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




Base de datos
Asunto de la revista
Intervalo de año de publicación
1.
PLoS Med ; 7(7): e1000315, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20668658

RESUMEN

BACKGROUND: Traditionally, non-small cell lung cancer is treated as a single disease entity in terms of systemic therapy. Emerging evidence suggests the major subtypes--adenocarcinoma (AC) and squamous cell carcinoma (SqCC)--respond differently to therapy. Identification of the molecular differences between these tumor types will have a significant impact in designing novel therapies that can improve the treatment outcome. METHODS AND FINDINGS: We used an integrative genomics approach, combing high-resolution comparative genomic hybridization and gene expression microarray profiles, to compare AC and SqCC tumors in order to uncover alterations at the DNA level, with corresponding gene transcription changes, which are selected for during development of lung cancer subtypes. Through the analysis of multiple independent cohorts of clinical tumor samples (>330), normal lung tissues and bronchial epithelial cells obtained by bronchial brushing in smokers without lung cancer, we identified the overexpression of BRF2, a gene on Chromosome 8p12, which is specific for development of SqCC of lung. Genetic activation of BRF2, which encodes a RNA polymerase III (Pol III) transcription initiation factor, was found to be associated with increased expression of small nuclear RNAs (snRNAs) that are involved in processes essential for cell growth, such as RNA splicing. Ectopic expression of BRF2 in human bronchial epithelial cells induced a transformed phenotype and demonstrates downstream oncogenic effects, whereas RNA interference (RNAi)-mediated knockdown suppressed growth and colony formation of SqCC cells overexpressing BRF2, but not AC cells. Frequent activation of BRF2 in >35% preinvasive bronchial carcinoma in situ, as well as in dysplastic lesions, provides evidence that BRF2 expression is an early event in cancer development of this cell lineage. CONCLUSIONS: This is the first study, to our knowledge, to show that the focal amplification of a gene in Chromosome 8p12, plays a key role in squamous cell lineage specificity of the disease. Our data suggest that genetic activation of BRF2 represents a unique mechanism of SqCC lung tumorigenesis through the increase of Pol III-mediated transcription. It can serve as a marker for lung SqCC and may provide a novel target for therapy. Please see later in the article for the Editors' Summary.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias Pulmonares/genética , Factor de Transcripción TFIIIB/genética , Factor de Transcripción TFIIIB/fisiología , Adenocarcinoma/genética , Adenocarcinoma/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/fisiología , Carcinoma de Células Escamosas/patología , Linaje de la Célula/genética , Hibridación Genómica Comparativa , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genómica/métodos , Humanos , Neoplasias Pulmonares/patología , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Oncogenes/fisiología , Especificidad de Órganos/genética , Integración de Sistemas , Células Tumorales Cultivadas
2.
BMC Genomics ; 8: 32, 2007 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-17257419

RESUMEN

BACKGROUND: Prostate cancer is the most frequently diagnosed cancer in American men, and few effective treatment options are available to patients who develop hormone-refractory prostate cancer. The molecular changes that occur to allow prostate cells to proliferate in the absence of androgens are not fully understood. RESULTS: Subtractive hybridization experiments performed with samples from an in vivo model of hormonal progression identified 25 expressed sequences representing novel human transcripts. Intriguingly, these 25 sequences have small open-reading frames and are not highly conserved through evolution, suggesting many of these novel expressed sequences may be derived from untranslated regions of novel transcripts or from non-coding transcripts. Examination of a large metalibrary of human Serial Analysis of Gene Expression (SAGE) tags demonstrated that only three of these novel sequences had been previously detected. RT-PCR experiments confirmed that the 6 sequences tested were expressed in specific human tissues, as well as in clinical samples of prostate cancer. Further RT-PCR experiments for five of these fragments indicated they originated from large untranslated regions of unannotated transcripts. CONCLUSION: This study underlines the value of using complementary techniques in the annotation of the human genome. The tissue-specific expression of 4 of the 6 clones tested indicates the expression of these novel transcripts is tightly regulated, and future work will determine the possible role(s) these novel transcripts may play in the progression of prostate cancer.


Asunto(s)
Resistencia a Antineoplásicos/genética , Genes Relacionados con las Neoplasias , Neoplasias de la Próstata/metabolismo , Andrógenos/farmacología , Animales , Regulación Neoplásica de la Expresión Génica , Biblioteca de Genes , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Neoplasias de la Próstata/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN , Células Tumorales Cultivadas , Regiones no Traducidas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA