Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 357(1): 134-44, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26791601

RESUMO

Trace amine-associated receptor 1 (TAAR1) has been implicated in the behavioral effects of amphetamine-type stimulant drugs in rodents. TAAR1 has also been suggested as a target for novel medications to treat psychostimulant addiction. We previously reported that binding affinities at TAAR1 can differ between structural analogs of psychostimulants, and species differences have been observed. In this study, we complement our previous findings with additional substances and the determination of functional activation potencies. In summary, we present here pharmacological in vitro profiles of 101 psychoactive substances at human, rat, and mouse TAAR1. p-Tyramine, ß-phenylethylamine, and tryptamine were included as endogenous comparator compounds. Functional cAMP measurements and radioligand displacement assays were conducted with human embryonic kidney 293 cells that expressed human, rat, or mouse TAAR1. Most amphetamines, phenethylamine, and aminoindanes exhibited potentially physiologically relevant rat and mouse TAAR1 activation (EC50 < 5 µM) and showed full or partial (Emax < 80%) agonist properties. Cathinone derivatives, including mephedrone and methylenedioxypyrovalerone, exhibited weak (EC50 = 5-10 µM) to negligible (EC50 > 10 µM) binding properties at TAAR1. Pipradrols, including methylphenidate, exhibited no affinity for TAAR1. We found considerable species differences in activity at TAAR1 among the highly active ligands, with a rank order of rat > mouse > human. This characterization provides information about the pharmacological profile of psychoactive substances. The species differences emphasize the relevance of clinical studies to translationally complement rodent studies on the role of TAAR1 activity for psychoactive substances.


Assuntos
Psicotrópicos/farmacologia , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Anfetaminas/farmacologia , Animais , Células HEK293 , Humanos , Camundongos , Fenetilaminas/farmacologia , Ratos , Receptores Acoplados a Proteínas G/agonistas , Especificidade da Espécie , Relação Estrutura-Atividade , Triptaminas/farmacologia , Tiramina/farmacologia
2.
Eur Neuropsychopharmacol ; 25(11): 2049-61, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26372541

RESUMO

Trace amine-associated receptor 1 (TAAR1) activation by selective endogenous agonists modulates dopaminergic neurotransmission. This results in antipsychotic-like behavior in vivo which might be initiated by an interaction of TAAR1 and dopamine D2L receptor (D2R). Here we analyzed the functional link between TAAR1 and D2R using highly potent and selective TAAR1 agonists, and newly generated tools such as TAAR1 knock-out and TAAR1 overexpressing rats as well as specific anti-rat TAAR1 antibodies. We provide data from co-immunoprecipitation experiments supporting a functional interaction of the two receptors in heterologous cells and in brain tissue. Interaction of TAAR1 with D2R altered the subcellular localization of TAAR1 and increased D2R agonist binding affinity. Using specific ß-arrestin 2 (ßArr2) complementation assays we show that the interaction of TAAR1 with D2R reduced ßArr2 recruitment to D2R. In addition, we report that besides Gαs-protein signaling TAAR1 also signals via ßArr2. In the presence of D2R, cAMP signaling of TAAR1 was reduced while its ßArr2 signaling was enhanced, resulting in reduced GSK3ß activation. These results demonstrate that ßArr2 signaling may be an important pathway for TAAR1 function and that the activation of the TAAR1-D2R complex negatively modulates GSK3ß signaling. Given that patients with schizophrenia or bipolar disorder show increased GSK3ß signaling, such a reduction of GSK3ß signaling triggered by the interaction of D2R with activated TAAR1 further supports TAAR1 as a target for the treatment of psychiatric disorders.


Assuntos
Arrestinas/metabolismo , Encéfalo/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetulus , AMP Cíclico/metabolismo , Técnicas de Inativação de Genes , Glicogênio Sintase Quinase 3 beta , Células HEK293 , Humanos , Ratos Transgênicos , Ratos Wistar , Receptores de Dopamina D2/agonistas , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , beta-Arrestina 2 , beta-Arrestinas
3.
Neuropharmacology ; 99: 546-53, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26318099

RESUMO

BACKGROUND: N-2-methoxybenzyl-phenethylamines (NBOMe drugs) are newly used psychoactive substances with poorly defined pharmacological properties. The aim of the present study was to characterize the receptor binding profiles of a series of NBOMe drugs compared with their 2,5-dimethoxy-phenethylamine analogs (2C drugs) and lysergic acid diethylamide (LSD) in vitro. METHODS: We investigated the binding affinities of 2C drugs (2C-B, 2C-C, 2C-D, 2C-E, 2C-H, 2C-I, 2C-N, 2C-P, 2C-T-2, 2C-T-4, 2C-T-7, and mescaline), their NBOMe analogs, and LSD at monoamine receptors and determined functional 5-hydroxytryptamine-2A (5-HT2A) and 5-HT2B receptor activation. Binding at and the inhibition of monoamine uptake transporters were also determined. Human cells that were transfected with the respective human receptors or transporters were used (with the exception of trace amine-associated receptor-1 [TAAR1], in which rat/mouse receptors were used). RESULTS: All of the compounds potently interacted with serotonergic 5-HT2A, 5-HT2B, 5-HT2C receptors and rat TAAR1 (most Ki and EC50: <1 µM). The N-2-methoxybenzyl substitution of 2C drugs increased the binding affinity at serotonergic 5-HT2A, 5-HT2C, adrenergic α1, dopaminergic D1-3, and histaminergic H1 receptors and monoamine transporters but reduced binding to 5-HT1A receptors and TAAR1. As a result, NBOMe drugs were very potent 5-HT2A receptor agonists (EC50: 0.04-0.5 µM) with high 5-HT2A/5-HT1A selectivity and affinity for adrenergic α1 receptors (Ki: 0.3-0.9 µM) and TAAR1 (Ki: 0.06-2.2 µM), similar to LSD, but not dopaminergic D1-3 receptors (most Ki:>1 µM), unlike LSD. CONCLUSION: The binding profile of NBOMe drugs predicts strong hallucinogenic effects, similar to LSD, but possibly more stimulant properties because of α1 receptor interactions.


Assuntos
Fenetilaminas/farmacologia , Psicotrópicos/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Camundongos , Estrutura Molecular , Células NIH 3T3 , Inibidores da Captação de Neurotransmissores/farmacologia , Fenetilaminas/química , Ligação Proteica , Psicotrópicos/química , Ensaio Radioligante , Receptor 5-HT1A de Serotonina/metabolismo , Receptores 5-HT2 de Serotonina/metabolismo
4.
Proc Natl Acad Sci U S A ; 106(47): 20081-6, 2009 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-19892733

RESUMO

Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor (GPCR) that is nonselectively activated by endogenous metabolites of amino acids. TAAR1 is considered a promising drug target for the treatment of psychiatric and neurodegenerative disorders. However, no selective ligand to identify TAAR1-specific signaling mechanisms is available yet. Here we report a selective TAAR1 antagonist, EPPTB, and characterize its physiological effects at dopamine (DA) neurons of the ventral tegmental area (VTA). We show that EPPTB prevents the reduction of the firing frequency of DA neurons induced by p-tyramine (p-tyr), a nonselective TAAR1 agonist. When applied alone, EPPTB increases the firing frequency of DA neurons, suggesting that TAAR1 either exhibits constitutive activity or is tonically activated by ambient levels of endogenous agonist(s). We further show that EPPTB blocks the TAAR1-mediated activation of an inwardly rectifying K(+) current. When applied alone, EPPTB induces an apparent inward current, suggesting the closure of tonically activated K(+) channels. Importantly, these EPPTB effects were absent in Taar1 knockout mice, ruling out off-target effects. We additionally found that both the acute application of EPPTB and the constitutive genetic lack of TAAR1 increase the potency of DA at D2 receptors in DA neurons. In summary, our data support that TAAR1 tonically activates inwardly rectifying K(+) channels, which reduces the basal firing frequency of DA neurons in the VTA. We hypothesize that the EPPTB-induced increase in the potency of DA at D2 receptors is part of a homeostatic feedback mechanism compensating for the lack of inhibitory TAAR1 tone.


Assuntos
Benzamidas/química , Benzamidas/metabolismo , Dopamina/metabolismo , Sistema Límbico , Neurônios/metabolismo , Pirrolidinas/química , Pirrolidinas/metabolismo , Receptores Acoplados a Proteínas G , Área Tegmentar Ventral , Potenciais de Ação/fisiologia , Animais , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Humanos , Sistema Límbico/citologia , Sistema Límbico/metabolismo , Camundongos , Camundongos Knockout , Estrutura Molecular , Neurônios/citologia , Oócitos/citologia , Oócitos/fisiologia , Técnicas de Patch-Clamp , Ratos , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...