during pregnancy on early postnatal outcomes. Females were trained to intravenously self-administer oxycodone several weeks prior to mating and then continuously throughout pregnancy followed by withdrawal around the time of parturition. Offspring were monitored for weight gain and separation-induced ultrasonic vocalizations (i.e. number of calls) while dams were examined for motivated maternal responding. Neural expression of the mu opioid receptor gene OPRM1 was examined in offspring on postnatal day 1 (PND1). Results indicate that females self-administer oxycodone during pregnancy at levels similar to those observed in cycling females. Postpartum, oxycodone withdrawn females demonstrate impaired maternal responding. In offspring, while no significant group effects were observed on body weight or call number, age-dependent alterations in weight gain and call number correlated with the dams cumulative oxycodone dose during pregnancy. In addition, offspring demonstrated region specific effects of oxycodone exposure on OPRM1 on PND1. Overall, these findings demonstrate that pregnant females will voluntarily self-administer oxycodone at levels similar to cycling females when using a short access model. Further, maternal oxycodone self-administration alters the maternal-offspring dyad in a manner that is dose-dependent and results in sex- and region-specific effects on OPRM1 expression.
Collapse
Affiliation(s)
- Fair M Vassoler
- Cummings School of Veterinary Medicine, Tuft University, North Grafton, MA 02536, United States of America
| | - Michelle L Oranges
- Cummings School of Veterinary Medicine, Tuft University, North Grafton, MA 02536, United States of America
| | - Anika M Toorie
- Cummings School of Veterinary Medicine, Tuft University, North Grafton, MA 02536, United States of America
| | - Elizabeth M Byrnes
- Cummings School of Veterinary Medicine, Tuft University, North Grafton, MA 02536, United States of America.
| |
Collapse
|
31
|
Animal models of binge drinking, current challenges to improve face validity. Neurosci Biobehav Rev 2018; 106:112-121. [PMID: 29738795 DOI: 10.1016/j.neubiorev.2018.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/29/2018] [Accepted: 05/01/2018] [Indexed: 01/08/2023]
Abstract
Binge drinking (BD), i.e., consuming a large amount of alcohol in a short period of time, is an increasing public health issue. Though no clear definition has been adopted worldwide the speed of drinking seems to be a keystone of this behavior. Developing relevant animal models of BD is a priority for gaining a better characterization of the neurobiological and psychobiological mechanisms underlying this dangerous and harmful behavior. Until recently, preclinical research on BD has been conducted mostly using forced administration of alcohol, but more recent studies used scheduled access to alcohol, to model more voluntary excessive intakes, and to achieve signs of intoxications that mimic the human behavior. The main challenges for future research are discussed regarding the need of good face validity, construct validity and predictive validity of animal models of BD.
Collapse
|
32
|
Persons AL, Bradaric BD, Dodiya HB, Ohene-Nyako M, Forsyth CB, Keshavarzian A, Shaikh M, Napier TC. Colon dysregulation in methamphetamine self-administering HIV-1 transgenic rats. PLoS One 2018; 13:e0190078. [PMID: 29293553 PMCID: PMC5749763 DOI: 10.1371/journal.pone.0190078] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023] Open
Abstract
The integrity and function of the gut is impaired in HIV-infected individuals, and gut pathogenesis may play a role in several HIV-associated disorders. Methamphetamine is a popular illicit drug abused by HIV-infected individuals. However, the effect of methamphetamine on the gut and its potential to exacerbate HIV-associated gut pathology is not known. To shed light on this scenario, we evaluated colon barrier pathology in a rat model of the human comorbid condition. Intestinal barrier integrity and permeability were assessed in drug-naïve Fischer 344 HIV-1 transgenic (Tg) and non-Tg rats, and in Tg and non-Tg rats instrumented with jugular cannulae trained to self-administer methamphetamine or serving as saline-yoked controls. Intestinal permeability was determined by measuring the urine content of orally gavaged sugars. Intestinal barrier integrity was evaluated by immunoblotting or immunofluorescence of colon claudin-1 and zonula occludens-1 (ZO-1), two major tight junction proteins that regulate gut epithelial paracellular permeability. Both non-Tg and Tg rats self-administered moderate amounts of methamphetamine. These amounts were sufficient to increase colon permeability, reduce protein level of claudin-1, and reduce claudin-1 and ZO-1 immunofluorescence in Tg rats relative to non-Tg rats. Methamphetamine decreased tight junction immunofluorescence in non-Tg rats, with a similar, but non-significant trend observed in Tg rats. However, the effect of methamphetamine on tight junction proteins was subthreshold to gut leakiness. These findings reveal that both HIV-1 proteins and methamphetamine alter colon barrier integrity, and indicate that the gut may be a pathogenic site for these insults.
Collapse
Affiliation(s)
- Amanda L. Persons
- Department of Psychiatry, Rush University Medical Center, Chicago, IL, United States of America
- Department of Physician Assistant Studies, Rush University Medical Center, Chicago, IL, United States of America
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- * E-mail:
| | - Brinda D. Bradaric
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Health Sciences, Rush University Medical Center, Chicago, IL, United States of America
| | - Hemraj B. Dodiya
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, United States of America
| | - Michael Ohene-Nyako
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, United States of America
| | - Christopher B. Forsyth
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States of America
| | - Ali Keshavarzian
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States of America
| | - Maliha Shaikh
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States of America
| | - T. Celeste Napier
- Department of Psychiatry, Rush University Medical Center, Chicago, IL, United States of America
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States of America
- Department of Pharmacology, Rush University Medical Center, Chicago, IL, United States of America
| |
Collapse
|
33
|
Tolerance to rewarding brain electrical stimulation: Differential effects of contingent and non-contingent activation of parabrachial complex and lateral hypothalamus. Behav Brain Res 2018; 336:15-21. [DOI: 10.1016/j.bbr.2017.08.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 08/14/2017] [Accepted: 08/17/2017] [Indexed: 01/03/2023]
|
34
|
Abstract
Compulsion and impulsivity are both primary features of drug addiction. Based on decades of animal research, we have a detailed understanding of the factors (both environmental and physiological) that influence compulsive drug use, but still know relatively little about the impulsive aspects of drug addiction. This review outlines our current knowledge of the relationship between impulsivity and drug addiction, focusing on cognitive and motor impulsivity, which are particularly relevant to this disorder. Topics to be discussed include the influence of chronic drug administration on impulsivity, the mechanisms that may explain drug-induced impulsivity, and the role of individual differences in the development of impulsive drug use. In addition, the manner in which contemporary theories of drug addiction conceptualize the relationship between impulsivity and compulsion is examined. Most importantly, this review emphasizes a critical role for animal research in understanding the role of impulsivity in the development and maintenance of drug addiction.
Collapse
Affiliation(s)
- Mary C Olmstead
- Department of Psychology, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
35
|
Bosse KE, Oginsky MF, Susick LL, Ramalingam S, Ferrario CR, Conti AC. Adenylyl Cyclase 1 Is Required for Ethanol-Induced Locomotor Sensitization and Associated Increases in NMDA Receptor Phosphorylation and Function in the Dorsal Medial Striatum. J Pharmacol Exp Ther 2017; 363:148-155. [PMID: 28838956 PMCID: PMC5625283 DOI: 10.1124/jpet.117.242321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/08/2017] [Indexed: 11/30/2022] Open
Abstract
Neuroadaptive responses to chronic ethanol, such as behavioral sensitization, are associated with N-methyl-D-aspartate receptor (NMDAR) recruitment. Ethanol enhances GluN2B-containing NMDAR function and phosphorylation (Tyr-1472) of the GluN2B-NMDAR subunit in the dorsal medial striatum (DMS) through a protein kinase A (PKA)-dependent pathway. Ethanol-induced phosphorylation of PKA substrates is partially mediated by calcium-stimulated adenylyl cyclase 1 (AC1), which is enriched in the dorsal striatum. As such, AC1 is poised as an upstream modulator of ethanol-induced DMS neuroadaptations that promote drug responding, and thus represents a therapeutic target. Our hypothesis is that loss of AC1 activity will prevent ethanol-induced locomotor sensitization and associated DMS GluN2B-NMDAR adaptations. We evaluated AC1's contribution to ethanol-evoked locomotor responses and DMS GluN2B-NMDAR phosphorylation and function using AC1 knockout (AC1KO) mice. Results were mechanistically validated with the AC1 inhibitor, NB001. Acute ethanol (2.0 g/kg) locomotor responses in AC1KO and wild-type (WT) mice pretreated with NB001 (10 mg/kg) were comparable to WT ethanol controls. However, repeated ethanol treatment (10 days, 2.5 g/kg) failed to produce sensitization in AC1KO or NB001 pretreated mice, as observed in WT ethanol controls, following challenge exposure (2.0 g/kg). Repeated exposure to ethanol in the sensitization procedure significantly increased pTyr-1472 GluN2B levels and GluN2B-containing NMDAR transmission in the DMS of WT mice. Loss of AC1 signaling impaired ethanol-induced increases in DMS pGluN2B levels and NMDAR-mediated transmission. Together, these data support a critical and specific role for AC1 in striatal signaling that mediates ethanol-induced behavioral sensitization, and identify GluN2B-containing NMDARs as an important AC1 target.
Collapse
Affiliation(s)
- Kelly E Bosse
- Research & Development, John D. Dingell VA Medical Center, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (M.F.O., C.R.F.)
| | - Max F Oginsky
- Research & Development, John D. Dingell VA Medical Center, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (M.F.O., C.R.F.)
| | - Laura L Susick
- Research & Development, John D. Dingell VA Medical Center, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (M.F.O., C.R.F.)
| | - Sailesh Ramalingam
- Research & Development, John D. Dingell VA Medical Center, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (M.F.O., C.R.F.)
| | - Carrie R Ferrario
- Research & Development, John D. Dingell VA Medical Center, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (M.F.O., C.R.F.)
| | - Alana C Conti
- Research & Development, John D. Dingell VA Medical Center, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan (K.E.B., L.L.S., S.R., A.C.C.); and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (M.F.O., C.R.F.)
| |
Collapse
|
36
|
Nona CN, Nobrega JN. A role for nucleus accumbens glutamate in the expression but not the induction of behavioural sensitization to ethanol. Behav Brain Res 2017; 336:269-281. [PMID: 28919158 DOI: 10.1016/j.bbr.2017.09.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/23/2017] [Accepted: 09/13/2017] [Indexed: 12/27/2022]
Abstract
Mechanisms underlying differential sensitivity to behavioural sensitization to ethanol (EtOH) remain poorly understood, although accumulating evidence suggests a role for glutamatergic processes in the ventral striatum. Efforts to address this issue can benefit from the well-documented fact that in any given cohort, some of the mice (High sensitized; HS) show robust sensitization, while others (Low sensitized; LS) show little, if any, sensitization. Here, we examined whether this variability might be differentially associated with nucleus accumbens (NAc) glutamate processes. Male DBA mice received 5 EtOH (2.2g/kg) or saline injections twice a week and were challenged with EtOH (1.8g/kg) 2 weeks after injection 5. When an EtOH challenge was administered 2 weeks following the induction of sensitization, HS, but not LS, mice showed a robust increase in glutamate levels (67%, P<0.01) as measured by in vivo microdialysis. In a separate cohort, the mGlu2/3 agonist LY354740 (10mg/kg), given prior to the EtOH challenge, abolished the expression of sensitization. To ascertain whether enhanced release could also be observed during the induction of sensitization, glutamate levels were measured after the 1st and 5th EtOH injection and were found to be unchanged in HS mice, although briefly elevated in LS mice at injection 5. To further assess possible glutamate involvement during the induction of sensitization, sensitizing EtOH injections were co-administered with NMDAR antagonists. At the doses used, MK-801 (0.25mg/kg) and CGS 19755 (10mg/kg) blocked the expression of sensitization, but did not significantly interfere with the development of EtOH sensitization. Within the limitations of the present design, the results suggest an important role for EtOH-induced glutamate release in the NAc when sensitization is well established, but not necessarily during the development of sensitization.
Collapse
Affiliation(s)
- Christina N Nona
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Behavioural Neurobiology Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | - José N Nobrega
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Behavioural Neurobiology Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Departments of Psychiatry and Psychology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
37
|
Rüedi-Bettschen D, Platt DM. Detrimental effects of self-administered methamphetamine during pregnancy on offspring development in the rat. Drug Alcohol Depend 2017; 177:171-177. [PMID: 28600929 PMCID: PMC5701573 DOI: 10.1016/j.drugalcdep.2017.03.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 03/22/2017] [Accepted: 03/24/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Methamphetamine (METH) abuse by pregnant women is a commonly observed phenomenon. While the harmful effects of METH are well described for adults, there is only limited knowledge of the effects of METH use during pregnancy on the developing child. In the present study, we investigated how intraveneous (iv) METH self-administration throughout pregnancy affected rat dams and their offspring through weaning, compared to controls. METHODS Female rats (n=16) were trained to self-administer METH iv; every drug infusion by a dam also resulted in a saline injection to a yoked control (n=16). When stable levels of self-administration were reached, all females were mated. Daily, 2-h self-administration sessions continued until litters were born. General health and weight was assessed daily in dams and pups. In addition, pups were evaluated for achievement of age-appropriate developmental milestones (i.e., righting reflex, negative geotaxis, pinna detachment, fur appearance, incisor eruption and eye opening). RESULTS Dams self-administered 2-3mg/kg/day METH throughout gestation without consequence to dam health or weight gain during pregnancy. All females produced viable litters, and litter size and composition did not differ between saline and METH dams. Similarly, maternal pup-directed behavior was not affected by prior METH self-administration. However, despite a lack of weight difference in pups, METH-exposed pups were significantly delayed in reaching all assessed developmental milestones compared to controls. CONCLUSION These results indicate that in utero exposure to moderate METH doses can profoundly and adversely affect offspring development, suggesting that even recreational METH use during pregnancy has potential for harm.
Collapse
Affiliation(s)
- Daniela Rüedi-Bettschen
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA.
| | - Donna M. Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. Tel: +1 601-984-5890,Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
38
|
Ugur M, Kaya E, Gozen O, Koylu EO, Kanit L, Keser A, Balkan B. Chronic nicotine-induced changes in gene expression of delta and kappa-opioid receptors and their endogenous ligands in the mesocorticolimbic system of the rat. Synapse 2017; 71. [PMID: 28509375 DOI: 10.1002/syn.21985] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022]
Abstract
Delta and kappa opioid receptors (DOR and KOR, respectively) and their endogenous ligands, proenkephalin (PENK) and prodynorphin (PDYN)-derived opioid peptides are proposed as important mediators of nicotine reward. This study investigated the regulatory effect of chronic nicotine treatment on the gene expression of DOR, KOR, PENK and PDYN in the mesocorticolimbic system. Three groups of rats were injected subcutaneously with nicotine at doses of 0.2, 0.4, or 0.6 mg/kg/day for 6 days. Rats were decapitated 1 hr after the last dose on day six, as this timing coincides with increased dopamine release in the mesocorticolimbic system. mRNA levels in the ventral tegmental area (VTA), lateral hypothalamic area (LHA), amygdala (AMG), dorsal striatum (DST), nucleus accumbens, and medial prefrontal cortex were measured by quantitative real-time PCR. Our results showed that nicotine upregulated DOR mRNA in the VTA at all of the doses employed, in the AMG at the 0.4 and 0.6 mg/kg doses, and in the DST at the 0.4 mg/kg dose. Conversely, PDYN mRNA was reduced in the LHA with 0.6 mg/kg nicotine and in the AMG with 0.4 mg/kg nicotine. KOR mRNA was also decreased in the DST with 0.6 mg/kg nicotine. Nicotine did not regulate PENK mRNA in any brain region studied.
Collapse
Affiliation(s)
- Muzeyyen Ugur
- Department of Physiology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Egemen Kaya
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Oguz Gozen
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Ersin O Koylu
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Lutfiye Kanit
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Aysegul Keser
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Burcu Balkan
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| |
Collapse
|
39
|
Lee BH, Park TY, Lin E, Li H, Yang CH, Choi KH. Altered Acoustic Startle Reflex, Prepulse Inhibition, and Peripheral Brain-Derived Neurotrophic Factor in Morphine Self-Administered Rats. Int J Neuropsychopharmacol 2016; 20:383-391. [PMID: 27927738 PMCID: PMC5417055 DOI: 10.1093/ijnp/pyw107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/23/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Previous studies suggested that opiate withdrawal may increase anxiety and disrupt brain-derived neurotrophic factor function, but the effects of i.v. morphine self-administration on these measures remain unclear. METHODS Adult male Sprague-Dawley rats were implanted with a catheter in the jugular vein. After 1 week of recovery, the animals were allowed to self-administer either i.v. morphine (0.5 mg/kg per infusion, 4 h/d) or saline in the operant conditioning chambers. The acoustic startle reflex and prepulse inhibition were measured at a baseline and on self-administration days 1, 3, 5, and 7 (1- and 3-hour withdrawal). Blood samples were collected on self-administration days 3, 5, and 7 from separate cohorts of animals, and the levels of brain-derived neurotrophic factor and corticosterone were assayed using the enzyme-linked immunosorbent assay method. RESULTS Compared with the saline group, the morphine self-administration group showed hyper-locomotor activity and reduced defecation during the self-administration. The morphine self-administration increased acoustic startle reflex at 1-hour but not 3-hour withdrawal from morphine and disrupted prepulse inhibition at 3-hour but not 1-hour withdrawal. The blood brain-derived neurotrophic factor levels were decreased in the morphine self-administration group at self-administration days 3 and 5, while the corticosterone levels remained unchanged throughout the study. CONCLUSIONS The current findings suggest that spontaneous withdrawal from i.v. morphine self-administration may have transient effects on acoustic startle, sensorimotor gating, and peripheral brain-derived neurotrophic factor levels, and these changes may contribute to the adverse effects of opiate withdrawal.
Collapse
Affiliation(s)
- Bong Hyo Lee
- Department of Acupuncture, Moxibustion and Acupoint, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Lee); Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Lee, Mr Park, Ms Lin, Dr Li, and Dr Choi); Center for Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD (Mr Park, Dr Li, and Dr Choi); Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Yang); Daniel K. Inouye Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Choi)
| | - Thomas Y. Park
- Department of Acupuncture, Moxibustion and Acupoint, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Lee); Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Lee, Mr Park, Ms Lin, Dr Li, and Dr Choi); Center for Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD (Mr Park, Dr Li, and Dr Choi); Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Yang); Daniel K. Inouye Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Choi)
| | - Erica Lin
- Department of Acupuncture, Moxibustion and Acupoint, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Lee); Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Lee, Mr Park, Ms Lin, Dr Li, and Dr Choi); Center for Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD (Mr Park, Dr Li, and Dr Choi); Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Yang); Daniel K. Inouye Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Choi)
| | - He Li
- Department of Acupuncture, Moxibustion and Acupoint, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Lee); Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Lee, Mr Park, Ms Lin, Dr Li, and Dr Choi); Center for Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD (Mr Park, Dr Li, and Dr Choi); Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Yang); Daniel K. Inouye Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Choi)
| | - Chae Ha Yang
- Department of Acupuncture, Moxibustion and Acupoint, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Lee); Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Lee, Mr Park, Ms Lin, Dr Li, and Dr Choi); Center for Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD (Mr Park, Dr Li, and Dr Choi); Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Yang); Daniel K. Inouye Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Choi)
| | - Kwang H. Choi
- Department of Acupuncture, Moxibustion and Acupoint, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Lee); Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Lee, Mr Park, Ms Lin, Dr Li, and Dr Choi); Center for Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD (Mr Park, Dr Li, and Dr Choi); Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea (Dr Yang); Daniel K. Inouye Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, MD (Dr Choi)
| |
Collapse
|
40
|
Response contingency directs long-term cocaine-induced neuroplasticity in prefrontal and striatal dopamine terminals. Eur Neuropsychopharmacol 2016; 26:1667-72. [PMID: 27593624 DOI: 10.1016/j.euroneuro.2016.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/18/2016] [Accepted: 08/22/2016] [Indexed: 11/22/2022]
Abstract
Exposure to addictive substances such as cocaine is well-known to alter brain organisation. Cocaine-induced neuroadaptations depend on several factors, including drug administration paradigm. To date, studies addressing the consequences of cocaine exposure on dopamine transmission have either not been designed to investigate the role of response contingency or focused only on short-term neuroplasticity. We demonstrate a key role of response contingency in directing long-term cocaine-induced neuroplasticity throughout projection areas of the mesocorticolimbic dopamine system. We found enhanced electrically-evoked [(3)H]dopamine release from superfused brain slices of nucleus accumbens shell and core, dorsal striatum and medial prefrontal cortex three weeks after cessation of cocaine self-administration. In yoked cocaine rats receiving the same amount of cocaine passively, sensitised dopamine terminal reactivity was only observed in the nucleus accumbens core. Control sucrose self-administration experiments demonstrated that the observed neuroadaptations were not the result of instrumental learning per se. Thus, long-term withdrawal from cocaine self-administration is associated with widespread sensitisation of dopamine terminals throughout frontostriatal circuitries.
Collapse
|
41
|
Orrù A, Caffino L, Moro F, Cassina C, Giannotti G, Di Clemente A, Fumagalli F, Cervo L. Contingent and non-contingent recreational-like exposure to ethanol alters BDNF expression and signaling in the cortico-accumbal network differently. Psychopharmacology (Berl) 2016; 233:3149-60. [PMID: 27370019 DOI: 10.1007/s00213-016-4358-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 06/09/2016] [Indexed: 01/03/2023]
Abstract
RATIONALE Although brain-derived neurotrophic factor (BDNF) is part of a homeostatic pathway involved in the development of alcohol dependence, it is not clear whether this is also true after recreational ethanol consumption. OBJECTIVES We examined BDNF expression and signaling in the cortico-striatal network immediately and 24 h after either a single intravenous (i.v.) ethanol operant self-administration session or the last of 14 sessions. METHODS To compare contingent and non-contingent ethanol exposure, we incorporated the "yoked control-operant paradigm" in which rats actively taking ethanol (S-Et) were paired with two yoked controls receiving passive infusions of ethanol (Y-Et) or saline. RESULTS A single ethanol exposure transiently reduced BDNF mRNA levels in the medial prefrontal cortex (mPFC) of Y-Et. Immediately after the last of 14 sessions, mRNA and mature BDNF protein levels (mBDNF) were reduced in the mPFC in both S-Et and Y-Et while mBDNF expression was raised in the nucleus accumbens (NAc), suggesting enhanced anterograde transport from the mPFC. Conversely, 24 h later mBDNF expression and signaling were raised in the mPFC and NAc of S-Et rats but reduced in the NAc of Y-Et rats, with concomitant reduction of downstream signaling pathways. CONCLUSIONS Our findings indicate that recreational-like i.v. doses of ethanol promote early changes in neurotrophin expression, depending on the length and modality of administration, the brain region investigated, and the presence of the drug. A rapid intervention targeting the BDNF system might be useful to prevent escalation to alcohol abuse.
Collapse
Affiliation(s)
- Alessandro Orrù
- Experimental Psychopharmacology, Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche 'Mario Negri', Via Giuseppe La Masa 19, 20156, Milan, Italy.
- Institute of Translational Pharmacology (C.N.R.), Parco Scientifico e Tecnologico della Sardegna, Polaris - Edificio 5 - Località, Piscinamanna, 09010, Pula, Cagliari, Italy.
| | - Lucia Caffino
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Federico Moro
- Experimental Psychopharmacology, Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche 'Mario Negri', Via Giuseppe La Masa 19, 20156, Milan, Italy
| | - Chiara Cassina
- Experimental Psychopharmacology, Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche 'Mario Negri', Via Giuseppe La Masa 19, 20156, Milan, Italy
| | - Giuseppe Giannotti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Angelo Di Clemente
- Experimental Psychopharmacology, Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche 'Mario Negri', Via Giuseppe La Masa 19, 20156, Milan, Italy
| | - Fabio Fumagalli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Luigi Cervo
- Experimental Psychopharmacology, Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche 'Mario Negri', Via Giuseppe La Masa 19, 20156, Milan, Italy.
| |
Collapse
|
42
|
Enga RM, Jackson A, Damaj MI, Beardsley PM. Oxycodone physical dependence and its oral self-administration in C57BL/6J mice. Eur J Pharmacol 2016; 789:75-80. [PMID: 27393461 DOI: 10.1016/j.ejphar.2016.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/01/2016] [Accepted: 07/05/2016] [Indexed: 11/30/2022]
Abstract
Abuse of prescription opioids, such as oxycodone, has markedly increased in recent decades. While oxycodone's antinociceptive effects have been detailed in several preclinical reports, surprisingly few preclinical reports have elaborated its abuse-related effects. This is particularly surprising given that oxycodone has been in clinical use since 1917. In a novel oral operant self-administration procedure, C57BL/6J mice were trained to self-administer water before introducing increasing concentrations of oxycodone (0.056-1.0mg/ml) under post-prandial conditions during daily, 3-h test sessions. As the concentration of oxycodone increased, the numbers of deliveries first increased, then decreased in an inverted U-shape fashion characteristic of the patterns of other drugs self-administered during limited access conditions. After post-prandial conditions were removed, self-administration at the highest concentration was maintained suggesting oral oxycodone served as a positive reinforcer. In other mice, using a novel regimen of physical dependence, mice were administered increasing doses of oxycodone (9.0-33.0mg/kg, s.c.) over 9 days, challenged with naloxone (0.1-10.0mg/kg, s.c.), and then observed for 30min. Naloxone dose-dependently increased the observed number of somatic signs of withdrawal, suggesting physical dependence of oxycodone was induced under this regimen. This is the first report demonstrating induction of oral operant self-administration of oxycodone and dose-dependent precipitations of oxycodone withdrawal in C57BL/6J mice. The use of oral operant self-administration as well as the novel physical dependence regimen provides useful approaches to further examine the abuse- and dependence-related effects of this highly abused prescription opioid.
Collapse
Affiliation(s)
- Rachel M Enga
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, VA 23298, USA
| | - Asti Jackson
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, VA 23298, USA
| | - M Imad Damaj
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, VA 23298, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, P.O. Box 980310, Richmond, VA 23298-0310, USA
| | - Patrick M Beardsley
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, VA 23298, USA; Institute for Drug and Alcohol Studies, Virginia Commonwealth University, P.O. Box 980310, Richmond, VA 23298-0310, USA; Center for Biomarker Research and Personalized Medicine, School of Pharmacy, Virginia Commonwealth University, 1112 East Clay Street, Richmond, VA 23298, USA.
| |
Collapse
|
43
|
Le Foll B, Chefer SI, Kimes AS, Stein EA, Goldberg SR, Mukhin AG. Impact of short access nicotine self-administration on expression of α4β2* nicotinic acetylcholine receptors in non-human primates. Psychopharmacology (Berl) 2016; 233:1829-35. [PMID: 26911381 PMCID: PMC4846528 DOI: 10.1007/s00213-016-4250-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 02/16/2016] [Indexed: 10/22/2022]
Abstract
RATIONALE Although nicotine exposure upregulates the α4β2* subtype of nicotinic acetylcholine receptors (nAChRs), the upregulation of nAChRs in non-human primates voluntarily self-administering nicotine has never been demonstrated. OBJECTIVES The objective of the study is to determine if short access to nicotine in a non-human primate model of nicotine self-administration is sufficient to induce nAChRs upregulation. METHODS We combined a nicotine self-administration paradigm with in vivo measure of α4β2* nAChRs using 2-[(18)F]fluoro-A-85380 (2-FA) and positron emission tomography (PET) in six squirrel monkeys. PET measurement was performed before and after intravenous nicotine self-administration (unit dose 10 μg/kg per injection). Monkeys were trained to self-administer nicotine under a fixed-ratio (FR) schedule of reinforcement. Intermittent access (1 h daily per weekday) to nicotine was allowed for 4 weeks and levels of α4β2* nAChRs were measured 4 days later. RESULTS This intermittent access was sufficient to induce upregulation of α4β2* receptors in the whole brain (31 % upregulation) and in specific brain areas (+36 % in amygdala and +62 % in putamen). CONCLUSIONS These results indicate that intermittent nicotine exposure is sufficient to produce change in nAChRs expression.
Collapse
Affiliation(s)
- Bernard Le Foll
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 33 Russell Street, Toronto, ON, Canada, M5S 2S1. .,Departments of Family and Community Medicine, Pharmacology, Psychiatry, Institute of Medical Sciences, University of Toronto, Toronto, Canada. .,Ambulatory Care and Structured Treatment Program, Centre for Addiction and Mental Health, Toronto, Canada. .,Preclinical Pharmacology Section, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, MD, 21224, USA.
| | - Svetlana I. Chefer
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224,The Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Disease, NIH, Fort. Detrick, Frederick, MD, 21702
| | - Alane S. Kimes
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224,Office of the Clinical Director (retired), Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Elliot A. Stein
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Steven R. Goldberg
- Preclinical Pharmacology Section, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Alexey G. Mukhin
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224,Department Psychiatry and Behavioral Sciences and Center for Smoking Cessation, Duke University Medical Center, 2424 Erwin Road, Suite 201, Durham, NC 27705
| |
Collapse
|
44
|
Pascale A, Osera C, Moro F, Di Clemente A, Giannotti G, Caffino L, Govoni S, Fumagalli F, Cervo L. Abstinence from cocaine-self-administration activates the nELAV/GAP -43 pathway in the hippocampus: A stress-related effect? Hippocampus 2016; 26:700-4. [PMID: 26850084 DOI: 10.1002/hipo.22572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2016] [Indexed: 11/06/2022]
Abstract
We previously demonstrated that nELAV/GAP-43 pathway is pivotal for learning and its hippocampal expression is up-regulated by acute stress following repeated cocaine administration. We therefore hypothesized that abstinence-induced stress may sustain nELAV/GAP-43 pathway during early abstinence following 2 weeks of cocaine self-administration. We found that contingent, but not non-contingent, cocaine exposure selectively increases hippocampal nELAV, but not GAP-43, expression immediately after the last self-administration session, an effect that wanes after 24 h and that comes back 7 days later when nELAV activation becomes associated with increased expression of GAP-43, an effect again observed only in animals self-administering the psychostimulant. Such effect is specific for nELAV since the ubiquitous ELAV/HuR is unchanged. This nELAV profile suggests that its initial transient alteration is perhaps related to the daily administration of cocaine, while the increase in the nELAV/GAP-43 pathway following a week of abstinence may reflect the activation of this cascade as a target of stressful conditions associated with drug-related memories. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Cecilia Osera
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Federico Moro
- Experimental Psychopharmacology, Department of Neuroscience, IRCCS-Istituto Di Ricerche Farmacologiche "Mario Negri,", Milan, Italy
| | - Angelo Di Clemente
- Experimental Psychopharmacology, Department of Neuroscience, IRCCS-Istituto Di Ricerche Farmacologiche "Mario Negri,", Milan, Italy
| | - Giuseppe Giannotti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| | - Lucia Caffino
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Fabio Fumagalli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| | - Luigi Cervo
- Experimental Psychopharmacology, Department of Neuroscience, IRCCS-Istituto Di Ricerche Farmacologiche "Mario Negri,", Milan, Italy
| |
Collapse
|
45
|
The pharmacokinetics of methamphetamine self-administration in male and female rats. Drug Alcohol Depend 2015; 150:164-9. [PMID: 25796510 PMCID: PMC4387073 DOI: 10.1016/j.drugalcdep.2015.02.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/25/2015] [Accepted: 02/26/2015] [Indexed: 11/22/2022]
Abstract
BACKGROUND Because methamphetamine (METH) pharmacokinetics after single iv doses show significant differences between male and female rats, we hypothesized that pharmacokinetic differences in METH disposition could be a contributing factor to the patterns of METH self-administration behaviors in rats. METHODS For the studies, we used a passive (non-contingent) METH dosing schedule consisting of 27 METH iv bolus injections (0.048mg/kg) over 2h derived from a previous active (contingent) METH self-administration behavioral study in male rats. After METH dosing of male and female Sprague-Dawley rats (n=5/group), METH and amphetamine serum concentrations were determined by LC-MS/MS. Pharmacokinetic analysis, including predictive mathematical simulations of the data, was then conducted. RESULTS Male and female rats achieved relatively stable METH serum concentrations within 20min, which remained constant from 20 to 120min. While not statistically different, METH clearance and volume of distribution values for females were 25% and 33% lower (respectively) than males. Linear regression analysis of predicted METH concentrations from pharmacokinetic simulations versus observed concentrations showed a substantially better correlation with male data than female data (r(2)=0.71 vs. 0.56; slope=0.95 vs. 0.45, respectively). At 120min, the time of predicted peak METH serum concentrations, female values were 42% higher than expected, while male values were within 3%. CONCLUSIONS Unlike METH male pharmacokinetic data, the female data was less predictable during multiple METH administrations and produced overall higher than expected METH concentrations. These findings demonstrate that METH pharmacokinetics could contribute to differences in METH self-administration behaviors in rats.
Collapse
|
46
|
Mata MM, Napier TC, Graves SM, Mahmood F, Raeisi S, Baum LL. Methamphetamine decreases CD4 T cell frequency and alters pro-inflammatory cytokine production in a model of drug abuse. Eur J Pharmacol 2015; 752:26-33. [PMID: 25678251 DOI: 10.1016/j.ejphar.2015.02.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 01/28/2015] [Accepted: 02/03/2015] [Indexed: 01/10/2023]
Abstract
The reason co-morbid methamphetamine use and HIV infection lead to more rapid progression to AIDS is unclear. We used a model of methamphetamine self-administration to measure the effect of methamphetamine on the systemic immune system to better understand the co-morbidity of methamphetamine and HIV. Catheters were implanted into the jugular veins of male, Sprague Dawley rats so they could self-administer methamphetamine (n=18) or be given saline (control; n=16) for 14 days. One day after the last operant session, blood and spleens were collected. We measured serum levels of pro-inflammatory cytokines, intracellular IFN-γ and TNF-α, and frequencies of CD4(+), CD8(+), CD200(+) and CD11b/c(+) lymphocytes in the spleen. Rats that self-administered methamphetamine had a lower frequency of CD4(+) T cells, but more of these cells produced IFN-γ. Methamphetamine did not alter the frequency of TNF-α-producing CD4(+) T cells. Methamphetamine using rats had a higher frequency of CD8(+) T cells, but fewer of them produced TNF-α. CD11b/c and CD200 expression were unchanged. Serum cytokine levels of IFN-γ, TNF-α and IL-6 in methamphetamine rats were unchanged. Methamphetamine lifetime dose inversely correlated with serum TNF-α levels. Our data suggest that methamphetamine abuse may exacerbate HIV disease progression by activating CD4 T cells, making them more susceptible to HIV infection, and contributing to their premature demise. Methamphetamine may also increase susceptibility to HIV infection, explaining why men who have sex with men (MSM) and frequently use methamphetamine are at the highest risk of HIV infection.
Collapse
Affiliation(s)
- Mariana M Mata
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, United States
| | - T Celeste Napier
- Department of Pharmacology and Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL 60612, United States
| | - Steven M Graves
- Department of Pharmacology and Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL 60612, United States
| | - Fareeha Mahmood
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, United States
| | - Shohreh Raeisi
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, United States
| | - Linda L Baum
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, United States.
| |
Collapse
|
47
|
|
48
|
Brennan KA, Laugesen M, Truman P. Whole tobacco smoke extracts to model tobacco dependence in animals. Neurosci Biobehav Rev 2014; 47:53-69. [PMID: 25064817 DOI: 10.1016/j.neubiorev.2014.07.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/12/2014] [Accepted: 07/14/2014] [Indexed: 01/01/2023]
Abstract
Smoking tobacco is highly addictive and a leading preventable cause of death. The main addictive constituent is nicotine; consequently it has been administered to laboratory animals to model tobacco dependence. Despite extensive use, this model might not best reflect the powerful nature of tobacco dependence because nicotine is a weak reinforcer, the pharmacology of smoke is complex and non-pharmacological factors have a critical role. These limitations have led researchers to expose animals to smoke via the inhalative route, or to administer aqueous smoke extracts to produce more representative models. The aim was to review the findings from molecular/behavioural studies comparing the effects of nicotine to tobacco/smoke extracts to determine whether the extracts produce a distinct model. Indeed, nicotine and tobacco extracts yielded differential effects, supporting the initiative to use extracts as a complement to nicotine. Of the behavioural tests, intravenous self-administration experiments most clearly revealed behavioural differences between nicotine and extracts. Thus, future applications for use of this behavioural model were proposed that could offer new insights into tobacco dependence.
Collapse
Affiliation(s)
- Katharine A Brennan
- School of Psychology, Victoria University of Wellington, PO Box 600, Wellington 6140, New Zealand.
| | - Murray Laugesen
- Health New Zealand Ltd, 36 Winchester St, Lyttelton, Christchurch, New Zealand
| | - Penelope Truman
- Institute of Environmental Science and Research Ltd, PO Box 50348, Porirua 5240, New Zealand
| |
Collapse
|
49
|
Kousik SM, Napier TC, Ross RD, Sumner DR, Carvey PM. Dopamine receptors and the persistent neurovascular dysregulation induced by methamphetamine self-administration in rats. J Pharmacol Exp Ther 2014; 351:432-9. [PMID: 25185214 DOI: 10.1124/jpet.114.217802] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Recently abstinent methamphetamine (Meth) abusers showed neurovascular dysregulation within the striatum. The factors that contribute to this dysregulation and the persistence of these effects are unclear. The current study addressed these knowledge gaps. First, we evaluated the brains of rats with a history of Meth self-administration following various periods of forced abstinence. Micro-computed tomography revealed a marked reduction in vessel diameter and vascular volume uniquely within the striatum between 1 and 28 days after Meth self-administration. Microvessels showed a greater impairment than larger vessels. Subsequently, we determined that dopamine (DA) D2 receptors regulated Meth-induced striatal vasoconstriction via acute noncontingent administration of Meth. These receptors likely regulated the response to striatal hypoxia, as hypoxia inducible factor 1α was elevated. Acute Meth exposure also increased striatal levels of endothelin receptor A and decreased neuronal nitric oxide synthase. Collectively, the data provide novel evidence that Meth-induced striatal neurovascular dysregulation involves DA receptor signaling that results in vasoconstriction via endothelin receptor A and nitric oxide signaling. As these effects can lead to hypoxia and trigger neuronal damage, these findings provide a mechanistic explanation for the selective striatal toxicity observed in the brains of Meth-abusing humans.
Collapse
Affiliation(s)
- Sharanya M Kousik
- Center for Compulsive Behavior and Addiction (S.M.K., T.C.N., P.M.C.), Department of Pharmacology (S.M.K., T.C.N., P.M.C.), Department of Psychiatry (T.C.N.), Department of Neurologic Sciences (P.M.C.), and Department of Anatomy and Cell Biology (R.D.R., D.R.S.), Rush University Medical Center, Chicago, Illinois
| | - T Celeste Napier
- Center for Compulsive Behavior and Addiction (S.M.K., T.C.N., P.M.C.), Department of Pharmacology (S.M.K., T.C.N., P.M.C.), Department of Psychiatry (T.C.N.), Department of Neurologic Sciences (P.M.C.), and Department of Anatomy and Cell Biology (R.D.R., D.R.S.), Rush University Medical Center, Chicago, Illinois
| | - Ryan D Ross
- Center for Compulsive Behavior and Addiction (S.M.K., T.C.N., P.M.C.), Department of Pharmacology (S.M.K., T.C.N., P.M.C.), Department of Psychiatry (T.C.N.), Department of Neurologic Sciences (P.M.C.), and Department of Anatomy and Cell Biology (R.D.R., D.R.S.), Rush University Medical Center, Chicago, Illinois
| | - D Rick Sumner
- Center for Compulsive Behavior and Addiction (S.M.K., T.C.N., P.M.C.), Department of Pharmacology (S.M.K., T.C.N., P.M.C.), Department of Psychiatry (T.C.N.), Department of Neurologic Sciences (P.M.C.), and Department of Anatomy and Cell Biology (R.D.R., D.R.S.), Rush University Medical Center, Chicago, Illinois
| | - Paul M Carvey
- Center for Compulsive Behavior and Addiction (S.M.K., T.C.N., P.M.C.), Department of Pharmacology (S.M.K., T.C.N., P.M.C.), Department of Psychiatry (T.C.N.), Department of Neurologic Sciences (P.M.C.), and Department of Anatomy and Cell Biology (R.D.R., D.R.S.), Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
50
|
Marshell R, Kearney-Ramos T, Brents LK, Hyatt WS, Tai S, Prather PL, Fantegrossi WE. In vivo effects of synthetic cannabinoids JWH-018 and JWH-073 and phytocannabinoid Δ9-THC in mice: inhalation versus intraperitoneal injection. Pharmacol Biochem Behav 2014; 124:40-7. [PMID: 24857780 DOI: 10.1016/j.pbb.2014.05.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/06/2014] [Accepted: 05/14/2014] [Indexed: 11/20/2022]
Abstract
Human users of synthetic cannabinoids (SCBs) JWH-018 and JWH-073 typically smoke these drugs, but preclinical studies usually rely on injection for drug delivery. We used the cannabinoid tetrad and drug discrimination to compare in vivo effects of inhaled drugs with injected doses of these two SCBs, as well as with the phytocannabinoid Δ(9)-tetrahydrocannabinol (Δ(9)-THC). Mice inhaled various doses of Δ(9)-THC, JWH-018 or JWH-073, or were injected intraperitoneally (IP) with these same compounds. Rectal temperature, tail flick latency in response to radiant heat, horizontal bar catalepsy, and suppression of locomotor activity were assessed in each animal. In separate studies, mice were trained to discriminate Δ(9)-THC (IP) from saline, and tests were performed with inhaled or injected doses of the SCBs. Both SCBs elicited Δ(9)-THC-like effects across both routes of administration, and effects following inhalation were attenuated by pretreatment with the CB1 antagonist/inverse agonist rimonabant. No cataleptic effects were observed following inhalation, but all compounds induced catalepsy following injection. Injected JWH-018 and JWH-073 fully substituted for Δ(9)-THC, but substitution was partial (JWH-073) or required relatively higher doses (JWH-018) when drugs were inhaled. These studies demonstrate that the SCBs JWH-018 and JWH-073 elicit dose-dependent, CB1 receptor-mediated Δ(9)-THC-like effects in mice when delivered via inhalation or via injection. Across these routes of administration, differences in cataleptic effects and, perhaps, discriminative stimulus effects, may implicate the involvement of active metabolites of these compounds.
Collapse
Affiliation(s)
- R Marshell
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W, Markham Street - Mail 638, Little Rock, AR 72205-7199, United States
| | - T Kearney-Ramos
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W, Markham Street - Mail 638, Little Rock, AR 72205-7199, United States
| | - L K Brents
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W, Markham Street - Mail 638, Little Rock, AR 72205-7199, United States
| | - W S Hyatt
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W, Markham Street - Mail 638, Little Rock, AR 72205-7199, United States
| | - S Tai
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W, Markham Street - Mail 638, Little Rock, AR 72205-7199, United States
| | - P L Prather
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W, Markham Street - Mail 638, Little Rock, AR 72205-7199, United States
| | - W E Fantegrossi
- Department of Pharmacology & Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W, Markham Street - Mail 638, Little Rock, AR 72205-7199, United States.
| |
Collapse
|