Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
2.
Oncogene ; 26(18): 2621-34, 2007 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-17043644

RESUMO

Lung cancer is the leading cause of cancer-related deaths in the United States due, in large part, to the lack of early detection methods. Lung cancer arises from a complex series of genetic and epigenetic changes leading to uncontrolled cell growth and metastasis. Unlike genetic changes, epigenetic changes, such as DNA methylation and histone acetylation, are reversible with currently available pharmaceuticals and are early events in lung tumorigenesis detectable by non-invasive methods. In order to better understand how epigenetic changes contribute to lung cancer, and to identify new disease biomarkers, we combined pharmacologic inhibition of DNA methylation and histone deacetylation in non-small cell lung cancer (NSCLC) cell lines, with genome-wide expression profiling. Of the more than 200 genes upregulated by these treatments, three of these, neuronatin, metallothionein 3 and cystatin E/M, were frequently hypermethylated and transcriptionally downregulated in NSCLC cell lines and tumors. Interestingly, four other genes, cylindromatosis, CD9, activating transcription factor 3 and oxytocin receptor, were dominantly regulated by histone deacetylation and were also frequently downregulated in lung tumors. The majority of these genes also suppressed NSCLC growth in culture when ectopically expressed. This study therefore reveals new putative NSCLC growth regulatory genes and epigenetic disease biomarkers that may enhance early detection strategies and serve as therapeutic targets.


Assuntos
Azacitidina/farmacologia , Carcinoma Pulmonar de Células não Pequenas/genética , Metilação de DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Epigênese Genética/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ácidos Hidroxâmicos/farmacologia , Acetilação , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Azacitidina/análogos & derivados , Biomarcadores Tumorais , Carcinoma de Células Grandes/tratamento farmacológico , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/patologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Imunoprecipitação da Cromatina , Ensaio de Unidades Formadoras de Colônias , Inibidores de Histona Desacetilases , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
3.
Oncogene ; 20(24): 3139-55, 2001 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-11420731

RESUMO

The field of epigenetics has recently moved to the forefront of studies relating to diverse processes such as transcriptional regulation, chromatin structure, genome integrity, and tumorigenesis. Recent work has revealed how DNA methylation and chromatin structure are linked at the molecular level and how methylation anomalies play a direct causal role in tumorigenesis and genetic disease. Much new information has also come to light regarding the cellular methylation machinery, known as the DNA methyltransferases, in terms of their roles in mammalian development and the types of proteins they are known to interact with. This information has forced a new view for the role of DNA methyltransferases. Rather than enzymes that act in isolation to copy methylation patterns after replication, the types of interactions discovered thus far indicate that DNA methyltransferases may be components of larger complexes actively involved in transcriptional control and chromatin structure modulation. These new findings will likely enhance our understanding of the myriad roles of DNA methylation in disease as well as point the way to novel therapies to prevent or repair these defects.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Neoplasias/genética , Animais , DNA (Citosina-5-)-Metiltransferases/genética , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Humanos , Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/patologia
4.
Cancer Res ; 60(17): 4907-12, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10987305

RESUMO

A DNA fragment frequently hypermethylated in tumor cells was isolated using a novel screening strategy termed methylation-sensitive arbitrarily primed PCR. The isolated sequence corresponded to a CpG island at the 5' end of a previously unknown gene, TPEF (transmembrane protein containing epidermal growth factor and follistatin domains). Expression of TPEF was observed using Northern master blot analysis of a variety of normal tissues including colon, bladder, and prostate tissue. TPEF maps to human chromosome 2q33, where frequent loss of heterozygosity is seen in various human tumors, and TPEF was not expressed in most human colon and various other tumor cell lines examined by reverse transcription-PCR. Nine of 11 tumor cell lines were highly methylated in the 5' region and the first exon of the gene that demonstrated features characteristic of a CpG island. However the other two cell lines, which expressed TPEF, were hypomethylated in the 5' end of the gene. The region was also hypermethylated in 11 of 16 primary bladder tumors and in 3 of 4 primary colon tumors when compared with adjacent normal tissue. Our results suggest that potential tumor suppressor genes can be isolated from human tumors by virtue of their altered methylation patterns.


Assuntos
Metilação de DNA , Fator de Crescimento Epidérmico/genética , Glicoproteínas/genética , Proteínas de Membrana/genética , Proteínas de Neoplasias , Neoplasias/genética , Sequência de Aminoácidos , Sequência de Bases , Cromossomos Humanos Par 2/genética , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Ilhas de CpG/genética , Ilhas de CpG/fisiologia , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Fator de Crescimento Epidérmico/biossíntese , Folistatina , Regulação Neoplásica da Expressão Gênica/fisiologia , Inativação Gênica/fisiologia , Glicoproteínas/biossíntese , Humanos , Proteínas de Membrana/biossíntese , Dados de Sequência Molecular , Neoplasias/metabolismo , Estrutura Terciária de Proteína/genética , Homologia de Sequência de Aminoácidos , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/metabolismo
5.
Nat Genet ; 25(3): 338-42, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10888886

RESUMO

Methylation of CpG islands is associated with transcriptional silencing and the formation of nuclease-resistant chromatin structures enriched in hypoacetylated histones. Methyl-CpG-binding proteins, such as MeCP2, provide a link between methylated DNA and hypoacetylated histones by recruiting histone deacetylase, but the mechanisms establishing the methylation patterns themselves are unknown. Whether DNA methylation is always causal for the assembly of repressive chromatin or whether features of transcriptionally silent chromatin might target methyltransferase remains unresolved. Mammalian DNA methyltransferases show little sequence specificity in vitro, yet methylation can be targeted in vivo within chromosomes to repetitive elements, centromeres and imprinted loci. This targeting is frequently disrupted in tumour cells, resulting in the improper silencing of tumour-suppressor genes associated with CpG islands. Here we show that the predominant mammalian DNA methyltransferase, DNMT1, co-purifies with the retinoblastoma (Rb) tumour suppressor gene product, E2F1, and HDAC1 and that DNMT1 cooperates with Rb to repress transcription from promoters containing E2F-binding sites. These results establish a link between DNA methylation, histone deacetylase and sequence-specific DNA binding activity, as well as a growth-regulatory pathway that is disrupted in nearly all cancer cells.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , DNA (Citosina-5-)-Metiltransferases/metabolismo , Proteínas de Ligação a DNA , Histona Desacetilases/metabolismo , Regiões Promotoras Genéticas , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Células 3T3 , Animais , Inibidor p16 de Quinase Dependente de Ciclina/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Células HeLa , Histona Desacetilase 1 , Histona Desacetilases/genética , Humanos , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Elementos de Resposta , Proteína do Retinoblastoma/genética , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/genética
7.
Nucleic Acids Res ; 28(10): 2108-13, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10773079

RESUMO

DNA methylation is essential for mammalian development, X-chromosome inactivation, and imprinting yet aberrant methylation patterns are one of the most common features of transformed cells. One of the proposed causes for these defects in the methylation machinery is overexpression of one or more of the three known catalytically active DNA methyltransferases (DNMTs) 1, 3a and 3b, yet there are clearly examples in which overexpression is minimal or non-existent but global methylation anomalies persist. An alternative mechanism which could give rise to global methylation errors is the improper expression of one or more of the DNMTs during the cell cycle. To begin to study the latter possibility we examined the expression of the mRNAs for DNMT1, 3a and 3b during the cell cycle of normal and transformed cells. We found that DNMT1 and 3b levels were significantly downregulated in G(0)/G(1)while DNMT3a mRNA levels were less sensitive to cell cycle alterations and were maintained at a slightly higher level in tumor lines compared to normal cell strains. Enzymatic activity assays revealed a similar decrease in the overall methylation capacity of the cells during G(0)/G(1)arrest and again revealed that a tumor cell line maintained a higher methylation capacity during arrest than a normal cell strain. These results reveal a new level of control exerted over the cellular DNA methylation machinery, the loss of which provides an alternative mechanism for the genesis of the aberrant methylation patterns observed in tumor cells.


Assuntos
Ciclo Celular , DNA (Citosina-5-)-Metiltransferases/genética , Regulação Enzimológica da Expressão Gênica , RNA Mensageiro/genética , Transcrição Gênica , Sequência de Bases , Neoplasias da Mama , Linhagem Celular , DNA (Citosina-5-)-Metiltransferase 1 , DNA Metiltransferase 3A , Feminino , Fase G1 , Humanos , Cinética , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Fase de Repouso do Ciclo Celular , Fase S , Células Tumorais Cultivadas , Bexiga Urinária , Neoplasias da Bexiga Urinária , DNA Metiltransferase 3B
8.
Carcinogenesis ; 21(3): 461-7, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10688866

RESUMO

DNA methylation, or the covalent addition of a methyl group to cytosine within the context of the CpG dinucleotide, has profound effects on the mammalian genome. These effects include transcriptional repression via inhibition of transcription factor binding or the recruitment of methyl-binding proteins and their associated chromatin remodeling factors, X chromosome inactivation, imprinting and the suppression of parasitic DNA sequences. DNA methylation is also essential for proper embryonic development; however, its presence can add an additional burden to the genome. Normal methylation patterns are frequently disrupted in tumor cells with global hypomethylation accompanying region-specific hypermethylation. When these hypermethylation events occur within the promoter of a tumor suppressor gene they will silence the gene and provide the cell with a growth advantage in a manner akin to deletions or mutations. Recent work indicating that DNA methylation is an important player in both DNA repair and genome stability as well as the discovery of a new family of DNA methyltransferases makes now a very exciting period for the methylation field. This review will highlight the major findings in the methylation field over the past 20 years then summarize the most important and interesting future directions the field is likely to take in the next millennium.


Assuntos
Metilação de DNA , Neoplasias/genética , 5-Metilcitosina , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/farmacologia , Carcinógenos/metabolismo , Cromatina/química , Citosina/análogos & derivados , Citosina/metabolismo , Reparo do DNA , Previsões , Expressão Gênica , Humanos
9.
Nat Rev Genet ; 1(1): 11-9, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11262868

RESUMO

DNA methylation has recently moved to centre stage in the aetiology of human neurodevelopmental syndromes such as the fragile X, ICF and Rett syndromes. These diseases result from the misregulation of genes that occurs with the loss of appropriate epigenetic controls during neuronal development. Recent advances have connected DNA methylation to chromatin-remodelling enzymes, and understanding this link will be central to the design of new therapeutic tools.


Assuntos
Metilação de DNA , Doenças Genéticas Inatas/genética , Genoma , Humanos
10.
Semin Cancer Biol ; 9(5): 369-75, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10547345

RESUMO

EBV is a ubiquitous herpesvirus associated with a variety of lymphoid and epithelial tumors. In healthy lymphocytes and in tumors immune surveillance is evaded by suppression of a family of immunodominant viral antigens. Methylation of a viral promoter plays a crucial role in this suppression. Methylation of the viral genome in the latent state over evolutionary time is believed to account for CpG suppression that distinguishes this virus from most other large DNA viruses. Pharmacologic manipulation of methylation may offer an opportunity to unmask viral antigens and expose tumors to immune surveillance.


Assuntos
Metilação de DNA , Herpesvirus Humano 4/genética , Regulação Viral da Expressão Gênica/genética , Herpesvirus Humano 4/fisiologia , Humanos
11.
Mol Cell Biol ; 19(10): 6690-8, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10490608

RESUMO

De novo methylation of CpG islands within the promoters of eukaryotic genes is often associated with their transcriptional repression, yet the methylation of CpG islands located downstream of promoters does not block transcription. We investigated the kinetics of mRNA induction, demethylation, and remethylation of the p16 promoter and second-exon CpG islands in T24 cells after 5-aza-2'-deoxycytidine (5-Aza-CdR) treatment to explore the relationship between CpG island methylation and gene transcription. The rates of remethylation of both CpG islands were associated with time but not with the rate of cell division, and remethylation of the p16 exon 2 CpG island occurred at a higher rate than that of the p16 promoter. We also examined the relationship between the remethylation of coding sequence CpG islands and gene transcription. The kinetics of remethylation of the p16 exon 2, PAX-6 exon 5, c-ABL exon 11, and MYF-3 exon 3 loci were examined following 5-Aza-CdR treatment because these genes contain exonic CpG islands which are hypermethylated in T24 cells. Remethylation occurred most rapidly in the p16, PAX-6, and c-ABL genes, shown to be transcribed prior to drug treatment. These regions also exhibited higher levels of remethylation in single-cell clones and subclones derived from 5-Aza-CdR-treated T24 cells. Our data suggest that de novo methylation is not restricted to the S phase of the cell cycle and that transcription through CpG islands does not inhibit their remethylation.


Assuntos
Ilhas de CpG , Metilação de DNA , Proteínas de Homeodomínio , Transcrição Gênica , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Divisão Celular , Inibidor p16 de Quinase Dependente de Ciclina/genética , Metilação de DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Decitabina , Éxons , Proteínas do Olho , Regulação da Expressão Gênica , Modelos Genéticos , Proteína MyoD/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Repressoras , Células Tumorais Cultivadas
12.
Am J Pathol ; 155(2): 619-25, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10433954

RESUMO

The Epstein-Barr virus (EBV) major latent promoter C drives the expression of viral nuclear proteins important in lymphocyte immortalization and as targets for immune surveillance by cytotoxic T cells. Hypermethylation of the C promoter silences its transcription. This promoter is methylated and silent in Burkitt's lymphoma, Hodgkin's disease, nasopharyngeal carcinoma, and nasal lymphoma. However, it is never methylated in the EBV-immortalized lymphoblastoid cell lines that serve as a model for EBV-associated lymphoproliferative disease. We have analyzed C promoter methylation in iatrogenic EBV-associated B-cell lymphoproliferative disease, mainly posttransplant lymphoma, using a sensitive polymerase chain reaction-based C promoter methylation assay. Our results showed heterogeneity in lymphoproliferative disease with methylation of viral DNA in specimens from 3 of 13 patients. In specimens from two of these patients, only methylated viral DNA was detected and viral nuclear antigen expression was correspondingly restricted. Heterogeneity in C promoter methylation and expression of associated transcripts may be an important determinant of the growth properties of lymphoproliferative lesions and may provide an explanation for the failure of some tumors to respond to withdrawal or reduction of immunosuppressive therapy.


Assuntos
Herpesvirus Humano 4/genética , Linfoma de Células B/etiologia , Linfoma de Células B/virologia , Reação em Cadeia da Polimerase/métodos , Regiões Promotoras Genéticas , Linfoma de Burkitt/virologia , Carcinoma/virologia , Linhagem Celular , Doença de Hodgkin/virologia , Humanos , Terapia de Imunossupressão/efeitos adversos , Linfoma/virologia , Metilação , Modelos Genéticos , Neoplasias Nasofaríngeas/virologia , Células Tumorais Cultivadas
13.
Oncogene ; 18(26): 3810-20, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10445844

RESUMO

The INK4a/ARF locus on human chromosome 9p resides at the nexus of two critical cell cycle regulatory pathways, the p53 pathway and the retinoblastoma (pRb) gene pathway. Through the use of shared coding regions and alternative reading frames two distinct proteins are produced: INK4a is a cyclin-dependent kinase inhibitor whereas ARF binds the MDM2 proto-oncogene and stabilizes p53. We have examined the expression patterns of the INK4a/ARF locus at the RNA level in normal human and murine tissues to determine if these genes are coordinately regulated. We found that both INK4a and ARF were expressed in most tissues at low levels detectable only by RT-PCR. The pancreas was an exception in that it expressed no detectable ARF mRNA but expressed high levels of INK4a mRNA. Furthermore, human pancreas expressed an additional previously unrecognized splice variant of INK4a, termed p12, through the use of an alternative splice donor site within intron 1. The p12 transcript produced a 12 kD protein composed of INK4a exon 1alpha and a novel intron-derived C-terminus. This novel protein did not interact with cdk4 but was capable of suppressing growth in a pRb-independent manner. The implications of the capacity of the INK4a/ARF locus to encode a third transcript, and for pancreatic cancer, in which the INK4a/ARF locus is nearly always altered, are considered.


Assuntos
Ciclo Celular/genética , Cromossomos Humanos Par 9/genética , Genes p16 , Splicing de RNA , RNA Mensageiro/metabolismo , Fases de Leitura/genética , Sequências Reguladoras de Ácido Nucleico , Animais , Sequência de Bases , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/imunologia , Linhagem Celular , Clonagem Molecular , DNA Complementar/genética , Feminino , Regulação da Expressão Gênica , Genes do Retinoblastoma , Genes p53 , Humanos , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , Pâncreas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Regiões Promotoras Genéticas , Proto-Oncogene Mas , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Deleção de Sequência , Homologia de Sequência do Ácido Nucleico , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/patologia
14.
Nucleic Acids Res ; 27(11): 2291-8, 1999 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10325416

RESUMO

DNA methylation in mammals is required for embryonic development, X chromosome inactivation and imprinting. Previous studies have shown that methylation patterns become abnormal in malignant cells and may contribute to tumorigenesis by improper de novo methylation and silencing of the promoters for growth-regulatory genes. RNA and protein levels of the DNA methyltransferase DNMT1 have been shown to be elevated in tumors, however murine stem cells lacking Dnmt1 are still able to de novo methylate viral DNA. The recent cloning of a new family of DNA methyltransferases (Dnmt3a and Dnmt3b) in mouse which methylate hemimethylated and unmethylated templates with equal efficiencies make them candidates for the long sought de novo methyltransferases. We have investigated the expression of human DNMT1, 3a and 3b and found widespread, coordinate expression of all three transcripts in most normal tissues. Chromosomal mapping placed DNMT3a on chromosome 2p23 and DNMT3b on chromosome 20q11.2. Significant overexpression of DNMT3b was seen in tumors while DNMT1 and DNMT3a were only modestly over-expressed and with lower frequency. Lastly, several novel alternatively spliced forms of DNMT3b, which may have altered enzymatic activity, were found to be expressed in a tissue-specific manner.


Assuntos
Neoplasias do Colo/enzimologia , DNA (Citosina-5-)-Metiltransferases/genética , Neoplasias Renais/enzimologia , Neoplasias Pancreáticas/enzimologia , Neoplasias da Bexiga Urinária/enzimologia , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Mapeamento Cromossômico , Cromossomos Humanos Par 2 , Cromossomos Humanos Par 20 , Neoplasias do Colo/genética , DNA Metiltransferase 3A , DNA Complementar , Expressão Gênica , Humanos , Neoplasias Renais/genética , Camundongos , Dados de Sequência Molecular , Neoplasias Pancreáticas/genética , RNA Mensageiro , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Neoplasias da Bexiga Urinária/genética , DNA Metiltransferase 3B
15.
Mol Cell Biol ; 18(11): 6457-73, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9774662

RESUMO

The INK4a/ARF locus encodes two proteins involved in tumor suppression in a manner virtually unique in mammalian cells. Distinct first exons, driven from separate promoters, splice onto a common exon 2 and 3 but utilize different reading frames to produce two completely distinct proteins, both of which play roles in cell cycle control. INK4a, a critical element of the retinoblastoma gene pathway, binds to and inhibits the activities of CDK4 and CDK6, while ARF, a critical element of the p53 pathway, increases the level of functional p53 via interaction with MDM2. Here we clone and characterize the promoter of the human ARF gene and show that it is a CpG island characteristic of a housekeeping gene which contains numerous Sp1 sites. Both ARF and INK4a are coordinately expressed in cells except when their promoter regions become de novo methylated. In one of these situations, ARF transcription could be reactivated by treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine, and the reactivation kinetics of ARF and INK4a were found to differ slightly in a cell line in which both genes were silenced by methylation. The ARF promoter was also found to be highly responsive to E2F1 expression, in keeping with previous results at the RNA level. Lastly, transcription from the ARF promoter was down-regulated by wild-type p53 expression, and the magnitude of the effect correlated with the status of the endogenous p53 gene. This finding points to the existence of an autoregulatory feedback loop between p53, MDM2, and ARF, aimed at keeping p53 levels in check.


Assuntos
Ciclo Celular/fisiologia , Ilhas de CpG/genética , Metilação de DNA , Genes Supressores de Tumor/genética , Genes p53/genética , Regiões Promotoras Genéticas/genética , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Inibidor p16 de Quinase Dependente de Ciclina , Regulação para Baixo/fisiologia , Regulação da Expressão Gênica/genética , Genes Reporter/genética , Humanos , Dados de Sequência Molecular , Análise de Sequência de DNA , Deleção de Sequência/genética , Transfecção/genética , Regulação para Cima/fisiologia
16.
J Virol ; 72(9): 7075-83, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9696800

RESUMO

Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1) is indispensable for viral DNA replication and episome maintenance in latency. Four promoters, Cp, Wp, Qp, and Fp, are known to drive EBNA1 expression. Here we show that the TATA-less Qp is constitutively active in a variety of EBV-positive [EBV(+)] tumors and cell lines, irrespective of the activities of other EBNA1 promoters, the type of viral latency, and the cell type. The transcription of highly regulated promoters such as the EBV Cp is known to be directly regulated by CpG methylation. To characterize the role of CpG methylation in the regulation of the constitutively active Qp, we performed bisulfite genomic sequencing and functional analyses using a methylation cassette transcriptional reporter assay. Twenty consecutive CpG sites (16 proximal to the Qp initiation site and 4 upstream of the adjacent Fp initiation site) were studied by bisulfite sequencing of DNA extracted from EBV(+) tumors and cell lines. Eighteen EBV(+) tumors of lymphoid (B, T, and NK cell) or epithelial origin and five Burkitt's lymphoma cell lines were studied. The 16 CpG sites proximal to Qp were virtually all unmethylated, but the 4 CpG sites upstream of the Fp initiation site were variably methylated. The methylation cassette assay showed that in vitro methylation of the Qp cassette (-172 to +32) resulted in strong repression of Qp activity in transient transfection. Thus, Qp is susceptible to repression by methylation but was found to be consistently hypomethylated and expressed in all tumors and tumor-derived cell lines studied.


Assuntos
Metilação de DNA , DNA Viral , Antígenos Nucleares do Vírus Epstein-Barr/genética , Herpesvirus Humano 4/genética , Regiões Promotoras Genéticas , Sequência de Bases , Linhagem Celular Transformada , Sequência Conservada , Humanos , Dados de Sequência Molecular , Transcrição Gênica , Células Tumorais Cultivadas
17.
Blood ; 91(4): 1373-81, 1998 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9454768

RESUMO

Many aspects of Epstein-Barr virus (EBV) and tumor biology have been studied in Burkitt's lymphoma (BL)-derived cell lines. However, in tissue culture, patterns of gene expression and CpG [corrected] methylation often change and viral strain selection may occur. In this report, 10 cases of snap-frozen endemic BL tumors are characterized in terms of viral gene expression, promoter usage, methylation, and viral strain. EBNA1 and BamHI-A rightward transcripts (BART) were detected in 7 of 7 and LMP2A transcripts in 5 of 7 tumors with well-preserved RNA. Transcripts for the other EBNAs and for LMP1 were not detected in any tumor. These tumors differ from BL cell lines in that they lack a variety of lytic cycle transcripts. This pattern of viral gene expression in endemic BL is similar to that reported in peripheral blood mononuclear cells (PBMCs) from healthy EBV-seropositive individuals. EBNA1 transcripts originated from the Q promoter (Qp) but not C, W, or F promoters that drive transcription of EBNA1 in other circumstances. Whereas Cp has been previously shown to be entirely CpG methylated in BL, bisulfite genomic sequencing showed virtually no methylation in Qp. Type-A EBV was detected in 6 of 10 and type B in 4 of 10 cases. A previously reported 30bp deletion variant in the carboxyl terminal of LMP1 gene was detected in 5 of 10 cases. The association with both A and B strains contrasts with EBV-associated Hodgkin's disease, nasopharyngeal carcinoma, and post-transplant lymphoproliferative disease, which are much more consistently associated with A strain virus.


Assuntos
Linfoma de Burkitt/virologia , Genes Virais , Herpesvirus Humano 4/isolamento & purificação , Primers do DNA , Herpesvirus Humano 4/genética , Humanos , Reação em Cadeia da Polimerase
19.
Blood ; 90(11): 4480-4, 1997 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9373258

RESUMO

Epstein-Barr virus (EBV) latent infection in B cells persists over years or decades despite a sustained cytotoxic immune response to viral antigens. We present data that methylated EBV DNA can be detected in the normal lymphocytes of healthy volunteers. Whereas methylation of foreign DNA has been recognized as a potential cellular defense mechanism, methylation of EBV DNA may be an essential part of the virus life cycle in vivo, explaining the persistence of virus-infected B cells in the face of immune surveillance. Methylation of the C promoter helps to prevent expression of the immunodominant antigens expressed from this promoter. First recognized in tumors, methylation-associated evasion of immune surveillance is not an aberration restricted to tumor tissue but is detected in normal EBV-infected lymphocytes. Methylation of the viral genome in latency also provides an explanation for the CpG suppression associated with EBV but not other large DNA viruses.


Assuntos
Metilação de DNA , DNA Viral/metabolismo , Herpesvirus Humano 4/genética , Linfócitos/virologia , Linhagem Celular , Humanos , Linfócitos/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Análise de Sequência de DNA
20.
J Virol ; 71(9): 6445-54, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9261362

RESUMO

Methylation-associated transcriptional repression is recognized in many settings and may play a role in normal differentiation and in tumorigenesis. Both sequence-specific and nonspecific mechanisms have been elaborated. Recently, we have presented evidence that methylation-associated inhibition of the Epstein-Barr virus (EBV) major latency promoter (BamHI C promoter or Cp) in Burkitt's lymphoma and Hodgkin's disease may play an important role in the pathogenesis of these tumors by protecting them from CD8+ cytotoxic T-cell immunosurveillance. The mechanism of transcriptional repression may relate to specific inhibition of the binding of a cellular transcription factor by methylation. To dissect the viral promoter with regard to transcriptional sensitivity to methylation, we have devised an assay that allows the methylation of discrete regions of reporter plasmids. During the course of the assay, methylation patterns appeared to be stable; there was no evidence of either spread or reversal of the imposed methylation pattern. Application of the assay to the 3.8-kb region upstream of the major EBV latency promoter with natural Cp reporter plasmids showed that sensitivity to methylation is not homogeneously distributed but is concentrated in two discrete regions. The first of these methylation-hypersensitive regions (MHRI) is the previously identified EBNA-2 response element, which includes the methylation-sensitive CBF2 binding site. The second (MHRII) is a sequence further downstream whose potential role in methylation-mediated transcriptional repression had been previously unsuspected. In chimeric enhancer/promoter plasmids, methylation of this downstream region was sufficient to virtually abolish simian virus 40 enhancer-driven transcription. Further dissection indicated that methylation of the EBNA-2 response element (MHRI) was sufficient to abolish EBNA-2-mediated Cp activity while methylation of a region including the EBNA-2 response element and downstream sequence (MHRI and MHRII) was sufficient to abolish all Cp-mediated reporter activity, including that driven by the EBNA-1-dependent enhancer in the origin of plasmid replication, oriP.


Assuntos
Mapeamento Cromossômico , Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/genética , Regiões Promotoras Genéticas , Sítios de Ligação , Metilação de DNA , Humanos , Transcrição Gênica , Ativação Transcricional , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA