Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 135(7): 1167-9, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-19109887

RESUMEN

Whether 5-methylcytosine (meC) can be enzymatically removed from vertebrate DNA has been the subject of extensive study and also some controversy. Rai et al. (2008) now report that cytosine demethylation can be accomplished in a one-cell zebrafish embryo by the combined action of a cytidine deaminase and a thymine DNA glycosylase.


Asunto(s)
Citosina/metabolismo , Metilación de ADN , Animales , Citidina Desaminasa/metabolismo , Timina ADN Glicosilasa/metabolismo , Pez Cebra
2.
BMC Cancer ; 20(1): 290, 2020 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-32252665

RESUMEN

BACKGROUND: Identifying molecular differences between primary and metastatic colorectal cancers-now possible with the aid of omics technologies-can improve our understanding of the biological mechanisms of cancer progression and facilitate the discovery of novel treatments for late-stage cancer. We compared the DNA methylomes of primary colorectal cancers (CRCs) and CRC metastases to the liver. Laser microdissection was used to obtain epithelial tissue (10 to 25 × 106 µm2) from sections of fresh-frozen samples of primary CRCs (n = 6), CRC liver metastases (n = 12), and normal colon mucosa (n = 3). DNA extracted from tissues was enriched for methylated sequences with a methylCpG binding domain (MBD) polypeptide-based protocol and subjected to deep sequencing. The performance of this protocol was compared with that of targeted enrichment for bisulfite sequencing used in a previous study of ours. RESULTS: MBD enrichment captured a total of 322,551 genomic regions (249.5 Mb or ~ 7.8% of the human genome), which included over seven million CpG sites. A few of these regions were differentially methylated at an expected false discovery rate (FDR) of 5% in neoplastic tissues (primaries: 0.67%, i.e., 2155 regions containing 279,441 CpG sites; liver metastases: 1%, i.e., 3223 regions containing 312,723 CpG sites) as compared with normal mucosa samples. Most of the differentially methylated regions (DMRs; 94% in primaries; 70% in metastases) were hypermethylated, and almost 80% of these (1882 of 2396) were present in both lesion types. At 5% FDR, no DMRs were detected in liver metastases vs. primary CRC. However, short regions of low-magnitude hypomethylation were frequent in metastases but rare in primaries. Hypermethylated DMRs were far more abundant in sequences classified as intragenic, gene-regulatory, or CpG shelves-shores-island segments, whereas hypomethylated DMRs were equally represented in extragenic (mainly, open-sea) and intragenic (mainly, gene bodies) sequences of the genome. Compared with targeted enrichment, MBD capture provided a better picture of the extension of CRC-associated DNA hypermethylation but was less powerful for identifying hypomethylation. CONCLUSIONS: Our findings demonstrate that the hypermethylation phenotype in CRC liver metastases remains similar to that of the primary tumor, whereas CRC-associated DNA hypomethylation probably undergoes further progression after the cancer cells have migrated to the liver.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Epigenoma , Neoplasias Hepáticas/secundario , Neoplasias Colorrectales/metabolismo , Epigénesis Genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Captura por Microdisección con Láser/métodos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Fenotipo , Regiones Promotoras Genéticas
3.
Chem Res Toxicol ; 30(3): 769-776, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28076683

RESUMEN

Bioactivation as well as DNA repair affects the susceptibility of cancer cells to the action of DNA-alkylating chemotherapeutic drugs. However, information is limited with regard to the relative contributions of these processes to the biological outcome of metabolically activated DNA alkylating agents. We evaluated the influence of cellular bioactivation capacity and DNA repair on cytotoxicity of the DNA alkylating agent acylfulvene (AF). We compared the cytotoxicity and RNA synthesis inhibition by AF and its synthetic activated analogue iso-M0 in a panel of fibroblast cell lines with deficiencies in transcription-coupled (TC-NER) or global genome nucleotide excision repair (GG-NER). We related these data to the inherent bioactivation capacity of each cell type on the basis of mRNA levels. We demonstrated that specific inactivation of TC-NER by siRNA had the largest positive impact on AF activity in a cancer cell line. These findings establish that transcription-coupled DNA repair reduces cellular sensitivity to AF, independent of the requirement for bioactivation.


Asunto(s)
Reparación del ADN , Sesquiterpenos/farmacología , Compuestos de Espiro/farmacología , Transcripción Genética/efectos de los fármacos , Activación Metabólica , Línea Celular , Humanos , Sesquiterpenos/farmacocinética , Compuestos de Espiro/farmacocinética
4.
BMC Cancer ; 14: 46, 2014 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24472434

RESUMEN

BACKGROUND: Biological processes are controlled by transcription networks. Expression changes of transcription factor (TF) genes in precancerous lesions are therefore crucial events in tumorigenesis. Our aim was to obtain a comprehensive picture of these changes in colorectal adenomas. METHODS: Using a 3-pronged selection procedure, we analyzed transcriptomic data on 34 human tissue samples (17 adenomas and paired samples of normal mucosa, all collected with ethics committee approval and written, informed patient consent) to identify TFs with highly significant tumor-associated gene expression changes whose potential roles in colorectal tumorigenesis have been under-researched. Microarray data were subjected to stringent statistical analysis of TF expression in tumor vs. normal tissues, MetaCore-mediated identification of TF networks displaying enrichment for genes that were differentially expressed in tumors, and a novel quantitative analysis of the publications examining the TF genes' roles in colorectal tumorigenesis. RESULTS: The 261 TF genes identified with this procedure included DACH1, which plays essential roles in the proper proliferation and differentiation of retinal and leg precursor cell populations in Drosophila melanogaster. Its possible roles in colorectal tumorigenesis are completely unknown, but it was found to be markedly overexpressed (mRNA and protein) in all colorectal adenomas and in most colorectal carcinomas. However, DACH1 expression was absent in some carcinomas, most of which were DNA mismatch-repair deficient. When networks were built using the set of TF genes identified by all three selection procedures, as well as the entire set of transcriptomic changes in adenomas, five hub genes (TGFB1, BIRC5, MYB, NR3C1, and TERT) where identified as putatively crucial components of the adenomatous transformation process. CONCLUSION: The transcription-regulating network of colorectal adenomas (compared with that of normal colorectal mucosa) is characterized by significantly altered expression of over 250 TF genes, many of which have never been investigated in relation to colorectal tumorigenesis.


Asunto(s)
Adenoma/genética , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Perfilación de la Expresión Génica , Factores de Transcripción/genética , Adenoma/metabolismo , Adenoma/patología , Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Análisis por Conglomerados , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Genes myb , Humanos , Inmunohistoquímica , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Survivin , Telomerasa/genética , Telomerasa/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
5.
Nucleic Acids Res ; 38(22): 8131-40, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20705653

RESUMEN

It is known that transcription can induce DNA recombination, thus compromising genomic stability. RECQ5 DNA helicase promotes genomic stability by regulating homologous recombination. Recent studies have shown that RECQ5 forms a stable complex with RNA polymerase II (RNAPII) in human cells, but the cellular role of this association is not understood. Here, we provide evidence that RECQ5 specifically binds to the Ser2,5-phosphorylated C-terminal repeat domain (CTD) of the largest subunit of RNAPII, RPB1, by means of a Set2-Rpb1-interacting (SRI) motif located at the C-terminus of RECQ5. We also show that RECQ5 associates with RNAPII-transcribed genes in a manner dependent on the SRI motif. Notably, RECQ5 density on transcribed genes correlates with the density of Ser2-CTD phosphorylation, which is associated with the productive elongation phase of transcription. Furthermore, we show that RECQ5 negatively affects cell viability upon inhibition of spliceosome assembly, which can lead to the formation of mutagenic R-loop structures. These data indicate that RECQ5 binds to the elongating RNAPII complex and support the idea that RECQ5 plays a role in the maintenance of genomic stability during transcription.


Asunto(s)
ARN Polimerasa II/metabolismo , RecQ Helicasas/metabolismo , Transcripción Genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular , Supervivencia Celular , Secuencia Conservada , Humanos , Datos de Secuencia Molecular , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , ARN Polimerasa II/química , RecQ Helicasas/química , Secuencias Repetitivas de Aminoácido , Empalmosomas/metabolismo
6.
Epigenetics Chromatin ; 13(1): 25, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32487212

RESUMEN

BACKGROUND: DNA methylation is a highly studied epigenetic signature that is associated with regulation of gene expression, whereby genes with high levels of promoter methylation are generally repressed. Genomic imprinting occurs when one of the parental alleles is methylated, i.e., when there is inherited allele-specific methylation (ASM). A special case of imprinting occurs during X chromosome inactivation in females, where one of the two X chromosomes is silenced, to achieve dosage compensation between the sexes. Another more widespread form of ASM is sequence dependent (SD-ASM), where ASM is linked to a nearby heterozygous single nucleotide polymorphism (SNP). RESULTS: We developed a method to screen for genomic regions that exhibit loss or gain of ASM in samples from two conditions (treatments, diseases, etc.). The method relies on the availability of bisulfite sequencing data from multiple samples of the two conditions. We leverage other established computational methods to screen for these regions within a new R package called DAMEfinder. It calculates an ASM score for all CpG sites or pairs in the genome of each sample, and then quantifies the change in ASM between conditions. It then clusters nearby CpG sites with consistent change into regions. In the absence of SNP information, our method relies only on reads to quantify ASM. This novel ASM score compares favorably to current methods that also screen for ASM. Not only does it easily discern between imprinted and non-imprinted regions, but also females from males based on X chromosome inactivation. We also applied DAMEfinder to a colorectal cancer dataset and observed that colorectal cancer subtypes are distinguishable according to their ASM signature. We also re-discover known cases of loss of imprinting. CONCLUSION: We have designed DAMEfinder to detect regions of differential ASM (DAMEs), which is a more refined definition of differential methylation, and can therefore help in breaking down the complexity of DNA methylation and its influence in development and disease.


Asunto(s)
Alelos , Metilación de ADN , Epigenómica/métodos , Análisis de Secuencia de ADN/métodos , Neoplasias Colorrectales/genética , Islas de CpG , Femenino , Impresión Genómica , Humanos , Masculino , Programas Informáticos
7.
Mol Cancer ; 8: 124, 2009 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20015382

RESUMEN

BACKGROUND: Tumor development in the human colon is commonly accompanied by epigenetic changes, such as DNA methylation and chromatin modifications. These alterations result in significant, inheritable changes in gene expression that contribute to the selection of tumor cells with enhanced survival potential. RESULTS: A recent high-throughput gene expression analysis conducted by our group identified numerous genes whose transcription was markedly diminished in colorectal tumors. One of these, the protein-tyrosine phosphatase receptor type R (PTPRR) gene, was dramatically downregulated from the earliest stages of cellular transformation. Here, we show that levels of both major PTPRR transcript variants are markedly decreased (compared with normal mucosal levels) in precancerous and cancerous colorectal tumors, as well in colorectal cancer cell lines. The expression of the PTPRR-1 isoform was inactivated in colorectal cancer cells as a result of de novo CpG island methylation and enrichment of transcription-repressive histone-tail marks, mainly H3K27me3. De novo methylation of the PTPRR-1 transcription start site was demonstrated in 29/36 (80%) colorectal adenomas, 42/44 (95%) colorectal adenocarcinomas, and 8/8 (100%) liver metastases associated with the latter tumors. CONCLUSIONS: Epigenetic downregulation of PTPRR seems to be an early alteration in colorectal cell transformation, which is maintained during the clonal selection associated with tumor progression. It may represent a preliminary step in the constitutive activation of the RAS/RAF/MAPK/ERK signalling, an effect that will later be consolidated by mutations in genes encoding key components of this pathway.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Colorrectales/genética , Silenciador del Gen , Proteínas Tirosina Fosfatasas Clase 7 Similares a Receptores/genética , Adenocarcinoma/patología , Neoplasias Colorrectales/patología , Metilación de ADN , Humanos , ARN Mensajero/genética
8.
Mol Cancer Res ; 5(12): 1263-75, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18171984

RESUMEN

Colorectal cancers are believed to arise predominantly from adenomas. Although these precancerous lesions have been subjected to extensive clinical, pathologic, and molecular analyses, little is currently known about the global gene expression changes accompanying their formation. To characterize the molecular processes underlying the transformation of normal colonic epithelium, we compared the transcriptomes of 32 prospectively collected adenomas with those of normal mucosa from the same individuals. Important differences emerged not only between the expression profiles of normal and adenomatous tissues but also between those of small and large adenomas. A key feature of the transformation process was the remodeling of the Wnt pathway reflected in patent overexpression and underexpression of 78 known components of this signaling cascade. The expression of 19 Wnt targets was closely correlated with clear up-regulation of KIAA1199, whose function is currently unknown. In normal mucosa, KIAA1199 expression was confined to cells in the lower portion of intestinal crypts, where Wnt signaling is physiologically active, but it was markedly increased in all adenomas, where it was expressed in most of the epithelial cells, and in colon cancer cell lines, it was markedly reduced by inactivation of the beta-catenin/T-cell factor(s) transcription complex, the pivotal mediator of Wnt signaling. Our transcriptomic profiles of normal colonic mucosa and colorectal adenomas shed new light on the early stages of colorectal tumorigenesis and identified KIAA1199 as a novel target of the Wnt signaling pathway and a putative marker of colorectal adenomatous transformation.


Asunto(s)
Adenoma/genética , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Adenoma/patología , Anciano , Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/patología , Femenino , Marcadores Genéticos , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia , ARN Mensajero/metabolismo , Transcripción Genética
9.
Oncol Rep ; 17(6): 1421-7, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17487400

RESUMEN

Epigenetic alterations have been reported in colorectal neoplasia which can either complement or in some cases be predisposed to genetic alterations such as K-ras mutations. We examined the promoter methylation status of the CDKN2A and O6-methylguanine-DNA methyltransferase (MGMT) genes, after sodium bisulfite conversion and DNA amplification with methylation specific PCR. Moreover, we searched for G to A transitions in codons 12 and 13 of the K-ras oncogene in normal colorectal mucosae, aberrant crypt foci (ACF, early premalignant lesions) and carcinomas. CDKN2A hypermethylation was an infrequent event in ACF (2 of 26, 7.7%). On the contrary, MGMT hypermethylation was found in the normal mucosae (3 of the 12 samples, 25%), in 14 of the 26 ACF (53.8%) and in 7 of the 9 (77.8%) carcinomas examined. K-ras mutations were evident in 6 ACF (23%) and in 3 carcinomas (33.3%), mostly associated with MGMT promoter hypermethylation. These findings strongly support the hypothesis that epigenetic mechanisms play an important role in the early steps of colorectal carcinogenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , O(6)-Metilguanina-ADN Metiltransferasa/genética , Anciano , Anciano de 80 o más Años , Carcinoma/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Metilación de ADN , Femenino , Genes ras/genética , Humanos , Mucosa Intestinal/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Sulfitos/química
10.
Cancer Res ; 65(23): 10759-66, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16322221

RESUMEN

The human mismatch repair (MMR) proteins hMLH1 and hPMS2 function in MMR as a heterodimer. Cells lacking either protein have a strong mutator phenotype and display microsatellite instability, yet mutations in the hMLH1 gene account for approximately 50% of hereditary nonpolyposis colon cancer families, whereas hPMS2 mutations are substantially less frequent and less penetrant. Similarly, in the mouse model, Mlh1-/- animals are highly cancer prone and present with gastrointestinal tumors at an early age, whereas Pms2-/- mice succumb to cancer much later in life and do not present with gastrointestinal tumors. This evidence suggested that MLH1 might functionally interact with another MutL homologue, which compensates, at least in part, for a deficiency in PMS2. Sterility of Mlh1-/-, Pms2-/-, and Mlh3-/- mice implicated the Mlh1/Pms2 and Mlh1/Mlh3 heterodimers in meiotic recombination. We now show that the hMLH1/hMLH3 heterodimer, hMutLgamma, can also assist in the repair of base-base mismatches and single extrahelical nucleotides in vitro. Analysis of hMLH3 expression in colon cancer cell lines indicated that the protein levels vary substantially and independently of hMLH1. If hMLH3 participates in MMR in vivo, its partial redundancy with hPMS2, coupled with the fluctuating expression levels of hMLH3, may help explain the low penetrance of hPMS2 mutations in hereditary nonpolyposis colon cancer families.


Asunto(s)
Disparidad de Par Base , Proteínas Portadoras/fisiología , Reparación del ADN/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Baculoviridae/genética , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Humanos , Homólogo 1 de la Proteína MutL , Proteínas MutL , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Spodoptera/genética , Spodoptera/metabolismo
11.
Clin Cancer Res ; 11(23): 8332-40, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16322293

RESUMEN

PURPOSE: Many studies have evaluated the role of high levels of microsatellite instability (MSI) as a prognostic marker and predictor of the response to chemotherapy in colorectal cancer (CRC); however, the results are not conclusive. The aim of this study was to analyze the prognostic significance of high levels of MSI (MSI-H) in CRC patients in relation to fluorouracil-based chemotherapy. EXPERIMENTAL DESIGN: In three different institutions, 1,263 patients with CRC were tested for the presence of MSI, and CRC-specific survival was then analyzed in relation to MSI status, chemotherapy, and other clinical and pathologic variables. RESULTS: Two hundred and fifty-six tumors were MSI-H (20.3%): these were more frequently at a less advanced stage, right-sided, poorly differentiated, with mucinous phenotype, and expansive growth pattern than microsatellite stable carcinomas. Univariate and multivariate analyses of 5-year-specific survival revealed stage, tumor location, grade of differentiation, MSI, gender, and age as significant prognostic factors. The prognostic advantage of MSI tumors was particularly evident in stages II and III in which chemotherapy did not significantly affect the survival of MSI-H patients. Finally, we analyzed survival in MSI-H patients in relation to the presence of mismatch repair gene mutations. MSI-H patients with hereditary non-polyposis colorectal cancer showed a better prognosis as compared with sporadic MSI-H; however, in multivariate analysis, this difference disappeared. CONCLUSIONS: The type of genomic instability could influence the prognosis of CRC, in particular in stages II and III. Fluorouracil-based chemotherapy does not seem to improve survival among MSI-H patients. The survival benefit for patients with hereditary non-polyposis colorectal cancer is mainly determined by younger age and less advanced stage as compared with sporadic MSI-H counterpart.


Asunto(s)
Neoplasias Colorrectales/genética , Inestabilidad Genómica , Repeticiones de Microsatélite/genética , Proteínas Adaptadoras Transductoras de Señales , Adenocarcinoma/diagnóstico , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma Mucinoso/diagnóstico , Adenocarcinoma Mucinoso/tratamiento farmacológico , Adenocarcinoma Mucinoso/genética , Antimetabolitos Antineoplásicos/uso terapéutico , Proteínas Portadoras/genética , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas de Unión al ADN/genética , Femenino , Fluorouracilo/uso terapéutico , Humanos , Masculino , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS/genética , Estadificación de Neoplasias , Proteínas Nucleares/genética , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia , Resultado del Tratamiento
12.
Cancer Res ; 63(23): 8158-66, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14678970

RESUMEN

DNA mismatch repair (MMR) plays a key role in the cytotoxic response of human cells to methylating agents, however, the cascade of events leading to cell cycle arrest and cell death has yet to be characterized. We studied the role of MMR in the transcriptional response to DNA methylation damage in two human cellular models: (a). the lymphoblastoid cell line TK6 and its derivative MT1, which is mutated in the MMR gene hMSH6; and (b). the epithelial cell line 293T Lalpha in which the expression of the MMR gene hMLH1 can be tightly regulated and p53 is inactivated. Upon N-methyl-N'-nitro-N-nitrosoguanidine treatment, only cells with functional MMR were killed, but the type of cytotoxic response differed. In TK6 cells, S-phase arrest and apoptosis were accompanied by a dramatic change in gene expression, notably, an up-regulation of several genes encoding growth inhibitors and proapoptotic factors both p53 dependent and independent. In contrast, the MMR-dependent transcriptional response in 293T Lalpha cells was substantially less pronounced than in TK6 cells, despite an efficient induction of a G(2)-M checkpoint and nonapoptotic cell death. Thus, we demonstrate that in human cells of different origin, MMR-mediated killing by methylating agents occurs through different pathways and regardless of the p53 status. Moreover, once DNA methylation damage has been processed by the MMR system, tumor cells might be committed to die, although one or more of their signaling pathways are impaired.


Asunto(s)
Disparidad de Par Base/fisiología , Metilación de ADN/efectos de los fármacos , Reparación del ADN/fisiología , Metilnitronitrosoguanidina/toxicidad , Proteínas Adaptadoras Transductoras de Señales , Linfocitos B/efectos de los fármacos , Linfocitos B/fisiología , Proteínas Portadoras , Línea Celular , Reparación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Humanos , Homólogo 1 de la Proteína MutL , Mutación Missense , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/deficiencia , Proteínas Nucleares , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/fisiología , Regulación hacia Arriba/efectos de los fármacos
13.
Pathol Res Pract ; 200(5): 371-7, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15239345

RESUMEN

The main purpose of this study was to examine the expression of mucins and mismatch repair proteins in colorectal carcinomas. The immunohistochemical distribution of apomucins MUC2, MUC5AC, and the expression of MLH1 and MSH2 proteins were examined in 76 mucinous and 60 non-mucinous colorectal carcinomas. MUC2 was noted in all mucinous carcinomas, whereas MUC5AC was present in 41 cases only (54%). In non-mucinous carcinomas, MUC2 was expressed in 61.7% of the tumors; by contrast, MUC5AC was present in 20% of the cases. The expression level of apomucins was significantly different in mucinous and non-mucinous lesions (p<0.001). Twenty-seven (35.5%) of the mucinous carcinomas showed no MLH1 expression, whereas 11 (18.3%) of the non-mucinous tumors did. This difference was statistically significant (p<0.005). Altered expression of MSH2 protein was never observed. The lack of MLH1 expression was considerably more frequent in carcinomas with secretion of MUC5AC (p<0.005). Our study has demonstrated this close relationship by immunohistochemical methods. In summary, our data show: (1) differences in the expression of mucins between mucinous and non-mucinous tumors; (2) a high frequency of altered MLH1 protein expression (35.5%) in mucinous carcinomas; (3) a significant relationship between the presence of MUC5AC and the altered expression of MLH1 protein in colorectal carcinomas.


Asunto(s)
Adenocarcinoma Mucinoso/patología , Neoplasias Colorrectales/patología , Mucinas/biosíntesis , Proteínas de Neoplasias/biosíntesis , Proteínas Adaptadoras Transductoras de Señales , Adenocarcinoma Mucinoso/metabolismo , Adulto , Anciano , Proteínas Portadoras , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mucina 5AC , Mucina 2 , Homólogo 1 de la Proteína MutL , Proteínas Nucleares
14.
Genome Biol ; 15(2): R35, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24517713

RESUMEN

Affinity capture of DNA methylation combined with high-throughput sequencing strikes a good balance between the high cost of whole genome bisulfite sequencing and the low coverage of methylation arrays. We present BayMeth, an empirical Bayes approach that uses a fully methylated control sample to transform observed read counts into regional methylation levels. In our model, inefficient capture can readily be distinguished from low methylation levels. BayMeth improves on existing methods, allows explicit modeling of copy number variation, and offers computationally efficient analytical mean and variance estimators. BayMeth is available in the Repitools Bioconductor package.


Asunto(s)
Teorema de Bayes , Metilación de ADN/genética , Estudios de Evaluación como Asunto , Secuenciación de Nucleótidos de Alto Rendimiento , Islas de CpG/genética , Variaciones en el Número de Copia de ADN , Genoma Humano , Humanos
15.
PLoS One ; 8(7): e69473, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936024

RESUMEN

We previously reported that the expression of KIAA1199 in human colorectal tumors (benign and malignant) is markedly higher than that in the normal colonic mucosa. In this study, we investigated the functions of the protein encoded by this gene, which are thus far unknown. Immunostaining studies were used to reveal its subcellular localization, and proteomic and gene expression experiments were conducted to identify proteins that might interact with KIAA1199 and molecular pathways in which it might play roles. Using colon cancer cell lines, we showed that both endogenous and ectopically expressed KIAA1199 is secreted into the extracellular environment. In the cells, it was found mainly in the perinuclear space (probably the ER) and cell membrane. Both cellular compartments were also over-represented in lists of proteins identified by mass spectrometry as putative KIAA1199 interactors and/or proteins encoded by genes whose transcription was significantly changed by KIAA1199 expression. These proteomic and transcriptomic datasets concordantly link KIAA1199 to several genes/proteins and molecular pathways, including ER processes like protein binding, transport, and folding; and Ca(2+), G-protein, ephrin, and Wnt signaling. Immunoprecipitation experiments confirmed KIAA1199's interaction with the cell-membrane receptor ephrin A2 and with the ER receptor ITPR3, a key player in Ca(2+) signaling. By modulating Ca(2+) signaling, KIAA1199 could affect different branches of the Wnt network. Our findings suggest it may negatively regulate the Wnt/CTNNB1 signaling, and its expression is associated with decreased cell proliferation and invasiveness.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteínas/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Forma de la Célula/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Humanos , Hialuronoglucosaminidasa , Invasividad Neoplásica , Unión Proteica/genética , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Vía de Señalización Wnt/genética
16.
Toxicol In Vitro ; 25(2): 446-53, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21095227

RESUMEN

Cigarette smoke causes lung tumorigenesis; however, the mechanisms underlying transformation are unknown. We investigated if tobacco compounds induce DNA promoter hypermethylation in BEAS-2B cells treated with low doses of cigarette smoke condensate (CSC) for one month. Transcriptional profiles and anchorage-independent growth were explored using Affymetrix microarray and soft agar assay, respectively. To investigate if tobacco compounds induce hypermethylation, CSC/dimethyl sulfoxide (DMSO)-treated cells were further treated with 5-Aza-2'-deoxycytidine (5AzaC) and trychostatin A (TSA). This treatment was followed by transcriptional profiling. CSC-exposed cells acquired a fibroblast-like shape with enhanced anchorage-independent growth. Silencing of epithelial cadherin, the hallmark of epithelial to mesenchymal transition (EMT), was observed upon exposure to CSC. Changes in the expression of genes involved in epidermal development, intercellular junction formation, and cytoskeleton formation were identified. Gene expression profiles from 5AzaC- and TSA-treated cells revealed 130 genes possibly methylated due to chronic CSC exposure. Our results suggest that E-cadherin may also be silenced by hypermethylation in an in vitro model of chronic exposure to low doses of CSC. This study demonstrates evidence for a tobacco compound induced EMT-like process in vitro and provides insight into possible mechanisms of gene silencing occurring during this treatment.


Asunto(s)
Bronquios/patología , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Nicotiana/toxicidad , Humo/efectos adversos , Azacitidina/análogos & derivados , Azacitidina/farmacología , Células Cultivadas , Citoesqueleto/fisiología , Metilación de ADN , Decitabina , Perfilación de la Expresión Génica , Glucósidos/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Uniones Intercelulares/fisiología
17.
EMBO Mol Med ; 3(6): 334-47, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21538994

RESUMEN

Improved colonoscopy is revealing precancerous lesions that were frequently missed in the past, and ∼30% of those detected today have nonpolypoid morphologies ranging from slightly raised to depressed. To characterize these lesions molecularly, we assessed transcription of 23,768 genes in 42 precancerous lesions (25 slightly elevated nonpolypoid and 17 pedunculated polypoid), each with corresponding samples of normal mucosa. Nonpolypoid versus polypoid morphology explained most gene expression variance among samples; histology, size, and degree of dysplasia were also linked to specific patterns. Expression changes in polypoid lesions frequently affected cell-cycling pathways, whereas cell-survival dysregulation predominated in nonpolypoid lesions. The latter also displayed fewer and less dramatic expression changes than polypoid lesions. Paradigmatic of this trend was progressive loss through the normal > nonpolypoid > polypoid > cancer sequence of TMIGD1 mRNA and protein. This finding, along with TMIGD1 protein expression patterns in tissues and cell lines, suggests that TMIGD1 might be associated with intestinal-cell differentiation. We conclude that molecular dysregulation in slightly elevated, nonpolypoid, precancerous colorectal lesions may be somewhat less severe than that observed in classic adenomatous polyps.


Asunto(s)
Neoplasias Colorrectales/patología , Perfilación de la Expresión Génica , Glicoproteínas de Membrana/genética , Lesiones Precancerosas/patología , Pólipos Adenomatosos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Colonoscopía , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Análisis de Componente Principal
18.
Cell Cycle ; 9(22): 4592-9, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21099343

RESUMEN

We have previously shown that the DNA damage-induced G2 arrest is contributed by inhibition of Aurora A (AurA) and that transduction of active AurA into arrested cells allows bypassing the block through reactivation of CDK1. In this study, we investigated the mechanism of DNA damage-induced AurA inhibition. We provide evidence that ionizing radiation (IR) administered in mitosis, a time when AurA protein and enzymatic activity reach peak levels, impairs interaction with the partner TPX2, leading to inactivation of the kinase through dephosphorylation of AurA T-loop residue, T288. We find that decreased AurA-TPX2 complex formation in response to irradiation results from reduced cellular levels of TPX2, an effect that is both contributed by increased APC/CDH1-dependent protein degradation and decreased translation of TPX2 mRNA.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/efectos de la radiación , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Aurora Quinasas , Proteínas de Ciclo Celular/genética , Regulación de la Expresión Génica , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Nucleares/genética , ARN Mensajero/metabolismo , Radiación Ionizante
19.
Gastroenterology ; 129(3): 1047-59, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16143142

RESUMEN

BACKGROUND & AIMS: Colon cancers with defective DNA mismatch repair (MMR) have peculiar molecular, pathologic, and clinical features, including high-level microsatellite instability, conspicuous lymphocytic infiltration, preferential location in the proximal colon, and better prognosis. Our aim was to characterize the transcriptional profile of this colon cancer subset. METHODS: An oligonucleotide microarray containing 12,625 probes was used to evaluate gene expression in 25 proximal colon cancers, 10 samples of normal colon mucosa, and 14 colon cancer cell lines. Transcriptional profiles of MMR-deficient cancers and cell lines were compared with those of their MMR-proficient counterparts. RESULTS: Unsupervised analysis of microarray data showed that MMR status exerts a predominant influence on the gene expression profile of proximal colon cancers. Hierarchical clustering divided the cancers into 2 groups corresponding almost perfectly with their MMR status. Supervised analysis identified numerous gene expression changes that represent a genetic signature of MMR-deficient colon cancers. Changes in genes involved in apoptosis and the immune response were consistent with the better prognosis of MMR-deficient cancers. In MMR-deficient cancers and cell lines, 4-1BBL, a crucial gene in the anti-tumor immune response, was, respectively, 2.4 and 6.0 times more expressed than in their MMR-proficient counterparts. This difference was confirmed by quantitative reverse-transcription polymerase chain reaction and flow cytometric assessment of 4-1BBL protein expression in colon cancer cell lines. Our analysis also showed novel possible gene targets of microsatellite instability. CONCLUSIONS: MMR inactivation produces distinct changes in the cellular messenger RNA pool, which is consistent with a unique tumorigenesis pathway.


Asunto(s)
Disparidad de Par Base , Neoplasias del Colon/genética , Reparación del ADN , Perfilación de la Expresión Génica , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Transcripción Genética , Proteínas Adaptadoras Transductoras de Señales , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Portadoras , Colon/patología , Colon/fisiología , ADN de Neoplasias/genética , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Mucosa Intestinal/patología , Mucosa Intestinal/fisiología , Masculino , Persona de Mediana Edad , Homólogo 1 de la Proteína MutL , Eliminación de Secuencia
20.
Dis Colon Rectum ; 48(3): 567-71, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15719192

RESUMEN

PURPOSE: Familial adenomatous polyposis is an autosomal dominant disease characterized by the presence of 100 or more colorectal adenomatous polyps. Mutations in the adenomatous polyposis coli gene are primarily responsible for the development of this disease. This study was designed to investigation of adenomatous polyposis coli (APC) gene mutations in members of familial adenomatous polyposis family to identify individuals at risk of the disease. METHODS: We examined one patient with familial adenomatous polyposis and 21 family members including one affected person from familial adenomatous polyposis and 20 nonsymptomatic persons. We studied E, D, F, and G segments of exon 15 of the adenomatous polyposis coli gene by heteroduplex analysis. RESULTS: We used silver staining method for staining. We found a mutation for five persons at segment F of exon 15 of the adenomatous polyposis coli gene. Two of them were affected by colorectal cancer, one of whom was the proband, and the other three were nonsymptomatic family members. The pathogenetic mutation was a T deletion at codon 1172, causing a frameshift in the adenomatous polyposis coli gene, as a result of the sequencing analysis of these cases. CONCLUSIONS: Investigation of adenomatous polyposis coli gene mutations is very important for the identification of genetic susceptibility to colorectal cancer and for the definition of tumor developing at an early stage. Furthermore, the identification of this mutation for the first time in a Turkish family will be useful to foster further studies on familial adenomatous polyposis in Turkey.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Genes APC , Predisposición Genética a la Enfermedad , Adulto , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN , Femenino , Análisis Heterodúplex , Humanos , Masculino , Linaje , Turquía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA