1
|
Apryatin SA. The Neurometabolic Function of the Dopamine-Aminotransferase System. Metabolites 2025; 15:21. [PMID: 39852364 DOI: 10.3390/metabo15010021] [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: 12/16/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES The neurometabolic function is controlled by a complex multi-level physiological system that includes neurochemical, hormonal, immunological, sensory, and metabolic components. Functional disorders of monoamine systems are often detected in clinical practice together with metabolic dysfunctions. An important part of the mentioned pathological conditions are associated with disturbances in protein metabolism, some of the most important biomarkers which are aminotransferases and transcription factors that regulate and direct the most important metabolic reactions. Another important part of energy metabolism is the dopamine-mediated regulation of protein metabolism. METHODS The review describes research results into the dopamine-mediated mechanism of metabolic regulation in humans and animals. Particular attention is paid to the neurometabolic mechanisms of protein metabolism. RESULTS The dopamine-aminotransferase system of the energy metabolism regulation is a separate, independent, regulatory and diagnostically significant biochemical pathway controlled by the hormonal system, the key hormone is cortisol, the key neurotransmitter is dopamine, the key transcription factor is CREB, and the key regulatory enzymes are alanine aminotransferase, aspartate aminotransferase, and tyrosine aminotransferase. CONCLUSIONS This review presents an original study describing the discovery of a new regulatory mechanism for neurometabolic physiological function in humans and animals. A key part of this mechanism is the dopamine-aminotransferase system.
Collapse
Affiliation(s)
- Sergey A Apryatin
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| |
Collapse
|
2
|
Jiang K, Zheng Y, Zeng L, Wang L, Li F, Pu J, Lu Y, Zhao S, Xu F. The versatile binding landscape of the TAAR1 pocket for LSD and other antipsychotic drug molecules. Cell Rep 2024; 43:114505. [PMID: 39002128 DOI: 10.1016/j.celrep.2024.114505] [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: 03/21/2024] [Revised: 05/01/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024] Open
Abstract
Increasing global concerns about psychoactive substance addiction and psychotic disorders highlight the need for comprehensive research into the structure-function relationship governing ligand recognition between these substances and their receptors in the brain. Recent studies indicate the significant involvement of trace amine-associated receptor 1 (TAAR1) in the signaling regulation of the hallucinogen lysergic acid diethylamide (LSD) and other antipsychotic drugs. This study presents structures of the TAAR1-Gs protein complex recognizing LSD, which exhibits a polypharmacological profile, and the partial agonist RO5263397, which is a drug candidate for schizophrenia and addiction. Moreover, we elucidate the cross-species recognition and partial activation mechanism for TAAR1, which holds promising implications from a drug discovery perspective. Through mutagenesis, functional studies, and molecular dynamics (MD) simulations, we provide a comprehensive understanding of a versatile TAAR1 pocket in recognizing various ligands as well as in the ligand-free state, underpinning the structural basis of its high adaptability. These findings offer valuable insights for the design of antipsychotic drugs.
Collapse
Affiliation(s)
- Kexin Jiang
- iHuman Institute, ShanghaiTech University, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - You Zheng
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Liting Zeng
- iHuman Institute, ShanghaiTech University, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ling Wang
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Fei Li
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Jun Pu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Fei Xu
- iHuman Institute, ShanghaiTech University, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
3
|
Shemiakova TS, Efimova EV, Gainetdinov RR. TAARs as Novel Therapeutic Targets for the Treatment of Depression: A Narrative Review of the Interconnection with Monoamines and Adult Neurogenesis. Biomedicines 2024; 12:1263. [PMID: 38927470 PMCID: PMC11200894 DOI: 10.3390/biomedicines12061263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Depression is a common mental illness of great concern. Current therapy for depression is only suitable for 80% of patients and is often associated with unwanted side effects. In this regard, the search for and development of new antidepressant agents remains an urgent task. In this review, we discuss the current available evidence indicating that G protein-coupled trace amine-associated receptors (TAARs) might represent new targets for depression treatment. The most frequently studied receptor TAAR1 has already been investigated in the treatment of schizophrenia, demonstrating antidepressant and anxiolytic properties. In fact, the TAAR1 agonist Ulotaront is currently undergoing phase 2/3 clinical trials testing its safety and efficacy in the treatment of major depressive disorder and generalized anxiety disorder. Other members of the TAAR family (TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) are not only involved in the innate olfaction of volatile amines, but are also expressed in the limbic brain areas. Furthermore, animal studies have shown that TAAR2 and TAAR5 regulate emotional behaviors and thus may hold promise as potential antidepressant targets. Of particular interest is their connection with the dopamine and serotonin systems of the brain and their involvement in the regulation of adult neurogenesis, known to be affected by the antidepressant drugs currently in use. Further non-clinical and clinical studies are necessary to validate TAAR1 (and potentially other TAARs) as novel therapeutic targets for the treatment of depression.
Collapse
Affiliation(s)
- Taisiia S. Shemiakova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia; (T.S.S.); (E.V.E.)
| | - Evgeniya V. Efimova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia; (T.S.S.); (E.V.E.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia; (T.S.S.); (E.V.E.)
- Saint-Petersburg University Hospital, Saint-Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
4
|
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
|
5
|
Dedic N, Wang L, Hajos-Korcsok E, Hecksher-Sørensen J, Roostalu U, Vickers SP, Wu S, Anacker C, Synan C, Jones PG, Milanovic S, Hopkins SC, Bristow LJ, Koblan KS. TAAR1 agonists improve glycemic control, reduce body weight and modulate neurocircuits governing energy balance and feeding. Mol Metab 2024; 80:101883. [PMID: 38237896 PMCID: PMC10839149 DOI: 10.1016/j.molmet.2024.101883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVE Metabolic Syndrome, which can be induced or exacerbated by current antipsychotic drugs (APDs), is highly prevalent in schizophrenia patients. Recent preclinical and clinical evidence suggest that agonists at trace amine-associated receptor 1 (TAAR1) have potential as a new treatment option for schizophrenia. Intriguingly, preclinical tudies have also identified TAAR1 as a novel regulator of metabolic control. Here we evaluated the effects of three TAAR1 agonists, including the clinical development candidate ulotaront, on body weight, metabolic parameters and modulation of neurocircuits implicated in homeostatic and hedonic feeding. METHODS Effects of TAAR1 agonists (ulotaront, RO5166017 and/or RO5263397) on body weight, food intake and/or metabolic parameters were investigated in rats fed a high-fat diet (HFD) and in a mouse model of diet-induced obesity (DIO). Body weight effects were also determined in a rat and mouse model of olanzapine-, and corticosterone-induced body weight gain, respectively. Glucose tolerance was assessed in lean and diabetic db/db mice and fasting plasma glucose and insulin examined in DIO mice. Effects on gastric emptying were evaluated in lean mice and rats. Drug-induced neurocircuit modulation was evaluated in mice using whole-brain imaging of c-fos protein expression. RESULTS TAAR1 agonists improved oral glucose tolerance by inhibiting gastric emptying. Sub-chronic administration of ulotaront in rats fed a HFD produced a dose-dependent reduction in body weight, food intake and liver triglycerides compared to vehicle controls. In addition, a more rapid reversal of olanzapine-induced weight gain and food intake was observed in HFD rats switched to ulotaront or RO5263397 treatment compared to those switched to vehicle. Chronic ulotaront administration also reduced body weight and improved glycemic control in DIO mice, and normalized corticosterone-induced body weight gain in mice. TAAR1 activation increased neuronal activity in discrete homeostatic and hedonic feeding centers located in the dorsal vagal complex and hypothalamus with concurrent activation of several limbic structures. CONCLUSION The current data demonstrate that TAAR1 agonists, as a class, not only lack APD-induced metabolic liabilities but can reduce body weight and improve glycemic control in rodent models. The underlying mechanisms likely include TAAR1-mediated peripheral effects on glucose homeostasis and gastric emptying as well as central regulation of energy balance and food intake.
Collapse
Affiliation(s)
- Nina Dedic
- Sumitomo Pharma America, Inc., Marlborough, MA, USA.
| | - Lien Wang
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | | | | | | | | | - Serena Wu
- Department of Psychiatry, New York State Psychiatric Institute (NYSPI), Columbia University, NY, New York City, USA
| | - Christoph Anacker
- Department of Psychiatry, New York State Psychiatric Institute (NYSPI), Columbia University, NY, New York City, USA
| | | | | | | | | | | | | |
Collapse
|
6
|
Landini L, Dadson P, Gallo F, Honka MJ, Cena H. Microbiota in anorexia nervosa: potential for treatment. Nutr Res Rev 2023; 36:372-391. [PMID: 35875979 DOI: 10.1017/s0954422422000130] [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] [Indexed: 11/07/2022]
Abstract
Anorexia nervosa (AN) is characterised by the restriction of energy intake in relation to energy needs and a significantly lowered body weight than normally expected, coupled with an intense fear of gaining weight. Treatment of AN is currently based on psychological and refeeding approaches, but their efficacy remains limited since 40% of patients after 10 years of medical care still present symptoms of AN. The intestine hosts a large community of microorganisms, called the "microbiota", which live in symbiosis with the human host. The gut microbiota of a healthy human is dominated by bacteria from two phyla: Firmicutes and, majorly, Bacteroidetes. However, the proportion in their representation differs on an individual basis and depends on many external factors including medical treatment, geographical location and hereditary, immunological and lifestyle factors. Drastic changes in dietary intake may profoundly impact the composition of the gut microbiota, and the resulting dysbiosis may play a part in the onset and/or maintenance of comorbidities associated with AN, such as gastrointestinal disorders, anxiety and depression, as well as appetite dysregulation. Furthermore, studies have reported the presence of atypical intestinal microbial composition in patients with AN compared with healthy normal-weight controls. This review addresses the current knowledge about the role of the gut microbiota in the pathogenesis and treatment of AN. The review also focuses on the bidirectional interaction between the gastrointestinal tract and the central nervous system (microbiota-gut-brain axis), considering the potential use of the gut microbiota manipulation in the prevention and treatment of AN.
Collapse
Affiliation(s)
- Linda Landini
- S.S.D. Dietetics and Clinical Nutrition ASL 4 Chiavarese Liguria-Sestri Levante Hospital, Sestri Levante, Italy
| | - Prince Dadson
- Turku PET Centre, University of Turku, Turku, Finland
| | - Fabrizio Gallo
- S.S.D. Dietetics and Clinical Nutrition ASL 4 Chiavarese Liguria-Sestri Levante Hospital, Sestri Levante, Italy
| | | | - Hellas Cena
- Dietetics and Clinical Nutrition Laboratory, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
- Clinical Nutrition and Dietetics Service, Unit of Internal Medicine and Endocrinology, ICS Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
7
|
Vaganova AN, Shemyakova TS, Lenskaia KV, Rodionov RN, Steenblock C, Gainetdinov RR. Trace Amine-Associated Receptors and Monoamine-Mediated Regulation of Insulin Secretion in Pancreatic Islets. Biomolecules 2023; 13:1618. [PMID: 38002300 PMCID: PMC10669413 DOI: 10.3390/biom13111618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Currently, metabolic syndrome treatment includes predominantly pharmacological symptom relief and complex lifestyle changes. Trace amines and their receptor systems modulate signaling pathways of dopamine, norepinephrine, and serotonin, which are involved in the pathogenesis of this disorder. Trace amine-associated receptor 1 (TAAR1) is expressed in endocrine organs, and it was revealed that TAAR1 may regulate insulin secretion in pancreatic islet β-cells. For instance, accumulating data demonstrate the positive effect of TAAR1 agonists on the dynamics of metabolic syndrome progression and MetS-associated disease development. The role of other TAARs (TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) in the islet's function is much less studied. In this review, we summarize the evidence of TAARs' contribution to the metabolic syndrome pathogenesis and regulation of insulin secretion in pancreatic islets. Additionally, by the analysis of public transcriptomic data, we demonstrate that TAAR1 and other TAAR receptors are expressed in the pancreatic islets. We also explore associations between the expression of TAARs mRNA and other genes in studied samples and demonstrate the deregulation of TAARs' functional associations in patients with metabolic diseases compared to healthy donors.
Collapse
Affiliation(s)
- Anastasia N. Vaganova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.N.V.); (T.S.S.)
- St. Petersburg State University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Taisiia S. Shemyakova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.N.V.); (T.S.S.)
| | - Karina V. Lenskaia
- Department of Medicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia;
| | - Roman N. Rodionov
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (R.N.R.); (C.S.)
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (R.N.R.); (C.S.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.N.V.); (T.S.S.)
- St. Petersburg State University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
8
|
Katolikova NV, Vaganova AN, Shafranskaya DD, Efimova EV, Malashicheva AB, Gainetdinov RR. Expression Pattern of Trace Amine-Associated Receptors during Differentiation of Human Pluripotent Stem Cells to Dopaminergic Neurons. Int J Mol Sci 2023; 24:15313. [PMID: 37894992 PMCID: PMC10607858 DOI: 10.3390/ijms242015313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Trace amine-associated receptors (TAARs), which were discovered only in 2001, are known to be involved in the regulation of a spectrum of neuronal processes and may play a role in the pathogenesis of a number of neuropsychiatric diseases, such as schizophrenia and others. We have previously shown that TAARs also have interconnections with the regulation of neurogenesis and, in particular, with the neurogenesis of dopamine neurons, but the exact mechanisms of this are still unknown. In our work we analyzed the expression of TAARs (TAAR1, TAAR2, TAAR5, TAAR6, TAAR8 and TAAR9) in cells from the human substantia nigra and ventral tegmental areas and in human pluripotent stem cells at consecutive stages of their differentiation to dopaminergic neurons, using RNA sequencing data from open databases, and TaqMan PCR data from the differentiation of human induced pluripotent stem cells in vitro. Detectable levels of TAARs expression were found in cells at the pluripotent stages, and the dynamic of their expression had a trend of increasing with the differentiation and maturation of dopamine neurons. The expression of several TAAR types (particularly TAAR5) was also found in human dopaminergic neuron-enriched zones in the midbrain. This is the first evidence of TAARs expression during neuronal differentiation, which can help to approach an understanding of the role of TAARs in neurogenesis.
Collapse
Affiliation(s)
- Nataliia V. Katolikova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia (R.R.G.)
| | - Anastasia N. Vaganova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia (R.R.G.)
- Saint-Petersburg University Hospital, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia
| | - Daria D. Shafranskaya
- Center for Algorithmic Biotechnology, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia
| | - Evgeniya V. Efimova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia (R.R.G.)
| | - Anna B. Malashicheva
- Department of Embryology, Faculty of Biology, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia;
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia (R.R.G.)
- Saint-Petersburg University Hospital, Saint-Petersburg State University, 199034 Saint-Petersburg, Russia
| |
Collapse
|
9
|
Achtyes ED, Hopkins SC, Dedic N, Dworak H, Zeni C, Koblan K. Ulotaront: review of preliminary evidence for the efficacy and safety of a TAAR1 agonist in schizophrenia. Eur Arch Psychiatry Clin Neurosci 2023; 273:1543-1556. [PMID: 37165101 PMCID: PMC10465394 DOI: 10.1007/s00406-023-01580-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/26/2023] [Indexed: 05/12/2023]
Abstract
Ulotaront is a trace amine-associated receptor 1 (TAAR1) agonist in Phase 3 clinical development for the treatment of schizophrenia. Ulotaront was discovered through a unique, target-agnostic approach optimized to identify drug candidates lacking D2 and 5-HT2A receptor antagonism, while demonstrating an antipsychotic-like phenotypic profile in vivo. The mechanism of action (MOA) of ulotaront is thought to be mediated by agonism at TAAR1 and serotonin 5-HT1A receptors. Ulotaront has completed two Phase 2 trials (4-week acute study and 26-week open-label extension) which led to Breakthrough Therapy Designation from the US Food and Drug Administration for the treatment of schizophrenia. In the double-blind, placebo-controlled, acute study, ulotaront was associated with significant (p < 0.001) improvement in Positive and Negative Syndrome Scale (PANSS) total score (effect size [ES]: 0.45), with improvements vs. placebo also observed across secondary endpoints. Post-hoc analyses of the acute trial revealed additional evidence to support the effect of ulotaront on negative symptoms. In the 4-week study, ulotaront was well-tolerated, with an incidence of adverse events (AEs) numerically lower compared to placebo (45.8% vs. 50.4%; with a number needed to harm [NNH] for individual ulotaront AEs all > 40). The open-label extension demonstrated further improvement across schizophrenia symptoms and confirmed the tolerability of ulotaront, with a 6-month completion rate of 67%. Based on current data, ulotaront shows potential to be a first-in-class TAAR1 agonist for the treatment of schizophrenia with a safety and efficacy profile distinct from current antipsychotics.
Collapse
Affiliation(s)
- Eric D Achtyes
- WMU Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | | | - Nina Dedic
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| | | | - Courtney Zeni
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA.
| | | |
Collapse
|
10
|
Shabani S, Houlton S, Ghimire B, Tonello D, Reed C, Baba H, Aldrich S, Phillips TJ. Robust aversive effects of trace amine-associated receptor 1 activation in mice. Neuropsychopharmacology 2023; 48:1446-1454. [PMID: 37055488 PMCID: PMC10425385 DOI: 10.1038/s41386-023-01578-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/03/2023] [Accepted: 03/26/2023] [Indexed: 04/15/2023]
Abstract
Drugs that stimulate the trace amine-associated receptor 1 (TAAR1) are under clinical investigation as treatments for several neuropsychiatric disorders. Previous studies in a genetic mouse model of voluntary methamphetamine intake identified TAAR1, expressed by the Taar1 gene, as a critical mediator of aversive methamphetamine effects. Methamphetamine is a TAAR1 agonist, but also has actions at monoamine transporters. Whether exclusive activation of TAAR1 has aversive effects was not known at the time we conducted our studies. Mice were tested for aversive effects of the selective TAAR1 agonist, RO5256390, using taste and place conditioning procedures. Hypothermic and locomotor effects were also examined, based on prior evidence of TAAR1 mediation. Male and female mice of several genetic models were used, including lines selectively bred for high and low methamphetamine drinking, a knock-in line in which a mutant form of Taar1 that codes for a non-functional TAAR1 was replaced by the reference Taar1 allele that codes for functional TAAR1, and their matched control line. RO5256390 had robust aversive, hypothermic and locomotor suppressing effects that were found only in mice with functional TAAR1. Knock-in of the reference Taar1 allele rescued these phenotypes in a genetic model that normally lacks TAAR1 function. Our study provides important data on TAAR1 function in aversive, locomotor, and thermoregulatory effects that are important to consider when developing TAAR1 agonists as therapeutic drugs. Because other drugs can have similar consequences, potential additive effects should be carefully considered as these treatment agents are being developed.
Collapse
Affiliation(s)
- Shkelzen Shabani
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
- Department of Biology, Minot State University, Minot, ND, USA
- Biomedical Sciences at Grand Valley State University, Allendale, MI, USA
| | - Sydney Houlton
- Department of Biology, Minot State University, Minot, ND, USA
| | - Bikalpa Ghimire
- Department of Biology, Minot State University, Minot, ND, USA
| | - Derek Tonello
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
| | - Cheryl Reed
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Harue Baba
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Sara Aldrich
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Tamara J Phillips
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA.
- VA Portland Health Care System, Portland, OR, USA.
| |
Collapse
|
11
|
Frycz BA, Nowicka K, Konopka A, Hoener MC, Bulska E, Kaczmarek L, Stefaniuk M. Activation of trace amine-associated receptor 1 (TAAR1) transiently reduces alcohol drinking in socially housed mice. Addict Biol 2023; 28:e13285. [PMID: 37369127 DOI: 10.1111/adb.13285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 06/29/2023]
Abstract
Alcohol dependence is characterized by the abnormal release of dopamine in the brain reward-related areas. Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that negatively regulates dopamine neurotransmission and thus is a promising target in the treatment of drug addiction. However, the role of TAAR1 in the regulation of alcohol abuse remains understudied. Here, we assessed the effect of TAAR1 activation on alcohol drinking behaviours of C57Bl/6J female mice housed in IntelliCages. The animals were administered with either vehicle or TAAR1 full selective agonist, RO5256390, and tested for alcohol consumption, alcohol preference and motivation for alcohol seeking. We found that mice with the highest preference for alcohol (high drinkers) in the RO5256390 group consumed less alcohol and had lower alcohol preference in comparison with high drinkers in the vehicle group, during 20 h of free alcohol access (FAA). We also found decreased alcohol consumption and alcohol preference comparing all animals in the RO5256390 to all animals in the vehicle group, during 20 h of FAA performed after the abstinence. These effects of RO5256390 lasted for the first 24 h after administration that roughly corresponded to the compound level in the brain, measured by mass spectrometry. Finally, we found that administration of RO5256390 may attenuate motivation for alcohol seeking. Taken together, our findings reveal that activation of TAAR1 may transiently reduce alcohol drinking; thus, TAAR1 is a promising target for the treatment of alcohol abuse and relapse.
Collapse
Affiliation(s)
- Bartosz Adam Frycz
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Klaudia Nowicka
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Konopka
- Faculty of Chemistry, Biological, and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Marius Christian Hoener
- Neuroscience and Rare Diseases Discovery and Translational Area, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Ewa Bulska
- Faculty of Chemistry, Biological, and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Marzena Stefaniuk
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
12
|
Berger G, Corris JD, Fields SE, Hao L, Scarpa LL, Bello NT. Systematic Review of Binge Eating Rodent Models for Developing Novel or Repurposing Existing Pharmacotherapies. Biomolecules 2023; 13:742. [PMID: 37238615 PMCID: PMC10216509 DOI: 10.3390/biom13050742] [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: 02/10/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Recent advances in developing and screening candidate pharmacotherapies for psychiatric disorders have depended on rodent models. Eating disorders are a set of psychiatric disorders that have traditionally relied on behavioral therapies for effective long-term treatment. However, the clinical use of Lisdexamfatamine for binge eating disorder (BED) has furthered the notion of using pharmacotherapies for treating binge eating pathologies. While there are several binge eating rodent models, there is not a consensus on how to define pharmacological effectiveness within these models. Our purpose is to provide an overview of the potential pharmacotherapies or compounds tested in established rodent models of binge eating behavior. These findings will help provide guidance for determining pharmacological effectiveness for potential novel or repurposed pharmacotherapies.
Collapse
Affiliation(s)
- Gregory Berger
- Endocrinology and Animal Biosciences Graduate Program, Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Joshua D. Corris
- Endocrinology and Animal Biosciences Graduate Program, Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Spencer E. Fields
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Nutritional Sciences Graduate Program, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Lihong Hao
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Lori L. Scarpa
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Nicholas T. Bello
- Endocrinology and Animal Biosciences Graduate Program, Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Rutgers Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA
| |
Collapse
|
13
|
Sarkar S, Saika-Voivod I, Berry MD. Modelling of p-tyramine transport across human intestinal epithelial cells predicts the presence of additional transporters. Front Physiol 2022; 13:1009320. [PMID: 36505075 PMCID: PMC9733674 DOI: 10.3389/fphys.2022.1009320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/22/2022] [Indexed: 11/12/2022] Open
Abstract
p-Tyramine (TYR) is an endogenous trace amine, which can also be synthesized by intestinal microbiota, and is present in commonly consumed diets. TYR is an agonist for the intracellular trace amine-associated receptor 1, which has been implicated in psychiatric, metabolic, and immune-related disorders. We have previously demonstrated TYR readily diffuses across lipid bilayers, while transport across Caco-2 cell membranes involves Organic Cation Transporter 2 (OCT2) and a Na+-dependent active transporter. Here we developed mathematical models to determine whether known kinetics for these processes are sufficient to explain observed transcellular TYR passage. Ordinary differential equations were developed for known TYR transport processes to predict concentration-time relationships. Michaelis-Menten kinetics were assumed for all transporter-mediated processes and a one phase exponential function used for simple diffusion. Modelled concentration-time plots were compared to published experimental results. Additional transporter functions were sequentially added to models to improve consistency, and a least squares error minimization approach utilized to determine added transporter kinetics. Finally, possible TYR compartmentalization was also modelled. Following apical loading, transport across the apical, but not the basolateral, membrane was modelled without additional transporters, suggesting a basolateral transporter was missing. Consistent with this, models of basolateral compartment loading did not match experimental observations, indicating missing basolateral transporters were bidirectional. Addition of a transporter with the kinetic characteristics of OCT2 did not improve models. Varying the kinetic parameters of the added transporter improved models of basolateral, but worsened apical, loading models, suggesting the need for either a directional preference in transporters, or intracellular TYR compartmentalization. Experimental parameters were recapitulated by introducing asymmetry into the apical OCT2 (Kt_OCT2_apicaltocell = 110.4 nM, Kt_OCT2_celltoapical = 1,227.9 nM), and a symmetric basolateral facilitated diffusion transporter (Vmax = 6.0 nM/s, Kt = 628.3 nM). The apparent directionality of OCT2 may reflect altered TYR ionization due to known pH differences between compartments. Models for asymmetry and compartmentalization were compared by root mean square deviation from experimental data, and it was found that TYR compartmentalization could only partially replace the need for asymmetry of OCT2. In conclusion, modelling indicates that known TYR transport processes are insufficient to explain experimental concentration-time profiles and that asymmetry of the apical membrane OCT2 combined with additional, low affinity, basolateral membrane facilitated diffusion transporters are required.
Collapse
Affiliation(s)
- Shreyasi Sarkar
- Department of Biochemistry, Memorial University of Newfoundland, St. John’s, NL, Canada,*Correspondence: Shreyasi Sarkar,
| | - Ivan Saika-Voivod
- Department of Physics and Physical Oceanography, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Mark D. Berry
- Department of Biochemistry, Memorial University of Newfoundland, St. John’s, NL, Canada
| |
Collapse
|
14
|
Liu J, Wu R, Johnson B, Zhang Y, Zhu Q, Li JX. Selective TAAR1 agonists induce conditioned taste aversion. Psychopharmacology (Berl) 2022; 239:3345-3353. [PMID: 36056214 DOI: 10.1007/s00213-022-06222-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/24/2022] [Indexed: 01/07/2023]
Abstract
RATIONALE Trace amine-associated receptor 1 (TAAR1) is the best-studied receptor of trace amines, a group of biogenic amines expressed at a relatively low level in the mammalian brain. Growing evidence suggests that TAAR1 plays a critical role in various neuropsychiatric disorders. Given that selective TAAR1 agonists were shown to produce pro-cognition and antipsychotic-like effects as well as to suppress drug use and relapse, they have been proposed to be novel treatments for mental disorders such as schizophrenia and addiction. However, the aversive effects of selective TAAR1 agonists remain largely unknown. OBJECTIVES Here, we evaluated whether the selective TAAR1 full agonist RO5166017 and partial agonist RO5263397 could induce conditioned taste aversion (CTA). RESULTS We found that RO5166017 and RO5263397 produced significant aversions to both saccharin and NaCl taste novelty. Furthermore, RO5166017 produced CTA to saccharin in TAAR1 heterozygous knockout (taar1±) and wild-type rats but not in TAAR1 homozygous knockout rats (taar1-/-), suggesting that TAAR1 was sufficient for the taste aversive stimulus property of RO5166017. CONCLUSIONS Taken together, our data indicate that selective TAAR1 agonists could produce strong CTA. Our study urges careful evaluations of the aversive effects of TAAR1 agonists before translating them to clinical use for the treatment of mental disorders.
Collapse
Affiliation(s)
- Jianfeng Liu
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China.,Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Ruyan Wu
- Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Bernard Johnson
- Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, Program in Neuroscience, University at Buffalo, The State University of New York, 955 Main Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
15
|
O'Connor RM, Kenny PJ. Utility of 'substance use disorder' as a heuristic for understanding overeating and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2022; 118:110580. [PMID: 35636576 DOI: 10.1016/j.pnpbp.2022.110580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
Abstract
Rates of obesity and obesity-associated diseases have increased dramatically in countries with developed economies. Substance use disorders (SUDs) are characterized by the persistent use of the substance despite negative consequences. It has been hypothesized that overconsumption of palatable energy dense food can elicit SUD-like maladaptive behaviors that contribute to persistent caloric intake beyond homeostatic need even in the face of negative consequences. Palatable food and drugs of abuse act on many of the same motivation-related circuits in the brain, and can induce, at least superficially, similar molecular, cellular, and physiological adaptations on these circuits. As such, applying knowledge about the neurobiological mechanisms of SUDs may serve as useful heuristic to better understand the persistent overconsumption of palatable food that contributes to obesity. However, many important differences exist between the actions of drugs of abuse and palatable food in the brain. This warrants caution when attributing weight gain and obesity to the manifestation of a putative SUD-related behavioral disorder. Here, we describe similarities and differences between compulsive drug use in SUDs and overconsumption in obesity and consider the merit of the concept of "food addiction".
Collapse
Affiliation(s)
- Richard M O'Connor
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States of America
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States of America.
| |
Collapse
|
16
|
Raony Í, Domith I, Lourenco MV, Paes-de-Carvalho R, Pandolfo P. Trace amine-associated receptor 1 modulates motor hyperactivity, cognition, and anxiety-like behavior in an animal model of ADHD. Prog Neuropsychopharmacol Biol Psychiatry 2022; 117:110555. [PMID: 35346791 DOI: 10.1016/j.pnpbp.2022.110555] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 03/03/2022] [Accepted: 03/22/2022] [Indexed: 02/03/2023]
Abstract
Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that has recently been implicated in several psychiatric conditions related to monoaminergic dysfunction, such as schizophrenia, substance use disorders, and mood disorders. Although attention-deficit/hyperactivity disorder (ADHD) is also related to changes in monoaminergic neurotransmission, studies that assess whether TAAR1 participates in the neurobiology of ADHD are lacking. We hypothesized that TAAR1 plays an important role in ADHD and might represent a potential therapeutic target. Here, we investigate if TAAR1 modulates behavioral phenotypes in Spontaneously Hypertensive Rats (SHR), the most validated animal model of ADHD, and Wistar Kyoto rats (WKY, used as a control strain). Our results showed that TAAR1 is downregulated in ADHD-related brain regions in SHR compared with WKY. While intracerebroventricular (i.c.v.) administration of the selective TAAR1 antagonist EPPTB impaired cognitive performance in SHR, i.c.v. administration of highly selective TAAR1 full agonist RO5256390 decreased motor hyperactivity, novelty-induced locomotion, and induced an anxiolytic-like behavior. Overall, our findings show that changes in TAAR1 levels/activity underlie behavior in SHR, suggesting that TAAR1 plays a role in the neurobiology of ADHD. Although additional confirmatory studies are required, TAAR1 might be a potential pharmacological target for individuals with this disorder.
Collapse
Affiliation(s)
- Ícaro Raony
- Laboratory of Neurobiology of Animal Behavior, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Ivan Domith
- Laboratory of Cellular Neurobiology, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Roberto Paes-de-Carvalho
- Laboratory of Cellular Neurobiology, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil
| | - Pablo Pandolfo
- Laboratory of Neurobiology of Animal Behavior, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói 24020-141, Brazil.
| |
Collapse
|
17
|
Expression of Trace Amine-Associated Receptors in the Murine and Human Hippocampus Based on Public Transcriptomic Data. Cells 2022; 11:cells11111813. [PMID: 35681508 PMCID: PMC9180029 DOI: 10.3390/cells11111813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Hippocampus is one of the neurogenic zones where adult neurogenesis takes place. This process is quite complex and has a multicomponent regulation. A family of G protein-coupled trace amine-associated receptors (TAARs) was discovered only in 2001, and most of them (TAAR2-TAAR9) were primarily considered olfactory. Recent studies have shown, however, that they are also expressed in the mouse brain, particularly in limbic formations, and can play a role in the regulation of emotional behaviors. The observations in knockout mice indicate that at least two members of the family, TAAR2 and TAAR5, have an impact on the regulation of adult neurogenesis. In the present study, we analyzed the expression of TAARs in the murine and human hippocampus using public RNAseq datasets. Our results indicate a low but detectable level of certain TAARs expression in the hippocampal cells in selected high-quality transcriptomic datasets from both mouse and human samples. At the same time, we observed the difference between humans, where TAAR6 expression was the highest, and murine samples, where TAAR1, TAAR2, TAAR3, TAAR4 and TAAR5 are more pronouncedly expressed. These observations provide further support to the data gained in knockout mice, indicating a role of TAARs in the regulation of adult neurogenesis in the hippocampus.
Collapse
|
18
|
Heal DJ, Smith SL. Prospects for new drugs to treat binge-eating disorder: Insights from psychopathology and neuropharmacology. J Psychopharmacol 2022; 36:680-703. [PMID: 34318734 PMCID: PMC9150143 DOI: 10.1177/02698811211032475] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Binge-eating disorder (BED) is a common psychiatric condition with adverse psychological and metabolic consequences. Lisdexamfetamine (LDX) is the only approved BED drug treatment. New drugs to treat BED are urgently needed. METHODS A comprehensive review of published psychopathological, pharmacological and clinical findings. RESULTS The evidence supports the hypothesis that BED is an impulse control disorder with similarities to ADHD, including responsiveness to catecholaminergic drugs, for example LDX and dasotraline. The target product profile (TPP) of the ideal BED drug combines treating the psychopathological drivers of the disorder with an independent weight-loss effect. Drugs with proven efficacy in BED have a common pharmacology; they potentiate central noradrenergic and dopaminergic neurotransmission. Because of the overlap between pharmacotherapy in attention deficit hyperactivity disorder (ADHD) and BED, drug-candidates from diverse pharmacological classes, which have already failed in ADHD would also be predicted to fail if tested in BED. The failure in BED trials of drugs with diverse pharmacological mechanisms indicates many possible avenues for drug discovery can probably be discounted. CONCLUSIONS (1) The efficacy of drugs for BED is dependent on reducing its core psychopathologies of impulsivity, compulsivity and perseveration and by increasing cognitive control of eating. (2) The analysis revealed a large number of pharmacological mechanisms are unlikely to be productive in the search for effective new BED drugs. (3) The most promising areas for new treatments for BED are drugs, which augment noradrenergic and dopaminergic neurotransmission and/or those which are effective in ADHD.
Collapse
Affiliation(s)
- David J Heal
- David J Heal, DevelRx Ltd, BioCity, Nottingham, NG1 1GF, UK.
| | | |
Collapse
|
19
|
Synan C, Bowen C, Heal DJ, Froger-Colléaux C, Beardsley PM, Dedic N, Hopkins SC, Campbell U, Koblan KS. Ulotaront, a novel TAAR1 agonist with 5-HT1A agonist activity, lacks abuse liability and attenuates cocaine cue-induced relapse in rats. Drug Alcohol Depend 2022; 231:109261. [PMID: 35033729 DOI: 10.1016/j.drugalcdep.2021.109261] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Ulotaront (SEP-363856) is a trace amine-associated receptor 1 (TAAR1) agonist with 5-hydroxytryptamine type 1A (5-HT1A) agonist activity that is currently in Phase 3 clinical development for the treatment of schizophrenia. Unlike available antipsychotics, the efficacy of ulotaront is not mediated by blockade of dopamine D2 or serotonin 5-HT2A receptors. In a short-term randomized clinical trial, ulotaront has demonstrated significant efficacy in the treatment of adults with an acute exacerbation of schizophrenia. Given ulotaront's novel mechanism of action a series of preclinical studies were performed to evaluate its potential abuse liability. METHODS A battery of studies were conducted in male and female rats to evaluate whether ulotaront produces behavioral changes suggestive of human abuse potential. In addition, studies were undertaken to probe the potential for ulotaront to block reinstatement of cocaine-seeking behavior in male rats. RESULTS Ulotaront was not self-administered by rats trained to self-administer amphetamine, cocaine, or heroin. The subjective qualities of ulotaront were distinct from those produced by amphetamine in a drug discrimination procedure. Ulotaront, and buspirone, a non-scheduled anxiolytic with 5-HT1A agonism, partially generalized to the interoceptive cue elicited by 3, 4-methylenedioxymethamphetamine (MDMA). In addition, ulotaront demonstrated a trend to reduce cocaine-primed induced reinstatement, and dose-dependently reduced cue-reinstated responding. CONCLUSION The current results suggest that the TAAR1/5-HT1A agonist ulotaront is not likely to pose a risk for recreational abuse in humans and may have potential therapeutic utility as a treatment of substance use disorders.
Collapse
Affiliation(s)
- Colleen Synan
- Sunovion Pharmaceuticals, Inc., Marlborough, MA, USA
| | - Carrie Bowen
- Sunovion Pharmaceuticals, Inc., Marlborough, MA, USA
| | - David J Heal
- DevelRx Ltd, BioCity, Nottingham, United Kingdom
| | | | - Patrick M Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Nina Dedic
- Sunovion Pharmaceuticals, Inc., Marlborough, MA, USA
| | | | - Una Campbell
- Sunovion Pharmaceuticals, Inc., Marlborough, MA, USA
| | | |
Collapse
|
20
|
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: 44] [Impact Index Per Article: 11.0] [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
|
21
|
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
|
22
|
Caron A, Jane Michael N. New Horizons: Is Obesity a Disorder of Neurotransmission? J Clin Endocrinol Metab 2021; 106:e4872-e4886. [PMID: 34117881 DOI: 10.1210/clinem/dgab421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Indexed: 11/19/2022]
Abstract
Obesity is a disease of the nervous system. While some will view this statement as provocative, others will take it as obvious. Whatever our side is, the pharmacology tells us that targeting the nervous system works for promoting weight loss. It works, but at what cost? Is the nervous system a safe target for sustainable treatment of obesity? What have we learned-and unlearned-about the central control of energy balance in the last few years? Herein we provide a thought-provoking exploration of obesity as a disorder of neurotransmission. We discuss the state of knowledge on the brain pathways regulating energy homeostasis that are commonly targeted in anti-obesity therapy and explore how medications affecting neurotransmission such as atypical antipsychotics, antidepressants, and antihistamines relate to body weight. Our goal is to provide the endocrine community with a conceptual framework that will help expending our understanding of the pathophysiology of obesity, a disease of the nervous system.
Collapse
Affiliation(s)
- Alexandre Caron
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
- Montreal Diabetes Research Center, Montreal, QC, Canada
| | - Natalie Jane Michael
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| |
Collapse
|
23
|
Rehn S, Raymond JS, Boakes RA, Leenaars CHC. A systematic review and meta-analysis of animal models of binge eating - Part 1: Definitions and food/drink intake outcomes. Neurosci Biobehav Rev 2021; 132:1137-1156. [PMID: 34742923 DOI: 10.1016/j.neubiorev.2021.10.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 12/09/2022]
Abstract
Binge eating involves consuming excessive amounts of food within a discrete period of time and is associated with significant impairments in binge-eating disorder and bulimia nervosa. While research on clinical binge eating has provided valuable aetiological insights, animal models allow for closer examination of environmental, biological, and developmental risk factors. Numerous animal models of binge eating exist and differ widely in operational definitions of bingeing, animal characteristics and methodological parameters. The current review aimed to synthesise the available published evidence on these models. A systematic review of binge definitions in 170 articles found most studies displayed good face validity. Meta-analyses on 150 articles confirmed that the amount of food or drink consumed by animals under binge conditions was larger than that of non-binge conditions across many protocols. The meta-regression revealed species, strain, and sex moderated binge effect size, with the largest effect observed in studies with female animals and mice. Risk of bias assessment identified that improved reporting of allocation, baseline characteristics and outcome assessment is required in future studies.
Collapse
Affiliation(s)
- Simone Rehn
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, 2006 NSW, Australia.
| | - Joel S Raymond
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, 2006 NSW, Australia; The University of Sydney, Brain and Mind Centre, 94 Mallett Street, Camperdown, Sydney, NSW 2050, Australia
| | - Robert A Boakes
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, 2006 NSW, Australia
| | - Cathalijn H C Leenaars
- Institute for Laboratory Animal Science, Hannover Medical School, 30625, Hannover, Germany; Department for Health Evidence, Radboud University Medical Centre, 6600, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Trace Amine-Associated Receptor 1 as a Target for the Development of New Antipsychotics: Current Status of Research and Future Directions. CNS Drugs 2021; 35:1153-1161. [PMID: 34655036 DOI: 10.1007/s40263-021-00864-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Schizophrenia is a mental illness associated with an array of symptoms that often result in disability. The primary treatments for schizophrenia are termed antipsychotics. Although antipsychotics modulate a number of different receptor types and subtypes, all currently regulatory agency-approved antipsychotics share in common direct or functional antagonism at the dopamine type 2 receptor (D2R). The majority of people with schizophrenia do not achieve full resolution of their symptoms with antipsychotics, suggesting the need for alternative or complementary approaches. The primary focus of this review is to assess the evidence for the role of the trace amine-associated receptor 1 (TAAR-1) in schizophrenia and the role of TAAR-1 modulators as novel-mechanism antipsychotics. Topics include an overview of TAAR-1 physiology and pathophysiology in schizophrenia, interaction with other neurotransmitter systems, including the dopaminergic, glutamatergic and serotonergic system, and finally, a review of investigational TAAR-1 compounds that have reached Phase II clinical studies in schizophrenia: SEP-363856 (ulotaront) and RO6889450 (ralmitaront). Thus far, results are publicly available only for ulotaront in a relatively young (18-40 years) and acutely exacerbated cohort. These results showed positive effects for overall schizophrenia symptoms without significant tolerability concerns. An ongoing study of ralmitaront will assess specific efficacy in patients with persistent negative symptoms. If trials of TAAR-1 modulators, and other novel-mechanism targets for schizophrenia that are under active study, continue to show positive results, the definition of an antipsychotic may need to be expanded beyond the D2R target in the near future.
Collapse
|
25
|
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: 8] [Impact Index Per Article: 2.0] [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
|
26
|
Kong Q, Zhang H, Wang M, Zhang J, Zhang Y. The TAAR1 inhibitor EPPTB suppresses neuronal excitability and seizure activity in mice. Brain Res Bull 2021; 171:142-149. [PMID: 33811954 DOI: 10.1016/j.brainresbull.2021.03.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 10/21/2022]
Abstract
Epilepsy is a common neurological disease. G protein-coupled receptors (GPCRs) are extensively distributed and play an important role in human health by serving as therapeutic targets for various diseases. As one of the GPCRs, trace amine-associated receptor 1 (TAAR1) has recently aroused increasing interest as a potential therapeutic target for psychiatric disorders. However, the effect of TAAR1 on epileptic seizures remains unclear. We hypothesized that TAAR1 plays an important role in epilepsy and might represent a potential therapeutic target. In this study, we analyzed a mouse epilepsy model and patients with temporal lobe epilepsy (TLE) and observed substantially increased TAAR1 expression compared with the control group. In recordings of hippocampal slices, the TAAR1-specific inhibitor N-(3-ethoxyphenyl)-4-(pyrrolidin-1-yl)-3-(trifluoromethyl) benzamide (EPPTB) suppressed the excitability of hippocampal pyramidal neurons. EPPTB also reduced seizure-like events (SLEs) and seizure activity. Our results suggest that EPPTB attenuates seizure activity and that TAAR1 might be a potential drug target for individuals with epilepsy.
Collapse
Affiliation(s)
- Qingxia Kong
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China; Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China
| | - Hao Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China
| | - Min Wang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China; Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China
| | - Junchen Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China.
| | - Yanke Zhang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China; Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China.
| |
Collapse
|
27
|
Liu J, Seaman R, Johnson B, Wu R, Vu J, Tian J, Zhang Y, Li JX. Activation of trace amine-associated receptor 1 selectively attenuates the reinforcing effects of morphine. Br J Pharmacol 2021; 178:933-945. [PMID: 33247948 DOI: 10.1111/bph.15335] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Trace amine-associated TA1 receptors play critical roles in regulating dopamine transmission. Previous studies showed that pharmacologically or genetically manipulating the activity of TA1 receptors modulates addiction-like behaviours associated with psychostimulants. However, little is known about whether TA1 receptor modulation would regulate the behavioural effects of opioids. EXPERIMENTAL APPROACH Effects of the selective TA1 receptor partial agonist RO5263397 on the addiction-related and antinociceptive effects of morphine were systematically assessed in male rats and mice. KEY RESULTS RO5263397 attenuated the expression of morphine-induced behavioural sensitization in wildtype but not TA1 receptor knockout mice. RO5263397 shifted the dose-effect curve of morphine self-administration downward and reduced the breakpoint in a progressive ratio schedule of reinforcement but did not affect food self-administration in rats. RO5263397 decreased the cue- and drug-induced reinstatement of morphine-seeking behaviour in rats. RO5263397 alone did not trigger reinstatement of morphine-seeking behaviour or change locomotor activity in rats with a history of morphine self-administration. However, RO5263397 did not affect the expression of morphine-induced conditioned place preference in mice or rats. RO5263397 did not affect naltrexone-precipitated jumping behaviour or naltrexone-induced conditioned place aversion in morphine-dependent mice. Furthermore, RO5263397 did not affect the analgesic effects of morphine in an acute nociception model in mice and a chronic pain model in rats. CONCLUSION AND IMPLICATIONS These results indicated that TA1 receptor activation selectively attenuated the reinforcing, but not withdrawal or antinociceptive effects of morphine, suggesting that selective TA1 receptor agonists might be useful to combat opioid addiction, while sparing the analgesic effects.
Collapse
Affiliation(s)
- Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Robert Seaman
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Bernard Johnson
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Jimmy Vu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Jingwei Tian
- School of Pharmacy, Yantai University, Yantai, China
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, North Carolina, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
28
|
Zhukov DA, Vinogradova EP. Trace Amines and Behavior. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420040108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
29
|
Novel 1-Amidino-4-Phenylpiperazines as Potent Agonists at Human TAAR1 Receptor: Rational Design, Synthesis, Biological Evaluation and Molecular Docking Studies. Pharmaceuticals (Basel) 2020; 13:ph13110391. [PMID: 33202687 PMCID: PMC7697893 DOI: 10.3390/ph13110391] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/19/2022] Open
Abstract
Targeting trace amine-associated receptor 1 (TAAR1) receptor continues to offer an intriguing opportunity to develop innovative therapies in different pharmacological settings. Pursuing our endeavors in the search for effective and safe human TAAR1 (hTAAR1) ligands, we synthesized a new series of 1-amidino-4-phenylpiperazine derivatives (1–16) based on the application of a combined pharmacophore model/scaffold simplification strategy for an in-house series of biguanide-based TAAR1 agonists. Most of the novel compounds proved to be more effective than their prototypes, showing nanomolar EC50 values in functional activity at hTAAR1 and low general cytotoxicity (CC50 > 80 µM) when tested on the Vero-76 cell line. In this new series, the main determinant for TAAR1 agonism ability appears to result from the appropriate combination between the steric size and position of the substituents on the phenyl ring rather than from their different electronic nature, since both electron-withdrawing and electron donor groups are permitted. In particular, the ortho-substitution seems to impose a more appropriate spatial geometry to the molecule that entails an enhanced TAAR1 potency profile, as experienced, in the following order, by compounds 15 (2,3-diCl, EC50 = 20 nM), 2 (2-CH3, EC50 = 30 nM), 6 (2-OCH3, EC50 = 93 nM) and 3 (2-Cl, EC50 = 160 nM). Apart from the interest in them as valuable leads for the development of promising hTAAR1 agonists, these simple small molecules have further allowed us to identify the minimal structural requirements for producing an efficient hTAAR1 targeting ability.
Collapse
|
30
|
Zendehdel M, Hassanpour S, Movahedi N. Central and peripheral methylamine-induced hypophagia is mediated via nitric oxide and TAAR 1 in neonatal layer-type chicken. Neurosci Lett 2020; 739:135408. [PMID: 33027685 DOI: 10.1016/j.neulet.2020.135408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 11/28/2022]
Abstract
The aim of the current study was to determine effects of intracerebroventricular (ICV) and intraperitoneal (i.p.) administration of Methylamine (MET) and possible interactions with nitric oxide (NO) and TAAR1 pathways in 24-h fasted (FD24) and ad libitum layer-type chicken. In experiment 1, FD24 chicken ICV injected with MET (15, 30, 45, 60 and 75 μg). In experiment 2, ICV injection of MET (15, 30, 45, 60 and 75 μg) was injected in the ad libitum birds. Experiments 3-4 were similar to experiments 1-2, except chicken i.p. injected with MET (15, 30, 45, 60 and 75 mg/kg). In experiment 5, FD24 birds ICV injected with l-NAME (NO synthesis inhibitor, 100 nmol), MET (75 μg) and co-injection of l-NAME + MET. Experiment 6 was similar to experiment 5, except, ad libitum birds received injections. In experiment 7, FD24 chicken i.p. injected with l-NAME (100 mg/kg), MET (75 mg/kg) and co-injection of l-NAME + MET. In experiment 8, FD24 birds ICV injected with RO5256390 (selective TAAR1 agonist, 10, 20 and 40 μg). In experiment 9, ad libitum birds ICV injected with RO5256390 (10, 20 and 40 μg). In experiment 10, FD24 birds ICV injected with RO5256390 (10 μg), MET (75 μg) and their co-injection. Experiment 11 was similar to experiment 10, except, ad libitum birds received ICV injections. In experiment 12, FD24 chicken i.p. injected with RO5256390 (2.5, 5 and 10 mg/kg). In experiment 13, FD24 chicken i.p. injected with RO5256390 (2.5 mg/kg), MET (75 mg/kg) and RO5256390 + MET. Then cumulative food intake was determined until 120 min after injection. According to the results, ICV injection of MET decreased food intake in FD24 and ad libitum chicken (P < 0.05). MET (i.p.) diminished food consumption in fasted (P < 0.05) but not in ad libitum chicken (P> 0.05). Co-injection of the l-NAME + MET significantly decreased MET-induced hypophagia in FD24 and ad libitum chicken (P < 0.05). MET-induced hypophagia (i.p.) weakened by l-NAME in FD24 chicken (P < 0.05). RO5256390 decreased food intake in FD24 and ad libitum chicken (P < 0.05). Co-injection of RO5256390 + MET increased MET-induced hypophagia in FD24 and ad libitum chicken (P < 0.05). RO5256390 decreased food intake in FD24 chicken (P < 0.05). Co-injection of the RO5256390 + MET amplified MET-induced hypophagia in FD24 chicken (P < 0.05). Based on the findings, MET-induced hypophagia is mediated via NO and TAAR1 pathways on food intake in layer chicken.
Collapse
Affiliation(s)
- Morteza Zendehdel
- Division of Physiology, Faculty of Veterinary Medicine, University of Tehran, 14155-6453, Tehran, Iran
| | - Shahin Hassanpour
- Division of Physiology, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Nima Movahedi
- Division of Physiology, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
31
|
Lee HS, Giunti E, Sabino V, Cottone P. Consummatory, Feeding Microstructural, and Metabolic Effects Induced by Limiting Access to Either a High-Sucrose or a High-Fat Diet. Nutrients 2020; 12:nu12061610. [PMID: 32486207 PMCID: PMC7352440 DOI: 10.3390/nu12061610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Binge eating disorder (BED) is characterized by recurrent binge eating episodes consisting of rapid consumption of excessive amounts of highly palatable, energy-dense food within discrete periods of time. The aim of this study was to test the consummatory, food microstructural, and metabolic effects of a one hour limited access to either a high-sucrose diet (HSD) or a high-fat diet (HFD) in an operant rat model of binge-like eating. Methods: Female rats were subject to a binge-like eating procedure in which a HSD, a HFD, or a standard chow diet were provided in a fixed ratio 1 (FR1) operant schedule of reinforcement. Results: Limiting access to either a HSD or a HFD promoted binge-like eating as compared to the control chow diet. However, binge-like eating of HSD, but not HFD, was based on a true increase in the amount of food consumed, an increased eating rate, and a decrease in the intake of the home-cage standard chow, altogether suggesting an increase in palatability. Moreover, while HSD rats consumed overall less energy than HFD rats, the former were more energy efficient and gained more body weight than the latter. Conclusions: These results provide information on how the quality of food can deeply influence the behavioral and metabolic outcomes of binge-like eating.
Collapse
|
32
|
Hicks C, Sabino V, Cottone P. The Alpha-1 Adrenergic Receptor Antagonist Prazosin Reduces Binge-Like Eating in Rats. Nutrients 2020; 12:nu12061569. [PMID: 32481494 PMCID: PMC7352795 DOI: 10.3390/nu12061569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 01/19/2023] Open
Abstract
Background: Binge-eating disorder is a pervasive addiction-like disorder that is defined by excessive and uncontrollable consumption of food within brief periods of time. The aim of the current study was to examine the role of the brain noradrenergic system in binge-like eating through the use of the alpha-1 adrenergic receptor antagonist prazosin. Methods: For this purpose, we employed a limited access model whereby male Wistar rats were allowed to nosepoke for either chow (Chow rats) or a sugary, highly palatable food (Palatable rats) for 1 h/day. The effects of prazosin (0, 0.5, 1 and 2 mg/kg, i.p.) were tested in a fixed ratio 1 (FR1) and progressive ratio (PR) schedule of reinforcement. Results: The results show that prazosin preferentially reduced the responses for palatable food in a FR1 reinforcement schedule; when tested in a PR schedule of reinforcement, prazosin increased breakpoint in both Chow and Palatable rats, but more potently and more efficaciously in the latter. Our results suggest that prazosin treatment preferentially increased the motivational properties of the palatable diet. Conclusions: The current findings provide the characterization of the effects of prazosin on binge-like eating and offer support to the existing literature showing the important role of the noradrenergic system in addiction-like behavior.
Collapse
|
33
|
Liu J, Johnson B, Wu R, Seaman R, Vu J, Zhu Q, Zhang Y, Li JX. TAAR1 agonists attenuate extended-access cocaine self-administration and yohimbine-induced reinstatement of cocaine-seeking. Br J Pharmacol 2020; 177:3403-3414. [PMID: 32246467 DOI: 10.1111/bph.15061] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 03/06/2020] [Accepted: 03/21/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE The trace amine-associated receptor 1 (TAAR1) negatively modulates dopamine transmission. Our previous studies demonstrated that TAAR1 agonists attenuated cue- and drug-induced cocaine-seeking and increased the elasticity of the cocaine demand curve, in the short-access cocaine self-administration model. Compulsive use of cocaine, which is an essential criterion of cocaine use disorder, can be induced by extended access to cocaine self-administration. EXPERIMENTAL APPROACH To characterize the role of TAAR1 in compulsive cocaine use, we evaluated the effects of activation of TAAR1 on cocaine intake, cocaine binge and cue-induced cocaine-seeking using the extended-access cocaine self-administration model in adult male Sprague-Dawley rats. We also investigated the role of TAAR1 in stress-triggered cocaine relapse by using the α2 -adrenoceptor antagonist yohimbine-induced reinstatement of cocaine-seeking. KEY RESULTS The selective TAAR1 partial agonist RO5263397 attenuated cocaine intake and did not develop tolerance during the 10-day extended-access cocaine self-administration. RO5263397 reduced a 12-h binge intake of cocaine after forced abstinence. RO5263397 also decreased cue-induced cocaine-seeking after prolonged abstinence from extended-access cocaine self-administration. Furthermore, RO5263397 and the selective TAAR1 full agonist RO5166017 reduced yohimbine-induced reinstatement of cocaine-seeking behaviour. CONCLUSION AND IMPLICATIONS Activation of TAAR1 attenuated extended-access cocaine self-administration and stress-induced cocaine reinstatement. These results suggest that TAAR1 agonists are promising pharmacological interventions to treat cocaine use disorder and relapse.
Collapse
Affiliation(s)
- Jianfeng Liu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Bernard Johnson
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Ruyan Wu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA.,School of Medicine, Yangzhou University, Yangzhou, China
| | - Robert Seaman
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Jimmy Vu
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong, China
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, North Carolina, USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
34
|
Dorotenko A, Tur M, Dolgorukova A, Bortnikov N, Belozertseva IV, Zvartau EE, Gainetdinov RR, Sukhanov I. The Action of TAAR1 Agonist RO5263397 on Executive Functions in Rats. Cell Mol Neurobiol 2020; 40:215-228. [PMID: 31734895 DOI: 10.1007/s10571-019-00757-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/31/2019] [Indexed: 12/26/2022]
Abstract
Trace amine-associated receptor 1 (TAAR1) is a widely recognized new perspective target for the neuropsychiatric pharmacological treatment. Despite a growing number of studies investigating TAAR1 role in the animal models of different pathologies, information of TAAR1 agonists impact on executive cognitive functions is limited. The goal of the present study was to evaluate the activity of highly selective partial TAAR1 agonist RO5263397 on various executive cognitive functions. The results of the present study demonstrated that the pretreatment with RO5263397 was able to increase attention and decrease cognitive flexibility in rats. The analysis of the RO5263397 action on impulsivity demonstrated that the TAAR1 activation failed to affect premature responding but was able to slightly modify impulsive choice. Problem solving was resistant to the pharmacological intervention.
Collapse
Affiliation(s)
- Artem Dorotenko
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy str. 6-8, St. Petersburg, Russia, 197022
| | - Margarita Tur
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy str. 6-8, St. Petersburg, Russia, 197022
| | - Antonina Dolgorukova
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy str. 6-8, St. Petersburg, Russia, 197022
| | - Nikita Bortnikov
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy str. 6-8, St. Petersburg, Russia, 197022
| | - Irina V Belozertseva
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy str. 6-8, St. Petersburg, Russia, 197022
| | - Edwin E Zvartau
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy str. 6-8, St. Petersburg, Russia, 197022
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya Emb. 7-9, St. Petersburg, Russia, 199034
- St. Petersburg University Hospital, St. Petersburg State University, Universitetskaya Emb. 7-9, St. Petersburg, Russia, 199034
| | - Ilya Sukhanov
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, Lev Tolstoy str. 6-8, St. Petersburg, Russia, 197022.
| |
Collapse
|
35
|
Abstract
Trace amine-associated receptors (TAARs) are a family of G protein-coupled receptors (GPCRs) that are evolutionarily conserved in vertebrates. The first discovered TAAR1 is mainly expressed in the brain, and is able to detect low abundant trace amines. TAAR1 is also activated by several synthetic compounds and psychostimulant drugs like amphetamine. Activation of TAAR1 by specific agonists can regulate the classical monoaminergic systems in the brain. Further studies have revealed that other TAAR family members are highly expressed in the olfactory system which are termed olfactory TAARs. In vertebrates, olfactory TAARs can specifically recognize volatile or water-soluble amines. Some of these TAAR agonists are produced by decarboxylation of amino acids. In addition, some TAAR agonists are ethological odors that mediate animal innate behaviors. In this study, we provide a comprehensive review of TAAR agonists, including their structures, biosynthesis pathways, and functions.
Collapse
Affiliation(s)
- Zhengrong Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Qian Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China.
| |
Collapse
|
36
|
Cripps MJ, Bagnati M, Jones TA, Ogunkolade BW, Sayers SR, Caton PW, Hanna K, Billacura MP, Fair K, Nelson C, Lowe R, Hitman GA, Berry MD, Turner MD. Identification of a subset of trace amine-associated receptors and ligands as potential modulators of insulin secretion. Biochem Pharmacol 2020; 171:113685. [DOI: 10.1016/j.bcp.2019.113685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022]
|
37
|
Loftis JM, Lasarev M, Shi X, Lapidus J, Janowsky A, Hoffman WF, Huckans M. Trace amine-associated receptor gene polymorphism increases drug craving in individuals with methamphetamine dependence. PLoS One 2019; 14:e0220270. [PMID: 31600226 PMCID: PMC6786581 DOI: 10.1371/journal.pone.0220270] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/27/2019] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Methamphetamine (MA) is a potent agonist at the trace amine-associated receptor 1 (TAAR1). This study evaluated a common variant (CV) in the human TAAR1 gene, synonymous single nucleotide polymorphism (SNP) V288V, to determine the involvement of TAAR1 in MA dependence. METHODS Participants (n = 106) with active MA dependence (MA-ACT), in remission from MA dependence (MA-REM), with active polysubstance dependence, in remission from polysubstance dependence, and with no history of substance dependence completed neuropsychiatric symptom questionnaires and provided blood samples. In vitro expression and function of CV and wild type TAAR1 receptors were also measured. RESULTS The V288V polymorphism demonstrated a 40% increase in TAAR1 protein expression in cell culture, but message sequence and protein function were unchanged, suggesting an increase in translation efficiency. Principal components analysis resolved neuropsychiatric symptoms into four components, PC1 (depression, anxiety, memory, and fatigue), PC2 (pain), PC3 (drug and alcohol craving), and PC4 (sleep disturbances). Analyses of study group and TAAR1 genotype revealed a significant interaction for PC3 (craving response) (p = 0.003). The control group showed no difference in PC3 associated with TAAR1, while adjusted mean craving for the MA-ACT and MA-REM groups, among those with at least one copy of V288V, was estimated to be, respectively, 1.55 (p = 0.036) and 1.77 (p = 0.071) times the adjusted mean craving for those without the TAAR1 SNP. CONCLUSIONS Neuroadaptation to chronic MA use may be altered by TAAR1 genotype and result in increased dopamine signaling and craving in individuals with the V288V genotype.
Collapse
Affiliation(s)
- Jennifer M. Loftis
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
| | - Michael Lasarev
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Oregon Health & Science University and Portland State University School of Public Health, Portland, OR, United States of America
| | - Xiao Shi
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
| | - Jodi Lapidus
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Oregon Health & Science University and Portland State University School of Public Health, Portland, OR, United States of America
| | - Aaron Janowsky
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States of America
| | - William F. Hoffman
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States of America
- Mental Health and Clinical Neurosciences Division, VA Portland Health Care System, Portland, OR, United States of America
| | - Marilyn Huckans
- Research & Development Service, VA Portland Health Care System, Portland, OR, United States of America
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, United States of America
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States of America
- Mental Health and Clinical Neurosciences Division, VA Portland Health Care System, Portland, OR, United States of America
| |
Collapse
|
38
|
Michael ES, Covic L, Kuliopulos A. Trace amine-associated receptor 1 (TAAR1) promotes anti-diabetic signaling in insulin-secreting cells. J Biol Chem 2019; 294:4401-4411. [PMID: 30670596 DOI: 10.1074/jbc.ra118.005464] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/16/2019] [Indexed: 12/25/2022] Open
Abstract
Pancreatic β-cell failure in type 2 diabetes mellitus is a serious challenge that results in an inability of the pancreas to produce sufficient insulin to properly regulate blood glucose levels. Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor expressed by β-cells that has recently been proposed as a potential target for improving glycemic control and suppressing binge eating behaviors. We discovered that TAAR1 is coupled to Gαs-signaling pathways in insulin-secreting β-cells to cause protein kinase A (PKA)/exchange protein activated by cAMP (Epac)-dependent release of insulin, activation of RAF proto-oncogene, Ser/Thr kinase (Raf)-mitogen-activated protein kinase (MAPK) signaling, induction of cAMP response element-binding protein (CREB)-insulin receptor substrate 2 (Irs-2), and increased β-cell proliferation. Interestingly, TAAR1 triggered cAMP-mediated calcium influx and release from internal stores, both of which were required for activation of a MAPK cascade utilizing calmodulin-dependent protein kinase II (CaMKII), Raf, and MAPK/ERK kinase 1/2 (MEK1/2). Together, these data identify TAAR1/Gαs-mediated signaling pathways that promote insulin secretion, improved β-cell function and proliferation, and highlight TAAR1 as a promising new target for improving β-cell health in type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Emily S Michael
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Graduate Biomedical Sciences/Biochemistry, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Lidija Covic
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Graduate Biomedical Sciences/Biochemistry, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Athan Kuliopulos
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Department of Medicine, Tufts University School of Graduate Biomedical Sciences/Biochemistry, Tufts University School of Medicine, Boston, Massachusetts 02111
| |
Collapse
|
39
|
Moore CF, Panciera JI, Sabino V, Cottone P. Neuropharmacology of compulsive eating. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0024. [PMID: 29352024 DOI: 10.1098/rstb.2017.0024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2017] [Indexed: 12/22/2022] Open
Abstract
Compulsive eating behaviour is a transdiagnostic construct observed in certain forms of obesity and eating disorders, as well as in the proposed construct of 'food addiction'. Compulsive eating can be conceptualized as comprising three elements: (i) habitual overeating, (ii) overeating to relieve a negative emotional state, and (iii) overeating despite adverse consequences. Neurobiological processes that include maladaptive habit formation, the emergence of a negative affect, and dysfunctions in inhibitory control are thought to drive the development and persistence of compulsive eating behaviour. These complex psychobehavioural processes are under the control of various neuropharmacological systems. Here, we describe the current evidence implicating these systems in compulsive eating behaviour, and contextualize them within the three elements. A better understanding of the neuropharmacological substrates of compulsive eating behaviour has the potential to significantly advance the pharmacotherapy for feeding-related pathologies.This article is part of a discussion meeting issue 'Of mice and mental health: facilitating dialogue between basic and clinical neuroscientists'.
Collapse
Affiliation(s)
- Catherine F Moore
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA.,Graduate Program for Neuroscience, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA
| | - Julia I Panciera
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA.,MS in Medical Sciences Program, Graduate Medical Sciences, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA.,Master of Public Health Program, Department of Health Policy and Management, Boston University School of Public Health, 715 Albany Street, Boston, MA, USA
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, 72 E. Concord Street, R-618, Boston, MA 02118, USA
| |
Collapse
|
40
|
Activation of trace amine-associated receptor 1 attenuates schedule-induced polydipsia in rats. Neuropharmacology 2019; 144:184-192. [DOI: 10.1016/j.neuropharm.2018.10.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 10/20/2018] [Accepted: 10/22/2018] [Indexed: 11/23/2022]
|
41
|
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
|
42
|
Liu JF, Seaman R, Siemian JN, Bhimani R, Johnson B, Zhang Y, Zhu Q, Hoener MC, Park J, Dietz DM, Li JX. Role of trace amine-associated receptor 1 in nicotine's behavioral and neurochemical effects. Neuropsychopharmacology 2018; 43:2435-2444. [PMID: 29472642 PMCID: PMC6180004 DOI: 10.1038/s41386-018-0017-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/18/2018] [Accepted: 01/20/2018] [Indexed: 12/21/2022]
Abstract
Nicotine addiction and abuse remains a global health issue. To date, the fundamental neurobiological mechanism of nicotine addiction remains incompletely understood. Trace amine-associated receptor 1 (TAAR1) is thought to directly modulate dopaminergic system and are thought to be a neural substrate underlying addictive-like behaviors. We aimed to investigate the role of TAAR1 in nicotine addictive-like behaviors. TAAR1 expression after nicotine treatment was evaluated by western blotting. c-Fos immunofluorescence and in vivo fast-scan cyclic voltammetry were used to examine the activation of brain regions and dopamine release, respectively. We then thoroughly and systematically examined the role of TAAR1 in mediating nicotine-induced sensitization, nicotine discrimination, nicotine self-administration, nicotine demand curve, and the reinstatement of nicotine-seeking. Local pharmacological manipulation was conducted to determine the role of TAAR1 in the nucleus accumbens (NAcs) in the reinstatement of nicotine-seeking. We found that the expression of TAAR1 protein was selectively downregulated in the NAc, with no change in either dorsal striatum or prefrontal cortex. TAAR1 activation was sufficient to block nicotine-induced c-Fos expression in the NAc, while also reducing nicotine-induced dopamine release in the NAc. Systemic administration of TAAR1 agonists attenuated the expression and development of nicotine-induced sensitization, nicotine self-administration, the reinstatement of nicotine-seeking, and increased the elasticity of nicotine demand curve, while intra-NAc infusions of a TAAR1 agonist was sufficient to attenuate nicotine reinstatement. Moreover, TAAR1-knockout rats showed augmented cue-induced and drug-induced reinstatement of nicotine-seeking. These results indicated that modulation of TAAR1 activity regulates nicotine addictive-like behaviors and TAAR1 represents a novel target towards the treatment of nicotine addiction.
Collapse
Affiliation(s)
- Jian-Feng Liu
- 0000 0004 1936 9887grid.273335.3Department of Pharmacology and Toxicology; Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA ,0000 0000 9530 8833grid.260483.bSchool of Pharmacy, Nantong University, 226001 Nantong, China
| | - Robert Seaman
- 0000 0004 1936 9887grid.273335.3Department of Pharmacology and Toxicology; Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Justin N. Siemian
- 0000 0004 1936 9887grid.273335.3Department of Pharmacology and Toxicology; Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Rohan Bhimani
- 0000 0004 1936 9887grid.273335.3Department of Biotechnical and Clinical Laboratory Sciences, University at Buffalo, Buffalo, NY 14214 USA
| | - Bernard Johnson
- 0000 0004 1936 9887grid.273335.3Department of Pharmacology and Toxicology; Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Yanan Zhang
- 0000000100301493grid.62562.35Research Triangle Institute, Research Triangle Park, NC 27709 USA
| | - Qing Zhu
- 0000 0004 1936 9887grid.273335.3Department of Pharmacology and Toxicology; Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA ,0000 0000 9530 8833grid.260483.bSchool of Pharmacy, Nantong University, 226001 Nantong, China
| | - Marius C. Hoener
- 0000 0004 0374 1269grid.417570.0Neuroscience, Ophthalmology and Rare Disease DTA, pRED, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jinwoo Park
- 0000 0004 1936 9887grid.273335.3Department of Biotechnical and Clinical Laboratory Sciences, University at Buffalo, Buffalo, NY 14214 USA
| | - David M. Dietz
- 0000 0004 1936 9887grid.273335.3Department of Pharmacology and Toxicology; Program in Neuroscience, University at Buffalo, Buffalo, NY 14214 USA
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology; Program in Neuroscience, University at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
43
|
Espinoza S, Leo D, Sotnikova TD, Shahid M, Kääriäinen TM, Gainetdinov RR. Biochemical and Functional Characterization of the Trace Amine-Associated Receptor 1 (TAAR1) Agonist RO5263397. Front Pharmacol 2018; 9:645. [PMID: 29977204 PMCID: PMC6022153 DOI: 10.3389/fphar.2018.00645] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/30/2018] [Indexed: 12/13/2022] Open
Abstract
Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor, which signals through elevating intracellular cAMP levels, and expressed in most vertebrates, including rodents and humans. In recent years, several lines of evidence indicated the role of TAAR1 in the regulation of dopaminergic system and its importance in physiological processes such as locomotion, control of emotional states and cognition. In our study, we used RO5263397, a selective TAAR1 agonist, as a tool and characterized its pharmacology in vitro in HEK293 cells and its effects in vivo in tests assessing potential antidepressant and antipsychotic actions. We found that RO5263397 not only increases cAMP levels at very low concentrations but also can induce the phosphorylation of ERK and CREB in a concentration- and time-dependent manner. Like other TAAR1 agonists, RO5263397 potently suppressed high dopamine-dependent hyperactivity in mice lacking the dopamine transporter. Moreover, RO5263397 produced a strong antidepressant-like effect in the forced swim test comparable to fluoxetine. Furthermore, the antidepressant-like activity was blocked by pretreatment with SCH23390 (dopamine D1 receptor antagonist) or NBQX (glutamate AMPA receptor antagonist) but only in part by WAY100635 (serotonin 5HT1A receptor antagonist). In conclusion, our study confirms some previous in vitro and in vivo findings in relation to the pharmacological effects of RO5263397 but more importantly provides new insight on intracellular signaling pathway and other neurotransmitter receptors modulated by TAAR1 receptor activation.
Collapse
Affiliation(s)
- Stefano Espinoza
- Fondazione Istituto Italiano di Tecnologia, Department of Neuroscience and Brain Technologies, Genoa, Italy
| | - Damiana Leo
- Fondazione Istituto Italiano di Tecnologia, Department of Neuroscience and Brain Technologies, Genoa, Italy.,Department of Neurosciences, University of Mons, Mons, Belgium
| | - Tatyana D Sotnikova
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia
| | | | | | - Raul R Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia.,Skolkovo Institute of Science and Technology, Moscow, Russia
| |
Collapse
|
44
|
Schwartz MD, Canales JJ, Zucchi R, Espinoza S, Sukhanov I, Gainetdinov RR. Trace amine-associated receptor 1: a multimodal therapeutic target for neuropsychiatric diseases. Expert Opin Ther Targets 2018; 22:513-526. [DOI: 10.1080/14728222.2018.1480723] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Juan J. Canales
- Division of Psychology, School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | | | - Stefano Espinoza
- Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies Dept., Genoa, Italy
| | - Ilya Sukhanov
- Institute of Pharmacology, Pavlov Medical University, St. Petersburg, Russia
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Center for Translational Biomedicine, Skolkovo Institute of Science and Technology, Moscow, Russia
| |
Collapse
|
45
|
Sukhanov I, Dorofeikova M, Dolgorukova A, Dorotenko A, Gainetdinov RR. Trace Amine-Associated Receptor 1 Modulates the Locomotor and Sensitization Effects of Nicotine. Front Pharmacol 2018; 9:329. [PMID: 29681856 PMCID: PMC5898227 DOI: 10.3389/fphar.2018.00329] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/21/2018] [Indexed: 11/23/2022] Open
Abstract
Trace amine-associated receptor 1 (TAAR1) has emerged as a promising target for addiction treatments because it affects dopamine transmission in the mesolimbic pathway. TAAR1 is involved in the effects of addictive drugs, such as amphetamines, cocaine and ethanol, but the impact of TAAR1 on the effects of nicotine, the psychoactive drug responsible for the development and maintenance of tobacco smoking, has not yet been studied. This study was performed to investigate the possible modulatory action of TAAR1 on the effects of nicotine on locomotor behaviors in rats and mice. Pretreatment with the TAAR1 agonist RO5263397 dose-dependently decreased nicotine-induced hyperlocomotion in rats habituated to locomotor boxes, prevented the development of nicotine sensitization and blocked hypermotility in nicotine-sensitized rats at the highest tested dose (10 mg/kg). The lack of TAAR1 failed to affect the effects of nicotine on the locomotion of mutant mice. Based on the results of the present study, TAAR1 activation attenuates the locomotion-stimulating effects of nicotine on rats. These results further support the previously proposed hypothesis that TAAR1 is a promising target for the prevention and treatment of drug addiction. Further studies aimed at analyzing the effects of TAAR1 agonists on animal models of nicotine addiction are warranted.
Collapse
Affiliation(s)
- Ilya Sukhanov
- Laboratory of Behavioral Pharmacology, Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russia.,Laboratory of Neurochemical Pharmacology, Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Technologia, Genoa, Italy.,Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia
| | - Mariia Dorofeikova
- Laboratory of Behavioral Pharmacology, Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russia
| | - Antonina Dolgorukova
- Laboratory of Behavioral Pharmacology, Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russia
| | - Artem Dorotenko
- Laboratory of Behavioral Pharmacology, Valdman Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russia
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia.,Skolkovo Institute of Science and Technology, Skolkovo Innovation Center, Moscow, Russia
| |
Collapse
|
46
|
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
|
47
|
Abstract
Eating disorders and some forms of obesity are characterized by addictive-like, compulsive eating behavior which contains numerous similarities with compulsive drug use. Food intake is in part mediated by reward and reinforcement processes that can become dysregulated in these disorders. Additionally, impairments in inhibitory control regulation of reward-related responding can cause or further exacerbate binge and compulsive eating. Dysfunctions in two neurotransmitter systems in the mesocorticolimbic pathway, dopamine and glutamate, are thought to contribute to maladaptive eating behaviors. The trace amine associated receptor 1 (TAAR1) system is a promising therapeutic target for compulsive eating behavior due to the role of TAAR1 in synaptic transmission and in the modulation of dopaminergic and glutamatergic signaling. In support of this notion, the TAAR1 agonist RO5256390 was found to decrease the reinforcing effects of palatable food-cues and to reduce binge-like and compulsive-like eating of palatable food. Additionally, prolonged, intermittent access to palatable food was shown to downregulate TAAR1 in the prefrontal cortex, suggesting a potential role for TAAR1 signaling in inhibitory control processes. Research into the role of TAAR1 in addiction, including TAAR1’s ability to modulate psychostimulant reward and reinforcement, bolsters support for TAAR1 agonism as a pharmacological treatment for compulsive eating and other addictive behaviors. This review summarizes the evidence for TAAR1 agonism as a promising therapeutic for compulsive eating behavior as well as the hypothesized mechanism responsible for these effects.
Collapse
Affiliation(s)
- Catherine F Moore
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, United States.,The Graduate Program for Neuroscience, Boston University School of Medicine, Boston, MA, United States
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, United States
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
48
|
Schwartz MD, Palmerston JB, Lee DL, Hoener MC, Kilduff TS. Deletion of Trace Amine-Associated Receptor 1 Attenuates Behavioral Responses to Caffeine. Front Pharmacol 2018; 9:35. [PMID: 29456505 PMCID: PMC5801540 DOI: 10.3389/fphar.2018.00035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/12/2018] [Indexed: 12/18/2022] Open
Abstract
Trace amines (TAs), endogenous amino acid metabolites that are structurally similar to the biogenic amines, are endogenous ligands for trace amine-associated receptor 1 (TAAR1), a GPCR that modulates dopaminergic, serotonergic, and glutamatergic activity. Selective TAAR1 full and partial agonists exhibit similar pro-cognitive, antidepressant- and antipsychotic-like properties in rodents and non-human primates, suggesting TAAR1 as a novel target for the treatment of neurological and psychiatric disorders. We previously reported that TAAR1 partial agonists are wake-promoting in rats and mice, and that TAAR1 knockout (KO) and overexpressing mice exhibit altered sleep-wake and EEG spectral composition. Here, we report that locomotor and EEG spectral responses to the psychostimulants modafinil and caffeine are attenuated in TAAR1 KO mice. TAAR1 KO mice and WT littermates were instrumented for EEG and EMG recording and implanted with telemetry transmitters for monitoring locomotor activity (LMA) and core body temperature (Tb). Following recovery, mice were administered modafinil (25, 50, 100 mg/kg), caffeine (2.5, 10, 20 mg/kg) or vehicle p.o. at ZT6 in balanced order. In WT mice, both modafinil and caffeine dose-dependently increased LMA for up to 6 h following dosing, whereas only the highest dose of each drug increased LMA in KO mice, and did so for less time after dosing. This effect was particularly pronounced following caffeine, such that total LMA response was significantly attenuated in KO mice compared to WT at all doses of caffeine and did not differ from Vehicle treatment. Tb increased comparably in both genotypes in a dose-dependent manner. TAAR1 deletion was associated with reduced wake consolidation following both drugs, but total time in wakefulness did not differ between KO and WT mice. Furthermore, gamma band EEG activity following both modafinil and caffeine treatment was attenuated in TAAR1 KO compared to WT mice. Our results show that TAAR1 is a critical component of the behavioral and cortical arousal associated with two widely used psychostimulants with very different mechanisms of action. Together with our previous findings, these data suggest that TAAR1 is a previously unrecognized component of an endogenous wake-modulating system.
Collapse
Affiliation(s)
- Michael D Schwartz
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| | - Jeremiah B Palmerston
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| | - Diana L Lee
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| | - Marius C Hoener
- Neuroscience, Ophthalmology and Rare Diseases Discovery and Translational Area, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Ltd., Basel, Switzerland
| | - Thomas S Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| |
Collapse
|
49
|
Guariento S, Tonelli M, Espinoza S, Gerasimov AS, Gainetdinov RR, Cichero E. Rational design, chemical synthesis and biological evaluation of novel biguanides exploring species-specificity responsiveness of TAAR1 agonists. Eur J Med Chem 2018; 146:171-184. [DOI: 10.1016/j.ejmech.2018.01.059] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 11/28/2022]
|
50
|
Novelle MG, Diéguez C. Food Addiction and Binge Eating: Lessons Learned from Animal Models. Nutrients 2018; 10:E71. [PMID: 29324652 PMCID: PMC5793299 DOI: 10.3390/nu10010071] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 12/26/2017] [Accepted: 01/09/2018] [Indexed: 01/10/2023] Open
Abstract
The feeding process is required for basic life, influenced by environment cues and tightly regulated according to demands of the internal milieu by regulatory brain circuits. Although eating behaviour cannot be considered "addictive" under normal circumstances, people can become "addicted" to this behaviour, similarly to how some people are addicted to drugs. The symptoms, cravings and causes of "eating addiction" are remarkably similar to those experienced by drug addicts, and both drug-seeking behaviour as eating addiction share the same neural pathways. However, while the drug addiction process has been highly characterised, eating addiction is a nascent field. In fact, there is still a great controversy over the concept of "food addiction". This review aims to summarize the most relevant animal models of "eating addictive behaviour", emphasising binge eating disorder, that could help us to understand the neurobiological mechanisms hidden under this behaviour, and to improve the psychotherapy and pharmacological treatment in patients suffering from these pathologies.
Collapse
Affiliation(s)
- Marta G Novelle
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria (IDIS), CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 15786 Santiago de Compostela, Spain.
| | - Carlos Diéguez
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria (IDIS), CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 15786 Santiago de Compostela, Spain.
| |
Collapse
|