Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 32(43): 5144-55, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-23178493

RESUMO

Although radiotherapy improves survival in patients, glioblastoma multiformes (GBMs) tend to relapse with augmented tumor migration and invasion even after ionizing radiation (IR). Aberrant nuclear factor-κB (NF-κB) and signal transducer and activator of transcription factor 3 (Stat3) activation and interaction have been suggested in several human tumors. However, possible NF-κB/Stat3 interaction and the role of Stat3 in maintenance of NF-κB nuclear retention in GBM still remain unknown. Stat3 and NF-κB (p65) physically interact with one another in the nucleus in glioma tumors. Most importantly, glutathione S-transferase pull-down assays identified that Stat3 binds to the p65 transactivation domain and is present in the NF-κB DNA-binding complex. Irradiation significantly elevated nuclear phospho-p65/phospho-Stat3 interaction in correlation with increased intercellular adhesion molecule-1 (ICAM-1) and soluble-ICAM-1 levels, migration and invasion in human glioma xenograft cell lines 4910 and 5310. Chromatin immunopreicipitation and promoter luciferase activity assays confirmed the critical role of adjacent NF-κB (+399) and Stat3 (+479) binding motifs in the proximal intron-1 in elevating IR-induced ICAM-1 expression. Specific inhibition of Stat3 or NF-κB with Stat3.siRNA or JSH-23 severely inhibited IR-induced p65 recruitment onto ICAM-1 intron-1 and suppressed migratory properties in both the cell lines. On the other hand, Stat3C- or IR-induced Stat3 promoter recruitment was significantly decreased in p65-knockdown cells, thereby suggesting the reciprocal regulation between p65 and Stat3. We also observed a significant increase in NF-κB enrichment on ICAM-1 intron-1 and ICAM-1 transactivation in Stat3C overexpressing cells. In in vivo orthotopic experiments, suppression of tumor growth in Stat3.si+IR-treated mice was associated with the inhibition of IR-induced p-p65/p-Stat3 nuclear colocalization and ICAM-1 levels. To our knowledge, this is the first study showing the crucial role of NF-κB/Stat3 nuclear association in IR-induced ICAM-1 regulation and implies that targeting NF-κB/Stat3 interaction may have future therapeutic significance in glioma treatment.


Assuntos
Neoplasias Encefálicas/genética , Glioma/genética , Molécula 1 de Adesão Intercelular/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos da radiação , Sequência Consenso , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioma/patologia , Humanos , Molécula 1 de Adesão Intercelular/genética , Íntrons/genética , Camundongos , NF-kappa B/metabolismo , Invasividade Neoplásica/genética , Radiação , Fator de Transcrição STAT3/genética , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Cell Death Dis ; 3: e445, 2012 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-23254288

RESUMO

Gliomas display anoikis resistance, enhanced invasion in to the adjacent brain parenchyma and eventually recur despite using the standard therapies. Our studies on increased anoikis sensitization in matrix metalloproteinase-2 (MMP-2)-knockdown 4910 and 5310 human glioma xenograft cells were interestingly correlated with p21-activated kinase 4 (PAK4) inhibition, prompting us to further investigate the role of PAK4 in glioma. Here, we report the PAK4 upregulation in positive correlation with increasing glioma pathological grades. The siRNA-mediated PAK4 knockdown elevated anoikis, and inhibited invasion and migration by downregulating MMP-2, αvß3-integrin and phospho-epidermal growth factor receptor (phospho-EGFR). The cDNA-PCR arrays revealed a transcriptional suppression of essential proteins involved in cell proliferation and adhesion in PAK4-knockdown cells. Most importantly, glutathione S-transferase pull-down assays demonstrated the MMP-2 as a new PAK4-interacting protein which binds to PAK4 kinase domain. Individual EGFR/ErbB2 inhibitor and αvß3 antibody treatments in PAK4si-treated cells indicated the regulation of αvß3/EGFR survival signaling by PAK4. Overexpression of PAK4 significantly reversed the MMP2si-induced cell death in both cell lines. Codepletion of PAK4 and MMP-2 resulted in robust anoikis-mediated cell death, and severely inhibited invasive and migratory properties in these cells. PAK4si inhibited in vivo tumor growth in nude mice by inhibiting MMP-2, ß3-integrin and phospho-EGFR levels in tumors. Our findings indicate a physical association between PAK4 and MMP-2, and suggest the future therapeutic potential of PAK4/MMP-2 dual targeting in glioma treatment.


Assuntos
Anoikis , Movimento Celular , Glioma/metabolismo , Glioma/patologia , Metaloproteinase 2 da Matriz/metabolismo , Quinases Ativadas por p21/metabolismo , Animais , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Feminino , Glioma/genética , Glioma/fisiopatologia , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Metaloproteinase 2 da Matriz/genética , Camundongos , Camundongos Nus , Invasividade Neoplásica , Ligação Proteica , Transdução de Sinais , Transplante Heterólogo , Quinases Ativadas por p21/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...