Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell ; 171(2): 372-384.e12, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28942920

RESUMEN

MiRNAs are regulatory molecules that can be packaged into exosomes and secreted from cells. Here, we show that adipose tissue macrophages (ATMs) in obese mice secrete miRNA-containing exosomes (Exos), which cause glucose intolerance and insulin resistance when administered to lean mice. Conversely, ATM Exos obtained from lean mice improve glucose tolerance and insulin sensitivity when administered to obese recipients. miR-155 is one of the miRNAs overexpressed in obese ATM Exos, and earlier studies have shown that PPARγ is a miR-155 target. Our results show that miR-155KO animals are insulin sensitive and glucose tolerant compared to controls. Furthermore, transplantation of WT bone marrow into miR-155KO mice mitigated this phenotype. Taken together, these studies show that ATMs secrete exosomes containing miRNA cargo. These miRNAs can be transferred to insulin target cell types through mechanisms of paracrine or endocrine regulation with robust effects on cellular insulin action, in vivo insulin sensitivity, and overall glucose homeostasis.


Asunto(s)
Tejido Adiposo/citología , Resistencia a la Insulina , Macrófagos/metabolismo , MicroARNs/metabolismo , Adipocitos/metabolismo , Animales , Células Cultivadas , Glucosa/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Transducción de Señal
2.
Cell ; 147(4): 815-26, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22078880

RESUMEN

Insulin resistance, tissue inflammation, and adipose tissue dysfunction are features of obesity and Type 2 diabetes. We generated adipocyte-specific Nuclear Receptor Corepressor (NCoR) knockout (AKO) mice to investigate the function of NCoR in adipocyte biology, glucose and insulin homeostasis. Despite increased obesity, glucose tolerance was improved in AKO mice, and clamp studies demonstrated enhanced insulin sensitivity in liver, muscle, and fat. Adipose tissue macrophage infiltration and inflammation were also decreased. PPARγ response genes were upregulated in adipose tissue from AKO mice and CDK5-mediated PPARγ ser-273 phosphorylation was reduced, creating a constitutively active PPARγ state. This identifies NCoR as an adaptor protein that enhances the ability of CDK5 to associate with and phosphorylate PPARγ. The dominant function of adipocyte NCoR is to transrepress PPARγ and promote PPARγ ser-273 phosphorylation, such that NCoR deletion leads to adipogenesis, reduced inflammation, and enhanced systemic insulin sensitivity, phenocopying the TZD-treated state.


Asunto(s)
Adipocitos/metabolismo , Proteínas Co-Represoras/genética , Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina , Co-Represor 1 de Receptor Nuclear/metabolismo , PPAR gamma/metabolismo , Animales , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/antagonistas & inhibidores , Fosforilación , Tiazolidinedionas
3.
Am J Physiol Endocrinol Metab ; 300(1): E164-74, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20959535

RESUMEN

Peroxisome proliferator-activated receptor-γ (PPARγ) ligands, including the insulin-sensitizing thiazolidinedione drugs, transcriptionally regulate hundreds of genes. Little is known about the relationship between PPARγ ligand-specific modulation of cellular mechanisms and insulin sensitization. We characterized the insulin sensitivity and multitissue gene expression profiles of lean and insulin-resistant, obese Zucker rats untreated or treated with one of four PPARγ ligands (pioglitazone, rosiglitazone, troglitazone, and AG-035029). We analyzed the transcriptional profiles of adipose tissue, skeletal muscle, and liver from the rats and determined whether ligand treatment insulin-sensitizing potency was related to ligand treatment-induced alteration of functional pathways. Ligand treatments improved insulin sensitivity in obese rats to varying degrees. Adipose tissue profiles revealed ligand treatment-selective modulation of inflammatory and branched-chain amino acid (BCAA) metabolic pathways, which correlated with ligand treatment-specific insulin-sensitizing potency. Skeletal muscle profiles showed that obese rats exhibited elevated expression of adipocyte and slow-twitch fiber markers, which further increased after ligand treatment, but the magnitude of the treatment-induced changes was not correlated with insulin sensitization. Although PPARγ ligand treatments heterogeneously improved dysregulated expression of cholesterol and fatty acid biosynthetic pathways in obese rat liver, these alterations were not correlated with ligand insulin-sensitizing potency. PPARγ ligand treatment-specific insulin-sensitizing potency correlated with modulation of adipose tissue inflammatory and BCAA metabolic pathways, suggesting a functional relationship between these pathways and whole body insulin sensitivity. Other PPARγ ligand treatment-induced functional pathway changes were detected in adipose tissue, skeletal muscle, and liver profiles but were not related to degree of insulin sensitization.


Asunto(s)
Hipoglucemiantes/farmacología , Resistencia a la Insulina , Obesidad/metabolismo , PPAR gamma/agonistas , PPAR gamma/metabolismo , Tejido Adiposo Blanco/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Perfilación de la Expresión Génica , Técnica de Clampeo de la Glucosa , Mediadores de Inflamación/metabolismo , Ligandos , Hígado/metabolismo , Macrófagos/metabolismo , Masculino , Músculo Esquelético/metabolismo , Obesidad/sangre , Obesidad/tratamiento farmacológico , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Distribución Aleatoria , Ratas , Ratas Zucker
4.
Cell Metab ; 33(4): 781-790.e5, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33450179

RESUMEN

Insulin resistance is a major pathophysiologic defect in type 2 diabetes and obesity, while anti-inflammatory M2-like macrophages are important in maintaining normal metabolic homeostasis. Here, we show that M2 polarized bone marrow-derived macrophages (BMDMs) secrete miRNA-containing exosomes (Exos), which improve glucose tolerance and insulin sensitivity when given to obese mice. Depletion of their miRNA cargo blocks the ability of M2 BMDM Exos to enhance insulin sensitivity. We found that miR-690 is highly expressed in M2 BMDM Exos and functions as an insulin sensitizer both in vivo and in vitro. Expressing an miR-690 mimic in miRNA-depleted BMDMs generates Exos that recapitulate the effects of M2 BMDM Exos on metabolic phenotypes. Nadk is a bona fide target mRNA of miR-690, and Nadk plays a role in modulating macrophage inflammation and insulin signaling. Taken together, these data suggest miR-690 could be a new therapeutic insulin-sensitizing agent for metabolic disease.


Asunto(s)
Exosomas/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Animales , Antagomirs/metabolismo , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Dieta Alta en Grasa , Hepatocitos/citología , Hepatocitos/metabolismo , Insulina/metabolismo , Resistencia a la Insulina , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Obesidad/metabolismo , Obesidad/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ribonucleasa III/deficiencia , Ribonucleasa III/genética
5.
Sci Rep ; 9(1): 14779, 2019 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-31611602

RESUMEN

Insulin resistance is a key feature of obesity and type 2 diabetes. PU.1 is a master transcription factor predominantly expressed in macrophages but after HFD feeding PU.1 expression is also significantly increased in adipocytes. We generated adipocyte specific PU.1 knockout mice using adiponectin cre to investigate the role of PU.1 in adipocyte biology, insulin and glucose homeostasis. In HFD-fed obese mice systemic glucose tolerance and insulin sensitivity were improved in PU.1 AKO mice and clamp studies indicated improvements in both adipose and liver insulin sensitivity. At the level of adipose tissue, macrophage infiltration and inflammation was decreased and glucose uptake was increased in PU.1 AKO mice compared with controls. While PU.1 deletion in adipocytes did not affect the gene expression of PPARg itself, we observed increased expression of PPARg target genes in eWAT from HFD fed PU.1 AKO mice compared with controls. Furthermore, we observed decreased phosphorylation at serine 273 in PU.1 AKO mice compared with fl/fl controls, indicating that PPARg is more active when PU.1 expression is reduced in adipocytes. Therefore, in obesity the increased expression of PU.1 in adipocytes modifies the adipocyte PPARg cistrome resulting in impaired glucose tolerance and insulin sensitivity.


Asunto(s)
Adipocitos/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Obesidad/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Células 3T3-L1 , Animales , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Técnicas de Inactivación de Genes , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/sangre , Obesidad/etiología , Obesidad/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Regulación hacia Arriba
6.
Diabetes ; 68(7): 1415-1426, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31010956

RESUMEN

The composition of the gastrointestinal microbiota and associated metabolites changes dramatically with diet and the development of obesity. Although many correlations have been described, specific mechanistic links between these changes and glucose homeostasis remain to be defined. Here we show that blood and intestinal levels of the microbiota-produced N-formyl peptide, formyl-methionyl-leucyl-phenylalanine, are elevated in high-fat diet-induced obese mice. Genetic or pharmacological inhibition of the N-formyl peptide receptor Fpr1 leads to increased insulin levels and improved glucose tolerance, dependent upon glucagon-like peptide 1. Obese Fpr1 knockout mice also display an altered microbiome, exemplifying the dynamic relationship between host metabolism and microbiota. Overall, we describe a new mechanism by which the gut microbiota can modulate glucose metabolism, providing a potential approach for the treatment of metabolic disease.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Microbiota/fisiología , Obesidad/metabolismo , Oligopéptidos/metabolismo , Animales , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Cromatografía Liquida , Dieta Alta en Grasa , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Glucosa/farmacología , Intolerancia a la Glucosa , Hibridación Fluorescente in Situ , Insulina/metabolismo , Masculino , Espectrometría de Masas , Ratones , Ratones Noqueados , Ratones Obesos , Obesidad/inducido químicamente
7.
Biochem Biophys Res Commun ; 364(3): 515-21, 2007 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-17963725

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a nuclear receptor regulated by the insulin-sensitizing thiazolidinediones (TZDs). We studied selective modulation of endogenous genes by PPARgamma ligands using microarray, RNA expression kinetics, and chromatin immunoprecipitation (ChIP) in 3T3-L1 adipocytes. We found over 300 genes that were significantly regulated the TZDs pioglitazone, rosiglitazone, and troglitazone. TZD-mediated expression profiles were unique but overlapping. Ninety-one genes were commonly regulated by all three ligands. TZD time course and dose-response studies revealed gene- and TZD-specific expression kinetics. PEPCK expression was induced rapidly but PDK4 expression was induced gradually. Troglitazone EC50 values for PEPCK, PDK4, and RGS2 regulation were greater than those for pioglitazone and rosiglitazone. TZDs differentially induced histone acetylation of and PPARgamma recruitment to target gene promoters. Selective modulation of PPARgamma by TZDs resulted in distinct expression profiles and transcription kinetics which may be due to differential promoter activation and chromatin remodeling of target genes.


Asunto(s)
Regulación de la Expresión Génica/genética , PPAR gamma/genética , Regiones Promotoras Genéticas/genética , Activación Transcripcional/genética , Células 3T3-L1 , Animales , Ratones
8.
J Clin Invest ; 127(3): 1019-1030, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28192375

RESUMEN

Tissue inflammation is a key component of obesity-induced insulin resistance, with a variety of immune cell types accumulating in adipose tissue. Here, we have demonstrated increased numbers of B2 lymphocytes in obese adipose tissue and have shown that high-fat diet-induced (HFD-induced) insulin resistance is mitigated in B cell-deficient (Bnull) mice. Adoptive transfer of adipose tissue B2 cells (ATB2) from wild-type HFD donor mice into HFD Bnull recipients completely restored the effect of HFD to induce insulin resistance. Recruitment and activation of ATB2 cells was mediated by signaling through the chemokine leukotriene B4 (LTB4) and its receptor LTB4R1. Furthermore, the adverse effects of ATB2 cells on glucose homeostasis were partially dependent upon T cells and macrophages. These results demonstrate the importance of ATB2 cells in obesity-induced insulin resistance and suggest that inhibition of the LTB4/LTB4R1 axis might be a useful approach for developing insulin-sensitizing therapeutics.


Asunto(s)
Tejido Adiposo/inmunología , Subgrupos de Linfocitos B/inmunología , Resistencia a la Insulina/inmunología , Leucotrieno B4/inmunología , Receptores de Leucotrieno B4/inmunología , Transducción de Señal/inmunología , Tejido Adiposo/patología , Animales , Subgrupos de Linfocitos B/patología , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Resistencia a la Insulina/genética , Leucotrieno B4/genética , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/inmunología , Obesidad/patología , Receptores de Leucotrieno B4/genética , Transducción de Señal/genética , Linfocitos T/inmunología , Linfocitos T/patología
9.
Nat Med ; 21(3): 239-247, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25706874

RESUMEN

Insulin resistance results from several pathophysiologic mechanisms, including chronic tissue inflammation and defective insulin signaling. We found that liver, muscle and adipose tissue exhibit higher levels of the chemotactic eicosanoid LTB4 in obese high-fat diet (HFD)-fed mice. Inhibition of the LTB4 receptor Ltb4r1, through either genetic or pharmacologic loss of function, led to an anti-inflammatory phenotype with protection from insulin resistance and hepatic steatosis. In vitro treatment with LTB4 directly enhanced macrophage chemotaxis, stimulated inflammatory pathways, reduced insulin-stimulated glucose uptake in L6 myocytes, and impaired insulin-mediated suppression of hepatic glucose output in primary mouse hepatocytes. This was accompanied by lower insulin-stimulated Akt phosphorylation and higher Irs-1/2 serine phosphorylation, and all of these events were dependent on Gαi and Jnk1, two downstream mediators of Ltb4r1 signaling. These observations elucidate a novel role of the LTB4-Ltb4r1 signaling pathway in hepatocyte and myocyte insulin resistance, and they show that in vivo inhibition of Ltb4r1 leads to robust insulin-sensitizing effects.


Asunto(s)
Hepatocitos/inmunología , Resistencia a la Insulina/inmunología , Leucotrieno B4/inmunología , Macrófagos/inmunología , Fibras Musculares Esqueléticas/inmunología , Obesidad/inmunología , Receptores de Leucotrieno B4/inmunología , Animales , Glucemia/metabolismo , Dieta Alta en Grasa , Hígado Graso/inmunología , Hígado Graso/metabolismo , Hepatocitos/metabolismo , Inflamación/inmunología , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Ratones Obesos , Fibras Musculares Esqueléticas/metabolismo , Obesidad/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Leucotrieno B4/antagonistas & inhibidores , Receptores de Leucotrieno B4/genética , Transducción de Señal
10.
Mol Metab ; 4(5): 378-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25973386

RESUMEN

OBJECTIVE: Adipose tissue is the primary site for lipid deposition that protects the organisms in cases of nutrient excess during obesogenic diets. The histone deacetylase Sirtuin 1 (SIRT1) inhibits adipocyte differentiation by targeting the transcription factor peroxisome proliferator activated-receptor gamma (PPARγ). METHODS: To assess the specific role of SIRT1 in adipocytes, we generated Sirt1 adipocyte-specific knockout mice (ATKO) driven by aP2 promoter onto C57BL/6 background. Sirt1 (flx/flx) aP2Cre (+) (ATKO) and Sirt1 (flx/flx) aP2Cre (-) (WT) mice were fed high-fat diet for 5 weeks (short-term) or 15 weeks (chronic-term). Metabolic studies were combined with gene expression analysis and phosphorylation/acetylation patterns in adipose tissue. RESULTS: On standard chow, ATKO mice exhibit low-grade chronic inflammation in adipose tissue, along with glucose intolerance and insulin resistance compared with control fed mice. On short-term HFD, ATKO mice become more glucose intolerant, hyperinsulinemic, insulin resistant and display increased inflammation. During chronic HFD, WT mice developed a metabolic dysfunction, higher than ATKO mice, and thereby, knockout mice are more glucose tolerant, insulin sensitive and less inflamed relative to control mice. SIRT1 attenuates adipogenesis through PPARγ repressive acetylation and, in the ATKO mice adipocyte PPARγ was hyperacetylated. This high acetylation was associated with a decrease in Ser273-PPARγ phosphorylation. Dephosphorylated PPARγ is constitutively active and results in higher expression of genes associated with increased insulin sensitivity. CONCLUSION: Together, these data establish that SIRT1 downregulation in adipose tissue plays a previously unknown role in long-term inflammation resolution mediated by PPARγ activation. Therefore, in the context of obesity, the development of new therapeutics that activate PPARγ by targeting SIRT1 may provide novel approaches to the treatment of T2DM.

11.
J Clin Invest ; 122(7): 2444-53, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22653059

RESUMEN

Obesity-induced inflammation is a key component of systemic insulin resistance, which is a hallmark of type 2 diabetes. A major driver of this inflammation/insulin resistance syndrome is the accumulation of proinflammatory macrophages in adipose tissue and liver. We found that the orphan GPCR Gpr21 was highly expressed in the hypothalamus and macrophages of mice and that whole-body KO of this receptor led to a robust improvement in glucose tolerance and systemic insulin sensitivity and a modest lean phenotype. The improvement in insulin sensitivity in the high-fat diet-fed (HFD-fed) Gpr21 KO mouse was traced to a marked reduction in tissue inflammation caused by decreased chemotaxis of Gpr21 KO macrophages into adipose tissue and liver. Furthermore, mice lacking macrophage expression of Gpr21 were protected from HFD-induced inflammation and displayed improved insulin sensitivity. Results of in vitro chemotaxis studies in human monocytes suggested that the defect in chemotaxis observed ex vivo and in vivo in mice is also translatable to humans. Cumulatively, our data indicate that GPR21 has a critical function in coordinating macrophage proinflammatory activity in the context of obesity-induced insulin resistance.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/genética , Animales , Trasplante de Médula Ósea , Ingestión de Alimentos , Metabolismo Energético , Epidídimo/metabolismo , Perfilación de la Expresión Génica , Glucosa/metabolismo , Hipotálamo/metabolismo , Mediadores de Inflamación/metabolismo , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Hígado/metabolismo , Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/patología , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/metabolismo , Eliminación de Secuencia , Transcripción Genética , Aumento de Peso
12.
PLoS One ; 5(11): e13959, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-21103061

RESUMEN

BACKGROUND: Insulin resistance is manifested in muscle, adipose tissue, and liver and is associated with adipose tissue inflammation. The cellular components and mechanisms that regulate the onset of diet-induced insulin resistance are not clearly defined. METHODOLOGY AND PRINCIPAL FINDINGS: We initially observed osteopontin (OPN) mRNA over-expression in adipose tissue of obese, insulin resistant humans and rats which was normalized by thiazolidinedione (TZD) treatment in both species. OPN regulates inflammation and is implicated in pathogenic maladies resulting from chronic obesity. Thus, we tested the hypothesis that OPN is involved in the early development of insulin resistance using a 2-4 week high fat diet (HFD) model. OPN KO mice fed HFD for 2 weeks were completely protected from the severe skeletal muscle, liver and adipose tissue insulin resistance that developed in wild type (WT) controls, as determined by hyperinsulinemic euglycemic clamp and acute insulin-stimulation studies. Although two-week HFD did not alter body weight or plasma free fatty acids and cytokines in either strain, HFD-induced hyperleptinemia, increased adipose tissue inflammation (macrophages and cytokines), and adipocyte hypertrophy were significant in WT mice and blunted or absent in OPN KO mice. Adipose tissue OPN protein isoform expression was significantly altered in 2- and 4-week HFD-fed WT mice but total OPN protein was unchanged. OPN KO bone marrow stromal cells were more osteogenic and less adipogenic than WT cells in vitro. Interestingly, the two differentiation pathways were inversely affected by HFD in WT cells in vitro. CONCLUSIONS: The OPN KO phenotypes we report reflect protection from insulin resistance that is associated with changes in adipocyte biology and adipose tissue inflammatory status. OPN is a key component in the development of HFD-induced insulin resistance.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Resistencia a la Insulina , Osteopontina/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Adulto , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipoglucemiantes/farmacología , Insulina/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Osteopontina/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Zucker , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Tiazolidinedionas/farmacología
13.
PLoS One ; 4(9): e7250, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19787041

RESUMEN

BACKGROUND: Recent understanding that insulin resistance is an inflammatory condition necessitates searching for genes that regulate inflammation in insulin sensitive tissues. 12/15-lipoxygenase (12/15LO) regulates the expression of proinflammatory cytokines and chemokines and is implicated in the early development of diet-induced atherosclerosis. Thus, we tested the hypothesis that 12/15LO is involved in the onset of high fat diet (HFD)-induced insulin resistance. METHODOLOGY/PRINCIPAL FINDINGS: Cells over-expressing 12/15LO secreted two potent chemokines, MCP-1 and osteopontin, implicated in the development of insulin resistance. We assessed adipose tissue inflammation and whole body insulin resistance in wild type (WT) and 12/15LO knockout (KO) mice after 2-4 weeks on HFD. In adipose tissue from WT mice, HFD resulted in recruitment of CD11b(+), F4/80(+) macrophages and elevated protein levels of the inflammatory markers IL-1beta, IL-6, IL-10, IL-12, IFNgamma, Cxcl1 and TNFalpha. Remarkably, adipose tissue from HFD-fed 12/15LO KO mice was not infiltrated by macrophages and did not display any increase in the inflammatory markers compared to adipose tissue from normal chow-fed mice. WT mice developed severe whole body (hepatic and skeletal muscle) insulin resistance after HFD, as measured by hyperinsulinemic euglycemic clamp. In contrast, 12/15LO KO mice exhibited no HFD-induced change in insulin-stimulated glucose disposal rate or hepatic glucose output during clamp studies. Insulin-stimulated Akt phosphorylation in muscle tissue from HFD-fed mice was significantly greater in 12/15LO KO mice than in WT mice. CONCLUSIONS: These results demonstrate that 12/15LO mediates early stages of adipose tissue inflammation and whole body insulin resistance induced by high fat feeding.


Asunto(s)
Tejido Adiposo/metabolismo , Araquidonato 12-Lipooxigenasa/fisiología , Araquidonato 15-Lipooxigenasa/fisiología , Grasas de la Dieta/metabolismo , Resistencia a la Insulina , Alimentación Animal , Animales , Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Antígeno CD11b/biosíntesis , Cruzamientos Genéticos , Humanos , Inflamación , Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA