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










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 33(1): 668-682, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30024789

RESUMO

Nogo-A is a key inhibitory molecule of axon regeneration in oligodendrocytes. However, little is known about its role in adult neurons. In this study, we showed an important function of Nogo-A on regulation of inflammatory pain in dorsal root ganglion (DRG) neurons. In adult rats with complete Freund's adjuvant (CFA) hind paw inflammation, DRG neurons showed a significant increase in Nogo-A expression. Disruption of Nogo-A signaling with Nogo-66 receptor antagonist peptide, Nogo-A blocking antibody, Nogo-A short hairpin RNA, or Nogo-A gene knockout attenuated CFA-induced inflammatory heat hyperalgesia. Moreover, disruption of Nogo-A signaling suppressed the function and expression in DRG neurons of the transient receptor potential vanilloid subfamily member (TRPV)-1 channel, which is known to be the endogenous transducer of noxious heat during inflammation. These effects were accompanied with a reduction in LIM domain kinase (LIMK)/cofilin phosphorylation and actin polymerization. Similar disruption of actin filament architecture by direct action of Latrunculin A reduced the TRPV-1 activity and up-regulation of TRPV-1 protein caused by CFA. We conclude that Nogo-A plays an essential role in the development of inflammatory heat hyperalgesia, partly through maintaining TRPV-1 function via activation of the LIMK/cofilin pathway, which regulates actin filament dynamics. These findings support a therapeutic potential of modulating Nogo-A signaling in pain management.-Hu, F., Liu, H.-C., Su, D.-Q., Chen, H.-J., Chan, S.-O., Wang, Y., Wang, J. Nogo-A promotes inflammatory heat hyperalgesia by maintaining TRPV-1 function in the rat dorsal root ganglion neuron.


Assuntos
Gânglios Espinais/patologia , Temperatura Alta/efeitos adversos , Hiperalgesia/etiologia , Inflamação Neurogênica/complicações , Neurônios/patologia , Proteínas Nogo/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cofilina 1/metabolismo , Gânglios Espinais/imunologia , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Quinases Lim/metabolismo , Masculino , Inflamação Neurogênica/metabolismo , Inflamação Neurogênica/patologia , Neurônios/imunologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
2.
Sci Signal ; 7(331): ra61, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24962708

RESUMO

Changes in the actin cytoskeleton in neurons are associated with synaptic plasticity and may also be involved in mechanisms of nociception. We found that the LIM motif-containing protein kinases (LIMKs), which regulate actin dynamics, promoted the development of inflammatory hyperalgesia (excessive sensitivity to painful stimuli). Pain is sensed by the primary sensory neurons of dorsal root ganglion (DRG). In rats injected with complete Freund's adjuvant (CFA), which induces inflammatory heat hyperalgesia, DRG neurons showed an increase in LIMK activity and in the phosphorylation and thus inhibition of the LIMK substrate cofilin, an actin-severing protein. Manipulations that reduced LIMK activity or abundance, prevented the phosphorylation of cofilin, or disrupted actin filaments in DRG neurons attenuated CFA-induced heat hyperalgesia. Inflammatory stimuli stimulated actin polymerization and enhanced the response of the cation channel TRPV1 (transient receptor potential V1) to capsaicin in DRG neurons, effects that were reversed by the knockdown of LIMK or preventing cofilin phosphorylation. Furthermore, inflammatory stimuli caused the serine phosphorylation of TRPV1, which was abolished by preventing cofilin phosphorylation in DRG neurons. We conclude that LIMK-dependent actin rearrangement in primary sensory neurons, leading to altered TRPV1 sensitivity, is involved in the development of inflammatory hyperalgesia.


Assuntos
Actinas/metabolismo , Hiperalgesia/metabolismo , Quinases Lim/metabolismo , Células Receptoras Sensoriais/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Western Blotting , Células Cultivadas , Cofilina 1/metabolismo , Adjuvante de Freund/administração & dosagem , Adjuvante de Freund/toxicidade , Gânglios Espinais/citologia , Temperatura Alta , Hiperalgesia/induzido quimicamente , Inflamação/induzido quimicamente , Inflamação/metabolismo , Quinases Lim/genética , Masculino , Fosforilação , Polimerização , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo
3.
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
4.
Neurosci Bull ; 28(2): 182-92, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22466129

RESUMO

OBJECTIVE: Our previous study identified Threonine 161 (Thr-161), located in the second intracellular loop of the δ-opioid receptor (DOR), as the only consensus phosphorylation site for cyclin-dependent kinase 5 (Cdk5). The aim of this study was to assess the function of DOR phosphorylation by Cdk5 in complete Freund's adjuvant (CFA)-induced inflammatory pain and morphine tolerance. METHODS: Dorsal root ganglion (DRG) neurons of rats with CFA-induced inflammatory pain were acutely dissociated and the biotinylation method was used to explore the membrane localization of phosphorylated DOR at Thr-161 (pThr-161-DOR), and paw withdrawal latency was measured after intrathecal delivery of drugs or Tat-peptide, using a radiant heat stimulator in rats with CFA-induced inflammatory pain. RESULTS: Both the total amount and the surface localization of pThr-161-DOR were significantly enhanced in the ipsilateral DRG following CFA injection. Intrathecal delivery of the engineered Tat fusion-interefering peptide corresponding to the second intracellular loop of DOR (Tat-DOR-2L) increased inflammatory hypersensitivity, and inhibited DOR- but not µ-opioid receptor-mediated spinal analgesia in CFA-treated rats. However, intrathecal delivery of Tat-DOR-2L postponed morphine antinociceptive tolerance in rats with CFA-induced inflammatory pain. CONCLUSION: Phosphorylation of DOR at Thr-161 by Cdk5 attenuates hypersensitivity and potentiates morphine tolerance in rats with CFA-induced inflammatory pain, while disruption of the phosphorylation of DOR at Thr-161 attenuates morphine tolerance.


Assuntos
Tolerância a Medicamentos/fisiologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/patologia , Hipersensibilidade/tratamento farmacológico , Hipersensibilidade/patologia , Inflamação/patologia , Morfina/efeitos adversos , Receptores Opioides delta/antagonistas & inibidores , Sequência de Aminoácidos/efeitos dos fármacos , Sequência de Aminoácidos/fisiologia , Analgésicos Opioides/efeitos adversos , Animais , Modelos Animais de Doenças , Hiperalgesia/etiologia , Hipersensibilidade/etiologia , Inflamação/induzido quimicamente , Inflamação/complicações , Masculino , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/química , Receptores Opioides delta/metabolismo , Treonina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...