Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 5: e1114, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24625973

RESUMO

The serine/threonine kinase AKT is generally accepted as a promising anticancer therapeutic target. However, the relief of feedback inhibition and enhancement of other survival pathways often attenuate the anticancer effects of AKT inhibitors. These compensatory mechanisms are very complicated and remain poorly understood. In the present study, we found a novel 2-pyrimidyl-5-amidothiazole compound, DC120, as an ATP competitive AKT kinase inhibitor that suppressed proliferation and induced apoptosis in liver cancer cells both in vitro and in vivo. DC120 blocked the phosphorylation of downstream molecules in the AKT signal pathway in dose- and time-dependent manners both in vitro and in vivo. However, unexpectedly, DC120 activated mammalian target of rapamycin complex 1 (mTORC1) pathway that was suggested by increased phosphorylation of 70KD ribosomal protein S6 kinase (P70S6K) and eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1). The activated mTORC1 signal was because of increase of intracellular Ca(2+) via Ca(2+)/calmodulin (CaM)/ signaling to human vacuolar protein sorting 34 (hVps34) upon AKT inhibition. Meanwhile, DC120 attenuated the inhibitory effect of AKT on CRAF by decreasing phosphorylation of CRAF at Ser259 and thus activated the mitogen-activated protein kinase (MAPK) pathway. The activation of the mTORC1 and MAPK pathways by DC120 was not mutually dependent, and the combination of DC120 with mTORC1 inhibitor and/or MEK inhibitor induced significant apoptosis and growth inhibition both in vitro and in vivo. Taken together, the combination of AKT, mTORC1 and/or MEK inhibitors would be a promising therapeutic strategy for liver cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Pirimidinas/farmacologia , Tiazóis/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Calmodulina/metabolismo , Proteínas de Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Classe III de Fosfatidilinositol 3-Quinases/genética , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Relação Dose-Resposta a Droga , Retroalimentação Fisiológica , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Interferência de RNA , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo , Transfecção , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Oncogene ; 30(8): 944-55, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-20956948

RESUMO

An elevated DNA-repair capacity in cancer cells leads to radiation resistance and severely limits the efficacy of radiation therapy. Activation of Akt is tightly associated with resistance to radiotherapy, and Mre11 protein has important role during the repair of DNA double-strand breaks (DSBs). In this report, our results showed that inhibition of Akt activity impaired the repair of DSBs in CNE2 cells, whereas activated Akt promoted the repair of DSBs in HeLa cells. Knockdown of Mre11 also impaired the process of DSB repair in both these two cell lines. More importantly, we found that Akt could regulate Mre11 expression. Inhibition of Akt activity by small interfering RNA or LY294002 efficiently downregulated the Mre11 expression in CNE2 cells, and transfection with myr-Akt plasmid in HeLa cells upregulated the Mre11 expression. In addition, luciferase reporter analysis revealed that Mre11 reporter activity increased after transfection with myr-Akt1 plasmids, and this myr-Akt1-induced transcriptional activity was blocked in the presence of LY294002. Further study showed GSK3ß/ß-catenin/LEF-1 pathway was involved in this regulation. Knockdown of ß-catenin or LEF-1 led to the downregulation of Mre11, whereas overexpression of ß-catenin led to upregulation of Mre11. The chromatin immunoprecipitation assay assay showed ß-catenin/LEF-1 heterodimer could directly bind to the promoter of Mre11 in vivo. And the luciferase activity of the pGL3-Mre11 and pGL3-Lef increased in HeLa cells following ß-catenin plasmid co-transfected, but was abolished when the LEF-1-binding conserved sequences of Mre11 promoter were mutated. These results together support Akt can upregulate the expression of Mre11 through GSK3ß/ ß-catenin/LEF pathway to elevate DSB-repair capacity in cancer cells.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/genética , Proteínas de Ligação a DNA/biossíntese , Neoplasias/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tolerância a Radiação/genética , Ativação Enzimática/fisiologia , Expressão Gênica , Regulação Neoplásica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Técnicas de Silenciamento de Genes , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células HeLa , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Proteína Homóloga a MRE11 , Neoplasias/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Radiação Ionizante , Transdução de Sinais/fisiologia , Transfecção , beta Catenina/genética , beta Catenina/metabolismo
3.
Oncogene ; 28(6): 886-98, 2009 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-19060920

RESUMO

The c-Jun NH2-terminal kinase (JNK) pathway represents one subgroup of MAP kinases that are activated primarily by cytokines and exposure to environmental stress. Autophagy is a protein-degradation system characterized by the formation of double-membrane vacuoles termed autophagosomes. Autophagy-related gene beclin 1 plays a key role in autophagosome formation. However, the relationships between activation of JNK pathway, autophagy induction and Beclin 1 expression remain elusive. In this study, we used human cancer cell lines CNE2 and Hep3B to investigate the role of JNK-mediated Beclin 1 expression in ceramide-induced autophagic cell death. Ceramide-treated cells exhibited the characteristics of autophagy (that is, acidic vesicular organelle formation and the LC3-II generation). JNK was activated in these two cell lines exposed to ceramide and the phosphorylation of c-Jun also increased. In the meantime, we found that ceramide upregulated Beclin 1 expression in cancer cells. The upregulation of Beclin 1 expression could be blocked by SP600125 (a specific inhibitor of JNK) or a small interfering RNA (siRNA) directed against JNK1/2 or c-Jun. Chromatin immunoprecipitation and luciferase reporter analysis revealed that c-Jun was involved in the regulation of beclin 1 transcription in response to ceramide treatment. In addition, inhibition of JNK activity by SP600125 could inhibit autophagy induction by ceramide. Furthermore, Beclin 1 knockdown by siRNA also inhibited ceramide-mediated autophagic cell death. JNK-mediated Beclin 1 expression was also observed in topotecan-induced autophagy. These data suggest that activation of JNK pathway can mediate Beclin 1 expression, which plays a key role in autophagic cell death in cancer cells.


Assuntos
Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Regulação Neoplásica da Expressão Gênica , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas de Membrana/metabolismo , Antracenos/farmacologia , Proteína Beclina-1 , Caspase 3/metabolismo , Linhagem Celular Tumoral , Ceramidas/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , MAP Quinase Quinase 4/metabolismo , Fagossomos/metabolismo , Fosforilação , RNA Interferente Pequeno/metabolismo
5.
Oncogene ; 25(53): 7070-7, 2006 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-16715131

RESUMO

It has been shown that acetylcholinesterase (AChE) expression was induced during apoptosis and the anti-sense oligonucleotides and siRNA of AChE may prevent apoptosis in various cell types. However, the mechanisms underlying AChE upregulation remain elusive. We demonstrated here that c-Jun NH2-terminal kinase (JNK) could mediate AChE expression. In this study, both etoposide and excisanin A, two anticancer agents, induced apoptosis in colon cancer cell line SW620 as determined by Annexin V staining, the cleavage of caspase-3 and the proteolytic degradation of poly (ADP-ribose) polymerase (PARP). The results showed that both the agents upregulated AChE in SW620 cells. In the meantime, JNK was also activated and the expression and phosphorylation of c-Jun increased in SW620 cells exposed to the two agents. The induced AChE mRNA and protein expression could be blocked by SP600125, a specific inhibitor of SAPK/JNK, and small interfering RNA directed against JNK1/2. Transfection with adenovirus-mediated dominant negative c-Jun also blocked the upregulation of AChE expression. Together, these results suggest that AChE expression may be mediated by the activation of JNK pathway during apoptosis through a c-Jun-dependent mechanism.


Assuntos
Acetilcolinesterase/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Diterpenos/farmacologia , Etoposídeo/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Acetilcolinesterase/genética , Adenoviridae/genética , Antracenos/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Interferente Pequeno/genética , Regulação para Cima/efeitos dos fármacos
6.
Cancer Lett ; 169(1): 27-32, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11410322

RESUMO

Nasopharyngeal carcinoma (NPC), which occurs with a high incidence in southern China and southeast Asia, is of epithelial origin with overexpression of EGF receptor. To study the effect of inhibition of EGFR signaling on nasopharyngeal carcinoma cell proliferation and cell cycle distribution, EGFR tyrosine kinase inhibitor AG1478 was employed to treat Nasopharyngeal Carcinoma CNE2 cells. The results showed that AG1478 inhibited proliferation of CNE2 cells. Immunoblot showed that AG1478 inhibited EGFR phosphorylation in CNE2 cells without reduced expression of EGFR protein. The activation of Akt and MAPK which are downstream molecules of EGFR signaling pathway, were also inhibited by AG1478. AG1478 induced cell cycle arrest in G1 phase, and the levels of protein p27 were significantly up-regulated. We concluded that inhibition of the EGFR signaling induced cell cycle arrest in G1 phase in CNE2 cells and p27 up-regulation was involved in this process. The EGFR kinase specific inhibitor is of potential to be developed into drugs for NPC treatment.


Assuntos
Proteínas de Ciclo Celular , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Neoplasias Nasofaríngeas/patologia , Proteínas Serina-Treonina Quinases , Proteínas Supressoras de Tumor , Tirfostinas/farmacologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27 , Ativação Enzimática/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Inibidores do Crescimento/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Nasofaríngeas/enzimologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Quinazolinas , Células Tumorais Cultivadas/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
7.
Yao Xue Xue Bao ; 36(7): 498-501, 2001 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-12585079

RESUMO

AIM: To investigate the mechanism of apoptosis of HL60 cells induced by the annonaceous acetogenin, squamocin. METHODS: Induction of apoptosis was determined through Hoechst33258 dye staining and DNA agarose gel electrophoresis. Expression of the proteins was detected using Western blot analysis. Caspase-3 activity was detected using caspase-3 kit. RESULTS: Treatment of HL-60 cells with squamocin resulted in extensive nuclear condensation, DNA fragmentation, cleavage of the death substrate poly(ADP-ribose) polymerase (PARP) and induction of caspase-3 activity. Pretreatment of HL-60 cells with caspase-3 specific inhibitor DEVD-CHO prevented squamocin-induced DNA fragmentation, PARP cleavage and cell death. Stress-activated protein kinase (SAPK/JNK) was activated after treatment with squamocin in HL-60 cells. CONCLUSION: These results suggest that apoptosis of HL-60 cells induced by squamocin require caspase-3 activation, and could be related to SAPK activation.


Assuntos
Apoptose , Caspases/metabolismo , Furanos/farmacologia , Lactonas/farmacologia , Annona/química , Antineoplásicos Fitogênicos/isolamento & purificação , Antineoplásicos Fitogênicos/farmacologia , Caspase 3 , Medicamentos de Ervas Chinesas/isolamento & purificação , Medicamentos de Ervas Chinesas/farmacologia , Furanos/isolamento & purificação , Células HL-60 , Humanos , Lactonas/isolamento & purificação , Plantas Medicinais/química
8.
Oncology ; 57(1): 36-41, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10394123

RESUMO

The efficacy and targeting cells of angiogenesis inhibitor TNP-470 on human squamous cell nasopharyngeal carcinoma (NPC) were investigated. The colorimetric MTT assay was used to evaluate the IC50 values of NPC/HK1 cells and human dermal microvascular endothelial cells (HDMEC) for TNP-470. An NPC human tumor model was built by tumor-bearing nude mice using the NPC cell line of NPC/HK1. TNP-470 (30 mg/kg s.c.) was injected every other day. The results showed that the IC50 of NPC/HK1 cells for TNP-470 was 3.8 times higher than that of HDMEC. A significant difference in tumor volume between control and treatment groups was found after 7 days of treatment and increased thereafter. At the end of the treatment, tumor volume was 773.7 +/- 287.1 mm3 (n = 8) in the control group versus 454.5 +/- 132.8 mm3 (n = 8) in the treatment group (p = 0. 013); the ratio of the mean tumor volume in treated animals to that of control animals was 0.587, resulting a 41.3% decrease in tumor growth. The necrotic area was larger in the treatment group. Physical toxicity did not result from the treatment. These studies suggest that angiogenesis inhibitor TNP-470 is effective in the treatment of squamous cell NPC without obvious toxicity.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Neoplasias Nasofaríngeas/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Sesquiterpenos/farmacologia , Cicloexanos , Humanos , Neoplasias Nasofaríngeas/fisiopatologia , O-(Cloroacetilcarbamoil)fumagilol , Células Tumorais Cultivadas
9.
Med Lab Sci ; 47(2): 132-4, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2381269

RESUMO

Blood selenium concentrations in 111 nasopharyngeal cancer patients and 56 normal subjects were determined using a modified 2,3-diamino naphthalene fluorometric method. Cancer subjects in this study were divided into two groups and blood Se levels (mean +/- SD) were 0.105 +/- 0.024 for untreated cases, 0.113 +/- 0.034 for those who had received treatment, and 0.109 +/- 0.030 microgram/ml for all subjects. The results suggest that low blood Se levels in nasopharyngeal cancer patients are a consequence of their disease rather than the cause of the cancer.


Assuntos
Neoplasias Nasofaríngeas/sangue , Selênio/sangue , Adulto , China , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...