Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
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
2.
Endocr Rev ; 43(3): 441-468, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35552682

RESUMEN

During the last decade, there has been great interest in elucidating the biological role of extracellular vesicles (EVs), particularly, their hormone-like role in cell-to-cell communication. The field of endocrinology is uniquely placed to provide insight into the functions of EVs, which are secreted from all cells into biological fluids and carry endocrine signals to engage in paracellular and distal interactions. EVs are a heterogeneous population of membrane-bound vesicles of varying size, content, and bioactivity. EVs are specifically packaged with signaling molecules, including lipids, proteins, and nucleic acids, and are released via exocytosis into biofluid compartments. EVs regulate the activity of both proximal and distal target cells, including translational activity, metabolism, growth, and development. As such, EVs signaling represents an integral pathway mediating intercellular communication. Moreover, as the content of EVs is cell-type specific, it is a "fingerprint" of the releasing cell and its metabolic status. Recently, changes in the profile of EV and bioactivity have been described in several endocrine-related conditions including diabetes, obesity, cardiovascular diseases, and cancer. The goal of this statement is to highlight relevant aspects of EV research and their potential role in the field of endocrinology.


Asunto(s)
Endocrinología , Vesículas Extracelulares , Transporte Biológico , Comunicación Celular/fisiología , Vesículas Extracelulares/fisiología , Humanos , Transducción de Señal
3.
Nat Cell Biol ; 24(6): 954-967, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35637408

RESUMEN

Epidemiological studies demonstrate an association between breast cancer (BC) and systemic dysregulation of glucose metabolism. However, how BC influences glucose homeostasis remains unknown. We show that BC-derived extracellular vesicles (EVs) suppress pancreatic insulin secretion to impair glucose homeostasis. EV-encapsulated miR-122 targets PKM in ß-cells to suppress glycolysis and ATP-dependent insulin exocytosis. Mice receiving high-miR-122 EVs or bearing BC tumours exhibit suppressed insulin secretion, enhanced endogenous glucose production, impaired glucose tolerance and fasting hyperglycaemia. These effects contribute to tumour growth and are abolished by inhibiting EV secretion or miR-122, restoring PKM in ß-cells or supplementing insulin. Compared with non-cancer controls, patients with BC have higher levels of circulating EV-encapsulated miR-122 and fasting glucose concentrations but lower fasting insulin; miR-122 levels are positively associated with glucose and negatively associated with insulin. Therefore, EV-mediated impairment of whole-body glycaemic control may contribute to tumour progression and incidence of type 2 diabetes in some patients with BC.


Asunto(s)
Neoplasias de la Mama , Diabetes Mellitus Tipo 2 , Vesículas Extracelulares , MicroARNs , Animales , Neoplasias de la Mama/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Vesículas Extracelulares/metabolismo , Femenino , Glucosa/metabolismo , Homeostasis , Humanos , Insulina/metabolismo , Secreción de Insulina , Ratones , MicroARNs/genética , MicroARNs/metabolismo
4.
Nat Metab ; 3(9): 1163-1174, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34489604

RESUMEN

In chronic obesity, hepatocytes become insulin resistant and exert important effects on systemic metabolism. Here we show that in early onset obesity (4 weeks high-fat diet), hepatocytes secrete exosomes that enhance insulin sensitivity both in vitro and in vivo. These beneficial effects were due to exosomal microRNA miR-3075, which is enriched in these hepatocyte exosomes. FA2H is a direct target of miR-3075 and small interfering RNA depletion of FA2H in adipocytes, myocytes and primary hepatocytes leads to increased insulin sensitivity. In chronic obesity (16-18 weeks of a high-fat diet), hepatocyte exosomes promote a state of insulin resistance. These chronic obese hepatocyte exosomes do not directly cause impaired insulin signalling in vitro but do promote proinflammatory activation of macrophages. Taken together, these studies show that in early onset obesity, hepatocytes produce exosomes that express high levels of the insulin-sensitizing miR-3075. In chronic obesity, this compensatory effect is lost and hepatocyte-derived exosomes from chronic obese mice promote insulin resistance.


Asunto(s)
Exosomas/metabolismo , Hepatocitos/metabolismo , Resistencia a la Insulina/genética , Obesidad/metabolismo , Adipocitos/metabolismo , Animales , Dieta Alta en Grasa , Macrófagos/metabolismo , Ratones , Células Musculares/metabolismo , ARN Interferente Pequeño/genética
5.
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
6.
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
7.
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
8.
J Immunol ; 203(12): 3427-3435, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31712385

RESUMEN

Obesity impacts over 30% of the United States population, resulting in a wide array of complications. Included among these is the deterioration of the intestinal barrier, which has been implicated in type 2 diabetes and susceptibility to bacterial transepithelial migration. The intestinal epithelium is maintained by αß and γδ intraepithelial T lymphocytes, which migrate along the epithelia, support epithelial homeostasis, and protect from infection. In this study, we investigate how obesity impacts intraepithelial lymphocyte (IEL) persistence and function in intestinal homeostasis and repair. Mice were fed a high-fat diet to induce obesity and to study immunomodulation in the intestine. There is a striking reduction in αß and γδ IEL persistence as obesity progresses with a different mechanism in αß versus γδ IEL populations. CD4+ and CD4+CD8+ αß intraepithelial T lymphocytes exhibit reduced homeostatic proliferation in obesity, whereas both αß and γδ IELs downregulate CD103 and CCR9. The reduction in intraepithelial T lymphocytes occurs within 7 wk of high-fat diet administration and is not dependent on chronic inflammation via TNF-α. Young mice administered a high-fat diet upon weaning exhibit the most dramatic phenotype, showing that childhood obesity has consequences on intestinal IEL seeding. Together, this dysfunction in the intestinal epithelium renders obese mice more susceptible to dextran sulfate sodium-induced colitis. Diet-induced weight loss restores IEL number and CD103/CCR9 expression and improves outcome in colitis. Together, these data confirm that obesity has immunomodulatory consequences in intestinal tissues that can be improved with weight loss.


Asunto(s)
Colitis/etiología , Colitis/metabolismo , Inmunomodulación , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Factores de Edad , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores , Colitis/patología , Sulfato de Dextran/efectos adversos , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Inmunohistoquímica , Cadenas alfa de Integrinas/genética , Cadenas alfa de Integrinas/metabolismo , Masculino , Ratones , Obesidad/complicaciones , Receptores CCR/genética , Receptores CCR/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Bazo/inmunología , Bazo/metabolismo , Timo/inmunología , Timo/metabolismo
9.
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
10.
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
11.
J Natl Cancer Inst ; 111(1): 52-59, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30202902

RESUMEN

Background: GPR120, a G protein-coupled receptor for long-chain polyunsaturated fatty acids (FAs), mediates the anti-inflammatory effects of omega-3 (ω-3) FAs. We investigated whether host or tumor GPR120 plays a role in the anti-prostate cancer effects of ω-3 FAs. Methods: MycCap prostate cancer allografts were grown in immunocompetent wild-type (WT) and GPR120 knockout (KO) mice fed ω-3 (fish oil) or ω-6 (corn oil) diets. Immune cell infiltration was quantified by flow cytometry, and gene expression of immune cell markers in isolated tumor-associated macrophages (TAMs) was quantified by quantitative real-time polymerase chain reaction. Archived tissue from a fish oil intervention trial was used to correlate gene expression of GPR120 with cell cycle progression (CCP) genes and Ki67 index (n = 11-15 per group). All statistical tests were two-sided. Results: In WT mice (n = 7 per group), dietary ω-3 FAs decreased MycCap allograft tumor growth (mean [SD] final tumor volume ω-6 = 491 [437] mm3 vs ω-3 = 127 [77] mm3, P = .04), whereas in global GPR120KO mice (n = 7 per group) ω-3 FAs had no anticancer effects. Dietary ω-3 FAs inhibited GPR120KO-MycCaP allografts grown in WT mice (n = 8 per group; mean [SD] final tumor volume ω-6 = 776 [767] mm3 vs ω-3 = 36 [34] mm3, P = .02). Omega-3 FA treatment decreased the number of M2-like TAMs in tumor tissue and gene expression of M2 markers in isolated TAMs compared with ω-6 controls in WT (n = 7 per group) but not in GPR120KO mice (n = 7 per group). In human tissue, higher expression of stromal GPR120 correlated with greater reduction in expression of CCP genes in men with prostate cancer on a high-ω-3 diet (r = -.57, P = .04). Conclusions: Host GPR120 plays a central role in the anti-prostate cancer effects of dietary ω-3 FAs. Future studies are required to determine if the anticancer effects of ω-3 FAs are mediated through inhibition of M2-like macrophages and if host GPR120 status predicts anticancer effects of dietary ω-3 FAs in men with prostate cancer.


Asunto(s)
Dieta , Ácidos Grasos Omega-3/antagonistas & inhibidores , Macrófagos/patología , Neoplasias de la Próstata/patología , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Animales , Estudios de Casos y Controles , Progresión de la Enfermedad , Ácidos Grasos Omega-3/administración & dosificación , Estudios de Seguimiento , Humanos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo
12.
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
13.
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
14.
J Clin Invest ; 127(1): 1-4, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28045402

RESUMEN

There are currently over 1.9 billion people who are obese or overweight, leading to a rise in related health complications, including insulin resistance, type 2 diabetes, cardiovascular disease, liver disease, cancer, and neurodegeneration. The finding that obesity and metabolic disorder are accompanied by chronic low-grade inflammation has fundamentally changed our view of the underlying causes and progression of obesity and metabolic syndrome. We now know that an inflammatory program is activated early in adipose expansion and during chronic obesity, permanently skewing the immune system to a proinflammatory phenotype, and we are beginning to delineate the reciprocal influence of obesity and inflammation. Reviews in this series examine the activation of the innate and adaptive immune system in obesity; inflammation within diabetic islets, brain, liver, gut, and muscle; the role of inflammation in fibrosis and angiogenesis; the factors that contribute to the initiation of inflammation; and therapeutic approaches to modulate inflammation in the context of obesity and metabolic syndrome.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Síndrome Metabólico/metabolismo , Obesidad/metabolismo , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Diabetes Mellitus Tipo 2/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Síndrome Metabólico/patología , Neoplasias/metabolismo , Neoplasias/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Obesidad/patología , Especificidad de Órganos
15.
Cell ; 167(4): 973-984.e12, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27814523

RESUMEN

In obesity, macrophages and other immune cells accumulate in insulin target tissues, promoting a chronic inflammatory state and insulin resistance. Galectin-3 (Gal3), a lectin mainly secreted by macrophages, is elevated in both obese subjects and mice. Administration of Gal3 to mice causes insulin resistance and glucose intolerance, whereas inhibition of Gal3, through either genetic or pharmacologic loss of function, improved insulin sensitivity in obese mice. In vitro treatment with Gal3 directly enhanced macrophage chemotaxis, reduced insulin-stimulated glucose uptake in myocytes and 3T3-L1 adipocytes and impaired insulin-mediated suppression of glucose output in primary mouse hepatocytes. Importantly, we found that Gal3 can bind directly to the insulin receptor (IR) and inhibit downstream IR signaling. These observations elucidate a novel role for Gal3 in hepatocyte, adipocyte, and myocyte insulin resistance, suggesting that Gal3 can link inflammation to decreased insulin sensitivity. Inhibition of Gal3 could be a new approach to treat insulin resistance.


Asunto(s)
Galectina 3/sangre , Galectina 3/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Animales , Quimiotaxis , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Galectina 3/antagonistas & inhibidores , Galectina 3/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Insulina/sangre , Resistencia a la Insulina , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Células Musculares/metabolismo , Células Musculares/patología , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología
16.
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
17.
Diabetes ; 64(4): 1120-30, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25315009

RESUMEN

The current dogma is that obesity-associated hepatic inflammation is due to increased Kupffer cell (KC) activation. However, recruited hepatic macrophages (RHMs) were recently shown to represent a sizable liver macrophage population in the context of obesity. Therefore, we assessed whether KCs and RHMs, or both, represent the major liver inflammatory cell type in obesity. We used a combination of in vivo macrophage tracking methodologies and adoptive transfer techniques in which KCs and RHMs are differentially labeled with fluorescent markers. With these approaches, the inflammatory phenotype of these distinct macrophage populations was determined under lean and obese conditions. In vivo macrophage tracking revealed an approximately sixfold higher number of RHMs in obese mice than in lean mice, whereas the number of KCs was comparable. In addition, RHMs comprised smaller size and immature, monocyte-derived cells compared with KCs. Furthermore, RHMs from obese mice were more inflamed and expressed higher levels of tumor necrosis factor-α and interleukin-6 than RHMs from lean mice. A comparison of the MCP-1/C-C chemokine receptor type 2 (CCR2) chemokine system between the two cell types showed that the ligand (MCP-1) is more highly expressed in KCs than in RHMs, whereas CCR2 expression is approximately fivefold greater in RHMs. We conclude that KCs can participate in obesity-induced inflammation by causing the recruitment of RHMs, which are distinct from KCs and are not precursors to KCs. These RHMs then enhance the severity of obesity-induced inflammation and hepatic insulin resistance.


Asunto(s)
Gluconeogénesis/fisiología , Hígado/metabolismo , Macrófagos/metabolismo , Obesidad/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Hígado Graso/metabolismo , Hígado Graso/patología , Interleucina-6/metabolismo , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/patología , Hígado/patología , Macrófagos/patología , Masculino , Ratones , Ratones Obesos , Obesidad/etiología , Obesidad/patología , Receptores CCR2/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
18.
Immunity ; 41(1): 36-48, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25035952

RESUMEN

Chronic, low-grade adipose tissue inflammation is a key etiological mechanism linking the increasing incidence of type 2 diabetes (T2D) and obesity. It is well recognized that the immune system and metabolism are highly integrated, and macrophages, in particular, have been identified as critical effector cells in the initiation of inflammation and insulin resistance. Recent advances have been made in the understanding of macrophage recruitment and retention to adipose tissue and the participation of other immune cell populations in the regulation of this inflammatory process. Here we discuss the pathophysiological link between macrophages, obesity, and insulin resistance, highlighting the dynamic immune cell regulation of adipose tissue inflammation. We also describe the mechanisms by which inflammation causes insulin resistance and the new therapeutic targets that have emerged.


Asunto(s)
Tejido Adiposo/inmunología , Diabetes Mellitus Tipo 2/inmunología , Resistencia a la Insulina/inmunología , Macrófagos/inmunología , Obesidad/inmunología , Tejido Adiposo/citología , Animales , Citocinas/inmunología , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Hígado/citología , Hígado/inmunología , Activación de Macrófagos/inmunología , Ratones , Músculo Esquelético/citología , Músculo Esquelético/inmunología , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Transducción de Señal/inmunología
19.
J Clin Invest ; 124(8): 3566-78, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24983318

RESUMEN

The MAP kinase kinase kinase TGFß-activated kinase 1 (TAK1) is activated by TLRs, IL-1, TNF, and TGFß and in turn activates IKK-NF-κB and JNK, which regulate cell survival, growth, tumorigenesis, and metabolism. TAK1 signaling also upregulates AMPK activity and autophagy. Here, we investigated TAK1-dependent regulation of autophagy, lipid metabolism, and tumorigenesis in the liver. Fasted mice with hepatocyte-specific deletion of Tak1 exhibited severe hepatosteatosis with increased mTORC1 activity and suppression of autophagy compared with their WT counterparts. TAK1-deficient hepatocytes exhibited suppressed AMPK activity and autophagy in response to starvation or metformin treatment; however, ectopic activation of AMPK restored autophagy in these cells. Peroxisome proliferator-activated receptor α (PPARα) target genes and ß-oxidation, which regulate hepatic lipid degradation, were also suppressed in hepatocytes lacking TAK1. Due to suppression of autophagy and ß-oxidation, a high-fat diet challenge aggravated steatohepatitis in mice with hepatocyte-specific deletion of Tak1. Notably, inhibition of mTORC1 restored autophagy and PPARα target gene expression in TAK1-deficient livers, indicating that TAK1 acts upstream of mTORC1. mTORC1 inhibition also suppressed spontaneous liver fibrosis and hepatocarcinogenesis in animals with hepatocyte-specific deletion of Tak1. These data indicate that TAK1 regulates hepatic lipid metabolism and tumorigenesis via the AMPK/mTORC1 axis, affecting both autophagy and PPARα activity.


Asunto(s)
Autofagia/fisiología , Ácidos Grasos/metabolismo , Hígado Graso/prevención & control , Neoplasias Hepáticas Experimentales/prevención & control , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia/efectos de los fármacos , Hígado Graso/genética , Hígado Graso/metabolismo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Quinasas Quinasa Quinasa PAM/deficiencia , Quinasas Quinasa Quinasa PAM/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Oxidación-Reducción , PPAR alfa/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
20.
Nat Med ; 20(8): 942-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24997608

RESUMEN

It is well known that the ω-3 fatty acids (ω-3-FAs; also known as n-3 fatty acids) can exert potent anti-inflammatory effects. Commonly consumed as fish products, dietary supplements and pharmaceuticals, ω-3-FAs have a number of health benefits ascribed to them, including reduced plasma triglyceride levels, amelioration of atherosclerosis and increased insulin sensitivity. We reported that Gpr120 is the functional receptor for these fatty acids and that ω-3-FAs produce robust anti-inflammatory, insulin-sensitizing effects, both in vivo and in vitro, in a Gpr120-dependent manner. Indeed, genetic variants that predispose to obesity and diabetes have been described in the gene encoding GPR120 in humans (FFAR4). However, the amount of fish oils that would have to be consumed to sustain chronic agonism of Gpr120 is too high to be practical, and, thus, a high-affinity small-molecule Gpr120 agonist would be of potential clinical benefit. Accordingly, Gpr120 is a widely studied drug discovery target within the pharmaceutical industry. Gpr40 is another lipid-sensing G protein-coupled receptor, and it has been difficult to identify compounds with a high degree of selectivity for Gpr120 over Gpr40 (ref. 11). Here we report that a selective high-affinity, orally available, small-molecule Gpr120 agonist (cpdA) exerts potent anti-inflammatory effects on macrophages in vitro and in obese mice in vivo. Gpr120 agonist treatment of high-fat diet-fed obese mice causes improved glucose tolerance, decreased hyperinsulinemia, increased insulin sensitivity and decreased hepatic steatosis. This suggests that Gpr120 agonists could become new insulin-sensitizing drugs for the treatment of type 2 diabetes and other human insulin-resistant states in the future.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ácidos Grasos Omega-3/metabolismo , Resistencia a la Insulina/fisiología , Receptores Acoplados a Proteínas G/agonistas , Animales , Arginasa/biosíntesis , Linfocitos B Reguladores/inmunología , Secuencia de Bases , Diabetes Mellitus Tipo 2/genética , Ácidos Docosahexaenoicos/farmacología , Hígado Graso/tratamiento farmacológico , Hiperinsulinismo/tratamiento farmacológico , Inflamación , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Datos de Secuencia Molecular , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Obesidad/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA