Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 32(42): 14709-21, 2012 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-23077056

RESUMO

The number of functional transient receptor potential vanilloid 1 (TRPV1) channels at the surface, especially at the peripheral terminals of primary sensory neurons, regulates heat sensitivity, and increased surface localization of TRPV1s contributes to heat hyperalgesia. However, the mechanisms for regulating TRPV1 surface localization are essentially unknown. Here, we show that cyclin-dependent kinase 5 (Cdk5), a new player in thermal pain sensation, positively regulates TRPV1 surface localization. Active Cdk5 was found to promote TRPV1 anterograde transport in vivo, suggesting a regulatory role of Cdk5 in TRPV1 membrane trafficking. TRPV1-containing vesicles bind to the forkhead-associated (FHA) domain of the KIF13B (kinesin-3 family member 13B) and are thus delivered to the cell surface. Overexpression of Cdk5 or its activator p35 promoted and inhibition of Cdk5 activity prevented the KIF13B-TRPV1 association, indicating that Cdk5 promotes TRPV1 anterograde transport by mediating the motor-cargo association. Cdk5 phosphorylates KIF13B at Thr-506, a residue located in the FHA domain. T506A mutation reduced the motor-cargo interaction and the cell-permeable TAT-T506 peptide, targeting to the Thr-506, decreased TRPV1 surface localization, demonstrating the essential role of Thr-506 phosphorylation in TRPV1 transport. Moreover, complete Freund's adjuvant (CFA) injection-induced activation of Cdk5 increased the anterograde transport of TRPV1s, contributing to the development and possibly the maintenance of heat hyperalgesia, whereas intrathecal delivery of the TAT-T506 peptide alleviated CFA-induced heat hyperalgesia in rats. Thus, Cdk5 regulation of TRPV1 membrane trafficking is a fundamental mechanism controlling the heat sensitivity of nociceptors, and moderate inhibition of Thr-506 phosphorylation during inflammation might be helpful for the treatment of inflammatory thermal pain.


Assuntos
Quinase 5 Dependente de Ciclina/fisiologia , Temperatura Alta/efeitos adversos , Cinesinas/fisiologia , Glicoproteínas de Membrana/metabolismo , Nociceptores/metabolismo , Limiar da Dor/fisiologia , Canais de Cátion TRPV/metabolismo , Animais , Animais Recém-Nascidos , Células CHO , Linhagem Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Masculino , Fosforilação , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/antagonistas & inibidores , Treonina/metabolismo
2.
J Neurosci ; 29(11): 3551-64, 2009 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-19295160

RESUMO

Morphine is the most commonly used and most effective analgesic in the clinic. However, its use is limited by the tolerance. Evidence indicates that the delta-opioid receptor (DOR) is essential for morphine antinociceptive tolerance; however, their underlying mechanisms are poorly understood. Here, we show that cyclin-dependent kinase 5 (Cdk5), activated in morphine antinociceptive tolerance, directly phosphorylates DOR at Thr-161 in DRG neurons. Cdk5 was found to phosphorylate Thr-161 in the second loop of DOR, but not the corresponding residue in the mu-opioid receptor (MOR). Phosphorylation at Thr-161 is required for normal cell surface expression of DOR, and the formation of DOR-MOR heterodimers. Our studies indicated that inhibition of Cdk5 activity or overexpression of a DOR mutant lacking the Cdk5 phosphorylation site displayed relatively low cell surface expression and relatively low abilities to form heterodimers of DOR and MOR; intrathecal delivery of a construct expressing the T161A mutant of DOR attenuated morphine antinociceptive tolerance in rats, suggesting that Thr-161 phosphorylation of DOR contributed to Cdk5-mediated morphine antinociceptive tolerance. Furthermore, an engineered Tat fusion-interfering peptide corresponding to the second intracellular loop of DOR (Tat-DOR-2L), reduced the cell surface expression of DOR, disrupted the formation of DOR-MOR heterodimers, and significantly attenuated the development of morphine antinociceptive tolerance after intrathecal injection. The present study indicates that Cdk5-mediated phosphorylation of DOR at Thr-161 plays a crucial role in the development of morphine tolerance and suggests the possibility of targeting DOR phosphorylation at Thr-161 to attenuate morphine antinociceptive tolerance during pain management.


Assuntos
Quinase 5 Dependente de Ciclina/antagonistas & inibidores , Quinase 5 Dependente de Ciclina/metabolismo , Tolerância a Medicamentos/fisiologia , Morfina/farmacologia , Medição da Dor/métodos , Receptores Opioides delta/antagonistas & inibidores , Receptores Opioides delta/metabolismo , Treonina/metabolismo , Sequência de Aminoácidos , Analgésicos Opioides/farmacologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Quinase 5 Dependente de Ciclina/genética , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Medição da Dor/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/genética , Treonina/genética
3.
Neurochem Res ; 32(9): 1592-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17562172

RESUMO

A previous knockout study revealed the critical role of downstream regulatory element antagonist modulator (DREAM) in pain processing in the spinal cord by transcriptional regulation of prodynorphin (PPD) gene. Here, we report that, in contrast to the nuclear localization of other transcription factors, DREAM showed a punctate staining pattern in rat spinal dorsal horn in immunofluorescent analysis, with a membrane localization profile in some neurons and its expression accumulated in the inner zone of lamina II. In an inflammatory pain model induced by complete Freund's adjuvant (CFA) injection, we used Western blot analysis and detected transient upregulation of DREAM in the nuclear fraction of ipsilateral spinal dorsal horn at 2 h and 6 h post-injection, and a slow upregulation in the membrane fraction for 7 days. These studies suggest that DREAM might have other roles in pain modulation in the spinal cord in addition to its well-known role as a transcriptional repressor.


Assuntos
Inflamação/metabolismo , Proteínas Interatuantes com Canais de Kv/metabolismo , Dor/metabolismo , Proteínas Repressoras/metabolismo , Medula Espinal/metabolismo , Animais , Adjuvante de Freund , Masculino , Dor/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Regulação para Cima
4.
Pain ; 127(1-2): 109-20, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16996690

RESUMO

Cyclin-dependent kinase 5 (Cdk5) is a unique member of the CDK family. It is predominantly expressed in postmitotic neurons and has been implicated in neuronal plasticity. The present study showed that Cdk5 and p35 were expressed in primary sensory and dorsal horn neurons, while p25, an N-terminal truncated derivative of p35, could only be detected in the dorsal horn neurons. Importantly, in the case of control rats, the p35 protein level was much higher in small- and medium-diameter DRG neurons than it was in large neurons. Following CFA injection, Cdk5 activity was upregulated in both primary sensory and dorsal horn neurons. Cdk5 activation in DRG neurons required p35, whereas p25 was required in the dorsal horn. Intrathecal pretreatment with Roscovitine, a specific inhibitor of Cdk5 activity, and intrathecal delivery of the DN-Cdk5(N144) gene both alleviated CFA-induced heat hyperalgesia but not mechanical allodynia. In contrast, overexpression of Cdk5, p35 or p25 in primary sensory and dorsal horn neurons significantly enhanced heat hyperalgesia. We conclude that Cdk5/p35 and Cdk5/p25 complexes in primary sensory and dorsal horn neurons may potentially be involved in nociceptive transmission after inflammation and may be employed in synaptic plasticity underlying pain hypersensitization.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Temperatura Alta/efeitos adversos , Hiperalgesia/fisiopatologia , Neurite (Inflamação)/fisiopatologia , Neurônios Aferentes/enzimologia , Células do Corno Posterior/enzimologia , Animais , Ativação Enzimática , Hiperalgesia/etiologia , Masculino , Neurite (Inflamação)/etiologia , Ratos , Ratos Sprague-Dawley
5.
Exp Neurol ; 202(2): 313-23, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16879821

RESUMO

Cyclin-dependent kinase 5 (CDK5), a unique member of the CDK family of cyclin-dependent kinases, is predominantly expressed in postmitotic neurons with proposed roles in both cell survival and programmed cell death. To understand how CDK5 participates in such disparate cellular outcomes, we investigated whether activation of CDK5 could mediate neuroprotection from serum deprivation by mu-opioid receptor agonist in differentiated SH-SY5Y cells and primary hippocampal neurons. We found that CDK5 kinase activity decreased following serum deprivation in differentiated SH-SY5Y cells coincident with increased cell loss and activation of caspases cascade activation, which was reversed by opioid antagonist. Overexpression of CDK5 in serum-free medium reversed activation of caspase cascade and augmented DAMGO neuroprotection. Blocking CDK5 activity by pharmacologic inhibitor, roscovitine or overexpression of dominant negative CDK5 augmented activation of cell death markers and diminished mu-opioid receptor agonist protection. Reduction in CDK5 activity corresponded to reduction in protein levels of CDK5 activator p35 during serum deprivation which was also reversed by mu-opioid receptor agonist. Phosphorylation of STAT3 at Serine 727 by CDK5 decreased during serum deprivation, and partly recovered by mu-opioid agonist. PI3K signaling pathway was not required for CDK5-mediated mu-opioid neuroprotection against serum deprivation. These findings indicate that neuroprotection by mu-opioid receptor agonist against serum deprivation is mediated by activation of CDK5 through up-regulation of p35 and phosphorylation of STAT3 by CDK5 may contribute to the neuroprotection.


Assuntos
Quinase 5 Dependente de Ciclina/fisiologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptores Opioides mu/agonistas , Soro/metabolismo , Animais , Western Blotting/métodos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Interações Medicamentosas , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipocampo/citologia , Humanos , Mutagênese/fisiologia , Naloxona/farmacologia , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/antagonistas & inibidores , Sais de Tetrazólio , Tiazóis , Fatores de Tempo , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...