1
|
Bushramol S, Ravikumar C. Vibrational spectroscopic interpretation, solvent effect and molecular docking studies of TAAR1 partial agonist RO5263397. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 325:125137. [PMID: 39303337 DOI: 10.1016/j.saa.2024.125137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Density functional theory studies of TAAR1 (trace amine associated receptor 1) partial agonist RO5263397 carried out with precise and detailed spectroscopic investigation as well as validated experimentally. FT-IR, confocal Raman and UV-visible spectroscopic techniques were used to characterize the compound and corresponding theoretical calculations were carried out using DFT/B3LYP method with 6-311++G (d,p) basis set. Estimated and observed vibrational wavenumbers of the compound were assigned. UV-visible spectrum and FMOs (frontier molecular orbital) analysis reveals that the polarity affects the molecular reactivity and stability of the compound. Donor - acceptor interaction and second order perturbation energy have been explained using natural bond orbital analysis clarify the presence hydrogen bonds in the system. ELF and LOL studies visualises the localized and delocalized electrons in the title compound. RDG analysis evidences the various interactions present in the monomer and dimer of RO5263397. The structural importance of the compound were clearly examined using NMR spectral analysis. The existence of hydrogen bonding is validated by reactive site findings from Mulliken atomic charge distribution and molecule electrostatic potential surface studies. Information about distinct drug-receptor interactions obtained from molecular docking investigation offers the path of further study of molecular activity in various drug-receptor mechanism.
Collapse
Affiliation(s)
- S Bushramol
- Nanotechnology and Advanced Materials Research Centre, Department of Physics, CMS College, Kottayam 686 001, Kerala, India
| | - C Ravikumar
- Nanotechnology and Advanced Materials Research Centre, Department of Physics, CMS College, Kottayam 686 001, Kerala, India.
| |
Collapse
|
2
|
Kitaichi M, Kato T, Oki H, Tatara A, Kawada T, Miyazaki K, Ishikawa C, Kaneda K, Shimizu I. DSP-6745, a novel 5-hydroxytryptamine modulator with rapid antidepressant, anxiolytic, antipsychotic and procognitive effects. Psychopharmacology (Berl) 2024; 241:2223-2239. [PMID: 38856765 DOI: 10.1007/s00213-024-06629-2] [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: 01/24/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Current treatment of major depressive disorder is facing challenges, including a low remission rate, late onset of efficacy, and worsening severity due to comorbid symptoms such as psychosis and cognitive dysfunction. Serotonin (5-HT) neurotransmission is involved in a wide variety of psychiatric diseases and its potential as a drug target continues to attract attention. OBJECTIVES The present study elucidates the effects of a novel 5-HT modulator, DSP-6745, on depression and its comorbid symptoms. RESULTS In vitro radioligand binding and functional assays showed that DSP-6745 is a potent inhibitor of 5-HT transporter and 5-HT2A, 5-HT2C, and 5-HT7 receptors. In vivo, DSP-6745 (6.4 and 19.1 mg/kg as free base, p.o.) increased the release of not only 5-HT, norepinephrine, and dopamine, but also glutamate in the medial prefrontal cortex. The results of in vivo mouse phenotypic screening by SmartCube® suggested that DSP-6745 has a behavioral signature combined with antidepressant-, anxiolytic-, and antipsychotic-like signals. A single oral dose of DSP-6745 (6.4 and 19.1 mg/kg) showed rapid antidepressant-like efficacy in the rat forced swim test, even at 24 h post-dosing, and anxiolytic activity in the rat social interaction test. Moreover, DSP-6745 (12.7 mg/kg, p.o.) led to an improvement in the apomorphine-induced prepulse inhibition deficit in rats. In the marmoset object retrieval with detour task, which is used to assess cognitive functions such as attention and behavioral inhibition, DSP-6745 (7.8 mg/kg, p.o.) enhanced cognition. CONCLUSIONS These data suggest that DSP-6745 is a multimodal 5-HT receptor antagonist and a 5-HT transporter inhibitor and has the potential to be a rapid acting antidepressant with efficacies in mitigating the comorbid symptoms of depression.
Collapse
Affiliation(s)
- Maiko Kitaichi
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Taro Kato
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan.
| | - Hitomi Oki
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Ayaka Tatara
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Takuya Kawada
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Kenji Miyazaki
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Chihiro Ishikawa
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Isao Shimizu
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| |
Collapse
|
3
|
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
|
4
|
Paramonova P, Lebedev R, Sokolov A, Dar'in D, Kanov E, Murtazina R, Gainetdinov R, Kalinin S, Bakulina O. Azide-based in situ preparation of fused heterocyclic imines and their multicomponent reactions. Org Biomol Chem 2024; 22:8328-8336. [PMID: 39315616 DOI: 10.1039/d4ob01321b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Structurally diverse pyrroles, indoles and imidazoles bearing an N-ω-azidoalkyl moiety and an aldehyde or ketone function were prepared and successfully introduced into in situ imine generation via the intramolecular Staudinger/aza-Wittig tandem reaction. Reduction of the generated imines led to medicinally relevant nitrogen-containing fused heterocycles such as tetrahydropyrrolo[1,2-a]pyrazines and diazepines. Rare 8-membered hexahydropyrrolo[1,2-a][1,4]diazocine and 9-membered dihydro-4,8-(metheno)pyrrolo[1,2-a][1,4]diazacycloundecine were also synthesized. In addition, several one-pot transformations involving cyclic anhydrides or isocyanides (Castagnoli-Cushman, Ugi and azido-Ugi reactions) were added to the Staudinger/aza-Wittig sequence to afford novel fused polyheterocyclic delta-lactams, cyclic bisamides and tetrazoles in a multicomponent fashion. The synthesized compounds were profiled against human Trace Amine-Associated Receptor 1 (hTAAR1), and the best performing compound showed low nanomolar agonistic activity with an EC50 of 0.025 μM.
Collapse
Affiliation(s)
- Polina Paramonova
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia.
| | - Rodion Lebedev
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia.
| | - Alexander Sokolov
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia.
| | - Dmitry Dar'in
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia.
- Saint Petersburg Research Institute of Phthisiopulmonology, Saint Petersburg, 191036, Russia
| | - Evgeny Kanov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Saint Petersburg University Hospital, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| | - Ramilya Murtazina
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Raul Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Saint Petersburg University Hospital, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| | - Stanislav Kalinin
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia.
| | - Olga Bakulina
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg 199034, Russia.
| |
Collapse
|
5
|
Sun M, Zhang Y, Zhang XQ, Zhang Y, Wang XD, Li JT, Si TM, Su YA. Dopamine D1 receptor in medial prefrontal cortex mediates the effects of TAAR1 activation on chronic stress-induced cognitive and social deficits. Neuropsychopharmacology 2024; 49:1341-1351. [PMID: 38658737 PMCID: PMC11224251 DOI: 10.1038/s41386-024-01866-7] [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: 01/14/2024] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024]
Abstract
Trace amine-associated receptor 1 (TAAR1) is an intracellular expressed G-protein-coupled receptor that is widely expressed in major dopaminergic areas and plays a crucial role in modulation of central dopaminergic neurotransmission and function. Pharmacological studies have clarified the roles of dopamine D1 receptor (D1R) in the medial prefrontal cortex (mPFC) in cognitive function and social behaviors, and chronic stress can inhibit D1R expression due to its susceptibility. Recently, we identified TAAR1 in the mPFC as a potential target for treating chronic stress-induced cognitive and social dysfunction, but whether D1R is involved in mediating the effects of TAAR1 agonist remains unclear. Combined genomics and transcriptomic studies revealed downregulation of D1R in the mPFC of TAAR1-/- mice. Molecular dynamics simulation showed that hydrogen bond, salt bridge, and Pi-Pi stacking interactions were formed between TAAR1 and D1R indicating a stable TAAR1-D1R complex structure. Using pharmacological interventions, we found that D1R antagonist disrupted therapeutic effect of TAAR1 partial agonist RO5263397 on stress-related cognitive and social dysfunction. Knockout TAAR1 in D1-type dopamine receptor-expressing neurons reproduced adverse effects of chronic stress, and TAAR1 conditional knockout in the mPFC led to similar deficits, along with downregulation of D1R expression, all of these effects were ameliorated by viral overexpression of D1R in the mPFC, suggesting the functional interaction between TAAR1 and D1R. Collectively, our data elucidate the possible molecular mechanism that D1R in the mPFC mediates the effects of TAAR1 activation on chronic stress-induced cognitive and social deficits.
Collapse
Affiliation(s)
- Meng Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Yue Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xian-Qiang Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| |
Collapse
|
6
|
Zhang Y, Zhang XQ, Niu WP, Sun M, Zhang Y, Li JT, Si TM, Su YA. TAAR1 in dentate gyrus is involved in chronic stress-induced impairments in hippocampal plasticity and cognitive function. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110995. [PMID: 38514038 DOI: 10.1016/j.pnpbp.2024.110995] [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: 12/08/2023] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Multiple lines of evidence suggest that the trace amine-associated receptor 1 (TAAR1) holds promise as a potential target for stress-related disorders, such as treating major depressive disorder (MDD). The role of TAAR1 in the regulation of adult neurogenesis is recently supported by transcriptomic data. However, it remains unknown whether TAAR1 in dentate gyrus (DG) mediate chronic stress-induced negative effects on hippocampal plasticity and related behavior in mice. The present study consisted of a series of experiments using RNAscope, genetic approaches, behavioral tests, immunohistochemical staining, Golgi-Cox technique to unravel the effects of TAAR1 on alterations of dentate neuronal plasticity and cognitive function in the chronic social defeat stress model. The mice subjected to chronic defeat stress exhibited a noteworthy decrease in the mRNA level of TAAR1 in DG. Additionally, they exhibited compromised social memory and spatial object recognition memory, as well as impaired proliferation and maturation of adult-born dentate granule cells. Moreover, the selective knockout TAAR1 in DG mostly mimicked the cognitive function deficits and neurogenesis impairment induced by chronic stress. Importantly, the administration of the selective TAAR1 partial agonist RO5263397 during stress exposure attenuated the adverse effects of chronic stress on cognitive function, adult neurogenesis, dendritic arborization, and the synapse number of dentate neurons in DG. In summary, our findings suggest that TAAR1 plays a crucial role in mediating the detrimental effects of chronic stress on hippocampal plasticity and cognition. TAAR1 agonists exhibit therapeutic potential for individuals suffering from cognitive impairments associated with MDD.
Collapse
Affiliation(s)
- Yue Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xian-Qiang Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Wei-Pan Niu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Meng Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| |
Collapse
|
7
|
Savchenko A, Targa G, Fesenko Z, Leo D, Gainetdinov RR, Sukhanov I. Dopamine Transporter Deficient Rodents: Perspectives and Limitations for Neuroscience. Biomolecules 2023; 13:806. [PMID: 37238676 PMCID: PMC10216310 DOI: 10.3390/biom13050806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
The key element of dopamine (DA) neurotransmission is undoubtedly DA transporter (DAT), a transmembrane protein responsible for the synaptic reuptake of the mediator. Changes in DAT's function can be a key mechanism of pathological conditions associated with hyperdopaminergia. The first strain of gene-modified rodents with a lack of DAT were created more than 25 years ago. Such animals are characterized by increased levels of striatal DA, resulting in locomotor hyperactivity, increased levels of motor stereotypes, cognitive deficits, and other behavioral abnormalities. The administration of dopaminergic and pharmacological agents affecting other neurotransmitter systems can mitigate those abnormalities. The main purpose of this review is to systematize and analyze (1) known data on the consequences of changes in DAT expression in experimental animals, (2) results of pharmacological studies in these animals, and (3) to estimate the validity of animals lacking DAT as models for discovering new treatments of DA-related disorders.
Collapse
Affiliation(s)
- Artem Savchenko
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy Str. 6-8, 197022 St. Petersburg, Russia;
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Zoia Fesenko
- Institute of Translational Biomedicine, St. Petersburg State University, 7/9 Universitetskaya Emb., 199034 St. Petersburg, Russia
| | - Damiana Leo
- Department of Neurosciences, University of Mons, 7000 Mons, Belgium
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, 7/9 Universitetskaya Emb., 199034 St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, Fontanka River Emb. 154, 190121 St. Petersburg, Russia
| | - Ilya Sukhanov
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy Str. 6-8, 197022 St. Petersburg, Russia;
- St. Petersburg University Hospital, St. Petersburg State University, Fontanka River Emb. 154, 190121 St. Petersburg, Russia
| |
Collapse
|
8
|
Cai X, Wu M, Zhang Z, Liu H, Huang S, Song J, Ren S, Huang Y. Electroacupuncture alleviated depression‐like behaviors in ventromedial prefrontal cortex of chronic unpredictable mild stress‐induced rats: Increasing synaptic transmission and phosphorylating dopamine transporter. CNS Neurosci Ther 2023. [PMID: 37002793 PMCID: PMC10401110 DOI: 10.1111/cns.14200] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
AIMS Electroacupuncture (EA) shows advantages in both clinical practice and depression animal models. Dopaminergic-related dysfunction in the prefrontal cortex (PFC) may be a hidden antidepressant mechanism of EA, where dopamine transporter (DAT) plays an essential role. This study aimed to investigate the synaptic transmission and DAT-related changes of EA in depression. METHODS Male Sprague-Dawley rats were subjected to 3-week chronic unpredictable mild stress (CUMS). The successfully modeled rats were then randomly and equally assigned to CUMS, selective serotonin reuptake inhibitor (SSRI), and EA or SSRI + EA groups, followed by a 2-week treatment respectively. After monitoring body weight and behavioral tests of all rats, the ventromedial PFC (vmPFC) tissue was collected for electrophysiology and the expression detection of DAT, phosphorylated DAT (p-DAT), cyclic adenosine monophosphate (cAMP), protein kinase A (PKA), and trace amine-associated receptor 1 (TAAR1). RESULTS Depressive-like behaviors induced by CUMS were alleviated by EA, SSRI, and SSRI + EA treatments through behavioral tests. Compared with CUMS group, EA improved synaptic transmission in vmPFC by upregulating spontaneous excitatory postsynaptic currents amplitude. Molecularly, EA reversed the increased total DAT and p-DAT expression as well as the decreased ratio of p-DAT/total DAT along with the activation of TAAR1, cAMP, and PKA in vmPFC. CONCLUSION We speculated that the antidepressant effect of EA was associated with enhanced synaptic transmission in vmPFC, and the upregulated phosphorylation of DAT relevant to TAAR1, cAMP, and PKA may be the potential mechanism.
Collapse
Affiliation(s)
- Xiaowen Cai
- School of Traditional Chinese Medicine Southern Medical University Guangzhou 510515 Guangdong China
| | - Mei Wu
- School of Traditional Chinese Medicine Southern Medical University Guangzhou 510515 Guangdong China
| | - Zhinan Zhang
- School of Traditional Chinese Medicine Southern Medical University Guangzhou 510515 Guangdong China
| | - Huacong Liu
- School of Traditional Chinese Medicine Southern Medical University Guangzhou 510515 Guangdong China
| | - Shengtao Huang
- School of Traditional Chinese Medicine Southern Medical University Guangzhou 510515 Guangdong China
| | - Jia Song
- Guangdong‐Hong Kong‐Macao Greater Bay Area Center for Brain Science and Brain‐Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders Southern Medical University Guangzhou 510515 Guangdong China
| | - Siqiang Ren
- Guangdong‐Hong Kong‐Macao Greater Bay Area Center for Brain Science and Brain‐Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders Southern Medical University Guangzhou 510515 Guangdong China
| | - Yong Huang
- School of Traditional Chinese Medicine Southern Medical University Guangzhou 510515 Guangdong China
| |
Collapse
|
9
|
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
|
10
|
Giménez-Palomo A, Vieta E. The potential of TAAR1 agonists in bipolar disorder. Eur Neuropsychopharmacol 2022; 62:4-6. [PMID: 35803120 DOI: 10.1016/j.euroneuro.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/15/2022]
Affiliation(s)
- Anna Giménez-Palomo
- Bipolar and Depressive Disorders Unit, Hospital Clinic, Institute of Neuroscience, University of Barcelona, IDIBAPS, CIBERSAM, ISCIII, 170 Villarroel St, 08036 Barcelona, Catalonia, Spain
| | - Eduard Vieta
- Bipolar and Depressive Disorders Unit, Hospital Clinic, Institute of Neuroscience, University of Barcelona, IDIBAPS, CIBERSAM, ISCIII, 170 Villarroel St, 08036 Barcelona, Catalonia, Spain.
| |
Collapse
|
11
|
Raony Í, Domith I, Lourenco MV, Paes-de-Carvalho R, Pandolfo P. Trace amine-associated receptor 1 modulates motor hyperactivity, cognition, and anxiety-like behavior in an animal model of ADHD. Prog Neuropsychopharmacol Biol Psychiatry 2022; 117:110555. [PMID: 35346791 DOI: 10.1016/j.pnpbp.2022.110555] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 03/03/2022] [Accepted: 03/22/2022] [Indexed: 02/03/2023]
Abstract
Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that has recently been implicated in several psychiatric conditions related to monoaminergic dysfunction, such as schizophrenia, substance use disorders, and mood disorders. Although attention-deficit/hyperactivity disorder (ADHD) is also related to changes in monoaminergic neurotransmission, studies that assess whether TAAR1 participates in the neurobiology of ADHD are lacking. We hypothesized that TAAR1 plays an important role in ADHD and might represent a potential therapeutic target. Here, we investigate if TAAR1 modulates behavioral phenotypes in Spontaneously Hypertensive Rats (SHR), the most validated animal model of ADHD, and Wistar Kyoto rats (WKY, used as a control strain). Our results showed that TAAR1 is downregulated in ADHD-related brain regions in SHR compared with WKY. While intracerebroventricular (i.c.v.) administration of the selective TAAR1 antagonist EPPTB impaired cognitive performance in SHR, i.c.v. administration of highly selective TAAR1 full agonist RO5256390 decreased motor hyperactivity, novelty-induced locomotion, and induced an anxiolytic-like behavior. Overall, our findings show that changes in TAAR1 levels/activity underlie behavior in SHR, suggesting that TAAR1 plays a role in the neurobiology of ADHD. Although additional confirmatory studies are required, TAAR1 might be a potential pharmacological target for individuals with this disorder.
Collapse
Affiliation(s)
- Ícaro Raony
- Laboratory of Neurobiology of Animal Behavior, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ivan Domith
- Laboratory of Cellular Neurobiology, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Roberto Paes-de-Carvalho
- Laboratory of Cellular Neurobiology, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil
| | - Pablo Pandolfo
- Laboratory of Neurobiology of Animal Behavior, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil.
| |
Collapse
|
12
|
Decker AM, Brackeen MF, Mohammadkhani A, Kormos CM, Hesk D, Borgland SL, Blough BE. Identification of a Potent Human Trace Amine-Associated Receptor 1 Antagonist. ACS Chem Neurosci 2022; 13:1082-1095. [PMID: 35325532 DOI: 10.1021/acschemneuro.2c00086] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human trace amine-associated receptor subtype 1 (hTAAR1) is a G protein-coupled receptor that has therapeutic potential for multiple diseases, including schizophrenia, drug addiction, and Parkinson's disease (PD). Although several potent agonists have been identified and have shown positive results in various clinical trials for schizophrenia, the discovery of potent hTAAR1 antagonists remains elusive. Herein, we report the results of structure-activity relationship studies that have led to the discovery of a potent hTAAR1 antagonist (RTI-7470-44, 34). RTI-7470-44 exhibited an IC50 of 8.4 nM in an in vitro cAMP functional assay, a Ki of 0.3 nM in a radioligand binding assay, and showed species selectivity for hTAAR1 over the rat and mouse orthologues. RTI-7470-44 displayed good blood-brain barrier permeability, moderate metabolic stability, and a favorable preliminary off-target profile. Finally, RTI-7470-44 increased the spontaneous firing rate of mouse VTA dopaminergic neurons and blocked the effects of the known TAAR1 agonist RO5166017. Collectively, this work provides a promising hTAAR1 antagonist probe that can be used to study TAAR1 pharmacology and the potential therapeutic role in hypodopaminergic diseases such as PD.
Collapse
Affiliation(s)
- Ann M. Decker
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - Marcus F. Brackeen
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - Aida Mohammadkhani
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Chad M. Kormos
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - David Hesk
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| | - Stephanie L. Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N4N1, Canada
| | - Bruce E. Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
13
|
TAAR1 regulates drug-induced reinstatement of cocaine-seeking via negatively modulating CaMKIIα activity in the NAc. Mol Psychiatry 2022; 27:2136-2145. [PMID: 35079125 PMCID: PMC9829124 DOI: 10.1038/s41380-022-01448-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 01/12/2023]
Abstract
Relapse remains a major challenge to the treatment of cocaine addiction. Recent studies suggested that the trace amine-associated receptor 1 (TAAR1) could be a promising target to treat cocaine addiction and relapse; however, the underlying mechanism remains unclear. Here, we aimed to investigate the neural mechanism underlying the role of TAAR1 in the drug priming-induced reinstatement of cocaine-seeking behavior in rats, an animal model of cocaine relapse. We focused on the shell subregion of nucleus accumbens (NAc), a key brain region of the brain reward system. We found that activation of TAAR1 by systemic and intra-NAc shell administration of the selective TAAR1 agonist RO5166017 attenuated drug-induced reinstatement of cocaine-seeking and prevented drug priming-induced CaMKIIα activity in the NAc shell. Activation of TAAR1 dampened the CaMKIIα/GluR1 signaling pathway in the NAc shell and reduced AMPAR-EPSCs on the NAc slice. Microinjection of the selective TAAR1 antagonist EPPTB into the NAc shell enhanced drug-induced reinstatement as well as potentiated CaMKIIα activity in the NAc shell. Furthermore, viral-mediated expression of CaMKIIα in the NAc shell prevented the behavioral effects of TAAR1 activation. Taken together, our findings indicate that TAAR1 regulates drug-induced reinstatement of cocaine-seeking by negatively regulating CaMKIIα activity in the NAc. Our findings elucidate a novel mechanism of TAAR1 in regulating drug-induced reinstatement of cocaine-seeking and further suggests that TAAR1 is a promising target for the treatment of cocaine relapse.
Collapse
|
14
|
Bon C, Chern TR, Cichero E, O’Brien TE, Gustincich S, Gainetdinov RR, Espinoza S. Discovery of Novel Trace Amine-Associated Receptor 5 (TAAR5) Antagonists Using a Deep Convolutional Neural Network. Int J Mol Sci 2022; 23:3127. [PMID: 35328548 PMCID: PMC8954676 DOI: 10.3390/ijms23063127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/06/2022] [Accepted: 03/10/2022] [Indexed: 12/10/2022] Open
Abstract
Trace amine-associated receptor 5 (TAAR5) is a G protein-coupled receptor that belongs to the TAARs family (TAAR1-TAAR9). TAAR5 is expressed in the olfactory epithelium and is responsible for sensing 3-methylamine (TMA). However, recent studies showed that TAAR5 is also expressed in the limbic brain regions and is involved in the regulation of emotional behaviour and adult neurogenesis, suggesting that TAAR5 antagonism may represent a novel therapeutic strategy for anxiety and depression. We used the AtomNet® model, the first deep learning neural network for structure-based drug discovery, to identify putative TAAR5 ligands and tested them in an in vitro BRET assay. We found two mTAAR5 antagonists with low to submicromolar activity that are able to inhibit the cAMP production induced by TMA. Moreover, these two compounds also inhibited the mTAAR5 downstream signalling, such as the phosphorylation of CREB and ERK. These two hits exhibit drug-like properties and could be used to further develop more potent TAAR5 ligands with putative anxiolytic and antidepressant activity.
Collapse
Affiliation(s)
- Carlotta Bon
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), 16132 Genova, Italy; (C.B.); (S.G.)
| | - Ting-Rong Chern
- Atomwise Inc., San Francisco, CA 94103, USA; (T.-R.C.); (T.E.O.)
| | - Elena Cichero
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 16132 Genoa, Italy;
| | | | - Stefano Gustincich
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), 16132 Genova, Italy; (C.B.); (S.G.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia;
- Saint-Petersburg University Hospital, Saint-Petersburg State University, 199034 St. Petersburg, Russia
| | - Stefano Espinoza
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), 16132 Genova, Italy; (C.B.); (S.G.)
- Department of Health Sciences and Research, Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), 28100 Novara, Italy
| |
Collapse
|
15
|
Jarończyk M, Walory J. Novel Molecular Targets of Antidepressants. Molecules 2022; 27:533. [PMID: 35056845 PMCID: PMC8778443 DOI: 10.3390/molecules27020533] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Antidepressants target a variety of proteins in the central nervous system (CNS), the most important belonging to the family of G-protein coupled receptors and the family of neurotransmitter transporters. The increasing number of crystallographic structures of these proteins have significantly contributed to the knowledge of their mechanism of action, as well as to the design of new drugs. Several computational approaches such as molecular docking, molecular dynamics, and virtual screening are useful for elucidating the mechanism of drug action and are important for drug design. This review is a survey of molecular targets for antidepressants in the CNS and computer based strategies to discover novel compounds with antidepressant activity.
Collapse
|
16
|
Trace Amine-Associated Receptor 1 as a Target for the Development of New Antipsychotics: Current Status of Research and Future Directions. CNS Drugs 2021; 35:1153-1161. [PMID: 34655036 DOI: 10.1007/s40263-021-00864-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Schizophrenia is a mental illness associated with an array of symptoms that often result in disability. The primary treatments for schizophrenia are termed antipsychotics. Although antipsychotics modulate a number of different receptor types and subtypes, all currently regulatory agency-approved antipsychotics share in common direct or functional antagonism at the dopamine type 2 receptor (D2R). The majority of people with schizophrenia do not achieve full resolution of their symptoms with antipsychotics, suggesting the need for alternative or complementary approaches. The primary focus of this review is to assess the evidence for the role of the trace amine-associated receptor 1 (TAAR-1) in schizophrenia and the role of TAAR-1 modulators as novel-mechanism antipsychotics. Topics include an overview of TAAR-1 physiology and pathophysiology in schizophrenia, interaction with other neurotransmitter systems, including the dopaminergic, glutamatergic and serotonergic system, and finally, a review of investigational TAAR-1 compounds that have reached Phase II clinical studies in schizophrenia: SEP-363856 (ulotaront) and RO6889450 (ralmitaront). Thus far, results are publicly available only for ulotaront in a relatively young (18-40 years) and acutely exacerbated cohort. These results showed positive effects for overall schizophrenia symptoms without significant tolerability concerns. An ongoing study of ralmitaront will assess specific efficacy in patients with persistent negative symptoms. If trials of TAAR-1 modulators, and other novel-mechanism targets for schizophrenia that are under active study, continue to show positive results, the definition of an antipsychotic may need to be expanded beyond the D2R target in the near future.
Collapse
|
17
|
Role of trace amine‑associated receptor 1 in the medial prefrontal cortex in chronic social stress-induced cognitive deficits in mice. Pharmacol Res 2021; 167:105571. [PMID: 33753244 DOI: 10.1016/j.phrs.2021.105571] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022]
Abstract
Emerging evidence supports an essential role of trace amine-associated receptor 1 (TAAR1) in neuropsychiatric disorders such as depression and schizophrenia. Stressful events are critical contributors to various neuropsychiatric disorders. This study examined the role of TAAR1 in mediating the negative outcomes of stressful events. In mice that experienced chronic social defeat stress but not acute stress, a significant reduction in the TAAR1 mRNA level was found in the medial prefrontal cortex (mPFC), a brain region that is known to be vulnerable to stress experience. Conditional TAAR1 knockout in the mPFC mimicked the cognitive deficits induced by chronic stress. In addition, chronic treatment with the selective TAAR1 partial agonist RO5263397 ameliorated chronic stress-induced changes in cognitive function, dendritic arborization, and the synapse number of pyramidal neurons in the mPFC but did not affect chronic stress-induced anxiety-like behaviors. Biochemically, chronic stress reduced the ratio of vesicular transporters of glutamate-1 (VGluT1) / vesicular GABA transporter (VGAT) in the mPFC,most prominently in the prelimbic cortex, and RO5263397 restored the excitatory-inhibitory (E/I) imbalance. Together, the results of this study reveal an essential role of TAAR1 in mediating chronic stress-induced cognitive impairments and suggest that TAAR1 agonists may be uniquely useful to treat MDD-related cognitive impairments.
Collapse
|
18
|
Dodd S, F Carvalho A, Puri BK, Maes M, Bortolasci CC, Morris G, Berk M. Trace Amine-Associated Receptor 1 (TAAR1): A new drug target for psychiatry? Neurosci Biobehav Rev 2020; 120:537-541. [PMID: 33031817 DOI: 10.1016/j.neubiorev.2020.09.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/31/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023]
Abstract
There are nine subfamilies of TAARs. They are predominantly intracellular, located in the central nervous system and peripherally. They have a role in homeostasis and rheostasis, and also in olfaction. They demonstrate significant cross-talk with the monoamine system and are involved in the regulation of cAMP signalling and K+ channels. There is evidence to suggest that TAAR1 may be a promising therapeutic target for the treatment of schizophrenia, psychosis in Parkinson's disease, substance use disorders, and the metabolic syndrome and obesity. TAAR1 expression may also be a prognostic biomarker for cancers. A number of TAAR modulators have been identified, including endogenous ligands and new chemical entities. Some of these agents have shown efficacy in animal models of addiction behaviours, depression and anxiety. Only one agent, SEP-363856, has progressed to randomised clinical trials in humans; however further, larger studies with SEP-363856 are required to clarify its suitability as a new treatment for schizophrenia spectrum disorders. SEP-363856 is an agonist of TAAR1 and 5HT1A and it is not clear to what extent its efficacy can be attributed to TAAR1 rather than to other drug targets. However, current research suggests that TAAR1 has an important role in human physiology and pathophysiology. TAAR1 modulators may become an important new drug class for the management of a wide array of mental disorders in the future.
Collapse
Affiliation(s)
- Seetal Dodd
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia; Centre for Youth Mental Health, University of Melbourne, Parkville, Australia; Department of Psychiatry, University of Melbourne, Parkville, Australia; University Hospital Geelong, Barwon Health, PO Box 281, Geelong, Victoria, 3220, Australia.
| | - André F Carvalho
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, Toronto, ON, Canada
| | | | - Michael Maes
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Chiara C Bortolasci
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia; Centre for Molecular and Medical Research, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Gerwyn Morris
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Australia; Centre for Youth Mental Health, University of Melbourne, Parkville, Australia; Department of Psychiatry, University of Melbourne, Parkville, Australia; University Hospital Geelong, Barwon Health, PO Box 281, Geelong, Victoria, 3220, Australia
| |
Collapse
|
19
|
Decker AM, Mathews KM, Blough BE, Gilmour BP. Validation of a High-Throughput Calcium Mobilization Assay for the Human Trace Amine-Associated Receptor 1. SLAS DISCOVERY 2020; 26:140-150. [PMID: 32734809 DOI: 10.1177/2472555220945279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The human trace amine-associated receptor 1 (hTAAR1) is a G protein-coupled receptor (GPCR) that is widely expressed in monoaminergic nuclei in the central nervous system and has therapeutic potential for multiple diseases, including drug addiction and schizophrenia. Thus, identification of novel hTAAR1 ligands is critical to advancing our knowledge of hTAAR1 function and to the development of therapeutics for a wide range of diseases. Herein we describe the development of a robust, 3-addition high-throughput screening (HTS) calcium mobilization assay using stable CHO-Gαq16-hTAAR1 cells, which functionally couple hTAAR1 to the promiscuous Gαq16 protein and thus allow signal transduction to occur through mobilization of internal calcium. Our previously established 96-well hTAAR1 assay was first miniaturized to the 384-well format and optimized to provide an assay with a Z' factor of 0.84, which is indicative of a robust HTS assay. Using the 3-addition protocol, 22,000 compounds were screened and yielded a ~1% agonist hit rate and a ~0.2% antagonist hit rate. Of the antagonist hits, two confirmed hits are the most potent hTAAR1 antagonists identified to date (IC50 = 206 and 281 nM). While scientists have been studying hTAAR1 for years, the lack of suitable hTAAR1 antagonists has been a major roadblock for studying the basic pharmacology of hTAAR1. Thus, these new ligands will serve as valuable tools to study hTAAR1-mediated signaling mechanisms, therapeutic potential, and in vivo functions.
Collapse
Affiliation(s)
- Ann M Decker
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| | - Kelly M Mathews
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| | - Brian P Gilmour
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| |
Collapse
|
20
|
Wu R, Liu J, Wang K, Huang Y, Zhang Y, Li JX. Effects of a trace amine-associated receptor 1 agonist RO 5263397 on ethanol-induced behavioral sensitization. Behav Brain Res 2020; 390:112641. [PMID: 32407821 DOI: 10.1016/j.bbr.2020.112641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alcohol dependence is a chronic and severe health problem which puts a heavy burden on society. Alcohol activates mesolimbic dopamine circuity to achieve its reinforcing effect. While TAAR1 is critically involved in the modulation of dopamine, there is little evidence indicating that TAAR1 could play a role in behavioral effects of ethanol. METHODS By using the animal model of behavioral sensitization induced by ethanol in mice, the present study was performed to investigate whether the activation of TAAR1 would affect the behavioral plasticity of ethanol. RESULTS Repeated administration with ethanol induced a significant increased locomotion in WT mice with females showing higher level of sensitization to ethanol than male mice. The TAAR1 agonist RO5263397 significantly decreased the expression of ethanol-induced behavioral sensitization both in male and female WT mice (0.1 and 0.32 mg/kg). Repeated RO5263397 exposure also prevented the development of behavioral sensitization to ethanol both in male and female WT mice. Moreover, while TAAR1-KO mice developed normal levels of ethanol-induced behavioral sensitization, RO5263397 did not affect this behavior in TAAR1-KO mice. CONCLUSIONS These results indicated that the TAAR1 agonist RO5263397 negatively regulated the expression and development of ethanol-elicited behavioral sensitization in WT but not in TAAR1-KO mice. The present study suggests that TAAR1 is probably involved in certain addiction-like effects of alcohol and could be a useful drug target for the development of new medications to treat alcohol dependence.
Collapse
Affiliation(s)
- Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States; School of Medicine, Yangzhou University, Yangzhou, China
| | - Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Kaixuan Wang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Yufei Huang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| | - Yanan Zhang
- Research Triangle Institute, NC, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
21
|
Abstract
Trace amine-associated receptor 1 is one of the best-characterized receptors of trace amines. Growing evidence shows that TAAR1 negatively regulates the monoaminergic activity, including dopamine transmission in the mesocorticolimbic system. Neurochemical assays demonstrated that selective TAAR1 full and partial agonists were effective to prevent psychostimulants-induced dopamine transmission in vitro and in vivo. In the last decade, many preclinical models of psychostimulant addiction such as drug-induced behavioral sensitization, drug-induced conditioned place preference, drug self-administration, drug discrimination, and relapse models were used to assess the effects of TAAR1 agonists on psychostimulants' behavioral effects. In general, activation of TAAR1 attenuated while knockout of TAAR1 potentiated psychostimulant abuse-related behaviors. Here, we review the advances in TAAR1 and its agonists in modulating psychostimulant addiction. We discuss the similarities and differences between the neurochemical and behavioral effects of TAAR1 full and partial agonists. We also discuss several concerns including the abuse liability, sleep reduction, and species-dependent effects that might affect the successful translation of TAAR1 agonists from preclinical studies to clinical application. In conclusion, although further investigations are in need to address certain concerns and the underlying neural mechanisms, TAAR1 agonists appear to be a promising pharmacotherapy to treat psychostimulant addiction and prevent relapse.
Collapse
|
22
|
Shi X, Swanson TL, Miner NB, Eshleman AJ, Janowsky A. Activation of Trace Amine-Associated Receptor 1 Stimulates an Antiapoptotic Signal Cascade via Extracellular Signal-Regulated Kinase 1/2. Mol Pharmacol 2019; 96:493-504. [PMID: 31409621 DOI: 10.1124/mol.119.116798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/02/2019] [Indexed: 12/30/2022] Open
Abstract
Methamphetamine (MA) is highly addictive and neurotoxic, causing cell death in humans and in rodent models. MA, along with many of its analogs, is an agonist at the G protein-coupled trace amine-associated receptor 1 (TAAR1). TAAR1 activation protects against MA-induced degeneration of dopaminergic neurons, suggesting that TAAR1 plays a role in regulating MA-induced neurotoxicity. However, the mechanisms involved in TAAR1's role in neurotoxicity and cell death have not been described in detail. In this study, we investigated the apoptosis pathway in Taar1 wild-type (WT) and knockout (KO) mice and in cells expressing the recombinant receptor. Bcl-2, an antiapoptotic protein, was upregulated ∼3-fold in the midbrain area (substantial nigra and ventral tegmental area) in Taar1 KO compared with WT mice, and MA significantly increased Bcl-2 expression in WT mice but decreased Bcl-2 expression in KO mice. The proapoptotic protein Bax did not differ across genotype or in response to MA. Bcl-2 expression was significantly upregulated by the TAAR1 agonist RO5166017 ((S)-4-[(ethyl-phenyl-amino)-methyl]-4,5-dihydro-oxazol-2-ylamine) in cells expressing the recombinant mouse TAAR1. Additionally, activation of TAAR1 by RO5166017 increased phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, and protein kinase B (AKT), but only inhibition of ERK1/2 phosphorylation prevented TAAR1-induced increases in Bcl-2 levels, indicating that TAAR1 activation increases Bcl-2 through an ERK1/2-dependent pathway. All changes to ERK1/2 pathway intermediates were blocked by the TAAR1 antagonist, N-(3-ethoxyphenyl)-4-(1-pyrrolidinyl)-3-(trifluoromethyl) benzamide. These findings suggest that TAAR1 activation protects against MA-induced cell apoptosis and TAAR1 may play a role in cell death in neurodegenerative diseases. SIGNIFICANCE STATEMENT: Methamphetamine stimulates TAAR1, a G protein-coupled receptor. The role and mechanisms for TAAR1 in methamphetamine-induced neurotoxicity are not known. Here, we report that, in genetic mouse models and cells expressing the recombinant receptor, TAAR1 activates the ERK1/2 pathway but not the AKT pathway to upregulate the antiapoptotic protein Bcl-2, which protects cells from drug-induced toxicity.
Collapse
Affiliation(s)
- Xiao Shi
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| | - Tracy L Swanson
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| | - Nicholas B Miner
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| | - Amy J Eshleman
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| | - Aaron Janowsky
- Research Service, Veterans Affairs Portland Health Care System, Portland, Oregon (X.S., T.L.S., N.B.M., A.J.E., A.J.); and The Methamphetamine Abuse Research Center (X.S., A.J.) and Departments of Psychiatry (X.S., T.L.S., A.J.E., A.J.) and Behavioral Neuroscience (N.B.M., A.J.E., A.J.), Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
23
|
Keeping up with the therapeutic advances in schizophrenia: a review of novel and emerging pharmacological entities. CNS Spectr 2019; 24:38-69. [PMID: 31482779 DOI: 10.1017/s109285291900124x] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Schizophrenia remains one of the most severe medical diseases. Current dopamine modulating first-generation and second-generation antipsychotics target mainly positive symptoms, but not/inadequately negative and cognitive symptoms. Additional challenges include non-adherence and adverse effects, especially cardiometabolic dysregulation. This review evaluates new/emerging pharmacological treatments for schizophrenia. Therapies targeting total symptoms include cannabidiol, D3 antagonist/5-HT1A partial agonist F17464, lumateperone (ITI-007), phosphodiesterase 10A (PDE10A) inhibitors MK-8189 and TAK-063, sodium nitroprusside, and trace amine-associated receptor-1 (TAAR1) agonist RO5263397 and SEP-363856. Treatments targeting negative symptoms include the PDE10A inhibitor LuAF-11167, 5-HT2A inverse agonist pimavanserin, sigma-2/5-HT2A antagonist roluperidone (MIN-101), and d-amino acid oxidase (DAAO) inhibitor TAK-831. Agents targeting primarily cognitive dysfunction are the glycine transporter-1 inhibitor BI-425809 and cannabidiol. Therapies targeting residual positive symptoms/treatment-resistant schizophrenia include pimavanserin, dopamine D1/D2 antagonist LuAF-35700, and DAAO inhibitor sodium benzoate. Two new long-acting injectable antipsychotic formulations, Aripiprazole Lauroxil NanoCrystal® and the first subcutaneous injectable LAI Perseris (RBP-7000), were recently approved by U.S. Food and Drug Administration, and positive results were announced for Risperidone ISM®, each achieving therapeutic levels within 24 hours, without need for initial oral cotreatment/loading injection-strategies. Paliperidone palmitate 6-monthly intramuscularly injectable and Risperidone subcutaneously injectable TV46000 are currently under investigation. Finally, the samidorphan+olanzapine combination targets reduced weight gain liability, while maintaining olanzapine's efficacy. Most of these trial programs are still ongoing or have yielded mixed or even negative results. Thus, additional mechanisms of action and agents require study to improve schizophrenia outcomes for total/positive symptoms with reduced adverse effects, but also cognitive symptoms, negative symptoms, and treatment resistance, the areas of greatest need in schizophrenia currently.
Collapse
|
24
|
Stafford AM, Reed C, Baba H, Walter NAR, Mootz JRK, Williams RW, Neve KA, Fedorov LM, Janowsky AJ, Phillips TJ. Taar1 gene variants have a causal role in methamphetamine intake and response and interact with Oprm1. eLife 2019; 8:e46472. [PMID: 31274109 PMCID: PMC6682400 DOI: 10.7554/elife.46472] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/04/2019] [Indexed: 12/17/2022] Open
Abstract
We identified a locus on mouse chromosome 10 that accounts for 60% of the genetic variance in methamphetamine intake in mice selectively bred for high versus low methamphetamine consumption. We nominated the trace amine-associated receptor 1 gene, Taar1, as the strongest candidate and identified regulation of the mu-opioid receptor 1 gene, Oprm1, as another contributor. This study exploited CRISPR-Cas9 to test the causal role of Taar1 in methamphetamine intake and a genetically-associated thermal response to methamphetamine. The methamphetamine-related traits were rescued, converting them to levels found in methamphetamine-avoiding animals. We used a family of recombinant inbred mouse strains for interval mapping and to examine independent and epistatic effects of Taar1 and Oprm1. Both methamphetamine intake and the thermal response mapped to Taar1 and the independent effect of Taar1 was dependent on genotype at Oprm1. Our findings encourage investigation of the contribution of Taar1 and Oprm1 variants to human methamphetamine addiction.
Collapse
Affiliation(s)
- Alexandra M Stafford
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Cheryl Reed
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Harue Baba
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Nicole AR Walter
- Division of NeuroscienceOregon National Primate Research CenterPortlandUnited States
| | - John RK Mootz
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Robert W Williams
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Sciences CenterMemphisUnited States
| | - Kim A Neve
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
- Veterans Affairs Portland Health Care SystemPortlandUnited States
| | - Lev M Fedorov
- Transgenic Mouse Models Shared Resource, Knight Cancer InstituteOregon Health & Science UniversityPortlandUnited States
| | - Aaron J Janowsky
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
- Veterans Affairs Portland Health Care SystemPortlandUnited States
- Department of PsychiatryOregon Health & Science UniversityPortlandUnited States
| | - Tamara J Phillips
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
- Veterans Affairs Portland Health Care SystemPortlandUnited States
| |
Collapse
|