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
1.
Methods Mol Biol ; 2129: 149-160, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32056176

RESUMEN

Orthotopic xenograft model recapitulates the faithful organ-specific microenvironment and facilitates analyses involving tumor-stromal interactions that are crucial for developing new-generation cancer therapy. Herein, we describe the detailed rationales and protocols of a versatile orthotopic xenograft model for esophageal squamous cell carcinoma.


Asunto(s)
Carcinoma de Células Escamosas de Esófago/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/clasificación , Esófago/patología , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Neoplasias de la Boca/patología , Trasplante Heterólogo , Microambiente Tumoral
2.
Proc Natl Acad Sci U S A ; 108(20): 8390-5, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21540330

RESUMEN

Chromosome 14 was transferred into tumorigenic nasopharyngeal carcinoma and esophageal carcinoma cell lines by a microcell-mediated chromosome transfer approach. Functional complementation of defects present in the cancer cells suppressed tumor formation. A candidate tumor-suppressor gene, cysteine-rich intestinal protein 2 (CRIP2), located in the hot spot for chromosomal loss at 14q32.3, was identified as an important candidate gene capable of functionally suppressing tumor formation. Previous studies have shown that CRIP2 is associated with development. To date, no report has provided functional evidence supporting a role for CRIP2 in tumor development. The present study provides unequivocal evidence that CRIP2 can functionally suppress tumorigenesis. CRIP2 is significantly down-regulated in nasopharyngeal carcinoma cell lines and tumors. CRIP2 reexpression functionally suppresses in vivo tumorigenesis and angiogenesis; these effects are induced by its transcription-repressor capability. It interacts with the NF-κB/p65 to inhibit its DNA-binding ability to the promoter regions of the major proangiogenesis cytokines critical for tumor progression, including IL6, IL8, and VEGF. In conclusion, we provide compelling evidence that CRIP2 acts as a transcription repressor of the NF-κB-mediated proangiogenic cytokine expression and thus functionally inhibits tumor formation and angiogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Transformación Celular Neoplásica/genética , Citocinas/genética , FN-kappa B/metabolismo , Neovascularización Patológica/genética , Transcripción Genética , Proteínas Supresoras de Tumor/genética , Proteínas Angiogénicas/análisis , Línea Celular , Línea Celular Tumoral , Cromosomas Humanos Par 14 , Citocinas/fisiología , Humanos , Proteínas con Dominio LIM , Proteínas Represoras/fisiología
3.
Mol Cancer Res ; 6(4): 592-603, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18403638

RESUMEN

Loss of chromosome 13q regions in esophageal squamous cell carcinoma (ESCC) is a frequent event. Monochromosome transfer approaches provide direct functional evidence for tumor suppression by chromosome 13 in SLMT-1, an ESCC cell line, and identify critical regions at 13q12.3, 13q14.11, and 13q14.3. Differential gene expression profiles of three tumor-suppressing microcell hybrids (MCH) and their tumorigenic parental SLMT-1 cell line were revealed by competitive hybridization using 19k cDNA oligonucleotide microarrays. Nine candidate 13q14 tumor-suppressor genes (TSG), including RB1, showed down-regulation in SLMT-1, compared with NE1, an immortalized normal esophageal epithelial cell line; their average gene expression was restored in MCHs compared with SLMT-1. Reverse transcription-PCR validated gene expression levels in MCHs and a panel of ESCC cell lines. Results suggest that the tumor-suppressing effect is not attributed to RB1, but instead likely involves thrombospondin type I domain-containing 1 (THSD1), a novel candidate TSG mapping to 13q14. Quantitative reverse transcription-PCR detected down-regulation of THSD1 expression in 100% of ESCC and other cancer cell lines. Mechanisms for THSD1 silencing in ESCC involved loss of heterozygosity and promoter hypermethylation, as analyzed by methylation-specific PCR and clonal bisulfite sequencing. Transfection of wild-type THSD1 into SLMT-1 resulted in significant reduction of colony-forming ability, hence providing functional evidence for its growth-suppressive activity. These findings suggest that THSD1 is a good candidate TSG.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 13/genética , Neoplasias Esofágicas/genética , Genes Supresores de Tumor , Análisis por Micromatrices , Trombospondinas/genética , Alelos , Línea Celular Transformada , Línea Celular Tumoral , Segregación Cromosómica , Metilación de ADN/efectos de los fármacos , Desoxicitidina/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma Humano/genética , Humanos , Ácidos Hidroxámicos/farmacología , Hibridación Fluorescente in Situ , Repeticiones de Microsatélite/genética , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombospondinas/metabolismo , Transfección , Ensayo de Tumor de Célula Madre
4.
Oncogene ; 24(4): 697-705, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15580306

RESUMEN

The key genes involved in the development of esophageal squamous cell carcinoma (ESCC) remain to be elucidated. Previous studies indicate extensive genomic alterations occur on chromosome 9 in ESCC. Using a monochromosome transfer approach, this study provides functional evidence and narrows down the critical region (CR) responsible for chromosome 9 tumor suppressing activity to a 2.4 Mb region mapping to 9q33-q34 between markers D9S1798 and D9S61. Interestingly, a high prevalence of allelic loss in this CR is also observed in primary ESCC tumors by microsatellite typing. Allelic loss is found in 30/34 (88%) tumors and the loss of heterozygosity (LOH) frequency ranges from 67 to 86%. Absent to low expression of a 9q32 candidate tumor suppressor gene (TSG), DEC1 (deleted in esophageal cancer 1), is detected in four Asian ESCC cell lines. Stably expressing DEC1 transfectants provide functional evidence for inhibition of tumor growth in nude mice and DEC1 expression is decreased in tumor segregants arising after long-term selection in vivo. There is 74% LOH in the DEC1 region of ESCC primary tumors. This study provides the first functional evidence for the presence of critical tumor suppressive regions on 9q33-q34. DEC1 is a candidate TSG that may be involved in ESCC development.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Cromosomas Humanos Par 9/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Pruebas de Carcinogenicidad , Carcinoma de Células Escamosas/patología , Deleción Cromosómica , Cromosomas Humanos Par 18/genética , ADN Complementario/genética , Neoplasias Esofágicas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Células Tumorales Cultivadas
5.
Genes Chromosomes Cancer ; 37(4): 359-68, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12800147

RESUMEN

In many cancers, including nasopharyngeal carcinoma (NPC), extensive and multiple regions of allelic loss occur on chromosome 14. However, to date no functionally conclusive tumor suppressor genes have yet been identified on this chromosome. Through use of the monochromosome transfer technique, this study provides functional evidence for the importance of two discrete regions of chromosome 14. A newly established A9 mouse donor cell line containing an intact copy of chromosome 14 was used for transfer of this intact chromosome into the NPC HONE1 cell line. Twelve independently established microcell hybrids demonstrated uniform loss of specific chromosome 14 loci from both endogenous and exogenous alleles. By microsatellite typing and fluorescence in situ hybridization with BAC probes, the two critical regions were localized to 14q11.2-13.1 and 14q32.1. Selective elimination of these regions during hybrid selection was strongly associated with both hybrid survival and tumor growth in vivo. This functional evidence now narrows down the candidate areas for further studies and suggests that at least two hitherto unidentified growth-related genes localized on two critical regions of chromosome arm 14q play an important role in tumorigenesis.


Asunto(s)
Carcinoma/genética , Carcinoma/patología , Cromosomas Humanos Par 14/genética , Técnicas de Transferencia de Gen , Genes Supresores de Tumor , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Animales , Línea Celular , Línea Celular Transformada , Deleción Cromosómica , Dosificación de Gen , Humanos , Células Híbridas , Hibridación Fluorescente in Situ , Ratones , Repeticiones de Microsatélite/genética , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA