1
|
Shajan B, Bastiampillai T, Hellyer SD, Nair PC. Unlocking the secrets of trace amine-associated receptor 1 agonists: new horizon in neuropsychiatric treatment. Front Psychiatry 2024; 15:1464550. [PMID: 39553890 PMCID: PMC11565220 DOI: 10.3389/fpsyt.2024.1464550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/07/2024] [Indexed: 11/19/2024] Open
Abstract
For over seven decades, dopamine receptor 2 (D2 receptor) antagonists remained the mainstay treatment for neuropsychiatric disorders. Although it is effective for treating hyperdopaminergic symptoms, it is often ineffective for treating negative and cognitive deficits. Trace amine-associated receptor 1 (TAAR1) is a novel, pharmacological target in the treatment of schizophrenia and other neuropsychiatric conditions. Several TAAR1 agonists are currently being developed and are in various stages of clinical and preclinical development. Previous efforts to identify TAAR1 agonists have been hampered by challenges in pharmacological characterisation, the absence of experimentally determined structures, and species-specific preferences in ligand binding and recognition. Further, poor insights into the functional selectivity of the receptor led to the characterisation of ligands with analogous signalling mechanisms. Such approaches limited the understanding of divergent receptor signalling and their potential clinical utility. Recent cryogenic electron microscopic (cryo-EM) structures of human and mouse TAAR1 (hTAAR1 and mTAAR1, respectively) in complex with agonists and G proteins have revealed detailed atomic insights into the binding pockets, binding interactions and binding modes of several agonists including endogenous trace amines (β-phenylethylamine, 3-Iodothyronamine), psychostimulants (amphetamine, methamphetamine), clinical compounds (ulotaront, ralmitaront) and repurposed drugs (fenoldopam). The in vitro screening of drug libraries has also led to the discovery of novel TAAR1 agonists (asenapine, guanabenz, guanfacine) which can be used in clinical trials or further developed to treat different neuropsychiatric conditions. Furthermore, an understanding of unappreciated signalling mechanisms (Gq, Gs/Gq) by TAAR1 agonists has come to light with the discovery of selective compounds to treat schizophrenia-like phenotypes. In this review, we discuss the emergence of structure-based approaches in the discovery of novel TAAR1 agonists through drug repurposing strategies and structure-guided designs. Additionally, we discuss the functional selectivity of TAAR1 signalling, which provides important clues for developing disorder-specific compounds.
Collapse
Affiliation(s)
- Britto Shajan
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Tarun Bastiampillai
- Department of Psychiatry, Monash University, Parkville, Melbourne, VIC, Australia
- Flinders Health and Medical Research Institute (FHMRI) College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Shane D. Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Pramod C. Nair
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Flinders Health and Medical Research Institute (FHMRI) College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, SA, Australia
- Discipline of Medicine, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
2
|
Faquih T, Potts K, Yu B, Kaplan R, Isasi CR, Qi Q, Taylor KD, Liu PY, Tracy RP, Johnson C, Rich SS, Clish CB, Gerzsten RE, Rotter JI, Redline S, Sofer T, Wang H. Steroid Hormone Biosynthesis and Dietary Related Metabolites Associated with Excessive Daytime Sleepiness. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.12.24313561. [PMID: 39314973 PMCID: PMC11419218 DOI: 10.1101/2024.09.12.24313561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background Excessive daytime sleepiness (EDS) is a complex sleep problem that affects approximately 33% of the United States population. Although EDS usually occurs in conjunction with insufficient sleep, and other sleep and circadian disorders, recent studies have shown unique genetic markers and metabolic pathways underlying EDS. Here, we aimed to further elucidate the biological profile of EDS using large scale single- and pathway-level metabolomics analyses. Methods Metabolomics data were available for 877 metabolites in 6,071 individuals from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL) and EDS was assessed using the Epworth Sleepiness Scale (ESS) questionnaire. We performed linear regression for each metabolite on continuous ESS, adjusting for demographic, lifestyle, and physiological confounders, and in sex specific groups. Subsequently, gaussian graphical modelling was performed coupled with pathway and enrichment analyses to generate a holistic interactive network of the metabolomic profile of EDS associations. Findings We identified seven metabolites belonging to steroids, sphingomyelin, and long chain fatty acids sub-pathways in the primary model associated with EDS, and an additional three metabolites in the male-specific analysis. The identified metabolites particularly played a role in steroid hormone biosynthesis. Interpretation Our findings indicate that an EDS metabolomic profile is characterized by endogenous and dietary metabolites within the steroid hormone biosynthesis pathway, with some pathways that differ by sex. Our findings identify potential pathways to target for addressing the causes or consequences of EDS and related sleep disorders. Funding Details regarding funding supporting this work and all studies involved are provided in the acknowledgments section.
Collapse
Affiliation(s)
- Tariq Faquih
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- CardioVascular Institute (CVI), Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
| | - Kaitlin Potts
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
| | - Bing Yu
- Department of Epidemiology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Carmen R Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Qibin Qi
- Department of Epidemiology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kent D Taylor
- Department of Paediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Centre, Torrance, CA, USA
| | - Peter Y Liu
- Department of Paediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Centre, Torrance, CA, USA
| | - Russell P Tracy
- Department of Biochemistry, University of Vermont, Burlington, VT, USA
| | - Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, USA
| | - Stephen S Rich
- Centre for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Clary B Clish
- Metabolite Profiling Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert E Gerzsten
- Broad Institute, Cambridge, MA, USA
- CardioVascular Institute (CVI), Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jerome I Rotter
- Department of Paediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Centre, Torrance, CA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- CardioVascular Institute (CVI), Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Shemiakova TS, Efimova EV, Gainetdinov RR. TAARs as Novel Therapeutic Targets for the Treatment of Depression: A Narrative Review of the Interconnection with Monoamines and Adult Neurogenesis. Biomedicines 2024; 12:1263. [PMID: 38927470 PMCID: PMC11200894 DOI: 10.3390/biomedicines12061263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Depression is a common mental illness of great concern. Current therapy for depression is only suitable for 80% of patients and is often associated with unwanted side effects. In this regard, the search for and development of new antidepressant agents remains an urgent task. In this review, we discuss the current available evidence indicating that G protein-coupled trace amine-associated receptors (TAARs) might represent new targets for depression treatment. The most frequently studied receptor TAAR1 has already been investigated in the treatment of schizophrenia, demonstrating antidepressant and anxiolytic properties. In fact, the TAAR1 agonist Ulotaront is currently undergoing phase 2/3 clinical trials testing its safety and efficacy in the treatment of major depressive disorder and generalized anxiety disorder. Other members of the TAAR family (TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) are not only involved in the innate olfaction of volatile amines, but are also expressed in the limbic brain areas. Furthermore, animal studies have shown that TAAR2 and TAAR5 regulate emotional behaviors and thus may hold promise as potential antidepressant targets. Of particular interest is their connection with the dopamine and serotonin systems of the brain and their involvement in the regulation of adult neurogenesis, known to be affected by the antidepressant drugs currently in use. Further non-clinical and clinical studies are necessary to validate TAAR1 (and potentially other TAARs) as novel therapeutic targets for the treatment of depression.
Collapse
Affiliation(s)
- Taisiia S. Shemiakova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia; (T.S.S.); (E.V.E.)
| | - Evgeniya V. Efimova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia; (T.S.S.); (E.V.E.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia; (T.S.S.); (E.V.E.)
- Saint-Petersburg University Hospital, Saint-Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
4
|
Osaka H, Kanazawa T. Emerging trends in antipsychotic and antidepressant drug development: Targeting nonmonoamine receptors and innovative mechanisms. PCN REPORTS : PSYCHIATRY AND CLINICAL NEUROSCIENCES 2023; 2:e157. [PMID: 38868733 PMCID: PMC11114387 DOI: 10.1002/pcn5.157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/26/2023] [Accepted: 10/30/2023] [Indexed: 06/14/2024]
Abstract
The domain of psychiatric drug development is currently witnessing a notable transformation, with a paramount emphasis on targeting nonmonoamine receptors and exploring inventive mechanisms of action. This paper presents an overview of the ongoing advancements in antipsychotic and antidepressant drug development. Historically, antipsychotics predominantly targeted dopamine receptors, but there is now an escalating interest in drugs that act on alternative receptors, exemplified by the TAAR1 receptor. One noteworthy candidate is Ulotaront (SEP-363856), an agent acting as a TAAR1 agonist with 5-HT1A agonist activity, demonstrating promising outcomes in the treatment of schizophrenia, devoid of extrapyramidal symptoms or metabolic side-effects. Similarly, MIN-101 (Roluperidone) and KarXT are currently in development, with its focus on addressing the symptoms in schizophrenia. In the domain of antidepressants, novel therapeutic approaches have surfaced, such as Auvelity, a Food and Drug Administration (FDA)-approved NMDA receptor antagonist synergistically combined with Bupropion to enhance its effects. Another notable candidate is Zuranolone, operating as a GABA A receptor-positive allosteric modulator, showcasing efficacy in treating major depressive disorder (MDD) and postpartum depression. Additionally, TAK-653 (NBI-1065845) and MJI821 (Onfasprodil) have emerged as potential antidepressants targeting AMPA receptors and NMDA receptor 2B (NR2B) negative allosteric modulation, respectively. This paper underscores the transformative potential of these novel drug candidates in psychiatric treatment and their ability to address cases that were previously treatment-resistant. By focusing on nonmonoamine receptors and introducing innovative mechanisms, these drugs offer a promising prospect of improved outcomes for individuals suffering from schizophrenia and MDD. Thus, sustained attention and dedication to the development of such drugs are essential to augmenting the therapeutic options available for psychiatric patients.
Collapse
Affiliation(s)
- Hitoshi Osaka
- Department of NeuropsychiatryOsaka Medical and Pharmaceutical UniversityTakatsuki‐cityOsakaJapan
| | - Tetsufumi Kanazawa
- Department of NeuropsychiatryOsaka Medical and Pharmaceutical UniversityTakatsuki‐cityOsakaJapan
| |
Collapse
|
5
|
Shang P, Rong N, Jiang JJ, Cheng J, Zhang MH, Kang D, Qi L, Guo L, Yang GM, Liu Q, Zhou Z, Li XB, Zhu KK, Meng QB, Han X, Yan W, Kong Y, Yang L, Wang X, Lei D, Feng X, Liu X, Yu X, Wang Y, Li Q, Shao ZH, Yang F, Sun JP. Structural and signaling mechanisms of TAAR1 enabled preferential agonist design. Cell 2023; 186:5347-5362.e24. [PMID: 37963465 DOI: 10.1016/j.cell.2023.10.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/09/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023]
Abstract
Trace amine-associated receptor 1 (TAAR1) senses a spectrum of endogenous amine-containing metabolites (EAMs) to mediate diverse psychological functions and is useful for schizophrenia treatment without the side effects of catalepsy. Here, we systematically profiled the signaling properties of TAAR1 activation and present nine structures of TAAR1-Gs/Gq in complex with EAMs, clinical drugs, and synthetic compounds. These structures not only revealed the primary amine recognition pocket (PARP) harboring the conserved acidic D3.32 for conserved amine recognition and "twin" toggle switch for receptor activation but also elucidated that targeting specific residues in the second binding pocket (SBP) allowed modulation of signaling preference. In addition to traditional drug-induced Gs signaling, Gq activation by EAM or synthetic compounds is beneficial to schizophrenia treatment. Our results provided a structural and signaling framework for molecular recognition by TAAR1, which afforded structural templates and signal clues for TAAR1-targeted candidate compounds design.
Collapse
Affiliation(s)
- Pan Shang
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China
| | - Naikang Rong
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China
| | - Jing-Jing Jiang
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Jie Cheng
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China
| | - Ming-Hui Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, Shandong 250012, China
| | - Lei Qi
- Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China; Biomedical Research Center for Structural Analysis, Shandong University, No.44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Lulu Guo
- Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China
| | - Gong-Ming Yang
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Qun Liu
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China
| | - Zhenzhen Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, Shandong 250012, China
| | - Xiao-Bing Li
- Medical Science and Technology Innovation Center, Shandong Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Kong-Kai Zhu
- Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China
| | - Qing-Biao Meng
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xiang Han
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Wenqi Yan
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yalei Kong
- Songjiang Institute and Shanghai Songjiang District Central Hospital, Center for Brain Science in Shanghai Children's Medical Center, Department of Anatomy and Physiology, Ministry of Education, Shanghai Key Laboratory of Children's Environmental Health in Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lejin Yang
- Department of Psychology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Dapeng Lei
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xin Feng
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan, Shandong 250012, China
| | - Xiao Yu
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yue Wang
- Medical Science and Technology Innovation Center, Shandong Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| | - Qian Li
- Songjiang Institute and Shanghai Songjiang District Central Hospital, Center for Brain Science in Shanghai Children's Medical Center, Department of Anatomy and Physiology, Ministry of Education, Shanghai Key Laboratory of Children's Environmental Health in Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhen-Hua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Fan Yang
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China.
| | - Jin-Peng Sun
- NHC Key Laboratory of Otorhinolaryngology, Qilu hospital and School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China; Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
6
|
Vaganova AN, Shemyakova TS, Lenskaia KV, Rodionov RN, Steenblock C, Gainetdinov RR. Trace Amine-Associated Receptors and Monoamine-Mediated Regulation of Insulin Secretion in Pancreatic Islets. Biomolecules 2023; 13:1618. [PMID: 38002300 PMCID: PMC10669413 DOI: 10.3390/biom13111618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Currently, metabolic syndrome treatment includes predominantly pharmacological symptom relief and complex lifestyle changes. Trace amines and their receptor systems modulate signaling pathways of dopamine, norepinephrine, and serotonin, which are involved in the pathogenesis of this disorder. Trace amine-associated receptor 1 (TAAR1) is expressed in endocrine organs, and it was revealed that TAAR1 may regulate insulin secretion in pancreatic islet β-cells. For instance, accumulating data demonstrate the positive effect of TAAR1 agonists on the dynamics of metabolic syndrome progression and MetS-associated disease development. The role of other TAARs (TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) in the islet's function is much less studied. In this review, we summarize the evidence of TAARs' contribution to the metabolic syndrome pathogenesis and regulation of insulin secretion in pancreatic islets. Additionally, by the analysis of public transcriptomic data, we demonstrate that TAAR1 and other TAAR receptors are expressed in the pancreatic islets. We also explore associations between the expression of TAARs mRNA and other genes in studied samples and demonstrate the deregulation of TAARs' functional associations in patients with metabolic diseases compared to healthy donors.
Collapse
Affiliation(s)
- Anastasia N. Vaganova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.N.V.); (T.S.S.)
- St. Petersburg State University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Taisiia S. Shemyakova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.N.V.); (T.S.S.)
| | - Karina V. Lenskaia
- Department of Medicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Roman N. Rodionov
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (R.N.R.); (C.S.)
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (R.N.R.); (C.S.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.N.V.); (T.S.S.)
- St. Petersburg State University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
7
|
Shabani S, Houlton S, Ghimire B, Tonello D, Reed C, Baba H, Aldrich S, Phillips TJ. Robust aversive effects of trace amine-associated receptor 1 activation in mice. Neuropsychopharmacology 2023; 48:1446-1454. [PMID: 37055488 PMCID: PMC10425385 DOI: 10.1038/s41386-023-01578-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/03/2023] [Accepted: 03/26/2023] [Indexed: 04/15/2023]
Abstract
Drugs that stimulate the trace amine-associated receptor 1 (TAAR1) are under clinical investigation as treatments for several neuropsychiatric disorders. Previous studies in a genetic mouse model of voluntary methamphetamine intake identified TAAR1, expressed by the Taar1 gene, as a critical mediator of aversive methamphetamine effects. Methamphetamine is a TAAR1 agonist, but also has actions at monoamine transporters. Whether exclusive activation of TAAR1 has aversive effects was not known at the time we conducted our studies. Mice were tested for aversive effects of the selective TAAR1 agonist, RO5256390, using taste and place conditioning procedures. Hypothermic and locomotor effects were also examined, based on prior evidence of TAAR1 mediation. Male and female mice of several genetic models were used, including lines selectively bred for high and low methamphetamine drinking, a knock-in line in which a mutant form of Taar1 that codes for a non-functional TAAR1 was replaced by the reference Taar1 allele that codes for functional TAAR1, and their matched control line. RO5256390 had robust aversive, hypothermic and locomotor suppressing effects that were found only in mice with functional TAAR1. Knock-in of the reference Taar1 allele rescued these phenotypes in a genetic model that normally lacks TAAR1 function. Our study provides important data on TAAR1 function in aversive, locomotor, and thermoregulatory effects that are important to consider when developing TAAR1 agonists as therapeutic drugs. Because other drugs can have similar consequences, potential additive effects should be carefully considered as these treatment agents are being developed.
Collapse
Affiliation(s)
- Shkelzen Shabani
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
- Department of Biology, Minot State University, Minot, ND, USA
- Biomedical Sciences at Grand Valley State University, Allendale, MI, USA
| | - Sydney Houlton
- Department of Biology, Minot State University, Minot, ND, USA
| | - Bikalpa Ghimire
- Department of Biology, Minot State University, Minot, ND, USA
| | - Derek Tonello
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
| | - Cheryl Reed
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Harue Baba
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Sara Aldrich
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Tamara J Phillips
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA.
- VA Portland Health Care System, Portland, OR, USA.
| |
Collapse
|
8
|
Polini B, Ricardi C, Bertolini A, Carnicelli V, Rutigliano G, Saponaro F, Zucchi R, Chiellini G. T1AM/TAAR1 System Reduces Inflammatory Response and β-Amyloid Toxicity in Human Microglial HMC3 Cell Line. Int J Mol Sci 2023; 24:11569. [PMID: 37511328 PMCID: PMC10380917 DOI: 10.3390/ijms241411569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Microglial dysfunction is one of the hallmarks and leading causes of common neurodegenerative diseases (NDDs), including Alzheimer's disease (AD) and Parkinson's disease (PD). All these pathologies are characterized by aberrant aggregation of disease-causing proteins in the brain, which can directly activate microglia, trigger microglia-mediated neuroinflammation, and increase oxidative stress. Inhibition of glial activation may represent a therapeutic target to alleviate neurodegeneration. Recently, 3-iodothyronamine (T1AM), an endogenous derivative of thyroid hormone (TH) able to interact directly with a specific GPCR known as trace amine-associated receptor 1 (TAAR1), gained interest for its ability to promote neuroprotection in several models. Nevertheless, T1AM's effects on microglial disfunction remain still elusive. In the present work we investigated whether T1AM could inhibit the inflammatory response of human HMC3 microglial cells to LPS/TNFα or β-amyloid peptide 25-35 (Aβ25-35) stimuli. The results of ELISA and qPCR assays revealed that T1AM was able to reduce microglia-mediated inflammatory response by inhibiting the release of proinflammatory factors, including IL-6, TNFα, NF-kB, MCP1, and MIP1, while promoting the release of anti-inflammatory mediators, such as IL-10. Notably, T1AM anti-inflammatory action in HMC3 cells turned out to be a TAAR1-mediated response, further increasing the relevance of the T1AM/TAAR1 system in the management of NDDs.
Collapse
Affiliation(s)
- Beatrice Polini
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Caterina Ricardi
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Andrea Bertolini
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Vittoria Carnicelli
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Grazia Rutigliano
- Institute of Clinical Sciences, Imperial College London, London SW7 2AZ, UK;
| | - Federica Saponaro
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Riccardo Zucchi
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Grazia Chiellini
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| |
Collapse
|
9
|
Kuvarzin SR, Sukhanov I, Onokhin K, Zakharov K, Gainetdinov RR. Unlocking the Therapeutic Potential of Ulotaront as a Trace Amine-Associated Receptor 1 Agonist for Neuropsychiatric Disorders. Biomedicines 2023; 11:1977. [PMID: 37509616 PMCID: PMC10377193 DOI: 10.3390/biomedicines11071977] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
All antipsychotics currently used in clinic block D2 dopamine receptors. Trace amine-associated receptor 1 is emerging as a new therapeutic target for schizophrenia and several other neuropsychiatric disorders. SEP-363856 (International Nonproprietary Name: Ulotaront) is an investigational antipsychotic drug with a novel mechanism of action that does not involve antagonism of dopamine D2 receptors. Ulotaront is an agonist of trace amine-associated receptor 1 and serotonin 5-HT1A receptors, but can modulate dopamine neurotransmission indirectly. In 2019, the United States Food and Drug Administration granted Breakthrough Therapy Designation for ulotaront for the treatment of schizophrenia. Phase 2 clinical studies indicated that ulotaront can reduce both positive and negative symptoms of schizophrenia without causing the extrapyramidal or metabolic side effects that are inherent to most currently used antipsychotics. At present, it is in phase 3 clinical development for the treatment of schizophrenia and is expected to be introduced into clinical practice in 2023-2024. Clinical studies evaluating the potential efficacy of ulotaront in Parkinson's disease psychosis, generalized anxiety disorder, and major depressive disorder have also been started. The aim of this scoping review is to summarize all currently available preclinical and clinical evidence on the utility of ulotaront in the treatment of schizophrenia. Here, we show the main characteristics and distinctive features of this drug. Perspectives and limitations on the potential use of ulotaront in the pharmacotherapy of several other neuropsychiatric disorders are also discussed.
Collapse
Affiliation(s)
- Savelii R Kuvarzin
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| | - Ilya Sukhanov
- Valdman Institute of Pharmacology, Pavlov Medical University, 197022 Saint Petersburg, Russia
| | - Kirill Onokhin
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 Saint Petersburg, Russia
- Accellena Research and Development Inc., 199106 Saint Petersburg, Russia
| | | | - Raul R Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 Saint Petersburg, Russia
- Saint Petersburg University Hospital, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| |
Collapse
|
10
|
de Bartolomeis A, Ciccarelli M, De Simone G, Mazza B, Barone A, Vellucci L. Canonical and Non-Canonical Antipsychotics' Dopamine-Related Mechanisms of Present and Next Generation Molecules: A Systematic Review on Translational Highlights for Treatment Response and Treatment-Resistant Schizophrenia. Int J Mol Sci 2023; 24:ijms24065945. [PMID: 36983018 PMCID: PMC10051989 DOI: 10.3390/ijms24065945] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Schizophrenia is a severe psychiatric illness affecting almost 25 million people worldwide and is conceptualized as a disorder of synaptic plasticity and brain connectivity. Antipsychotics are the primary pharmacological treatment after more than sixty years after their introduction in therapy. Two findings hold true for all presently available antipsychotics. First, all antipsychotics occupy the dopamine D2 receptor (D2R) as an antagonist or partial agonist, even if with different affinity; second, D2R occupancy is the necessary and probably the sufficient mechanism for antipsychotic effect despite the complexity of antipsychotics' receptor profile. D2R occupancy is followed by coincident or divergent intracellular mechanisms, implying the contribution of cAMP regulation, β-arrestin recruitment, and phospholipase A activation, to quote some of the mechanisms considered canonical. However, in recent years, novel mechanisms related to dopamine function beyond or together with D2R occupancy have emerged. Among these potentially non-canonical mechanisms, the role of Na2+ channels at the dopamine at the presynaptic site, dopamine transporter (DAT) involvement as the main regulator of dopamine concentration at synaptic clefts, and the putative role of antipsychotics as chaperones for intracellular D2R sequestration, should be included. These mechanisms expand the fundamental role of dopamine in schizophrenia therapy and may have relevance to considering putatively new strategies for treatment-resistant schizophrenia (TRS), an extremely severe condition epidemiologically relevant and affecting almost 30% of schizophrenia patients. Here, we performed a critical evaluation of the role of antipsychotics in synaptic plasticity, focusing on their canonical and non-canonical mechanisms of action relevant to the treatment of schizophrenia and their subsequent implication for the pathophysiology and potential therapy of TRS.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Mariateresa Ciccarelli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Benedetta Mazza
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
11
|
Chatterjee I, Chatterjee S. Investigating the symptomatic and morphological changes in the brain based on pre and post-treatment: A critical review from clinical to neuroimaging studies on schizophrenia. IBRO Neurosci Rep 2023. [DOI: 10.1016/j.ibneur.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
|
12
|
de Bartolomeis A, Ciccarelli M, Vellucci L, Fornaro M, Iasevoli F, Barone A. Update on novel antipsychotics and pharmacological strategies for treatment resistant schizophrenia. Expert Opin Pharmacother 2022; 23:2035-2052. [DOI: 10.1080/14656566.2022.2145884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Dentistry, University of Naples “Federico II”, Naples, Italy
| | - Mariateresa Ciccarelli
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Dentistry, University of Naples “Federico II”, Naples, Italy
| | - Licia Vellucci
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Dentistry, University of Naples “Federico II”, Naples, Italy
| | - Michele Fornaro
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Dentistry, University of Naples “Federico II”, Naples, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Dentistry, University of Naples “Federico II”, Naples, Italy
| | - Annarita Barone
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Dentistry, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
13
|
Improved cognitive performance in trace amine-associated receptor 5 (TAAR5) knock-out mice. Sci Rep 2022; 12:14708. [PMID: 36038766 PMCID: PMC9424310 DOI: 10.1038/s41598-022-18924-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Trace amine-associated receptors (TAARs) are a family of G protein-coupled receptors present in mammals in the brain and several peripheral organs. Apart from its olfactory role, TAAR5 is expressed in the major limbic brain areas and regulates brain serotonin functions and emotional behaviours. However, most of its functions remain undiscovered. Given the role of serotonin and limbic regions in some aspects of cognition, we used a temporal decision-making task to unveil a possible role of TAAR5 in cognitive processes. We found that TAAR5 knock-out mice showed a generally better performance due to a reduced number of errors and displayed a greater rate of improvement at the task than WT littermates. However, task-related parameters, such as time accuracy and uncertainty have not changed significantly. Overall, we show that TAAR5 modulates specific domains of cognition, highlighting a new role in brain physiology.
Collapse
|
14
|
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: 3.3] [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
|
15
|
TAAR1 regulates drug-induced reinstatement of cocaine-seeking via negatively modulating CaMKIIα activity in the NAc. Mol Psychiatry 2022; 27:2136-2145. [PMID: 35079125 PMCID: PMC9829124 DOI: 10.1038/s41380-022-01448-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 01/12/2023]
Abstract
Relapse remains a major challenge to the treatment of cocaine addiction. Recent studies suggested that the trace amine-associated receptor 1 (TAAR1) could be a promising target to treat cocaine addiction and relapse; however, the underlying mechanism remains unclear. Here, we aimed to investigate the neural mechanism underlying the role of TAAR1 in the drug priming-induced reinstatement of cocaine-seeking behavior in rats, an animal model of cocaine relapse. We focused on the shell subregion of nucleus accumbens (NAc), a key brain region of the brain reward system. We found that activation of TAAR1 by systemic and intra-NAc shell administration of the selective TAAR1 agonist RO5166017 attenuated drug-induced reinstatement of cocaine-seeking and prevented drug priming-induced CaMKIIα activity in the NAc shell. Activation of TAAR1 dampened the CaMKIIα/GluR1 signaling pathway in the NAc shell and reduced AMPAR-EPSCs on the NAc slice. Microinjection of the selective TAAR1 antagonist EPPTB into the NAc shell enhanced drug-induced reinstatement as well as potentiated CaMKIIα activity in the NAc shell. Furthermore, viral-mediated expression of CaMKIIα in the NAc shell prevented the behavioral effects of TAAR1 activation. Taken together, our findings indicate that TAAR1 regulates drug-induced reinstatement of cocaine-seeking by negatively regulating CaMKIIα activity in the NAc. Our findings elucidate a novel mechanism of TAAR1 in regulating drug-induced reinstatement of cocaine-seeking and further suggests that TAAR1 is a promising target for the treatment of cocaine relapse.
Collapse
|
16
|
Tonelli M. One step away from the breakthrough of TAAR1 agonists for the treatment of neuropsychiatric disorders. Curr Med Chem 2022; 29:4893-4895. [PMID: 35170404 DOI: 10.2174/0929867329666220216111512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 12/31/2021] [Accepted: 01/11/2022] [Indexed: 11/22/2022]
Affiliation(s)
- Michele Tonelli
- Department of Pharmacy, University of Genoa, Viale Benedetto XV n. 3, 16132, Genoa, Italy
| |
Collapse
|
17
|
Jarończyk M, Walory J. Novel Molecular Targets of Antidepressants. Molecules 2022; 27:533. [PMID: 35056845 PMCID: PMC8778443 DOI: 10.3390/molecules27020533] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Antidepressants target a variety of proteins in the central nervous system (CNS), the most important belonging to the family of G-protein coupled receptors and the family of neurotransmitter transporters. The increasing number of crystallographic structures of these proteins have significantly contributed to the knowledge of their mechanism of action, as well as to the design of new drugs. Several computational approaches such as molecular docking, molecular dynamics, and virtual screening are useful for elucidating the mechanism of drug action and are important for drug design. This review is a survey of molecular targets for antidepressants in the CNS and computer based strategies to discover novel compounds with antidepressant activity.
Collapse
|
18
|
Wu R, Liu J, Johnson B, Huang Y, Zhang Y, Li JX. Activation of trace amine-associated receptor 1 attenuates nicotine withdrawal-related effects. Addict Biol 2022; 27:e13075. [PMID: 34170054 PMCID: PMC8709869 DOI: 10.1111/adb.13075] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 01/03/2023]
Abstract
Nicotine addiction is a leading avoidable brain disorder globally. Although nicotine induces a modest reinforcing effect, which is important for the initial drug use, the transition from nicotine use to nicotine addiction involves the mechanisms responsible for the negative consequences of drug abstinence. Recent study suggested that trace amine-associated receptor 1 (TAAR1) is a promising pharmacological target for the modulation of positive reinforcing effects of nicotine. However, whether TAAR1 plays a part in the negative reinforcement of nicotine withdrawal remains to be determined. Here, using a long-access (LA) self-administration model, we investigated whether LA rats show increased nicotine intake and withdrawal symptoms in comparison with saline and ShA rats and then tested the effect of TAAR1 partial agonist RO5263397 on nicotine withdrawal effects. We found that rats from long-access group showed significant abstinence-induced anxiety-like behaviour, mechanic hypersensitivity, increased number of precipitated withdrawal signs and higher motivation for the drug, while rats from short-access did not differ from saline group. TAAR1 partial agonist RO5263397 significantly reduced the physical and motivational withdrawal effects of nicotine in LA rats, as reflected by increased time spent on the open arm in the elevated plus maze (EPM) test, normalized paw withdrawal threshold, decreased withdrawal signs and motivation to self-administer nicotine. This study indicates that activation of TAAR1 attenuates the negative-reinforcing effects of nicotine withdrawal and further suggests TAAR1 as a promising target to treat nicotine addiction.
Collapse
Affiliation(s)
- Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
- School of Medicine, Yangzhou University, Yangzhou, China
| | - 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
| | - Yufei Huang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - 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
|
19
|
Abstract
PURPOSE OF REVIEW Over the last ten years, the treatment of psychosis has seen a near explosion of creative development in both novel agents and new delivery modalities. The current review summarizes these developments over the past decade (2011-2020). We performed a systematic review utilizing PubMed and PsychInfo with the aim of identifying all the RCT and related analyses in adults with psychosis (schizophrenia and mania). RECENT FINDINGS We identified 11 significant developments: the introduction of new antipsychotics cariprazine, brexpiprazole, lumateperone, and pimavanserin; introduction of new delivery methods: subcutaneous long-acting risperidone, aripiprazole lauroxil, transdermal asenapine, and inhaled loxapine; and the introduction of new approaches such as olanzapine/samidorphan for olanzapine-associated weight gain, examination of the TAAR1 agonist SEP 363,856 as a test of concept, and the combination of Xanomeline/Trospium, an M1 and M4 muscarinic receptor agonist in conjunction with a peripheral anticholinergic. Last decade has seen a tremendous development in second-generation antipsychotics which provides unprecedented treatment options for clinicians in treating psychosis.
Collapse
|
20
|
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: 1.3] [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
|
21
|
Trace Amine-Associated Receptor 1 as a Target for the Development of New Antipsychotics: Current Status of Research and Future Directions. CNS Drugs 2021; 35:1153-1161. [PMID: 34655036 DOI: 10.1007/s40263-021-00864-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Schizophrenia is a mental illness associated with an array of symptoms that often result in disability. The primary treatments for schizophrenia are termed antipsychotics. Although antipsychotics modulate a number of different receptor types and subtypes, all currently regulatory agency-approved antipsychotics share in common direct or functional antagonism at the dopamine type 2 receptor (D2R). The majority of people with schizophrenia do not achieve full resolution of their symptoms with antipsychotics, suggesting the need for alternative or complementary approaches. The primary focus of this review is to assess the evidence for the role of the trace amine-associated receptor 1 (TAAR-1) in schizophrenia and the role of TAAR-1 modulators as novel-mechanism antipsychotics. Topics include an overview of TAAR-1 physiology and pathophysiology in schizophrenia, interaction with other neurotransmitter systems, including the dopaminergic, glutamatergic and serotonergic system, and finally, a review of investigational TAAR-1 compounds that have reached Phase II clinical studies in schizophrenia: SEP-363856 (ulotaront) and RO6889450 (ralmitaront). Thus far, results are publicly available only for ulotaront in a relatively young (18-40 years) and acutely exacerbated cohort. These results showed positive effects for overall schizophrenia symptoms without significant tolerability concerns. An ongoing study of ralmitaront will assess specific efficacy in patients with persistent negative symptoms. If trials of TAAR-1 modulators, and other novel-mechanism targets for schizophrenia that are under active study, continue to show positive results, the definition of an antipsychotic may need to be expanded beyond the D2R target in the near future.
Collapse
|
22
|
Galluppi GR, Polhamus DG, Fisher JM, Hopkins SC, Koblan KS. Population pharmacokinetic analysis of ulotaront in subjects with schizophrenia. CPT Pharmacometrics Syst Pharmacol 2021; 10:1245-1254. [PMID: 34292664 PMCID: PMC8520744 DOI: 10.1002/psp4.12692] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/23/2021] [Accepted: 07/06/2021] [Indexed: 01/02/2023] Open
Abstract
Ulotaront (SEP-363856) is a trace amine-associated receptor 1 agonist with 5-HT1A agonist activity in phase III development for the treatment of schizophrenia. The efficacy of ulotaront is not mediated by blockade of D2 or 5-HT2A receptors. The aim of this study was to evaluate the population pharmacokinetics (PopPKs) of ulotaront in adult subjects using pooled data from seven phase I studies, one phase II acute study, and one 6-month extension study. Single and multiple (up to 7 days) oral doses (5-150 mg/day) were studied in both healthy adult subjects (with intensive serial plasma sampling) and adult patients with schizophrenia (some with intensive and some with sparse plasma sampling). Ulotaront was well-absorbed and exhibited dose-proportionality in doses ranging from 10 to 100 mg, in mean maximum concentration, area under the concentration-time curve, and minimum concentration. Moderate interindividual variability was observed in concentration-time profiles. The estimated median time to maximal concentration was 2.8 h and the median effective half-life was 7 h, corresponding to an exposure accumulation ratio of 1.10 at steady-state with daily dosing. There was no indication of time-dependent changes in PKs after up to 12 weeks of daily dose administration. No clinically meaningful effects on ulotaront PK parameters were observed based on race, age, sex, formulation (capsule or tablet), or clinical status (healthy volunteer vs. patient with schizophrenia); body weight was the only meaningful covariate.
Collapse
|
23
|
Nataf S, Pays L. Molecular Insights into SARS-CoV2-Induced Alterations of the Gut/Brain Axis. Int J Mol Sci 2021; 22:10440. [PMID: 34638785 PMCID: PMC8508788 DOI: 10.3390/ijms221910440] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 12/16/2022] Open
Abstract
For a yet unknown reason, a substantial share of patients suffering from COVID-19 develop long-lasting neuropsychiatric symptoms ranging from cognitive deficits to mood disorders and/or an extreme fatigue. We previously reported that in non-neural cells, angiotensin-1 converting enzyme 2 (ACE2), the gene coding for the SARS-CoV2 host receptor, harbors tight co-expression links with dopa-decarboxylase (DDC), an enzyme involved in the metabolism of dopamine. Here, we mined and integrated data from distinct human expression atlases and found that, among a wide range of tissues and cells, enterocytes of the small intestine express the highest expression levels of ACE2, DDC and several key genes supporting the metabolism of neurotransmitters. Based on these results, we performed co-expression analyses on a recently published set of RNA-seq data obtained from SARS-CoV2-infected human intestinal organoids. We observed that in SARS-CoV2-infected enterocytes, ACE2 co-regulates not only with DDC but also with a specific group of genes involved in (i) the dopamine/trace amines metabolic pathway, (ii) the absorption of microbiota-derived L-DOPA and (iii) the absorption of neutral amino acids serving as precursors to neurotransmitters. We conclude that in patients with long COVID, a chronic infection and inflammation of small intestine enterocytes might be indirectly responsible for prolonged brain alterations.
Collapse
Affiliation(s)
- Serge Nataf
- INSERM, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France;
- Bank of Tissues and Cells, Lyon University Hospital (Hospices Civils de Lyon), 69003 Lyon, France
| | - Laurent Pays
- INSERM, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France;
- Bank of Tissues and Cells, Lyon University Hospital (Hospices Civils de Lyon), 69003 Lyon, France
| |
Collapse
|
24
|
Docherty JR, Alsufyani HA. Pharmacology of Drugs Used as Stimulants. J Clin Pharmacol 2021; 61 Suppl 2:S53-S69. [PMID: 34396557 DOI: 10.1002/jcph.1918] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/22/2021] [Indexed: 12/21/2022]
Abstract
Psychostimulant, cardiovascular, and temperature actions of stimulants involve adrenergic (norepinephrine), dopaminergic (dopamine), and serotonergic (serotonin) pathways. Stimulants such as amphetamine, 3,4-methylenedioxymethamphetamine (MDMA), or mephedrone can act on the neuronal membrane monoamine transporters NET, DAT, and SERT and/or the vesicular monoamine transporter 2 to inhibit reuptake of neurotransmitter or cause release by reverse transport. Stimulants may have additional effects involving pre- and postsynaptic/junctional receptors for norepinephrine, dopamine, and serotonin and other receptors. As a result, stimulants may have a wide range of possible actions. Agents with cocaine or MDMA-like actions can induce serious and potentially fatal adverse events via thermodysregulatory, cardiovascular, or other mechanisms. MDMA-like stimulants may cause hyperthermia that can be life threathening. Recreational users of stimulants should be aware of the dangers of hyperthermia in a rave/club environment.
Collapse
Affiliation(s)
| | - Hadeel A Alsufyani
- Department of Physiology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
25
|
Effect of TAAR1/5-HT 1A agonist SEP-363856 on REM sleep in humans. Transl Psychiatry 2021; 11:228. [PMID: 33879769 PMCID: PMC8058073 DOI: 10.1038/s41398-021-01331-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/02/2021] [Accepted: 03/23/2021] [Indexed: 02/05/2023] Open
Abstract
SEP-363856 is a trace amine-associated receptor 1 (TAAR1) and 5-hydroxytryptamine type 1A (5-HT1A) agonist, currently in Phase 3 clinical trials for the treatment of schizophrenia. Although SEP-363856 activates TAAR1 and 5-HT1A receptors in vitro, an accessible marker of time- and concentration-dependent effects of SEP-363856 in humans is lacking. In rodents, SEP-363856 has been shown to suppress rapid eye movement (REM) sleep. The aim of the current study was to translate the REM sleep effects to humans and determine pharmacokinetic/pharmacodynamic (PK/PD) relationships of SEP-363856 on a measure of brain activity. The effects of SEP-363856 were evaluated in a randomized, double-blind, placebo-controlled, 2-way crossover study of single oral doses (50 and 10 mg) on REM sleep in healthy male subjects (N = 12 at each dose level). Drug concentrations were sampled during sleep to interpolate individual subject's pharmacokinetic trajectories. SEP-363856 suppressed REM sleep parameters with very large effect sizes (>3) following single doses of 50 mg and plasma concentrations ≥100 ng/mL. Below that effective concentration, the 10 mg dose elicited much smaller effects, increasing only the latency to REM sleep (effect size = 1). The PK/PD relationships demonstrated that REM sleep probability increased as drug concentrations declined below 100 ng/mL over the course of the night. SEP-363856 was generally safe and well tolerated at both doses. The REM sleep-suppressing effects of SEP-363856 provide an accessible marker of brain activity, which can aid in dose selection and help elucidate its therapeutic potential in further clinical trials.
Collapse
|
26
|
Ryan RM, Ingram SL, Scimemi A. Regulation of Glutamate, GABA and Dopamine Transporter Uptake, Surface Mobility and Expression. Front Cell Neurosci 2021; 15:670346. [PMID: 33927596 PMCID: PMC8076567 DOI: 10.3389/fncel.2021.670346] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/15/2021] [Indexed: 01/31/2023] Open
Abstract
Neurotransmitter transporters limit spillover between synapses and maintain the extracellular neurotransmitter concentration at low yet physiologically meaningful levels. They also exert a key role in providing precursors for neurotransmitter biosynthesis. In many cases, neurons and astrocytes contain a large intracellular pool of transporters that can be redistributed and stabilized in the plasma membrane following activation of different signaling pathways. This means that the uptake capacity of the brain neuropil for different neurotransmitters can be dynamically regulated over the course of minutes, as an indirect consequence of changes in neuronal activity, blood flow, cell-to-cell interactions, etc. Here we discuss recent advances in the mechanisms that control the cell membrane trafficking and biophysical properties of transporters for the excitatory, inhibitory and modulatory neurotransmitters glutamate, GABA, and dopamine.
Collapse
Affiliation(s)
- Renae M. Ryan
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Susan L. Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | | |
Collapse
|
27
|
Sun J, Chen J, Kumata K, Xiao Z, Rong J, Haider A, Shao T, Wang L, Xu H, Zhang MR, Liang SH. Imaging the trace amine-associated receptor 1 by positron emission tomography. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2021.153007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
28
|
Role of trace amine‑associated receptor 1 in the medial prefrontal cortex in chronic social stress-induced cognitive deficits in mice. Pharmacol Res 2021; 167:105571. [PMID: 33753244 DOI: 10.1016/j.phrs.2021.105571] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022]
Abstract
Emerging evidence supports an essential role of trace amine-associated receptor 1 (TAAR1) in neuropsychiatric disorders such as depression and schizophrenia. Stressful events are critical contributors to various neuropsychiatric disorders. This study examined the role of TAAR1 in mediating the negative outcomes of stressful events. In mice that experienced chronic social defeat stress but not acute stress, a significant reduction in the TAAR1 mRNA level was found in the medial prefrontal cortex (mPFC), a brain region that is known to be vulnerable to stress experience. Conditional TAAR1 knockout in the mPFC mimicked the cognitive deficits induced by chronic stress. In addition, chronic treatment with the selective TAAR1 partial agonist RO5263397 ameliorated chronic stress-induced changes in cognitive function, dendritic arborization, and the synapse number of pyramidal neurons in the mPFC but did not affect chronic stress-induced anxiety-like behaviors. Biochemically, chronic stress reduced the ratio of vesicular transporters of glutamate-1 (VGluT1) / vesicular GABA transporter (VGAT) in the mPFC,most prominently in the prelimbic cortex, and RO5263397 restored the excitatory-inhibitory (E/I) imbalance. Together, the results of this study reveal an essential role of TAAR1 in mediating chronic stress-induced cognitive impairments and suggest that TAAR1 agonists may be uniquely useful to treat MDD-related cognitive impairments.
Collapse
|
29
|
Kubicskó K, Farkas Ö. Quantum chemical (QM:MM) investigation of the mechanism of enzymatic reaction of tryptamine and N,N-dimethyltryptamine with monoamine oxidase A. Org Biomol Chem 2020; 18:9660-9674. [PMID: 33215182 DOI: 10.1039/d0ob01118e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The endogenous psychedelic (mind-altering) N,N-dimethyltryptamine (DMT) molecule has an important role in tissue protection, regeneration, and immunity via sigma-1 receptor activation as its natural ligand. The immunologic properties of DMT suggest this biogenic compound should be investigated thoroughly in other aspects as well. In our in silico project, we examined the metabolism of DMT and its primary analogue, the tryptamine (T), by the monoamine oxidase (MAO) flavoenzyme. MAO has two isoforms, MAO-A and MAO-B. MAOs perform the oxidation of various monoamines by their flavin adenine dinucleotide (FAD) cofactor. Two-layer QM:MM calculations at the ONIOM(M06-2X/6-31++G(d,p):UFF=QEq) level were performed including the whole enzyme to explore the potential energy surface (PES) of the reactions. Our findings reinforced that a hybrid mechanism, a mixture of pure H+ and H- transfer pathways, describes precisely the rate-determining step of amine oxidation as suggested by earlier works. Additionally, our results show that the oxidation of tertiary amine DMT requires a lower activation barrier than the primary amine T. This may reflect a general rule, thus we recommend further investigations. Furthermore, we demonstrated that at pH 7.4 the protonated form of these substrates enter the enzyme. As the deprotonation of substrates is crucial, we presumed protonated cofactor, FADH+, may form. Surprisingly, the activation barriers are much lower compared to FAD with both substrates. Therefore, we suggest further investigations in this direction.
Collapse
Affiliation(s)
- Károly Kubicskó
- Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary.
| | | |
Collapse
|
30
|
Grinchii D, Dremencov E. Mechanism of Action of Atypical Antipsychotic Drugs in Mood Disorders. Int J Mol Sci 2020; 21:ijms21249532. [PMID: 33333774 PMCID: PMC7765178 DOI: 10.3390/ijms21249532] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/03/2020] [Accepted: 12/11/2020] [Indexed: 01/07/2023] Open
Abstract
Atypical antipsychotic drugs were introduced in the early 1990s. Unlike typical antipsychotics, which are effective only against positive symptoms of schizophrenia, atypical antipsychotics are effective against negative and cognitive symptoms as well. Furthermore, they are effective not only in psychotic but also in affective disorders, on their own or as adjuncts to antidepressant drugs. This review presents the neural mechanisms of currently existing atypical antipsychotics and putative antipsychotics currently being investigated in preclinical and clinical studies and how these relate to their effectiveness in mood disorders such as depression, anxiety, and post-traumatic stress disorder (PTSD). Typical antipsychotics act almost exclusively on the dopamine system. Atypical drugs, however, modulate serotonin (5-HT), norepinephrine, and/or histamine neurotransmission as well. This multimodal mechanism of action putatively underlies the beneficial effect of atypical antipsychotics in mood and anxiety disorders. Interestingly, novel experimental drugs having dual antipsychotic and antidepressant therapeutic potential, such as histamine, adenosine, and trace amine-associated receptors (TAAR) ligand, are also characterized by a multimodal stimulatory effect on central 5-HT, norepinephrine, and/or histamine transmission. The multimodal stimulatory effect on central monoamine neurotransmission may be thus primarily responsible for the combined antidepressant and antipsychotic therapeutic potential of certain central nervous system (CNS) drugs.
Collapse
|
31
|
Novel 1-Amidino-4-Phenylpiperazines as Potent Agonists at Human TAAR1 Receptor: Rational Design, Synthesis, Biological Evaluation and Molecular Docking Studies. Pharmaceuticals (Basel) 2020; 13:ph13110391. [PMID: 33202687 PMCID: PMC7697893 DOI: 10.3390/ph13110391] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/19/2022] Open
Abstract
Targeting trace amine-associated receptor 1 (TAAR1) receptor continues to offer an intriguing opportunity to develop innovative therapies in different pharmacological settings. Pursuing our endeavors in the search for effective and safe human TAAR1 (hTAAR1) ligands, we synthesized a new series of 1-amidino-4-phenylpiperazine derivatives (1–16) based on the application of a combined pharmacophore model/scaffold simplification strategy for an in-house series of biguanide-based TAAR1 agonists. Most of the novel compounds proved to be more effective than their prototypes, showing nanomolar EC50 values in functional activity at hTAAR1 and low general cytotoxicity (CC50 > 80 µM) when tested on the Vero-76 cell line. In this new series, the main determinant for TAAR1 agonism ability appears to result from the appropriate combination between the steric size and position of the substituents on the phenyl ring rather than from their different electronic nature, since both electron-withdrawing and electron donor groups are permitted. In particular, the ortho-substitution seems to impose a more appropriate spatial geometry to the molecule that entails an enhanced TAAR1 potency profile, as experienced, in the following order, by compounds 15 (2,3-diCl, EC50 = 20 nM), 2 (2-CH3, EC50 = 30 nM), 6 (2-OCH3, EC50 = 93 nM) and 3 (2-Cl, EC50 = 160 nM). Apart from the interest in them as valuable leads for the development of promising hTAAR1 agonists, these simple small molecules have further allowed us to identify the minimal structural requirements for producing an efficient hTAAR1 targeting ability.
Collapse
|
32
|
Cadaverine and Spermine Elicit Ca 2+ Uptake in Human CP Cells via a Trace Amine-Associated Receptor 1 Dependent Pathway. J Mol Neurosci 2020; 71:625-637. [PMID: 32816235 DOI: 10.1007/s12031-020-01684-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/11/2020] [Indexed: 12/24/2022]
Abstract
The choroid plexus (CP) constitutes a barrier between the blood and the cerebrospinal fluid (CSF) which regulates the exchange of substances between these two fluids through mechanisms that are not completely understood. Polyamines as spermine, spermidine and putrescine are produced by all cells and are present in the CSF. Interestingly, their levels are altered in some neuronal disorders as Alzheimer's and Parkinson's diseases, thus increasing the interest in their signalling in the central nervous system (CNS). Cadaverine, on the other hand, is synthetized by the intestinal microbiome, suggesting that the presence of this bacterial metabolite in the CSF requires that it is up taken to the CNS across brain barriers. We knew that polyamines are detected by the olfactory signalling cascade operating at the CP, but the receptor involved had not been identified. The zebrafish TAAR13c was the only receptor known to bind a polyamine-cadaverine. Thus, we searched for a human receptor with homology to TAAR13c and found that some human TAARs including TAAR1 showed great homology. Then, we confirmed the expression of TAAR1 mRNA and protein in a human cell line of the CP, and in human CP samples. Calcium imaging assays after TAAR1 knockdown in these cells with a specific siRNA against TAAR1 showed a consistent reduction in the responses of these cells to cadaverine and spermidine, but not to spermine, suggesting that TAAR1 is activated by cadaverine and spermidine, but not spermine.
Collapse
|
33
|
Decker AM, Mathews KM, Blough BE, Gilmour BP. Validation of a High-Throughput Calcium Mobilization Assay for the Human Trace Amine-Associated Receptor 1. SLAS DISCOVERY 2020; 26:140-150. [PMID: 32734809 DOI: 10.1177/2472555220945279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The human trace amine-associated receptor 1 (hTAAR1) is a G protein-coupled receptor (GPCR) that is widely expressed in monoaminergic nuclei in the central nervous system and has therapeutic potential for multiple diseases, including drug addiction and schizophrenia. Thus, identification of novel hTAAR1 ligands is critical to advancing our knowledge of hTAAR1 function and to the development of therapeutics for a wide range of diseases. Herein we describe the development of a robust, 3-addition high-throughput screening (HTS) calcium mobilization assay using stable CHO-Gαq16-hTAAR1 cells, which functionally couple hTAAR1 to the promiscuous Gαq16 protein and thus allow signal transduction to occur through mobilization of internal calcium. Our previously established 96-well hTAAR1 assay was first miniaturized to the 384-well format and optimized to provide an assay with a Z' factor of 0.84, which is indicative of a robust HTS assay. Using the 3-addition protocol, 22,000 compounds were screened and yielded a ~1% agonist hit rate and a ~0.2% antagonist hit rate. Of the antagonist hits, two confirmed hits are the most potent hTAAR1 antagonists identified to date (IC50 = 206 and 281 nM). While scientists have been studying hTAAR1 for years, the lack of suitable hTAAR1 antagonists has been a major roadblock for studying the basic pharmacology of hTAAR1. Thus, these new ligands will serve as valuable tools to study hTAAR1-mediated signaling mechanisms, therapeutic potential, and in vivo functions.
Collapse
Affiliation(s)
- Ann M Decker
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| | - Kelly M Mathews
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| | - Brian P Gilmour
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| |
Collapse
|
34
|
Koblan KS, Kent J, Hopkins SC, Krystal JH, Cheng H, Goldman R, Loebel A. A Non-D2-Receptor-Binding Drug for the Treatment of Schizophrenia. N Engl J Med 2020; 382:1497-1506. [PMID: 32294346 DOI: 10.1056/nejmoa1911772] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND An oral compound, SEP-363856, that does not act on dopamine D2 receptors but has agonist activity at trace amine-associated receptor 1 (TAAR1) and 5-hydroxytryptamine type 1A (5-HT1A) receptors, may represent a new class of psychotropic agent for the treatment of psychosis in schizophrenia. METHODS We performed a randomized, controlled trial to evaluate the efficacy and safety of SEP-363856 in adults with an acute exacerbation of schizophrenia. The patients were randomly assigned in a 1:1 ratio to receive once-daily treatment with SEP-363856 (50 mg or 75 mg) or placebo for 4 weeks. The primary end point was the change from baseline in the total score on the Positive and Negative Symptom Scale (PANSS; range, 30 to 210; higher scores indicate more severe psychotic symptoms) at week 4. There were eight secondary end points, including the changes from baseline in the scores on the Clinical Global Impressions Severity (CGI-S) scale and the Brief Negative Symptom Scale (BNSS). RESULTS A total of 120 patients were assigned to the SEP-363856 group and 125 to the placebo group. The mean total score on the PANSS at baseline was 101.4 in the SEP-363856 group and 99.7 in the placebo group, and the mean change at week 4 was -17.2 points and -9.7 points, respectively (least-squares mean difference, -7.5 points; 95% confidence interval, -11.9 to -3.0; P = 0.001). The reductions in the CGI-S and BNSS scores at week 4 were generally in the same direction as those for the primary outcome, but the results were not adjusted for multiple comparisons. Adverse events with SEP-363856 included somnolence and gastrointestinal symptoms; one sudden cardiac death occurred in the SEP-363856 group. The incidence of extrapyramidal symptoms and changes in the levels of lipids, glycated hemoglobin, and prolactin were similar in the trial groups. CONCLUSIONS In this 4-week trial involving patients with an acute exacerbation of schizophrenia, SEP-363856, a non-D2-receptor-binding antipsychotic drug, resulted in a greater reduction from baseline in the PANSS total score than placebo. Longer and larger trials are necessary to confirm the effects and side effects of SEP-363856, as well as its efficacy relative to existing drug treatments for patients with schizophrenia. (Funded by Sunovion Pharmaceuticals; ClinicalTrials.gov number, NCT02969382.).
Collapse
Affiliation(s)
- Kenneth S Koblan
- From Sunovion Pharmaceuticals, Marlborough, MA (K.S.K., J.K., S.C.H., H.C., R.G., A.L.); and the Department of Psychiatry, Yale University, the Department of Neuroscience, Yale University School of Medicine, and Behavioral Health Services, Yale New Haven Hospital, New Haven (J.H.K.), and the Clinical Neurosciences Division, Veterans Affairs National Center for PTSD, Veterans Affairs Connecticut Healthcare System, West Haven (J.H.K.) - all in Connecticut
| | - Justine Kent
- From Sunovion Pharmaceuticals, Marlborough, MA (K.S.K., J.K., S.C.H., H.C., R.G., A.L.); and the Department of Psychiatry, Yale University, the Department of Neuroscience, Yale University School of Medicine, and Behavioral Health Services, Yale New Haven Hospital, New Haven (J.H.K.), and the Clinical Neurosciences Division, Veterans Affairs National Center for PTSD, Veterans Affairs Connecticut Healthcare System, West Haven (J.H.K.) - all in Connecticut
| | - Seth C Hopkins
- From Sunovion Pharmaceuticals, Marlborough, MA (K.S.K., J.K., S.C.H., H.C., R.G., A.L.); and the Department of Psychiatry, Yale University, the Department of Neuroscience, Yale University School of Medicine, and Behavioral Health Services, Yale New Haven Hospital, New Haven (J.H.K.), and the Clinical Neurosciences Division, Veterans Affairs National Center for PTSD, Veterans Affairs Connecticut Healthcare System, West Haven (J.H.K.) - all in Connecticut
| | - John H Krystal
- From Sunovion Pharmaceuticals, Marlborough, MA (K.S.K., J.K., S.C.H., H.C., R.G., A.L.); and the Department of Psychiatry, Yale University, the Department of Neuroscience, Yale University School of Medicine, and Behavioral Health Services, Yale New Haven Hospital, New Haven (J.H.K.), and the Clinical Neurosciences Division, Veterans Affairs National Center for PTSD, Veterans Affairs Connecticut Healthcare System, West Haven (J.H.K.) - all in Connecticut
| | - Hailong Cheng
- From Sunovion Pharmaceuticals, Marlborough, MA (K.S.K., J.K., S.C.H., H.C., R.G., A.L.); and the Department of Psychiatry, Yale University, the Department of Neuroscience, Yale University School of Medicine, and Behavioral Health Services, Yale New Haven Hospital, New Haven (J.H.K.), and the Clinical Neurosciences Division, Veterans Affairs National Center for PTSD, Veterans Affairs Connecticut Healthcare System, West Haven (J.H.K.) - all in Connecticut
| | - Robert Goldman
- From Sunovion Pharmaceuticals, Marlborough, MA (K.S.K., J.K., S.C.H., H.C., R.G., A.L.); and the Department of Psychiatry, Yale University, the Department of Neuroscience, Yale University School of Medicine, and Behavioral Health Services, Yale New Haven Hospital, New Haven (J.H.K.), and the Clinical Neurosciences Division, Veterans Affairs National Center for PTSD, Veterans Affairs Connecticut Healthcare System, West Haven (J.H.K.) - all in Connecticut
| | - Antony Loebel
- From Sunovion Pharmaceuticals, Marlborough, MA (K.S.K., J.K., S.C.H., H.C., R.G., A.L.); and the Department of Psychiatry, Yale University, the Department of Neuroscience, Yale University School of Medicine, and Behavioral Health Services, Yale New Haven Hospital, New Haven (J.H.K.), and the Clinical Neurosciences Division, Veterans Affairs National Center for PTSD, Veterans Affairs Connecticut Healthcare System, West Haven (J.H.K.) - all in Connecticut
| |
Collapse
|
35
|
Freyberg Z, Saavedra JM. Trace Amines and Trace Amine-Associated Receptors: A New Frontier in Cell Signaling. Cell Mol Neurobiol 2020; 40:189-190. [PMID: 32006222 DOI: 10.1007/s10571-020-00800-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 10/25/2022]
Abstract
Trace amines, including β-phenylethylamine, p-octopamine, p-tyramine, and tryptamine, are produced in high levels in invertebrates where they play major roles in homeostasis regulation in a manner similar to that of adrenergic systems in mammals (Rutigliano et al. in Front Pharmacol 8:987, 2017; Gainetdinov et al. in Pharmacol Rev 70(3):549-620, 2018; Nagaya et al. in Neurosci Lett 329(3):324-328, 2002). In mammals, however, their levels are very low, initially prompting these molecules to be termed "trace" or "minor" amines in mammals with only a secondary role in the regulation of more abundant biogenic amines including catecholamines and serotonin (Gainetdinov et al. in Pharmacol Rev 70(3):549-620, 2018). The more recent discovery of trace amine-associated receptors (TAARs) revealed major, previously unsuspected roles of the trace amines and has led to increasing interest within the scientific community. For example, TAARs have been proposed to modulate signaling through dopamine (Schwartz et al. in Expert Opin Ther Targets 22(6):513-526, 2018). Furthermore, these receptors are implicated in both numerous physiological functions including regulation of sleep, olfaction, metabolism, and immunity as well in disease (e.g., substance abuse, neuropsychiatric disorders) (Gainetdinov et al. in Pharmacol Rev 70(3):549-620, 2018; Rutigliano et al. in Front Pharmacol 8:987, 2017). Consequently, trace amine and TAAR research is rapidly growing and is of great translational relevance. In this Special Issue, leaders in trace amine and TAAR research offer both reviews and original research papers that cover a wide range of topics from involvement of TAAR signaling in metabolic regulation and neurophysiology to implications of this signaling in neuropsychiatric diseases including substance abuse and schizophrenia. While a diverse range of topics is covered by these works, the common theme running through all of them is the increasing awareness that trace amine and TAAR signaling represent novel signaling mechanisms in the brain and periphery. These topics are both highly timely and of considerable importance not only for those working in the field but also for the neuroscience community at large.
Collapse
Affiliation(s)
- Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Juan M Saavedra
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, 20057, USA.
| |
Collapse
|
36
|
Abstract
Trace amine-associated receptors (TAARs) are a family of G protein-coupled receptors (GPCRs) that are evolutionarily conserved in vertebrates. The first discovered TAAR1 is mainly expressed in the brain, and is able to detect low abundant trace amines. TAAR1 is also activated by several synthetic compounds and psychostimulant drugs like amphetamine. Activation of TAAR1 by specific agonists can regulate the classical monoaminergic systems in the brain. Further studies have revealed that other TAAR family members are highly expressed in the olfactory system which are termed olfactory TAARs. In vertebrates, olfactory TAARs can specifically recognize volatile or water-soluble amines. Some of these TAAR agonists are produced by decarboxylation of amino acids. In addition, some TAAR agonists are ethological odors that mediate animal innate behaviors. In this study, we provide a comprehensive review of TAAR agonists, including their structures, biosynthesis pathways, and functions.
Collapse
Affiliation(s)
- Zhengrong Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Qian Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China.
| |
Collapse
|
37
|
Bugda Gwilt K, González DP, Olliffe N, Oller H, Hoffing R, Puzan M, El Aidy S, Miller GM. Actions of Trace Amines in the Brain-Gut-Microbiome Axis via Trace Amine-Associated Receptor-1 (TAAR1). Cell Mol Neurobiol 2020; 40:191-201. [PMID: 31836967 DOI: 10.1007/s10571-019-00772-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022]
Abstract
Trace amines and their primary receptor, Trace Amine-Associated Receptor-1 (TAAR1) are widely studied for their involvement in the pathogenesis of neuropsychiatric disorders despite being found in the gastrointestinal tract at physiological levels. With the emergence of the "brain-gut-microbiome axis," we take the opportunity to review what is known about trace amines in the brain, the defined sources of trace amines in the gut, and emerging understandings on the levels of trace amines in various gastrointestinal disorders. Similarly, we discuss localization of TAAR1 expression in the gut, novel findings that TAAR1 may be implicated in inflammatory bowel diseases, and the reported comorbidities of neuropsychiatric disorders and gastrointestinal disorders. With the emergence of TAAR1 specific compounds as next-generation therapeutics for schizophrenia (Roche) and Parkinson's related psychoses (Sunovion), we hypothesize a therapeutic benefit of these compounds in clinical trials in the brain-gut-microbiome axis, as well as a potential for thoughtful manipulation of the brain-gut-microbiome axis to modulate symptoms of neuropsychiatric disease.
Collapse
Affiliation(s)
- Katlynn Bugda Gwilt
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA.
- Center for Drug Discovery, Northeastern University, Boston, MA, USA.
- Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, USA.
| | - Dulce Pamela González
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Neva Olliffe
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Department of Biology, College of Science, Northeastern University, Boston, MA, USA
| | - Haley Oller
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Rachel Hoffing
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Department of Biology, College of Science, Northeastern University, Boston, MA, USA
| | - Marissa Puzan
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, USA
| | - Sahar El Aidy
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Gregory M Miller
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
38
|
Accorroni A, Rutigliano G, Sabatini M, Frascarelli S, Borsò M, Novelli E, Bandini L, Ghelardoni S, Saba A, Zucchi R, Origlia N. Exogenous 3-Iodothyronamine Rescues the Entorhinal Cortex from β-Amyloid Toxicity. Thyroid 2020; 30:147-160. [PMID: 31709926 DOI: 10.1089/thy.2019.0255] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: A novel form of thyroid hormone (TH) signaling is represented by 3-iodothyronamine (T1AM), an endogenous TH derivative that interacts with specific molecular targets, including trace amine-associated receptor 1 (TAAR1), and induces pro-learning and anti-amnestic effects in mice. Dysregulation of TH signaling has long been hypothesized to play a role in Alzheimer's disease (AD). In the present investigation, we explored the neuroprotective role of T1AM in beta amyloid (Aβ)-induced synaptic and behavioral impairment, focusing on the entorhinal cortex (EC), an area that is affected early by AD pathology. Methods: Field potentials were evoked in EC layer II, and long-term potentiation (LTP) was elicited by high frequency stimulation (HFS). T1AM (5 μM) and/or Aβ(1-42) (200 nM), were administered for 10 minutes, starting 5 minutes before HFS. Selective TAAR1 agonist RO5166017 (250 nM) and TAAR1 antagonist EPPTB (5 nM) were also used. The electrophysiological experiments were repeated in EC-slices taken from a mouse model of AD (mutant human amyloid precursor protein [mhAPP], J20 line). We also assessed the in vivo effects of T1AM on EC-dependent associative memory deficits, which were detected in mhAPP mice by behavioral evaluations based on the novel-object recognition paradigm. TAAR1 expression was determined by Western blot, whereas T1AM and its metabolite 3-iodothyroacetic acid (TA1) were assayed by high-performance liquid chromatography coupled to mass spectrometry. Results: We demonstrate the presence of endogenous T1AM and TAAR1 in the EC of wild-type and mhAPP mice. Exposure to Aβ(1-42) inhibited LTP, and T1AM perfusion (at a concentration of 5 μM, leading to an actual concentration in the perfusion buffer ranging from 44 to 298 nM) restored it, whereas equimolar amounts of 3,5,3'-triiodo-L-thyronine (T3) and TA1 were ineffective. The response to T1AM was abolished by the TAAR1 antagonist EPPTB, whereas it was mimicked by the TAAR1 agonist RO5166017. In the EC of APPJ20 mice, LTP could not be elicited, but it was rescued by T1AM. The intra-cerebro-ventricular administration of T1AM (0.89 μg/kg) also restored recognition memory that was impaired in mhAPP mice. Conclusions: Our results suggest that T1AM and TAAR1 are part of an endogenous system that can be modulated to prevent synaptic and behavioral deficits associated with Aβ-related toxicity.
Collapse
Affiliation(s)
- Alice Accorroni
- Scuola Superiore di Studi Universitari e di Perfezionamento Sant'Anna, Pisa, Italy
- Institute of Neuroscience of the Italian National Research Council (CNR), Pisa, Italy
| | - Grazia Rutigliano
- Scuola Superiore di Studi Universitari e di Perfezionamento Sant'Anna, Pisa, Italy
| | | | | | - Marco Borsò
- Department of Pathology, University of Pisa, Pisa, Italy
| | - Elena Novelli
- Institute of Neuroscience of the Italian National Research Council (CNR), Pisa, Italy
| | | | | | | | | | - Nicola Origlia
- Institute of Neuroscience of the Italian National Research Council (CNR), Pisa, Italy
| |
Collapse
|
39
|
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: 1.5] [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
|
40
|
Molecular Variants in Human Trace Amine-Associated Receptors and Their Implications in Mental and Metabolic Disorders. Cell Mol Neurobiol 2019; 40:239-255. [PMID: 31643000 PMCID: PMC7028809 DOI: 10.1007/s10571-019-00743-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023]
Abstract
We provide a comprehensive review of the available evidence on the pathophysiological implications of genetic variants in the human trace amine-associated receptor (TAAR) superfamily. Genes coding for trace amine-associated receptors (taars) represent a multigene family of G-protein-coupled receptors, clustered to a small genomic region of 108 kb located in chromosome 6q23, which has been consistently identified by linkage analyses as a susceptibility locus for schizophrenia and affective disorders. Most TAARs are expressed in brain areas involved in emotions, reward and cognition. TAARs are activated by endogenous trace amines and thyronamines, and evidence for a modulatory action on other monaminergic systems has been reported. Therefore, linkage analyses were followed by fine mapping association studies in schizophrenia and affective disorders. However, none of these reports has received sufficient universal replication, so their status remains uncertain. Single nucleotide polymorphisms in taars have emerged as susceptibility loci from genome-wide association studies investigating migraine and brain development, but none of the detected variants reached the threshold for genome-wide significance. In the last decade, technological advances enabled single-gene or whole-exome sequencing, thus allowing the detection of rare genetic variants, which may have a greater impact on the risk of complex disorders. Using these approaches, several taars (especially taar1) variants have been detected in patients with mental and metabolic disorders, and in some cases, defective receptor function has been demonstrated in vitro. Finally, with the use of transcriptomic and peptidomic techniques, dysregulations of TAARs (especially TAAR6) have been identified in brain disorders characterized by cognitive impairment.
Collapse
|
41
|
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: 2.3] [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
|
42
|
Rutigliano G, Bräunig J, Del Grande C, Carnicelli V, Masci I, Merlino S, Kleinau G, Tessieri L, Pardossi S, Paisdzior S, Dell'Osso L, Biebermann H, Zucchi R. Non-Functional Trace Amine-Associated Receptor 1 Variants in Patients With Mental Disorders. Front Pharmacol 2019; 10:1027. [PMID: 31572197 PMCID: PMC6753877 DOI: 10.3389/fphar.2019.01027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
Background: The G protein–coupled receptor (GPCR) trace amine-associated receptor 1 (TAAR1) is expressed across brain areas involved in emotions, reward and cognition, and modulates monoaminergic and glutamatergic neurotransmissions. TAAR1 is stimulated with nanomolar affinity by 3-iodothyronamine (T1AM), an endogenous messenger considered a novel branch of thyroid hormone signaling. The human gene for TAAR1 maps to locus 6q23, within a region associated with major mental disorders. Materials and Methods: We screened a cohort of patients with major mental disorders (n = 104) and a group of healthy controls (n = 130) for TAAR1 variants. HEK293 cells were transiently transfected with: i) wild-type TAAR1 and ii) mutated TAAR1, either in homozygous or heterozygous state. Cell surface expression and Gs/adenylyl cyclase activation upon administration of β-phenylethylamine (PEA), T1AM, and RO5166017, were assessed. Results: We detected 13 missense variants in TAAR1 coding region, with a significant enrichment in patients as compared to healthy controls (11 vs. 1, 1 variant in both groups, p < 0.01). In silico analysis identified four dysfunctional variants, all in patients. Three of these—R23C, Y131C, and C263R—were functionally characterized. In cells co-transfected with wild-type and mutated TAAR1, we observed a significant reduction of cell surface expression. In heterozygosity, the three TAAR1 variants substantially dampened Gs signaling in response to PEA, and, more robustly, to T1AM. Co-stimulation with PEA and RO5166017 did not yield any improvement in Gs signaling. R23C, Y131C, and C263R are rare in the general population and map in functionally important highly conserved positions across TAAR1 orthologous and paralogous genes. Conclusions: Our findings suggest that disruptions of TAAR1 activity may be relevant to the pathophysiology of mental disorders, thereby providing a promising target for novel psychopharmacological interventions.
Collapse
Affiliation(s)
- Grazia Rutigliano
- Scuola Superiore Sant'Anna, Pisa, Italy.,National Research Council (CNR), Institute of Clinical Physiology (IFC), Pisa, Italy
| | - Julia Bräunig
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute für Experimentelle Pädiatrische Endokrinology, Berlin, Germany
| | - Claudia Del Grande
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Pisa, Italy
| | | | - Isabella Masci
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Pisa, Italy
| | - Sergio Merlino
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Pisa, Italy
| | - Gunnar Kleinau
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Physics and Biophysics, Group Protein X-ray Crystallography and Signal Transduction, Berlin, Germany
| | | | | | - Sarah Paisdzior
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute für Experimentelle Pädiatrische Endokrinology, Berlin, Germany
| | - Liliana Dell'Osso
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Pisa, Italy
| | - Heike Biebermann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute für Experimentelle Pädiatrische Endokrinology, Berlin, Germany
| | | |
Collapse
|
43
|
Keeping up with the therapeutic advances in schizophrenia: a review of novel and emerging pharmacological entities. CNS Spectr 2019; 24:38-69. [PMID: 31482779 DOI: 10.1017/s109285291900124x] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Schizophrenia remains one of the most severe medical diseases. Current dopamine modulating first-generation and second-generation antipsychotics target mainly positive symptoms, but not/inadequately negative and cognitive symptoms. Additional challenges include non-adherence and adverse effects, especially cardiometabolic dysregulation. This review evaluates new/emerging pharmacological treatments for schizophrenia. Therapies targeting total symptoms include cannabidiol, D3 antagonist/5-HT1A partial agonist F17464, lumateperone (ITI-007), phosphodiesterase 10A (PDE10A) inhibitors MK-8189 and TAK-063, sodium nitroprusside, and trace amine-associated receptor-1 (TAAR1) agonist RO5263397 and SEP-363856. Treatments targeting negative symptoms include the PDE10A inhibitor LuAF-11167, 5-HT2A inverse agonist pimavanserin, sigma-2/5-HT2A antagonist roluperidone (MIN-101), and d-amino acid oxidase (DAAO) inhibitor TAK-831. Agents targeting primarily cognitive dysfunction are the glycine transporter-1 inhibitor BI-425809 and cannabidiol. Therapies targeting residual positive symptoms/treatment-resistant schizophrenia include pimavanserin, dopamine D1/D2 antagonist LuAF-35700, and DAAO inhibitor sodium benzoate. Two new long-acting injectable antipsychotic formulations, Aripiprazole Lauroxil NanoCrystal® and the first subcutaneous injectable LAI Perseris (RBP-7000), were recently approved by U.S. Food and Drug Administration, and positive results were announced for Risperidone ISM®, each achieving therapeutic levels within 24 hours, without need for initial oral cotreatment/loading injection-strategies. Paliperidone palmitate 6-monthly intramuscularly injectable and Risperidone subcutaneously injectable TV46000 are currently under investigation. Finally, the samidorphan+olanzapine combination targets reduced weight gain liability, while maintaining olanzapine's efficacy. Most of these trial programs are still ongoing or have yielded mixed or even negative results. Thus, additional mechanisms of action and agents require study to improve schizophrenia outcomes for total/positive symptoms with reduced adverse effects, but also cognitive symptoms, negative symptoms, and treatment resistance, the areas of greatest need in schizophrenia currently.
Collapse
|
44
|
Dedic N, Jones PG, Hopkins SC, Lew R, Shao L, Campbell JE, Spear KL, Large TH, Campbell UC, Hanania T, Leahy E, Koblan KS. SEP-363856, a Novel Psychotropic Agent with a Unique, Non-D2 Receptor Mechanism of Action. J Pharmacol Exp Ther 2019; 371:1-14. [DOI: 10.1124/jpet.119.260281] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/10/2019] [Indexed: 01/03/2023] Open
|
45
|
Takahashi T, Noriaki S, Matsumura M, Li C, Takahashi K, Nishino S. Advances in pharmaceutical treatment options for narcolepsy. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1521267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Tatsunori Takahashi
- Stanford University Sleep and Circadian Neurobiology Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sakai Noriaki
- Stanford University Sleep and Circadian Neurobiology Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Mari Matsumura
- Stanford University Sleep and Circadian Neurobiology Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Chenyu Li
- Stanford University Sleep and Circadian Neurobiology Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kayo Takahashi
- Stanford University Sleep and Circadian Neurobiology Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Seiji Nishino
- Stanford University Sleep and Circadian Neurobiology Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|