Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Rep ; 27(12): 3547-3560.e5, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31130414

RESUMO

Orchestrating cell-cycle-dependent mRNA oscillations is critical to cell proliferation in multicellular organisms. Even though our understanding of cell-cycle-regulated transcription has improved significantly over the last three decades, the mechanisms remain untested in vivo. Unbiased transcriptomic profiling of G0, G1-S, and S-G2-M sorted cells from FUCCI mouse embryos suggested a central role for E2Fs in the control of cell-cycle-dependent gene expression. The analysis of gene expression and E2F-tagged knockin mice with tissue imaging and deep-learning tools suggested that post-transcriptional mechanisms universally coordinate the nuclear accumulation of E2F activators (E2F3A) and canonical (E2F4) and atypical (E2F8) repressors during the cell cycle in vivo. In summary, we mapped the spatiotemporal expression of sentinel E2F activators and canonical and atypical repressors at the single-cell level in vivo and propose that two distinct E2F modules relay the control of gene expression in cells actively cycling (E2F3A-8-4) and exiting the cycle (E2F3A-4) during mammalian development.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Diferenciação Celular , Fator de Transcrição E2F3/fisiologia , Fator de Transcrição E2F4/fisiologia , Regulação da Expressão Gênica , Proteínas Repressoras/fisiologia , Animais , Proteínas de Ciclo Celular/genética , Proliferação de Células , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Transcriptoma
2.
Biochem Pharmacol ; 76(12): 1653-68, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18823950

RESUMO

Structurally diverse chemotherapeutic and chemopreventive drugs, including camptothecin, doxorubicin, sanguinarine, and others, were found to cause covalent crosslinking of proliferating cell nuclear antigen (PCNA) trimers in mammalian cells exposed to fluorescent light. This PCNA damage was caused by both nuclear and cytoplasmically localizing drugs. For some drugs, the PCNA crosslinking was evident even with very brief exposures to laboratory room lighting. In the absence of drugs, there was no detectable covalent crosslinking of PCNA trimers. Other proteins were photo-crosslinked to PCNA at much lower levels, including crosslinking of additional PCNA to the PCNA trimer. The proteins photo-crosslinked to PCNA did not vary with cell type or drug. PCNA was not crosslinked to itself or to other proteins by superoxide, hydrogen peroxide or hydroxyl radicals, but hydrogen peroxide caused monoubiquitination of PCNA. Quenching of PCNA photo-crosslinking by histidine, and enhancement by deuterium oxide, suggest a role for singlet oxygen in the crosslinking. SV40 large T antigen hexamers were also efficiently covalently photo-crosslinked by drugs and light. Photodynamic crosslinking of nuclear proteins by cytoplasmically localizing drugs, together with other evidence, argues that these drugs may reach the nucleoplasm in amounts sufficient to photodamage important chromosomal enzymes. The covalent crosslinking of PCNA trimers provides an extremely sensitive biomarker for photodynamic damage. The damage to PCNA and large T antigen raises the possibility that DNA damage signaling and repair mechanisms may be compromised when cells treated with antineoplastic drugs are exposed to visible light.


Assuntos
Antineoplásicos/efeitos adversos , Luz/efeitos adversos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Linhagem Celular Tumoral , Dimerização , Histidina/metabolismo , Humanos , Fotoquímica , Oxigênio Singlete
3.
J Clin Oncol ; 24(23): 3771-9, 2006 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-16801630

RESUMO

Epigenetic editing of gene expression by aberrant methylation of DNA may help tumor cells escape attack from the innate and acquired immune systems. Resistance to antiproliferative effects and apoptosis induction by interferons (IFNs) was postulated to result from silencing of IFN response genes by promoter hypermethylation. Treatment of human ACHN renal cell carcinoma (RCC) and A375 melanoma cells with the DNA demethylating nucleoside analog 5-AZA-2'-deoxycytidine (5-AZA-dC) synergistically augmented antiproliferative effects of IFN- alpha (alpha) 2 and IFN-beta (beta). Either 5-AZA-dC or an antisense to DNA methyltransferase 1 (DNMT1) overcame resistance to apoptosis induction by IFNs with up to 85% apoptotic cells resulting from the combinations. No similar potentiation occurred in normal kidney epithelial cells. IFN response genes were augmented more than 10 times in expression by 5-AZA-dC. Demethylation by 5-AZA-dC of the promoter of the prototypic, apoptosis-associated IFN response gene XAF1 was confirmed by methylation-specific polymerase chain reaction. siRNA to XAF1 inhibited IFN-induced apoptosis; conversely, overexpression of XAF1 overcame resistance to apoptosis induction by IFN-beta. As occurred with apoptosis-resistant melanoma cells in vitro, tumor growth inhibition in the nude mouse of human A375 melanoma xenografts resulted from treatment with 5-AZA-dC in combination with IFN-beta, an effect not resulting from either single agent. The importance of epigenetic remodeling of expression of immune-modifying genes in tumor cells was further suggested by identifying reactivation of the cancer-testis antigens MAGE and RAGE in ACHN cells after DNMT1 depletion. Thus, inhibitors of DNMT1 may have clinical relevance for immune modulation by augmentation of cytokine effects and/or expression of tumor-associated antigens.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Azacitidina/análogos & derivados , Carcinoma de Células Renais/tratamento farmacológico , Metilação de DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Interferons/farmacologia , Melanoma Experimental/tratamento farmacológico , Proteínas de Neoplasias/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antimetabólitos Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose , Azacitidina/farmacologia , Western Blotting , Carcinoma de Células Renais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilases de Modificação do DNA/farmacologia , Decitabina , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias Renais/tratamento farmacológico , Melanoma Experimental/metabolismo , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase , Transplante Heterólogo , Regulação para Cima
4.
Cancer Res ; 66(5): 2785-93, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16510600

RESUMO

Resistance of human renal cell carcinoma (RCC) and melanoma to the apoptosis-inducing effects of IFNs was postulated to result from epigenetic silencing of genes by DNA methylation, a common feature of human cancers. To reverse silencing, 5-AZA-deoxycytidine (5-AZA-dC) or selective depletion of DNA methyltransferase 1 (DNMT1) by phosphorothioate oligonucleotide antisense (DNMT1 AS) were employed in cells resistant (<5% terminal deoxynucleotidyl transferase-mediated nick-end labeling positive) to apoptosis induction by IFN-alpha2 and IFN-beta (ACHN, SK-RC-45, and A375). 5-AZA-dC and DNMT1 AS similarly depleted available DNMT1 protein and, at doses that did not cause apoptosis alone, resulted in apoptotic response to IFNs. The proapoptotic tumor suppressor RASSF1A was reactivated by DNMT1 inhibitors in all three cell lines. This was associated with demethylation of its promoter region. IFNs augmented RASSF1A protein expression after reactivation by DNMT1 inhibition. In IFN-sensitive WM9 melanoma cells, expression of RASSF1A was constitutive but also augmented by IFNs. RASSF1A small interfering RNA reduced IFN-induced apoptosis in WM9 cells and in DNMT1-depleted ACHN cells. Conversely, lentiviral expression of RASSF1A but not transduction with empty virus enabled IFN-induced apoptosis. IFN induced tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and TRAIL-neutralizing antibody inhibited apoptotic response to IFN in RASSF1A-expressing ACHN cells. Accordingly, RASSF1A markedly sensitized to recombinant TRAIL. Normal kidney epithelial cells, although expressing RASSF1A, did not undergo apoptosis in response to IFN or TRAIL but had >400-fold higher TRAIL decoy receptor 1 expression than transduced ACHN cells (real-time reverse transcription-PCR). Results identified RASSF1A as regulated by IFNs and participating in IFN-induced apoptosis at least in part by sensitization to TRAIL.


Assuntos
Apoptose/efeitos dos fármacos , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Proteínas Supressoras de Tumor/biossíntese , Apoptose/genética , Proteínas Reguladoras de Apoptose/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/deficiência , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética , Inativação Gênica , Células HeLa , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Lentivirus/genética , Lentivirus/metabolismo , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Glicoproteínas de Membrana/farmacologia , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética
5.
Anticancer Res ; 25(6B): 4013-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16309193

RESUMO

BACKGROUND: The GSTPI gene encodes for glutathione S-transferase pi (GST-pi), which protects cells from cytotoxic agents. The carcinogenic role of this enzyme is at issue because functional polymorphisms have been shown to be a risk factor of human cancer. Moreover, GST-pi protein loss has frequently been reported in various human cancers. MATERIALS AND METHODS: The expression of GST-pi and the methylation status of the promoter area of GST-pi were investigated in gastric carcinomas. Eleven human SNUgastric cancer cell lines, PC-3 prostate cancer cell lines, various cancer tissues and normal gastric mucosa tissues were analyzed by immunohistochemistry, in situ hybridization, Western blot and methylation specific PCR. RESULTS: Only 22 (2.0%o) out of 1081 cases showed loss of GST-pi expression. Interestingly, 16 out of 22 GST-pi-negative cases were Epstein-Barr virus (EBV)-associated gastric carcinomas. The loss of expression of GST-pi among EBV-associated gastric carcinomas was found to be 27.1% (16/59), but to be 0.6% (6/1022) in EBV-negative gastric carcinomas (p < 0.001). Eight out of 16 cases with loss of GST-pi expression showed CpG island methylation in the GSTP1 promoter region, while none of the normal gastric mucosa or EBV-negative gastric carcinomas showed methylation (p < 0.001). CONCLUSION: These findings demonstrate that the loss of GST-pi expression is clustered in a subset of gastric carcinomas with EBV incorporation, and that the methylation of the promoter of the GSTP1 gene is correlated with this loss of GST-pi expression. Our results suggest that GST-pi abrogation by CpG island hypermethylation may account for EBV-associated gastric carcinoma.


Assuntos
Infecções por Vírus Epstein-Barr/genética , Glutationa S-Transferase pi/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/virologia , Infecções Tumorais por Vírus/genética , Linhagem Celular Tumoral , Ilhas de CpG , Metilação de DNA , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/enzimologia , Feminino , Inativação Gênica , Glutationa S-Transferase pi/biossíntese , Herpesvirus Humano 4 , Humanos , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/patologia , Infecções Tumorais por Vírus/complicações , Infecções Tumorais por Vírus/enzimologia , Infecções Tumorais por Vírus/virologia
6.
Int J Oncol ; 26(5): 1265-71, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15809717

RESUMO

The caspases are a family of aspartic acid-specific proteases that fulfill varied and often critical roles in mammalian apoptosis or in the proteolytic activation of cytokines. Caspase-1 (interleukin-1beta-converting enzyme) is a member of the cysteine protease family, which cleaves target proteins following aspartic acid residues. We investigated caspase-1 expression in stomach cancer, tissues and cell lines. Of 301 consecutive gastric carcinomas, 58 cases (19.3%) showed the expressional loss of caspase-1. Loss of caspase-1 expression was significantly associated with pTNM stage (p=0.03), lymph node metastasis (p=0.01) and patient survival (p<0.01). Caspase-1 expression was also significantly correlated in an inverse manner with p53 expression (p<0.01). Among the 11 gastric cancer cell lines examined, three cell lines showed loss of expression at the protein and mRNA levels. On treatment with 5-aza-2'-deoxycytidine (5-aza-C), and/or trichostatin A (TSA), all three cell lines re-expressed caspase-1 mRNA. The above findings suggest that epigenetic events such as DNA methylation and histone deacetylation play important roles in the regulation of caspase-1, and that loss of caspase-1 expression is associated with poor survival in gastric carcinoma.


Assuntos
Azacitidina/análogos & derivados , Carcinoma/genética , Caspase 1/biossíntese , Caspase 1/genética , Metilação de DNA , Perfilação da Expressão Gênica , Metástase Linfática/genética , Neoplasias Gástricas/genética , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/farmacologia , Carcinoma/patologia , Decitabina , Feminino , Histonas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Neoplasias Gástricas/patologia , Análise de Sobrevida , Células Tumorais Cultivadas
7.
Clin Cancer Res ; 9(15): 5768-75, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14654562

RESUMO

PURPOSE: The purpose is to investigate the potential correlation between antioxidant enzyme (AOE) levels and resistance to anticancer drugs in human gastric carcinoma cell lines. EXPERIMENTAL DESIGN: Protein contents of AOEs such as manganese superoxide dismutase (MnSOD), copper/zinc superoxide dismutase, catalase, glutathione S-transferase-pi, p-glycoprotein, and multidrug resistance-associated protein were observed by Western blot analysis, and MnSOD activity was measured in six Korean gastric cancer cell lines. The direct correlation between AOEs and the chemosensitivity to doxorubicin (DOX), mitomycin C, 5-fluorouracil, and vinblastine was analyzed by cytotoxicity test. MnSOD was overexpressed by transient transfection of human MnSOD cDNA. RESULTS: Expressions of AOEs in gastric cancer cell lines were variable. MnSOD expression was related with the resistance to DOX and mitomycin C but not with that to 5-fluorouracil and vinblastine. In comparison, expressions of other AOEs, p-glycoprotein and multidrug resistance-associated protein, were not correlated with tumor sensitivity to any of the drugs used. Cell lines with a high MnSOD protein content showed higher MnSOD activity than those with a low MnSOD protein content. In addition, MnSOD overexpression increased the resistance of gastric carcinoma cells to DOX. CONCLUSIONS: MnSOD is an important factor of drug response to reactive oxygen species-generating anticancer drugs in the gastric cancer cells. Thus, measurement of MnSOD levels in clinical samples may provide an indication of subsequent treatment response of gastric cancer patients.


Assuntos
Antineoplásicos/toxicidade , Neoplasias Gástricas/patologia , Superóxido Dismutase/genética , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/análise , Coreia (Geográfico) , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/metabolismo
8.
Mol Carcinog ; 37(1): 32-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12720298

RESUMO

DNA repair failure is known to be a critical event during carcinogenesis of colorectal cancers. To investigate whether O(6)-methylguanine-DNA methyltransferase (MGMT) is altered during colorectal carcinogenesis, we performed immunohistochemical staining on 265 sporadic colorectal cancers, 113 sporadic adenomas, 33 familial adenomatous polyposis (FAP) colorectal cancers, and 93 FAP adenomas. Sixty-seven of 265 sporadic colorectal cancer cases and five of 113 sporadic adenoma cases showed loss of MGMT expression (P < 0.001). Among FAP patients, four of 33 cancers and six of 93 adenomas showed loss of MGMT protein expression. When we compared the association between MGMT promoter hypermethylation and protein expression, almost all cases without a methylated allele were positive for the expression of MGMT. In contrast, cases with promoter methylation frequently showed loss of MGMT expression (P < 0.01). Loss of MGMT was correlated with some clinicopathological characteristics, i.e., tumor invasion (P = 0.013) and stage (P = 0.035) in sporadic colorectal cancer, and degree of atypism (P = 0.042) in sporadic adenoma. Our results show that loss of expression of MGMT occurs more frequently in cancer than in adenoma in both sporadic and FAP patients, and that loss of expression of MGMT is associated with hypermethylation of the promoter area of MGMT gene.


Assuntos
Polipose Adenomatosa do Colo/enzimologia , Polipose Adenomatosa do Colo/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Adenoma/enzimologia , Adenoma/genética , Adenoma/metabolismo , Polipose Adenomatosa do Colo/patologia , Regulação Enzimológica da Expressão Gênica , Humanos , Estadiamento de Neoplasias , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA