1
|
Blockade of dopamine D3 receptor in ventral tegmental area attenuating contextual fear memory. Biomed Pharmacother 2023; 158:114179. [PMID: 36592493 DOI: 10.1016/j.biopha.2022.114179] [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: 11/05/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
The abnormal fear memory will lead to the onset of stress disorders, such as post-traumatic stress disorder (PTSD) and so on. Therefore, the intervention in the formation of abnormal fear memory will provide a new strategy for the prevention and treatment of PTSD. In our previous studies, we found that blockade of dopamine D3 receptor (DRD3) with highly selective antagonist YQA14 or knockout of DRD3 was able to attenuate the expression or retrieval of fear memory in PTSD animal models. However, the neurobiological mechanism of regulation of DRD3 in fear is unclear. In the present research, we clarified that DRD3 was expressed in the dopaminergic (DAergic) neurons in the ventral tegmental area (VTA). Then, we identified that microinjection of YQA14 (1 μg/0.2 μl/side) in VTA before the aversive stimuli in the training session or during days subsequent to the shock significantly meliorated the freezing behaviors in the inescapable electric foot-shock model. At last, using fiber photometry system, we found that microinjection of YQA14 in VTA promoted the dopamine neurotransmitter release in the basolateral amygdala (BLA), and pre-training YQA14 infusion in VTA lowered the increase of dopamine (DA) in BLA induced by shock during the training session or by context during the retrieval session. All above the results demonstrated that YQA14 attenuated the fear learning through the blockade of DRD3 in VTA decreasing the excitability of the projection to BLA. This study may provide new mechanisms and potential intervention targets for stress disorders with abnormal fear memory.
Collapse
|
2
|
Vamvakopoulou IA, Fonville L, Hayes A, McGonigle J, Elliott R, Ersche KD, Flechais R, Orban C, Murphy A, Smith DG, Suckling J, Taylor EM, Deakin B, Robbins TW, Nutt DJ, Lingford-Hughes AR, Paterson LM. Selective D3 receptor antagonism modulates neural response during negative emotional processing in substance dependence. Front Psychiatry 2022; 13:998844. [PMID: 36339857 PMCID: PMC9627287 DOI: 10.3389/fpsyt.2022.998844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Negative affective states contribute to the chronic-relapsing nature of addiction. Mesolimbic dopamine D3 receptors are well placed to modulate emotion and are dysregulated in substance dependence. Selective antagonists might restore dopaminergic hypofunction, thus representing a potential treatment target. We investigated the effects of selective D3 antagonist, GSK598809, on the neural response to negative emotional processing in substance dependent individuals and healthy controls. Methodology Functional MRI BOLD response was assessed during an evocative image task, 2 h following acute administration of GSK598809 (60 mg) or placebo in a multi-site, double-blind, pseudo-randomised, cross-over design. Abstinent drug dependent individuals (DD, n = 36) comprising alcohol-only (AO, n = 19) and cocaine-alcohol polydrug (PD, n = 17) groups, and matched controls (n = 32) were presented with aversive and neutral images in a block design (contrast of interest: aversive > neutral). Whole-brain mixed-effects and a priori ROI analyses tested for group and drug effects, with identical models exploring subgroup effects. Results No group differences in task-related BOLD signal were identified between DD and controls. However, subgroup analysis revealed greater amygdala/insular BOLD signal in PD compared with AO groups. Following drug administration, GSK598809 increased BOLD response across HC and DD groups in thalamus, caudate, putamen, and pallidum, and reduced BOLD response in insular and opercular cortices relative to placebo. Multivariate analyses in a priori ROIs revealed differential effects of D3 antagonism according to subgroup in substantia nigra; GSK598809 increased BOLD response in AO and decreased response in PD groups. Conclusion Acute GSK598809 modulates the BOLD response to aversive image processing, providing evidence that D3 antagonism may impact emotional regulation. Enhanced BOLD response within D3-rich mesolimbic regions is consistent with its pharmacology and with attenuation of substance-related hypodopaminergic function. However, the lack of group differences in task-related BOLD response and the non-specific effect of GSK598809 between groups makes it difficult to ascertain whether D3 antagonism is likely to be normalising or restorative in our abstinent populations. The suggestion of differential D3 modulation between AO and PD subgroups is intriguing, raising the possibility of divergent treatment responses. Further study is needed to determine whether D3 antagonism should be recommended as a treatment target in substance dependence.
Collapse
Affiliation(s)
- Ioanna A. Vamvakopoulou
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Leon Fonville
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Alexandra Hayes
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - John McGonigle
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Rebecca Elliott
- Neuroscience and Psychiatry Unit, Institute of Brain, Behaviour and Mental Health, The University of Manchester, Manchester, United Kingdom
| | - Karen D. Ersche
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Remy Flechais
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Csaba Orban
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Anna Murphy
- Neuroscience and Psychiatry Unit, Institute of Brain, Behaviour and Mental Health, The University of Manchester, Manchester, United Kingdom
| | - Dana G. Smith
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| | - John Suckling
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Eleanor M. Taylor
- Neuroscience and Psychiatry Unit, Institute of Brain, Behaviour and Mental Health, The University of Manchester, Manchester, United Kingdom
| | - Bill Deakin
- Neuroscience and Psychiatry Unit, Institute of Brain, Behaviour and Mental Health, The University of Manchester, Manchester, United Kingdom
| | - Trevor W. Robbins
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| | - David J. Nutt
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Anne R. Lingford-Hughes
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Louise M. Paterson
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Pribiag H, Shin S, Wang EHJ, Sun F, Datta P, Okamoto A, Guss H, Jain A, Wang XY, De Freitas B, Honma P, Pate S, Lilascharoen V, Li Y, Lim BK. Ventral pallidum DRD3 potentiates a pallido-habenular circuit driving accumbal dopamine release and cocaine seeking. Neuron 2021; 109:2165-2182.e10. [PMID: 34048697 PMCID: PMC9013317 DOI: 10.1016/j.neuron.2021.05.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/01/2021] [Accepted: 05/04/2021] [Indexed: 01/18/2023]
Abstract
Drugs of abuse induce persistent remodeling of reward circuit function, a process thought to underlie the emergence of drug craving and relapse to drug use. However, how circuit-specific, drug-induced molecular and cellular plasticity can have distributed effects on the mesolimbic dopamine reward system to facilitate relapse to drug use is not fully elucidated. Here, we demonstrate that dopamine receptor D3 (DRD3)-dependent plasticity in the ventral pallidum (VP) drives potentiation of dopamine release in the nucleus accumbens during relapse to cocaine seeking after abstinence. We show that two distinct VP DRD3+ neuronal populations projecting to either the lateral habenula (LHb) or the ventral tegmental area (VTA) display different patterns of activity during drug seeking following abstinence from cocaine self-administration and that selective suppression of elevated activity or DRD3 signaling in the LHb-projecting population reduces drug seeking. Together, our results uncover how circuit-specific DRD3-mediated plasticity contributes to the process of drug relapse.
Collapse
Affiliation(s)
- Horia Pribiag
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sora Shin
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA 24016, USA; Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA 24061, USA
| | - Eric Hou-Jen Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fangmiao Sun
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871 10 Beijing, China; PKU-IDG/McGovern Institute for Brain Research, 100871 Beijing, China
| | - Paul Datta
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alexander Okamoto
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hayden Guss
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Akanksha Jain
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xiao-Yun Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Bruna De Freitas
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Patrick Honma
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stefan Pate
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Varoth Lilascharoen
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Biological Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871 10 Beijing, China; PKU-IDG/McGovern Institute for Brain Research, 100871 Beijing, China
| | - Byung Kook Lim
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Biological Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
4
|
Groman SM, Hillmer AT, Liu H, Fowles K, Holden D, Morris ED, Lee D, Taylor J. Midbrain D 3 Receptor Availability Predicts Escalation in Cocaine Self-administration. Biol Psychiatry 2020; 88:767-776. [PMID: 32312578 PMCID: PMC8954711 DOI: 10.1016/j.biopsych.2020.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Results from neuroimaging studies suggest that disruptions in flexible decision-making functions in substance-dependent individuals are a consequence of drug-induced neural adaptations. In addicted populations, however, the causal relationship between biobehavioral phenotypes of susceptibility and addiction consequence is difficult to dissociate. Indeed, evidence from animals suggests that poor decision making due to preexisting biological factors can independently enhance the risk for developing addiction-like behaviors. Neuroimaging studies in animals provide a unique translational approach for the identification of the neurobiological mechanisms that mediate susceptibility to addiction. METHODS We used positron emission tomography in rats to quantify regional dopamine D2/3 receptors and metabotropic glutamate receptor 5 (mGluR5) and assessed decision making using a probabilistic reversal learning task. Susceptibility to self-administer cocaine was then quantified for 21 days followed by tests of motivation and relapse-like behaviors. RESULTS We found that deficits specifically in reward-guided choice behavior on the probabilistic reversal learning task predicted greater escalation of cocaine self-administration behavior and greater motivation for cocaine and, critically, were associated with higher midbrain D3 receptor availability. Additionally, individual differences in midbrain D3 receptor availability independently predicted the rate of escalation in cocaine-taking behaviors. No differences in mGluR5 availability, responses during tests of extinction, or cue-induced reinstatement were observed between the groups. CONCLUSIONS These findings indicate that our identified D3-mediated decision-making phenotype can be used as a behavioral biomarker for assessment of cocaine use susceptibility in human populations.
Collapse
Affiliation(s)
- Stephanie M. Groman
- Department of Psychiatry Yale University,Correspondence should be addressed to: Stephanie M. Groman, Ph.D. (), Jane R. Taylor, Ph.D. (), 34 Park Street, New Haven CT 06515
| | - Ansel T. Hillmer
- Department of Psychiatry Yale University,Department of Radiology and Biomedical Imaging Yale University,Department of Yale Positron Emission Tomography Center Yale University
| | - Heather Liu
- Department of Radiology and Biomedical Imaging Yale University
| | - Krista Fowles
- Department of Yale Positron Emission Tomography Center Yale University
| | - Daniel Holden
- Department of Yale Positron Emission Tomography Center Yale University
| | - Evan D. Morris
- Department of Radiology and Biomedical Imaging Yale University,Department of Yale Positron Emission Tomography Center Yale University,Invicro, LLC
| | - Daeyeol Lee
- The Zanvyl Krieger Mind/Brain Institute, Department of Neuroscience, Department of Psychological and Brain Sciences, Johns Hopkins University
| | - Jane Taylor
- Department of Psychiatry Yale University,Department of Neuroscience Yale University,Correspondence should be addressed to: Stephanie M. Groman, Ph.D. (), Jane R. Taylor, Ph.D. (), 34 Park Street, New Haven CT 06515
| |
Collapse
|
5
|
Jordan CJ, He Y, Bi GH, You ZB, Cao J, Xi ZX, Newman AH. (±)VK4-40, a novel dopamine D 3 receptor partial agonist, attenuates cocaine reward and relapse in rodents. Br J Pharmacol 2020; 177:4796-4807. [PMID: 32851643 DOI: 10.1111/bph.15244] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Despite widespread abuse of cocaine, there are no approved treatments for cocaine use disorder. Chronic cocaine use is associated with up-regulated dopamine D3 receptor expression in the brain. Therefore, most D3 -based medication development has focused on D3 antagonists. However, D3 antagonists do not attenuate cocaine intake under "easy" self-administration conditions, when response requirements are low. We evaluated a novel, highly selective and metabolically stable D3 partial agonist, (±)VK4-40, for its efficacy in reducing cocaine intake and relapse to drug seeking. EXPERIMENTAL APPROACH The impact of (±)VK4-40 on cocaine intake and relapse was evaluated using intravenous self-administration procedures under a fixed-ratio 2 reinforcement schedule and cocaine-primed reinstatement conditions in rats. Optogenetic brain-stimulation reward procedures were used to evaluate the interaction of (±)VK4-40 and cocaine in the mesolimbic dopamine system in mice. Sucrose self-administration in rats and a conditioned place preference paradigm in mice were used to evaluate the abuse potential of (±)VK4-40 alone and other unwanted effects. KEY RESULTS (±)VK4-40 dose-dependently reduced cocaine self-administration and cocaine-primed reinstatement of drug-seeking behaviour. (±)VK4-40 also inhibited cocaine-enhanced brain-stimulation reward caused by optogenetic stimulation of dopamine neurons in the ventral tegmental area. (±)VK4-40 alone decreased brain-stimulation reward but produced neither conditioned place preference nor place aversion. This new D3 partial agonist also failed to alter oral sucrose self-administration. CONCLUSION AND IMPLICATIONS The novel D3 partial agonist, (±)VK4-40 attenuates cocaine reward and relapse in rodents, without significant unwanted effects. These findings support further investigation of D3 partial agonists as putative treatments for cocaine use disorder.
Collapse
Affiliation(s)
- Chloe J Jordan
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Yi He
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA.,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guo-Hua Bi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Zhi-Bing You
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
6
|
Bennett A, Barrera E, Namballa H, Harding W, Ranaldi R. (-)-Stepholidine blocks expression, but not development, of cocaine conditioned place preference in rats. Neurosci Lett 2020; 734:135151. [PMID: 32531529 PMCID: PMC7368993 DOI: 10.1016/j.neulet.2020.135151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 01/08/2023]
Abstract
The purpose of this study was to investigate the effects of (-)-stepholidine (SPD), a compound with dopamine D1 partial agonist and D2/D3 antagonist properties, on the development and expression of cocaine conditioned place preference (CPP). Subjects (N = 65; male Long Evans rats) were tested using a CPP procedure consisting of 3 phases: (1) a 15-min pre-exposure session where animals could explore each compartment freely, (2) eight 30-min conditioning sessions where animals were restricted to one side or the other with cocaine (10 mg/kg) or saline, respectively, on alternating days and (3) a 15-minute preference test session where animals could explore each compartment freely. To test the effects of SPD on expression of cocaine CPP, rats were administered vehicle (distilled water with 20 % DMSO), 10, 15 or 20 mg/kg SPD (intraperitoneally) 30 min prior to the test session. We found that 20 mg/kg of SPD significantly blocked the expression of cocaine CPP. To test the effects of SPD on the development of CPP, 0 (vehicle), 10, 15 or 20mg/kg SPD were administered 30 min prior to each cocaine conditioning session and vehicle before each saline conditioning session; no treatment was given prior to the test session. A preference test showed that each SPD group maintained a CPP similar to the vehicle group. These data indicate that SPD can block the expression of a cocaine CPP but has no effect on its development, suggesting that it inhibits the effects of cocaine cues on cocaine incentive motivated behavior. These results suggest that SPD may be a potential treatment for cue-driven aspects of cocaine use disorder.
Collapse
Affiliation(s)
- A Bennett
- Queens College of the City University of New York, Department of Psychology, USA
| | - E Barrera
- Queens College of the City University of New York, Department of Psychology, USA
| | - H Namballa
- Hunter College of the City University of New York, Department of Chemistry, USA; CUNY Graduate Center, 365 5thAvenue, New York, NY 10016, USA; CUNY Graduate Center, 365 5thAvenue, New York, NY 10016, USA
| | - W Harding
- Hunter College of the City University of New York, Department of Chemistry, USA; CUNY Graduate Center, 365 5thAvenue, New York, NY 10016, USA; CUNY Graduate Center, 365 5thAvenue, New York, NY 10016, USA
| | - R Ranaldi
- Queens College of the City University of New York, Department of Psychology, USA; CUNY Graduate Center, 365 5thAvenue, New York, NY 10016, USA.
| |
Collapse
|
7
|
Galaj E, Newman AH, Xi ZX. Dopamine D3 receptor-based medication development for the treatment of opioid use disorder: Rationale, progress, and challenges. Neurosci Biobehav Rev 2020; 114:38-52. [PMID: 32376243 PMCID: PMC7252042 DOI: 10.1016/j.neubiorev.2020.04.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 01/11/2023]
Abstract
Opioid abuse and related overdose deaths continue to rise in the United States, contributing to the current national opioid crisis. Although several opioid-based pharmacotherapies are available (e.g., methadone, buprenorphine, naloxone), they show limited effectiveness in long-term relapse prevention. In response to the opioid crisis, the National Institute on Drug Abuse proposed a list of pharmacological targets of highest priority for medication development for the treatment of opioid use disorders (OUD). Among these are antagonists of dopamine D3 receptors (D3R). In this review, we first review recent progress in research of the dopamine hypothesis of opioid reward and abuse and then describe the rationale and recent development of D3R ligands for the treatment of OUD. Herein, an emphasis is placed on the effectiveness of newly developed D3R antagonists in the animal models of OUD. These new drug candidates may also potentiate the analgesic effects of clinically used opioids, making them attractive as adjunctive medications for pain management and treatment of OUD.
Collapse
Affiliation(s)
- Ewa Galaj
- Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| | - Amy Hauck Newman
- Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
| | - Zheng-Xiong Xi
- Molecular Targets and Medication Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States.
| |
Collapse
|
8
|
Yang G, Liu L, Zhang R, Li J, Leung CK, Huang J, Li Y, Shen B, Zeng X, Zhang D. Cannabidiol attenuates methamphetamine-induced conditioned place preference via the Sigma1R/AKT/GSK-3β/CREB signaling pathway in rats. Toxicol Res (Camb) 2020; 9:202-211. [PMID: 32670551 DOI: 10.1093/toxres/tfaa021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/27/2020] [Accepted: 03/23/2020] [Indexed: 01/07/2023] Open
Abstract
Methamphetamine (METH) is a highly addictive psychostimulant. Cannabidiol (CBD) is an exogenous cannabinoid without psychostimulating activity, which has potential therapeutic effects on opioid addiction. However, it is unclear whether CBD has therapeutic effects on METH-induced motivational effects. The present study examines whether CBD has a protective effect on METH-induced conditioned place preference (CPP) in rats by regulating the Sigma1R and AKT-GSK3β-CREB signaling pathway. Seventy rats were equally and randomly divided into seven groups. The rat CPP model was established via the intraperitoneal injection (IP) of 2 mg/kg of METH. Next, the intraperitoneal injection of 10, 20, 40, and 80 mg/kg CBD was performed 1 h prior to the injection of saline or METH. The protein expression levels of Sigma1R, AKT, p-AKT, GSK-3β, p-GSK-3β, CREB, and p-CREB in the rats' prefrontal cortex, nucleus accumbens, and hippocampus and ventral tegmental were detected using western blot analysis. CBD was found to inhibit METH-induced CPP in a dose-dependent fashion. The expression levels of Sigma1R, p-AKT, p-GSK3β, and p-CREB increased significantly in the METH-induced CPP model. Treatment involving different doses of CBD caused differential inhibitory responses in the cellular protein abundance of Sigma1R, p-AKT, p-GSK3β, and p-CREB across various brain regions. The present study found that METH can induce CPP in rats. When a pretreatment of CBD is applied, the CBD can weaken CPP in METH-induced rats by regulating the SigmaR1/AKT/GSK-3β/CREB signaling pathway. The results of this study indicate that CBD has a potential therapeutic effect on METH-induced rewarding effects.
Collapse
Affiliation(s)
- Genmeng Yang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan 650500, China
| | - Liu Liu
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan 650500, China
| | - Ruilin Zhang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan 650500, China
| | - Juan Li
- School of Basic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan 650500, China
| | - Chi-Kwan Leung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,CUHK-SDU Joint Laboratory of Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jian Huang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan 650500, China
| | - Yuanyuan Li
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan 650500, China
| | - Baoyu Shen
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan 650500, China
| | - Xiaofeng Zeng
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan 650500, China
| | - Dongxian Zhang
- School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan 650500, China
| |
Collapse
|
9
|
Huang J, Yang G, Li Z, Leung CK, Wang W, Li Y, Liu L, Shen B, He C, He Y, Zeng X, Li J. Involvement of dopamine D3 receptor and dopamine transporter in methamphetamine-induced behavioral sensitization in tree shrews. Brain Behav 2020; 10:e01533. [PMID: 31943832 PMCID: PMC7010569 DOI: 10.1002/brb3.1533] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/15/2019] [Accepted: 12/29/2019] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION This study aims to establish a methamphetamine (METH)-induced behavioral sensitization model using tree shrews, as well as to measure the protein expression of the dopamine D3 receptor (D3R) and dopamine transporter (DAT). METHODS Forty tree shrews were equally and randomly divided into four experimental groups: those administered with 1, 2, and 4 mg/kg METH and a control group (treated with an equal amount of normal saline). Each experimental group was repeatedly exposed to METH for nine consecutive days to induce the development of behavioral sensitization, followed by four days of withdrawal (without the METH treatment) to induce the transfer of behavioral sensitization, then given 0.5 mg/kg of METH to undergo the expression of behavioral sensitization. Altered locomotor and stereotypic behaviors were measured daily via open-field experiments during the development and expression stages, and weight changes were also recorded. Then, the Western blot method was used to detect the expression levels of D3R and DAT in three brain regions: the nucleus accumbens, prefrontal cortex, and dorsal striatum 24 hr after the last behavioral test. RESULTS METH administration augmented motor-stimulant responses and stereotypic behaviors in all experimental groups, and stereotypic behaviors intensified more in the groups treated with 2 and 4 mg/kg METH. Motion distance, speed, and trajectory were significantly elevated in all experimental, however, METH at 4 mg/kg induced more stereotypic behaviors, decreasing these locomotor activities as compared with the 2 mg/kg METH group. 2 and 4 mg/kg METH significantly upregulated and downregulated D3R and DAT expression levels, respectively, in three brain regions, and these changes are more pronounced in 2 mg/kg METH. CONCLUSIONS These results indicated that this animal model may be used to study the neurobiological mechanisms that underly the development and expression of behavioral sensitization to METH. Deregulated D3R and DAT expression may be involved in the METH-induced behavioral sensitization.
Collapse
Affiliation(s)
- Jian Huang
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Genmeng Yang
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Zhen Li
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Chi-Kwan Leung
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China.,CUHK-SDU Joint Laboratory of Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Wenguang Wang
- Center of Tree Shrew Germplasm Resources, Institute of Medical Biology, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Yunnan Innovation Team of Standardization and Application Research in Tree Shrew, the Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Yuanyuan Li
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Liu Liu
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Baoyu Shen
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Cuihua He
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Yongwang He
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Xiaofeng Zeng
- School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Juan Li
- School of Basic Medicine, Kunming Medical University, Kunming, China
| |
Collapse
|
10
|
Abstract
How do brain systems evaluate the affective valence of a stimulus - that is, its quality of being good or bad? One possibility is that a neural subsystem, or 'module' (such as a subregion of the brain, a projection pathway, a neuronal population or an individual neuron), is permanently dedicated to mediate only one affective function, or at least only one specific valence - an idea that is termed here the 'affective modules' hypothesis. An alternative possibility is that a given neural module can exist in multiple neurobiological states that give it different affective functions - an idea termed here the 'affective modes' hypothesis. This suggests that the affective function or valence mediated by a neural module need not remain permanently stable but rather can change dynamically across different situations. An evaluation of evidence for the 'affective modules' versus 'affective modes' hypotheses may be useful for advancing understanding of the affective organization of limbic circuitry.
Collapse
|
11
|
Jordan CJ, Humburg BA, Thorndike EB, Shaik AB, Xi ZX, Baumann MH, Newman AH, Schindler CW. Newly Developed Dopamine D 3 Receptor Antagonists, R-VK4-40 and R-VK4-116, Do Not Potentiate Cardiovascular Effects of Cocaine or Oxycodone in Rats. J Pharmacol Exp Ther 2019; 371:602-614. [PMID: 31562201 PMCID: PMC6863462 DOI: 10.1124/jpet.119.259390] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/23/2019] [Indexed: 12/24/2022] Open
Abstract
Opioid and cocaine abuse are major public health burdens. Existing medications for opioid use disorder are limited by abuse liability and side effects, whereas no treatments are currently approved in the United States for cocaine use disorder. Dopamine D3 receptor (D3R) antagonists have shown promise in attenuating opioid and cocaine reward and mitigating relapse in preclinical models. However, translation of D3R antagonists to the clinic has been hampered by reports that the D3R antagonists GSK598,809 (5-(5-((3-((1S,5R)-1-(2-fluoro-4-(trifluoromethyl)phenyl)-3-azabicyclo[3.1.0]hexan-3-yl)propyl)thio)-4-methyl-4H-1,2,4-triazol-3-yl)-4-methyloxazole) and SB-277,011A (2-(2-((1r,4r)-4-(2-oxo-2-(quinolin-4-yl)ethyl)cyclohexyl)ethyl)-1,2,3,4-tetrahydroisoquinoline-6-carbonitrile) have adverse cardiovascular effects in the presence of cocaine. Recently, we developed two structurally novel D3R antagonists, R-VK4-40 and R-VK4-116, which are highly selective for D3R and display translational potential for treatment of opioid use disorder. Here, we tested whether R-VK4-40 ((R)-N-(4-(4-(2-Chloro-3-ethylphenyl)piperazin-1-yl)-3-hydroxybutyl)-1H-indole-2-carboxamide) and R-VK4-116 ((R)-N-(4-(4-(3-Chloro-5-ethyl-2-methoxyphenyl)piperazin-1-yl)-3-hydroxybutyl)-1H-indole-2-carboxamide) have unwanted cardiovascular effects in the presence of oxycodone, a prescription opioid, or cocaine in freely moving rats fitted with surgically implanted telemetry transmitters. We also examined cardiovascular effects of the D3R antagonist, SB-277,011A, and L-741,626 (1-((1H-indol-3-yl)methyl)-4-(4-chlorophenyl)piperidin-4-ol), a dopamine D2 receptor-selective antagonist, for comparison. Consistent with prior reports, SB-277,011A increased blood pressure, heart rate, and locomotor activity alone and in the presence of cocaine. L-741,626 increased blood pressure and heart rate. In contrast, R-VK4-40 alone dose-dependently reduced blood pressure and heart rate and attenuated oxycodone-induced increases in blood pressure and oxycodone or cocaine-induced increases in heart rate. Similarly, R-VK4-116 alone dose-dependently reduced cocaine-induced increases in blood pressure and heart rate. These results highlight the safety of new D3R antagonists and support the continued development of R-VK4-40 and R-VK4-116 for the treatment of opioid and cocaine use disorders. SIGNIFICANCE STATEMENT: Opioid and cocaine abuse are major public health challenges and new treatments that do not adversely impact the cardiovascular system are needed. Here, we show that two structurally novel dopamine D3 receptor antagonists, R-VK4-40 and R-VK4-116, do not potentiate, and may even protect against, oxycodone- or cocaine-induced changes in blood pressure and heart rate, supporting their further development for the treatment of opioid and/or cocaine use disorders.
Collapse
Affiliation(s)
- Chloe J Jordan
- Molecular Targets and Medications Discovery Branch (C.J.J., B.A.H., A.B.S., Z.-X.X., A.H.N.), Designer Drug Research Unit (M.H.B., C.W.S.), and Preclinical Pharmacology Section (E.B.T., C.W.S.), Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland
| | - Bree A Humburg
- Molecular Targets and Medications Discovery Branch (C.J.J., B.A.H., A.B.S., Z.-X.X., A.H.N.), Designer Drug Research Unit (M.H.B., C.W.S.), and Preclinical Pharmacology Section (E.B.T., C.W.S.), Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland
| | - Eric B Thorndike
- Molecular Targets and Medications Discovery Branch (C.J.J., B.A.H., A.B.S., Z.-X.X., A.H.N.), Designer Drug Research Unit (M.H.B., C.W.S.), and Preclinical Pharmacology Section (E.B.T., C.W.S.), Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland
| | - Anver Basha Shaik
- Molecular Targets and Medications Discovery Branch (C.J.J., B.A.H., A.B.S., Z.-X.X., A.H.N.), Designer Drug Research Unit (M.H.B., C.W.S.), and Preclinical Pharmacology Section (E.B.T., C.W.S.), Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch (C.J.J., B.A.H., A.B.S., Z.-X.X., A.H.N.), Designer Drug Research Unit (M.H.B., C.W.S.), and Preclinical Pharmacology Section (E.B.T., C.W.S.), Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland
| | - Michael H Baumann
- Molecular Targets and Medications Discovery Branch (C.J.J., B.A.H., A.B.S., Z.-X.X., A.H.N.), Designer Drug Research Unit (M.H.B., C.W.S.), and Preclinical Pharmacology Section (E.B.T., C.W.S.), Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch (C.J.J., B.A.H., A.B.S., Z.-X.X., A.H.N.), Designer Drug Research Unit (M.H.B., C.W.S.), and Preclinical Pharmacology Section (E.B.T., C.W.S.), Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland
| | - Charles W Schindler
- Molecular Targets and Medications Discovery Branch (C.J.J., B.A.H., A.B.S., Z.-X.X., A.H.N.), Designer Drug Research Unit (M.H.B., C.W.S.), and Preclinical Pharmacology Section (E.B.T., C.W.S.), Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland
| |
Collapse
|
12
|
Pal RK, Gadhiya S, Ramsey S, Cordone P, Wickstrom L, Harding WW, Kurtzman T, Gallicchio E. Inclusion of enclosed hydration effects in the binding free energy estimation of dopamine D3 receptor complexes. PLoS One 2019; 14:e0222902. [PMID: 31568493 PMCID: PMC6768453 DOI: 10.1371/journal.pone.0222902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/30/2019] [Indexed: 01/04/2023] Open
Abstract
Confined hydration and conformational flexibility are some of the challenges encountered for the rational design of selective antagonists of G-protein coupled receptors. We present a set of C3-substituted (-)-stepholidine derivatives as potent binders of the dopamine D3 receptor. The compounds are characterized biochemically, as well as by computer modeling using a novel molecular dynamics-based alchemical binding free energy approach which incorporates the effect of the displacement of enclosed water molecules from the binding site. The free energy of displacement of specific hydration sites is obtained using the Hydration Site Analysis method with explicit solvation. This work underscores the critical role of confined hydration and conformational reorganization in the molecular recognition mechanism of dopamine receptors and illustrates the potential of binding free energy models to represent these key phenomena.
Collapse
Affiliation(s)
- Rajat Kumar Pal
- Department of Chemistry, Brooklyn College, 2900 Bedford Avenue, Brooklyn, NY 11210, United States of America
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
| | - Satishkumar Gadhiya
- PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
- Department of Chemistry, Hunter College, 695 Park Avenue, NY 10065, United States of America
| | - Steven Ramsey
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
- Department of Chemistry, Lehman College, 250 Bedford Park Blvd. West, Bronx, NY 10468, United States of America
| | - Pierpaolo Cordone
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
- Department of Chemistry, Hunter College, 695 Park Avenue, NY 10065, United States of America
| | - Lauren Wickstrom
- Department of Science, Borough of Manhattan Community College, 199 Chambers Street, New York, NY 10007, United States of America
| | - Wayne W. Harding
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
- PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
- Department of Chemistry, Hunter College, 695 Park Avenue, NY 10065, United States of America
| | - Tom Kurtzman
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
- PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
- Department of Chemistry, Lehman College, 250 Bedford Park Blvd. West, Bronx, NY 10468, United States of America
| | - Emilio Gallicchio
- Department of Chemistry, Brooklyn College, 2900 Bedford Avenue, Brooklyn, NY 11210, United States of America
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
- PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States of America
- * E-mail:
| |
Collapse
|
13
|
Newman AH, Cao J, Keighron JD, Jordan CJ, Bi GH, Liang Y, Abramyan AM, Avelar AJ, Tschumi CW, Beckstead MJ, Shi L, Tanda G, Xi ZX. Translating the atypical dopamine uptake inhibitor hypothesis toward therapeutics for treatment of psychostimulant use disorders. Neuropsychopharmacology 2019; 44:1435-1444. [PMID: 30858517 PMCID: PMC6785152 DOI: 10.1038/s41386-019-0366-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 11/10/2022]
Abstract
Medication-assisted treatments are unavailable to patients with cocaine use disorders. Efforts to develop potential pharmacotherapies have led to the identification of a promising lead molecule, JJC8-091, that demonstrates a novel binding mode at the dopamine transporter (DAT). Here, JJC8-091 and a structural analogue, JJC8-088, were extensively and comparatively assessed to elucidate neurochemical correlates to their divergent behavioral profiles. Despite sharing significant structural similarity, JJC8-088 was more cocaine-like, increasing extracellular DA concentrations in the nucleus accumbens shell (NAS) efficaciously and more potently than JJC8-091. In contrast, JJC8-091 was not self-administered and was effective in blocking cocaine-induced reinstatement to drug seeking. Electrophysiology experiments confirmed that JJC8-091 was more effective than JJC8-088 at inhibiting cocaine-mediated enhancement of DA neurotransmission. Further, when VTA DA neurons in DAT-cre mice were optically stimulated, JJC8-088 produced a significant leftward shift in the stimulation-response curve, similar to cocaine, while JJC8-091 shifted the curve downward, suggesting attenuation of DA-mediated brain reward. Computational models predicted that JJC8-088 binds in an outward facing conformation of DAT, similar to cocaine. Conversely, JJC8-091 steers DAT towards a more occluded conformation. Collectively, these data reveal the underlying molecular mechanism at DAT that may be leveraged to rationally optimize leads for the treatment of cocaine use disorders, with JJC8-091 representing a compelling candidate for development.
Collapse
Affiliation(s)
- Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| | - Jianjing Cao
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Jacqueline D. Keighron
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Chloe J. Jordan
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Guo-Hua Bi
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Ying Liang
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Ara M. Abramyan
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Alicia J. Avelar
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX USA
| | - Christopher W. Tschumi
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX USA ,0000 0000 8527 6890grid.274264.1Aging & Metabolism Research Group, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Michael J. Beckstead
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX USA ,0000 0000 8527 6890grid.274264.1Aging & Metabolism Research Group, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Lei Shi
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Gianluigi Tanda
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Zheng-Xiong Xi
- 0000 0004 1936 8075grid.48336.3aMolecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224 USA
| |
Collapse
|
14
|
Selective dopamine D3 receptor antagonist YQA14 inhibits morphine-induced behavioral sensitization in wild type, but not in dopamine D3 receptor knockout mice. Acta Pharmacol Sin 2019; 40:583-588. [PMID: 30224637 DOI: 10.1038/s41401-018-0153-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/17/2018] [Indexed: 12/15/2022] Open
Abstract
Increasing preclinical evidence demonstrates that dopamine D3 receptor (D3R) antagonists are a potential option for the treatment of drug addiction. The reinstatement of the addiction can be triggered by environmental stimuli that acquire motivational salience through repeated associations with the drug's effects. YQA14 is a novel D3R antagonist that has exhibited pharmacotherapeutic efficacy in reducing cocaine and amphetamine reward and relapse to drug seeking in mice. In this study we investigated the effects of YQA14 on morphine-induced context-specific locomotor sensitization in mice. We showed that repeated injection of YQA14 (6.25-25 mg/kg every day ip) prior to morphine (10 mg/kg every day sc) not only inhibited the acquisition, but also significantly attenuated the expression of morphine-induced locomotor sensitization. Furthermore, in the expression phase, one single injection of YQA14 (6.25-25 mg/kg, ip) dose-dependently inhibited the expression of morphine-induced behavioral sensitization. Moreover, YQA14 inhibited the expression of morphine-induced behavioral sensitization in wild mice (WT), but not in D3R knockout (D3R-/-) mice in the expression phase. In addition, D3R-/- mice also displayed the reduction in the expression phase compared with WT mice. In summary, this study demonstrates that blockade or knockout of the D3R inhibits morphine-induced behavior sensitization, suggesting that D3R plays an important role in the pathogenesis and etiology of morphine addiction, and it might be a potential target for clinical management of opioid addiction.
Collapse
|
15
|
D'Souza MS. Brain and Cognition for Addiction Medicine: From Prevention to Recovery Neural Substrates for Treatment of Psychostimulant-Induced Cognitive Deficits. Front Psychiatry 2019; 10:509. [PMID: 31396113 PMCID: PMC6667748 DOI: 10.3389/fpsyt.2019.00509] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/28/2019] [Indexed: 01/10/2023] Open
Abstract
Addiction to psychostimulants like cocaine, methamphetamine, and nicotine poses a continuing medical and social challenge both in the United States and all over the world. Despite a desire to quit drug use, return to drug use after a period of abstinence is a common problem among individuals dependent on psychostimulants. Recovery for psychostimulant drug-dependent individuals is particularly challenging because psychostimulant drugs induce significant changes in brain regions associated with cognitive functions leading to cognitive deficits. These cognitive deficits include impairments in learning/memory, poor decision making, and impaired control of behavioral output. Importantly, these drug-induced cognitive deficits often impact adherence to addiction treatment programs and predispose abstinent addicts to drug use relapse. Additionally, these cognitive deficits impact effective social and professional rehabilitation of abstinent addicts. The goal of this paper is to review neural substrates based on animal studies that could be pharmacologically targeted to reverse psychostimulant-induced cognitive deficits such as impulsivity and impairment in learning and memory. Further, the review will discuss neural substrates that could be used to facilitate extinction learning and thus reduce emotional and behavioral responses to drug-associated cues. Moreover, the review will discuss some non-pharmacological approaches that could be used either alone or in combination with pharmacological compounds to treat the above-mentioned cognitive deficits. Psychostimulant addiction treatment, which includes treatment for cognitive deficits, will help promote abstinence and allow for better rehabilitation and integration of abstinent individuals into society.
Collapse
Affiliation(s)
- Manoranjan S D'Souza
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, Ada, OH, United States
| |
Collapse
|
16
|
Manuszak M, Harding W, Gadhiya S, Ranaldi R. (-)-Stepholidine reduces cue-induced reinstatement of cocaine seeking and cocaine self-administration in rats. Drug Alcohol Depend 2018; 189:49-54. [PMID: 29879681 PMCID: PMC6062455 DOI: 10.1016/j.drugalcdep.2018.04.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Dopamine receptors are implicated in cocaine reward and seeking. We hypothesize that (-)-stepholidine, a dopamine D1/D2/D3 multi-receptor agent, would be effective in reducing cocaine reward and seeking in an animal model. We investigated the effects of (-)-stepholidine in cue-induced reinstatement of cocaine seeking and cocaine self-administration (reward). METHODS Cue-induced reinstatement experiment: Rats were trained to press a lever reinforced by cocaine (1 mg/kg/injection) for 15 consecutive daily sessions, after which the response was extinguished by withholding cocaine and cocaine-paired cues (light and pump activation). This was followed by a cue-induced reinstatement test where subjects were exposed to two cocaine cue presentations and presses on the active lever produced cues. Subjects were treated with one of four (-)-stepholidine doses prior to the reinstatement test. Cocaine self-administration (reward) experiment: Rats were trained to self-administer cocaine under a progressive ratio schedule of reinforcement. After stable breakpoints were established, rats were injected with four doses of (-)-stepholidine prior to testing; each dose was injected prior to a separate test session with no-treatment sessions intervening to re-establish break points. RESULTS (-)-Stepholidine significantly reduced cue-induced reinstatement of cocaine seeking in a dose-related manner. Additionally, (-)-stepholidine significantly reduced break points for cocaine reward. (-)-Stepholidine did not significantly affect locomotor activity. CONCLUSIONS (-)-Stepholidine reduces cue-induced reinstatement of cocaine seeking and cocaine reward, suggesting that it may be useful in treating relapse in cocaine addiction.
Collapse
Affiliation(s)
- M. Manuszak
- Department of Psychology, Queens College, City University of New York, 65-30 Kissena Blvd, Flushing NY 11367, USA
| | - W. Harding
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, NY 10065, USA,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA,Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016
| | - S. Gadhiya
- Angion Biomedica Corp., 51 Charles Linndbergh Blvd, Uniondale, NY 11553, USA
| | - R. Ranaldi
- Department of Psychology, Queens College, City University of New York, 65-30 Kissena Blvd, Flushing NY 11367, USA,The Graduate Center of the City University of New York, New York, NY 10016, USA
| |
Collapse
|
17
|
Role of dopamine D3 receptor in alleviating behavioural deficits in animal models of post-traumatic stress disorder. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:190-200. [PMID: 29510167 DOI: 10.1016/j.pnpbp.2018.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 01/06/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a complicated psychiatric disorder, which occurs after exposure to a traumatic event. The main clinical manifestation of PTSD includes fear and stress dysregulation. In both animals and humans, dysregulation of dopamine function appears to be related to conditioned fear responses. Previous studies show that the dopamine D3 receptor (D3R) is involved in schizophrenia, autism, and substance use disorders and is related to emotional disorders. However, few studies have investigated the role of the D3R in the pathogenesis and aetiology of PTSD. In the current study, we have reported that D3R knockout (D3R-/-) mice displayed decreased freezing time of contextual fearing and anxiolytic effects following training sessions consisting of exposure to inescapable electric foot-shocks. Similarly, highly selective blockade of D3Rs by YQA14, a novel D3R antagonist, significantly ameliorated freezing and anxiogenic-like behaviours in the single-prolonged stress (SPS) model of PTSD in rats. And more, YQA14 selectively alleviated the symptoms of PTSD in WT mice but not in D3R-/- mice. In summary, this study demonstrates the anti-PTSD effects of blockade or knockout of the D3R, suggesting that the D3R might play an important role in the pathogenesis and aetiology of PTSD, and might be a potential target for the clinical management of PTSD.
Collapse
|
18
|
Rice OV, Ashby CR, Dixon C, Laurenzo W, Hayden J, Song R, Li J, Tiwari AK, Gardner EL. Selective dopamine D 3 receptor antagonism significantly attenuates stress-induced immobility in a rat model of post-traumatic stress disorder. Synapse 2018; 72:e22035. [PMID: 29704283 DOI: 10.1002/syn.22035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/16/2018] [Accepted: 04/25/2018] [Indexed: 01/02/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating psychiatric syndrome that occurs in individuals exposed to extremely threatening or traumatic events. In both animals and humans, dopamine (DA) function appears to be dysregulated in brain areas involved in the conditioned fear response(s) that underlie PTSD. In this study, we determined the effect of the selective DA D3 receptor antagonists YQA14A (6.25, 12.5 and 25 mg/kg i.p.) and SB-277011A (6 mg/kg i.p.) on tone-induced fear (assessed by measuring freeze time) in a modified version of the single-prolonged stress (SPS) model of PTSD in adult male Sprague-Dawley rats. Rats pretreated with vehicle and then subjected to restraint stress, forced swim and random foot shock (SPS) in the presence of a distinctive tone, displayed a significantly increased tone-induced contextual freeze time and fecal pellet mass following re-exposure to the tone. Rats pretreated with a single i.p. injection of 6.25 or 12.5 mg/kg of YQA14 or 6 mg/kg of SB-277011A showed significantly attenuated contextual freeze time in the presence of the tone when tested 14 days after exposure to SPS. Overall, our results indicate that selectively antagonizing DA D3 receptors significantly decreases freezing time caused by an environment previously associated with stress. If our findings can be extrapolated to humans with PTSD, they suggest that DA D3 receptors may play a role in the pathophysiology of PTSD, and may have therapeutic utility for the clinical management of PTSD.
Collapse
Affiliation(s)
- Onarae V Rice
- Psychology Department, Neuroscience Program, Furman University, 3300 Poinsett Highway, Greenville, South Carolina
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, 8000 Utopia Parkway, Jamaica, New York
| | - Clark Dixon
- Psychology Department, Neuroscience Program, Furman University, 3300 Poinsett Highway, Greenville, South Carolina
| | - William Laurenzo
- Psychology Department, Neuroscience Program, Furman University, 3300 Poinsett Highway, Greenville, South Carolina
| | - Jason Hayden
- Psychology Department, Neuroscience Program, Furman University, 3300 Poinsett Highway, Greenville, South Carolina
| | - Rui Song
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jin Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, Ohio
| | - Eliot L Gardner
- Neuropsychopharmacology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
19
|
Powell GL, Bonadonna JP, Vannan A, Xu K, Mach RH, Luedtke RR, Neisewander JL. Dopamine D3 receptor partial agonist LS-3-134 attenuates cocaine-motivated behaviors. Pharmacol Biochem Behav 2018; 171:46-53. [PMID: 29807065 DOI: 10.1016/j.pbb.2018.05.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/01/2018] [Accepted: 05/24/2018] [Indexed: 12/19/2022]
Abstract
AIMS The dopamine D3 receptor (D3R) is a pharmacotherapeutic target for drug dependence. We have successfully imaged human D3Rs using radiolabeled LS-3-134, an arylamide phenylpiperazine with moderate selectivity for the D3R over D2R and low efficacy at the D2 and D3R. In this study, we screened for effects of LS-3-134 as a potential anti-cocaine therapeutic. METHODS Male rats were pretreated with LS-3-134 (0, 1.0, 3.2, or 5.6 mg/kg, IP) 15 min prior to tests for its effects on spontaneous and cocaine-induced locomotion. We next investigated the effects of LS-3-134 (0, 1.0, 3.2, 5.6, or 10.0 mg/kg, IP) on operant responding on a multiple variable-interval (VI) 60-second schedule with alternating cocaine (0.375 mg/kg, IV) and sucrose (45 mg) reinforcer components. Additionally, we tested LS-3-134 (5.6 mg/kg, IP) effects on a progressive ratio (PR) schedule of cocaine reinforcement, on extinction of cocaine-seeking behavior, and on reinstatement of extinguished cocaine-seeking behavior by cocaine-associated light/tone cues. RESULTS LS-3-134 did not alter spontaneous locomotion, but at 5.6 mg/kg, it reduced cocaine-induced locomotion, break points on the high-effort progressive ratio schedule of reinforcement, and responding during extinction and cue reinstatement. In contrast, LS-3-134 did not alter cocaine or sucrose reinforcement on the low-effort multiple VI 60-second schedule. CONCLUSIONS The effects of LS-3-134 are similar to other dopamine D3 low efficacy partial agonists and antagonists in attenuating cocaine intake under high effort schedules of reinforcement and in attenuating cocaine-seeking behavior elicited by cocaine-associated cues. These findings are consistent with the anti-craving profile of other dopamine D3 drugs.
Collapse
Affiliation(s)
- Gregory L Powell
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Annika Vannan
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Kuiying Xu
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Robert H Mach
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Robert R Luedtke
- University of North Texas Health Science Center, the Department of Pharmacology and Neuroscience, Fort Worth, TX, United States
| | | |
Collapse
|
20
|
Dopamine D1 and D3 receptor polypharmacology as a potential treatment approach for substance use disorder. Neurosci Biobehav Rev 2018; 89:13-28. [PMID: 29577963 DOI: 10.1016/j.neubiorev.2018.03.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 03/19/2018] [Accepted: 03/19/2018] [Indexed: 12/29/2022]
Abstract
In the search for efficacious pharmacotherapies to treat cocaine addiction much attention has been given to agents targeting dopamine D1 or D3 receptors because of the involvement of these receptors in drug-related behaviors. D1-like and D3 receptor partial agonists and antagonists have been shown to reduce drug reward, reinstatement of drug seeking and conditioned place preference in rodents and non-human primates. However, translation of these encouraging results to clinical settings has been limited due to a number of factors including toxicity, poor pharmacokinetic properties and extrapyramidal and sedative side effects. This review highlights the role of D1 and D3 receptors in drug reward and seeking, the discovery of D1-D3 heteromers and their potential as targets in the treatment of addiction.
Collapse
|
21
|
Prieto GA. Abnormalities of Dopamine D 3 Receptor Signaling in the Diseased Brain. J Cent Nerv Syst Dis 2017; 9:1179573517726335. [PMID: 28855798 PMCID: PMC5562332 DOI: 10.1177/1179573517726335] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/19/2017] [Indexed: 01/15/2023] Open
Abstract
Dopamine D3 receptors (D3R) modulate neuronal activity in several brain regions including cortex, striatum, cerebellum, and hippocampus. A growing body of evidence suggests that aberrant D3R signaling contributes to multiple brain diseases, such as Parkinson’s disease, essential tremor, schizophrenia, and addiction. In line with these findings, D3R has emerged as a potential target in the treatment of neurological disorders. However, the mechanisms underlying neuronal D3R signaling are poorly understood, either in healthy or diseased brain. Here, I review the molecular mechanisms involved in D3R signaling via monomeric D3R and heteromeric receptor complexes (e.g., D3R-D1R, D3R-D2R, D3R-A2aR, and D3R-D3nf). I focus on D3R signaling pathways that, according to recent reports, contribute to pathological brain states. In particular, I describe evidence on both quantitative (e.g., increased number or affinity) and qualitative (e.g., switched signaling) changes in D3R that has been associated with brain dysfunction. I conclude with a description of basic mechanisms that modulate D3R signaling such as desensitization, as disruption of these mechanisms may underlie pathological changes in D3R signaling. Because several lines of evidence support the idea that imbalances in D3R signaling alter neural function, a better understanding of downstream D3R pathways is likely to reveal novel therapeutic strategies toward dopamine-related brain disorders.
Collapse
Affiliation(s)
- G Aleph Prieto
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| |
Collapse
|
22
|
The Novel Modafinil Analog, JJC8-016, as a Potential Cocaine Abuse Pharmacotherapeutic. Neuropsychopharmacology 2017; 42:1871-1883. [PMID: 28266501 PMCID: PMC5564383 DOI: 10.1038/npp.2017.41] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/15/2017] [Accepted: 02/20/2017] [Indexed: 12/22/2022]
Abstract
(±)Modafinil ((±)MOD) and its R-enantiomer (R-modafinil; R-MOD) have been investigated for their potential as treatments for psychostimulant addiction. We recently reported a series of (±)MOD analogs, of which JJC8-016 (N-(2-((bis(4-fluorophenyl)methyl)thio)ethyl)-3-phenylpropan-1-amine) was selected for further development. JJC8-016 and R-MOD were evaluated for binding across ~70 receptors, transporters, and enzymes. Although at a concentration of 10 μM, there were many hits for JJC8-016, binding affinities in the range of its DAT affinity were only observed at the serotonin transporter (SERT), dopamine D2-like, and sigma1 receptors. R-MOD was more selective, but had much lower affinity at the DAT (Ki=3 μM) than JJC8-016 (Ki=116 nM). In rats, systemic administration of R-MOD alone (10-30 mg/kg i.p.) dose-dependently increased locomotor activity and electrical brain-stimulation reward, whereas JJC8-016 (10-30 mg/kg i.p.) did not produce these effects. Strikingly, pretreatment with JJC8-016 dose-dependently inhibited cocaine-enhanced locomotion, cocaine self-administration, and cocaine-induced reinstatement of drug-seeking behavior, whereas R-MOD inhibited cocaine-induced reinstatement only at the high dose of 100 mg/kg. Notably, JJC8-016 alone neither altered extracellular dopamine in the nucleus accumbens nor maintained self-administration. It also failed to induce reinstatement of drug-seeking behavior. These findings suggest that JJC8-016 is a unique DAT inhibitor that has no cocaine-like abuse potential by itself. Moreover, pretreatment with JJC8-016 significantly inhibits cocaine-taking and cocaine-seeking behavior likely by interfering with cocaine binding to DAT. In addition, off-target actions may also contribute to its potential therapeutic utility in the treatment of cocaine abuse.
Collapse
|
23
|
Hassan HE, Kelly D, Honick M, Shukla S, Ibrahim A, Gorelick DA, Glassman M, McMahon RP, Wehring HJ, Kearns AM, Feldman S, Yu M, Bauer K, Wang JB. Pharmacokinetics and Safety Assessment of l-Tetrahydropalmatine in Cocaine Users: A Randomized, Double-Blind, Placebo-Controlled Study. J Clin Pharmacol 2016; 57:151-160. [PMID: 27363313 DOI: 10.1002/jcph.789] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/13/2016] [Accepted: 06/28/2016] [Indexed: 11/07/2022]
Abstract
Cocaine use disorder (CUD) remains a significant public health challenge. l-Tetrahydropalmatine (l-THP), a well-tolerated and nonaddictive compound, shows promise for the management of CUD. Its pharmacologic profile includes blockade at dopamine and other monoamine receptors and attenuation of cocaine self-administration, reinstatement, and rewarding properties in rats. This study evaluated the safety of l-THP in human cocaine users and its influence on the safety and pharmacokinetics (PK) of cocaine. Twenty-four cocaine-using adult men were randomized to receive l-THP (30 mg twice a day orally) or placebo double-blind for 4 days, with an intranasal cocaine (40 mg) challenge on the fourth day. Safety and tolerability were evaluated using vital signs, ECG, clinical laboratory tests, and standardized self-report instruments. Peripheral venous blood was collected periodically and later assayed for l-THP and cocaine using highly sensitive and specific ultraperformance liquid chromatography-fluorescence detection (UPLC-FLD) methods. Twenty subjects completed the study, of whom 19 provided complete PK data. The short 3.5-day course of l-THP was safe and well tolerated and did not affect cocaine's PK or its acute cardiovascular effects. The cocaine AUC0→∞ was 211.5 and 261.4 h·ng/mL, and the Cmax was 83.3 and 104.5 ng/mL for the l-THP and placebo groups, respectively. In addition there were no significant differences in the number of side effects reported in each group (l-THP group 22 [48%], placebo group 24 [52%]) or vital signs including, heart rate, blood pressure, complete blood count, or ECG. These findings suggest that oral THP has promise for further development as a treatment for CUD.
Collapse
Affiliation(s)
- Hazem E Hassan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Deanna Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Moshe Honick
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Sagar Shukla
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Ahmed Ibrahim
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - David A Gorelick
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew Glassman
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert P McMahon
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Heidi J Wehring
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ann Marie Kearns
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stephanie Feldman
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mingming Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Ken Bauer
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, USA
| | - Jia Bei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
24
|
Cao DN, Song R, Zhang SZ, Wu N, Li J. Nucleus accumbens hyperpolarization-activated cyclic nucleotide-gated channels modulate methamphetamine self-administration in rats. Psychopharmacology (Berl) 2016; 233:3017-29. [PMID: 27329413 DOI: 10.1007/s00213-016-4349-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/31/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE Methamphetamine addiction is believed to primarily result from increased dopamine release and the inhibition of dopamine uptake. Some evidence suggests that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play important roles in the functional modulation of dopaminergic neurons and the pathophysiology of related diseases. However, little is known about the effects of HCN channels on methamphetamine addiction. OBJECTIVES The present study investigated the role of brain HCN channels in methamphetamine addiction. RESULTS Acute intracerebroventricular (i.c.v.) injection or bilateral intra-accumbens microinjections of non-selective HCN channel blocker ZD7288 (0.3125 and 0.625 μg) significantly reduced both methamphetamine (0.0125 or 0.05 mg/kg/infusion)-induced self-administration under fixed ratio 2 reinforcement and the breakpoint of methamphetamine (0.05 mg/kg/infusion) under progressive ratio reinforcement in rats. Moreover, compared with i.c.v. injection, bilateral intra-accumbens microinjections of ZD7288 exerted stronger inhibitory effects, suggesting that blockade of HCN channels in the nucleus accumbens reduced the reinforcing effects of and motivation for methamphetamine. We also found that ZD7288 (0.625 and 1.25 μg, i.c.v.) significantly decreased methamphetamine (1 mg/kg, intraperitoneal (i.p.))-induced hyperactivity with no effect on the spontaneous activity in rats. Finally, in vivo microdialysis experiments showed that the HCN channel blockade using ZD7288 (0.625 and 1.25 μg, i.c.v.) decreased methamphetamine (1 mg/kg, i.p.)-induced elevation of extracellular dopamine levels in the nucleus accumbens. CONCLUSIONS These results indicate that HCN channels in the nucleus accumbens are involved in the reinforcing properties of methamphetamine and highlight the importance of HCN channels in the regulation of dopamine neurotransmission underlying methamphetamine addiction.
Collapse
Affiliation(s)
- Dan-Ni Cao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Rui Song
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Shu-Zhuo Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Ning Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Jin Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China.
| |
Collapse
|
25
|
Cortés A, Moreno E, Rodríguez-Ruiz M, Canela EI, Casadó V. Targeting the dopamine D3 receptor: an overview of drug design strategies. Expert Opin Drug Discov 2016; 11:641-64. [PMID: 27135354 DOI: 10.1080/17460441.2016.1185413] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Dopamine is a neurotransmitter widely distributed in both the periphery and the central nervous system (CNS). Its physiological effects are mediated by five closely related G protein-coupled receptors (GPCRs) that are divided into two major subclasses: the D1-like (D1, D5) and the D2-like (D2, D3, D4) receptors. D3 receptors (D3Rs) have the highest density in the limbic areas of the brain, which are associated with cognitive and emotional functions. These receptors are therefore attractive targets for therapeutic management. AREAS COVERED This review summarizes the functional and pharmacological characteristics of D3Rs, including the design and clinical relevance of full agonists, partial agonists and antagonists, as well as the capacity of these receptors to form active homodimers, heterodimers or higher order receptor complexes as pharmacological targets in several neurological and neurodegenerative disorders. EXPERT OPINION The high sequence homology between D3R and the D2-type challenges the development of D3R-selective compounds. The design of new D3R-preferential ligands with improved physicochemical properties should provide a better pharmacokinetic/bioavailability profile and lesser toxicity than is found with existing D3R ligands. It is also essential to optimize D3R affinity and, especially, D3R vs. D2-type binding and functional selectivity ratios. Developing allosteric and bitopic ligands should help to improve the D3R selectivity of these drugs. As most evidence points to the ability of GPCRs to form homomers and heteromers, the most promising therapeutic strategy in the future is likely to involve the application of heteromer-selective drugs. These selective ligands would display different affinities for a given receptor depending on the receptor partners within the heteromer. Therefore, designing novel compounds that specifically target and modulate D1R-D3R heteromers would be an interesting approach for the treatment of levodopa (L-DOPA)-induced dyskinesias.
Collapse
Affiliation(s)
- Antoni Cortés
- a Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain.,b Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB) , University of Barcelona , Barcelona , Spain
| | - Estefanía Moreno
- a Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain.,b Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB) , University of Barcelona , Barcelona , Spain
| | - Mar Rodríguez-Ruiz
- a Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain.,b Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB) , University of Barcelona , Barcelona , Spain
| | - Enric I Canela
- a Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain.,b Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB) , University of Barcelona , Barcelona , Spain
| | - Vicent Casadó
- a Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain.,b Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB) , University of Barcelona , Barcelona , Spain
| |
Collapse
|
26
|
Liu F, Wang X, Li Z, Li J, Zhuang X, Zhang Z. P-Glycoprotein (ABCB1) limits the brain distribution of YQA-14, a novel dopamine D3 receptor antagonist. Chem Pharm Bull (Tokyo) 2016; 63:512-8. [PMID: 26133067 DOI: 10.1248/cpb.c15-00089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
YQA-14 is a promising agent for treating addiction to cocaine and opioids. However, previous studies have showed there is marked contrast between the relatively small differences in pharmacological action in vivo and the large differences in their respective receptor binding properties in vitro. We hypothesized that the conflict between the in vivo and in vitro outcomes was attributable to poor brain exposure to YQA-14 caused by drug efflux transporters. To address this issue, we investigated the directional flux of YQA-14 across Caco-2 cells at 37°C or 4°C and the bidirectional transport in the presence and absence of transporter chemical inhibitors. These phenomena were further investigated by an in vivo determination of the brain and blood pharmacokinetics (PK) profile of YQA-14 following intraperitoneal administration with and without inhibitor. The efflux ratio of YQA-14 on Caco-2 cell monolayers was 2.39 and the efflux was temperature-dependent. When co-incubated with GF120918 or LY335979, the efflux of YQA-14 was markedly decreased. However, there was no significant difference in the permeability of YQA-14 when the cells were treated with Ko143. In vivo experiments showed that the brain-to-plasma ratio increased by more than 75-fold and 20-fold with co-administration of GF120918 and LY335979, respectively. Use of Ko143 did not change the brain-to-blood ratio of YQA-14. The results indicate that the brain distribution of YQA-14 was restricted because of active efflux transport at the blood brain barrier. In addition, P-glycoprotein (P-gp) played a dominant role in limiting the distribution of YQA-14 to the brain.
Collapse
Affiliation(s)
- Fei Liu
- Department of Pharmacy, The First Affiliated Hospital of PLA
| | | | | | | | | | | |
Collapse
|
27
|
Galaj E, Haynes J, Nisanov R, Ananthan S, Ranaldi R. The dopamine D3 receptor antagonist, SR 21502, facilitates extinction of cocaine conditioned place preference. Drug Alcohol Depend 2016; 159:263-6. [PMID: 26710978 PMCID: PMC4724481 DOI: 10.1016/j.drugalcdep.2015.11.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/07/2015] [Accepted: 11/24/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Pharmacotherapeutic agents that could facilitate extinction of cocaine cues would be useful in the treatment of cocaine addiction. We tested whether SR 21502, a selective dopamine (DA) D3 receptor antagonist, can facilitate extinction of cocaine conditioned place preference (CPP) in rats. METHODS In experiment 1, cocaine (10mg/kg) CPP was first established and then extinguished. During the extinction phase the rats were injected with SR 21502 and placed in the previously cocaine-paired compartment for four sessions and vehicle in the other compartment on four alternating sessions. The rats were then tested again for cocaine CPP. In experiment 2, different groups of rats were trained to associate SR 21502 with one compartment and saline with the other. RESULTS In experiment 1, the animals spent significantly more time in the cocaine-paired compartment after cocaine conditioning than they did before conditioning. Subsequently, the animals treated with SR 21502 during the extinction phase spent significantly less time in the cocaine-paired compartment than the vehicle group. In experiment 2, animals conditioned with SR 21502 preferred neither side of the CPP apparatus, indicating that SR 21502 produced no effects of its own. CONCLUSIONS These findings suggest that treatment with SR 21502, a DA D3 receptor antagonist, in the presence of cocaine cues can facilitate extinction of cocaine CPP and further suggest that this compound might be an effective cocaine addiction treatment.
Collapse
Affiliation(s)
- E Galaj
- CUNY, The Graduate Center, United States
| | - J Haynes
- Department of Psychology, Queens College of the City University of New York, United States
| | - R Nisanov
- CUNY, The Graduate Center, United States
| | - S Ananthan
- Department of Chemistry, Drug Discovery Division, Southern Research Institute, United States
| | - R Ranaldi
- CUNY, The Graduate Center, United States; Department of Psychology, Queens College of the City University of New York, United States.
| |
Collapse
|
28
|
Sun L, Song R, Chen Y, Yang RF, Wu N, Su RB, Li J. A selective D3 receptor antagonist YQA14 attenuates methamphetamine-induced behavioral sensitization and conditioned place preference in mice. Acta Pharmacol Sin 2016; 37:157-65. [PMID: 26687935 DOI: 10.1038/aps.2015.96] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/28/2015] [Indexed: 12/14/2022] Open
Abstract
AIM We have reported that a selective dopamine D3 receptor antagonist YQA14 attenuates cocaine reward and relapse to drug-seeking in mice. In the present study, we investigated whether YQA14 could inhibit methamphetamine (METH)-induced locomotor sensitization and conditioned place preference (CPP) in mice. METHODS Locomotor activity was monitored in mice treated with METH (1 mg/kg, ip) daily on d 4-13, followed by a challenge with METH (0.5 mg/kg) on d 21. CPP was examined in mice that were administered METH (1 mg/kg) or saline alternately on each other day for 8 days (METH conditioning). YQA14 was injected intraperitoneally 20 min prior to METH or saline. RESULTS Both repetitive (daily on d 4-13) and a single injection (on the day of challenge) of YQA14 (6.25, 12.5 and 25 mg/kg) dose-dependently inhibited the acquisition and expression of METH-induced locomotor sensitization. However, repetitive injection of YQA14 (daily during the METH conditioning) did not alter the acquisition of METH-induced CPP, whereas a single injection of YQA14 (prior to CPP test) dose-dependently attenuated the expression of METH-induced CPP. In addition, the repetitive injection of YQA14 dose-dependently facilitated the extinction and decreased the reinstatement of METH-induced CPP. CONCLUSION Brain D3 receptors are critically involved in the reward and psychomotor-stimulating effects of METH. Thus, YQA14 deserves further study as a potential medication for METH addiction.
Collapse
|
29
|
Ashby CR, Rice OV, Heidbreder CA, Gardner EL. The selective dopamine D₃ receptor antagonist SB-277011A attenuates drug- or food-deprivation reactivation of expression of conditioned place preference for cocaine in male Sprague-Dawley rats. Synapse 2016; 69:336-44. [PMID: 25851636 DOI: 10.1002/syn.21820] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 02/26/2015] [Accepted: 03/12/2015] [Indexed: 12/25/2022]
Abstract
We determined the effect of the selective dopamine D3 receptor antagonist SB-277011A on reactivation of conditioned place preference (CPP) to cocaine elicited by priming injections of cocaine or exposure to food deprivation stress (21 h) in male Sprague-Dawley rats. Animals paired with the cocaine-associated chamber displayed a robust and consistent CPP response. This CPP was extinguished after repeated pairings of the conditioned stimuli (cocaine-paired chamber contextual cues) in the absence of the unconditioned stimulus (cocaine). Twenty-four hours later, the administration of 5 mg kg(-1) i.p. of cocaine (immediately before the test) or exposure to 21 h of food deprivation reactivated the expression of the cocaine-induced CPP. In contrast, administration of 1 ml kg(-1) i.p. of vehicle did not reactivate the CPP response. Administration of the selective dopamine D3 receptor antagonist SB-277011A (3-24 mg kg(-1) i.p.) 30 min before cocaine administration on the test day produced a significant attenuation of CPP reactivation. Reactivation of the CPP response produced by food deprivation was also significantly attenuated by SB-277011A (6 or 12 mg kg(-1) i.p.) given 30 min before the test session. SB-277011A (12 or 24 mg kg(-1) i.p.) did not itself produce reactivation of the CPP response. Overall, these results suggest that the reactivation of the incentive value of drug-associated cues by cocaine or food deprivation is attenuated by selective antagonism of D3 receptors.
Collapse
Affiliation(s)
- Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, Jamaica, New York, 11439
| | | | | | | |
Collapse
|
30
|
Appel NM, Li SH, Holmes TH, Acri JB. Dopamine D3 Receptor Antagonist (GSK598809) Potentiates the Hypertensive Effects of Cocaine in Conscious, Freely-Moving Dogs. J Pharmacol Exp Ther 2015; 354:484-92. [PMID: 26177654 DOI: 10.1124/jpet.115.224121] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/14/2015] [Indexed: 12/12/2022] Open
Abstract
The chronic and relapsing nature of addiction presents unique challenges for ensuring the safety of a potential medication. A patient may use cocaine, for example, while taking the medication or take more medication than prescribed. Thus, a potential medication must be safe and not exacerbate the effects of cocaine. Multiple published studies support antagonism of brain dopamine D3 receptor function as a potential mechanism of action for an anti-addiction medication. Dopamine D3 receptors are widely distributed outside the central nervous system, however; for example, dopamine D3 receptors in the kidneys are implicated in regulating blood pressure. The selective dopamine D3 receptor antagonist GSK598809 [1-(2-fluoro-4-trifluoromethyl-phenyl)-3-{3-[4-methyl-5-(4-methyl-oxazol-5-yl)-4H-[1,2,4]triazol-3-ylsulfanyl]-propyl}-3-aza-bicyclo[3.1.0]hexane] has been proposed as a medication to treat cocaine and other substance use disorders. The US Food and Drug Administration has established guidelines recommending safety studies to investigate potential undesirable pharmacodynamic effects of a substance in relation to exposure in the therapeutic range and above. Hence, we assessed the interaction between this selective dopamine D3 receptor antagonist and cocaine on hemodynamics and cardiac function in freely-moving, telemetered dogs before conducting a clinical trial. GSK598809 increased the hemodynamic effect of cocaine in this model. Thus, the increase in blood pressure after intravenous cocaine was greater in animals that had been pretreated with GSK598809 compared with vehicle. This finding suggests that GSK598809 in particular, and perhaps dopamine D3 receptor antagonists as a class, may produce unacceptable cardiovascular risks as medications to treat cocaine use disorder.
Collapse
Affiliation(s)
- Nathan M Appel
- Division of Pharmacotherapies and Medical Consequences of Drug Abuse, National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland (N.M.A., S.-H.L., J.B.A.); and Stanford Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California (T.H.H.)
| | - Shou-Hua Li
- Division of Pharmacotherapies and Medical Consequences of Drug Abuse, National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland (N.M.A., S.-H.L., J.B.A.); and Stanford Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California (T.H.H.)
| | - Tyson H Holmes
- Division of Pharmacotherapies and Medical Consequences of Drug Abuse, National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland (N.M.A., S.-H.L., J.B.A.); and Stanford Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California (T.H.H.)
| | - Jane B Acri
- Division of Pharmacotherapies and Medical Consequences of Drug Abuse, National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland (N.M.A., S.-H.L., J.B.A.); and Stanford Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California (T.H.H.)
| |
Collapse
|
31
|
Ashby CR, Rice OV, Heidbreder CA, Gardner EL. The selective dopamine D3 receptor antagonist SB-277011A significantly accelerates extinction to environmental cues associated with cocaine-induced place preference in male Sprague-Dawley rats. Synapse 2015; 69:512-4. [PMID: 26138517 DOI: 10.1002/syn.21839] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/24/2015] [Accepted: 07/01/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St. John's University, Jamaica, New York, 11439
| | - Onarae V Rice
- Psychology Department, Furman University, Greenville, South Carolina, 29613
| | | | - Eliot L Gardner
- Neuropsychopharmacology Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, 21224
| |
Collapse
|
32
|
Pardo M, López-Cruz L, San Miguel N, Salamone JD, Correa M. Selection of sucrose concentration depends on the effort required to obtain it: studies using tetrabenazine, D1, D2, and D3 receptor antagonists. Psychopharmacology (Berl) 2015; 232:2377-91. [PMID: 25647696 DOI: 10.1007/s00213-015-3872-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/16/2015] [Indexed: 12/30/2022]
Abstract
RATIONALE Low doses of dopamine (DA) antagonists and accumbens DA depletions reduce food-reinforced instrumental behavior but do not impair primary food motivation, causing animals to reallocate behavior away from food-reinforced tasks with high response requirements and select less effortful alternatives. However, it is uncertain if this same pattern of effects would occur if sucrose was used as the reinforcer. OBJECTIVES These experiments studied the impact of DA depletion and antagonism on performance of an effort-related choice task using sucrose as the reinforcer, as well as sucrose consumption, preference, and taste reactivity tests. METHODS The effects of DA manipulations were assessed using a task in which rats chose between lever pressing on a fixed ratio 7 schedule for 5.0 % sucrose versus freely consuming a less concentrated solution (0.3 %). RESULTS The DA depleting agent tetrabenazine shifted effort-related choice, decreasing lever pressing for 5.0 % sucrose but increasing intake of the concurrently available 0.3 % sucrose. Tetrabenazine did not affect sucrose appetitive taste reactivity, or sucrose consumption or preference, in free consumption tests. The D1 antagonist ecopipam and the D2 antagonist haloperidol also shifted choice behavior at doses that did not alter sucrose consumption or preference. In contrast, sucrose pre-exposure reduced consumption across all conditions. D3 antagonism had no effects. CONCLUSIONS D1 and D2 receptor blockade and DA depletion reduce the tendency to work for sucrose under conditions that leave fundamental aspects of sucrose motivation (intake, preference, hedonic reactivity) intact. These findings have implications for studies employing sucrose intake or preference in animal models of depression.
Collapse
Affiliation(s)
- Marta Pardo
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, 12071, Castelló, Spain
| | - Laura López-Cruz
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, 12071, Castelló, Spain
| | - Noemí San Miguel
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, 12071, Castelló, Spain
| | - John D Salamone
- Department of Psychology, University of Connecticut, Storrs, CT, 06269-1020, USA
| | - Mercè Correa
- Àrea de Psicobiologia, Campus de Riu Sec, Universitat Jaume I, 12071, Castelló, Spain. .,Department of Psychology, University of Connecticut, Storrs, CT, 06269-1020, USA.
| |
Collapse
|
33
|
Rangel-Barajas C, Malik M, Mach RH, Luedtke RR. Pharmacological modulation of abnormal involuntary DOI-induced head twitch response movements in male DBA/2J mice: II. Effects of D3 dopamine receptor selective compounds. Neuropharmacology 2015; 93:179-90. [PMID: 25698528 DOI: 10.1016/j.neuropharm.2014.10.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 12/27/2022]
Abstract
We recently reported on the characterization of the hallucinogen 2,5-dimethoxy-4-methylamphetamine's (DOI) ability to elicit a head twitch response (HTR) in DBA/2J mice and the ability of D2 vs. D3 dopamine receptor selective compounds to modulate that response. For these studies, the ability of D3 vs. D2 dopamine receptor selective compounds to attenuate the DOI-dependent HTR was examined. WC 10, a D3 dopamine receptor weak partial agonist with 40-fold binding selectivity for D3 vs. D2 dopamine receptors, produced a dose-dependent decrease in the DOI-induced HTR (IC50 = 3.7 mg/kg). WC 44, a D3 receptor selective full agonist, also inhibited the DOI-induced HTR (IC50 = 5.1 mg/kg). The effect of two D3 receptor selective partial agonists, LAX-4-136 and WW-III-55, were also evaluated. These analogs exhibit 150-fold and 800-fold D3 vs. D2 binding selectivity, respectively. Both compounds inhibited the HTR with similar potency but with different maximum efficacies. At 10 mg/kg WW-III-55 inhibited the HTR by 95%, while LAX-4-136 administration resulted in a 50% reduction. In addition, DOI (5 mg/kg) was administered at various times after LAX-4-136 or WW-III-55 administration to compare the duration of action. The homopiperazine analog LAX-4-136 exhibited greater stability. An assessment of our test compounds on motor performance and coordination was performed using a rotarod test. None of the D3 dopamine receptor selective compounds significantly altered latency to fall, suggesting that these compounds a) did not attenuate the DOI-dependent HTR due to sedative or adverse motor effects and b) may have antipsychotic/antihallucinogenic activity.
Collapse
Affiliation(s)
- Claudia Rangel-Barajas
- University of North Texas Health Science Center, Department of Pharmacology and Neuroscience, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| | - Maninder Malik
- University of North Texas Health Science Center, Department of Pharmacology and Neuroscience, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| | - Robert H Mach
- Radiochemistry Laboratory, Neurology Department, University of Pennsylvania School of Medicine, Chemistry Building, 231 S. 34th Street, Philadelphia, PA 19104, USA
| | - Robert R Luedtke
- University of North Texas Health Science Center, Department of Pharmacology and Neuroscience, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA.
| |
Collapse
|
34
|
Chen Y, Song R, Yang RF, Wu N, Li J. A novel dopamine D3 receptor antagonist YQA14 inhibits methamphetamine self-administration and relapse to drug-seeking behaviour in rats. Eur J Pharmacol 2014; 743:126-32. [DOI: 10.1016/j.ejphar.2014.09.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/12/2014] [Accepted: 09/15/2014] [Indexed: 01/26/2023]
|
35
|
Galaj E, Manuszak M, Arastehmanesh D, Ranaldi R. Microinjections of a dopamine D1 receptor antagonist into the ventral tegmental area block the expression of cocaine conditioned place preference in rats. Behav Brain Res 2014; 272:279-85. [PMID: 25017572 DOI: 10.1016/j.bbr.2014.07.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/11/2014] [Accepted: 07/03/2014] [Indexed: 11/17/2022]
Abstract
Stimulation of dopamine (DA) D1 receptors in the ventral tegmental area (VTA) is involved in primary rewards. In the current study we investigated whether VTA D1 receptor stimulation likewise plays a role in mediating the rewarding effects of cocaine-associated stimuli, using the cocaine conditioned place preference (CPP) paradigm. Rats were prepared with cannulae so as to allow microinjections in the VTA and later conditioned to a cocaine-associated environment using the CPP paradigm. Prior to each conditioning session rats were injected with either saline or cocaine (10mg/kg, intraperitoneally) and then placed in one of the two sides of the CPP apparatus. Sessions lasted 30min a day over a period of eight days, such that rats alternated daily between consistently experiencing cocaine in one side and saline in the other. On the test day, which was conducted one day after conditioning, rats were given bilateral microinjections of one of four doses of the D1 antagonist, SCH 23390, (0, 2, 4 or 8μg/0.5μl) directly into the VTA and allowed free access to both sides of the apparatus. Preference for either side was measured as time spent in each side and compared to the same measures taken before conditioning. The D1 antagonist produced a dose-related, significant reduction in the preference for the cocaine-paired side compared to vehicle. These data suggest that the expression of cocaine conditioned place preference requires stimulation of VTA D1 receptors and, as such, are the first to suggest a role for VTA dendritically released DA in cocaine-, or other reward-, related learning.
Collapse
Affiliation(s)
- E Galaj
- Graduate Center of City University of New York, NY, United States
| | | | | | - R Ranaldi
- Graduate Center of City University of New York, NY, United States; Queens College, NY, United States.
| |
Collapse
|