Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
J Biol Chem ; 289(47): 32628-38, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25296756

RESUMEN

Human plasminogen kringle 5 (K5) is known to display its potent anti-angiogenesis effect through inducing endothelial cell (EC) apoptosis, and the voltage-dependent anion channel 1 (VDAC1) has been identified as a receptor of K5. However, the exact role and underlying mechanisms of VDAC1 in K5-induced EC apoptosis remain elusive. In the current study, we showed that K5 increased the protein level of VDAC1, which initiated the mitochondrial apoptosis pathway of ECs. Our findings also showed that K5 inhibited the ubiquitin-dependent degradation of VDAC1 by promoting the phosphorylation of VDAC1, possibly at Ser-12 and Thr-107. The phosphorylated VDAC1 was attenuated by the AKT agonist, glycogen synthase kinase (GSK) 3ß inhibitor, and siRNA, suggesting that K5 increased VDAC1 phosphorylation via the AKT-GSK3ß pathway. Furthermore, K5 promoted cell surface translocation of VDAC1, and binding between K5 and VDAC1 was observed on the plasma membrane. HKI protein blocked the impact of K5 on the AKT-GSK3ß pathway by competitively inhibiting the interaction of K5 and cell surface VDAC1. Moreover, K5-induced EC apoptosis was suppressed by VDAC1 antibody. These data show for the first time that K5-induced EC apoptosis is mediated by the positive feedback loop of "VDAC1-AKT-GSK3ß-VDAC1," which may provide new perspectives on the mechanisms of K5-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Fragmentos de Péptidos/farmacología , Plasminógeno/farmacología , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Apoptosis/genética , Western Blotting , Caspasas/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Retroalimentación Fisiológica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fragmentos de Péptidos/genética , Fosforilación/efectos de los fármacos , Plasminógeno/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Ubiquitina/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/genética
2.
J Biol Chem ; 289(44): 30785-30799, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25225287

RESUMEN

Pigment epithelium-derived factor (PEDF), a potent antiangiogenesis agent, has recently attracted attention for targeting tumor cells in several types of tumors. However, less is known about the apoptosis-inducing effect of PEDF on human lung cancer cells and the underlying molecular events. Here we report that PEDF has a growth-suppressive and proapoptotic effect on lung cancer xenografts. Accordingly, in vitro, PEDF apparently induced apoptosis in A549 and Calu-3 cells, predominantly via the Fas-L/Fas death signaling pathway. Interestingly, A549 and Calu-3 cells are insensitive to the Fas-L/Fas apoptosis pathway because of the low level of cell surface Fas. Our results revealed that, in addition to the enhancement of Fas-L expression, PEDF increased the sensitivity of A549 and Calu-3 cells to Fas-L-mediated apoptosis by triggering the translocation of Fas protein to the plasma membrane in a p53- and FAP-1-dependent manner. Similarly, the up-regulation of Fas-L by PEDF was also mediated by p53. Furthermore, peroxisome proliferator-activated receptor γ was determined to be the upstream regulator of p53. Together, these findings uncover a novel mechanism of tumor cell apoptosis induced by PEDF and provide a potential therapeutic strategy for tumors that are insensitive to Fas-L/Fas-dependent apoptosis because of a low level of cell surface Fas.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Proteínas del Ojo/farmacología , Proteína Ligando Fas/genética , Factores de Crecimiento Nervioso/farmacología , Serpinas/farmacología , Proteína p53 Supresora de Tumor/fisiología , Receptor fas/metabolismo , Animales , Antineoplásicos/uso terapéutico , Caspasa 8/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Proteínas del Ojo/fisiología , Proteínas del Ojo/uso terapéutico , Proteína Ligando Fas/metabolismo , Humanos , Neoplasias Pulmonares , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/prevención & control , Factores de Crecimiento Nervioso/fisiología , Factores de Crecimiento Nervioso/uso terapéutico , PPAR gamma/metabolismo , Transporte de Proteínas , Proteína Tirosina Fosfatasa no Receptora Tipo 13/metabolismo , Serpinas/fisiología , Serpinas/uso terapéutico , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
3.
World J Gastroenterol ; 22(17): 4345-53, 2016 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-27158203

RESUMEN

AIM: To investigate the anti-apoptotic capability of the hepatitis B virus (HBV) in the HepG2 hepatoma cell line and the underlying mechanisms. METHODS: Cell viability and apoptosis were measured by MTT assay and flow cytometry, respectively. Targeted knockdown of manganese superoxide dismutase (MnSOD), AMP-activated protein kinase (AMPK) and hepatitis B virus X protein (HBx) genes as well as AMPK agonist AICAR and antagonist compound C were employed to determine the correlations of expression of these genes. RESULTS: HBV markedly protected the hepatoma cells from growth suppression and cell death in the condition of serum deprivation. A decrease of superoxide anion production accompanied with an increase of MnSOD expression and activity was found in HepG2.215 cells. Moreover, AMPK activation contributed to the up-regulation of MnSOD. HBx protein was identified to induce the expression of AMPK and MnSOD. CONCLUSION: Our results suggest that HBV suppresses mitochondrial superoxide level and exerts an anti-apoptotic effect by activating AMPK/MnSOD signaling pathway, which may provide a novel pharmacological strategy to prevent HCC.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Apoptosis , Carcinoma Hepatocelular/etiología , Virus de la Hepatitis B/patogenicidad , Neoplasias Hepáticas/etiología , Transducción de Señal/fisiología , Superóxido Dismutasa/fisiología , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Especies Reactivas de Oxígeno/metabolismo
4.
Int J Pharm ; 438(1-2): 1-10, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22974524

RESUMEN

Pigment epithelium-derived factor (PEDF) recombinant protein has been investigated in many kinds of solid tumors due to its potent antiangiogenic activity. However, the complexity of protein purification, instability of recombinant protein and requirement of repeated injections are obstacles for the recombinant PEDF therapy for solid tumors. We successfully synthesized polyethyleneglycol-polyetherimide (PEG-PEI) and cRGD-PEG-PEI which was coupled with a cyclic RGD peptide, a special ligand for integrin αvß3 receptor, as the vehicle for PEDF gene therapy in this study. In vitro, the competitive binding assay showed that cRGD contributed to the enhanced gene transfection efficiency of PEG-PEI in human umbilical vein endothelial cells (HUVECs). PEDF gene delivered by cRGD-PEG-PEI apparently suppressed growth of tumor with a 67.4% reduction and decreased microvessel density in nude mice bearing SW620 human colorectal xenografts. Accordingly, SW620 tumors from cRGD-PEG-PEI/PEDF-pcDNA3.1 (+)-treated mice expressed more PEDF than that of the control groups. Our study demonstrated that cRGD-PEG-PEI transported the PEDF gene into endothelia cells more efficiently than PEG-PEI, resulting in more effective inhibitory effects on tumor growth by anti-angiogenesis. Therefore, for the first time, we have explored an effective non-viral vehicle for PEDF gene therapy by targeting endothelial cells.


Asunto(s)
Neoplasias Colorrectales/terapia , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/genética , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/genética , Serpinas/administración & dosificación , Serpinas/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/patología , ADN/administración & dosificación , ADN/química , Proteínas del Ojo/química , Técnicas de Transferencia de Gen , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Nanopartículas/química , Factores de Crecimiento Nervioso/química , Oligopéptidos/química , Polietilenglicoles/química , Polietileneimina/análogos & derivados , Polietileneimina/química , Serpinas/química , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA