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.
Acta Pharmacol Sin ; 37(9): 1178-89, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27498777

RESUMO

AIM: Increasing evidence has shown that environmental factors such as rotenone and paraquat induce neuroinflammation, which contributes to the pathogenesis of Parkinson's disease (PD). In this study, we investigated the molecular mechanisms underlying the repression by menaquinone-4 (MK-4), a subtype of vitamin K2, of rotenone-induced microglial activation in vitro. METHODS: A microglial cell line (BV2) was exposed to rotenone (1 µmol/L) with or without MK-4 treatment. The levels of TNF-α or IL-1ß in 100 µL of cultured media of BV2 cells were measured using ELISA kits. BV2 cells treated with rotenone with or without MK4 were subjected to mitochondrial membrane potential, ROS production, immunofluorescence or immunoblot assays. The neuroblastoma SH-SY5Y cells were treated with conditioned media (CM) of BV2 cells that were exposed to rotenone with or without MK-4 treatment, and the cell viability was assessed using MTT assay. RESULTS: In rotenone-treated BV2 cells, MK-4 (0.5-20 µmol/L) dose-dependently suppressed the upregulation in the expression of iNOS and COX-2 in the cells, as well as the production of TNF-α and IL-1ß in the cultured media. MK-4 (5-20 µmol/L) significantly inhibited rotenone-induced nuclear translocation of NF-κB in BV2 cells. MK-4 (5-20 µmol/L) significantly inhibited rotenone-induced p38 activation, ROS production, and caspase-1 activation in BV2 cells. MK-4 (5-20 µmol/L) also restored the mitochondrial membrane potential that had been damaged by rotenone. Exposure to CM from rotenone-treated BV2 cells markedly decreased the viability of SH-SY5Y cells. However, this rotenone-activated microglia-mediated death of SH-SY5Y cells was significantly attenuated when the BV2 cells were co-treated with MK-4 (5-20 µmol/L). CONCLUSION: Vitamin K2 can directly suppress rotenone-induced activation of microglial BV2 cells in vitro by repressing ROS production and p38 activation.


Assuntos
Poluentes Ambientais/toxicidade , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Microglia/efeitos dos fármacos , Rotenona/toxicidade , Vitamina K 2/análogos & derivados , Animais , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interleucina-1beta/metabolismo , Camundongos , Microglia/imunologia , NF-kappa B/metabolismo , Neuroimunomodulação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Vitamina K 2/farmacologia
2.
Guang Pu Xue Yu Guang Pu Fen Xi ; 36(2): 340-4, 2016 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-27209727

RESUMO

Eu³âº doped BaSrMg (PO4)2 were prepared by a hydrothermal method. The crystal structure and morphology of BaSrMg(PO4)2:Eu³âº phosphor were characterized by X-ray powder diffraction (XRD) and field emission scanning electron microscopy (FESEM). The effects of different pH values (5, 6, 7 and 8) and different reaction temperatures (120, 140, 160, 180 and 200 °C) on the crystal structure and morphology of BaSrMg(PO4)2:Eu³âº phosphor were studied in this paper. The results of XRD indicate that diffraction peaks are sharp and strong only when pH value is 6, meanwhile the FESEM shows the morphology is regular-shaped. The XRD patterns show amorphous halos superimposed with several weak sharp peaks for the samples preparing under the pH values of 5, 7 and 8. It indicates that these three samples are solid solution or mixed phases, which are in accord with the results of FESEM. From the fluorescence spectra, the peaks in the excitation spectra were assigned to the transition from 7F0 to 5D4, 5L8, 5L6 and 5D2, while the peaks of emission spectra corresponding to the transition of 5D1 --> 7F1 and 5D0-->7Fj (J = 0, 1, 2, 3 and 4). The strongest emission peak of the optimized phosphor located at 613 nm (5D0--> 7F2), excited by the main excitation peak with wavelength of 394 nm. The splitting of the emission peaks changes depends on pH values and temperatures, which indicating that luminescence properties is closely related to the crystal structure and morphology of particles.

3.
Stem Cell Res Ther ; 7(1): 73, 2016 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-27194135

RESUMO

BACKGROUND: Proliferation and migration of endothelial progenitor cells (EPCs) play important roles in restoring vascular injuries. ß2 adrenergic receptors (ß2ARs) are widely expressed in many tissues and have a beneficial impact on EPCs regulating neoangiogenesis. The aim of the present study was to determine the effect of overexpressing ß2ARs in infused peripheral blood (PB)-derived EPCs on the re-endothelialization in injured vessels. METHODS: Induction of endothelial injury was performed in male nude mice that were subjected to wire-mediated injury to the carotid artery. Human PB-derived EPCs were transfected with an adenovirus serotype 5 vector expressing ß2AR (Ad5/ß2AR-EPCs) and were examined 48 h later. ß2AR gene expression in EPCs was detected by real-time polymerase chain reaction and Western blot analysis. In vitro, the proliferation, migration, adhesion, and nitric oxide production of Ad5/ß2AR-EPCs were measured. Meanwhile, phosphorylated Akt and endothelial nitric oxide synthase (eNOS), which are downstream of ß2AR signaling, were also elevated. In an in vivo study, CM-DiI-labeled EPCs were injected intravenously into mice subjected to carotid injury. After 3 days, cells recruited to the injury sites were detected by fluorescent microscopy, and the re-endothelialization was assessed by Evans blue dye. RESULTS: In vitro, ß2AR overexpression augmented EPC proliferation, migration, and nitric oxide production and enhanced EPC adhesion to endothelial cell monolayers. In vivo, when cell tracking was used, the number of recruited CM-DiI-labeled EPCs was significantly higher in the injured zone in mice transfused with Ad5/ß2AR-EPCs compared with non-transfected EPCs. The degree of re-endothelialization was also higher in the mice transfused with Ad5/ß2AR-EPCs compared with non-transfected EPCs. We also found that the phosphorylation of Akt and eNOS was increased in Ad5/ß2AR-EPCs. Preincubation with ß2AR inhibitor (ICI118,551), Akt inhibitor (ly294002), or eNOS inhibitor (L-NAME) significantly attenuated the enhanced in vitro function and in vivo re-endothelialization capacity of EPCs induced by ß2AR overexpression. CONCLUSIONS: The present study demonstrates that ß2AR overexpression enhances EPC functions in vitro and enhances the vascular repair abilities of EPCs in vivo via the ß2AR/Akt/eNOS pathway. Upregulation of ß2AR gene expression through gene transfer may be a novel therapeutic target for endothelial repair.


Assuntos
Lesões das Artérias Carótidas/genética , Células Progenitoras Endoteliais/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Proteínas Proto-Oncogênicas c-akt/genética , Reepitelização/genética , Receptores Adrenérgicos beta 2/genética , Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Animais , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Adesão Celular , Movimento Celular , Proliferação de Células , Cromonas/farmacologia , Células Progenitoras Endoteliais/citologia , Regulação da Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Injeções Intravenosas , Masculino , Camundongos Nus , Morfolinas/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação/efeitos dos fármacos , Propanolaminas/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Transfecção
4.
Acta Pharmacol Sin ; 36(11): 1300-7, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26364802

RESUMO

AIM: Parkin has been shown to exert protective effects against 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in different models of Parkinson disease. In the present study we investigated the molecular mechanisms underlying the neuroprotective action of parkin in vitro. METHODS: HEK293, HeLa and PC12 cells were transfected with parkin, parkin mutants, p62 or si-p62. Protein expression and ubiquitination were assessed using immunoblot analysis. Immunoprecipitation assay was performed to identify the interaction between parkin and scaffold protein p62. PC12 and SH-SY5Y cells were treated with 6-OHDA (200 µmol/L), and cell apoptosis was detected using PI and Hoechst staining. RESULTS: In HEK293 cells co-transfected with parkin and p62, parkin was co-immunoprecipitated with p62, and parkin overexpression increased p62 protein levels. In parkin-deficient HeLa cells, transfection with wild-type pakin, but not with ligase activity-deficient pakin mutants, significantly increased p62 levels, suggesting that parkin stabilized p62 through its E3 ligase activity. Transfection with parkin or p62 significantly repressed ERK1/2 phosphorylation in HeLa cells, but transfection with parkin did not repress ERK1/2 phosphorylation in p62-knockdown HeLa cells, suggesting that p62 was involved in parkin-induced inhibition on ERK1/2 phosphorylation. Overexpression of parkin or p62 significantly repressed 6-OHDA-induced ERK1/2 phosphorylation in PC12 cells, and parkin overexpression inhibited 6-OHDA-induced apoptosis in PC12 and SH-SY5Y cells. CONCLUSION: Parkin protects PC12 cells against 6-OHDA-induced apoptosis via ubiquitinating and stabilizing scaffold protein p62, and repressing ERK1/2 activation.


Assuntos
Apoptose , Proteínas de Choque Térmico/metabolismo , Oxidopamina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células HEK293 , Células HeLa , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células PC12 , Ratos , Proteína Sequestossoma-1 , Ubiquitinação
5.
Acta Pharmacol Sin ; 36(9): 1043-52, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26299953

RESUMO

AIM: In the penumbra after focal cerebral ischemia, an increase of protease Omi is linked to a decrease of Hs1-associated protein X-1 (Hax-1), a protein belonging to the Bcl-2 family. In this study we investigated the mechanisms underlying the regulation of Hax-1 by protease Omi in cerebral ischemia/reperfusion (I/R) injury. METHODS: Mouse neuroblastoma N2a cells were subjected to oxygen-glucose deprivation and reoxygenation (OGD/R); cell viability was assessed with MTT assay. Mice underwent 2-h middle cerebral artery occlusion (MCAO) and reperfusion, and the infarct volume was determined with TTC staining. The expression of Omi and Hax-1 was detected using immunoblot and immunofluorescence assays. The mitochondrial membrane potential was measured using TMRM staining. RESULTS: In the brains of MCAO mice, the protein level of Omi was significantly increased, while the protein level of Hax-1 was decreased. Similar changes were observed in OGD/R-treated N2a cells, but the mRNA level of Hax-1 was not changed. Furthermore, in OGD/R-treated N2a cells, knockdown of Omi significantly increased Hax-1 protein level. Immunofluorescence assay showed that Omi and Hax-1 were co-localized in mitochondria of N2a cells. OGD/R caused marked mitochondrial damage and apoptosis in N2a cells, while inhibition of Omi protease activity with UCF-101 (10 µmol/L) or overexpression of Hax-1 could restore the mitochondrial membrane potential and attenuate cell apoptosis. Moreover, pretreatment of MCAO mice with UCF-101 (7.15 mg/kg, ip) could restore Hax-1 expression, inhibit caspase activation, and significantly reduce the infarct volume. CONCLUSION: Protease Omi impairs mitochondrial function by cleaving Hax-1, which induces apoptosis in OGD/R-treated N2a cells and causes I/R injury in MCAO mice.


Assuntos
Encéfalo/patologia , Infarto da Artéria Cerebral Média/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/metabolismo , Neuroblastoma/metabolismo , Proteínas/metabolismo , Traumatismo por Reperfusão/metabolismo , Serina Endopeptidases/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Glucose/metabolismo , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Infarto da Artéria Cerebral Média/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mitocôndrias/metabolismo , Neuroblastoma/patologia , Oxigênio/metabolismo , Traumatismo por Reperfusão/patologia
6.
Acta Pharmacol Sin ; 36(8): 966-75, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26238290

RESUMO

AIM: Omi is an ATP-independent serine protease that is necessary for neuronal function and survival. The aim of this study was to investigate the role of protease Omi in regulating differentiation of mouse neuroblastoma cells and to identify the substrate of Omi involved in this process. METHODS: Mouse neuroblastoma N2a cells and Omi protease-deficient mnd2 mice were used in this study. To modulate Omi and E2F1 expression, N2a cells were transfected with expression plasmids, shRNA plasmids or siRNA. Protein levels were detected using immunoblot assays. The interaction between Omi and E2F1 was studied using immunoprecipitation, GST pulldown and in vitro cleavage assays. N2a cells were treated with 20 µmol/L retinoic acid (RA) and 1% fetal bovine serum to induce neurite outgrowth, which was measured using Image J software. RESULTS: E2F1 was significantly increased in Omi knockdown cells and in brain lysates of mnd2 mice, and was decreased in cells overexpressing wild-type Omi, but not inactive Omi S276C. In brain lysates of mnd2 mice, endogenous E2F1 was co-immunoprecipitated with endogenous Omi. In vitro cleavage assay demonstrated that Omi directly cleaved E2F1. Treatment of N2a cells with RA induced marked differentiation and neurite outgrowth accompanied by significantly increased Omi and decreased E2F1 levels, which were suppressed by pretreatment with the specific Omi inhibitor UCF-101. Knockdown of Omi in N2a cells suppressed RA-induced neurite outgrowth, which was partially restored by knockdown of E2F1. CONCLUSION: Protease Omi facilitates neurite outgrowth by cleaving the transcription factor E2F1 in differentiated neuroblastoma cells; E2F1 is a substrate of Omi.


Assuntos
Fator de Transcrição E2F1/metabolismo , Proteínas Mitocondriais/metabolismo , Neuritos/metabolismo , Serina Endopeptidases/metabolismo , Animais , Linhagem Celular Tumoral , Células HEK293 , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neuritos/ultraestrutura , Neuroblastoma/metabolismo , Neurogênese
7.
Biomed Environ Sci ; 28(4): 253-62, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25966751

RESUMO

OBJECTIVE: This study was aimed to investigate the effects of carbon monoxide releasing molecule (CORM-2), a novel carbon monoxide carrier, on the reendothelialization of carotid artery in rat endothelial denudation model. METHODS: Male rats subjected to carotid artery balloon injury were treated with CORM-2, inactive CORM-2 (iCORM-2) or dimethyl sulfoxide (DMSO). The reendothelialization capacity was evaluated by Evans Blue dye and the immunostaining with anti-CD31 antibody. The number of circulating endothelial progenitor cells (EPCs) was detected by flow cytometry. The proliferation, migration, and adhesion of human umbilical vein endothelial cells (HUVECs) were assessed by using [3H]thymidine, Boyden chamber and human fibronectin respectively. The expressions of protein were detected by using western blot analysis. RESULTS: CORM-2 remarkably accelerated the re-endothelialization 5 d later and inhibited neointima formation 28 d later. In addition, the number of peripheral EPCs significantly increased in CORM-2-treated rats than that in iCORM-2 or DMSO-treated rats after 5 d later. In vitro experiments, CORM-2 significantly enhanced the proliferation, migration and adhesion of HUVECs. The levels of Akt, eNOS phosphorylation, and NO generation in HUVECs were also much higher in CORM-2 treated group. Blocking of PI3K/Akt/eNOS signaling pathway markedly suppressed the enhanced migration and adhesion of HUVECs induced by CORM-2. CONCLUSION: CORM-2 could promote endothelial repair, and inhibit neointima formation after carotid artery balloon injury, which might be associated with the function changes of HUVECs regulated by PI3K/Akt/eNOS pathway.


Assuntos
Monóxido de Carbono/farmacologia , Lesões das Artérias Carótidas/tratamento farmacológico , Artéria Carótida Primitiva/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/metabolismo , Animais , Monóxido de Carbono/metabolismo , Lesões das Artérias Carótidas/imunologia , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/imunologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Adesão Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
8.
Chin Med J (Engl) ; 126(13): 2536-42, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23823830

RESUMO

BACKGROUND: Whether an addition of OAC to double antiplatelet therapy for patients with an indication of chronic oral anticoagulation undergoing PCI-S may improve clinical outcomes is still debated. This meta-analysis aimed to update and re-compare the benefits and risks of triple antithrombotic therapy (TT) with double anti-platelet therapy (DAPT) after in patients who requiring oral anticoagulation after percutaneous coronary interventions with stenting (PCI-s). METHODS: Ten reports of observational retrospective or prospective studies were retrieved, including a total of 6296 patients, follow-up period ranging from 1 year to 2 years. RESULTS: Baseline characteristics were similar in both groups. The main finding of this study is the overall incidence of major adverse cardiovascular events (MACE), myocardial infarction (MI) and stent thrombosis was comparable between two groups. Patients with TT was associated with significant reduction in ischemic stroke (OR: 0.27; 95%CI: 0.13 - 0.57; P = 0.0006) as compared to DAPT. We reaffirmed triple therapy significantly increased the risk of major bleeding (OR: 1.47; 95%CI: 1.22 - 1.78; P < 0.0001) and minor bleeding (OR: 1.55; 95%CI: 1.07 - 2.24; P = 0.02). CONCLUSIONS: Triple therapy is more efficacious in reducing the occurrence of ischemic stroke in PCI-s patients with an indication of chronic oral anticoagulation (OAC), compared with DAPT. However, it significantly increased major and minor risk of bleeding. It is imperative that further prospective randomized controlled trials are required to defne the best therapeutic strategy for patients with an indication of chronic OAC undergoing PCI-s.


Assuntos
Anticoagulantes/uso terapêutico , Fibrinolíticos/administração & dosagem , Intervenção Coronária Percutânea , Inibidores da Agregação Plaquetária/administração & dosagem , Stents , Idoso , Quimioterapia Combinada , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Viés de Publicação
9.
Acta Pharmacol Sin ; 34(5): 651-6, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23564079

RESUMO

AIM: To investigate whether sequestosome 1/p62 (p62), a key cargo adaptor protein involved in both the ubiquitin-proteasome system and the autophagy-lysosome system, could directly regulate autophagy in vitro. METHODS: HEK 293 cells or HeLa cells were transfected with p62-expressing plasmids or siRNA targeting p62. The cells or the cell lysates were subsequently subjected to immunofluorescence assay, immunoprecipitation assay, or immunoblot analysis. In vitro pulldown assay was used to study the interaction of p62 with Bcl-2. RESULTS: Overexpression of p62 significantly increased the basal level of autophagy in both HEK 293 cells and HeLa cells, whereas knockdown of p62 significantly decreased the basal level of autophagy. In vitro pulldown assay showed that p62 directly interacted with Bcl-2. It was observed in HeLa cells that p62 co-localized with Bcl-2. Furthermore, knockdown of p62 in HEK 293 cells significantly increased the amount of Beclin 1 that co-immunoprecipitated with Bcl-2. CONCLUSION: p62 induces autophagy by disrupting the association between Bcl-2 and Beclin 1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Células HEK293 , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Mapas de Interação de Proteínas , RNA Interferente Pequeno/genética , Proteína Sequestossoma-1 , Transfecção , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...