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.
Cell Death Dis ; 10(10): 756, 2019 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-31591393

RESUMEN

Since the publication of this article the authors have noted that there were two typos that could have caused confusion to the readers:1) References to "integrin alpha chain V" should have been "integrin alpha 5". These appear in the Abstract and the first paragraph of the Results section.2) In the Results section under the header "Integrins activate Fyn tyrosine kinases" and the legend of Figure 6a, "tyr530" should be replaced with "tyr416".3) In the the legend of Figure 6b, "pSrc" with should be replaced with "pFyn".

2.
Cell Death Dis ; 8(4): e2737, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28383553

RESUMEN

Oxidative modification of the voltage-gated potassium (K+) channel KCNB1 promotes apoptosis in the neurons of cortex and hippocampus through a signaling pathway mediated by Src tyrosine kinases. How oxidation of the channel is transduced into Src recruitment and activation, however, was not known. Here we show that the apoptotic signal originates from integrins, which form macromolecular complexes with KCNB1 channels. The initial stimulus is transduced to Fyn and possibly other Src family members by focal adhesion kinase (FAK). Thus KCNB1 and integrin alpha chain V (integrin-α5) coimmunoprecipitated in the mouse brain and these interactions were retained upon channel's oxidation. Pharmacological inhibition of integrin signaling or FAK suppressed apoptosis induced by oxidation of KCNB1, as well as FAK and Src/Fyn activation. Most importantly, the activation of the integrin-FAK-Src/Fyn cascade was negligible in the presence of non-oxidizable C73A KCNB1 mutant channels, even though they normally interacted with integrin-α5. This leads us to conclude that the transition between the non-oxidized and oxidized state of KCNB1 activates integrin signaling. KCNB1 oxidation may favor integrin clustering, thereby facilitating the recruitment and activation of FAK and Src/Fyn kinases.


Asunto(s)
Apoptosis/fisiología , Encéfalo/metabolismo , Integrina alfa5/metabolismo , Canales de Potasio Shab/metabolismo , Transducción de Señal/fisiología , Sustitución de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Integrina alfa5/genética , Ratones , Mutación Missense , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Canales de Potasio Shab/genética , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
3.
J Neurosci ; 36(43): 11084-11096, 2016 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-27798188

RESUMEN

The delayed rectifier potassium (K+) channel KCNB1 (Kv2.1), which conducts a major somatodendritic current in cortex and hippocampus, is known to undergo oxidation in the brain, but whether this can cause neurodegeneration and cognitive impairment is not known. Here, we used transgenic mice harboring human KCNB1 wild-type (Tg-WT) or a nonoxidable C73A mutant (Tg-C73A) in cortex and hippocampus to determine whether oxidized KCNB1 channels affect brain function. Animals were subjected to moderate traumatic brain injury (TBI), a condition characterized by extensive oxidative stress. Dasatinib, a Food and Drug Administration-approved inhibitor of Src tyrosine kinases, was used to impinge on the proapoptotic signaling pathway activated by oxidized KCNB1 channels. Thus, typical lesions of brain injury, namely, inflammation (astrocytosis), neurodegeneration, and cell death, were markedly reduced in Tg-C73A and dasatinib-treated non-Tg animals. Accordingly, Tg-C73A mice and non-Tg mice treated with dasatinib exhibited improved behavioral outcomes in motor (rotarod) and cognitive (Morris water maze) assays compared to controls. Moreover, the activity of Src kinases, along with oxidative stress, were significantly diminished in Tg-C73A brains. Together, these data demonstrate that oxidation of KCNB1 channels is a contributing mechanism to cellular and behavioral deficits in vertebrates and suggest a new therapeutic approach to TBI. SIGNIFICANCE STATEMENT: This study provides the first experimental evidence that oxidation of a K+ channel constitutes a mechanism of neuronal and cognitive impairment in vertebrates. Specifically, the interaction of KCNB1 channels with reactive oxygen species plays a major role in the etiology of mouse model of traumatic brain injury (TBI), a condition associated with extensive oxidative stress. In addition, a Food and Drug Administration-approved drug ameliorates the outcome of TBI in mouse, by directly impinging on the toxic pathway activated in response to oxidation of the KCNB1 channel. These findings elucidate a basic mechanism of neurotoxicity in vertebrates and might lead to a new therapeutic approach to TBI in humans, which, despite significant efforts, is a condition that remains without effective pharmacological treatments.


Asunto(s)
Lesiones Traumáticas del Encéfalo/fisiopatología , Trastornos del Conocimiento/fisiopatología , Hipocampo/fisiopatología , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canales de Potasio Shab/metabolismo , Animales , Apoptosis , Lesiones Traumáticas del Encéfalo/patología , Trastornos del Conocimiento/patología , Dasatinib/administración & dosificación , Hipocampo/patología , Masculino , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas , Neuronas/patología , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA