Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 6: 35854, 2016 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-27775025

RESUMEN

Strategies to identify tumors at highest risk for treatment failure are currently under investigation for patients with bladder cancer. We demonstrate that flow cytometric detection of poorly differentiated basal tumor cells (BTCs), as defined by the co-expression of CD90, CD44 and CD49f, directly from patients with early stage tumors (T1-T2 and N0) and patient-derived xenograft (PDX) engraftment in locally advanced tumors (T3-T4 or N+) predict poor prognosis in patients with bladder cancer. Comparative transcriptomic analysis of bladder tumor cells isolated from PDXs indicates unique patterns of gene expression during bladder tumor cell differentiation. We found cell division cycle 25C (CDC25C) overexpression in poorly differentiated BTCs and determined that CDC25C expression predicts adverse survival independent of standard clinical and pathologic features in bladder cancer patients. Taken together, our findings support the utility of BTCs and bladder cancer PDX models in the discovery of novel molecular targets and predictive biomarkers for personalizing oncology care for patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Anciano , Animales , Biomarcadores de Tumor/genética , Diferenciación Celular/genética , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones SCID , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/cirugía , Fosfatasas cdc25/genética
2.
Oncogene ; 35(50): 6439-6445, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27212035

RESUMEN

Aberrant expression of the DNA methyltransferases (DNMTs) and disruption of DNA methylation patterns are associated with carcinogenesis and cancer cell survival. The oncogenic MUC1-C protein is aberrantly overexpressed in diverse carcinomas; however, there is no known link between MUC1-C and DNA methylation. Our results demonstrate that MUC1-C induces the expression of DNMT1 and DNMT3b, but not DNMT3a, in breast and other carcinoma cell types. We show that MUC1-C occupies the DNMT1 and DNMT3b promoters in complexes with NF-κB p65 and drives DNMT1 and DNMT3b transcription. In this way, MUC1-C controls global DNA methylation as determined by analysis of LINE-1 repeat elements. The results further demonstrate that targeting MUC1-C downregulates DNA methylation of the CDH1 tumor suppressor gene in association with induction of E-cadherin expression. These findings provide compelling evidence that MUC1-C is of functional importance to induction of DNMT1 and DNMT3b and, in turn, changes in DNA methylation patterns in cancer cells.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/fisiología , Metilación de ADN , Mucina-1/fisiología , Neoplasias/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Transición Epitelial-Mesenquimal , Humanos , Células MCF-7 , Factor de Transcripción ReIA/fisiología , ADN Metiltransferasa 3B
3.
Br J Cancer ; 113(2): 327-35, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26042934

RESUMEN

BACKGROUND: The CXCL10/CXCR3 signalling mediates paracrine interactions between tumour and stromal cells that govern leukocyte trafficking and angiogenesis. Emerging data implicate noncanonical CXCL10/CXCR3 signalling in tumourigenesis and metastasis. However, little is known regarding the role for autocrine CXCL10/CXCR3 signalling in regulating the metastatic potential of individual tumour clones. METHODS: We performed transcriptomic and cytokine profiling to characterise the functions of CXCL10 and CXCR3 in tumour cells with different metastatic abilities. We modulated the expression of the CXCL10/CXCR3 pathway using shRNA-mediated silencing in both in vitro and in vivo models of B16F1 melanoma. In addition, we examined the expression of CXCL10 and CXCR3 and their associations with clinical outcomes in clinical data sets derived from over 670 patients with melanoma and colon and renal cell carcinomas. RESULTS: We identified a critical role for autocrine CXCL10/CXCR3 signalling in promoting tumour cell growth, motility and metastasis. Analysis of publicly available clinical data sets demonstrated that coexpression of CXCL10 and CXCR3 predicted an increased metastatic potential and was associated with early metastatic disease progression and poor overall survival. CONCLUSION: These findings support the potential for CXCL10/CXCR3 coexpression as a predictor of metastatic recurrence and point towards a role for targeting of this oncogenic axis in the treatment of metastatic disease.


Asunto(s)
Quimiocina CXCL10/fisiología , Transducción de Señal/fisiología , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Receptores CXCR3/fisiología
4.
Oncogene ; 34(40): 5187-97, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25659581

RESUMEN

The mucin 1 (MUC1) oncoprotein has been linked to the inflammatory response by promoting cytokine-mediated activation of the NF-κB pathway. The TGF-ß-activated kinase 1 (TAK1) is an essential effector of proinflammatory NF-κB signaling that also regulates cancer cell survival. The present studies demonstrate that the MUC1-C transmembrane subunit induces TAK1 expression in colon cancer cells. MUC1 also induces TAK1 in a MUC1(+/-)/IL-10(-/-) mouse model of colitis and colon tumorigenesis. We show that MUC1-C promotes NF-κB-mediated activation of TAK1 transcription and, in a positive regulatory loop, MUC1-C contributes to TAK1-induced NF-κB signaling. In this way, MUC1-C binds directly to TAK1 and confers the association of TAK1 with TRAF6, which is necessary for TAK1-mediated activation of NF-κB. Targeting MUC1-C thus suppresses the TAK1NF-κB pathway, downregulates BCL-XL and in turn sensitizes colon cancer cells to MEK inhibition. Analysis of colon cancer databases further indicates that MUC1, TAK1 and TRAF6 are upregulated in tumors associated with decreased survival and that MUC1-C-induced gene expression patterns predict poor outcomes in patients. These results support a model in which MUC1-C-induced TAK1NF-κB signaling contributes to intestinal inflammation and colon cancer progression.


Asunto(s)
Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Quinasas Quinasa Quinasa PAM/metabolismo , Mucina-1/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Neoplasias del Colon/metabolismo , Neoplasias del Colon/mortalidad , Progresión de la Enfermedad , Humanos , Immunoblotting , Inmunoprecipitación , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa , Modelos de Riesgos Proporcionales
5.
Oncogene ; 29(6): 920-9, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19915608

RESUMEN

Signal transducer and activator of transcription 1 (STAT1) is activated in the inflammatory response to interferons. The MUC1 oncoprotein is overexpressed in human breast cancers. Analysis of genes differentially expressed in MUC1-transformed cells has identified a network linking MUC1 and STAT1 that is associated with cellular growth and inflammation. The results further show that the MUC1-C subunit associates with STAT1 in cells and the MUC1-C cytoplasmic domain binds directly to the STAT1 DNA-binding domain. The interaction between MUC1-C and STAT1 is inducible by IFNgamma in non-malignant epithelial cells and constitutive in breast cancer cells. Moreover, the MUC1-STAT1 interaction contributes to the activation of STAT1 target genes, including MUC1 itself. Analysis of two independent databases showed that MUC1 and STAT1 are coexpressed in about 15% of primary human breast tumors. Coexpression of MUC1 and the STAT1 pathway was found to be significantly associated with decreased recurrence-free and overall survival. These findings indicate that (i) MUC1 and STAT1 function in an auto-inductive loop, and (ii) activation of both MUC1 and the STAT1 pathway in breast tumors confers a poor prognosis for patients.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/metabolismo , Mucina-1/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Citoplasma/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Interferón gamma/farmacología , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Ratones , Datos de Secuencia Molecular , Mucina-1/química , Mucina-1/genética , Pronóstico , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína , Ratas , Factor de Transcripción STAT1/genética , Transducción de Señal/efectos de los fármacos
6.
Cancer Gene Ther ; 16(4): 373-81, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18974777

RESUMEN

TNFerade is a radioinducible adenoviral vector expressing tumor necrosis factor-alpha (TNF-alpha) (Ad.Egr-TNF) currently in a phase III trial for inoperable pancreatic cancer. We studied B16-F1 melanoma tumors in TNF receptor wild-type (C57BL/6) and deficient (TNFR1,2-/- and TNFR1-/-) mice. Ad.Egr-TNF+IR inhibited tumor growth compared with IR in C57BL/6 but not in receptor-deficient mice. Tumors resistant to TNF-alpha were also sensitive to Ad.Egr-TNF+IR in C57BL/6 mice. Ad.Egr-TNF+IR produced an increase in tumor-associated endothelial cell apoptosis not observed in receptor-deficient animals. Also, B16-F1 tumors in mice with germline deletions of TNFR1,2, TNFR1 or TNF-alpha, or in mice receiving anti-TNF-alpha exhibited radiosensitivity. These results show that tumor-associated endothelium is the principal target for Ad.Egr-TNF radiosensitization and implicate TNF-alpha signaling in tumor radiosensitivity.


Asunto(s)
Terapia Genética/métodos , Melanoma Experimental/terapia , Fármacos Sensibilizantes a Radiaciones , Factor de Necrosis Tumoral alfa/metabolismo , Terapia por Rayos X , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Etanercept , Humanos , Inmunoglobulina G/farmacología , Inmunosupresores/farmacología , Ratones , Trasplante de Neoplasias , Receptores del Factor de Necrosis Tumoral , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA