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










Base de datos
Intervalo de año de publicación
1.
Metabolism ; 151: 155759, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38101770

RESUMEN

BACKGROUND AND AIMS: Subcutaneous adipose tissue (SAT) dysfunction contributes to NAFLD pathogenesis and may be influenced by the gut microbiota. Whether transcript profiles of SAT are associated with liver fibrosis and are influenced by synbiotic treatment (that changes the gut microbiome) is unknown. We investigated: (a) whether the presence of clinically significant, ≥F2 liver fibrosis associated with adipose tissue (AT) dysfunction, differential gene expression in SAT, and/or a marker of tissue fibrosis (Composite collagen gene expression (CCGE)); and (b) whether synbiotic treatment modified markers of AT dysfunction and the SAT transcriptome. METHODS: Sixty-two patients with NAFLD (60 % men) were studied before and after 12 months of treatment with synbiotic or placebo and provided SAT samples. Vibration-controlled transient elastography (VCTE)-validated thresholds were used to assess liver fibrosis. RNA-sequencing and histological analysis of SAT were performed to determine differential gene expression, CCGE and the presence of collagen fibres. Regression modelling and receiver operator characteristic curve analysis were used to test associations with, and risk prediction for, ≥F2 liver fibrosis. RESULTS: Patients with ≥F2 liver fibrosis (n = 24) had altered markers of AT dysfunction and a SAT gene expression signature characterised by enrichment of inflammatory and extracellular matrix-associated genes, compared to those with

Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Simbióticos , Masculino , Humanos , Femenino , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/terapia , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Biomarcadores , Cirrosis Hepática/genética , Cirrosis Hepática/terapia , Cirrosis Hepática/complicaciones , Fibrosis , Tejido Adiposo/patología , Colágeno/genética , Hígado/patología
2.
Curr Opin Clin Nutr Metab Care ; 26(2): 72-77, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36728211

RESUMEN

PURPOSE OF REVIEW: This review aims to discuss the most recent evidence identifying the presence of distinct white adipocyte subpopulations in white adipose tissue (WAT) and how these may be altered with increasing adiposity and/or metabolic disease. We conceptualize how changes in adipocyte subpopulations may contribute to alterations in WAT function and the development of metabolic diseases such as type 2 diabetes mellitus (T2DM), nonalcoholic fatty liver disease (NAFLD) and cardiovascular disease (CVD). RECENT FINDINGS: Studies utilizing novel analytical approaches support the existence of distinct white adipocyte subpopulations in both human and murine WAT. Adipocyte subtypes are potentially functionally distinct and may have different roles in WAT function and obesity-associated metabolic diseases. SUMMARY: The exploration of white adipocyte heterogeneity using novel analytical technologies, has unveiled a new layer of complexity in the study of WAT biology. Interrogation of potential functional differences between adipocyte subpopulations and their role in the function of different WAT depots is now needed. Through understanding the mechanisms regulating white adipocyte subtype development and potential pathophysiological consequences of changes in the presence of adipocyte subpopulations, studies could provide novel therapeutic targets for the treatment of T2DM, NAFLD, and CVD.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Enfermedades Metabólicas , Enfermedad del Hígado Graso no Alcohólico , Humanos , Ratones , Animales , Adipocitos Blancos , Diabetes Mellitus Tipo 2/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Tejido Adiposo Blanco/metabolismo , Enfermedades Metabólicas/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo
3.
Proc Nutr Soc ; 81(2): 146-161, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35934688

RESUMEN

In recent years, a wealth of factors are associated with increased risk of developing non-alcoholic fatty liver disease (NAFLD) and NAFLD is now thought to increase the risk of multiple extra-hepatic diseases. The aim of this review is first to focus on the role of ageing and sex as key, poorly understood risk factors in the development and progression of NAFLD. Secondly, we aim to discuss the roles of white adipose tissue (WAT) and intestinal dysfunction, as producers of extra-hepatic factors known to further contribute to the pathogenesis of NAFLD. Finally, we aim to summarise the role of NAFLD as a multi-system disease affecting other organ systems beyond the liver. Both increased age and male sex increase the risk of NAFLD and this may be partly driven by alterations in the distribution and function of WAT. Similarly, changes in gut microbiota composition and intestinal function with ageing and chronic overnutrition are likely to contribute to the development of NAFLD both directly (i.e. by affecting hepatic function) and indirectly via exacerbating WAT dysfunction. Consequently, the presence of NAFLD significantly increases the risk of various extra-hepatic diseases including CVD, type 2 diabetes mellitus, chronic kidney disease and certain extra-hepatic cancers. Thus changes in WAT and intestinal function with ageing and chronic overnutrition contribute to the development of NAFLD - a multi-system disease that subsequently contributes to the development of other chronic cardiometabolic diseases.


Asunto(s)
Diabetes Mellitus Tipo 2 , Enfermedad del Hígado Graso no Alcohólico , Hipernutrición , Tejido Adiposo , Envejecimiento , Diabetes Mellitus Tipo 2/etiología , Humanos , Hígado , Masculino , Enfermedad del Hígado Graso no Alcohólico/etiología , Hipernutrición/complicaciones , Hipernutrición/patología
4.
Nat Metab ; 3(10): 1302-1312, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34650277

RESUMEN

Tumour necrosis factor (TNF) is a classical, pleiotropic pro-inflammatory cytokine. It is also the first 'adipokine' described to be produced from adipose tissue, regulated in obesity and proposed to contribute to obesity-associated metabolic disease. In this review, we provide an overview of TNF in the context of metabolic inflammation or metaflammation, its discovery as a metabolic messenger, its sites and mechanisms of action and some critical considerations for future research. Although we focus on TNF and the studies that elucidated its immunometabolic actions, we highlight a conceptual framework, generated by these studies, that is equally applicable to the complex network of pro-inflammatory signals, their biological activity and their integration with metabolic regulation, and to the field of immunometabolism more broadly.


Asunto(s)
Factor de Necrosis Tumoral alfa/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Resistencia a la Insulina , Obesidad/fisiopatología
5.
Nutr Diabetes ; 11(1): 32, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34663793

RESUMEN

BACKGROUND: Type 2 diabetes mellitus (T2DM) is a strong risk factor for liver fibrosis in non-alcoholic fatty liver disease (NAFLD). It remains uncertain why T2DM increases the risk of liver fibrosis. It has been suggested that growth differentiation factor-15 (GDF-15) concentrations increase the risk of liver fibrosis. We aimed to investigate (a) whether GDF-15 concentrations were associated with liver fibrosis and involved in the relationship between T2DM and liver fibrosis and (b) what factors linked with T2DM are associated with increased GDF-15 concentrations. METHODS: Ninety-nine patients with NAFLD (61% men, 42.4% T2DM) were studied. Serum GDF-15 concentrations were measured by electro-chemiluminescence immunoassay. Vibration-controlled transient elastography (VCTE)-validated thresholds were used to assess liver fibrosis. Regression modelling, receiver operator characteristic curve analysis and Sobel test statistics were used to test associations, risk predictors and the involvement of GDF-15 in the relationship between T2DM and liver fibrosis, respectively. RESULTS: Patients with NAFLD and T2DM (n = 42) had higher serum GDF-15 concentrations [mean (SD): 1271.0 (902.1) vs. 640.3 (332.5) pg/ml, p < 0.0001], and a higher proportion had VCTE assessed ≥F2 fibrosis (48.8 vs. 23.2%, p = 0.01) than those without T2DM. GDF-15 was independently associated with liver fibrosis (p = 0.001), and GDF-15 was the most important single factor predicting ≥F2 or ≥F3 fibrosis (≥F2 fibrosis AUROC 0.75, (95% CI 0.63-0.86), p < 0.001, with sensitivity, specificity, positive predictive (PPV) and negative predictive (NPV) values of 56.3%, 86.9%, 69.2% and 79.1%, respectively). GDF-15 was involved in the association between T2DM and ≥F2 fibrosis (Sobel test statistic 2.90, p = 0.004). Other factors associated with T2DM explained 60% of the variance in GDF-15 concentrations (p < 0.0001). HbA1c concentrations alone explained 30% of the variance (p < 0.0001). CONCLUSIONS: GDF-15 concentrations are a predictor of liver fibrosis and potentially involved in the association between T2DM and liver fibrosis in NAFLD. HbA1c concentrations explain a large proportion of the variance in GDF-15 concentrations.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Factor 15 de Diferenciación de Crecimiento/sangre , Cirrosis Hepática/sangre , Enfermedad del Hígado Graso no Alcohólico/sangre , Adulto , Anciano , Diabetes Mellitus Tipo 2/epidemiología , Diagnóstico por Imagen de Elasticidad/métodos , Femenino , Hemoglobina Glucada/análisis , Humanos , Inmunoensayo/métodos , Cirrosis Hepática/epidemiología , Cirrosis Hepática/metabolismo , Modelos Logísticos , Masculino , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Curva ROC , Ensayos Clínicos Controlados Aleatorios como Asunto , Factores de Riesgo , Adulto Joven
6.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34445165

RESUMEN

Macrophages and lymphocytes demonstrate metabolic plasticity, which is dependent partly on their state of activation and partly on the availability of various energy yielding and biosynthetic substrates (fatty acids, glucose, and amino acids). These substrates are essential to fuel-based metabolic reprogramming that supports optimal immune function, including the inflammatory response. In this review, we will focus on metabolism in macrophages and lymphocytes and discuss the role of fatty acids in governing the phenotype, activation, and functional status of these important cells. We summarize the current understanding of the pathways of fatty acid metabolism and related mechanisms of action and also explore possible new perspectives in this exciting area of research.


Asunto(s)
Ácidos Grasos/inmunología , Linfocitos/inmunología , Macrófagos/inmunología , Animales , Ácidos Grasos/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Activación de Linfocitos , Linfocitos/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo
7.
Placenta ; 113: 67-73, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-33994009

RESUMEN

Endocytosis is an essential mechanism for cellular uptake in many human tissues. A range of endocytic mechanisms occur including clathrin-dependent and -independent mechanisms. However, the role of endocytosis in the placenta and the spatial localisation of individual mechanisms is not well understood. The two principal cell layers that comprise the placental barrier to maternal-fetal transfer are the syncytiotrophoblast and fetal capillary endothelium. Endocytic uptake into the syncytiotrophoblast has been demonstrated for physiological maternal molecules such as transferrin-bound iron and low density lipoprotein (LDL) and may play an important role in the uptake of several other micronutrients, serum proteins, and therapeutics at both major placental cell barriers. These mechanisms may also mediate placental uptake of some viruses and nanoparticles. This review introduces the mechanisms of cargo-specific endocytosis and what is known about their localisation in the placenta, focussing predominantly on the syncytiotrophoblast. A fuller understanding of placental endocytosis is necessary to explain both fetal nutrition and the properties of the placental barrier. Characterising placental endocytic mechanisms and their regulation may allow us to identify their role in pregnancy pathologies and provide new avenues for therapeutic intervention.


Asunto(s)
Endocitosis , Placenta/fisiología , Femenino , Humanos , Técnicas In Vitro , Embarazo
8.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1865(10): 158776, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32738301

RESUMEN

Palmitoleic acid (POA, 16:1n-7) is a lipokine that has potential nutraceutical use to treat non-alcoholic fatty liver disease. We tested the effects of POA supplementation (daily oral gavage, 300 mg/Kg, 15 days) on murine liver inflammation induced by a high fat diet (HFD, 59% fat, 12 weeks). In HFD-fed mice, POA supplementation reduced serum insulin and improved insulin tolerance compared with oleic acid (OA, 300 mg/Kg). The livers of POA-treated mice exhibited less steatosis and inflammation than those of OA-treated mice with lower inflammatory cytokine levels and reduced toll-like receptor 4 protein content. The anti-inflammatory effects of POA in the liver were accompanied by a reduction in liver macrophages (LM, CD11c+; F4/80+; CD86+), an effect that could be triggered by peroxisome proliferator activated receptor (PPAR)-γ, a lipogenic transcription factor upregulated in livers of POA-treated mice. We also used HFD-fed mice with selective deletion of PPAR-γ in myeloid cells (PPAR-γ KOLyzCre+) to test whether the beneficial anti-inflammatory effects of POA are dependent on macrophages PPAR-γ. POA-mediated improvement of insulin tolerance was tightly dependent on myeloid PPAR-γ, while POA anti-inflammatory actions including the reduction in liver inflammatory cytokines were preserved in mice bearing myeloid cells deficient in PPAR-γ. This overlapped with increased CD206+ (M2a) cells and downregulation of CD86+ and CD11c+ liver macrophages. Moreover, POA supplementation increased hepatic AMPK activity and decreased expression of the fatty acid binding scavenger receptor, CD36. We conclude that POA controls liver inflammation triggered by fat accumulation through induction of M2a macrophages independently of myeloid cell PPAR-γ.


