Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 4: e621, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23640463

RESUMO

Although the recruitment of fibroblasts to areas of injury is critical for wound healing, their subsequent apoptosis is necessary in order to prevent excessive scarring. Fibroproliferative diseases, such as pulmonary fibrosis, are often characterized by fibroblast resistance to apoptosis, but the mechanism(s) for this resistance remains elusive. Here, we employed a murine model of pulmonary fibrosis and cells from patients with idiopathic pulmonary fibrosis (IPF) to explore epigenetic mechanisms that may be responsible for the decreased expression of Fas, a cell surface death receptor whose expression has been observed to be decreased in pulmonary fibrosis. Murine pulmonary fibrosis was elicited by intratracheal injection of bleomycin. Fibroblasts cultured from bleomycin-treated mice exhibited decreased Fas expression and resistance to Fas-mediated apoptosis compared with cells from saline-treated control mice. Although there were no differences in DNA methylation, the Fas promoter in fibroblasts from bleomycin-treated mice exhibited decreased histone acetylation and increased histone 3 lysine 9 trimethylation (H3K9Me3). This was associated with increased histone deacetylase (HDAC)-2 and HDAC4 expression. Treatment with HDAC inhibitors increased Fas expression and restored susceptibility to Fas-mediated apoptosis. Fibroblasts from patients with IPF likewise exhibited decreased histone acetylation and increased H3K9Me3 at the Fas promoter and increased their expression of Fas in the presence of an HDAC inhibitor. These findings demonstrate the critical role of histone modifications in the development of fibroblast resistance to apoptosis in both a murine model and in patients with pulmonary fibrosis and suggest novel approaches to therapy for progressive fibroproliferative disorders.


Assuntos
Apoptose , Fibroblastos/metabolismo , Histonas/metabolismo , Receptor fas/metabolismo , Acetilação , Animais , Apoptose/efeitos dos fármacos , Bleomicina/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/citologia , Histona Desacetilase 2/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Humanos , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Receptor fas/genética
2.
Nat Med ; 5(7): 810-6, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10395327

RESUMO

The mitogen-activated protein kinase pathway is thought to be essential in cellular growth and differentiation. Here we report the discovery of a highly potent and selective inhibitor of the upstream kinase MEK that is orally active. Tumor growth was inhibited as much as 80% in mice with colon carcinomas of both mouse and human origin after treatment with this inhibitor. Efficacy was achieved with a wide range of doses with no signs of toxicity, and correlated with a reduction in the levels of activated mitogen-activated protein kinase in excised tumors. These data indicate that MEK inhibitors represent a promising, noncytotoxic approach to the clinical management of colon cancer.


Assuntos
Benzamidas/farmacologia , Ciclo Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Neoplasias do Colo/fisiopatologia , Inibidores Enzimáticos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Benzamidas/uso terapêutico , Caderinas/análise , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/uso terapêutico , Feminino , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Nus , Invasividade Neoplásica/prevenção & controle , Transplante Heterólogo , Células Tumorais Cultivadas
4.
Invest New Drugs ; 13(4): 295-302, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8824347

RESUMO

PD153035 is a potent (Ki = 6 pm) and specific inhibitor of the epidermal growth factor (EGF) receptor tyrosine kinase that suppresses tyrosine phosphorylation of the EGF receptor in A431 cells at nanomolar concentrations in cell culture. We have examined the pharmacokinetics of this compound and its ability to rapidly suppress phosphorylation of the EGF receptor in A431 human epidermoid tumors grown as xenografts in immunodeficient nude mice. Following a single i.p. dose of 80 mg/kg, the drug levels in the plasma and tumor rose to 50 and 22 microM within 15 minutes. While the plasma levels of PD153035 fell below 1 microM by 3 hours, in the tumors it remained at micromolar concentrations for at least 12 hours. The tyrosine phosphorylation of the EGF receptor was rapidly suppressed by 80-90% in the tumors. However receptor phosphorylation returned to control levels after 3 hours despite the continued presence of the drug at concentrations which, based on previous in vitro results, were predicted to maintain inhibition. EGF-stimulated tyrosine kinase activity in tumor extracts was decreased and recovered in parallel with the effects of PD153035 on receptor phosphorylation though the activity had reached only about half of the control activity after three hours. These results demonstrate the potential for using small molecule inhibitors to inhibit the EGF receptor tyrosine kinase in vivo, though a fair evaluation of their potential anti-cancer activity will have to wait for solutions to problems with sustained delivery which may allow us to maintain suppression of EGF receptor phosphorylation.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/enzimologia , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Quinazolinas/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacocinética , Humanos , Camundongos , Camundongos Nus , Neoplasias/metabolismo , Quinazolinas/farmacocinética , Transplante Heterólogo , Células Tumorais Cultivadas
5.
Invest New Drugs ; 14(4): 341-7, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-9157068

RESUMO

CI-980, originally synthesized as a potential folate antagonist, is a tubulin-binding mitotic inhibitor currently in pediatric phase I and adult phase II clinical trials. Because of its extensive tissue distribution in animals and its favorable activity against multidrug resistant (MDR)-cells compared with other mitotic inhibitors, such as vincristine, we examined the membrane transport properties of CI-980. CI-980 accumulated rapidly in L1210 and CHO/K1 cells, reaching intracellular levels 40- and 8-fold higher, respectively, than those in the extracellular medium. Efflux was also quite rapid, but a small fraction of drug remained associated with the cells in drug-free medium. The uptake of CI-980 was not temperature or energy dependent, nor was it saturable up to an extracellular concentration of 100 microM. Inhibitors of nucleoside transport had no effect on CI-980 uptake. A cell line deficient in the transport of reduced folate was not resistant to CI-980, nor did it exhibit reduced CI-980 uptake. A 100-fold excess of the R-enantiomer inhibited CI-980 uptake by only 50%. These results are consistent with a model of CI-980 uptake involving passive diffusion followed by significant but largely reversible binding to intracellular or membrane components.


Assuntos
Antineoplásicos/farmacocinética , Carbamatos/farmacocinética , Antagonistas do Ácido Fólico/farmacocinética , Pirazinas/farmacocinética , Piridinas/farmacocinética , Animais , Transporte Biológico , Células CHO/metabolismo , Proteínas de Transporte/fisiologia , Bovinos , Cricetinae , Dipiridamol/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Leucemia L1210/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA