Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 32(12): 2929-37, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23042820

RESUMEN

OBJECTIVE: Macrophage endothelial lipase (EL) is associated with increased atherosclerosis and inflammation. Because of their anti-inflammatory properties we hypothesized that n-3 fatty acids, in contrast to saturated fatty acids, would lower macrophages and arterial EL and inflammatory markers. METHODS AND RESULTS: Murine J774 and peritoneal macrophages were incubated with eicosapentaenoic acid or palmitic acid in the presence or absence of lipopolysaccaride (LPS). LPS increased EL mRNA and protein. Palmitic acid alone or with LPS dose-dependently increased EL mRNA and protein. In contrast, eicosapentaenoic acid dose-dependently abrogated effects of LPS or palmitic acid on increasing EL expression. EL expression closely linked to peroxisome proliferator activated receptor (PPAR)γ expression. Eicosapentaenoic acid blocked rosiglitazone (a PPARγ agonist)-mediated EL activation and GW9662 (a PPARγ antagonist)-blocked palmitic acid-mediated EL stimulation. Eicosapentaenoic acid alone or with LPS blunted LPS-mediated stimulation of macrophage proinflammatory interleukin-6, interleukin-12p40, and toll-like receptor-4 mRNA and increased anti-inflammatory interleukin-10 and mannose receptor mRNA. In vivo studies in low density lipoprotein receptor knockout mice showed that high saturated fat rich diets, but not n-3 diets, increased arterial EL, PPARγ, and proinflammatory cytokine mRNA. CONCLUSIONS: n-3 fatty acids, in contrast to saturated fatty acids, decrease EL in parallel with modulating pro- and anti-inflammatory markers, and these effects on EL link to PPARγ.


Asunto(s)
Aorta/metabolismo , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos/farmacología , Interleucina-6/metabolismo , Lipasa/metabolismo , Macrófagos Peritoneales/metabolismo , PPAR gamma/metabolismo , Animales , Aorta/efectos de los fármacos , Biomarcadores/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Ácido Eicosapentaenoico/farmacología , Técnicas In Vitro , Subunidad p40 de la Interleucina-12/metabolismo , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Ácido Palmítico/farmacología , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores de LDL/metabolismo , Receptor Toll-Like 4/metabolismo
2.
J Clin Invest ; 120(10): 3443-54, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20852389

RESUMEN

Excess lipid accumulation in the heart is associated with decreased cardiac function in humans and in animal models. The reasons are unclear, but this is generally believed to result from either toxic effects of intracellular lipids or excessive fatty acid oxidation (FAO). PPARγ expression is increased in the hearts of humans with metabolic syndrome, and use of PPARγ agonists is associated with heart failure. Here, mice with dilated cardiomyopathy due to cardiomyocyte PPARγ overexpression were crossed with PPARα-deficient mice. Surprisingly, this cross led to enhanced expression of several PPAR-regulated genes that mediate fatty acid (FA) uptake/oxidation and triacylglycerol (TAG) synthesis. Although FA oxidation and TAG droplet size were increased, heart function was preserved and survival improved. There was no marked decrease in cardiac levels of triglyceride or the potentially toxic lipids diacylglycerol (DAG) and ceramide. However, long-chain FA coenzyme A (LCCoA) levels were increased, and acylcarnitine content was decreased. Activation of PKCα and PKCδ, apoptosis, ROS levels, and evidence of endoplasmic reticulum stress were also reduced. Thus, partitioning of lipid to storage and oxidation can reverse cardiolipotoxicity despite increased DAG and ceramide levels, suggesting a role for other toxic intermediates such as acylcarnitines in the toxic effects of lipid accumulation in the heart.


Asunto(s)
Ácidos Grasos/metabolismo , Lípidos/toxicidad , Miocardio/metabolismo , PPAR alfa/fisiología , PPAR gamma/fisiología , Animales , Apoptosis , Ácidos Grasos no Esterificados/sangre , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Miocitos Cardíacos/ultraestructura , Oxidación-Reducción , PPAR alfa/deficiencia , Especies Reactivas de Oxígeno/metabolismo , Triglicéridos/biosíntesis
3.
PLoS One ; 5(6): e11004, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20543983

RESUMEN

The inflammatory milieu in the respiratory tract in cystic fibrosis (CF) has been linked to the defective expression of the cystic transmembrane regulator (CFTR) in epithelial cells. Alveolar macrophages (AM), important contibutors to inflammatory responses in the lung, also express CFTR. The present study analyzes the phenotype of human AM with silenced CFTR. Expression of CFTR mRNA and the immature form of the CFTR protein decreased 100-fold and 5.2-fold, respectively, in AM transfected with a CFTR specific siRNA (CFTR-siRNA) compared to controls. Reduction of CFTR expression in AM resulted in increased secretion of IL-8, increased phosphorylation of NF-kappaB, a positive regulator of IL-8 expression, and decreased expression of IkappaB-alpha, the inhibitory protein of NF-kappaB activation. AM with silenced CFTR expression also showed increased apoptosis. We hypothesized that caveolin-1 (Cav1), a membrane protein that is co-localized with CFTR in lipid rafts and that is related to inflammation and apoptosis in macrophages, may be affected by decreased CFTR expression. Messenger RNA and protein levels of Cav1 were increased in AM with silenced CFTR. Expression and transcriptional activity of sterol regulatory element binding protein (SREBP), a negative transcriptional regulator of Cav1, was decreased in AM with silenced CFTR, but total and free cholesterol mass did not change. These findings indicate that silencing of CFTR in human AM results in an inflammatory phenotype and apoptosis, which is associated to SREBP-mediated regulation of Cav1.


Asunto(s)
Caveolina 1/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Silenciador del Gen , Macrófagos Alveolares/metabolismo , ARN Mensajero/genética , Adulto , Anciano , Apoptosis , Células Cultivadas , Femenino , Humanos , Macrófagos Alveolares/citología , Masculino , Persona de Mediana Edad , Fenotipo
4.
J Lipid Res ; 50(6): 1101-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19144995

RESUMEN

Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR) that affect protein structure and channel function. CFTR, localized in the apical membrane within cholesterol and sphingomyelin rich regions, is an ABC transporter that functions as a chloride channel. Here, we report that expression of defective CFTR (DeltaF508CFTR or decreased CFTR) in human lung epithelial cell lines increases sphingolipid synthesis and mass of sphinganine, sphingosine, four long-chain saturated ceramide species, C16 dihydroceramide, C22, C24, C26-ceramide, and sphingomyelin, and decreases mass of C18 and unsaturated C18:1 ceramide species. Decreased expression of CFTR is associated with increased expression of long-chain base subunit 1 of serine-palmitoyl CoA, the rate-limiting enzyme of de novo sphingolipid synthesis and increased sphingolipid synthesis. Overexpression of DeltaF508CFTR in bronchoalveolar cells that do not express CFTR increases sphingolipid synthesis and mass, whereas overexpression of wild-type CFTR, but not of an unrelated ABC transporter, ABCA7, decreases sphingolipid synthesis and mass. The data are consistent with a model in which CFTR functions within a feedback system that affects sphingolipid synthesis and in which increased sphingolipid synthesis could reflect a physiological response to sequestration of sphingolipids or altered membrane structure.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Esfingolípidos/metabolismo , Línea Celular , Membrana Celular/metabolismo , Ceramidas/metabolismo , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Humanos , Metabolismo de los Lípidos , Mutación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina C-Palmitoiltransferasa/metabolismo , Transducción de Señal , Esfingolípidos/biosíntesis , Transducción Genética
5.
Diabetes ; 57(6): 1674-82, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18346984

RESUMEN

OBJECTIVE: Patients with diabetes often have dyslipidemia and increased postprandial lipidmia. Induction of diabetes in LDL receptor (Ldlr(-/-)) knockout mice also leads to marked dyslipidemia. The reasons for this are unclear. RESEARCH DESIGN AND METHODS: We placed Ldlr(-/-) and heterozygous LDL receptor knockout (Ldlr(+/-)) mice on a high-cholesterol (0.15%) diet, induced diabetes with streptozotocin (STZ), and assessed reasons for differences in plasma cholesterol. RESULTS: STZ-induced diabetic Ldlr(-/-) mice had plasma cholesterol levels more than double those of nondiabetic controls. Fast-performance liquid chromatography and ultracentrifugation showed an increase in both VLDL and LDL. Plasma VLDL became more cholesterol enriched, and both VLDL and LDL had a greater content of apolipoprotein (apo)E. In LDL the ratio of apoB48 to apoB100 was increased. ApoB production, assessed using [(35)S]methionine labeling in Triton WR1339-treated mice, was not increased in fasting STZ-induced diabetic mice. Similarly, postprandial lipoprotein production was not increased. Reduction of cholesterol in the diet to normalize the amount of cholesterol intake by the control and STZ-induced diabetic animals reduced plasma cholesterol levels in STZ-induced diabetic mice, but plasma cholesterol was still markedly elevated compared with nondiabetic controls. LDL from STZ-induced diabetic mice was cleared from the plasma and trapped more rapidly by livers of control mice. STZ treatment reduced liver expression of the proteoglycan sulfation enzyme, heparan sulfate N-deacetylase/N-sulfotrasferase-1, an effect that was reproduced in cultured hepatocytyes by a high glucose-containing medium. CONCLUSIONS: STZ-induced diabetic, cholesterol-fed mice developed hyperlipidemia due to a non-LDL receptor defect in clearance of circulating apoB-containing lipoproteins.


Asunto(s)
Colesterol/sangre , Diabetes Mellitus Experimental/sangre , Lípidos/sangre , Lipoproteínas/sangre , Receptores de LDL/deficiencia , Triglicéridos/sangre , Animales , Apolipoproteínas B/sangre , Apolipoproteínas E/sangre , Glucemia/metabolismo , Colesterol en la Dieta , Cruzamientos Genéticos , Diabetes Mellitus Experimental/fisiopatología , Dislipidemias/genética , Hígado/fisiopatología , Neoplasias Hepáticas , Neoplasias Hepáticas Experimentales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Tumorales Cultivadas
6.
Lipids ; 42(10): 885-91, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17680293

RESUMEN

Sterol regulatory element binding proteins (SREBPs) are key transcription proteins that bind to sterol regulatory elements (SRE) of genes essential for cellular cholesterol and fatty acid homeostasis. Polyunsaturated fatty acids (PUFA) strongly inhibit SREBP processing at post-transcriptional levels. We questioned if delivering PUFA as part of a triglyceride (TG) molecule would have similar effects and efficiency as free non-esterified PUFA. CHO cells stably transfected with an SRE-promoter linked to the luciferase reporter gene were incubated for 8-24 h with linoleic acid (LA) complexed to BSA (molar ratios 0.5-4:1), VLDL-sized trilinolein emulsions (TL, 25-200 microg/ml), and chylomicron-sized soy oil emulsions in the presence and absence of apoE. Effects of LA and TL on decreasing SRE-luciferase activity were similar and dose and time dependent. Both TL and LA significantly and rapidly (

Asunto(s)
Ácidos Grasos no Esterificados/farmacología , Ácidos Grasos Insaturados/farmacología , Expresión Génica/efectos de los fármacos , Elementos Reguladores de la Transcripción , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Esteroles/metabolismo , Triglicéridos/farmacología , Animales , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Genes Reporteros , Modelos Biológicos , Regiones Promotoras Genéticas , Proteínas de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA