Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Undersea Hyperb Med ; 48(3): 287-295, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34390633

RESUMO

OBJECTIVE: Decompression sickness (DCS) causes serious brain hypoxic-ischemic injury. This experiment was designed to observe whether hyperbaric oxygen (HBO2) pretreatment played a neuroprotective effect in decompression sickness rat models and to explore the mechanism of protective effects. METHODS: Sprague-Dawley (SD) male rats were pretreated with HBO2 and then underwent decompression to establish the DCS rat model. Antioxidant capacities were evaluated by detecting peroxides (GPx), superoxide dismutase (SOD), catalase (CAT) activity and malondialdehyde (MDA) content in brains. The levels of metal elements manganese (Mn), zinc (Zn), iron (Fe) and magnesium (Mg) in brain tissues were assessed by flame atomic absorption spectrometry. Necrosis and apoptosis of neurons were assessed by H-E staining and immunohistochemical staining. RESULTS: HBO2 pretreatment reduced the degree of necrosis and apoptosis in brain tissues of decompression sickness rat models. In addition, HBO2 pretreatment increased GPx, SOD and CAT activities and reduced MDA accumulation. It also increased the content of Mn, Zn, Fe and Mg in brain tissue, which are all related to free radical metabolism. CONCLUSION: These results suggested that HBO2 pretreatment has protective effects on brain injury of rats with decompression sickness. The mechanism of the protective effects may be related to reducing oxidative damage by affecting metal elements in vivo.


Assuntos
Encéfalo/metabolismo , Doença da Descompressão/complicações , Oxigenoterapia Hiperbárica/métodos , Animais , Apoptose , Encéfalo/patologia , Química Encefálica , Caspase 3/análise , Catalase/análise , Catalase/metabolismo , Descompressão , Doença da Descompressão/metabolismo , Hipóxia-Isquemia Encefálica/etiologia , Ferro/análise , Ferro/metabolismo , Magnésio/análise , Magnésio/metabolismo , Masculino , Malondialdeído/análise , Malondialdeído/metabolismo , Manganês/análise , Manganês/metabolismo , Necrose , Neurônios/patologia , Proteínas Proto-Oncogênicas c-bcl-2/análise , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/análise , Superóxido Dismutase/metabolismo , Zinco/análise , Zinco/metabolismo , Proteína X Associada a bcl-2/análise
2.
Cell Physiol Biochem ; 41(2): 555-568, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28214847

RESUMO

BACKGROUND: Autophagy disorder contributes to dedifferentiation of arterial smooth muscle cells, but the mechanisms are poorly understood. Here, we sought to investigate the role of scaffolding adaptor p62/SQSTM1 (p62) in phenotype switching of mouse coronary arterial myocytes (CAMs) induced by CD38 gene deficiency or lysosomal dysfunction which blocks autophagic flux in the cells. METHODS: Protein expression was measured by western blot analysis and immunofluorescent staining. Cell cycle and proliferation rate were analyzed by flow cytometry and MTS assay respectively. mRNA abundance was tested by qRT-PCR. RESULTS: CD38 gene deficiency or bafilomycin A1 (baf), a selective lysosomal inhibitor treatment increased proliferation rate and vimentin expression in CAMs which was prevented by p62 gene silencing. Cell percentage in G2/M and G0/G1 phase was decreased and increased by CD38 deficiency or baf treatment, respectively which was accompanied by accrual of cyclin-dependent kinase 1 (CDK1) protein. Although free ubiquitin content was increased, the colocalization of it to CDK1 was markedly decreased in CD38-/- or baf treated CAMs. Furthermore, the changes in both cell cycle and CDK1 ubiquitinylation could be restored by p62 gene silencing. CONCLUSION: The results suggest in CD38-/- or baf treated CAMs, p62 accumulation promotes phenotype transition and proliferation by accelerating cell cycle progress through G2/M which might relate to the compromised ubiquitinylation and degradation of CDK1.


Assuntos
Autofagia , Miócitos Cardíacos/metabolismo , Proteína Sequestossoma-1/metabolismo , ADP-Ribosil Ciclase 1/deficiência , ADP-Ribosil Ciclase 1/genética , Animais , Autofagia/efeitos dos fármacos , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cloroquina/farmacologia , Vasos Coronários/citologia , Lisossomos/metabolismo , Macrolídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/citologia , Fenótipo , RNA Interferente Pequeno/metabolismo , Proteína Sequestossoma-1/antagonistas & inibidores , Proteína Sequestossoma-1/genética , Ubiquitinação/efeitos dos fármacos , Vimentina/genética , Vimentina/metabolismo
3.
Front Biosci (Landmark Ed) ; 22(4): 558-569, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27814632

RESUMO

Collagen deposition is a hallmark of atherosclerosis. Although compromised collagen I degradation has been implied in the pathogenesis of atherosclerosis, the molecular mechanisms are still unclear. Thus, we determined the role of CD38, an enzyme involved in cellular calcium modulation and autophagic flux, in the regulation of collagen I degradation in coronary arterial myocytes (CAMs).In primary cultured CAMs from CD38-/- mice, collagen I protein accumulation but not mRNA abundance was significantly increased compared with cells from CD38+/+ mice either under control or upon TGF-Beta stimulation. Pharmacological inhibition of the formation of autophagosomes with 3-methyladenine or of autophagolysosomes with a lysosomal functional blocker, bafilomycin A1, induced a similar increase in collagen protein levels, while inhibition of the proteasome by MG132 had no effects on collagen I accumulation. In addition, CD38-deficiency did not change the protein expression of matrix metalloprotein-9 (MMP-9) or tissue inhibitor of metalloproteinase-1 (TIMP-1) in CAMs. Confocal microscopy showed that collagen I deposition was mainly lied within lysosomes or autophagosomes in CD38-/- or TGF-Beta treated CAMs. Collagen I deposition increased when CAMs lack CD38 expression or if autophagy was blocked, which is associated with impaired autophagic degradation of collagen I. This CD38 regulation of autophagic flux may represent a novel mechanism for extracellular matrix (ECM) plasticity of coronary arteries upon atherogenic stimulation.


Assuntos
ADP-Ribosil Ciclase 1/genética , Colágeno Tipo I/metabolismo , Vasos Coronários/metabolismo , Glicoproteínas de Membrana/genética , Miócitos Cardíacos/metabolismo , ADP-Ribosil Ciclase 1/deficiência , ADP-Ribosil Ciclase 1/metabolismo , Animais , Autofagia , Células Cultivadas , Colágeno Tipo I/genética , Doença da Artéria Coronariana/etiologia , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Vasos Coronários/citologia , Dieta Ocidental/efeitos adversos , Lisossomos/metabolismo , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/citologia , Fagossomos/metabolismo , Inibidores de Proteassoma/farmacologia , Proteólise , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta/metabolismo
4.
Clin Exp Pharmacol Physiol ; 42(5): 510-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740656

RESUMO

Microgravity-induced vascular remodelling may play an important role in post-spaceflight orthostatic intolerance. In this study, we aimed to investigate the effects of simulated microgravity on monocyte adhesion to aortic endothelium in hindlimb unweighted rats and to elucidate the underlying mechanisms associated with this event. Sprague-Dawley rats were subjected to 4-week hindlimb unweighting to simulate microgravity. The recruitment of monocytes to the abdominal aorta was investigated by en face immunofluorescence staining and monocyte binding assays. The expression of the adhesion molecules E-selectin and vascular cell adhesion molecule-1 as well as the cytokine monocyte chemoattractant protein (MCP)-1 was evaluated by immunohistochemical staining, western blot, and quantitative reverse transcription polymerase chain reaction analyses. Additionally, nuclear factor-κB (NF-κB) activation and the messenger RNA expression levels of E-selectin, vascular cell adhesion molecule-1, and MCP-1 were assessed with the administration of an NF-κB inhibitor, pyrrolidine dithiocarbamate. Results showed that simulated microgravity significantly increased monocyte recruitment to the aortic endothelium, protein expression of E-selectin and MCP-1, and NF-κB activation in the abdominal aorta of rats. Pyrrolidine dithiocarbamate treatment not only significantly inhibited NF-κB activity but also reduced the messenger RNA levels of E-selectin, vascular cell adhesion molecule-1, and MCP-1 as well as monocyte recruitment in the abdominal aorta of hindlimb unweighted rats. These results suggest that simulated microgravity increases monocyte adhesion to rat aortic endothelium via the NF-κB-mediated expression of the adhesion molecule E-selectin and the cytokine MCP-1. Therefore, an NF-κB-mediated inflammatory response may be one of the cellular mechanisms responsible for arterial remodelling during exposure to microgravity.


Assuntos
Aorta Abdominal/citologia , Endotélio Vascular/citologia , Monócitos/citologia , NF-kappa B/metabolismo , Simulação de Ausência de Peso , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Adesão Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Quimiocina CCL2/genética , Selectina E/genética , Endotélio Vascular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Monócitos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Pirrolidinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Tiocarbamatos/farmacologia , Molécula 1 de Adesão de Célula Vascular/genética
5.
Am J Physiol Heart Circ Physiol ; 298(3): H992-H1002, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20061541

RESUMO

The present study determined whether activation of acid sphingomyelinase (ASM) drives membrane proximal lysosomes to fuse to the cell surface, facilitating membrane lipid rafts (LRs) clustering in coronary arterial endothelial cells (CAECs) and leading to endothelial dysfunction. By confocal microscopy, the activators of ASM, phosphatidylinositol (PI), and bis (monoacylglyceryl) phosphate (Bis), and an inducer of ASM, butyrate, were found to increase LRs clustering in bovine CAECs, which was blocked by lysosome fusion inhibitor vacuolin-1. However, arsenic trioxide (Ars), an inducer of de novo synthesis of ceramide, had no such effect. Similarly, vacuolin-1-blockable effects were observed using fluorescence resonance energy transfer detection. Liquid chromatography-electrospray ionization-tandem mass spectrometry analysis demonstrated that all of these treatments, even Ars, increased ceramide production in CAECs. When ASM gene was silenced, all treatments except Ars no longer increased ceramide levels. Furthermore, dynamic fluorescence monitoring in live cells showed that PI and Bis stimulated lysosome-membrane fusion in CAECs. Functionally, PI and Bis impaired endothelium-dependent vasodilation in perfused coronary arteries, which was blocked by vacuolin-1 and a lysosome function inhibitor, bafilomycine. FasL (Fas ligand), a previously confirmed lysosome fusion stimulator as a comparison, also produced a similar effect. It is concluded that ASM activation serves as a triggering mechanism and driving force, leading to fusion of membrane proximal lysosomes into LR clusters on the cell membrane of CAECs, which represents a novel mechanism mediating endothelial dysfunction during death receptor activation or other pathological situation.


Assuntos
Membrana Celular/fisiologia , Vasos Coronários/fisiopatologia , Endotélio Vascular/fisiopatologia , Lisossomos/fisiologia , Fusão de Membrana/fisiologia , Esfingomielina Fosfodiesterase/fisiologia , Animais , Bovinos , Células Cultivadas , Ceramidas/metabolismo , Vasos Coronários/citologia , Vasos Coronários/ultraestrutura , Endotélio Vascular/citologia , Endotélio Vascular/ultraestrutura , Proteína Ligante Fas/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Fusão de Membrana/efeitos dos fármacos , Microdomínios da Membrana/fisiologia , Modelos Animais
6.
Antioxid Redox Signal ; 12(6): 703-12, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19761405

RESUMO

This study explored the mechanism mediating the aggregation of membrane NADPH oxidase (NOX) subunits and subsequent activation of this enzyme in bovine coronary arterial endothelial cells (CAECs). With confocal microscopy, we found that FasL stimulated lipid rafts (LRs) clustering with NOX subunit aggregation and acid sphingomyelinase (ASM) gathering, which was blocked by the siRNA of sortilin, an intracellular protein responsible for the binding and targeting of ASM to lysosomes. Correspondingly, FasL-induced O(2)(.-) production through NOX in LRs fractions was abolished by sortilin siRNA. Further, with flow-cytometry and fluorescence resonance energy transfer (FRET) analysis, we surprisingly demonstrated that after FasL stimulation, sortilin was exposed to cell membranes from lysosomes together with Lamp-1 and ASM, and these lysosomal components were aggregated and form a signaling complex in cell membranes. With co-immunoprecipitation, lysosomal sortilin and ASM were found to interact more strongly when CAECs were stimulated by FasL. Functionally, inhibition of either sortilin expression, lysosome function, LRs clustering, or NOX activity significantly attenuated FasL-induced decrease in nitric oxide (NO) levels. It is concluded that lysosome-targeted ASM, through sortilin, is able to traffic to and expose to cell-membrane surface, which may lead to LRs clustering and NOX activation in CAECs.


Assuntos
Membrana Celular/enzimologia , Vasos Coronários/citologia , Células Endoteliais/enzimologia , Lisossomos/enzimologia , NADPH Oxidases/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/antagonistas & inibidores , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Vasos Coronários/enzimologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Ativação Enzimática , Proteína Ligante Fas/metabolismo , Inativação Gênica , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Lisossomos/metabolismo , RNA Interferente Pequeno/farmacologia
7.
J Cell Mol Med ; 13(9B): 3303-14, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20196779

RESUMO

The present study tested the hypothesis that homocysteine (Hcys)-induced ceramide production stimulates lipid rafts (LRs) clustering on the membrane of glomerular endothelial cells (GECs) to form redox signalling platforms by aggregation and activation of NADPH oxidase subunits and thereby enhances superoxide (O2*-) production, leading to glomerular endothelial dysfunction and ultimate injury or sclerosis. Using confocal microscopy, we first demonstrated a co-localization of LR clusters with NADPH oxidase subunits, gp91(phox) and p47(phox) in the GECs membrane upon Hcys stimulation. Immunoblot analysis of floated detergent-resistant membrane fractions found that in LR fractions NADPH oxidase subunits gp91(phox) and p47(phox) are enriched and that the activity of this enzyme dramatically increased. We also examined the effect of elevated Hcys on the cell monolayer permeability in GECs. It was found that Hcys significantly increased GEC permeability, which was blocked by inhibition of LR redox signalling platform formation. Finally, we found that Hcys-induced enhancement of GEC permeability is associated with the regulation of microtubule stability through these LR-redox platforms. It is concluded that the early injurious effect of Hcys on the glomerular endothelium is associated with the formation of redox signalling platforms via LR clustering, which may lead to increases in glomerular permeability by disruption of microtubule network in GECs.


Assuntos
Células Endoteliais/citologia , Homocisteína/metabolismo , Glomérulos Renais/metabolismo , Rim/patologia , Lipídeos/química , Microdomínios da Membrana/metabolismo , Oxirredução , Animais , Masculino , Glicoproteínas de Membrana/metabolismo , Microscopia Confocal/métodos , Microtúbulos/metabolismo , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Permeabilidade , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA