1
|
Vaganova AN, Fesenko ZS, Efimova EV, Chekrygin SA, Shafranskaya DD, Prjibelski AD, Katolikova NV, Gainetdinov RR. Knocking Out TAAR5: A Pathway to Enhanced Neurogenesis and Dopamine Signaling in the Striatum. Cells 2024; 13:1910. [PMID: 39594659 PMCID: PMC11592834 DOI: 10.3390/cells13221910] [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: 09/23/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The member of trace-amine associated receptor family, TAAR5 receptor was suggested to recognize tertiary amines, mostly in the olfactory system; however, knocking out the receptor TAAR5 in mice showed an enhancing effect on adult neurogenesis and dopamine neurotransmission in the striatum. To estimate the role of the TAAR5, we performed gene expression profiling of striatal samples from TAAR5 knockout (KO) mice and their wild-type littermates. The higher expression of several genes involved in dopaminergic signaling and the downregulation of genes associated with gliogenesis were revealed in TAAR5-KO mice. Meanwhile, the upregulating effect of TAAR5 knockout on genes was associated with neurogenesis and synaptogenesis. The estimation of cell-type relative abundance through the deconvolution of RNA sequencing data demonstrated that TAAR5-KO striatum samples contain more D2 dopamine receptor-expressing medium spiny neurons but fewer astrocytes than wild-type mice. Our findings indicate that previously identified improvement in cognitive functions and motor coordination in TAAR5-KO mice may activate genes involved in neurogenesis, synaptogenesis, and synapse organization in the striatum. These data suggest that the pharmaceutical targeting of TAAR5 may improve striatum-dependent cognitive or motor functions. At the same time, a more detailed investigation of future TAAR5 antagonists' effect on glia development is necessary.
Collapse
Affiliation(s)
- Anastasia N. Vaganova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia; (A.N.V.)
- Saint-Petersburg University Hospital, 199034 Saint-Petersburg, Russia
| | - Zoia S. Fesenko
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia; (A.N.V.)
| | - Evgeniya V. Efimova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia; (A.N.V.)
| | - Sergei A. Chekrygin
- Resource Center “Bio-Bank Center”, Research Park of Saint-Petersburg State University, 198504 Saint-Petersburg, Russia;
| | - Daria D. Shafranskaya
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia; (A.N.V.)
| | | | - Nataliia V. Katolikova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia; (A.N.V.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia; (A.N.V.)
- Saint-Petersburg University Hospital, 199034 Saint-Petersburg, Russia
| |
Collapse
|
2
|
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
|
3
|
Lin Y, Wang J, Shi F, Yang L, Wu S, Qiao A, Ye S. Molecular Mechanisms of Methamphetamine-Induced Addiction via TAAR1 Activation. J Med Chem 2024; 67:18593-18605. [PMID: 39358311 PMCID: PMC11513891 DOI: 10.1021/acs.jmedchem.4c01961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Trace amine-associated receptor 1 (TAAR1), a member of the trace amine receptor family, recognizes various trace amines in the brain, including endogenous β-phenylethylamine (PEA) and methamphetamine (METH). TAAR1 is a novel target for several neurological disorders, including schizophrenia, depression, and substance abuse. Herein, we report the structure of the human TAAR1-Gs protein complex bound to METH. Using functional studies, we reveal the molecular basis of METH recognition by TAAR1, and potential mechanisms underlying the selectivity of TAAR1 for different ligands. Molecular dynamics simulations further elucidated possible mechanisms for the binding of chiral amphetamine (AMPH)-like psychoactive drugs to TAAR1. Additionally, we discovered a hydrophobic core on the transmembrane helices (TM), TM5 and TM6, explaining the unique mechanism of TAAR1 activation. These findings reveal the ligand recognition pattern and activation mechanism of TAAR1, which has important implications for the development of next-generation treatments for substance abuse and various neurological disorders.
Collapse
Affiliation(s)
- Yun Lin
- Tianjin
Key Laboratory of Function and Application of Biological Macromolecular
Structures, School of Life Sciences, Tianjin
University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Jiening Wang
- State
Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative
Innovation Center for Green Transformation of Bio-Resources, Hubei
Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Fan Shi
- Department
of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Linlin Yang
- Department
of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shan Wu
- State
Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative
Innovation Center for Green Transformation of Bio-Resources, Hubei
Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Anna Qiao
- Tianjin
Key Laboratory of Function and Application of Biological Macromolecular
Structures, School of Life Sciences, Tianjin
University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Sheng Ye
- Tianjin
Key Laboratory of Function and Application of Biological Macromolecular
Structures, School of Life Sciences, Tianjin
University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| |
Collapse
|
4
|
Ye N, Wang Q, Li Y, Zhen X. Current emerging therapeutic targets and clinical investigational agents for schizophrenia: Challenges and opportunities. Med Res Rev 2024. [PMID: 39300769 DOI: 10.1002/med.22086] [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: 06/04/2024] [Revised: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Since the first discovery of antipsychotics in the 1950s, targeting dopaminergic drugs has manifested to well manage the positive symptoms of schizophrenia with limited efficacy for the negative and cognitive symptoms. In past decades, extensive efforts have been undertaken towards the development of innovative agents that can effectively stabilize the dopamine and serotonin systems or target to nondopaminergic pathways, leading to various promising drug candidates entering into clinical trials. Notably, the sigma-2, 5-HT2A, and α1A receptor antagonist roluperidone, as well as a fixed-dose combination of the M1/4 receptor agonist KarXT, have been submitted for NDA applications. The dual agonist ulotaront, which targets TAAR1 and 5-HT1A receptors, and the GlyT1 inhibitor iclepertin have advanced into phase 3 clinical trials. Nevertheless, satisfactory therapeutic strategies for schizophrenia remain elusive. This review highlights current clinical endeavors in developing novel chemical small-molecule entities and fixed-dose combinations for the treatment of schizophrenia since 2017, thus facilitating the efficient development of the next generation of antipsychotics.
Collapse
Affiliation(s)
- Na Ye
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Qi Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yue Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| |
Collapse
|
5
|
Díaz-Holguín A, Saarinen M, Vo DD, Sturchio A, Branzell N, Cabeza de Vaca I, Hu H, Mitjavila-Domènech N, Lindqvist A, Baranczewski P, Millan MJ, Yang Y, Carlsson J, Svenningsson P. AlphaFold accelerated discovery of psychotropic agonists targeting the trace amine-associated receptor 1. SCIENCE ADVANCES 2024; 10:eadn1524. [PMID: 39110804 PMCID: PMC11305387 DOI: 10.1126/sciadv.adn1524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Artificial intelligence is revolutionizing protein structure prediction, providing unprecedented opportunities for drug design. To assess the potential impact on ligand discovery, we compared virtual screens using protein structures generated by the AlphaFold machine learning method and traditional homology modeling. More than 16 million compounds were docked to models of the trace amine-associated receptor 1 (TAAR1), a G protein-coupled receptor of unknown structure and target for treating neuropsychiatric disorders. Sets of 30 and 32 highly ranked compounds from the AlphaFold and homology model screens, respectively, were experimentally evaluated. Of these, 25 were TAAR1 agonists with potencies ranging from 12 to 0.03 μM. The AlphaFold screen yielded a more than twofold higher hit rate (60%) than the homology model and discovered the most potent agonists. A TAAR1 agonist with a promising selectivity profile and drug-like properties showed physiological and antipsychotic-like effects in wild-type but not in TAAR1 knockout mice. These results demonstrate that AlphaFold structures can accelerate drug discovery.
Collapse
Affiliation(s)
- Alejandro Díaz-Holguín
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Marcus Saarinen
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
| | - Duc Duy Vo
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Andrea Sturchio
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
- Department of Neurology, James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, University of Cincinnati, Cincinnati, OH, USA
| | - Niclas Branzell
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
| | - Israel Cabeza de Vaca
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Huabin Hu
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Núria Mitjavila-Domènech
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Annika Lindqvist
- Department of Pharmacy, SciLifeLab Drug Discovery and Development Platform, Uppsala University, Box 580, SE-751 23 Uppsala, Sweden
| | - Pawel Baranczewski
- Department of Pharmacy, SciLifeLab Drug Discovery and Development Platform, Uppsala University, Box 580, SE-751 23 Uppsala, Sweden
| | - Mark J. Millan
- Neuroinflammation Therapeutic Area, Institut de Recherches Servier, Centre de Recherches de Croissy, Paris, France and Institute of Neuroscience and Psychology, College of Medicine, Vet and Life Sciences, Glasgow University, Scotland, Glasgow, UK
| | - Yunting Yang
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Box 596, SE-751 24 Uppsala, Sweden
| | - Per Svenningsson
- Neuro Svenningsson, Department of Clinical Neuroscience, Karolinska Institute, SE-171 76 Stockholm, Sweden
| |
Collapse
|
6
|
Scarano N, Espinoza S, Brullo C, Cichero E. Computational Methods for the Discovery and Optimization of TAAR1 and TAAR5 Ligands. Int J Mol Sci 2024; 25:8226. [PMID: 39125796 PMCID: PMC11312273 DOI: 10.3390/ijms25158226] [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: 07/02/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
G-protein-coupled receptors (GPCRs) represent a family of druggable targets when treating several diseases and continue to be a leading part of the drug discovery process. Trace amine-associated receptors (TAARs) are GPCRs involved in many physiological functions with TAAR1 having important roles within the central nervous system (CNS). By using homology modeling methods, the responsiveness of TAAR1 to endogenous and synthetic ligands has been explored. In addition, the discovery of different chemo-types as selective murine and/or human TAAR1 ligands has helped in the understanding of the species-specificity preferences. The availability of TAAR1-ligand complexes sheds light on how different ligands bind TAAR1. TAAR5 is considered an olfactory receptor but has specific involvement in some brain functions. In this case, the drug discovery effort has been limited. Here, we review the successful computational efforts developed in the search for novel TAAR1 and TAAR5 ligands. A specific focus on applying structure-based and/or ligand-based methods has been done. We also give a perspective of the experimental data available to guide the future drug design of new ligands, probing species-specificity preferences towards more selective ligands. Hints for applying repositioning approaches are also discussed.
Collapse
Affiliation(s)
- Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (C.B.)
| | - Stefano Espinoza
- Department of Health Sciences and Research Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), 28100 Novara, Italy;
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (C.B.)
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (C.B.)
| |
Collapse
|
7
|
Vaganova AN, Markina AA, Belousov AM, Lenskaia KV, Gainetdinov RR. Dopamine Receptors and TAAR1 Functional Interaction Patterns in the Duodenum Are Impaired in Gastrointestinal Disorders. Biomedicines 2024; 12:1590. [PMID: 39062162 PMCID: PMC11274761 DOI: 10.3390/biomedicines12071590] [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: 05/24/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Currently, there is a growing amount of evidence for the involvement of dopamine receptors and the functionally related trace amine-associated receptor, TAAR1, in upper intestinal function. In the present study, we analyzed their expression in the duodenum using publicly accessible transcriptomic data. We revealed the expression of DRD1, DRD2, DRD4, DRD5, and TAAR1 genes in different available datasets. The results of the gene ontology (GO) enrichment analysis for DRD2 and especially TAAR1 co-expressed genes were consistent with the previously described localization of D2 and TAAR1 in enteric neurons and secretory cells, respectively. Considering that co-expressed genes are more likely to be involved in the same biological processes, we analyzed genes that are co-expressed with TAAR1, DRD2, DRD4, and DRD5 genes in healthy mucosa and duodenal samples from patients with functional dyspepsia (FD) or diabetes-associated gastrointestinal symptoms. Both pathological conditions showed a deregulation of co-expression patterns, with a high discrepancy between DRDs and TAAR1 co-expressed gene sets in normal tissues and patients' samples and a loss of these genes' functional similarity. Meanwhile, we discovered specific changes in co-expression patterns that may suggest the involvement of TAAR1 and D5 receptors in pathologic or compensatory processes in FD or diabetes accordingly. Despite our findings suggesting the possible role of TAAR1 and dopamine receptors in functional diseases of the upper intestine, underlying mechanisms need experimental exploration and validation.
Collapse
Affiliation(s)
- Anastasia N. Vaganova
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.)
- St. Petersburg State University Hospital, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Alisa A. Markina
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.)
| | - Aleksandr M. Belousov
- St. Petersburg State University Hospital, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Karina V. Lenskaia
- Department of Medicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.)
- St. Petersburg State University Hospital, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| |
Collapse
|
8
|
Shajan B, Marri S, Bastiampillai T, Gregory KJ, Hellyer SD, Nair PC. Trace amine associated receptor 1: predicted effects of single nucleotide variants on structure-function in geographically diverse populations. Hum Genomics 2024; 18:61. [PMID: 38863077 PMCID: PMC11165750 DOI: 10.1186/s40246-024-00620-w] [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: 03/27/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024] Open
Abstract
Trace Amine Associated Receptor 1 (TAAR1) is a novel pharmaceutical target under investigation for the treatment of several neuropsychiatric conditions. TAAR1 single nucleotide variants (SNV) have been found in patients with schizophrenia and metabolic disorders. However, the frequency of variants in geographically diverse populations and the functional effects of such variants are unknown. In this study, we aimed to characterise the distribution of TAAR1 SNVs in five different WHO regions using the Database of Genotypes and Phenotypes (dbGaP) and conducted a critical computational analysis using available TAAR1 structural data to identify SNVs affecting ligand binding and/or functional regions. Our analysis shows 19 orthosteric, 9 signalling and 16 micro-switch SNVs hypothesised to critically influence the agonist induced TAAR1 activation. These SNVs may non-proportionally influence populations from discrete regions and differentially influence the activity of TAAR1-targeting therapeutics in genetically and geographically diverse populations. Notably, our dataset presented with orthosteric SNVs D1033.32N (found only in the South-East Asian Region and Western Pacific Region) and T1945.42A (found only in South-East Asian Region), and 2 signalling SNVs (V1253.54A/T2526.36A, found in African Region and commonly, respectively), all of which have previously demonstrated to influence ligand induced functions of TAAR1. Furthermore, bioinformatics analysis using SIFT4G, MutationTaster 2, PROVEAN and MutationAssessor predicted all 16 micro-switch SNVs are damaging and may further influence the agonist activation of TAAR1, thereby possibly impacting upon clinical outcomes. Understanding the genetic basis of TAAR1 function and the impact of common mutations within clinical populations is important for the safe and effective utilisation of novel and existing pharmacotherapies.
Collapse
Affiliation(s)
- Britto Shajan
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Shashikanth Marri
- Flinders Health and Medical Research Institute (FHMRI) College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Tarun Bastiampillai
- Department of Psychiatry, Monash University, Parkville, Melbourne, VIC, Australia
- Discipline of Psychiatry, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Shane D Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, 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, University of Adelaide, Adelaide, South Australia, Australia.
- Discipline of Medicine, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
9
|
Li M, Liu Y, Sun M, Yang Y, Zhang L, Liu Y, Li F, Liu H. SEP-363856 exerts neuroprotection through the PI3K/AKT/GSK-3β signaling pathway in a dual-hit neurodevelopmental model of schizophrenia-like mice. Drug Dev Res 2024; 85:e22225. [PMID: 38879781 DOI: 10.1002/ddr.22225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 10/11/2024]
Abstract
Schizophrenia (SZ) is a serious, destructive neurodevelopmental disorder. Antipsychotic medications are the primary therapy approach for this illness, but it's important to pay attention to the adverse effects as well. Clinical studies for SZ are currently in phase ΙΙΙ for SEP-363856 (SEP-856)-a new antipsychotic that doesn't work on dopamine D2 receptors. However, the underlying action mechanism of SEP-856 remains unknown. This study aimed to evaluate the impact and underlying mechanisms of SEP-856 on SZ-like behavior in a perinatal MK-801 treatment combined with social isolation from the weaning to adulthood model (MK-SI). First, we created an animal model that resembles SZ that combines the perinatal MK-801 with social isolation from weaning to adulthood. Then, different classical behavioral tests were used to evaluate the antipsychotic properties of SEP-856. The levels of proinflammatory cytokines (tumor necrosis factor-α, interleukin-6, and interleukin-1β), apoptosis-related genes (Bax and Bcl-2), and synaptic plasticity-related genes (brain-derived neurotrophic factor [BDNF] and PSD-95) in the hippocampus were analyzed by quantitative real-time PCR. Hematoxylin and eosin staining were used to observe the morphology of neurons in the hippocampal DG subregions. Western blot was performed to detect the protein expression levels of BDNF, PSD-95, Bax, Bcl-2, PI3K, p-PI3K, AKT, p-AKT, GSK-3β, p-GSK-3β in the hippocampus. MK-SI neurodevelopmental disease model studies have shown that compared with sham group, MK-SI group exhibit higher levels of autonomic activity, stereotyped behaviors, withdrawal from social interactions, dysregulated sensorimotor gating, and impaired recognition and spatial memory. These findings imply that the MK-SI model can mimic symptoms similar to those of SZ. Compared with the MK-SI model, high doses of SEP-856 all significantly reduced increased activity, improved social interaction, reduced stereotyping behavior, reversed sensorimotor gating dysregulation, and improved recognition memory and spatial memory impairment in MK-SI mice. In addition, SEP-856 can reduce the release of proinflammatory factors in the MK-SI model, promote the expression of BDNF and PSD-95 in the hippocampus, correct the Bax/Bcl-2 imbalance, turn on the PI3K/AKT/GSK-3β signaling pathway, and ultimately help the MK-SI mice's behavioral abnormalities. SEP-856 may play an antipsychotic role in MK-SI "dual-hit" model-induced SZ-like behavior mice by promoting synaptic plasticity recovery, decreasing death of hippocampal neurons, lowering the production of pro-inflammatory substances in the hippocampal region, and subsequently initiating the PI3K/AKT/GSK-3β signaling cascade.
Collapse
Affiliation(s)
- Mengdie Li
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yunxiao Liu
- Department of Psychiatry, School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Meng Sun
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yating Yang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ling Zhang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuexia Liu
- The Second People's Hospital of Huizhou, Huizhou, Guangdong, China
| | - Fujin Li
- The Second People's Hospital of Huizhou, Huizhou, Guangdong, China
| | - Huanzhong Liu
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
10
|
Barabássy Á, Dombi ZB, Németh G. D3 Receptor-Targeted Cariprazine: Insights from Lab to Bedside. Int J Mol Sci 2024; 25:5682. [PMID: 38891871 PMCID: PMC11172134 DOI: 10.3390/ijms25115682] [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: 04/18/2024] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
Until the late 1800s, drug development was a chance finding based on observations and repeated trials and errors. Today, drug development must go through many iterations and tests to ensure it is safe, potent, and effective. This process is a long and costly endeavor, with many pitfalls and hurdles. The aim of the present review article is to explore what is needed for a molecule to move from the researcher bench to the patients' bedside, presented from an industry perspective through the development program of cariprazine. Cariprazine is a relatively novel antipsychotic medication, approved for the treatment of schizophrenia, bipolar mania, bipolar depression, and major depression as an add-on. It is a D3-preferring D3-D2 partial agonist with the highest binding to the D3 receptors compared to all other antipsychotics. Based on the example of cariprazine, there are several key factors that are needed for a molecule to move from the researcher bench to the patients' bedside, such as targeting an unmet medical need, having a novel mechanism of action, and a smart implementation of development plans.
Collapse
Affiliation(s)
| | | | - György Németh
- Medical Division, Gedeon Richter Plc., 1103 Budapest, Hungary; (Á.B.)
| |
Collapse
|
11
|
Garisetti V, Dhanabalan AK, Dasararaju G. Discovery of potential TAAR1 agonist targeting neurological and psychiatric disorders: An in silico approach. Int J Biol Macromol 2024; 264:130528. [PMID: 38431013 DOI: 10.1016/j.ijbiomac.2024.130528] [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: 09/13/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
Trace amine-associated receptor 1 (TAAR1) is a G-protein-coupled receptor which is primarily expressed in the brain. It is activated by trace amines which play a role in regulating neurotransmitters like dopamine, serotonin and norepinephrine. TAAR1 agonists have potential applications in the treatment of neurological and psychiatric disorders, especially schizophrenia. In this study, we have used a structure-based virtual screening approach to identify potential TAAR1 agonist(s). We have modelled the structure of TAAR1 and predicted the binding pocket. Further, molecular docking of a few well-known antipsychotic drugs was carried out with TAAR1 model, which showed key interactions with the binding pocket. From screening a library of 5 million compounds from the Enamine REAL Database using structure-based virtual screening method, we shortlisted 12 compounds which showed good docking score, glide energy and interactions with the key residues. One lead compound (Z31378290) was finally selected. The lead compound showed promising binding affinity and stable interactions with TAAR1 during molecular dynamics simulations and demonstrated better van der Waals and binding energy than the known agonist, ulotaront. Our findings suggest that the lead compound may serve as a potential TAAR1 agonist, offering a promising avenue for the development of new therapies for neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Vasavi Garisetti
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai 600025, Tamil Nadu, India
| | - Anantha Krishnan Dhanabalan
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai 600025, Tamil Nadu, India
| | - Gayathri Dasararaju
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai 600025, Tamil Nadu, India.
| |
Collapse
|
12
|
Zhou Z, Zhang W, Zhao F, Sun Y, Wang N, Cheng J, Zhan P, Yang F, Sun JP, Liu X, Kang D. Structure-Based Design of Novel G-Protein-Coupled Receptor TAAR1 Agonists as Potential Antipsychotic Drug Candidates. J Med Chem 2024; 67:4234-4249. [PMID: 38416116 DOI: 10.1021/acs.jmedchem.4c00195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
The existing available antipsychotics have failed to manage the cognitive impairment of schizophrenia and induced a number of seriously undesirable effects. Trace amine-associated receptor 1 (TAAR1) has emerged as an ideal target for the design of antischizophrenia drugs, with the ability to mediate multiple psychological functions by sensing endogenous amine-containing metabolites without the side effects of catalepsy. In this work, a series of novel TAAR1 agonists were designed based on the structural analysis of the TAAR1 activation pocket. Among them, 6e displayed a potent TAAR1-Gs/Gq dual-pathway activation property, being different from that of the clinical drug candidate SEP-363856 with only TAAR1-Gs pathway activation. In rodent models, 6e significantly alleviated MK-801-induced schizophrenia-like cognitive phenotypes without inducing catalepsy. Furthermore, 6e·HCl exhibited favorable pharmacokinetic (T1/2 = 2.31 h, F = 39%) and safety properties. All these demonstrated that 6e·HCl may be used as a novel drug candidate for schizophrenia treatment.
Collapse
Affiliation(s)
- 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
| | - Weifeng Zhang
- Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China
| | - Fabao Zhao
- 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
| | - Yanying Sun
- 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
| | - Na Wang
- 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
| | - Jie Cheng
- Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China
| | - Peng Zhan
- 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
| | - Fan Yang
- Advanced Medical Research Institute and Meili Lake Translational Research Park, Shandong University, Jinan, Shandong 250012, China
| | - Jin-Peng Sun
- Advanced Medical Research Institute and Meili Lake Translational Research Park, 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
| | - 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
| |
Collapse
|
13
|
Dedic N, Wang L, Hajos-Korcsok E, Hecksher-Sørensen J, Roostalu U, Vickers SP, Wu S, Anacker C, Synan C, Jones PG, Milanovic S, Hopkins SC, Bristow LJ, Koblan KS. TAAR1 agonists improve glycemic control, reduce body weight and modulate neurocircuits governing energy balance and feeding. Mol Metab 2024; 80:101883. [PMID: 38237896 PMCID: PMC10839149 DOI: 10.1016/j.molmet.2024.101883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVE Metabolic Syndrome, which can be induced or exacerbated by current antipsychotic drugs (APDs), is highly prevalent in schizophrenia patients. Recent preclinical and clinical evidence suggest that agonists at trace amine-associated receptor 1 (TAAR1) have potential as a new treatment option for schizophrenia. Intriguingly, preclinical tudies have also identified TAAR1 as a novel regulator of metabolic control. Here we evaluated the effects of three TAAR1 agonists, including the clinical development candidate ulotaront, on body weight, metabolic parameters and modulation of neurocircuits implicated in homeostatic and hedonic feeding. METHODS Effects of TAAR1 agonists (ulotaront, RO5166017 and/or RO5263397) on body weight, food intake and/or metabolic parameters were investigated in rats fed a high-fat diet (HFD) and in a mouse model of diet-induced obesity (DIO). Body weight effects were also determined in a rat and mouse model of olanzapine-, and corticosterone-induced body weight gain, respectively. Glucose tolerance was assessed in lean and diabetic db/db mice and fasting plasma glucose and insulin examined in DIO mice. Effects on gastric emptying were evaluated in lean mice and rats. Drug-induced neurocircuit modulation was evaluated in mice using whole-brain imaging of c-fos protein expression. RESULTS TAAR1 agonists improved oral glucose tolerance by inhibiting gastric emptying. Sub-chronic administration of ulotaront in rats fed a HFD produced a dose-dependent reduction in body weight, food intake and liver triglycerides compared to vehicle controls. In addition, a more rapid reversal of olanzapine-induced weight gain and food intake was observed in HFD rats switched to ulotaront or RO5263397 treatment compared to those switched to vehicle. Chronic ulotaront administration also reduced body weight and improved glycemic control in DIO mice, and normalized corticosterone-induced body weight gain in mice. TAAR1 activation increased neuronal activity in discrete homeostatic and hedonic feeding centers located in the dorsal vagal complex and hypothalamus with concurrent activation of several limbic structures. CONCLUSION The current data demonstrate that TAAR1 agonists, as a class, not only lack APD-induced metabolic liabilities but can reduce body weight and improve glycemic control in rodent models. The underlying mechanisms likely include TAAR1-mediated peripheral effects on glucose homeostasis and gastric emptying as well as central regulation of energy balance and food intake.
Collapse
Affiliation(s)
- Nina Dedic
- Sumitomo Pharma America, Inc., Marlborough, MA, USA.
| | - Lien Wang
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | | | | | | | | | - Serena Wu
- Department of Psychiatry, New York State Psychiatric Institute (NYSPI), Columbia University, NY, New York City, USA
| | - Christoph Anacker
- Department of Psychiatry, New York State Psychiatric Institute (NYSPI), Columbia University, NY, New York City, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Zilberg G, Parpounas AK, Warren AL, Yang S, Wacker D. Molecular basis of human trace amine-associated receptor 1 activation. Nat Commun 2024; 15:108. [PMID: 38168118 PMCID: PMC10762035 DOI: 10.1038/s41467-023-44601-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
The human trace amine-associated receptor 1 (hTAAR1, hTA1) is a key regulator of monoaminergic neurotransmission and the actions of psychostimulants. Despite preclinical research demonstrating its tractability as a drug target, its molecular mechanisms of activation remain unclear. Moreover, poorly understood pharmacological differences between rodent and human TA1 complicate the translation of findings from preclinical disease models into novel pharmacotherapies. To elucidate hTA1's mechanisms on the molecular scale and investigate the underpinnings of its divergent pharmacology from rodent orthologs, we herein report the structure of the human TA1 receptor in complex with a Gαs heterotrimer. Our structure reveals shared structural elements with other TAARs, as well as with its closest monoaminergic orthologue, the serotonin receptor 5-HT4R. We further find that a single mutation dramatically shifts the selectivity of hTA1 towards that of its rodent orthologues, and report on the effects of substituting residues to those found in serotonin and dopamine receptors. Strikingly, we also discover that the atypical antipsychotic medication and pan-monoaminergic antagonist asenapine potently and efficaciously activates hTA1. Together our studies provide detailed insight into hTA1 structure and function, contrast its molecular pharmacology with that of related receptors, and uncover off-target activities of monoaminergic drugs at hTA1.
Collapse
Affiliation(s)
- Gregory Zilberg
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Alexandra K Parpounas
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Audrey L Warren
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Shifan Yang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Daniel Wacker
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
15
|
Tsukada H, Chen YL, Xiao G, Lennek L, Milanovic SM, Worden M, Polhamus DG, Chiu YY, Hopkins SC, Galluppi GR. A Phase I, Open-Label, Fixed Sequence Study to Investigate the Effect of Cytochrome P450 2D6 Inhibition on the Pharmacokinetics of Ulotaront in Healthy Subjects. Clin Pharmacokinet 2023; 62:1755-1763. [PMID: 37882999 PMCID: PMC10684410 DOI: 10.1007/s40262-023-01317-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND Ulotaront is a novel psychotropic agent with agonist activity at trace amine-associated receptor 1 (TAAR1) and 5-hydroxytryptamine type 1A (5-HT1A) receptors in phase III clinical development for the treatment of schizophrenia. OBJECTIVE This study aimed to investigate the effect of paroxetine, a strong cytochrome P450 (CYP) 2D6 inhibitor, on ulotaront pharmacokinetics (PK) in healthy volunteers. METHODS Subjects received a single oral dose of 25 mg ulotaront on Day 1 and an oral dose of 20 mg paroxetine once daily from Days 5 to 10 to achieve steady-state plasma paroxetine levels. On Day 11, subjects received another single oral dose of 25 mg ulotaront, with continued daily oral dosing of 20 mg paroxetine from Days 11 to 14. All 24 subjects were CYP2D6 normal metabolizers. RESULTS Coadministration of paroxetine increased ulotaront maximum observed plasma concentration (Cmax) and area under the plasma concentration-time curve from time zero to infinity (AUC∞) by 31% and 72%, respectively, and decreased ulotaront apparent clearance (CL/F) by approximately 42%. While coadministration of paroxetine increased AUC∞ of active but minor metabolite SEP-363854 by 32%, it had no effect on SEP-363854 Cmax, or on SEP-363854 to the ulotaront AUC from time zero to the last quantifiable concentration (AUClast) ratio. Based on the acceptable adverse event profile of ulotaront across previous phase II studies, the increase in ulotaront exposure is unlikely to be clinically meaningful. CONCLUSIONS Weak drug-drug interactions were observed between ulotaront and the strong CYP2D6 inhibitor paroxetine; however, dose adjustment as a precondition when ulotaront is coadministered with strong CYP2D6 inhibitors or administered to CYP2D6 poor metabolizers should not be necessary.
Collapse
Affiliation(s)
- Hironobu Tsukada
- Sumitomo Pharma America, 84 Waterford Dr., Marlborough, MA, 01752, USA
- Sumitomo Pharma Co., Ltd, Tokyo, Japan
| | - Yu-Luan Chen
- Sumitomo Pharma America, 84 Waterford Dr., Marlborough, MA, 01752, USA
| | - Guangqing Xiao
- Sumitomo Pharma America, 84 Waterford Dr., Marlborough, MA, 01752, USA
| | - Lisa Lennek
- Sumitomo Pharma America, 84 Waterford Dr., Marlborough, MA, 01752, USA
| | | | - MaryAlice Worden
- Sumitomo Pharma America, 84 Waterford Dr., Marlborough, MA, 01752, USA
| | | | - Yu-Yuan Chiu
- Sumitomo Pharma America, 84 Waterford Dr., Marlborough, MA, 01752, USA
| | - Seth C Hopkins
- Sumitomo Pharma America, 84 Waterford Dr., Marlborough, MA, 01752, USA
| | - Gerald R Galluppi
- Sumitomo Pharma America, 84 Waterford Dr., Marlborough, MA, 01752, USA.
| |
Collapse
|
16
|
Liu H, Zheng Y, Wang Y, Wang Y, He X, Xu P, Huang S, Yuan Q, Zhang X, Wang L, Jiang K, Chen H, Li Z, Liu W, Wang S, Xu HE, Xu F. Recognition of methamphetamine and other amines by trace amine receptor TAAR1. Nature 2023; 624:663-671. [PMID: 37935377 DOI: 10.1038/s41586-023-06775-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
Trace amine-associated receptor 1 (TAAR1), the founding member of a nine-member family of trace amine receptors, is responsible for recognizing a range of biogenic amines in the brain, including the endogenous β-phenylethylamine (β-PEA)1 as well as methamphetamine2, an abused substance that has posed a severe threat to human health and society3. Given its unique physiological role in the brain, TAAR1 is also an emerging target for a range of neurological disorders including schizophrenia, depression and drug addiction2,4,5. Here we report structures of human TAAR1-G-protein complexes bound to methamphetamine and β-PEA as well as complexes bound to RO5256390, a TAAR1-selective agonist, and SEP-363856, a clinical-stage dual agonist for TAAR1 and serotonin receptor 5-HT1AR (refs. 6,7). Together with systematic mutagenesis and functional studies, the structures reveal the molecular basis of methamphetamine recognition and underlying mechanisms of ligand selectivity and polypharmacology between TAAR1 and other monoamine receptors. We identify a lid-like extracellular loop 2 helix/loop structure and a hydrogen-bonding network in the ligand-binding pockets, which may contribute to the ligand recognition in TAAR1. These findings shed light on the ligand recognition mode and activation mechanism for TAAR1 and should guide the development of next-generation therapeutics for drug addiction and various neurological disorders.
Collapse
Affiliation(s)
- Heng Liu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - You Zheng
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yue Wang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yumeng Wang
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecule Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xinheng He
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Peiyu Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sijie Huang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qingning Yuan
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinyue Zhang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ling Wang
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kexin Jiang
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hong Chen
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai, China
- Shanghai Yuansi Standard Science and Technology Co., Ltd, Shanghai, China
| | - Zhen Li
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenbin Liu
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai, China.
- Shanghai Yuansi Standard Science and Technology Co., Ltd, Shanghai, China.
| | - Sheng Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecule Cell Science, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - H Eric Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Fei Xu
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
17
|
Robles A, Dinamarca-Villarroel L, Torres GE, Fierro A. Collective and Coordinated Conformational Changes Determine Agonism or Antagonism at the Human Trace Amine-Associated Receptor 1. ACS OMEGA 2023; 8:43051-43059. [PMID: 38024694 PMCID: PMC10652269 DOI: 10.1021/acsomega.3c06315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023]
Abstract
The human trace amine-associated receptor (hTAAR1), a G protein-coupled receptor, has been postulated as a new target in the treatment of neuropsychiatric conditions. The mechanism associated with activation or inactivation by agonists or antagonists in hTAAR1 and other GPCRs has not yet been fully elucidated. In this study, we combined computational methods including homology modeling, docking, and molecular dynamic simulations to reveal novel conformational changes associated with agonist and antagonist interactions in hTAAR1. Our findings suggest a differential cascade of coordinated movements based on the presence of either an agonist or antagonist and primarily involving the second extracellular loop, transmembrane domain 5, and the third intracellular domains of hTAAR1. Our study provides an opportunity to predict the effects on new ligands with agonistic or antagonistic activity at hTAAR1 based on the reported conformational changes.
Collapse
Affiliation(s)
- Agustín
I. Robles
- Departamento
de Química Orgánica, Escuela de Química, Facultad
de Química y de Farmacia, Pontificia
Universidad Católica de Chile, Santiago 7820436, Chile
- Department
of Molecular Pharmacology & Neuroscience, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60611-2001, United States
| | - Luis Dinamarca-Villarroel
- Departamento
de Química Orgánica, Escuela de Química, Facultad
de Química y de Farmacia, Pontificia
Universidad Católica de Chile, Santiago 7820436, Chile
- Department
of Molecular Pharmacology & Neuroscience, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60611-2001, United States
| | - Gonzalo E. Torres
- Department
of Molecular Pharmacology & Neuroscience, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60611-2001, United States
| | - Angélica Fierro
- Departamento
de Química Orgánica, Escuela de Química, Facultad
de Química y de Farmacia, Pontificia
Universidad Católica de Chile, Santiago 7820436, Chile
| |
Collapse
|
18
|
Vaganova AN, Maslennikova DD, Konstantinova VV, Kanov EV, Gainetdinov RR. The Expression of Trace Amine-Associated Receptors (TAARs) in Breast Cancer Is Coincident with the Expression of Neuroactive Ligand-Receptor Systems and Depends on Tumor Intrinsic Subtype. Biomolecules 2023; 13:1361. [PMID: 37759760 PMCID: PMC10526748 DOI: 10.3390/biom13091361] [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: 07/12/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Currently, the contribution of trace amine-associated receptors (TAARs) to breast cancer (BC) is recognized, but their associations with various pathological characteristics are not yet understood. There is accumulated transcriptomic data for BC tumors, which are represented in publicly accessible databases. We estimated TAARs' (including TAAR1, TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) associations with BC stage, grade, and molecular subtypes in these data and identified that the expression of all TAARs was associated with more unfavorable cancer subtypes, including basal-like and HER2-positive tumors. Also, the significant upregulation of all TAARs was demonstrated in circulating tumor cells compared to the metastatic lesions. Considering that co-expressed genes are more likely to be involved in the same biologic processes, we analyzed genes that are co-expressed with TAARs in BC. These gene sets were enriched with the genes of the olfactory transduction pathway and neuroactive ligand-receptor interaction participants. TAARs are co-expressed with G-protein-coupled receptors of monoamine neurotransmitters including dopamine, norepinephrine, and serotonin as well as with other neuroactive ligand-specific receptors. Since TAAR1 is able to modulate the activity of monoamine receptors that are involved in the regulation of BC growth, TAAR1 and potentially other TAARs may be regarded as prospective therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Anastasia N. Vaganova
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.); (E.V.K.)
- St. Petersburg University Hospital, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Daria D. Maslennikova
- Faculty of Biology, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Valeria V. Konstantinova
- St. Petersburg University Hospital, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Evgeny V. Kanov
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.); (E.V.K.)
- St. Petersburg University Hospital, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.); (E.V.K.)
- St. Petersburg University Hospital, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| |
Collapse
|
19
|
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
|
20
|
Isaacson SH, Goldstein M, Pahwa R, Singer C, Klos K, Pucci M, Zhang Y, Crandall D, Koblan KS, Navia B. Ulotaront, a Trace Amine-Associated Receptor 1/Serotonin 5-HT 1A Agonist, in Patients With Parkinson Disease Psychosis: A Pilot Study. Neurol Clin Pract 2023; 13:e200175. [PMID: 37273942 PMCID: PMC10238151 DOI: 10.1212/cpj.0000000000200175] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 04/04/2023] [Indexed: 06/06/2023]
Abstract
Background and Objectives Ulotaront (SEP-363856) is a trace amine-associated receptor 1 agonist with 5-HT1A receptor agonist activity currently in phase 3 clinical development for the treatment of schizophrenia. In this exploratory, flexibly dosed study, ulotaront was evaluated for the treatment of Parkinson disease psychosis (PDP). Methods Patients with PDP requiring antipsychotic therapy were randomized, double-blind to ulotaront (25, 50, or 75 mg/d) or placebo. Mixed Model for Repeated Measures was used to assess change from baseline in the Scale for the Assessment of Positive Symptoms for Parkinson Disease (SAPS-PD) at 6 weeks (primary end point). Results The efficacy analysis sample comprised 38 patients (ulotaront, n = 24; placebo, n = 14). SAPS-PD total scores were numerically reduced in ulotaront-treated vs placebo-treated patients from week 1 to week 6: Least squares mean (95% confidence interval) difference in change from baseline at week 6 was -1.1 (-6.5, 4.3, p = 0.681). PDP symptom complete remission (≥100% improvement [reduction] from baseline in SAPS-PD total score) was observed in 25% of ulotaront-treated vs 0% of placebo-treated patients. SAPS-PD and Neuropsychiatric Inventory hallucinations subscales were numerically reduced vs placebo, and SAPS-PD total scores were reduced in patients with greater cognitive impairment (baseline Mini-Mental State Examination [MMSE] scores ≤24). Ulotaront improved Scales for Outcomes in Parkinson Disease Sleep Scale - Daytime Sleepiness scores (p = 0.022). There was no worsening of Unified Parkinson Disease Rating Scale Part III motor score, MMSE, or vital signs. Adverse events (≥10%) with ulotaront vs placebo included hallucinations (24% vs 14%), confusional state (20% vs 14%), dizziness (16% vs 7%), nausea (12% vs 7%), and falls (12% vs 21%). Discussion In this exploratory pilot study, ulotaront may decrease PDP symptoms without worsening motor function, particularly in patients with cognitive impairment. Trial Registration Information ClinicalTrials.gov identifier: NCT02969369; submitted: November 17, 2016; study start date: December 31, 2016. Classification of Evidence This Class II study was an exploratory pilot study that was underpowered to detect a statistically significant difference between ulotaront and placebo in the treatment of patients with Parkinson disease psychosis without worsening motor function.
Collapse
Affiliation(s)
- Stuart H Isaacson
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Mark Goldstein
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Rajesh Pahwa
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Carlos Singer
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Kevin Klos
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Michael Pucci
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Yi Zhang
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - David Crandall
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Kenneth S Koblan
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| | - Bradford Navia
- Parkinson's Disease and Movement Disorders Center of Boca Raton (SHI), FL; JEM Research Institute (MG), Lake Worth, FL; University of Kansas (RP), Kansas City; University of Miami Health System (CS), FL; The Movement Disorder Clinic of Oklahoma (KK), Tulsa; The Lockwood Group (MP), Stamford, CT; and Sunovion Pharmaceuticals Inc. (YZ, DC, KSK, BN), Marlborough, MA
| |
Collapse
|
21
|
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
|
22
|
Meyer JM, Correll CU. Increased Metabolic Potential, Efficacy, and Safety of Emerging Treatments in Schizophrenia. CNS Drugs 2023; 37:545-570. [PMID: 37470979 PMCID: PMC10374807 DOI: 10.1007/s40263-023-01022-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
Patients with schizophrenia experience a broad range of detrimental health outcomes resulting from illness severity, heterogeneity of disease, lifestyle behaviors, and adverse effects of antipsychotics. Because of these various factors, patients with schizophrenia have a much higher risk of cardiometabolic abnormalities than people without psychiatric illness. Although exposure to many antipsychotics increases cardiometabolic risk factors, mortality is higher in patients who are not treated versus those who are treated with antipsychotics. This indicates both direct and indirect benefits of adequately treated illness, as well as the need for beneficial medications that result in fewer cardiometabolic risk factors and comorbidities. The aim of the current narrative review was to outline the association between cardiometabolic dysfunction and schizophrenia, as well as discuss the confluence of factors that increase cardiometabolic risk in this patient population. An increased understanding of the pathophysiology of schizophrenia has guided discovery of novel treatments that do not directly target dopamine and that not only do not add, but may potentially minimize relevant cardiometabolic burden for these patients. Key discoveries that have advanced the understanding of the neural circuitry and pathophysiology of schizophrenia now provide possible pathways toward the development of new and effective treatments that may mitigate the risk of metabolic dysfunction in these patients. Novel targets and preclinical and clinical data on emerging treatments, such as glycine transport inhibitors, nicotinic and muscarinic receptor agonists, and trace amine-associated receptor-1 agonists, offer promise toward relevant therapeutic advancements. Numerous areas of investigation currently exist with the potential to considerably progress our knowledge and treatment of schizophrenia.
Collapse
Affiliation(s)
- Jonathan M Meyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
| | - Christoph U Correll
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
- Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
23
|
Barnes DA, Hoener MC, Moore CS, Berry MD. TAAR1 Regulates Purinergic-induced TNF Secretion from Peripheral, But Not CNS-resident, Macrophages. J Neuroimmune Pharmacol 2023; 18:100-111. [PMID: 36380156 DOI: 10.1007/s11481-022-10053-8] [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: 06/20/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Abstract
Trace amine-associated receptor 1 (TAAR1) is an established neuroregulatory G protein-coupled receptor with recent studies suggesting additional functions related to immunomodulation. Our lab has previously investigated TAAR1 expression within cells of the innate immune system and herein we aim to further elucidate TAAR1 function in both peripherally-derived and CNS-resident macrophages. The selective TAAR1 agonist RO5256390 was used in combination with common damage associated molecular patterns (ATP and ADP) to observe the effect of TAAR1 agonism on modulating cytokine secretion and metabolic profiles. In mouse bone-marrow derived macrophages, TAAR1 agonism inhibited TNF secretion following ATP stimulation, which appeared to be downstream of an associated pro-inflammatory shift in metabolic profile and transcriptional regulation of TNF synthesis. In contrast, TAAR1 agonism had no effect on ADP-induced TNF and IL-6 secretion in mouse microglia in either the presence or absence of astrocytes. In summary, we report a novel interaction between TAAR1 and purinergic signaling in peripherally-derived, but not CNS-resident, macrophages. These findings provide the first evidence of trace aminergic and purinergic crosstalk, and support the potential for TAAR1 as a novel therapeutic target in inflammatory disorders.
Collapse
Affiliation(s)
- David A Barnes
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John's, NL, A1B 3X9, Canada
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Marius C Hoener
- Neuroscience and Rare Diseases Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, 4070, Basel, Switzerland
| | - Craig S Moore
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Mark D Berry
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John's, NL, A1B 3X9, Canada.
| |
Collapse
|
24
|
Le GH, Gillissie ES, Rhee TG, Cao B, Alnefeesi Y, Guo Z, Di Vincenzo JD, Jawad MY, March AM, Ramachandra R, Lui LMW, McIntyre RS. Efficacy, safety, and tolerability of ulotaront (SEP-363856, a trace amine-associated receptor 1 agonist) for the treatment of schizophrenia and other mental disorders with similar pathophysiology: a systematic review of preclinical and clinical trials. Expert Opin Investig Drugs 2023:1-15. [PMID: 37096491 DOI: 10.1080/13543784.2023.2206559] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
INTRODUCTION Schizophrenia is a mental illness that can disrupt emotions, perceptions, cognition, and reduce quality of life. The classical approach to treat schizophrenia uses typical and atypical antipsychotics; however, limitations include low efficacy in mitigating negative symptoms and cognitive dysfunctions, and a range of adverse effects. Evidence has accumulated on trace amine-associated receptor 1 (TAAR1) as a novel therapeutic target for treating schizophrenia. This systematic review investigates the available evidence on a TAAR1 agonist, ulotaront, as a treatment for schizophrenia. METHODS A systematic search was conducted on PubMed/MEDLINE, and Ovid databases for English-published articles from inception to December 18, 2022. Literature focusing on the association between ulotaront and schizophrenia were evaluated based on an inclusion/exclusion criterion. Selected studies were assessed for risk of bias, using Cochrane Collaboration tool, and summarized in a table to generate discussion topics. RESULTS Three clinical, two comparative, and five preclinical studies examining ulotaront's pharmacology, tolerability and safety, and/or efficacy were identified. Results indicate that ulotaront has a differing adverse effects profile from other antipsychotics, may mitigate metabolic-related adverse effects commonly associated with antipsychotics, and may be effective for treating positive and negative symptoms. CONCLUSIONS Findings from available literature present ulotaront as a potential and promising alternative treatment method for schizophrenia. Despite this, our results were limited due to lack of clinical trials on ulotaront's long-term efficacy and mechanisms of action. Future research should focus on these limitations to elucidate ulotaront's efficacy and safety for the treatment of schizophrenia and other mental disorders with similar pathophysiology.
Collapse
Affiliation(s)
- Gia Han Le
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Emily S Gillissie
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Taeho Greg Rhee
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
- VA New England Mental Illness, Research, Education and Clinical Center (MIRECC), VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Public Health Sciences, School of Medicine, University of Connecticut, Farmington, CT, USA
| | - Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, 400715, P. R. China
| | - Yazen Alnefeesi
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Ziji Guo
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Joshua D Di Vincenzo
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Muhammad Youshay Jawad
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Andrew M March
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Ranuk Ramachandra
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Leanna M W Lui
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| |
Collapse
|
25
|
Chen YL, Tsukada H, Milanovic S, Shi L, Li Y, Mao Y, Koblan KS, Galluppi GR. Comparative Bioequivalence of Tablet and Capsule Formulations of Ulotaront and the Effect of Food on the Pharmacokinetics of the Tablet Form in Humans. Neurol Ther 2023; 12:815-832. [PMID: 36932300 DOI: 10.1007/s40120-023-00459-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
INTRODUCTION Ulotaront (SEP-363856), a dual trace animeassociated receptor 1 (TAAR1) and 5-HT1A receptor agonist, is in phase 3 clinical development for the treatment of schizophrenia. This study evaluated the comparative bioequivalence (BE) between tablet and capsule formulations of ulotaront and the food effect (FE) on pharmacokinetics (PK) of tablet form in healthy adult human subjects. METHODS The BE study applied an open-label two-period crossover design in 24 healthy volunteers. Subjects were randomly assigned (1:1) to dosing sequence AB or BA (A, 25 mg ulotaront tablet; B, 25 mg ulotaront capsule). The FE study also used an open-label randomized two-period crossover design in 20 healthy volunteers. Subjects were fasted overnight then randomly assigned (1:1) to dosing sequence AB or BA (A, fasted condition; B, fed condition). Dosing periods were separated by 1 week for both studies. Serial plasma samples from each period were collected and analyzed by LC-MS/MS. PK parameters were calculated using Phoenix WinNonlin® software. RESULTS For the BE study, geometric mean ulotaront Cmax values were 93.28 and 86.98 ng/mL for tablet and capsule, respectively. Cmax ratio was 107.25% (90% CI 101.84-112.94%). Geometric mean ulotaront area under the plasma concentration-time curve from time 0 to infinity (AUC0-∞) values were 868.8 and 829.3 ng·h/mL for tablet and capsule, respectively. AUC0-∞ ratio was 104.76% (90% CI 100.68109.01%). For the FE study, geometric mean ulotaront Cmax was 157.89 and 157.95 ng/mL under fed and fasted conditions, respectively. Geometric mean ratio of Cmax was 99.96% (90% CI 94.48-105.77%). Geometric mean ulotaront AUC0-∞ was 1584.2 ng·h/mL fed and 1589.2 ng·h/mL fasted. Geometric mean ratio for AUC0-∞ was 99.69% (90% CI 95.02-104.58%). There was a delay in tmax (median difference 1.47 h) in the fed condition. CONCLUSIONS The results showed geometric mean ratios and 90% CIs for both Cmax and AUC0-∞ for ulotaront were well within typical bioequivalence criteria of 80-125% for both the BE and FE studies, thereby confirming the bioequivalence of the two dosage forms and no significant food effect.
Collapse
Affiliation(s)
- Yu-Luan Chen
- Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA.
| | - Hironobu Tsukada
- Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Snezana Milanovic
- Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Lei Shi
- Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Yan Li
- Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Yongcai Mao
- Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Kenneth S Koblan
- Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| | - Gerald R Galluppi
- Sunovion Pharmaceuticals Inc., 84 Waterford Drive, Marlborough, MA, 01752, USA
| |
Collapse
|
26
|
Recent Advances in Psychopharmacology: From Bench to Bedside Novel Trends in Schizophrenia. J Pers Med 2023; 13:jpm13030411. [PMID: 36983593 PMCID: PMC10058851 DOI: 10.3390/jpm13030411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Research in the field of psychopharmacology is ongoing to develop novel compounds which can revolutionize the treatment of psychiatric disorders. The concept of bench-to-bedside is a tedious process, transforming the initial research performed in the laboratories into novel treatment options. Schizophrenia (SCZ) is a chronic psychiatric illness with significant morbidity and mortality. SCZ not only presents with psychotic symptoms including hallucinations and delusions but also with negative and cognitive symptoms. The negative symptoms include the diminished ability to express emotions, loss of pleasure, and motivation with minimal social interactions. Conventional antipsychotics primarily target positive symptoms with minimal therapeutic benefits for negative and cognitive symptoms along with metabolic side effects. Researchers have explored novel targets to develop new compounds to overcome the above limitations. The glutamatergic system has provided new hope in treating schizophrenia by targeting negative and cognitive symptoms. Other receptor modulators, including serotonergic, phosphodiesterase, trans-amine-associated receptors, etc., are novel targets for developing new compounds. Future research is required in this field to explore novel compounds and establish their efficacy and safety for the treatment of schizophrenia. Last but not least, pharmacogenomics has effectively utilized genetic information to develop novel compounds by minimizing the risk of failure of the clinical trials and enhancing efficacy and safety.
Collapse
|
27
|
Trace amine-associated receptor 1 (TAAR1) agonism as a new treatment strategy for schizophrenia and related disorders. Trends Neurosci 2023; 46:60-74. [PMID: 36369028 DOI: 10.1016/j.tins.2022.10.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/08/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
Abstract
Schizophrenia remains a major health burden, highlighting the need for new treatment approaches. We consider the potential for targeting the trace amine (TA) system. We first review genetic, preclinical, and clinical evidence for the role of TAs in the aetiopathology of schizophrenia. We then consider how the localisation and function of the trace amine-associated receptor 1 (TAAR1) position it to modulate key brain circuits for the disorder. Studies in rodents using Taar1 knockout (TAAR1-KO) and overexpression models show that TAAR1 agonism inhibits midbrain dopaminergic and serotonergic activity, and enhances prefrontal glutamatergic function. TAAR1 agonists also reduce hyperactivity, attenuate prepulse inhibition (PPI) deficits and social withdrawal, and improve cognitive measures in animal models. Finally, we consider findings from clinical trials of TAAR1 agonists and how this approach may address psychotic and negative symptoms, tolerability issues, and other unmet needs in the treatment of schizophrenia.
Collapse
|
28
|
Ceskova E. Has the utilization of serotonin receptor antagonism made an impact on schizophrenia treatment? Expert Opin Pharmacother 2022; 23:1865-1868. [PMID: 36250483 DOI: 10.1080/14656566.2022.2137403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Eva Ceskova
- Department of Neurology and Psychiatry, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
29
|
TAAR1 dependent and independent actions of the potential antipsychotic and dual TAAR1/5-HT 1A receptor agonist SEP-383856. Neuropsychopharmacology 2022; 47:2319-2329. [PMID: 36100653 PMCID: PMC9630386 DOI: 10.1038/s41386-022-01421-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 07/04/2022] [Accepted: 07/30/2022] [Indexed: 11/09/2022]
Abstract
SEP-383856 (SEP-856) is a novel antipsychotic under clinical development. It displays a unique pattern of receptor interaction, with only weak (partial agonist) activity at dopamine D2 receptors, yet more potent agonist activity at the trace amine associated receptor (TAAR1) and 5-hydroxytryptamine 1 A receptor (5-HT1A). Nonetheless, these observations await independent confirmation and more detailed characterization of the in vitro and in vivo actions of SEP-856 at TAAR1 and 5-HT1A receptors would be instructive. Herein, we employed luminescence complementation technology in heterologous live cell systems, confocal microscopy, voltage clamp electrophysiology, behavioral readouts and TAAR1 knockout (KO) mice to study SEP-856 in further detail. We provide evidence for the ability of SEP-856 to activate TAAR1 at the surface plasma membrane, and show that this interaction results in Gαs recruitment (pEC50: 6.08 ± 0.22 EMAX: 96.41% ± 15.26) and by extension, to G-protein inwardly rectifying potassium (GIRK) channel activation. Using TAAR1-KO mice, we find TAAR1 to be indispensable for SEP-856 control of body temperature, baseline locomotion reduction and for "antipsychotic-like" efficacy as characterized by a reversal of dizocilipine (MK-801) mediated disruption of pre-pulse inhibition. Conversely, the inhibition by SEP-856 of MK-801 induced locomotion was unaffected in TAAR1 KO mice. SEP-856 behaved as a low-potency, partial agonist at the 5-HT1A receptor, while it partially inhibited recruitment of D2 receptor-coupled Gα and GIRK by DA and acted as a weak partial agonist with low potency at the same receptor when applied alone. Our findings corroborate and extend previous observations on the molecular substrates engaged by this unique, dual TAAR1/5-HT1A receptor agonist and potential antipsychotic that could prove to have major advantages in the treatment of schizophrenia and other psychotic disorders.
Collapse
|
30
|
Stereoselectivity in the Membrane Transport of Phenylethylamine Derivatives by Human Monoamine Transporters and Organic Cation Transporters 1, 2, and 3. Biomolecules 2022; 12:biom12101507. [PMID: 36291716 PMCID: PMC9599461 DOI: 10.3390/biom12101507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/24/2022] Open
Abstract
Stereoselectivity is well known and very pronounced in drug metabolism and receptor binding. However, much less is known about stereoselectivity in drug membrane transport. Here, we characterized the stereoselective cell uptake of chiral phenylethylamine derivatives by human monoamine transporters (NET, DAT, and SERT) and organic cation transporters (OCT1, OCT2, and OCT3). Stereoselectivity differed extensively between closely related transporters. High-affinity monoamine transporters (MATs) showed up to 2.4-fold stereoselective uptake of norepinephrine and epinephrine as well as of numerous analogs. While NET and DAT preferentially transported (S)-norepinephrine, SERT preferred the (R)-enantiomer. In contrast, NET and DAT showed higher transport for (R)-epinephrine and SERT for (S)-epinephrine. Generally, MAT stereoselectivity was lower than expected from their high affinity to several catecholamines and from the high stereoselectivity of some inhibitors used as antidepressants. Additionally, the OCTs differed strongly in their stereoselectivity. While OCT1 showed almost no stereoselective uptake, OCT2 was characterized by a roughly 2-fold preference for most (R)-enantiomers of the phenylethylamines. In contrast, OCT3 transported norphenylephrine and phenylephrine with 3.9-fold and 3.3-fold preference for their (R)-enantiomers, respectively, while the para-hydroxylated octopamine and synephrine showed no stereoselective OCT3 transport. Altogether, our data demonstrate that stereoselectivity is highly transporter-to-substrate specific and highly diverse even between homologous transporters.
Collapse
|
31
|
Krasavin M, Peshkov AA, Lukin A, Komarova K, Vinogradova L, Smirnova D, Kanov EV, Kuvarzin SR, Murtazina RZ, Efimova EV, Gureev M, Onokhin K, Zakharov K, Gainetdinov RR. Discovery and In Vivo Efficacy of Trace Amine-Associated Receptor 1 (TAAR1) Agonist 4-(2-Aminoethyl)- N-(3,5-dimethylphenyl)piperidine-1-carboxamide Hydrochloride (AP163) for the Treatment of Psychotic Disorders. Int J Mol Sci 2022; 23:ijms231911579. [PMID: 36232878 PMCID: PMC9569940 DOI: 10.3390/ijms231911579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022] Open
Abstract
Starting from a screening hit, a set of analogs was synthesized based on a 4-(2-aminoethyl)piperidine core not associated previously with trace amine-associated receptor 1 (TAAR1) modulation in the literature. Several structure–activity relationship generalizations have been drawn from the observed data, some of which were corroborated by molecular modeling against the crystal structure of TAAR1. The four most active compounds (EC50 for TAAR1 agonistic activity ranging from 0.033 to 0.112 μM) were nominated for evaluation in vivo. The dopamine transporter knockout (DAT-KO) rat model of dopamine-dependent hyperlocomotion was used to evaluate compounds’ efficacy in vivo. Out of four compounds, only one compound (AP163) displayed a statistically significant and dose-dependent reduction in hyperlocomotion in DAT-KO rats. As such, compound AP163 represents a viable lead for further preclinical characterization as a potential novel treatment option for disorders associated with increased dopaminergic function, such as schizophrenia.
Collapse
Affiliation(s)
- Mikhail Krasavin
- Department of Medicinal Chemistry, Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Correspondence: (M.K.); (R.R.G.)
| | - Anatoly A. Peshkov
- Department of Medicinal Chemistry, Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Alexey Lukin
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia
| | - Kristina Komarova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia
| | - Lyubov Vinogradova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Moscow 119454, Russia
| | - Daria Smirnova
- Department of Medicinal Chemistry, Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Evgeny V. Kanov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Savelii R. Kuvarzin
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Ramilya Z. Murtazina
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Evgeniya V. Efimova
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Maxim Gureev
- Center of Bio- and Chemoinformatics, Sechenov First Moscow State Medical University, Moscow 119435, Russia
| | - Kirill Onokhin
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Konstantin Zakharov
- Accellena Research and Development Inc., 88A Sredniy pr. V.O., Saint Petersburg 199106, Russia
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Correspondence: (M.K.); (R.R.G.)
| |
Collapse
|
32
|
Vaganova AN, Katolikova NV, Murtazina RZ, Kuvarzin SR, Gainetdinov RR. Public Transcriptomic Data Meta-Analysis Demonstrates TAAR6 Expression in the Mental Disorder-Related Brain Areas in Human and Mouse Brain. Biomolecules 2022; 12:biom12091259. [PMID: 36139098 PMCID: PMC9496192 DOI: 10.3390/biom12091259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/24/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
G protein-coupled trace amine-associated receptors (TAAR) recognize different classes of amine compounds, including trace amines or other exogenous and endogenous molecules. Yet, most members of the TAAR family (TAAR2-TAAR9) are considered olfactory receptors involved in sensing innate odors. In this study, TAAR6 mRNA expression was evaluated in the brain transcriptomic datasets available in the GEO, Allen Brain Atlas, and GTEx databases. Transcriptomic data analysis demonstrated ubiquitous weak TAAR6 mRNA expression in the brain, especially in the prefrontal cortex and nucleus accumbens. RNA sequencing of isolated cells from the nucleus accumbens showed that the expression of TAAR6 in some cell populations may be more pronounced than in whole-tissue samples. Curiously, in D1 and D2 medium spiny neurons of the nucleus accumbens, TAAR6 expression was co-regulated with genes involved in G protein-coupled receptor signaling. However, in cholinergic interneurons of the nucleus accumbens, TAAR6 expression was not associated with the activation of any specific biological process. Finally, TAAR6 expression in the mouse prefrontal cortex was validated experimentally by RT-PCR analysis. These data demonstrated that TAAR6 is expressed at low levels in the human and mouse brain, particularly in limbic structures involved in the pathogenesis of mental disorders, and thus might represent a new pharmacotherapeutic target.
Collapse
Affiliation(s)
- Anastasia N. Vaganova
- Institute of Translational Biomedicine, Saint Petersburg State University, Universitetskaya nab. 7/9, 199034 Saint Petersburg, Russia
| | - Nataliia V. Katolikova
- Institute of Translational Biomedicine, Saint Petersburg State University, Universitetskaya nab. 7/9, 199034 Saint Petersburg, Russia
| | - Ramilya Z. Murtazina
- Institute of Translational Biomedicine, Saint Petersburg State University, Universitetskaya nab. 7/9, 199034 Saint Petersburg, Russia
| | - Savelii R. Kuvarzin
- Institute of Translational Biomedicine, Saint Petersburg State University, Universitetskaya nab. 7/9, 199034 Saint Petersburg, Russia
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Universitetskaya nab. 7/9, 199034 Saint Petersburg, Russia
- St. Petersburg University Hospital, Saint Petersburg State University, Universitetskaya nab. 7/9, 199034 Saint Petersburg, Russia
- Correspondence:
| |
Collapse
|
33
|
Risk of Drug-induced Movement Disorders with Newer Antipsychotic Agents. Tremor Other Hyperkinet Mov (N Y) 2022; 12:19. [PMID: 35836971 PMCID: PMC9187243 DOI: 10.5334/tohm.695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/27/2022] [Indexed: 01/19/2023] Open
Abstract
Background: The last decade has seen development of numerous novel antipsychotic drugs with unique mechanisms including long-acting formulations for clinical use. A comparative assessment of these new drugs with each other and previous antipsychotics have not been performed with regards to risk for drug-induced movement disorders (DIMD). Methods: Medline was searched from January 2010 to February 2022 for primary research articles and review articles in English using the search terms “extrapyramidal” and “tardive” with individual drug names of novel antipsychotics. Results: We identified articles describing the risk of DIMD with 6 novel antipsychotics, 4 novel formulations, and 3 experimental antipsychotics. Both short- and long-term data generally showed comparable to lower risk of DIMD with novel antipsychotics and recent long-acting formulations compared to previously marketed antipsychotics. Discussion: Several novel antipsychotics, particularly lumateperone and pimavanserin, show promise in being able to treat psychosis while reducing the risk of DIMD. Long-acting paliperidone may reduce risk of DIMD while other long-acting injectable formulations of SGA have similar risk of DIMD compared to oral formulations. New drug targets for treating psychosis without dopamine blockade also show promise.
Collapse
|
34
|
Grinevich VP, Zakirov AN, Berseneva UV, Gerasimova EV, Gainetdinov RR, Budygin EA. Applying a Fast-Scan Cyclic Voltammetry to Explore Dopamine Dynamics in Animal Models of Neuropsychiatric Disorders. Cells 2022; 11:cells11091533. [PMID: 35563838 PMCID: PMC9100021 DOI: 10.3390/cells11091533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 02/07/2023] Open
Abstract
Progress in the development of technologies for the real-time monitoring of neurotransmitter dynamics has provided researchers with effective tools for the exploration of etiology and molecular mechanisms of neuropsychiatric disorders. One of these powerful tools is fast-scan cyclic voltammetry (FSCV), a technique which has progressively been used in animal models of diverse pathological conditions associated with alterations in dopamine transmission. Indeed, for several decades FSCV studies have provided substantial insights into our understanding of the role of abnormal dopaminergic transmission in pathogenetic mechanisms of drug and alcohol addiction, Parkinson’s disease, schizophrenia, etc. Here we review the applications of FSCV to research neuropsychiatric disorders with particular attention to recent technological advances.
Collapse
Affiliation(s)
- Vladimir P. Grinevich
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
| | - Amir N. Zakirov
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
| | - Uliana V. Berseneva
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
| | - Elena V. Gerasimova
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
| | - Raul R. Gainetdinov
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
- Institute of Translational Biomedicine and St. Petersburg State University Hospital, St. Petersburg State University, Universitetskaya Emb. 7-9, St. Petersburg 199034, Russia
| | - Evgeny A. Budygin
- Department of Neurobiology, Sirius University, 1 Olympic Ave., Sirius, Sochi 353340, Russia; (V.P.G.); (A.N.Z.); (U.V.B.); (E.V.G.); (R.R.G.)
- Correspondence:
| |
Collapse
|
35
|
Spark DL, Fornito A, Langmead CJ, Stewart GD. Beyond antipsychotics: a twenty-first century update for preclinical development of schizophrenia therapeutics. Transl Psychiatry 2022; 12:147. [PMID: 35393394 PMCID: PMC8991275 DOI: 10.1038/s41398-022-01904-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/02/2022] [Accepted: 03/15/2022] [Indexed: 11/15/2022] Open
Abstract
Despite 50+ years of drug discovery, current antipsychotics have limited efficacy against negative and cognitive symptoms of schizophrenia, and are ineffective-with the exception of clozapine-against any symptom domain for patients who are treatment resistant. Novel therapeutics with diverse non-dopamine D2 receptor targets have been explored extensively in clinical trials, yet often fail due to a lack of efficacy despite showing promise in preclinical development. This lack of translation between preclinical and clinical efficacy suggests a systematic failure in current methods that determine efficacy in preclinical rodent models. In this review, we critically evaluate rodent models and behavioural tests used to determine preclinical efficacy, and look to clinical research to provide a roadmap for developing improved translational measures. We highlight the dependence of preclinical models and tests on dopamine-centric theories of dysfunction and how this has contributed towards a self-reinforcing loop away from clinically meaningful predictions of efficacy. We review recent clinical findings of distinct dopamine-mediated dysfunction of corticostriatal circuits in patients with treatment-resistant vs. non-treatment-resistant schizophrenia and suggest criteria for establishing rodent models to reflect such differences, with a focus on objective, translational measures. Finally, we review current schizophrenia drug discovery and propose a framework where preclinical models are validated against objective, clinically informed measures and preclinical tests of efficacy map onto those used clinically.
Collapse
Affiliation(s)
- Daisy L Spark
- Drug Discovery Biology, Neuroscience & Mental Health Therapeutic Program Area, and Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, Monash Biomedical Imaging, and School of Psychological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Christopher J Langmead
- Drug Discovery Biology, Neuroscience & Mental Health Therapeutic Program Area, and Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| | - Gregory D Stewart
- Drug Discovery Biology, Neuroscience & Mental Health Therapeutic Program Area, and Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|