Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Cancer ; 130(7): 1558-66, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21544814

RESUMEN

Defects in the mismatch repair system lead to microsatellite instability (MSI), a feature observed in ∼ 15% of all colorectal cancers (CRCs). Microsatellite mutations that drive tumourigenesis, typically inactivation of tumour suppressors, are selected for and are frequently detected in MSI cancers. Here, we evaluated somatic mutations in microsatellite repeats of 790 genes chosen based on reduced expression in MSI CRC and existence of a coding mononucleotide repeat of 6-10 bp in length. All the repeats were initially sequenced in 30 primary MSI CRC samples and whenever frameshift mutations were identified in >20%, additional 70 samples were sequenced. To distinguish driver mutations from passengers, we similarly analyzed the occurrence of frameshift mutations in 121 intronic control repeats and utilized a statistical regression model to determine cut-off mutation frequencies for repeats of all types (A/T and C/G, 6-10 bp). Along with several know target genes, including TGFBR2, ACVR2, and MSH3, six novel candidate driver genes emerged that harbored significantly more mutations than identical control repeats. The mutation frequencies in 100 MSI CRC samples were 51% in G8 of GLYR1, 47% in T9 of ABCC5, 43% in G8 of WDTC1, 33% in A8 of ROCK1, 30% in T8 of OR51E2, and 28% in A8 of TCEB3. Immunohistochemical staining of GLYR1 revealed defective protein expression in tumors carrying biallelic mutations, supporting a loss of function hypothesis. This is a large scale, unbiased effort to identify genes that when mutated are likely to contribute to MSI CRC development.


Asunto(s)
Neoplasias Colorrectales/genética , ADN de Neoplasias/genética , Inestabilidad de Microsatélites , Línea Celular Tumoral , Mutación del Sistema de Lectura , Células HCT116 , Humanos , Inmunohistoquímica/métodos , Repeticiones de Microsatélite , Tasa de Mutación , Análisis de Regresión
2.
Proc Natl Acad Sci U S A ; 105(14): 5513-8, 2008 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-18391202

RESUMEN

A recent study described a recessive ATPase activating germ-line mutation in smooth-muscle myosin (smmhc/myh11) underlying the zebrafish meltdown (mlt) phenotype. The mlt zebrafish develops intestinal abnormalities reminiscent of human Peutz-Jeghers syndrome (PJS) and juvenile polyposis (JP). To examine the role of MYH11 in human intestinal neoplasia, we searched for MYH11 mutations in patients with colorectal cancer (CRC), PJS and JP. We found somatic protein-elongating frameshift mutations in 55% of CRCs displaying microsatellite instability and in the germ-line of one individual with PJS. Additionally, two somatic missense mutations were found in one microsatellite stable CRC. These two missense mutations, R501L and K1044N, and the frameshift mutations were functionally evaluated. All mutations resulted in unregulated molecules displaying constitutive motor activity, similar to the mutant myosin underlying mlt. Thus, MYH11 mutations appear to contribute also to human intestinal neoplasia. Unregulated MYH11 may affect the cellular energy balance or disturb cell lineage decisions in tumor progenitor cells. These data challenge our view on MYH11 as a passive differentiation marker functioning in muscle contraction and add to our understanding of intestinal neoplasia.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Intestinales/genética , Mutación , Cadenas Pesadas de Miosina/genética , Miosina Tipo II/genética , Miosinas del Músculo Liso/genética , Poliposis Adenomatosa del Colon/genética , Secuencia de Aminoácidos , Estudios de Casos y Controles , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN , Humanos , Datos de Secuencia Molecular , Síndrome de Peutz-Jeghers/genética , Miosinas del Músculo Liso/fisiología
3.
Genes Chromosomes Cancer ; 48(7): 544-51, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19373782

RESUMEN

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a tumor predisposition syndrome with cutaneous and uterine leiomyomatosis as well as renal cell cancer (RCC) as its clinical manifestations. HLRCC is caused by heterozygous germline mutations in the fumarate hydratase (fumarase) gene. In this study, we used array comparative genomic hybridization to identify the specific copy number changes characterizing the HLRCC-associated RCCs. The study material comprised formalin-fixed paraffin-embedded renal tumors obtained from Finnish patients with HLRCC. All 11 investigated tumors displayed the papillary type 2 histopathology typical for HLRCC renal tumors. The most frequent copy number changes detected in at least 3/11 (27%) of the tumors were gains in chromosomes 2, 7, and 17, and losses in 13q12.3-q21.1, 14, 18, and X. These findings provide genetic evidence for a distinct copy number profile in HLRCC renal tumors compared with sporadic RCC tumors of the same histopathological subtype, and delineate chromosomal regions that associate with this very aggressive form of RCC.


Asunto(s)
Carcinoma de Células Renales/genética , Dosificación de Gen , Neoplasias Renales/genética , Leiomiomatosis/genética , Adulto , Anciano , Aberraciones Cromosómicas , Hibridación Genómica Comparativa , Femenino , Eliminación de Gen , Humanos , Masculino , Persona de Mediana Edad , Mutagénesis Insercional
4.
Cancer Res ; 67(12): 5691-8, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17575135

RESUMEN

Microsatellite instability (MSI) is observed in approximately 12% of colorectal cancers. Genes containing a mononucleotide microsatellite in the coding sequence are particularly prone to inactivation in MSI tumorigenesis, and much work has been conducted to identify genes with high repetitive tract mutation rates in these tumors. Much less attention has been paid to background mutation frequencies, and no work has focused on nontranscribed regions. Here, we studied 114 nontranscribed intergenic A/T and C/G repeats 6 to 10 bp in length, located distant from known genes, to examine background mutation frequencies in MSI colorectal cancers. A strong correlation with tract length was observed, and mutation frequencies of up to 87% were observed in 8 to 10 bp tracts. Subsequently, to compare the background mutation rate in transcribed and nontranscribed noncoding repeats, we screened nine randomly selected intronic C/G8 repeats. In addition, the coding repeats of seven suggested MSI target genes, and nine previously published intronic A8 and G8 repeats were analyzed. Intronic repeats seemed to mutate less frequently than nontranscribed intergenic repeats. Our results show that strand slippage mutations in mismatch repair-deficient cells are as abundant in short intergenic repeats as in many proposed MSI target genes. However, under mismatch repair deficiency, strand slippage mutations in transcribed sequences seem to be repaired more efficiently than in intergenic nontranscribed sequences. The mechanisms causing these differences are not yet understood and should be a subject for further studies. For MSI target gene identification, repeats in transcribed sequences seem to be the most appropriate reference group for coding region repeat mutations.


Asunto(s)
Neoplasias Colorrectales/genética , ADN de Neoplasias/genética , Inestabilidad de Microsatélites , Secuencias Repetitivas de Ácidos Nucleicos/genética , Análisis Mutacional de ADN , Humanos , Mutación , Reacción en Cadena de la Polimerasa
5.
Cancer Res ; 65(11): 4607-13, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15930278

RESUMEN

DNA mismatch repair (MMR)-deficient cells typically accumulate mutations in short repetitive DNA tracts. This microsatellite instability (MSI) facilitates malignant transformation when affecting genes with growth-related and caretaker functions. To date, several putative MSI target genes have been proposed mainly based on high mutation frequency within their coding regions. However, some intronic repeat mutations have also been suggested to associate with MSI tumorigenesis, indicating the need for additional analyses on noncoding repeats. Here we have analyzed an intronic T9 repeat of semenogelin I (SEMG1) and report mutation frequencies of 51% (75 of 146) and 62% (8 of 13) in MMR-deficient primary colorectal cancers and cell lines, respectively. The putative effect of the SEMG1 mutations was assessed by RNA and protein level analyses, but no differences were detected between colorectal cancer cell lines with different SEMG1 status. Subsequently, the general background mutation frequency of MSI colorectal cancers was assessed by screening for intergenic T9 repeat alterations. One of 10 examined repeats was mutated in 70% (102 of 145) of the colorectal cancers evaluated. The frequencies observed here are notably higher than previously published in noncoding repeats shorter than 10 bp in MMR-deficient primary tumors. Our results indicate that high mutation frequencies, similar or higher than those observed in proposed and approved target genes, can be detected in repeat tracts of MSI tumors without any apparent selection pressure. These data call for urgent and thorough large-scale evaluation of mutation frequencies in neutral short repetitive sequences in MMR-deficient tumors.


Asunto(s)
Neoplasias Colorrectales/genética , Mutación del Sistema de Lectura , Repeticiones de Microsatélite/genética , Proteínas de Secreción de la Vesícula Seminal/genética , Alelos , Disparidad de Par Base/genética , Secuencia de Bases , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Reparación del ADN/genética , ADN de Neoplasias/genética , Inestabilidad Genómica , Humanos , Intrones/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Secreción de la Vesícula Seminal/biosíntesis , Proteínas de Secreción de la Vesícula Seminal/metabolismo
6.
Clin Cancer Res ; 11(17): 6311-6, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16144935

RESUMEN

We have recently reported that low tumor levels of SMAD4, a key mediator of transforming growth factor-beta superfamily signaling, can predict the probability of recurrence in patients with Dukes C colorectal cancer who had surgery as the only form of treatment. However, standard treatment for Dukes C colorectal cancer patients currently involves the administration of 5-fluorouracil (5-FU)-based adjuvant chemotherapy after surgery. Approximately 30% to 40% of these patients present with recurrence and die within 5 years, and there is great need for markers capable of predicting poor prognosis after the combined surgery/adjuvant treatment. In this study, we evaluate the prognostic value of SMAD4 in patients treated with surgery and 5-FU-based adjuvant therapy. We used immunohistochemistry and quantitative real-time reverse transcription-PCR to measure the levels of SMAD4 protein and mRNA expression in the primary tumors and a number of lymph node metastases from a series of 75 Dukes C colorectal cancer patients with at least 6 years of follow-up. Patients with tumors expressing low levels of SMAD4 protein or mRNA showed significantly shorted disease-free and overall survival than patients with high tumor levels of SMAD4. The median survival of patients with low SMAD4 protein or mRNA tumor levels was 1.4 and 1.2 years, respectively, whereas patients with high SMAD4 tumor level had a median survival of >9.3 years. In addition, the protein and mRNA levels of SMAD4 in lymph node metastases was significantly lower than in primary tumors (P = 0.006). In contrast, allelic imbalance in chromosome 18q21 was of no prognostic significance in these patients. In conclusion, low SMAD4 tumor levels identified a subset of patients with poor prognosis following surgery and 5-FU-based adjuvant therapy; therefore, these patients could be good candidates to receive combined treatment with additional chemotherapeutic agents such as CPT-11 and/or oxaliplatin.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas de Unión al ADN/metabolismo , Fluorouracilo/uso terapéutico , Transactivadores/metabolismo , Desequilibrio Alélico , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Quimioterapia Adyuvante , Cromosomas Humanos Par 18/genética , Neoplasias Colorrectales/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Masculino , Persona de Mediana Edad , Mutación , Pronóstico , ARN Mensajero/metabolismo , Proteína Smad4 , Tasa de Supervivencia , Transactivadores/genética
7.
Clin Cancer Res ; 11(7): 2606-11, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15814640

RESUMEN

More than 50% of patients with Dukes C colorectal cancer have disease recurrence and die within 5 years after surgical removal of their primary tumor. It is currently not possible to distinguish patients with good and bad prognosis. SMAD4 is an important tumor suppressor gene that mediates transforming growth factor-beta superfamily signaling and is located in chromosome 18q21, a region with frequent genetic losses in these tumors. Allelic imbalance in 18q has been linked to poor prognosis in a subset of colorectal cancer patients. Therefore, we generated a tissue microarray containing triplicate tumor samples from 86 Dukes C patients and used immunohistochemistry to assess the relative expression level of SMAD4 and its value as a prognostic marker. In addition, SMAD4 was screened for mutations and two polymorphic microsatellite markers were used to assess the presence of allelic imbalance in these tumors. Patients with tumors expressing high SMAD4 levels had significantly better overall (P < 0.025) and disease-free (P < 0.013) survival than patients with low levels. This identifies SMAD4 as a prognostic marker for Dukes C colorectal cancer. Although all tumors with absent SMAD4 staining showed allelic imbalance in 18q21, tumors with 18q21 allelic imbalance as a group showed no difference in SMAD4 levels compared with tumors without allelic imbalance, suggesting that additional mechanisms of SMAD4 down-regulation exist. In addition, although SMAD4 mutations were found in five tumors, they were not associated with shorter survival. In conclusion, the level of expression of SMAD4 was found to be a more sensitive marker than 18q21 allelic imbalance and SMAD4 mutations, which were of no prognostic significance for these patients.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/genética , Transactivadores/genética , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Cromosomas Humanos Par 18/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Análisis Mutacional de ADN , ADN de Neoplasias/química , ADN de Neoplasias/genética , Proteínas de Unión al ADN/análisis , Femenino , Humanos , Inmunohistoquímica , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Mutación , Pronóstico , Proteína Smad4 , Análisis de Supervivencia , Transactivadores/análisis
8.
Mol Cancer Res ; 8(7): 952-60, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20551151

RESUMEN

The circadian clock regulates daily variations in physiologic processes. CLOCK acts as a regulator in the circadian apparatus controlling the expression of other clock genes, including PER1. Clock genes have been implicated in cancer-related functions; in this work, we investigated CLOCK as a possible target of somatic mutations in microsatellite unstable colorectal cancers. Combining microarray gene expression data and public gene sequence information, we identified CLOCK as 1 of 790 putative novel microsatellite instability (MSI) target genes. A total of 101 MSI colorectal carcinomas (CRC) were sequenced for a coding microsatellite in CLOCK. The effect of restoring CLOCK expression was studied in LS180 cells lacking wild-type CLOCK by stably expressing GST-CLOCK or glutathione S-transferase empty vector and testing the effects of UV-induced apoptosis and radiation by DNA content analysis using flow cytometry. Putative novel CLOCK target genes were searched by using ChIP-seq. CLOCK mutations occurred in 53% of MSI CRCs. Restoring CLOCK expression in cells with biallelic CLOCK inactivation resulted in protection against UV-induced apoptosis and decreased G(2)-M arrest in response to ionizing radiation. Using ChIP-Seq, novel CLOCK-binding elements were identified near DNA damage genes p21, NBR1, BRCA1, and RAD50. CLOCK is shown to be mutated in cancer, and altered response to DNA damage provides one plausible mechanism of tumorigenesis.


Asunto(s)
Adenocarcinoma/genética , Proteínas CLOCK/genética , Neoplasias Colorrectales/genética , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular Tumoral , Mutación del Sistema de Lectura , Perfilación de la Expresión Génica , Células HCT116 , Humanos , Repeticiones de Microsatélite , Datos de Secuencia Molecular , Mutación
9.
Gastroenterology ; 129(3): 874-84, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16143127

RESUMEN

BACKGROUND & AIMS: Although approximately 50% of Dukes' C colorectal cancer patients are surgically cured, it is currently not possible to distinguish these patients from those at high risk of recurrence. The recent advent of routine adjuvant chemotherapy for these patients has greatly complicated the identification of new markers predicting the response to surgery, which is now reliant on archived materials. Microarray analysis allows fine tumor classification but cannot be used with paraffin-embedded archival samples. METHODS: We used microarray analysis of a unique set of fresh-frozen tumor samples from Dukes' C patients who had surgery as the only form of treatment to identify molecular signatures that characterize tumors from patients with good and bad prognosis. RESULTS: Unsupervised hierarchical clustering and a K-nearest neighbors-based classifier identified groups of patients with significantly different survival (P = .019 and P = .0001). Expression profiling outperformed previously reported genetic markers of prognosis such as TP53 and K-RAS mutational status and allelic imbalance in chromosome 18q, which were of limited prognostic power in this study. Functional categories significantly enriched in gene-expression differences included protein transport and folding. The prognostic potential of the RAS homologue RHOA, one of the most differentially expressed genes, was further investigated using immunohistochemistry and a tissue microarray containing 137 independent Dukes' C tumor samples. Reduced RHOA expression was associated with significantly shorter survival (P = .01). CONCLUSIONS: This study shows that gene-expression profiling of surgical tumor samples can predict recurrence in Dukes' C patients. Therefore, this approach could be used to guide decisions concerning the clinical management of these patients.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Perfilación de la Expresión Génica , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/mortalidad , Supervivencia sin Enfermedad , Genes p53 , Genes ras , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Valor Predictivo de las Pruebas , Pronóstico , ARN Neoplásico/genética , ARN Neoplásico/aislamiento & purificación , Recurrencia , Análisis de Supervivencia , Factores de Tiempo , Proteína de Unión al GTP rhoA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA