1
|
Xi ZX, Bocarsly ME, Galaj E, Hempel B, Teresi C, Shaw M, Bi GH, Jordan C, Linz E, Alton H, Tanda G, Freyberg Z, Alvarez VA, Newman AH. Presynaptic and Postsynaptic Mesolimbic Dopamine D 3 Receptors Play Distinct Roles in Cocaine Versus Opioid Reward in Mice. Biol Psychiatry 2024; 96:752-765. [PMID: 38838841 PMCID: PMC11446657 DOI: 10.1016/j.biopsych.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Past research has illuminated pivotal roles of dopamine D3 receptors (D3R) in the rewarding effects of cocaine and opioids. However, the cellular and neural circuit mechanisms that underlie these actions remain unclear. METHODS We employed Cre-LoxP techniques to selectively delete D3R from presynaptic dopamine neurons or postsynaptic dopamine D1 receptor (D1R)-expressing neurons in male and female mice. We utilized RNAscope in situ hybridization, immunohistochemistry, real-time polymerase chain reaction, voltammetry, optogenetics, microdialysis, and behavioral assays (n ≥ 8 animals per group) to functionally characterize the roles of presynaptic versus postsynaptic D3R in cocaine and opioid actions. RESULTS Our results revealed D3R expression in ∼25% of midbrain dopamine neurons and ∼70% of D1R-expressing neurons in the nucleus accumbens. While dopamine D2 receptors (D2R) were expressed in ∼80% dopamine neurons, we found no D2R and D3R colocalization among these cells. Selective deletion of D3R from dopamine neurons increased exploratory behavior in novel environments and enhanced pulse-evoked nucleus accumbens dopamine release. Conversely, deletion of D3R from D1R-expressing neurons attenuated locomotor responses to D1-like and D2-like agonists. Strikingly, deletion of D3R from either cell type reduced oxycodone self-administration and oxycodone-enhanced brain-stimulation reward. In contrast, neither of these D3R deletions impacted cocaine self-administration, cocaine-enhanced brain-stimulation reward, or cocaine-induced hyperlocomotion. Furthermore, D3R knockout in dopamine neurons reduced oxycodone-induced hyperactivity and analgesia, while deletion from D1R-expressing neurons potentiated opioid-induced hyperactivity without affecting analgesia. CONCLUSIONS We dissected presynaptic versus postsynaptic D3R function in the mesolimbic dopamine system. D2R and D3R are expressed in different populations of midbrain dopamine neurons, regulating dopamine release. Mesolimbic D3R are critically involved in the actions of opioids but not cocaine.
Collapse
Affiliation(s)
- Zheng-Xiong Xi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland.
| | - Miriam E Bocarsly
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland
| | - Ewa Galaj
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Briana Hempel
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Catherine Teresi
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland
| | - Marlisa Shaw
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland
| | - Guo-Hua Bi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland; Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Chloe Jordan
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Emily Linz
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland; Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Hannah Alton
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland; Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Veronica A Alvarez
- Laboratory on Neurobiology of Compulsive Behaviors, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, Bethesda, Maryland; National Institute of Mental Health, Center on Compulsive Behaviors, Intramural Research Program, Bethesda, Maryland
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, Maryland.
| |
Collapse
|
2
|
Shafiee A, Beiky M, Mohammadi I, Rajai S, Jafarabady K, Moradi S, Beikmohamadi M, Teixeira AL. Effect of smoking on Brain-Derived Neurotrophic Factor (BDNF) blood levels: A systematic review and meta-analysis. J Affect Disord 2024; 349:525-533. [PMID: 38199418 DOI: 10.1016/j.jad.2024.01.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024]
Abstract
BACKGROUND Brain-Derived Neurotrophic Factor (BDNF) is a neurotrophin that plays a crucial role in neuronal survival and plasticity. Previous studies have suggested that smoking may influence BDNF levels, but the findings have been inconsistent. METHODS A comprehensive search of electronic databases was conducted to identify relevant studies. Inclusion criteria were applied to select studies that investigated the relationship between smoking and blood levels of BDNF. A random-effects model was used to estimate the overall effect size. RESULTS A total of 23 studies were included. The meta-analysis revealed a significant association between smoking and increased blood levels of BDNF (standardized mean difference [SMD] = -0.38, 95 % confidence interval [CI] 0.15 to 0.62, p = 0.002). Subgroup analyses based on BDNF source showed a significant increase in plasma-derived BDNF levels (SMD = 1.02, 95 % CI 0.50 to 1.53, p = 0.0001), while no significant difference was observed in serum-derived BDNF levels (SMD = 0.02, 95 % CI -0.19 to 0.22, p = 0.87). The pooled analysis revealed a non-significant difference in blood levels of BDNF between former smokers and non-smokers (random-effects model, SMD = 0.21, 95 % CI -0.04 to 0.46, p = 0.1). CONCLUSION Smokers exhibited significantly higher plasma levels of BDNF compared to non-smokers. Further research is needed to elucidate the underlying mechanisms and explore the potential therapeutic implications of targeting BDNF in smoking.
Collapse
Affiliation(s)
- Arman Shafiee
- Department of Psychiatry and Mental Health, Alborz University of Medical Sciences, Karaj, Iran; Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| | - Maryam Beiky
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Ida Mohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahryar Rajai
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kyana Jafarabady
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Shayan Moradi
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahya Beikmohamadi
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Antonio Lucio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
3
|
Tan X, Neslund EM, Ding ZM. The involvement of dopamine and D2 receptor-mediated transmission in effects of cotinine in male rats. Neuropharmacology 2023; 230:109495. [PMID: 36914092 PMCID: PMC10071274 DOI: 10.1016/j.neuropharm.2023.109495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
Previous studies indicated that cotinine, the major metabolite of nicotine, supported intravenous self-administration and exhibited relapse-like drug-seeking behaviors in rats. Subsequent studies started to reveal an important role of the mesolimbic dopamine system in cotinine's effects. Passive administration of cotinine elevated extracellular dopamine levels in the nucleus accumbens (NAC) and the D1 receptor antagonist SCH23390 attenuated cotinine self-administration. The objective of the current study was to further investigate the role of mesolimbic dopamine system in mediating cotinine's effects in male rats. Conventional microdialysis was conducted to examine NAC dopamine changes during active self-administration. Quantitative microdialysis and Western blot were used to determine cotinine-induced neuroadaptations within the NAC. Behavioral pharmacology was performed to investigate potential involvement of D2-like receptors in cotinine self-administration and relapse-like behaviors. NAC extracellular dopamine levels increased during active self-administration of cotinine and nicotine with less robust increase during cotinine self-administration. Repeated subcutaneous injections of cotinine reduced basal extracellular dopamine concentrations without altering dopamine reuptake in the NAC. Chronic self-administration of cotinine led to reduced protein expression of D2 receptors within the core but not shell subregion of the NAC, but did not change either D1 receptors or tyrosine hydroxylase in either subregion. On the other hand, chronic nicotine self-administration had no significant effect on any of these proteins. Systemic administration of eticlopride, a D2-like receptor antagonist attenuated both cotinine self-administration and cue-induced reinstatement of cotinine seeking. These results further support the hypothesis that the mesolimbic dopamine transmission plays a critical role in mediating reinforcing effects of cotinine.
Collapse
Affiliation(s)
- Xiaoying Tan
- Department of Anesthesiology & Perioperative Medicine, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, 17033, USA
| | - Elizabeth M Neslund
- Department of Anesthesiology & Perioperative Medicine, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, 17033, USA
| | - Zheng-Ming Ding
- Department of Anesthesiology & Perioperative Medicine, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, 17033, USA; Department of Pharmacology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, 17033, USA.
| |
Collapse
|
4
|
Kim KM. Unveiling the Differences in Signaling and Regulatory Mechanisms between Dopamine D2 and D3 Receptors and Their Impact on Behavioral Sensitization. Int J Mol Sci 2023; 24:ijms24076742. [PMID: 37047716 PMCID: PMC10095578 DOI: 10.3390/ijms24076742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/09/2023] Open
Abstract
Dopamine receptors are classified into five subtypes, with D2R and D3R playing a crucial role in regulating mood, motivation, reward, and movement. Whereas D2R are distributed widely across the brain, including regions responsible for motor functions, D3R are primarily found in specific areas related to cognitive and emotional functions, such as the nucleus accumbens, limbic system, and prefrontal cortex. Despite their high sequence homology and similar signaling pathways, D2R and D3R have distinct regulatory properties involving desensitization, endocytosis, posttranslational modification, and interactions with other cellular components. In vivo, D3R is closely associated with behavioral sensitization, which leads to increased dopaminergic responses. Behavioral sensitization is believed to result from D3R desensitization, which removes the inhibitory effect of D3R on related behaviors. Whereas D2R maintains continuous signal transduction through agonist-induced receptor phosphorylation, arrestin recruitment, and endocytosis, which recycle and resensitize desensitized receptors, D3R rarely undergoes agonist-induced endocytosis and instead is desensitized after repeated agonist exposure. In addition, D3R undergoes more extensive posttranslational modifications, such as glycosylation and palmitoylation, which are needed for its desensitization. Overall, a series of biochemical settings more closely related to D3R could be linked to D3R-mediated behavioral sensitization.
Collapse
Affiliation(s)
- Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea
| |
Collapse
|
5
|
Becker G, Lespine LF, Bahri MA, Serrano ME, Lemaire C, Luxen A, Tirelli E, Plenevaux A. Exercise against cocaine sensitization in mice: a [18F]fallypride micro-PET study. Brain Commun 2022; 4:fcab294. [PMID: 35169698 PMCID: PMC8833578 DOI: 10.1093/braincomms/fcab294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/18/2021] [Accepted: 12/12/2021] [Indexed: 11/14/2022] Open
Abstract
Abstract
Wheel-running exercise in laboratory rodents (animal model useful to study the neurobiology of aerobic exercise) decreases behavioural markers of vulnerability to addictive properties of various drugs of abuse including cocaine. However, neurobiological mechanisms underpinning this protective effect are far from fully characterized. Here, 28-day-old female C57BL/6J mice were housed with (n = 48) or without (n = 48) a running wheel for 6 weeks before being tested for acute locomotor responsiveness and initiation of locomotor sensitization to intraperitoneal injections of 8 mg/kg cocaine. The long-term expression of sensitization took place 3 weeks after the last session. On the day after, all mice underwent a micro-PET imaging session with [18F]fallypride radiotracer (dopamine 2/3 receptors antagonist). Exercised mice were less sensitive to acute and sensitized cocaine hyperlocomotor effects, such attenuation being particularly well marked for long-term expression of sensitization (η2P = 0.262). Chronic administration of cocaine was associated with a clear-cut increase of [18F]fallypride binding potential in mouse striatum (η2P = 0.170) while wheel-running exercise was associated with a moderate decrease in dopamine 2/3 receptors density in striatum (η2P = 0.075), a mechanism that might contribute to protective properties of exercise against drugs of abuse vulnerability.
Collapse
Affiliation(s)
- Guillaume Becker
- GIGA—Cyclotron Research Center—In Vivo Imaging, University of Liège, 4000 Liege, Belgium
| | - Louis-Ferdinand Lespine
- Department of Psychology, University of Liège, 4000 Liege, Belgium
- Pôle MOPHA, Pôle Est, Centre Hospitalier Le Vinatier, Bron, France
| | - Mohamed Ali Bahri
- GIGA—Cyclotron Research Center—In Vivo Imaging, University of Liège, 4000 Liege, Belgium
| | - Maria Elisa Serrano
- GIGA—Cyclotron Research Center—In Vivo Imaging, University of Liège, 4000 Liege, Belgium
| | - Christian Lemaire
- GIGA—Cyclotron Research Center—In Vivo Imaging, University of Liège, 4000 Liege, Belgium
| | - André Luxen
- GIGA—Cyclotron Research Center—In Vivo Imaging, University of Liège, 4000 Liege, Belgium
| | - Ezio Tirelli
- Department of Psychology, University of Liège, 4000 Liege, Belgium
| | - Alain Plenevaux
- GIGA—Cyclotron Research Center—In Vivo Imaging, University of Liège, 4000 Liege, Belgium
| |
Collapse
|
6
|
The Role of Dopamine D3 Receptors in Tobacco Use Disorder: A Synthesis of the Preclinical and Clinical Literature. Curr Top Behav Neurosci 2022; 60:203-228. [PMID: 36173599 DOI: 10.1007/7854_2022_392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tobacco smoking is a significant cause of preventable morbidity and mortality globally. Current pharmacological approaches to treat tobacco use disorder (TUD) are only partly effective and novel approaches are needed. Dopamine has a well-established role in substance use disorders, including TUD, and there has been a long-standing interest in developing agents that target the dopaminergic system to treat substance use disorders. Dopamine has 5 receptor subtypes (DRD1 to DRD5). Given the localization and safety profile of the dopamine receptor D3 (DRD3), it is of therapeutic potential for TUD. In this chapter, the preclinical and clinical literature investigating the role of DRD3 in processes relevant to TUD will be reviewed, including in nicotine reinforcement, drug reinstatement, conditioned stimuli and cue-reactivity, executive function, and withdrawal. Similarities and differences in findings from the animal and human work will be synthesized and findings will be discussed in relation to the therapeutic potential of targeting DRD3 in TUD.
Collapse
|
7
|
Sokoloff P, Le Foll B. A Historical Perspective on the Dopamine D3 Receptor. Curr Top Behav Neurosci 2022; 60:1-28. [PMID: 35467293 DOI: 10.1007/7854_2022_315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Before 1990, the multiplicity of dopamine receptors beyond D1 and D2 had remained a controversial concept, despite its substantial clinical implications, at a time when it was widely accepted that dopamine interacted with only two receptor subtypes, termed D1 and D2, differing one from the other by their pharmacological specificity and opposite effects on adenylyl cyclase. It was also generally admitted that the therapeutic efficacy of antipsychotics resulted from blockade of D2 receptors. Thanks to molecular biology techniques, the D3 receptor could be characterized as a distinct molecular entity having a restricted anatomical gene expression and different signaling, which could imply peculiar functions in controlling cognitive and emotional behaviors. Due to the structural similarities of D2 and D3 receptors, the search for D3-selective compounds proved to be difficult, but nevertheless led to the identification of fairly potent and in vitro and in vivo selective compounds. The latter permitted to confirm a role of D3 receptors in motor functions, addiction, cognition, and schizophrenia, which paved the way for the development of new drugs for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON, Canada. .,Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada. .,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. .,Departments of Family and Community Medicine, University of Toronto, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada. .,Waypoint Research Institute, Waypoint Centre for Mental Health Care, 5, Penetanguishene, ON, Canada.
| |
Collapse
|
8
|
Adolescent nicotine treatment causes robust locomotor sensitization during adolescence but impedes the spontaneous acquisition of nicotine intake in adult female Wistar rats. Pharmacol Biochem Behav 2021; 207:173224. [PMID: 34197844 DOI: 10.1016/j.pbb.2021.173224] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 11/20/2022]
Abstract
Very few people are able to quit smoking, and therefore it is essential to know which factors contribute to the development of compulsive nicotine use. These studies aimed to investigate if early-adolescent nicotine exposure causes locomotor sensitization and affects anxiety-like behavior and the spontaneous acquisition of intravenous nicotine self-administration. Early-adolescent male and female rats were treated with nicotine from postnatal (P) days 24 to 42, and anxiety-like behavior and locomotor activity were investigated one day after the cessation of nicotine treatment and in adulthood (>P75). The spontaneous acquisition of nicotine self-administration was also investigated in adulthood. The rats self-administered 0.03 mg/kg/infusion of nicotine for six days under a fixed-ratio (FR) 1 schedule and four days under an FR2 schedule (3-h sessions). Repeated nicotine administration increased locomotor activity, rearing, and stereotypies in a small open field in adolescent male and female rats. One day after the last nicotine injection, the percentage of open arm entries in the elevated plus-maze test was decreased in the males and increased in the females. However, locomotor activity in the small open field was unaffected. Adolescent nicotine treatment did not affect anxiety-like behavior and locomotor activity in adulthood. During the 10-day nicotine self-administration period, the females had a higher level of nicotine intake than the males. Adolescent nicotine treatment decreased nicotine intake in the females. In conclusion, these findings indicate that repeated nicotine administration during adolescence causes robust behavioral sensitization and leads to lower nicotine intake in females throughout the acquisition period in adulthood in rats.
Collapse
|
9
|
Roles of the Functional Interaction between Brain Cholinergic and Dopaminergic Systems in the Pathogenesis and Treatment of Schizophrenia and Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22094299. [PMID: 33919025 PMCID: PMC8122651 DOI: 10.3390/ijms22094299] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Most physiologic processes in the brain and related diseases involve more than one neurotransmitter system. Thus, elucidation of the interaction between different neurotransmitter systems could allow for better therapeutic approaches to the treatments of related diseases. Dopaminergic (DAergic) and cholinergic neurotransmitter system regulate various brain functions that include cognition, movement, emotion, etc. This review focuses on the interaction between the brain DAergic and cholinergic systems with respect to the pathogenesis and treatment of schizophrenia and Parkinson’s disease (PD). We first discussed the selection of motor plans at the level of basal ganglia, the major DAergic and cholinergic pathways in the brain, and the receptor subtypes involved in the interaction between the two signaling systems. Next, the roles of each signaling system were discussed in the context of the negative symptoms of schizophrenia, with a focus on the α7 nicotinic cholinergic receptor and the dopamine D1 receptor in the prefrontal cortex. In addition, the roles of the nicotinic and dopamine receptors were discussed in the context of regulation of striatal cholinergic interneurons, which play crucial roles in the degeneration of nigrostriatal DAergic neurons and the development of L-DOPA-induced dyskinesia in PD patients. Finally, we discussed the general mechanisms of nicotine-induced protection of DAergic neurons.
Collapse
|
10
|
Huang Z, Wu D, Qu X, Li M, Zou J, Tan S. BDNF and nicotine dependence: associations and potential mechanisms. Rev Neurosci 2020; 32:/j/revneuro.ahead-of-print/revneuro-2020-0044/revneuro-2020-0044.xml. [PMID: 32887210 DOI: 10.1515/revneuro-2020-0044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/18/2020] [Indexed: 12/30/2022]
Abstract
Smoking is the leading preventable cause of death worldwide and tobacco addiction has become a serious public health problem. Nicotine is the main addictive component of tobacco, and the majority of people that smoke regularly develop nicotine dependence. Nicotine addiction is deemed to be a chronic mental disorder. Although it is well known that nicotine binds to the nicotinic acetylcholine receptors (nAChRs) and activates the mesolimbic dopaminergic system (MDS) to generate the pleasant and rewarding effects, the molecular mechanisms of nicotine addiction are not fully understood. Brain-derived neurotrophic factor (BDNF) is the most prevalent growth factor in the brain, which regulates neuron survival, differentiation, and synaptic plasticity, mainly through binding to the high affinity receptor tyrosine kinase receptor B (TrkB). BDNF gene polymorphisms are associated with nicotine dependence and blood BDNF levels are altered in smokers. In this review, we discussed the effects of nicotine on BDNF expression in the brain and summarized the underlying signaling pathways, which further indicated BDNF as a key regulator in nicotine dependence. Further studies that aim to understand the neurobiological mechanism of BDNF in nicotine addcition would provide a valuable reference for quitting smoking and developing the treatment of other addictive substances.
Collapse
Affiliation(s)
- Zeyi Huang
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, 28 W. Changsheng Road, Hengyang421001,Hunan, China
| | - Daichao Wu
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, 28 W. Changsheng Road, Hengyang421001,Hunan, China
| | - Xilin Qu
- Grade 2017 of Clinical Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang421001,Hunan, China
| | - Meixiang Li
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, 28 W. Changsheng Road, Hengyang421001,Hunan, China
| | - Ju Zou
- Department of Parasitology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang421001,Hunan, China
| | - Sijie Tan
- Department of Histology and Embryology, Institute of Clinical Anatomy & Reproductive Medicine, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, 28 W. Changsheng Road, Hengyang421001,Hunan, China
| |
Collapse
|
11
|
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: 6.5] [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
|
12
|
Acharya S, Kim KM. α4β2 nicotinic acetylcholine receptor downregulates D 3 dopamine receptor expression through protein kinase C activation. Biochem Biophys Res Commun 2019; 514:665-671. [PMID: 31078264 DOI: 10.1016/j.bbrc.2019.05.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/03/2019] [Indexed: 02/08/2023]
Abstract
Receptor transactivation or crosstalk refers to instances in which the signaling of a given receptor is regulated by different classes of receptors. Functional crosstalk between α4β2 nicotinic acetylcholine receptor (nAChR) and D3 dopamine receptor (D3R) that belong to the family of ligand-gated ion channels and G protein-coupled receptors, respectively, has been reported from brain dopaminergic neurons. For example, D3R is involved in the development of reward-related behaviors induced by α4β2 nAChR stimulation. However, the molecular mechanisms involved in their crosstalk remain unclear. Among PKC isoforms (α, βII, γ, and δ) evaluated in this study, PKCβII interacted with D3R and potentiated D3R endocytosis. Following α4β2 nAChR stimulation, activated PKCβII translocated to the plasma membrane to induce clathrin-mediated endocytosis of D3R, resulting in downregulation and signal inhibition. Considering that D3R plays important roles in mediating reward-related physiological actions of α4β2 nAChR, this study could provide a new insight into the regulatory mechanism involved in nicotine addiction.
Collapse
Affiliation(s)
- Srijan Acharya
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea.
| |
Collapse
|
13
|
Schmidt HD, Rupprecht LE, Addy NA. Neurobiological and Neurophysiological Mechanisms Underlying Nicotine Seeking and Smoking Relapse. MOLECULAR NEUROPSYCHIATRY 2019; 4:169-189. [PMID: 30815453 PMCID: PMC6388439 DOI: 10.1159/000494799] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/23/2018] [Indexed: 12/19/2022]
Abstract
Tobacco-related morbidity and mortality continue to be a significant public health concern. Unfortunately, current FDA-approved smoking cessation pharmacotherapies have limited efficacy and are associated with high rates of relapse. Therefore, a better understanding of the neurobiological and neurophysiological mechanisms that promote smoking relapse is needed to develop novel smoking cessation medications. Here, we review preclinical studies focused on identifying the neurotransmitter and neuromodulator systems that mediate nicotine relapse, often modeled in laboratory animals using the reinstatement paradigm, as well as the plasticity-dependent neurophysiological mechanisms that facilitate nicotine reinstatement. Particular emphasis is placed on how these neuroadaptations relate to smoking relapse in humans. We also highlight a number of important gaps in our understanding of the neural mechanisms underlying nicotine reinstatement and critical future directions, which may lead toward the development of novel, target pharmacotherapies for smoking cessation.
Collapse
Affiliation(s)
- Heath D. Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laura E. Rupprecht
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nii A. Addy
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
- Interdepartmental Neuroscience Program, Yale Graduate School of Arts and Sciences, New Haven, Connecticut, USA
| |
Collapse
|
14
|
Borroto-Escuela DO, Perez De La Mora M, Manger P, Narváez M, Beggiato S, Crespo-Ramírez M, Navarro G, Wydra K, Díaz-Cabiale Z, Rivera A, Ferraro L, Tanganelli S, Filip M, Franco R, Fuxe K. Brain Dopamine Transmission in Health and Parkinson's Disease: Modulation of Synaptic Transmission and Plasticity Through Volume Transmission and Dopamine Heteroreceptors. Front Synaptic Neurosci 2018; 10:20. [PMID: 30042672 PMCID: PMC6048293 DOI: 10.3389/fnsyn.2018.00020] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 06/19/2018] [Indexed: 01/04/2023] Open
Abstract
This perspective article provides observations supporting the view that nigro-striatal dopamine neurons and meso-limbic dopamine neurons mainly communicate through short distance volume transmission in the um range with dopamine diffusing into extrasynaptic and synaptic regions of glutamate and GABA synapses. Based on this communication it is discussed how volume transmission modulates synaptic glutamate transmission onto the D1R modulated direct and D2R modulated indirect GABA pathways of the dorsal striatum. Each nigro-striatal dopamine neuron was first calculated to form large numbers of neostriatal DA nerve terminals and then found to give rise to dense axonal arborizations spread over the neostriatum, from which dopamine is released. These neurons can through DA volume transmission directly influence not only the striatal GABA projection neurons but all the striatal cell types in parallel. It includes the GABA nerve cells forming the island-/striosome GABA pathway to the nigral dopamine cells, the striatal cholinergic interneurons and the striatal GABA interneurons. The dopamine modulation of the different striatal nerve cell types involves the five dopamine receptor subtypes, D1R to D5R receptors, and their formation of multiple extrasynaptic and synaptic dopamine homo and heteroreceptor complexes. These features of the nigro-striatal dopamine neuron to modulate in parallel the activity of practically all the striatal nerve cell types in the dorsal striatum, through the dopamine receptor complexes allows us to understand its unique and crucial fine-tuning of movements, which is lost in Parkinson's disease. Integration of striatal dopamine signals with other transmitter systems in the striatum mainly takes place via the receptor-receptor interactions in dopamine heteroreceptor complexes. Such molecular events also participate in the integration of volume transmission and synaptic transmission. Dopamine modulation of the glutamate synapses on the dorsal striato-pallidal GABA pathway involves D2R heteroreceptor complexes such as D2R-NMDAR, A2AR-D2R, and NTSR1-D2R heteroreceptor complexes. The dopamine modulation of glutamate synapses on the striato-entopeduncular/nigral pathway takes place mainly via D1R heteroreceptor complexes such as D1R-NMDAR, A2R-D1R, and D1R-D3R heteroreceptor complexes. Dopamine modulation of the island/striosome compartment of the dorsal striatum projecting to the nigral dopamine cells involve D4R-MOR heteroreceptor complexes. All these receptor-receptor interactions have relevance for Parkinson's disease and its treatment.
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Section of Physiology, Department of Biomolecular Science, University of Urbino, Urbino, Italy
- Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Yaguajay, Cuba
| | - Miguel Perez De La Mora
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paul Manger
- Faculty of Health Sciences, School of Anatomical Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Manuel Narváez
- Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - Sarah Beggiato
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Minerva Crespo-Ramírez
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gemma Navarro
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biomedicine, University of Barcelona, Barcelona, Spain
| | - Karolina Wydra
- Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Zaida Díaz-Cabiale
- Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - Alicia Rivera
- Department of Cell Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
| | - Luca Ferraro
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Sergio Tanganelli
- Department of Life Sciences and Biotechnology (SVEB), University of Ferrara, Ferrara, Italy
| | - Małgorzata Filip
- Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biomedicine, University of Barcelona, Barcelona, Spain
- CiberNed: Centro de Investigación en Red Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Vousooghi N, Sadat‐Shirazi M, Safavi P, Zeraati R, Akbarabadi A, Makki SM, Nazari S, Zarrindast MR. Adult rat morphine exposure changes morphine preference, anxiety, and the brain expression of dopamine receptors in male offspring. Int J Dev Neurosci 2018; 69:49-59. [DOI: 10.1016/j.ijdevneu.2018.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/27/2018] [Accepted: 06/24/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Nasim Vousooghi
- Department of NeuroscienceSchool of Advanced Technologies in Medicine, Tehran University of Medical SciencesTehranIran
- Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical SciencesTehranIran
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical SciencesTehranIran
| | - Mitra‐Sadat Sadat‐Shirazi
- Department of NeuroscienceSchool of Advanced Technologies in Medicine, Tehran University of Medical SciencesTehranIran
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical SciencesTehranIran
| | - Payam Safavi
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical SciencesTehranIran
| | - Ramin Zeraati
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical SciencesTehranIran
| | - Ardeshir Akbarabadi
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical SciencesTehranIran
- Department of Veterinary MedicineGarmsar Branch, Islamic Azad UniversityGarmsarIran
| | - Seyed Mohammad Makki
- Department of PsychiatrySchool of Medicine, Shahid Beheshti University of Medical SciencesTehranIran
| | - Shahrzad Nazari
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical SciencesTehranIran
| | - Mohammad Reza Zarrindast
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical SciencesTehranIran
- Department of PharmacologySchool of Medicine, Tehran University of Medical SciencesTehranIran
- Institute for Studies in Theoretical Physics and Mathematics, School of Cognitive SciencesTehranIran
- Institute for Cognitive Science StudiesTehranIran
| |
Collapse
|
16
|
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: 4.8] [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
|
17
|
Morud J, Strandberg J, Andrén A, Ericson M, Söderpalm B, Adermark L. Progressive modulation of accumbal neurotransmission and anxiety-like behavior following protracted nicotine withdrawal. Neuropharmacology 2017; 128:86-95. [PMID: 28986279 DOI: 10.1016/j.neuropharm.2017.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/25/2017] [Accepted: 10/01/2017] [Indexed: 12/18/2022]
Abstract
Due to the highly addictive properties of nicotine, a low percentage of users successfully maintain cessation for longer periods of time. This might be linked to neuroadaptations elicited by the drug, and understanding progressive changes in neuronal function might provide critical insight into nicotine addiction. We have previously shown that neurotransmission in the nucleus accumbens (nAc), a key brain region with respect to drug reinforcement and relapse, is suppressed for as long as seven months after a brief period of nicotine treatment. Studies were therefore performed to define the temporal properties of these effects, and to assess behavioral correlates to altered neurotransmission. Ex vivo electrophysiology revealed progressive depression of synaptic efficacy in the nAc of rats previously receiving nicotine. In addition, following three months of nicotine withdrawal, the responses to GABAA receptor modulating drugs were blunted together with downregulation of several GABAA receptor subunits. In correlation to reduced accumbal neurotransmission, a reduced anxiety-like behavior; assessed in the elevated plus-maze and marble burying tests, were identified in animals pre-treated with nicotine. Lastly, to test the causal relationship between suppressed excitability in the nAc and reduced anxiety-like behavior, rats received local administration of diazepam in the nAc while monitoring behavioral effects on the elevated plus-maze. These results show that nicotine produces long-lasting changes in the GABAergic system, which are observed first after extended withdrawal. Our data also suggest that nicotine produces a progressive suppression of accumbal excitability, which could result in behavioral alterations that may have implications for further drug intake.
Collapse
Affiliation(s)
- Julia Morud
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Box 410, 405 30 Gothenburg, Sweden.
| | - Joakim Strandberg
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Anna Andrén
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Box 410, 405 30 Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Box 410, 405 30 Gothenburg, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Box 410, 405 30 Gothenburg, Sweden; Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Box 410, 405 30 Gothenburg, Sweden
| |
Collapse
|
18
|
You ZB, Gao JT, Bi GH, He Y, Boateng C, Cao J, Gardner EL, Newman AH, Xi ZX. The novel dopamine D3 receptor antagonists/partial agonists CAB2-015 and BAK4-54 inhibit oxycodone-taking and oxycodone-seeking behavior in rats. Neuropharmacology 2017; 126:190-199. [PMID: 28888944 DOI: 10.1016/j.neuropharm.2017.09.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/02/2017] [Accepted: 09/05/2017] [Indexed: 12/30/2022]
Abstract
The use of prescription opioid analgesics, particularly oxycodone, has dramatically increased, and parallels escalated opioid abuse and drug-related deaths worldwide. Understanding the molecular mechanisms underlying the development of opioid dependence and expanding treatment options to counter prescription opioid abuse has become a critical public health matter. In the present study, we first evaluated the reinforcing effects of oxycodone in a rat model of self-administration and then explored the potential utility of two novel high affinity dopamine D3 receptor (D3R) antagonists/partial agonists, CAB2-015 and BAK4-54, for treatment of prescription opioid abuse and dependence. We found that rats acquired oxycodone self-administration rapidly within a range of unit doses that was similar to that for heroin, confirming that oxycodone has significant abuse potential. Strikingly, pretreatment with either CAB2-015 or BAK4-54 (0.4-10 mg/kg, i.p.) dose-dependently decreased oxycodone self-administration, and shifted the oxycodone dose-response curve downward. Repeated pretreatment with CAB2-015 or BAK4-54 (0.4-4 mg/kg) facilitated extinction and inhibited oxycodone-induced reinstatement of drug-seeking behavior. In addition, pretreatment with CAB2-015 or BAK4-54 (4-10 mg/kg) also dose-dependently decreased oxycodone-enhanced locomotor activity, but only CAB2-015 decreased oral sucrose self-administration. These data suggest that D3R antagonists may be suitable alternatives or adjunctive to opioid-based medications currently used clinically in treating opioid addiction and that the D3R-selective ligands (CAB2-015 or BAK4-54) provide new lead molecules for development.
Collapse
Affiliation(s)
- Zhi-Bing You
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA
| | - Jun-Tao Gao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA
| | - Guo-Hua Bi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA
| | - Yi He
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA
| | - Comfort Boateng
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA
| | - Eliot L Gardner
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA.
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA.
| |
Collapse
|
19
|
Ugur M, Kaya E, Gozen O, Koylu EO, Kanit L, Keser A, Balkan B. Chronic nicotine-induced changes in gene expression of delta and kappa-opioid receptors and their endogenous ligands in the mesocorticolimbic system of the rat. Synapse 2017; 71. [PMID: 28509375 DOI: 10.1002/syn.21985] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022]
Abstract
Delta and kappa opioid receptors (DOR and KOR, respectively) and their endogenous ligands, proenkephalin (PENK) and prodynorphin (PDYN)-derived opioid peptides are proposed as important mediators of nicotine reward. This study investigated the regulatory effect of chronic nicotine treatment on the gene expression of DOR, KOR, PENK and PDYN in the mesocorticolimbic system. Three groups of rats were injected subcutaneously with nicotine at doses of 0.2, 0.4, or 0.6 mg/kg/day for 6 days. Rats were decapitated 1 hr after the last dose on day six, as this timing coincides with increased dopamine release in the mesocorticolimbic system. mRNA levels in the ventral tegmental area (VTA), lateral hypothalamic area (LHA), amygdala (AMG), dorsal striatum (DST), nucleus accumbens, and medial prefrontal cortex were measured by quantitative real-time PCR. Our results showed that nicotine upregulated DOR mRNA in the VTA at all of the doses employed, in the AMG at the 0.4 and 0.6 mg/kg doses, and in the DST at the 0.4 mg/kg dose. Conversely, PDYN mRNA was reduced in the LHA with 0.6 mg/kg nicotine and in the AMG with 0.4 mg/kg nicotine. KOR mRNA was also decreased in the DST with 0.6 mg/kg nicotine. Nicotine did not regulate PENK mRNA in any brain region studied.
Collapse
Affiliation(s)
- Muzeyyen Ugur
- Department of Physiology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Egemen Kaya
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Oguz Gozen
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Ersin O Koylu
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Lutfiye Kanit
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Aysegul Keser
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Burcu Balkan
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| |
Collapse
|
20
|
Norman H, D'Souza MS. Endogenous opioid system: a promising target for future smoking cessation medications. Psychopharmacology (Berl) 2017; 234:1371-1394. [PMID: 28285326 DOI: 10.1007/s00213-017-4582-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 02/24/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Nicotine addiction continues to be a health challenge across the world. Despite several approved medications, smokers continue to relapse. Several human and animal studies have evaluated the role of the endogenous opioid system as a potential target for smoking cessation medications. METHODS In this review, studies that have elucidated the role of the mu (MORs), delta (DORs), and kappa (KORs) opioid receptors in nicotine reward, nicotine withdrawal, and reinstatement of nicotine seeking will be discussed. Additionally, the review will discuss discrepancies in the literature and therapeutic potential of the endogenous opioid system, and suggest studies to address gaps in knowledge with respect to the role of the opioid receptors in nicotine dependence. RESULTS Data available till date suggest that blockade of the MORs and DORs decreased the rewarding effects of nicotine, while activation of the MORs and DORs decreased nicotine withdrawal-induced aversive effects. In contrast, activation of the KORs decreased the rewarding effects of nicotine, while blockade of the KORs decreased nicotine withdrawal-induced aversive effects. Interestingly, blockade of the MORs and KORs attenuated reinstatement of nicotine seeking. In humans, MOR antagonists have shown benefits in select subpopulations of smokers and further investigation is required to realize their full therapeutic potential. CONCLUSION Future work must assess the influence of polymorphisms in opioid receptor-linked genes in nicotine dependence, which will help in both identifying individuals vulnerable to nicotine addiction and the development of opioid-based smoking cessation medications. Overall, the endogenous opioid system continues to be a promising target for future smoking cessation medications.
Collapse
Affiliation(s)
- Haval Norman
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH, 45810, USA
| | - Manoranjan S D'Souza
- Department of Pharmaceutical and Biomedical Sciences, The Raabe College of Pharmacy, Ohio Northern University, 525 S Main Street, Ada, OH, 45810, USA.
| |
Collapse
|
21
|
Adermark L, Morud J, Lotfi A, Danielsson K, Ulenius L, Söderpalm B, Ericson M. Temporal Rewiring of Striatal Circuits Initiated by Nicotine. Neuropsychopharmacology 2016; 41:3051-3059. [PMID: 27388328 PMCID: PMC5101553 DOI: 10.1038/npp.2016.118] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 05/18/2016] [Accepted: 06/30/2016] [Indexed: 01/24/2023]
Abstract
Drug addiction has been conceptualized as maladaptive recruitment of integrative circuits coursing through the striatum, facilitating drug-seeking and drug-taking behavior. The aim of this study was to define temporal neuroadaptations in striatal subregions initiated by 3 weeks of intermittent nicotine exposure followed by protracted abstinence. Enhanced rearing activity was assessed in motor activity boxes as a measurement of behavioral change induced by nicotine (0.36 mg/kg), whereas electrophysiological field potential recordings were performed to evaluate treatment effects on neuronal activity. Dopamine receptor mRNA expression was quantified by qPCR, and nicotine-induced dopamine release was measured in striatal subregions using in vivo microdialysis. Golgi staining was performed to assess nicotine-induced changes in spine density of medium spiny neurons. The data presented here show that a brief period of nicotine exposure followed by abstinence leads to temporal changes in synaptic efficacy, dopamine receptor expression, and spine density in a subregion-specific manner. Nicotine may thus initiate a reorganization of striatal circuits that continues to develop despite protracted abstinence. We also show that the response to nicotine is modulated in previously exposed rats even after 6 months of abstinence. The data presented here suggests that, even though not self-administered, nicotine may produce progressive neuronal alterations in brain regions associated with goal-directed and habitual performance, which might contribute to the development of compulsive drug seeking and the increased vulnerability to relapse, which are hallmarks of drug addiction.
Collapse
Affiliation(s)
- Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, Gothenburg, Sweden
| | - Julia Morud
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, Gothenburg, Sweden
| | - Amir Lotfi
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, Gothenburg, Sweden
| | - Klara Danielsson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, Gothenburg, Sweden
| | - Lisa Ulenius
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, Gothenburg, Sweden
- Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, Gothenburg, Sweden
- Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at The University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
Thiruchselvam T, Malik S, Le Foll B. A review of positron emission tomography studies exploring the dopaminergic system in substance use with a focus on tobacco as a co-variate. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 43:197-214. [PMID: 27901585 DOI: 10.1080/00952990.2016.1257633] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
With the evolving sensitivity of positron emission tomography (PET) and the emergence of novel radiotracers, greater insight has been gained into the dopaminergic system as it relates to substance use. In this review, we summarize PET investigations from the last ten years that explore the dopaminergic system in tobacco, alcohol, stimulant, opiates, and cannabis addiction. In light of the prevalence of substance co-use, this review will also explore the effect of tobacco and other substance abuse co-morbidity on the dopaminergic system across study samples in the reviewed literature. In non-dependence, increased DA transmission following acute stimulant administration is a robust and consistent observation but is less detectable following acute alcohol and tobacco, where it likely represents a conditioned effect mediating reward expectation. Chronic drug exposure is generally associated with a hypo-functioning pre-synaptic dopamine system and lower D2/D3 receptor availability relative to healthy controls. Emerging evidence also shows that stimulant use disorders in particular may also be associated with greater D3 receptor availability relative to controls. A defined role for the dopaminergic system in cannabis and opiate use is yet to be elucidated. Future work is also needed to delineate the potential interactive effects of acute and chronic tobacco and substance co-use on the dopaminergic system.
Collapse
Affiliation(s)
- Thulasi Thiruchselvam
- b Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute , Centre for Addiction and Mental Health , Toronto , ON , Canada
| | - Saima Malik
- b Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute , Centre for Addiction and Mental Health , Toronto , ON , Canada
| | - Bernard Le Foll
- a Addiction Medicine Service, Ambulatory Care and Structured Treatments , Centre for Addiction and Mental Health , Toronto , ON , Canada.,b Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute , Centre for Addiction and Mental Health , Toronto , ON , Canada.,c Department of Family and Community Medicine , Pharmacology and Toxicology, Psychiatry, Institute of Medical Sciences, University of Toronto , Toronto , ON , Canada.,d Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health , Toronto , ON , Canada
| |
Collapse
|
23
|
Sex Differences in Midbrain Dopamine D2-Type Receptor Availability and Association with Nicotine Dependence. Neuropsychopharmacology 2016; 41:2913-2919. [PMID: 27329684 PMCID: PMC5061883 DOI: 10.1038/npp.2016.105] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 11/09/2022]
Abstract
Women differ from men in smoking-related behaviors, among them a greater difficulty in quitting smoking. Unlike female smokers, male smokers have lower striatal dopamine D2-type receptor availability (binding potential, BPND) than nonsmokers and exhibit greater smoking-induced striatal dopamine release. Because dopamine D2-type autoreceptors in the midbrain influence striatal dopamine release, a function that has been linked to addiction, we tested for sex differences in midbrain dopamine D2-type receptor BPND and in relationships between midbrain BPND, nicotine dependence and striatal dopamine D2-type receptor BPND. Positron emission tomography was used with [18F]fallypride to measure BPND in a midbrain region, encompassing the substantia nigra and ventral tegmental area, in 18 daily smokers (7 women, 11 men) and 19 nonsmokers (10 women, 9 men). A significant sex-by-group interaction reflected greater midbrain BPND in female but not male smokers than in corresponding nonsmokers (F1, 32=5.089, p=0.03). Midbrain BPND was positively correlated with BPND in the caudate nucleus and putamen in nonsmokers and female smokers but not in male smokers and with nicotine dependence in female but not in male smokers. Striatal BPND was correlated negatively with nicotine dependence and smoking exposure. These findings extend observations on dopamine D2-type receptors in smokers and suggest a sex difference in how midbrain dopamine D2-type autoreceptors influence nicotine dependence.
Collapse
|
24
|
Carboni L, Romoli B, Romualdi P, Zoli M. Repeated nicotine exposure modulates prodynorphin and pronociceptin levels in the reward pathway. Drug Alcohol Depend 2016; 166:150-8. [PMID: 27430399 DOI: 10.1016/j.drugalcdep.2016.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 01/02/2023]
Abstract
BACKGROUND Nicotine dependence is maintained by neurobiological adaptations in the dopaminergic brain reward pathway with the contribution of opioidergic circuits. This study assessed the role of opioid peptides and receptors on the molecular changes associated with nicotine dependence. To this aim we analysed nicotine effects on opioid gene and receptor expression in the reward pathway in a nicotine sensitization model. METHODS Sprague-Dawley rats received nicotine administrations for five days and locomotor activity assessment showed the development of sensitization. The mRNA expression of prodynorphin (pdyn), pronociceptin (pnoc) and the respective receptors was measured by quantitative PCR in the ventral midbrain (VM), the nucleus accumbens (NAc), the caudate-putamen (CPu), the pre-frontal cortex (PFCx), and the hippocampus. RESULTS A significant positive effect of sensitization on pdyn mRNA levels was detected in the CPu. This effect was supported by a significant and selective correlation between the two parameters in this region. Moreover, chronic but not acute nicotine treatment significantly decreased pdyn mRNA levels in the NAc and increased expression in the PFCx. Pnoc mRNA was significantly increased in the VM and the PFCx after sub-chronic administration of nicotine, whereas no alterations were observed after acute treatment. No treatment associated changes were detected in κ-opioid receptor or nociceptin receptor mRNAs. CONCLUSIONS This experiment revealed an effect of nicotine administration that was distinguishable from the effect of nicotine sensitization. While several pnoc and pdyn changes were associated to nicotine administration, the only significant effect of sensitization was a significant increase in pdyn in the CPu.
Collapse
Affiliation(s)
- Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy.
| | - Benedetto Romoli
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Michele Zoli
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
25
|
Kumar V, Bonifazi A, Ellenberger MP, Keck TM, Pommier E, Rais R, Slusher BS, Gardner E, You ZB, Xi ZX, Newman AH. Highly Selective Dopamine D3 Receptor (D3R) Antagonists and Partial Agonists Based on Eticlopride and the D3R Crystal Structure: New Leads for Opioid Dependence Treatment. J Med Chem 2016; 59:7634-50. [PMID: 27508895 DOI: 10.1021/acs.jmedchem.6b00860] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The recent and precipitous increase in opioid analgesic abuse and overdose has inspired investigation of the dopamine D3 receptor (D3R) as a target for therapeutic intervention. Metabolic instability or predicted toxicity has precluded successful translation of previously reported D3R-selective antagonists to clinical use for cocaine abuse. Herein, we report a series of novel and D3R crystal structure-guided 4-phenylpiperazines with exceptionally high D3R affinities and/or selectivities with varying efficacies. Lead compound 19 was selected based on its in vitro profile: D3R Ki = 6.84 nM, 1700-fold D3R versus D2R binding selectivity, and its metabolic stability in mouse microsomes. Compound 19 inhibited oxycodone-induced hyperlocomotion in mice and reduced oxycodone-induced locomotor sensitization. In addition, pretreatment with 19 also dose-dependently inhibited the acquisition of oxycodone-induced conditioned place preference (CPP) in rats. These findings support the D3R as a target for opioid dependence treatment and compound 19 as a new lead molecule for development.
Collapse
Affiliation(s)
- Vivek Kumar
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Michael P Ellenberger
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Thomas M Keck
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States.,Department of Chemistry & Biochemistry, Department of Biomedical & Translational Sciences, College of Science and Mathematics, Rowan University , 201 Mullica Hill Road, Glassboro, New Jersey 08028, United States
| | - Elie Pommier
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States.,Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine , 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine , 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Barbara S Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine , 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Eliot Gardner
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Zhi-Bing You
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health , 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - 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, Maryland 21224, United States
| |
Collapse
|
26
|
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: 4.4] [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
|
27
|
Kaya E, Gozen O, Ugur M, Koylu EO, Kanit L, Balkan B. Nicotine regulates cocaine-amphetamine-Regulated Transcript (Cart) in the mesocorticolimbic system. Synapse 2016; 70:283-92. [PMID: 26990424 DOI: 10.1002/syn.21903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/03/2016] [Accepted: 03/10/2016] [Indexed: 12/21/2022]
Abstract
Cocaine-and-Amphetamine Regulated Transcript (CART) mRNA and peptides are intensely expressed in the brain regions comprising mesocorticolimbic system. Studies suggest that CART peptides may have a role in the regulation of reward circuitry. The present study aimed to examine the effect of nicotine on CART expression in the mesocorticolimbic system. Three different doses of nicotine (0.2, 0.4, 0.6 mg/kg free base) were injected subcutaneously for 5 days, and on day 6, rats were decapitated following a challenge dose. CART mRNA and peptide levels in medial prefrontal cortex (mPFC), nucleus accumbens (NAc), dorsal striatum (DST), amygdala (AMG), lateral hypothalamic area (LHA), and ventral tegmental area (VTA) were measured by quantitative real-time PCR (qPCR) and Western Blot analysis, respectively. In the mPFC, 0.4 and 0.6 mg/kg nicotine, decreased CART peptide levels whereas there was no effect on CART mRNA levels. In the VTA, a down-regulation of CART peptide expression was observed with 0.2 and 0.6 mg/kg nicotine. Conversely, 0.4 and 0.6 mg/kg nicotine increased CART mRNA levels in the AMG without affecting the CART peptide expression. Nicotine did not regulate CART mRNA or CART peptide expression in the NAc, DST, and LHA. We conclude that nicotine regulates CART expression in the mesocorticolimbic system and this regulation may play an important role in nicotine reward. Synapse 70:283-292, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Egemen Kaya
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Oguz Gozen
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Muzeyyen Ugur
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey
| | - Ersin O Koylu
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Lutfiye Kanit
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| | - Burcu Balkan
- Department of Physiology, School of Medicine, Ege University, Izmir, Turkey.,Center for Brain Research, Ege University, Izmir, Turkey
| |
Collapse
|
28
|
Morud J, Adermark L, Perez-Alcazar M, Ericson M, Söderpalm B. Nicotine produces chronic behavioral sensitization with changes in accumbal neurotransmission and increased sensitivity to re-exposure. Addict Biol 2016; 21:397-406. [PMID: 25581387 DOI: 10.1111/adb.12219] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tobacco use is often associated with long-term addiction as well as high risk of relapse following cessation. This is suggestive of persistent neural adaptations, but little is known about the long-lasting effects of nicotine on neural circuits. In order to investigate the long-term effects of nicotine exposure, Wistar rats were treated for 3 weeks with nicotine (0.36 mg/kg), and the duration of behavioral and neurophysiological adaptations was evaluated 7 months later. We found that increased drug-induced locomotion persisted 7 months after the initial behavioral sensitization. In vitro analysis of synaptic activity in the core and shell of the nucleus accumbens (nAc) revealed a decrease in input/output function in both regions of nicotine-treated rats as compared to vehicle-treated control rats. In addition, administration of the dopamine D2 receptor agonist quinpirole (5 μM) significantly increased evoked population spike amplitude in the nAc shell of nicotine-treated rats as compared to vehicle-treated control rats. To test whether nicotine exposure creates long-lasting malleable circuits, animals were re-exposed to nicotine 7 months after the initial exposure. This treatment revealed an increased sensitivity to nicotine among animals previously exposed to nicotine, with higher nicotine-induced locomotion responses than observed initially. In vitro electrophysiological recordings in re-exposed rats detected an increased sensitivity to dopamine D2 receptor activation. These results suggest that nicotine produces persistent neural adaptations that make the system sensitive and receptive to future nicotine re-exposure.
Collapse
Affiliation(s)
- Julia Morud
- Addiction Biology Unit; Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at University of Gothenburg; Sweden
| | - Louise Adermark
- Addiction Biology Unit; Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at University of Gothenburg; Sweden
| | - Marta Perez-Alcazar
- Department of Physiology; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at the University of Gothenburg; Sweden
| | - Mia Ericson
- Addiction Biology Unit; Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at University of Gothenburg; Sweden
| | - Bo Söderpalm
- Addiction Biology Unit; Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at University of Gothenburg; Sweden
- Beroendekliniken; Sahlgrenska University Hospital; Sweden
| |
Collapse
|
29
|
Smith LN, Penrod RD, Taniguchi M, Cowan CW. Assessment of Cocaine-induced Behavioral Sensitization and Conditioned Place Preference in Mice. J Vis Exp 2016:53107. [PMID: 26967472 DOI: 10.3791/53107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
It is thought that rewarding experiences with drugs create strong contextual associations and encourage repeated intake. In turn, repeated exposures to drugs of abuse make lasting alterations in the brain function of vulnerable individuals, and these persistent alterations likely serve to maintain the maladaptive drug seeking and taking behaviors characteristic of addiction/dependence(2). In rodents, reward experience and contextual associations are frequently measured using the conditioned place preference assay, or CPP, wherein preference for a previously drug-paired context is measured. Behavioral sensitization, on the other hand, is an increase in a drug-induced behavior that develops progressively over repeated exposures. Since sensitized behaviors can often be measured after several months of drug abstinence, depending on the dose and length of initial exposure, they are considered observable correlates of lasting drug-induced plasticity. Researchers have found these assays useful in determining the neurobiological substrates mediating aspects of addiction as well as assessing the potential of different interventions in disrupting these behaviors. This manuscript describes basic, effective protocols for mouse CPP and locomotor behavioral sensitization to cocaine.
Collapse
Affiliation(s)
- Laura N Smith
- Department of Psychiatry, Harvard Medical School, McLean Hospital
| | - Rachel D Penrod
- Department of Psychiatry, Harvard Medical School, McLean Hospital
| | - Makoto Taniguchi
- Department of Psychiatry, Harvard Medical School, McLean Hospital
| | | |
Collapse
|
30
|
Galaj E, Manuszak M, Babic S, Ananthan S, Ranaldi R. The selective dopamine D3 receptor antagonist, SR 21502, reduces cue-induced reinstatement of heroin seeking and heroin conditioned place preference in rats. Drug Alcohol Depend 2015; 156:228-233. [PMID: 26429728 PMCID: PMC4633332 DOI: 10.1016/j.drugalcdep.2015.09.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/13/2015] [Accepted: 09/14/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Because the role of dopamine (DA) D3 receptors has been investigated primarily in relation to cocaine-related behaviors little is known of the role of these receptors in heroin seeking. PURPOSES To investigate the effect of the selective DA D3 receptor antagonist, SR 21502, on cue-induced reinstatement of heroin seeking and heroin conditioned place preference (CPP). METHODS In experiment 1, rats were trained to self-administer intravenous heroin for 15 days followed by extinction. Following extinction animals were treated with one of several SR 21502 doses (0, 7.5, 10 or 15mg/kg) and a cue-induced reinstatement test was conducted. In experiment 2, animals were conditioned to experience heroin in one compartment of a CPP apparatus and saline in the other. On the test day animals were treated with 0, 3.75, 7.5, 10 or 15mg/kg of SR 21502 and tested for their CPP. RESULTS The results from experiment 1 showed a significant dose-related reduction in cue-induced reinstatement of active lever pressing in the 7.5 and 10mg groups and an absence of the reinstatement effect in the 15mg group. In experiment 2, animals treated with vehicle or 3.75mg of SR 21502 showed significant heroin place preferences but those treated with the higher doses showed no CPP. CONCLUSIONS Our findings suggest that DA D3 receptors play a significant role in heroin approach behaviors driven by conditioned stimuli. As such, we propose that SR 21502 holds potential as an effective pharmacotherapeutic agent for relapse prevention and should be studied further.
Collapse
MESH Headings
- Animals
- Conditioning, Classical/drug effects
- Conditioning, Classical/physiology
- Cues
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Extinction, Psychological/drug effects
- Extinction, Psychological/physiology
- Female
- Heroin/administration & dosage
- Heroin Dependence/physiopathology
- Heroin Dependence/rehabilitation
- Imidazoles/pharmacology
- Male
- Pyridines/pharmacology
- Rats
- Rats, Long-Evans
- Receptors, Dopamine D3/antagonists & inhibitors
- Receptors, Dopamine D3/physiology
- Recurrence
- Self Administration
- Substance Abuse, Intravenous/physiopathology
- Substance Abuse, Intravenous/rehabilitation
Collapse
Affiliation(s)
- Ewa Galaj
- CUNY Graduate Center, New York, NY, United States
| | - Monica Manuszak
- Queens College of the City University of New York, Department of Psychology, Flushing, NY, United States
| | - Sandra Babic
- Queens College of the City University of New York, Department of Psychology, Flushing, NY, United States
| | - Subramaniam Ananthan
- Department of Chemistry, Drug Discovery Division, Southern Research Institute, Birmingham, AL, United States
| | - Robert Ranaldi
- CUNY Graduate Center, New York, NY, United States; Queens College of the City University of New York, Department of Psychology, Flushing, NY, United States.
| |
Collapse
|
31
|
Le Foll B, Di Ciano P. Neuronal circuitry underlying the impact of D3 receptor ligands in drug addiction. Eur Neuropsychopharmacol 2015; 25:1401-9. [PMID: 25266821 PMCID: PMC4362926 DOI: 10.1016/j.euroneuro.2014.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 08/11/2014] [Accepted: 08/16/2014] [Indexed: 01/31/2023]
Abstract
Since the cloning of the D3 receptor in the early 1990s, there has been a great deal of interest in this receptor as a possible therapeutic target for drug addiction. The development of a D3 ligand suitable for use in humans has remained elusive, so the study of the function of the D3 receptor and its possible therapeutic efficacy has largely been restricted to animals. Pre-clinical studies have established that systemic administration of D3 ligands, particularly antagonists and partial agonists, can alter drug-seeking in animals. Despite over a decade of research, few studies have investigated the effects of intra-cerebral infusion of D3 ligands on drug-seeking. In the present review, these studies are summarized, which have largely focused on stimulus-controlled behaviors. Converging evidence from studies of D3 receptor expression, Fos and pharmacological Magnetic Resonance Imaging (phMRI) is also provided to delineate some of the D3 brain systems involved in drug-seeking and taking. The data so far indicate that different brain systems may be involved in different types of stimulus control as well as drug taking.
Collapse
Affiliation(s)
- Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON, Canada M5S 2S1; Alcohol Research and Treatment Clinic, Addiction Medicine Services, Ambulatory Care and Structured Treatments, Centre for Addiction and Mental Health, Toronto, Ontario, Canada M6J 1H4; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON, Canada M5S 2S1; Department of Family and Community Medicine, University of Toronto, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada; Department of Psychiatry, Division of Brain and Therapeutics, University of Toronto, Toronto, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Canada.
| | - Patricia Di Ciano
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, 33 Russell Street, Toronto, ON, Canada M5S 2S1
| |
Collapse
|
32
|
Boileau I, Nakajima S, Payer D. Imaging the D3 dopamine receptor across behavioral and drug addictions: Positron emission tomography studies with [(11)C]-(+)-PHNO. Eur Neuropsychopharmacol 2015; 25:1410-20. [PMID: 26141509 DOI: 10.1016/j.euroneuro.2015.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 04/07/2015] [Accepted: 06/12/2015] [Indexed: 12/21/2022]
Abstract
Chronic drug use has been associated with dopaminergic abnormalities, detectable in humans with positron emission tomography (PET). Among these, a hallmark feature is low D2 dopamine receptor availability, which has been linked to clinical outcomes, but has not yet translated into a therapeutic strategy. The D3 dopamine receptor on the other hand has gained increasing attention, as, in contrast to D2, chronic exposure to drugs has been shown to up-regulate this receptor subtype in preclinical models of addiction-a phenomenon linked to dopamine system sensitization and drug-seeking. The present article summarizes the literature to date in humans, suggesting that the D3 receptor may indeed contribute to core features of addiction such as impulsiveness and cognitive impairment. A particularly useful tool in investigating this question is the PET imaging probe [(11)C]-(+)-PHNO, which binds to D2/3 dopamine receptors but has preferential affinity for D3. This technique has been used to demonstrate D3 up-regulation in humans, and can be applied to assess pharmacological interventions for development of D3-targeted strategies in addiction treatment.
Collapse
Affiliation(s)
- Isabelle Boileau
- Addiction Imaging Research Group, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada; Campbell Family Mental Health Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| | - Shinichiro Nakajima
- Multimodal Imaging Group & Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, ON, Canada; Keio University, School of Medicine, Department of Neuropsychiatry, Tokyo, Japan
| | - Doris Payer
- Addiction Imaging Research Group, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Goutier W, O'Connor JJ, Lowry JP, McCreary AC. The effect of nicotine induced behavioral sensitization on dopamine D1 receptor pharmacology: An in vivo and ex vivo study in the rat. Eur Neuropsychopharmacol 2015; 25:933-43. [PMID: 25795518 DOI: 10.1016/j.euroneuro.2015.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/19/2014] [Accepted: 02/21/2015] [Indexed: 01/07/2023]
Abstract
Behavioral sensitization is a phenomenon which can develop following repeated intermittent administration of a range of psychostimulants, and other compounds, and may model neuroplastic changes seen in addictive processes and neuropsychiatric disease. The aim of the present study was to investigate the effect of dopamine D1 receptor (D1R) ligands on nicotine-induced behavioral sensitization and their molecular consequences in the striatum. Wistar rats were chronically treated (5 days) with vehicle or nicotine (0.4 mg/kg; s.c.) and locomotor activity was measured. Following a 5 day withdrawal period, rats were pretreated with vehicle or the D1R antagonist SCH-23390 (0.03 mg/kg; i.p.) and challenged with nicotine. Either 45 min or 24h post-challenge, the striatum was isolated and ex vivo receptor binding and cAMP accumulation (using LC-MS/MS) were assessed. It was shown that chronic nicotine administration induced the development and expression of locomotor sensitization, of which the latter was blocked by SCH-23390. Nicotine-induced sensitization had no effect on forskolin stimulated cAMP accumulation but increased the efficacy of dopamine for the D1R and decreased the potency of D1R agonists. These effects were antagonized by in vivo pre-challenge with SCH-23390. No effect on D1 receptor binding was observed. Moreover, time dependent effects were observed between tissue taken 45 min and 24h post-challenge. The present findings provide a connection between behavioral sensitization and intracellular cAMP accumulation through the D1R. Together these data suggest that changes in D1R signaling in the dorsal striatum may play an important role in the underlying mechanisms of nicotine-induced behavioral sensitization.
Collapse
Affiliation(s)
- W Goutier
- Abbott Healthcare Products B.V. (formerly Solvay Pharmaceuticals B.V.), C.J. van Houtenlaan 36, 1381 CP Weesp, The Netherlands; Department of Chemistry, National University of Ireland Maynooth, Maynooth, Co. Kildare, Ireland
| | - J J O'Connor
- UCD School of Biomolecular and Biomedical Science, Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin 4, Ireland; Department of Chemistry, National University of Ireland Maynooth, Maynooth, Co. Kildare, Ireland
| | - J P Lowry
- Department of Chemistry, National University of Ireland Maynooth, Maynooth, Co. Kildare, Ireland
| | - A C McCreary
- Abbott Healthcare Products B.V. (formerly Solvay Pharmaceuticals B.V.), C.J. van Houtenlaan 36, 1381 CP Weesp, The Netherlands.
| |
Collapse
|
34
|
Vousooghi N, Zarei SZ, Sadat-Shirazi MS, Eghbali F, Zarrindast MR. mRNA expression of dopamine receptors in peripheral blood lymphocytes of computer game addicts. J Neural Transm (Vienna) 2015; 122:1391-8. [DOI: 10.1007/s00702-015-1408-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/04/2015] [Indexed: 12/24/2022]
|
35
|
Smith LN, Bachus SE, McDonald CG, Smith RF. Role of the D3 dopamine receptor in nicotine sensitization. Behav Brain Res 2015; 289:92-104. [PMID: 25907750 DOI: 10.1016/j.bbr.2015.04.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 04/06/2015] [Accepted: 04/13/2015] [Indexed: 11/29/2022]
Abstract
Adolescent cigarette use is associated with reduced quitting success and continued smoking in adulthood. Interestingly, polymorphisms of the dopamine D3 receptor (DRD3) gene have been associated with smoking behavior, and the receptor is expressed in an age- and brain region-dependent manner that suggests relevance to addiction. Here, we investigate the possible role of dopamine-related receptors, including DRD3 and an intriguing splice variant known as D3nf, in nicotine-induced sensitization. In adolescent and adult male rats, we examined (1) alterations occurring in dopamine receptor-related mRNAs (DRD1, DRD2, DRD3 and D3nf) at two time points during a sensitizing regimen of nicotine and (2) whether DRD3 antagonism either during the initial treatment (induction) or at a later challenge exposure (expression) is able to block nicotine sensitization. Nicotine-induced changes were seen for DRD3 and D3nf mRNAs in the nucleus accumbens shell early in repeated exposure in both age groups. DRD3 antagonism only blocked the induction of sensitization in adolescents and did not block the expression of sensitization in either age group. Adolescents and adults showed opposite DRD1 mRNA responses to nicotine treatment, while no age- and nicotine-related changes in DRD2 mRNA were observed. These data reveal important age-dependent regulation of DRD1- and DRD3-related mRNAs during the course of nicotine exposure. Furthermore, they highlight a requirement for DRD3 signaling in the development of adolescent nicotine sensitization, suggesting it may represent an appropriate target in the prevention of nicotine dependence initiated at this age.
Collapse
Affiliation(s)
- Laura N Smith
- George Mason University, Department of Psychology, MSN 3F5, 4400 University Drive, Fairfax, VA 22030, USA.
| | - Susan E Bachus
- George Mason University, Department of Psychology, MSN 3F5, 4400 University Drive, Fairfax, VA 22030, USA
| | - Craig G McDonald
- George Mason University, Department of Psychology, MSN 3F5, 4400 University Drive, Fairfax, VA 22030, USA
| | - Robert F Smith
- George Mason University, Department of Psychology, MSN 3F5, 4400 University Drive, Fairfax, VA 22030, USA
| |
Collapse
|
36
|
Dopamine D3 receptors in the basolateral amygdala and the lateral habenula modulate cue-induced reinstatement of nicotine seeking. Neuropsychopharmacology 2014; 39:3049-58. [PMID: 24998621 PMCID: PMC4229576 DOI: 10.1038/npp.2014.158] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 05/29/2014] [Accepted: 05/30/2014] [Indexed: 01/27/2023]
Abstract
Dopamine D3 receptors are implicated in cue-induced relapse to drug seeking. We have previously shown that systemic administration of a selective D3 antagonist reduces cue-induced reinstatement of nicotine seeking in rats. The current study sought to investigate potential neural substrates mediating this effect. The D3 antagonist SB-277011-A (0.01-1 μg/0.5 μl/side) infused into the basolateral amygdala or the lateral habenula, but not the nucleus accumbens, significantly attenuated cue-induced reinstatement of nicotine seeking. Moreover, infusion of SB-277011-A (1 μg/0.5 μl/side) into the basolateral amygdala or lateral habenula had no effect on food self-administration. Together with the finding that systemic SB-277011-A had no effect on extinction responding, this suggests that the effects observed here were on reinstatement and cue seeking, and not due to nonspecific motor activation or contextual-modified residual responding. The further finding of binding of [(125)I]7-OH-PIPAT to D3 receptors in the lateral habenula and in the basolateral amygdala is consistent with an important role of D3 receptors in these areas in nicotine seeking. It was also found that systemic administration of the selective D2 antagonist L741626 decreased cue-induced reinstatement, consistent with a role of D2 and D3 receptors in modulating this behavior. The current study supports an important role for D3 receptors in the basolateral amygdala and lateral habenula in cue-induced reinstatement.
Collapse
|
37
|
In vivo evidence for greater amphetamine-induced dopamine release in pathological gambling: a positron emission tomography study with [(11)C]-(+)-PHNO. Mol Psychiatry 2014; 19:1305-13. [PMID: 24322203 DOI: 10.1038/mp.2013.163] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 08/06/2013] [Accepted: 09/30/2013] [Indexed: 01/09/2023]
Abstract
Drug addiction has been associated with deficits in mesostriatal dopamine (DA) function, but whether this state extends to behavioral addictions such as pathological gambling (PG) is unclear. Here we used positron emission tomography and the D3 receptor-preferring radioligand [(11)C]-(+)-PHNO during a dual-scan protocol to investigate DA release in response to oral amphetamine in pathological gamblers (n=12) and healthy controls (n=11). In contrast with human neuroimaging findings in drug addiction, we report the first evidence that PG is associated with greater DA release in dorsal striatum (54-63% greater [(11)C]-(+)-PHNO displacement) than controls. Importantly, dopaminergic response to amphetamine in gamblers was positively predicted by D3 receptor levels (measured in substantia nigra), and related to gambling severity, allowing for construction of a mechanistic model that could help explain DA contributions to PG. Our results are consistent with a hyperdopaminergic state in PG, and support the hypothesis that dopaminergic sensitization involving D3-related mechanisms might contribute to the pathophysiology of behavioral addictions.
Collapse
|
38
|
Keck TM, Burzynski C, Shi L, Newman AH. Beyond small-molecule SAR: using the dopamine D3 receptor crystal structure to guide drug design. ADVANCES IN PHARMACOLOGY 2014; 69:267-300. [PMID: 24484980 DOI: 10.1016/b978-0-12-420118-7.00007-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The dopamine D3 receptor is a target of pharmacotherapeutic interest in a variety of neurological disorders including schizophrenia, restless leg syndrome, and drug addiction. The high protein sequence homology between the D3 and D2 receptors has posed a challenge to developing D3 receptor-selective ligands whose behavioral actions can be attributed to D3 receptor engagement, in vivo. However, through primarily small-molecule structure-activity relationship (SAR) studies, a variety of chemical scaffolds have been discovered over the past two decades that have resulted in several D3 receptor-selective ligands with high affinity and in vivo activity. Nevertheless, viable clinical candidates remain limited. The recent determination of the high-resolution crystal structure of the D3 receptor has invigorated structure-based drug design, providing refinements to the molecular dynamic models and testable predictions about receptor-ligand interactions. This chapter will highlight recent preclinical and clinical studies demonstrating potential utility of D3 receptor-selective ligands in the treatment of addiction. In addition, new structure-based rational drug design strategies for D3 receptor-selective ligands that complement traditional small-molecule SAR to improve the selectivity and directed efficacy profiles are examined.
