Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Medicine (Baltimore) ; 99(6): e19125, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32028438

RESUMO

Pain, the main symptom of osteoarthritis (OA), can lead to functional disability in patients with knee OA. Understanding the association factors related to knee pain is important since preventing OA-induced disabilities can be achieved by modifying these pain-associated issues. Therefore, this study was aimed to investigate the association factors for OA-induced knee pain in Taiwanese patients who received total knee replacements (TKR).In this retrospective study, 357 subjects who had undergone TKR at the Taipei Municipal Wan-Fang Hospital were recruited. The distribution of pain severity among patients with knee OA was evaluated. Demographic data and clinical parameters were analyzed to determine relationships between these variables and the severity of knee OA pain.Of the 357 patients studied, 54% and 33% had moderate and severe knee pain, respectively. Furthermore, a multivariate logistic regression analysis revealed that serum creatinine (>1.5 mg/dL) and an estimated glomerular filtration rate (eGFR) (<60 mL/min/1.73 m) were significantly associated with severe knee pain in OA patients. A significant correlation between severe knee pain and serum creatinine or eGFR was demonstrated by Pearson correlations.Taken together, the renal insufficiency defined by an elevated serum creatinine or a low eGFR in OA patients who required TKR was associated with severe knee pain. These variables must be considered while treating knee OA pain, especially in those patients with severe pain.


Assuntos
Artralgia/etiologia , Artroplastia do Joelho/efeitos adversos , Osteoartrite do Joelho/complicações , Insuficiência Renal/complicações , Idoso , Creatinina/sangue , Feminino , Taxa de Filtração Glomerular , Humanos , Masculino , Medição da Dor , Estudos Retrospectivos
2.
J Neurosurg ; 124(6): 1654-64, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26613168

RESUMO

OBJECT Hypoxia can induce cell death or trigger adaptive mechanisms to guarantee cell survival. Neuron-derived orphan receptor 1 (NOR-1) works as an early-response protein in response to a variety of environmental stresses. In this study, the authors evaluated the roles of NOR-1 in hypoxia-induced neuronal insults. METHODS Neuro-2a cells were exposed to oxygen/glucose deprivation (OGD). Cell viability, cell morphology, cas-pase-3 activity, DNA fragmentation, and cell apoptosis were assayed to determine the mechanisms of OGD-induced neuronal insults. RNA and protein analyses were carried out to evaluate the effects of OGD on expressions of NOR-1, cAMP response element-binding (CREB), and cellular inhibitor of apoptosis protein 2 (cIAP2) genes. Translations of these gene expressions were knocked down using RNA interference. Mice subjected to traumatic brain injury (TBI) and NOR-1 was immunodetected. RESULTS Exposure of neuro-2a cells to OGD decreased cell viability in a time-dependent manner. Additionally, OGD led to cell shrinkage, DNA fragmentation, and cell apoptosis. In parallel, treatment of neuro-2a cells with OGD time dependently increased cellular NOR-1 mRNA and protein expressions. Interestingly, administration of TBI also augmented NOR-1 levels in the impacted regions of mice. As to the mechanism, exposure to OGD increased nuclear levels of the transcription factor CREB protein. Downregulating CREB expression using RNA interference simultaneously inhibited OGD-induced NOR-1 mRNA expression. Also, levels of cIAP2 mRNA and protein in neuro-2a cells were augmented by OGD. After reducing cIAP2 translation, OGD-induced cell death was reduced. Sequentially, application of NOR-1 small interfering RNA to neuro-2a cells significantly inhibited OGD-induced cIAP2 mRNA expression and concurrently alleviated hypoxia-induced alterations in cell viability, caspase-3 activation, DNA damage, and cell apoptosis. CONCLUSIONS This study shows that NOR-1 can transduce survival signals in neuronal cells responsible for hypoxiainduced apoptotic insults through activation of a CREB/cIAP2-dependent mechanism.


Assuntos
Apoptose/fisiologia , Hipóxia Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Glucose/deficiência , Hipóxia/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos ICR , RNA Mensageiro/metabolismo
3.
Toxicology ; 339: 40-50, 2016 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-26655082

RESUMO

Glutamate can activate NMDA receptor (NMDAR) and subsequently induces excitotoxic neuron loss. However, roles of NMDARs in the blood-brain barrier (BBB) are little known. This study used a mouse cerebrovascular endothelial cell (MCEC) model to evaluate the effects of NMDAR activation on maintenance of the BBB and its possible mechanisms. Analysis of confocal microscopy revealed expressions of NMDAR subunits, GluN1 and GLUN2B, in MCECs. An immunoblot assay further showed the existence of GluN1 in plasma membranes of MCECs. In brain tissues, a confocal microscopic analysis demonstrated co-localization of GluN1 and factor VIII, a biomarker of MCECs. In addition, GluN1 mRNA was detected in MCECs and the brain. Functional assays showed that exposure of MCECs to NMDA increased calcium influx. Separately, NMDA suppressed transendothelial electrical resistance values, levels of occludin, and occludin tight junctions. As to the mechanism, NMDA stimulated sequential phosphorylations of extracellular signal-regulated kinase (ERK)1/2 and mitogen-activated ERK (MEK)1. Interestingly, amounts of matrix metalloproteinase (MMP)2 and MMP9 in MCECs were augmented by NMDA. The NMDA-induced alterations in ERK1/2 phosphorylation and occludin levels were reversed by pretreatment with PD98059, a MEK inhibitor, and MK-801, a NMDAR antagonist, respectively. Therefore, this study shows the functional presence of NMDARs in MCECs, and NMDAR activation can disrupt the MCEC-constructed tight junction barrier via activation of the MEK1/2-ERK1/2 signaling pathway and upregulation of MMP2/9 expressions.


Assuntos
Circulação Cerebrovascular , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Junções Íntimas/efeitos dos fármacos , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Endotélio Vascular/citologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Fator VIII/análise , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Ocludina/biossíntese , Receptores de N-Metil-D-Aspartato/biossíntese , Receptores de N-Metil-D-Aspartato/genética
4.
PLoS One ; 8(8): e72404, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23940812

RESUMO

Lipopolysaccharide (LPS) is a critical factor for inducing acute lung injury. GATA-2, a transcription factor, contributes to the control of cell activity and function. Exposure of RAW 264.7 cells to LPS induced interleukin (IL)-1ß mRNA and protein expression and GATA-2 translocation from the cytoplasm to nuclei in concentration- and time-dependent manners. A bioinformatic search revealed that GATA-2-specific binding elements exist in the 5'-promoter region of the il-1ß gene. LPS could enhance the transactivation activity of GATA-2 in macrophages. Knocking-down translation of GATA-2 mRNA using RNA interference significantly alleviated LPS-induced IL-1ß mRNA and protein expression. As to the mechanism, transfection of toll-like receptor (TLR) 4 small interfering (si)RNA into macrophages concurrently decreased LPS-caused increases in nuclear GATA-2 levels. Sequentially, treatment with myeloid differentiation factor 88 (MyD88) siRNA decreased LPS-induced phosphorylation of mitogen-activated protein kinases (MAPKs) kinase 1/2 and subsequent translocation of GATA-2. Reducing MAPK activities using specific inhibitors simultaneously decreased GATA-2 activation. Furthermore, exposure of primary macrophages to LPS significantly increased the transactivation activities of GATA-2 and IL-1ß mRNA and protein expression. Transfection of GATA-2 siRNA inhibited LPS-induced IL-1ß mRNA expression. Results of this study show that LPS induction of il-1ß gene expression in macrophages is mediated by GATA-2 via activation of TLR4, MyD88, and MAPKs.


Assuntos
Fator de Transcrição GATA2/fisiologia , Interleucina-1beta/genética , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/fisiologia , Receptor 4 Toll-Like/fisiologia , Animais , Células Cultivadas , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-1beta/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Macrófagos Peritoneais/metabolismo , Camundongos , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
5.
Surgery ; 154(1): 58-68, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23684363

RESUMO

BACKGROUND: Cerebrovascular endothelial cells (CECs), major component cells of the blood-brain barrier, can be injured by oxidative stress. Propofol can protect cells from oxidative injury. The aim of this study was to evaluate the effects of propofol on nitrosative stress-induced insults to CECs and its possible mechanisms. METHODS: Primary CECs isolated from mouse cerebral capillaries were exposed to2 nitric oxide (NO) donors: sodium nitroprusside (SNP) or S-nitrosoglutathione (GSNO). Cellular NO levels, cell morphologies, and cell viabilities were analyzed. DNA fragmentation and apoptotic cells were quantified using flow cytometry. Proapoptotic Bcl2-antagonist-killer (Bak) and cytochrome c were immunodetected. Bak translocation was analyzed using confocal microscopy. Caspases-9 and -3 activities were measured fluorometrically. Permeability of the CEC monolayer was assayed by measuring the transendothelial electrical resistance. RESULTS: Exposure of CECs to SNP increased cellular NO levels and simultaneously decreased cell viability (P < .01). Meanwhile, treatment of CECs with propofol at a therapeutic concentration (50 µM) decreased SNP-induced cell death (P < .01). SNP induced DNA fragmentation and cell apoptosis, but propofol decreased the cell injury (P < .01). Sequentially, propofol decreased SNP-enhanced Bak levels and translocation from the cytoplasm to mitochondria (P < .05). Exposure of CECs to propofol attenuated GSNO-induced cell death, apoptosis, and caspase-3 activation (P < .01). Additionally, propofol protected CECs against SNP-induced disruption of the CEC monolayer (P < .05). Consequently, SNP-enhanced cascade activation of caspases-9 and -3 was decreased by propofol (P < .01). CONCLUSION: This study suggested that propofol at a therapeutic concentration can protect against nitrosative stress-induced apoptosis of CECs due to downregulation of the intrinsic Bak-mitochondrion-cytochrome c-caspase protease pathway.


Assuntos
Apoptose/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Citoproteção , Células Endoteliais/efeitos dos fármacos , Mitocôndrias/fisiologia , Propofol/farmacologia , Espécies Reativas de Nitrogênio/metabolismo , S-Nitrosoglutationa/farmacologia , Animais , Caspases/fisiologia , Citocromos c/fisiologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Camundongos , Camundongos Endogâmicos ICR , Óxido Nítrico/metabolismo , Nitroprussiato/farmacologia , Estresse Fisiológico , Proteína Killer-Antagonista Homóloga a bcl-2/fisiologia
6.
Respir Res ; 13: 88, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23031213

RESUMO

BACKGROUND: Lipoteichoic acid (LTA), a gram-positive bacterial outer membrane component, can cause septic shock. Our previous studies showed that the gram-negative endotoxin, lipopolysaccharide (LPS), could induce surfactant protein-A (SP-A) production in human alveolar epithelial (A549) cells. OBJECTIVES: In this study, we further evaluated the effect of LTA on SP-A biosynthesis and its possible signal-transducing mechanisms. METHODS: A549 cells were exposed to LTA. Levels of SP-A, nuclear factor (NF)-κB, extracellular signal-regulated kinase 1/2 (ERK1/2), and mitogen-activated/extracellular signal-regulated kinase kinase (MEK)1 were determined. RESULTS: Exposure of A549 cells to 10, 30, and 50 µg/ml LTA for 24 h did not affect cell viability. Meanwhile, when exposed to 30 µg/ml LTA for 1, 6, and 24 h, the biosynthesis of SP-A mRNA and protein in A549 cells significantly increased. As to the mechanism, LTA enhanced cytosolic and nuclear NF-κB levels in time-dependent manners. Pretreatment with BAY 11-7082, an inhibitor of NF-κB activation, significantly inhibited LTA-induced SP-A mRNA expression. Sequentially, LTA time-dependently augmented phosphorylation of ERK1/2. In addition, levels of phosphorylated MEK1 were augmented following treatment with LTA. CONCLUSIONS: Therefore, this study showed that LTA can increase SP-A synthesis in human alveolar type II epithelial cells through sequentially activating the MEK1-ERK1/2-NF-κB-dependent pathway.


Assuntos
Células Epiteliais Alveolares/metabolismo , Lipopolissacarídeos/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteína A Associada a Surfactante Pulmonar/biossíntese , Ácidos Teicoicos/farmacologia , Células Epiteliais Alveolares/efeitos dos fármacos , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Humanos , Immunoblotting , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
7.
J Cereb Blood Flow Metab ; 31(3): 842-54, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20940732

RESUMO

Cerebrovascular endothelial cells (CECs) are crucial components of the blood-brain barrier. Our previous study showed that oxidized low-density lipoprotein (oxLDL) induces apoptosis of CECs. This study was designed to further evaluate the effects of resveratrol on oxLDL-induced CEC insults and its possible molecular mechanisms. Resveratrol decreased the oxidation of LDL into oxLDL. Additionally, the oxLDL-caused oxidative stress and cell damage were attenuated by resveratrol. Exposure of CECs to oxLDL induced cell shrinkage, DNA fragmentation, and cell apoptosis, but resveratrol defended against such injuries. Application of Lox-1 small interference (si)RNA into CECs reduced the translation of this membrane receptor, and simultaneously increased resveratrol protection from oxLDL-induced cell apoptosis. By comparison, overexpression of Lox-1 attenuated resveratrol protection. Resveratrol inhibited oxLDL-induced Lox-1 mRNA and protein expressions. Both resveratrol and Lox-1 siRNA decreased oxLDL-enhanced translocation of proapoptotic Bcl-2-associated X protein (Bax) from the cytoplasm to mitochondria. Sequentially, oxLDL-induced alterations in the mitochondrial membrane potential, cytochrome c release, and activities of caspases-9, -3, and -6 were decreased by resveratrol. Pretreatment with Z-VEID-FMK (benzyloxycarbonyl-Leu-Glu-His-Asp-fluoromethyl ketone) synergistically promoted resveratrol's protection against DNA fragmentation and cell apoptosis. Therefore, this study shows that resveratrol can protect CECs from oxLDL-induced apoptotic insults via downregulating Lox-1-mediated activation of the Bax-mitochondria-cytochrome c-caspase protease pathway.


Assuntos
Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Circulação Cerebrovascular , Células Endoteliais/fisiologia , Lipoproteínas LDL/farmacologia , Receptores Depuradores Classe E/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estilbenos/farmacologia , Animais , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocromos c/metabolismo , Citoproteção , Regulação para Baixo , Células Endoteliais/efeitos dos fármacos , Lipoproteínas LDL/antagonistas & inibidores , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Mitocôndrias/fisiologia , Peptídeo Hidrolases/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo , Resveratrol , Receptores Depuradores Classe E/antagonistas & inibidores , Receptores Depuradores Classe E/genética , Distribuição Tecidual/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
8.
Arch Toxicol ; 85(3): 209-18, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20848084

RESUMO

Alveolar type II epithelial cells can regulate immune responses to sepsis-induced acute lung injury. Lipopolysaccharide (LPS), an outer membrane component of Gram-negative bacteria, can cause septic shock. This study was designed to evaluate the cytotoxic effects of LPS on human alveolar epithelial A549 cells and its possible molecular mechanisms. Exposure of A549 cells to LPS decreased cell viability in concentration- and time-dependent manners. In parallel, LPS concentration- and time-dependently induced apoptosis of A549 cells. Meanwhile, LPS only at a high concentration of 10 µg/ml caused mildly necrotic insults to A549 cells. In terms of the mechanism, exposure of A549 cells to LPS increased the levels of cellular nitric oxide and reactive oxygen species (ROS). Pretreatment with N-acetylcysteine (NAC), an antioxidant, significantly lowered LPS-caused enhancement of intracellular ROS in A549 cells and simultaneously attenuated the apoptotic insults. Sequentially, treatment of A549 cells with LPS caused significant decreases in the mitochondrial membrane potential and biosynthesis of adenosine triphosphate. In succession, LPS triggered the release of cytochrome c from the mitochondria to the cytoplasm. Activities of caspase-9 and caspase-6 were subsequently augmented following LPS administration. Consequently, exposure of A549 cells induced DNA fragmentation in a time-dependent manner. Pretreatment of A549 cells with NAC significantly ameliorated LPS-caused alterations in caspase-9 activation and DNA damage. Therefore, this study shows that LPS specifically induces apoptotic insults to human alveolar epithelial cells through ROS-mediated activation of the intrinsic mitochondrion-cytochrome c-caspase protease mechanism.


Assuntos
Apoptose/fisiologia , Lipopolissacarídeos/farmacocinética , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/metabolismo , Células Epiteliais Alveolares/metabolismo , Caspase 6/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Células Cultivadas , Citocromos c/metabolismo , Fragmentação do DNA , Ativação Enzimática , Células Epiteliais/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias/fisiologia , Óxido Nítrico/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Fatores de Tempo
9.
Immunobiology ; 216(6): 707-14, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21112663

RESUMO

Surfactant proteins (SPs) produced by pulmonary epithelial cells participate in the regulation of sepsis-induced acute lung injury. Our previous study has shown that lipopolysaccharide (LPS), a Gram-negative bacterial outer membrane component, can regulate sp-a gene expression in human lung carcinoma type II epithelial A549 cells. This study was further designed to evaluate the signal-transducing mechanisms of LPS-induced sp-a gene expression. Exposure of A549 cells to LPS induced SP-A mRNA and protein production in time-dependent manners. Application of toll-like receptor 2 (TLR2) siRNA into A549 cells decreased the levels of this receptor and simultaneously inhibited LPS-induced SP-A mRNA expression. Sequentially, LPS enhanced phosphorylation of mitogen-activated protein kinase (MEK) 4 and c-Jun NH(2) terminal kinase 1 (JNK1) in time-dependent manners. Application of TLR2 siRNA decreased LPS-enhanced phosphorylation of MEK4 and JNK1. After knocking-down the translation of MyD88 by RNA interference, the LPS-triggered MEK4 phosphorylation was attenuated. Consequently, LPS augmented the translocation of c-Jun from the cytoplasm to nuclei without affecting c-Fos. Pretreatment of A549 cells with SP600125, an inhibitor of JNK1, significantly lowered LPS-induced SP-A mRNA production. Analyses of an electrophoretic mobility shift assay and a reporter gene further showed that LPS increased the transactivation activity of AP-1 in A549 cells. Therefore, the present study demonstrates that LPS can induce sp-a gene expression in human type II epithelial A549 cells through TLR2-mediated sequential activation of MyD88-MEK4-JNK1-AP-1.


Assuntos
Células Epiteliais Alveolares/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Proteína A Associada a Surfactante Pulmonar/genética , Receptor 2 Toll-Like/metabolismo , Linhagem Celular Tumoral , Humanos , MAP Quinase Quinase 4/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína A Associada a Surfactante Pulmonar/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo
10.
Chin J Physiol ; 50(3): 121-6, 2007 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-17867432

RESUMO

The aim of this study was to examine the inhibitory mechanisms of caffeic acid phenethyl ester (CAPE), which is derived from the propolis of honeybee, in platelet activation. In this study, CAPE (15 and 25 microM) markedly inhibited platelet aggregation stimulated by collagen (2 microg/ml). CAPE (15 and 25 microM) increased cyclic GMP level, and cyclic GMP-induced vasodilator-stimulated phosphoprotein (VASP) Ser157 phosphorylation, but did not increase cyclic AMP in washed human platelets. Rapid phosphorylation of a platelet protein of Mw. 47,000 (P47), a marker of protein kinase C activation, was triggered by phorbol-12, 13-dibutyrate (150 nM). This phosphorylation was markedly inhibited by CAPE (15 and 25 microM). The present study reports a novel and potent antiplatelet agent, CAPE, which involved in the following inhibitory pathways: CAPE increases cyclic GMP/VASP Ser157 phosphorylation, and subsequently inhibits protein kinase C activity, resulting in inhibition of P47 phosphorylation, which ultimately inhibits platelet aggregation. These results strongly indicate that CAPE appears to represent a novel and potent antiplatelet agent for treatment of arterial thromboembolism.


Assuntos
Plaquetas/metabolismo , Ácidos Cafeicos/farmacologia , GMP Cíclico/metabolismo , Álcool Feniletílico/análogos & derivados , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Plaquetas/efeitos dos fármacos , Ácidos Cafeicos/uso terapêutico , Moléculas de Adesão Celular/metabolismo , AMP Cíclico/metabolismo , Humanos , Proteínas dos Microfilamentos/metabolismo , Álcool Feniletílico/farmacologia , Álcool Feniletílico/uso terapêutico , Dibutirato de 12,13-Forbol/farmacologia , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Ativação Plaquetária/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Inibidores da Agregação Plaquetária/uso terapêutico , Proteína Quinase C/antagonistas & inibidores , Tromboembolia/tratamento farmacológico
11.
J Cell Biochem ; 101(6): 1520-31, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17492650

RESUMO

During inflammation, overproduction of nitric oxide (NO) can damage chondrocytes. In this study, we separately evaluated the toxic effects of exogenous and endogenous NO on human chondrocytes and their possible mechanisms. Human chondrocytes were exposed to sodium nitroprusside (SNP), an NO donor, or a combination of lipopolysaccharide (LPS) and interferon-gamma (IFN-gamma) as the exogenous and endogenous sources of NO, respectively. Administration of SNP or a combination of LPS and IFN-gamma in human chondrocytes increased cellular NO levels but decreased cell viability. Exposure to exogenous or endogenous NO significantly induced apoptosis of human chondrocytes. When treated with exogenous or endogenous NO, the mitochondrial membrane potential time-dependently decreased. Exposure to exogenous or endogenous NO significantly enhanced cellular reactive oxygen species (ROS) and cytochrome c (Cyt c) levels. Administration of exogenous or endogenous NO increased caspase-3 activity and consequently induced DNA fragmentation. Suppression of caspase-3 activation by Z-DEVD-FMK decreased NO-induced DNA fragmentation and cell apoptosis. Similar to SNP, exposure of human chondrocytes to S-nitrosoglutathione (GSNO), another NO donor, caused significant increases in Cyt c levels, caspase-3 activity, and DNA fragmentation, and induced cell apoptosis. Pretreatment with N-monomethyl arginine (NMMA), an inhibitor of NO synthase, significantly decreased cellular NO levels, and lowered endogenous NO-induced alterations in cellular Cyt c amounts, caspase-3 activity, DNA fragmentation, and cell apoptosis. Results of this study show that NO from exogenous and endogenous sources can induce apoptotic insults to human chondrocytes via a mitochondria-dependent mechanism.


Assuntos
Condrócitos/imunologia , Mitocôndrias/metabolismo , Óxido Nítrico/metabolismo , Apoptose/fisiologia , Caspase 3/metabolismo , Sobrevivência Celular , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Citocromos c/metabolismo , Fragmentação do DNA , Humanos , Interferon gama/imunologia , Interferon gama/farmacologia , L-Lactato Desidrogenase/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Potenciais da Membrana/fisiologia , Doadores de Óxido Nítrico/farmacologia , Nitroprussiato/farmacologia , Espécies Reativas de Oxigênio/metabolismo , S-Nitrosoglutationa/metabolismo
12.
Toxicol Appl Pharmacol ; 219(1): 42-53, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17239413

RESUMO

Cerebral endothelial cells (CECs) are crucial components of the blood-brain barrier. Oxidized low-density lipoprotein (oxLDL) can induce cell injuries. In this study, we attempted to evaluate the effects of oxLDL on mouse CECs and its possible mechanisms. Mouse CECs were isolated from brain tissues and identified by immunocytochemical staining of vimentin and Factor VIII. oxLDL was prepared from LDL oxidation by copper sulfate. Exposure of mouse CECs to oxLDL decreased cell viability in concentration- and time-dependent manners. oxLDL time-dependently caused shrinkage of cell morphologies. Administration of oxLDL to CECs induced DNA fragmentation in concentration- and time-dependent manners. Analysis of the cell cycle revealed that oxLDL concentration- and time-dependently increased the proportion of CECs which underwent apoptosis. Analysis of confocal microscopy and immunoblot revealed that oxLDL significantly increased cellular and mitochondrial Bax levels as well as the translocation of this proapoptotic protein from the cytoplasm to mitochondria. In parallel with the increase in the levels and translocation of Bax, oxLDL time-dependently decreased the mitochondrial membrane potential. Exposure of mouse CECs to oxLDL decreased the amounts of mitochondrial cytochrome c, but enhanced cytosolic cytochrome c levels. The amounts of intracellular reactive oxygen species were significantly augmented after oxLDL administration. Sequentially, oxLDL increased activities of caspase-9, -3, and -6 in time-dependent manners. Pretreatment with Z-VEID-FMK, an inhibitor of caspase-6, significantly decreased caspase-6 activity and the oxLDL-induced DNA fragmentation and cell apoptosis. This study showed that oxLDL induces apoptotic insults to CECs via signal-transducing events, including enhancing Bax translocation, mitochondrial dysfunction, cytochrome c release, increases in intracellular reactive oxygen species, and cascade activation of caspase-9, -3, and -6. Therefore, oxLDL can damage the blood-brain barrier through induction of CEC apoptosis via a Bax-mitochondria-caspase protease pathway.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Células Endoteliais/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Mitocôndrias/metabolismo , Peptídeo Hidrolases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocromos c/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Imuno-Histoquímica , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Microscopia Confocal , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Membranas Mitocondriais/efeitos dos fármacos , NADH Desidrogenase/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo
13.
Gynecol Obstet Invest ; 63(1): 11-4, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-16837786

RESUMO

Pulmonary hemorrhage is a rare but sometimes fatal complication of hysteroscopy. We present the first case report in which a healthy patient developed lung collapse induced by pulmonary hemorrhage after operative hysteroscopy. The possible etiologies of this rare complication are also discussed.


Assuntos
Hemorragia/etiologia , Histeroscopia/efeitos adversos , Pneumopatias/etiologia , Adulto , Dextranos/efeitos adversos , Feminino , Humanos
14.
Ann N Y Acad Sci ; 1042: 262-71, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15965071

RESUMO

Sepsis is a serious and life-threatening syndrome that often occurs in intensive care unit (ICU) patients. During sepsis, inflammatory cytokines and nitric oxide (NO) can be overproduced, causing tissue and cell injury. Propofol is an intravenous agent used for sedation of ICU patients. Our previous study showed that propofol has immunosuppressive effects on macrophage functions. This study was designed to evaluate the anti-inflammatory and antioxidative effects of propofol on the biosyntheses of tumor necrosis factor alpha (TNF-alpha), interleukin 1beta (IL-1beta), IL-6, and NO in lipopolysaccharide (LPS)- activated macrophages. Exposure to a therapeutic concentration of propofol (50 microM), LPS (1 ng/mL), or a combination of these two drugs for 1, 6, and 24 h was not cytotoxic to the macrophages. ELISA revealed that LPS increased macrophage TNF-alpha, IL-1beta, and IL-6 protein levels in a time-dependent manner, whereas propofol significantly reduced the levels of LPS-enhanced TNF-alpha, IL-1beta, and IL-6 proteins. Data from RT-PCR showed that LPS induced TNF-alpha, IL-1beta, and IL-6 mRNA, but propofol inhibited these effects. LPS also increased NO production and inducible nitric oxide synthase (iNOS) expression in macrophages. Exposure of macrophages to propofol significantly inhibited the LPS-induced NO biosynthesis. The present study shows that propofol, at a therapeutic concentration, has anti-inflammatory and antioxidative effects on the biosyntheses of TNF-alpha, IL-1beta, IL-6, and NO in LPS-activated macro-phages and that the suppressive effects are exerted at the pretranslational level.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Propofol/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular , Regulação da Expressão Gênica , Inflamação , Interleucina-1/biossíntese , Interleucina-1/genética , Interleucina-6/biossíntese , Interleucina-6/genética , Macrófagos/citologia , Camundongos , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/metabolismo , Oxirredução/efeitos dos fármacos , RNA Mensageiro , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
15.
Crit Care Med ; 33(5): 1044-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15891334

RESUMO

OBJECTIVE: Ketamine, an intravenous anesthetic agent, can modulate vascular tone. Nitric oxide (NO), constitutively produced in endothelial cells, contributes to vasoregulation. In this study, we attempted to evaluate the effects of ketamine on NO biosynthesis in human umbilical vein endothelial cells and its possible mechanism. DESIGN: Controlled laboratory study SETTINGS: Research laboratory in a universal hospital. SUBJECTS: Human umbilical vein endothelial cells prepared from human umbilical cord veins were exposed to 1, 10, 100, and 1000 microM ketamine for 1, 6, and 24 hrs. MEASUREMENTS AND MAIN RESULTS: Exposure to 1, 10, and 100 microM ketamine for 1, 6, and 24 hrs was not cytotoxic to human umbilical vein endothelial cells. However, ketamine at 1000 microM significantly caused cell apoptosis. A therapeutic concentration of ketamine (100 microM) time-dependently reduced the levels of nitrite in human umbilical vein endothelial cells. Immunoblot analysis revealed that ketamine time-dependently decreased endothelial NO synthase protein production in human umbilical vein endothelial cells. Results of an assay by reverse-transcription polymerase chain reaction showed that ketamine significantly inhibited levels of endothelial NO synthase messenger RNA. Ketamine time-dependently reduced bradykinin-enhanced intracellular calcium concentrations. Analysis by confocal microscopy further demonstrated the suppressive effects of ketamine on bradykinin-induced calcium mobilization. CONCLUSIONS: A clinically relevant concentration of ketamine can reduce NO biosynthesis. The suppressive mechanisms occur not only by pretranslational inhibition of eNOS expression but also by a posttranslational decrease in endothelial NO synthase activity due to a reduction in intracellular calcium levels.


Assuntos
Analgésicos/farmacologia , Endotélio Vascular/efeitos dos fármacos , Ketamina/farmacologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/biossíntese , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/enzimologia , Endotélio Vascular/metabolismo , Feminino , Humanos , Veias Umbilicais
16.
Acta Anaesthesiol Taiwan ; 42(1): 33-40, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15148692

RESUMO

Evolving from conventional microscopic technologies, confocal microscopy has proved itself to play an important role in the biomedical research during the past decade. Confocal microscope has many advantages over traditional microscope including the ability to look deeply into inside cells with less photodamage and photobleach, reconstruct three-dimensional images, and chart intracellular dynamic events in the living cells. With these remarkable properties and the availability of fluorescent dyes for living cells, the confocal microscopy has been widely used in solving many unknown questions in biological and pharmacological fields. In clinics, confocal microscope has also served as an important tool to observe the living cells in skins and eyes. For anesthesiologists, confocal microscope has made possible novel experimental approaches for the effects of multiple anesthetic agents on cells. Furthermore, the technology of fibreoptical confocal endomicroscopy is now on its way of maturation. It will soon be the era for confocal microscopy to explore the "cell behavior" inside of intact living tissues.


Assuntos
Microscopia Confocal , Anestésicos/farmacologia
17.
J Trauma ; 55(3): 540-5, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14501900

RESUMO

BACKGROUND: Sepsis is a serious and life-threatening syndrome that occurs in intensive care unit patients. Lipopolysaccharide (LPS) has been implicated as one of major causes of sepsis. Nitric oxide (NO) and cytokines are involved in sepsis-induced inflammatory responses. This study is aimed at evaluating the effects of NO on the modulation of pro- and anti-inflammatory cytokines in LPS-activated macrophages and its possible mechanism. METHODS: N-Monomethyl arginine (NMMA), an inhibitor of NO synthase, was used in this study to suppress NO production. Mouse macrophage-like Raw 264.7 cells were exposed to LPS, NMMA, or a combination of NMMA and LPS. Cell viability was determined by the colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-di-phenyltetrazolium bromide assay. The amounts of nitrite, an oxidative product of NO, in the culture medium were quantified according to the Griess reaction method. Enzyme-linked immunosorbent assay and reverse-transcriptase polymerase chain reaction were carried out to determine the expression of tumor necrosis factor (TNF)-alpha, interleukin (IL)-1 beta, and IL-10 in macrophages. RESULTS: Exposure of macrophages to LPS, NMMA, and a combination of NMMA and LPS for 24 hours did not affect cell viability. LPS significantly increased the amounts of nitrite in macrophages (p < 0.01). Treatment with NMMA decreased LPS-enhanced nitrite (p < 0.01) in a concentration-dependent manner. Analyses of enzyme-linked immunosorbent assays and reverse-transcriptase polymerase chain reaction revealed that LPS significantly induced TNF-alpha, IL-1 beta, and IL-10 proteins and mRNA (p < 0.01). A combined treatment with NMMA and LPS significantly blocked LPS-induced TNF-alpha and IL-1 beta (p < 0.01), but synergistically enhanced LPS-induced IL-10 (p < 0.05) protein and RNA. CONCLUSION: This study has shown that NO suppression can inhibit LPS-induced TNF-alpha and IL-1 beta but enhance IL-10, and the modulation occurs at a pretranslational level.


Assuntos
Lipopolissacarídeos/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Óxido Nítrico/farmacologia , ômega-N-Metilarginina/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Interações Medicamentosas , Ensaio de Imunoadsorção Enzimática , Inflamação/metabolismo , Interleucina-1 , Interleucina-10/biossíntese , Interleucina-6/biossíntese , Lipopolissacarídeos/efeitos adversos , Macrófagos/metabolismo , Camundongos , RNA Mensageiro/genética , Fator de Necrose Tumoral alfa/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...