Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cells ; 12(15)2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37566001

RESUMEN

Glioblastoma (GBM) is the most common and aggressive primary brain tumor. GBM contains a small subpopulation of glioma stem cells (GSCs) that are implicated in treatment resistance, tumor infiltration, and recurrence, and are thereby considered important therapeutic targets. Recent clinical studies have suggested that the choice of general anesthetic (GA), particularly propofol, during tumor resection, affects subsequent tumor response to treatments and patient prognosis. In this study, we investigated the molecular mechanisms underlying propofol's anti-tumor effects on GSCs and their interaction with microglia cells. Propofol exerted a dose-dependent inhibitory effect on the self-renewal, expression of mesenchymal markers, and migration of GSCs and sensitized them to both temozolomide (TMZ) and radiation. At higher concentrations, propofol induced a large degree of cell death, as demonstrated using microfluid chip technology. Propofol increased the expression of the lncRNA BDNF-AS, which acts as a tumor suppressor in GBM, and silencing of this lncRNA partially abrogated propofol's effects. Propofol also inhibited the pro-tumorigenic GSC-microglia crosstalk via extracellular vesicles (EVs) and delivery of BDNF-AS. In conclusion, propofol exerted anti-tumor effects on GSCs, sensitized these cells to radiation and TMZ, and inhibited their pro-tumorigenic interactions with microglia via transfer of BDNF-AS by EVs.


Asunto(s)
Neoplasias Encefálicas , Vesículas Extracelulares , Glioblastoma , Glioma , Propofol , ARN Largo no Codificante , Humanos , Neoplasias Encefálicas/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Vesículas Extracelulares/metabolismo , Glioblastoma/metabolismo , Glioma/metabolismo , Microglía/metabolismo , Células Madre Neoplásicas/patología , Propofol/farmacología , ARN Largo no Codificante/genética , Temozolomida/farmacología
3.
Cell Death Dis ; 11(10): 899, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-33093452

RESUMEN

Glioblastoma (GBM) is a highly aggressive tumor with poor prognosis. A small subpopulation of glioma stem cells (GSCs) has been implicated in radiation resistance and tumor recurrence. In this study we analyzed the expression of miRNAs associated with the functions of GSCs using miRNA microarray analysis of these cells compared with human neural stem cells. These analyses identified gene clusters associated with glioma cell invasiveness, axonal guidance, and TGF-ß signaling. miR-504 was significantly downregulated in GSCs compared with NSCs, its expression was lower in GBM compared with normal brain specimens and further decreased in the mesenchymal glioma subtype. Overexpression of miR-504 in GSCs inhibited their self-renewal, migration and the expression of mesenchymal markers. The inhibitory effect of miR-504 was mediated by targeting Grb10 expression which acts as an oncogene in GSCs and GBM. Overexpression of exogenous miR-504 resulted also in its delivery to cocultured microglia by GSC-secreted extracellular vesicles (EVs) and in the abrogation of the GSC-induced polarization of microglia to M2 subtype. Finally, miR-504 overexpression prolonged the survival of mice harboring GSC-derived xenografts and decreased tumor growth. In summary, we identified miRNAs and potential target networks that play a role in the stemness and mesenchymal transition of GSCs and the miR-504/Grb10 pathway as an important regulator of this process. Overexpression of miR-504 exerted antitumor effects in GSCs as well as bystander effects on the polarization of microglia via delivery by EVs.


Asunto(s)
Neoplasias Encefálicas/genética , Vesículas Extracelulares/fisiología , Glioblastoma/genética , MicroARNs/fisiología , Microglía/citología , Células Madre Neoplásicas/citología , Animales , Neoplasias Encefálicas/metabolismo , Proteína Adaptadora GRB10/fisiología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Humanos , Ratones , Ratones Desnudos , Análisis por Micromatrices , Células-Madre Neurales/citología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Oncotarget ; 8(19): 31785-31801, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28423669

RESUMEN

Despite advances in novel therapeutic approaches for the treatment of glioblastoma (GBM), the median survival of 12-14 months has not changed significantly. Therefore, there is an imperative need to identify molecular mechanisms that play a role in patient survival. Here, we analyzed the expression and functions of a novel lncRNA, TALNEC2 that was identified using RNA seq of E2F1-regulated lncRNAs. TALNEC2 was localized to the cytosol and its expression was E2F1-regulated and cell-cycle dependent. TALNEC2 was highly expressed in GBM with poor prognosis, in GBM specimens derived from short-term survivors and in glioma cells and glioma stem cells (GSCs). Silencing of TALNEC2 inhibited cell proliferation and arrested the cells in the G1\S phase of the cell cycle in various cancer cell lines. In addition, silencing of TALNEC2 decreased the self-renewal and mesenchymal transformation of GSCs, increased sensitivity of these cells to radiation and prolonged survival of mice bearing GSC-derived xenografts. Using miRNA array analysis, we identified specific miRNAs that were altered in the silenced cells that were associated with cell-cycle progression, proliferation and mesenchymal transformation. Two of the downregulated miRNAs, miR-21 and miR-191, mediated some of TALNEC2 effects on the stemness and mesenchymal transformation of GSCs. In conclusion, we identified a novel E2F1-regulated lncRNA that is highly expressed in GBM and in tumors from patients of short-term survival. The expression of TALNEC2 is associated with the increased tumorigenic potential of GSCs and their resistance to radiation. We conclude that TALNEC2 is an attractive therapeutic target for the treatment of GBM.


Asunto(s)
Autorrenovación de las Células/genética , Glioma/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , ARN Largo no Codificante/genética , Tolerancia a Radiación/genética , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/genética , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Silenciador del Gen , Glioma/mortalidad , Glioma/patología , Glioma/radioterapia , Humanos , Ratones , MicroARNs/genética , Pronóstico , Transporte de ARN , Resultado del Tratamiento , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Oncotarget ; 7(35): 56456-56470, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27486821

RESUMEN

Glioblastoma (GBM) is the most aggressive primary brain tumor with poor prognosis. Here, we studied the effects of phenformin, a mitochondrial complex I inhibitor and more potent chemical analog of the diabetes drug metformin on the inhibition of cell growth and induction of apoptosis of glioma stem cells (GSCs) using both in vitro and in vivo models. Phenformin inhibited the self-renewal of GSCs, decreased the expression of stemness and mesenchymal markers and increased the expression of miR-124, 137 and let-7. Silencing of let-7 abrogated phenformin effects on the self-renewal of GSCs via a pathway associated with inhibition of H19 and HMGA2 expression. Moreover, we demonstrate that phenformin inhibited tumor growth and prolonged the overall survival of mice orthotopically transplanted with GSCs. Combined treatments of phenformin and temozolomide exerted an increased antitumor effect on GSCs in vitro and in vivo. In addition, dichloroacetate, an inhibitor of the glycolysis enzyme pyruvate dehydrogenase kinase, that decreases lactic acidosis induced by biguanides, enhanced phenformin effects on the induction of cell death in GSCs and prolonged the survival of xenograft-bearing mice. Our results demonstrate for the first time that phenformin targets GSCs and can be efficiently combined with current therapies for GBM treatment and GSC eradication.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Glioma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Fenformina/farmacología , Animales , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Proliferación Celular , Ácido Dicloroacético/farmacología , Reposicionamiento de Medicamentos , Silenciador del Gen , Glioblastoma/patología , Glioma/patología , Proteína HMGA2/antagonistas & inhibidores , Humanos , Hipoglucemiantes/química , Lentivirus , Ratones , Ratones Desnudos , MicroARNs/metabolismo , Recurrencia Local de Neoplasia , Trasplante de Neoplasias , Células Madre Neoplásicas/patología , ARN Largo no Codificante/antagonistas & inhibidores
6.
Oncotarget ; 6(26): 22680-97, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26267319

RESUMEN

Glioblastomas (GBMs), the most aggressive primary brain tumors, exhibit increased invasiveness and resistance to anti-tumor treatments. We explored the role of RTVP-1, a glioma-associated protein that promotes glioma cell migration, in the mesenchymal transformation of GBM. Analysis of The Cancer Genome Atlas (TCGA) demonstrated that RTVP-1 expression was higher in mesenchymal GBM and predicted tumor recurrence and poor clinical outcome. ChiP analysis revealed that the RTVP-1 promoter binds STAT3 and C/EBPß, two master transcription factors that regulate mesenchymal transformation of GBM. In addition, IL-6 induced RTVP-1 expression in a STAT3-dependent manner. RTVP-1 increased the migration and mesenchymal transformation of glioma cells. Similarly, overexpression of RTVP-1 in human neural stem cells induced mesenchymal differentiation, whereas silencing of RTVP-1 in glioma stem cells (GSCs) decreased the mesenchymal transformation and stemness of these cells. Silencing of RTVP-1 also increased the survival of mice bearing GSC-derived xenografts. Using gene array analysis of RTVP-1 silenced glioma cells we identified IL-6 as a mediator of RTVP-1 effects on the mesenchymal transformation and migration of GSCs, therefore acting in a positive feedback loop by upregulating RTVP-1 expression via the STAT3 pathway. Collectively, these results implicate RTVP-1 as a novel prognostic marker and therapeutic target in GBM.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioma/metabolismo , Glioma/patología , Interleucina-6/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Proliferación Celular/fisiología , Transición Epitelial-Mesenquimal , Glioma/genética , Xenoinjertos , Humanos , Proteínas de la Membrana , Ratones , Ratones Desnudos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Transducción de Señal , Activación Transcripcional , Transfección
7.
Oncotarget ; 6(23): 19826-40, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26305187

RESUMEN

Glioblastoma (GBM) are characterized by increased invasion into the surrounding normal brain tissue. RTVP-1 is highly expressed in GBM and regulates the migration and invasion of glioma cells. To further study RTVP-1 effects we performed a pull-down assay using His-tagged RTVP-1 followed by mass spectrometry and found that RTVP-1 was associated with the actin polymerization regulator, N-WASP. This association was further validated by co-immunoprecipitation and FRET analysis. We found that RTVP-1 increased cell spreading, migration and invasion and these effects were at least partly mediated by N-WASP. Another protein which was found by the pull-down assay to interact with RTVP-1 is hnRNPK. This protein has been recently reported to associate with and to inhibit the effect of N-WASP on cell spreading. hnRNPK decreased cell migration, spreading and invasion in glioma cells. Using co-immunoprecipitation we validated the interactions of hnRNPK with N-WASP and RTVP-1 in glioma cells. In addition, we found that overexpression of RTVP-1 decreased the association of N-WASP and hnRNPK. In summary, we report that RTVP-1 regulates glioma cell spreading, migration and invasion and that these effects are mediated via interaction with N-WASP and by interfering with the inhibitory effect of hnRNPK on the function of this protein.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Movimiento Celular , Glioblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ribonucleoproteínas/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Forma de la Célula , Transferencia Resonante de Energía de Fluorescencia , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo K , Humanos , Inmunoprecipitación , Espectrometría de Masas , Proteínas de la Membrana , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/genética , Unión Proteica , Proteómica/métodos , Interferencia de ARN , Ribonucleoproteínas/genética , Transducción de Señal , Transfección , Células Tumorales Cultivadas , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética
8.
Oncotarget ; 6(3): 1850-64, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25682201

RESUMEN

Glioblastoma (GBM), the most aggressive primary brain tumors, are highly infiltrative. Although GBM express high Ras activity and Ras proteins have been implicated in gliomagenesis, Ras-activating mutations are not frequent in these tumors. RasGRP3, an important signaling protein responsive to diacylglycerol (DAG), increases Ras activation. Here, we examined the expression and functions of RasGRP3 in GBM and glioma cells. RasGRP3 expression was upregulated in GBM specimens and glioma stem cells compared with normal brains and neural stem cells, respectively. RasGRP3 activated Ras and Rap1 in glioma cells and increased cell migration and invasion partially via Ras activation. Using pull-down assay and mass spectroscopy we identified the actin-related protein, Arp3, as a novel interacting protein of RasGRP3. The interaction of RasGRP3 and Arp3 was validated by immunofluorescence staining and co-immunoprecipitation, and PMA, which activates RasGRP3 and induces its translocation to the peri-nuclear region, increased the association of Arp3 and RasGRP3. Arp3 was upregulated in GBM, regulated cell spreading and migration and its silencing partially decreased these effects of RasGRP3 in glioma cells. In summary, RasGRP3 acts as an important integrating signaling protein of the DAG and Ras signaling pathways and actin polymerization and represents an important therapeutic target in GBM.


Asunto(s)
Proteína 3 Relacionada con la Actina/metabolismo , Neoplasias Encefálicas/patología , Movimiento Celular/fisiología , Glioma/patología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteína 3 Relacionada con la Actina/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Glioma/genética , Glioma/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Transducción de Señal , Transfección , Factores de Intercambio de Guanina Nucleótido ras
9.
Stem Cells Dev ; 23(23): 2851-61, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25036385

RESUMEN

MicroRNAs (miRNAs) are potential therapeutic targets in a variety of pathological conditions in the brain; however, their clinical application is hampered by lack of efficient delivery modes. Mesenchymal stromal stem cells (MSCs) migrate to sites of injury and inflammation and exert therapeutic effects in various neurological disorders. Here, we examined the ability of MSCs to deliver exogenous miRNA mimics and pre-miRNAs to human neural progenitor cells (NPCs) and astrocytes and characterized the functional impact of this delivery. We found that MSCs efficiently delivered fluorescent-labeled miR-124 and miR-145 mimics to cocultured NPCs and astrocytes. We further demonstrated the delivery of the miRNAs using novel reporter plasmids that contain a sequence complementary to miR-124 or miR-145 downstream of luciferase or mCherry. Binding of the specific miRNAs to these sequences results in decreased luciferase activity or mCherry fluorescence and therefore enable analysis of miRNA delivery in living cells. The delivered exogenous miR-124 significantly decreased the expression of the target gene Sox9 by targeting its 3'-UTR, and increased the neuronal differentiation of the NPCs. In addition, the delivered miR-124 increased the expression of the glutamate transporters, EAAT1 in NPCs and EAAT2 in both NPCs and astrocytes. Similar results were obtained with MSCs transfected with pre-miR-124. The miRNA delivery was mediated by MSC-derived exosomes and was cell contact independent. These results suggest that MSCs can functionally deliver exogenous miRNAs to neural cells and provide an efficient route of therapeutic miRNA delivery to the brain in pathological conditions with clinical implications for regenerative medicine.


Asunto(s)
Transportador 1 de Aminoácidos Excitadores/biosíntesis , Proteínas de Transporte de Glutamato en la Membrana Plasmática/biosíntesis , Células Madre Mesenquimatosas/metabolismo , MicroARNs , Células-Madre Neurales/metabolismo , Regiones no Traducidas 3' , Diferenciación Celular , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores , Regulación de la Expresión Génica/genética , Proteínas de Transporte de Glutamato en la Membrana Plasmática/genética , Humanos , Células Madre Mesenquimatosas/citología , MicroARNs/genética , MicroARNs/metabolismo , Células-Madre Neurales/citología , Factor de Transcripción SOX9/biosíntesis
10.
Oncol Lett ; 7(4): 1209-1212, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24944694

RESUMEN

Related to testes-specific, vespid and pathogenesis protein-1 (RTVP-1), also known as glioma pathogenesis-related protein 1, is highly expressed and has oncogenic features in glioblastoma (GBM; World Health Organization class IV). Promoter methylation has been found to control RTVP-1 expression in prostate carcinoma, Wilms' tumor, acute myeloid leukemia and melanoma. In this bi-institutional study, the methylation status of RTVP-1 in astrocytic brain malignancies (GBM and oligodendroglioma) was examined. The RTVP-1 promoter was hypomethylated in GBM compared with non-tumor brain samples, but was hypermethylated in oligodendroglioma. RTVP-1 methylation correlated with RTVP-1 expression at the mRNA level. In GBM, hypermethylation of the RTVP-1 promoter was associated with improved overall survival although with no statistical significance.

11.
Oncotarget ; 4(5): 665-76, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23714687

RESUMEN

Glioblastomas (GBM), the most common and aggressive malignant astrocytic tumors, contain a small subpopulation of cancer stem cells (GSCs) that are implicated in therapeutic resistance and tumor recurrence. Here, we study the expression and function of miR-137, a putative suppressor miRNA, in GBM and GSCs. We found that the expression of miR-137 was significantly lower in GBM and GSCs compared to normal brains and neural stem cells (NSCs) and that the miR-137 promoter was hypermethylated in the GBM specimens. The expression of miR-137 was increased in differentiated NSCs and GSCs and overexpression of miR-137 promoted the neural differentiation of both cell types. Moreover, pre-miR-137 significantly decreased the self-renewal of GSCs and the stem cell markers Oct4, Nanog, Sox2 and Shh. We identified RTVP-1 as a novel target of miR-137 in GSCs; transfection of the cells with miR-137 decreased the expression of RTVP-1 and the luciferase activity of RTVP-1 3'-UTR reporter plasmid. Furthermore, overexpression of RTVP-1 plasmid lacking its 3'-UTR abrogated the inhibitory effect of miR-137 on the self-renewal of GSCs. Silencing of RTVP-1 decreased the self-renewal of GSCs and the expression of CXCR4 and overexpression of CXCR4 abrogated the inhibitory effect of RTVP-1 silencing on GSC self-renewal. These results demonstrate that miR-137 is downregulated in GBM probably due to promoter hypermethylation. miR-137 inhibits GSC self-renewal and promotes their differentiation by targeting RTVP-1 which downregulates CXCR4. Thus, miR-137 and RTVP-1 are attractive therapeutic targets for the eradication of GSCs and for the treatment of GBM.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/citología , Proteínas del Tejido Nervioso/metabolismo , Receptores CXCR4/biosíntesis , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Diferenciación Celular , Movimiento Celular/genética , Proliferación Celular , Metilación de ADN , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Proteínas Hedgehog/biosíntesis , Proteínas de Homeodominio/biosíntesis , Humanos , Proteínas de la Membrana , MicroARNs/biosíntesis , MicroARNs/genética , Proteína Homeótica Nanog , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Regiones Promotoras Genéticas/genética , Factores de Transcripción SOXB1/biosíntesis , Transducción de Señal/genética , Células Tumorales Cultivadas
12.
Oncotarget ; 4(2): 346-61, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23548312

RESUMEN

MicroRNAs (miRNAs) have emerged as potential cancer therapeutics; however, their clinical use is hindered by lack of effective delivery mechanisms to tumor sites. Mesenchymal stem cells (MSCs) have been shown to migrate to experimental glioma and to exert anti-tumor effects by delivering cytotoxic compounds. Here, we examined the ability of MSCs derived from bone marrow, adipose tissue, placenta and umbilical cord to deliver synthetic miRNA mimics to glioma cells and glioma stem cells (GSCs). We examined the delivery of miR-124 and miR-145 mimics as glioma cells and GSCs express very low levels of these miRNAs. Using fluorescently labeled miRNA mimics and in situ hybridization, we demonstrated that all the MSCs examined delivered miR-124 and miR-145 mimics to co-cultured glioma cells and GSCs via gap junction- dependent and independent processes. The delivered miR-124 and miR-145 mimics significantly decreased the luciferase activity of their respected reporter target genes, SCP-1 and Sox2, and decreased the migration of glioma cells and the self-renewal of GSCs. Moreover, MSCs delivered Cy3-miR-124 mimic to glioma xenografts when administered intracranially. These results suggest that MSCs can deliver synthetic exogenous miRNA mimics to glioma cells and GSCs and may provide an efficient route of therapeutic miRNA delivery in vivo.


Asunto(s)
Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Movimiento Celular/genética , Glioma/patología , Glioma/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , MicroARNs/administración & dosificación , Células Madre Neoplásicas/patología , Animales , Neoplasias Encefálicas/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Humanos , Ratones , Ratones Desnudos , MicroARNs/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
PLoS One ; 8(2): e54652, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23390502

RESUMEN

Glioblastomas (GBM), the most common and aggressive type of malignant glioma, are characterized by increased invasion into the surrounding brain tissues. Despite intensive therapeutic strategies, the median survival of GBM patients has remained dismal over the last decades. In this study we examined the expression of miR-145 in glial tumors and its function in glioma cells. Using TCGA analysis and real-time PCR we found that the expression of miR-145/143 cluster was downregulated in astrocytic tumors compared to normal brain specimens and in glioma cells and glioma stem cells (GSCs) compared to normal astrocytes and neural stem cells. Moreover, the low expression of both miR-145 and miR-143 in GBM was correlated with poor patient prognosis. Transfection of glioma cells with miR-145 mimic or transduction with a lentivirus vector expressing pre-miR 145 significantly decreased the migration and invasion of glioma cells. We identified connective tissue growth factor (CTGF) as a novel target of miR-145 in glioma cells; transfection of the cells with this miRNA decreased the expression of CTGF as determined by Western blot analysis and the expression of its 3'-UTR fused to luciferase. Overexpression of a CTGF plasmid lacking the 3'-UTR and administration of recombinant CTGF protein abrogated the inhibitory effect of miR-145 on glioma cell migration. Similarly, we found that silencing of CTGF decreased the migration of glioma cells. CTGF silencing also decreased the expression of SPARC, phospho-FAK and FAK and overexpression of SPARC abrogated the inhibitory effect of CTGF silencing on cell migration. These results demonstrate that miR-145 is downregulated in glial tumors and its low expression in GBM predicts poor patient prognosis. In addition miR-145 regulates glioma cell migration by targeting CTGF which downregulates SPARC expression. Therefore, miR-145 is an attractive therapeutic target for anti-invasive treatment of astrocytic tumors.


Asunto(s)
Neoplasias Encefálicas/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , MicroARNs/genética , Proteínas Supresoras de Tumor/genética , Regiones no Traducidas 3' , Astrocitos/citología , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Silenciador del Gen , Genes Reporteros , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Luciferasas , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Osteonectina , ARN Interferente Pequeño/genética , Transducción de Señal , Transfección , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo
14.
Cell Signal ; 23(12): 1936-43, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21777672

RESUMEN

Gliomas are characterized by increased infiltration into the surrounding normal brain tissue. We recently reported that RTVP-1 is highly expressed in gliomas and plays a role in the migration of these cells, however the regulation of RTVP-1 expression in these cells is not yet described. In this study we examined the role of PKC in the regulation of RTVP-1 expression and found that PMA and overexpression of PKCα and PKCε increased the expression of RTVP-1, whereas PKCδ exerted an opposite effect. Using the MatInspector software, we identified a SRF binding site on the RTVP-1 promoter. Chromatin immunoprecipitation (ChIP) assay revealed that SRF binds to the RTVP-1 promoter in U87 cells, and that this binding was significantly increased in response to serum addition. Moreover, silencing of SRF blocked the induction of RTVP-1 expression in response to serum. We found that overexpression of PKCα and PKCε increased the activity of the RTVP-1 promoter and the binding of SRF to the promoter. In contrast, overexpression of PKCδ blocked the increase in RTVP-1 expression in response to serum and the inhibitory effect of PKCδ was abrogated in cells expressing a SRFT160A mutant. SRF regulated the migration of glioma cells and its effect was partially mediated by RTVP-1. We conclude that RTVP-1 is a PKC-regulated gene and that this regulation is at least partly mediated by SRF. Moreover, RTVP-1 plays a role in the effect of SRF on glioma cell migration.


Asunto(s)
Glioma/fisiopatología , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C/metabolismo , Factor de Respuesta Sérica/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular , Glioma/metabolismo , Humanos , Isoenzimas/metabolismo , Proteínas de la Membrana , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/genética , Fosforilación , Regiones Promotoras Genéticas , Transcripción Genética , Activación Transcripcional
15.
Neuro Oncol ; 13(8): 857-65, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21788343

RESUMEN

We studied the effect of the integrin inhibitor cilengitide in glioma cells. Cilengitide induced cell detachment and decreased cell viability, and induction of autophagy followed by cell apoptosis. In addition, cilengitide decreased the cell renewal of glioma stem-like cells (GSCs). Inhibition of autophagy decreased the cytotoxic effect of cilengitide. Pretreatment of glioma cells and GSCs with cilengitide prior to γ-irradiation resulted in a larger increase in autophagy and a more significant decrease in cell survival. We found that cilengitide induced autophagy collectively in glioma cells, xenografts, and GSCs, which contributed to its cytotoxic effects and sensitized these cells to γ-radiation.


Asunto(s)
Autofagia/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Venenos de Serpiente/uso terapéutico , Animales , Autofagia/efectos de la radiación , Western Blotting , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Terapia Combinada , Rayos gamma , Glioma/patología , Glioma/radioterapia , Humanos , Células Madre Neoplásicas/efectos de la radiación , Ratas , Ratas Desnudas , Trasplante Heterólogo
16.
Cell Signal ; 23(8): 1348-57, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21440622

RESUMEN

In this study we examined the effects of proteasome inhibitors on cell apoptosis in TRAIL-resistant glioma cells and glioma stem cells (GSCs). Treatment with proteasome inhibitors and TRAIL induced apoptosis in all the resistant glioma cells and GSCs, but not in astrocytes and neural progenitor cells. Since PKCε has been implicated in the resistance of glioma cells to TRAIL, we examined its role in TRAIL and proteasome inhibitor-induced apoptosis. We found that TRAIL did not induce significant changes in the expression of PKCε, whereas a partial decrease in PKCε expression was obtained by proteasome inhibitors. A combined treatment of TRAIL and proteasome inhibitors induced accumulation of the catalytic fragment of PKCε and significantly and selectively decreased its protein and mRNA levels in the cancer but not in normal cells. Overexpression of PKCε partially inhibited the apoptotic effect of the proteasome inhibitors and TRAIL, and the caspase-resistant PKCεD383A mutant exerted a stronger inhibitory effect. Silencing of PKCε induced cell apoptosis in both glioma cells and GSCs, further supporting its role in cell survival. TRAIL and the proteasome inhibitors decreased the expression of AKT and XIAP in a PKCε-dependent manner and overexpression of these proteins abolished the apoptotic effect of this treatment. Moreover, silencing of XIAP sensitized glioma cells to TRAIL. Our results indicate that proteasome inhibitors sensitize glioma cells and GSCs to TRAIL by decreasing the expression of PKCε, AKT and XIAP. Combining proteasome inhibitors with TRAIL may be useful therapeutically in the treatment of gliomas and the eradication of GSCs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Glioma/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Proteína Quinasa C-epsilon/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Apoptosis , Astrocitos/efectos de los fármacos , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular Tumoral , Regulación hacia Abajo , Glioma/enzimología , Humanos , Leupeptinas/farmacología , Mutagénesis Sitio-Dirigida , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Quinasa C-epsilon/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
17.
Int J Cancer ; 125(3): 717-22, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19431142

RESUMEN

Malignant gliomas are characterized by a short median survival which is largely impacted by the resistance of these tumors tochemo- and radiotherapy. Recent studies suggest that a small subpopulation of cancer stem cells, which are highly resistant to gamma-radiation, has the capacity to repopulate the tumors and contribute to their malignant progression. gamma-radiation activates the process of autophagy and inhibition of this process increases the radiosensitivity of glioma cells; however, the role of autophagy in the resistance of glioma stem cells (GSCs) to radiation has not been yet reported. In this study we examined the induction of autophagy by gamma-radiation in CD133+ GSCs. Irradiation of CD133+ cells induced autophagy within 24-48 hr and slightly decreased the viability of the cells. gamma-radiation induced a larger degree of autophagy in the CD133+ cells as compared with CD133- cells and the CD133+ cells expressed higher levels of the autophagy-related proteins LC3, ATG5 and ATG12. The autophagy inhibitor bafilomycin A1 and silencing of ATG5 and beclin1 sensitized the CD133+ cells to gamma-radiation and significantly decreased the viability of the irradiated cells and their ability to form neurospheres. Collectively, these results indicate that the induction of autophagy contributes to the radioresistance of these cells and autophagy inhibitors may be employed to increase the sensitivity of CD133+ GSCs to gamma-radiation.


Asunto(s)
Antígenos CD/análisis , Autofagia/efectos de los fármacos , Autofagia/efectos de la radiación , Neoplasias Encefálicas/radioterapia , Rayos gamma/uso terapéutico , Glioma/radioterapia , Glicoproteínas/análisis , Péptidos/análisis , Antígeno AC133 , Proteínas Reguladoras de la Apoptosis/genética , Proteína 12 Relacionada con la Autofagia , Proteína 5 Relacionada con la Autofagia , Beclina-1 , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/fisiopatología , Electroquimioterapia , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Glioma/inmunología , Glioma/fisiopatología , Humanos , Macrólidos/farmacología , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/efectos de la radiación , Fármacos Sensibilizantes a Radiaciones/farmacología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Regulación hacia Arriba
18.
Neoplasia ; 10(12): 1335-42, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19048112

RESUMEN

Insulin-like growth factor binding protein 7 (IGFBP-7) is the only member of the IGFBP superfamily that binds strongly to insulin, suggesting that IGFBP-7 may have different functions from other IGFBPs. Unlike other IGFBPs, the expression and functions of IGFBP-7 in glioma tumors have not been reported. Using cDNA microarray analysis, we found that expression of IGFBP-7 correlated with the grade of glioma tumors and the overall patient survival. This finding was further validated by real-time reverse transcription-polymerase chain reaction and Western blot analysis. We used RNAi to examine the role of IGFBP-7 in glioma cells, inhibiting IGFBP-7 expression by short interfering RNA transfection. Cell proliferation was suppressed after IGFBP-7 expression was inhibited for 5 days, and glioma cell growth was stimulated consistently by the addition of recombinant IGFBP-7 protein. Moreover, glioma cell migration was attenuated by IGFBP-7 depletion but enhanced by IGFBP-7 overexpression and addition. Overexpression of AKT1 in IGFBP-7-overxpressed cells attenuated the IGFBP-7-promoted migration and further enhanced inhibition of IGFBP-7 depletion on the migration. Phosphorylation of AKT and Erk1/2 was also inversely regulated by IGFBP-7 expression. These two factors together suggest that IGFBP-7 can regulate glioma cell migration through the AKT-ERK pathway, thereby playing an important role in glioma growth and migration.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glioma/patología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Glioma/metabolismo , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Sistema de Señalización de MAP Quinasas , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Neuro Oncol ; 10(3): 236-43, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18403493

RESUMEN

We characterized the expression and function of the endoplasmic reticulum protein GRP78 in glial tumors. GRP78 is highly expressed in glioblastomas but not in oligodendrogliomas, and its expression is inversely correlated with median patient survival. Overexpression of GRP78 in glioma cells decreases caspase 7 activation and renders the cells resistant to etoposide- and cisplatin-induced apoptosis, whereas silencing of GRP78 decreases cell growth and sensitizes glioma cells to etoposide, cisplatin, and gamma-radiation. Thus, GRP78 contributes to the increased apoptosis resistance and growth of glioma cells and may provide a target for enhancing the therapeutic responsiveness of these tumors.


Asunto(s)
Apoptosis/fisiología , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Glioma/metabolismo , Proteínas de Choque Térmico/biosíntesis , Chaperonas Moleculares/biosíntesis , Biomarcadores de Tumor/análisis , Western Blotting , Neoplasias Encefálicas/mortalidad , Caspasa 7 , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Chaperón BiP del Retículo Endoplásmico , Activación Enzimática/fisiología , Expresión Génica , Perfilación de la Expresión Génica , Glioma/mortalidad , Humanos , Inmunohistoquímica , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba
20.
J Biol Chem ; 283(25): 17731-9, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18434324

RESUMEN

The mechanism underlying the important role of protein kinase Cdelta (PKCdelta) in the apoptotic effect of etoposide in glioma cells is incompletely understood. Here, we examined the role of PKCdelta in the activation of Erk1/2 by etoposide. We found that etoposide induced persistent activation of Erk1/2 and nuclear translocation of phospho-Erk1/2. MEK1 inhibitors decreased the apoptotic effect of etoposide, whereas inhibitors of p38 and JNK did not. The activation of Erk1/2 by etoposide was downstream of PKCdelta since the phosphorylation of Erk1/2 was inhibited by a PKCdelta-KD mutant and PKCdelta small interfering RNA. We recently reported that phosphorylation of PKCdelta on tyrosines 64 and 187 was essential for the apoptotic effect of etoposide. Using PKCdeltatyrosine mutants, we found that the phosphorylation of PKCdeltaon these tyrosine residues, but not on tyrosine 155, was also essential for the activation of Erk1/2 by etoposide. In contrast, nuclear translocation of PKCdelta was independent of its tyrosine phosphorylation and not necessary for the phosphorylation of Erk1/2. Etoposide induced down-regulation of kinase phosphatase-1 (MKP-1), which correlated with persistent phosphorylation of Erk1/2 and was dependent on the tyrosine phosphorylation of PKCdelta. Moreover, silencing of MKP-1 increased the phosphorylation of Erk1/2 and the apoptotic effect of etoposide. Etoposide induced polyubiquitylation and degradation of MKP-1 that was dependent on PKCdelta and on its tyrosine phosphorylation. These results indicate that distinct phosphorylation of PKCdeltaon tyrosines 64 and 187 specifically activates the Erk1/2 pathway by the down-regulation of MKP-1, resulting in the persistent phosphorylation of Erk1/2 and cell apoptosis.


Asunto(s)
Apoptosis , Fosfatasa 1 de Especificidad Dual/metabolismo , Etopósido/farmacología , Regulación Enzimológica de la Expresión Génica , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa C-delta/metabolismo , Tirosina/química , Antineoplásicos Fitogénicos/farmacología , Humanos , Microscopía Confocal , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA