1
|
Liu H, Zheng Y, Wang Y, Wang Y, He X, Xu P, Huang S, Yuan Q, Zhang X, Wang L, Jiang K, Chen H, Li Z, Liu W, Wang S, Xu HE, Xu F. Recognition of methamphetamine and other amines by trace amine receptor TAAR1. Nature 2023; 624:663-671. [PMID: 37935377 DOI: 10.1038/s41586-023-06775-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
Trace amine-associated receptor 1 (TAAR1), the founding member of a nine-member family of trace amine receptors, is responsible for recognizing a range of biogenic amines in the brain, including the endogenous β-phenylethylamine (β-PEA)1 as well as methamphetamine2, an abused substance that has posed a severe threat to human health and society3. Given its unique physiological role in the brain, TAAR1 is also an emerging target for a range of neurological disorders including schizophrenia, depression and drug addiction2,4,5. Here we report structures of human TAAR1-G-protein complexes bound to methamphetamine and β-PEA as well as complexes bound to RO5256390, a TAAR1-selective agonist, and SEP-363856, a clinical-stage dual agonist for TAAR1 and serotonin receptor 5-HT1AR (refs. 6,7). Together with systematic mutagenesis and functional studies, the structures reveal the molecular basis of methamphetamine recognition and underlying mechanisms of ligand selectivity and polypharmacology between TAAR1 and other monoamine receptors. We identify a lid-like extracellular loop 2 helix/loop structure and a hydrogen-bonding network in the ligand-binding pockets, which may contribute to the ligand recognition in TAAR1. These findings shed light on the ligand recognition mode and activation mechanism for TAAR1 and should guide the development of next-generation therapeutics for drug addiction and various neurological disorders.
Collapse
Affiliation(s)
- Heng Liu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - You Zheng
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yue Wang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yumeng Wang
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecule Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xinheng He
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Peiyu Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sijie Huang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qingning Yuan
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinyue Zhang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ling Wang
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kexin Jiang
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hong Chen
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai, China
- Shanghai Yuansi Standard Science and Technology Co., Ltd, Shanghai, China
| | - Zhen Li
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenbin Liu
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai, China.
- Shanghai Yuansi Standard Science and Technology Co., Ltd, Shanghai, China.
| | - Sheng Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecule Cell Science, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - H Eric Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Fei Xu
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
2
|
Zhukov IS, Karpova IV, Krotova NA, Tissen IY, Demin KA, Shabanov PD, Budygin EA, Kalueff AV, Gainetdinov RR. Enhanced Aggression, Reduced Self-Grooming Behavior and Altered 5-HT Regulation in the Frontal Cortex in Mice Lacking Trace Amine-Associated Receptor 1 (TAAR1). Int J Mol Sci 2022; 23:ijms232214066. [PMID: 36430544 PMCID: PMC9695497 DOI: 10.3390/ijms232214066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The Trace Amine-Associated Receptor 1 (TAAR1) is one of the six functional receptors belonging to the family of monoamine-related G protein-coupled receptors (TAAR1-TAAR9) found in humans. However, the exact biological mechanisms of TAAR1 central and peripheral action remain to be fully understood. TAAR1 is widely expressed in the prefrontal cortex and several limbic regions, interplaying with the dopamine system to modulate the reward circuitry. Recent clinical trials suggest the efficacy of TAAR1 agonists as potential novel antipsychotic agents. Here, we characterize behavioral and neurochemical phenotypes of TAAR1 knockout mice, focusing on aggression and self-grooming behavior that both strongly depend on the monoaminergic signaling and cortico-striatal and cortico-limbic circuits. Overall, we report increased aggression in these knockout mice in the resident-intruder test, accompanied by reduced self-grooming behavior in the novelty-induced grooming test, and by higher cortical serotonin (5-HT) tissue levels. Further studies are necessary to explore whether TAAR1-based therapies can become potential novel treatments for a wide range of neuropsychiatric disorders associated with aggression.
Collapse
Affiliation(s)
- Ilya S. Zhukov
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- Institute of Experimental Medicine, Acad. Pavlov str. 12, 197376 St. Petersburg, Russia
| | - Inessa V. Karpova
- Institute of Experimental Medicine, Acad. Pavlov str. 12, 197376 St. Petersburg, Russia
| | - Nataliya A. Krotova
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, 197341 St. Petersburg, Russia
| | - Ilya Y. Tissen
- Institute of Experimental Medicine, Acad. Pavlov str. 12, 197376 St. Petersburg, Russia
| | - Konstantin A. Demin
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, 197341 St. Petersburg, Russia
| | - Petr D. Shabanov
- Institute of Experimental Medicine, Acad. Pavlov str. 12, 197376 St. Petersburg, Russia
| | - Evgeny A. Budygin
- Neurobiology Program, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Allan V. Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, 197341 St. Petersburg, Russia
- Neurobiology Program, Sirius University of Science and Technology, 354340 Sochi, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, 197758 St. Petersburg, Russia
- Neurobiology Laboratory, Ural Federal University, 620002 Yekaterinburg, Russia
- Laboratory of Cell and Molecular Biology and Neurobiology, School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141701 Moscow, Russia
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
- Correspondence:
| |
Collapse
|
3
|
Decker AM, Brackeen MF, Mohammadkhani A, Kormos CM, Hesk D, Borgland SL, Blough BE. Identification of a Potent Human Trace Amine-Associated Receptor 1 Antagonist. ACS Chem Neurosci 2022; 13:1082-1095. [PMID: 35325532 DOI: 10.1021/acschemneuro.2c00086] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human trace amine-associated receptor subtype 1 (hTAAR1) is a G protein-coupled receptor that has therapeutic potential for multiple diseases, including schizophrenia, drug addiction, and Parkinson's disease (PD). Although several potent agonists have been identified and have shown positive results in various clinical trials for schizophrenia, the discovery of potent hTAAR1 antagonists remains elusive. Herein, we report the results of structure-activity relationship studies that have led to the discovery of a potent hTAAR1 antagonist (RTI-7470-44, 34). RTI-7470-44 exhibited an IC50 of 8.4 nM in an in vitro cAMP functional assay, a Ki of 0.3 nM in a radioligand binding assay, and showed species selectivity for hTAAR1 over the rat and mouse orthologues. RTI-7470-44 displayed good blood-brain barrier permeability, moderate metabolic stability, and a favorable preliminary off-target profile. Finally, RTI-7470-44 increased the spontaneous firing rate of mouse VTA dopaminergic neurons and blocked the effects of the known TAAR1 agonist RO5166017. Collectively, this work provides a promising hTAAR1 antagonist probe that can be used to study TAAR1 pharmacology and the potential therapeutic role in hypodopaminergic diseases such as PD.
Collapse
Affiliation(s)
- Ann M. Decker
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - Marcus F. Brackeen
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - Aida Mohammadkhani
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Chad M. Kormos
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - David Hesk
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - Stephanie L. Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Bruce E. Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
4
|
Garey JD, Lusskin SI, Scialli AR. Teratogen update: Amphetamines. Birth Defects Res 2020; 112:1171-1182. [PMID: 32755038 DOI: 10.1002/bdr2.1774] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/19/2020] [Accepted: 07/06/2020] [Indexed: 01/09/2023]
Abstract
Amphetamines are synthetic noncatecholamine sympathomimetic amines that act as psychostimulants. They have been prescribed for the treatment of attention-deficit/hyperactivity disorder (ADHD), narcolepsy, and additional health conditions. Amphetamines are also drugs of abuse. Some experimental animal studies suggested adverse developmental effects of amphetamines, including structural malformations. These effects were most often observed in experimental animals at higher dose levels than those used for treatment or abuse and at dose levels that produce maternal toxicity. Controlled studies of amphetamine use for the treatment of ADHD and other indications did not suggest that amphetamines are likely to cause structural malformations, although there are three studies associating medication for ADHD or methamphetamine abuse with gastroschisis. We did not locate studies on the neurobehavioral effects of prenatal exposures to therapeutic amphetamine use. Amphetamine abuse was associated with offspring neurobehavioral abnormalities, but lack of adequate adjustment for confounding interferes with interpretation of the associations. Adverse effects of methamphetamine abuse during pregnancy may be due to factors associated with drug abuse rather than methamphetamine itself. The adverse effects observed in methamphetamine abuse studies may not be extrapolatable to amphetamine medication use.
Collapse
Affiliation(s)
- Joan D Garey
- Reproductive Toxicology Center, A Non-Profit Foundation, Washington, District of Columbia, USA
| | - Shari I Lusskin
- Reproductive Toxicology Center, A Non-Profit Foundation, Washington, District of Columbia, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Obstetrics, Gynecology, and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anthony R Scialli
- Reproductive Toxicology Center, A Non-Profit Foundation, Washington, District of Columbia, USA
| |
Collapse
|
5
|
Tonelli M, Cichero E. Trace amine associated receptor 1 (TAAR1) modulators: a patent review (2010-present). Expert Opin Ther Pat 2019; 30:137-145. [DOI: 10.1080/13543776.2020.1708900] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Michele Tonelli
- Dipartimento di Farmacia, Università di Genova, Viale Benedetto XV 3, Genova, Italy
| | - Elena Cichero
- Dipartimento di Farmacia, Università di Genova, Viale Benedetto XV 3, Genova, Italy
| |
Collapse
|
6
|
Cheng J, Wang S, Lin W, Wu N, Wang Y, Chen M, Xie XQ, Feng Z. Computational Systems Pharmacology-Target Mapping for Fentanyl-Laced Cocaine Overdose. ACS Chem Neurosci 2019; 10:3486-3499. [PMID: 31257858 DOI: 10.1021/acschemneuro.9b00109] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The United States of America is fighting against one of its worst-ever drug crises. Over 900 people a week die from opioid- or heroin-related overdoses, while millions more suffer from opioid prescription addiction. Recently, drug overdoses caused by fentanyl-laced cocaine specifically are on the rise. Due to drug synergy and an increase in side effects, polydrug addiction can cause more risk than addiction to a single drug. In the present work, we systematically analyzed the overdose and addiction mechanism of cocaine and fentanyl. First, we applied our established chemogenomics knowledgebase and machine-learning-based methods to map out the potential and known proteins, transporters, and metabolic enzymes and the potential therapeutic target(s) for cocaine and fentanyl. Sequentially, we looked into the detail of (1) the addiction to cocaine and fentanyl by binding to the dopamine transporter and the μ opioid receptor (DAT and μOR, respectively), (2) the potential drug-drug interaction of cocaine and fentanyl via p-glycoprotein (P-gp) efflux, (3) the metabolism of cocaine and fentanyl in CYP3A4, and (4) the physiologically based pharmacokinetic (PBPK) model for two drugs and their drug-drug interaction at the absorption, distribution, metabolism, and excretion (ADME) level. Finally, we looked into the detail of JWH133, an agonist of cannabinoid 2-receptor (CB2) with potential as a therapy for cocaine and fentanyl overdose. All these results provide a better understanding of fentanyl and cocaine polydrug addiction and future drug abuse prevention.
Collapse
Affiliation(s)
- Jin Cheng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu 224005, China
| | - Siyi Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Weiwei Lin
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Nan Wu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Yuanqiang Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Maozi Chen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
7
|
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
|