Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunometabolism (Cobham) ; 4(4): e00008, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36337734

RESUMEN

An increase in glucose uptake driving aerobic glycolysis is a robust hallmark of immune cell activation. The glycolytic response supports functional alterations of the innate immune cells including the production and release of cytokines. Large inter-individual differences in the magnitude of this cytokine response are known to exist. In addition, the presence of disease is known to impact on immune cell function. Whether variation in metabolic responses of immune cells exist between individuals during health or disease is currently unknown. Here, we explore inter-individual differences in the glycolytic rate of immune cells using lactate production as readout upon activation using a variety of different stimuli. Glycolytic responses are subsequently associated to functional immune cell responses in healthy humans. In addition, we determined the glycolytic rate of immune cells and its association with immune function using patients diagnosed with diabetes mellitus. Based on the relative increase in lactate production after activation, distinct clusters of low, intermediate, and high responders could be identified, illustrating the existence of variation in glycolytic responses in healthy subjects. Interestingly, the production of cytokines mirrored these high-, intermediate-, and low-lactate patterns after pathogenic stimulation. In patients with diabetes mellitus, a reduced correlation was found between lactate and cytokine production, specifically for IL-6. Furthermore, based on the relative increase in lactate production, variability in the glycolytic response was reduced compared to healthy subjects. In conclusion, our results show a specific association between the glycolytic rate and function in human immune cells after stimulation with different pathogens. In addition to demonstrating the existence of glycolytic variability and specificity depending on the type of stimulus, the association between glycolysis and function in innate immune cells is altered during the presence of diabetes.

2.
Diab Vasc Dis Res ; 17(1): 1479164119892140, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31868000

RESUMEN

BACKGROUND: C-type lectin receptors, including Dectin-2, are pattern recognition receptors on monocytes and macrophages that mainly recognize sugars and sugar-like structures present on fungi. Activation of C-type lectin receptors induces downstream CARD9 signalling, leading to the production of cytokines. We hypothesized that under hyperglycaemic conditions, as is the case in diabetes mellitus, glycosylated protein (sugar-like) structures activate C-type lectin receptors, leading to immune cell activation and increased atherosclerosis development. METHODS: Low-density lipoprotein receptor-deficient mice were lethally irradiated and transplanted with bone marrow from control wild-type, Dectin-2-/- or Card9-/- mice. After 6 weeks of recovery, mice received streptozotocin injections (50 mg/g BW; 5 days) to induce hyperglycaemia. After an additional 2 weeks, mice were fed a Western-type diet (0.1% cholesterol) for 10 weeks. RESULTS AND CONCLUSION: Deletion of haematopoietic Dectin-2 reduced the number of circulating Ly6Chi monocytes, increased pro-inflammatory cytokine production, but did not affect atherosclerosis development. Deletion of haematopoietic CARD9 tended to reduce macrophage and collagen content in atherosclerotic lesions, again without influencing the lesion size. Deletion of haematopoietic Dectin-2 did not influence atherosclerosis development under hyperglycaemic conditions, despite some minor effects on inflammation. Deletion of haematopoietic CARD9 induced minor alterations in plaque composition under hyperglycaemic conditions, without affecting lesion size.


Asunto(s)
Enfermedades de la Aorta/etiología , Aterosclerosis/etiología , Glucemia/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Diabetes Mellitus Experimental/complicaciones , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Lectinas Tipo C/genética , Animales , Antígenos Ly/metabolismo , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores/sangre , Trasplante de Médula Ósea , Proteínas Adaptadoras de Señalización CARD/deficiencia , Células Cultivadas , Colágeno/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/sangre , Dieta Occidental , Predisposición Genética a la Enfermedad , Lectinas Tipo C/deficiencia , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/patología , Placa Aterosclerótica , Receptores de LDL/deficiencia , Receptores de LDL/genética
3.
Sci Rep ; 9(1): 4337, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30867470

RESUMEN

Inflammatory reactions activated by pattern recognition receptors (PRRs) on the membrane of innate immune cells play an important role in atherosclerosis. Whether the PRRs of the C-type lectin receptor (CLR) family including Dectin-2 may be involved in the pathogenesis of atherosclerosis remains largely unknown. Recently, the CLR-adaptor molecule caspase recruitment domain family member 9 (CARD9) has been suggested to play a role in cardiovascular pathologies as it provides the link between CLR activation and transcription of inflammatory cytokines as well as immune cell recruitment. We therefore evaluated whether hematopoietic deletion of Dectin-2 or CARD9 reduces inflammation and atherosclerosis development. Low-density lipoprotein receptor (Ldlr)-knockout mice were transplanted with bone marrow from wild-type, Dectin-2- or Card9-knockout mice and fed a Western-type diet containing 0.1% (w/w) cholesterol. After 10 weeks, lipid and inflammatory parameters were measured and atherosclerosis development was determined. Deletion of hematopoietic Dectin-2 or CARD9 did not influence plasma triglyceride and cholesterol levels. Deletion of hematopoietic Dectin-2 did not affect atherosclerotic lesion area, immune cell composition, ex vivo cytokine secretion by peritoneal cells or bone marrow derived macrophages. Unexpectedly, deletion of hematopoietic CARD9 increased atherosclerotic lesion formation and lesion severity. Deletion of hematopoietic CARD9 did also not influence circulating immune cell composition and peripheral cytokine secretion. Besides a tendency to a reduced macrophage content within these lesions, plasma MCP-1 levels decreased upon WTD feeding. Deletion of hematopoietic Dectin-2 did not influence atherosclerosis development in hyperlipidemic mice. The absence of CARD9 unexpectedly increased atherosclerotic lesion size and severity, suggesting that the presence of CARD9 may protect against initiation of atherosclerosis development.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Sistema Hematopoyético/metabolismo , Hiperlipidemias/patología , Lectinas Tipo C/genética , Placa Aterosclerótica/prevención & control , Animales , Hiperlipidemias/sangre , Ratones , Ratones Noqueados , Placa Aterosclerótica/patología
4.
Front Immunol ; 9: 2564, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30483253

RESUMEN

Lactate, the end product of anaerobic glycolysis, is produced in high amounts by innate immune cells during inflammatory activation. Although immunomodulating effects of lactate have been reported, evidence from human studies is scarce. Here we show that expression of genes involved in lactate metabolism and transport is modulated in human immune cells during infection and upon inflammatory activation with TLR ligands in vitro, indicating an important role for lactate metabolism in inflammation. Extracellular lactate induces metabolic reprogramming in innate immune cells, as evidenced by reduced glycolytic and increased oxidative rates of monocytes immediately after exposure to lactate. A short-term infusion of lactate in humans in vivo increased ex vivo glucose consumption of PBMCs, but effects on metabolic rates and cytokine production were limited. Interestingly, long-term treatment with lactate ex vivo, reflecting pathophysiological conditions in local microenvironments such as tumor or adipose tissue, significantly modulated cytokine production with predominantly anti-inflammatory effects. We found time- and stimuli-dependent effects of extracellular lactate on cytokine production, further emphasizing the complex interplay between metabolism and immune cell function. Together, our findings reveal lactate as a modulator of immune cell metabolism which translates to reduced inflammation and may ultimately function as a negative feedback signal to prevent excessive inflammatory responses.


Asunto(s)
Tejido Adiposo/fisiología , Anaerobiosis/genética , Glucólisis/genética , Ácido Láctico/metabolismo , Leucocitos Mononucleares/metabolismo , Monocitos/metabolismo , Células Cultivadas , Microambiente Celular , Citocinas/metabolismo , Humanos , Inmunidad Innata/genética , Inmunomodulación , Inflamación/genética , Leucocitos Mononucleares/inmunología , Monocitos/inmunología , Oxidación-Reducción , Cultivo Primario de Células
5.
Cytokine ; 108: 205-212, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29698883

RESUMEN

Toll like receptors (TLRs) are expressed in adipose tissue and promote adipose tissue inflammation during obesity. Recently, anti-inflammatory properties have been attributed to TLR10 in myeloid cells, the only member of the TLR family with inhibitory activity. In order to assess whether TLR10-induced inhibition of inflammation may be protective during the development of obesity and metabolic abnormalities we used transgenic human TLR10 mice (hTLR10tg) and wild type (WT) controls on a C57B6J background. HFD-feeding enhanced TLR10 expression in the adipose tissue, and HFD-fed hTLR10tg mice displayed reduced adipocyte size, adipose tissue weight, and a trend toward lower plasma insulin levels compared to WT mice. In humans, obese individuals with polymorphisms in the TLR10 gene displayed reduced macrophage infiltration in the adipose tissue accompanied by a trend to lower leptin levels and higher adiponectin levels in plasma. In healthy individuals with the same polymorphisms in the TLR10 gene we did not observe any difference in plasma concentrations of leptin and adiponectin. We conclude that TLR10 impacts adipose tissue morphology in obesity. Larger studies in humans are warranted to assess its potential value as therapeutic target in metabolic syndrome and type 2 diabetes.


Asunto(s)
Tejido Adiposo/patología , Leptina/sangre , Obesidad/metabolismo , Receptor Toll-Like 10/metabolismo , Adipocitos/citología , Adipoquinas/sangre , Adiponectina/sangre , Animales , Biopsia , Estudios de Cohortes , Técnicas de Sustitución del Gen , Humanos , Inflamación , Macrófagos/inmunología , Masculino , Ratones Transgénicos , Adhesión en Parafina , Polimorfismo de Nucleótido Simple , Receptor Toll-Like 10/genética , Regulación hacia Arriba
6.
Diabetologia ; 60(7): 1304-1313, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28382382

RESUMEN

AIMS/HYPOTHESIS: Obesity induces macrophages to drive inflammation in adipose tissue, a crucial step towards the development of type 2 diabetes. The tricarboxylic acid (TCA) cycle intermediate succinate is released from cells under metabolic stress and has recently emerged as a metabolic signal induced by proinflammatory stimuli. We therefore investigated whether succinate receptor 1 (SUCNR1) could play a role in the development of adipose tissue inflammation and type 2 diabetes. METHODS: Succinate levels were determined in human plasma samples from individuals with type 2 diabetes and non-diabetic participants. Succinate release from adipose tissue explants was studied. Sucnr1 -/- and wild-type (WT) littermate mice were fed a high-fat diet (HFD) or low-fat diet (LFD) for 16 weeks. Serum metabolic variables, adipose tissue inflammation, macrophage migration and glucose tolerance were determined. RESULTS: We show that hypoxia and hyperglycaemia independently drive the release of succinate from mouse adipose tissue (17-fold and up to 18-fold, respectively) and that plasma levels of succinate were higher in participants with type 2 diabetes compared with non-diabetic individuals (+53%; p < 0.01). Sucnr1 -/- mice had significantly reduced numbers of macrophages (0.56 ± 0.07 vs 0.92 ± 0.15 F4/80 cells/adipocytes, p < 0.05) and crown-like structures (0.06 ± 0.02 vs 0.14 ± 0.02, CLS/adipocytes p < 0.01) in adipose tissue and significantly improved glucose tolerance (p < 0.001) compared with WT mice fed an HFD, despite similarly increased body weights. Consistently, macrophages from Sucnr1 -/- mice showed reduced chemotaxis towards medium collected from apoptotic and hypoxic adipocytes (-59%; p < 0.05). CONCLUSIONS/INTERPRETATION: Our results reveal that activation of SUCNR1 in macrophages is important for both infiltration and inflammation of adipose tissue in obesity, and suggest that SUCNR1 is a promising therapeutic target in obesity-induced type 2 diabetes. DATA AVAILABILITY: The dataset generated and analysed during the current study is available in GEO with the accession number GSE64104, www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE64104 .


Asunto(s)
Diabetes Mellitus/metabolismo , Inflamación/metabolismo , Macrófagos/citología , Obesidad/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adulto , Anciano , Animales , Glucemia/metabolismo , Peso Corporal , Movimiento Celular , Quimiotaxis , Ciclo del Ácido Cítrico , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Prueba de Tolerancia a la Glucosa , Humanos , Hiperglucemia/metabolismo , Hipoxia , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Receptores Acoplados a Proteínas G/genética , Transducción de Señal
7.
Diabetes ; 66(4): 1052-1061, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28115398

RESUMEN

Severe hypoglycemic events have been associated with increased cardiovascular mortality in patients with diabetes, which may be explained by hypoglycemia-induced inflammation. We used ex vivo stimulations of peripheral blood mononuclear cells (PBMCs) and monocytes obtained during hyperinsulinemic-euglycemic (5.0 mmol/L)-hypoglycemic (2.6 mmol/L) clamps in 11 healthy participants, 10 patients with type 1 diabetes and normal awareness of hypoglycemia (NAH), and 10 patients with type 1 diabetes and impaired awareness (IAH) to test whether the composition and inflammatory function of immune cells adapt to a more proinflammatory state after hypoglycemia. Hypoglycemia increased leukocyte numbers in healthy control participants and patients with NAH but not in patients with IAH. Leukocytosis strongly correlated with the adrenaline response to hypoglycemia. Ex vivo, PBMCs and monocytes displayed a more robust cytokine response to microbial stimulation after hypoglycemia compared with euglycemia, although it was less pronounced in patients with IAH. Of note, hypoglycemia increased the expression of markers of demargination and inflammation in PBMCs. We conclude that hypoglycemia promotes mobilization of specific leukocyte subsets from the marginal pool and induces proinflammatory functional changes in immune cells. Inflammatory responses were less pronounced in IAH, indicating that counterregulatory hormone responses are key modulators of hypoglycemia-induced proinflammatory effects. Hypoglycemia-induced proinflammatory changes may promote a sustained inflammatory state.


Asunto(s)
Diabetes Mellitus Tipo 1 , Epinefrina/metabolismo , Hipoglucemia/inmunología , Leucocitosis/inmunología , Monocitos/inmunología , ARN Mensajero/metabolismo , Adulto , Concienciación , Estudios de Casos y Controles , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/inmunología , Citocinas/efectos de los fármacos , Citocinas/inmunología , Femenino , Expresión Génica , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Humanos , Hipoglucemia/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/inmunología , Ácido Láctico/metabolismo , Lipopolisacáridos/farmacología , Masculino , Monocitos/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología , Adulto Joven
8.
Nat Microbiol ; 2: 16246, 2016 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-27991883

RESUMEN

Microbial stimuli such as lipopolysaccharide (LPS) induce robust metabolic rewiring in immune cells known as the Warburg effect. It is unknown whether this increase in glycolysis and decrease in oxidative phosphorylation (OXPHOS) is a general characteristic of monocytes that have encountered a pathogen. Using CD14+ monocytes from healthy donors, we demonstrated that most microbial stimuli increased glycolysis, but that only stimulation of Toll-like receptor (TLR) 4 with LPS led to a decrease in OXPHOS. Instead, activation of other TLRs, such as TLR2 activation by Pam3CysSK4 (P3C), increased oxygen consumption and mitochondrial enzyme activity. Transcriptome and metabolome analysis of monocytes stimulated with P3C versus LPS confirmed the divergent metabolic responses between both stimuli, and revealed significant differences in the tricarboxylic acid cycle, OXPHOS and lipid metabolism pathways following stimulation of monocytes with P3C versus LPS. At a functional level, pharmacological inhibition of complex I of the mitochondrial electron transport chain diminished cytokine production and phagocytosis in P3C- but not LPS-stimulated monocytes. Thus, unlike LPS, complex microbial stimuli and the TLR2 ligand P3C induce a specific pattern of metabolic rewiring that involves upregulation of both glycolysis and OXPHOS, which enables activation of host defence mechanisms such as cytokine production and phagocytosis.

9.
Mol Med ; 222016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27261776

RESUMEN

Activation of inflammatory pathways is known to accompany development of obesity-induced non-alcoholic fatty liver disease (NAFLD), insulin resistance and type 2 diabetes. In addition to caspase-1, the neutrophil serine proteases proteinase 3, neutrophil elastase and cathepsin G are able to process the inactive pro-inflammatory mediators IL-1ß and IL-18 to their bioactive forms, thereby regulating inflammatory responses. In the present study, we investigated whether proteinase 3 is involved in obesity-induced development of insulin resistance and NAFLD. We investigated the development of NAFLD and insulin resistance in mice deficient for neutrophil elastase/proteinase 3 and neutrophil elastase/cathepsin G and in wild-type mice treated with the neutrophil serine proteinase inhibitor human alpha-1 antitrypsin. Expression profiling of metabolically relevant tissues obtained from insulin resistant mice showed that expression of proteinase 3 was specifically upregulated in the liver, whereas neutrophil elastase, cathepsin G and caspase-1 were not. Neutrophil elastase/proteinase 3 deficient mice showed strongly reduced levels of lipids in the liver after fed a high fat diet. Moreover, these mice were resistant to high fat diet-induced weight gain, inflammation and insulin resistance. Injection of proteinase 3 exacerbated insulin resistance in caspase-1(-/-) mice, indicating that proteinase 3 acts independently of caspase-1. Treatment with alpha-1 antitrypsin during the last 10 days of a 16 week high fat diet reduced hepatic lipid content and decreased fasting glucose levels. We conclude that proteinase 3 is involved in NAFLD and insulin resistance and that inhibition of proteinase 3 may have therapeutic potential.


Asunto(s)
Resistencia a la Insulina/genética , Hígado/metabolismo , Mieloblastina/genética , Mieloblastina/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Elastasa de Leucocito/genética , Masculino , Ratones , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/inducido químicamente , Obesidad/genética , Regulación hacia Arriba
10.
Sci Rep ; 6: 21906, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26932716

RESUMEN

Vanins are enzymes that convert pantetheine to pantothenic acid (vitamin B5). Insights into the function of vanins have evolved lately, indicating vanin-1 to play a role in inflammation, oxidative stress and cell migration. Moreover, vanin-1 has recently gained attention as a novel modulator of hepatic glucose and lipid metabolism. In the present study, we investigated the role of vanin-1 in the development of hepatic steatosis and insulin resistance in animal models of obesity and diabetes. In addition, we evaluated the potency of RR6, a novel pharmacological vanin-1 inhibitor, as an anti-diabetic drug. Increased vanin activity was observed in plasma and liver of high fat diet (HFD)-induced obese mice, as well as ZDF-diabetic rats. Ablation of vanin-1 (Vnn1(-/-) mice) mildly improved glucose tolerance and insulin sensitivity in HFD-fed mice, but had no effects on body weight, hepatic steatosis or circulating lipid levels. Oral administration of RR6 for 8 days completely inhibited plasma vanin activity, but did not affect hepatic glucose production, insulin sensitivity or hepatic steatosis in ZDF-diabetes rats. In conclusion, absence of vanin-1 activity improves insulin sensitivity in HFD-fed animals, yet short-term inhibition of vanin activity may have limited value as an anti-diabetic strategy.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/genética , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Tipo 2/enzimología , Inhibidores Enzimáticos/farmacología , Amidohidrolasas/metabolismo , Animales , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Insulina/farmacología , Resistencia a la Insulina , Lípidos/sangre , Masculino , Ratones , Ratones Noqueados , Obesidad/sangre , Obesidad/enzimología , Obesidad/patología , Ratas Wistar , Ratas Zucker
11.
PLoS One ; 10(4): e0123152, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25867514

RESUMEN

Innate immunity plays a pivotal role in obesity-induced low-grade inflammation originating from adipose tissue. Key receptors of the innate immune system including Toll-like receptors-2 and -4 (TLRs) are triggered by nutrient excess to promote inflammation. The role of other TLRs in this process is largely unknown. In addition to double-stranded viral mRNA, TLR-3 can also recognize mRNA from dying endogenous cells, a process that is frequently observed within obese adipose tissue. Here, we identified profound expression of TLR-3 in adipocytes and investigated its role during diet-induced obesity. Human adipose tissue biopsies (n=80) and an adipocyte cell-line were used to study TLR-3 expression and function. TLR-3-/- and WT animals were exposed to a high-fat diet (HFD) for 16 weeks to induce obesity. Expression of TLR-3 was significantly higher in human adipocytes compared to the non-adipocyte cells part of the adipose tissue. In vitro, TLR-3 expression was induced during differentiation of adipocytes and stimulation of the receptor led to elevated expression of pro-inflammatory cytokines. In vivo, TLR-3 deficiency did not significantly influence HFD-induced obesity, insulin sensitivity or inflammation. In humans, TLR-3 expression in adipose tissue did not correlate with BMI or insulin sensitivity (HOMA-IR). Together, our results demonstrate that TLR-3 is highly expressed in adipocytes and functionally active. However, TLR-3 appears to play a redundant role in obesity-induced inflammation and insulin resistance.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/patología , Inflamación/complicaciones , Obesidad/complicaciones , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Adulto , Anciano , Animales , Dieta Alta en Grasa , Femenino , Humanos , Resistencia a la Insulina , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Receptor Toll-Like 3/genética , Aumento de Peso
13.
Nat Commun ; 5: 4711, 2014 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-25182023

RESUMEN

Cytokines of the IL-1 family are important modulators of obesity-induced inflammation and the development of systemic insulin resistance. Here we show that IL-1 family member IL-37, recently characterized as an anti-inflammatory cytokine, ameliorates obesity-induced inflammation and insulin resistance. Mice transgenic for human IL-37 (IL-37tg) exhibit reduced numbers of adipose tissue macrophages, increased circulating levels of adiponectin and preserved glucose tolerance and insulin sensitivity after 16 weeks of HFD. In vitro treatment of adipocytes with recombinant IL-37 reduces adipogenesis and activates AMPK signalling. In humans, elevated steady-state IL-37 adipose tissue mRNA levels are positively correlated with insulin sensitivity and a lower inflammatory status of the adipose tissue. These findings reveal IL-37 as an important anti-inflammatory modulator during obesity-induced inflammation and insulin resistance in both mice and humans, and suggest that IL-37 is a potential target for the treatment of obesity-induced insulin resistance and type 2 diabetes.


Asunto(s)
Adipocitos/efectos de los fármacos , Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina , Interleucina-1/metabolismo , Obesidad/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Adiponectina/sangre , Adiponectina/genética , Tejido Adiposo/patología , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa , Grasas de la Dieta/efectos adversos , Expresión Génica , Prueba de Tolerancia a la Glucosa , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Insulina/sangre , Insulina/genética , Interleucina-1/genética , Interleucina-1/farmacología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Transgénicos , Obesidad/etiología , Obesidad/genética , Obesidad/patología , Cultivo Primario de Células
14.
J Hepatol ; 61(2): 366-72, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24751833

RESUMEN

BACKGROUND & AIMS: Peroxisome proliferator-activated receptor alpha (PPARα) is a key regulator of hepatic fat oxidation that serves as an energy source during starvation. Vanin-1 has been described as a putative PPARα target gene in liver, but its function in hepatic lipid metabolism is unknown. METHODS: We investigated the regulation of vanin-1, and total vanin activity, by PPARα in mice and humans. Furthermore, the function of vanin-1 in the development of hepatic steatosis in response to starvation was examined in Vnn1 deficient mice, and in rats treated with an inhibitor of vanin activity. RESULTS: Liver microarray analyses reveals that Vnn1 is the most prominently regulated gene after modulation of PPARα activity. In addition, activation of mouse PPARα regulates hepatic- and plasma vanin activity. In humans, consistent with regulation by PPARα, plasma vanin activity increases in all subjects after prolonged fasting, as well as after treatment with the PPARα agonist fenofibrate. In mice, absence of vanin-1 exacerbates the fasting-induced increase in hepatic triglyceride levels. Similarly, inhibition of vanin activity in rats induces accumulation of hepatic triglycerides upon fasting. Microarray analysis reveal that the absence of vanin-1 associates with gene sets involved in liver steatosis, and reduces pathways involved in oxidative stress and inflammation. CONCLUSIONS: We show that hepatic vanin-1 is under extremely sensitive regulation by PPARα and that plasma vanin activity could serve as a readout of changes in PPARα activity in human subjects. In addition, our data propose a role for vanin-1 in regulation of hepatic TG levels during fasting.


Asunto(s)
Amidohidrolasas/fisiología , Metabolismo de los Lípidos , Hígado/metabolismo , PPAR alfa/fisiología , Animales , Hígado Graso/etiología , Proteínas Ligadas a GPI/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Inanición/metabolismo
15.
PLoS One ; 9(2): e89615, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586913

RESUMEN

Chronic low-grade inflammation in adipose tissue often accompanies obesity, leading to insulin resistance and increasing the risk for metabolic diseases. MAP3K8 (TPL2/COT) is an important signal transductor and activator of pro-inflammatory pathways that has been linked to obesity-induced adipose tissue inflammation. We used human adipose tissue biopsies to study the relationship of MAP3K8 expression with markers of obesity and expression of pro-inflammatory cytokines (IL-1ß, IL-6 and IL-8). Moreover, we evaluated obesity-induced adipose tissue inflammation and insulin resistance in mice lacking MAP3K8 and WT mice on a high-fat diet (HFD) for 16 weeks. Individuals with a BMI >30 displayed a higher mRNA expression of MAP3K8 in adipose tissue compared to individuals with a normal BMI. Additionally, high mRNA expression levels of IL-1ß, IL-6 and IL-8, but not TNF -α, in human adipose tissue were associated with higher expression of MAP3K8. Moreover, high plasma SAA and CRP did not associate with increased MAP3K8 expression in adipose tissue. Similarly, no association was found for MAP3K8 expression with plasma insulin or glucose levels. Mice lacking MAP3K8 had similar bodyweight gain as WT mice, yet displayed lower mRNA expression levels of IL-1ß, IL-6 and CXCL1 in adipose tissue in response to the HFD as compared to WT animals. However, MAP3K8 deficient mice were not protected against HFD-induced adipose tissue macrophage infiltration or the development of insulin resistance. Together, the data in both human and mouse show that MAP3K8 is involved in local adipose tissue inflammation, specifically for IL-1ß and its responsive cytokines IL-6 and IL-8, but does not seem to have systemic effects on insulin resistance.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/patología , Proteínas Sanguíneas/análisis , Inflamación/etiología , Quinasas Quinasa Quinasa PAM/metabolismo , Quinasas Quinasa Quinasa PAM/fisiología , Obesidad/complicaciones , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Tejido Adiposo/metabolismo , Animales , Western Blotting , Índice de Masa Corporal , Células Cultivadas , Dieta Alta en Grasa , Prueba de Tolerancia a la Glucosa , Humanos , Técnicas para Inmunoenzimas , Inflamación/metabolismo , Inflamación/patología , Insulina/metabolismo , Resistencia a la Insulina , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Obesidad/patología , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
16.
Cytokine ; 62(1): 96-103, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23478176

RESUMEN

Obesity-induced inflammation is associated with insulin resistance and morphologically characterized by macrophage influx into the adipose tissue. Recently, various other immune cells, including B- and T-cells, have been shown to participate in modulating adipose tissue inflammation during the development of obesity. We show that HFD-feeding modulates the influx of B and T-cells into adipose tissue of obese animals, suggestive of a role of the adaptive immune system in the development of adipose tissue inflammation. Despite a lower bodyweight after HFD-feeding, gene expression levels of CD68, F4/80 and MCP-1 in white adipose tissue were enhanced in SCID animals that lack B- and T-cells. Moreover, conditioned medium from adipose tissue explants of HFD-fed SCID mice revealed increased release of IL-6 and CXCL1 compared to WT animals. Compared to WT mice, glucose tolerance was impaired in B- and T-cell deficient mice after HFD-feeding. Thus, complete B- and T-cell deficiency does not protect against HFD-induced adipose tissue inflammation and glucose intolerance. In contrast, SCID mice showed an increased pro-inflammatory status at the level of the adipose tissue in some cytokines. Our findings suggest that a delicate balance between various B- and T-cell populations controls adipose tissue inflammation.


Asunto(s)
Linfocitos B/metabolismo , Intolerancia a la Glucosa/complicaciones , Inflamación/complicaciones , Depleción Linfocítica , Obesidad/complicaciones , Obesidad/prevención & control , Linfocitos T/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Peso Corporal , Dieta Alta en Grasa , Epidídimo/patología , Conducta Alimentaria , Intolerancia a la Glucosa/patología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID
17.
Proc Natl Acad Sci U S A ; 108(37): 15324-9, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21876127

RESUMEN

Inflammation plays a key role in the pathogenesis of obesity. Chronic overfeeding leads to macrophage infiltration in the adipose tissue, resulting in proinflammatory cytokine production. Both microbial and endogenous danger signals trigger assembly of the intracellular innate immune sensor Nlrp3, resulting in caspase-1 activation and production of proinflammatory cytokines IL-1ß and IL-18. Here, we showed that mice deficient in Nlrp3, apoptosis-associated speck-like protein, and caspase-1 were resistant to the development of high-fat diet-induced obesity, which correlated with protection from obesity-induced insulin resistance. Furthermore, hepatic triglyceride content, adipocyte size, and macrophage infiltration in adipose tissue were all reduced in mice deficient in inflammasome components. Monocyte chemoattractant protein (MCP)-1 is a key molecule that mediates macrophage infiltration. Indeed, defective inflammasome activation was associated with reduced MCP-1 production in adipose tissue. Furthermore, plasma leptin and resistin that affect energy use and insulin sensitivity were also changed by inflammasome-deficiency. Detailed metabolic and molecular phenotyping demonstrated that the inflammasome controls energy expenditure and adipogenic gene expression during chronic overfeeding. These findings reveal a critical function of the inflammasome in obesity and insulin resistance, and suggest inhibition of the inflammasome as a potential therapeutic strategy.


Asunto(s)
Inflamasomas/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo/patología , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Movimiento Celular/efectos de los fármacos , Colesterol/sangre , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/metabolismo , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Metabolismo Energético/efectos de los fármacos , Hígado Graso/complicaciones , Hígado Graso/patología , Hígado Graso/prevención & control , Técnica de Clampeo de la Glucosa , Humanos , Hiperinsulinismo/complicaciones , Hiperinsulinismo/patología , Hipertrofia , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Obesidad/sangre , Obesidad/complicaciones , Triglicéridos/sangre
18.
Endocrinology ; 152(10): 3769-78, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21862623

RESUMEN

The immune competent abdominal adipose tissue, either stored viscerally [visceral adipose tissue (VAT)] or sc [sc adipose tissue (SAT)], has been identified as a source of IL-1ß and IL-18. To become active, the proforms of these cytokines require processing by caspase-1, which itself is mediated by the inflammasome. In this descriptive study, we investigate the expression of inflammasome components and caspase-1 in human fat and determine whether caspase-1 activity contributes to the enhanced inflammatory status of VAT. Paired SAT and VAT biopsies from 10 overweight subjects (body mass index, 25-28 kg/m(2)) were used to study the cellular composition and the intrinsic inflammatory capacity of both adipose tissue depots. The percentage of CD8(+) T cells within the lymphocyte fraction was significantly higher in VAT compared with SAT (41.6 vs. 30.4%; P < 0.05). Adipose tissue cultures showed a higher release of IL-1ß (10-fold; P < 0.05), IL-18 (3-fold; P < 0.05), and IL-6 and IL-8 (3-fold, P < 0.05; and 4-fold, P < 0.05, respectively) from VAT compared with SAT that was significantly reduced by inhibiting caspase-1 activity. In addition, caspase-1 activity was 3-fold (P < 0.05) higher in VAT compared with SAT, together with an increase in the protein levels of the inflammasome members apoptosis-associated speck-like protein containing a C-terminal caspase-recruitment domain (2-fold; P < 0.05) and nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 (2-fold; nonsignificant). Finally, caspase-1 activity levels were positively correlated with the percentage of CD8(+) T cells present in adipose tissue. Our results show that caspase-1 and nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 inflammasome members are abundantly present in human VAT. The increased intrinsic caspase-1 activity in VAT represents a novel and specific inflammatory pathway that may determine the proinflammatory character of this specific depot.


Asunto(s)
Caspasa 1/fisiología , Inflamasomas/fisiología , Inflamación/etiología , Grasa Intraabdominal/inmunología , Adulto , Linfocitos T CD8-positivos/inmunología , Activación Enzimática , Femenino , Humanos , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Grasa Intraabdominal/enzimología , Masculino , Persona de Mediana Edad , Grasa Subcutánea/inmunología
19.
Cell Metab ; 12(6): 593-605, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21109192

RESUMEN

Obesity-induced inflammation originating from expanding adipose tissue interferes with insulin sensitivity. Important metabolic effects have been recently attributed to IL-1ß and IL-18, two members of the IL-1 family of cytokines. Processing of IL-1ß and IL-18 requires cleavage by caspase-1, a cysteine protease regulated by a protein complex called the inflammasome. We demonstrate that the inflammasome/caspase-1 governs adipocyte differentiation and insulin sensitivity. Caspase-1 is upregulated during adipocyte differentiation and directs adipocytes toward a more insulin-resistant phenotype. Treatment of differentiating adipocytes with recombinant IL-1ß and IL-18, or blocking their effects by inhibitors, reveals that the effects of caspase-1 on adipocyte differentiation are largely conveyed by IL-1ß. Caspase-1 and IL-1ß activity in adipose tissue is increased both in diet-induced and genetically induced obese animal models. Conversely, mice deficient in caspase-1 are more insulin sensitive as compared to wild-type animals. In addition, differentiation of preadipocytes isolated from caspase-1(-/-) or NLRP3(-/-) mice resulted in more metabolically active fat cells. In vivo, treatment of obese mice with a caspase-1 inhibitor significantly increases their insulin sensitivity. Indirect calorimetry analysis revealed higher fat oxidation rates in caspase-1(-/-) animals. In conclusion, the inflammasome is an important regulator of adipocyte function and insulin sensitivity, and caspase-1 inhibition may represent a novel therapeutic target in clinical conditions associated with obesity and insulin resistance.


Asunto(s)
Adipocitos/fisiología , Caspasa 1/metabolismo , Diferenciación Celular/fisiología , Inflamasomas/metabolismo , Resistencia a la Insulina/fisiología , Obesidad/metabolismo , Adipocitos/metabolismo , Animales , Calorimetría Indirecta , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/genética , Activación Enzimática/fisiología , Técnicas Histológicas , Immunoblotting , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Reacción en Cadena de la Polimerasa
20.
Proc Natl Acad Sci U S A ; 104(42): 16645-50, 2007 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-17925445

RESUMEN

Infectious diseases exert a constant evolutionary pressure on the genetic makeup of our innate immune system. Polymorphisms in Toll-like receptor 4 (TLR4) have been related to susceptibility to Gram-negative infections and septic shock. Here we show that two polymorphisms of TLR4, Asp299Gly and Thr399Ile, have unique distributions in populations from Africa, Asia, and Europe. Genetic and functional studies are compatible with a model in which the nonsynonymous polymorphism Asp299Gly has evolved as a protective allele against malaria, explaining its high prevalence in subSaharan Africa. However, the same allele could have been disadvantageous after migration of modern humans into Eurasia, putatively because of increased susceptibility to severe bacterial infections. In contrast, the Asp299Gly allele, when present in cosegregation with Thr399Ile to form the Asp299Gly/Thr399Ile haplotype, shows selective neutrality. Polymorphisms in TLR4 exemplify how the interaction between our innate immune system and the infectious pressures in particular environments may have shaped the genetic variations and function of our immune system during the out-of-Africa migration of modern humans.


Asunto(s)
Emigración e Inmigración , Evolución Molecular , Infecciones/genética , Polimorfismo Genético , Población/genética , Receptor Toll-Like 4/genética , Adulto , Alelos , Secuencia de Aminoácidos , Femenino , Haplotipos , Humanos , Inmunidad Innata/genética , Masculino , Persona de Mediana Edad , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...