Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 306: 64-75, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29715474

RESUMEN

Paclitaxel is a cytotoxic drug which frequently causes sensory peripheral neuropathy in patients. Increasing evidence suggests that altered intracellular calcium (Ca2+) signals play an important role in the pathogenesis of this condition. In the present study, we examined the interplay between Ca2+ release channels in the endoplasmic reticulum (ER) and Ca2+ permeable channels in the plasma membrane in the context of paclitaxel mediated neurotoxicity. We observed that in small to medium size dorsal root ganglia neurons (DRGN) the inositol-trisphosphate receptor (InsP3R) type 1 was often concentrated in the periphery of cells, which is in contrast to homogenous ER distribution. G protein-coupled designer receptors were used to further elucidate phosphoinositide mediated Ca2+ signaling: This approach showed strong InsP3 mediated Ca2+ signals close to the plasma membrane, which can be amplified by Ca2+ entry through TRPV4 channels. In addition, our results support a physical interaction and partial colocalization of InsP3R1 and TRPV4 channels. In the context of paclitaxel-induced neurotoxicity, blocking Ca2+ influx through TRPV4 channels reduced cell death in cultured DRGN. Pretreatment of mice with the pharmacological TRPV4 inhibitor HC067047 prior to paclitaxel injections prevented electrophysiological and behavioral changes associated with paclitaxel-induced neuropathy. In summary, these results underline the relevance of TRPV4 signaling for the pathogenesis of paclitaxel-induced neuropathy and suggest novel preventive strategies.


Asunto(s)
Antineoplásicos Fitogénicos/toxicidad , Síndromes de Neurotoxicidad/prevención & control , Paclitaxel/toxicidad , Canales Catiónicos TRPV/antagonistas & inhibidores , Animales , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Ganglios Espinales/patología , Inmunohistoquímica , Receptores de Inositol 1,4,5-Trifosfato/genética , Ratones Endogámicos C57BL , Síndromes de Neurotoxicidad/patología , Ratas , Ratas Wistar , Transfección
2.
J Neurosci ; 36(31): 8132-48, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27488634

RESUMEN

UNLABELLED: The aim of this study was to explore the signaling and neuroprotective effect of transactivator of transcription (TAT) protein transduction of the apoptosis repressor with CARD (ARC) in in vitro and in vivo models of cerebral ischemia in mice. In mice, transient focal cerebral ischemia reduced endogenous ARC protein in neurons in the ischemic striatum at early reperfusion time points, and in primary neuronal cultures, RNA interference resulted in greater neuronal susceptibility to oxygen glucose deprivation (OGD). TAT.ARC protein delivery led to a dose-dependent better survival after OGD. Infarct sizes 72 h after 60 min middle cerebral artery occlusion (MCAo) were on average 30 ± 8% (mean ± SD; p = 0.005; T2-weighted MRI) smaller in TAT.ARC-treated mice (1 µg intraventricularly during MCAo) compared with controls. TAT.ARC-treated mice showed better performance in the pole test compared with TAT.ß-Gal-treated controls. Importantly, post-stroke treatment (3 h after MCAo) was still effective in affording reduced lesion volume by 20 ± 7% (mean ± SD; p < 0.05) and better functional outcome compared with controls. Delayed treatment in mice subjected to 30 min MCAo led to sustained neuroprotection and functional behavior benefits for at least 28 d. Functionally, TAT.ARC treatment inhibited DAXX-ASK1-JNK signaling in the ischemic brain. ARC interacts with DAXX in a CARD-dependent manner to block DAXX trafficking and ASK1-JNK activation. Our work identifies for the first time ARC-DAXX binding to block ASK1-JNK activation as an ARC-specific endogenous mechanism that interferes with neuronal cell death and ischemic brain injury. Delayed delivery of TAT.ARC may present a promising target for stroke therapy. SIGNIFICANCE STATEMENT: Up to now, the only successful pharmacological target of human ischemic stroke is thrombolysis. Neuroprotective pharmacological strategies are needed to accompany therapies aiming to achieve reperfusion. We describe that apoptosis repressor with CARD (ARC) interacts and inhibits DAXX and proximal signals of cell death. In a murine stroke model mimicking human malignant infarction in the territory of the middle cerebral artery, TAT.ARC salvages brain tissue when given during occlusion or 3 h delayed with sustained functional benefits (28 d). This is a promising novel therapeutic approach because it appears to be effective in a model producing severe injury by interfering with an array of proximal signals and effectors of the ischemic cascade, upstream of JNK, caspases, and BIM and BAX activation.


Asunto(s)
Apoptosis , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Productos del Gen tat/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Animales , Proteínas Co-Represoras , Masculino , Ratones , Ratones Endogámicos C57BL , Chaperonas Moleculares , Unión Proteica , Mapas de Interacción de Proteínas
3.
J Cereb Blood Flow Metab ; 31(11): 2152-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21863037

RESUMEN

Small ubiquitin-like modifier (SUMO)2/3 but not SUMO1 conjugation is activated after transient cerebral ischemia. To investigate its function, we blocked neuronal SUMO2/3 translation through lentiviral microRNA delivery in primary cortical neurons. Viability was unaffected by SUMO2/3 silencing unless neurons were stressed by transient oxygen-glucose deprivation (OGD). Both 15 and 45 minutes of OGD were tolerated by control microRNA-expressing neurons but damaged >60% of neurons expressing SUMO2/3 microRNA. Damaging OGD (75 minutes) increased neuronal loss to 54% (control microRNA) and to 99% (SUMO2/3 microRNA). This suggests that activation of SUMO2/3 conjugation is an endogenous neuroprotective stress response.


Asunto(s)
Ataque Isquémico Transitorio/prevención & control , Neuronas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Técnicas de Cultivo de Célula , Hipoxia de la Célula , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Vectores Genéticos , Glucosa/metabolismo , Inmunohistoquímica , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/patología , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/administración & dosificación , MicroARNs/genética , MicroARNs/farmacología , Neuronas/patología , Oxígeno/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Sumoilación/efectos de los fármacos , Sumoilación/genética , Ubiquitinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA