1
|
Pretorius L, Coetzee JA, Santos APD, Smith C. Modulating autism spectrum disorder pathophysiology using a trace amine-focused approach: targeting the gut. Mol Med 2025; 31:198. [PMID: 40394473 DOI: 10.1186/s10020-025-01232-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025] Open
Abstract
Autism spectrum disorder (ASD) affects approximately 1% of the population directly, but also a much higher proportion (family and caregivers) indirectly. Although ASD is characterized by high prevalence of anxiety and poor gastrointestinal health, current treatment strategies are mainly focused on neurological symptomatic treatment, with little to no attention to gut health. Furthermore, many psychiatric drugs used for management of secondary neurological symptoms, are known to exacerbate gut health issues and neurological dysregulation across the gut-brain axis.Trace amines are neurotransmitter-like substances synthesized endogenously in the human brain - in trace amounts - but also in high abundance by the microbiome. Emerging evidence suggests dysregulation of the trace amine system in ASD. Since trace aminergic signalling is central to regulatory system homeostasis, we hypothesize targeting this system in the ASD context. Given the various sources of trace amines, we suggest that normalization of functional dysbiosis in terms of trace aminergic signalling - rather than microbial compositional dysbiosis - should be a focus in medicines development. In addition, a holistic consideration including also other factors at play in determining trace aminergic signalling outcome - such as receptor binding, enzymatic role players, etc. - is required to fully elucidate and therapeutically modify the pathophysiology of regulatory systems implicated in ASD.This review firstly provides a brief overview of trace amine dysregulation in ASD for context. Secondly, we formulate our hypothesis on how this may therapeutically address symptomology, with consideration of cellular and molecular mechanism interplay across the gut-brain axis. Finally, we provide a critical assessment of advances in therapeutics development and drug re-purposing, gaps in knowledge and priorities for medicines development going forward.
Collapse
Affiliation(s)
- L Pretorius
- Experimental Medicine Group, Dept Medicine, Stellenbosch University, Parow, South Africa
| | - J A Coetzee
- Experimental Medicine Group, Dept Medicine, Stellenbosch University, Parow, South Africa
| | - A P Dos Santos
- Experimental Medicine Group, Dept Medicine, Stellenbosch University, Parow, South Africa
| | - C Smith
- Experimental Medicine Group, Dept Medicine, Stellenbosch University, Parow, South Africa.
| |
Collapse
|
2
|
Yang Y, Qiao X, Yu S, Zhao X, Jin Y, Liu R, Li J, Wang L, Song L. A trace amine associated receptor mediates antimicrobial immune response in the oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 156:105171. [PMID: 38537729 DOI: 10.1016/j.dci.2024.105171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/01/2024]
Abstract
Trace amine-associated receptors (TAARs) are a class of G protein-coupled receptors, playing an immunomodulatory function in the neuroinflammatory responses. In the present study, a TAAR homologue with a 7tm_classA_rhodopsin-like domain (designated as CgTAAR1L) was identified in oyster Crassostrea gigas. The abundant CgTAAR1L transcripts were detected in visceral ganglia and haemocytes compared to other tissues, which were 55.35-fold and 32.95-fold (p < 0.01) of those in adductor muscle, respectively. The mRNA expression level of CgTAAR1L in haemocytes significantly increased and reached the peak level at 3 h after LPS or Poly (I:C) stimulation, which was 4.55-fold and 12.35-fold of that in control group, respectively (p < 0.01). After the expression of CgTAAR1L was inhibited by the injection of its targeted siRNA, the mRNA expression levels of interleukin17s (CgIL17-1, CgIL17-5 and CgIL17-6), and defensin (Cgdefh1) significantly decreased at 3 h after LPS stimulation, which was 0.51-fold (p < 0.001), 0.39-fold (p < 0.01), 0.48-fold (p < 0.05) and 0.41-fold (p < 0.05) of that in the control group, respectively. The nuclear translocation of Cgp65 protein was suppressed in the CgTAAR1L-RNAi oysters. Furthermore, the number of Vibrio splendidus in the haemolymph of CgTAAR1L-RNAi oysters significantly increased (4.11-fold, p < 0.001) compared with that in the control group. In contrast, there was no significant difference in phagocytic rate of haemocytes to V. splendidus in the CgTAAR1L-RNAi oysters. These results indicated that CgTAAR1L played an important role in the immune defense against bacterial infection by inducing the expressions of interleukin and defensin.
Collapse
Affiliation(s)
- Yuehong Yang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Xue Qiao
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Simiao Yu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Xinyu Zhao
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Yuhao Jin
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Rui Liu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Jie Li
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| |
Collapse
|
3
|
Moiseenko VI, Apryatina VA, Gainetdinov RR, Apryatin SA. Trace Amine-Associated Receptors' Role in Immune System Functions. Biomedicines 2024; 12:893. [PMID: 38672247 PMCID: PMC11047934 DOI: 10.3390/biomedicines12040893] [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: 02/29/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Trace amines are a separate, independent group of biogenic amines, close in structure to classical monoamine neurotransmitters such as dopamine, serotonin, and norepinephrine that include many products of the endogenous or bacteria-mediated decarboxylation of amino acids. A family of G protein-coupled trace amine-associated receptors (in humans, TAAR1, TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) that senses trace amines was discovered relatively recently. They are mostly investigated for their involvement in the olfaction of volatile amines encoding innate behaviors and their potential contribution to the pathogenesis of neuropsychiatric disorders, but the expression of the TAAR family of receptors is also observed in various populations of cells in the immune system. This review is focused on the basic information of the interaction of trace amines and their receptors with cells of the general immune systems of humans and other mammals. We also overview the available data on TAARs' role in the function of individual populations of myeloid and lymphoid cells. With further research on the regulatory role of the trace amine system in immune functions and on uncovering the contribution of these processes to the pathogenesis of the immune response, a significant advance in the field could be expected. Furthermore, the determination of the molecular mechanisms of TAARs' involvement in immune system regulation and the further investigation of their potential chemotactic role could bring about the development of new approaches for the treatment of disorders related to immune system dysfunctions.
Collapse
Affiliation(s)
| | | | | | - Sergey A. Apryatin
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| |
Collapse
|
4
|
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
|
5
|
Sakanoue W, Yokoyama T, Hirakawa M, Maesawa S, Sato K, Saino T. 3-Iodothyronamine, a trace amine-associated receptor agonist, regulates intracellular Ca2+ increases via CaMK II through Epac2 in rat cerebral arterioles. Biomed Res 2023; 44:219-232. [PMID: 37779034 DOI: 10.2220/biomedres.44.219] [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: 10/03/2023]
Abstract
Trace amines (TAs) in the nervous system bind to TA-associated receptors (TAARs) and are involved in the regulation of monoaminergic functions. Among TAAR subtypes, TAAR1 has been implicated in the development of neurological disorders, such as schizophrenia. The present study investigated the effects of the TAAR1 agonist, 3-iodothyronamine (T1AM) on cerebral arterioles using fluctuations in the intracellular concentration of Ca2+ ([Ca2+]i) as an index of contractile responses. In cerebral arterioles, most of the TAAR agonists did not increase [Ca2+]i, while only T1AM elevated [Ca2+]i in vascular smooth muscle cells. This increase involved extracellular Ca2+ influx through T-type Ca2+ channels and inositol trisphosphate- and ryanodine-receptor-mediated Ca2+ release from intracellular stores. The inhibition of the cAMP sensor, exchange protein directly activated by cAMP (Epac) 2, and calmodulin kinase (CaMK) II strongly inhibited Ca2+ elevations. The present study revealed that T1AM acted not only on the TAAR1 receptor as previously suggested, but also on other G-protein coupled receptors and/or signal transduction systems to increase intracellular Ca2+ in cerebral arteriole smooth muscle cells. These results suggest that when using T1AM in clinical practice, attention should be paid to the early rise in blood pressure.
Collapse
Affiliation(s)
- Wakana Sakanoue
- Division of Dental Anesthesiology, Department of Reconstructive Oral and Maxillofacial Surgery, School of Dentistry, Iwate Medical University, Yahaba, Japan
| | - Takuya Yokoyama
- Laboratory of Veterinary Anatomy and Cell Biology, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Masato Hirakawa
- Department of Anatomy (Cell Biology), Iwate Medical University, Yahaba, Japan
| | - Satsuki Maesawa
- Division of Dental Anesthesiology, Department of Reconstructive Oral and Maxillofacial Surgery, School of Dentistry, Iwate Medical University, Yahaba, Japan
| | - Kenichi Sato
- Division of Dental Anesthesiology, Department of Reconstructive Oral and Maxillofacial Surgery, School of Dentistry, Iwate Medical University, Yahaba, Japan
| | - Tomoyuki Saino
- Department of Anatomy (Cell Biology), Iwate Medical University, Yahaba, Japan
| |
Collapse
|
6
|
Chilunda V, Weiselberg J, Martinez-Meza S, Mhamilawa LE, Cheney L, Berman JW. Methamphetamine induces transcriptional changes in cultured HIV-infected mature monocytes that may contribute to HIV neuropathogenesis. Front Immunol 2022; 13:952183. [PMID: 36059515 PMCID: PMC9433802 DOI: 10.3389/fimmu.2022.952183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
HIV-associated neurocognitive impairment (HIV-NCI) persists in 15-40% of people with HIV (PWH) despite effective antiretroviral therapy. HIV-NCI significantly impacts quality of life, and there is currently no effective treatment for it. The development of HIV-NCI is complex and is mediated, in part, by the entry of HIV-infected mature monocytes into the central nervous system (CNS). Once in the CNS, these cells release inflammatory mediators that lead to neuroinflammation, and subsequent neuronal damage. Infected monocytes may infect other CNS cells as well as differentiate into macrophages, thus contributing to viral reservoirs and chronic neuroinflammation. Substance use disorders in PWH, including the use of methamphetamine (meth), can exacerbate HIV neuropathogenesis. We characterized the effects of meth on the transcriptional profile of HIV-infected mature monocytes using RNA-sequencing. We found that meth mediated an upregulation of gene transcripts related to viral infection, cell adhesion, cytoskeletal arrangement, and extracellular matrix remodeling. We also identified downregulation of several gene transcripts involved in pathogen recognition, antigen presentation, and oxidative phosphorylation pathways. These transcriptomic changes suggest that meth increases the infiltration of mature monocytes that have a migratory phenotype into the CNS, contributing to dysregulated inflammatory responses and viral reservoir establishment and persistence, both of which contribute to neuronal damage. Overall, our results highlight potential molecules that may be targeted for therapy to limit the effects of meth on HIV neuropathogenesis.
Collapse
Affiliation(s)
- Vanessa Chilunda
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jessica Weiselberg
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Samuel Martinez-Meza
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Lwidiko E. Mhamilawa
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Parasitology and Medical Entomology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Department of Women’s and Children’s Health, International Maternal and Child Health (IMCH), Uppsala University, Uppsala, Sweden
| | - Laura Cheney
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Medicine, Division of Infectious Diseases, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, United States
| | - Joan W. Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
7
|
Wu R, Liu J, Li JX. Trace amine-associated receptor 1 and drug abuse. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 93:373-401. [PMID: 35341572 PMCID: PMC9826737 DOI: 10.1016/bs.apha.2021.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Trace amine-associated receptor 1 (TAAR1) is the best characterized receptor selectively activated by trace amines. It is broadly expressed in the monoaminergic system in the brain including ventral tegmental area (VTA), nucleus accumbens (NAc), dorsal raphe (DR) and substantial nigra (SN). Extensive studies have suggested that TAAR1 plays an important role in the modulation of monoaminergic system, especially dopamine (DA) transmission which may underlie the mechanisms by which TAAR1 interventions affect drug abuse-like behaviors. TAAR1 activation inhibits the rewarding and reinforcing effects of drugs from different classes including psychostimulants, opioid and alcohol as well as drug-induced increase in DA accumulation. The mechanisms of TAAR1's function in mediating drug abuse-like behaviors are not clear. However, it is hypothesized that TAAR1 interaction with DA transporter (DAT) and dopamine D2 receptor (D2) and the subsequent modulation of cellular cascades may contribute to the effects of TAAR1 in regulating drug abuse. Further studies are needed to investigate the role of TAAR1 in other drugs of abuse-related behaviors and its safety and efficacy for prolonged medications. Together, TAAR1 inhibits drug-induced DA transmission and drug abuse-related behaviors. Therefore, TAAR1 may be a promising therapeutic target for the treatment of drug addiction.
Collapse
Affiliation(s)
- Ruyan Wu
- Medical College of Yangzhou University, Yangzhou, China,Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Jianfeng Liu
- Department of Psychological and Brain Sciences, College of Liberal Arts, Texas A&M University, College Station, TX, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA,Corresponding authors: Dr. Jun-Xu Li, , Department of Pharmacology and Toxicology, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY 14214. Tel: +1 716 829 2482; Fax: +1 716 829 2801
| |
Collapse
|
8
|
Glyakina AV, Pavlov CD, Sopova JV, Gainetdinov RR, Leonova EI, Galzitskaya OV. Search for Structural Basis of Interactions of Biogenic Amines with Human TAAR1 and TAAR6 Receptors. Int J Mol Sci 2021; 23:ijms23010209. [PMID: 35008636 PMCID: PMC8745718 DOI: 10.3390/ijms23010209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
The identification and characterization of ligand-receptor binding sites are important for drug development. Trace amine-associated receptors (TAARs, members of the class A GPCR family) can interact with different biogenic amines and their metabolites, but the structural basis for their recognition by the TAARs is not well understood. In this work, we have revealed for the first time a group of conserved motifs (fingerprints) characterizing TAARs and studied the docking of aromatic (β-phenylethylamine, tyramine) and aliphatic (putrescine and cadaverine) ligands, including gamma-aminobutyric acid, with human TAAR1 and TAAR6 receptors. We have identified orthosteric binding sites for TAAR1 (Asp68, Asp102, Asp284) and TAAR6 (Asp78, Asp112, Asp202). By analyzing the binding results of 7500 structures, we determined that putrescine and cadaverine bind to TAAR1 at one site, Asp68 + Asp102, and to TAAR6 at two sites, Asp78 + Asp112 and Asp112 + Asp202. Tyramine binds to TAAR6 at the same two sites as putrescine and cadaverine and does not bind to TAAR1 at the selected Asp residues. β-Phenylethylamine and gamma-aminobutyric acid do not bind to the TAAR1 and TAAR6 receptors at the selected Asp residues. The search for ligands targeting allosteric and orthosteric sites of TAARs has excellent pharmaceutical potential.
Collapse
Affiliation(s)
- Anna V. Glyakina
- Institute of Mathematical Problems of Biology, Russian Academy of Sciences, Keldysh Institute of Applied Mathematics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia;
| | - Constantine D. Pavlov
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia;
| | - Julia V. Sopova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (J.V.S.); (R.R.G.)
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (J.V.S.); (R.R.G.)
| | - Elena I. Leonova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (J.V.S.); (R.R.G.)
- Animal Genetic Technologies Department, University of Science and Technology, 1 Olympic Ave, 354340 Sochi, Russia
- Correspondence: (E.I.L.); (O.V.G.)
| | - Oxana V. Galzitskaya
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia;
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
- Correspondence: (E.I.L.); (O.V.G.)
| |
Collapse
|
9
|
Dedic N, Dworak H, Zeni C, Rutigliano G, Howes OD. Therapeutic Potential of TAAR1 Agonists in Schizophrenia: Evidence from Preclinical Models and Clinical Studies. Int J Mol Sci 2021; 22:ijms222413185. [PMID: 34947997 PMCID: PMC8704992 DOI: 10.3390/ijms222413185] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 12/14/2022] Open
Abstract
Trace amine-associated receptor 1 (TAAR1) has emerged as a promising therapeutic target for neuropsychiatric disorders due to its ability to modulate monoaminergic and glutamatergic neurotransmission. In particular, agonist compounds have generated interest as potential treatments for schizophrenia and other psychoses due to TAAR1-mediated regulation of dopaminergic tone. Here, we review unmet needs in schizophrenia, the current state of knowledge in TAAR1 circuit biology and neuropharmacology, including preclinical behavioral, imaging, and cellular evidence in glutamatergic, dopaminergic and genetic models linked to the pathophysiology of psychotic, negative and cognitive symptoms. Clinical trial data for TAAR1 drug candidates are reviewed and contrasted with antipsychotics. The identification of endogenous TAAR1 ligands and subsequent development of small-molecule agonists has revealed antipsychotic-, anxiolytic-, and antidepressant-like properties, as well as pro-cognitive and REM-sleep suppressing effects of TAAR1 activation in rodents and non-human primates. Ulotaront, the first TAAR1 agonist to progress to randomized controlled clinical trials, has demonstrated efficacy in the treatment of schizophrenia, while another, ralmitaront, is currently being evaluated in clinical trials in schizophrenia. Coupled with the preclinical findings, this provides a rationale for further investigation and development of this new pharmacological class for the treatment of schizophrenia and other psychiatric disorders.
Collapse
Affiliation(s)
- Nina Dedic
- Sunovion Pharmaceuticals, Marlborough, MA 01752, USA; (H.D.); (C.Z.)
- Correspondence:
| | - Heather Dworak
- Sunovion Pharmaceuticals, Marlborough, MA 01752, USA; (H.D.); (C.Z.)
| | - Courtney Zeni
- Sunovion Pharmaceuticals, Marlborough, MA 01752, USA; (H.D.); (C.Z.)
| | - Grazia Rutigliano
- Department of Pathology, University of Pisa, via Savi 10, 56126 Pisa, Italy;
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK;
| | - Oliver D. Howes
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK;
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London SE5 8AF, UK
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK
| |
Collapse
|
10
|
Strassheim D, Sullivan T, Irwin DC, Gerasimovskaya E, Lahm T, Klemm DJ, Dempsey EC, Stenmark KR, Karoor V. Metabolite G-Protein Coupled Receptors in Cardio-Metabolic Diseases. Cells 2021; 10:3347. [PMID: 34943862 PMCID: PMC8699532 DOI: 10.3390/cells10123347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
G protein-coupled receptors (GPCRs) have originally been described as a family of receptors activated by hormones, neurotransmitters, and other mediators. However, in recent years GPCRs have shown to bind endogenous metabolites, which serve functions other than as signaling mediators. These receptors respond to fatty acids, mono- and disaccharides, amino acids, or various intermediates and products of metabolism, including ketone bodies, lactate, succinate, or bile acids. Given that many of these metabolic processes are dysregulated under pathological conditions, including diabetes, dyslipidemia, and obesity, receptors of endogenous metabolites have also been recognized as potential drug targets to prevent and/or treat metabolic and cardiovascular diseases. This review describes G protein-coupled receptors activated by endogenous metabolites and summarizes their physiological, pathophysiological, and potential pharmacological roles.
Collapse
Affiliation(s)
- Derek Strassheim
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Timothy Sullivan
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - David C. Irwin
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Evgenia Gerasimovskaya
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Tim Lahm
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health Denver, Denver, CO 80206, USA;
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| | - Dwight J. Klemm
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Edward C. Dempsey
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kurt R. Stenmark
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Vijaya Karoor
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health Denver, Denver, CO 80206, USA;
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
11
|
Barnes DA, Galloway DA, Hoener MC, Berry MD, Moore CS. TAAR1 Expression in Human Macrophages and Brain Tissue: A Potential Novel Facet of MS Neuroinflammation. Int J Mol Sci 2021; 22:ijms222111576. [PMID: 34769007 PMCID: PMC8584001 DOI: 10.3390/ijms222111576] [Citation(s) in RCA: 17] [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: 09/28/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
TAAR1 is a neuroregulator with emerging evidence suggesting a role in immunomodulation. Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system. Here, we investigate TAAR1 expression in human primary monocytes, peripherally-derived macrophages, and MS brain tissue. RT-qPCR was used to assess TAAR1 levels in MS monocytes. Using a previously validated anti-human TAAR1 antibody and fluorescence microscopy, TAAR1 protein was visualized in lipopolysaccharide-stimulated or basal human macrophages, as well as macrophage/microglia populations surrounding, bordering, and within a mixed active/inactive MS lesion. In vivo, TAAR1 mRNA expression was significantly lower in MS monocytes compared to age- and sex-matched healthy controls. In vitro, TAAR1 protein showed a predominant nuclear localization in quiescent/control macrophages with a shift to a diffuse intracellular distribution following lipopolysaccharide-induced activation. In brain tissue, TAAR1 protein was predominantly expressed in macrophages/microglia within the border region of mixed active/inactive MS lesions. Considering that TAAR1-mediated anti-inflammatory effects have been previously reported, decreased mRNA in MS patients suggests possible pathophysiologic relevance. A shift in TAAR1 localization following pro-inflammatory activation suggests its function is altered in pro-inflammatory states, while TAAR1-expressing macrophages/microglia bordering an MS lesion supports TAAR1 as a novel pharmacological target in cells directly implicated in MS neuroinflammation.
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; (D.A.B.); (M.D.B.)
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John’s, NL A1B 3V6, Canada;
| | - Dylan A. Galloway
- 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, Ophthalmology and Rare Diseases DTA, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche, 4070 Basel, Switzerland;
| | - Mark D. Berry
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John’s, NL A1B 3X9, Canada; (D.A.B.); (M.D.B.)
| | - 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;
- Correspondence: ; Tel.: +1-709-864-4955
| |
Collapse
|
12
|
Alessio N, Squillaro T, Lettiero I, Galano G, De Rosa R, Peluso G, Galderisi U, Di Bernardo G. Biomolecular Evaluation of Piceatannol's Effects in Counteracting the Senescence of Mesenchymal Stromal Cells: A New Candidate for Senotherapeutics? Int J Mol Sci 2021; 22:11619. [PMID: 34769049 PMCID: PMC8583715 DOI: 10.3390/ijms222111619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Several investigations on senescence and its causative role in aging have underscored the importance of developing senotherapeutics, a field focused on killing senescent cells and/or preventing their accumulation within tissues. Using polyphenols in counteracting senescence may facilitate the development of senotherapeutics given their presence in the human diet, their confirmed tolerability and absence of severe side effects, and their role in preventing senescence and inducing the death of senescent cells. Against that background, we evaluated the effect of piceatannol, a natural polyphenol, on the senescence of mesenchymal stromal cells (MSCs), which play a key role in the body's homeostasis. Among our results, piceatannol reduced the number of senescent cells both after genotoxic stress that induced acute senescence and in senescent replicative cultures. Such senotherapeutics activity, moreover, promoted the recovery of cell proliferation and the stemness properties of MSCs. Altogether, our findings demonstrate piceatannol's effectiveness in counteracting senescence by targeting its associated pathways and detecting and affecting P53-dependent and P53-independent senescence. Our study thus suggests that, given piceatannol's various mechanisms to accomplish its pleiotropic activities, it may be able to counteract any senescent phenotypes.
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
| | - Tiziana Squillaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
| | - Ida Lettiero
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
| | - Giovanni Galano
- ASL Napoli 1 Centro P.S.I. Napoli Est-Barra, 80147 Naples, Italy; (G.G.); (R.D.R.)
| | - Roberto De Rosa
- ASL Napoli 1 Centro P.S.I. Napoli Est-Barra, 80147 Naples, Italy; (G.G.); (R.D.R.)
| | - Gianfranco Peluso
- Research Institute on Terrestrial Ecosystems, CNR, 80131 Naples, Italy;
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
13
|
Trace amine-associated receptor 1 (TAAR1): Potential application in mood disorders: A systematic review. Neurosci Biobehav Rev 2021; 131:192-210. [PMID: 34537265 DOI: 10.1016/j.neubiorev.2021.09.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 12/29/2022]
Abstract
There is a need for innovation with respect to therapeutics in psychiatry. Available evidence indicates that the trace amine-associated receptor 1 (TAAR1) agonist SEP-363856 is promising, as it improves measures of cognitive and reward function in schizophrenia. Hedonic and cognitive impairments are transdiagnostic and constitute major burdens in mood disorders. Herein, we systematically review the behavioural and genetic literature documenting the role of TAAR1 in reward and cognitive function, and propose a mechanistic model of TAAR1's functions in the brain. Notably, TAAR1 activity confers antidepressant-like effects, enhances attention and response inhibition, and reduces compulsive reward seeking without impairing normal function. Further characterization of the responsible mechanisms suggests ion-homeostatic, metabolic, neurotrophic, and anti-inflammatory enhancements in the limbic system. Multiple lines of evidence establish the viability of TAAR1 as a biological target for the treatment of mood disorders. Furthermore, the evidence suggests a role for TAAR1 in reward and cognitive function, which is attributed to a cascade of events that are relevant to the cellular integrity and function of the central nervous system.
Collapse
|
14
|
Markin PA, Brito A, Moskaleva NE, Tagliaro F, La Frano MR, Savitskii MV, Appolonova SA. Short- and long-term exposures of the synthetic cannabinoid 5F-APINAC induce metabolomic alterations associated with neurotransmitter systems and embryotoxicity confirmed by teratogenicity in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2021; 243:109000. [PMID: 33561556 DOI: 10.1016/j.cbpc.2021.109000] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/21/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Synthetic cannabinoids are abused substances with strong psychoactive effects. Little is known about the effects on neurotransmission and the toxicity of the second-generation cannabinoid 5F-APINAC. The objective was to assess the influence of short- and long-term exposures of 5F-APINAC on metabolites associated with neurotransmission on zebrafish. METHODS Short-term ("acute", 4 h) and long-term ("chronic", 96 h) exposures to 5F-APINAC were performed at 0.001, 0.01, 0.1, 1.0 and 10 μM. Intervention groups were compared with a vehicle control. Each group n = 20 zebrafish eggs/larvae. Metabolites related to neurotransmission were determined. RESULTS In chronic exposure, larvae exposed to 10 μM 5F-APINAC presented morphological and developmental alterations. GABA had the lowest concentrations at higher exposure in acute (p < 0.01) and chronic (p < 0.001) experiments. Glutamine showed a descending trend in the acute experiment, but an ascending trend in the chronic exposure (p < 0.05). In chronic exposure, tryptophan presented an overall descending trend, but with a neat increase at 10 μM 5F-APINAC (p < 0.001). Tryptamine in acute exposure presented lower (p < 0.05) concentrations at higher doses. Dopamine and acetylcholine presented highest (p < 0.05) concentrations in the acute and chronic exposures, but with a drop at the highest doses in the chronic experiments. In chronic exposure, xanthurenic acid decreased, except for the highest dose. Picolinic acid was increased at the highest doses in the chronic experiment (p < 0.001). CONCLUSIONS Short- and long-term exposures induced metabolomic alterations associated with the gamma-aminobutyric acid/glutamic acid, dopaminergic/adrenergic, cholinergic neurotransmitter systems, and the kynurenine pathway. Chronic exposure at 10 μM 5F-APINAC was associated with embryotoxicity confirmed by teratogenesis.
Collapse
Affiliation(s)
- Pavel A Markin
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; PhD Program in Nanosciences and Advanced Technologies, University of Verona, Verona, Italy; I.M. Sechenov First Moscow State Medical University, Russia
| | - Alex Brito
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Natalia E Moskaleva
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; World-Class Research Center "Digital biodesign and personalized healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Franco Tagliaro
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Michael R La Frano
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, CA, USA; Cal Poly Metabolomics Service Center, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Mark V Savitskii
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; I.M. Sechenov First Moscow State Medical University, Russia
| | - Svetlana A Appolonova
- Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
15
|
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
|
16
|
Zhukov IS, Kubarskaya LG, Tissen IY, Kozlova AA, Dagayev SG, Kashuro VA, Vlasova OL, Sinitca EL, Karpova IV, Gainetdinov RR. Minimal Age-Related Alterations in Behavioral and Hematological Parameters in Trace Amine-Associated Receptor 1 (TAAR1) Knockout Mice. Cell Mol Neurobiol 2020; 40:273-282. [PMID: 31399838 PMCID: PMC11448943 DOI: 10.1007/s10571-019-00721-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/04/2019] [Indexed: 10/26/2022]
Abstract
Since the discovery in 2001, the G protein-coupled trace amine-associated receptor 1 (TAAR1) has become an important focus of research targeted on evaluation of its role in the central nervous system (CNS). Meanwhile, impact of TAAR1 in the peripheral organs is less investigated. Expression of TAAR1 was demonstrated in different peripheral tissues: pancreatic β-cells, stomach, intestines, white blood cells (WBC), and thyroid. However, the role of TAAR1 in regulation of hematological parameters has not been investigated yet. In this study, we performed analysis of anxiety-related behaviors, a complete blood count (CBC), erythrocyte fragility, as well as FT3/FT4 thyroid hormones levels in adult and middle-aged TAAR1 knockout mice. Complete blood count analysis was performed on a Siemens Advia 2120i hematology analyzer and included more than 35 measured and calculated parameters. Erythrocyte fragility test evaluated spherocytosis pathologies of red blood cells (RBC). No significant alterations in essentially all these parameters were found in mice without TAAR1. However, comparative aging analysis has revealed a decreased neutrophils level in the middle-aged TAAR1 knockout mouse group. Minimal alterations in these parameters observed in TAAR1 knockout mice suggest that future TAAR1-based therapies should exert little hematological effect and thus will likely have a good safety profile.
Collapse
Affiliation(s)
- I S Zhukov
- Institute of Translational Biomedicine and Saint Petersburg University Hospital, Saint Petersburg State University, Universitetskaya nab., 7-9, Saint Petersburg, Russia, 199034.
- Institute of Experimental Medicine, Acad. Pavlov str. 12, Saint Petersburg, Russia, 197376.
| | - L G Kubarskaya
- Institute of Toxicology of Federal Medical-Biological Agency, Bekhtereva str. 1., Saint Petersburg, Russia, 192019
| | - I Y Tissen
- Institute of Experimental Medicine, Acad. Pavlov str. 12, Saint Petersburg, Russia, 197376
| | - A A Kozlova
- Institute of Translational Biomedicine and Saint Petersburg University Hospital, Saint Petersburg State University, Universitetskaya nab., 7-9, Saint Petersburg, Russia, 199034
| | - S G Dagayev
- Institute of Toxicology of Federal Medical-Biological Agency, Bekhtereva str. 1., Saint Petersburg, Russia, 192019
| | - V A Kashuro
- Institute of Toxicology of Federal Medical-Biological Agency, Bekhtereva str. 1., Saint Petersburg, Russia, 192019
| | - O L Vlasova
- Peter the Great Saint Petersburg Polytechnic University, Polytechnicheskaya str. 29, Saint Petersburg, Russia, 195251
| | - E L Sinitca
- Institute of Experimental Medicine, Acad. Pavlov str. 12, Saint Petersburg, Russia, 197376
| | - I V Karpova
- Institute of Experimental Medicine, Acad. Pavlov str. 12, Saint Petersburg, Russia, 197376
| | - R R Gainetdinov
- Institute of Translational Biomedicine and Saint Petersburg University Hospital, Saint Petersburg State University, Universitetskaya nab., 7-9, Saint Petersburg, Russia, 199034
| |
Collapse
|
17
|
Bugda Gwilt K, González DP, Olliffe N, Oller H, Hoffing R, Puzan M, El Aidy S, Miller GM. Actions of Trace Amines in the Brain-Gut-Microbiome Axis via Trace Amine-Associated Receptor-1 (TAAR1). Cell Mol Neurobiol 2020; 40:191-201. [PMID: 31836967 PMCID: PMC11448870 DOI: 10.1007/s10571-019-00772-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022]
Abstract
Trace amines and their primary receptor, Trace Amine-Associated Receptor-1 (TAAR1) are widely studied for their involvement in the pathogenesis of neuropsychiatric disorders despite being found in the gastrointestinal tract at physiological levels. With the emergence of the "brain-gut-microbiome axis," we take the opportunity to review what is known about trace amines in the brain, the defined sources of trace amines in the gut, and emerging understandings on the levels of trace amines in various gastrointestinal disorders. Similarly, we discuss localization of TAAR1 expression in the gut, novel findings that TAAR1 may be implicated in inflammatory bowel diseases, and the reported comorbidities of neuropsychiatric disorders and gastrointestinal disorders. With the emergence of TAAR1 specific compounds as next-generation therapeutics for schizophrenia (Roche) and Parkinson's related psychoses (Sunovion), we hypothesize a therapeutic benefit of these compounds in clinical trials in the brain-gut-microbiome axis, as well as a potential for thoughtful manipulation of the brain-gut-microbiome axis to modulate symptoms of neuropsychiatric disease.
Collapse
Affiliation(s)
- Katlynn Bugda Gwilt
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA.
- Center for Drug Discovery, Northeastern University, Boston, MA, USA.
- Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, USA.
| | - Dulce Pamela González
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Neva Olliffe
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Department of Biology, College of Science, Northeastern University, Boston, MA, USA
| | - Haley Oller
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Rachel Hoffing
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Department of Biology, College of Science, Northeastern University, Boston, MA, USA
| | - Marissa Puzan
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, USA
| | - Sahar El Aidy
- Department of Molecular Immunology and Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Gregory M Miller
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
18
|
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 PMCID: PMC6744391 DOI: 10.1124/mol.119.116798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
19
|
Bugda Gwilt K, Olliffe N, Hoffing RA, Miller GM. Trace amine associated receptor 1 (TAAR1) expression and modulation of inflammatory cytokine production in mouse bone marrow-derived macrophages: a novel mechanism for inflammation in ulcerative colitis. Immunopharmacol Immunotoxicol 2019; 41:577-585. [DOI: 10.1080/08923973.2019.1672178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Katlynn Bugda Gwilt
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - Neva Olliffe
- Department of Biology, Northeastern University, Boston, MA, USA
- Honors Program, Northeastern University, Boston, MA, USA
| | | | - Gregory M. Miller
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
20
|
TAAR1 levels and sub-cellular distribution are cell line but not breast cancer subtype specific. Histochem Cell Biol 2019; 152:155-166. [PMID: 31111198 DOI: 10.1007/s00418-019-01791-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2019] [Indexed: 12/23/2022]
Abstract
Trace amine-associated receptors are G protein-coupled receptors of which TAAR1 is the most well-studied. Recently, Vattai et al. (J Cancer Res Clin Oncol 143:1637-1647 https://doi.org/10.1007/s00432-017-2420-8 , 2017) reported that expression of TAAR1 may be a marker of breast cancer (BC) survival, with a positive correlation also suggested between TAAR1 expression and HER2 positivity. Neither a role for TAAR1 in breast tissue, nor in cancer, had previously been suspected. We, therefore, sought to provide independent validation and to further examine these putative relationships. First, a bioinformatic analysis on 58 total samples including normal breast tissue, BC-related cell lines, and tumour samples representing different BC sub-types found no clear correlation between TAAR1 mRNA levels and any BC subtype, including HER2 + . We next confirmed the bioinformatics data correlated to protein expression using a well validated anti-human TAAR1 antibody. TAAR1 mRNA levels correlated with the relative intensity of immunofluorescence staining in six BC cell lines (MCF-7, T47D, MDA-MB-231, SKBR3, MDA-MB-468, BT-474), but not in the MCF-10A immortalized mammary gland line, which had high mRNA but low protein levels. As expected, TAAR1 protein was intracellular in all cell lines. Surprisingly MCF-7, SKBR3, and MDA-MB-468 showed pronounced nuclear localization. The relative protein expression in MCF-7, MDA-MB-231, and MCF-10A lines was further confirmed by semi-quantitative flow cytometry. Finally, we demonstrate that the commercially available anti-TAAR1 antibody has poor selectivity, which likely explains the lack of correlation with the previous study. Therefore, while we clearly demonstrate variable expression and sub-cellular localization of TAAR1 across BC cell lines, we find no evidence for association with BC subtype.
Collapse
|
21
|
Yatham VR, Bellotti P, König B. Decarboxylative hydrazination of unactivated carboxylic acids by cerium photocatalysis. Chem Commun (Camb) 2019; 55:3489-3492. [PMID: 30829348 PMCID: PMC6498423 DOI: 10.1039/c9cc00492k] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We report the cerium photocatalyzed radical decarboxylative hydrazination of carboxylic acids with di-tert-butylazodicarboxylate (DBAD). The operationally simple protocol provides rapid access to synthetically useful hydrazine derivatives and overcomes current scope limitations in the photoredox-catalyzed decarboxylation of carboxylic acids.
Collapse
Affiliation(s)
- Veera Reddy Yatham
- Institut für Organische Chemie, Fakultät für Chemie und Pharmazie, Universität Regensburg, Universitätstraße 31, D-93053 Regensburg, Germany.
| | | | | |
Collapse
|
22
|
Li X, Pongkitwitoon S, Lu H, Lee C, Gelberman R, Thomopoulos S. CTGF induces tenogenic differentiation and proliferation of adipose-derived stromal cells. J Orthop Res 2019; 37:574-582. [PMID: 30756417 PMCID: PMC6467286 DOI: 10.1002/jor.24248] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 01/21/2019] [Indexed: 02/04/2023]
Abstract
Intrasynovial tendons are paucicellular and hypovascular, resulting in a poor response to injury. Surgical repair of ruptured or lacerated tendons often lead to complications such as adhesions, repair site gapping, and repair site rupture. Adipose-derived stem cells (ASCs) have shown promise for enhancing tendon repair, as they have the capacity to differentiate into tendon fibroblasts and augment the healing response. Furthermore, connective tissue growth factor (CTGF) has been shown to promote tendon regeneration via the stimulation of endogenous tendon stem cells. Here, we evaluated the potential of CTGF to promote tenogenic differentiation of ASCs in vitro. Gene and protein expression, cell proliferation, and FAK and ERK1/2 signaling were assessed. CTGF increased tenogenic genes in mouse ASCs in a dose- and time-dependent manner. Western blot and immunostaining analyses demonstrated increases in tenogenic protein expression in CTGF-treated ASCs at all timepoints studied. CTGF increased ASC proliferation in a dose-dependent manner. CTGF induced phosphorylation of ERK1/2 within 5 min and FAK within 15 min; both signals persisted for 120 min. Blocking FAK and ERK1/2 pathways by selective inhibitors SCH772984 and PF573228, respectively, attenuated the CTGF-induced tenogenic differentiation and proliferation of ASCs. These results suggest that CTGF induces tenogenic differentiation of ASCs via the FAK and ERK1/2 pathway. Statement of clinical significance: Although prior research has led to advances in tendon operative techniques and rehabilitation methods, clinical outcomes after tendon repair remain variable, with high rates of repair site gapping or rupture. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Xiaoning Li
- Xiangya Hospital, Central South University, Changsha, PR China,Department of Orthopedic Surgery, Columbia University, 650W 168th St, New York 10032 New York,Department of Biomedical Engineering, Columbia University, 650W 168th St, New York 10032 New York
| | - Suphannee Pongkitwitoon
- Department of Orthopedic Surgery, Columbia University, 650W 168th St, New York 10032 New York,Department of Biomedical Engineering, Columbia University, 650W 168th St, New York 10032 New York
| | - Hongbin Lu
- Xiangya Hospital, Central South University, Changsha, PR China
| | - Chang Lee
- College of Dental Medicine, Columbia University, New York, New York
| | - Richard Gelberman
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, 650W 168th St, New York 10032 New York,Department of Biomedical Engineering, Columbia University, 650W 168th St, New York 10032 New York
| |
Collapse
|
23
|
Maßberg D, Hatt H. Human Olfactory Receptors: Novel Cellular Functions Outside of the Nose. Physiol Rev 2018; 98:1739-1763. [PMID: 29897292 DOI: 10.1152/physrev.00013.2017] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Olfactory receptors (ORs) are not exclusively expressed in the olfactory sensory neurons; they are also observed outside of the olfactory system in all other human tissues tested to date, including the testis, lung, intestine, skin, heart, and blood. Within these tissues, certain ORs have been determined to be exclusively expressed in only one tissue, whereas other ORs are more widely distributed in many different tissues throughout the human body. For most of the ectopically expressed ORs, limited data are available for their functional roles. They have been shown to be involved in the modulation of cell-cell recognition, migration, proliferation, the apoptotic cycle, exocytosis, and pathfinding processes. Additionally, there is a growing body of evidence that they have the potential to serve as diagnostic and therapeutic tools, as ORs are highly expressed in different cancer tissues. Interestingly, in addition to the canonical signaling pathways activated by ORs in olfactory sensory neurons, alternative pathways have been demonstrated in nonolfactory tissues. In this review, the existing data concerning the expression, as well as the physiological and pathophysiological functions, of ORs outside of the nose are highlighted to provide insights into future lines of research.
Collapse
Affiliation(s)
- Désirée Maßberg
- Ruhr-University Bochum, Department of Cell Physiology , Bochum , Germany
| | - Hanns Hatt
- Ruhr-University Bochum, Department of Cell Physiology , Bochum , Germany
| |
Collapse
|
24
|
Dave S, Chen L, Yu C, Seaton M, Khodr CE, Al-Harthi L, Hu XT. Methamphetamine decreases K + channel function in human fetal astrocytes by activating the trace amine-associated receptor type-1. J Neurochem 2018; 148:29-45. [PMID: 30295919 DOI: 10.1111/jnc.14606] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 12/22/2022]
Abstract
Methamphetamine (Meth) is a potent and commonly abused psychostimulant. Meth alters neuron and astrocyte activity; yet the underlying mechanism(s) is not fully understood. Here we assessed the impact of acute Meth on human fetal astrocytes (HFAs) using whole-cell patch-clamping. We found that HFAs displayed a large voltage-gated K+ efflux (IKv ) through Kv /Kv -like channels during membrane depolarization, and a smaller K+ influx (Ikir ) via inward-rectifying Kir /Kir -like channels during membrane hyperpolarization. Meth at a 'recreational' (20 μM) or toxic/fatal (100 μM) concentration depolarized resting membrane potential (RMP) and suppressed IKv/Kv-like . These changes were associated with a decreased time constant (Ƭ), and mimicked by blocking the two-pore domain K+ (K2P )/K2P -like and Kv /Kv -like channels, respectively. Meth also diminished IKir/Kir-like , but only at toxic/fatal levels. Given that Meth is a potent agonist for the trace amine-associated receptor type-1 (TAAR1), and TAAR1-coupled cAMP/cAMP-activated protein kinase (PKA) cascade, we further evaluated whether the Meth impact on K+ efflux was mediated by this pathway. We found that antagonizing TAAR1 with N-(3-Ethoxyphenyl)-4-(1-pyrrolidinyl)-3-(trifluoromethyl)benzamide (EPPTB) reversed Meth-induced suppression of IKv/Kv-like ; and inhibiting PKA activity by H89 abolished Meth effects on suppressing IKv/Kv-like . Antagonizing TAAR1 might also attenuate Meth-induced RMP depolarization. Voltage-gated Ca2+ currents were not detected in HFAs. These novel findings demonstrate that Meth suppresses IKv/Kv-like by facilitating the TAAR1/Gs /cAMP/PKA cascade and altering the kinetics of Kv /Kv -like channel gating, but reduces K2P /K2P -like channel activity through other pathway(s), in HFAs. Given that Meth-induced decrease in astrocytic K+ efflux through K2P /K2P -like and Kv /Kv -like channels reduces extracellular K+ levels, such reduction could consequently contribute to a decreased excitability of surrounding neurons. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Sonya Dave
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Lihua Chen
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Chunjiang Yu
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Melanie Seaton
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Christina E Khodr
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Lena Al-Harthi
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| | - Xiu-Ti Hu
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
25
|
Abstract
Trace amines are endogenous compounds classically regarded as comprising β-phenylethyalmine, p-tyramine, tryptamine, p-octopamine, and some of their metabolites. They are also abundant in common foodstuffs and can be produced and degraded by the constitutive microbiota. The ability to use trace amines has arisen at least twice during evolution, with distinct receptor families present in invertebrates and vertebrates. The term "trace amine" was coined to reflect the low tissue levels in mammals; however, invertebrates have relatively high levels where they function like mammalian adrenergic systems, involved in "fight-or-flight" responses. Vertebrates express a family of receptors termed trace amine-associated receptors (TAARs). Humans possess six functional isoforms (TAAR1, TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9), whereas some fish species express over 100. With the exception of TAAR1, TAARs are expressed in olfactory epithelium neurons, where they detect diverse ethological signals including predators, spoiled food, migratory cues, and pheromones. Outside the olfactory system, TAAR1 is the most thoroughly studied and has both central and peripheral roles. In the brain, TAAR1 acts as a rheostat of dopaminergic, glutamatergic, and serotonergic neurotransmission and has been identified as a novel therapeutic target for schizophrenia, depression, and addiction. In the periphery, TAAR1 regulates nutrient-induced hormone secretion, suggesting its potential as a novel therapeutic target for diabetes and obesity. TAAR1 may also regulate immune responses by regulating leukocyte differentiation and activation. This article provides a comprehensive review of the current state of knowledge of the evolution, physiologic functions, pharmacology, molecular mechanisms, and therapeutic potential of trace amines and their receptors in vertebrates and invertebrates.
Collapse
Affiliation(s)
- Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); Skolkovo Institute of Science and Technology (Skoltech), Moscow, Russia (R.R.G.); Neuroscience, Ophthalmology, and Rare Diseases Discovery and Translational Area, pRED, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (M.C.H.); and Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada (M.D.B.)
| | - Marius C Hoener
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); Skolkovo Institute of Science and Technology (Skoltech), Moscow, Russia (R.R.G.); Neuroscience, Ophthalmology, and Rare Diseases Discovery and Translational Area, pRED, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (M.C.H.); and Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada (M.D.B.)
| | - Mark D Berry
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); Skolkovo Institute of Science and Technology (Skoltech), Moscow, Russia (R.R.G.); Neuroscience, Ophthalmology, and Rare Diseases Discovery and Translational Area, pRED, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (M.C.H.); and Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada (M.D.B.)
| |
Collapse
|
26
|
Fleischer LM, Somaiya RD, Miller GM. Review and Meta-Analyses of TAAR1 Expression in the Immune System and Cancers. Front Pharmacol 2018; 9:683. [PMID: 29997511 PMCID: PMC6029583 DOI: 10.3389/fphar.2018.00683] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 06/06/2018] [Indexed: 12/29/2022] Open
Abstract
Since its discovery in 2001, the major focus of TAAR1 research has been on its role in monoaminergic regulation, drug-induced reward and psychiatric conditions. More recently, TAAR1 expression and functionality in immune system regulation and immune cell activation has become a topic of emerging interest. Here, we review the immunologically-relevant TAAR1 literature and incorporate open-source expression and cancer survival data meta-analyses. We provide strong evidence for TAAR1 expression in the immune system and cancers revealed through NCBI GEO datamining and discuss its regulation in a spectrum of immune cell types as well as in numerous cancers. We discuss connections and logical directions for further study of TAAR1 in immunological function, and its potential role as a mediator or modulator of immune dysregulation, immunological effects of psychostimulant drugs of abuse, and cancer progression.
Collapse
Affiliation(s)
- Lisa M Fleischer
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Rachana D Somaiya
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Gregory M Miller
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States.,Department of Chemical Engineering, Northeastern University, Boston, MA, United States.,Center for Drug Discovery, Northeastern University, Boston, MA, United States
| |
Collapse
|
27
|
Liu JF, Li JX. TAAR1 in Addiction: Looking Beyond the Tip of the Iceberg. Front Pharmacol 2018; 9:279. [PMID: 29636691 PMCID: PMC5881156 DOI: 10.3389/fphar.2018.00279] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/12/2018] [Indexed: 11/23/2022] Open
Abstract
Trace-amine associated receptor 1 (TAAR1) is the best-characterized member of the family of TAARs. TAAR1 is broadly expressed in the brain, especially within the monoaminergic systems. Evidence from electrophysiological and neurochemical studies evaluating the effects of genetic and pharmacological interventions on TAAR1 revealed that TAAR1 modulates transmission of monoamines, especially dopamine. TAAR1 agonists dampened drugs of abuse-induced dopamine accumulation. In general, TAAR1 agonists specifically inhibited the rewarding and reinforcing effects of drugs of abuse and drug-abuse related behaviors. Details of the mechanism of TAAR1 remain elusive; however, it is thought to be regulated by its interactions with D2 receptors. In addition, the alternative cellular mechanism such as an interaction between TAAR1 and D3 may also participate in the action of TAAR1 agonists. Further studies are required to investigate the role of TAAR1 in other drugs of abuse-related behaviors and the underlying neural mechanisms. Collectively, TAAR1 negatively modulates dopaminergic systems and dopamine-related behaviors and TAAR1 agonists are promising pharmacotherapy to treat drug addiction and relapse.
Collapse
Affiliation(s)
- Jian-Feng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States.,School of Pharmacy, Yantai University, Yantai, China
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
28
|
Zhang X, Mantas I, Alvarsson A, Yoshitake T, Shariatgorji M, Pereira M, Nilsson A, Kehr J, Andrén PE, Millan MJ, Chergui K, Svenningsson P. Striatal Tyrosine Hydroxylase Is Stimulated via TAAR1 by 3-Iodothyronamine, But Not by Tyramine or β-Phenylethylamine. Front Pharmacol 2018; 9:166. [PMID: 29545750 PMCID: PMC5837966 DOI: 10.3389/fphar.2018.00166] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 02/14/2018] [Indexed: 01/16/2023] Open
Abstract
The trace amine-associated receptor 1 (TAAR1) is expressed by dopaminergic neurons, but the precise influence of trace amines upon their functional activity remains to be fully characterized. Here, we examined the regulation of tyrosine hydroxylase (TH) by tyramine and beta-phenylethylamine (β-PEA) compared to 3-iodothyronamine (T1AM). Immunoblotting and amperometry were performed in dorsal striatal slices from wild-type (WT) and TAAR1 knockout (KO) mice. T1AM increased TH phosphorylation at both Ser19 and Ser40, actions that should promote functional activity of TH. Indeed, HPLC data revealed higher rates of L-dihydroxyphenylalanine (DOPA) accumulation in WT animals treated with T1AM after the administration of a DOPA decarboxylase inhibitor. These effects were abolished both in TAAR1 KO mice and by the TAAR1 antagonist, EPPTB. Further, they were specific inasmuch as Ser845 phosphorylation of the post-synaptic GluA1 AMPAR subunit was unaffected. The effects of T1AM on TH phosphorylation at both Ser19 (CamKII-targeted), and Ser40 (PKA-phosphorylated) were inhibited by KN-92 and H-89, inhibitors of CamKII and PKA respectively. Conversely, there was no effect of an EPAC analog, 8-CPT-2Me-cAMP, on TH phosphorylation. In line with these data, T1AM increased evoked striatal dopamine release in TAAR1 WT mice, an action blunted in TAAR1 KO mice and by EPPTB. Mass spectrometry imaging revealed no endogenous T1AM in the brain, but detected T1AM in several brain areas upon systemic administration in both WT and TAAR1 KO mice. In contrast to T1AM, tyramine decreased the phosphorylation of Ser40-TH, while increasing Ser845-GluA1 phosphorylation, actions that were not blocked in TAAR1 KO mice. Likewise, β-PEA reduced Ser40-TH and tended to promote Ser845-GluA1 phosphorylation. The D1 receptor antagonist SCH23390 blocked tyramine-induced Ser845-GluA1 phosphorylation, but had no effect on tyramine- or β-PEA-induced Ser40-TH phosphorylation. In conclusion, by intracellular cascades involving CaMKII and PKA, T1AM, but not tyramine and β-PEA, acts via TAAR1 to promote the phosphorylation and functional activity of TH in the dorsal striatum, supporting a modulatory influence on dopamine transmission.
Collapse
Affiliation(s)
- Xiaoqun Zhang
- Section of Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine L8:01, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Mantas
- Section of Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine L8:01, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Alexandra Alvarsson
- Section of Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine L8:01, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Takashi Yoshitake
- Section of Pharmacological Neurochemistry, Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Mohammadreza Shariatgorji
- Biomolecular Mass Spectrometry Imaging, National Resource for Mass Spectrometry Imaging, Science for Life Laboratory, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Marcela Pereira
- Section of Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine L8:01, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Nilsson
- Biomolecular Mass Spectrometry Imaging, National Resource for Mass Spectrometry Imaging, Science for Life Laboratory, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Jan Kehr
- Section of Pharmacological Neurochemistry, Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Per E Andrén
- Biomolecular Mass Spectrometry Imaging, National Resource for Mass Spectrometry Imaging, Science for Life Laboratory, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Mark J Millan
- Centre for Therapeutic Innovation-CNS, Institut de Recherches Servier, Centre de Recherches de Croissy, Paris, France
| | - Karima Chergui
- Section of Molecular Neurophysiology, Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Per Svenningsson
- Section of Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine L8:01, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
29
|
Bellusci L, Laurino A, Sabatini M, Sestito S, Lenzi P, Raimondi L, Rapposelli S, Biagioni F, Fornai F, Salvetti A, Rossi L, Zucchi R, Chiellini G. New Insights into the Potential Roles of 3-Iodothyronamine (T1AM) and Newly Developed Thyronamine-Like TAAR1 Agonists in Neuroprotection. Front Pharmacol 2017; 8:905. [PMID: 29311919 PMCID: PMC5732922 DOI: 10.3389/fphar.2017.00905] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/28/2017] [Indexed: 12/30/2022] Open
Abstract
3-Iodothyronamine (T1AM) is an endogenous high-affinity ligand of the trace amine-associated receptor 1 (TAAR1), detected in mammals in many organs, including the brain. Recent evidence indicates that pharmacological TAAR1 activation may offer a novel therapeutic option for the treatment of a wide range of neuropsychiatric and metabolic disorders. To assess potential neuroprotection by TAAR1 agonists, in the present work, we initially investigated whether T1AM and its corresponding 3-methylbiaryl-methane analog SG-2 can improve learning and memory when systemically administered to mice at submicromolar doses, and whether these effects are modified under conditions of MAO inhibition by clorgyline. Our results revealed that when i.p. injected to mice, both T1AM and SG-2 produced memory-enhancing and hyperalgesic effects, while increasing ERK1/2 phosphorylation and expression of transcription factor c-fos. Notably, both compounds appeared to rely on the action of ubiquitous enzymes MAO to produce the corresponding oxidative metabolites that were then able to activate the histaminergic system. Since autophagy is key for neuronal plasticity, in a second line of experiments we explored whether T1AM and synthetic TAAR1 agonists SG1 and SG2 were able to induce autophagy in human glioblastoma cell lines (U-87MG). After treatment of U-87MG cells with 1 μM T1AM, SG-1, SG-2 transmission electron microscopy (TEM) and immunofluorescence (IF) showed a significant time-dependent increase of autophagy vacuoles and microtubule-associated protein 1 light chain 3 (LC3). Consistently, Western blot analysis revealed a significant increase of the LC3II/LC3I ratio, with T1AM and SG-1 being the most effective agents. A decreased level of the p62 protein was also observed after treatment with T1AM and SG-1, which confirms the efficacy of these compounds as autophagy inducers in U-87MG cells. In the process to dissect which pathway induces ATG, the effects of these compounds were evaluated on the PI3K-AKT-mTOR pathway. We found that 1 μM T1AM, SG-1 and SG-2 decreased pAKT/AKT ratio at 0.5 and 4 h after treatment, suggesting that autophagy is induced by inhibiting mTOR phosphorylation by PI3K-AKT-mTOR pathway. In conclusion, our study shows that T1AM and thyronamine-like derivatives SG-1 and SG-2 might represent valuable tools to therapeutically intervene with neurological disorders.
Collapse
Affiliation(s)
- Lorenza Bellusci
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Pisa, Italy
| | - Annunziatina Laurino
- Section of Pharmacology and Toxicology, Department of Psychology, Neurology, Drug Sciences, Health of the Child, Pharmacology, University of Florence, Florence, Italy
| | - Martina Sabatini
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Pisa, Italy
| | - Simona Sestito
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Paola Lenzi
- Unit of Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Laura Raimondi
- Section of Pharmacology and Toxicology, Department of Psychology, Neurology, Drug Sciences, Health of the Child, Pharmacology, University of Florence, Florence, Italy
| | - Simona Rapposelli
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Francesco Fornai
- Unit of Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Alessandra Salvetti
- Unit of Experimental Biology and Genetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Leonardo Rossi
- Unit of Experimental Biology and Genetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Riccardo Zucchi
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Pisa, Italy
| | - Grazia Chiellini
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Pisa, Italy
| |
Collapse
|
30
|
Miner NB, Elmore JS, Baumann MH, Phillips TJ, Janowsky A. Trace amine-associated receptor 1 regulation of methamphetamine-induced neurotoxicity. Neurotoxicology 2017; 63:57-69. [PMID: 28919515 PMCID: PMC5683899 DOI: 10.1016/j.neuro.2017.09.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/13/2017] [Accepted: 09/14/2017] [Indexed: 12/20/2022]
Abstract
Trace amine-associated receptor 1 (TAAR1) is activated by methamphetamine (MA) and modulates dopaminergic (DA) function. Although DA dysregulation is the hallmark of MA-induced neurotoxicity leading to behavioral and cognitive deficits, the intermediary role of TAAR1 has yet to be characterized. To investigate TAAR1 regulation of MA-induced neurotoxicity, Taar1 transgenic knock-out (KO) and wildtype (WT) mice were administered saline or a neurotoxic regimen of 4 i.p. injections, 2h apart, of MA (2.5, 5, or 10mg/kg). Temperature data were recorded during the treatment day. Additionally, striatal tissue was collected 2 or 7days following MA administration for analysis of DA, 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), and tyrosine hydroxylase (TH) levels, as well as glial fibrillary acidic protein (GFAP) expression. MA elicited an acute hypothermic drop in body temperature in Taar1-WT mice, but not in Taar1-KO mice. Two days following treatment, DA and TH levels were lower in Taar1-KO mice compared to Taar1-WT mice, regardless of treatment, and were dose-dependently decreased by MA. GFAP expression was significantly increased by all doses of MA at both time points and greater in Taar1-KO compared to Taar1-WT mice receiving MA 2.5 or 5mg/kg. Seven days later, DA levels were decreased in a similar pattern: DA was significantly lower in Taar1-KO compared to Taar1-WT mice receiving MA 2.5 or 5mg/kg. TH levels were uniformly decreased by MA, regardless of genotype. These results indicate that activation of TAAR1 potentiates MA-induced hypothermia and TAAR1 confers sustained neuroprotection dependent on its thermoregulatory effects.
Collapse
Affiliation(s)
- Nicholas B Miner
- Research Service, VA Portland Health Care System, Portland, OR, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Josh S Elmore
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Tamara J Phillips
- Research Service, VA Portland Health Care System, Portland, OR, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; The Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Aaron Janowsky
- Research Service, VA Portland Health Care System, Portland, OR, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; The Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
31
|
Interaction Between the Trace Amine-Associated Receptor 1 and the Dopamine D 2 Receptor Controls Cocaine's Neurochemical Actions. Sci Rep 2017; 7:13901. [PMID: 29066851 PMCID: PMC5655641 DOI: 10.1038/s41598-017-14472-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/11/2017] [Indexed: 02/07/2023] Open
Abstract
Recent evidence suggests that the trace amine-associated receptor 1 (TAAR1) plays a pivotal role in the regulation of dopamine (DA) transmission and cocaine’s actions. However, the underlying mechanisms through which TAAR1 activation mediates these effects have not yet been elucidated. Here, we used fast-scan cyclic voltammetry to measure DA dynamics and explore such mechanisms. We show, first, that the full TAAR1 agonist, RO5256390, dose-dependently blocked cocaine-induced inhibition of DA clearance in slices of the nucleus accumbens. Second, subthreshold inhibition of PKA or PKC phosphorylation did not prevent TAAR1 suppression of cocaine effects whereas subeffective doses of the DA D2 receptor antagonist, L-741,626, rescued cocaine’s ability to produce changes in DA uptake in the presence of full TAAR1 activation, thus indicating that TAAR1 modulation of cocaine effects requires simultaneous DA D2 receptor activation. Predictably, inhibition of glycogen synthase kinase-3 (GSK-3), which results from activation of D2/TAAR1 heterodimers, fully reproduced the inhibitory effects of TAAR1 activation on cocaine-induced changes in DA transmission. Collectively, the present observations reveal that the ability of TAAR1 to regulate cocaine effects is linked to cooperative interactions with D2 autoreceptors and associated downstream molecular targets converging on GSK-3 and suggest a new mechanism to disrupt cocaine neurochemical actions.
Collapse
|
32
|
Methamphetamine: Effects on the brain, gut and immune system. Pharmacol Res 2017; 120:60-67. [DOI: 10.1016/j.phrs.2017.03.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/08/2017] [Accepted: 03/07/2017] [Indexed: 12/31/2022]
|
33
|
Khan MZ, Nawaz W. The emerging roles of human trace amines and human trace amine-associated receptors (hTAARs) in central nervous system. Biomed Pharmacother 2016; 83:439-449. [PMID: 27424325 DOI: 10.1016/j.biopha.2016.07.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/28/2016] [Accepted: 07/01/2016] [Indexed: 02/06/2023] Open
Abstract
Human trace amines (TAs) are endogenous compounds, previously almost ignored in human pathology for many reasons (difficulty of their measurement in biological fluids, unknown receptors for elusive amines), are now considered to play a significant role in synaptic transmission within the central nervous system (CNS) acting as neuromodulators. The recent discovery of a novel family of G-protein-coupled receptors (GPCRs) that includes individual members that are highly specific for TAs indicates a potential role for TAs as vertebrate neurotransmitters or neuromodulators, although the majority of these GPCRs so far have not been demonstrated to be activated by TAs. Human trace amine receptors (including TAAR1 TAAR2 TAAR5 TAAR6 TAAR8 TAAR9) are expressed in the brain and play significant physiological and neuropathological roles by activation of trace amines. We herein discuss the recent findings that provide insights into the functional roles of human trace amines (including P-Octopamine, β phenylethylamine, Tryptamine, Tyramine, Synephrine, 3-Iodothyronamine, 3-Methoxytyramine, N-Methyltyramine, N-Methylphenethylamine) in brain. Furthermore, we discuss the known functions of human trace amine receptors in brain.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| | - Waqas Nawaz
- School of basic medicine and clinical pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
34
|
Pei Y, Asif-Malik A, Canales JJ. Trace Amines and the Trace Amine-Associated Receptor 1: Pharmacology, Neurochemistry, and Clinical Implications. Front Neurosci 2016; 10:148. [PMID: 27092049 PMCID: PMC4820462 DOI: 10.3389/fnins.2016.00148] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 03/21/2016] [Indexed: 01/30/2023] Open
Abstract
Biogenic amines are a collection of endogenous molecules that play pivotal roles as neurotransmitters and hormones. In addition to the "classical" biogenic amines resulting from decarboxylation of aromatic acids, including dopamine (DA), norepinephrine, epinephrine, serotonin (5-HT), and histamine, other biogenic amines, present at much lower concentrations in the central nervous system (CNS), and hence referred to as "trace" amines (TAs), are now recognized to play significant neurophysiological and behavioral functions. At the turn of the century, the discovery of the trace amine-associated receptor 1 (TAAR1), a phylogenetically conserved G protein-coupled receptor that is responsive to both TAs, such as β-phenylethylamine, octopamine, and tyramine, and structurally-related amphetamines, unveiled mechanisms of action for TAs other than interference with aminergic pathways, laying the foundations for deciphering the functional significance of TAs and its mammalian CNS receptor, TAAR1. Although, its molecular interactions and downstream targets have not been fully elucidated, TAAR1 activation triggers accumulation of intracellular cAMP, modulates PKA and PKC signaling and interferes with the β-arrestin2-dependent pathway via G protein-independent mechanisms. TAAR1 is uniquely positioned to exert direct control over DA and 5-HT neuronal firing and release, which has profound implications for understanding the pathophysiology of, and therefore designing more efficacious therapeutic interventions for, a range of neuropsychiatric disorders that involve aminergic dysregulation, including Parkinson's disease, schizophrenia, mood disorders, and addiction. Indeed, the recent development of novel pharmacological tools targeting TAAR1 has uncovered the remarkable potential of TAAR1-based medications as new generation pharmacotherapies in neuropsychiatry. This review summarizes recent developments in the study of TAs and TAAR1, their intricate neurochemistry and pharmacology, and their relevance for neurodegenerative and neuropsychiatric disease.
Collapse
Affiliation(s)
| | | | - Juan J. Canales
- Department of Neuroscience, Psychology and Behaviour, University of LeicesterLeicester, UK
| |
Collapse
|
35
|
Shi X, Walter NAR, Harkness JH, Neve KA, Williams RW, Lu L, Belknap JK, Eshleman AJ, Phillips TJ, Janowsky A. Genetic Polymorphisms Affect Mouse and Human Trace Amine-Associated Receptor 1 Function. PLoS One 2016; 11:e0152581. [PMID: 27031617 PMCID: PMC4816557 DOI: 10.1371/journal.pone.0152581] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/16/2016] [Indexed: 12/15/2022] Open
Abstract
Methamphetamine (MA) and neurotransmitter precursors and metabolites such as tyramine, octopamine, and β-phenethylamine stimulate the G protein-coupled trace amine-associated receptor 1 (TAAR1). TAAR1 has been implicated in human conditions including obesity, schizophrenia, depression, fibromyalgia, migraine, and addiction. Additionally TAAR1 is expressed on lymphocytes and astrocytes involved in inflammation and response to infection. In brain, TAAR1 stimulation reduces synaptic dopamine availability and alters glutamatergic function. TAAR1 is also expressed at low levels in heart, and may regulate cardiovascular tone. Taar1 knockout mice orally self-administer more MA than wild type and are insensitive to its aversive effects. DBA/2J (D2) mice express a non-synonymous single nucleotide polymorphism (SNP) in Taar1 that does not respond to MA, and D2 mice are predisposed to high MA intake, compared to C57BL/6 (B6) mice. Here we demonstrate that endogenous agonists stimulate the recombinant B6 mouse TAAR1, but do not activate the D2 mouse receptor. Progeny of the B6XD2 (BxD) family of recombinant inbred (RI) strains have been used to characterize the genetic etiology of diseases, but contrary to expectations, BXDs derived 30-40 years ago express only the functional B6 Taar1 allele whereas some more recently derived BXD RI strains express the D2 allele. Data indicate that the D2 mutation arose subsequent to derivation of the original RIs. Finally, we demonstrate that SNPs in human TAAR1 alter its function, resulting in expressed, but functional, sub-functional and non-functional receptors. Our findings are important for identifying a predisposition to human diseases, as well as for developing personalized treatment options.
Collapse
Affiliation(s)
- Xiao Shi
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States of America
- The Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Nicole A. R. Walter
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States of America
| | - John H. Harkness
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Kim A. Neve
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States of America
- The Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Robert W. Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - John K. Belknap
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States of America
- The Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Amy J. Eshleman
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Tamara J. Phillips
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States of America
- The Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Aaron Janowsky
- Veterans Affairs Portland Health Care System, Portland, Oregon, United States of America
- The Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Psychiatry, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
36
|
Grandy DK, Miller GM, Li JX. "TAARgeting Addiction"--The Alamo Bears Witness to Another Revolution: An Overview of the Plenary Symposium of the 2015 Behavior, Biology and Chemistry Conference. Drug Alcohol Depend 2016; 159:9-16. [PMID: 26644139 PMCID: PMC4724540 DOI: 10.1016/j.drugalcdep.2015.11.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/13/2015] [Accepted: 11/14/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND In keeping with the free-thinking tradition San Antonians are known for, the Scientific Program Committee of the Behavior, Biology and Chemistry: Translational Research in Addiction Conference chose trace amine-associated receptor 1 (TAAR1) as the focus of the plenary symposium for its 7th annual meeting held at the University of Texas Health Science Center at San Antonio on March 14 and 15, 2015. The timing of the meeting's plenary session on TAAR1 coincided with the Ides of March, an apt concurrence given the long association of this date with the overthrow of the status quo. And whether aware of the coincidence or not, those in attendance witnessed the plunging of the metaphorical dagger into the heart of the dopamine (DA) transporter (DAT)-centric view of psychostimulant action. METHODS The symposium's four plenary presentations focused on the molecular and cellular biology, genetics, medicinal chemistry and behavioral pharmacology of the TAAR1 system and the experimental use of newly developed selective TAAR1 ligands. RESULTS The consensus was that TAAR1 is a DA and methamphetamine receptor, interacts with DAT and DA D2 receptors, and is essential in modulating addiction-related effects of psychostimulants. CONCLUSIONS Collectively the findings presented during the symposium constitute a significant challenge to the current view that psychostimulants such as methamphetamine and amphetamine solely target DAT to interfere with normal DA signaling and provide a novel conceptual framework from which a more complete understanding of the molecular mechanisms underlying the actions of DA and METH is likely to emerge.
Collapse
Affiliation(s)
- David K. Grandy
- Department of Physiology and Pharmacology, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Gregory M. Miller
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
37
|
Marcinek P, Geithe C, Krautwurst D. Chemosensory G Protein-Coupled Receptors (GPCR) in Blood Leukocytes. TOPICS IN MEDICINAL CHEMISTRY 2016. [DOI: 10.1007/7355_2016_101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
38
|
Seo JW, Jones SM, Hostetter TA, Iliff JJ, West GA. Methamphetamine induces the release of endothelin. J Neurosci Res 2015; 94:170-8. [PMID: 26568405 DOI: 10.1002/jnr.23697] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 10/09/2015] [Accepted: 10/28/2015] [Indexed: 11/09/2022]
Abstract
Methamphetamine is a potent psychostimulant drug of abuse that increases release and blocks reuptake of dopamine, producing intense euphoria, factors that may contribute to its widespread abuse. It also produces severe neurotoxicity resulting from oxidative stress, DNA damage, blood-brain barrier disruption, microgliosis, and mitochondrial dysfunction. Intracerebral hemorrhagic and ischemic stroke have been reported after intravenous and oral abuse of methamphetamine. Several studies have shown that methamphetamine causes vasoconstriction of vessels. This study investigates the effect of methamphetamine on endothelin-1 (ET-1) release in mouse brain endothelial cells by ELISA. ET-1 transcription as well as endothelial nitric oxide synthase (eNOS) activation and transcription were measured following methamphetamine treatment. We also examine the effect of methamphetamine on isolated cerebral arteriolar vessels from C57BL/6 mice. Penetrating middle cerebral arterioles were cannulated at both ends with a micropipette system. Methamphetamine was applied extraluminally, and the vascular response was investigated. Methamphetamine treatment of mouse brain endothelial cells resulted in ET-1 release and a transient increase in ET-1 message. The activity and transcription of eNOS were only slightly enhanced after 24 hr of treatment with methamphetamine. In addition, methamphetamine caused significant vasoconstriction of isolated mouse intracerebral arterioles. The vasoconstrictive effect of methamphetamine was attenuated by coapplication of the endothelin receptor antagonist PD145065. These findings suggest that vasoconstriction induced by methamphetamine is mediated through the endothelin receptor and may involve an endothelin-dependent pathway.
Collapse
Affiliation(s)
- Jeong-Woo Seo
- Neurotrauma Research, Swedish Medical Center, Englewood, Colorado
| | - Susan M Jones
- Neurotrauma Research, Swedish Medical Center, Englewood, Colorado
| | | | - Jeffrey J Iliff
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
39
|
Borgmann K, Ghorpade A. HIV-1, methamphetamine and astrocytes at neuroinflammatory Crossroads. Front Microbiol 2015; 6:1143. [PMID: 26579077 PMCID: PMC4621459 DOI: 10.3389/fmicb.2015.01143] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/05/2015] [Indexed: 12/30/2022] Open
Abstract
As a popular psychostimulant, methamphetamine (METH) use leads to long-lasting, strong euphoric effects. While METH abuse is common in the general population, between 10 and 15% of human immunodeficiency virus-1 (HIV-1) patients report having abused METH. METH exacerbates the severity and onset of HIV-1-associated neurocognitive disorders (HAND) through direct and indirect mechanisms. Repetitive METH use impedes adherence to antiretroviral drug regimens, increasing the likelihood of HIV-1 disease progression toward AIDS. METH exposure also directly affects both innate and adaptive immunity, altering lymphocyte numbers and activity, cytokine signaling, phagocytic function and infiltration through the blood brain barrier. Further, METH triggers the dopamine reward pathway and leads to impaired neuronal activity and direct toxicity. Concurrently, METH and HIV-1 alter the neuroimmune balance and induce neuroinflammation, which modulates a wide range of brain functions including neuronal signaling and activity, glial activation, viral infection, oxidative stress, and excitotoxicity. Pathologically, reactive gliosis is a hallmark of both HIV-1- and METH-associated neuroinflammation. Significant commonality exists in the neurotoxic mechanisms for both METH and HAND; however, the pathways dysregulated in astroglia during METH exposure are less clear. Thus, this review highlights alterations in astrocyte intracellular signaling pathways, gene expression and function during METH and HIV-1 comorbidity, with special emphasis on HAND-associated neuroinflammation. Importantly, this review carefully evaluates interventions targeting astrocytes in HAND and METH as potential novel therapeutic approaches. This comprehensive overview indicates, without a doubt, that during HIV-1 infection and METH abuse, a complex dialog between all neural cells is orchestrated through astrocyte regulated neuroinflammation.
Collapse
Affiliation(s)
- Kathleen Borgmann
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Anuja Ghorpade
- Department of Cell Biology and Immunology, University of North Texas Health Science Center Fort Worth, TX, USA
| |
Collapse
|
40
|
Ozburn AR, Janowsky AJ, Crabbe JC. Commonalities and Distinctions Among Mechanisms of Addiction to Alcohol and Other Drugs. Alcohol Clin Exp Res 2015; 39:1863-77. [PMID: 26431116 PMCID: PMC4594192 DOI: 10.1111/acer.12810] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/10/2015] [Indexed: 01/25/2023]
Abstract
BACKGROUND Alcohol abuse is comorbid with abuse of many other drugs, some with similar pharmacology and others quite different. This leads to the hypothesis of an underlying, unitary dysfunctional neurobiological basis for substance abuse risk and consequences. METHODS In this review, we discuss commonalities and distinctions of addiction to alcohol and other drugs. We focus on recent advances in preclinical studies using rodent models of drug self-administration. RESULTS While there are specific behavioral and molecular manifestations common to alcohol, psychostimulant, opioid, and nicotine dependence, attempts to propose a unifying theory of the addictions inevitably face details where distinctions are found among classes of drugs. CONCLUSIONS For alcohol, versus other drugs of abuse, we discuss and compare advances in: (i) neurocircuitry important for the different stages of drug dependence; (ii) transcriptomics and genetical genomics; and (iii) enduring effects, noting in particular the contributions of behavioral genetics and animal models.
Collapse
Affiliation(s)
- Angela R. Ozburn
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA
- Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Aaron J. Janowsky
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA
- Department of Psychiatry, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - John C. Crabbe
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA
- Portland Alcohol Research Center, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
41
|
Phillips TJ, Shabani S. An animal model of differential genetic risk for methamphetamine intake. Front Neurosci 2015; 9:327. [PMID: 26441502 PMCID: PMC4585292 DOI: 10.3389/fnins.2015.00327] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/31/2015] [Indexed: 11/13/2022] Open
Abstract
The question of whether genetic factors contribute to risk for methamphetamine (MA) use and dependence has not been intensively investigated. Compared to human populations, genetic animal models offer the advantages of control over genetic family history and drug exposure. Using selective breeding, we created lines of mice that differ in genetic risk for voluntary MA intake and identified the chromosomal addresses of contributory genes. A quantitative trait locus was identified on chromosome 10 that accounts for more than 50% of the genetic variance in MA intake in the selected mouse lines. In addition, behavioral and physiological screening identified differences corresponding with risk for MA intake that have generated hypotheses that are testable in humans. Heightened sensitivity to aversive and certain physiological effects of MA, such as MA-induced reduction in body temperature, are hallmarks of mice bred for low MA intake. Furthermore, unlike MA-avoiding mice, MA-preferring mice are sensitive to rewarding and reinforcing MA effects, and to MA-induced increases in brain extracellular dopamine levels. Gene expression analyses implicate the importance of a network enriched in transcription factor genes, some of which regulate the mu opioid receptor gene, Oprm1, in risk for MA use. Neuroimmune factors appear to play a role in differential response to MA between the mice bred for high and low intake. In addition, chromosome 10 candidate gene studies provide strong support for a trace amine-associated receptor 1 gene, Taar1, polymorphism in risk for MA intake. MA is a trace amine-associated receptor 1 (TAAR1) agonist, and a non-functional Taar1 allele segregates with high MA consumption. Thus, reduced TAAR1 function has the potential to increase risk for MA use. Overall, existing findings support the MA drinking lines as a powerful model for identifying genetic factors involved in determining risk for harmful MA use. Future directions include the development of a binge model of MA intake, examining the effect of withdrawal from chronic MA on MA intake, and studying potential Taar1 gene × gene and gene × environment interactions. These and other studies are intended to improve our genetic model with regard to its translational value to human addiction.
Collapse
Affiliation(s)
- Tamara J. Phillips
- VA Portland Health Care SystemPortland, OR, USA
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science UniversityPortland, OR, USA
| | | |
Collapse
|
42
|
Lam VM, Espinoza S, Gerasimov AS, Gainetdinov RR, Salahpour A. In-vivo pharmacology of Trace-Amine Associated Receptor 1. Eur J Pharmacol 2015; 763:136-42. [DOI: 10.1016/j.ejphar.2015.06.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/26/2015] [Accepted: 06/15/2015] [Indexed: 11/30/2022]
|
43
|
Sriram U, Cenna JM, Haldar B, Fernandes NC, Razmpour R, Fan S, Ramirez SH, Potula R. Methamphetamine induces trace amine-associated receptor 1 (TAAR1) expression in human T lymphocytes: role in immunomodulation. J Leukoc Biol 2015; 99:213-23. [PMID: 26302754 DOI: 10.1189/jlb.4a0814-395rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/05/2015] [Indexed: 01/18/2023] Open
Abstract
The novel transmembrane G protein-coupled receptor, trace amine-associated receptor 1 (TAAR1), represents a potential, direct target for drugs of abuse and monoaminergic compounds, including amphetamines. For the first time, our studies have illustrated that there is an induction of TAAR1 mRNA expression in resting T lymphocytes in response to methamphetamine. Methamphetamine treatment for 6 h significantly increased TAAR1 mRNA expression (P < 0.001) and protein expression (P < 0.01) at 24 h. With the use of TAAR1 gene silencing, we demonstrate that methamphetamine-induced cAMP, a classic response to methamphetamine stimulation, is regulated via TAAR1. We also show by TAAR1 knockdown that the down-regulation of IL-2 in T cells by methamphetamine, which we reported earlier, is indeed regulated by TAAR1. Our results also show the presence of TAAR1 in human lymph nodes from HIV-1-infected patients, with or without a history of methamphetamine abuse. TAAR1 expression on lymphocytes was largely in the paracortical lymphoid area of the lymph nodes with enhanced expression in lymph nodes of HIV-1-infected methamphetamine abusers rather than infected-only subjects. In vitro analysis of HIV-1 infection of human PBMCs revealed increased TAAR1 expression in the presence of methamphetamine. In summary, the ability of methamphetamine to activate trace TAAR1 in vitro and to regulate important T cell functions, such as cAMP activation and IL-2 production; the expression of TAAR1 in T lymphocytes in peripheral lymphoid organs, such as lymph nodes; and our in vitro HIV-1 infection model in PBMCs suggests that TAAR1 may play an important role in methamphetamine -mediated immune-modulatory responses.
Collapse
Affiliation(s)
- Uma Sriram
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jonathan M Cenna
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Bijayesh Haldar
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nicole C Fernandes
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Roshanak Razmpour
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shongshan Fan
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Servio H Ramirez
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Raghava Potula
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
44
|
Zhang J, Yang J, Jang R, Zhang Y. GPCR-I-TASSER: A Hybrid Approach to G Protein-Coupled Receptor Structure Modeling and the Application to the Human Genome. Structure 2015; 23:1538-1549. [PMID: 26190572 DOI: 10.1016/j.str.2015.06.007] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 06/03/2015] [Accepted: 06/10/2015] [Indexed: 12/31/2022]
Abstract
Experimental structure determination remains difficult for G protein-coupled receptors (GPCRs). We propose a new hybrid protocol to construct GPCR structure models that integrates experimental mutagenesis data with ab initio transmembrane (TM) helix assembly simulations. The method was tested on 24 known GPCRs where the ab initio TM-helix assembly procedure constructed the correct fold for 20 cases. When combined with weak homology and sparse mutagenesis restraints, the method generated correct folds for all the tested cases with an average Cα root-mean-square deviation 2.4 Å in the TM regions. The new hybrid protocol was applied to model all 1,026 GPCRs in the human genome, where 923 have a high confidence score and are expected to have correct folds; these contain many pharmaceutically important families with no previously solved structures, including Trace amine, Prostanoids, Releasing hormones, Melanocortins, Vasopressin, and Neuropeptide Y receptors. The results demonstrate new progress on genome-wide structure modeling of TM proteins.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA
| | - Jianyi Yang
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA; School of Mathematical Sciences and LPMC, Nankai University, Tianjin 300071, China
| | - Richard Jang
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA
| | - Yang Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA; Department of Biological Chemistry, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, MI 48109, USA.
| |
Collapse
|
45
|
Jasiulewicz A, Lisowska KA, Pietruczuk K, Frąckowiak J, Fulop T, Witkowski JM. Homeostatic 'bystander' proliferation of human peripheral blood B cells in response to polyclonal T-cell stimulation in vitro. Int Immunol 2015; 27:579-88. [PMID: 25995267 DOI: 10.1093/intimm/dxv032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 05/15/2015] [Indexed: 12/15/2022] Open
Abstract
The mechanisms of maintenance of adequate numbers of B lymphocytes and of protective levels of immunoglobulins in the absence of antigenic (re)stimulation remain not fully understood. Meanwhile, our results presented here show that both peripheral blood naive and memory B cells can be activated strongly and non-specifically (in a mitogen-like fashion) in 5-day in vitro cultures of anti-CD3- or concanavalin A (Con A)-stimulated peripheral blood mononuclear cells of healthy people. This polyclonal, bystander activation of the B cells includes multiple divisions of most of them (assessed here by the flow cytometric technique of dividing cell tracking) and significant antibody [immunoglobulin M (IgM) and IgG] secretion. Observed proliferation of the CD19(+) B cells depends on contact with stimulated T helper (Th) cells (via CD40-CD40L interaction) and on the response of B cells to secreted interleukins IL-5, IL-10 and IL-4, and is correlated with the levels of these Th-derived molecules, while it does not involve the ligation of the BCR/CD19 complex. We suggest that the effect might reflect the situation occurring in vivo as the homeostatic proliferation of otherwise non-stimulated, peripheral B lymphocytes, providing an always ready pool for efficient antibody production to any new (or cognate) antigen challenge.
Collapse
Affiliation(s)
- Aleksandra Jasiulewicz
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Katarzyna A Lisowska
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Krzysztof Pietruczuk
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Joanna Frąckowiak
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Tamas Fulop
- Research Center on Aging, Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| |
Collapse
|
46
|
Loftis JM, Janowsky A. Neuroimmune basis of methamphetamine toxicity. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 118:165-97. [PMID: 25175865 DOI: 10.1016/b978-0-12-801284-0.00007-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although it is not known which antigen-specific immune responses (or if antigen-specific immune responses) are relevant or required for methamphetamine's neurotoxic effects, it is apparent that methamphetamine exposure is associated with significant effects on adaptive and innate immunity. Alterations in lymphocyte activity and number, changes in cytokine signaling, impairments in phagocytic functions, and glial activation and gliosis have all been reported. These drug-induced changes in immune response, particularly within the CNS, are now thought to play a critical role in the addiction process for methamphetamine dependence as well as for other substance use disorders. In Section 2, methamphetamine's effects on glial cell (e.g., microglia and astrocytes) activity and inflammatory signaling cascades are summarized, including how alterations in immune cell function can induce the neurotoxic and addictive effects of methamphetamine. Section 2 also describes neurotransmitter involvement in the modulation of methamphetamine's inflammatory effects. Section 3 discusses the very recent use of pharmacological and genetic animal models which have helped elucidate the behavioral effects of methamphetamine's neurotoxic effects and the role of the immune system. Section 4 is focused on the effects of methamphetamine on blood-brain barrier integrity and associated immune consequences. Clinical considerations such as the combined effects of methamphetamine and HIV and/or HCV on brain structure and function are included in Section 4. Finally, in Section 5, immune-based treatment strategies are reviewed, with a focus on vaccine development, neuroimmune therapies, and other anti-inflammatory approaches.
Collapse
Affiliation(s)
- Jennifer M Loftis
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA; Department of Psychiatry, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA.
| | - Aaron Janowsky
- Research & Development Service, Portland VA Medical Center, Portland, Oregon, USA; Department of Psychiatry, Oregon Health & Science University, School of Medicine, Portland, Oregon, USA; Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, Oregon, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
47
|
Dinter J, Mühlhaus J, Wienchol CL, Yi CX, Nürnberg D, Morin S, Grüters A, Köhrle J, Schöneberg T, Tschöp M, Krude H, Kleinau G, Biebermann H. Inverse agonistic action of 3-iodothyronamine at the human trace amine-associated receptor 5. PLoS One 2015; 10:e0117774. [PMID: 25706283 PMCID: PMC4382497 DOI: 10.1371/journal.pone.0117774] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/30/2014] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Application of 3-iodothyronamine (3-T1AM) results in decreased body temperature and body weight in rodents. The trace amine-associated receptor (TAAR) 1, a family A G protein-coupled receptor, is a target of 3-T1AM. However, 3-T1AM effects still persist in mTaar1 knockout mice, which suggest so far unknown further receptor targets that are of physiological relevance. TAAR5 is a highly conserved TAAR subtype among mammals and we here tested TAAR5 as a potential 3-T1AM target. First, we investigated mouse Taar5 (mTaar5) expression in several brain regions of the mouse in comparison to mTaar1. Secondly, to unravel the full spectrum of signaling capacities, we examined the distinct Gs-, Gi/o-, G12/13-, Gq/11- and MAP kinase-mediated signaling pathways of mouse and human TAAR5 under ligand-independent conditions and after application of 3-T1AM. We found overlapping localization of mTaar1 and mTaar5 in the amygdala and ventromedial hypothalamus of the mouse brain. Second, the murine and human TAAR5 (hTAAR5) display significant basal activity in the Gq/11 pathway but show differences in the basal activity in Gs and MAP kinase signaling. In contrast to mTaar5, 3-T1AM application at hTAAR5 resulted in significant reduction in basal IP3 formation and MAP kinase signaling. In conclusion, our data suggest that the human TAAR5 is a target for 3-T1AM, exhibiting inhibitory effects on IP3 formation and MAP kinase signaling pathways, but does not mediate Gs signaling effects as observed for TAAR1. This study also indicates differences between TAAR5 orthologs with respect to their signaling profile. In consequence, 3-T1AM-mediated effects may differ between rodents and humans.
Collapse
Affiliation(s)
- Juliane Dinter
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jessica Mühlhaus
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carolin Leonie Wienchol
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Chun-Xia Yi
- Institute for Diabetes and Obesity, Helmholtz-Zentrum München, German Research Center for Environmental Health, München, Germany
| | - Daniela Nürnberg
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Silke Morin
- Institute for Diabetes and Obesity, Helmholtz-Zentrum München, German Research Center for Environmental Health, München, Germany
| | - Annette Grüters
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Torsten Schöneberg
- Institut für Biochemie, Molekulare Biochemie, Medizinische Fakultät, University of Leipzig, Leipzig, Germany
| | - Matthias Tschöp
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heiko Krude
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gunnar Kleinau
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heike Biebermann
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
48
|
Mühlhaus J, Dinter J, Nürnberg D, Rehders M, Depke M, Golchert J, Homuth G, Yi CX, Morin S, Köhrle J, Brix K, Tschöp M, Kleinau G, Biebermann H. Analysis of human TAAR8 and murine Taar8b mediated signaling pathways and expression profile. Int J Mol Sci 2014; 15:20638-55. [PMID: 25391046 PMCID: PMC4264187 DOI: 10.3390/ijms151120638] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 10/25/2014] [Accepted: 11/04/2014] [Indexed: 12/04/2022] Open
Abstract
The thyroid hormone derivative 3-iodothyronamine (3-T1AM) exerts metabolic effects in vivo that contradict known effects of thyroid hormones. 3-T1AM acts as a trace amine-associated receptor 1 (TAAR1) agonist and activates Gs signaling in vitro. Interestingly, 3-T1AM-meditated in vivo effects persist in Taar1 knockout-mice indicating that further targets of 3-T1AM might exist. Here, we investigated another member of the TAAR family, the only scarcely studied mouse and human trace-amine-associated receptor 8 (Taar8b, TAAR8). By RT-qPCR and locked-nucleic-acid (LNA) in situ hybridization, Taar8b expression in different mouse tissues was analyzed. Functionally, we characterized TAAR8 and Taar8b with regard to cell surface expression and signaling via different G-protein-mediated pathways. Cell surface expression was verified by ELISA, and cAMP accumulation was quantified by AlphaScreen for detection of Gs and/or Gi/o signaling. Activation of G-proteins Gq/11 and G12/13 was analyzed by reporter gene assays. Expression analyses revealed at most marginal Taar8b expression and no gender differences for almost all analyzed tissues. In heart, LNA-in situ hybridization demonstrated the absence of Taar8b expression. We could not identify 3-T1AM as a ligand for TAAR8 and Taar8b, but both receptors were characterized by a basal Gi/o signaling activity, a so far unknown signaling pathway for TAARs.
Collapse
Affiliation(s)
- Jessica Mühlhaus
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Juliane Dinter
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Daniela Nürnberg
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Maren Rehders
- School of Engineering and Science, Research Center MOLIFE-Molecular Life Science, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany.
| | - Maren Depke
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt-University Greifswald, Fr iedrich-Ludwig-Jahn-Str. 15a, 17487 Greifswald, Germany.
| | - Janine Golchert
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt-University Greifswald, Fr iedrich-Ludwig-Jahn-Str. 15a, 17487 Greifswald, Germany.
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt-University Greifswald, Fr iedrich-Ludwig-Jahn-Str. 15a, 17487 Greifswald, Germany.
| | - Chun-Xia Yi
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute for Diabetes and Obesity, Business Campus Garching, Parkring 13, 85748 Garching, Germany.
| | - Silke Morin
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute for Diabetes and Obesity, Business Campus Garching, Parkring 13, 85748 Garching, Germany.
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Klaudia Brix
- School of Engineering and Science, Research Center MOLIFE-Molecular Life Science, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany.
| | - Matthias Tschöp
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute for Diabetes and Obesity, Business Campus Garching, Parkring 13, 85748 Garching, Germany.
| | - Gunnar Kleinau
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Heike Biebermann
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
49
|
Gozal EA, O'Neill BE, Sawchuk MA, Zhu H, Halder M, Chou CC, Hochman S. Anatomical and functional evidence for trace amines as unique modulators of locomotor function in the mammalian spinal cord. Front Neural Circuits 2014; 8:134. [PMID: 25426030 PMCID: PMC4224135 DOI: 10.3389/fncir.2014.00134] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 10/22/2014] [Indexed: 12/18/2022] Open
Abstract
The trace amines (TAs), tryptamine, tyramine, and β-phenylethylamine, are synthesized from precursor amino acids via aromatic-L-amino acid decarboxylase (AADC). We explored their role in the neuromodulation of neonatal rat spinal cord motor circuits. We first showed that the spinal cord contains the substrates for TA biosynthesis (AADC) and for receptor-mediated actions via trace amine-associated receptors (TAARs) 1 and 4. We next examined the actions of the TAs on motor activity using the in vitro isolated neonatal rat spinal cord. Tyramine and tryptamine most consistently increased motor activity with prominent direct actions on motoneurons. In the presence of N-methyl-D-aspartate, all applied TAs supported expression of a locomotor-like activity (LLA) that was indistinguishable from that ordinarily observed with serotonin, suggesting that the TAs act on common central pattern generating neurons. The TAs also generated distinctive complex rhythms characterized by episodic bouts of LLA. TA actions on locomotor circuits did not require interaction with descending monoaminergic projections since evoked LLA was maintained following block of all Na+-dependent monoamine transporters or the vesicular monoamine transporter. Instead, TA (tryptamine and tyramine) actions depended on intracellular uptake via pentamidine-sensitive Na+-independent membrane transporters. Requirement for intracellular transport is consistent with the TAs having much slower LLA onset than serotonin and for activation of intracellular TAARs. To test for endogenous actions following biosynthesis, we increased intracellular amino acid levels with cycloheximide. LLA emerged and included distinctive TA-like episodic bouts. In summary, we provided anatomical and functional evidence of the TAs as an intrinsic spinal monoaminergic modulatory system capable of promoting recruitment of locomotor circuits independent of the descending monoamines. These actions support their known sympathomimetic function.
Collapse
Affiliation(s)
| | | | | | - Hong Zhu
- Physiology Department, Emory University Atlanta, GA, USA
| | - Mallika Halder
- Physiology Department, Emory University Atlanta, GA, USA
| | | | - Shawn Hochman
- Physiology Department, Emory University Atlanta, GA, USA
| |
Collapse
|
50
|
TAAR1 transforms thinking about a plant alkaloid that transformed the practice of medicine. Int J Neuropsychopharmacol 2014; 17:1545-7. [PMID: 24901712 DOI: 10.1017/s1461145714000911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|