Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(14)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35887290

RESUMEN

Although osteosarcoma is the most common primary malignant bone tumor, chemotherapeutic drugs and treatment have failed to increase the five-year survival rate over the last three decades. We previously demonstrated that type 5 metabotropic glutamate receptor, mGluR5, is required to proliferate metastatic osteosarcoma cells. In this work, we delivered mGluR5 siRNAs in vitro using superparamagnetic iron oxide nanocages (IO-nanocages) as delivery vehicles and applied alternating magnetic fields (AMFs) to improve mGluR5 siRNAs release. We observed functional outcomes when mGluR5 expression is silenced in human and mouse osteosarcoma cell lines. The results elucidated that the mGluR5 siRNAs were successfully delivered by IO-nanocages and their release was enhanced by AMFs, leading to mGluR5 silencing. Moreover, we observed that the proliferation of both human and mouse osteosarcoma cells decreased significantly when mGluR5 expression was silenced in the cells. This novel magnetic siRNA delivery methodology was capable of silencing mGluR5 expression significantly in osteosarcoma cell lines under the AMFs, and our data suggested that this method can be further used in future clinical applications in cancer therapy.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/terapia , Línea Celular Tumoral , Proliferación Celular , Compuestos Férricos , Humanos , Campos Magnéticos , Ratones , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/terapia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico
2.
Int J Oncol ; 59(5)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34713302

RESUMEN

Riluzole, a glutamate release inhibitor, has been in use for the treatment of amyotrophic lateral sclerosis for over two decades since its approval by the Food and Drug Administration. Recently, riluzole has been evaluated in cancer cells and indicated to block cell proliferation and/or induce cell death. Riluzole has been proven effective as an anti­neoplastic drug in cancers of various tissue origins, including the skin, breast, pancreas, colon, liver, bone, brain, lung and nasopharynx. While cancer cells expressing glutamate receptors frequently respond to riluzole treatment, numerous types of cancer cell lacking glutamate receptors unexpectedly responded to riluzole treatment as well. Riluzole was demonstrated to interfere with glutamate secretion, growth signaling pathways, Ca2+ homeostasis, glutathione synthesis, reactive oxygen species generation and integrity of DNA, as well as autophagic and apoptotic pathways. Of note, riluzole is highly effective in inducing cell death in cisplatin­resistant lung cancer cells. Furthermore, riluzole pretreatment sensitizes glioma and melanoma to radiation therapy. In addition, in triple­negative breast cancer, colorectal cancer, melanoma and glioblastoma, riluzole has synergistic effects in combination with select drugs. In an effort to highlight the therapeutic potential of riluzole, the current study reviewed the effect and outcome of riluzole treatment on numerous cancer types investigated thus far. The mechanism of action and the various molecular pathways affected by riluzole are discussed.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Riluzol/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Neoplasias/patología , Riluzol/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Sci Rep ; 11(1): 20974, 2021 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-34697383

RESUMEN

Our lab has previously demonstrated Riluzole to be an effective drug in inhibiting proliferation and inducing apoptosis in both human and mouse osteosarcoma. Yes-associated protein is a transcription co-activator, known to be involved in cell proliferation or apoptosis depending on its protein partner. In the present study we investigated the role of YAP in apoptosis in osteosarcoma, we hypothesized that YAP may be activated by Riluzole to induce apoptosis in osteosarcoma. By knocking down the expression of YAP, we have demonstrated that Riluzole failed to induce apoptosis in YAP deficient osteosarcoma cells. Riluzole caused translocation of YAP from the cytoplasm to the nucleus, indicating YAP's role in apoptosis. Both Riluzole-induced phosphorylation of YAP at tyrosine 357 and Riluzole-induced apoptosis were blocked by inhibitors of c-Abl kinase. In addition, knockdown of c-Abl kinase prevented Riluzole-induced apoptosis in LM7 cells. We further demonstrated that Riluzole promoted interaction between YAP and p73, while c-Abl kinase inhibitors abolished the interaction. Subsequently, we demonstrated that Riluzole enhanced activity of the Bax promoter in a luciferase reporter assay and enhanced YAP/p73 binding on endogenous Bax promoter in a ChIP assay. Our data supports a novel mechanism in which Riluzole activates c-Abl kinase to regulate pro-apoptotic activity of YAP in osteosarcoma.


Asunto(s)
Neoplasias Óseas/genética , Osteosarcoma/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , Riluzol/farmacología , Proteínas Señalizadoras YAP/genética , Apoptosis , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Citoplasma/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Osteosarcoma/metabolismo , Fosforilación , Transporte de Proteínas , Proteína Tumoral p73/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Proteína X Asociada a bcl-2/genética
4.
Oncol Rep ; 43(1): 169-176, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31789402

RESUMEN

Osteosarcomas are the most commonly occurring malignant bone cancer in young individuals. The survival rate of patients with metastatic osteosarcoma is low and has been stagnant for over two decades. We previously demonstrated that the glutamate release inhibitor, riluzole inhibits osteosarcoma cell growth. Towards the development of more effective therapy, we investigated the delivery of riluzole in human metastatic osteosarcoma xenografts in mice. We compared the efficacy of riluzole delivery by intraperitoneally injecting either free riluzole or riluzole released via two different shapes of iron oxide nanoparticles (nanocage or nanosphere) of size 15±2.5 nm. We monitored tumor size using Vernier calipers and bioluminescence assay and found a significant reduction in tumor size in the riluzole­treated groups when injected, either in free form or via nanoparticles, compared to the control groups (PBS, nanosphere or nanocage). Importantly, nanocage­delivered riluzole was most effective in reducing tumor size in the xenograft nude mice. While riluzole delivery induced apoptosis in tumor tissues in all three groups of riluzole­treated animals, it was highest in tumors from the nanocage­delivered riluzole group. Therefore, we conclude that riluzole is an effective drug to reduce tumor size in osteosarcoma and the efficacy of riluzole as a apoptotic and tumor­reducing drug is enhanced when delivered via nanocage.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Compuestos Férricos/química , Osteosarcoma/tratamiento farmacológico , Riluzol/administración & dosificación , Animales , Neoplasias Óseas/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos , Humanos , Inyecciones Intraperitoneales , Masculino , Nanopartículas del Metal , Ratones , Ratones Desnudos , Osteosarcoma/patología , Riluzol/química , Riluzol/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
PLoS One ; 12(2): e0171256, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28231291

RESUMEN

Osteosarcomas are malignant tumors of bone, most commonly seen in children and adolescents. Despite advances in modern medicine, the poor survival rate of metastatic osteosarcoma has not improved in two decades. In the present study we have investigated the effect of Riluzole on a human and mouse metastatic osteosarcoma cells. We show that LM7 cells secrete glutamate in the media and that mGluR5 receptors are required for the proliferation of LM7 cells. Riluzole, which is known to inhibit glutamate release, inhibits proliferation, induces apoptosis and prevents migration of LM7 cells. This is also seen with Fenobam, a specific blocker of mGluR5. We also show that Riluzole alters the phosphorylation status of AKT/P70 S6 kinase, ERK1/2 and JNK1/2. Thus Riluzole is an effective drug to inhibit proliferation and survival of osteosarcoma cells and has therapeutic potential for the treatment of osteosarcoma exhibiting autocrine glutamate signaling.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Osteosarcoma/tratamiento farmacológico , Receptor del Glutamato Metabotropico 5/metabolismo , Riluzol/farmacología , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Ácido Glutámico/metabolismo , Humanos , Ratones , Osteosarcoma/metabolismo , Osteosarcoma/patología , Receptor del Glutamato Metabotropico 5/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
6.
Nano Lett ; 16(12): 7357-7363, 2016 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-27960523

RESUMEN

Although a range of nanoparticles have been developed as drug delivery systems in cancer therapeutics, this approach faces several important challenges concerning nanocarrier circulation, clearance, and penetration. The impact of reducing nanoparticle size on penetration through leaky blood vessels around tumor microenvironments via enhanced permeability and retention (EPR) effect has been extensively examined. Recent research has also investigated the effect of nanoparticle shape on circulation and target binding affinity. However, how nanoparticle shape affects drug release and therapeutic efficacy has not been previously explored. Here, we compared the drug release and efficacy of iron oxide nanoparticles possessing either a cage shape (IO-NCage) or a solid spherical shape (IO-NSP). Riluzole cytotoxicity against metastatic cancer cells was enhanced 3-fold with IO-NCage. The shape of nanoparticles (or nanocages) affected the drug release point and cellular internalization, which in turn influenced drug efficacy. Our study provides evidence that the shape of iron oxide nanoparticles has a significant impact on drug release and efficacy.


Asunto(s)
Dextranos , Portadores de Fármacos , Compuestos Férricos , Nanopartículas , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Humanos , Riluzol/administración & dosificación
7.
PLoS One ; 8(5): e64588, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23724064

RESUMEN

Glioblastomas exploit various molecular pathways to promote glutamate- dependent growth by activating the AMPA (2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl) propanoic acid) receptor, the group II metabotropic glutamate receptor, mGluR, and the epidermal growth factor receptor, EGFR. We hypothesized that targeting more than one of these pathways would be more effective in inhibiting glutamate-dependent growth. Using a model of U87 cell line, we show that blocking glutamate release by Riluzole inhibits cell proliferation. Glutamate-dependent growth is effectively inhibited by a combination of Iressa, an inhibitor of EGFR activation and LY341495, a group II mGluR inhibitor. Treatment of U87 cells with a combination of Iressa and LY341495 inhibits proliferation as indicated by Ki-67 staining, induces apoptosis and inhibits migration of U87 cells more effectively than the treatment by Iressa or LY341495 alone. These results demonstrate that a combinatorial therapy with Iressa and LY341495 is more effective due to synergistic effects of these drugs in inhibiting the growth of glioblastoma.


Asunto(s)
Aminoácidos/farmacología , Movimiento Celular/efectos de los fármacos , Quinazolinas/farmacología , Xantenos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Gefitinib , Glioblastoma/metabolismo , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Humanos , Riluzol/farmacología
8.
J Neurosci ; 33(14): 6123-32, 2013 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-23554493

RESUMEN

The mechanisms by which natural rewards such as sugar affect synaptic transmission and behavior are largely unexplored. Here, we investigate regulation of nucleus accumbens synapses by sucrose intake. Previous studies have shown that AMPA receptor (AMPAR) trafficking is a major mechanism for regulating synaptic strength, and that in vitro, trafficking of AMPARs containing the GluA1 subunit takes place by a two-step mechanism involving extrasynaptic and then synaptic receptor transport. We report that in rat, repeated daily ingestion of a 25% sucrose solution transiently elevated spontaneous locomotion and potentiated accumbens core synapses through incorporation of Ca(2+)-permeable AMPA receptors (CPARs), which are GluA1-containing, GluA2-lacking AMPARs. Electrophysiological, biochemical, and quantitative electron microscopy studies revealed that sucrose training (7 d) induced a stable (>24 h) intraspinous GluA1 population, and that in these rats a single sucrose stimulus rapidly (5 min) but transiently (<24 h) elevated GluA1 at extrasynaptic sites. CPARs and dopamine D1 receptors were required in vivo for elevated locomotion after sucrose ingestion. Significantly, a 7 d protocol of daily ingestion of a 3% solution of saccharin, a noncaloric sweetener, induced synaptic GluA1 similarly to 25% sucrose ingestion. These findings identify multistep GluA1 trafficking, previously described in vitro, as a mechanism for acute regulation of synaptic transmission in vivo by a natural orosensory reward. Trafficking is stimulated by a chemosensory pathway that is not dependent on the caloric value of sucrose.


Asunto(s)
Neuronas/metabolismo , Receptores AMPA/metabolismo , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación , Animales , Proteínas Portadoras , Condicionamiento Operante/fisiología , Dopamina beta-Hidroxilasa/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Técnicas In Vitro , Locomoción/fisiología , Masculino , Microscopía Electrónica de Transmisión , Neuronas/efectos de los fármacos , Núcleo Accumbens/citología , Fosfoproteínas/metabolismo , Densidad Postsináptica/metabolismo , Densidad Postsináptica/ultraestructura , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo , Sinaptosomas/metabolismo , Sinaptosomas/ultraestructura
9.
Nucleic Acids Res ; 39(Database issue): D691-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21067998

RESUMEN

Reactome (http://www.reactome.org) is a collaboration among groups at the Ontario Institute for Cancer Research, Cold Spring Harbor Laboratory, New York University School of Medicine and The European Bioinformatics Institute, to develop an open source curated bioinformatics database of human pathways and reactions. Recently, we developed a new web site with improved tools for pathway browsing and data analysis. The Pathway Browser is an Systems Biology Graphical Notation (SBGN)-based visualization system that supports zooming, scrolling and event highlighting. It exploits PSIQUIC web services to overlay our curated pathways with molecular interaction data from the Reactome Functional Interaction Network and external interaction databases such as IntAct, BioGRID, ChEMBL, iRefIndex, MINT and STRING. Our Pathway and Expression Analysis tools enable ID mapping, pathway assignment and overrepresentation analysis of user-supplied data sets. To support pathway annotation and analysis in other species, we continue to make orthology-based inferences of pathways in non-human species, applying Ensembl Compara to identify orthologs of curated human proteins in each of 20 other species. The resulting inferred pathway sets can be browsed and analyzed with our Species Comparison tool. Collaborations are also underway to create manually curated data sets on the Reactome framework for chicken, Drosophila and rice.


Asunto(s)
Bases de Datos Factuales , Modelos Biológicos , Fenómenos Biológicos , Gráficos por Computador , Bases de Datos Genéticas , Bases de Datos de Proteínas , Regulación de la Expresión Génica , Humanos , Internet , Redes y Vías Metabólicas , Transducción de Señal
10.
Nucleic Acids Res ; 37(Database issue): D619-22, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18981052

RESUMEN

Reactome (http://www.reactome.org) is an expert-authored, peer-reviewed knowledgebase of human reactions and pathways that functions as a data mining resource and electronic textbook. Its current release includes 2975 human proteins, 2907 reactions and 4455 literature citations. A new entity-level pathway viewer and improved search and data mining tools facilitate searching and visualizing pathway data and the analysis of user-supplied high-throughput data sets. Reactome has increased its utility to the model organism communities with improved orthology prediction methods allowing pathway inference for 22 species and through collaborations to create manually curated Reactome pathway datasets for species including Arabidopsis, Oryza sativa (rice), Drosophila and Gallus gallus (chicken). Reactome's data content and software can all be freely used and redistributed under open source terms.


Asunto(s)
Bases de Datos de Proteínas , Fenómenos Fisiológicos , Proteínas/metabolismo , Animales , Humanos , Redes y Vías Metabólicas , Modelos Animales , Proteínas/genética , Proteínas/fisiología , Transducción de Señal , Programas Informáticos , Integración de Sistemas
11.
J Cell Physiol ; 194(2): 117-26, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12494450

RESUMEN

HCF-1 functions as a coactivator for herpes simplex virus VP16 and a number of mammalian transcription factors. Mature HCF-1 is composed of two subunits generated by proteolytic cleavage of a larger precursor at six centrally-located HCF(PRO) repeats. The resulting N- and C-terminal subunits remain tightly associated via two complementary pairs of self-association domains: termed SAS1N-SAS1C and SAS2N-SAS2C. Additional HCF proteins have been identified in mammals (HCF-2) and Caenorhabditis elegans (CeHCF). Both contain well-conserved SAS1 domains but do not undergo proteolytic processing. Thus, the significance of the cleavage and self-association of HCF-1 remains enigmatic. Here, we describe the isolation of the Drosophila HCF homologue (dHCF) using a genetic screen based on conservation of the SAS1 interaction. The N-terminal beta-propeller domain of dHCF supports VP16-induced complex formation and is more similar to mammalian HCF-1 than other homologues. We show that full-length dHCF expressed in Drosophila cells undergoes proteolytic cleavage giving rise to tightly associated N- and C-terminal subunits. As with HCF-1, the SAS1N and SAS1C elements of dHCF are separated by a large central region, however, this sequence lacks obvious homology to the HCF(PRO) repeats required for HCF-1 cleavage. The conservation of HCF processing in insect cells argues that formation of separate N- and C-terminal subunits is important for HCF function.


Asunto(s)
Drosophila/genética , Péptido Hidrolasas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Factores de Transcripción , Envejecimiento/fisiología , Secuencia de Aminoácidos/genética , Animales , Clonación Molecular , Secuencia Conservada , ADN Complementario/aislamiento & purificación , Proteína Vmw65 de Virus del Herpes Simple/fisiología , Factor C1 de la Célula Huésped , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína/fisiología , Proteínas/química , Temperatura , Transcripción Genética
12.
J Biol Chem ; 277(46): 44292-9, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12235138

RESUMEN

HCF-1 is a cellular protein required by VP16 to activate the herpes simplex virus (HSV) immediate-early genes. VP16 is a component of the viral tegument and, after release into the cell, binds to HCF-1 and translocates to the nucleus to form a complex with the POU domain protein Oct-1 and a VP16-responsive DNA sequence. This VP16-induced complex boosts transcription of the viral immediate-early genes and initiates lytic replication. In uninfected cells, HCF-1 functions as a coactivator for the cellular transcription factors LZIP and GABP and also plays an essential role in cell proliferation. VP16 and LZIP share a tetrapeptide HCF-binding motif recognized by the beta-propeller domain of HCF-1. Here we describe a new cellular HCF-1 beta-propeller domain binding protein, termed HPIP, which contains a functional HCF-binding motif and a leucine-rich nuclear export sequence. We show that HPIP shuttles between the nucleus and cytoplasm in a CRM1-dependent manner and that overexpression of HPIP leads to accumulation of HCF-1 in the cytoplasm. These data suggest that HPIP regulates HCF-1 activity by modulating its subcellular localization. Furthermore, HPIP-mediated export may provide the pool of cytoplasmic HCF-1 required for import of virion-derived VP16 into the nucleus.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Proteínas/metabolismo , Factores de Transcripción , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Northern Blotting , Células COS , Citoplasma/metabolismo , ADN Complementario/metabolismo , Proteínas Fluorescentes Verdes , Proteína Vmw65 de Virus del Herpes Simple/metabolismo , Factor C1 de la Célula Huésped , Humanos , Péptidos y Proteínas de Señalización Intracelular , Leucina/metabolismo , Proteínas Luminiscentes/metabolismo , Ratones , Microscopía Fluorescente , Modelos Biológicos , Datos de Secuencia Molecular , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Homología de Secuencia de Aminoácido , Distribución Tisular , Transfección , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA