Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Sci ; 101(3): 728-34, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20059476

RESUMO

Methotrexate (MTX) has been used to treat various hematological malignancies. Since MTX prevents tumor cells from proliferating by inhibiting dihydrofolate reductase (DHFR), DHFR expression is a key determinant of resistance to MTX in malignant hematological tumor cells. The antiproliferative effect of MTX was significantly enhanced by the knockdown of DHFR expression by siRNA in Jurkat cells. Therefore, a novel strategy down-regulating DHFR expression seems promising for enhancing sensitivity to MTX. We found that SU9516, a cyclin-dependent kinase inhibitor, reduced the expression of both DHFR mRNA and protein. Moreover, we found that DHFR promoter activity was attenuated by SU9516 dependent on the E2F site. Finally, pretreatment with SU9516 significantly enhanced sensitivity to MTX in a colony formation assay. We conclude that a combination of cyclin-dependent kinase inhibitors and MTX may be useful for overcoming resistance to MTX.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Imidazóis/farmacologia , Indóis/farmacologia , Metotrexato/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Células Jurkat , Regiões Promotoras Genéticas , Purinas/farmacologia , Tetra-Hidrofolato Desidrogenase/genética
2.
Biochem Genet ; 48(11-12): 970-86, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20862607

RESUMO

Gene promoter regions of p15(INK4B), a cyclin-dependent kinase inhibitor, and phosphatase and tensin homolog (PTEN), a dual-function protein and lipid phosphatase, interact with regulatory factors for gene transcription and methylation. Normal individuals exhibit sequence polymorphisms in these regulatory genes. We isolated genomic DNA from whole blood of healthy Japanese individuals and sequenced promoter regions of the p15 ( INK4B ) and PTEN genes. We also examined the influence of polymorphisms on promoter activity in several cell lines. We identified polymorphisms at positions -699, -394, and -242 and an insertion at position -320 in the p15 ( INK4B ) gene and a polymorphism at position -1142 in the PTEN gene. Reporter gene analysis revealed that these polymorphisms influenced transcriptional regulation in their cell lines. Our results indicate for the first time that promoter sequences of the p15 ( INK4B ) and PTEN genes differ among normal Japanese individuals and that promoter polymorphisms can influence gene transcription.


Assuntos
Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Regulação da Expressão Gênica , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Polimorfismo Genético , Regiões Promotoras Genéticas , Sequência de Bases/genética , Linhagem Celular Transformada/química , Inibidor de Quinase Dependente de Ciclina p15/sangue , Inibidor de Quinase Dependente de Ciclina p15/química , DNA/sangue , DNA/genética , Genes Reporter , Humanos , Japão , Luciferases/análise , Dados de Sequência Molecular , Mutação/genética , PTEN Fosfo-Hidrolase/sangue , PTEN Fosfo-Hidrolase/química , Fatores de Transcrição/metabolismo
3.
Int J Oncol ; 32(5): 1105-10, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18425338

RESUMO

Thymidylate synthase (TS) is a dNTP synthetic enzyme and is also a target enzyme of 5-fluorouracil (5-FU). 5-FU is one of the anticancer agents most frequently used for the treatment of colorectal cancers. However, the clinical rate of response to its use as a single agent is not exceptionally high. Therefore, various combination chemotherapies have been devised. The elevated expression of TS in cancer cells is a serious obstacle in the clinical use of 5-FU. In the present study, TS expression was up-regulated by the knockout of the p21WAF1/CIP1 gene in human colorectal cancer HCT116 cells, suggesting that TS expression is mediated through the inhibition of cyclin-dependent kinase (CDK). Based on these findings, we tested whether the CDK inhibitor (CDKI) SU9516, acted as a suppressor of TS. SU9516 effectively reduced the expression of TS in a dose-dependent manner. Furthermore, the reduction of TS expression resulted in enhancement of the sensitivity to 5-FU in human colon cancer DLD-1 cells. Thus, SU9516 might be a promising compound for combination chemotherapy with 5-FU.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Colorretais/enzimologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Timidilato Sintase/antagonistas & inibidores , Antimetabólitos Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Relação Dose-Resposta a Droga , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Repressão Enzimática , Fluoruracila/análogos & derivados , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Células HCT116 , Humanos , Imidazóis/farmacologia , Indóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/metabolismo , Timidilato Sintase/biossíntese , Timidilato Sintase/genética , Transfecção
4.
Int J Oncol ; 31(4): 915-22, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17786325

RESUMO

We previously established a bioassay method to screen for compounds that activate the promoter activity of p21(WAF1/CIP1), a potent inhibitor of cyclin-dependent kinases, in a p53-independent manner. As an activator of p21(WAF1/CIP1) promoter activity, we isolated cryptolepine (CLP: 5-methyl indolo (2,3b)-quiniine), an indoloquinoline alkaloid, from the traditional Ayurvedic medicinal plant Sida cordifolia. We show here that CLP induces the expression of p21(WAF1/CIP1) with growth arrest in p53-mutated human osteosarcoma MG63 cells. Four micromolar of CLP completely inhibited the growth of MG63 cells and caused G2/M-phase arrest. CLP up-regulated the expression of p21(WAF1/CIP1) at both mRNA and protein levels in a dose-dependent manner. Using several mutant p21(WAF1/CIP1) promoter constructs, we found that the CLP-responsive element is an Sp1 site at -82 relative to the transcription start site of the p21(WAF1/CIP1) promoter. These findings suggest that CLP arrests the growth of MG63 cells by activating the p21(WAF1/CIP1) promoter through the specific Sp1 site in a p53-independent manner. In addition, CLP-mediated cell cycle arrest was reduced by the knockout of the p21(WAF1/CIP1) gene in human colon cancer HCT116 cells, suggesting that the cell cycle arrest by CLP was at least partially mediated through the induction of p21(WAF1/CIP1) expression. Although we need further study of chemotherapeutic effect in vivo, these results raise the possibility that CLP might be a suitable chemotherapeutic agent for treatment of osteosarcoma.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Divisão Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fase G2/efeitos dos fármacos , Alcaloides Indólicos/farmacologia , Osteossarcoma/tratamento farmacológico , Quinolinas/farmacologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Western Blotting , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Luciferases , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Elementos de Resposta , Fator de Transcrição Sp1/metabolismo , Inibidores da Topoisomerase II , Transcrição Gênica , Células Tumorais Cultivadas/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
5.
Oncol Rep ; 18(5): 1263-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17914583

RESUMO

Sulforaphane (SFN), a naturally occurring isothiocyanate, is an attractive agent due to its potent anticancer effects. SFN suppresses the proliferation of various cancer cells in vitro and in vivo. In this study, we report that SFN inhibited the proliferation of cultured murine osteosarcoma LM8 cells. Twenty micromolar SFN completely inhibited the growth of LM8 cells and caused G2/M-phase arrest. SFN induced the expression of p21(WAF1/CIP1) protein causing the cell cycle arrest in a dose-dependent manner. SFN induced apoptosis which was characterized by the appearance of cells with sub-G1 DNA content and the cleavage and activation of caspase-3. We showed that SFN induced the growth arrest and up-regulated the expression of p21(WAF1/CIP1) protein in a p53-independent manner in human osteosarcoma MG63 cells. We found that intraperitoneal administration of SFN (1 or 2 mg, 5 times/week) significantly inhibited the growth of LM8 xenografts to <30% of the controls in a preclinical animal model without causing any toxicity. In osteosarcoma cells, our findings provide in vivo evidence for the efficacy of SFN against the advanced growth of tumor. We showed that SFN induces cell cycle arrest and apoptosis in osteosarcoma cells and inhibits tumor xenograft growth. Furthermore, SFN is a potent inducer of p21(WAF1/CIP1) in osteosarcoma cells. These results raise the possibility that SFN may be a promising candidate for molecular-targeting chemotherapy against osteosarcoma.


Assuntos
Anticarcinógenos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/patologia , Divisão Celular/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Osteossarcoma/patologia , Tiocianatos/farmacologia , Animais , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Humanos , Isotiocianatos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Sulfóxidos
6.
Toxicol Rep ; 4: 172-180, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28959638

RESUMO

Chemotherapy-induced taste disorder is one of the critical issues in cancer therapy. Bortezomib, a proteasome inhibitor, is a key agent in multiple myeloma therapy, but it induces a taste disorder. In this study, we investigated the characteristics of bortezomib-induced taste disorder and the underlying mechanism in mice. Among the five basic tastes, the sour taste sensitivity of mice was significantly increased by bortezomib administration. In bortezomib-administered mice, protein expression of PKD2L1 was increased. The increased sour taste sensitivity induced by bortezomib returned to the control level on cessation of its administration. These results suggest that an increase in protein expression of PKD2L1 enhances the sour taste sensitivity in bortezomib-administered mice, and this alteration is reversed on cessation of its administration.

7.
Oncogene ; 24(48): 7180-9, 2005 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-16007131

RESUMO

Luteolin, a naturally occurring flavonoid, induces apoptosis in various cancer cells. Little is known however concerning the underlying molecular mechanisms responsible for this activity. In this report, we reveal a novel mechanism by which luteolin-induced apoptosis occurs, and show for the first time that the apoptosis by luteolin is mediated through death receptor 5 (DR5) upregulation. Luteolin markedly induced the expression of DR5, along with Bcl-2-interacting domain cleavage and the activation of caspase-8, -10, -9 and -3. In addition, suppression of DR5 expression with siRNA efficiently reduced luteolin-induced caspase activation and apoptosis. Human recombinant DR5/Fc also inhibited luteolin-induced apoptosis. On the other hand, luteolin induced neither DR5 protein expression nor apoptosis in normal human peripheral blood mononuclear cells. These results suggest that DR5 induced by luteolin plays a role in luteolin-induced apoptosis, and raises the possibility that treatment with luteolin might be promising as a new therapy against cancer.


Assuntos
Apoptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Luteolina/farmacologia , Receptores do Fator de Necrose Tumoral/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Western Blotting , Inibidores de Caspase , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Luciferases/metabolismo , Masculino , Regiões Promotoras Genéticas , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/genética , Proteínas Recombinantes/metabolismo
8.
Int J Oncol ; 26(1): 185-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15586239

RESUMO

Apigenin, a common dietary flavonoid, has been shown to induce cell growth-inhibition and cell cycle arrest in many cancer cell lines. One important effect of apigenin is to increase the stability of the tumor suppressor p53 in normal cells. Therefore, apigenin is expected to play a large role in cancer prevention by modifying the effects of p53 protein. However, the mechanisms of apigenin's effects on p53-mutant cancer cells have not been revealed yet. We assessed the influence of apigenin on cell growth and the cell cycle in p53-mutant cell lines. Treatment with apigenin resulted in growth-inhibition and G2/M phase arrest in two p53-mutant cancer cell lines, HT-29 and MG63. These effects were associated with a marked increase in the protein expression of p21/WAF1. We have shown that p21/WAF1 mRNA expression was also markedly increased by treatment with apigenin in a dose- and time-dependent manner. However, we could not detect p21/WAF1 promoter activity following treatment with apigenin. Similarly, promoter activity from pG13-Luc, a p53-responsive promoter plasmid, was not activated by treatment with apigenin with or without p53 protein expression. These results suggest that there is a p53-independent pathway for apigenin in p53-mutant cell lines, which induces p21/WAF1 expression and growth-inhibition. Apigenin may be a useful chemopreventive agent not only in wild-type p53 status, but also in cancer with mutant p53.


Assuntos
Anticarcinógenos/farmacologia , Apigenina/farmacologia , Proteínas de Ciclo Celular/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Anticarcinógenos/uso terapêutico , Apigenina/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Quimioprevenção , Inibidor de Quinase Dependente de Ciclina p21 , Humanos , Mutação/genética , Neoplasias/prevenção & controle , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Proteína Supressora de Tumor p53/genética
9.
FEBS Lett ; 577(1-2): 55-9, 2004 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-15527761

RESUMO

Genistein is the most abundant isoflavone of soybeans and has been shown to cause growth arrest in various human cancer cell lines. However, the precise mechanism for this is still unclear. We report here that the growth arrest and DNA damage-inducible gene 45 (gadd45) gene is induced by genistein via its promoter in a DU145 human prostate cancer cell line. The binding of transcription factor nuclear factor-Y to the CCAAT site of the gadd45 promoter appears to be important for this activation by genistein.


Assuntos
Divisão Celular/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genisteína/farmacologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Masculino
10.
J Interferon Cytokine Res ; 23(11): 667-76, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14651781

RESUMO

2'-5'-Oligoadenylate synthetase (OAS), an interferon (IFN) induced enzyme, synthesizes 2'-5'-oligoadenylate (2-5A) from ATP when activated by dsRNA. Chicken OAS (ChOAS) has a ubiquitin-like (UbL) domain of two consecutive sequences (UbL1 and UbL2) at its carboxyl-terminus. The OAS gene has at least two alleles, OAS*A and OAS*B. OAS-A is the wild-type (wt) and OAS-B is a mutant deleted of a highly hydrophobic region of UbL1. To study the function of the UbL domain, enzymatic and physiologic properties were compared between OAS-A and OAS-B. OAS-B was more susceptible to trypsin than OAS-A and was converted very quickly into p38, deleting a greater part of the UbL domain. The p38 has the enzymatic activity to synthesize 2-5A. Thermal inactivation of OAS-B occurred at a lower temperature than that of OAS-A and p38, with loss of the ability to bind dsRNA. In contrast to OAS-A, the content of OAS-B in erythrocytes decreased during growth to a very low level. However, red blood cells (RBC) from anemic B/B chickens synthesized OAS-B at a high level comparable to A/A, although OAS-B levels decreased sharply again during maturation to erythrocytes. Thus, OAS-B carrying the mutated UbL domain is unstable compared with OAS-A in vitro and in vivo, and the wt UbL domain may contribute to the stability of the protein structure of ChOAS.


Assuntos
2',5'-Oligoadenilato Sintetase/química , 2',5'-Oligoadenilato Sintetase/genética , Alelos , Conformação Proteica , Ubiquitinas/química , 2',5'-Oligoadenilato Sintetase/metabolismo , Sequência de Aminoácidos , Animais , Galinhas , Estabilidade Enzimática , Eritrócitos/enzimologia , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína
11.
Chem Biol Interact ; 194(2-3): 120-6, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21964418

RESUMO

Perfluorooctane sulfonate (PFOS) is a chemically stable compound extensively used as oil and water repellent, surface active agents in our daily life. Accumulative research evidence gradually appears the toxicity of PFOS against mammals, but the whole figure remains to be elucidated. The present study was conducted to know the effects of PFOS on human hepatic drug metabolizing-type cytochrome P450 (CYP) isoenzymes such as CYP1A2 (7-ethoxyresorufin as a substrate), CYP2A6 (coumarin), CYP2B6 (7-ethoxy-4-trifluoromethylcoumarin), CYP2C8 (paclitaxel), CYP2C9 (diclofenac), CYP2C19 (S-mephenytoin), CYP2D6 (bufuralol), CYP2E1 (chlorzoxazone) and CYP3A4 (testosterone) in human livers employing their typical substrates. Although all of the oxidation reactions tested were more or less inhibited by PFOS, diclofenac 4'-hydroxylation mediated mainly by CYP2C9 was most strongly inhibited (K(i) value of 40 nM), followed by paclitaxel 6α-hydroxylation mediated mainly by CYP2C8 (K(i) value of 4 µM). The substrate oxidation reactions catalyzed by CYP2A6, CYP2B6, CYP2C19 and CYP3A4 were moderately (K(i) values of 35 to 45 µM), and those by CYP1A2, CYP2D6 and CYP2E1 were weakly inhibited by PFOS (K(i) values of 190-300 µM). The inhibition by PFOS for coumarin 7-hydroxylation mainly catalyzed by human liver microsomal CYP2A6 as well as by the recombinant enzyme was found to be enhanced by the preincubation of PFOS with human liver microsomes and NADPH as compared to the case without preincubation. The inhibition of the human liver microsomal cumarin 7-hydroxylation was PFOS concentration-dependent, and exhibited pseudo-first-order kinetics with respect to preincubation time, yielding K(inact) and K(I) values of 0.06 min(-1) and 23 µM, respectively. These results suggest that the metabolism of medicines which are substrates for CYP2C9 may be altered by PFOS in human bodies, and that PFOS is a mechanism-based inhibitor of CYP2A6.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Fluorocarbonos/farmacologia , Hidrocarboneto de Aril Hidroxilases/metabolismo , Biocatálise , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP2A6 , Humanos , Concentração Inibidora 50 , Cinética , Espécies Reativas de Oxigênio/metabolismo
12.
Carcinogenesis ; 27(9): 1768-77, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16571651

RESUMO

Sulforaphane (SFN), a naturally occurring isothiocyanate, is an attractive agent because of its potent anticancer effects. SFN suppresses the proliferation of various cancer cells in vitro and in vivo. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is also one of the most promising candidates for cancer therapeutics owing to its ability to selectively induce apoptosis in tumor cells. In this study, we report that SFN enhances TRAIL-induced apoptosis in human osteosarcoma cells, Saos2 and MG63. The apoptosis induced by co-treatment with SFN and TRAIL was markedly blocked by a dominant negative form of the TRAIL receptor or caspase inhibitors. The combined use of SFN and TRAIL effectively induced Bid cleavage and the activation of caspases 8, 10, 9 and 3 at ineffective concentrations for each agent. SFN upregulated the expression of death receptor 5 (DR5), a receptor for TRAIL, at mRNA and protein levels in a dose-dependent manner. In addition, the SFN-mediated sensitization to TRAIL was reduced by DR5 siRNA, suggesting that the sensitization was at least partially mediated through the induction of DR5 expression. Furthermore, SFN sensitized TRAIL-induced apoptosis in a p53-independent manner. On the other hand, SFN neither induced DR5 protein expression or enhanced TRAIL-induced apoptosis in normal human peripheral blood mononuclear cells. Thus, combined treatment with SFN and TRAIL might be a promising therapy for osteosarcoma.


Assuntos
Anticarcinógenos/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Regulação Neoplásica da Expressão Gênica , Glicoproteínas de Membrana/metabolismo , Osteossarcoma/patologia , Receptores do Fator de Necrose Tumoral/biossíntese , Tiocianatos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Antineoplásicos/farmacologia , Apoptose , Caspases/biossíntese , Linhagem Celular , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , Isotiocianatos , Leucócitos Mononucleares/metabolismo , Osteossarcoma/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Sulfóxidos , Ligante Indutor de Apoptose Relacionado a TNF
13.
Biochem Biophys Res Commun ; 333(3): 833-8, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-15963948

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is one of the most promising candidates for cancer therapeutics. However, some tumor cells are resistant to TRAIL-induced apoptosis. Our previous studies have shown that luteolin, a naturally occurring flavonoid, induces the up-regulation of death receptor 5 (DR5), which is a receptor for TRAIL. Here, we show for the first time that luteolin synergistically acts with exogenous soluble recombinant human TRAIL to induce apoptosis in HeLa cells, but not in normal human peripheral blood mononuclear cells. The combined use of luteolin and TRAIL induced Bid cleavage and the activation of caspase-8. Also, human recombinant DR5/Fc chimera protein, caspase inhibitors, and DR5 siRNA efficiently reduced apoptosis induced by co-treatment with luteolin and TRAIL. These results raise the possibility that this combined treatment with luteolin and TRAIL might be promising as a new therapy against cancer.


Assuntos
Apoptose/efeitos dos fármacos , Luteolina/farmacologia , Glicoproteínas de Membrana/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Neoplasias do Colo do Útero/patologia , Proteínas Reguladoras de Apoptose , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Proteínas de Transporte/metabolismo , Caspase 8 , Caspases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Ativação Enzimática , Feminino , Células HeLa , Humanos , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA