Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cells ; 12(15)2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37566001

RESUMO

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.


Assuntos
Neoplasias Encefálicas , Vesículas Extracelulares , Glioblastoma , Glioma , Propofol , RNA Longo não Codificante , Humanos , Neoplasias Encefálicas/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Vesículas Extracelulares/metabolismo , Glioblastoma/metabolismo , Glioma/metabolismo , Microglia/metabolismo , Células-Tronco Neoplásicas/patologia , Propofol/farmacologia , RNA Longo não Codificante/genética , Temozolomida/farmacologia
2.
Pharmaceutics ; 15(3)2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36986821

RESUMO

We have developed new formulations of nanohydrogels (NHGs) complexed with DNA devoid of cell toxicity, which, together with their tuned sizes, makes them of great interest for delivering DNA/RNA for foreign protein expression. Transfection results demonstrate that, unlike classical lipo/polyplexes, the new NHGs can be incubated indefinitely with cells without apparent cellular toxicity, resulting in the high expression of foreign proteins for long periods of time. Although protein expression starts with a delay as compared to classical systems, it is sustained for a long period of time, even after passing cells without observation of toxicity. A fluorescently labelled NHG used for gene delivery was detected inside cells very early after incubation, but the protein expression was delayed by many days, demonstrating that there is a time-dependent release of genes from the NHGs. We suggest that this delay is due to the slow but continuous release of DNA from the particles concomitantly with slow but continuous protein expression. Additionally, results obtained after the in vivo administration of m-Cherry/NHG complexes indicated a delayed but prolonged expression of the marker gene in the tissue of administration. Overall, we have demonstrated gene delivery and foreign protein expression using GFP and m-Cherry marker genes complexed with biocompatible nanohydrogels.

3.
Pharmaceutics ; 15(2)2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36839655

RESUMO

The advent of protein expression using m-RNA applied lately for treating the COVID pandemic, and gene editing using CRISPR/Cas9 technology for introducing DNA sequences at a specific site in the genome, are milestones for the urgent need of developing new nucleic acid delivery systems with improved delivery properties especially for in vivo applications. We have designed, synthesized, and characterized novel cross-linked monodispersed nanohydrogels (NHG's) with well-defined sizes ranging between 50-400 nm. The synthesis exploits the formation of self-assemblies generated upon heating a thermo-responsive mixture of monomers. Self-assemblies are formed and polymerized at high temperatures resulting in NHGs with sizes that are predetermined by the sizes of the intermediate self-assemblies. The obtained NHGs were chemically reduced to lead particles with highly positive zeta potential and low cell toxicity. The NHGs form complexes with DNA, and at optimal charge ratio the size of the complexes is concomitant with the size of the NHG's. Thus, the DNA is fully embedded inside the NHGs. The new NHGs and their DNA complexes are devoid of cell toxicity which together with their tunned sizes, make them potential tools for gene delivery and foreign protein expression.

4.
Elife ; 102021 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-34355696

RESUMO

Inactivating mutations in the Methyl-CpG Binding Protein 2 (MECP2) gene are the main cause of Rett syndrome (RTT). Despite extensive research into MECP2 function, no treatments for RTT are currently available. Here, we used an evolutionary genomics approach to construct an unbiased MECP2 gene network, using 1028 eukaryotic genomes to prioritize proteins with strong co-evolutionary signatures with MECP2. Focusing on proteins targeted by FDA-approved drugs led to three promising targets, two of which were previously linked to MECP2 function (IRAK, KEAP1) and one that was not (EPOR). The drugs targeting these three proteins (Pacritinib, DMF, and EPO) were able to rescue different phenotypes of MECP2 inactivation in cultured human neural cell types, and appeared to converge on Nuclear Factor Kappa B (NF-κB) signaling in inflammation. This study highlights the potential of comparative genomics to accelerate drug discovery, and yields potential new avenues for the treatment of RTT.


Assuntos
Proteína 2 de Ligação a Metil-CpG/uso terapêutico , Síndrome de Rett/terapia , Genômica , Humanos , Síndrome de Rett/genética
5.
Cancers (Basel) ; 13(7)2021 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-33916466

RESUMO

Glioblastoma multiforme (GBM) is the most lethal subtype of glioma. Cannabis sativa is used for the treatment of various medical conditions. Around 150 phytocannabinoids have been identified in C. sativa, among them Δ-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) that trigger GBM cell death. However, the optimal combinations of cannabis molecules for anti-GBM activity are unknown. Chemical composition was determined using high-performance liquid chromatography (HPLC) and gas chromatography mass spectrometry (GC/MS). Cytotoxic activity was determined by XTT and lactate dehydrogenase (LDH) assays and apoptosis and cell cycle by fluorescence-activated cell sorting (FACS). F-actin structures were observed by confocal microscopy, gene expression by quantitative PCR, and cell migration and invasion by scratch and transwell assays, respectively. Fractions of a high-THC cannabis strain extract had significant cytotoxic activity against GBM cell lines and glioma stem cells derived from tumor specimens. A standard mix (SM) of the active fractions F4 and F5 induced apoptosis and expression of endoplasmic reticulum (ER)-stress associated-genes. F4 and F5 inhibited cell migration and invasion, altered cell cytoskeletons, and inhibited colony formation in 2 and 3-dimensional models. Combinations of cannabis compounds exert cytotoxic, anti-proliferative, and anti-migratory effects and should be examined for efficacy on GBM in pre-clinical studies and clinical trials.

6.
Cell Death Dis ; 10(2): 82, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30692507

RESUMO

Duchenne muscular dystrophy (DMD) is a progressive, lethal, X-linked disease of skeletal and cardiac muscles caused by mutations in the dystrophin gene. Loss of dystrophin leads to muscle fiber damage and impairment of satellite cell asymmetric division, which are essential for muscle regeneration. These processes ultimately result in muscle wasting and the replacement of the degenerating muscles by fibrogenic cells, a process that leads to the generation of fibrotic tissues. Preimplantation factor (PIF) is an evolutionary conserved 15-amino acid peptide secreted by viable mammalian embryos. Synthetic PIF (sPIF) reproduces the protective/regenerative effects of the endogenous peptide in immune disorders and transplantation models. In this study, we demonstrated that sPIF treatment promoted mouse and human myoblast differentiation and inhibited the expression of collagen 1A1, collagen 1A2, and TGF-ß in DMD patient-derived myoblasts. Additionally, sPIF increased the expression of utrophin, a homolog of dystrophin protein. sPIF effects were mediated via the upregulation of lncRNA H19 and miR-675 and downregulation of let-7. sPIF also inhibited the expression of miR-21, a major fibrosis regulator. The administration of sPIF in mdx mice significantly decreased serum creatine kinase and collagen I and collagen IV expression in the diaphragm, whereas it increased utrophin expression in the diaphragm, heart and quadriceps muscles. In conclusion, sPIF promoted the differentiation of DMD myoblasts, increased utrophin expression via the H19/miRNA-675/let-7 pathway, and reduced muscle fibrosis possibly via the upregulation of miR-675 and inhibition of miR-21 expression. These findings strongly support pursuing sPIF as a potential therapeutic agent for DMD. Moreover, the completion of an sPIF phase I safety trial will further promote the use of sPIF for the treatment of muscular dystrophies.


Assuntos
Proteínas de Transporte/genética , MicroRNAs/genética , Distrofia Muscular de Duchenne/genética , Mioblastos/metabolismo , Utrofina/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos
7.
Biomaterials ; 174: 67-78, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29783118

RESUMO

Duchenne muscular dystrophy (DMD) is a degenerative lethal, X-linked disease of skeletal and cardiac muscles caused by mutations in the dystrophin gene. Cell therapy using different cell types, including mesenchymal stromal cells (MSCs), has been considered as a potential approach for the treatment of DMD. MSCs can be obtained from autologous sources such as bone marrow and adipose tissues or from allogeneic placenta and umbilical cord. The safety and therapeutic impact of these cells has been demonstrated in pre-clinical and clinical studies and their functions are attributed to paracrine effects that are mediated by secreted cytokines and extracellular vesicles. Here, we studied the therapeutic effects of placenta-derived MSCs (PL-MSCs) and their secreted exosomes using mouse and human myoblasts from healthy controls, Duchenne patients and mdx mice. Treatment of myoblasts with conditioned medium or exosomes secreted by PL-MSCs increased the differentiation of these cells and decreased the expression of fibrogenic genes in DMD patient myoblasts. In addition, these treatments also increased the expression of utrophin in these cells. Using a quantitative miR-29c reporter, we demonstrated that the PL-MSC effects were partly mediated by the transfer of exosomal miR-29c. Intramuscular transplantation of PL-MSCs in mdx mice resulted in decreased creatine kinase levels. PL-MSCs significantly decreased the expression of TGF-ß and the level of fibrosis in the diaphragm and cardiac muscles, inhibited inflammation and increased utrophin expression. In vivo imaging analyses using MSCs labeled with gold nanoparticles or fluorescent dyes demonstrated localization of the cells in the muscle tissues up to 3 weeks post treatment. Altogether, these results demonstrate that PL-MSCs and their secreted exosomes have important clinical applications in cell therapy of DMD partly via the targeted delivery of exosomal miR-29c.


Assuntos
Exossomos/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Distrofia Muscular de Duchenne/tratamento farmacológico , Placenta/citologia , Tecido Adiposo/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/metabolismo , Distrofina/metabolismo , Vesículas Extracelulares/metabolismo , Feminino , Corantes Fluorescentes/química , Regulação da Expressão Gênica/efeitos dos fármacos , Ouro/química , Humanos , Nanopartículas Metálicas/química , Camundongos Endogâmicos mdx , MicroRNAs/metabolismo , Mioblastos/efeitos dos fármacos , Placenta/efeitos dos fármacos , Gravidez , Transfecção/métodos , Fator de Crescimento Transformador beta/metabolismo , Cordão Umbilical/metabolismo , Utrofina/metabolismo
8.
Theranostics ; 7(10): 2690-2703, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28819456

RESUMO

Recent studies have proposed that abnormal glutamatergic neurotransmission and glial pathology play an important role in the etiology and manifestation of depression. It was postulated that restoration of normal glutamatergic transmission, by enhancing glutamate uptake, may have a beneficial effect on depression. We examined this hypothesis using unique human glial-like mesenchymal stem cells (MSCs), which in addition to inherent properties of migration to regions of injury and secretion of neurotrophic factors, were differentiated to express high levels of functional glutamate transporters (excitatory amino acid transporters; EAAT). Additionally, gold nanoparticles (GNPs), which serve as contrast agents for CT imaging, were loaded into the cells for non-invasive, real-time imaging and tracking of MSC migration and final location within the brain. MSC-EAAT (2×105; 10 µl) were administered (i.c.v.) to Flinder Sensitive Line rats (FSLs), a genetic model for depression, and longitudinal behavioral and molecular changes were monitored. FSL rats treated with MSC-EAAT showed attenuated depressive-like behaviors (measured by the forced swim test, novelty exploration test and sucrose self-administration paradigm), as compared to controls. CT imaging, Flame Atomic Absorption Spectroscopy analysis and immunohistochemistry showed that the majority of MSCs homed specifically to the dentate gyrus of the hippocampus, a region showing structural brain changes in depression, including loss of glial cells. mRNA and protein levels of EAAT1 and BDNF were significantly elevated in the hippocampus of MSC-EAAT-treated FSLs. Our findings indicate that MSC-EAATs effectively improve depressive-like manifestations, possibly in part by increasing both glutamate uptake and neurotropic factor secretion in the hippocampus.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/biossíntese , Depressão/terapia , Expressão Gênica , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Animais , Comportamento Animal , Giro Denteado/patologia , Depressão/patologia , Modelos Animais de Doenças , Humanos , Estudos Longitudinais , Ratos , Usos Terapêuticos
9.
Stem Cell Res Ther ; 7(1): 149, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27724977

RESUMO

BACKGROUND: Newcastle disease virus (NDV) is an avian paramyxovirus, which selectively exerts oncolytic effects in cancer cells. Mesenchymal stem cells (MSCs) have been reported to affect tumor growth and deliver anti-tumor agents to experimental glioblastoma (GBM). Here, we explored the effects of NDV-infected MSCs derived from different sources, on glioma cells and glioma stem cells (GSCs) and the mechanisms involved in their effects. METHODS: The glioma cell lines (A172 and U87) and primary GSCs that were generated from GBM tumors were used in this study. MSCs derived from bone marrow, adipose tissue or umbilical cord were infected with NDV (MTH-68/H). The ability of these cells to deliver the virus to glioma cell lines and GSCs and the effects of NDV-infected MSCs on cell death and on the stemness and self-renewal of GSCs were examined. The mechanisms involved in the cytotoxic effects of the NDV-infected MSCs and their influence on the radiation sensitivity of GSCs were examined as well. RESULTS: NDV induced a dose-dependent cell death in glioma cells and a low level of apoptosis and inhibition of self-renewal in GSCs. MSCs derived from bone marrow, adipose and umbilical cord that were infected with NDV delivered the virus to co-cultured glioma cells and GSCs. Conditioned medium of NDV-infected MSCs induced higher level of apoptosis in the tumor cells compared with the apoptosis induced by their direct infection with similar virus titers. These results suggest that factor(s) secreted by the infected MSCs sensitized the glioma cells to the cytotoxic effects of NDV. We identified TRAIL as a mediator of the cytotoxic effects of the infected MSCs and demonstrated that TRAIL synergized with NDV in the induction of cell death in glioma cells and GSCs. Moreover, conditioned medium of infected MSCs enhanced the sensitivity of GSCs to γ-radiation. CONCLUSIONS: NDV-infected umbilical cord-derived MSCs may provide a novel effective therapeutic approach for targeting GSCs and GBM and for sensitizing these tumors to γ-radiation.


Assuntos
Glioma/terapia , Glioma/virologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Neoplásicas/citologia , Vírus da Doença de Newcastle/fisiologia , Vírus Oncolíticos/fisiologia , Apoptose/fisiologia , Morte Celular/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura/métodos , Glioblastoma/metabolismo , Glioblastoma/terapia , Glioblastoma/virologia , Glioma/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/virologia , Células-Tronco Neoplásicas/metabolismo , Terapia Viral Oncolítica/métodos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Cordão Umbilical/citologia , Cordão Umbilical/metabolismo
10.
Nanoscale ; 7(48): 20489-96, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26583784

RESUMO

Diabetes mellitus is a chronic metabolic disease, characterized by high blood glucose levels, affecting millions of people around the world. Currently, the main treatment for diabetes requires multiple daily injections of insulin and self-monitoring of blood glucose levels, which markedly affect patients' quality of life. In this study we present a novel strategy for controlled and prolonged glucose regulation, based on the administration of insulin-coated gold nanoparticles (INS-GNPs). We show that both intravenous and subcutaneous injection of INS-GNPs into a mouse model of type 1 diabetes decreases blood glucose levels for periods over 3 times longer than free insulin. We further showed that conjugation of insulin to GNPs prevented its rapid degradation by the insulin-degrading-enzyme, and thus allows controlled and adjustable bio-activity. Moreover, we assessed different sizes and concentrations of INS-GNPs, and found that both parameters have a critical effect in vivo, enabling specific adjustment of blood glucose levels. These findings have the potential to improve patient compliance in diabetes mellitus.


Assuntos
Glicemia/metabolismo , Materiais Revestidos Biocompatíveis , Ouro , Hipoglicemiantes , Insulina , Nanopartículas Metálicas/química , Animais , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Diabetes Mellitus/sangue , Diabetes Mellitus/tratamento farmacológico , Ouro/química , Ouro/farmacologia , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Insulina/química , Insulina/farmacologia , Masculino , Camundongos Endogâmicos NOD
11.
ACS Nano ; 8(9): 9274-85, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25133802

RESUMO

A critical problem in the development and implementation of stem cell-based therapy is the lack of reliable, noninvasive means to image and trace the cells post-transplantation and evaluate their biodistribution, final fate, and functionality. In this study, we developed a gold nanoparticle-based CT imaging technique for longitudinal mesenchymal stem cell (MSC) tracking within the brain. We applied this technique for noninvasive monitoring of MSCs transplanted in a rat model for depression. Our research reveals that cell therapy is a potential approach for treating neuropsychiatric disorders. Our results, which demonstrate that cell migration could be detected as early as 24 h and up to one month post-transplantation, revealed that MSCs specifically navigated and homed to distinct depression-related brain regions. We further developed a noninvasive quantitative CT ruler, which can be used to determine the number of cells residing in a specific brain region, without tissue destruction or animal scarification. This technique may have a transformative effect on cellular therapy, both for basic research and clinical applications.


Assuntos
Encéfalo/diagnóstico por imagem , Rastreamento de Células/métodos , Depressão/diagnóstico por imagem , Depressão/patologia , Células-Tronco Mesenquimais/citologia , Nanopartículas Metálicas , Tomografia Computadorizada por Raios X , Animais , Comportamento Animal , Encéfalo/patologia , Movimento Celular , Depressão/terapia , Ouro/química , Humanos , Transplante de Células-Tronco Mesenquimais , Ratos
12.
Neuro Oncol ; 15(1): 29-40, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23144078

RESUMO

Glioblastomas (GBM) are characterized by resistance to chemotherapy and radiotherapy, and therefore, alternative therapeutic approaches are needed. TRAIL induces apoptosis in cancer but not in normal cells and is considered to be a promising anti-tumor agent. However, its short in vivo half-life and lack of efficient administration modes are serious impediments to its therapeutic efficacy. Nanoparticles (NP) have been used as effective delivery tools for various anticancer drugs. TRAIL was conjugated to magnetic ferric oxide NP by binding the TRAIL primary amino groups to activated double bonds on the surface of the NP. The effect of NP-TRAIL was examined on the apoptosis of glioma cells and self-renewal of glioma stem cells (GSCs). In addition, the ability of the NP-TRAIL to track U251 cell-derived glioma xenografts and to affect cell apoptosis, tumor volume, and survival among xenografted rats was also examined. Conjugation of TRAIL to NP increased its apoptotic activity against different human glioma cells and GSCs, as compared with free recombinant TRAIL. Combined treatment with NP-TRAIL and γ-radiation or bortezomib sensitized TRAIL-resistant GSCs to NP-TRAIL. Using rhodamine-labeled NP and U251 glioma cell-derived xenografts, we demonstrated that the NP-TRAIL were found in the tumor site and induced a significant increase in glioma cell apoptosis, a decrease in tumor volume, and increased animal survival. In summary, conjugation of TRAIL to NP increased its apoptotic activity both in vitro and in vivo. Therefore, NP-TRAIL represents a targeted anticancer agent with more efficient action for the treatment of GBM and the eradication of GSCs.


Assuntos
Apoptose , Glioma/prevenção & controle , Nanopartículas , Células-Tronco Neoplásicas/patologia , Proteínas Recombinantes/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Antineoplásicos/uso terapêutico , Western Blotting , Ácidos Borônicos/uso terapêutico , Bortezomib , Proliferação de Células , Terapia Combinada , Feminino , Compostos Férricos/química , Raios gama , Glioma/mortalidade , Glioma/patologia , Humanos , Técnicas Imunoenzimáticas , Técnicas In Vitro , Células-Tronco Neoplásicas/metabolismo , Pirazinas/uso terapêutico , Ratos , Ratos Nus , Taxa de Sobrevida , Ligante Indutor de Apoptose Relacionado a TNF/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cell Signal ; 23(12): 1936-43, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21777672

RESUMO

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.


Assuntos
Glioma/fisiopatologia , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteína Quinase C/metabolismo , Fator de Resposta Sérica/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Movimento Celular , Glioma/metabolismo , Humanos , Isoenzimas/metabolismo , Proteínas de Membrana , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas do Tecido Nervoso/genética , Fosforilação , Regiões Promotoras Genéticas , Transcrição Gênica , Ativação Transcricional
14.
Neuro Oncol ; 10(3): 236-43, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18403493

RESUMO

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.


Assuntos
Apoptose/fisiologia , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Glioma/metabolismo , Proteínas de Choque Térmico/biossíntese , Chaperonas Moleculares/biossíntese , Biomarcadores Tumorais/análise , Western Blotting , Neoplasias Encefálicas/mortalidade , Caspase 7 , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/fisiologia , Chaperona BiP do Retículo Endoplasmático , Ativação Enzimática/fisiologia , Expressão Gênica , Perfilação da Expressão Gênica , Glioma/mortalidade , Humanos , Imuno-Histoquímica , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Regulação para Cima
15.
Glia ; 55(2): 224-32, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17091491

RESUMO

In this study, we examined the role of PKC in the differentiation of multipotential neural precursor cells (NPCs). We found that the NPCs expressed PKCalpha,beta2,delta,epsilon,zeta and low levels of PKCgamma. The PKC activator, PMA, selectively increased the number of astrocytes, whereas it decreased the generation of neurons and oligodendrocytes. Similarly, overexpression of PKCepsilon increased the differentiation of astrocytes and a PKCepsilonKD mutant abolished PMA effect. PMA phosphorylates PKCepsilon on serine 729. Using a PKCepsilonS729A mutant, we found that the phosphorylation of PKCepsilon on serine 729 was essential for the differentiation of astrocytes induced by PMA. To delineate the mechanisms involved in PMA and PKCepsilon effects, we examined the expression of Notch1, which has been associated with astrocytic differentiation. We found that PMA and PKCepsilon induced a large increase in Notch1 expression and the PKCepsilonS729A mutant abolished PMA effect. Moreover, the PKCepsilonS729A mutant also inhibited the effect of CNTF on astrocytic differentiation and Notch 1 expression. Finally, Notch1 mediated the effect of PMA on astrocytic differentiation, since the gamma-secretase inhibitor L-685,458, and Notch1 silencing abolished PMA effect. Our data suggest an important role of PKCepsilon in astrocytic differentiation and implicate Notch1 as a possible mediator of this effect.


Assuntos
Astrócitos/enzimologia , Encéfalo/enzimologia , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular/fisiologia , Células-Tronco Multipotentes/enzimologia , Proteína Quinase C-épsilon/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Fator Neurotrófico Ciliar/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Células-Tronco Multipotentes/efeitos dos fármacos , Regeneração Nervosa/fisiologia , Fosforilação , Proteína Quinase C-épsilon/efeitos dos fármacos , Proteína Quinase C-épsilon/genética , Interferência de RNA , Ratos , Receptor Notch1/genética , Receptor Notch1/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
16.
Cancer Res ; 66(8): 4139-48, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16618735

RESUMO

In this study, we examined the expression and functions of related to testes-specific, vespid, and pathogenesis protein 1 (RTVP-1) in glioma cells. RTVP-1 was expressed in high levels in glioblastomas, whereas its expression in low-grade astrocytomas and normal brains was very low. Transfection of glioma cells with small interfering RNAs targeting RTVP-1 decreased cell proliferation in all the cell lines examined and induced cell apoptosis in some of them. Overexpression of RTVP-1 increased astrocyte and glioma cell proliferation and the anchorage-independent growth of the cells. In addition, overexpression of RTVP-1 rendered glioma cells more resistant to the apoptotic effect of tumor necrosis factor-related apoptosis-inducing ligand and serum deprivation. To delineate the molecular mechanisms involved in the survival effects of RTVP-1, we examined the expression and phosphorylation of various apoptosis-related proteins. We found that overexpression of RTVP-1 decreased the phosphorylation of c-Jun-NH2-kinase and increased the expression of Bcl2 and that the protective effect of RTVP-1 was partially mediated by Bcl2. Finally, we found that RTVP-1 regulated the invasion of glioma cells as was evident by their enhanced migration through Matrigel and by their increased invasion in a spheroid confrontation assay. The increased invasive potential of the RTVP-1 overexpressors was also shown by the increased activity of matrix metalloproteinase 2 in these cells. Our results suggest that the expression of RTVP-1 is correlated with the degree of malignancy of astrocytic tumors and that RTVP-1 is involved in the regulation of the growth, survival, and invasion of glioma cells. Collectively, these findings suggest that RTVP-1 is a potential therapeutic target in gliomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioma/metabolismo , Glioma/patologia , Proteínas de Neoplasias/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Sequência de Aminoácidos , Apoptose/fisiologia , Astrocitoma/enzimologia , Astrocitoma/metabolismo , Astrocitoma/patologia , Neoplasias Encefálicas/enzimologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Glioblastoma/enzimologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioma/enzimologia , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Proteínas de Membrana , Dados de Sequência Molecular , Invasividade Neoplásica
17.
Cancer Res ; 65(16): 7301-9, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16103081

RESUMO

In this study, we examined the role of protein kinase C (PKC)-epsilon in the apoptosis and survival of glioma cells using tumor necrosis factor-related apoptosis inducing ligand (TRAIL)-stimulated cells and silencing of PKCepsilon expression. Treatment of glioma cells with TRAIL induced activation, caspase-dependent cleavage, and down-regulation of PKCepsilon within 3 to 5 hours of treatment. Overexpression of PKCepsilon inhibited the apoptosis induced by TRAIL, acting downstream of caspase 8 and upstream of Bid cleavage and cytochrome c release from the mitochondria. A caspase-resistant PKCepsilon mutant (D383A) was more protective than PKCepsilon, suggesting that both the cleavage of PKCepsilon and its down-regulation contributed to the apoptotic effect of TRAIL. To further study the role of PKCepsilon in glioma cell apoptosis, we employed short interfering RNAs directed against the mRNA of PKCepsilon and found that silencing of PKCepsilon expression induced apoptosis of various glioma cell lines and primary glioma cultures. To delineate the molecular mechanisms involved in the apoptosis induced by silencing of PKCepsilon, we examined the expression and phosphorylation of various apoptosis-related proteins. We found that knockdown of PKCepsilon did not affect the expression of Bcl2 and Bax or the phosphorylation and expression of Erk1/2, c-Jun-NH2-kinase, p38, or STAT, whereas it selectively reduced the expression of AKT. Similarly, TRAIL reduced the expression of AKT in glioma cells and this decrease was abolished in cells overexpressing PKCepsilon. Our results suggest that the cleavage of PKCepsilon and its down-regulation play important roles in the apoptotic effect of TRAIL. Moreover, PKCepsilon regulates AKT expression and is essential for the survival of glioma cells.


Assuntos
Apoptose/fisiologia , Glioma/enzimologia , Glioma/patologia , Proteína Quinase C/fisiologia , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Caspases/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Ativação Enzimática/efeitos dos fármacos , Inativação Gênica , Glioma/genética , Humanos , Glicoproteínas de Membrana/farmacologia , Dados de Sequência Molecular , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/biossíntese , Proteína Quinase C/genética , Proteína Quinase C-épsilon , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-akt , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
18.
J Biol Chem ; 280(25): 23643-52, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15774464

RESUMO

Protein kinase Cdelta (PKCdelta) regulates cell apoptosis in a cell- and stimulus-specific manner. Here, we studied the role of PKCdelta in the apoptotic effect of TRAIL in glioma cells. We found that transfection of the cells with a PKCdelta kinase-dead mutant (K376R) or with a small interfering RNA targeting the PKCdelta mRNA increased the apoptotic effect of tumor necrosis factor-related apoptosis inducing ligand (TRAIL), whereas overexpression of PKCdelta decreased it. PKCdelta acted downstream of caspase 8 and upstream of cytochrome c release from the mitochondria. TRAIL induced cleavage of PKCdelta within 2-3 h of treatment, which was abolished by caspase 3, 8, and 9 inhibitors. The cleavage of PKCdelta was essential for its protective effect because overexpression of a caspase-resistant mutant (PKCdeltaD327A) did not protect glioma cells from TRAIL-induced apoptosis but rather increased it. TRAIL induced translocation of PKCdelta to the perinuclear region and the endoplasmic reticulum and phosphorylation of PKCdelta on tyrosine 155. Using a PKCdeltaY155F mutant, we found that the phosphorylation of PKCdelta on tyrosine 155 was essential for the cleavage of PKCdelta in response to TRAIL and for its translocation to the endoplasmic reticulum. In addition, phosphorylation of PKCdelta on tyrosine 155 was necessary for the activation of AKT in response to TRAIL. Our results indicate that PKCdelta protects glioma cells from the apoptosis induced by TRAIL and implicate the phosphorylation of PKCdelta on tyrosine 155 and its cleavage as essential factors in the anti-apoptotic effect of PKCdelta.


Assuntos
Glicoproteínas de Membrana/metabolismo , Proteína Quinase C/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Tirosina/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Imunofluorescência , Glioma/patologia , Humanos , Hidrólise , Camundongos , Mutagênese Sítio-Dirigida , Fosforilação , Proteína Quinase C/genética , Proteína Quinase C-delta , Ligante Indutor de Apoptose Relacionado a TNF
19.
J Virol ; 78(18): 10187-92, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15331751

RESUMO

TPA (12-O-tetradecanoylphorbol-13-acetate), a well-known activator of protein kinase C (PKC), can experimentally induce reactivation of Kaposi's sarcoma-associated herpesvirus (KSHV) in certain latently infected cells. We selectively blocked the activity of PKC isoforms by using GF 109203X or rottlerin and demonstrated that this inhibition largely decreased lytic KSHV reactivation by TPA. Translocation of the PKCdelta isoform was evident shortly after TPA stimulation. Overexpression of the dominant-negative PKCdelta mutant supported an essential role for the PKCdelta isoform in virus reactivation, yet overexpression of PKCdelta alone was not sufficient to induce lytic reactivation of KSHV, suggesting that additional signaling molecules participate in this pathway.


Assuntos
Herpesvirus Humano 8/fisiologia , Proteína Quinase C/fisiologia , Acetofenonas/farmacologia , Benzopiranos/farmacologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Herpesvirus Humano 8/efeitos dos fármacos , Herpesvirus Humano 8/patogenicidade , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C-delta , Acetato de Tetradecanoilforbol/farmacologia , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos
20.
Mol Pharmacol ; 66(1): 76-84, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15213298

RESUMO

RasGRP is a family of guanine nucleotide exchange factors that activate small GTPases and contain a C1 domain similar to the one present in protein kinase C (PKC). In this study, we examined the interaction of RasGRP3 and PKC in response to the phorbol ester PMA. In Chinese hamster ovary or LN-229 cells heterologously expressing RasGRP3, phorbol 12-myristate 13-acetate (PMA) induced translocation of RasGRP3 to the perinuclear region and a decrease in the electrophoretic mobility of RasGRP3. The mobility shift was associated with phosphorylation of RasGRP3 on serine residues and seemed to be PKCdelta-dependent because it was blocked by the PKCdelta inhibitor rottlerin as well as by a PKCdelta kinase-dead mutant. Using coimmunoprecipitation, we found that PMA induced the physical association of RasGRP3 with PKCdelta and, using in situ methods, we showed colocalization of PKCdelta and RasGRP3 in the perinuclear region. PKCdelta phosphorylated RasGRP3 in vitro. Previous studies suggest that ectopic expression of RasGRP3 increases activation of Erk1/2. We found that overexpression of either PKCdelta or RasGRP3 increased the activation of Erk1/2 by PMA. In contrast, coexpression of PKCdelta and RasGRP3 yielded a level of phosphorylation of Erk1/2 similar to that of control vector cells. Our results suggest that PKCdelta may act as an upstream kinase associating with and phosphorylating RasGRP3 in response to PMA. The interaction between RasGRP3 and PKCdelta points to the existence of complex cross-talk between various members of the phorbol ester receptors which can have important impact on major signal transduction pathways and cellular processes induced by phorbol esters or DAG


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteína Quinase C/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Animais , Transporte Biológico , Células CHO , Cricetinae , Eletroforese , Feminino , Fatores de Troca do Nucleotídeo Guanina/efeitos dos fármacos , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C-delta , Serina/metabolismo , Transfecção , Fatores ras de Troca de Nucleotídeo Guanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA