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











Base de datos
Intervalo de año de publicación
1.
Oncogene ; 32(36): 4214-21, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23045272

RESUMEN

Viral and pharmacological inducers of protein kinase RNA-activated (PKR)-like ER kinase (PERK) were shown to accelerate the phosphorylation-dependent degradation of the IFNAR1 chain of the Type 1 interferon (IFN) receptor and to limit cell sensitivity to IFN. Here we report that hypoxia can elicit these effects in a PERK-dependent manner. The altered fate of IFNAR1 affected by signaling downstream of PERK depends on phosphorylation of eIF2α (eukaryotic translational initiation factor 2-α) and ensuing activation of p38α kinase. Activators of other eIF2α kinases such as PKR or GCN2 (general control nonrepressed-2) are also capable of eliminating IFNAR1 and blunting IFN responses. Modulation of constitutive PKR activity in human breast cancer cells stabilizes IFNAR1 and sensitizes these cells to IFNAR1-dependent anti-tumorigenic effects. Although downregulation of IFNAR1 and impaired IFNAR1 signaling can be elicited in response to amino-acid deficit, the knockdown of GCN2 in melanoma cells reverses these phenotypes. We propose that, in cancer cells and the tumor microenvironment, activation of diverse eIF2α kinases followed by IFNAR1 downregulation enables multiple cellular components of tumor tissue to evade the direct and indirect anti-tumorigenic effects of Type 1 IFN.


Asunto(s)
Interferón Tipo I/metabolismo , Estrés Fisiológico , Animales , Línea Celular , Cricetinae , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Ratones , Neoplasias/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , eIF-2 Quinasa/metabolismo
2.
Oncogene ; 31(2): 161-72, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-21666722

RESUMEN

Interferon alpha (IFNα) is widely used for treatment of melanoma and certain other malignancies. This cytokine as well as the related IFNß exerts potent anti-tumorigenic effects; however, their efficacy in patients is often suboptimal. Here, we report that inflammatory signaling impedes the effects of IFNα/ß. Melanoma cells can secrete pro-inflammatory cytokines that inhibit cellular responses to IFNα/ß via activating the ligand-independent pathway for the phosphorylation and subsequent ubiquitination and accelerated degradation of the IFNAR1 chain of type I IFN receptor. Catalytic activity of the p38 protein kinase was required for IFNAR1 downregulation and inhibition of IFNα/ß signaling induced by proinflammatory cytokines such as interleukin 1 (IL-1). Activation of p38 kinase inversely correlated with protein levels of IFNAR1 in clinical melanoma specimens. Inhibition of p38 kinase augmented the inhibitory effects of IFNα/ß on cell viability and growth in vitro and in vivo. The roles of inflammation and p38 protein kinase in regulating cellular responses to IFNα/ß in normal and tumor cells are discussed.


Asunto(s)
Inflamación/fisiopatología , Interferón-alfa/fisiología , Transducción de Señal , Animales , Línea Celular Tumoral , Citocinas/fisiología , Humanos , Mediadores de Inflamación/fisiología , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones SCID , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA