Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1204126, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37711626

RESUMEN

In obesity, adipose tissue infiltrating macrophages acquire a unique pro-inflammatory polarization, thereby playing a key role in the development of chronic inflammation and Type 2 diabetes. Increased saturated fatty acids (SFAs) levels have been proposed to drive this specific polarization. Accordingly, we investigated the immunometabolic reprogramming in SFA-treated human macrophages. As expected, RNA sequencing highlighted a pro-inflammatory profile but also metabolic signatures including glycolysis and hypoxia as well as a strong unfolded protein response. Glycolysis upregulation was confirmed in SFA-treated macrophages by measuring glycolytic gene expression, glucose uptake, lactate production and extracellular acidification rate. Like in LPS-stimulated macrophages, glycolysis activation in SFA-treated macrophages was dependent on HIF-1α activation and fueled the production of pro-inflammatory cytokines. SFAs and LPS both induced IRE1α endoribonuclease activity, as demonstrated by XBP1 mRNA splicing, but with different kinetics matching HIF-1α activation and the glycolytic gene expression. Interestingly, the knockdown of IRE1α and/or the pharmacological inhibition of its RNase activity prevented HIF-1α activation and significantly decreased glycolysis upregulation. Surprisingly, XBP1s appeared to be dispensable, as demonstrated by the lack of inhibiting effect of XBP1s knockdown on glycolytic genes expression, glucose uptake, lactate production and HIF-1α activation. These experiments demonstrate for the first time a key role of IRE1α in HIF-1α-mediated glycolysis upregulation in macrophages stimulated with pro-inflammatory triggers like LPS or SFAs through XBP1s-independent mechanism. IRE1 could mediate this novel function by targeting other transcripts (mRNA or pre-miRNA) through a mechanism called regulated IRE1-dependent decay or RIDD. Deciphering the underlying mechanisms of this novel IRE1 function might lead to novel therapeutic targets to curtail sterile obesity- or infection-linked inflammation.


Asunto(s)
Diabetes Mellitus Tipo 2 , Endorribonucleasas , Humanos , Glucosa , Glucólisis , Lipopolisacáridos/farmacología , Proteínas Serina-Treonina Quinasas , Ribonucleasa Pancreática , Ribonucleasas , Regulación hacia Arriba , Proteína 1 de Unión a la X-Box/genética
3.
Int J Obes (Lond) ; 45(7): 1607-1617, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33934108

RESUMEN

BACKGROUND/OBJECTIVES: Innate lymphoid cells (ILCs) play an important role in the maintenance of immune and metabolic homeostasis in adipose tissue (AT). The crosstalk between AT ILCs and adipocytes and other immune cells coordinates adipocyte differentiation, beiging, glucose metabolism and inflammation. Although the metabolic and homeostatic functions of mouse ILCs have been extensively investigated, little is known about human adipose ILCs and their roles in obesity and insulin resistance (IR). SUBJECTS/METHODS: Here we characterized T and NK cell populations in omental AT (OAT) from women (n = 18) with morbid obesity and varying levels of IR and performed an integrated analysis of metabolic parameters and adipose tissue transcriptomics. RESULTS: In OAT, we found a distinct population of CD56-NKp46+EOMES+ NK cells characterized by expression of cytotoxic molecules, pro-inflammatory cytokines, and markers of cell activation. AT IFNγ+ NK cells, but not CD4, CD8 or γδ T cells, were positively associated with glucose levels, glycated hemoglobin (HbA1c) and IR. AT NK cells were linked to a pro-inflammatory gene expression profile in AT and developed an effector phenotype in response to IL-12 and IL-15. Moreover, integrated transcriptomic analysis revealed a potential implication of AT IFNγ+ NK cells in controlling adipose tissue inflammation, remodeling, and lipid metabolism. CONCLUSIONS: Our results suggest that a distinct IFNγ-producing NK cell subset is involved in metabolic homeostasis in visceral AT in humans with obesity and may be a potential target for therapy of IR.


Asunto(s)
Hiperglucemia/metabolismo , Resistencia a la Insulina/fisiología , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Obesidad Mórbida/metabolismo , Adulto , Células Cultivadas , Femenino , Humanos , Grasa Intraabdominal/citología , Grasa Intraabdominal/metabolismo , Masculino , Persona de Mediana Edad , Adulto Joven
4.
Nutrients ; 12(12)2020 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-33302552

RESUMEN

Growth differentiation factor-15 (GDF-15) and its receptor GFRAL are both involved in the development of obesity and insulin resistance. Plasmatic GDF-15 level increases with obesity and is positively associated with disease progression. Despite macrophages have been recently suggested as a key source of GDF-15 in obesity, little is known about the regulation of GDF-15 in these cells. In the present work, we sought for potential pathophysiological activators of GDF15 expression in human macrophages and identified saturated fatty acids (SFAs) as strong inducers of GDF15 expression and secretion. SFAs increase GDF15 expression through the induction of an ER stress and the activation of the PERK/eIF2/CHOP signaling pathway in both PMA-differentiated THP-1 cells and in primary monocyte-derived macrophages. The transcription factor CHOP directly binds to the GDF15 promoter region and regulates GDF15 expression. Unlike SFAs, unsaturated fatty acids do not promote GDF15 expression and rather inhibit both SFA-induced GDF15 expression and ER stress. These results suggest that free fatty acids may be involved in the control of GDF-15 and provide new molecular insights about how diet and lipid metabolism may regulate the development of obesity and T2D.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Ácidos Grasos/farmacología , Factor 15 de Diferenciación de Crecimiento/metabolismo , Macrófagos/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo , eIF-2 Quinasa/metabolismo , Supervivencia Celular/efectos de los fármacos , Dieta , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ácidos Grasos no Esterificados , Ácidos Grasos Insaturados , Regulación de la Expresión Génica/efectos de los fármacos , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Metabolismo de los Lípidos , Obesidad/metabolismo , ARN Interferente Pequeño , Células THP-1
5.
Sci Rep ; 10(1): 21095, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33273527

RESUMEN

Retinoic acid receptor-related orphan receptor-alpha (RORα) is a transcription factor from the nuclear receptor family expressed by immune cells and involved in the development of obesity, insulin resistance (IR) and non-alcoholic steatohepatitis (NASH). It was recently reported that mice deficient for RORα in macrophages develop more severe NASH upon high fat diet (HFD) feeding due to altered Kupffer cell function. To better understand the role of RORα in obesity and IR, we independently generated a macrophage RORα-deficient mouse line. We report that RORα deletion in macrophages does not impact on HFD-induced obesity and IR. Surprisingly, we did not confirm an effect on NASH development upon HFD feeding nor in the more severe and obesity-independent choline-deficient, L-amino acid-defined diet model. Our results therefore show that RORα deletion in macrophages does not alter the development of obesity and IR and question its role in NASH.


Asunto(s)
Resistencia a la Insulina , Macrófagos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Obesidad/metabolismo , Animales , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Eliminación de Gen , Macrófagos del Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Obesidad/etiología
7.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(7): 1017-1030, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30953761

RESUMEN

NLRP3 inflammasome plays a key role in Western diet-induced systemic inflammation and was recently shown to mediate long-lasting trained immunity in myeloid cells. Saturated fatty acids (SFAs) are sterile triggers able to induce the assembly of the NLRP3 inflammasome in macrophages, leading to IL-1ß secretion while unsaturated ones (UFAs) prevent SFAs-mediated NLRP3 activation. Unlike previous studies using LPS-primed bone marrow derived macrophages, we do not see any ROS or IRE-1α involvement in SFAs-mediated NLRP3 activation in human monocytes-derived macrophages. Rather we show that SFAs need to enter the cells and to be activated into acyl-CoA to lead to NLRP3 activation in human macrophages. However, their ß-oxidation is dispensable. Instead, they are channeled towards phospholipids but redirected towards lipid droplets containing triacylglycerol in the presence of UFAs. Lipidomic analyses and Laurdan fluorescence experiments demonstrate that SFAs induce a dramatic saturation of phosphatidylcholine (PC) correlated with a loss of membrane fluidity, both events inhibited by UFAs. The silencing of CCTα, the key enzyme in PC synthesis, prevents SFA-mediated NLRP3 activation, demonstrating the essential role of the de novo PC synthesis. This SFA-induced membrane remodeling promotes a disruption of the plasma membrane Na, K-ATPase, instigating a K+ efflux essential and sufficient for NLRP3 activation. This work opens novel therapeutic avenues to interfere with Western diet-associated diseases such as those targeting the glycerolipid pathway.


Asunto(s)
Ácidos Grasos/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Potasio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Transporte Biológico , Células Cultivadas , Humanos , Inflamasomas/metabolismo , Fosfolípidos/metabolismo
8.
Cell ; 177(5): 1201-1216.e19, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31031005

RESUMEN

Innate immune responses are intricately linked with intracellular metabolism of myeloid cells. Toll-like receptor (TLR) stimulation shifts intracellular metabolism toward glycolysis, while anti-inflammatory signals depend on enhanced mitochondrial respiration. How exogenous metabolic signals affect the immune response is unknown. We demonstrate that TLR-dependent responses of dendritic cells (DCs) are exacerbated by a high-fatty-acid (FA) metabolic environment. FAs suppress the TLR-induced hexokinase activity and perturb tricarboxylic acid cycle metabolism. These metabolic changes enhance mitochondrial reactive oxygen species (mtROS) production and, in turn, the unfolded protein response (UPR), leading to a distinct transcriptomic signature with IL-23 as hallmark. Interestingly, chemical or genetic suppression of glycolysis was sufficient to induce this specific immune response. Conversely, reducing mtROS production or DC-specific deficiency in XBP1 attenuated IL-23 expression and skin inflammation in an IL-23-dependent model of psoriasis. Thus, fine-tuning of innate immunity depends on optimization of metabolic demands and minimization of mtROS-induced UPR.


Asunto(s)
Microambiente Celular/inmunología , Células Dendríticas/inmunología , Inmunidad Innata , Mitocondrias/inmunología , Especies Reactivas de Oxígeno/inmunología , Respuesta de Proteína Desplegada/inmunología , Animales , Microambiente Celular/genética , Ciclo del Ácido Cítrico/genética , Ciclo del Ácido Cítrico/inmunología , Células Dendríticas/patología , Hexoquinasa/genética , Hexoquinasa/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Noqueados , Mitocondrias/genética , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Respuesta de Proteína Desplegada/genética , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/inmunología
9.
Mol Metab ; 23: 82-87, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30904385

RESUMEN

OBJECTIVES: The nuclear receptor superfamily is a potential target for the development of new treatments for obesity and metabolic diseases. Increasing evidence has pointed towards the retinoic acid-related orphan receptor-alpha (RORα) as an important nuclear receptor involved in several biological processes. RORα full body knockout mice display improved metabolic phenotypes on both chow and high fat (60% fat, 20% carbohydrate) diets, but also have severe behavioral abnormalities. Here we investigated the effect of hepatic RORα by generating mice with liver-specific RORα deletion to elucidate the role of this nuclear receptor on host metabolism. METHODS: 8 week-old mice with liver-specific RORα deletion and littermate controls were fed either chow or western-style diets (40% fat, 40% carbohydrate) for 12 weeks. Metabolic phenotyping was performed at the end of the dietary intervention. RESULTS: Here, we show that hepatic RORα deletion does not affect the metabolic susceptibility to either chow or western-style diet in terms of glucose metabolism and adiposity. CONCLUSIONS: Our data indicate that liver deletion of RORα does not have a pivotal role in the regulation of hepatic glucose and lipid metabolism on chow or western-style diet.


Asunto(s)
Dieta Occidental , Glucosa/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Tejido Adiposo Blanco/metabolismo , Adiposidad/genética , Animales , Dieta Vegetariana , Femenino , Técnicas de Inactivación de Genes , Hepatocitos/metabolismo , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo
10.
J Virol ; 92(10)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29491159

RESUMEN

Hepatitis C virus (HCV) infection causes 500,000 deaths annually, in association with end-stage liver diseases. Investigations of the HCV life cycle have widened the knowledge of virology, and here we discovered that two piperazinylbenzenesulfonamides inhibit HCV entry into liver cells. The entry of HCV into host cells is a complex process that is not fully understood but is characterized by multiple spatially and temporally regulated steps involving several known host factors. Through a high-content virus infection screening analysis with a library of 1,120 biologically active chemical compounds, we identified SB258585, an antagonist of serotonin receptor 6 (5-HT6), as a new inhibitor of HCV entry in liver-derived cell lines as well as primary hepatocytes. A functional characterization suggested a role for this compound and the compound SB399885, which share similar structures, as inhibitors of a late HCV entry step, modulating the localization of the coreceptor tight junction protein claudin-1 (CLDN1) in a 5-HT6-independent manner. Both chemical compounds induced an intracellular accumulation of CLDN1, reflecting export impairment. This regulation correlated with the modulation of protein kinase A (PKA) activity. The PKA inhibitor H89 fully reproduced these phenotypes. Furthermore, PKA activation resulted in increased CLDN1 accumulation at the cell surface. Interestingly, an increase of CLDN1 recycling did not correlate with an increased interaction with CD81 or HCV entry. These findings reinforce the hypothesis of a common pathway, shared by several viruses, which involves G-protein-coupled receptor-dependent signaling in late steps of viral entry.IMPORTANCE The HCV entry process is highly complex, and important details of this structured event are poorly understood. By screening a library of biologically active chemical compounds, we identified two piperazinylbenzenesulfonamides as inhibitors of HCV entry. The mechanism of inhibition was not through the previously described activity of these inhibitors as antagonists of serotonin receptor 6 but instead through modulation of PKA activity in a 5-HT6-independent manner, as proven by the lack of 5-HT6 in the liver. We thus highlighted the involvement of the PKA pathway in modulating HCV entry at a postbinding step and in the recycling of the tight junction protein claudin-1 (CLDN1) toward the cell surface. Our work underscores once more the complexity of HCV entry steps and suggests a role for the PKA pathway as a regulator of CLDN1 recycling, with impacts on both cell biology and virology.


Asunto(s)
Claudina-1/metabolismo , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Receptores de Serotonina/metabolismo , Sulfonamidas/farmacología , Internalización del Virus/efectos de los fármacos , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hepacivirus/fisiología , Hepatocitos/virología , Humanos , Isoquinolinas/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Tetraspanina 28/metabolismo , Uniones Estrechas/metabolismo
12.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 113-124, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27984090

RESUMEN

Glioblastomas are the deadliest type of brain cancer and are frequently associated with poor prognosis and a high degree of recurrence despite removal by surgical resection and treatment by chemo- and radio-therapy. Photodynamic therapy (PDT) is a treatment well known to induce mainly necrotic and apoptotic cell death in solid tumors. 5-Aminolevulinic acid (5-ALA)-based PDT was recently shown to sensitize human glioblastoma cells (LN-18) to a RIP3 (Receptor Interacting Protein 3)-dependent cell death which is counter-acted by activation of autophagy. These promising results led us to investigate the pathways involved in cell death and survival mechanisms occurring in glioblastoma following PDT. In the present study, we describe a new TSC2 (Tuberous Sclerosis 2)-dependent survival pathway implicating MK2 (MAPKAPK2) kinase and 14-3-3 proteins which conducts to the activation of a pro-survival autophagy. Moreover, we characterized a new RIP3/TSC2 complex where RIP3 is suggested to promote cell death by targeting TSC2-dependent survival pathway. These results highlight (i) a new role of TSC2 to protect glioblastoma against PDT-induced cell death and (ii) TSC2 and 14-3-3 as new RIP3 partners.


Asunto(s)
Proteínas 14-3-3/genética , Ácido Aminolevulínico/farmacología , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Neuroglía/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética , Proteínas 14-3-3/antagonistas & inhibidores , Proteínas 14-3-3/metabolismo , Ácido Aminolevulínico/metabolismo , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Proteína 7 Relacionada con la Autofagia/antagonistas & inhibidores , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Silenciador del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Luz , Neuroglía/metabolismo , Neuroglía/patología , Fotoquimioterapia , Fármacos Fotosensibilizantes/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo
13.
J Innate Immun ; 9(2): 162-180, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27855374

RESUMEN

The cell wall of mycobacteria is characterised by glycolipids composed of different classes of mycolic acids (MAs; alpha-, keto-, and methoxy-) and sugars (trehalose, glucose, and arabinose). Studies using mutant Mtb strains have shown that the structure of MAs influences the inflammatory potential of these glycolipids. As mutant Mtb strains possess a complex mixture of glycolipids, we analysed the inflammatory potential of single classes of mycolate esters of the Mtb cell wall using 38 different synthetic analogues. Our results show that synthetic trehalose dimycolate (TDM) and trehalose, glucose, and arabinose monomycolates (TMM, GMM, and AraMM) activate bone marrow-derived dendritic cells in terms of the production of pro-inflammatory cytokines (IL-6 and TNF-α) and reactive oxygen species, upregulation of costimulatory molecules, and activation of NLRP3 inflammasome by a mechanism dependent on Mincle. These findings demonstrate that Mincle receptor can also recognise pentose esters and seem to contradict the hypothesis that production of GMM is an escape mechanism used by pathogenic mycobacteria to avoid recognition by the innate immune system. Finally, our experiments indicate that TMM and GMM, as well as TDM, can promote Th1 and Th17 responses in mice in an OVA immunisation model, and that further analysis of their potential as novel adjuvants for subunit vaccines is warranted.


Asunto(s)
Pared Celular/inmunología , Células Dendríticas/fisiología , Glucolípidos/inmunología , Inflamación/inmunología , Mycobacterium tuberculosis/inmunología , Ácidos Micólicos/química , Tuberculosis/inmunología , Adyuvantes Inmunológicos , Animales , Células de la Médula Ósea/fisiología , Diferenciación Celular , Células Cultivadas , Ésteres/química , Glucosa , Glucolípidos/síntesis química , Inflamasomas/metabolismo , Interleucina-6/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Trehalosa/química , Trehalosa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Virol ; 89(4): 2436-41, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25473054

RESUMEN

The protein encoded by ORF9 is essential for varicella-zoster virus (VZV) replication. Previous studies documented its presence in the trans-Golgi network and its involvement in secondary envelopment. In this work, we deleted the ORF9p acidic cluster, destroying its interaction with ORF47p, and this resulted in a nuclear accumulation of both proteins. This phenotype results in an accumulation of primary enveloped capsids in the perinuclear space, reflecting a capsid de-envelopment defect.


Asunto(s)
Cápside/metabolismo , Herpesvirus Humano 3/fisiología , Eliminación de Secuencia , Proteínas Virales/genética , Liberación del Virus , Replicación Viral , Núcleo Celular/virología , Herpesvirus Humano 3/genética
15.
Biochem Pharmacol ; 92(1): 131-41, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25175736

RESUMEN

Free fatty acids (FFAs) are metabolic intermediates that may be obtained through the diet or synthesized endogenously. In addition to serving as an important source of energy, they produce a variety of both beneficial and detrimental effects. They play essential roles as structural components of all cell membranes and as signaling molecules regulating metabolic pathways through binding to nuclear or membrane receptors. However, under conditions of FFAs overload, they become toxic, inducing ROS production, ER stress, apoptosis and inflammation. SFAs (saturated fatty acids), unlike UFAs (unsaturated fatty acids), have recently been proposed as triggers of the NLRP3 inflammasome, a molecular platform mediating the processing of IL-1ß in response to infection and stress conditions. Interestingly, UFAs, especially ω-3 FAs, inhibit NLRP3 inflammasome activation in various settings. We focus on emerging models of NLRP3 inflammasome activation with a special emphasis on the molecular mechanisms by which FFAs modulate the activation of this complex. Taking into consideration the current literature and FFA properties, we discuss the putative involvement of mitochondria and the role of cardiolipin, a mitochondrial phospholipid, proposed to be sensed by NLRP3 after release, exposure and/or oxidation. Finally, we review how this SFA-mediated NLRP3 inflammasome activation contributes to the development of both insulin resistance and deficiency associated with obesity/type 2 diabetes. In this context, we highlight the potential clinical use of ω-3 FAs as anti-inflammatory compounds.


Asunto(s)
Proteínas Portadoras/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Obesidad/metabolismo , Proteínas Portadoras/genética , Diabetes Mellitus Tipo 2/genética , Regulación de la Expresión Génica , Humanos , Inflamasomas/efectos de los fármacos , Resistencia a la Insulina/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR , Obesidad/genética
16.
Diabetologia ; 56(11): 2487-97, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24013717

RESUMEN

AIMS/HYPOTHESIS: Obesity is a heterogeneous condition comprising both individuals who remain metabolically healthy (MHO) and those who develop metabolic disorders (metabolically unhealthy, MUO). Adipose tissue is also heterogeneous in that its visceral component is more frequently associated with metabolic dysfunction than its subcutaneous component. The development of metabolic disorders is partly mediated by the NLR family pyrin domain containing-3 (NLRP3) inflammasome, which increases the secretion of inflammatory cytokines via activation of caspase-1. We compared the immunological profile and NLRP3 activity in adipose tissue between MUO and MHO individuals. METHODS: MHO and MUO phenotypes were defined, respectively, as the absence and the presence of the metabolic syndrome. Cellular composition and intrinsic inflammasome activity were investigated by flow cytometry, quantitative RT-PCR and tissue culture studies in subcutaneous and visceral adipose tissue from 23 MUO, 21 MHO and nine lean individuals. RESULTS: We found significant differences between the three study groups, including an increased secretion of IL-1ß, increased expression of IL1B and NLRP3, increased number of adipose tissue macrophages and decreased number of regulatory T cells in the visceral adipose tissue of MUO patients compared with MHO and lean participants. In macrophages derived from visceral adipose tissue, both caspase-1 activity and IL-1ß levels were higher in MUO patients than in MHO patients. Furthermore, caspase-1 activity was higher in CD11c(+)CD206(+) adipose tissue macrophages than in CD11c(-)CD206(+) cells. CONCLUSIONS/INTERPRETATION: The MUO phenotype seems to be associated with an increased activation of the NLPR3 inflammasome in macrophages infiltrating visceral adipose tissue, and a less favourable inflammatory profile compared with the MHO phenotype.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/metabolismo , Obesidad/metabolismo , Adulto , Proteínas Portadoras/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Masculino , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR , Obesidad/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Lipid Res ; 54(11): 2998-3008, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24006511

RESUMEN

The NLRP3 inflammasome is involved in many obesity-associated diseases, such as type 2 diabetes, atherosclerosis, and gouty arthritis, through its ability to induce interleukin (IL)-1ß release. The molecular link between obesity and inflammasome activation is still unclear, but free fatty acids have been proposed as one triggering event. Here we reported opposite effects of saturated fatty acids (SFAs) compared with unsaturated fatty acids (UFAs) on NLRP3 inflammasome in human monocytes/macrophages. Palmitate and stearate, both SFAs, triggered IL-1ß secretion in a caspase-1/ASC/NLRP3-dependent pathway. Unlike SFAs, the UFAs oleate and linoleate did not lead to IL-1ß secretion. In addition, they totally prevented the IL-1ß release induced by SFAs and, with less efficiency, by a broad range of NLRP3 inducers, including nigericin, alum, and monosodium urate. UFAs did not affect the transcriptional effect of SFAs, suggesting a specific effect on the NLRP3 activation. These results provide a new anti-inflammatory mechanism of UFAs by preventing the activation of the NLRP3 inflammasome and, therefore, IL-1ß processing. By this way, UFAs might play a protective role in NLRP3-associated diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Ácidos Grasos Insaturados/farmacología , Inflamasomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Línea Celular , Humanos , Interleucina-1beta/metabolismo , Macrófagos/citología , Monocitos/citología , Proteína con Dominio Pirina 3 de la Familia NLR , Palmitatos/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estearatos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA