Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Cell ; 172(1-2): 22-40, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29328913

RESUMEN

The worldwide obesity epidemic has emerged as a major cause of insulin resistance and Type 2 diabetes. Chronic tissue inflammation is a well-recognized feature of obesity, and the field of immunometabolism has witnessed many advances in recent years. Here, we review the major features of our current understanding with respect to chronic obesity-related inflammation in metabolic tissues and focus on how these inflammatory changes affect insulin sensitivity, insulin secretion, food intake, and glucose homeostasis. There is a growing appreciation of the varied and sometimes integrated crosstalk between cells within a tissue (intraorgan) and tissues within an organism (interorgan) that supports inflammation in the context of metabolic dysregulation. Understanding these pathways and modes of communication has implications for translational studies. We also briefly summarize the state of this field with respect to potential current and developing therapeutics.


Asunto(s)
Trastornos del Metabolismo de la Glucosa/metabolismo , Inmunidad Innata , Integración de Sistemas , Animales , Trastornos del Metabolismo de la Glucosa/etiología , Trastornos del Metabolismo de la Glucosa/inmunología , Humanos , Inflamación/inmunología , Inflamación/metabolismo
2.
Immunity ; 56(1): 3-5, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36630915

RESUMEN

Nonalcoholic steatohepatitis (NASH) is a common liver disease involving interactions between a variety of liver cell types. In this issue of Immunity, Wang et al. show that efferocytosis of dying lipid-laden hepatocytes by hepatic macrophages protects against the development of NASH.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Hígado/metabolismo , Inflamación/metabolismo , Macrófagos , Hepatocitos/metabolismo , Ratones Endogámicos C57BL
3.
Cell ; 157(6): 1339-1352, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24906151

RESUMEN

Adipose tissue hypoxia and inflammation have been causally implicated in obesity-induced insulin resistance. Here, we report that, early in the course of high-fat diet (HFD) feeding and obesity, adipocyte respiration becomes uncoupled, leading to increased oxygen consumption and a state of relative adipocyte hypoxia. These events are sufficient to trigger HIF-1α induction, setting off the chronic adipose tissue inflammatory response characteristic of obesity. At the molecular level, these events involve saturated fatty acid stimulation of the adenine nucleotide translocase 2 (ANT2), an inner mitochondrial membrane protein, which leads to the uncoupled respiratory state. Genetic or pharmacologic inhibition of either ANT2 or HIF-1α can prevent or reverse these pathophysiologic events, restoring a state of insulin sensitivity and glucose tolerance. These results reveal the sequential series of events in obesity-induced inflammation and insulin resistance.


Asunto(s)
Adipocitos/metabolismo , Dieta Alta en Grasa , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Oxígeno/metabolismo , Translocador 2 del Nucleótido Adenina/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia de la Célula , Ácidos Grasos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inflamación/metabolismo , Ácido Láctico/metabolismo , Ratones , Ratones Noqueados , Óxido Nítrico/metabolismo
4.
Genes Dev ; 35(5-6): 307-328, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33649162

RESUMEN

Obesity is the most common cause of insulin resistance, and the current obesity epidemic is driving a parallel rise in the incidence of T2DM. It is now widely recognized that chronic, subacute tissue inflammation is a major etiologic component of the pathogenesis of insulin resistance and metabolic dysfunction in obesity. Here, we summarize recent advances in our understanding of immunometabolism. We discuss the characteristics of chronic inflammation in the major metabolic tissues and how obesity triggers these events, including a focus on the role of adipose tissue hypoxia and macrophage-derived exosomes. Last, we also review current and potential new therapeutic strategies based on immunomodulation.


Asunto(s)
Inflamación , Enfermedades Metabólicas/fisiopatología , Tejido Adiposo/citología , Tejido Adiposo/fisiopatología , Hipoxia de la Célula , Enfermedad Crónica , Exosomas/metabolismo , Humanos , Inmunomodulación , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/terapia
5.
Cell ; 153(2): 413-25, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23582329

RESUMEN

Here, we demonstrate that the fractalkine (FKN)/CX3CR1 system represents a regulatory mechanism for pancreatic islet ß cell function and insulin secretion. CX3CR1 knockout (KO) mice exhibited a marked defect in glucose and GLP1-stimulated insulin secretion, and this defect was also observed in vitro in isolated islets from CX3CR1 KO mice. In vivo administration of FKN improved glucose tolerance with an increase in insulin secretion. In vitro treatment of islets with FKN increased intracellular Ca(2+) and potentiated insulin secretion in both mouse and human islets. The KO islets exhibited reduced expression of a set of genes necessary for the fully functional, differentiated ß cell state, whereas treatment of wild-type (WT) islets with FKN led to increased expression of these genes. Lastly, expression of FKN in islets was decreased by aging and high-fat diet/obesity, suggesting that decreased FKN/CX3CR1 signaling could be a mechanism underlying ß cell dysfunction in type 2 diabetes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores de Quimiocina/metabolismo , Transducción de Señal , Adulto , Envejecimiento , Animales , Receptor 1 de Quimiocinas CX3C , Cadáver , Quimiocina CX3CL1/administración & dosificación , Quimiocina CX3CL1/metabolismo , Dieta Alta en Grasa , Expresión Génica , Glucosa/metabolismo , Humanos , Hiperglucemia/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Receptores de Quimiocina/genética
6.
Mol Cell Proteomics ; 12(12): 3744-58, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24030101

RESUMEN

Insulin resistance plays a major role in the development of type 2 diabetes and obesity and affects a number of biological processes such as mitochondrial biogenesis. Though mitochondrial dysfunction has been linked to the development of insulin resistance and pathogenesis of type 2 diabetes, the precise mechanism linking the two is not well understood. We used high fat diet (HFD)-induced obesity dependent diabetes mouse models to gain insight into the potential pathways altered with metabolic disease, and carried out quantitative proteomic analysis of liver mitochondria. As previously reported, proteins involved in fatty acid oxidation, branched chain amino acid degradation, tricarboxylic acid cycle, and oxidative phosphorylation were uniformly up-regulated in the liver of HFD fed mice compared with that of normal diet. Further, our studies revealed that retinol metabolism is distinctly down-regulated and the mitochondrial structural proteins-components of mitochondrial inter-membrane space bridging (MIB) complex (Mitofilin, Sam50, and ChChd3), and Tim proteins-essential for protein import, are significantly up-regulated in HFD fed mice. Structural and functional studies on HFD and normal diet liver mitochondria revealed remodeling of HFD mitochondria to a more condensed form with increased respiratory capacity and higher ATP levels compared with normal diet mitochondria. Thus, it is likely that the structural remodeling is essential to accommodate the increased protein content in presence of HFD: the mechanism could be through the MIB complex promoting contact site and crista junction formation and in turn facilitating the lipid and protein uptake.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Proteoma/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Ciclo del Ácido Cítrico/genética , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Dieta Alta en Grasa , Grasas de la Dieta/administración & dosificación , Regulación de la Expresión Génica , Resistencia a la Insulina , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/ultraestructura , Proteínas Mitocondriales/genética , Anotación de Secuencia Molecular , Obesidad/inducido químicamente , Obesidad/genética , Fosforilación Oxidativa , Mapeo de Interacción de Proteínas , Proteoma/genética , Transducción de Señal , Espectrometría de Masas en Tándem , Vitamina A/metabolismo
7.
Nat Metab ; 6(5): 880-898, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38605183

RESUMEN

The obesity epidemic continues to worsen worldwide, driving metabolic and chronic inflammatory diseases. Thiazolidinediones, such as rosiglitazone (Rosi), are PPARγ agonists that promote 'M2-like' adipose tissue macrophage (ATM) polarization and cause insulin sensitization. As ATM-derived small extracellular vesicles (ATM-sEVs) from lean mice are known to increase insulin sensitivity, we assessed the metabolic effects of ATM-sEVs from Rosi-treated obese male mice (Rosi-ATM-sEVs). Here we show that Rosi leads to improved glucose and insulin tolerance, transcriptional repolarization of ATMs and increased sEV secretion. Administration of Rosi-ATM-sEVs rescues obesity-induced glucose intolerance and insulin sensitivity in vivo without the known thiazolidinedione-induced adverse effects of weight gain or haemodilution. Rosi-ATM-sEVs directly increase insulin sensitivity in adipocytes, myotubes and primary mouse and human hepatocytes. Additionally, we demonstrate that the miRNAs within Rosi-ATM-sEVs, primarily miR-690, are responsible for these beneficial metabolic effects. Thus, using ATM-sEVs with specific miRNAs may provide a therapeutic path to induce insulin sensitization.


Asunto(s)
Tejido Adiposo , Vesículas Extracelulares , Resistencia a la Insulina , Macrófagos , Rosiglitazona , Animales , Rosiglitazona/farmacología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Ratones , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/efectos de los fármacos , Masculino , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Obesidad/metabolismo , Insulina/metabolismo , Adipocitos/metabolismo , Adipocitos/efectos de los fármacos , Ratones Endogámicos C57BL
8.
Diabetes ; 71(7): 1508-1524, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35472707

RESUMEN

In obesity, increased mitochondrial metabolism with the accumulation of oxidative stress leads to mitochondrial damage and ß-cell dysfunction. In particular, ß-cells express antioxidant enzymes at relatively low levels and are highly vulnerable to oxidative stress. Early in the development of obesity, ß-cells exhibit increased glucose-stimulated insulin secretion in order to compensate for insulin resistance. This increase in ß-cell function under the condition of enhanced metabolic stress suggests that ß-cells possess a defense mechanism against increased oxidative damage, which may become insufficient or decline at the onset of type 2 diabetes. Here, we show that metabolic stress induces ß-cell hypoxia inducible factor 2α (HIF-2α), which stimulates antioxidant gene expression (e.g., Sod2 and Cat) and protects against mitochondrial reactive oxygen species (ROS) and subsequent mitochondrial damage. Knockdown of HIF-2α in Min6 cells exaggerated chronic high glucose-induced mitochondrial damage and ß-cell dysfunction by increasing mitochondrial ROS levels. Moreover, inducible ß-cell HIF-2α knockout mice developed more severe ß-cell dysfunction and glucose intolerance on a high-fat diet, along with increased ROS levels and decreased islet mitochondrial mass. Our results provide a previously unknown mechanism through which ß-cells defend against increased metabolic stress to promote ß-cell compensation in obesity.


Asunto(s)
Antioxidantes , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diabetes Mellitus Tipo 2 , Animales , Antioxidantes/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Glucosa/farmacología , Ratones , Ratones Noqueados , Obesidad , Especies Reactivas de Oxígeno/metabolismo
9.
J Biol Chem ; 285(29): 22174-85, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20442402

RESUMEN

Orosomucoid (ORM), also called alpha-1 acid glycoprotein, is an abundant plasma protein that is an immunomodulator induced by stressful conditions such as infections. In this study, we reveal that Orm is induced selectively in the adipose tissue of obese mice to suppress excess inflammation that otherwise disturbs energy homeostasis. Adipose Orm levels were elevated by metabolic signals, including insulin, high glucose, and free fatty acid, as well as by the proinflammatory cytokine tumor necrosis factor-alpha, which is found in increased levels in the adipose tissue of morbid obese subjects. In both adipocytes and macrophages, ORM suppressed proinflammatory gene expression and pathways such as NF-kappaB and mitogen-activated protein kinase signalings and reactive oxygen species generation. Concomitantly, ORM relieved hyperglycemia-induced insulin resistance as well as tumor necrosis factor-alpha-mediated lipolysis in adipocytes. Accordingly, ORM improved glucose and insulin tolerance in obese and diabetic db/db mice. Taken together, our results suggest that ORM integrates inflammatory and metabolic signals to modulate immune responses to protect adipose tissue from excessive inflammation and thereby from metabolic dysfunction.


Asunto(s)
Adipoquinas/metabolismo , Metabolismo Energético , Homeostasis , Inflamación/metabolismo , Orosomucoide/metabolismo , Transducción de Señal , Adipocitos/efectos de los fármacos , Adipocitos/enzimología , Adipocitos/patología , Adipoquinas/genética , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/metabolismo , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Inflamación/sangre , Inflamación/patología , Insulina/metabolismo , Resistencia a la Insulina , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Ratones Obesos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Orosomucoide/genética , Transducción de Señal/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
10.
JCI Insight ; 6(20)2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34676827

RESUMEN

Macrophage proinflammatory activation is an important etiologic component of the development of insulin resistance and metabolic dysfunction in obesity. However, the underlying mechanisms are not clearly understood. Here, we demonstrate that a mitochondrial inner membrane protein, adenine nucleotide translocase 2 (ANT2), mediates proinflammatory activation of adipose tissue macrophages (ATMs) in obesity. Ant2 expression was increased in ATMs of obese mice compared with lean mice. Myeloid-specific ANT2-knockout (ANT2-MKO) mice showed decreased adipose tissue inflammation and improved insulin sensitivity and glucose tolerance in HFD/obesity. At the molecular level, we found that ANT2 mediates free fatty acid-induced mitochondrial permeability transition, leading to increased mitochondrial reactive oxygen species production and damage. In turn, this increased HIF-1α expression and NF-κB activation, leading to proinflammatory macrophage activation. Our results provide a previously unknown mechanism for how obesity induces proinflammatory activation of macrophages with propagation of low-grade chronic inflammation (metaflammation).


Asunto(s)
Translocador 2 del Nucleótido Adenina/metabolismo , Inflamación/genética , Activación de Macrófagos/genética , Obesidad/genética , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones
11.
JCI Insight ; 6(21)2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34747371

RESUMEN

Patients with diabetes with coronary microvascular disease (CMD) exhibit higher cardiac mortality than patients without CMD. However, the molecular mechanism by which diabetes promotes CMD is poorly understood. RNA-binding protein human antigen R (HuR) is a key regulator of mRNA stability and translation; therefore, we investigated the role of HuR in the development of CMD in mice with type 2 diabetes. Diabetic mice exhibited decreases in coronary flow velocity reserve (CFVR; a determinant of coronary microvascular function) and capillary density in the left ventricle. HuR levels in cardiac endothelial cells (CECs) were significantly lower in diabetic mice and patients with diabetes than the controls. Endothelial-specific HuR-KO mice also displayed significant reductions in CFVR and capillary density. By examining mRNA levels of 92 genes associated with endothelial function, we found that HuR, Cx40, and Nox4 levels were decreased in CECs from diabetic and HuR-KO mice compared with control mice. Cx40 expression and HuR binding to Cx40 mRNA were downregulated in CECs from diabetic mice. Cx40-KO mice exhibited decreased CFVR and capillary density, whereas endothelium-specific Cx40 overexpression increased capillary density and improved CFVR in diabetic mice. These data suggest that decreased HuR contributes to the development of CMD in diabetes through downregulation of gap junction protein Cx40 in CECs.


Asunto(s)
Conexinas/metabolismo , Diabetes Mellitus Tipo 2/genética , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Masculino , Ratones
12.
Biochem Biophys Res Commun ; 392(3): 323-8, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20060380

RESUMEN

microRNAs (miRNAs) are non-coding small RNAs regulating gene expression, cell growth, and differentiation. Although several miRNAs have been implicated in cell growth and differentiation, it is barely understood their roles in adipocyte differentiation. In the present study, we reveal that miR-27a is involved in adipocyte differentiation by binding to the PPARgamma 3'-UTR whose sequence motifs are highly conserved in mammals. During adipogenesis, the expression level of miR-27a was inversely correlated with that of adipogenic marker genes such as PPARgamma and adiponectin. In white adipose tissue, miR-27a was more abundantly expressed in stromal vascular cell fraction than in mature adipocyte fraction. Ectopic expression of miR-27a in 3T3-L1 pre-adipocytes repressed adipocyte differentiation by reducing PPARgamma expression. Interestingly, the level of miR-27a in mature adipocyte fraction of obese mice was down-regulated than that of lean mice. Together, these results suggest that miR-27a would suppress adipocyte differentiation through targeting PPARgamma and thereby down-regulation of miR-27a might be associated with adipose tissue dysregulation in obesity.


Asunto(s)
Adipocitos/citología , Adipogénesis/genética , Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , PPAR gamma/genética , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Secuencia de Bases , Regulación hacia Abajo , Ratones , MicroARNs/genética , Obesidad/genética
13.
Mol Cell Biol ; 27(2): 438-52, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17074803

RESUMEN

Insulin plays a critical role in whole-body energy homeostasis by regulating lipid and glucose metabolism. In fat and liver tissues, ADD1/SREBP1c is a key transcription factor to mediate insulin-dependent regulation of gene expression. Although transcriptional and proteolytic activation of ADD1/SREBP1c has been studied intensively, the mechanism by which insulin regulates expression of its target genes with ADD1/SREBP1c at the chromatin level is unclear. Here, we reveal that SWI/SNF chromatin remodeling factors interact with the ADD1/SREBP1c and actively regulate insulin-dependent gene expression. Insulin enhanced recruitment of SWI/SNF chromatin remodeling factors to its target gene promoters with concomitant changes in the chromatin structures as well as gene expression. Furthermore, in vivo overexpression of BAF155/SRG3, a component of the SWI/SNF complex, substantially promoted insulin target gene expression and insulin sensitivity. Taken together, our results suggest that the SWI/SNF chromatin remodeling complexes confer not only insulin-dependent gene expression but also insulin sensitivity in vivo via interaction with ADD1/SREBP1c.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Insulina/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Regulación de la Expresión Génica , Insulina/farmacología , Masculino , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo
14.
Nat Rev Endocrinol ; 16(2): 81-90, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31836875

RESUMEN

Chronic, unresolved tissue inflammation is a well-described feature of obesity, type 2 diabetes mellitus (T2DM) and other insulin-resistant states. In this context, adipose tissue and liver inflammation have been particularly well studied; however, abundant evidence demonstrates that inflammatory processes are also activated in pancreatic islets from obese animals and humans with obesity and/or T2DM. In this Review, we focus on the characteristics of immune cell-mediated inflammation in islets and the consequences of this with respect to ß-cell function. In contrast to type 1 diabetes mellitus, the dominant immune cell type causing inflammation in obese and T2DM islets is the macrophage. The increased macrophage accumulation in T2DM islets primarily arises through local proliferation of resident macrophages, which then provide signals (such as platelet-derived growth factor) that drive ß-cell hyperplasia (a classic feature of obesity). In addition, islet macrophages also impair the insulin secretory capacity of ß-cells. Through these mechanisms, islet-resident macrophages underlie the inflammatory response in obesity and mechanistically participate in the ß-cell hyperplasia and dysfunction that characterizes this insulin-resistant state. These findings point to the possibility of therapeutics that target islet inflammation to elicit beneficial effects on ß-cell function and glycaemia.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Macrófagos/fisiología , Obesidad/metabolismo , Animales , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Células Secretoras de Insulina/inmunología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Obesidad/inmunología
15.
Mol Metab ; 41: 101039, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32534258

RESUMEN

OBJECTIVE: Recent evidence indicates that inhibition of prolyl hydroxylase domain (PHD) proteins can exert beneficial effects to improve metabolic abnormalities in mice and humans. However, the underlying mechanisms are not clearly understood. This study was designed to address this question. METHODS: A pan-PHD inhibitor compound was injected into WT and liver-specific hypoxia-inducible factor (HIF)-2α KO mice, after onset of obesity and glucose intolerance, and changes in glucose and glucagon tolerance were measured. Tissue-specific changes in basal glucose flux and insulin sensitivity were also measured by hyperinsulinemic euglycemic clamp studies. Molecular and cellular mechanisms were assessed in normal and type 2 diabetic human hepatocytes, as well as in mouse hepatocytes. RESULTS: Administration of a PHD inhibitor compound (PHDi) after the onset of obesity and insulin resistance improved glycemic control by increasing insulin and decreasing glucagon sensitivity in mice, independent of body weight change. Hyperinsulinemic euglycemic clamp studies revealed that these effects of PHDi treatment were mainly due to decreased basal hepatic glucose output and increased liver insulin sensitivity. Hepatocyte-specific deletion of HIF-2α markedly attenuated these effects of PHDi treatment, showing PHDi effects are HIF-2α dependent. At the molecular level, HIF-2α induced increased Irs2 and cyclic AMP-specific phosphodiesterase gene expression, leading to increased and decreased insulin and glucagon signaling, respectively. These effects of PHDi treatment were conserved in human and mouse hepatocytes. CONCLUSIONS: Our results elucidate unknown mechanisms for how PHD inhibition improves glycemic control through HIF-2α-dependent regulation of hepatic insulin and glucagon sensitivity.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Insulina/metabolismo , Inhibidores de Prolil-Hidroxilasa/farmacología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Glucagón/metabolismo , Glucosa/metabolismo , Hepatocitos/metabolismo , Humanos , Resistencia a la Insulina/fisiología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Prolil Hidroxilasas/metabolismo , Inhibidores de Prolil-Hidroxilasa/metabolismo , Transducción de Señal
16.
Cell Metab ; 31(1): 162-173.e5, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31708444

RESUMEN

Insulin resistance is a major factor in obesity-linked type 2 diabetes. PPARγ is a master regulator of adipogenesis, and small molecule agonists, termed thiazolidinediones, are potent therapeutic insulin sensitizers. Here, we studied the role of transcriptional co-activator with PDZ-binding motif (TAZ) as a transcriptional co-repressor of PPARγ. We found that adipocyte-specific TAZ knockout (TAZ AKO) mice demonstrate a constitutively active PPARγ state. Obese TAZ AKO mice show improved glucose tolerance and insulin sensitivity compared to littermate controls. PPARγ response genes are upregulated in adipose tissue from TAZ AKO mice and adipose tissue inflammation was also decreased. In vitro and in vivo mechanistic studies revealed that the TAZ-PPARγ interaction is partially dependent on ERK-mediated Ser112 PPARγ phosphorylation. As adipocyte PPARγ Ser112 phosphorylation is increased in obesity, repression of PPARγ activity by TAZ could contribute to insulin resistance. These results identify TAZ as a new factor in the development of obesity-induced insulin resistance.


Asunto(s)
Adipocitos/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina/genética , PPAR gamma/metabolismo , Transactivadores/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Adipocitos/enzimología , Adipogénesis/genética , Animales , Línea Celular , Dieta Alta en Grasa , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Inmunohistoquímica , Inflamación/genética , Inflamación/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Obesos , PPAR gamma/genética , Fosforilación , Transactivadores/genética
17.
Mol Endocrinol ; 22(9): 2176-89, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18562625

RESUMEN

Glutathione peroxidase 3 (GPx3) accounts for the major antioxidant activity in the plasma. Here, we demonstrate that down-regulation of GPx3 in the plasma of obese subjects is associated with adipose GPx3 dysregulation, resulting from the increase of inflammatory signals and oxidative stress. Although GPx3 was abundantly expressed in kidney, lung, and adipose tissue, we observed that GPx3 expression was reduced selectively in the adipose tissue of several obese animal models as decreasing plasma GPx3 level. Adipose GPx3 expression was greatly suppressed by prooxidative conditions such as high levels of TNFalpha and hypoxia. In contrast, the antioxidant N-acetyl cysteine and the antidiabetic drug rosiglitazone increased adipose GPx3 expression in obese and diabetic db/db mice. Moreover, GPx3 overexpression in adipocytes improved high glucose-induced insulin resistance and attenuated inflammatory gene expression whereas GPx3 neutralization in adipocytes promoted expression of proinflammatory genes. Taken together, these data suggest that suppression of GPx3 expression in the adipose tissue of obese subjects might constitute a vicious cycle to expand local reactive oxygen species accumulation in adipose tissue potentially into systemic oxidative stress and obesity-related metabolic complications.


Asunto(s)
Tejido Adiposo/enzimología , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Obesidad/enzimología , Obesidad/genética , Células 3T3-L1 , Acetilcisteína/farmacología , Tejido Adiposo/efectos de los fármacos , Animales , Antioxidantes/farmacología , Secuencia de Bases , Regulación Enzimológica de la Expresión Génica/genética , Glutatión Peroxidasa/sangre , Humanos , Hipoxia/enzimología , Hipoxia/genética , Riñón/enzimología , Lipopolisacáridos/farmacología , Pulmón/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/sangre , Estrés Oxidativo , PPAR gamma/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Rosiglitazona , Tiazolidinedionas/farmacología , Factor de Necrosis Tumoral alfa/farmacología
18.
Sci Adv ; 5(7): eaaw4176, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31281892

RESUMEN

The decrease in incretin effects is an important etiologic component of type 2 diabetes with unknown mechanisms. In an attempt to understand obesity-induced changes in liver oxygen homeostasis, we found that liver HIF-1α expression was increased mainly by soluble factors released from obese adipocytes, leading to decreased incretin effects. Deletion of hepatocyte HIF-1α protected obesity-induced glucose intolerance without changes in body weight, liver steatosis, or insulin resistance. In-depth mouse metabolic phenotyping revealed that obesity increased first-pass degradation of an incretin hormone GLP-1 with increased liver DPP4 expression and decreased sinusoidal blood flow rate, reducing active GLP-1 levels in peripheral circulation. Hepatocyte HIF-1α KO blocked these changes induced by obesity. Deletion of hepatocyte HIF-2α did not change liver DPP4 expression but improved hepatic steatosis. Our results identify a previously unknown pathway for obesity-induced impaired beta cell glucose response (incretin effects) and the development of glucose intolerance through inter-organ communications.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Hepatocitos/metabolismo , Obesidad/etiología , Tejido Adiposo/metabolismo , Tejido Adiposo/fisiopatología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Dieta Alta en Grasa/efectos adversos , Dipeptidil Peptidasa 4/metabolismo , Hígado Graso/etiología , Hígado Graso/metabolismo , Glucosa/metabolismo , Hepatitis/etiología , Resistencia a la Insulina , Hígado/irrigación sanguínea , Hígado/metabolismo , Masculino , Ratones Noqueados
19.
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
20.
Nat Metab ; 1(1): 86-97, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-31528845

RESUMEN

Decreased adipose tissue oxygen tension and increased HIF-1α expression can trigger adipose tissue inflammation and dysfunction in obesity. Our current understanding of obesity-associated decreased adipose tissue oxygen tension is mainly focused on changes in oxygen supply and angiogenesis. Here, we demonstrate that increased adipocyte O2 demand, mediated by ANT2 activity, is the dominant cause of adipocyte hypoxia. Deletion of adipocyte Ant2 improves obesity-induced intracellular adipocyte hypoxia by decreasing obesity-induced adipocyte oxygen demand, without effects on mitochondrial number or mass, or oligomycin-sensitive respiration. This led to decreased adipose tissue HIF-1α expression and inflammation with improved glucose tolerance and insulin resistance in both a preventative or therapeutic setting. Our results suggest that ANT2 may be a target for the development of insulin sensitizing drugs and that ANT2 inhibition might have clinical utility.


Asunto(s)
Translocador 2 del Nucleótido Adenina/deficiencia , Adipocitos/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Resistencia a la Insulina/genética , Obesidad/etiología , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Animales , Apoptosis , Fibrosis , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA