Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Diabetes ; 65(8): 2380-91, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27207527

RESUMEN

In the current study, we used muscle-specific TRIB3 overexpressing (MOE) and knockout (MKO) mice to determine whether TRIB3 mediates glucose-induced insulin resistance in diabetes and whether alterations in TRIB3 expression as a function of nutrient availability have a regulatory role in metabolism. In streptozotocin diabetic mice, TRIB3 MOE exacerbated, whereas MKO prevented, glucose-induced insulin resistance and impaired glucose oxidation and defects in insulin signal transduction compared with wild-type (WT) mice, indicating that glucose-induced insulin resistance was dependent on TRIB3. In response to a high-fat diet, TRIB3 MOE mice exhibited greater weight gain and worse insulin resistance in vivo compared with WT mice, coupled with decreased AKT phosphorylation, increased inflammation and oxidative stress, and upregulation of lipid metabolic genes coupled with downregulation of glucose metabolic genes in skeletal muscle. These effects were prevented in the TRIB3 MKO mice relative to WT mice. In conclusion, TRIB3 has a pathophysiological role in diabetes and a physiological role in metabolism. Glucose-induced insulin resistance and insulin resistance due to diet-induced obesity both depend on muscle TRIB3. Under physiological conditions, muscle TRIB3 also influences energy expenditure and substrate metabolism, indicating that the decrease and increase in muscle TRIB3 under fasting and nutrient excess, respectively, are critical for metabolic homeostasis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Glucosa/toxicidad , Músculo Esquelético/metabolismo , Animales , Composición Corporal/genética , Composición Corporal/fisiología , Calorimetría Indirecta , Proteínas de Ciclo Celular/genética , Colesterol/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/metabolismo , Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología
2.
Atherosclerosis ; 228(1): 124-35, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23510830

RESUMEN

OBJECTIVE: Adiponectin is one of several important, metabolically active cytokines secreted from adipose tissue. Epidemiologic studies have associated low circulating levels of this adipokine with multiple metabolic disorders including obesity, insulin resistance, type II diabetes, and cardiovascular disease. To investigate how enhanced adiponectin-mediated changes in metabolism in vivo, we generated transgenic mice which specifically overexpress the gene coding for adiponectin receptor 1 (AdipoR1) in mouse macrophages using the human scavenger receptor A-I gene (SR-AI) enhancer/promoter. We found that macrophage-specific AdipoR1 transgenic mice (AdR1-TG) presented reduced whole body weight, fat accumulation and liver steatosis when these transgenic mice were fed with a high fat diet. Moreover, these macrophage AdR1-TG mice exhibited enhanced whole-body glucose tolerance and insulin sensitivity with reduced proinflammatory cytokines, MCP-1 and TNF-α, both in the serum and in the insulin target metabolic tissues. Additional studies demonstrated that these macrophage AdR1-TG animals exhibited reduced macrophage foam cell formation in the arterial wall when these transgenic mice were crossed with a low-density lipoprotein receptor (Ldlr) deficient mouse model. CONCLUSIONS: These results suggest that AdipoR1 overexpressed in macrophages can physiologically modulate metabolic activities in vivo by enhancing adiponectin actions in distal metabolically active tissues. The AdipoR1 modified macrophages provide unique interactions with the residented tissues/cells, suggesting a novel role of macrophage adiponectin receptor in improving metabolic disorders in vivo.


Asunto(s)
Macrófagos Peritoneales/fisiología , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo , Adiponectina/sangre , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Proteínas Portadoras/genética , Células Cultivadas , Colesterol/sangre , Modelos Animales de Enfermedad , Hígado Graso/genética , Hígado Graso/metabolismo , Células Espumosas/citología , Células Espumosas/fisiología , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Insulina/sangre , Hígado/metabolismo , Macrófagos Peritoneales/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Empalme Serina-Arginina , Triglicéridos/sangre
3.
Biochem Biophys Res Commun ; 433(4): 567-72, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23523784

RESUMEN

BACKGROUND: Overexpression of adiponectin receptor 1 in macrophages can physiologically modulate metabolic activities in vivo by enhancing adiponectin actions in distal metabolically active tissues. To investigate the effects of enhanced adiponectin actions in TALLYHO (TH) diabetic mouse model, we crossed the adiponectin receptor 1 macrophage-specific transgenic mice (AdR1-TG) with the TALLYHO diabetic mice (TH) to examine the changes of lipid accumulation and insulin sensitivity in these mice. METHODS: AdR1-TG/TH and the control WT/TH mice were fed either normal diet or high fat diet for 28weeks. Whole body weights of these mice were measured and mouse sera were analyzed for the levels of cholesterol, triglyceride, and free fatty acids. Glucose tolerance testing (GTT) and insulin tolerance testing (ITT) in these mice were performed to investigate systemic insulin sensitivity in vivo. Molecular markers for insulin signaling pathway in mouse skeletal muscle tissues, IRS-1 and AKT, were examined. Mouse serum insulin levels were measured and Sirt1 gene expression in mouse pancreatic tissues was also quantified related to the insulin secretion. The Caspase 3 protein levels were analyzed by Western blot methods. RESULTS: Compared to the control WT/TH mice, AdR1-TG/TH mice showed significantly lower body weights under either normal diet or high fat diet and the mouse serum levels of cholesterol, triglyceride and free fatty acids were significantly decreased in the transgenic crossed mice when compared to those from the control mice. Improved GTT and ITT tests indicating increased systemic insulin sensitivity in the transgenic crossed mice demonstrated the enhanced adiponectin actions on the systemic metabolism in vivo. The increases of insulin secretion and its related gene expression were also detected in the transgenic crossed mice. In contrast, the control mice showed hypertrophy pancreases companying with high apoptosis gene expression. These results suggest that enhanced adiponectin actions by overexpressing adiponectin receptor 1 in macrophages can provide unique interactions with the metabolic tissues/cells, improving lipid accumulation and insulin sensitivity in TALLYHO diabetic mice.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Resistencia a la Insulina , Receptores de Adiponectina/metabolismo , Triglicéridos/metabolismo , Animales , Apoptosis , Biomarcadores/sangre , Biomarcadores/metabolismo , Western Blotting , Peso Corporal , Caspasa 3/análisis , Colesterol/sangre , Cruzamientos Genéticos , Diabetes Mellitus Experimental/dietoterapia , Diabetes Mellitus Experimental/patología , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos no Esterificados/sangre , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Hipertrofia/genética , Hipertrofia/metabolismo , Hipertrofia/patología , Insulina/sangre , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Secreción de Insulina , Masculino , Ratones , Ratones Transgénicos/genética , Ratones Transgénicos/metabolismo , Músculo Esquelético/metabolismo , Páncreas/metabolismo , Páncreas/patología , Receptores de Adiponectina/genética , Transducción de Señal , Sirtuina 1/genética , Sirtuina 1/metabolismo , Triglicéridos/sangre
4.
Atherosclerosis ; 221(1): 66-75, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22227293

RESUMEN

OBJECTIVE: Adiponectin is an adipokine that exerts anti-inflammatory and anti-atherogenic effects during macrophage transformation into foam cells. To further understand the signaling pathways of adiponectin involved in macrophage foam cell transformation, we investigated the roles of two adiponectin receptors (AdipoR1 and AdipoR2) and their downstream adaptor protein, phosphotyrosine interaction, PH domain and leucine zipper containing 1 (APPL1) in mediating adiponectin action on foam cell transformation. METHODS AND RESULTS: Transfections were performed to overexpress or knockdown AdipoR1 or AdipoR2 genes in human THP-1 monocytes. Lentiviral-shRNAs were also used to knockdown APPL1 gene in these cells. Foam cell transformation was induced via exposure to oxidized low-density lipoprotein (oxLDL). Our results showed that both AdipoR1 and AdipoR2 were critical for transducing the adiponectin signal that suppresses lipid accumulation and inhibits transformation from macrophage to foam cell. However, AdipoR1 and AdipoR2 were found to have differential effects in diminishing proinflammatory responses. While AdipoR1 was required by adiponectin to suppress tumor necrosis factor alpha (TNFα) and monocyte chemotactic protein 1 (MCP-1) gene expression, AdipoR2 served as the dominant receptor for adiponectin suppression of scavenger receptor A type 1 (SR-AI) and upregulation of interleukin-1 receptor antagonist (IL-1Ra). Knockdown of APPL1 significantly abrogated the ability of adiponectin to inhibit lipid accumulation, SR-AI and nuclear factor-κB (NF-κB) gene expression, and Akt phosphorylation in macrophage foam cells. CONCLUSIONS: In current studies, we have demonstrated that adiponectin's abilty to suppress macrophage lipid accumulation and foam cell formation is mediated through AdipoR1 and AdipoR2 and the APPL1 docking protein. However, AdipoR1 and AdipoR2 exhibited a differential ability to regulate inflammatory cytokines and SR-A1. These novel data support the idea that the adiponectin-AdipoR1/2-APPL1 axis may serve as a potential therapeutic target for preventing macrophage foam cell formation and atherosclerosis.


Asunto(s)
Adiponectina/metabolismo , Aterosclerosis/metabolismo , Citocinas/metabolismo , Células Espumosas/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Receptores de Adiponectina/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aterosclerosis/genética , Aterosclerosis/inmunología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Colesterol/metabolismo , Citocinas/genética , Células Espumosas/inmunología , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/inmunología , Lipoproteínas LDL/metabolismo , FN-kappa B/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Receptores de Adiponectina/genética , Transfección
5.
Am J Physiol Endocrinol Metab ; 301(1): E180-6, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21505149

RESUMEN

Epidemiological studies have associated low circulating levels of the adipokine adiponectin with multiple metabolic disorders, including metabolic syndrome, obesity, insulin resistance, type II diabetes, and cardiovascular disease. Recently, we reported that adiponectin selectively overexpressed in mouse macrophages can improve insulin sensitivity and protect against inflammation and atherosclerosis. To further investigate the role of adiponectin and macrophages on lipid and lipometabolism in vivo, we engineered the expression of adiponectin in mouse macrophages (Ad-TG mice) and examined effects on plasma lipoproteins and on the expression levels of genes involved in lipoprotein metabolism in tissues. Compared with the wild-type (WT) mice, Ad-TG mice exhibited significantly lower levels of plasma total cholesterol (-21%, P < 0.05) due to significantly decreased LDL (-34%, P < 0.05) and VLDL (-32%, P < 0.05) cholesterol concentrations together with a significant increase in HDL cholesterol (+41%, P < 0.05). Further studies investigating potential mechanisms responsible for the change in lipoprotein cholesterol profile revealed that adiponectin-producing macrophages altered expression of key genes in liver tissue, including apoA1, apoB, apoE, the LDL receptor, (P < 0.05), and ATP-binding cassette G1 (P < 0.01). In addition, Ad-TG mice also exhibited higher total and high-molecular-weight adipnection levels in plasma and increased expression of the anti-inflammatory cytokine IL-10 as well as a decrease in the proinflammatory cytokine IL-6 in adipose tissue. These results indicate that macrophages engineered to produce adiponectin can influence in vivo gene expression in adipose tissue in a manner that reduces inflammation and macrophage infiltration and in liver tissue in a manner that alters the circulating lipoprotein profile, resulting in a decrease in VLDL and LDL and an increase in HDL cholesterol. The data support further study addressing the use of genetically manipulated macrophages as a novel therapeutic approach for treatment of cardiometabolic disease.


Asunto(s)
Metabolismo de los Lípidos/genética , Macrófagos Peritoneales/metabolismo , Adiponectina/sangre , Adiponectina/genética , Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Citocinas/sangre , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Metabolismo de los Lípidos/fisiología , Lípidos/análisis , Lípidos/sangre , Lipoproteínas/análisis , Lipoproteínas/metabolismo , Hígado/metabolismo , Macrófagos Peritoneales/fisiología , Ratones , Ratones Transgénicos , Especificidad de Órganos/genética
6.
Diabetes ; 59(4): 791-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20350970

RESUMEN

OBJECTIVE: Adiponectin is one of several important metabolically active cytokines secreted from adipose tissue. Epidemiologic studies have associated low-circulating levels of this adipokine with multiple metabolic disorders including obesity, insulin resistance, type 2 diabetes, and cardiovascular disease. To investigate adiponectin-mediated changes in metabolism in vivo, we generated transgenic mice that specifically express the gene coding for human adiponectin in mouse macrophages using the human scavenger receptor A-I gene enhancer/promoter. METHODS AND RESULTS: Using this transgenic mouse model, we found that adiponectin expression was associated with reduced whole-animal body and fat-pad weight and an improved lipid accumulation in macrophages when these transgenic mice were fed with a high-fat diet. Moreover, these macrophage Ad-TG mice exhibit enhanced whole-body glucose tolerance and insulin sensitivity with reduced proinflammatory cytokines, MCP-1 and TNF-a (both in the serum and in the metabolic active macrophage), adipose tissue, and skeletal muscle under the high-fat diet condition. Additional studies demonstrated that these macrophage adiponectin transgenic animals exhibit reduced macrophage foam cell formation in the arterial wall when these transgenic mice were crossed with an LDL receptor-deficient mouse model and were fed a high-fat diet. CONCLUSIONS: These results suggest that adiponectin expressed in macrophages can physiologically modulate metabolic activities in vivo by improving metabolism in distal tissues. The use of macrophages as carriers for adiponectin, a molecule with antidiabetes, anti-inflammatory, and antiatherogenic properties, provides a novel and unique strategy for studying the mechanisms of adiponectin-mediated alterations in body metabolism in vivo.


Asunto(s)
Adiponectina/genética , Aterosclerosis/prevención & control , Inflamación/prevención & control , Insulina/fisiología , Macrófagos Peritoneales/fisiología , Animales , Proteínas Portadoras/genética , ADN Complementario/genética , Elementos de Facilitación Genéticos , Vectores Genéticos , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Hormona del Crecimiento/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/metabolismo , Plásmidos , Regiones Promotoras Genéticas , Empalme del ARN , Factores de Empalme Serina-Arginina
7.
Atherosclerosis ; 202(1): 152-61, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18511057

RESUMEN

Adiponectin is one of several, important metabolically active cytokines secreted from adipocytes. Low circulating levels of this adipokine have been associated epidemiologically with obesity, insulin resistance, type II diabetes, and cardiovascular disease. To determine if adiponectin can modulate lipid metabolism in macrophages, we expressed the adiponectin gene in human THP-1 macrophage foam cells using a lentiviral vector expression system and demonstrated that macrophages transduced with the adiponectin gene had decreased lipid accumulation compared with control macrophages transduced with the LacZ gene. Macrophages transduced with the adiponectin gene also exhibited decreased oxidized low-density lipoprotein (oxLDL) uptake and increased HDL-mediated cholesterol efflux. Additional studies suggest two potential mechanisms for the reduced lipid accumulation in these adiponectin-transduced macrophage foam cells. The first mechanism involves the PPARgamma and LXR signaling pathways which up-regulate the expression of ABCA1 and promote lipid efflux from these cells. The second mechanism involves decreased lipid uptake and increased lipid hydrolysis which may result from decreased SR-AI and increased SR-BI and HSL gene activities in the transformed macrophage foam cells. We also demonstrated that the expression of two proatherogenic cytokines, MCP-1 and TNFalpha, were decreased in the adiponectin-transduced macrophage foam cells. These results suggest that adiponectin may modulate multiple pathways of lipid metabolism in macrophages. Our studies provide new insights into potential mechanisms of adiponectin-mediated alterations in lipid metabolism and macrophage foam cell formation which may impact the development of atherosclerosis.


Asunto(s)
Adiponectina/metabolismo , Células Espumosas/citología , Lípidos/química , Macrófagos/citología , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Adiponectina/genética , Aterosclerosis/metabolismo , Transporte Biológico , Línea Celular Tumoral , Colesterol/química , Colesterol/metabolismo , Células Espumosas/metabolismo , Humanos , Lentivirus/genética , Metabolismo de los Lípidos , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
8.
J Biol Chem ; 282(43): 31525-33, 2007 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-17785466

RESUMEN

After observing that expression of two NR4A orphan nuclear receptors, NR4A3 and NR4A1, was altered by insulin in cDNA microarray analyses of human skeletal muscle, we studied whether these receptors could modulate insulin sensitivity. We found that both NR4A3 and NR4A1 were induced by insulin and by thiazolidinedione drugs (pioglitazone and troglitazone) in 3T3-L1 adipocytes. Furthermore, gene expression of NR4A3 and NR4A1 was reduced in skeletal muscles and adipose tissues from multiple rodent models of insulin resistance. To determine whether NR4A3 could modulate insulin sensitivity, 3T3-L1 adipocytes were stably transduced with NR4A3 or LacZ (control) lentiviral vectors. Compared with LacZ expressing cells, hyperexpression of NR4A3 increased the ability of insulin to augment glucose transport activity, and the mechanism involved increased recruitment of GLUT4 glucose transporters to the plasma membrane. NR4A3 hyperexpression also led to an increase in insulin-mediated tyrosine phosphorylation of insulin receptor substrate-1 as well as Akt phosphorylation. Suppression of NR4A3 using lentiviral short hairpin RNA constructs reduced the ability of insulin to stimulate glucose transport and phosphorylate Insulin receptor substrate-1 and Akt. Thus, NR4A3 and NR4A1 are attractive novel therapeutic targets for potential amelioration of insulin resistance, and treatment and prevention of type 2 diabetes and the metabolic syndrome.


Asunto(s)
Adipocitos/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Insulina/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Factores de Transcripción/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Diferenciación Celular , Vectores Genéticos , Transportador de Glucosa de Tipo 1/análisis , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 4/análisis , Transportador de Glucosa de Tipo 4/metabolismo , Operón Lac/genética , Lentivirus/genética , Ratones , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Factores de Tiempo , Transducción Genética , Transfección
9.
Nutr Metab (Lond) ; 3: 28, 2006 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-16854242

RESUMEN

Resistin is secreted from adipocytes, and high circulating levels have been associated with obesity and insulin resistance. To investigate whether resistin could exert autocrine effects in adipocytes, we expressed resistin gene in 3T3-L1 fibroblasts using a lentiviral vector, and selected several stably-transduced cell lines under blasticidin selection. We observed that 3T3-L1 adipocytes expressing resistin have a decreased gene expression for related transcriptional factors (CCAAT/enhancer binding protein alpha(C/EBPalpha), peroxisome proliferator-activated receptor gamma (PPARgamma), and adipocyte lipid binding protein (ALBP/aP2) which is one of target genes for the PPARgamma during adipocyte differentiation,. Overexpression of resistin increased the levels of three proinflammatory cytokines, tumor necrosis factor alpha (TNFalpha), interleukin 6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1), which play important roles for insulin resistance, glucose and lipid metabolisms during adipogenesis. Furthermore, overexpressing resistin in adipocytes inhibits glucose transport 4 (GLUT4) activity and its gene expression, reducing insulin's ability for glucose uptake by 30%. In conclusion, resistin overexpression in stably transduced 3T3-L1 cells resulted in: 1) Attenuation of programmed gene expression responsible for adipogenesis; 2) Increase in expression of proinflammatory cytokines; 3) Decrease in insulin responsiveness of the glucose transport system. These data suggest a new role for resistin as an autocrine/paracrine factor affecting inflammation and insulin sensitivity in adipose tissue.

10.
Atherosclerosis ; 188(1): 102-11, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16313911

RESUMEN

The critical initiating event in atherogenesis involves the invasion of monocytes through the endothelial wall of arteries, and their transformation from macrophages into foam cells. Human THP-1 monocytic cells can be induced to differentiate into macrophages by phorbol myristate acetate (PMA) treatment, and can then be converted into foam cells by exposure to oxidized low-density lipoprotein (oxLDL). We previously reported that adipocyte lipid binding protein (ALBP/aP2) is a gene that is highly up-regulated in foam cells in response to oxLDL. Here, we showed that overexpression of the ALBP gene using a lentiviral construct in macrophage foam cells enhanced the accumulations of cholesterol and triglyceride, probably due to an increased expression of the scavenger receptor type AI (SR-AI), which plays an important role in cell lipid metabolism. Moreover, we determined that the expression of acyl-coenzyme A: cholesterol-acyltransferase 1 (ACAT1) gene was up-regulated by the overexpression of ALBP gene, and on the other hand, the ATP-binding cassette A1 (ABCA1) gene and hormone sensitive lipase (HSL) gene, which mediate separately cholesterol efflux and cholesterol ester hydrolysis in the macrophage cells, were down-regulated by the overexpression of ALBP gene in these cells. Finally, our data indicated that oxLDL regulates expression of ALBP related to two peroxisome proliferator-responsive elements (PPREs) which are located in ALBP promoter region. These results have determined that ALBP gene expression accelerates cholesterol and triglyceride accumulation in macrophage foam cells and affects some key gene expression for lipid metabolism, suggesting some pivotal roles of ALBP in lipid metabolism for macrophage foam cell formation.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/fisiología , Células Espumosas/metabolismo , Regulación de la Expresión Génica , Metabolismo de los Lípidos/genética , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Diferenciación Celular , Línea Celular , Células Cultivadas , Colesterol/análisis , Colesterol/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Células Espumosas/química , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Lipoproteínas LDL/farmacología , Macrófagos/química , Macrófagos/metabolismo , Proliferadores de Peroxisomas/metabolismo , Elementos de Respuesta , Receptores Depuradores de Clase A/genética , Esterol Esterasa/genética , Esterol O-Aciltransferasa/genética , Triglicéridos/análisis , Triglicéridos/metabolismo
11.
Diabetes ; 55(1): 249-59, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16380500

RESUMEN

Adiponectin circulates in human plasma mainly as a 180-kDa low molecular weight (LMW) hexamer and a high molecular weight (HMW) multimer of approximately 360 kDa. We comprehensively examined the relationships between circulating levels of total adiponectin, adiponectin multimers, and the relative distribution (i.e., ratio) of multimeric forms with key features of the metabolic syndrome. Total adiponectin (r = 0.45), HMW (r = 0.47), LMW (r = 0.31), and HMW-to-total adiponectin ratio (r = 0.29) were significantly correlated with insulin-stimulated glucose disposal rate. Similarly, total (r = -0.30), HMW (r = -0.38), and HMW-to-total adiponectin ratio (r = -0.34) were correlated with central fat distribution but not with total fat mass or BMI. Regarding energy metabolism, although there were no effects on resting metabolic rate, total (r = 0.41) and HMW (r = 0.44) were associated with increasing rates of fat oxidation. HMW-to-total adiponectin ratio increased as a function of total adiponectin, and it was HMW quantity (not total or HMW-to-total adiponectin ratio or LMW) that was primarily responsible for all of these relationships. Impact on nuclear magnetic resonance lipoprotein subclasses was assessed. HMW and total adiponectin were correlated with decreases in large VLDL (r = -0.44 and -0.41); decreases in small LDL (r = -0.41 and -0.36) and increases in large LDL (r = 0.36 and 0.30) particle concentrations accompanied by increased LDL particle size (r = 0.47 and 0.39); and increases in large HDL (r = 0.45 and 0.37) and HDL particle size (r = 0.53 and 0.47). Most of these correlations persisted after adjustment for metabolic covariables. In conclusion, first, serum adiponectin is associated with increased insulin sensitivity, reduced abdominal fat, and high basal lipid oxidation; however, it is HMW quantity, not total or HMW-to-total adiponectin ratio, that is primarily responsible for these relationships. Second, reduced quantities of HMW independently recapitulate the lipoprotein subclass profile associated with insulin resistance after correcting for glucose disposal rate and BMI. Finally, HMW adiponectin is an important factor in explaining the metabolic syndrome.


Asunto(s)
Adiponectina/química , Adiponectina/metabolismo , Síndrome Metabólico/metabolismo , Tejido Adiposo/metabolismo , Adulto , Presión Sanguínea , Distribución de la Grasa Corporal , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Peso Molecular , Estructura Cuaternaria de Proteína
12.
J Lipid Res ; 46(7): 1369-79, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15834118

RESUMEN

Adiponectin is secreted from adipocytes, and low circulating levels have been epidemiologically associated with obesity, insulin resistance, type 2 diabetes, and cardiovascular disease. To investigate whether adiponectin could exert autocrine effects in adipocytes, we expressed the adiponectin gene in 3T3-L1 fibroblasts. We observed that 3T3-L1 fibroblasts expressing adiponectin have a fast growth phase and reach confluence more rapidly compared with control cells or LacZ-transduced cells. Furthermore, cells with overexpressed adiponectin were observed to differentiate into adipocytes more rapidly, and during adipogenesis, they exhibited more prolonged and robust gene expression for related transcriptional factors, CCAAT/enhancer binding protein alpha (C/EBP2), peroxisome proliferator-activated receptor gamma (PPARgamma), and adipocyte determination and differentiation factor 1/sterol-regulatory element binding protein 1c (ADD1/SREBP1c) and earlier suppression of PPARgamma coactivator-1alpha (PGC-1alpha). In fully differentiated adipocytes, adiponectin-overexpressing cells accumulated more and larger lipid droplets compared with control cells. Also, adiponectin increased insulin's ability to maximally stimulate glucose uptake by 78% through increased glucose transporter 4 (GLUT4) gene expression and increased GLUT4 recruitment to the plasma membrane. These data suggest a new role for adiponectin as an autocrine factor in adipose tissues: promoting cell proliferation and differentiation from preadipocytes into adipocytes, augmenting programmed gene expression responsible for adipogenesis, and increasing lipid content and insulin responsiveness of the glucose transport system in adipocytes.


Asunto(s)
Adipocitos/citología , Resistencia a la Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Metabolismo de los Lípidos , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adiponectina , Animales , Proteína alfa Potenciadora de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Portadoras/biosíntesis , Diferenciación Celular/efectos de los fármacos , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión a Ácidos Grasos , Transportador de Glucosa de Tipo 4 , Insulina/farmacología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Ratones , Proteínas de Transporte de Monosacáridos/biosíntesis , Proteínas Musculares/biosíntesis , PPAR gamma/biosíntesis , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Transactivadores/biosíntesis , Factores de Transcripción/biosíntesis
13.
Atherosclerosis ; 165(2): 259-69, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12417276

RESUMEN

The critical initiating event in atherogenesis involves the invasion of monocytes through the endothelial wall of arteries, and their transformation from macrophages into foam cells. Human THP-1 monocytic cells can be induced to differentiate into macrophages by phorbol myristate acetate (PMA) treatment, and can then be converted into foam cells by exposure to oxidized low-density lipoprotein (oxLDL). To define genes that are specifically expressed during the transformation of macrophages into foam cells, we have performed a subtractive library screening utilizing mRNA isolated from THP-1 macrophages and foam cells. From this analysis, we have identified adipocyte lipid binding protein (ALBP/aP2) as a gene that is highly upregulated in foam cells in response to oxLDL. Furthermore, overexpression the ALBP gene using an adenovirus construct enhanced the accumulation of cholesterol ester in macrophage foam cells, probably due to an increase in transcription since oxLDL enhanced ALBP promoter activity in experiments using a promoter-luciferase reporter gene construct. The induction of ALBP by oxLDL probably involved activation of peroxisome proliferator-activated receptor gamma (PPARgamma) transcription factors, since four different endogenous PPARgamma ligands, including 9-hydroxyoctadecadienoic acid (9-HODE) and 13-hydroxyoctadecadienoic acid (13-HODE), two oxidized lipid components of oxLDL, as well as 15-deoxy-delta12,14 prostaglandin J2 (15d-PGJ2) and retinoic acid (RA), all induced ALBP expression in macrophage/foam cells. Finally, ALBP was found to be highly expressed in vivo in macrophage/foam cells of human atherosclerotic plaques. These observations suggest that oxLDL-mediated increase in ALBP gene expression accelerate cholesterol ester accumulation, and that this is an important component of the genetic program regulating conversion of macrophages to foam cells. The observation that ALBP is readily detected in foam cells in active atherosclerotic lesions implicates a role for ALBP in human vascular disease. The induction of ALPB expression by oxLDL likely involves activation of PPARgamma by components of oxLDL (9-HODE and 13-HODE) that also function as PPARgamma ligands. Our results add to the concern that the clinical use of insulin-sensitizing PPARgamma agonists (i.e. thiazolidinediones) to treat Type 2 Diabetes could exacerbate atherosclerosis, and highlight the need for clinical trials that address this issue.


Asunto(s)
Arteriosclerosis/genética , Proteínas Portadoras/genética , Endotelio Vascular/metabolismo , Células Espumosas/metabolismo , Lipoproteínas LDL/genética , Proteínas de Neoplasias , Proteínas Supresoras de Tumor , Arteriosclerosis/metabolismo , Northern Blotting , Proteínas Portadoras/metabolismo , Células Cultivadas , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos , Biblioteca de Genes , Humanos , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo
14.
Atherosclerosis ; 160(1): 11-20, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11755918

RESUMEN

Interleukin-8 (IL-8) is one of cytokines detected at sites of inflammation and in macrophage-foam cells of atherosclerotic lesions. The expression of IL-8 gene can be induced in cholesterol loaded THP-1 macrophages by oxidized low density lipoprotein. We report for the first time that the expression of human IL-8 gene in THP-1 macrophages is upregulated in a time- and concentration-dependent manner by prostaglandin D2 metabolite 15-deoxy-delta12, 14 prostaglandin J2 (15d-PGJ2), which is a natural ligand for activation of peroxisome proliferator-activated receptor-gamma transcription factor. Studies to identify the signal transduction pathways involved showed that IL-8 upregulation-mediated by 15d-PGJ2 was markedly inhibited when the THP-1 macrophages were incubated with a highly selective and cell-permeable inhibitor of the mitogen-activated protein kinase (MAPK) signaling pathway, 2'-amino-3'-methoxyflavone (PD98059). This inhibition was concentration-dependent, suggesting that 15d-PGJ2 regulates the expression of IL-8 gene in THP-1 macrophages through a MAPK signaling pathway. In contrast, THP-1 macrophages when treated with pyrrolidine dithiocarbamate, an anti-oxidant and the selective inhibitor for nuclear factor kappaB, showed an enhanced 15d-PGJ2-mediated upregulation of IL-8 gene expression. The data presented in this report may contribute to unravel some of the mechanisms behind the inflammatory component of atherosclerosis.


Asunto(s)
Factores Inmunológicos/farmacología , Interleucina-8/biosíntesis , Interleucina-8/genética , Macrófagos/metabolismo , Prostaglandina D2/análogos & derivados , Prostaglandina D2/metabolismo , Prostaglandina D2/farmacología , Regulación hacia Arriba/genética , LDL-Colesterol/efectos de los fármacos , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , FN-kappa B/biosíntesis , FN-kappa B/efectos de los fármacos , FN-kappa B/genética , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Tretinoina/farmacología , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...