Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
2.
Cancer Res ; 55(11): 2279-83, 1995 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-7538900

RESUMO

Approximately one third of breast cancers grow independently of estrogen, lack detectable estrogen receptor (ER) protein, and rarely respond to hormonal treatment. Previous studies correlated the lack of ER gene expression in ER-negative breast tumor cells with hypermethylation of a CpG island in the 5' region of the ER gene. In order to determine whether demethylation of the ER gene in the ER-negative human breast cancer cell line MDA-MB-231 could affect ER transcription, cells were treated with two inhibitors of DNA methylation, 5-azacytidine or 5-aza-2'-deoxycytidine. DNA from cells treated with either drug became partially demethylated at several methylation-sensitive restriction enzyme sites, including HhaI, NotI, and SacII, within the ER CpG island. This demethylation correlated with reexpression of the ER gene as detected by reverse transcriptase-PCR and production of ER protein as detected by Western blot analysis. ER produced in drug-treated cells was functionally active as demonstrated by its ability to activate transcription of estrogen-responsive genes. These results suggest that DNA methylation of the ER CpG island may play a role in suppression of ER gene expression in ER-negative breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/ultraestrutura , Regulação Neoplásica da Expressão Gênica/fisiologia , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sequência de Bases , Neoplasias da Mama/metabolismo , Citosina/metabolismo , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Decitabina , Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Guanina/metabolismo , Humanos , Metilação , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Estrogênio/fisiologia , Células Tumorais Cultivadas
3.
Cancer Res ; 61(19): 7025-9, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11585728

RESUMO

Formation of transcriptional repression complexes such as DNA methyltransferase (DNMT) 1/histone deacetylase (HDAC) or methyl-CpG binding protein/HDAC is emerging as an important mechanism in silencing a variety of methylated tissue-specific and imprinted genes. Our previous studies showed that treatment of estrogen receptor (ER)-alpha-negative human breast cancer cells with the DNMT inhibitor 5-aza-2'-deoxycytidine (5-aza-dC) led to ER mRNA and protein re-expression. Also, the HDAC inhibitor trichostatin A (TSA) could induce ER transcript about 5-fold. Here we show that 5-aza-dC alone induced ER transcript about 30-40-fold, and the addition of TSA elevated ER mRNA expression about 10-fold more in the human ER-negative breast cancer cell lines MDA-MB-231 and MDA-MB-435. Overall, the combination of 5-aza-dC and TSA induced a 300-400-fold increase in ER transcript. Restoration of estrogen responsiveness was demonstrated by the ability of the induced ER protein to elicit estrogen response element-regulated reporter activity from an exogenous plasmid as well as induce expression of the ER target gene, progesterone receptor. The synergistic activation of ER occurs concomitantly with markedly reduced soluble DNMT1 expression and activity, partial demethylation of the ER CpG island, and increased acetylation of histones H(3) and H(4). These data suggest that the activities of both DNMT1 and HDAC are key regulators of methylation-mediated ER gene silencing.


Assuntos
Azacitidina/farmacologia , Neoplasias da Mama/enzimologia , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/farmacologia , Receptores de Estrogênio/fisiologia , Acetilação/efeitos dos fármacos , Azacitidina/análogos & derivados , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA/efeitos dos fármacos , Decitabina , Sinergismo Farmacológico , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Fulvestranto , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Humanos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética , Células Tumorais Cultivadas
4.
Cancer Res ; 60(24): 6890-4, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11156387

RESUMO

Recent findings have established a connection between DNA methylation and transcriptionally inactive chromatin characterized by deacetylated histones. Because the absence of estrogen receptor alpha (ERalpha) gene expression has been associated with aberrant methylation of its CpG island in a significant fraction of breast cancers, we tested whether histone deacetylase activity contributes to the transcriptional inactivation of the methylated ER gene in a panel of ER-negative human breast cancer cells. Treatment of these cells with trichostatin A, a specific histone deacetylase inhibitor, led to dose- and time-dependent re-expression of ER mRNA as detected by reverse transcription-PCR without alteration in ERalpha CpG island methylation. Trichostatin A-induced ER re-expression was associated with increased sensitivity to DNase I at the ER locus in MDA-MB-231 cells. These data implicate inactive chromatin mediated by histone deacetylation as a critical component of ER gene silencing in human breast cancer cells. Therefore, histone deacetylation may be a potential target for therapeutic intervention in the treatment of a subset of ER-negative breast cancers.


Assuntos
Neoplasias da Mama/genética , Inibidores de Histona Desacetilases , Receptores de Estrogênio/genética , Ativação Transcricional , Sequência de Bases , Neoplasias da Mama/metabolismo , Cromatina/metabolismo , Ilhas de CpG/genética , Metilação de DNA , Desoxirribonuclease I/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos , Receptor alfa de Estrogênio , Inativação Gênica , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Receptores de Estrogênio/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Células Tumorais Cultivadas
5.
Cancer Res ; 59(21): 5464-70, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10554019

RESUMO

Several methods have been used recently to determine gene expression profiles of cell populations. Here we demonstrate the strength of combining two approaches, serial analysis of gene expression (SAGE) and DNA arrays, to help elucidate pathways in breast cancer progression by finding genes consistently expressed at different levels in primary breast cancers, metastatic breast cancers, and normal mammary epithelial cells. SAGE profiles of 21PT and 21MT, two well-characterized breast tumor cell lines, were compared with SAGE profiles of normal breast epithelial cells to identify differentially expressed genes. A subset of these candidates was then placed on an array and screened with clinical breast tumor samples to find genes and expressed sequence tags that are consistently expressed at different levels in diseased and normal tissues. In addition to finding the predicted overexpression of known breast cancer markers HER-2/neu and MUC-1, the powerful coupling of SAGE and DNA arrays resulted in the identification of genes and potential pathways not implicated previously in breast cancer. Moreover, these techniques also generated information about the differences and similarities of expression profiles in primary and metastatic breast tumors. Thus, combining SAGE and custom array technology allowed for the rapid identification and validation of the clinical relevance of many genes potentially involved in breast cancer progression. These differentially expressed genes may be useful as tumor markers and prognostic indicators and may be suitable targets for various forms of therapeutic intervention.


Assuntos
Neoplasias da Mama/genética , Análise Mutacional de DNA/métodos , Regulação Neoplásica da Expressão Gênica/genética , Análise de Sequência com Séries de Oligonucleotídeos , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , DNA Complementar/análise , Feminino , Biblioteca Gênica , Humanos , RNA Mensageiro/análise , Transcrição Gênica , Células Tumorais Cultivadas
6.
Oncogene ; 17(5): 577-83, 1998 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-9704923

RESUMO

Progesterone receptor (PR) is an estrogen-stimulated gene which has a CpG island that is heavily methylated in a significant fraction of estrogen receptor (ER)-negative/PR-negative human breast cancers and cell lines, including MDA-MB-231 cells. Treatment of MDA-MB-231 cells with the demethylating agent, 5-aza-2'-deoxycytidine (deoxyC) led to demethylation and expression of ER and PR. However, simultaneous treatment with antiestrogen prevented PR transcription, suggesting that demethylation of PR alone is not sufficient to reactivate the PR gene. To examine the effects of ER on the methylation status of the PR CpG island, we stably transfected MDA-MB-231 cells with an inducible expression vector for ER. Surprisingly, in two cell clones, we found that induction of PR gene expression by ligand-bound ER does not require demethylation of the PR CpG island. In contrast, induction of PR transcription was inhibited by blocking the interaction of ER with SRC-1A, a coactivator of ER function. For the first time, we show that a transcription factor with the potential to remodel heterochromatin can activate gene expression without altering the methylation status of the CpG island. These results raise the possibility that demethylation and histone acetylation are distinct but complementary mechanisms for destabilizing heterochromatin and activating transcription.


