Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Biochem Biophys ; 67(2): 399-414, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22183615

RESUMEN

Adiponectin (Ad), an adipokine exclusively secreted by the adipose tissue, has emerged as a paracrine metabolic regulator as well as a protectant against oxidative stress. Pharmacological approaches of protecting against clinical hyperoxic lung injury during oxygen therapy/treatment are limited. We have previously reported that Ad inhibits the NADPH oxidase-catalyzed formation of superoxide from molecular oxygen in human neutrophils. Based on this premise, we conducted studies to determine whether (i) exogenous Ad would protect against the hyperoxia-induced barrier dysfunction in the lung endothelial cells (ECs) in vitro, and (ii) endogenously synthesized Ad would protect against hyperoxic lung injury in wild-type (WT) and Ad-overexpressing transgenic (AdTg) mice in vivo. The results demonstrated that exogenous Ad protected against the hyperoxia-induced oxidative stress, loss of glutathione (GSH), cytoskeletal reorganization, barrier dysfunction, and leak in the lung ECs in vitro. Furthermore, the hyperoxia-induced lung injury, vascular leak, and lipid peroxidation were significantly attenuated in AdTg mice in vivo. Also, AdTg mice exhibited elevated levels of total thiols and GSH in the lungs as compared with WT mice. For the first time, our studies demonstrated that Ad protected against the hyperoxia-induced lung damage apparently through attenuation of oxidative stress and modulation of thiol-redox status.


Asunto(s)
Adiponectina/metabolismo , Adiponectina/farmacología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Adiponectina/genética , Animales , Bovinos , Hipoxia de la Célula/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Glutatión/metabolismo , Humanos , Peroxidación de Lípido/efectos de los fármacos , Pulmón/patología , Masculino , Ratones , Ratones Transgénicos , Estrés Oxidativo/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo
2.
Cell Biochem Biophys ; 67(2): 415-29, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22183614

RESUMEN

The mechanisms of poultry particulate matter (PM)-induced agricultural respiratory disorders are not thoroughly understood. Hence, it is hypothesized in this article that poultry PM induces the release of interleukin-8 (IL-8) by lung epithelial cells that is regulated upstream by the concerted action of cytosolic phospholipase A2 (cPLA2) and extracellular signal-regulated kinase (ERK). To test this hypothesis, the widely used cultured human lung epithelial cells (A549) were chosen as the model system. Poultry PM caused a significant activation of PLA2 in A549 cells, which was attenuated by AACOCF3 (cPLA2 inhibitor) and PD98059 (ERK-1/2 upstream inhibitor). Poultry PM induced upstream ERK-1/2 phosphorylation and downstream cPLA2 serine phosphorylation, in a concerted fashion, in cells with enhanced association of ERK-1/2 and cPLA2. The poultry PM-induced cPLA2 serine phosphorylation and IL-8 release were attenuated by AACOCF3, PD98059, and by transfection with dominant-negative ERK-1/2 DNA in cells. The poultry PM-induced IL-8 release by the bone marrow-derived macrophages of cPLA2 knockout mice was significantly lower. For the first time, this study demonstrated that the poultry PM-induced IL-8 secretion by human lung epithelial cells was regulated by cPLA2 activation through ERK-mediated serine phosphorylation, suggesting a mechanism of airway inflammation among poultry farm workers.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Interleucina-8/metabolismo , Pulmón/citología , Material Particulado/farmacología , Fosfolipasas A2/metabolismo , Aves de Corral , Animales , Ácido Araquidónico/metabolismo , Proteínas Sanguíneas/farmacología , Células de la Médula Ósea/citología , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Técnicas de Inactivación de Genes , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfolipasas A2/química , Fosfolipasas A2/deficiencia , Fosfolipasas A2/genética , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Serina/metabolismo , Factores de Tiempo
3.
Cell Biochem Biophys ; 67(2): 317-29, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22020799

RESUMEN

Mercury, especially methylmercury (MeHg), is implicated in the etiology of cardiovascular diseases. Earlier, we have reported that MeHg induces phospholipase D (PLD) activation through oxidative stress and thiol-redox alteration. Hence, we investigated the mechanism of the MeHg-induced PLD activation through the upstream regulation by phospholipase A2 (PLA2) and lipid oxygenases such as cyclooxygenase (COX) and lipoxygenase (LOX) in the bovine pulmonary artery endothelial cells (BPAECs). Our results showed that MeHg significantly activated both PLA2 (release of [(3)H]arachidonic acid, AA) and PLD (formation of [(32)P]phosphatidylbutanol) in BPAECs in dose- (0-10 µM) and time-dependent (0-60 min) fashion. The cPLA2-specific inhibitor, arachidonyl trifluoromethyl ketone (AACOCF3), significantly attenuated the MeHg-induced [(3)H]AA release in ECs. MeHg-induced PLD activation was also inhibited by AACOCF3 and the COX- and LOX-specific inhibitors. MeHg also induced the formation of COX- and LOX-catalyzed eicosanoids in ECs. MeHg-induced cytotoxicity (based on lactate dehydrogenase release) was protected by PLA2-, COX-, and LOX-specific inhibitors and 1-butanol, the PLD-generated PA quencher. For the first time, our studies showed that MeHg activated PLD in vascular ECs through the upstream action of cPLA2 and the COX- and LOX-generated eicosanoids. These results offered insights into the mechanism(s) of the MeHg-mediated vascular endothelial cell lipid signaling as an underlying cause of mercury-induced cardiovascular diseases.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Eicosanoides/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Compuestos de Metilmercurio/farmacología , Fosfolipasa D/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Biocatálisis , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiopatología , Bovinos , Relación Dosis-Respuesta a Droga , Eicosanoides/biosíntesis , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , L-Lactato Deshidrogenasa/metabolismo , Lipooxigenasa/metabolismo , Fosfolipasas A2/metabolismo , Fosforilación/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/metabolismo , Arteria Pulmonar/citología , Serina/metabolismo , Factores de Tiempo
4.
Indian J Biochem Biophys ; 50(5): 387-401, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24772960

RESUMEN

The purpose of this study was to elucidate the mechanism of the airborne poultry dust (particulate matter, PM)-induced respiratory tract inflammation, a common symptom in agricultural respiratory diseases. The study was based on the hypothesis that poultry PM would induce the release of inflammatory cytokine interleukin-8 (IL-8) by respiratory epithelial cells under the upstream regulation by cytosolic phospholipase A2 (cPLA2) activation and subsequent formation of cyclooxygenase (COX)- and lipoxygenase (LOX)-catalyzed arachidonic acid (AA) metabolites (eicosanoids). Human lung epithelial cells (A549) in culture were treated with the poultry PM (0.1-1.0 mg) for different lengths of time, following which PLA2 activity, release of eicosanoids and secretion of IL-8 in cells were determined. Poultry PM (1.0 mg/ml) caused a significant activation of PLA2 in a time-dependent manner (15-60 min), which was significantly attenuated by the calcium-chelating agents, cPLA2-specific inhibitor (AACOCF3) and antioxidant (vitamin C) in A549 cells. Poultry PM also significantly induced the release of COX- and LOX-catalyzed eicosanoids (prostaglandins, thromboxane A2 and leukotrienes B4 and C4) and upstream activation of AA LOX in the cells. Poultry PM also significantly induced release of IL-8 by the cells in a dose- and time-dependent manner, which was significantly attenuated by the calcium chelating agents, antioxidants and COX- and LOX-specific inhibitors. The current study for the first time revealed that the poultry PM-induced IL-8 release from the respiratory epithelial cells was regulated upstream by reactive oxygen species, cPLA2-, COX- and LOX-derived eicosanoid lipid signal mediators.


Asunto(s)
Agricultura , Citocinas/metabolismo , Eicosanoides/metabolismo , Material Particulado/farmacología , Mucosa Respiratoria/citología , Transducción de Señal/efectos de los fármacos , Animales , Antioxidantes/farmacología , Ácido Araquidónico/metabolismo , Biocatálisis , Línea Celular , Relación Dosis-Respuesta a Droga , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Interleucina-8/metabolismo , Lipooxigenasas/metabolismo , Material Particulado/química , Fosfolipasas A2 Citosólicas/antagonistas & inhibidores , Fosfolipasas A2 Citosólicas/metabolismo , Aves de Corral , Prostaglandina-Endoperóxido Sintasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/metabolismo , Solventes/química , Factores de Tiempo
5.
Toxicol Mech Methods ; 22(5): 383-96, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22409285

RESUMEN

Lung vascular alterations and pulmonary hypertension associated with oxidative stress have been reported to be involved in idiopathic lung fibrosis (ILF). Therefore, here, we hypothesize that the widely used lung fibrosis inducer, bleomycin, would cause cytoskeletal rearrangement through thiol-redox alterations in the cultured lung vascular endothelial cell (EC) monolayers. We exposed the monolayers of primary bovine pulmonary artery ECs to bleomycin (10 µg) and studied the cytotoxicity, cytoskeletal rearrangements, and the macromolecule (fluorescein isothiocyanate-dextran, 70,000 mol. wt.) paracellular transport in the absence and presence of two thiol-redox protectants, the classic water-soluble N-acetyl-L-cysteine (NAC) and the novel hydrophobic N,N'-bis-2-mercaptoethyl isophthalamide (NBMI). Our results revealed that bleomycin induced cytotoxicity (lactate dehydrogenase leak), morphological alterations (rounding of cells and filipodia formation), and cytoskeletal rearrangement (actin stress fiber formation and alterations of tight junction proteins, ZO-1 and occludin) in a dose-dependent fashion. Furthermore, our study demonstrated the formation of reactive oxygen species, loss of thiols (glutathione, GSH), EC barrier dysfunction (decrease of transendothelial electrical resistance), and enhanced paracellular transport (leak) of macromolecules. The observed bleomycin-induced EC alterations were attenuated by both NAC and NBMI, revealing that the novel hydrophobic thiol-protectant, NBMI, was more effective at µM concentrations as compared to the water-soluble NAC that was effective at mM concentrations in offering protection against the bleomycin-induced EC alterations. Overall, the results of the current study suggested the central role of thiol-redox in vascular EC dysfunction associated with ILF.


Asunto(s)
Acetilcisteína/farmacología , Citoesqueleto de Actina/efectos de los fármacos , Antioxidantes/farmacología , Bleomicina/farmacología , Cisteamina/análogos & derivados , Endotelio Vascular/efectos de los fármacos , Fibrosis Pulmonar Idiopática/prevención & control , Pulmón/efectos de los fármacos , Ácidos Ftálicos/farmacología , Compuestos de Sulfhidrilo/farmacología , Acetilcisteína/química , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Animales , Antioxidantes/química , Bovinos , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cisteamina/química , Cisteamina/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Glutatión/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Pulmón/patología , Microscopía Fluorescente , Estructura Molecular , Oxidación-Reducción , Ácidos Ftálicos/química , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad , Compuestos de Sulfhidrilo/química
6.
Int J Toxicol ; 30(6): 619-38, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21994240

RESUMEN

Here, we investigated thiol-redox-mediated phospholipase D (PLD) signaling as a mechanism of mercury cytotoxicity in mouse aortic endothelial cell (MAEC) in vitro model utilizing the novel lipid-soluble thiol-redox antioxidant and heavy metal chelator, N,N'-bis(2-mercaptoethyl)isophthalamide (NBMI) and the novel PLD-specific inhibitor, 5-fluoro-2-indolyl des-chlorohalopemide (FIPI). Our results demonstrated (i) mercury in the form of mercury(II) chloride, methylmercury, and thimerosal induced PLD activation in a dose- and time-dependent manner; (ii) NBMI and FIPI completely attenuated mercury- and oxidant-induced PLD activation; (iii) mercury induced upstream phosphorylation of extracellular-regulated kinase 1/2 (ERK1/2) leading to downstream threonine phosphorylation of PLD(1) which was attenuated by NBMI; (iv) mercury caused loss of intracellular glutathione which was restored by NBMI; and (v) NBMI and FIPI attenuated mercury- and oxidant-induced cytotoxicity in MAECs. For the first time, this study demonstrated that redox-dependent and PLD-mediated bioactive lipid signaling was involved in mercury-induced vascular EC cytotoxicity which was protected by NBMI and FIPI.


Asunto(s)
Antioxidantes/farmacología , Quelantes/farmacología , Células Endoteliales/efectos de los fármacos , Mercurio/toxicidad , Fosfolipasa D/antagonistas & inhibidores , Ácidos Ftálicos/farmacología , Animales , Antioxidantes/síntesis química , Aorta/citología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quelantes/síntesis química , Domperidona/análogos & derivados , Domperidona/farmacología , Células Endoteliales/metabolismo , Contaminantes Ambientales/toxicidad , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , L-Lactato Deshidrogenasa/metabolismo , Metabolismo de los Lípidos , Ratones , Oxidación-Reducción , Fosfolipasa D/metabolismo , Ácidos Ftálicos/síntesis química , Transducción de Señal/efectos de los fármacos , Compuestos de Sulfhidrilo/metabolismo
7.
Environ Health Perspect ; 119(3): 312-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20980218

RESUMEN

BACKGROUND: Mechanisms of cardiovascular injuries from exposure to gas and particulate air pollutants are unknown. OBJECTIVE: We sought to determine whether episodic exposure of rats to ozone or diesel exhaust particles (DEP) causes differential cardiovascular impairments that are exacerbated by ozone plus DEP. METHODS AND RESULTS: Male Wistar Kyoto rats (10-12 weeks of age) were exposed to air, ozone (0.4 ppm), DEP (2.1 mg/m(3)), or ozone (0.38 ppm) + DEP (2.2 mg/m(3)) for 5 hr/day, 1 day/week for 16 weeks, or to air, ozone (0.51 or 1.0 ppm), or DEP (1.9 mg/m(3)) for 5 hr/day for 2 days. At the end of each exposure period, we examined pulmonary and cardiovascular biomarkers of injury. In the 16-week study, we observed mild pulmonary pathology in the ozone, DEP, and ozone + DEP exposure groups, a slight decrease in circulating lymphocytes in the ozone and DEP groups, and decreased platelets in the DEP group. After 16 weeks of exposure, mRNA biomarkers of oxidative stress (hemeoxygenase-1), thrombosis (tissue factor, plasminogen activator inhibitor-1, tissue plasminogen activator, and von Willebrand factor), vasoconstriction (endothelin-1, endothelin receptors A and B, endothelial NO synthase) and proteolysis [matrix metalloprotease (MMP)-2, MMP-3, and tissue inhibitor of matrix metalloprotease-2] were increased by DEP and/or ozone in the aorta, but not in the heart. Aortic LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1) mRNA and protein increased after ozone exposure, and LOX-1 protein increased after exposure to ozone + DEP. RAGE (receptor for advanced glycation end products) mRNA increased in the ozone + DEP group. Exposure to ozone or DEP depleted cardiac mitochondrial phospholipid fatty acids (DEP > ozone). The combined effect of ozone and DEP exposure was less pronounced than exposure to either pollutant alone. Exposure to ozone or DEP for 2 days (acute) caused mild changes in the aorta. CONCLUSIONS: In animals exposed to ozone or DEP alone for 16 weeks, we observed elevated biomarkers of vascular impairments in the aorta, with the loss of phospholipid fatty acids in myocardial mitochondria. We conclude that there is a possible role of oxidized lipids and protein through LOX-1 and/or RAGE signaling.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Sistema Cardiovascular/efectos de los fármacos , Ozono/toxicidad , Material Particulado/toxicidad , Emisiones de Vehículos/toxicidad , Animales , Biomarcadores/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas WKY , Trombosis/inducido químicamente , Trombosis/metabolismo , Vasoconstricción/efectos de los fármacos
8.
Int J Toxicol ; 30(1): 69-90, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21131602

RESUMEN

The mechanisms of lung microvascular complications and pulmonary hypertension known to be associated with idiopathic pulmonary fibrosis (IPF), a debilitating lung disease, are not known. Therefore, we investigated whether bleomycin, the widely used experimental IPF inducer, would be capable of activating phospholipase D (PLD) and generating the bioactive lipid signal-mediator phosphatidic acid (PA) in our established bovine lung microvascular endothelial cell (BLMVEC) model. Our results revealed that bleomycin induced the activation of PLD and generation of PA in a dose-dependent (5, 10, and 100 µg) and time-dependent (2-12 hours) fashion that were significantly attenuated by the PLD-specific inhibitor, 5-fluoro-2-indolyl des-chlorohalopemide (FIPI). PLD activation and PA generation induced by bleomycin (5 µg) were significantly attenuated by the thiol protectant (N-acetyl-L-cysteine), antioxidants, and iron chelators suggesting the role of reactive oxygen species (ROS), lipid peroxidation, and iron therein. Furthermore, our study demonstrated the formation of ROS and loss of glutathione (GSH) in cells following bleomycin treatment, confirming oxidative stress as a key player in the bleomycin-induced PLD activation and PA generation in ECs. More noticeably, PLD activation and PA generation were observed to happen upstream of bleomycin-induced cytotoxicity in BLMVECs, which was protected by FIPI. This was also supported by our current findings that exposure of cells to exogenous PA led to internalization of PA and cytotoxicity in BLMVECs. For the first time, this study revealed novel mechanism of the bleomycin-induced redox-sensitive activation of PLD that led to the generation of PA, which was capable of inducing lung EC cytotoxicity, thus suggesting possible bioactive lipid-signaling mechanism/mechanisms of microvascular disorders encountered in IPF.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Endotelio Vascular/efectos de los fármacos , Fibrosis Pulmonar Idiopática/inducido químicamente , Ácidos Fosfatidicos/metabolismo , Fosfolipasa D/metabolismo , Supervivencia Celular/efectos de los fármacos , Domperidona/análogos & derivados , Domperidona/farmacología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Activación Enzimática/efectos de los fármacos , Fibrosis Pulmonar Idiopática/enzimología , Fibrosis Pulmonar Idiopática/patología , Indoles/farmacología , Pulmón/irrigación sanguínea , Microvasos/citología , Microvasos/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosfolipasa D/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...