1
|
Laskowski CS, Dorchak DL, Ward KM, Christensen DR, Euston DR. Can Slot-Machine Reward Schedules Induce Gambling Addiction in Rats? J Gambl Stud 2019; 35:887-914. [PMID: 31049772 DOI: 10.1007/s10899-019-09852-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Similar to drugs of abuse, random-ratio reward schedules are highly motivating and, in humans, are thought to foster gambling addiction. Animal gambling models, however, have not yet demonstrated the compulsivity so characteristic of drug addiction. Three criteria have been used to evaluate addiction-like behavior in drug models: (1) response inhibition when reward is not available, (2) persistence under a progressive ratio schedule, in which the response-to-reward ratio is stretched, and (3) persistence in spite of punishment. We tested whether prolonged exposure (6 weeks) to a gambling-like reward schedule would induce addiction-like symptoms in rats. In two studies, separate groups were trained to respond to either random- or fixed-ratio schedules for food reward. We found that rats trained on random-ratio schedules showed higher response rates and dramatically shorter pauses after rewards. Tests of addiction-like behavior, however, were largely negative. Response rates were not different during cued no-reward periods nor when reward was coupled with punishment. We also found no group differences when food was devalued nor in reinstatement of reward-seeking after a 1-week delay. The sole exception to this pattern was that rats in the second experiment showed greater persistence on a progressive ratio test. After experiment two, subjects were also orally administered pramipexole, which caused increased perseveration during progressive ratio testing, especially in the random ratio group. While, it is possible that longer training or more appetitive rewards might have led to addiction-like behavior, our results, on the surface, suggest that random-ratio schedules are motivating but not addictive.
Collapse
Affiliation(s)
- Catherine S Laskowski
- Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, T1K 3M4, Canada
| | - Danika L Dorchak
- Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, T1K 3M4, Canada
| | - Kathleen M Ward
- Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, T1K 3M4, Canada
| | - Darren R Christensen
- Faculty of Health Sciences, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
| | - David R Euston
- Department of Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB, T1K 3M4, Canada.
| |
Collapse
|
2
|
Phillips TJ, Mootz JRK, Reed C. Identification of Treatment Targets in a Genetic Mouse Model of Voluntary Methamphetamine Drinking. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:39-85. [PMID: 27055611 DOI: 10.1016/bs.irn.2016.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Methamphetamine has powerful stimulant and euphoric effects that are experienced as rewarding and encourage use. Methamphetamine addiction is associated with debilitating illnesses, destroyed relationships, child neglect, violence, and crime; but after many years of research, broadly effective medications have not been identified. Individual differences that may impact not only risk for developing a methamphetamine use disorder but also affect treatment response have not been fully considered. Human studies have identified candidate genes that may be relevant, but lack of control over drug history, the common use or coabuse of multiple addictive drugs, and restrictions on the types of data that can be collected in humans are barriers to progress. To overcome some of these issues, a genetic animal model comprised of lines of mice selectively bred for high and low voluntary methamphetamine intake was developed to identify risk and protective alleles for methamphetamine consumption, and identify therapeutic targets. The mu opioid receptor gene was supported as a target for genes within a top-ranked transcription factor network associated with level of methamphetamine intake. In addition, mice that consume high levels of methamphetamine were found to possess a nonfunctional form of the trace amine-associated receptor 1 (TAAR1). The Taar1 gene is within a mouse chromosome 10 quantitative trait locus for methamphetamine consumption, and TAAR1 function determines sensitivity to aversive effects of methamphetamine that may curb intake. The genes, gene interaction partners, and protein products identified in this genetic mouse model represent treatment target candidates for methamphetamine addiction.
Collapse
Affiliation(s)
- T J Phillips
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States; Veterans Affairs Portland Health Care System, Portland, OR, United States.
| | - J R K Mootz
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States
| | - C Reed
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
3
|
Kitanaka N, Kitanaka J, Hall FS, Kayama M, Sugimori H, Uhl GR, Takemura M. Pretreatment or Posttreatment with Aripiprazole Attenuates Methamphetamine-induced Stereotyped Behavior in Mice. J Exp Neurosci 2015; 9:1-10. [PMID: 26525833 PMCID: PMC4623557 DOI: 10.4137/jen.s27733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/15/2015] [Accepted: 06/22/2015] [Indexed: 12/03/2022] Open
Abstract
Aripiprazole is a third-generation atypical antipsychotic and a dopamine D2 receptor partial agonist. In the present study, we investigated whether a single administration of aripiprazole to mice, either as a pretreatment or as a posttreatment, would affect stereotypy induced by methamphetamine (METH). Pretreatment of male ICR mice with aripiprazole (1 or 10 mg/kg, i.p.) attenuated the incidence of METH-induced stereotypical behavior in a dose-dependent manner. Pretreatment of mice with 1 mg/kg aripiprazole produced an increase in the locomotor activity in mice treated with METH compared with mice treated with vehicle plus METH and with 10 mg/kg aripiprazole plus METH. This increase in locomotion is indicative of a rightward shift in the dose–response curve for METH, consistent with a shift in the type of stereotypical behavior observed from biting to sniffing. Aripiprazole posttreatment, after METH-induced stereotypical behavior, was fully expressed and also significantly attenuated overall stereotypy in an aripiprazole dose-dependent manner. These data suggest that the antagonism of METH effects by aripiprazole should be investigated as a potential treatment for acute METH overdose.
Collapse
Affiliation(s)
- Nobue Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - Junichi Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | - F Scott Hall
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Masaru Kayama
- Department of Pharmacology, Hyogo College of Medicine, Hyogo, Japan
| | | | - George R Uhl
- Research Service, New Mexico VA Healthcare System, Albuquerque, NM, USA
| | | |
Collapse
|
4
|
Jupp B, Dalley JW. Convergent pharmacological mechanisms in impulsivity and addiction: insights from rodent models. Br J Pharmacol 2014; 171:4729-66. [PMID: 24866553 PMCID: PMC4209940 DOI: 10.1111/bph.12787] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/02/2014] [Accepted: 05/12/2014] [Indexed: 01/15/2023] Open
Abstract
Research over the last two decades has widely demonstrated that impulsivity, in its various forms, is antecedent to the development of drug addiction and an important behavioural trait underlying the inability of addicts to refrain from continued drug use. Impulsivity describes a variety of rapidly and prematurely expressed behaviours that span several domains from impaired response inhibition to an intolerance of delayed rewards, and is a core symptom of attention deficit hyperactivity disorder (ADHD) and other brain disorders. Various theories have been advanced to explain how impulsivity interacts with addiction both causally and as a consequence of chronic drug abuse; these acknowledge the strong overlaps in neural circuitry and mechanisms between impulsivity and addiction and the seemingly paradoxical treatment of ADHD with stimulant drugs with high abuse potential. Recent years have witnessed unprecedented progress in the elucidation of pharmacological mechanisms underpinning impulsivity. Collectively, this work has significantly improved the prospect for new therapies in ADHD as well as our understanding of the neural mechanisms underlying the shift from recreational drug use to addiction. In this review, we consider the extent to which pharmacological interventions that target impulsive behaviour are also effective in animal models of addiction. We highlight several promising examples of convergence based on empirical findings in rodent-based studies.
Collapse
Affiliation(s)
- B Jupp
- Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of CambridgeCambridge, UK
- Florey Institute of Neuroscience and Mental Health, University of MelbourneParkville, Australia
| | - J W Dalley
- Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of CambridgeCambridge, UK
- Department of Psychiatry, University of CambridgeCambridge, UK
| |
Collapse
|
5
|
Ellenbroek BA. Histamine H₃ receptors, the complex interaction with dopamine and its implications for addiction. Br J Pharmacol 2014; 170:46-57. [PMID: 23647606 DOI: 10.1111/bph.12221] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 04/10/2013] [Accepted: 04/18/2013] [Indexed: 01/08/2023] Open
Abstract
Histamine H₃ receptors are best known as presynaptic receptors inhibiting the release of histamine, as well as other neurotransmitters including acetylcholine and dopamine. However, in the dorsal and ventral striatum, the vast majority of H₃ receptors are actually located postsynaptically on medium sized spiny output neurons. These cells also contain large numbers of dopamine (D₁ and D₂) receptors and it has been shown that H₃ receptors form heterodimers with both D₁ and D₂ receptors. Thus, the anatomical localization of H₃ receptors suggests a complex interaction that could both enhance and inhibit dopaminergic neurotransmission. Dopamine, especially within the striatal complex, plays a crucial role in the development of addiction, both in the initial reinforcing effects of drugs of abuse, as well as in maintenance, relapse and reinstatement of drug taking behaviour. It is, therefore, conceivable that H₃ receptors can moderate the development and maintenance of drug addiction. In the present review, we appraise the current literature on the involvement of H₃ receptors in drug addiction and try to explain these data within a theoretical framework, as well as provide suggestions for further research.
Collapse
Affiliation(s)
- B A Ellenbroek
- School of Psychology, Victoria University, Wellington, New Zealand.
| |
Collapse
|
6
|
Neisewander JL, Cheung THC, Pentkowski NS. Dopamine D3 and 5-HT1B receptor dysregulation as a result of psychostimulant intake and forced abstinence: Implications for medications development. Neuropharmacology 2013; 76 Pt B:301-19. [PMID: 23973315 DOI: 10.1016/j.neuropharm.2013.08.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 07/24/2013] [Accepted: 08/05/2013] [Indexed: 10/26/2022]
Abstract
Addiction to psychostimulants, including cocaine and amphetamine, is associated with dysregulation of dopamine and serotonin (5-HT) neurotransmitter systems. Neuroadaptations in these systems vary depending on the stage of the drug taking-abstinence-relapse cycle. Consequently, the effects of potential treatments that target these systems may vary depending on whether they are given during abstinence or relapse. In this review, we discuss evidence that dopamine D3 receptors (D3Rs) and 5-HT1B receptors (5-HT1BRs) are dysregulated in response to both chronic psychostimulant use and subsequent abstinence. We then review findings from preclinical self-administration models which support targeting D3Rs and 5-HT1BRs as potential medications for psychostimulant dependence. Potential side effects of the treatments are discussed and attention is given to studies reporting positive treatment outcomes that depend on: 1) whether testing occurs during self-administration versus abstinence, 2) whether escalation of drug self-administration has occurred, 3) whether the treatments are given repeatedly, and 4) whether social factors influence treatment outcomes. We conclude that D3/D2 agonists may decrease psychostimulant intake; however, side effects of D3/D2R full agonists may limit their therapeutic potential, whereas D3/D2R partial agonists have fewer undesirable side effects. D3-selective antagonists may not reduce psychostimulant intake during relapse, but nonetheless, may decrease motivation for seeking psychostimulants with relatively few side-effects. 5-HT1BR agonists provide a striking example of treatment outcomes that are dependent on the stage of the addiction cycle. Specifically, these agonists initially increase cocaine's reinforcing effects during maintenance of self-administration, but after a period of abstinence they reduce psychostimulant seeking and the resumption of self-administration. In conclusion, we suggest that factors contributing to dysregulation of monoamine systems, including drug history, abstinence, and social context, should be considered when evaluating potential treatments to better model treatment effects in humans. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Janet L Neisewander
- School of Life Sciences, P.O. Box 874501, Arizona State University, Tempe, AZ 85287-4501, USA.
| | | | | |
Collapse
|
7
|
Coffin PO, Santos GM, Das M, Santos DM, Huffaker S, Matheson T, Gasper J, Vittinghoff E, Colfax GN. Aripiprazole for the treatment of methamphetamine dependence: a randomized, double-blind, placebo-controlled trial. Addiction 2013. [PMID: 23186131 PMCID: PMC3602333 DOI: 10.1111/add.12073] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIMS To test aripiprazole for efficacy in decreasing use in methamphetamine-dependent adults, compared to placebo. DESIGN Participants were randomized to receive 12 weeks of aripiprazole or placebo, with a 3-month follow-up and a platform of weekly 30-minute substance abuse counseling. SETTING The trial was conducted from January 2009 to March 2012 at the San Francisco Department of Public Health. PARTICIPANTS Ninety actively using, methamphetamine-dependent, sexually active adults were recruited from community venues. MEASUREMENTS The primary outcome was regression estimated reductions in weekly methamphetamine-positive urines. Secondary outcomes were study medication adherence [by self-report and medication event monitoring systems (MEMS)], sexual risk behavior and abstinence from methamphetamine. FINDINGS Participant mean age was 38.7 years, 87.8% were male, 50.0% white, 18.9% African American, and 16.7% Latino. Eighty-three per cent of follow-up visits and final visits were completed. By intent-to-treat, participants assigned to aripiprazole had similar reductions in methamphetamine-positive urines as participants assigned to placebo [risk ratio (RR) 0.88, 95% confidence interval (CI): 0.66-1.19, P = 0.41]. Urine positivity declined from 73% (33 of 45 participants) to 45% (18 of 40) in the placebo arm and from 77% (34 of 44) to 44% (20 of 35) in the aripiprazole arm. Adherence by MEMS and self-report was 42 and 74%, respectively, with no significant difference between arms (MEMS P = 0.31; self-report P = 0.17). Most sexual risk behaviors declined similarly among participants in both arms (all P > 0.05). There were no serious adverse events related to study drug, although participants randomized to aripiprazole reported more akathisia, fatigue and drowsiness (P < 0.05). CONCLUSION Compared with placebo, aripiprazole did not reduce methamphetamine use significantly among actively using, dependent adults.
Collapse
Affiliation(s)
- PO Coffin
- San Francisco Department of Public Health,University of California, San Francisco
| | - GM Santos
- San Francisco Department of Public Health,University of California, San Francisco
| | - M Das
- San Francisco Department of Public Health,University of California, San Francisco
| | - DM Santos
- San Francisco Department of Public Health
| | - S Huffaker
- San Francisco Department of Public Health
| | - T Matheson
- San Francisco Department of Public Health
| | - J Gasper
- San Francisco Department of Public Health
| | | | - GN Colfax
- San Francisco Department of Public Health,University of California, San Francisco
| |
Collapse
|
8
|
Baracz SJ, Cornish JL. Oxytocin modulates dopamine-mediated reward in the rat subthalamic nucleus. Horm Behav 2013; 63:370-5. [PMID: 23238104 DOI: 10.1016/j.yhbeh.2012.12.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 12/03/2012] [Accepted: 12/04/2012] [Indexed: 11/29/2022]
Abstract
The subthalamic nucleus (STh) is increasingly recognized as an important region involved in the motivation for drug reward. It is not yet known if dopamine, the neurotransmitter primarily responsible for reward signaling, is also involved in mediating reward-related activity in the STh. The neuropeptide oxytocin acts within the STh to reduce the rewarding effects of the psychostimulant methamphetamine, through a proposed interaction with dopamine. However, the mechanisms of this interaction are unclear. The current study aimed to determine whether (i) dopamine microinjected into the STh would result in a significant place preference following a single-trial conditioning session, (ii) co-administered dopamine receptor antagonist would block the formation of a conditioned place preference (CPP) for dopamine, (iii) co-administered oxytocin would prevent CPP for dopamine and (iv) whether the selective oxytocin antagonist desGly-NH(2),d(CH(2))(5)[D-Tyr(2),Thr(4)]OVT, when co-administered with oxytocin and dopamine, would reverse the effects of oxytocin and result in a CPP for dopamine. Results showed that male Sprague Dawley rats i) formed a preference for the context paired with dopamine (100 nmol/side) administration into the STh, which was prevented by co-administration of ii) the mixed dopamine receptor antagonist fluphenazine (10 nmol/side) or iii) oxytocin (0.6 pmol/side), [corrected] with the oxytocin effect on dopamine CPP reversed by the co-administration of the oxytocin receptor antagonist (3 nmol/side). These data suggest that dopamine neurotransmission in the STh produces rewarding effects that can be reduced by activation of local oxytocin receptors.
Collapse
Affiliation(s)
- Sarah J Baracz
- Department of Psychology, Macquarie University, Sydney, Australia
| | | |
Collapse
|
9
|
Bäckström P, Etelälahti TJ, Hyytiä P. Attenuation of reinforcing and psychomotor stimulant effects of amphetamine by aripiprazole. Addict Biol 2011; 16:55-63. [PMID: 21158016 DOI: 10.1111/j.1369-1600.2010.00223.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Partial dopamine agonists are potential medications for the treatment of amphetamine addiction. They have been hypothesized to stabilize the dopamine system by acting as antagonists during high dopaminergic tone resulting from amphetamine use and as agonists during withdrawal. Aripiprazole is an atypical antipsychotic that acts as a partial D2 dopamine and a serotonin 5-HT₁(A) agonist and a serotonin 5-HT₂(A) antagonist. The aim of the present study was to examine the effects of aripiprazole on behaviors induced and maintained by d-amphetamine. To this end, intravenous d-amphetamine self-administration [fixed ratio 3 (FR3) schedule, 0.02 mg/infusion] and d-amphetamine-induced (0, 1.5 mg/kg intraperitoneally) locomotor activity, as well as spontaneous locomotor activity and sucrose pellet selfadministration (FR3 schedule) were studied in male Wistar rats after aripiprazole (0, 0.3, 1, 3 mg/kg i.p.) administration. Aripiprazole pre-treatment resulted in bidirectional effects on amphetamine self-administration. The 1 mg/kg dose increased, and the highest dose decreased the number of amphetamine infusions. In the locomotor activity experiments, aripiprazole attenuated amphetamine-induced activity dose-dependently and tended to suppress spontaneous activity. The highest aripiprazole doses decreased also sucrose pellet self-administration. The increase in amphetamine self-administration with the intermediate aripiprazole dose, as well as the decrease in amphetamine-induced locomotor activity, suggests that aripiprazole acted as a dopamine antagonist. Suppression of amphetamine and sucrose self-administration by the highest aripiprazole dose was probably caused by non-specific effects. Together, these results indicate that under conditions of dopaminergic stimulation, aripiprazole attenuates the reinforcing and psychomotor stimulant effects of d-amphetamine, but the dose range for this effect is rather limited.
Collapse
Affiliation(s)
- Pia Bäckström
- National Institute for Health and Welfare, Helsinki, Finland.
| | | | | |
Collapse
|
10
|
Tang J, Dani JA. Dopamine enables in vivo synaptic plasticity associated with the addictive drug nicotine. Neuron 2009; 63:673-82. [PMID: 19755109 DOI: 10.1016/j.neuron.2009.07.025] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 05/15/2009] [Accepted: 07/30/2009] [Indexed: 11/29/2022]
Abstract
Addictive drugs induce a dopamine signal that contributes to the initiation of addiction, and the dopamine signal influences drug-associated memories that perpetuate drug use. The addiction process shares many commonalities with the synaptic plasticity mechanisms normally attributed to learning and memory. Environmental stimuli repeatedly linked to addictive drugs become learned associations, and those stimuli come to elicit memories or sensations that motivate continued drug use. Applying in vivo recording techniques to freely moving mice, we show that physiologically relevant concentrations of the addictive drug nicotine directly cause in vivo hippocampal synaptic potentiation of the kind that underlies learning and memory. The drug-induced long-term synaptic plasticity required a local hippocampal dopamine signal. Disrupting general dopamine signaling prevented the nicotine-induced synaptic plasticity and conditioned place preference. These results suggest that dopaminergic signaling serves as a functional label of salient events by enabling and scaling synaptic plasticity that underlies drug-induced associative memory.
Collapse
Affiliation(s)
- Jianrong Tang
- Department of Neuroscience, Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
11
|
Koob GF, Kenneth Lloyd G, Mason BJ. Development of pharmacotherapies for drug addiction: a Rosetta stone approach. Nat Rev Drug Discov 2009; 8:500-15. [PMID: 19483710 DOI: 10.1038/nrd2828] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Current pharmacotherapies for addiction represent opportunities for facilitating treatment and are forming a foundation for evaluating new medications. Furthermore, validated animal models of addiction and a surge in understanding of neurocircuitry and neuropharmacological mechanisms involved in the development and maintenance of addiction - such as the neuroadaptive changes that account for the transition to dependence and the vulnerability to relapse - have provided numerous potential therapeutic targets. Here, we emphasize a 'Rosetta Stone approach', whereby existing pharmacotherapies for addiction are used to validate and improve animal and human laboratory models to identify viable new treatment candidates. This approach will promote translational research and provide a heuristic framework for developing efficient and effective pharmacotherapies for addiction.
Collapse
Affiliation(s)
- George F Koob
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, 10550 North Torrey Pines Road, SP30-2400 La Jolla, California 92037, USA.
| | | | | |
Collapse
|
12
|
Subjective, cognitive/psychomotor, and physiological effects of aripiprazole in Chinese light and heavy smokers. Drug Alcohol Depend 2009; 101:42-52. [PMID: 19070440 DOI: 10.1016/j.drugalcdep.2008.10.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Revised: 10/27/2008] [Accepted: 10/29/2008] [Indexed: 11/20/2022]
Abstract
BACKGROUND Drug addiction researchers have begun to study dopamine partial agonists as potential therapeutic agents. The partial dopamine D(2) receptor agonist aripiprazole has recently been tested as a treatment for stimulant and alcohol dependence in both animal and clinical studies. METHODS A randomized and placebo-controlled pilot clinical study was conducted in a population of Chinese light and heavy smokers to assess the effect of aripiprazole on various responses to smoking. The primary outcomes were subject's ratings on questionnaires of smoking urge, withdrawal syndromes, and cigarette evaluation. Placebo, 5, and 10mg aripiprazole were acutely administered in all participants, with administrations at least 7 days apart. Subjective responses to a smoked cigarette, working memory, and attention/psychomotor performance were assessed before and after drug administration in each experimental session. Abstinence-induced smoking urge, withdrawal symptoms, blood pressure, and heart rate were also measured every 45 min after drug administration. Finally, a cue-testing session was carried out 4h after each drug administration. RESULTS Administration of 10mg aripiprazole significantly decreased both the subjective response and psychological reward derived from smoking a cigarette in heavy smokers. While neither 5 nor 10mg aripiprazole significantly decreased abstinence-induced smoking urges or withdrawal symptoms in light and heavy smokers, these doses substantially attenuated drug cue-induced smoking urges in heavy smokers. Aripiprazole did not affect working memory or attention/psychomotor performance. CONCLUSIONS Light and heavy smokers responded differently to aripiprazole across various dependent measures. Aripiprazole may potentially affect various subjective responses to smoking in heavy smokers.
Collapse
|
13
|
Koob GF. Neurobiological substrates for the dark side of compulsivity in addiction. Neuropharmacology 2008; 56 Suppl 1:18-31. [PMID: 18725236 PMCID: PMC2637927 DOI: 10.1016/j.neuropharm.2008.07.043] [Citation(s) in RCA: 301] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2008] [Revised: 07/29/2008] [Accepted: 07/30/2008] [Indexed: 11/27/2022]
Abstract
Drug addiction can be defined by a compulsion to seek and take drug, loss of control in limiting intake, and the emergence of a negative emotional state when access to the drug is prevented. Drug addiction impacts multiple motivational mechanisms and can be conceptualized as a disorder that progresses from impulsivity (positive reinforcement) to compulsivity (negative reinforcement). The construct of negative reinforcement is defined as drug taking that alleviates a negative emotional state. The negative emotional state that drives such negative reinforcement is hypothesized to derive from dysregulation of key neurochemical elements involved in reward and stress within the basal forebrain structures involving the ventral striatum and extended amygdala. Specific neurochemical elements in these structures include not only decreases in reward neurotransmission, such as decreases in dopamine and opioid peptide function in the ventral striatum, but also recruitment of brain stress systems, such as corticotropin-releasing factor (CRF), in the extended amygdala. Acute withdrawal from all major drugs of abuse produces increases in reward thresholds, increases in anxiety-like responses, and increases in extracellular levels of CRF in the central nucleus of the amygdala. CRF receptor antagonists also block excessive drug intake produced by dependence. A brain stress response system is hypothesized to be activated by acute excessive drug intake, to be sensitized during repeated withdrawal, to persist into protracted abstinence, and to contribute to the compulsivity of addiction. Other components of brain stress systems in the extended amygdala that interact with CRF and may contribute to the negative motivational state of withdrawal include norepinephrine, dynorphin, and neuropeptide Y. The combination of loss of reward function and recruitment of brain stress systems provides a powerful neurochemical basis for a negative emotional state that is responsible for the negative reinforcement driving, at least in part, the compulsivity of addiction.
Collapse
Affiliation(s)
- George F Koob
- Committee on the Neurobiology of Addictive Disorders, Scripps Research Institute, 10550 North Torrey Pines Road, SP30-2400, La Jolla, CA 92037, USA.
| |
Collapse
|
14
|
Abstract
Amfetamine abuse has grown into a worldwide epidemic. Methamfetamine, a derivative of amfetamine made from readily accessible chemicals, has plagued the US since the 1960s, with an alarming recent surge in the numbers of those meeting the criteria for amfetamine abuse and dependence. We review this problem using a computerised literature search (PubMed 1964-2007) to summarise knowledge from animal and human studies about treatments for amfetamine dependence, while exploring the potential of pharmacogenetics to help uncover new treatment targets. Several promising therapeutic targets have come from animal models of reward, drug-taking behaviour and withdrawal. Although preclinical and selected clinical results have been promising, clinical studies have yielded inconsistent results. To improve these outcomes, pharmacogenetic studies may be used to identify candidate alleles that predict therapeutic response. Exciting preclinical findings and a steady progression of clinical results offer hope for the development of a treatment for amfetamine dependence.
Collapse
Affiliation(s)
- Kevin P Hill
- Alcohol and Drug Abuse Treatment Program, McLean Hospital, Belmont, Massachusetts 02478-9106, USA.
| | | |
Collapse
|
15
|
Wee S, Wang Z, Woolverton WL, Pulvirenti L, Koob GF. Effect of aripiprazole, a partial dopamine D2 receptor agonist, on increased rate of methamphetamine self-administration in rats with prolonged session duration. Neuropsychopharmacology 2007; 32:2238-47. [PMID: 17327886 PMCID: PMC2747088 DOI: 10.1038/sj.npp.1301353] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aripiprazole is a dopamine (DA) D(2) receptor partial agonist, approved by the Food and Drug Administration (FDA) for the treatment of schizophrenia. DA receptor partial agonists have been previously assessed as potential therapeutic agents for cocaine dependence. The present experiment examined the effect of aripiprazole on methamphetamine self-administration in a rodent model of an increasing drug self-administration with prolonged session duration. Wistar rats were allowed to self-administer methamphetamine (0.05 mg/kg/injection, intravenously) in either 1-h (short access: ShA rats) or 6-h sessions (long access: LgA rats). After 15 sessions, the dose-response function of methamphetamine was determined under either a progressive- or a fixed-ratio schedule. Next, the effect of aripiprazole (0.3-10 mg/kg, subcutaneuously (s.c.)) on the dose-response function was examined. LgA rats exhibited an increasing rate of methamphetamine self-administration. Responding for methamphetamine by LgA rats was higher than that of ShA rats under both schedules. Pretreatment with aripiprazole shifted the dose-response function of methamphetamine to the right in both LgA and ShA rats. However, the effect of aripiprazole was greater in LgA than ShA rats. In in vitro receptor binding assay, no change in the level of D(2) DA receptors in the nucleus accumbens and the striatum was found in any group. The present data suggest increased sensitivity of the dopaminergic system to aripiprazole in LgA rats compared with ShA rats. However, mechanisms other than downregulation of D(2) DA receptors in the nucleus accumbens and the striatum may be responsible for the increased sensitivity of the dopaminergic function in LgA rats.
Collapse
Affiliation(s)
- Sunmee Wee
- Committee on Neurobiology of Addictive Disorder, The Scripps Research Institute, La Jolla, CA92037, USA.
| | | | | | | | | |
Collapse
|
16
|
Abstract
BACKGROUND Methamphetamine abuse has become an increasing problem in both the United States and globally with concomitant increases in adverse medical, social and environmental sequelae. Behavioral therapies have been used with some success to treat methamphetamine abusers and dependent individuals, but are not universally efficacious. Methamphetamine has a rich pharmacology that theoretically provides many opportunities for potential pharmacotherapeutic intervention. Nevertheless, there are no approved medications with an indication for treating methamphetamine abusers or addicts at this time. AIM To describe briefly how methamphetamine functions and affects function in brain and report how basic researchers and clinicians are attempting to exploit and exploiting this knowledge to discover and develop effective pharmacotherapies. RESULTS Scientifically based approaches to medications development by evaluating medications that limit brain exposure to methamphetamine; modulate methamphetamine effects at vesicular monoamine transporter-2 (VMAT-2); or affect dopaminergic, serotonergic, GABAergic, and/or glutamatergic brain pathways that participate in methamphetamine's reinforcing effects are presented. CONCLUSION The evidence supports the rationale that pharmacotherapies to decrease methamphetamine use, or reduce craving during abstinence may be developed from altering the pharmacokinetics and pharmacodynamics of methamphetamine or its effects on appetitive systems in the brain.
Collapse
Affiliation(s)
- Frank J Vocci
- Division of Pharmacotherapies and Medical Consequences of Drug Abuse, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | | |
Collapse
|
17
|
Farley CM, Baella SA, Wacan JJ, Crawford CA, McDougall SA. Pre- and postsynaptic actions of a partial D2 receptor agonist in reserpinized young rats: Longevity of agonistic effects. Brain Res 2006; 1124:37-44. [PMID: 17070785 DOI: 10.1016/j.brainres.2006.09.068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Revised: 09/19/2006] [Accepted: 09/20/2006] [Indexed: 10/24/2022]
Abstract
Partial D2 receptor agonists (e.g., terguride, preclamol, and aripiprazole) have antagonist-like effects at normosensitive D2 postsynaptic receptors and synthesis modulating autoreceptors. In reserpine-pretreated adult and young rats, however, partial D2 agonists function like high efficacy agonists at D2 postsynaptic receptors and autoreceptors (i.e., terguride increases locomotor activity and decreases dopamine synthesis). The purpose of the present study was to examine the time-course of these pharmacological effects. In all experiments, preweanling rats were given daily injections of reserpine (1 mg/kg, i.p.) or vehicle on postnatal day (PD) 16-PD 20. In the dopamine synthesis experiments, the ability of terguride (0.8 mg/kg) to reduce striatal DOPA accumulation (in NSD-1015 treated rats) was assessed either 5 h or 1, 2, 4, or 8 days (Experiment 1) or 4, 8, 12, 16, 20, or 24 days (Experiment 2) after reserpine pretreatment. In the behavioral experiments, locomotor activity of vehicle or terguride (0.8 mg/kg, i.p.) treated rats was assessed 5 h or 1, 2, 4, or 8 days after the 5-day reserpine regimen. Results from the dopamine synthesis experiments showed that terguride caused agonist-like effects (i.e., decreased DOPA accumulation) at only the 5 h and 1 day time points, although terguride did not induce its normal antagonist-like effects even 20 days after reserpine pretreatment. In the behavioral experiments, terguride stimulated locomotor activity for only the initial 2 days after reserpine pretreatment. The results of the present study show that the agonistic effects of terguride at pre- and postsynaptic receptors are short-lived, but terguride may not exhibit normal antagonistic effects, at least at synthesis modulating autoreceptors, until long after conclusion of reserpine pretreatment.
Collapse
Affiliation(s)
- Cristal M Farley
- Department of Psychology, California State University, San Bernardino, CA 92407, USA
| | | | | | | | | |
Collapse
|
18
|
Hoefer ME, Voskanian SJ, Koob GF, Pulvirenti L. Effects of terguride, ropinirole, and acetyl-l-carnitine on methamphetamine withdrawal in the rat. Pharmacol Biochem Behav 2006; 83:403-9. [PMID: 16647107 DOI: 10.1016/j.pbb.2006.02.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Revised: 02/26/2006] [Accepted: 02/28/2006] [Indexed: 11/30/2022]
Abstract
Withdrawal from psychostimulants, including methamphetamine, induces a depressive state associated with lethargy, dysphoria, hyperphagia and psychomotor retardation. Previous work with repeated administration of amphetamine in rats has shown that amphetamine withdrawal produces decreased motivation to work for a non-drug reward, and this withdrawal is reversed by administration of a dopamine partial agonist. The purpose of the present study was to examine decreased motivation to work for a non-drug reward during methamphetamine withdrawal and explore the effects of a dopamine agonist, dopamine partial agonist, and indirect monoamine agonist on methamphetamine withdrawal. During withdrawal from repeated methamphetamine administration, rats showed reduced responding for a sweet solution in a progressive-ratio schedule of reinforcement, and this effect was significantly more pronounced than previously observed with amphetamine. Repeated systemic treatment with the dopamine partial agonist terguride (0.2 and 0.4 mg/kg, i.p., twice daily), the full dopamine agonist ropinirole (1 mg/kg, i.p., twice daily), and acetyl-L-carnitine (60 and 100 mg/kg, i.p.), a compound with a potential antidepressant effect, during methamphetamine withdrawal restored responding for the sweet solution, suggesting that these drugs may represent potential therapeutic strategies for the treatment of methamphetamine addiction during the withdrawal phase.
Collapse
Affiliation(s)
- Michael E Hoefer
- Molecular and Integrative Neurosciences Department, SP30-2400, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
19
|
Lile JA, Stoops WW, Vansickel AR, Glaser PEA, Hays LR, Rush CR. Aripiprazole attenuates the discriminative-stimulus and subject-rated effects of D-amphetamine in humans. Neuropsychopharmacology 2005; 30:2103-14. [PMID: 15988473 DOI: 10.1038/sj.npp.1300803] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The results of animal research suggest that the use of partial agonists at dopamine (DA) D2 receptors may be an effective strategy for the treatment of stimulant dependence. Aripiprazole is an atypical antipsychotic that has partial agonist activity at D2 receptors. In this experiment, seven human participants with a history of nontherapeutic stimulant use learned to discriminate 15 mg oral D-amphetamine. After acquiring the discrimination (ie > or =80% correct responding on four consecutive sessions), the effects of a range of doses of D-amphetamine (0, 2.5, 5, 10, and 15 mg), alone and in combination with aripiprazole (0 and 20 mg), were assessed. D-Amphetamine alone functioned as a discriminative stimulus, produced prototypical subject-rated drug effects (eg increased ratings of Active, Alert, Energetic) and elevated cardiovascular indices. These effects were generally a function of dose. Aripiprazole alone did not occasion D-amphetamine-appropriate responding or produce subject-rated effects, but modestly impaired performance. Administration of aripiprazole significantly attenuated the discriminative-stimulus and cardiovascular effects of D-amphetamine, as well as some of the subject-rated drug effects. These data are consistent with previous preclinical findings and suggest that DA partial agonists deserve further evaluation as potential pharmacotherapies in the management of stimulant dependence. Future studies should investigate the ability of aripiprazole or related compounds to attenuate the behavioral effects of stimulants associated with a greater degree of dependence, such as methamphetamine or cocaine, in dependent individuals.
Collapse
Affiliation(s)
- Joshua A Lile
- Department of Behavioral Science, College of Medicine, University of Kentucky, Lexington, KY 40536-0086, USA
| | | | | | | | | | | |
Collapse
|
20
|
Vocci F, Ling W. Medications development: Successes and challenges. Pharmacol Ther 2005; 108:94-108. [PMID: 16083966 DOI: 10.1016/j.pharmthera.2005.06.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2005] [Accepted: 06/07/2005] [Indexed: 01/03/2023]
Abstract
The National Institute on Drug Abuse has funded a medications program that has concentrated on the development of medications for opiate and cocaine dependence. Levomethadyl acetate (LAAM) and buprenorphine and buprenorphine/naloxone sublingual tablets were developed in conjunction with pharmaceutical partners and approved by the Food and Drug Administration. The remaining challenges for medications development for opiate dependence involves Phase IV studies in special populations, for example, pregnant opiate-dependent patients, and to translate neuroscience-based findings into treatments. Several marketed medications have shown initial efficacy to reduce cocaine use in well-controlled clinical trials. Disulfiram has been shown to reduce cocaine use in several clinical trials, while baclofen, modafinil, naltrexone, ondansetron, tiagabine, and topiramate have shown preliminary efficacy in initial clinical studies. Confirmatory studies of many of these medications is underway. More recently, the NIDA medications program has evaluated medications for their ability to reduce methamphetamine use. To date, no medications tested have shown efficacy to reduce methamphetamine use. Both marketed medications and investigational agents will be tested. Finally, NIDA has begun to test medications for efficacy to reduce cannabis use. Initial studies are underway. Both agonist and antagonist approaches will be evaluated. Additionally, medications will be tested in cannabis-dependent patients for the management of insomnia, withdrawal, and concurrent depression.
Collapse
Affiliation(s)
- Frank Vocci
- Division of Pharmacotherapies and Medical Consequences of Drug Abuse, National Institute on Drug Abuse, National Institutes of Health, 6001 Executive Boulevard, Ste 4123, MSC 9551, Bethesda, MD 20892, USA.
| | | |
Collapse
|
21
|
Neurotoxins and medicinals for the treatment of Parkinson’s disease. Part 2: dopamine receptors and their agonists. Pharm Chem J 2005. [DOI: 10.1007/s11094-006-0014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
22
|
McDougall SA, Hernandez RM, Reichel CM, Farley CM. The partial D2-like dopamine receptor agonist terguride acts as a functional antagonist in states of high and low dopaminergic tone: evidence from preweanling rats. Psychopharmacology (Berl) 2005; 178:431-9. [PMID: 15765258 DOI: 10.1007/s00213-004-2033-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2004] [Accepted: 09/07/2004] [Indexed: 10/26/2022]
Abstract
RATIONALE In adult rats, the partial D(2)-like agonist terguride acts as an antagonist at normosensitive D(2)-like post-synaptic receptors, while it acts as an agonist at the same receptors during states of low dopaminergic tone. OBJECTIVE The purpose of the present study was to determine whether partial D(2)-like agonists exhibit both antagonistic and agonistic actions during the preweanling period. METHODS In experiments 1 and 2 (examining the agonistic actions of terguride), preweanling rats were either given an escalating regimen of amphetamine to induce a state of amphetamine withdrawal or pretreated with the tyrosine hydroxylase inhibitor AMPT. Distance traveled was measured after rats were injected with saline, terguride (0.4-1.6 mg/kg), or the full D(2)-like receptor agonist NPA (0.01 mg/kg). In experiment 3 (examining the antagonistic actions of terguride), preweanling rats were pretreated with terguride 30 min before they were tested with saline, NPA (0.05 mg/kg), or amphetamine (1.5 mg/kg). RESULTS NPA had an exaggerated locomotor activating effect when tested under conditions of amphetamine withdrawal, while the partial D(2)-like agonist did not enhance distance traveled under any circumstance. Similarly, NPA increased and terguride did not affect the distance-traveled scores of AMPT-pretreated rats. In experiment 3, terguride pretreatment significantly reduced the distance traveled of amphetamine-treated and NPA-treated rats. CONCLUSIONS The behavioral evidence indicates that, during the preweanling period, terguride antagonizes D(2)-like post-synaptic receptors in a state of high dopaminergic tone; however, there is no evidence that terguride is capable of stimulating D(2)-like post-synaptic receptors during states of low dopaminergic tone.
Collapse
Affiliation(s)
- Sanders A McDougall
- Department of Psychology, California State University, San Bernardino, CA, 92407, USA.
| | | | | | | |
Collapse
|
23
|
Bowyer JF, Delongchamp RR, Jakab RL. Glutamate N-methyl-d-aspartate and dopamine receptors have contrasting effects on the limbic versus the somatosensory cortex with respect to amphetamine-induced neurodegeneration. Brain Res 2004; 1030:234-46. [PMID: 15571672 DOI: 10.1016/j.brainres.2004.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2004] [Indexed: 01/02/2023]
Abstract
The roles that glutamate N-methyl-D-aspartate (NMDA) and dopamine D1-like and D2-like receptors play in the cortical neurotoxicity occurring in rats exposed to multiple doses of amphetamine (AMPH) for 2 days was evaluated. Neurodegeneration in rats that did not become hyperthermic during AMPH exposure was quantified by counting isolectin B4-labeled phagocytic microglia and Fluoro-Jade (F-J)-labeled neurons in the somatosensory parietal cortex, piriform cortex and posterolateral cortical amygdaloid nucleus (PLCo). The NMDA receptor antagonist, dizocilpine (0.63 mg/kg day) blocked AMPH-induced neurodegeneration in the somatosensory cortex. However, it did not affect degeneration in the piriform cortex and PLCo indicating that limbic degeneration was not NMDA-mediated. The dopamine antagonists, eticlopride (D2/3, 0.25 mg/kg day) and SCH-23390 (D1, 0.25 mg/kg day), blocked the stereotypic behavior and neurodegeneration in the somatosensory cortex. However, eticlopride had a lesser protective effect in the limbic regions. As well, the dopamine D2/D3 agonist quinpirole (1.5 mg/kg day) protected against cortical neurodegeneration when it was given during AMPH exposure and continued until sacrifice. The dopamine D1 agonist (SKF-38393, 12.5 mg/kg day) had no significant effect on neurodegeneration. These data indicate that there are significant differences in NMDA and dopamine D2 modulation of AMPH-induced neurodegeneration in the somatosensory cortex compared to the limbic cortices, and limbic cortical degeneration is not necessarily dependent on excessive stimulation of NMDA receptors as it is in the somatosensory cortex. Although excessive dopamine receptor stimulation during amphetamine exposure may trigger the neurodegenerative processes, continued D2 stimulation after AMPH exposure is neuroprotective in the cortex.
Collapse
Affiliation(s)
- John F Bowyer
- Division of Neurotoxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079, USA.
| | | | | |
Collapse
|
24
|
Segregation of amphetamine reward and locomotor stimulation between nucleus accumbens medial shell and core. J Neurosci 2003. [PMID: 12867514 DOI: 10.1523/jneurosci.23-15-06295.2003] [Citation(s) in RCA: 171] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Convergent evidence suggests that amphetamine (AMPH) exerts its rewarding and locomotor stimulating effects via release of dopamine in the nucleus accumbens. However, there is no consensus as to the relative contributions of core and medial shell subregions to these effects. Moreover, the literature is based primarily on intracranial administration, which cannot fully mimic the drug distribution achieved by systemic administration. In the present study, the effects of bilateral 6-hydroxydopamine lesions of the accumbens core or medial shell on rewarding and locomotor stimulating effects of systemically administered amphetamine (0.75 mg/kg, i.p.) were examined in a conditioned place preference (CPP) procedure relying solely on tactile cues (floor texture). Residual dopamine innervation was quantified by [125I]-RTI-55 binding to the dopamine transporter. When lesions were performed before the conditioning phase, AMPH-induced locomotor stimulation and CPP magnitude were positively correlated with residual dopamine transporter binding in core and medial shell, respectively. Medial shell lesions did not affect morphine CPP, arguing that a sensory or mnemonic deficit was not responsible for the lesion-induced reduction in AMPH CPP. Medial shell lesions performed between the conditioning phase and the test day reduced the expression of amphetamine CPP. These results suggest that after systemic amphetamine administration, rewarding and locomotor stimulating effects of the drug are anatomically dissociated within the nucleus accumbens: the medial shell contributes to rewarding effects, whereas the core contributes to behavioral activation.
Collapse
|
25
|
De Vries TJ, Homberg JR, Binnekade R, Raasø H, Schoffelmeer ANM. Cannabinoid modulation of the reinforcing and motivational properties of heroin and heroin-associated cues in rats. Psychopharmacology (Berl) 2003; 168:164-169. [PMID: 12669182 DOI: 10.1007/s00213-003-1422-1] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2002] [Accepted: 01/26/2003] [Indexed: 10/26/2022]
Abstract
RATIONALE Recently, we provided evidence for a cannabinoid mechanism in relapse to cocaine seeking in rats. There is also increasing evidence for functional cross-talk between cannabinoid and opioid systems in several physiological processes. OBJECTIVES This study was designed to evaluate whether the cannabinoid system plays a role in mediating the reinforcing and motivational effects of heroin and heroin-paired stimuli. METHODS Male Wistar rats were trained to self-administer heroin (50 microg/kg per infusion) on fixed (FR5) or progressive ratio schedules of reinforcement in the presence of a discriminative and discrete heroin-associated cue. The selective cannabinoid CB1 antagonist SR141716A was given 30 min before the session to determine its effect on responding for heroin. Separate groups of rats were subjected to extinction training during which heroin-associated cues were absent and no heroin was delivered. During subsequent reinstatement tests, the effects of the cannabinoid agonist HU210 and the antagonist SR141716A on reinstatement of heroin seeking were evaluated. RESULTS The cannabinoid antagonist dose-dependently reduced responding for heroin on the FR5 schedule and to a greater extent on the progressive ratio schedule. HU210 (20 microg/kg) reinstated heroin seeking behaviour following a 2-week extinction period, whereas SR141716A dose-dependently attenuated heroin seeking that was provoked by a priming injection of heroin (0.25 mg/kg) and heroin seeking that was triggered by re-exposure to heroin paired stimuli. CONCLUSIONS The results show that the reinforcing and motivational effects of heroin and heroin-paired stimuli are mediated, at least in part, by activation of cannabinoid CB1 receptors. Therefore, the present study provides a rationale for the use of cannabinoid antagonists in the treatment of opiate addiction.
Collapse
MESH Headings
- Animals
- Behavior, Addictive/chemically induced
- Behavior, Addictive/prevention & control
- Cannabinoids/pharmacology
- Cues
- Dose-Response Relationship, Drug
- Heroin/administration & dosage
- Male
- Motivation
- Rats
- Rats, Wistar
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/physiology
- Reinforcement, Psychology
- Self Administration
Collapse
Affiliation(s)
- Taco J De Vries
- Research Institute Neurosciences Vrije Universiteit, Drug Abuse Program, Department of Medical Pharmacology, VU Medical Center, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands.
| | - Judith R Homberg
- Research Institute Neurosciences Vrije Universiteit, Drug Abuse Program, Department of Medical Pharmacology, VU Medical Center, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
| | - Rob Binnekade
- Research Institute Neurosciences Vrije Universiteit, Drug Abuse Program, Department of Medical Pharmacology, VU Medical Center, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
| | - Halfdan Raasø
- Research Institute Neurosciences Vrije Universiteit, Drug Abuse Program, Department of Medical Pharmacology, VU Medical Center, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
| | - Anton N M Schoffelmeer
- Research Institute Neurosciences Vrije Universiteit, Drug Abuse Program, Department of Medical Pharmacology, VU Medical Center, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Platt DM, Rodefer JS, Rowlett JK, Spealman RD. Suppression of cocaine- and food-maintained behavior by the D2-like receptor partial agonist terguride in squirrel monkeys. Psychopharmacology (Berl) 2003; 166:298-305. [PMID: 12589523 DOI: 10.1007/s00213-002-1347-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2002] [Accepted: 11/08/2002] [Indexed: 10/20/2022]
Abstract
RATIONALE The D2-like receptor partial agonist terguride has a profile of behavioral effects in rats that suggests potential benefit as a pharmacotherapy for cocaine addiction. OBJECTIVES The present study investigated the effects of terguride on cocaine- and food-maintained behavior in squirrel monkeys. METHODS Squirrel monkeys were trained to respond on a second-order schedule (FI 10 min, FR 10 or 30:S) of either i.v. cocaine injection or food pellet delivery. Additional monkeys were studied using quantitative observational techniques to construct side effect profiles. Under each procedure, the effects of terguride were compared with those of the reference D2-like receptor antagonist nemonapride and the D2-like receptor full agonist quinpirole. RESULTS Terguride and nemonapride, but not quinpirole, dose-dependently reduced cocaine-maintained behavior. In animals self-administering food, terguride decreased response rates at doses lower than those required to suppress cocaine-maintained behavior. Effective doses of terguride had no systematic effect on motor activity or muscle rigidity, whereas effective doses of nemonapride virtually eliminated motor activity and induced severe catalepsy. The primary observable effects of terguride were a modest increase in self-directed behavior (a D2-receptor agonist-like effect) at intermediate doses and a small increase in static posture (a D2-receptor antagonist-like effect) at the highest dose tested. CONCLUSIONS The results suggest that terguride has advantages over conventional D2-like receptor antagonists and agonists as a candidate pharmacotherapy for cocaine abuse; however, terguride significantly reduces food-maintained behavior at lower doses than those needed to decrease cocaine-maintained behavior suggesting limitations on the utility of terguride as a medication for cocaine addiction.
Collapse
Affiliation(s)
- Donna M Platt
- New England Regional Primate Research Center, Harvard Medical School, One Pine Hill Drive, PO Box 9102, MA 01772-9102, Southborough, USA.
| | | | | | | |
Collapse
|
27
|
Stuber GD, Evans SB, Higgins MS, Pu Y, Figlewicz DP. Food restriction modulates amphetamine-conditioned place preference and nucleus accumbens dopamine release in the rat. Synapse 2002; 46:83-90. [PMID: 12211086 DOI: 10.1002/syn.10120] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Food restriction has been shown to increase self-administration of psychostimulants, including cocaine and amphetamine (AMPH). Consistent with this, food-restricted rats are more sensitized to the rewarding effects of cocaine as measured by conditioned place preference (CPP). This study investigated whether moderate food restriction in rats (15 g/day) results in an increased CPP, relative to ad libitum-fed controls, to a second psychostimulant, AMPH. Conditioning trials consisted of six alternating injections of i.p. AMPH (0.425-6.8 mg/kg) and i.p. saline, paired with distinct environments. On Day 7, a drug-free 20-min choice test for environment was carried out to assess CPP. 0.85 mg/kg AMPH significantly increased CPP in food-restricted vs. ad libitum-fed rats. At 1.7 and 3.4 mg/kg AMPH, food-restricted rats showed decreased CPP, but increased locomotor activity, relative to ad libitum fed controls. To evaluate whether an alteration in extracellular fluid DA levels in the nucleus accumbens (NAc) core could account for these behavioral alterations, DA release was measured by microdialysis. DA release to a single acute i.p. injection of either 0.85 or 1.7 mg/kg AMPH was comparable in food-restricted and ad libitum fed rats. However, ad libitum fed rats demonstrated conditioned DA release after an AMPH conditioning paradigm analogous to the CPP paradigm, whereas food-restricted rats demonstrated no conditioned DA release. In conclusion, altered DA release in the nucleus accumbens core is not a primary effect of moderate food restriction and cannot completely account for either the altered CPP behavior or enhanced locomotor activity observed in this study.
Collapse
Affiliation(s)
- Garret D Stuber
- Department of Psychology, University of Washington, Seattle, Washington 98195-1525, USA
| | | | | | | | | |
Collapse
|
28
|
Homberg JR, van den Akker M, Raasø HS, Wardeh G, Binnekade R, Schoffelmeer ANM, de Vries TJ. Enhanced motivation to self-administer cocaine is predicted by self-grooming behaviour and relates to dopamine release in the rat medial prefrontal cortex and amygdala. Eur J Neurosci 2002; 15:1542-50. [PMID: 12028365 DOI: 10.1046/j.1460-9568.2002.01976.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Rats, like humans, show strong individual differences in their response to anxiogenic and stressful stimuli. In the present study we evaluated whether differences in stress-induced self-grooming behaviour may predict an individual's vulnerability to engage in drug self-administration behaviour. From a population of Wistar rats, the lower and upper quartile with respect to time spent self-grooming on an elevated plus maze (EPM) were selected and trained to intravenously self-administer cocaine under fixed and progressive ratio schedules of reinforcement. High grooming (HG) rats reached considerably higher breakpoints than low grooming (LG) rats but showed no differences in acquisition rate and dose-response relationships. Further, EPM exposure elicited higher anxiety levels and enhanced plasma corticosterone secretion in HG rats. In addition, HG rats did not display enhanced novelty-seeking and still spent more time self-grooming during an EPM re-test following the cocaine self-administration procedure, indicating that stress-induced self-grooming is a stable behavioural trait marker. Neurochemically, electrically evoked [(3)H]dopamine release in vitro was profoundly lower in brain slices from the substantia nigra, medial prefrontal cortex and amygdala of naive HG rats as compared to LG rats, whereas no differences were found in the nucleus accumbens shell and core, the ventral tegmental area and caudate putamen. In conclusion, stress-induced self-grooming specifically predicts enhanced motivation to self-administer cocaine rather than sensitivity to its reinforcing effects. Responsiveness of dopaminergic nerve terminals in the medial prefrontal cortex and amygdala may represent pre-existing underlying factors.
Collapse
Affiliation(s)
- Judith R Homberg
- Graduate School Neuroscience Amsterdam, Research Institute Neurosciences Vrije Universiteit, Drug Abuse Program, Department of Medical Pharmacology, VU Medical Center, Van Der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Orsini C, Izzo E, Koob GF, Pulvirenti L. Blockade of nitric oxide synthesis reduces responding for cocaine self-administration during extinction and reinstatement. Brain Res 2002; 925:133-40. [PMID: 11792361 DOI: 10.1016/s0006-8993(01)03267-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Nitric oxide is a gaseous neurotransmitter that plays a significant role in various forms of synaptic plasticity and may play a role in the behavioral effects of psychostimulant drugs and in cocaine addiction. The course of drug addiction consists of different phases. Relapse into drug-seeking behavior following a period of abstinence is believed to represent one of the major factors leading to the perpetuation of the addictive cycle. In this respect, experimental extinction procedures provide a measure of the motivational properties of drugs as reflected by the persistence of drug-seeking behavior in the absence of the drug and by the reinstatement of responding by non-contingent drug administration. Pretreatment with the nitric oxide synthase inhibitor L-NG-nitroarginine methyl ester (L-NAME, 50 mg/kg IP twice daily for 4 days) impaired responding for cocaine self-administration when the drug was available and the increase of drug-seeking behavior upon abrupt cessation of cocaine availability observed in control rats was significantly reduced after treatment with L-NAME. In addition, the priming effect of a non-contingent injection of cocaine on extinguished cocaine self-administration was also diminished by the same treatment. The acquisition of cocaine self-administration, in contrast, was not affected by treatment with L-NAME. These observations lend further support to the hypothesis of the involvement of nitric oxide in cocaine addiction and extend previous findings to components of the cocaine addictive cycle associated with relapse.
Collapse
Affiliation(s)
- Cristina Orsini
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|