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











Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 47(2): 811-20, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23288365

RESUMO

Mesenchymal stromal cells (MSCs) are emerging as candidate cells for the treatment of neurological diseases because of their neural replacement, neuroprotective, and neurotrophic effects. However, the majority of MSCs transplanted by various routes fail to reach the site of injury, and they have demonstrated only minimal therapeutic benefit in clinical trials. Therefore, enhancing the migration of MSCs to target sites is essential for this therapeutic strategy to be effective. In this study, we assessed whether inhibition of glycogen synthase kinase-3ß (GSK-3ß) increases the migration capacity of MSCs during ex vivo expansion. Human bone marrow MSCs (hBM-MSCs) were cultured with various GSK-3ß inhibitors (LiCl, SB-415286, and AR-A014418). Using a migration assay kit, we found that the motility of hBM-MSCs was significantly enhanced by GSK-3ß inhibition. Western blot analysis revealed increased levels of migration-related signaling proteins such as phospho-GSK-3ß, ß-catenin, phospho-c-Raf, phospho-extracellular signal-regulated kinase (ERK), phospho-ß-PAK-interacting exchange factor (PIX), and CXC chemokine receptor 4 (CXCR4). In addition, real-time polymerase chain reaction demonstrated increased expression of matrix metalloproteinase-2 (MMP-2), membrane-type MMP-1 (MT1-MMP), and ß-PIX. In the reverse approach, treatment with ß-PIX shRNA or CXCR4 inhibitor (AMD 3100) reduced hBM-MSC migration. These findings suggest that inhibition of GSK-3ß during ex vivo expansion of hBM-MSCs may enhance their migration capacity by increasing expression of ß-catenin, phospho-c-Raf, phospho-ERK, and ß-PIX and the subsequent up-regulation of CXCR4. Enhancing the migration capacity of hBM-MSCs by treating these cells with GSK-3ß inhibitors may increase their therapeutic potential.


Assuntos
Movimento Celular/fisiologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/fisiologia , Células-Tronco Mesenquimais/citologia , Receptores CXCR4/biossíntese , Fatores de Troca de Nucleotídeo Guanina Rho/biossíntese , Regulação para Cima/fisiologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Glicogênio Sintase Quinase 3 beta , Humanos , Células-Tronco Mesenquimais/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Receptores CXCR4/fisiologia , Fatores de Troca de Nucleotídeo Guanina Rho/fisiologia , Regulação para Cima/efeitos dos fármacos
2.
Mol Neurobiol ; 47(2): 757-69, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23254998

RESUMO

Phosphatidylinositol 3-kinase (PI3K) plays several important roles in neuronal survival. Activation of the pathway is essential for the neuroprotective mechanisms of materials that shield neuronal cells from many stressful conditions. However, there have been no reports to date about the effect of the direct activation of the pathway in hypoxic injury of neuronal cells. We investigated whether the direct activation of the PI3K pathway inhibits neuronal cell death induced by hypoxia. Primary cultured cortical neurons (PCCNs) were exposed to hypoxic conditions (less than 1 mol% O2) and/or treated with PI3K activator. Hypoxia reduced the viability of PCCNs in a time-dependent manner, but treatment with PI3K significantly restored viability in a concentration-dependent manner. Among the signaling proteins involved in the PI3K pathway, those associated with survival, including Akt and glycogen synthase kinase-3ß, were decreased shortly after exposure to hypoxia and those associated with cell death, including BAX, apoptosis-induced factor, cytochrome c, caspase-9, caspase-3, and poly(ADP-ribose) polymerase (PARP), were increased. However, treatment with PI3K activator normalized the expression levels of those signaling proteins. PARP activity and levels of ATP and NAD(+) altered by hypoxia were also normalized with direct PI3K activation. All these findings suggest that direct and early activation is important for protecting neuronal cells from hypoxic injury.


Assuntos
Trifosfato de Adenosina/metabolismo , Córtex Cerebral/metabolismo , NAD/metabolismo , Neurônios/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Regulação para Cima/fisiologia , Animais , Hipóxia Celular/fisiologia , Células Cultivadas , Córtex Cerebral/patologia , Regulação para Baixo/fisiologia , Ativação Enzimática/fisiologia , Neurônios/patologia , Fosfatidilinositol 3-Quinase/fisiologia , Poli(ADP-Ribose) Polimerases/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
3.
Brain Res ; 1478: 64-73, 2012 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23046589

RESUMO

Recanalization and secondary prevention are the main therapeutic strategies for acute ischemic stroke. Neuroprotective therapies have also been investigated despite unsuccessful clinical results. Coenzyme Q10 (CoQ10), which is an essential cofactor for electron transport in mitochondria, is known to have an antioxidant effect. We investigated the protective effects of CoQ10 against hypoxia in neural stem cells (NSCs). We measured cell viability and levels of intracellular signaling proteins after treatment with several concentrations of CoQ10 under hypoxia-reperfusion. CoQ10 protected NSCs against hypoxia-reperfusion in a concentration-dependent manner by reducing growth inhibition and inhibiting free radical formation. It increased the expression of a number of survival-related proteins such as phosphorylated Akt (pAkt), phosphorylated glycogen synthase kinase 3-ß (pGSK3-ß), and B-cell lymphoma 2 (Bcl-2) in NSCs injured by hypoxia-reperfusion and reduced the expression of death-related proteins such as cleaved caspase-3. We conclude that CoQ10 has effects against hypoxia-reperfusion induced damage to NSCs by enhancing survival signals and decreasing death signals.


Assuntos
Antioxidantes/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ubiquinona/análogos & derivados , Animais , Western Blotting , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Radicais Livres/metabolismo , Marcação In Situ das Extremidades Cortadas , Células-Tronco Neurais/citologia , Ratos , Traumatismo por Reperfusão/metabolismo , Ubiquinona/farmacologia
4.
Neurotoxicology ; 32(6): 879-87, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21683736

RESUMO

The neurotoxicity of L-3,4-dihydroxyphenylalanine (L-DOPA), one of the most important drugs for the treatment of Parkinson's disease, still remains controversial, although much more data on L-DOPA neurotoxicity have been presented. Considering the well known neuroprotective effects of erythropoietin (EPO), the inhibitory effects of EPO on L-DOPA neurotoxicity need to be evaluated. Neuronally differentiated PC12 (nPC12) cells were treated with different concentrations of L-DOPA and/or EPO for 24h. Cell viability was evaluated using trypan blue, 4',6-diamidino-2-phenylindole (DAPI) and TUNEL staining, and cell counting. Free radicals and intracellular signaling protein levels were measured with 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) and Western blotting, respectively. L-DOPA reduced nPC12 cell viability at higher concentrations, but combined treatment with EPO and L-DOPA significantly restored cell viability. Free radicals and hydroxyl radical levels increased by L-DOPA were decreased after combined treatment of L-DOPA and EPO. Levels of survival-related intracellular signaling proteins decreased in nPC12 cells treated with 200 µM L-DOPA but increased significantly in cells treated with 200µM L-DOPA and 5 µM EPO. However, cleaved caspase-3, a death-related protein, increased in nPC12 cells treated with 200 µM L-DOPA but decreased significantly in cells treated with 200 µM L-DOPA and 5 µM EPO. Pretreatment with LY294002, a phosphatidylinositol 3-kinase inhibitor, prior to combined treatment with EPO and L-DOPA almost completely blocked the protective effects of EPO. These results indicate that EPO can prevent L-DOPA neurotoxicity by activating the PI3K pathway as well as reducing oxidative stress.


Assuntos
Eritropoetina/farmacologia , Levodopa/toxicidade , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoproteção , Relação Dose-Resposta a Droga , Humanos , Radical Hidroxila/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Ratos , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
5.
Amyloid ; 12(1): 62-4, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16076613

RESUMO

A 39-year-old man with progressive peripheral neuropathy and autonomic failure showed amyloid deposition on sural nerve biopsy. Direct DNA sequencing of the TTR gene revealed a G to T mutation, causing a Lys to Asn substitution at position 35. This is the first FAP case in Korea which was diagnosed by a DNA test.


Assuntos
Neuropatias Amiloides Familiares/genética , Amiloide/genética , Mutação/genética , Pré-Albumina/genética , Adolescente , Adulto , Idade de Início , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem
6.
Brain Res Mol Brain Res ; 133(2): 176-86, 2005 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-15710234

RESUMO

The effects of diallyl disulfide (DADS), a garlic-derived compound, on the viability of neuronal cells and cell signals, including phosphatidylinositol 3-kinase (PI3K)/Akt, glycogen synthase kinase-3 (GSK-3), cytochrome c, caspase-3, and poly(ADP-ribose) polymerase (PARP), were investigated in PC12 cells neuronally differentiated by nerve growth factor. To evaluate the toxicity of DADS itself, nPC12 cells were treated with several concentrations of DADS, and 3,(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and trypan blue stain revealed that the viability was not affected by low concentration of DADS, up to 20 microM, but it was decreased at higher than this concentration. The levels of free radicals and membrane lipid peroxidation were significantly increased in nPC12 cells when treated with more than 50 microM DADS, and treatment of PC12 cells with 100 microM DADS killed the cells by inhibiting PI3K/Akt and by promoting activation of GSK-3 and caspase-3, release of cytochrome c, and cleavage of PARP. To evaluate the protective effects of low concentration of DADS on oxidative stress-injured nPC12 cells, the viability of the cells (pretreated with DADS for 2 h vs. not pretreated) was evaluated 24 h after exposure to 100 microM H2O2 for 30 min. Compared to the cells treated with 100 microM H2O2 only, pretreatment of the cells with 20 microM DADS before exposure to 100 microM H2O2 increased the viability and induced activation of PI3K and Akt, inactivation of GSK-3, and inhibition of cytochrome c release, caspase-3 activation, and PARP cleavage. These results indicate that low concentration of DADS has neuroprotective effects by activating PI3K/Akt and by inhibiting GSK-3 activation, cytochrome c release, caspase-3 activation, and PARP cleavage, whereas high concentration is rather cytotoxic. Therefore, some specific optimum concentration of DADS may be a new potential therapeutic strategy for oxidative stress injured in vitro model of neurodegenerative diseases.


Assuntos
Compostos Alílicos/farmacologia , Apoptose/efeitos dos fármacos , Dissulfetos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Células PC12/efeitos dos fármacos , Animais , Anti-Hipertensivos/farmacologia , Western Blotting/métodos , Caspase 3 , Caspases/metabolismo , Contagem de Células/métodos , Diferenciação Celular , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Fluoresceínas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Peróxido de Hidrogênio/toxicidade , Indóis , Glicoproteínas de Membrana/metabolismo , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Células PC12/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Sais de Tetrazólio , Tiazóis , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Azul Tripano
7.
Eur J Neurosci ; 20(6): 1461-72, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15355313

RESUMO

Poly(ADP-ribose) polymerase (PARP) plays an important role in ischaemic cell death, and 3-aminobenzamide (3-AB), one of the PARP inhibitors, has a protective effect on ischaemic stroke. We investigated the neuroprotective mechanisms of 3-AB in ischaemic stroke. The occlusion of middle cerebral artery (MCA) was made in 170 Sprague-Dawley rats, and reperfusion was performed 2 h after the occlusion. Another 10 Sprague-Dawley rats were used for sham operation. 3-AB was administered to 85 rats 10 min before the occlusion [3-AB group (n = 85) vs. control group without 3-AB (n = 85)]. Infarct volume and water content were measured, brain magnetic resonance imaging, terminal deoxynucleotidyltransferase (TdT)-mediated dUTP-biotin nick end-labelling (TUNEL) and Cresyl violet staining were performed, and immunoreactivities (IRs) of poly(ADP-ribose) polymer (PAR), cleaved caspase-3, CD11b, intercellular adhesion molecule-1 (ICAM-1), cyclooxygenase-2 (COX-2), phospho-Akt (pAkt) and phospho-glycogen synthase kinase-3 (pGSK-3) were compared in the peri-infarcted region of the 3-AB group and its corresponding ischaemic region of the control group at 2, 8, 24 and 72 h after the occlusion. In the 3-AB group, the infarct volume and the water content were decreased (about 45% and 3.6%, respectively, at 24 h), the number of TUNEL-positive cells was decreased (about 36% at 24 h), and the IRs of PAR, cleaved caspase-3, CD11b, ICAM-1 and COX-2 were significantly reduced, while the IRs of pAkt and pGSK-3 were increased. These results suggest that 3-AB treatment could reduce the infarct volume by reducing ischaemic cell death, its related inflammation and increasing survival signals. The inhibition of PARP could be another potential neuroprotective strategy in ischaemic stroke.


Assuntos
Benzamidas/farmacologia , Isquemia Encefálica/patologia , Morte Celular , Epoprostenol/análogos & derivados , Fármacos Neuroprotetores/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Benzamidas/uso terapêutico , Western Blotting/métodos , Encéfalo/metabolismo , Encéfalo/patologia , Edema Encefálico/etiologia , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Infarto Encefálico/prevenção & controle , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Antígeno CD11b/metabolismo , Caspase 3 , Caspases/metabolismo , Contagem de Células/métodos , Morte Celular/efeitos dos fármacos , Ciclo-Oxigenase 2 , Feminino , Lateralidade Funcional/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Imuno-Histoquímica/métodos , Marcação In Situ das Extremidades Cortadas , Infarto da Artéria Cerebral Média/complicações , Inflamação/etiologia , Inflamação/prevenção & controle , Molécula 1 de Adesão Intercelular/metabolismo , Isoenzimas/metabolismo , Imageamento por Ressonância Magnética/métodos , Masculino , Fármacos Neuroprotetores/uso terapêutico , Poli(ADP-Ribose) Polimerases/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley , Coloração e Rotulagem , Fatores de Tempo
8.
Neurotoxicology ; 25(5): 793-802, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15288510

RESUMO

Epigallocatechin gallate (EGCG) is one of most famous compounds of green tea. EGCG suppresses apoptosis induced by oxidative radical stress through several mechanisms. This study was designed to investigate whether EGCG plays a cytoprotective role by activating phosphatidylinositol-3 kinase (PI3K)/Akt-dependent anti-apoptotic pathway and inhibiting glycogen synthase kinase-3 (GSK-3) activity in oxidative stressed N18D3 neural cells. N18D3 cells, mouse neuroblastoma X dorsal root ganglion hybrid cell line, were pre-treated with EGCG or z-VAD-fmk, non-selective caspase inhibitor used as a control substance, for 2 h. The N18D3 cells were then exposed to low concentration of H(2)O(2) (100 microM) for 30 min, and further incubated for 24 h. MTT (3,[4,5-dimethylthiazol]-2-yl) assay and trypan blue staining were used to identify cell viability. Immunoreactivity (IR) of PI3K, Akt, and GSK-3 beta were measured by Western blotting. MTT assay and trypan blue staining showed that EGCG and z-VAD-fmk significantly increased cell viability, and IR of PI3K, phospho-Akt and phospho-GSK-3 beta was significantly increased in the cells treated with EGCG, but not in z-VAD-fmk treated. These results imply that EGCG has neuroprotective effect by increasing PI3K/Akt-dependent anti-apoptotic signals.


Assuntos
Catequina/análogos & derivados , Catequina/farmacologia , Quinase 3 da Glicogênio Sintase/fisiologia , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Oncogênicas de Retroviridae/fisiologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Inibidores de Caspase , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Proteína Oncogênica v-akt , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
9.
Brain Res Mol Brain Res ; 118(1-2): 72-81, 2003 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-14559356

RESUMO

The effects of epigallocatechin gallate (EGCG) on the phosphoinositide 3-kinase (PI3K)/Akt and glycogen synthase kinase-3 (GSK-3) pathway during oxidative-stress-induced injury were studied using H2O2-treated PC12 cells, which were differentiated by nerve growth factor (NGF). Following 100 microM H2O2 exposure, the viability of differentiated PC12 cells (EGCG or z-VAD-fmk pretreated vs. not pretreated) was evaluated the number of viable cell with Trypan blue and 3,4,5-dimethylthiazol-2-yl (MTT). Additionally, expression of cytochrome c, caspase-3, poly(ADP-ribose) polymerase (PARP), PI3K/Akt and GSK-3 was examined using Western blot analyses. EGCG or z-VAD-fmk-pretreated PC12 cells showed an increase of viability compared to untreated PC12 cells, and pretreatment of PC12 cells with either agent induced a dose-dependent inhibition of caspase-3 activation and PARP cleavage. However, inhibition of cytochrome c release was only detected in EGCG-pretreated cells. Upon examination of the PI3K/Akt and GSK-3 upstream pathway, Western blots of EGCG pretreated cells showed decreased immunoreactivity (IR) of Akt and GSK-3 and increased IR of p85a PI3K, phosphorylated Akt and phosphorylated GSK-3. In contrast, no changes were seen in z-VAD-fmk-pretreated cells. These results show that EGCG affects the PI3K/Akt, GSK-3 pathway as well as downstream signaling, including the cytochrome c and caspase-3 pathways. Therefore, it is suggested that EGCG-mediated activation of PI3K/Akt and inhibition of GSK-3 could be a new potential therapeutic strategy for neurodegenerative diseases associated with oxidative injury.


Assuntos
Apoptose/efeitos dos fármacos , Catequina/análogos & derivados , Catequina/farmacologia , Doenças Neurodegenerativas/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas Serina-Treonina Quinases , Animais , Apoptose/fisiologia , Caspase 3 , Inibidores de Caspase , Caspases/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Fator de Crescimento Neural/farmacologia , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/enzimologia , Estresse Oxidativo/fisiologia , Células PC12 , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA