Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Differ ; 28(4): 1333-1346, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33168956

RESUMO

T-LAK-originated protein kinase (TOPK) overexpression is a feature of multiple cancers, yet is absent from most phenotypically normal tissues. As such, TOPK expression profiling and the development of TOPK-targeting pharmaceutical agents have raised hopes for its future potential in the development of targeted therapeutics. Results presented in this paper confirm the value of TOPK as a potential target for the treatment of solid tumours, and demonstrate the efficacy of a TOPK inhibitor (OTS964) when used in combination with radiation treatment. Using H460 and Calu-6 lung cancer xenograft models, we show that pharmaceutical inhibition of TOPK potentiates the efficacy of fractionated irradiation. Furthermore, we provide in vitro evidence that TOPK plays a hitherto unknown role during S phase, showing that TOPK depletion increases fork stalling and collapse under conditions of replication stress and exogenous DNA damage. Transient knockdown of TOPK was shown to impair recovery from fork stalling and to increase the formation of replication-associated single-stranded DNA foci in H460 lung cancer cells. We also show that TOPK interacts directly with CHK1 and Cdc25c, two key players in the checkpoint signalling pathway activated after replication fork collapse. This study thus provides novel insights into the mechanism by which TOPK activity supports the survival of cancer cells, facilitating checkpoint signalling in response to replication stress and DNA damage.


Assuntos
Quinase 1 do Ponto de Checagem/efeitos dos fármacos , Neoplasias Pulmonares/radioterapia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Tolerância a Radiação/efeitos dos fármacos , Fosfatases cdc25/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem/genética , Quinase 1 do Ponto de Checagem/efeitos da radiação , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Inibidores de Proteínas Quinases/farmacologia , Quinolonas/farmacologia , Tolerância a Radiação/genética , Transdução de Sinais , Taxa de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto , Fosfatases cdc25/genética , Fosfatases cdc25/efeitos da radiação
2.
Br J Cancer ; 117(8): 1154-1163, 2017 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-28873083

RESUMO

BACKGROUND: Overexpression of epidermal growth factor receptor (EGFR) occurs in approximately 90% of head and neck squamous cell carcinoma (HNSCC), and is correlated with poor prognosis. Thus, targeting EGFR is a promising strategy for treatment of HNSCC. Several small molecule EGFR inhibitors have been tested in clinical trials for treatment of HNSCC, but none of them are more effective than the current chemotherapeutic drugs. Thus, it is urgently needed to develop novel EGFR inhibitors for HNSCC treatment. METHODS: By screening an in-house focused library containing approximately 650 000 known kinase inhibitors and kinase inhibitor-like compounds containing common kinase inhibitor core scaffolds, we identified SKLB188 as a lead compound for inhibition of EGFR. The anticancer effects of SKLB188 on HNSCC cells were investigated by in vitro cell growth, cell cycle and apoptosis assays, as well as in vivo FaDu xenograft mouse model. Molecular docking, in vitro kinase profiling and western blotting were performed to characterise EGFR as the molecular target. RESULTS: SKLB188 inhibited HNSCC cell proliferation by inducing G1 cell cycle arrest, which was associated with downregulating the expression of Cdc25A, cyclins D1/A and cyclin-dependent kinases (CDK2/4), and upregulating the expression of cyclin-dependent kinase (CDK) inhibitors (p21Cip1 and p27Kip1), leading to decreased phosphorylation of Rb. SKLB188 also induced caspase-dependent apoptosis of HNSCC cells by downregulating the expression of Mcl-1 and survivin. Molecular docking revealed that SKLB188 could bind to the kinase domain of EGFR through hydrogen bonds and hydrophobic interactions. In vitro kinase assay showed that SKLB188 inhibited the activity of a recombinant human EGFR very potently (IC50=5 nM). Western blot analysis demonstrated that SKLB188 inhibited the phosphorylation of EGFR and its downstream targets, extracellular signal-regulated protein kinases 1 and 2 (Erk1/2) and Akt in the cells. In addition, SKLB188 dose-dependently inhibited FaDu xenograft growth in nude mice, and concurrently inhibited the phosphorylation of Erk1/2 and Akt in the tumours. CONCLUSIONS: SKLB188 potently inhibits the growth of HNSCC cells in vitro and in vivo by targeting EGFR signalling. The results provide a basis for further clinical investigation of SKLB188 as a targeted therapy for HNSCC. Our findings may open a new avenue for development of novel EGFR inhibitors for treatment of HNSCC and other cancers.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/metabolismo , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/metabolismo , Purinas/farmacologia , Animais , Western Blotting , Ciclina A/efeitos dos fármacos , Ciclina A/metabolismo , Ciclina D1/efeitos dos fármacos , Ciclina D1/metabolismo , Quinase 2 Dependente de Ciclina/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Regulação para Baixo , Receptores ErbB/metabolismo , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Camundongos , Camundongos Nus , Simulação de Acoplamento Molecular , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/metabolismo
3.
Oncol Rep ; 32(4): 1748-56, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25175641

RESUMO

Esophageal squamous cell carcinoma (ESCC) is an aggressive tumor with high incidence and mortality worldwide. Diallyl disulfide (DADS) is a natural organosulfur compound, isolated from garlic. In this study, MTT assay showed that DADS significantly reduced cell viability in a dose- and time-dependent manner in ESCC cells, with lower toxicity in normal liver cells. Cell cycle analysis revealed that DADS made G2/M phase arrest. Molecular analysis suggested that this cell cycle arrest was likely made by the decrease of cyclin B1, cdc2, p-cdc2, cdc25c in concomitance with activation of the p53/p21 pathway. Apoptosis was detected by Annexin V/PI staining. The molecule markers showed that DADS induced apoptosis through activating caspases, altering the Bax/Bcl-2 balance and suppressing the MEK-ERK pathway. Our data indicated that DADS has the potential to be an effective and safe anticancer agent for ESCC therapy in the near future.


Assuntos
Compostos Alílicos/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas , Dissulfetos/farmacologia , Neoplasias Esofágicas , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Proteína Quinase CDC2 , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclina B1/efeitos dos fármacos , Ciclina B1/genética , Quinases Ciclina-Dependentes/efeitos dos fármacos , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Carcinoma de Células Escamosas do Esôfago , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/genética , Proteínas rho de Ligação ao GTP/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/metabolismo
4.
Asian Pac J Cancer Prev ; 15(8): 3651-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24870773

RESUMO

Hepatocellular carcinoma (HCC) has a relatively higher incidence in many countries of Asia. Globally, HCC has a high fatality rate and short survival. Epirubicin, a doxorubicin analogue, may be administered alone or in combination with other agents to treat primary liver cancer and metastatic diseases. However, the toxic effects of epirubicin to normal tissues and cells have been one of the major obstacles to successful cancer chemotherapy. Here, we investigated the effects of epirubicin in combination with kappa-selenocarrageenan on mice with H22 implanted tumors and HepG-2 cell proliferation, immune organ index, morphology, cell cycle and related protein expressions in vivo and in vitro with sequential drug exposure. The inhibitory rate of tumor growth in vivo was calculated. Drug sensitivity was measured by MTT assay, and the King's principle was used to evaluate the interaction of drug combination. Morphological changes were observed by fluorescent microscopy. Cell cycle changes were analyzed by flow cytometry. Expression of cyclin A, Cdc25A and Cdk2 were detected by Western blotting. In vivo results demonstrated that the inhibitory rate of EPI combined with KSC was higher than that of KSC or EPI alone, and the Q value indicated an additive effect. In addition, KSC could significantly raise the thymus and spleen indices of mice with H22 implanted tumors. In the drug sensitivity assay in vitro, exposure to KSC and EPI simultaneously was more effective than exposure sequentially in HepG-2 cells, while exposure to KSC prior to EPI was more effective than exposure to EPI prior to KSC. Q values showed an additive effect in the simultaneous group and antagonistic effects in the sequential groups. Morphological analysis showed similar results to the drug sensitivity assay. Cell cycle analysis revealed that exposure to KSC or EPI alone arrested the cells in S phase in HepG-2 cells, exposure to KSC and EPI simultaneously caused accumulation in the S phase, an effect caused by either KSC or EPI. Expression of cyclin A, Cdc25A and Cdk2 protein was down-regulated following exposure to KSC and EPI alone or in combination, exposure to KSC and EPI simultaneously resulting in the lowest values. Taken together, our findings suggest that KSC in combination with EPI might have potential as a new therapeutic regimen against HCC.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Carcinoma Hepatocelular , Carragenina/farmacologia , Epirubicina/farmacologia , Neoplasias Hepáticas , Compostos Organosselênicos/farmacologia , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Animais , Ciclo Celular/efeitos dos fármacos , Ciclina A/efeitos dos fármacos , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/metabolismo , Esquema de Medicação , Células Hep G2 , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/metabolismo
5.
J Periodontal Res ; 48(1): 66-73, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22834967

RESUMO

BACKGROUND AND OBJECTIVE: Short-chain fatty acids, such as butyric acid and propionic acid, are metabolic by-products generated by periodontal microflora such as Porphyromonas gingivalis, and contribute to the pathogenesis of periodontitis. However, the effects of butyrate on the biological activities of gingival fibroblasts (GFs) are not well elucidated. MATERIAL AND METHODS: Human GFs were exposed to various concentrations of butyrate (0.5-16 mm) for 24 h. Viable cells that excluded trypan blue were counted. Cell cycle distribution of GFs was analyzed by propidium iodide-staining flow cytometry. Cellular reactive oxygen species (ROS) production was measured by flow cytometry using 2',7'-dichlorofluorescein (DCF). Total RNA and protein lysates were isolated and subjected to RT-PCR using specific primers or to western blotting using specific antibodies, respectively. RESULTS: Butyrate inhibited the growth of GFs, as indicated by a decrease in the number of viable cells. This event was associated with an induction of G0/G1 and G2/M cell cycle arrest by butyrate (4-16 mm) in GFs. However, no marked apoptosis of GFs was noted in this experimental condition. Butyrate (> 2 mm) inhibited the expression of cdc2, cdc25C and cyclinB1 mRNAs and reduced the levels of Cdc2, Cdc25C and cyclinB1 proteins in GFs, as determined using RT-PCR and western blotting, respectively. This toxic effect of butyrate was associated with the production of ROS. CONCLUSION: These results suggest that butyrate generated by periodontal pathogens may be involved in the pathogenesis of periodontal diseases via the induction of ROS production and the impairment of cell growth, cell cycle progression and expression of cell cycle-related genes in GFs. These events are important in the initiation and prolongation of inflammatory processes in periodontal diseases.


Assuntos
Butiratos/farmacologia , Fibroblastos/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Apoptose/efeitos dos fármacos , Butiratos/toxicidade , Proteína Quinase CDC2 , Técnicas de Cultura de Células , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Corantes , Ciclina B/efeitos dos fármacos , Ciclina B1/efeitos dos fármacos , Quinases Ciclina-Dependentes , Fibroblastos/citologia , Citometria de Fluxo , Fluoresceína-5-Isotiocianato , Fluoresceínas , Corantes Fluorescentes , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Gengiva/citologia , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Propídio , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fosfatases cdc25/efeitos dos fármacos
6.
Int Endod J ; 45(9): 848-58, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22486746

RESUMO

AIM: To evaluate the effect of TEGDMA on cell cycle progression as well as alterations of cell cycle-related gene and protein expression. METHODOLOGY: Human dental pulp cells were exposed to 0-5 mmol L(-1) TEGDMA for 24 h. Cytotoxicity was evaluated by 3-(4, 5-dimethyl-thiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay. Cell cycle progression was analysed by propidium iodide (PI) flow cytometry. Cell death pathway was surveyed by annexin V/PI dual-staining flow cytometry. The mRNA expression of cell cycle-related genes (cdc2, cyclinB1 and p21) and COX-2 was evaluated by reverse transcriptase-polymerase chain reaction, and their protein expression was evaluated by Western blotting. The production of PGE(2) and PGF(2α) in the culture medium was determined by enzyme-linked immunosorbent assay. RESULTS: Triethylene glycol dimethacrylate inhibited cellular growth and induced cell cycle deregulation in dental pulp cells. High-dose exposure provoked both necrotic and apoptotic cell death. The gene and protein expression of cdc2, cyclin B1 and cdc25C declined obviously whilst cells treated with 2.5 mmol L(-1) TEGDMA concurrent with the elevated expression of p21. The mRNA and protein expression of COX-2, along with production of PGE(2) and PGF(2α), are drastically raised by 2.5-5 mmol L(-1) TEGDMA. CONCLUSIONS: Triethylene glycol dimethacrylate induced cytotoxicity, cell cycle arrest and apoptosis in dental pulp cells, which was associated with the decline of cdc2, cyclin B1, cdc25C expression and elevation of p21 expression. Concomitantly, COX-2 expression, PGE(2) and PGF(2α) production increased. These effects may contribute to explain the pulpal damage and inflammation induced by TEGDMA after operative procedures.


Assuntos
Ciclo-Oxigenase 2/efeitos dos fármacos , Materiais Dentários/toxicidade , Polpa Dentária/efeitos dos fármacos , Polietilenoglicóis/toxicidade , Ácidos Polimetacrílicos/toxicidade , Prostaglandinas/biossíntese , Anexina A5/farmacologia , Apoptose/efeitos dos fármacos , Proteína Quinase CDC2 , Técnicas de Cultura de Células , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Corantes , Ciclina B/efeitos dos fármacos , Ciclina B1/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/efeitos dos fármacos , Quinases Ciclina-Dependentes , Polpa Dentária/citologia , Dinoprosta/análise , Dinoprostona/análise , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo/métodos , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Humanos , Necrose , Propídio , Sais de Tetrazólio , Tiazóis , Fatores de Tempo , Fosfatases cdc25/efeitos dos fármacos
7.
Anticancer Drugs ; 21(9): 814-22, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20724916

RESUMO

3,3'-Diindolylmethane (DIM) is a potential chemopreventive phytochemical derived from Brassica vegetables. In this study, we assessed the effects of DIM on cell cycle regulation in both estrogen-dependent MCF-7 and estrogen receptor negative p53 mutant MDA-MB-468 human breast cancer cells. In-vitro culture studies showed that DIM dose dependently inhibited the proliferation of both cells. In addition, in-vivo xenograft model showed that DIM strongly inhibited the development of human breast tumors. Fluorescence activated cell sorter analysis showed a DIM-mediated G2/M cell cycle arrest in MCF-7 and MDA-MB-468 cells. Western blot analysis showed that DIM downregulated the expression of cyclin-dependent kinases 2 and 4 and Cdc25A, which plays an important role in G2/M phase. Furthermore, treatment of MCF-7 cells with DIM, which increased microRNA 21 expression, caused a downregulation of Cdc25A, resulting in an inhibition of breast cancer cell proliferation. Taken together, our data show that DIM is able to stop the cell cycle progression of human breast cancer cells regardless of their estrogen-dependence and p53 status, by differentially modulating cell cycle regulatory pathways. The modulation of microRNA 21 mediates the DIM cell cycle regulator effect in MCF-7 cells.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Indóis/farmacologia , MicroRNAs/metabolismo , Animais , Anticarcinógenos/administração & dosagem , Anticarcinógenos/farmacologia , Antineoplásicos/administração & dosagem , Western Blotting , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Fase G2/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Indóis/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/genética
8.
Carcinogenesis ; 29(6): 1258-66, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18495657

RESUMO

Regular consumption of mesalazine has been associated with a reduced risk of colorectal cancer (CRC) in patients with inflammatory bowel disease. The molecular mechanisms underlying the antineoplastic effect of 5-aminosalicylic acid remain, however, poorly characterized. In this study, we examined whether mesalazine affects cell cycle progression and analyzed specific checkpoint pathways in experimental models of CRC. Mesalazine inhibited the growth of HCT-116 and HT-29 cells, two CRC cell lines that express either a wild-type or mutated p53. Cell cycle analysis revealed that mesalazine induced cells to accumulate in S phase. This effect was associated with a sustained phosphorylation of the cyclin-dependent kinase (CDK)2 at threonine 14 and tyrosine 15 residues, an event that inactivates the CDK2-cyclin complex and blocks S-G(2) phase cell cycle transition. Consistently, mesalazine reduced the protein content of CDC25A, a phosphatase that regulates CDK2 phosphorylation status. Analysis of upstream kinases that negatively control CDC25A expression showed that mesalazine enhanced the activation of CHK1 and CHK2. However, silencing of CHK1 and CHK2 did not prevent the mesalazine-induced CDC25A protein downregulation. In contrast, CDC25A protein ubiquitination and degradation and accumulation of cells in S phase following mesalazine exposure were reverted by proteasome inhibitors. Notably, mesalazine also inhibited CDC25A in human CRC explants. Finally, we showed that mesalazine downregulated CDC25A in CT26, a murine CRC cell line, and prevented the formation of CT26-derived tumors in mice. Data show that mesalazine negatively regulates CDC25A protein expression, thus delaying CRC cell progression.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Neoplasias do Colo/metabolismo , Mesalamina/farmacologia , Fase S/efeitos dos fármacos , Fosfatases cdc25/efeitos dos fármacos , Animais , Western Blotting , Proliferação de Células/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Imunoprecipitação , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fosfatases cdc25/biossíntese
9.
Int J Cancer ; 123(1): 41-50, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18435416

RESUMO

Prostate cancer (PCa) is the leading cause of cancer-related deaths in men; urgent measures are warranted to lower this deadly malignancy. Silymarin is a known cancer chemopreventive agent, but the relative anticancer efficacy of its constituents is still unknown. Here, we compared the efficacy of 7 pure flavonolignan compounds isolated from silymarin, namely silybin A, silybin B, isosilybin A, isosilybin B, silydianin, isosilydianin, silychristin and isosilychristin, in advanced human PCa PC3 cells. Silybin A, silybin B, isosilybin A, isosilybin B, silibinin and silymarin strongly inhibited the colony formation by PC3 cells (p < 0.001), while silydianin, silychristin and isosilychristin had marginal effect (p < 0.05). Using cell growth and death assays, we identified isosilybin B as the most effective isomer. FACS analysis for cell cycle also showed that silybin A, silybin B, isosilybin A, isosilybin B, silibinin and silymarin treatment resulted in strong cell cycle arrest in PC3 cells after 72 hr of treatment, while the effect of silydianin, silychristin and isosilychristin was marginal (if any). Western blot analysis also showed the differential effect of these compounds on the levels of cell cycle regulators-cyclins (D, E, A and B), CDKs (Cdk2, 4 and Cdc2), CDKIs (p21 and p27) and other cell cycle regulators (Skp2, Cdc25A, B, C and Chk2). This study provided further evidence for differential anticancer potential among each silymarin constituent, which would have potential implications in devising better formulations of silymarin against prostate and other cancers.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/prevenção & controle , Substâncias Protetoras/farmacologia , Silimarina/farmacologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proteínas Inibidoras de Quinase Dependente de Ciclina/efeitos dos fármacos , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/efeitos dos fármacos , Ciclinas/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Flavonolignanos/farmacologia , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Isomerismo , Masculino , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Silibina , Silimarina/análogos & derivados , Silimarina/química , Fatores de Tempo , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/metabolismo
10.
Toxicology ; 246(2-3): 180-7, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18321631

RESUMO

Our previous results have indicated that Cdc25A is involved in benzo(a)pyrene (BaP)-induced S-phase checkpoint in 16HBE cells and A549 cells. In this paper, we reported the changes of the downstream molecular pathway of Cdc25A and the effects of over-expression and suppression of Cdc25A on BaP-induced S-phase checkpoint. In the S-phase checkpoint induced by BaP the reduction of Cdc25A contributes to cyclin A inhibition. Over-expression of Cdc25A abrogated BaP-induced S-phase arrest in 16HBE cells and concomitantly the expression levels of Cdk2 and cyclin A were not obviously changed by BaP when compared with the control. Cdc25A down-regulation by RNA interference (RNAi) prolonged the S-phase arrest induced by BaP and decreased clearly the expression levels of cyclin A and cyclin E. Therefore, our results further demonstrated that Cdc25A was an effector in Chk1-Cdc25A-cyclin A/Cdk2 pathway of S-phase checkpoint elicited by the carcinogen BaP in 16HBE cells.


Assuntos
Benzo(a)pireno/toxicidade , Carcinógenos Ambientais/toxicidade , Fase S/efeitos dos fármacos , Fosfatases cdc25/metabolismo , Animais , Linhagem Celular Tumoral , Ciclina A/genética , Ciclina A/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Humanos , Interferência de RNA/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Transfecção , Fosfatases cdc25/efeitos dos fármacos
11.
Nat Rev Cancer ; 7(3): 202-11, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17287826

RESUMO

Transient protein-protein interactions have key regulatory functions in many of the cellular processes that are implicated in cancerous growth, particularly the cell cycle. Targeting these transient interactions as therapeutic targets for anticancer drug development seems like a good idea, but it is not a trivial task. This Review discusses the issues and difficulties that are encountered when considering these transient interactions as drug targets, using the example of the cell division cycle 25 (Cdc25) phosphatases and their cyclin-dependent kinase (CDK)-cyclin protein substrates.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Inibidores Enzimáticos/farmacologia , Transdução de Sinais/fisiologia , Fosfatases cdc25/metabolismo , Animais , Ciclo Celular/fisiologia , Quinases Ciclina-Dependentes/química , Quinases Ciclina-Dependentes/efeitos dos fármacos , Humanos , Neoplasias/enzimologia , Ligação Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína , Fosfatases cdc25/química , Fosfatases cdc25/efeitos dos fármacos
12.
Mol Cancer Ther ; 5(8): 1935-43, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16928813

RESUMO

Mammalian cells initiate cell cycle arrest at different phases of the cell cycle in response to various forms of genotoxic stress to allow time for DNA repair, and thus preserving their genomic integrity. The protein kinases checkpoint kinase 1 (Chk1), checkpoint kinase 2 (Chk2), and mitogen-activated protein kinase-activated protein kinase 2 (MK2) have all been shown to be involved in cell cycle checkpoint control. Recently, cell cycle checkpoint abrogation has been proposed as one way to sensitize cancer cells to DNA-damaging agents due to the expected induction of mitotic catastrophe. Due to their overlapping substrate spectra and redundant functions, it is still not clear which kinase is mainly responsible for the cell cycle arrests conferred by clinically relevant chemotherapeutics. Thus, the issue remains about which kinase is the most therapeutically relevant target and, more importantly, whether multiple kinases might need to be targeted to achieve the best efficacy in light of recent studies showing superior efficacy for pan-receptor tyrosine kinase inhibitors. To clarify this issue, we investigated the roles of the three kinases in response to different genotoxic stresses through small interfering RNA-mediated specific target knockdowns. Our result showed that only the down-regulation of Chk1, but not of Chk2 or MK2, abrogated camptothecin- or 5-fluorouracil-induced S-phase arrest or doxorubicin-induced G(2)-phase arrest. This was followed by mitotic catastrophe and apoptosis. Moreover, double inhibition of Chk1 and Chk2 failed to achieve better efficacy than Chk1 inhibition alone; surprisingly, inhibition of MK2, in addition to Chk1 suppression, partially reversed the checkpoint abrogation and negated mitotic catastrophe. We further showed that this is due to the fact that in MK2-deficient cells, Cdc25A protein, which is critically required for the mitotic progression following checkpoint abrogation, becomes greatly depleted. In summary, our findings show that Chk1 is the only relevant checkpoint kinase as a cancer drug target and inhibition of other checkpoint kinases in addition to Chk1 would be nonproductive.


Assuntos
Ciclo Celular/fisiologia , Dano ao DNA/fisiologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Antineoplásicos/farmacologia , Camptotecina/farmacologia , Ciclo Celular/efeitos dos fármacos , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Doxorrubicina/farmacologia , Feminino , Fluoruracila/farmacologia , Células HeLa , Histonas/efeitos dos fármacos , Histonas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias/tratamento farmacológico , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno , Células Tumorais Cultivadas , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/metabolismo
13.
J Cell Biochem ; 99(1): 209-20, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16598773

RESUMO

Cdc25A is a potent tyrosine phosphatase that catalyzes specific dephosphorylation of cyclin/cyclin-dependent kinase (cdk) complexes to regulate G1 to S-phase cell cycle progression. Cdc25A mRNA levels are induced by 17beta-estradiol (E2) in ZR-75 breast cancer cells, and deletion analysis of the cdc25A promoter identified the -151 to -12 region as the minimal E2-responsive sequence. Subsequent mutation/deletion analysis showed that at least three different cis-elements were involved in activation of cdc25A by E2, namely, GC-rich Sp1 binding sites, CCAAT motifs that bind NF-Y, and E2F sites that bind DP/E2F1 proteins. Studies with inhibitors and dominant negative expression plasmids show that E2 activates cdc25A expression through activation of genomic ERalpha/Sp1 and E2F1 and cAMP-dependent activation of NF-YA. Thus, both genomic and non-genomic pathways of estrogen action are involved in induction of cdc25A in breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Estradiol/farmacologia , Regulação Neoplásica da Expressão Gênica , Fosfatases cdc25/genética , Composição de Bases , Sítios de Ligação , Neoplasias da Mama/tratamento farmacológico , Fator de Ligação a CCAAT/genética , Fator de Ligação a CCAAT/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Mutação , Regiões Promotoras Genéticas , Elementos de Resposta , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição TFIIB/genética , Fator de Transcrição TFIIB/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/metabolismo
14.
Free Radic Biol Med ; 40(2): 247-59, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16413407

RESUMO

In mammalian cells DNA damage activates a checkpoint that halts progression through S phase. To determine the ability of nitrating agents to induce S-phase arrest, mouse C10 cells synchronized in S phase were treated with nitrogen dioxide (NO(2)) or SIN-1, a generator of reactive nitrogen species (RNS). SIN-1 or NO(2) induced S-phase arrest in a dose- and time-dependent manner. As for the positive controls adozelesin and cisplatin, arrest was accompanied by phosphorylation of ATM kinase; dephosphorylation of pRB; decreases in RF-C, cyclin D1, Cdc25A, and Cdc6; and increases in p21. Comet assays indicated that RNS induce minimal DNA damage. Moreover, in a cell-free replication system, nuclei from cells treated with RNS were able to support control levels of DNA synthesis when incubated in cytosolic extracts from untreated cells, whereas nuclei from cells treated with cisplatin were not. Induction of phosphatase activity may represent one mechanism of RNS-induced arrest, for the PP1/PP2A phosphatase inhibitor okadaic acid inhibited dephosphorylation of pRB; prevented decreases in the levels of RF-C, cyclin D1, Cdc6, and Cdc25A; and bypassed arrest by SIN-1 or NO(2), but not cisplatin or adozelesin. Our studies suggest that RNS may induce S-phase arrest through mechanisms that differ from those elicited by classical DNA-damaging agents.


Assuntos
Ácido Okadáico/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Espécies Reativas de Nitrogênio/farmacologia , Fase S/efeitos dos fármacos , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Células Cultivadas , Cisplatino/farmacologia , Ciclina D1/efeitos dos fármacos , Ciclina D1/metabolismo , DNA/biossíntese , DNA/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Técnicas In Vitro , Camundongos , Molsidomina/análogos & derivados , Molsidomina/farmacologia , Dióxido de Nitrogênio/farmacologia , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Nitrogênio/antagonistas & inibidores , Espécies Reativas de Nitrogênio/metabolismo , Proteína de Replicação C/efeitos dos fármacos , Proteína de Replicação C/metabolismo , Proteína do Retinoblastoma/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Fatores de Tempo , Proteínas Supressoras de Tumor/efeitos dos fármacos , Proteínas Supressoras de Tumor/metabolismo , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/metabolismo
15.
J Cancer Res Clin Oncol ; 132(3): 150-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16283383

RESUMO

PURPOSE: Hsp90, a molecular chaperone, is involved in folding, assembly, maturation, and stabilization of the client proteins which regulate survival of cancer cells, and thus Hsp90 inhibitors may be potential molecular targeting agents for cancer treatment. We investigated whether Hsp90 inhibitors have therapeutic value in lung cancer. METHODS: First, expression levels of Hsp90 in lung cancer cells were examined by western blotting and immunohistochemical analyses. Next, the effect of Hsp90 inhibitors, geldanamycin and 17-allylaminogeldanamycin (17-AAG), on lung cancer cell growth was examined. RESULTS: Remarkable high expression of Hsp90 protein in lung cancer cell lines and a more intense signal for Hsp90 by immunohistochemistry in males, patients with smoking index over 600, and squamous cell carcinoma were observed. Both Hsp90 inhibitors dose dependently inhibited the growth of lung cancer cell lines and induced G2/M arrest concomitant with decreased protein levels of Cdc25C and Cdc2. Moreover, combination of an Hsp90 inhibitor and irradiation had an additive effect on cell growth inhibition and reduction of Cdc25C and Cdc2 protein levels. CONCLUSION: Hsp90 inhibitor is thus a therapeutic tool for lung cancer based on its target proteins, which are involved in tumor progression and antiproliferative activity in lung cancer cells.


Assuntos
Benzoquinonas/farmacologia , Proteína Quinase CDC2/efeitos dos fármacos , Proteínas de Ciclo Celular/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Lactamas Macrocíclicas/farmacologia , Neoplasias Pulmonares/metabolismo , Fosfatases cdc25/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Western Blotting , Proteína Quinase CDC2/biossíntese , Proteínas de Ciclo Celular/biossíntese , Linhagem Celular Tumoral , Feminino , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/biossíntese , Proteínas de Choque Térmico HSP90/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Imunoprecipitação , Masculino , Fosfatases cdc25/biossíntese
17.
Nat Prod Res ; 18(1): 1-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14974610

RESUMO

The CDC25 phosphatases regulate the cell division cycle by controlling the activity of cyclin-dependent kinases. While screening for inhibitors of phosphatases among natural products we repeatedly found that some polyprenyl-hydroquinones and polyprenyl-furans (furanoterpenoids) (furospongins, furospinosulins) were potent CDC25 phosphatase inhibitors. These compounds were extracted, isolated and identified independently from three sponge species (Spongia officinalis, Ircinia spinulosa, Ircinia muscarum), collected at different locations in the Mediterranean Sea. The compounds were inactive on the Ser/Thr phosphatase PP2C-alpha and on three kinases (CDK1, CDK5, GSK-3), suggesting that some potent and selective CDC25 phosphatase might be designed from these initial structures.


Assuntos
Ciclo Celular/efeitos dos fármacos , Furanos/farmacologia , Hidroquinonas/farmacologia , Poríferos/química , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/farmacologia , Animais , Escherichia coli , Furanos/isolamento & purificação , Hidroquinonas/isolamento & purificação
19.
Oncol Res ; 13(6-10): 347-52, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12725524

RESUMO

Small molecules provide exceptionally useful tools for probing signaling targets relevant for cancer and stem cell differentiation. In contrast to genetic approaches, the application of small molecules generally offers a graded and reversible disruption of a particular pathway. The vast array of theoretical chemical entities that exist in the chemical universe are now becoming available through the production and distribution of chemical libraries generated by both traditional and combinatorial methods, which are suitable for pharmacological use. Convenient and inexpensive cell-free and cell-based assays can be used to identify chemicals that exhibit desirable antisignaling properties. We illustrate a model of how agents targeted against signaling macromolecules involved in cancer, namely dual specificity protein phosphatases, can be identified.


Assuntos
Inibidores Enzimáticos/farmacologia , Transdução de Sinais/fisiologia , Fosfatases cdc25/efeitos dos fármacos , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Humanos , Modelos Biológicos , Modelos Moleculares , Fosfatases cdc25/classificação
20.
Glia ; 40(1): 44-54, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12237842

RESUMO

Temozolomide (TZM) is a novel methylating agent currently under investigation for treatment of recurrent high-grade gliomas. Although TZM generates a wide spectrum of methyl adducts, its cytotoxicity has been attributed to mismatch repair (MR)-mediated processing of O(6)-methylguanine:T mispairs. N3-methyladenine and N7-methylguanine adducts are promptly repaired by the base excision repair system, unless a poly(ADP-ribose) polymerase (PARP) inhibitor is combined to TZM. In this case, the repair process of N-methylpurines cannot be completed and the deriving DNA strand breaks contribute to cytotoxicity. In this study, we investigated the influence on cell growth and cell cycle of treatment with TZM + PARP inhibitor in glioma cells characterized by different susceptibility to TZM. The results indicated that PARP inhibitor increases growth inhibition induced by TZM in either p53-wild-type or p53-mutant glioblastoma cells, as early as 24 h after drug exposure. The enhancing effect exerted by PARP inhibitor was particularly evident in glioma cells characterized by a defective expression of MR, since these cells are tolerant to O(6)-methylguanine damage and show low sensitivity to TZM. In O(6)-alkylguanine-DNA alkyltransferase (OGAT)-deficient and MR-proficient tumor cells bearing wild-type p53, the drug combination markedly reduced cell accumulation in the G(2)/M phase of cell cycle and induction of the G(2) checkpoint regulator Chk1 kinase. In short-term cultures of glioma cells derived from surgical specimens, PARP inhibitor enhanced chemosensitivity to TZM and this effect was especially evident in OGAT-proficient tumors. Thus, a pharmacological strategy based on the interruption of N-methylpurine repair might represent a novel strategy to restore or increase glioma sensitivity to TZM.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Benzamidas/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Inibidores Enzimáticos/farmacologia , Glioma/tratamento farmacológico , Proteínas/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinase 1 do Ponto de Checagem , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Interações Medicamentosas , Resistencia a Medicamentos Antineoplásicos/genética , Quimioterapia Combinada , Fase G2/efeitos dos fármacos , Fase G2/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/enzimologia , Glioblastoma/genética , Glioma/enzimologia , Glioma/genética , Humanos , Mitose/efeitos dos fármacos , Mitose/genética , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas/metabolismo , Temozolomida , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Fosfatases cdc25/efeitos dos fármacos , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...