Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Cancer Res ; 23(20): 6215-6226, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28679777

RESUMEN

Purpose: HSP90, a highly conserved molecular chaperone that regulates the function of several oncogenic client proteins, is altered in glioblastoma. However, HSP90 inhibitors currently in clinical trials are short-acting, have unacceptable toxicities, or are unable to cross the blood-brain barrier (BBB). We examined the efficacy of onalespib, a potent, long-acting novel HSP90 inhibitor as a single agent and in combination with temozolomide (TMZ) against gliomas in vitro and in vivoExperimental Design: The effect of onalespib on HSP90, its client proteins, and on the biology of glioma cell lines and patient-derived glioma-initiating cells (GSC) was determined. Brain and plasma pharmacokinetics of onalespib and its ability to inhibit HSP90 in vivo were assessed in non-tumor-bearing mice. Its efficacy as a single agent or in combination with TMZ was assessed in vitro and in vivo using zebrafish and patient-derived GSC xenograft mouse glioma models.Results: Onalespib-mediated HSP90 inhibition depleted several survival-promoting client proteins such as EGFR, EGFRvIII, and AKT, disrupted their downstream signaling, and decreased the proliferation, migration, angiogenesis, and survival of glioma cell lines and GSCs. Onalespib effectively crossed the BBB to inhibit HSP90 in vivo and extended survival as a single agent in zebrafish xenografts and in combination with TMZ in both zebrafish and GSC mouse xenografts.Conclusions: Our results demonstrate the long-acting effects of onalespib against gliomas in vitro and in vivo, which combined with its ability to cross the BBB support its development as a potential therapeutic agent in combination with TMZ against gliomas. Clin Cancer Res; 23(20); 6215-26. ©2017 AACR.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Dacarbazina/análogos & derivados , Glioma/metabolismo , Glioma/patología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Isoindoles/farmacología , Animales , Antineoplásicos/farmacocinética , Benzamidas/farmacocinética , Barrera Hematoencefálica/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dacarbazina/farmacocinética , Dacarbazina/farmacología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Expresión Génica , Glioma/tratamiento farmacológico , Glioma/mortalidad , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Isoindoles/farmacocinética , Ratones , Células Madre Neoplásicas/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/efectos de los fármacos , Tasa de Supervivencia , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
2.
Stem Cell Res ; 12(3): 716-29, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24699410

RESUMEN

In the present study, we investigated the effect of simultaneous downregulation of uPAR and cathepsin B (pUC), alone or in combination with radiation, on JNK-MAPK signaling pathway in regulating the migration of non-GICs (glioma-initiating cells) and GICs. The increase in the expression of p-JNK with pUC treatment was mostly localized to nucleus whereas increase in the expression of p-JNK with radiation and overexpression of uPAR and cathepsin B was confined to cytoplasm of the cells. Depletion of cytosolic p-JNK with pUC treatment inhibited migration by downregulating the expression of the adapter proteins of the focal adhesion complex. We also observed that knockdown of uPAR and cathepsin B regulated the Ras-Pak-1 pathway to induce the translocation of p-JNK from cytosol to nucleus. In control cells, Pak-1 served as a functional inhibitor for MEKK-1, which inhibits the complex formation of MEKK-1 and p-JNK and thus inhibits the translocation of this complex into nucleus. Hence, we conclude that glioma cells utilize the availability of cytosolic p-JNK in driving the cells towards migration. Finally, treating the cells with pUC alone or in combination with radiation induced the translocation of the MEKK-1-p-JNK complex from cytosol to nucleus, thereby inhibiting the migration of glioma cells.


Asunto(s)
Catepsina B/metabolismo , Movimiento Celular , Glioma/enzimología , MAP Quinasa Quinasa 4/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Animales , Catepsina B/genética , Núcleo Celular/enzimología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/enzimología , Citoplasma/genética , Citoplasma/metabolismo , Citosol/enzimología , Citosol/metabolismo , Femenino , Glioma/genética , Glioma/metabolismo , Glioma/fisiopatología , Humanos , MAP Quinasa Quinasa 4/genética , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Transporte de Proteínas , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética
3.
Neurochem Res ; 38(11): 2313-22, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24013885

RESUMEN

Glioblastoma, the most common and aggressive primary brain tumors, carry a bleak prognosis and often recur even after standard treatment modalities. Emerging evidence suggests that deregulation of the Wnt/ß-catenin/Tcf signaling pathway contributes to glioblastoma progression. Nonsteroidal anti-inflammatory drugs (NSAIDs) inhibit tumor cell proliferation by suppressing Wnt/ß-catenin/Tcf signaling in various human malignancies. In this study, we sought to inhibit Wnt/ß-catenin/Tcf signaling in glioblastoma cells by the NSAIDs diclofenac and celecoxib. Both diclofenac and celecoxib significantly reduced the proliferation, colony formation and migration of human glioblastoma cells. Diclofenac and celecoxib downregulated ß-catenin/Tcf reporter activity. Western and qRT-PCR analysis showed that diclofenac and celecoxib reduced the expression of ß-catenin target genes Axin2, cyclin D1 and c-Myc. In addition, the cytoplasmic accumulation and nuclear translocation of ß-catenin was significantly reduced following diclofenac and celecoxib treatment. Furthermore, diclofenac and celecoxib significantly increased phosphorylation of ß-catenin and reduced the phosphorylation of GSK3ß. These results clearly indicated that diclofenac and celecoxib are potential therapeutic agents against glioblastoma cells that act by suppressing the activation of Wnt/ß-catenin/Tcf signaling.


Asunto(s)
Diclofenaco/farmacología , Glioblastoma/metabolismo , Pirazoles/farmacología , Sulfonamidas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Antiinflamatorios no Esteroideos/farmacología , Celecoxib , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Factores de Transcripción TCF/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
4.
Biochem Biophys Res Commun ; 434(3): 627-33, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23583374

RESUMEN

MicroRNAs are a novel family of small non-coding RNAs that regulate the expression of several genes involved in normal development as well as human disorders including cancer. Here we show that miR-874 plays a tumor suppressor role in non-small cell lung cancer (NSCLC) in vitro and in vivo. In silico target prediction analysis revealed numerous genes associated with tumor progression including MMP-2 and uPA as the putative target genes of miR-874. Our preliminary in situ hybridization experiments demonstrated the diminution of miR-874 expression in lung cancer tissues compared to their normal counter parts. Overexpression of miR-874 in CD133-positive cancer stem cell (CSC) population led to a significant loss in CSC-phenotype and enhanced sphere de-differentiation into epithelial-like cells. Restoration of miR-874 expression drastically reduced cell invading ability in comparison to mock and control-miR-treated cells by suppressing the protein levels of MMP-2 and uPA. In in vivo experiments, miR-874 treatment decreased orthotopic tumor growth in nude mice compared to mock and control-miR treatments. Further, the immunoreactivity of human anti-MMP-2 and anti-uPA was significantly reduced in tumor sections from mice that received miR-874 treatment. In conclusion, our study highlights the possible tumor suppressor role of miR-874 in NSCLC-initiating cells and suggests miR-874 as a potential target in the treatment of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , División Celular/genética , Neoplasias Pulmonares/patología , MicroARNs/fisiología , Invasividad Neoplásica/genética , Secuencia de Bases , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Cartilla de ADN , Humanos , Inmunohistoquímica , Hibridación in Situ , Neoplasias Pulmonares/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Int J Oncol ; 42(4): 1279-88, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23381805

RESUMEN

The majority of glioblastoma multiforme (GBM) tumors recur after radiation (IR) treatment due to increased angiogenesis and IR-induced signaling events in endothelial cells (ECs) that are involved in tumor neovascularization; however, these signaling events have yet to be well characterized. In the present study, we observed that IR (8 Gy) significantly elevated MMP-2 expression and gelatinolytic activity in 4910 and 5310 human GBM xenograft cells. In addition, ECs treated with tumor-conditioned media (CM) obtained from IR-treated 4910 and 5310 cells showed increased microtubule formation. In view of this finding, we investigated the possible anti-angiogenic effects of MMP-2 downregulation using siRNA (pM.si) in IR-treated cells. We also determined the effect of CM obtained from mock, pSV (scrambled vector) and pMMP-2.si on endothelial cell growth and vessel formation. pM.si-CM-treated ECs showed inhibited IR-CM-induced SDF-1, CXCR4, phospho-PI3K and phospho-AKT and αvß3 expression levels. In vitro angiogenesis assays also showed that the pM.si+IR decreased IR-induced vessel formation in ECs. Immunofluorescence and immunoprecipitation experiments indicated the abrogation of αvß3-SDF-1 interaction in pM.si-CM-treated ECs when compared to mock or pSV treatments. External supplementation of either rhMMP-2 or rhSDF-1 counteracted and noticeably reversed pM.si-inhibited SDF-1, CXCR4, phospho-PI3K and phospho-AKT expression levels and angiogenesis, thereby confirming the role of MMP-2 in the regulation of αvß3-mediated SDF-1/CXCR4 signaling. In addition to the in vitro results, the in vivo mouse dorsal air sac model also showed reduced angiogenesis after injection of pM.si alone or in combination with IR-treated xenograft cells. In contrast, injection of mock or pSV-treated cells resulted in robust formation of characteristic neovascularization. Collectively, our data demonstrate the role of MMP-2 in the regulation of SDF-1/CXCR4 signaling-mediated angiogenesis in ECs and show the anti-angiogenic efficacy of combining MMP-2 downregulation and IR when treating patients with GBM in the future.


Asunto(s)
Células Endoteliales/enzimología , Glioblastoma/radioterapia , Integrina alfaVbeta3/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Neovascularización Patológica/enzimología , Transducción de Señal , Animales , Línea Celular Tumoral , Quimiocina CXCL12/metabolismo , Medios de Cultivo Condicionados , Células Endoteliales/efectos de la radiación , Endotelio Vascular/enzimología , Endotelio Vascular/patología , Técnicas de Silenciamiento del Gen , Glioblastoma/irrigación sanguínea , Glioblastoma/enzimología , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Tolerancia a Radiación , Receptores CXCR4/metabolismo
6.
Physiol Plant ; 146(2): 136-48, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22462603

RESUMEN

Based on high economic importance and nutritious value of tomato fruits and as previous studies employed E8 promoter in fruit ripening-specific gene expression, we have developed transgenic tomato plants overexpressing tomato anionic peroxidase cDNA (tap1) under E8 promoter. Stable transgene integration was confirmed by polymerase chain reaction (PCR) and Southern analysis for nptII. Northern blotting confirmed elevated tap1 levels in the breaker- and red-ripe stages of T(1) transgenic fruits, whereas wild-type (WT) plants did not show tap1 expression in these developmental stages. Further, tap1 expression levels were significantly enhanced in response to wounding in breaker- and red-ripe stages of transgenic fruits, whereas wound-induced expression of tap1 was not detected in WT fruits. Confocal microscopy revealed high accumulation of phenolic compounds at the wound site in transgenic fruits suggesting a role of tap1 in wound-induced phenolic polymerization. Total peroxidase activity has increased remarkably in transgenic pericarp tissues in response to wounding, while very less or minimal levels were recorded in WT pericarp tissues. Transgenic fruits also displayed reduced post-harvest decay and increased resistance toward Alternaria alternata and Fusarium solani infection with noticeable inhibition in lesion formation. Conidiospore germination and mycelial growth of F. solani were severely inhibited when treated with E8-tap1 fruit extracts compared to WT fruits. 3-(4,5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay showed reduced spore viability when incubated in E8-tap1 fruit extracts. Thus, fruit-specific expression of tap1 using E8 promoter is associated with enhanced total peroxidase activity and high phenolic accumulation in fruits with minimized post-harvest deterioration caused by wounding and fungal attack in tomato fruits.


Asunto(s)
Resistencia a la Enfermedad/genética , Peroxidasas/genética , Peroxidasas/metabolismo , Enfermedades de las Plantas/genética , Plantas Modificadas Genéticamente/microbiología , Solanum lycopersicum/genética , Solanum lycopersicum/microbiología , Adaptación Fisiológica , Alternaria/patogenicidad , Frutas/enzimología , Frutas/genética , Frutas/crecimiento & desarrollo , Fusarium/patogenicidad , Regulación de la Expresión Génica de las Plantas , Variación Genética , Genotipo , Solanum lycopersicum/enzimología , Fenol/metabolismo , Polimerizacion , Estrés Fisiológico/fisiología
7.
PLoS One ; 6(6): e20614, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21698233

RESUMEN

BACKGROUND: Our previous work and that of others strongly suggests a relationship between the infiltrative phenotype of gliomas and the expression of MMP-2. Radiation therapy, which represents one of the mainstays of glioma treatment, is known to increase cell invasion by inducing MMP-2. Thus, inhibition of MMP-2 provides a potential means for improving the efficacy of radiotherapy for malignant glioma. METHODOLOGY/PRINCIPAL FINDINGS: We have tested the ability of a plasmid vector-mediated MMP-2 siRNA (p-MMP-2) to modulate ionizing radiation-induced invasive phenotype in the human glioma cell lines U251 and U87. Cells that were transfected with p-MMP-2 with and without radiation showed a marked reduction of MMP-2 compared to controls and pSV-transfected cells. A significant reduction of proliferation, migration, invasion and angiogenesis of cells transfected with p-MMP-2 and in combination with radiation was observed compared to controls. Western blot analysis revealed that radiation-enhanced levels of VEGF, VEGFR-2, pVEGFR-2, p-FAK, and p-p38 were inhibited with p-MMP-2-transfected cells. TUNEL staining showed that radiation did not induce apoptosis in U87 and U251 cells while a significant increase in TUNEL-positive cells was observed when irradiated cells were simultaneously transfected with p-MMP-2 as compared to controls. Intracranial tumor growth was predominantly inhibited in the animals treated with p-MMP-2 alone or in combination with radiation compared to controls. CONCLUSION/SIGNIFICANCE: MMP-2 inhibition, mediated by p-MMP-2 and in combination with radiation, significantly reduced tumor cell migration, invasion, angiogenesis and tumor growth by modulating several important downstream signaling molecules and directing cells towards apoptosis. Taken together, our results demonstrate the efficacy of p-MMP-2 in inhibiting radiation-enhanced tumor invasion and progression and suggest that it may act as a potent adjuvant for radiotherapy in glioma patients.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Metaloproteinasa 2 de la Matriz/genética , Invasividad Neoplásica/genética , ARN Interferente Pequeño/genética , Radiación Ionizante , Western Blotting , Neoplasias Encefálicas/irrigación sanguínea , Línea Celular Tumoral , Glioma/irrigación sanguínea , Humanos , Neovascularización Patológica , Plásmidos
8.
PLoS One ; 6(5): e19341, 2011 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-21573233

RESUMEN

BACKGROUND: Abrogation of apoptosis for prolonged cell survival is essential in cancer progression. In our previous studies, we showed the MMP-2 downregulation induced apoptosis in cancer cell lines. Here, we attempt to investigate the exact molecular mechanism of how MMP-2 depletion leads to apoptosis in glioma xenograft cell lines. METHODOLOGY/PRINCIPAL FINDINGS: MMP-2 transcriptional suppression by MMP-2siRNA (pM) induces apoptosis associated with PARP, caspase-8 and -3 cleavage in human glioma xenograft cells 4910 and 5310. Western blotting and cytokine array showed significant decrease in the cellular and secreted levels of TNF-α with concomitant reduction in TNFR1, TRADD, TRAF2, RIP, IKKß and pIκBα expression levels resulting in inhibition of p65 phosphorylation and nuclear translocation in pM-treated cells when compared to mock and pSV controls. In addition MMP-2 suppression led to elevated Fas-L, Fas and FADD expression levels along with increased p38 and JNK phosphorylation. The JNK-activity assay showed prolonged JNK activation in pM-transfected cells. Specific inhibition of p38 with SB203580 did not show any effect whereas inhibition of JNK phosphorylation with SP600125 notably reversed pM-induced cleavage of PARP, caspase-8 and -3, demonstrating a significant role of JNK in pM-induced cell death. Supplementation of rhMMP-2 counteracted the effect of pM by remarkably elevating TNF-α, TRADD, IKKß and pIκBα expression and decreasing FADD, Fas-L, and phospho-JNK levels. The EMSA analysis indicated significant reversal of pM-inhibited NF-κB activity by rhMMP-2 treatment which rescued cells from pM-induced cell death. In vivo studies indicated that pM treatment diminished intracranial tumor growth and the immuno histochemical analysis showed decreased phospho-p65 and enhanced phospho-JNK levels that correlated with increased TUNEL-positive apoptotic cells in pM-treated tumor sections. CONCLUSION/SIGNIFICANCE: In summary, our study implies a role of MMP-2 in the regulation of TNF-α mediated constitutive NF-κB activation and Fas-mediated JNK mediated apoptosis in glioma xenograft cells in vitro and in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Glioma/metabolismo , Glioma/terapia , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Antracenos/farmacología , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Ensayo de Cambio de Movilidad Electroforética , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Glioma/genética , Humanos , Imidazoles/farmacología , Inmunohistoquímica , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Metaloproteinasa 2 de la Matriz/genética , Ratones , Piridinas/farmacología , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
9.
Biochem Biophys Res Commun ; 390(3): 427-33, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19766095

RESUMEN

NF-kappaB activity is tightly regulated by IkappaB class of proteins. IkappaB proteins possess ankyrin repeats for binding to and inhibiting NF-kappaB. The regulatory protein, NPR1 from Brassica juncea possesses ankyrin repeats with sequence similarity to IkappaBalpha subgroup. Therefore, we examined whether stably expressed BjNPR1 could function as IkappaB in inhibiting NF-kappaB in human glioblastoma cell lines. We observed that BjNPR1 bound to NF-kappaB and inhibited its nuclear translocation. Further, BjNPR1 expression down-regulated the NF-kappaB target genes iNOS, Cox-2, c-Myc and cyclin D1 and reduced the proliferation rate of U373 cells. Finally, BjNPR1 decreased the levels of pERK, pJNK and PKCalpha and increased the Caspase-3 and Caspase-8 activities. These results suggested that inhibition of NF-kappaB activation by BjNPR1 can be a promising therapy in NF-kappaB dependent pathologies.


Asunto(s)
Proteínas I-kappa B/metabolismo , Planta de la Mostaza/metabolismo , FN-kappa B/antagonistas & inhibidores , Proteínas de Plantas/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Repetición de Anquirina , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/genética , Ciclooxigenasa 2/genética , Regulación hacia Abajo , Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Proteínas I-kappa B/genética , Inflamación/terapia , Datos de Secuencia Molecular , Planta de la Mostaza/genética , FN-kappa B/metabolismo , Neoplasias/terapia , Óxido Nítrico Sintasa de Tipo II/genética , Proteínas de Plantas/genética , Proteína Quinasa C-alfa/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , eIF-2 Quinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...