Collapse
Affiliation(s)
- Thomas M Keck
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, Baltimore, Maryland, USA
| | - Caitlin Burzynski
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, Baltimore, Maryland, USA
| | - Lei Shi
- Department of Physiology and Biophysics and Institute for Computational Biomedicine, Weill Cornell Medical College, New York, USA
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, Baltimore, Maryland, USA.
| |
Collapse
|
39
|
Garção P, Szabó EC, Wopereis S, Castro AA, Tomé ÂR, Prediger RD, Cunha RA, Agostinho P, Köfalvi A. Functional interaction between pre-synaptic α6β2-containing nicotinic and adenosine A2A receptors in the control of dopamine release in the rat striatum. Br J Pharmacol 2014; 169:1600-11. [PMID: 23638679 DOI: 10.1111/bph.12234] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 04/17/2013] [Accepted: 04/19/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Pre-synaptic nicotinic ACh receptors (nAChRs) and adenosine A2A receptors (A2A Rs) are involved in the control of dopamine release and are putative therapeutic targets in Parkinson's disease and addiction. Since A2A Rs have been reported to interact with nAChRs, here we aimed at mapping the possible functional interaction between A2A Rs and nAChRs in rat striatal dopaminergic terminals. EXPERIMENTAL APPROACH We pharmacologically characterized the release of dopamine and defined the localization of nAChR subunits in rat striatal nerve terminals in vitro and carried out locomotor behavioural sensitization in rats in vivo. KEY RESULTS In striatal nerve terminals, the selective A2A R agonist CGS21680 inhibited, while the A2A R antagonist ZM241385 potentiated the nicotine-stimulated [(3) H]dopamine ([(3) H]DA) release. Upon blockade of the α6 subunit-containing nAChRs, the remaining nicotine-stimulated [(3) H]DA release was no longer modulated by A2A R ligands. In the locomotor sensitization experiments, nicotine enhanced the locomotor activity on day 7 of repeated nicotine injection, an effect that no longer persisted after 1 week of drug withdrawal. Notably, ZM241385-injected rats developed locomotor sensitization to nicotine already on day 2, which remained persistent upon nicotine withdrawal. CONCLUSIONS AND IMPLICATIONS These results provide the first evidence for a functional interaction between nicotinic and adenosine A2A R in striatal dopaminergic terminals, with likely therapeutic consequences for smoking, Parkinson's disease and other dopaminergic disorders.
Collapse
Affiliation(s)
- P Garção
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Le Foll B, Wilson AA, Graff A, Boileau I, Di Ciano P. Recent methods for measuring dopamine D3 receptor occupancy in vivo: importance for drug development. Front Pharmacol 2014; 5:161. [PMID: 25071579 PMCID: PMC4090596 DOI: 10.3389/fphar.2014.00161] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 06/19/2014] [Indexed: 01/09/2023] Open
Abstract
There is considerable interest in developing highly selective dopamine (DA) D3 receptor ligands for a variety of mental health disorders. DA D3 receptors have been implicated in Parkinson's disease, schizophrenia, anxiety, depression, and substance use disorders. The most concrete evidence suggests a role for the D3 receptor in drug-seeking behaviors. D3 receptors are a subtype of D2 receptors, and traditionally the functional role of these two receptors has been difficult to differentiate. Over the past 10-15 years a number of compounds selective for D3 over D2 receptors have been developed. However, translating these findings into clinical research has been difficult as many of these compounds cannot be used in humans. Therefore, the functional data involving the D3 receptor in drug addiction mostly comes from pre-clinical studies. Recently, with the advent of [(11)C]-(+)-PHNO, it has become possible to image D3 receptors in the human brain with increased selectivity and sensitivity. This is a significant innovation over traditional methods such as [(11)C]-raclopride that cannot differentiate between D2 and D3 receptors. The use of [(11)C]-(+)-PHNO will allow for further delineation of the role of D3 receptors. Here, we review recent evidence that the role of the D3 receptor has functional importance and is distinct from the role of the D2 receptor. We then introduce the utility of analyzing [(11)C]-(+)-PHNO binding by region of interest. This novel methodology can be used in pre-clinical and clinical approaches for the measurement of occupancy of both D3 and D2 receptors. Evidence that [(11)C]-(+)-PHNO can provide insights into the function of D3 receptors in addiction is also presented.
Collapse
Affiliation(s)
- Bernard Le Foll
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health Toronto, ON, Canada ; Alcohol Research and Treatment Clinic, Addiction Medicine Services, Ambulatory Care and Structured Treatments, Centre for Addiction and Mental Health Toronto, ON, Canada ; Department of Family and Community Medicine, University of Toronto Toronto, ON, Canada ; Department of Pharmacology, University of Toronto Toronto, ON, Canada ; Division of Brain and Therapeutics, Department of Psychiatry, University of Toronto Toronto, ON, Canada ; Institute of Medical Sciences, University of Toronto Toronto, ON, Canada
| | - Alan A Wilson
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health Toronto, ON, Canada ; Division of Brain and Therapeutics, Department of Psychiatry, University of Toronto Toronto, ON, Canada ; Research Imaging Centre, Centre for Addiction and Mental Health Toronto, ON, Canada
| | - Ariel Graff
- Division of Brain and Therapeutics, Department of Psychiatry, University of Toronto Toronto, ON, Canada ; Research Imaging Centre, Centre for Addiction and Mental Health Toronto, ON, Canada ; Multimodal Imaging Group, Research Imaging Centre, Centre for Addiction and Mental Health Toronto, ON, Canada
| | - Isabelle Boileau
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health Toronto, ON, Canada ; Division of Brain and Therapeutics, Department of Psychiatry, University of Toronto Toronto, ON, Canada ; Institute of Medical Sciences, University of Toronto Toronto, ON, Canada ; Research Imaging Centre, Centre for Addiction and Mental Health Toronto, ON, Canada ; Addiction Imaging Research Group, Centre for Addiction and Mental Health Toronto, ON, Canada
| | - Patricia Di Ciano
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health Toronto, ON, Canada
| |
Collapse
|
41
|
Dopamine D3 receptor is necessary for ethanol consumption: an approach with buspirone. Neuropsychopharmacology 2014; 39:2017-28. [PMID: 24584330 PMCID: PMC4059912 DOI: 10.1038/npp.2014.51] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 02/12/2014] [Accepted: 02/24/2014] [Indexed: 11/09/2022]
Abstract
Mesolimbic dopamine (DA) controls drug- and alcohol-seeking behavior, but the role of specific DA receptor subtypes is unclear. We tested the hypothesis that D3R gene deletion or the D3R pharmacological blockade inhibits ethanol preference in mice. D3R-deficient mice (D3R(-/-)) and their wild-type (WT) littermates, treated or not with the D3R antagonists SB277011A and U99194A, were tested in a long-term free choice ethanol-drinking (two-bottle choice) and in a binge-like ethanol-drinking paradigm (drinking in the dark, DID). The selectivity of the D3R antagonists was further assessed by molecular modeling. Ethanol intake was negligible in D3R(-/-) and robust in WT both in the two-bottle choice and DID paradigms. Treatment with D3R antagonists inhibited ethanol intake in WT but was ineffective in D3R(-/-) mice. Ethanol intake increased the expression of RACK1 and brain-derived neurotrophic factor (BDNF) in both WT and D3R(-/-); in WT there was also a robust overexpression of D3R. Thus, increased expression of D3R associated with activation of RACK1/BDNF seems to operate as a reinforcing mechanism in voluntary ethanol intake. Indeed, blockade of the BDNF pathway by the TrkB selective antagonist ANA-12 reversed chronic stable ethanol intake and strongly decreased the striatal expression of D3R. Finally, we evaluated buspirone, an approved drug for anxiety disorders endowed with D3R antagonist activity (confirmed by molecular modeling analysis), that resulted effective in inhibiting ethanol intake. Thus, DA signaling via D3R is essential for ethanol-related reward and consumption and may represent a novel therapeutic target for weaning.
Collapse
|
42
|
Hadad NA, Knackstedt LA. Addicted to palatable foods: comparing the neurobiology of Bulimia Nervosa to that of drug addiction. Psychopharmacology (Berl) 2014; 231:1897-912. [PMID: 24500676 PMCID: PMC4484591 DOI: 10.1007/s00213-014-3461-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 01/20/2014] [Indexed: 12/18/2022]
Abstract
RATIONALE Bulimia nervosa (BN) is highly comorbid with substance abuse and shares common phenotypic and genetic predispositions with drug addiction. Although treatments for the two disorders are similar, controversy remains about whether BN should be classified as addiction. OBJECTIVES Here, we review the animal and human literature with the goal of assessing whether BN and drug addiction share a common neurobiology. RESULTS Similar neurobiological features are present following administration of drugs and bingeing on palatable food, especially sugar. Specifically, both disorders involve increases in extracellular dopamine (DA), D1 binding, D3 messenger RNA (mRNA), and ΔFosB in the nucleus accumbens (NAc). Animal models of BN reveal increases in ventral tegmental area (VTA) DA and enzymes involved in DA synthesis that resemble changes observed after exposure to addictive drugs. Additionally, alterations in the expression of glutamate receptors and prefrontal cortex activity present in human BN or following sugar bingeing in animals are comparable to the effects of addictive drugs. The two disorders differ in regards to alterations in NAc D2 binding, VTA DAT mRNA expression, and the efficacy of drugs targeting glutamate to treat these disorders. CONCLUSIONS Although additional empirical studies are necessary, the synthesis of the two bodies of research presented here suggests that BN shares many neurobiological features with drug addiction. While few Food and Drug Administration-approved options currently exist for the treatment of drug addiction, pharmacotherapies developed in the future, which target the glutamate, DA, and opioid systems, may be beneficial for the treatment of both BN and drug addiction.
Collapse
Affiliation(s)
- Natalie A Hadad
- Department of Psychology, University of Florida, P.O. Box 112250, Gainesville, FL, 32611-2250, USA
| | | |
Collapse
|
43
|
Hachimine P, Seepersad N, Ananthan S, Ranaldi R. The novel dopamine D3 receptor antagonist, SR 21502, reduces cocaine conditioned place preference in rats. Neurosci Lett 2014; 569:137-41. [DOI: 10.1016/j.neulet.2014.03.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 03/13/2014] [Accepted: 03/24/2014] [Indexed: 01/15/2023]
|
44
|
Janes AC, Farmer S, Frederick BD, Nickerson LD, Lukas SE. An increase in tobacco craving is associated with enhanced medial prefrontal cortex network coupling. PLoS One 2014; 9:e88228. [PMID: 24505440 PMCID: PMC3914963 DOI: 10.1371/journal.pone.0088228] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/03/2014] [Indexed: 12/05/2022] Open
Abstract
Craving is a key aspect of drug dependence that is thought to motivate continued drug use. Numerous brain regions have been associated with craving, suggesting that craving is mediated by a distributed brain network. Whether an increase in subjective craving is associated with enhanced interactions among brain regions was evaluated using resting state functional magnetic imaging (fMRI) in nicotine dependent participants. We focused on craving-related changes in the orbital and medial prefrontal cortex (OMPFC) network, which also included the subgenual anterior cingulate cortex (sgACC) extending into the ventral striatum. Brain regions in the OMPFC network are not only implicated in addiction and reward, but, due to their rich anatomic interconnections, may serve as the site of integration across craving-related brain regions. Subjective craving and resting state fMRI were evaluated twice with an ∼1 hour delay between the scans. Cigarette craving was significantly increased at the end, relative to the beginning of the scan session. Enhanced craving was associated with heightened coupling between the OMPFC network and other cortical, limbic, striatal, and visceromotor brain regions that are both anatomically interconnected with the OMPFC, and have been implicated in addiction and craving. This is the first demonstration confirming that an increase in craving is associated with enhanced brain region interactions, which may play a role in the experience of craving.
Collapse
Affiliation(s)
- Amy C. Janes
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
| | - Stacey Farmer
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
| | - Blaise deB. Frederick
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
| | - Lisa D. Nickerson
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
| | - Scott E. Lukas
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
| |
Collapse
|
45
|
Le Foll B, Collo G, Rabiner EA, Boileau I, Merlo Pich E, Sokoloff P. Dopamine D3 receptor ligands for drug addiction treatment: update on recent findings. PROGRESS IN BRAIN RESEARCH 2014; 211:255-75. [PMID: 24968784 DOI: 10.1016/b978-0-444-63425-2.00011-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The dopamine D3 receptor is located in the limbic area and apparently mediates selective effects on motivation to take drugs and drug-seeking behaviors, so that there has been considerable interest on the possible use of D3 receptor ligands to treat drug addiction. However, only recently selective tools allowing studying this receptor have been developed. This chapter presents an overview of findings that were presented at a symposium on the conference Dopamine 2013 in Sardinia in May 2013. Novel neurobiological findings indicate that drugs of abuse can lead to significant structural plasticity in rodent brain and that this is dependent on the availability of functional dopamine D3 autoreceptor, whose activation increased phosphorylation in the ERK pathway and in the Akt/mTORC1 pathway indicating the parallel engagement of a series of intracellular signaling pathways all involved in cell growth and survival. Preclinical findings using animal models of drug-seeking behaviors confirm that D3 antagonists have a promising profile to treat drug addiction across drugs of abuse type. Imaging the D3 is now feasible in human subjects. Notably, the development of (+)-4-propyl-9-hydroxynaphthoxazine ligand used in positron emission tomography (PET) studies in humans allows to measure D3 and D2 receptors based on the area of the brain under study. This PET ligand has been used to confirm up-regulation of D3 sites in psychostimulant users and to reveal that tobacco smoking produces elevation of dopamine at the level of D3 sites. There are now novel antagonists being developed, but also old drugs such as buspirone, that are available to test the D3 hypothesis in humans. The first results of clinical investigations are now being provided. Overall, those recent findings support further exploration of D3 ligands to treat drug addiction.
Collapse
Affiliation(s)
- Bernard Le Foll
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Alcohol Research and Treatment Clinic, Addiction Medicine Services, Ambulatory Care and Structured Treatments, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Family and Community Medicine, Pharmacology and Toxicology, Psychiatry, Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
| | - Ginetta Collo
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Eugenii A Rabiner
- Imanova, Centre for Imaging Sciences, London, UK; Centre for Neuroimaging Sciences, Institute of Psychiatry, King's College, London, UK
| | - Isabelle Boileau
- Addiction Imaging Research Group, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | | | | |
Collapse
|
46
|
Ouachikh O, Dieb W, Durif F, Hafidi A. Differential behavioral reinforcement effects of dopamine receptor agonists in the rat with bilateral lesion of the posterior ventral tegmental area. Behav Brain Res 2013; 252:24-31. [DOI: 10.1016/j.bbr.2013.05.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 05/21/2013] [Accepted: 05/23/2013] [Indexed: 01/12/2023]
|
47
|
Nolan BC, Liu S, Hammerslag LR, Cheung THC, Lenz J, Mach RH, Luedtke RR, Neisewander JL. Fos expression in response to dopamine D3-preferring phenylpiperazine drugs given with and without cocaine. Synapse 2013; 67:847-55. [PMID: 23766142 DOI: 10.1002/syn.21691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 06/06/2013] [Indexed: 11/07/2022]
Abstract
WC 44 and WC 10 are phenylpiperazines with low (23 fold) to moderate (42 fold) selectivity for dopamine D3 receptors (D3Rs) over D2Rs, respectively. WC 44 is a full D3R agonist in the forskolin-stimulated adenylyl cyclase (AC) assay, whereas WC 10 has little efficacy. In contrast to their opposite effects in the AC assay, these drugs often produce similar behavioral effects, suggesting that the AC assay does not predict the efficacy of these drugs in vivo. Here, we examined whether Fos protein expression induced by these drugs would be more consistent with their behavioral effects in vivo. Rats received either vehicle, WC 10 (5.6 mg/kg, i.p.), WC 44 (10.0 mg/kg, i.p), cocaine (10.0 mg/kg, i.p.), or cocaine with WC 10 (5.6 mg/kg, i.p.) or with WC 44 (10.0 mg/kg, i.p). Locomotion was monitored for 90 min and the brains were harvested for immunohistochemistry. Both WC 10 and WC 44 decreased spontaneous and cocaine-induced locomotion. Both compounds also increased Fos expression relative to saline in the dorsal striatum and nucleus accumbens core and shell, and relative to cocaine alone in the nucleus accumbens shell. The findings suggest that even though these compounds have different efficacy in the AC bioassy, they produce similar brain activation and attenuation of cocaine hyperlocomotion. Together with our previous research demonstrating that these compounds down-shift the cocaine self-administration dose-effect function, the findings support the idea that D3R-selective compounds may be useful for cocaine dependence medications development.
Collapse
Affiliation(s)
- Brian C Nolan
- Department of Psychology, Arizona State University, Tempe, Arizona, 85287; School of Life Sciences, Arizona State University, Tempe, Arizona, 85287
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Boileau I, Payer D, Chugani B, Lobo D, Behzadi A, Rusjan PM, Houle S, Wilson AA, Warsh J, Kish SJ, Zack M. The D2/3 dopamine receptor in pathological gambling: a positron emission tomography study with [11C]-(+)-propyl-hexahydro-naphtho-oxazin and [11C]raclopride. Addiction 2013; 108:953-63. [PMID: 23167711 DOI: 10.1111/add.12066] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 07/05/2012] [Accepted: 11/09/2012] [Indexed: 11/27/2022]
Abstract
AIMS Pathological gambling (PG) shares diagnostic features with substance use disorder (SUD), but the neurochemical mechanisms underlying PG are poorly understood. Because dopamine (DA), a neurotransmitter implicated in reward and reinforcement, is probably involved, we used positron emission tomography (PET) to test whether PG is associated with abnormalities in D2 and D3 receptor levels, as observed in SUD. DESIGN Case-control study comparing PG to healthy control (HC) subjects. SETTING Academic research imaging centre. PARTICIPANTS Thirteen non-treatment-seeking males meeting DSM-IV criteria for PG, and 12 matched HC (11 of whom completed PET). MEASUREMENTS Two PET scans (one with the D3 receptor preferring agonist [11C]-(+)-propyl-hexahydro-naphtho-oxazin (PHNO) and the other with [11C]raclopride) to assess D(2/3) DA receptor availability, and behavioural measures (self-report questionnaires and slot-machine game) to assess subjective effects and relationships to PET measures. FINDINGS Binding of both radiotracers did not differ between groups in striatum or substantia nigra (SN) (all P > 0.1). Across PG, [11C]-(+)-PHNO binding in SN, where the signal is attributable primarily to D3 receptors, correlated with gambling severity (r = 0.57, P = 0.04) and impulsiveness (r = 0.65, P = 0.03). In HC, [11C]raclopride binding in dorsal striatum correlated inversely with subjective effects of gambling (r = -0.70, P = 0.03) and impulsiveness (r = -0.70, P = 0.03). CONCLUSIONS Unlike with substance use disorder, there appear to be no marked differences in D2 /D3 levels between healthy subjects and pathological gamblers, suggesting that low receptor availability may not be a necessary feature of addiction. However, relationships between [11C]-(+)-PHNO binding and gambling severity/impulsiveness suggests involvement of the D3 receptor in impulsive/compulsive behaviours.
Collapse
Affiliation(s)
- Isabelle Boileau
- Addiction Imaging Research Group, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gozen O, Balkan B, Yildirim E, Koylu EO, Pogun S. The epigenetic effect of nicotine on dopamine D1 receptor expression in rat prefrontal cortex. Synapse 2013; 67:545-52. [PMID: 23447334 DOI: 10.1002/syn.21659] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 02/22/2013] [Indexed: 12/14/2022]
Abstract
Nicotine is a highly addictive drug and exerts its effect partially through causing dopamine release, thereby increasing intrasynaptic dopamine levels in the brain reward systems. Dopaine D1 receptor (DRD1) mRNAs and receptors are localized in reward-related brain regions, which receive cholinergic input. The aim of this study is to evaluate whether nicotine administration affects the expression of DRD1s, and if so, whether epigenetic mechanisms, such as histone acetylation, are involved. Twenty Male Sprague Dawley rats received nicotine (0.4 mg/kg/day, s.c.) or saline injections for 15 days. After nicotine/saline treatment, rats were perfused with saline; prefrontal cortex (PFC), corpus striatum (STR), and ventral tegmental area (VTA) were dissected. Homogenates were divided into two parts for total RNA isolation and histone H4 acetylation studies. DRD1 mRNA expression was significantly higher in the PFC of the nicotine-treated group compared with controls; similar trends were observed in the VTA and STR. To study epigenetic regulation, the 2kb upstream region of the DRD1 gene promoter was investigated for histone H4 acetylation in PFC samples. After chromatin immunoprecipitation with anti-acetyl histone H4 antibody, we found an increase in histone acetylation by two different primer pairs which amplified the -1365 to -1202 (P < 0.005) and -170 to +12 (P < 0.05) upstream regions of the DRD1 promoter. Our results suggest that intermittent subcutaneous nicotine administration increases the expression of DRD1 mRNA in the PFC of rats, and this increase may be due to changes in histone H4 acetylation of the 2kb promoter of the DRD1 gene.
Collapse
Affiliation(s)
- Oguz Gozen
- Ege University School of Medicine Department of Physiology, Izmir, Turkey.
| | | | | | | | | |
Collapse
|
50
|
Gross G, Drescher K. The role of dopamine D(3) receptors in antipsychotic activity and cognitive functions. Handb Exp Pharmacol 2013:167-210. [PMID: 23027416 DOI: 10.1007/978-3-642-25758-2_7] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Dopamine D(3) receptors have a pre- and postsynaptic localization in brain stem nuclei, limbic parts of the striatum, and cortex. Their widespread influence on dopamine release, on dopaminergic function, and on several other neurotransmitters makes them attractive targets for therapeutic intervention. The signaling pathways of D(3) receptors are distinct from those of other members of the D(2)-like receptor family. There is increasing evidence that D(3) receptors can form heteromers with dopamine D(1), D(2), and probably other G-protein-coupled receptors. The functional consequences remain to be characterized in more detail but might open new interesting pharmacological insight and opportunities. In terms of behavioral function, D(3) receptors are involved in cognitive, social, and motor functions, as well as in filtering and sensitization processes. Although the role of D(3) receptor blockade for alleviating positive symptoms is still unsettled, selective D(3) receptor antagonism has therapeutic features for schizophrenia and beyond as demonstrated by several animal models: improved cognitive function, emotional processing, executive function, flexibility, and social behavior. D(3) receptor antagonism seems to contribute to atypicality of clinically used antipsychotics by reducing extrapyramidal motor symptoms; has no direct influence on prolactin release; and does not cause anhedonia, weight gain, or metabolic dysfunctions. Unfortunately, clinical data with new, selective D(3) antagonists are still incomplete; their cognitive effects have only been communicated in part. In vitro, virtually all clinically used antipsychotics are not D(2)-selective but also have affinity for D(3) receptors. The exact D(3) receptor occupancies achieved in patients, particularly in cortical areas, are largely unknown, mainly because only nonselective or agonist PET tracers are currently available. It is unlikely that a degree of D(3) receptor antagonism optimal for antipsychotic and cognitive function can be achieved with existing antipsychotics. Therefore, selective D(3) antagonism represents a promising mechanism still to be fully exploited for the treatment of schizophrenia, cognitive deficits in schizophrenia, and comorbid conditions such as substance abuse.
Collapse
Affiliation(s)
- Gerhard Gross
- Abbott, Neuroscience Research, Ludwigshafen, Germany.
| | | |
Collapse
|