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.
Carcinogenesis ; 38(11): 1092-1103, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-28968743

RESUMEN

The signalling adaptor p62 is frequently overexpressed in numerous cancer types. Here, we found that p62 expression was elevated in metastatic breast cancer and its overexpression correlated with reduced metastasis- and relapse-free survival times. Analysis of p62 expression in breast cancer cell lines demonstrated that high p62 expression was associated with the invasive phenotypes of breast cancer. Indeed, silencing p62 expression attenuated the invasive phenotypes of highly metastatic cells, whereas overexpressing p62 promoted the invasion of non-metastatic cells in in vitro microfluidic model. Moreover, MDA-MB-231 cells with p62 depletion which were grown in a three-dimensional culture system exhibited a loss of invasive protrusions. Consistently, genetic ablation of p62 suppressed breast cancer metastasis in both zebrafish embryo and immunodeficient mouse models, as well as decreased tumourigenicity in vivo. To explore the molecular mechanism by which p62 promotes breast cancer invasion, we performed a co-immunoprecipitation-mass spectrometry analysis and revealed that p62 interacted with vimentin, which mediated the function of p62 in promoting breast cancer invasion. Vimentin protein expression was downregulated upon p62 suppression and upregulated with p62 overexpression in breast cancer cells. Linear regression analysis of clinical breast cancer specimens showed a positive correlation between p62 and vimentin protein expression. Together, our findings provide strong evidence that p62 functions as a tumour metastasis promoter by binding vimentin and promoting its expression. This finding might help to develop novel molecular therapeutic strategies for breast cancer metastasis treatment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Metástasis de la Neoplasia/patología , Proteína Sequestosoma-1/genética , Vimentina/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Regulación hacia Abajo/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Células HEK293 , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/patología , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Regulación hacia Arriba/fisiología , Pez Cebra
2.
Clin Cancer Res ; 23(16): 4602-4616, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28424201

RESUMEN

Purpose: Mutations in KRAS are considered to be the main drivers of acquired resistance to epidermal growth factor receptor (EGFR) blockade in patients with metastatic colorectal cancer (mCRC). However, the potential role of other genes downstream of the EGFR signaling pathway in conferring acquired resistance has not been extensively investigated.Experimental Design: Using circulating tumor DNA (ctDNA) from patients with mCRC and with acquired cetuximab resistance, we developed a targeted amplicon ultra-deep sequencing method to screen for low-abundance somatic mutations in a panel of genes that encode components of the EGFR signaling pathway. Mutations with significantly increased variant frequencies upon disease progression were selected by using quartile analysis. The functional consequences of the identified mutations were validated in cultured cells.Results: We analyzed 32 patients with acquired cetuximab resistance in a development cohort. Of them, seven (22%) carried five novel PIK3CA mutations, whereas eight (25%) carried previously reported KRAS mutations. Functional studies showed that novel PIK3CA mutations (all in exon 19; p.K944N, p.F930S, p.V955G, p.V955I, and p.K966E) promote cell viability in the presence of cetuximab. Only one novel PIK3CA mutation (p.K944N) was verified in one of the 27 patients with acquired resistance in a validation cohort, simultaneous KRAS and PIK3CA hotspot mutations were detected in two patients. Among the above 59 acquired resistance patients, those with PIK3CA or RAS mutations detected in ctDNA showed a pronounced decrease in progression-free survival than patients with no mutation.Conclusions: The PIK3CA mutations may potentially contribute to acquired cetuximab resistance in patients with mCRC. Clin Cancer Res; 23(16); 4602-16. ©2017 AACR.


Asunto(s)
Cetuximab/uso terapéutico , Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Colorrectales/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Mutación , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Inmunológicos/uso terapéutico , ADN Tumoral Circulante/sangre , ADN Tumoral Circulante/química , ADN Tumoral Circulante/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Análisis Mutacional de ADN , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas p21(ras)/genética , Estudios Retrospectivos
3.
Oncol Rep ; 30(1): 520-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23670160

RESUMEN

Melanoma is a malignant tumor of the melanocytes. microRNAs (miRNAs) are emerging as important regulators of cancer-related processes. A thorough understanding of miRNAs in melanoma progression is important for developing new therapeutic targets. miRNA expression was detected by quantitative PCR. In vitro, MTT assay, colony formation assay, invasion assay and flow cytometry analysis were performed to test the effect of miR-573 on melanoma cells. The effect of miR-573 in vivo was validated using a murine xenograft model. Using quantitative PCR, we found that the expression levels of miR-573 were lower in melanoma tissues and cell lines compared to normal skin tissues. miR-573 upregulation inhibited melanoma cell proliferation and invasion, and overexpression of melanoma cell adhesion molecule (MCAM) could alleviate the effect of miR-573 on melanoma cells. In vivo, miR-573 overexpression groups showed lower rates of tumor growth compared with the control group. In conclusion, our results demonstrate that the elevated MCAM expression due to miR-573 reduction is essential in melanoma initiation and progression.


Asunto(s)
Antígeno CD146/metabolismo , Melanoma/genética , Melanoma/metabolismo , MicroARNs/metabolismo , Animales , Apoptosis , Antígeno CD146/biosíntesis , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , MicroARNs/genética , Invasividad Neoplásica/genética , Trasplante de Neoplasias , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...