Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
1.
PLoS Genet ; 9(4): e1003464, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23637631

RESUMEN

Gene fusions, like BCR/ABL1 in chronic myelogenous leukemia, have long been recognized in hematologic and mesenchymal malignancies. The recent finding of gene fusions in prostate and lung cancers has motivated the search for pathogenic gene fusions in other malignancies. Here, we developed a "breakpoint analysis" pipeline to discover candidate gene fusions by tell-tale transcript level or genomic DNA copy number transitions occurring within genes. Mining data from 974 diverse cancer samples, we identified 198 candidate fusions involving annotated cancer genes. From these, we validated and further characterized novel gene fusions involving ROS1 tyrosine kinase in angiosarcoma (CEP85L/ROS1), SLC1A2 glutamate transporter in colon cancer (APIP/SLC1A2), RAF1 kinase in pancreatic cancer (ATG7/RAF1) and anaplastic astrocytoma (BCL6/RAF1), EWSR1 in melanoma (EWSR1/CREM), CDK6 kinase in T-cell acute lymphoblastic leukemia (FAM133B/CDK6), and CLTC in breast cancer (CLTC/VMP1). Notably, while these fusions involved known cancer genes, all occurred with novel fusion partners and in previously unreported cancer types. Moreover, several constituted druggable targets (including kinases), with therapeutic implications for their respective malignancies. Lastly, breakpoint analysis identified new cell line models for known rearrangements, including EGFRvIII and FIP1L1/PDGFRA. Taken together, we provide a robust approach for gene fusion discovery, and our results highlight a more widespread role of fusion genes in cancer pathogenesis.


Asunto(s)
Fusión Génica , Proteínas Tirosina Quinasas , Genómica , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética
2.
Proc Natl Acad Sci U S A ; 109(46): E3196-205, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23112155

RESUMEN

The mutational activation of oncogenes drives cancer development and progression. Classic oncogenes, such as MYC and RAS, are active across many different cancer types. In contrast, "lineage-survival" oncogenes represent a distinct and emerging class typically comprising transcriptional regulators of a specific cell lineage that, when deregulated, support the proliferation and survival of cancers derived from that lineage. Here, in a large collection of colorectal cancer cell lines and tumors, we identify recurrent amplification of chromosome 13, an alteration highly restricted to colorectal-derived cancers. A minimal region of amplification on 13q12.2 pinpoints caudal type homeobox transcription factor 2 (CDX2), a regulator of normal intestinal lineage development and differentiation, as a target of the amplification. In contrast to its described role as a colorectal tumor suppressor, CDX2 when amplified is required for the proliferation and survival of colorectal cancer cells. Further, transcriptional profiling, binding-site analysis, and functional studies link CDX2 to Wnt/ß-catenin signaling, itself a key oncogenic pathway in colorectal cancer. These data characterize CDX2 as a lineage-survival oncogene deregulated in colorectal cancer. Our findings challenge a prevailing view that CDX2 is a tumor suppressor in colorectal cancer and uncover an additional piece in the multistep model of colorectal tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/metabolismo , Amplificación de Genes , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Factor de Transcripción CDX2 , Línea Celular Tumoral , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Cromosomas Humanos Par 13/genética , Cromosomas Humanos Par 13/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Humanos , Ratones , Células 3T3 NIH , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Vía de Señalización Wnt/genética
3.
PLoS One ; 8(9): e74562, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040285

RESUMEN

Clear cell carcinoma (CCC) is a histologically distinct carcinoma subtype that arises in several organ systems and is marked by cytoplasmic clearing, attributed to abundant intracellular glycogen. Previously, transcription factor hepatocyte nuclear factor 1-beta (HNF1B) was identified as a biomarker of ovarian CCC. Here, we set out to explore more broadly the relation between HNF1B and carcinomas with clear cell histology. HNF1B expression, evaluated by immunohistochemistry, was significantly associated with clear cell histology across diverse gynecologic and renal carcinomas (P<0.001), as was hypomethylation of the HNF1B promoter (P<0.001). From microarray analysis, an empirically-derived HNF1B signature was significantly enriched for computationally-predicted targets (with HNF1 binding sites) (P<0.03), as well as genes associated with glycogen metabolism, including glucose-6-phophatase, and strikingly the blood clotting cascade, including fibrinogen, prothrombin and factor XIII. Enrichment of the clotting cascade was also evident in microarray data from ovarian CCC versus other histotypes (P<0.01), and HNF1B-associated prothrombin expression was verified by immunohistochemistry (P = 0.015). Finally, among gynecologic carcinomas with cytoplasmic clearing, HNF1B immunostaining was linked to a 3.0-fold increased risk of clinically-significant venous thrombosis (P = 0.043), and with a 2.3-fold increased risk (P = 0.011) in a combined gynecologic and renal carcinoma cohort. Our results define HNF1B as a broad marker of clear cell phenotype, and support a mechanistic link to glycogen accumulation and thrombosis, possibly reflecting (for gynecologic CCC) derivation from secretory endometrium. Our findings also implicate a novel mechanism of tumor-associated thrombosis (a major cause of cancer mortality), based on the direct production of clotting factors by cancer cells.


Asunto(s)
Adenocarcinoma de Células Claras/genética , Carcinoma de Células Renales/genética , Neoplasias Endometriales/genética , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/genética , Neoplasias Renales/genética , Neoplasias Ováricas/genética , Trombosis de la Vena/genética , Adenocarcinoma de Células Claras/complicaciones , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Coagulación Sanguínea , Factores de Coagulación Sanguínea/genética , Factores de Coagulación Sanguínea/metabolismo , Carcinoma de Células Renales/complicaciones , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Biología Computacional , Metilación de ADN , Neoplasias Endometriales/complicaciones , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfatasa/metabolismo , Glucógeno/metabolismo , Factor Nuclear 1-beta del Hepatocito/metabolismo , Humanos , Inmunohistoquímica , Neoplasias Renales/complicaciones , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Riesgo , Trombosis de la Vena/complicaciones , Trombosis de la Vena/metabolismo , Trombosis de la Vena/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA