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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 135(7): 1167-9, 2008 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-19109887

RESUMO

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.


Assuntos
Citosina/metabolismo , Metilação de DNA , Animais , Citidina Desaminase/metabolismo , Timina DNA Glicosilase/metabolismo , Peixe-Zebra
2.
BMC Cancer ; 20(1): 290, 2020 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-32252665

RESUMO

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.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Epigenoma , Neoplasias Hepáticas/secundário , Neoplasias Colorretais/metabolismo , Epigênese Genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Microdissecção e Captura a Laser/métodos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Fenótipo , Regiões Promotoras Genéticas
3.
Chem Res Toxicol ; 30(3): 769-776, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28076683

RESUMO

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.


Assuntos
Reparo do DNA , Sesquiterpenos/farmacologia , Compostos de Espiro/farmacologia , Transcrição Gênica/efeitos dos fármacos , Ativação Metabólica , Linhagem Celular , Humanos , Sesquiterpenos/farmacocinética , Compostos de Espiro/farmacocinética
4.
BMC Cancer ; 14: 46, 2014 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-24472434

RESUMO

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.


Assuntos
Adenoma/genética , Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Perfilação da Expressão Gênica , Fatores de Transcrição/genética , Adenoma/metabolismo , Adenoma/patologia , Biomarcadores Tumorais/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Análise por Conglomerados , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Genes myb , Humanos , Imuno-Histoquímica , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Survivina , Telomerase/genética , Telomerase/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
5.
Nucleic Acids Res ; 38(22): 8131-40, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20705653

RESUMO

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.


Assuntos
RNA Polimerase II/metabolismo , RecQ Helicases/metabolismo , Transcrição Gênica , Motivos de Aminoácidos , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Sobrevivência Celular , Sequência Conservada , Humanos , Dados de Sequência Molecular , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , RNA Polimerase II/química , RecQ Helicases/química , Sequências Repetitivas de Aminoácidos , Spliceossomos/metabolismo
6.
Epigenetics Chromatin ; 13(1): 25, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32487212

RESUMO

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.


Assuntos
Alelos , Metilação de DNA , Epigenômica/métodos , Análise de Sequência de DNA/métodos , Neoplasias Colorretais/genética , Ilhas de CpG , Feminino , Impressão Genômica , Humanos , Masculino , Software
7.
Mol Cancer ; 8: 124, 2009 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-20015382

RESUMO

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.


Assuntos
Adenocarcinoma/genética , Neoplasias Colorretais/genética , Inativação Gênica , Proteínas Tirosina Fosfatases Classe 7 Semelhantes a Receptores/genética , Adenocarcinoma/patologia , Neoplasias Colorretais/patologia , Metilação de DNA , Humanos , RNA Mensageiro/genética
8.
Mol Cancer Res ; 5(12): 1263-75, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18171984

RESUMO

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.


Assuntos
Adenoma/genética , Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Adenoma/patologia , Idoso , Biomarcadores Tumorais/metabolismo , Transformação Celular Neoplásica/genética , Neoplasias Colorretais/patologia , Feminino , Marcadores Genéticos , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Filogenia , RNA Mensageiro/metabolismo , Transcrição Gênica
9.
Oncol Rep ; 17(6): 1421-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17487400

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/genética , Neoplasias Colorretais/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , O(6)-Metilguanina-DNA Metiltransferase/genética , Idoso , Idoso de 80 Anos ou mais , Carcinoma/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Metilação de DNA , Feminino , Genes ras/genética , Humanos , Mucosa Intestinal/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Sulfitos/química
10.
Cancer Res ; 65(23): 10759-66, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16322221

RESUMO

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.


Assuntos
Pareamento Incorreto de Bases , Proteínas de Transporte/fisiologia , Reparo do DNA/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Baculoviridae/genética , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Humanos , Proteína 1 Homóloga a MutL , Proteínas MutL , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Spodoptera/genética , Spodoptera/metabolismo
11.
Clin Cancer Res ; 11(23): 8332-40, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16322293

RESUMO

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.


Assuntos
Neoplasias Colorretais/genética , Instabilidade Genômica , Repetições de Microssatélites/genética , Proteínas Adaptadoras de Transdução de Sinal , Adenocarcinoma/diagnóstico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma Mucinoso/diagnóstico , Adenocarcinoma Mucinoso/tratamento farmacológico , Adenocarcinoma Mucinoso/genética , Antimetabólitos Antineoplásicos/uso terapêutico , Proteínas de Transporte/genética , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/tratamento farmacológico , Proteínas de Ligação a DNA/genética , Feminino , Fluoruracila/uso terapêutico , Humanos , Masculino , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS/genética , Estadiamento de Neoplasias , Proteínas Nucleares/genética , Prognóstico , Estudos Prospectivos , Taxa de Sobrevida , Resultado do Tratamento
12.
Cancer Res ; 63(23): 8158-66, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14678970

RESUMO

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.


Assuntos
Pareamento Incorreto de Bases/fisiologia , Metilação de DNA/efeitos dos fármacos , Reparo do DNA/fisiologia , Metilnitronitrosoguanidina/toxicidade , Proteínas Adaptadoras de Transdução de Sinal , Linfócitos B/efeitos dos fármacos , Linfócitos B/fisiologia , Proteínas de Transporte , Linhagem Celular , Reparo do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Humanos , Proteína 1 Homóloga a MutL , Mutação de Sentido Incorreto , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/deficiência , Proteínas Nucleares , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/fisiologia , Regulação para Cima/efeitos dos fármacos
13.
Pathol Res Pract ; 200(5): 371-7, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15239345

RESUMO

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.


Assuntos
Adenocarcinoma Mucinoso/patologia , Neoplasias Colorretais/patologia , Mucinas/biossíntese , Proteínas de Neoplasias/biossíntese , Proteínas Adaptadoras de Transdução de Sinal , Adenocarcinoma Mucinoso/metabolismo , Adulto , Idoso , Proteínas de Transporte , Neoplasias Colorretais/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mucina-5AC , Mucina-2 , Proteína 1 Homóloga a MutL , Proteínas Nucleares
14.
Genome Biol ; 15(2): R35, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24517713

RESUMO

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.


Assuntos
Teorema de Bayes , Metilação de DNA/genética , Estudos de Avaliação como Assunto , Sequenciamento de Nucleotídeos em Larga Escala , Ilhas de CpG/genética , Variações do Número de Cópias de DNA , Genoma Humano , Humanos
15.
PLoS One ; 8(7): e69473, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936024

RESUMO

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.


Assuntos
Neoplasias Colorretais/metabolismo , Proteínas/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Forma Celular/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Retroalimentação Fisiológica , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Humanos , Hialuronoglucosaminidase , Invasividade Neoplásica , Ligação Proteica/genética , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Via de Sinalização Wnt/genética
16.
Toxicol In Vitro ; 25(2): 446-53, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21095227

RESUMO

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.


Assuntos
Brônquios/patologia , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Nicotiana/toxicidade , Fumaça/efeitos adversos , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Células Cultivadas , Citoesqueleto/fisiologia , Metilação de DNA , Decitabina , Perfilação da Expressão Gênica , Glucosídeos/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Junções Intercelulares/fisiologia
17.
EMBO Mol Med ; 3(6): 334-47, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21538994

RESUMO

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.


Assuntos
Neoplasias Colorretais/patologia , Perfilação da Expressão Gênica , Glicoproteínas de Membrana/genética , Lesões Pré-Cancerosas/patologia , Pólipos Adenomatosos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Colonoscopia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Feminino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , Análise de Componente Principal
18.
Cell Cycle ; 9(22): 4592-9, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21099343

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose/efeitos da radiação , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Aurora Quinases , Proteínas de Ciclo Celular/genética , Regulação da Expressão Gênica , Células HeLa , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Nucleares/genética , RNA Mensageiro/metabolismo , Radiação Ionizante
19.
Gastroenterology ; 129(3): 1047-59, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16143142

RESUMO

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.


Assuntos
Pareamento Incorreto de Bases , Neoplasias do Colo/genética , Reparo do DNA , Perfilação da Expressão Gênica , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Transcrição Gênica , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas de Transporte , Colo/patologia , Colo/fisiologia , DNA de Neoplasias/genética , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiologia , Masculino , Pessoa de Meia-Idade , Proteína 1 Homóloga a MutL , Deleção de Sequência
20.
Gastroenterology ; 128(5): 1160-71, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15887099

RESUMO

BACKGROUND & AIMS: Germline mutations in the DNA mismatch repair (MMR) genes MSH2, MSH6, or MLH1 predispose to colorectal cancer (CRC) with an autosomal dominant inheritance pattern. The protein encoded by PMS2 is also essential for MMR; however, alterations in this gene have been documented only in extremely rare cases. We addressed this unexpected finding by analyzing a large series of CRCs. METHODS: Expression of MSH2, MSH6, MLH1, and PMS2 was studied by immunohistochemistry in 1048 unselected, consecutive CRCs. Where absence of MMR proteins was detected, microsatellite instability and cytosine methylation of the respective gene promoter were analyzed. The DNA of patients presenting with PMS2-deficient cancers was examined for germline and somatic alterations in the PMS2 gene. RESULTS: An aberrant pattern of MMR protein expression was detected in 13.2% of CRCs. Loss of expression of MSH2, MSH6, or MLH1 was found in 1.4%, 0.5%, and 9.8%, respectively. PMS2 deficiency accompanied by microsatellite instability was found in 16 cases (1.5%) with a weak family history of cancer. The PMS2 promoter was not hypermethylated in these cases. Despite interference of the PMS2 pseudogenes, we identified several heterozygous germline mutations in the PMS2 gene. CONCLUSIONS: PMS2 defects account for a small but significant proportion of CRCs and for a substantial fraction of tumors with microsatellite instability. However, the penetrance of heterozygous germline mutations in PMS2 is considerably lower than that of mutations in other MMR genes. The possible underlying causes of this unorthodox inheritance pattern are discussed.


Assuntos
Adenoma/genética , Adenoma/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas de Transporte , Metilação de DNA , Feminino , Mutação em Linhagem Germinativa , Heterozigoto , Humanos , Imuno-Histoquímica , Masculino , Repetições de Microssatélites , Pessoa de Meia-Idade , Endonuclease PMS2 de Reparo de Erro de Pareamento , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA