Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biosensors (Basel) ; 7(2)2017 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-28587260

RESUMEN

In this present work, a glassy carbon electrode (GCE) was modified primarily with multiwalled carbon nanotubes (MWCNTs) and a composite of MWCNTs and titanium oxide nanoparticles (TiO2NPs). The enzyme horseradish peroxidase (HRP) was immobilized to enhance the sensing ability of GCE. The proposed biosensor was used for the sensitive determination of isoniazid (INZ) in various pharmaceutical samples. The electrochemical behaviour of the developed MWCNT-TiO2NPs-HRP-GCE biosensor was studied by using cyclic voltammetry (CV) and differential pulse voltammetric (DPV) techniques. Fourier transform infrared spectroscopy (FT-IR), X-ray diffraction (XRD), thermogravimetry (TGA) and transmission electron microscopy (TEM) techniques were used to characterize the developed sensor. Phosphate buffer solution (PBS) with pH 7 was used as supporting electrolyte in the present investigation. The cyclic voltammetric results revealed that the increment of anodic peak currents for the enzyme-induced sensor was almost 8-fold greater than that of a bare GCE. The DPV technique exhibited good limit of detection and limit of quantification values, viz., 0.0335 µM and 0.1118 µM, respectively. Moreover, the developed sensor showed long-lasting stability and repeatability without any interferents. This strongly indicates that the fabricated sensor shows outstanding electrochemical performance towards INZ, with excellent selectivity and sensitivity. The developed sensor was successfully applied to pharmaceutical samples and gave good percentages of recoveries.


Asunto(s)
Técnicas Biosensibles/métodos , Isoniazida/aislamiento & purificación , Nanopartículas del Metal/química , Técnicas Electroquímicas , Peroxidasa de Rábano Silvestre/química , Límite de Detección , Nanotubos de Carbono/química , Espectroscopía Infrarroja por Transformada de Fourier , Titanio/química
2.
Tumour Biol ; 39(3): 1010428317695921, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28351321

RESUMEN

The phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling pathway is crucial for tumor survival, proliferation, and progression, making it an attractive target for therapeutic intervention. In glioblastoma, activated mammalian target of rapamycin promotes invasive phenotype and correlates with poor patient survival. A wide range of mammalian target of rapamycin inhibitors are currently being evaluated for cytotoxicity and anti-proliferative activity in various tumor types but are not explored sufficiently for controlling tumor invasion and recurrence. We recently reported that mammalian target of rapamycin inhibitors-rapamycin, temsirolimus, torin 1, and PP242-suppressed invasion and migration promoted by tumor necrosis factor-alpha and phorbol-myristate-acetate in glioblastoma cells. As aggressive invasion and migration of tumors are associated with mesenchymal and stem-like cell properties, this study aimed to examine the effect of mammalian target of rapamycin inhibitors on these features in glioblastoma cells. We demonstrate that temsirolimus and torin 1 effectively reduced the constitutive as well as phorbol-myristate-acetate/oncostatin-M-induced expression of mesenchymal markers (fibronectin, vimentin, and YKL40) and neural stem cell markers (Sox2, Oct4, nestin, and mushashi1). The inhibitors significantly abrogated the neurosphere-forming capacity induced by phorbol-myristate-acetate and oncostatin-M. Furthermore, we demonstrate that the drugs dephosphorylated signal transducer and activator transcription factor 3, a major regulator of mesenchymal and neural stem cell markers implicating the role of signal transducer and activator transcription factor 3 in the inhibitory action of these drugs. The findings demonstrate the potential of mammalian target of rapamycin inhibitors as "stemness-inhibiting drugs" and a promising therapeutic approach to target glioma stem cells.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Factor de Transcripción STAT3/genética , Serina-Treonina Quinasas TOR/genética , Línea Celular Tumoral , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibronectinas/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/patología , Humanos , Naftiridinas/administración & dosificación , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Células-Madre Neurales/efectos de los fármacos , Oncostatina M/administración & dosificación , Fosfatidilinositol 3-Quinasas , Factor de Transcripción STAT3/biosíntesis , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Acetato de Tetradecanoilforbol/administración & dosificación , Acetato de Tetradecanoilforbol/análogos & derivados , Vimentina/biosíntesis
3.
Sci Rep ; 6: 22455, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26940200

RESUMEN

Glioblastoma (GBM) is the most aggressive type of brain tumors in adults with survival period <1.5 years of patients. The role of mTOR pathway is documented in invasion and migration, the features associated with aggressive phenotype in human GBM. However, most of the preclinical and clinical studies with mTOR inhibitors are focused on antiproliferative and cytotoxic activity in GBM. In this study, we demonstrate that mTOR inhibitors-rapamycin (RAP), temisirolimus (TEM), torin-1 (TOR) and PP242 suppress invasion and migration induced by Tumor Necrosis Factor-α (TNFα) and tumor promoter, Phorbol 12-myristate 13-acetate (PMA) and also reduce the expression of the TNFα and IL1ß suggesting their potential to regulate factors in microenvironment that support tumor progression. The mTOR inhibitors significantly decreased MMP-2 and MMP-9 mRNA, protein and activity that was enhanced by TNFα and PMA. The effect was mediated through reduction of Protein kinase C alpha (PKC-α) activity and downregulation of NFκB. TNFα- induced transcripts of NFκB targets -VEGF, pentraxin-3, cathepsin-B and paxillin, crucial in invasion were restored to basal level by these inhibitors. With limited therapeutic interventions currently available for GBM, our findings are significant and suggest that mTOR inhibitors may be explored as anti-invasive drugs for GBM treatment.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , FN-kappa B/metabolismo , Invasividad Neoplásica/prevención & control , Proteína Quinasa C-alfa/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Glioblastoma/patología , Humanos , Indoles/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Naftiridinas/farmacología , Fenilacetatos/farmacología , Purinas/farmacología , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Microambiente Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Neoplasia ; 17(2): 225-37, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25748242

RESUMEN

Glioblastoma (GBM), the most malignant of the brain tumors is classified on the basis of molecular signature genes using TCGA data into four subtypes- classical, mesenchymal, proneural and neural. The mesenchymal phenotype is associated with greater aggressiveness and low survival in contrast to GBMs enriched with proneural genes. The proinflammatory cytokines secreted in the microenvironment of gliomas play a key role in tumor progression. The study focused on the role of Oncostatin-M (OSM), an IL-6 family cytokine in inducing mesenchymal properties in GBM. Analysis of TCGA and REMBRANDT data revealed that expression of OSMR but not IL-6R or LIFR is upregulated in GBM and has negative correlation with survival. Amongst the GBM subtypes, OSMR level was in the order of mesenchymal > classical > neural > proneural. TCGA data and RT-PCR analysis in primary cultures of low and high grade gliomas showed a positive correlation between OSMR and mesenchymal signature genes-YKL40/CHI3L1, fibronectin and vimentin and a negative correlation with proneural signature genes-DLL3, Olig2 and BCAN. OSM enhanced transcript and protein level of fibronectin and YKL-40 and reduced the expression of Olig2 and DLL3 in GBM cells. OSM-regulated mesenchymal phenotype was associated with enhanced MMP-9 activity, increased cell migration and invasion. Importantly, OSM induced mesenchymal markers and reduced proneural genes even in primary cultures of grade-III glioma cells. We conclude that OSM-mediated signaling contributes to aggressive nature associated with mesenchymal features via STAT3 signaling in glioma cells. The data suggest that OSMR can be explored as potential target for therapeutic intervention.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Células Madre Mesenquimatosas/metabolismo , Oncostatina M/fisiología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Western Blotting , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Técnica del Anticuerpo Fluorescente Indirecta , Regulación de la Expresión Génica/fisiología , Glioma/patología , Humanos , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Receptores de Interleucina-6/genética , Receptores de Oncostatina M/genética
5.
FEBS Open Bio ; 5: 8-19, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25685660

RESUMEN

Glioblastoma multiforme (GBM) is the most malignant form of brain tumor and is associated with resistance to conventional therapy and poor patient survival. Prostate apoptosis response (Par)-4, a tumor suppressor, is expressed as both an intracellular and secretory/extracellular protein. Though secretory Par-4 induces apoptosis in cancer cells, its potential in drug-resistant tumors remains to be fully explored. Multicellular spheroids (MCS) of cancer cells often acquire multi-drug resistance and serve as ideal experimental models. We investigated the role of Par-4 in Tamoxifen (TAM)-induced cell death in MCS of human cell lines and primary cultures of GBM tumors. TCGA and REMBRANT data analysis revealed that low levels of Par-4 correlated with low survival period (21.85 ± 19.30 days) in GBM but not in astrocytomas (59.13 ± 47.26 days) and oligodendrogliomas (58.04 ± 59.80 days) suggesting low PAWR expression as a predictive risk factor in GBM. Consistently, MCS of human cell lines and primary cultures displayed low Par-4 expression, high level of chemo-resistance genes and were resistant to TAM-induced cytotoxicity. In monolayer cells, TAM-induced cytotoxicity was associated with enhanced expression of Par-4 and was alleviated by silencing of Par-4 using specific siRNA. TAM effectively induced secretory Par-4 in conditioned medium (CM) of cells cultured as monolayer but not in MCS. Moreover, MCS were rendered sensitive to TAM-induced cell death by exposure to conditioned medium (CM)-containing Par-4 (derived from TAM-treated monolayer cells). Also TAM reduced the expression of Akt and PKCζ in GBM cells cultured as monolayer but not in MCS. Importantly, combination of TAM with inhibitors to PI3K inhibitor (LY294002) or PKCζ resulted in secretion of Par-4 and cell death in MCS. Since membrane GRP78 is overexpressed in most cancer cells but not normal cells, and secretory Par-4 induces apoptosis by binding to membrane GRP78, secretory Par-4 is an attractive candidate for potentially overcoming therapy-resistance not only in malignant glioma but in broad spectrum of cancers.

6.
PLoS One ; 9(2): e88505, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24523904

RESUMEN

Gliomas are the most common and aggressive of brain tumors in adults. Cancer stem cells (CSC) contribute to chemoresistance in many solid tumors including gliomas. The function of prostate apoptosis response-4 (Par-4) as a pro-apoptotic protein is well documented in many cancers; however, its role in CSC remains obscure. In this study, we aimed to explore the role of Par-4 in drug-induced cytotoxicity using human glioma stem cell line--HNGC-2 and primary culture (G1) derived from high grade glioma. We show that among the panel of drugs- lomustine, carmustine, UCN-01, oxaliplatin, temozolomide and tamoxifen (TAM) screened, only TAM induced cell death and up-regulated Par-4 levels significantly. TAM-induced apoptosis was confirmed by PARP cleavage, Annexin V and propidium iodide staining and caspase-3 activity. Knock down of Par-4 by siRNA inhibited cell death by TAM, suggesting the role of Par-4 in induction of apoptosis. We also demonstrate that the mechanism involves break down of mitochondrial membrane potential, down regulation of Bcl-2 and reduced activation of Akt and ERK 42/44. Secretory Par-4 and GRP-78 were significantly expressed in HNGC-2 cells on exposure to TAM and specific antibodies to these molecules inhibited cell death suggesting that extrinsic Par-4 is important in TAM-induced apoptosis. Interestingly, TAM decreased the expression of neural stem cell markers--Nestin, Bmi1, Vimentin, Sox2, and Musashi in HNGC-2 cell line and G1 cells implicating its potential as a stemness inhibiting drug. Based on these data and our findings that enhanced levels of Par-4 sensitize the resistant glioma stem cells to drug-induced apoptosis, we propose that Par-4 may be explored for evaluating anti-tumor agents in CSC.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Neoplasias Encefálicas/metabolismo , Resistencia a Antineoplásicos , Glioma/metabolismo , Células Madre Neoplásicas/citología , Antineoplásicos/farmacología , Neoplasias Encefálicas/patología , Caspasa 3/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/patología , Humanos , Membranas Mitocondriales/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Tamoxifeno/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA