Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 42015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26402461

RESUMO

The lateral amygdala (LA) acquires differential coding of predictive and non-predictive fear stimuli that is critical for proper fear memory assignment. The neurotransmitter dopamine is an important modulator of LA activity and facilitates fear memory formation, but whether dopamine neurons aid in the establishment of discriminative fear coding by the LA is unknown. NMDA-type glutamate receptors in dopamine neurons are critical for the prevention of generalized fear following an aversive experience, suggesting a potential link between a cell autonomous function of NMDAR in dopamine neurons and fear coding by the LA. Here, we utilized mice with a selective genetic inactivation functional NMDARs in dopamine neurons (DAT-NR1 KO mice) combined with behavior, in vivo electrophysiology, and ex vivo electrophysiology in LA neurons to demonstrate that plasticity underlying differential fear coding in the LA is regulated by NMDAR signaling in dopamine neurons and alterations in this plasticity is associated non-discriminative cued-fear responses.


Assuntos
Complexo Nuclear Basolateral da Amígdala/fisiologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/fisiologia , Medo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Técnicas de Inativação de Genes , Masculino , Camundongos Knockout , Plasticidade Neuronal , Receptores de N-Metil-D-Aspartato/genética
2.
J Neurosci ; 35(37): 12917-31, 2015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-26377476

RESUMO

The endogenous dynorphin-κ opioid receptor (KOR) system encodes the dysphoric component of the stress response and controls the risk of depression-like and addiction behaviors; however, the molecular and neural circuit mechanisms are not understood. In this study, we report that KOR activation of p38α MAPK in ventral tegmental (VTA) dopaminergic neurons was required for conditioned place aversion (CPA) in mice. Conditional genetic deletion of floxed KOR or floxed p38α MAPK by Cre recombinase expression in dopaminergic neurons blocked place aversion to the KOR agonist U50,488. Selective viral rescue by wild-type KOR expression in dopaminergic neurons of KOR(-/-) mice restored U50,488-CPA, whereas expression of a mutated form of KOR that could not initiate p38α MAPK activation did not. Surprisingly, while p38α MAPK inactivation blocked U50,488-CPA, p38α MAPK was not required for KOR inhibition of evoked dopamine release measured by fast scan cyclic voltammetry in the nucleus accumbens. In contrast, KOR activation acutely inhibited VTA dopaminergic neuron firing, and repeated exposure attenuated the opioid response. This adaptation to repeated exposure was blocked by conditional deletion of p38α MAPK, which also blocked KOR-induced tyrosine phosphorylation of the inwardly rectifying potassium channel (GIRK) subunit Kir3.1 in VTA dopaminergic neurons. Consistent with the reduced response, GIRK phosphorylation at this amino terminal tyrosine residue (Y12) enhances channel deactivation. Thus, contrary to prevailing expectations, these results suggest that κ opioid-induced aversion requires regulation of VTA dopaminergic neuron somatic excitability through a p38α MAPK effect on GIRK deactivation kinetics rather than by presynaptically inhibiting dopamine release. SIGNIFICANCE STATEMENT: Kappa opioid receptor (KOR) agonists have the potential to be effective, nonaddictive analgesics, but their therapeutic utility is greatly limited by adverse effects on mood. Understanding how KOR activation produces dysphoria is key to the development of better analgesics and to defining how the endogenous dynorphin opioids produce their depression-like effects. Results in this study show that the aversive effects of κ receptor activation required arrestin-dependent p38α MAPK activation in dopamine neurons but did not require inhibition of dopamine release in the nucleus accumbens. Thus, contrary to the prevailing view, inhibition of mesolimbic dopamine release does not mediate the aversive effects of KOR activation and functionally selective κ opioids that do not activate arrestin signaling may be effective analgesics lacking dysphoric effects.


Assuntos
Aprendizagem da Esquiva/fisiologia , Dopamina/fisiologia , Neurônios Dopaminérgicos/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Receptores Opioides kappa/fisiologia , Área Tegmentar Ventral/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Potenciais de Ação/efeitos dos fármacos , Analgésicos não Narcóticos/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Dopamina/metabolismo , Ativação Enzimática , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Técnicas de Silenciamento de Genes , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Accumbens/metabolismo , Fosforilação/efeitos dos fármacos , Potássio/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores Opioides kappa/deficiência , Receptores Opioides kappa/genética , Proteínas Recombinantes de Fusão/farmacologia , Teste de Desempenho do Rota-Rod , Neurônios Serotoninérgicos/fisiologia , Área Tegmentar Ventral/citologia , Proteínas Quinases p38 Ativadas por Mitógeno/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...