Asunto(s)
Ácidos Grasos Monoinsaturados/farmacología , Inflamación/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , PPAR gamma/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Antígeno B7-2/genética , Antígeno CD11c/genética , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos Monoinsaturados/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Resistencia a la Insulina/genética , Lectinas Tipo C/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Ratones , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ácido Oléico/metabolismo , Ácido Oléico/farmacología , Proteínas Quinasas/genética , Receptores de Superficie Celular/genética
9.
Gastroenterology ; 158(6): 1597-1610.e7, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31987796

RESUMEN

BACKGROUND & AIMS: Dysbiosis of the intestinal microbiota has been associated with nonalcoholic fatty liver disease (NAFLD). We investigated whether administration of a synbiotic combination of probiotic and prebiotic agents affected liver fat content, biomarkers of liver fibrosis, and the composition of the fecal microbiome in patients with NAFLD. METHODS: We performed a double-blind phase 2 trial of 104 patients with NAFLD in the United Kingdom. Participants (mean age, 50.8 ± 12.6 years; 65% men; 37% with diabetes) were randomly assigned to groups given the synbiotic agents (fructo-oligosaccharides, 4 g twice per day, plus Bifidobacterium animalis subspecies lactis BB-12; n = 55) or placebo (n = 49) for 10-14 months. Liver fat content was measured at the start and end of the study by magnetic resonance spectroscopy, and liver fibrosis was determined from a validated biomarker scoring system and vibration-controlled transient elastography. Fecal samples were collected at the start and end of the study, the fecal microbiome were analyzed by 16S ribosomal DNA sequencing. RESULTS: Mean baseline and end-of-study magnetic resonance spectroscopy liver fat percentage values were 32.3% ± 24.8% and 28.5% ± 20.1% in the synbiotic group and 31.3% ± 22% and 25.2% ± 17.2% in the placebo group. In the unadjusted intention-to-treat analysis, we found no significant difference in liver fat reduction between groups (ß = 2.8; 95% confidence interval, -2.2 to 7.8; P = .30). In a fully adjusted regression model (adjusted for baseline measurement of the outcome plus age, sex, weight difference, and baseline weight), only weight loss was associated with a significant decrease in liver fat (ß = 2; 95% confidence interval, 1.5-2.6; P = .03). Fecal samples from patients who received the synbiotic had higher proportions of Bifidobacterium and Faecalibacterium species, and reductions in Oscillibacter and Alistipes species, compared with baseline; these changes were not observed in the placebo group. Changes in the composition of fecal microbiota were not associated with liver fat or markers of fibrosis. CONCLUSIONS: In a randomized trial of patients with NAFLD, 1 year of administration of a synbiotic combination (probiotic and prebiotic) altered the fecal microbiome but did not reduce liver fat content or markers of liver fibrosis. (ClinicalTrials.gov, Number: NCT01680640).


Asunto(s)
Disbiosis/dietoterapia , Microbioma Gastrointestinal/efectos de los fármacos , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/dietoterapia , Simbióticos/administración & dosificación , Adulto , Bifidobacterium animalis , Biomarcadores/análisis , Biopsia , Método Doble Ciego , Disbiosis/complicaciones , Diagnóstico por Imagen de Elasticidad , Heces/microbiología , Femenino , Humanos , Lípidos/análisis , Hígado/química , Hígado/diagnóstico por imagen , Hígado/patología , Cirrosis Hepática/prevención & control , Espectroscopía de Resonancia Magnética , Masculino , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/patología , Oligosacáridos/administración & dosificación , Prueba de Estudio Conceptual , Reino Unido
10.
Biology (Basel) ; 8(1)2019 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-30736459

RESUMEN

The current knowledge of sex-dependent differences in adipose tissue biology remains in its infancy and is motivated in part by the desire to understand why menopause is linked to an increased risk of metabolic disease. However, the development and characterization of targeted genetically-modified rodent models are shedding new light on the physiological actions of sex hormones in healthy reproductive metabolism. In this review we consider the need for differentially regulating metabolic flexibility, energy balance, and immunity in a sex-dependent manner. We discuss the recent advances in our understanding of physiological roles of systemic estrogen in regulating sex-dependent adipose tissue distribution, form and function; and in sex-dependent healthy immune function. We also review the decline in protective properties of estrogen signaling in pathophysiological settings such as obesity-related metaflammation and metabolic disease. It is clear that the many physiological actions of estrogen on energy balance, immunity, and immunometabolism together with its dynamic regulation in females make it an excellent candidate for regulating metabolic flexibility in the context of reproductive metabolism.

11.
Int J Obes (Lond) ; 43(3): 556-566, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30006585

RESUMEN

BACKGROUND/OBJECTIVES: The increased prevalence of obesity has prompted great strides in our understanding of specific adipose depots and their involvement in cardio-metabolic health. However, the impact of obesity on dermal white adipose tissue (dWAT) and dermal microvascular functionality remains unclear. This study aimed to investigate the temporal changes that occur in dWAT and dermal microvascular functionality during the development of diet-induced obesity and type 2 diabetes in mice. METHODS: Metabolic phenotyping of a murine model of hypercaloric diet (HCD)-induced obesity and type 2 diabetes was performed at three time points that reflected three distinct stages of disease development; 2 weeks of HCD-overweight-metabolically healthy, 4 weeks of HCD-obese-prediabetic and 12 weeks of HCD-obese-type 2 diabetic mice. Expansion of dWAT was characterized histologically, and changes in dermal microvascular reactivity were assessed in response to pressure and the vasodilators SNP and Ach. RESULTS: HCD resulted in a progressive expansion of dWAT and increased expression of pro-inflammatory markers (IL1ß and COX-2). Impairments in pressure-induced (PIV) and Ach-induced (endothelium-dependent) vasodilation occurred early, in overweight-metabolically healthy mice. Residual vasodilatory responses were NOS-independent but sensitive to COX inhibition. These changes were associated with reductions in NO and adiponectin bioavailability, and rescued by exogenous adiponectin or hyperinsulinemia. Obese-prediabetic mice continued to exhibit impaired Ach-dependent vasodilation but PIV appeared normalized. This normalization coincided with elevated endogenous adiponectin and insulin levels, and was sensitive to NOS, COX and PI3K, inhibition. In obese-type 2 diabetic mice, both Ach-stimulated and pressure-induced vasodilatory responses were increased through enhanced COX-2-dependent prostaglandin response. CONCLUSIONS: We demonstrate that the development of obesity, metabolic dysfunction and type 2 diabetes, in HCD-fed mice, is accompanied by increased dermal adiposity and associated metaflammation in dWAT. Importantly, these temporal changes are also linked to disease stage-specific dermal microvascular reactivity, which may reflect adaptive mechanisms driven by metaflammation.


Asunto(s)
Tejido Adiposo Blanco , Diabetes Mellitus Tipo 2/fisiopatología , Inflamación , Obesidad/fisiopatología , Piel , Adiponectina/metabolismo , Tejido Adiposo Blanco/irrigación sanguínea , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Tejido Adiposo Blanco/fisiopatología , Animales , Citocinas/metabolismo , Diabetes Mellitus Experimental , Inflamación/metabolismo , Inflamación/patología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Microvasos/metabolismo , Microvasos/patología , Microvasos/fisiopatología , Piel/irrigación sanguínea , Piel/metabolismo , Piel/fisiopatología , Vasodilatación/fisiología
13.
Br J Pharmacol ; 173(14): 2182-94, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27128025

RESUMEN

In this review, we focus on the secreted form of nicotinamide phosphoribosyltransferase (NAMPT); extracellular NAMPT (eNAMPT), also known as pre-B cell colony-enhancing factor or visfatin. Although intracellular NAMPT is a key enzyme in controlling NAD metabolism, eNAMPT has been reported to function as a cytokine, with many roles in physiology and pathology. Circulating eNAMPT has been associated with several metabolic and inflammatory disorders, including cancer. Because cytokines produced in the tumour micro-environment play an important role in cancer pathogenesis, in part by reprogramming cellular metabolism, future improvements in cancer immunotherapy will require a better understanding of the crosstalk between cytokine action and tumour biology. In this review, the knowledge of eNAMPT in cancer will be discussed, focusing on its immunometabolic function as a metabokine, its secretion, its mechanism of action and possible roles in the cancer micro-environment.


Asunto(s)
Neoplasias/enzimología , Nicotinamida Fosforribosiltransferasa/metabolismo , Animales , Humanos , Neoplasias/metabolismo , Microambiente Tumoral
14.
Cell Metab ; 22(6): 949-53, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26636490

RESUMEN

The "Rosies" of Cell Metabolism are back for the third part of the "Women in Metabolism" 2015 series. We are closing our anniversary celebrations with 14 inspiring and engaging new stories from women scientists in the metabolism field. A round of applause to all who contributed and supported this project!


Asunto(s)
Investigación , Aniversarios y Eventos Especiales , Femenino , Humanos
15.
Proc Natl Acad Sci U S A ; 112(2): 506-11, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25540417

RESUMEN

Obesity increases the risk of developing life-threatening metabolic diseases including cardiovascular disease, fatty liver disease, diabetes, and cancer. Efforts to curb the global obesity epidemic and its impact have proven unsuccessful in part by a limited understanding of these chronic progressive diseases. It is clear that low-grade chronic inflammation, or metaflammation, underlies the pathogenesis of obesity-associated type 2 diabetes and atherosclerosis. However, the mechanisms that maintain chronicity and prevent inflammatory resolution are poorly understood. Here, we show that inhibitor of κB kinase epsilon (IKBKE) is a novel regulator that limits chronic inflammation during metabolic disease and atherosclerosis. The pathogenic relevance of IKBKE was indicated by the colocalization with macrophages in human and murine tissues and in atherosclerotic plaques. Genetic ablation of IKBKE resulted in enhanced and prolonged priming of the NLRP3 inflammasome in cultured macrophages, in hypertrophic adipose tissue, and in livers of hypercholesterolemic mice. This altered profile associated with enhanced acute phase response, deregulated cholesterol metabolism, and steatoheptatitis. Restoring IKBKE only in hematopoietic cells was sufficient to reverse elevated inflammasome priming and these metabolic features. In advanced atherosclerotic plaques, loss of IKBKE and hematopoietic cell restoration altered plaque composition. These studies reveal a new role for hematopoietic IKBKE: to limit inflammasome priming and metaflammation.


Asunto(s)
Quinasa I-kappa B/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Tejido Adiposo/metabolismo , Adulto , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Proteínas Portadoras/metabolismo , Femenino , Sistema Hematopoyético/metabolismo , Humanos , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/genética , Inflamación/etiología , Hígado/metabolismo , Macrófagos/metabolismo , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Proteína con Dominio Pirina 3 de la Familia NLR , Placa Aterosclerótica/etiología , Placa Aterosclerótica/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
16.
PLoS One ; 9(10): e108963, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25330241

RESUMEN

Chronic hyperglycemia induces insulin resistance by mechanisms that are incompletely understood. One model of hyperglycemia-induced insulin resistance involves chronic preincubation of adipocytes in the presence of high glucose and low insulin concentrations. We have previously shown that the mTOR complex 1 (mTORC1) plays a partial role in the development of insulin resistance in this model. Here, we demonstrate that treatment with Go-6976, a widely used "specific" inhibitor of cPKCs, alleviates hyperglycemia-induced insulin resistance. However, the effects of mTOR inhibitor, rapamycin and Go-6976 were not additive and only rapamycin restored impaired insulin-stimulated AKT activation. Although, PKCα, (but not -ß) was abundantly expressed in these adipocytes, our studies indicate cPKCs do not play a major role in causing insulin-resistance in this model. There was no evidence of changes in the expression or phosphorylation of PKCα, and PKCα knock-down did not prevent the reduction of insulin-stimulated glucose transport. This was also consistent with lack of IRS-1 phosphorylation on Ser-24 in hyperglycemia-induced insulin-resistant adipocytes. Treatment with Go-6976 did inhibit a component of the mTORC1 pathway, as evidenced by decreased phosphorylation of S6 ribosomal protein. Raptor knock-down enhanced the effect of insulin on glucose transport in insulin resistant adipocytes. Go-6976 had the same effect in control cells, but was ineffective in cells with Raptor knock-down. Taken together these findings suggest that Go-6976 exerts its effect in alleviating hyperglycemia-induced insulin-resistance independently of cPKC inhibition and may target components of the mTORC1 signaling pathway.


Asunto(s)
Adipocitos/efectos de los fármacos , Carbazoles/administración & dosificación , Hiperglucemia/genética , Resistencia a la Insulina/genética , Insulina/metabolismo , Células 3T3-L1 , Adipocitos/enzimología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hiperglucemia/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/genética , Proteína Quinasa C-alfa/biosíntesis , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética
17.
Diabetes ; 62(11): 3697-708, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23919961

RESUMEN

The epidemic of obesity imposes unprecedented challenges on human adipose tissue (WAT) storage capacity that may benefit from adaptive mechanisms to maintain adipocyte functionality. Here, we demonstrate that changes in the regulatory feedback set point control of Insig1/SREBP1 represent an adaptive response that preserves WAT lipid homeostasis in obese and insulin-resistant states. In our experiments, we show that Insig1 mRNA expression decreases in WAT from mice with obesity-associated insulin resistance and from morbidly obese humans and in in vitro models of adipocyte insulin resistance. Insig1 downregulation is part of an adaptive response that promotes the maintenance of SREBP1 maturation and facilitates lipogenesis and availability of appropriate levels of fatty acid unsaturation, partially compensating the antilipogenic effect associated with insulin resistance. We describe for the first time the existence of this adaptive mechanism in WAT, which involves Insig1/SREBP1 and preserves the degree of lipid unsaturation under conditions of obesity-induced insulin resistance. These adaptive mechanisms contribute to maintain lipid desaturation through preferential SCD1 regulation and facilitate fat storage in WAT, despite on-going metabolic stress.


Asunto(s)
Adaptación Fisiológica , Tejido Adiposo Blanco/metabolismo , Proteínas de la Membrana/biosíntesis , Obesidad/fisiopatología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Células 3T3-L1 , Animales , Regulación hacia Abajo , Humanos , Resistencia a la Insulina , Metabolismo de los Lípidos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Obesidad Mórbida/metabolismo , ARN Mensajero/metabolismo , Estearoil-CoA Desaturasa/metabolismo
18.
Diabetes ; 61(12): 3139-47, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22923471

RESUMEN

In this study, we define a new role for lipocalin prostaglandin D synthase (L-PGDS) in the control of metabolic fuel utilization by brown adipose tissue (BAT). We demonstrate that L-PGDS expression in BAT is positively correlated with BAT activity, upregulated by peroxisome proliferator-activated receptor γ coactivator 1α or 1ß and repressed by receptor-interacting protein 140. Under cold-acclimated conditions, mice lacking L-PGDS had elevated reliance on carbohydrate to provide fuel for thermogenesis and had increased expression of genes regulating glycolysis and de novo lipogenesis in BAT. These transcriptional differences were associated with increased lipid content in BAT and a BAT lipid composition enriched with de novo synthesized lipids. Consistent with the concept that lack of L-PGDS increases glucose utilization, mice lacking L-PGDS had improved glucose tolerance after high-fat feeding. The improved glucose tolerance appeared to be independent of changes in insulin sensitivity, as insulin levels during the glucose tolerance test and insulin, leptin, and adiponectin levels were unchanged. Moreover, L-PGDS knockout mice exhibited increased expression of genes involved in thermogenesis and increased norepinephrine-stimulated glucose uptake to BAT, suggesting that sympathetically mediated changes in glucose uptake may have improved glucose tolerance. Taken together, these results suggest that L-PGDS plays an important role in the regulation of glucose utilization in vivo.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Animales , Composición Corporal/genética , Composición Corporal/fisiología , Línea Celular , Femenino , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Termogénesis/genética , Termogénesis/fisiología , Triglicéridos/metabolismo
19.
PLoS One ; 7(7): e39512, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792179

RESUMEN

Mice lacking Peroxisome Proliferator-Activated Receptor γ2 (PPARγ2) have unexpectedly normal glucose tolerance and mild insulin resistance. Mice lacking PPARγ2 were found to have elevated levels of Lipocalin prostaglandin D synthase (L-PGDS) expression in BAT and subcutaneous white adipose tissue (WAT). To determine if induction of L-PGDS was compensating for a lack of PPARγ2, we crossed L-PGDS KO mice to PPARγ2 KO mice to generate Double Knock Out mice (DKO). Using DKO mice we demonstrated a requirement of L-PGDS for maintenance of subcutaneous WAT (scWAT) function. In scWAT, DKO mice had reduced expression of thermogenic genes, the de novo lipogenic program and the lipases ATGL and HSL. Despite the reduction in markers of lipolysis in scWAT, DKO mice had a normal metabolic rate and elevated serum FFA levels compared to L-PGDS KO alone. Analysis of intra-abdominal white adipose tissue (epididymal WAT) showed elevated expression of mRNA and protein markers of lipolysis in DKO mice, suggesting that DKO mice may become more reliant on intra-abdominal WAT to supply lipid for oxidation. This switch in depot utilisation from subcutaneous to epididymal white adipose tissue was associated with a worsening of whole organism metabolic function, with DKO mice being glucose intolerant, and having elevated serum triglyceride levels compared to any other genotype. Overall, L-PGDS and PPARγ2 coordinate to regulate carbohydrate and lipid metabolism.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Oxidorreductasas Intramoleculares/metabolismo , Metabolismo de los Lípidos , Lipocalinas/metabolismo , PPAR gamma/metabolismo , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Biomarcadores/metabolismo , Metabolismo de los Hidratos de Carbono/genética , Eicosanoides/metabolismo , Regulación de la Expresión Génica , Resistencia a la Insulina/genética , Oxidorreductasas Intramoleculares/genética , Metabolismo de los Lípidos/genética , Lipocalinas/genética , Lipogénesis/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , PPAR gamma/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Grasa Subcutánea/metabolismo
20.
Diabetes ; 61(4): 915-24, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22357965

RESUMEN

Low concentrations of insulin-like growth factor (IGF) binding protein-1 (IGFBP1) are associated with insulin resistance, diabetes, and cardiovascular disease. We investigated whether increasing IGFBP1 levels can prevent the development of these disorders. Metabolic and vascular phenotype were examined in response to human IGFBP1 overexpression in mice with diet-induced obesity, mice heterozygous for deletion of insulin receptors (IR(+/-)), and ApoE(-/-) mice. Direct effects of human (h)IGFBP1 on nitric oxide (NO) generation and cellular signaling were studied in isolated vessels and in human endothelial cells. IGFBP1 circulating levels were markedly suppressed in dietary-induced obese mice. Overexpression of hIGFBP1 in obese mice reduced blood pressure, improved insulin sensitivity, and increased insulin-stimulated NO generation. In nonobese IR(+/-) mice, overexpression of hIGFBP1 reduced blood pressure and improved insulin-stimulated NO generation. hIGFBP1 induced vasodilatation independently of IGF and increased endothelial NO synthase (eNOS) activity in arterial segments ex vivo, while in endothelial cells, hIGFBP1 increased eNOS Ser(1177) phosphorylation via phosphatidylinositol 3-kinase signaling. Finally, in ApoE(-/-) mice, overexpression of hIGFBP1 reduced atherosclerosis. These favorable effects of hIGFBP1 on insulin sensitivity, blood pressure, NO production, and atherosclerosis suggest that increasing IGFBP1 concentration may be a novel approach to prevent cardiovascular disease in the setting of insulin resistance and diabetes.


Asunto(s)
Aterosclerosis/prevención & control , Presión Sanguínea/fisiología , Diabetes Mellitus/metabolismo , Resistencia a la Insulina/fisiología , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Óxido Nítrico/biosíntesis , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Células Cultivadas , Células Endoteliales , Eliminación de Gen , Humanos , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratones , Ratones Transgénicos , Obesidad/metabolismo , Receptor de Insulina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...