1
|
Lin Y, Wang J, Shi F, Yang L, Wu S, Qiao A, Ye S. Molecular Mechanisms of Methamphetamine-Induced Addiction via TAAR1 Activation. J Med Chem 2024; 67:18593-18605. [PMID: 39358311 PMCID: PMC11513891 DOI: 10.1021/acs.jmedchem.4c01961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Trace amine-associated receptor 1 (TAAR1), a member of the trace amine receptor family, recognizes various trace amines in the brain, including endogenous β-phenylethylamine (PEA) and methamphetamine (METH). TAAR1 is a novel target for several neurological disorders, including schizophrenia, depression, and substance abuse. Herein, we report the structure of the human TAAR1-Gs protein complex bound to METH. Using functional studies, we reveal the molecular basis of METH recognition by TAAR1, and potential mechanisms underlying the selectivity of TAAR1 for different ligands. Molecular dynamics simulations further elucidated possible mechanisms for the binding of chiral amphetamine (AMPH)-like psychoactive drugs to TAAR1. Additionally, we discovered a hydrophobic core on the transmembrane helices (TM), TM5 and TM6, explaining the unique mechanism of TAAR1 activation. These findings reveal the ligand recognition pattern and activation mechanism of TAAR1, which has important implications for the development of next-generation treatments for substance abuse and various neurological disorders.
Collapse
Affiliation(s)
- Yun Lin
- Tianjin
Key Laboratory of Function and Application of Biological Macromolecular
Structures, School of Life Sciences, Tianjin
University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Jiening Wang
- State
Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative
Innovation Center for Green Transformation of Bio-Resources, Hubei
Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Fan Shi
- Department
of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Linlin Yang
- Department
of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shan Wu
- State
Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative
Innovation Center for Green Transformation of Bio-Resources, Hubei
Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Anna Qiao
- Tianjin
Key Laboratory of Function and Application of Biological Macromolecular
Structures, School of Life Sciences, Tianjin
University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Sheng Ye
- Tianjin
Key Laboratory of Function and Application of Biological Macromolecular
Structures, School of Life Sciences, Tianjin
University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| |
Collapse
|
2
|
Wang L, Clark EA, Hanratty L, Koblan KS, Foley A, Dedic N, Bristow LJ. TAAR1 and 5-HT 1B receptor agonists attenuate autism-like irritability and aggression in rats prenatally exposed to valproic acid. Pharmacol Biochem Behav 2024; 245:173862. [PMID: 39197535 DOI: 10.1016/j.pbb.2024.173862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/02/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Despite the rising prevalence of autism spectrum disorder (ASD), there remains a significant unmet need for pharmacotherapies addressing its core and associative symptoms. While some atypical antipsychotics have been approved for managing associated irritability and aggression, their use is constrained by substantial side effects. This study aimed firstly to develop behavioral measures to explore frustration, irritability and aggression phenotypes in the rat prenatal valproic acid (VPA) model of ASD. Additionally, we investigated the potential of two novel mechanisms, 5-HT1B and TAAR1 agonism, to alleviate these behaviors. Male offspring exposed to prenatal VPA were trained to achieve stable performance on a cued operant task, followed by pharmacological assessment in an operant frustration test, bottle brush test and resident intruder test. VPA exposed rats demonstrated behaviors indicative of frustration and irritability, as well as increased aggression compared to controls. The irritability-like behavior and aggression were further exacerbated in animals previously experiencing a frustrative event during the operant test. Single administration of the 5-HT1B agonist CP-94253 or TAAR1 agonist RO5263397 attenuated the frustration-like behavior compared to vehicle. Additionally, both agonists reduced irritability-like behavior under both normal and frustrative conditions. While CP-94253 reduced aggression in the resident intruder test under both conditions, RO5263397 only produced effects in rats that previously experienced a frustrative event. Our study describes previously uncharacterized phenotypes of frustration, irritability, and aggression in the rat prenatal VPA model of ASD. Administration of selective TAAR1 or 5-HT1B receptor agonists alleviated these deficits, warranting further exploration of both targets in ASD treatment.
Collapse
Affiliation(s)
- Lien Wang
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | - Erin A Clark
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | | | | | | | - Nina Dedic
- Sumitomo Pharma America, Inc., Marlborough, MA, USA.
| | | |
Collapse
|
3
|
Nair PC, Bastiampillai T. Letters to the Editor: Letter to the Editor regarding 'A challenge to the dopamine orthodoxy in schizophrenia'. Aust N Z J Psychiatry 2023; 57:1394-1395. [PMID: 37465954 DOI: 10.1177/00048674231187998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Affiliation(s)
- Pramod C Nair
- Discipline of Clinical Pharmacology, Flinders Health and Medical Research Institute (FHMRI), College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Discipline of Medicine, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Tarun Bastiampillai
- Discipline of Psychiatry, Flinders Health and Medical Research Institute (FHMRI), College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Psychiatry, Monash University, Clayton, VIC, Australia
| |
Collapse
|
4
|
Wong TS, Li G, Li S, Gao W, Chen G, Gan S, Zhang M, Li H, Wu S, Du Y. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Target Ther 2023; 8:177. [PMID: 37137892 PMCID: PMC10154768 DOI: 10.1038/s41392-023-01427-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Manzhan Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China.
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, 518116, Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China.
| |
Collapse
|
5
|
Fan FS. Residues of ractopamine, a livestock feed additive, in meat might alleviate misuse of cocaine, nicotine, methamphetamine, and morphine. Nutr Health 2022; 29:171-174. [PMID: 36266952 DOI: 10.1177/02601060221134139] [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: 11/17/2022]
Abstract
Background: Substance misuse brings tremendous harm to global health. Strategies for the treatment and prevention of drug addiction are in urgent need. Aim: Trace amine-associated receptor 1 (TAAR1) widely distributed in the central nervous system has been identified as a hopeful target in the management of certain substance abuse. Discovery of food ingredients that act on TAAR1 might help health care providers develop chemoprevention for substance misuse disorders. Methods: Animal experiments clearly demonstrated the capability of TAAR1 agonists in attenuating addictive behavior regarding cocaine, nicotine, methamphetamine, and morphine. Ractopamine, a livestock feed additive used in the United States for over 20 years, has proven to be a full TAAR1 agonist. Literature review and internet web database survey were performed to see if ractopamine residues in meat could affect substance addiction behavior. Results: Integrating all available epidemiologic studies revealed that the prevalence of cocaine, nicotine, methamphetamine, and opioid misuse showed steadily downward or stable trends coincidently during the same time period of ractopamine use in the United States. Conclusion: A hypothesis is thus raised here that ractopamine residues in meat might have contributed secretly to the smoothened prevalence curves of cocaine, nicotine, methamphetamine, and opioids addiction.
Collapse
Affiliation(s)
- Frank S Fan
- Department of Medicine, Ministry of Health and Welfare Taitung Hospital, Taitung County, Taiwan
| |
Collapse
|
6
|
Liu J, Wu R, Johnson B, Zhang Y, Zhu Q, Li JX. Selective TAAR1 agonists induce conditioned taste aversion. Psychopharmacology (Berl) 2022; 239:3345-3353. [PMID: 36056214 DOI: 10.1007/s00213-022-06222-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/24/2022] [Indexed: 01/07/2023]
Abstract
RATIONALE Trace amine-associated receptor 1 (TAAR1) is the best-studied receptor of trace amines, a group of biogenic amines expressed at a relatively low level in the mammalian brain. Growing evidence suggests that TAAR1 plays a critical role in various neuropsychiatric disorders. Given that selective TAAR1 agonists were shown to produce pro-cognition and antipsychotic-like effects as well as to suppress drug use and relapse, they have been proposed to be novel treatments for mental disorders such as schizophrenia and addiction. However, the aversive effects of selective TAAR1 agonists remain largely unknown. OBJECTIVES Here, we evaluated whether the selective TAAR1 full agonist RO5166017 and partial agonist RO5263397 could induce conditioned taste aversion (CTA). RESULTS We found that RO5166017 and RO5263397 produced significant aversions to both saccharin and NaCl taste novelty. Furthermore, RO5166017 produced CTA to saccharin in TAAR1 heterozygous knockout (taar1±) and wild-type rats but not in TAAR1 homozygous knockout rats (taar1-/-), suggesting that TAAR1 was sufficient for the taste aversive stimulus property of RO5166017. CONCLUSIONS Taken together, our data indicate that selective TAAR1 agonists could produce strong CTA. Our study urges careful evaluations of the aversive effects of TAAR1 agonists before translating them to clinical use for the treatment of mental disorders.
Collapse
Affiliation(s)
- Jianfeng Liu
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China.,Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Ruyan Wu
- Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Bernard Johnson
- Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
7
|
Leo D, Targa G, Espinoza S, Villers A, Gainetdinov RR, Ris L. Trace Amine Associate Receptor 1 (TAAR1) as a New Target for the Treatment of Cognitive Dysfunction in Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23147811. [PMID: 35887159 PMCID: PMC9318502 DOI: 10.3390/ijms23147811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 02/01/2023] Open
Abstract
Worldwide, approximately 27 million people are affected by Alzheimer’s disease (AD). AD pathophysiology is believed to be caused by the deposition of the β-amyloid peptide (Aβ). Aβ can reduce long-term potentiation (LTP), a form of synaptic plasticity that is closely associated with learning and memory and involves postsynaptic glutamate receptor phosphorylation and trafficking. Moreover, Aβ seems to be able to reduce glutamatergic transmission by increasing the endocytosis of NMDA receptors. Trace amines (TAs) are biogenic amines that are structurally similar to monoamine neurotransmitters. TAs bind to G protein-coupled receptors, called TAARs (trace amine-associated receptors); the best-studied member of this family, TAAR1, is distributed in the cortical and limbic structures of the CNS. It has been shown that the activation of TAAR1 can rescue glutamatergic hypofunction and that TAAR1 can modulate glutamate NMDA receptor-related functions in the frontal cortex. Several lines of evidence also suggest the pro-cognitive action of TAAR1 agonists in various behavioural experimental protocols. Thus, we studied, in vitro, the role of the TAAR1 agonist RO5256390 on basal cortical glutamatergic transmission and tested its effect on Aβ-induced dysfunction. Furthermore, we investigated, in vivo, the role of TAAR1 in cognitive dysfunction induced by Aβ infusion in Aβ-treated mice. In vitro data showed that Aβ 1–42 significantly decreased NMDA cell surface expression while the TAAR1 agonist RO5256390 promoted their membrane insertion in cortical cells. In vivo, RO5256390 showed a mild pro-cognitive effect, as demonstrated by the better performance in the Y maze test in mice treated with Aβ. Further studies are needed to better understand the interplay between TAAR1/Aβ and glutamatergic signalling, in order to evaluate the eventual beneficial effect in different experimental paradigms and animal models. Taken together, our data indicate that TAAR1 agonism may provide a novel therapeutic approach in the treatments of disorders involving Aβ-induced cognitive impairments, such as AD.
Collapse
Affiliation(s)
- Damiana Leo
- Department of Neuroscience, Research Institute for Health Science and Technology, University of Mons, 20 Place du Parc, 7000 Mons, Belgium; (D.L.); (A.V.)
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy;
| | - Stefano Espinoza
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy;
| | - Agnès Villers
- Department of Neuroscience, Research Institute for Health Science and Technology, University of Mons, 20 Place du Parc, 7000 Mons, Belgium; (D.L.); (A.V.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya Emb. 7-9, 199034 St. Petersburg, Russia;
- St. Petersburg University Hospital, St. Petersburg State University, Universitetskaya Emb. 7-9, 199034 St. Petersburg, Russia
| | - Laurence Ris
- Department of Neuroscience, Research Institute for Health Science and Technology, University of Mons, 20 Place du Parc, 7000 Mons, Belgium; (D.L.); (A.V.)
- Correspondence: ; Tel.: +32-6537-3570
| |
Collapse
|
8
|
Raony Í, Domith I, Lourenco MV, Paes-de-Carvalho R, Pandolfo P. Trace amine-associated receptor 1 modulates motor hyperactivity, cognition, and anxiety-like behavior in an animal model of ADHD. Prog Neuropsychopharmacol Biol Psychiatry 2022; 117:110555. [PMID: 35346791 DOI: 10.1016/j.pnpbp.2022.110555] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 03/03/2022] [Accepted: 03/22/2022] [Indexed: 02/03/2023]
Abstract
Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that has recently been implicated in several psychiatric conditions related to monoaminergic dysfunction, such as schizophrenia, substance use disorders, and mood disorders. Although attention-deficit/hyperactivity disorder (ADHD) is also related to changes in monoaminergic neurotransmission, studies that assess whether TAAR1 participates in the neurobiology of ADHD are lacking. We hypothesized that TAAR1 plays an important role in ADHD and might represent a potential therapeutic target. Here, we investigate if TAAR1 modulates behavioral phenotypes in Spontaneously Hypertensive Rats (SHR), the most validated animal model of ADHD, and Wistar Kyoto rats (WKY, used as a control strain). Our results showed that TAAR1 is downregulated in ADHD-related brain regions in SHR compared with WKY. While intracerebroventricular (i.c.v.) administration of the selective TAAR1 antagonist EPPTB impaired cognitive performance in SHR, i.c.v. administration of highly selective TAAR1 full agonist RO5256390 decreased motor hyperactivity, novelty-induced locomotion, and induced an anxiolytic-like behavior. Overall, our findings show that changes in TAAR1 levels/activity underlie behavior in SHR, suggesting that TAAR1 plays a role in the neurobiology of ADHD. Although additional confirmatory studies are required, TAAR1 might be a potential pharmacological target for individuals with this disorder.
Collapse
Affiliation(s)
- Ícaro Raony
- Laboratory of Neurobiology of Animal Behavior, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ivan Domith
- Laboratory of Cellular Neurobiology, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Roberto Paes-de-Carvalho
- Laboratory of Cellular Neurobiology, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil
| | - Pablo Pandolfo
- Laboratory of Neurobiology of Animal Behavior, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil.
| |
Collapse
|
9
|
Liao S, Pino MJ, Deleon C, Lindner-Jackson M, Wu C. Interaction analyses of hTAAR1 and mTAAR1 with antagonist EPPTB. Life Sci 2022; 300:120553. [PMID: 35452636 DOI: 10.1016/j.lfs.2022.120553] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022]
Abstract
Trace amine-associated receptor 1 (TAAR1) plays a critical role in regulating monoaminergic activity. EPPTB is the only known selective potent antagonist of the mouse (m) TAAR1 presently, while it was shown to be weak at antagonizing human (h) TAAR1. The lack of high-resolution structure of TAAR1 hinders the understanding of the differences in the interaction modes between EPPTB and m/hTARR1. The purpose of this study is to probe these interaction modes using homology modeling, molecular docking, molecular dynamics (MD) simulations, and molecular mechanics-generalized Born surface area (MM-GBSA) binding energy calculations. Eight populated conformers of hTAAR1-EPPTB complex were observed during the MD simulations and could be used in structure-based virtual screening in future. The MM-GBSA binding energy of hTAAR1-EPPTB complex (-96.5 kcal/mol) is larger than that of mTAAR1-EPPTB complex (-106.7 kcal/mol), which is consistent with the experimental finding that EPPTB has weaker binding affinity to hTAAR1. The several residues in binding site of hTAAR1 (F1544.56, T1945.42 and I2907.39) are different from these of mTAAR1 (Y1534.56, A1935.42 and Y2877.39), which might contribute to the binding affinity difference. Our docking analysis on another hTAAR1 antagonist Compound 3 has found that 1). this compound binds in different pockets of our mTAAR1 and hTAAR1 homology models with a slightly stronger binding affinity to hTAAR1; 2). both antagonists bind to a very similar pocket of hTAAR1.
Collapse
Affiliation(s)
- Siyan Liao
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Michael James Pino
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States of America
| | - Catherine Deleon
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States of America
| | - Maurice Lindner-Jackson
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States of America
| | - Chun Wu
- Department of Chemistry and Biochemistry, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, United States of America.
| |
Collapse
|
10
|
Potential of Ligands for Trace Amine-Associated Receptor 1 (TAAR1) in the Management of Substance Use Disorders. CNS Drugs 2021; 35:1239-1248. [PMID: 34766253 PMCID: PMC8787759 DOI: 10.1007/s40263-021-00871-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/17/2021] [Indexed: 10/19/2022]
Abstract
Trace amines, including β-phenylethylamine (β-PEA), p-tyramine (TYR), tryptamine (TRP), and p-octopamine (OCT), represent a group of amines expressed at low levels in the mammalian brain. Given the close structural similarities to traditional monoamines, links between trace amines and the monoaminergic system have long been suspected. Trace amine-associated receptor 1 (TAAR1), the most well characterized receptor in the TAAR family, has been shown to be potently activated by trace amines such as TYR and PEA. Further, catecholamine metabolites and amphetamine analogs are also potent agonists of TAAR1, implicating the receptor in mediating the monoaminergic system and in substance use disorders. In the central nervous system, TAAR1 is expressed in brain regions involved in dopaminergic, serotonergic, and glutamatergic transmission, and genetic animal models and electrophysiological studies have revealed that TAAR1 is a potent modulator of the monoaminergic system. Selective and potent engineered TAAR1 ligands, including full (RO5166017 and RO5256390) and partial (RO5203648, RO5263397 and RO5073012) agonists and the antagonist EPPTB (N-(3-ethoxyphenyl)-4-(1-pyrrolidinyl)-3-(trifluoromethyl) benzamide, RO5212773), serve as invaluable tools for the investigation of TAAR1 functions and display significant potential for the development of TAAR1-based pharmacotherapies for the treatment of substance use disorders. Despite a number of advances that have been made, more clinical studies are warranted in order to test the potential and efficacy of TAAR1 ligands in the treatment of psychiatric disorders, including substance use disorders.
Collapse
|
11
|
Kiguchi N, Ko MC. Potential therapeutic targets for the treatment of opioid abuse and pain. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:335-371. [PMID: 35341570 PMCID: PMC10948018 DOI: 10.1016/bs.apha.2021.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although μ-opioid peptide (MOP) receptor agonists are effective analgesics available in clinical settings, their serious adverse effects put limits on their use. The marked increase in abuse and misuse of prescription opioids for pain relief and opioid overdose mortality in the past decade has seriously impacted society. Therefore, safe analgesics that produce potent analgesic effects without causing MOP receptor-related adverse effects are needed. This review highlights the potential therapeutic targets for the treatment of opioid abuse and pain based on available evidence generated through preclinical studies and clinical trials. To ameliorate the abuse-related effects of opioids, orexin-1 receptor antagonists and mixed nociceptin/MOP partial agonists have shown promising results in translational aspects of animal models. There are several promising non-opioid targets for selectively inhibiting pain-related responses, including nerve growth factor inhibitors, voltage-gated sodium channel inhibitors, and cannabinoid- and nociceptin-related ligands. We have also discussed several emerging and novel targets. The current medications for opioid abuse are opioid receptor-based ligands. Although neurobiological studies in rodents have discovered several non-opioid targets, there is a translational gap between rodents and primates. Given that the neuroanatomical aspects underlying opioid abuse and pain are different between rodents and primates, it is pivotal to investigate the functional profiles of these non-opioid compounds compared to those of clinically used drugs in non-human primate models before initiating clinical trials. More pharmacological studies of the functional efficacy, selectivity, and tolerability of these newly discovered compounds in non-human primates will accelerate the development of effective medications for opioid abuse and pain.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan.
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
12
|
Wu R, Liu J, Seaman R, Johnson B, Zhang Y, Li JX. The selective TAAR1 partial agonist RO5263397 promoted novelty recognition memory in mice. Psychopharmacology (Berl) 2021; 238:3221-3228. [PMID: 34291306 PMCID: PMC8605990 DOI: 10.1007/s00213-021-05937-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/09/2021] [Indexed: 11/28/2022]
Abstract
RATIONALE Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that has a particular role in regulating dopaminergic, serotonergic, and glutamatergic transmission. TAAR1 agonists have shown pro-cognitive activities. However, it remains largely unknown of the effects of TAAR1 agonists on memory performance. OBJECTIVES Here, by using the mice novel object recognition (NOR) test, we examined the effects of the selective TAAR1 partial agonist RO5263397 on recognition memory. RESULTS We found that RO5263397 significantly enhanced the retrieval of short-term memory (STM; 20 min after training) both in male and female mice. RO5263397 promoted the retrieval of STM in the wild-type (WT) littermates but not TAAR1-KO mice, indicating that the effects of RO5263397 were dependent on TAAR1. Interestingly, compared to their WT litters, TAAR1-KO mice showed similar levels of STM, suggesting that genetic deletion of taar1 gene did not affect the STM retrieval. Furthermore, RO5263397 also promoted the retrieval of long-term NOR memory (24 h after training). CONCLUSIONS These results indicate that TAAR1 activation promotes NOR memory retrieval. Consistent with previous studies, our finding further suggests that TAAR1 agonists have pro-cognitive properties.
Collapse
Affiliation(s)
- Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA.,Medical college of Yangzhou University, Yangzhou, China
| | - Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Robert Seaman
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Bernard Johnson
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University At Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14214, USA.
| |
Collapse
|
13
|
Kozlowska U, Nichols C, Wiatr K, Figiel M. From psychiatry to neurology: Psychedelics as prospective therapeutics for neurodegenerative disorders. J Neurochem 2021; 162:89-108. [PMID: 34519052 DOI: 10.1111/jnc.15509] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/30/2021] [Accepted: 09/05/2021] [Indexed: 12/22/2022]
Abstract
The studies of psychedelics, especially psychedelic tryptamines like psilocybin, are rapidly gaining interest in neuroscience research. Much of this interest stems from recent clinical studies demonstrating that they have a unique ability to improve the debilitating symptoms of major depressive disorder (MDD) long-term after only a single treatment. Indeed, the Food and Drug Administration (FDA) has recently designated two Phase III clinical trials studying the ability of psilocybin to treat forms of MDD with "Breakthrough Therapy" status. If successful, the use of psychedelics to treat psychiatric diseases like depression would be revolutionary. As more evidence appears in the scientific literature to support their use in psychiatry to treat MDD on and substance use disorders (SUD), recent studies with rodents revealed that their therapeutic effects might extend beyond treating MDD and SUD. For example, psychedelics may have efficacy in the treatment and prevention of brain injury and neurodegenerative diseases such as Alzheimer's Disease. Preclinical work has highlighted psychedelics' ability to induce neuroplasticity and synaptogenesis, and neural progenitor cell proliferation. Psychedelics may also act as immunomodulators by reducing levels of proinflammatory biomarkers, including IL-1β, IL-6, and tumor necrosis factor-α (TNF-α). Their exact molecular mechanisms, and induction of cellular interactions, especially between neural and glial cells, leading to therapeutic efficacy, remain to be determined. In this review, we discuss recent findings and information on how psychedelics may act therapeutically on cells within the central nervous system (CNS) during brain injuries and neurodegenerative diseases.
Collapse
Affiliation(s)
- Urszula Kozlowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland.,Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Charles Nichols
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Kalina Wiatr
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Maciej Figiel
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
14
|
Sapegin AV, Peshkov AA, Kanov EV, Gainetdinov RR, Duszyńska B, Bojarski AJ, Krasavin M. Novel medium-sized di(het)areno-fused 1,4,7-(oxa)thiadiazecines as probes for aminergic receptors. MENDELEEV COMMUNICATIONS 2021. [DOI: 10.1016/j.mencom.2021.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Levashova E, Firsov A, Bakulina O, Peshkov A, Kanov E, Gainetdinov RR, Krasavin M. Rare cis-configured 2,4-disubstituted 1-alkylpiperidines: synthesized and tested against trace-amine-associated receptor 1 (TAAR1). MENDELEEV COMMUNICATIONS 2021. [DOI: 10.1016/j.mencom.2021.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
16
|
Metabolomics reveals biomarkers of opioid use disorder. Transl Psychiatry 2021; 11:103. [PMID: 33542199 PMCID: PMC7862627 DOI: 10.1038/s41398-021-01228-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/08/2021] [Accepted: 01/18/2021] [Indexed: 01/30/2023] Open
Abstract
Opioid use disorder (OUD) is diagnosed using the qualitative criteria defined by the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Diagnostic biomarkers for OUD do not currently exist. Our study focused on developing objective biological markers to differentiate chronic opiate users with OUD from chronic opiate users without OUD. Using biospecimens from the Golestan Cohort Study, we compared the metabolomics profiles of high opium users who were diagnosed as OUD positive with high opium users who were diagnosed as OUD negative. High opium use was defined as maximum weekly opium usage greater than or equal to the median usage (2.4 g per week), and OUD was defined as having 2 or more DSM-5 criteria in any 12-month period. Among the 218 high opium users in this study, 80 were diagnosed as OUD negative, while 138 were diagnosed as OUD positive. Seven hundred and twelve peaks differentiated high opium users diagnosed as OUD positive from high opium users diagnosed as OUD negative. Stepwise logistic regression modeling of subject characteristics data together with the 712 differentiating peaks revealed a signature that is 95% predictive of an OUD positive diagnosis, a significant (p < 0.0001) improvement over a 63% accurate prediction based on subject characteristic data for these samples. These results suggest that a metabolic profile can be used to predict an OUD positive diagnosis.
Collapse
|
17
|
Liu J, Seaman R, Johnson B, Wu R, Vu J, Tian J, Zhang Y, Li JX. Activation of trace amine-associated receptor 1 selectively attenuates the reinforcing effects of morphine. Br J Pharmacol 2021; 178:933-945. [PMID: 33247948 DOI: 10.1111/bph.15335] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Trace amine-associated TA1 receptors play critical roles in regulating dopamine transmission. Previous studies showed that pharmacologically or genetically manipulating the activity of TA1 receptors modulates addiction-like behaviours associated with psychostimulants. However, little is known about whether TA1 receptor modulation would regulate the behavioural effects of opioids. EXPERIMENTAL APPROACH Effects of the selective TA1 receptor partial agonist RO5263397 on the addiction-related and antinociceptive effects of morphine were systematically assessed in male rats and mice. KEY RESULTS RO5263397 attenuated the expression of morphine-induced behavioural sensitization in wildtype but not TA1 receptor knockout mice. RO5263397 shifted the dose-effect curve of morphine self-administration downward and reduced the breakpoint in a progressive ratio schedule of reinforcement but did not affect food self-administration in rats. RO5263397 decreased the cue- and drug-induced reinstatement of morphine-seeking behaviour in rats. RO5263397 alone did not trigger reinstatement of morphine-seeking behaviour or change locomotor activity in rats with a history of morphine self-administration. However, RO5263397 did not affect the expression of morphine-induced conditioned place preference in mice or rats. RO5263397 did not affect naltrexone-precipitated jumping behaviour or naltrexone-induced conditioned place aversion in morphine-dependent mice. Furthermore, RO5263397 did not affect the analgesic effects of morphine in an acute nociception model in mice and a chronic pain model in rats. CONCLUSION AND IMPLICATIONS These results indicated that TA1 receptor activation selectively attenuated the reinforcing, but not withdrawal or antinociceptive effects of morphine, suggesting that selective TA1 receptor agonists might be useful to combat opioid addiction, while sparing the analgesic effects.
Collapse
Affiliation(s)
- Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Robert Seaman
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Bernard Johnson
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Jimmy Vu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Jingwei Tian
- School of Pharmacy, Yantai University, Yantai, China
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, North Carolina, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
18
|
Dodd S, F Carvalho A, Puri BK, Maes M, Bortolasci CC, Morris G, Berk M. Trace Amine-Associated Receptor 1 (TAAR1): A new drug target for psychiatry? Neurosci Biobehav Rev 2020; 120:537-541. [PMID: 33031817 DOI: 10.1016/j.neubiorev.2020.09.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/31/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023]
Abstract
There are nine subfamilies of TAARs. They are predominantly intracellular, located in the central nervous system and peripherally. They have a role in homeostasis and rheostasis, and also in olfaction. They demonstrate significant cross-talk with the monoamine system and are involved in the regulation of cAMP signalling and K+ channels. There is evidence to suggest that TAAR1 may be a promising therapeutic target for the treatment of schizophrenia, psychosis in Parkinson's disease, substance use disorders, and the metabolic syndrome and obesity. TAAR1 expression may also be a prognostic biomarker for cancers. A number of TAAR modulators have been identified, including endogenous ligands and new chemical entities. Some of these agents have shown efficacy in animal models of addiction behaviours, depression and anxiety. Only one agent, SEP-363856, has progressed to randomised clinical trials in humans; however further, larger studies with SEP-363856 are required to clarify its suitability as a new treatment for schizophrenia spectrum disorders. SEP-363856 is an agonist of TAAR1 and 5HT1A and it is not clear to what extent its efficacy can be attributed to TAAR1 rather than to other drug targets. However, current research suggests that TAAR1 has an important role in human physiology and pathophysiology. TAAR1 modulators may become an important new drug class for the management of a wide array of mental disorders in the future.
Collapse
Affiliation(s)
- Seetal Dodd
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia; Centre for Youth Mental Health, University of Melbourne, Parkville, Australia; Department of Psychiatry, University of Melbourne, Parkville, Australia; University Hospital Geelong, Barwon Health, PO Box 281, Geelong, Victoria, 3220, Australia.
| | - André F Carvalho
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, Toronto, ON, Canada
| | | | - Michael Maes
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Chiara C Bortolasci
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia; Centre for Molecular and Medical Research, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Gerwyn Morris
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia; Centre for Youth Mental Health, University of Melbourne, Parkville, Australia; Department of Psychiatry, University of Melbourne, Parkville, Australia; University Hospital Geelong, Barwon Health, PO Box 281, Geelong, Victoria, 3220, Australia
| |
Collapse
|
19
|
Does Manganese Contribute to Methamphetamine-Induced Psychosis? CURRENT EMERGENCY AND HOSPITAL MEDICINE REPORTS 2020. [DOI: 10.1007/s40138-020-00221-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Tsapakis EM, Preti A, Mintzas MD, Fountoulakis KN. Adjunctive treatment with psychostimulants and stimulant-like drugs for resistant bipolar depression: a systematic review and meta-analysis. CNS Spectr 2020; 26:1-12. [PMID: 32641179 DOI: 10.1017/s109285292000156x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Depression is considered to be the most difficult to treat phase of bipolar disorder as patients experience residual symptoms causing long-term disability. This work aims to explore the role of add-on stimulant and stimulant-like medication in resistant bipolar depression patients. METHODS Systematic review of add-on stimulants and stimulant-like drugs in resistant bipolar depression by following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Analysis was performed using the random-effects models. Heterogeneity was evaluated with Cochran's Q and I2 statistics. RESULTS Six randomized controlled trials of add-on modafinil, armodafinil, and lisdexamphetamine (LDX) (n = 813) vs placebo (n = 815) in the treatment of resistant bipolar depression were included. These drugs were more likely to induce remission from an episode of resistant bipolar depression (relative risk [RR] = 1.37; 95% confidence interval [CI]: 1.06-1.77; number needed to treat for an additional beneficial outcome = 16). Moreover, they did not induce more dropouts than placebo (RR = 1.04; 95% CI: 0.91-1.18), nor did they increase the risk of adverse effects (53/772 vs 41/771) at the end of treatment (RR = 1.30; 95% CI: 0.81-2.10; number needed to treat for an additional harmful outcome = 62). Suicidality and manic switch were not affected by active treatment. Heterogeneity was low (Cochran's Q: P > .05), but sometimes with a large CI. CONCLUSIONS LDX, modafinil, and armodafinil seem to offer a reasonably well-tolerated and safe treatment in resistant bipolar depression. Treatment guidelines should, therefore, be revised to include these medications earlier in the therapeutic algorithm for resistant acute bipolar depression. Further research is, however, necessary for the elucidation of the clinical usefulness of these and other similar compounds.
Collapse
Affiliation(s)
- Evangelia Maria Tsapakis
- Agios Charalambos Mental Health Clinic, Heraklion, Greece
- First Department of Psychiatry, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Antonio Preti
- Genneruxi Medical Center, Cagliari, Italy
- Center for Consultation-Liaison Psychiatry and Psychosomatics, University Hospital of Cagliari, Cagliari, Italy
| | | | - Konstantinos N Fountoulakis
- Third Department of Psychiatry, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
21
|
Wu R, Liu J, Wang K, Huang Y, Zhang Y, Li JX. Effects of a trace amine-associated receptor 1 agonist RO 5263397 on ethanol-induced behavioral sensitization. Behav Brain Res 2020; 390:112641. [PMID: 32407821 DOI: 10.1016/j.bbr.2020.112641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alcohol dependence is a chronic and severe health problem which puts a heavy burden on society. Alcohol activates mesolimbic dopamine circuity to achieve its reinforcing effect. While TAAR1 is critically involved in the modulation of dopamine, there is little evidence indicating that TAAR1 could play a role in behavioral effects of ethanol. METHODS By using the animal model of behavioral sensitization induced by ethanol in mice, the present study was performed to investigate whether the activation of TAAR1 would affect the behavioral plasticity of ethanol. RESULTS Repeated administration with ethanol induced a significant increased locomotion in WT mice with females showing higher level of sensitization to ethanol than male mice. The TAAR1 agonist RO5263397 significantly decreased the expression of ethanol-induced behavioral sensitization both in male and female WT mice (0.1 and 0.32 mg/kg). Repeated RO5263397 exposure also prevented the development of behavioral sensitization to ethanol both in male and female WT mice. Moreover, while TAAR1-KO mice developed normal levels of ethanol-induced behavioral sensitization, RO5263397 did not affect this behavior in TAAR1-KO mice. CONCLUSIONS These results indicated that the TAAR1 agonist RO5263397 negatively regulated the expression and development of ethanol-elicited behavioral sensitization in WT but not in TAAR1-KO mice. The present study suggests that TAAR1 is probably involved in certain addiction-like effects of alcohol and could be a useful drug target for the development of new medications to treat alcohol dependence.
Collapse
Affiliation(s)
- Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States; School of Medicine, Yangzhou University, Yangzhou, China
| | - Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Kaixuan Wang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Yufei Huang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Yanan Zhang
- Research Triangle Institute, NC, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
22
|
Liu J, Johnson B, Wu R, Seaman R, Vu J, Zhu Q, Zhang Y, Li JX. TAAR1 agonists attenuate extended-access cocaine self-administration and yohimbine-induced reinstatement of cocaine-seeking. Br J Pharmacol 2020; 177:3403-3414. [PMID: 32246467 DOI: 10.1111/bph.15061] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 03/06/2020] [Accepted: 03/21/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE The trace amine-associated receptor 1 (TAAR1) negatively modulates dopamine transmission. Our previous studies demonstrated that TAAR1 agonists attenuated cue- and drug-induced cocaine-seeking and increased the elasticity of the cocaine demand curve, in the short-access cocaine self-administration model. Compulsive use of cocaine, which is an essential criterion of cocaine use disorder, can be induced by extended access to cocaine self-administration. EXPERIMENTAL APPROACH To characterize the role of TAAR1 in compulsive cocaine use, we evaluated the effects of activation of TAAR1 on cocaine intake, cocaine binge and cue-induced cocaine-seeking using the extended-access cocaine self-administration model in adult male Sprague-Dawley rats. We also investigated the role of TAAR1 in stress-triggered cocaine relapse by using the α2 -adrenoceptor antagonist yohimbine-induced reinstatement of cocaine-seeking. KEY RESULTS The selective TAAR1 partial agonist RO5263397 attenuated cocaine intake and did not develop tolerance during the 10-day extended-access cocaine self-administration. RO5263397 reduced a 12-h binge intake of cocaine after forced abstinence. RO5263397 also decreased cue-induced cocaine-seeking after prolonged abstinence from extended-access cocaine self-administration. Furthermore, RO5263397 and the selective TAAR1 full agonist RO5166017 reduced yohimbine-induced reinstatement of cocaine-seeking behaviour. CONCLUSION AND IMPLICATIONS Activation of TAAR1 attenuated extended-access cocaine self-administration and stress-induced cocaine reinstatement. These results suggest that TAAR1 agonists are promising pharmacological interventions to treat cocaine use disorder and relapse.
Collapse
Affiliation(s)
- Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Bernard Johnson
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA.,School of Medicine, Yangzhou University, Yangzhou, China
| | - Robert Seaman
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Jimmy Vu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong, China
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, North Carolina, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
23
|
Abstract
Trace amine-associated receptor 1 is one of the best-characterized receptors of trace amines. Growing evidence shows that TAAR1 negatively regulates the monoaminergic activity, including dopamine transmission in the mesocorticolimbic system. Neurochemical assays demonstrated that selective TAAR1 full and partial agonists were effective to prevent psychostimulants-induced dopamine transmission in vitro and in vivo. In the last decade, many preclinical models of psychostimulant addiction such as drug-induced behavioral sensitization, drug-induced conditioned place preference, drug self-administration, drug discrimination, and relapse models were used to assess the effects of TAAR1 agonists on psychostimulants' behavioral effects. In general, activation of TAAR1 attenuated while knockout of TAAR1 potentiated psychostimulant abuse-related behaviors. Here, we review the advances in TAAR1 and its agonists in modulating psychostimulant addiction. We discuss the similarities and differences between the neurochemical and behavioral effects of TAAR1 full and partial agonists. We also discuss several concerns including the abuse liability, sleep reduction, and species-dependent effects that might affect the successful translation of TAAR1 agonists from preclinical studies to clinical application. In conclusion, although further investigations are in need to address certain concerns and the underlying neural mechanisms, TAAR1 agonists appear to be a promising pharmacotherapy to treat psychostimulant addiction and prevent relapse.
Collapse
|
24
|
Loftis JM, Lasarev M, Shi X, Lapidus J, Janowsky A, Hoffman WF, Huckans M. Trace amine-associated receptor gene polymorphism increases drug craving in individuals with methamphetamine dependence. PLoS One 2019; 14:e0220270. [PMID: 31600226 PMCID: PMC6786581 DOI: 10.1371/journal.pone.0220270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/27/2019] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Methamphetamine (MA) is a potent agonist at the trace amine-associated receptor 1 (TAAR1). This study evaluated a common variant (CV) in the human TAAR1 gene, synonymous single nucleotide polymorphism (SNP) V288V, to determine the involvement of TAAR1 in MA dependence. METHODS Participants (n = 106) with active MA dependence (MA-ACT), in remission from MA dependence (MA-REM), with active polysubstance dependence, in remission from polysubstance dependence, and with no history of substance dependence completed neuropsychiatric symptom questionnaires and provided blood samples. In vitro expression and function of CV and wild type TAAR1 receptors were also measured. RESULTS The V288V polymorphism demonstrated a 40% increase in TAAR1 protein expression in cell culture, but message sequence and protein function were unchanged, suggesting an increase in translation efficiency. Principal components analysis resolved neuropsychiatric symptoms into four components, PC1 (depression, anxiety, memory, and fatigue), PC2 (pain), PC3 (drug and alcohol craving), and PC4 (sleep disturbances). Analyses of study group and TAAR1 genotype revealed a significant interaction for PC3 (craving response) (p = 0.003). The control group showed no difference in PC3 associated with TAAR1, while adjusted mean craving for the MA-ACT and MA-REM groups, among those with at least one copy of V288V, was estimated to be, respectively, 1.55 (p = 0.036) and 1.77 (p = 0.071) times the adjusted mean craving for those without the TAAR1 SNP. CONCLUSIONS Neuroadaptation to chronic MA use may be altered by TAAR1 genotype and result in increased dopamine signaling and craving in individuals with the V288V genotype.
Collapse
Affiliation(s)
- Jennifer M. Loftis
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
| | - Michael Lasarev
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Oregon Health & Science University and Portland State University School of Public Health, Portland, OR, United States of America
| | - Xiao Shi
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
| | - Jodi Lapidus
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Oregon Health & Science University and Portland State University School of Public Health, Portland, OR, United States of America
| | - Aaron Janowsky
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States of America
| | - William F. Hoffman
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States of America
- Mental Health and Clinical Neurosciences Division, VA Portland Health Care System, Portland, OR, United States of America
| | - Marilyn Huckans
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Mental Health and Clinical Neurosciences Division, VA Portland Health Care System, Portland, OR, United States of America
| |
Collapse
|
25
|
Stafford AM, Reed C, Baba H, Walter NAR, Mootz JRK, Williams RW, Neve KA, Fedorov LM, Janowsky AJ, Phillips TJ. Taar1 gene variants have a causal role in methamphetamine intake and response and interact with Oprm1. eLife 2019; 8:e46472. [PMID: 31274109 PMCID: PMC6682400 DOI: 10.7554/elife.46472] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/04/2019] [Indexed: 12/17/2022] Open
Abstract
We identified a locus on mouse chromosome 10 that accounts for 60% of the genetic variance in methamphetamine intake in mice selectively bred for high versus low methamphetamine consumption. We nominated the trace amine-associated receptor 1 gene, Taar1, as the strongest candidate and identified regulation of the mu-opioid receptor 1 gene, Oprm1, as another contributor. This study exploited CRISPR-Cas9 to test the causal role of Taar1 in methamphetamine intake and a genetically-associated thermal response to methamphetamine. The methamphetamine-related traits were rescued, converting them to levels found in methamphetamine-avoiding animals. We used a family of recombinant inbred mouse strains for interval mapping and to examine independent and epistatic effects of Taar1 and Oprm1. Both methamphetamine intake and the thermal response mapped to Taar1 and the independent effect of Taar1 was dependent on genotype at Oprm1. Our findings encourage investigation of the contribution of Taar1 and Oprm1 variants to human methamphetamine addiction.
Collapse
Affiliation(s)
- Alexandra M Stafford
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Cheryl Reed
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Harue Baba
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Nicole AR Walter
- Division of NeuroscienceOregon National Primate Research CenterPortlandUnited States
| | - John RK Mootz
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Robert W Williams
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Sciences CenterMemphisUnited States
| | - Kim A Neve
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
- Veterans Affairs Portland Health Care SystemPortlandUnited States
| | - Lev M Fedorov
- Transgenic Mouse Models Shared Resource, Knight Cancer InstituteOregon Health & Science UniversityPortlandUnited States
| | - Aaron J Janowsky
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
- Veterans Affairs Portland Health Care SystemPortlandUnited States
- Department of PsychiatryOregon Health & Science UniversityPortlandUnited States
| | - Tamara J Phillips
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
- Veterans Affairs Portland Health Care SystemPortlandUnited States
| |
Collapse
|
26
|
Regional Analysis of the Brain Transcriptome in Mice Bred for High and Low Methamphetamine Consumption. Brain Sci 2019; 9:brainsci9070155. [PMID: 31262025 PMCID: PMC6681006 DOI: 10.3390/brainsci9070155] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/20/2019] [Accepted: 06/26/2019] [Indexed: 01/08/2023] Open
Abstract
Transcriptome profiling can broadly characterize drug effects and risk for addiction in the absence of drug exposure. Modern large-scale molecular methods, including RNA-sequencing (RNA-Seq), have been extensively applied to alcohol-related disease traits, but rarely to risk for methamphetamine (MA) addiction. We used RNA-Seq data from selectively bred mice with high or low risk for voluntary MA intake to construct coexpression and cosplicing networks for differential risk. Three brain reward circuitry regions were explored, the nucleus accumbens (NAc), prefrontal cortex (PFC), and ventral midbrain (VMB). With respect to differential gene expression and wiring, the VMB was more strongly affected than either the PFC or NAc. Coexpression network connectivity was higher in the low MA drinking line than in the high MA drinking line in the VMB, oppositely affected in the NAc, and little impacted in the PFC. Gene modules protected from the effects of selection may help to eliminate certain mechanisms from significant involvement in risk for MA intake. One such module was enriched in genes with dopamine-associated annotations. Overall, the data suggest that mitochondrial function and glutamate-mediated synaptic plasticity have key roles in the outcomes of selective breeding for high versus low levels of MA intake.
Collapse
|
27
|
Rodrigues AVSL, Almeida FJ, Vieira-Coelho MA. Dimethyltryptamine: Endogenous Role and Therapeutic Potential. J Psychoactive Drugs 2019; 51:299-310. [DOI: 10.1080/02791072.2019.1602291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alexandra VSL Rodrigues
- Department of Biomedicine-Pharmacology and Therapeutics unit, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Francisco Jcg Almeida
- Department of Biomedicine-Pharmacology and Therapeutics unit, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Maria A Vieira-Coelho
- Department of Biomedicine-Pharmacology and Therapeutics unit, Faculty of Medicine, University of Porto, Porto, Portugal
- Psychiatry and Mental Health Clinic, Hospital de São João, Porto, Portugal
| |
Collapse
|
28
|
Drug addiction: a curable mental disorder? Acta Pharmacol Sin 2018; 39:1823-1829. [PMID: 30382181 DOI: 10.1038/s41401-018-0180-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/29/2018] [Indexed: 02/06/2023] Open
Abstract
Drug addiction is a chronic, relapsing brain disorder. Multiple neural networks in the brain including the reward system (e.g., the mesocorticolimbic system), the anti-reward/stress system (e.g., the extended amygdala), and the central immune system, are involved in the development of drug addiction and relapse after withdrawal from drugs of abuse. Preclinical and clinical studies have demonstrated that it is promising to control drug addiction by pharmacologically targeting the addiction-related systems in the brain. Here we review the pharmacological targets within the dopamine system, glutamate system, trace amine system, anti-reward system, and central immune system, which are of clinical interests. Furthermore, we discuss other potential therapies, e.g., brain stimulation, behavioral treatments, and therapeutic gene modulation, which could be effective to treat drug addiction. We conclude that, although drug addiction is a complex disorder that involves complicated neural mechanisms and psychological processes, this mental disorder is treatable and may be curable by therapies such as gene modulation in the future.
Collapse
|
29
|
Kostrzewa RM, Wydra K, Filip M, Crawford CA, McDougall SA, Brown RW, Borroto-Escuela DO, Fuxe K, Gainetdinov RR. Dopamine D 2 Receptor Supersensitivity as a Spectrum of Neurotoxicity and Status in Psychiatric Disorders. J Pharmacol Exp Ther 2018; 366:519-526. [PMID: 29921706 DOI: 10.1124/jpet.118.247981] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022] Open
Abstract
Abnormality of dopamine D2 receptor (D2R) function, often observed as D2R supersensitivity (D2RSS), is a commonality of schizophrenia and related psychiatric disorders in humans. Moreover, virtually all psychotherapeutic agents for schizophrenia target D2R in brain. Permanent D2RSS as a feature of a new animal model of schizophrenia was first reported in 1991, and then behaviorally and biochemically characterized over the next 15-20 years. In this model of schizophrenia characterized by production of D2RSS in ontogeny, there are demonstrated alterations of signaling processes, as well as functional links between the biologic template of the animal model and ability of pharmacotherapeutics to modulate or reverse biologic and behavioral modalities toward normality. Another such animal model, featuring knockout of trace amine-associated receptor 1 (TAAR1), demonstrates D2RSS with an increase in the proportion of D2R in the high-affinity state. Currently, TAAR1 agonists are being explored as a therapeutic option for schizophrenia. There is likewise an overlay of D2RSS with substance use disorder. The aspect of adenosine A2A-D2 heteroreceptor complexes in substance use disorder is highlighted, and the association of adenosine A2A receptor antagonists in discriminative and rewarding effects of psychostimulants is outlined. In summary, these new animal models of schizophrenia have face, construct, and predictive validity, and distinct advantages over earlier models. While the review summarizes elements of D2RSS in schizophrenia per se, and its interplay with substance use disorder, a major focus is on presumed new molecular targets attending D2RSS in schizophrenia and related clinical entities.
Collapse
Affiliation(s)
- Richard M Kostrzewa
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Karolina Wydra
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Malgorzata Filip
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Cynthia A Crawford
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Sanders A McDougall
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Russell W Brown
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Dasiel O Borroto-Escuela
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Kjell Fuxe
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Raul R Gainetdinov
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| |
Collapse
|