Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
FEBS J ; 285(12): 2161-2181, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29582565

RESUMEN

Nuclear factor κB (NF-κB) transcription factors are the central hubs of signaling pathways connecting proinflammatory signals to cell survival, proliferation and cytokine production. In cancers, NF-κB signaling influences many aspects of tumor development, from initiation to metastasis. These functions are mediated by tumor-induced plasticity that allows tumor cells to adapt and survive in changing conditions within the tumor microenvironment. Tumor cell plasticity is shaped by the inflammatory microenvironment in tumors. This review focuses on inhibitor of NF-κB kinases, the direct upstream elements of NF-κB regulation, specifically on their conventional and non-conventional functions in animal models of tumorigenesis from the recent literature.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Quinasa I-kappa B/genética , FN-kappa B/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Microambiente Tumoral/genética , Animales , Carcinogénesis , Transición Epitelial-Mesenquimal/genética , Redes Reguladoras de Genes , Humanos , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Especificidad de Órganos , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Transducción de Señal
3.
Cell Rep ; 7(6): 1914-25, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24882009

RESUMEN

The recruitment of immune cells into solid tumors is an essential prerequisite of tumor development. Depending on the prevailing polarization profile of these infiltrating leucocytes, tumorigenesis is either promoted or blocked. Here, we identify IκB kinase α (IKKα) as a central regulator of a tumoricidal microenvironment during intestinal carcinogenesis. Mice deficient in IKKα kinase activity are largely protected from intestinal tumor development that is dependent on the enhanced recruitment of interferon γ (IFNγ)-expressing M1-like myeloid cells. In IKKα mutant mice, M1-like polarization is not controlled in a cell-autonomous manner but, rather, depends on the interplay of both IKKα mutant tumor epithelia and immune cells. Because therapies aiming at the tumor microenvironment rather than directly at the mutated cancer cell may circumvent resistance development, we suggest IKKα as a promising target for colorectal cancer (CRC) therapy.


Asunto(s)
Carcinogénesis/metabolismo , Quinasa I-kappa B/metabolismo , Intestinos/inmunología , Células Asesinas Naturales/patología , Células Mieloides/citología , Células Mieloides/enzimología , Animales , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/patología , Carcinogénesis/patología , Polaridad Celular , Transformación Celular Neoplásica , Células HEK293 , Humanos , Células Asesinas Naturales/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/patología , Fosforilación , Transducción de Señal
4.
Cell ; 152(1-2): 25-38, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23273993

RESUMEN

Cell-type plasticity within a tumor has recently been suggested to cause a bidirectional conversion between tumor-initiating stem cells and nonstem cells triggered by an inflammatory stroma. NF-κB represents a key transcription factor within the inflammatory tumor microenvironment. However, NF-κB's function in tumor-initiating cells has not been examined yet. Using a genetic model of intestinal epithelial cell (IEC)-restricted constitutive Wnt-activation, which comprises the most common event in the initiation of colon cancer, we demonstrate that NF-κB modulates Wnt signaling and show that IEC-specific ablation of RelA/p65 retards crypt stem cell expansion. In contrast, elevated NF-κB signaling enhances Wnt activation and induces dedifferentiation of nonstem cells that acquire tumor-initiating capacity. Thus, our data support the concept of bidirectional conversion and highlight the importance of inflammatory signaling for dedifferentiation and generation of tumor-initiating cells in vivo.


Asunto(s)
Desdiferenciación Celular , Transformación Celular Neoplásica , Neoplasias del Colon/patología , Células Madre Neoplásicas/patología , Animales , Colon/patología , Células Epiteliales/patología , Femenino , Humanos , Masculino , Ratones , FN-kappa B/metabolismo , Vía de Señalización Wnt
5.
Cell ; 130(5): 918-31, 2007 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-17803913

RESUMEN

IKKbeta-dependent NF-kappaB activation plays a key role in innate immunity and inflammation, and inhibition of IKKbeta has been considered as a likely anti-inflammatory therapy. Surprisingly, however, mice with a targeted IKKbeta deletion in myeloid cells are more susceptible to endotoxin-induced shock than control mice. Increased endotoxin susceptibility is associated with elevated plasma IL-1beta as a result of increased pro-IL-1beta processing, which was also seen upon bacterial infection. In macrophages enhanced pro-IL-1beta processing depends on caspase-1, whose activation is inhibited by NF-kappaB-dependent gene products. In neutrophils, however, IL-1beta secretion is caspase-1 independent and depends on serine proteases, whose activity is also inhibited by NF-kappaB gene products. Prolonged pharmacologic inhibition of IKKbeta also augments IL-1beta secretion upon endotoxin challenge. These results unravel an unanticipated role for IKKbeta-dependent NF-kappaB signaling in the negative control of IL-1beta production and highlight potential complications of long-term IKKbeta inhibition.


Asunto(s)
Quinasa I-kappa B/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , FN-kappa B/metabolismo , Infiltración Neutrófila , Neutrófilos/metabolismo , Choque Séptico/metabolismo , Animales , Apoptosis , Carbolinas/farmacología , Caspasa 1/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/genética , Interleucina-1beta/sangre , Interleucina-1beta/genética , Interleucina-6/sangre , Lipopolisacáridos , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Neutrófilos/inmunología , Neutrófilos/patología , Niacinamida/análogos & derivados , Niacinamida/farmacología , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Receptores de Interleucina-1/metabolismo , Serina Endopeptidasas/metabolismo , Serpinas/metabolismo , Choque Séptico/inducido químicamente , Choque Séptico/enzimología , Choque Séptico/inmunología , Choque Séptico/patología , Factores de Tiempo , Transfección , Factor de Necrosis Tumoral alfa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...