Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
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
3.
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
4.
Diabetes ; 71(8): 1617-1619, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35881834
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
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
16.
Cell Metab ; 29(2): 457-474.e5, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30595478

RESUMEN

The nature of obesity-associated islet inflammation and its impact on ß cell abnormalities remains poorly defined. Here, we explore immune cell components of islet inflammation and define their roles in regulating ß cell function and proliferation. Islet inflammation in obese mice is dominated by macrophages. We identify two islet-resident macrophage populations, characterized by their anatomical distributions, distinct phenotypes, and functional properties. Obesity induces the local expansion of resident intra-islet macrophages, independent of recruitment from circulating monocytes. Functionally, intra-islet macrophages impair ß cell function in a cell-cell contact-dependent manner. Increased engulfment of ß cell insulin secretory granules by intra-islet macrophages in obese mice may contribute to restricting insulin secretion. In contrast, both intra- and peri-islet macrophage populations from obese mice promote ß cell proliferation in a PDGFR signaling-dependent manner. Together, these data define distinct roles and mechanisms for islet macrophages in the regulation of islet ß cells.


Asunto(s)
Inflamación/inmunología , Células Secretoras de Insulina/metabolismo , Macrófagos/inmunología , Obesidad/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/inmunología , Animales , Línea Celular , Proliferación Celular , Secreción de Insulina , Células Secretoras de Insulina/patología , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos
17.
Mol Metab ; 20: 89-101, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30553772

RESUMEN

OBJECTIVE: Atherosclerosis is a major cause of cardiovascular disease. Monocyte-endothelial cell interactions are partly mediated by expression of monocyte CX3CR1 and endothelial cell fractalkine (CX3CL1). Interrupting the interaction between this ligand-receptor pair should reduce monocyte binding to the endothelial wall and reduce atherosclerosis. We sought to reduce atherosclerosis by preventing monocyte-endothelial cell interactions through use of a long-acting CX3CR1 agonist. METHODS: In this study, the chemokine domain of CX3CL1 was fused to the mouse Fc region to generate a long-acting soluble form of CX3CL1 suitable for chronic studies. CX3CL1-Fc or saline was injected twice a week (30 mg/kg) for 4 months into Ldlr knockout (KO) mice on an atherogenic western diet. RESULTS: CX3CL1-Fc-treated Ldlr KO mice showed decreased en face aortic lesion surface area and reduced aortic root lesion size with decreased necrotic core area. Flow cytometry analyses of CX3CL1-Fc-treated aortic wall cell digests revealed a decrease in M1-like polarized macrophages and T cells. Moreover, CX3CL1-Fc administration reduced diet-induced atherosclerosis after switching from an atherogenic to a normal chow diet. In vitro monocyte adhesion studies revealed that CX3CL1-Fc treatment caused fewer monocytes to adhere to a human umbilical vein endothelial cell monolayer. Furthermore, a dorsal window chamber model demonstrated that CX3CL1-Fc treatment decreased in vivo leukocyte adhesion and rolling in live capillaries after short-term ischemia-reperfusion. CONCLUSION: These results indicate that CX3CL1-Fc can inhibit monocyte/endothelial cell adhesion as well as reduce atherosclerosis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Quimiocina CX3CL1/uso terapéutico , Placa Aterosclerótica/tratamiento farmacológico , Animales , Aorta/patología , Aterosclerosis/genética , Aterosclerosis/prevención & control , Células Cultivadas , Quimiocina CX3CL1/genética , Fragmentos Fc de Inmunoglobulinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Placa Aterosclerótica/prevención & control , Receptores de LDL/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico
18.
J Clin Invest ; 128(4): 1458-1470, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29504946

RESUMEN

We have previously reported that the fractalkine (FKN)/CX3CR1 system represents a novel regulatory mechanism for insulin secretion and ß cell function. Here, we demonstrate that chronic administration of a long-acting form of FKN, FKN-Fc, can exert durable effects to improve glucose tolerance with increased glucose-stimulated insulin secretion and decreased ß cell apoptosis in obese rodent models. Unexpectedly, chronic FKN-Fc administration also led to decreased α cell glucagon secretion. In islet cells, FKN inhibited ATP-sensitive potassium channel conductance by an ERK-dependent mechanism, which triggered ß cell action potential (AP) firing and decreased α cell AP amplitude. This results in increased glucose-stimulated insulin secretion and decreased glucagon secretion. Beyond its islet effects, FKN-Fc also exerted peripheral effects to enhance hepatic insulin sensitivity due to inhibition of glucagon action. In hepatocytes, FKN treatment reduced glucagon-stimulated cAMP production and CREB phosphorylation in a pertussis toxin-sensitive manner. Together, these results raise the possibility of use of FKN-based therapy to improve type 2 diabetes by increasing both insulin secretion and insulin sensitivity.


Asunto(s)
Glucemia/metabolismo , Quimiocina CX3CL1/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Animales , Glucemia/genética , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Quimiocina CX3CL1/genética , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Fragmentos Fc de Inmunoglobulinas/genética , Secreción de Insulina/genética , Células Secretoras de Insulina/patología , Ratones , Ratones Transgénicos , Proteínas Recombinantes de Fusión/genética
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...