Assuntos
Ilhas de CpG , Metilação de DNA , Regulação da Expressão Gênica , Receptores de Progesterona/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Fulvestranto , Histona Acetiltransferases , Humanos , Coativador 1 de Receptor Nuclear , Receptores de Estrogênio/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
7.
Oncogene ; 19(12): 1556-63, 2000 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-10734315

RESUMO

Retinoic acid (RA)-resistance in breast cancer cells has been associated with irreversible loss of retinoic acid receptor beta, RARbeta, gene expression. Search of the causes affecting RARbeta gene activity has been oriented at identifying possible differences either at the level of one of the RARbeta promoters, RARbeta2, or at regulatory factors. We hypothesized that loss of RARbeta2 activity occurs as a result of multiple factors, including epigenetic modifications, which can pattern RARbeta2 chromatin state. Using methylation-specific PCR, we found hypermethylation at RARbeta2 in a significant proportion of both breast cancer cell lines and primary breast tumors. Treatment of cells with a methylated RARbeta2 promoter, by means of the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine (5-Aza-CdR), led to demethylation within RARbeta2 and expression of RARbeta indicating that DNA methylation is at least one factor, contributing to RARbeta inactivity. However, identically methylated promoters can differentially respond to RA, suggesting that RARbeta2 activity may be associated to different repressive chromatin states. This supposition is supported by the finding that the more stable repressive RARbeta2 state in the RA-resistant MDA-MB-231 cell line can be alleviated by the HDAC inhibitor, trichostatin A (TSA), with restoration of RA-induced RARbeta transcription. Thus, chromatin-remodeling drugs might provide a strategy to restore RARbeta activity, and help to overcome the hurdle of RA-resistance in breast cancer.


Assuntos
Neoplasias da Mama/genética , Cromatina/genética , Receptores do Ácido Retinoico/genética , Antimetabólitos Antineoplásicos , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Mama/citologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular , Ilhas de CpG , Metilação de DNA , Decitabina , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases , Humanos , Ácidos Hidroxâmicos/farmacologia , Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Receptores do Ácido Retinoico/efeitos dos fármacos , Receptores do Ácido Retinoico/metabolismo , Tretinoína/farmacologia , Células Tumorais Cultivadas
8.
Oncogene ; 18(52): 7453-61, 1999 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-10602504

RESUMO

Estrogen receptor (ER)-negative breast cancer cells display extensive methylation of the ER gene CpG island and elevated DNA methyltransferase (DMT) expression compared to ER-positive cells. The present study demonstrates that DMT protein levels tightly correlate with S phase fraction in ER-positive cells, whereas ER-negative cells express DMT throughout the cell cycle. In addition, levels of p21CIP1, which disrupts DMT binding to PCNA, are inversely correlated with DMT levels. Therefore increased DMT expression in ER-negative cells is not simply due to elevated S-phase fraction, but rather to more complex changes that allow cells to escape normal cell cycle-dependent controls on DMT expression. Because ER-negative breast tumors often have activated growth factor pathways, the impact of these pathways on DMT expression was examined in ER-positive cells. Stable transfection with fibroblast growth factors (FGFs) 1 and 4 led to increased DMT expression that could not be accounted for by a shift in S phase fraction. Elevated DMT protein expression in FGF-transfectants was accompanied by a significant decrease in p21, again suggesting a reciprocal relationship between these two proteins. However, acquisition of an estrogen-independent phenotype, even in conjunction with elevated DMT levels, was not sufficient to promote ER gene silencing via methylation. These results indicate that multiple steps are required for de novo methylation of the ER CpG island.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Receptores de Estrogênio/metabolismo , Sequência de Aminoácidos , Ilhas de CpG , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , DNA Antissenso/genética , Feminino , Humanos , Dados de Sequência Molecular , Receptores de Estrogênio/genética , Fase S/fisiologia , Transfecção , Células Tumorais Cultivadas
9.
Clin Cancer Res ; 2(5): 805-10, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-9816234

RESUMO

Hormonal factors have a profound influence on the development, treatment, and outcome of breast cancer. The absence of steroid hormone receptors is highly correlated with resistance to antihormonal treatments. Work in cultured human breast cancer cell lines has shown that the absence of estrogen receptor (ER) gene expression in ER- cells is associated with extensive methylation of the ER gene 5' CpG island, and treatment with agents that demethylate the ER gene CpG island results in the production of functional ER protein. The current study shows that CpG islands in the 5' region of the ER and progesterone receptor (PR) genes are methylated in a significant fraction of primary human breast cancer tissues. The ER CpG island is methylated at the methylation-sensitive NotI restriction site in 9 of 39 (25%) of primary ER- breast cancers but remains unmethylated in 53 ER+ breast cancers and 9 normal breast specimens. Three methylation-sensitive restriction sites in the PR gene CpG island are not methylated in normal breast specimens and PR+ human breast cancers but are hypermethylated in 40% of PR- human breast tumors. These data demonstrate that methylation of the ER and PR gene CpG islands is associated with the lack of ER and PR gene expression in a significant fraction of human breast cancers.


Assuntos
Neoplasias da Mama/genética , Ilhas de CpG , Metilação de DNA , Receptores de Estrogênio/genética , Receptores de Progesterona/genética , Feminino , Expressão Gênica , Humanos , Células Tumorais Cultivadas
12.
J Virol ; 68(7): 4274-86, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8207802

RESUMO

BK virus is a human papovavirus that latently infects a majority of the world's population. There are more than 30 strains of the virus, most of which differ in the structure of the early enhancer region. The enhancer of the progenitor strain, WW, from which the other strains can be derived, consists of four conserved DNA domains, P, Q, R, and S. Rearrangement of the enhancer occurs upon passage in tissue culture and is thought to occur during virus replication. The strain under study, PQ, was selected upon passage of the Gardner strain (PPPQS) in the permissive cell line, Vero. Mutational analysis of the entire enhancer region demonstrates the importance of five cis-acting sequences: DNA sites B, C, and F, which have homology to the NF-1 protein binding sequence; one purine-rich motif designated A; and site D, which is similar to an SP-1 protein binding site. Two sites, B and C, appear to have a negative influence on gene activity. To study the functional interactions in more detail, promoter-enhancer constructions that contain different combinations of the five DNA sites linked to the chloramphenicol acetyltransferase gene were tested for early gene activity. The results reveal that the proteins binding to the enhancer functionally cooperate with each other. The effects of making mutations at the DNA sites are very similar to the effects of using excess enhancer DNA sequences to titrate the proteins that bind to the cis-acting DNA sites (in vivo competition). Moreover, the effects of changing the spacing between the DNA sites also demonstrate that there are cooperative interactions among the proteins that bind to the PQ strain enhancer. DNA sites B, C, and F are clearly protected from DNase I digestion by Vero cell nuclear proteins. In addition, mutation of each DNA site alters its sensitivity to DNase I in the presence of Vero cell proteins. Interestingly, mutation of site B affects protein binding to site B as well as to sites A, C, D, and F. These results suggest that cooperative functional and physical interactions occur at the early enhancer of the PQ strain.


Assuntos
Vírus BK/genética , Elementos Facilitadores Genéticos , Genes Virais , Animais , Sequência de Bases , Chlorocebus aethiops , DNA Viral/metabolismo , Desoxirribonuclease I , Expressão Gênica , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Fatores de Transcrição/metabolismo , Transcrição Gênica , Células Vero
13.
Crit Rev Oncog ; 8(1): 29-46, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9516085

RESUMO

Estrogen receptor alpha (ER) plays a key role in the development and progression of breast cancer as well as the treatment and outcome of breast cancer patients. In normal mammary epithelial cells, the level of ER fluctuates during the menstrual cycle in response to cyclical changes in estrogen. However, in breast cancer normal control of ER gene expression and/or function is lost. Of particular interest, the absence of ER in mammary carcinomas is associated with a less-differentiated phenotype and resistance to endocrine therapies. This review focuses on our current understanding of the mechanisms that regulate ER alpha gene expression and function in breast cancer. These include alteration of the ER gene, loss of gene expression, alternative splicing of ER RNA, posttranslational modification of the protein, and interaction of ER with other proteins that can modify its function.


Assuntos
Neoplasias da Mama/fisiopatologia , Receptores de Estrogênio/fisiologia , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo
14.
J Biol Chem ; 272(51): 32260-6, 1997 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-9405430

RESUMO

The cytosine analog 5-aza-2'-deoxycytidine is a potent inhibitor of DNA methyltransferase. Its cytotoxicity has been attributed to several possible mechanisms including reexpression of growth suppressor genes and formation of covalent adducts between DNA methyltransferase and 5-aza-2'-deoxycytidine-substituted DNA which may lead to steric inhibition of DNA function. In this study, we use a panel of human breast cancer cell lines as a model system to examine the relative contribution of two mechanisms, gene reactivation and adduct formation. Estrogen receptor-negative cells, which have a hypermethylated estrogen receptor gene promoter, are more sensitive than estrogen receptor-positive cells and underwent apoptosis in response to 5-aza-2'-deoxycytidine. For the first time, we show that reactivation of a gene silenced by methylation, estrogen receptor, plays a major role in this toxicity in one estrogen receptor-negative cell line as treatment of the cells with anti-estrogen-blocked cell death. However, drug sensitivity of other tumor cell lines correlated best with increased levels of DNA methyltransferase activity and formation DNA.DNA methyltransferase adducts as analyzed in situ. Therefore, both reexpression of genes like estrogen receptor and formation of covalent enzyme. DNA adducts can play a role in 5-aza-2'-deoxycytidine toxicity in cancer cells.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Neoplasias da Mama/patologia , Adutos de DNA/metabolismo , Metilação de DNA , Metilases de Modificação do DNA/metabolismo , Receptores de Estrogênio/metabolismo , Azacitidina/farmacologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Humanos , Células Tumorais Cultivadas
15.
Proc Natl Acad Sci U S A ; 97(11): 6049-54, 2000 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-10811911

RESUMO

Expression of 14-3-3 final sigma (final sigma) is induced in response to DNA damage, and causes cells to arrest in G(2). By SAGE (serial analysis of gene expression) analysis, we identified final sigma as a gene whose expression is 7-fold lower in breast carcinoma cells than in normal breast epithelium. We verified this finding by Northern blot analysis. Remarkably, final sigma mRNA was undetectable in 45 of 48 primary breast carcinomas. Genetic alterations at final sigma such as loss of heterozygosity were rare (1/20 informative cases), and no mutations were detected (0/34). On the other hand, hypermethylation of CpG islands in the final sigma gene was detected in 91% (75/82) of breast tumors and was associated with lack of gene expression. Hypermethylation of final sigma is functionally important, because treatment of final sigma-non-expressing breast cancer cell lines with the drug 5-aza-2'-deoxycytidine resulted in demethylation of the gene and synthesis of final sigma mRNA. Breast cancer cells lacking final sigma expression showed increased number of chromosomal breaks and gaps when exposed to gamma-irradiation. Therefore, it is possible that loss of final sigma expression contributes to malignant transformation by impairing the G(2) cell cycle checkpoint function, thus allowing an accumulation of genetic defects. Hypermethylation and loss of final sigma expression are the most consistent molecular alterations in breast cancer identified so far.


Assuntos
Neoplasias da Mama/genética , Proteínas de Ciclo Celular/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Proteínas/genética , Tirosina 3-Mono-Oxigenase , Proteínas 14-3-3 , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Mama/citologia , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular/efeitos dos fármacos , Linhagem Celular Transformada/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Ilhas de CpG , Metilação de DNA/efeitos dos fármacos , Reparo do DNA/genética , Decitabina , Células Epiteliais/metabolismo , Feminino , Fase G2/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Biossíntese de Proteínas , Proteínas/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Tolerância a Radiação/genética , Proteínas Recombinantes de Fusão/fisiologia , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA