Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Cancer ; 10: 85, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21777459

RESUMEN

BACKGROUND: The presence of cancer-specific DNA methylation patterns in epithelial colorectal cells in human feces provides the prospect of a simple, non-invasive screening test for colorectal cancer and its precursor, the adenoma. This study investigates a panel of epigenetic markers for the detection of colorectal cancer and adenomas. METHODS: Candidate biomarkers were subjected to quantitative methylation analysis in test sets of tissue samples from colorectal cancers, adenomas, and normal colonic mucosa. All findings were verified in independent clinical validation series. A total of 523 human samples were included in the study. Receiver operating characteristic (ROC) curve analysis was used to evaluate the performance of the biomarker panel. RESULTS: Promoter hypermethylation of the genes CNRIP1, FBN1, INA, MAL, SNCA, and SPG20 was frequent in both colorectal cancers (65-94%) and adenomas (35-91%), whereas normal mucosa samples were rarely (0-5%) methylated. The combined sensitivity of at least two positives among the six markers was 94% for colorectal cancers and 93% for adenoma samples, with a specificity of 98%. The resulting areas under the ROC curve were 0.984 for cancers and 0.968 for adenomas versus normal mucosa. CONCLUSIONS: The novel epigenetic marker panel shows very high sensitivity and specificity for both colorectal cancers and adenomas. Our findings suggest this biomarker panel to be highly suitable for early tumor detection.


Asunto(s)
Adenoma/diagnóstico , Adenoma/genética , Biomarcadores de Tumor/aislamiento & purificación , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Epigénesis Genética/genética , Adenoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Carcinoma/diagnóstico , Carcinoma/genética , Carcinoma/patología , Neoplasias Colorrectales/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Mucosa Intestinal/química , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad
2.
Hum Mutat ; 29(11): E252-62, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18781614

RESUMEN

PTEN regulates cell homeostasis by inhibiting growth signals transduced through PI3-kinases. The gene is mutated in several cancer types, but so far, only a limited number of mutations have been reported in colorectal cancer. In the present study, direct sequencing was used to analyze the whole coding region and exon-intron boundaries of PTEN in a series of microsatellite stable (n=34) and microsatellite unstable (n=30) colorectal carcinomas with known TP53 mutation status. We detected 21 PTEN mutations in altogether 13 tumors (20%), including 19 mutations in the coding sequence and two in the exon-intron boundaries. Sixteen of these alterations have not been previously reported in colorectal cancer. Furthermore, seven out of the 13 altered tumors harbored more than one mutation, potentially leading to loss of gene function. Finally, all PTEN mutations found were in tumors harboring wild-type TP53. In conclusion, PTEN is mutated in a significant subgroup of colorectal carcinomas, and our findings further extend the previously small spectrum of reported PTEN changes. Additionally, it seems that mutations in PTEN and TP53 are mutually exclusive for this cancer type.


Asunto(s)
Carcinoma/genética , Neoplasias Colorrectales/genética , Inestabilidad de Microsatélites , Mutación , Fosfohidrolasa PTEN/genética , Adulto , Anciano , Anciano de 80 o más Años , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Exones , Genes p53 , Humanos , Intrones , Persona de Mediana Edad
3.
Neoplasia ; 10(7): 680-6, 2 p following 686, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18592002

RESUMEN

More than half of all colorectal carcinomas are known to exhibit an activated mitogen-activated protein kinase pathway. The NF1 gene, a negative regulator of KRAS, has not previously been examined in a series of colorectal cancer. In the present study, primary colorectal carcinomas stratified according to microsatellite instability status were analyzed. The whole coding region of NF1 was analyzed for mutations using denaturing high-performance liquid chromatography and sequencing, and the copy number alterations of NF1 were examined using multiple ligation-dependent probe amplification and real-time polymerase chain reaction. The mutational hot spots in KRAS and BRAF were sequenced, and promoter hypermethylation status of RASSF1A was assessed with a methylation-specific polymerase chain reaction. One sample had two missense mutations in NF1, whereas nine additional tumors had intronic mutations likely to affect exon splicing. Interestingly, 8 of these 10 tumors were microsatellite-unstable. Four other tumors showed a duplication of NF1. Mutations in KRAS and BRAF were mutually exclusive and were present at 40% and 22%, respectively. RASSF1A was hypermethylated in 31% of the samples. We show that the RAS signaling network is extensively dysregulated in colorectal carcinomas, because more than 70% of the tumors had an alteration in one or more of the four examined components.


Asunto(s)
Carcinoma/genética , Neoplasias Colorrectales/genética , Genes de Neurofibromatosis 1 , Genes ras , Proteínas Supresoras de Tumor/genética , Quinasas raf/genética , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Análisis Mutacional de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Pruebas Genéticas , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Transducción de Señal/genética
4.
J Transl Med ; 6: 13, 2008 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-18346269

RESUMEN

BACKGROUND: Tumor-derived aberrantly methylated DNA might serve as diagnostic biomarkers for cancer, but so far, few such markers have been identified. The aim of the present study was to investigate the potential of the MAL (T-cell differentiation protein) gene as an early epigenetic diagnostic marker for colorectal tumors. METHODS: Using methylation-specific polymerase chain reaction (MSP) the promoter methylation status of MAL was analyzed in 218 samples, including normal mucosa (n = 44), colorectal adenomas (n = 63), carcinomas (n = 65), and various cancer cell lines (n = 46). Direct bisulphite sequencing was performed to confirm the MSP results. MAL gene expression was investigated with real time quantitative analyses before and after epigenetic drug treatment. Immunohistochemical analysis of MAL was done using normal colon mucosa samples (n = 5) and a tissue microarray with 292 colorectal tumors. RESULTS: Bisulphite sequencing revealed that the methylation was unequally distributed within the MAL promoter and by MSP analysis a region close to the transcription start point was shown to be hypermethylated in the majority of colorectal carcinomas (49/61, 80%) as well as in adenomas (45/63, 71%). In contrast, only a minority of the normal mucosa samples displayed hypermethylation (1/23, 4%). The hypermethylation of MAL was significantly associated with reduced or lost gene expression in in vitro models. Furthermore, removal of the methylation re-induced gene expression in colon cancer cell lines. Finally, MAL protein was expressed in epithelial cells of normal colon mucosa, but not in the malignant cells of the same type. CONCLUSION: Promoter hypermethylation of MAL was present in the vast majority of benign and malignant colorectal tumors, and only rarely in normal mucosa, which makes it suitable as a diagnostic marker for early colorectal tumorigenesis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Metilación de ADN , Proteínas de Transporte de Membrana/genética , Proteínas de la Mielina/genética , Proteolípidos/genética , Neoplasias Colorrectales/metabolismo , Humanos , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito , Regiones Promotoras Genéticas
5.
Mol Cancer ; 7: 94, 2008 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-19117505

RESUMEN

BACKGROUND: Multiple epigenetic and genetic changes have been reported in colorectal tumors, but few of these have clinical impact. This study aims to pinpoint epigenetic markers that can discriminate between non-malignant and malignant tissue from the large bowel, i.e. markers with diagnostic potential. The methylation status of eleven genes (ADAMTS1, CDKN2A, CRABP1, HOXA9, MAL, MGMT, MLH1, NR3C1, PTEN, RUNX3, and SCGB3A1) was determined in 154 tissue samples including normal mucosa, adenomas, and carcinomas of the colorectum. The gene-specific and widespread methylation status among the carcinomas was related to patient gender and age, and microsatellite instability status. Possible CIMP tumors were identified by comparing the methylation profile with microsatellite instability (MSI), BRAF-, KRAS-, and TP53 mutation status. RESULTS: The mean number of methylated genes per sample was 0.4 in normal colon mucosa from tumor-free individuals, 1.2 in mucosa from cancerous bowels, 2.2 in adenomas, and 3.9 in carcinomas. Widespread methylation was found in both adenomas and carcinomas. The promoters of ADAMTS1, MAL, and MGMT were frequently methylated in benign samples as well as in malignant tumors, independent of microsatellite instability. In contrast, normal mucosa samples taken from bowels without tumor were rarely methylated for the same genes. Hypermethylated CRABP1, MLH1, NR3C1, RUNX3, and SCGB3A1 were shown to be identifiers of carcinomas with microsatellite instability. In agreement with the CIMP concept, MSI and mutated BRAF were associated with samples harboring hypermethylation of several target genes. CONCLUSION: Methylated ADAMTS1, MGMT, and MAL are suitable as markers for early tumor detection.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Metilación de ADN , Detección Precoz del Cáncer , Genes Relacionados con las Neoplasias , Mucosa Intestinal/metabolismo , Adenoma/genética , Adulto , Anciano , Anciano de 80 o más Años , Análisis por Conglomerados , Neoplasias del Colon/diagnóstico , ADN de Neoplasias/metabolismo , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Inestabilidad de Microsatélites , Repeticiones de Microsatélite/genética , Persona de Mediana Edad , Regiones Promotoras Genéticas , Caracteres Sexuales
6.
Mol Cancer ; 6: 2, 2007 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-17201907

RESUMEN

BACKGROUND: Despite the fact that metastases are the leading cause of colorectal cancer deaths, little is known about the underlying molecular changes in these advanced disease stages. Few have studied the overall gene expression levels in metastases from colorectal carcinomas, and so far, none has investigated the peritoneal carcinomatoses by use of DNA microarrays. Therefore, the aim of the present study is to investigate and compare the gene expression patterns of primary carcinomas (n = 18), liver metastases (n = 4), and carcinomatoses (n = 4), relative to normal samples from the large bowel. RESULTS: Transcriptome profiles of colorectal cancer metastases independent of tumor site, as well as separate profiles associated with primary carcinomas, liver metastases, or peritoneal carcinomatoses, were assessed by use of Bayesian statistics. Gains of chromosome arm 5p are common in peritoneal carcinomatoses and several candidate genes (including PTGER4, SKP2, and ZNF622) mapping to this region were overexpressed in the tumors. Expression signatures stratified on TP53 mutation status were identified across all tumors regardless of stage. Furthermore, the gene expression levels for the in vivo tumors were compared with an in vitro model consisting of cell lines representing all three tumor stages established from one patient. CONCLUSION: By statistical analysis of gene expression data from primary colorectal carcinomas, liver metastases, and carcinomatoses, we are able to identify genetic patterns associated with the different stages of tumorigenesis.


Asunto(s)
Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Peritoneales/metabolismo , Línea Celular Tumoral , Análisis por Conglomerados , Perfilación de la Expresión Génica , Genes p53 , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Hepáticas/secundario , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Peritoneales/secundario , Análisis por Matrices de Proteínas
7.
Cell Oncol ; 28(5-6): 259-72, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17167179

RESUMEN

BACKGROUND: Gene silencing through CpG island hypermethylation is a major mechanism in cancer development. In the present study, we aimed to identify and validate novel target genes inactivated through promoter hypermethylation in colorectal tumor development. METHODS: With the use of microarrays, the gene expression profiles of colon cancer cell lines before and after treatment with the demethylating agent 5-aza-2'-deoxycytidine were identified and compared. The expression of the responding genes was compared with microarray expression data of primary colorectal carcinomas. Four of these down-regulated genes were subjected to methylation-specific PCR, bisulphite sequencing, and quantitative gene expression analysis using tumors (n=198), normal tissues (n=44), and cell lines (n=30). RESULTS: Twenty-one genes with a CpG island in their promoter responded to treatment in cell lines, and were simultaneously down-regulated in primary colorectal carcinomas. Among 20 colon cancer cell lines, hypermethylation was subsequently identified for three of four analyzed genes, ADAMTS1 (85%), CRABP1 (90%), and NR3C1 (35%). For the latter two genes, hypermethylation was significantly associated with absence or reduced gene expression. The methylation status of ADAMTS1, CRABP1, and NR3C1 was further investigated in 116 colorectal carcinomas and adenomas. Twenty-three of 63 (37%), 7/60 (12%), and 2/63 (3%) adenomas, as well as 37/52 (71%), 25/51 (49%), and 13/51 (25%) carcinomas were hypermethylated for the respective genes. These genes were unmethylated in tumors (n=82) from three other organs, prostate, testis, and kidney. Finally, analysis of normal colorectal mucosa demonstrated that the observed promoter hypermethylation was cancer-specific. CONCLUSION: By using a refined microarray screening approach we present three genes with cancer-specific hypermethylation in colorectal tumors, ADAMTS1, CRABP1, and NR3C1.


Asunto(s)
Proteínas ADAM/genética , Neoplasias Colorrectales/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Receptores de Glucocorticoides/genética , Receptores de Ácido Retinoico/genética , Proteína ADAMTS1 , Línea Celular Tumoral , Islas de CpG/genética , Metilación de ADN , Humanos , Análisis por Micromatrices , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ADN , Células Tumorales Cultivadas
8.
Neoplasia ; 7(2): 99-108, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15802015

RESUMEN

An unselected series of 310 colorectal carcinomas, stratified according to microsatellite instability (MSI) and DNA ploidy, was examined for mutations and/or promoter hypermethylation of five components of the WNT signaling cascade [APC, CTNNB1 (encoding beta-catenin), AXIN2, TCF4, and WISP3] and three genes indirectly affecting this pathway [CDH1 (encoding E-cadherin), PTEN, and TP53]. APC and TP53 mutations were each present more often in microsatellite-stable (MSS) tumors than in those with MSI (P < .001 for both). We confirmed that the aneuploid MSS tumors frequently contained TP53 mutations (P < .001), whereas tumors with APC mutations and/or promoter hypermethylation revealed no associations to ploidy. Mutations in APC upstream of codons 1020 to 1169, encoding the beta-catenin binding site, were found in 15/144 mutated tumors and these patients seemed to have poor clinical outcome (P = .096). Frameshift mutations in AXIN2, PTEN, TCF4, and WISP3 were found in 20%, 17%, 46%, and 28% of the MSI tumors, respectively. More than half of the tumors with heterozygote mutations in AXIN2 were concurrently mutated in APC. The present study showed that more than 90% of all samples had alteration in one or more of the genes investigated, adding further evidence to the vital importance of activated WNT signaling in colorectal carcinogenesis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Inestabilidad Cromosómica , Neoplasias Colorrectales/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Repeticiones de Microsatélite , Mutación/genética , Transducción de Señal , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Proteína Axina , Proteínas CCN de Señalización Intercelular , Cadherinas/genética , Proteínas del Citoesqueleto/genética , Metilación de ADN , Proteínas de Unión al ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/genética , Ploidias , Factores de Transcripción TCF , Transactivadores/genética , Proteína 2 Similar al Factor de Transcripción 7 , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Proteínas Wnt , beta Catenina
9.
Mol Cancer ; 3: 28, 2004 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-15476557

RESUMEN

BACKGROUND: Tumor cell lines are commonly used as experimental tools in cancer research, but their relevance for the in vivo situation is debated. In a series of 11 microsatellite stable (MSS) and 9 microsatellite unstable (MSI) colon cancer cell lines and primary colon carcinomas (25 MSS and 28 MSI) with known ploidy stem line and APC, KRAS, and TP53 mutation status, we analyzed the promoter methylation of the following genes: hMLH1, MGMT, p16INK4a (CDKN2A alpha-transcript), p14ARF (CDKN2A beta-transcript), APC, and E-cadherin (CDH1). We compared the DNA methylation profiles of the cell lines with those of the primary tumors. Finally, we examined if the epigenetic changes were associated with known genetic markers and/or clinicopathological variables. RESULTS: The cell lines and primary tumors generally showed similar overall distribution and frequencies of gene methylation. Among the cell lines, 15%, 50%, 75%, 65%, 20% and 15% showed promoter methylation for hMLH1, MGMT, p16INK4a, p14ARF, APC, and E-cadherin, respectively, whereas 21%, 40%, 32%, 38%, 32%, and 40% of the primary tumors were methylated for the same genes. hMLH1 and p14ARF were significantly more often methylated in MSI than in MSS primary tumors, whereas the remaining four genes showed similar methylation frequencies in the two groups. Methylation of p14ARF, which indirectly inactivates TP53, was seen more frequently in tumors with normal TP53 than in mutated samples, but the difference was not statistically significant. Methylation of p14ARF and p16INK4a was often present in the same primary tumors, but association to diploidy, MSI, right-sided location and female gender was only significant for p14ARF. E-cadherin was methylated in 14/34 tumors with altered APC further stimulating WNT signaling. CONCLUSIONS: The present study shows that colon cancer cell lines are in general relevant in vitro models, comparable with the in vivo situation, as the cell lines display many of the same molecular alterations as do the primary carcinomas. The combined pattern of epigenetic and genetic aberrations in the primary carcinomas reveals associations between them as well as to clinicopathological variables, and may aid in the future molecular assisted classification of clinically distinct stages.


Asunto(s)
Neoplasias del Colon/genética , Neoplasias Colorrectales/genética , Metilación de ADN , Proteínas Adaptadoras Transductoras de Señales , Cadherinas/genética , Proteínas Portadoras , Línea Celular Tumoral , Neoplasias del Colon/patología , Neoplasias Colorrectales/patología , Islas de CpG , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Femenino , Genes APC , Humanos , Masculino , Repeticiones de Microsatélite/genética , Homólogo 1 de la Proteína MutL , Proteínas de Neoplasias/genética , Proteínas Nucleares , O(6)-Metilguanina-ADN Metiltransferasa/genética , Regiones Promotoras Genéticas , Factores Sexuales , Proteína p14ARF Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA