Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Int J Obes (Lond) ; 41(5): 722-728, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28179648

RESUMEN

BACKGROUND/OBJECTIVES: Although adipose tissue (AT) hypoxia is present in rodent models of obesity, evidence for this in humans is limited. Here, we investigated the effects of diet-induced weight loss (WL) on abdominal subcutaneous AT oxygen tension (pO2), AT blood flow (ATBF), AT capillary density, AT morphology and transcriptome, systemic inflammatory markers and insulin sensitivity in humans. SUBJECTS/METHODS: Fifteen overweight and obese individuals underwent a dietary intervention (DI), consisting of a 5-week very-low-calorie diet (VLCD, 500 kcal day-1; WL), and a subsequent 4-week weight stable diet (WS). Body composition, AT pO2 (optochemical monitoring), ATBF (133Xe washout), and whole-body insulin sensitivity were determined, and AT biopsies were collected at baseline, end of WL (week 5) and end of WS (week 9). RESULTS: Body weight, body fat percentage and adipocyte size decreased significantly during the DI period. The DI markedly decreased AT pO2 and improved insulin sensitivity, but did not alter ATBF. Finally, the DI increased AT gene expression of pathways related to mitochondrial biogenesis and non-mitochondrial oxygen consumption. CONCLUSIONS: VLCD-induced WL markedly decreases abdominal subcutaneous AT pO2, which is paralleled by a reduction in adipocyte size, increased AT gene expression of mitochondrial biogenesis markers and non-mitochondrial oxygen consumption pathways, and improved whole-body insulin sensitivity in humans.


Asunto(s)
Inflamación/fisiopatología , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Obesidad/fisiopatología , Sobrepeso/fisiopatología , Oxígeno/metabolismo , Grasa Subcutánea Abdominal/metabolismo , Pérdida de Peso/fisiología , Adipocitos/fisiología , Hipoxia de la Célula/fisiología , Dieta Reductora , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/dietoterapia , Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Obesidad/dietoterapia , Obesidad/metabolismo , Sobrepeso/dietoterapia , Sobrepeso/metabolismo , Consumo de Oxígeno , Fenotipo , Resultado del Tratamiento
2.
Int J Obes (Lond) ; 40(3): 507-13, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26471343

RESUMEN

BACKGROUND/OBJECTIVES: Adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) are key enzymes involved in intracellular lipid catabolism. We have previously shown decreased expression and activity of these lipases in adipose tissue of obese insulin resistant individuals. Here we hypothesized that lipase deficiency might impact on insulin sensitivity and metabolic homeostasis in adipocytes not just by enhancing lipid accumulation, but also by altering lipid and carbohydrate catabolism in a peroxisome proliferator-activated nuclear receptor (PPAR)-dependent manner. METHODS: To address our hypothesis, we performed a series of in vitro experiments in a human white adipocyte model, the human multipotent adipose-derived stem (hMADS) cells, using genetic (siRNA) and pharmacological knockdown of ATGL and/or HSL. RESULTS: We show that ATGL and HSL knockdown in hMADS adipocytes disrupted mitochondrial respiration, which was accompanied by a decreased oxidative phosphorylation (OxPhos) protein content. This lead to a reduced exogenous and endogenous palmitate oxidation following ATGL knockdown, but not in HSL deficient adipocytes. ATGL deficiency was followed by excessive triacylglycerol accumulation, and HSL deficiency further increased diacylglycerol accumulation. Both single and double lipase knockdown reduced insulin-stimulated glucose uptake, which was attributable to impaired insulin signaling. These effects were accompanied by impaired activation of the nuclear receptor PPARα, and restored on PPARα agonist treatment. CONCLUSIONS: The present study indicates that lipase deficiency in human white adipocytes contributes to mitochondrial dysfunction and insulin resistance, in a PPARα-dependent manner. Therefore, modulation of adipose tissue lipases may provide a promising strategy to reverse insulin resistance in obese and type 2 diabetic patients.


Asunto(s)
Adipocitos Blancos/metabolismo , Adiposidad/fisiología , Resistencia a la Insulina/fisiología , Lipasa/deficiencia , Mitocondrias/metabolismo , Obesidad/metabolismo , PPAR alfa/agonistas , Células Cultivadas , Metabolismo Energético , Humanos , Lipasa/fisiología , Lipólisis/fisiología , Obesidad/complicaciones , Esterol Esterasa/metabolismo
3.
Int J Obes (Lond) ; 38(5): 698-706, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24317366

RESUMEN

BACKGROUND AND OBJECTIVES: Impaired regulation of lipid oxidation (metabolic inflexibility) is associated with obesity and type 2 diabetes mellitus. Recent evidence has indicated that dietary polyphenols may modulate mitochondrial function, substrate metabolism and energy expenditure in humans. The present study investigated the effects of short-term supplementation of two combinations of polyphenols on energy expenditure (EE) and substrate metabolism in overweight subjects. SUBJECTS AND METHODS: Eighteen healthy overweight volunteers (9 women, 9 men; age 35±2.5 years; body mass index 28.9±0.4 kg m(-2)) participated in a randomized, double-blind cross-over trial. Combinations of epigallocatechin-gallate (E, 282 mg day(-1))+resveratrol (R, 200 mg day(-1)) and E+R+80 mg day(-1) soy isoflavones (S) or placebo capsules (PLA) were supplemented twice daily for a period of 3 days. On day 3, circulating metabolite concentrations, EE and substrate oxidation (using indirect calorimetry) were measured during fasting and postprandial conditions for 6 h (high-fat-mixed meal (2.6 MJ, 61.2 E% fat)). RESULTS: Short-term supplementation of E+R increased resting EE (E+R vs PLA: 5.45±0.24 vs 5.23±0.25 kJ min(-1), P=0.039), whereas both E+R (699±18 kJ 120 min(-1) vs 676±20 kJ 120 min(-1), P=0.028) and E+R+S (704±18 kJ 120 min(-1) vs 676±20 kJ 120 min(-1), P=0.014) increased 2-4 h-postprandial EE compared with PLA. Metabolic flexibility, calculated as the postprandial increase to the highest respiratory quotient achieved, tended to be improved by E+R compared with PLA and E+R+S only in men (E+R vs PLA: 0.11±0.02 vs 0.06±0.02, P=0.059; E+R+S: 0.03±0.02, P=0.009). E+R+S significantly increased fasting plasma free fatty acid (P=0.064) and glycerol (P=0.021) concentrations compared with PLA. CONCLUSIONS: We demonstrated for the first time that combined E+R supplementation for 3 days significantly increased fasting and postprandial EE, which was accompanied by improved metabolic flexibility in men but not in women. Addition of soy isoflavones partially reversed these effects possibly due to their higher lipolytic potential. The present findings may imply that long-term supplementation of these dosages of epigallocatechin-gallate combined with resveratrol may improve metabolic health and body weight regulation.


Asunto(s)
Antioxidantes/uso terapéutico , Catequina/análogos & derivados , Metabolismo Energético/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Sobrepeso/dietoterapia , Polifenoles/uso terapéutico , Estilbenos/uso terapéutico , Adulto , Índice de Masa Corporal , Catequina/uso terapéutico , Suplementos Dietéticos , Método Doble Ciego , Quimioterapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sobrepeso/metabolismo , Oxidación-Reducción , Periodo Posprandial , Resveratrol , Resultado del Tratamiento
4.
Int J Obes (Lond) ; 38(3): 470-3, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23958793

RESUMEN

Polyphenolic compounds, such as resveratrol, have recently received widespread interest because of their ability to mimic effects of calorie restriction. The objective of the present study was to gain more insight into the effects of 30 days resveratrol supplementation on adipose tissue morphology and underlying processes. Eleven healthy obese men were supplemented with placebo and resveratrol for 30 days (150 mg per day), separated by a 4-week washout period in a double-blind randomized crossover design. A postprandial abdominal subcutaneous adipose tissue biopsy was collected to assess adipose tissue morphology and gene expression using microarray analysis. Resveratrol significantly decreased adipocyte size, with a shift toward a reduction in the proportion of large and very-large adipocytes and an increase in small adipocytes. Microarray analysis revealed downregulation of Wnt and Notch signaling pathways and upregulation of pathways involved in cell cycle regulation after resveratrol supplementation, suggesting enhanced adipogenesis. Furthermore, lysosomal/phagosomal pathway and transcription factor EB were upregulated reflecting an alternative pathway of lipid breakdown by autophagy. Further research is necessary to investigate whether resveratrol improves adipose tissue function.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Obesidad/tratamiento farmacológico , Estilbenos/uso terapéutico , Adipogénesis/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Adulto , Anciano , Estudios Cruzados , Método Doble Ciego , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Obesidad/genética , Obesidad/metabolismo , Receptores Notch/efectos de los fármacos , Receptores Notch/metabolismo , Resveratrol , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Vía de Señalización Wnt/efectos de los fármacos
5.
Int J Obes (Lond) ; 36(5): 709-17, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21712806

RESUMEN

OBJECTIVE: To determine insulin sensitivity and skeletal muscle fatty acid (FA) handling at baseline and after a high-fat mixed meal in impaired fasting glucose (IFG), impaired glucose tolerance (IGT), IFG/IGT and normal glucose tolerance (NGT) subjects. DESIGN: In this multi-center study, insulin sensitivity and ß-cell function were assessed (n=102), using a euglycemic-hyperinsulinemic and hyperglycemic clamp with additional arginine stimulation and a 75 g oral glucose tolerance test. Fasting and postprandial skeletal muscle FA handling was examined in a substudy using the forearm balance technique (n=35). SUBJECTS: A total of 102 subjects with IFG (n=48), IGT (n=12), IFG/IGT (n=26) and NGT (n=16). RESULTS: IFG, IGT and IFG/IGT subjects had lower insulin sensitivity with no differences between groups, and lower impaired ß-cell function compared with NGT controls. The early postprandial increase in triacylglycerol (TAG) concentration was higher (iAUC(0-2 h) IFG: 238.4±26.5, IGT: 234.0±41.0 and NGT: 82.6±13.8 µmol l(-1) min(-1), both P<0.05) and early TAG extraction was increased (AUC(0-2 h) IFG: 56.8±9.0, IGT: 52.2±12.0 and NGT: 3.8±15.4 nmol·100 ml(-1) min(-1), P<0.05 and P=0.057, respectively) in both IFG and IGT subjects. CONCLUSION: IFG, IGT and IFG/IGT subjects have lower insulin sensitivity and impaired ß-cell function compared with age- and BMI-matched NGT controls. The increased postprandial TAG response and higher muscle TAG extraction in both IFG and IGT compared with NGT may lead to ectopic fat accumulation in the skeletal muscle, thereby contributing to insulin resistance.


Asunto(s)
Glucemia/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Músculo Esquelético/metabolismo , Estado Prediabético/metabolismo , Índice de Masa Corporal , Ayuno , Ácidos Grasos , Femenino , Técnica de Clampeo de la Glucosa , Intolerancia a la Glucosa , Humanos , Masculino , Persona de Mediana Edad , Periodo Posprandial
6.
Diabetes Obes Metab ; 14(7): 586-95, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22226145

RESUMEN

Agents interfering with the renin-angiotensin system (RAS) were consistently shown to lower the incidence of type 2 diabetes mellitus (T2DM), as compared to other antihypertensive drugs, in hypertensive high-risk populations. The mechanisms underlying this protective effect of RAS blockade using angiotensin-converting enzyme inhibitors or angiotensin-receptor blockers on glucose metabolism are not fully understood. In this article, we will review the evidence from randomized controlled trials and discuss the proposed mechanisms as to how RAS interference may delay the onset of T2DM. In particular, as T2DM is characterized by ß-cell dysfunction and obesity-related insulin resistance, we address the mechanisms that underlie RAS blockade-induced improvement in ß-cell function and insulin sensitivity.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Antihipertensivos/uso terapéutico , Diabetes Mellitus Tipo 2/prevención & control , Angiopatías Diabéticas/prevención & control , Hipertensión/tratamiento farmacológico , Sistema Renina-Angiotensina/efectos de los fármacos , Adulto , Anciano , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Angiopatías Diabéticas/sangre , Angiopatías Diabéticas/metabolismo , Medicina Basada en la Evidencia , Femenino , Humanos , Hipertensión/sangre , Hipertensión/fisiopatología , Incidencia , Resistencia a la Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Persona de Mediana Edad , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto
7.
Benef Microbes ; 13(4): 355-363, 2022 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-35866596

RESUMEN

Results from high altitude studies in humans and controlled animal experiments suggest that hypoxia exposure induces alterations in gut microbiota composition, which may in turn affect host metabolism. However, well-controlled studies investigating the effects of normobaric hypoxia exposure on gut microbiota composition in humans are lacking. The aim of this study was to explore the impact of mild intermittent hypoxia (MIH) exposure on gut microbiota composition in men with overweight and/or obesity. We performed a randomised, single-blind crossover study, in which participants were exposed to MIH (FiO2: 15%, 3×2 h per day) and normoxia (FiO2: 21%) for seven consecutive days. Following the MIH and normoxia exposure regimens, faecal samples were collected for determination of faecal microbiota composition using 16S rRNA gene-amplicon sequencing in the morning of day 8. Paired faecal samples were available for five individuals. Furthermore, tissue-specific insulin sensitivity was determined using the gold-standard two-step hyperinsulinemic-euglycemic clamp. MIH did not affect microbial alpha and beta-diversity but reduced the relative abundance of Christensenellaceae and Clostridiaceae bacterial families. MIH significantly increased the abundances of obligate anaerobic bacterial genera including Fusicatenibacter, Butyricicoccus and Holdemania, whilst reducing Christensenellaceae R-7 group and Clostridium sensu stricto 1, although these findings were not statistically significant after correction for multiple testing. Furthermore, MIH-induced alterations in abundances of several genera were associated with changes in metabolic parameters such as adipose and peripheral insulin sensitivity, plasma levels of insulin, fatty acids, triacylglycerol and lactate, and substrate oxidation. In conclusion, we demonstrate for the first time that MIH exposure induces modest effects on faecal microbiota composition in humans, shifting several bacterial families and genera towards higher abundances of anaerobic butyrate-producing bacteria. Moreover, MIH-induced effects on faecal microbial composition were associated with parameters related to glucose and lipid homeostasis, supporting a link between MIH-induced alterations in faecal microbiota composition and host metabolism. The study was registered at the Netherlands Trial Register: NL7120/NTR7325.


Asunto(s)
Microbioma Gastrointestinal , Resistencia a la Insulina , Probióticos , Animales , Butiratos/análisis , Estudios Cruzados , Ácidos Grasos/análisis , Heces/microbiología , Glucosa/metabolismo , Humanos , Hipoxia , Insulina , Lactatos , Lípidos/análisis , Masculino , Obesidad/microbiología , Sobrepeso/complicaciones , ARN Ribosómico 16S/análisis , ARN Ribosómico 16S/genética , Método Simple Ciego , Triglicéridos/análisis
8.
Int J Obes (Lond) ; 33(6): 669-79, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19399022

RESUMEN

BACKGROUND: Part of the heterogeneity of the obesity phenotype may originate from genetic differences between obese individuals that may influence energy expenditure (EE). OBJECTIVE: To examine if common single-nucleotide polymorphisms (SNPs) in genes related to obesity-associated phenotypes are associated with postabsorptive resting energy expenditure (REE) and postprandial REE in obese individuals. DESIGN AND METHODS: Postabsorptive REE and 3-h postprandial REE (liquid test meal containing 95% fat, energy content 50% of estimated REE) were measured in 743 obese individuals from eight clinical centres in seven European countries. The analysis assessed the association of genotypes of 44 SNPs in 28 obesity-related candidate genes with postabsorptive REE and postprandial REE taking into consideration the influence of body composition, habitual physical activity, insulin sensitivity, circulating thermogenic hormones and metabolites. RESULTS: After adjustment for fat-free mass (FFM), age, sex and research centre, SNPs in CART, GAD2, PCSK1, PPARG3, HSD11B1 and LIPC were significantly associated with postabsorptive REE. SNPs in GAD2, HSD11B1 and LIPC remained significantly associated with postabsorptive REE after further adjustment for fat mass (FM). SNPs in CART, PPARG2 and IGF2 were significantly associated with postprandial REE after similar adjustments. These associations with postprandial REE remained significant after further adjustment for FM. FTO, UCP2 and UCP3 variants were not associated with postabsorptive or postprandial REE. CONCLUSIONS: Several gene polymorphisms associated with obesity-related phenotypes but not FTO and UCP variants may be responsible for some of the inter-individual variability in postabsorptive REE and fat-induced thermogenesis unaccounted for by FFM, FM, age and sex. The association between FTO and obesity that has been reported earlier may not be mediated directly through modulation of EE in obese individuals.


Asunto(s)
Metabolismo Energético/genética , Obesidad/genética , Polimorfismo de Nucleótido Simple/genética , Periodo Posprandial/genética , Termogénesis/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Adulto , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Metabolismo Energético/fisiología , Femenino , Genotipo , Glutamato Descarboxilasa/genética , Humanos , Canales Iónicos/genética , Lipasa/genética , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Obesidad/metabolismo , Fenotipo , Periodo Posprandial/fisiología , Proteínas/genética , Descanso/fisiología , Termogénesis/fisiología , Proteína Desacopladora 2 , Proteína Desacopladora 3 , Adulto Joven
9.
Sci Rep ; 9(1): 4202, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862933

RESUMEN

Body fat distribution is an important determinant of cardiometabolic health. Lower-body adipose tissue (AT) has protective characteristics as compared to upper-body fat, but the underlying depot-differences remain to be elucidated. Here, we compared the proteome and morphology of abdominal and femoral AT. Paired biopsies from abdominal and femoral subcutaneous AT were taken from eight overweight/obese (BMI ≥ 28 kg/m2) women with impaired glucose metabolism after an overnight fast. Proteins were isolated and quantified using liquid chromatography-mass spectrometry, and protein expression in abdominal and femoral subcutaneous AT was compared. Moreover, correlations between fat cell size and the proteome of both AT depots were determined. In total, 651 proteins were identified, of which 22 proteins tended to be differentially expressed between abdominal and femoral AT after removal of blood protein signals (p < 0.05). Proteins involved in cell structure organization and energy metabolism were differently expressed between AT depots. Fat cell size, which was higher in femoral AT, was significantly correlated with ADH1B, POSTN and LCP1. These findings suggest that there are only slight differences in protein expression between abdominal and femoral subcutaneous AT. It remains to be determined whether these differences, as well as differences in protein activity, contribute to functional and/or morphological differences between these fat depots.


Asunto(s)
Grasa Abdominal/metabolismo , Obesidad/metabolismo , Proteoma/metabolismo , Grasa Subcutánea/metabolismo , Grasa Abdominal/patología , Femenino , Humanos , Persona de Mediana Edad , Obesidad/patología , Proteómica , Grasa Subcutánea/patología
10.
Thorax ; 63(12): 1110-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19020276

RESUMEN

Chronic obstructive pulmonary disease (COPD) and obesity are common and disabling chronic health conditions with increasing prevalence worldwide. A relationship between COPD and obesity is increasingly recognised, although the nature of this association remains unknown. This review focuses on the epidemiology of obesity in COPD and the impact of excessive fat mass on lung function, exercise capacity and prognosis. The evidence for altered adipose tissue functions in obesity--including reduced lipid storage capacity, altered expression and secretion of inflammatory factors, adipose tissue hypoxia and macrophage infiltration in adipose tissue--is also reviewed. The interrelationship between these factors and their contribution to the development of insulin resistance in obesity is considered. It is proposed that, in patients with COPD, reduced oxidative capacity and systemic hypoxia may amplify these disturbances, not only in obese patients but also in subjects with hidden loss of fat-free mass. The potential interaction between abnormal adipose tissue function, systemic inflammation and COPD may provide more insight into the pathogenesis and reversibility of systemic pathology in this disease.


Asunto(s)
Obesidad/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/etiología , Tejido Adiposo/patología , Adulto , Anciano , Tolerancia al Ejercicio , Femenino , Volumen Espiratorio Forzado/fisiología , Humanos , Hipoxia/etiología , Resistencia a la Insulina/fisiología , Masculino , Persona de Mediana Edad , Obesidad/patología , Obesidad/fisiopatología , Neumonía/etiología , Neumonía/patología , Neumonía/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología
11.
Diabetes Obes Metab ; 10(11): 1029-38, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18435774

RESUMEN

AIM: Adipose tissue and skeletal muscle are endocrine organs, secreting substances that have been implicated in obesity-related disorders. This study examined short-term beta-adrenergic regulation of circulating leptin, adiponectin and interleukin-6 (IL-6) concentrations and secretion from abdominal subcutaneous adipose tissue and muscle (IL-6) in vivo in lean and obese subjects. METHODS: Systemic concentrations and net fluxes of leptin, adiponectin and IL-6 across abdominal subcutaneous adipose tissue and forearm skeletal muscle (IL-6) were assessed before and during beta-adrenergic stimulation (intravenous isoprenaline infusion) in 13 lean and 10 obese men. RESULTS: Basal circulating leptin concentrations were higher in the obese (p < 0.001), while circulating adiponectin (p = 0.45) and IL-6 concentrations (p = 0.41) were not different between groups. beta-Adrenergic stimulation decreased leptin concentrations in both groups (p < 0.01), but did not reduce net abdominal subcutaneous adipose tissue leptin release. Increased leptin clearance and/or decreased leptin secretion from other fat depots may explain the reduction in leptin concentrations. Adiponectin concentrations remained unchanged during beta-adrenergic stimulation in both groups. beta-Adrenergic stimulation increased IL-6 concentration, which was more pronounced in the obese (p = 0.01 vs. lean). This cannot be explained by increased IL-6 release per unit abdominal subcutaneous adipose tissue and muscle but might be because of the increased fat mass and fat-free mass at whole-body level. CONCLUSIONS: Short-term beta-adrenergic stimulation decreases leptin concentrations, which cannot be explained by reduced net leptin release from abdominal subcutaneous adipose tissue, while it elevates IL-6 concentration partly by increased release from this fat depot and muscle. Finally, beta-adrenergic stimulation has no short-term regulatory role in adiponectin secretion.


Asunto(s)
Adiponectina/metabolismo , Agonistas Adrenérgicos beta/farmacología , Interleucina-6/metabolismo , Isoproterenol/farmacología , Leptina/metabolismo , Obesidad/fisiopatología , Adiponectina/sangre , Adulto , Estudios de Casos y Controles , Antebrazo , Humanos , Interleucina-6/sangre , Leptina/sangre , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Obesidad/sangre , Estadísticas no Paramétricas , Estimulación Química , Grasa Subcutánea Abdominal/metabolismo
12.
Sci Rep ; 8(1): 4677, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29549282

RESUMEN

Obesity is associated with a disturbed adipose tissue (AT) function characterized by adipocyte hypertrophy, an impaired lipolysis and pro-inflammatory phenotype, which contributes to insulin resistance (IR). We investigated whether AT phenotype in different AT depots of obese individuals with and without type 2 diabetes mellitus (T2DM) is associated with whole-body IR. Subcutaneous (SC) and visceral (V) AT biopsies from 18 lean, 17 obese and 8 obese T2DM men were collected. AT phenotype was characterized by ex vivo measurement of basal and stimulated lipolysis (mature adipocytes), adipocyte size distribution (AT tissue sections) and AT immune cells (flow cytometry). In VAT, mean adipocyte size, CD45+ leukocytes and M1 macrophages were significantly increased in both obese groups compared to lean individuals. In SCAT, despite adipocyte hypertrophy, no significant differences in immune cell populations between groups were found. In SCAT, multiple linear regression analysis showed that none of the AT phenotype markers independently contributed to HOMA-IR while in VAT, mean adipocyte size was significantly related to HOMA-IR. In conclusion, beside adipocyte hypertrophy in VAT, M1 macrophage- or B-cell-mediated inflammation, may contribute to IR, while inflammation in hypertrophic SCAT does not seem to play a major role in IR.


Asunto(s)
Diabetes Mellitus Tipo 2/inmunología , Grasa Intraabdominal/patología , Obesidad/inmunología , Grasa Subcutánea/patología , Adipocitos , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/patología , Humanos , Resistencia a la Insulina , Grasa Intraabdominal/citología , Grasa Intraabdominal/inmunología , Antígenos Comunes de Leucocito/metabolismo , Modelos Lineales , Lipólisis , Macrófagos/inmunología , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/patología , Fenotipo , Grasa Subcutánea/citología , Grasa Subcutánea/inmunología
13.
Sci Rep ; 8(1): 3933, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29500454

RESUMEN

Increased activation of the renin-angiotensin system is involved in the onset and progression of cardiometabolic diseases, while natriuretic peptides (NP) may exert protective effects. We have recently demonstrated that sacubitril/valsartan (LCZ696), a first-in-class angiotensin receptor neprilysin inhibitor, which blocks the angiotensin II type-1 receptor and augments natriuretic peptide levels, improved peripheral insulin sensitivity in obese hypertensive patients. Here, we investigated the effects of sacubitril/valsartan (400 mg QD) treatment for 8 weeks on the abdominal subcutaneous adipose tissue (AT) phenotype compared to the metabolically neutral comparator amlodipine (10 mg QD) in 70 obese hypertensive patients. Abdominal subcutaneous AT biopsies were collected before and after intervention to determine the AT transcriptome and expression of proteins involved in lipolysis, NP signaling and mitochondrial oxidative metabolism. Both sacubitril/valsartan and amlodipine treatment did not significantly induce AT transcriptional changes in pathways related to lipolysis, NP signaling and oxidative metabolism. Furthermore, protein expression of adipose triglyceride lipase (ATGL) (Ptime*group = 0.195), hormone-sensitive lipase (HSL) (Ptime*group = 0.458), HSL-ser660 phosphorylation (Ptime*group = 0.340), NP receptor-A (NPRA) (Ptime*group = 0.829) and OXPHOS complexes (Ptime*group = 0.964) remained unchanged. In conclusion, sacubitril/valsartan treatment for 8 weeks did not alter the abdominal subcutaneous AT transcriptome and expression of proteins involved in lipolysis, NP signaling and oxidative metabolism in obese hypertensive patients.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Aminobutiratos/uso terapéutico , Antagonistas de Receptores de Angiotensina/uso terapéutico , Hipertensión/tratamiento farmacológico , Neprilisina/antagonistas & inhibidores , Obesidad/metabolismo , Proteínas/metabolismo , Tetrazoles/uso terapéutico , Transcriptoma , Tejido Adiposo/metabolismo , Adulto , Aminobutiratos/farmacología , Amlodipino/farmacología , Antagonistas de Receptores de Angiotensina/farmacología , Compuestos de Bifenilo , Método Doble Ciego , Combinación de Medicamentos , Femenino , Humanos , Hipertensión/complicaciones , Hipertensión/metabolismo , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Grasa Subcutánea/metabolismo , Tetrazoles/farmacología , Valsartán
14.
Eur J Clin Nutr ; 71(9): 1040-1045, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28589947

RESUMEN

BACKGROUND/OBJECTIVES: The intestinal microbiota may have a profound impact on host metabolism. As evidence suggests that polyphenols affect substrate utilization, the present study aimed to investigate the effects of polyphenol supplementation on intestinal microbiota composition in humans. Furthermore, we examined whether (changes in) gut microbiota composition may determine the metabolic response to polyphenol supplementation. SUBJECTS/METHODS: In this randomized, double-blind, placebo (PLA)-controlled trial, 37 overweight and obese men and women (18 males/19 females, 37.8±1.6 years, body mass index: 29.6±0.5 kg/m2) received either epigallocatechin-3-gallate and resveratrol (EGCG+RES, 282 and 80 mg/day, respectively) or PLA for 12 weeks. Before and after intervention, feces samples were collected to determine microbiota composition. Fat oxidation was assessed by indirect calorimetry during a high-fat mixed meal test (2.6 MJ, 61 energy% fat) and skeletal muscle mitochondrial oxidative capacity by means of ex vivo respirometry on isolated skeletal muscle fibers. Body composition was measured by dual-energy X-ray absorptiometry. RESULTS: Fecal abundance of Bacteroidetes was higher in men as compared with women, whereas other assessed bacterial taxa were comparable. EGCG+RES supplementation significantly decreased Bacteroidetes and tended to reduce Faecalibacterium prausnitzii in men (P=0.05 and P=0.10, respectively) but not in women (P=0.15 and P=0.77, respectively). Strikingly, baseline Bacteroidetes abundance was predictive for the EGCG+RES-induced increase in fat oxidation in men but not in women. Other bacterial genera and species were not affected by EGCG+RES supplementation. CONCLUSIONS: We demonstrated that 12-week EGCG+RES supplementation affected the gut microbiota composition in men but not in women. Baseline microbiota composition determined the increase in fat oxidation after EGCG+RES supplementation in men.


Asunto(s)
Catequina/análogos & derivados , Microbioma Gastrointestinal/efectos de los fármacos , Sobrepeso/tratamiento farmacológico , Polifenoles/administración & dosificación , Estilbenos/administración & dosificación , Absorciometría de Fotón , Adulto , Bacteroidetes/aislamiento & purificación , Catequina/administración & dosificación , Catequina/farmacología , Método Doble Ciego , Metabolismo Energético , Faecalibacterium prausnitzii/aislamiento & purificación , Heces/microbiología , Femenino , Humanos , Masculino , Músculo Esquelético/metabolismo , Sobrepeso/metabolismo , Sobrepeso/microbiología , Polifenoles/farmacología , Resveratrol , Estilbenos/farmacología , Resultado del Tratamiento
15.
Benef Microbes ; 8(4): 557-562, 2017 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-28618864

RESUMEN

Gut microbiota composition may play an important role in the development of obesity-related comorbidities. However, only few studies have investigated gender-differences in microbiota composition and gender-specific associations between microbiota or microbial products and insulin sensitivity. Insulin sensitivity (hyperinsulinemic-euglycemic clamp), body composition (dual energy X-ray absorptiometry), substrate oxidation (indirect calorimetry), systemic inflammatory markers and microbiota composition (PCR) were determined in male (n=15) and female (n=14) overweight and obese subjects. Bacteroidetes/Firmicutes-ratio was higher in men than in women (P=0.001). Bacteroidetes/Firmicutes-ratio was inversely related to peripheral insulin sensitivity only in men (men: P=0.003, women: P=0.882). This association between Bacteroidetes/Firmicutes-ratio and peripheral insulin sensitivity did not change after adjustment for dietary fibre and saturated fat intake, body composition, fat oxidation and markers of inflammation. Bacteroidetes/Firmicutes-ratio was not associated with hepatic insulin sensitivity. Men and women differ in microbiota composition and its impact on insulin sensitivity, implying that women might be less sensitive to gut microbiota-induced metabolic aberrations than men. This trial was registered at clinicaltrials.gov as NCT02381145.


Asunto(s)
Bacteroidetes/aislamiento & purificación , Firmicutes/aislamiento & purificación , Microbioma Gastrointestinal , Insulina/metabolismo , Obesidad/microbiología , Adulto , Bacteroidetes/clasificación , Bacteroidetes/genética , Heces/microbiología , Femenino , Firmicutes/clasificación , Firmicutes/genética , Glucosa/metabolismo , Humanos , Resistencia a la Insulina , Masculino , Obesidad/metabolismo
16.
Chem Phys Lipids ; 207(Pt B): 239-245, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28647339

RESUMEN

BACKGROUND: Changes in the microbiota composition have been implicated in the development of obesity and type 2 diabetes. However, not much is known on the involvement of gut microbiota in lipid and cholesterol metabolism. In addition, the gut microbiota might also be a potential source of plasma oxyphytosterol and oxycholesterol concentrations (oxidation products of plant sterols and cholesterol). Therefore, the aim of this study was to modulate the gut microbiota by antibiotic therapy to investigate effects on parameters reflecting cholesterol metabolism and oxyphytosterol concentrations. DESIGN: A randomized, double blind, placebo-controlled trial was performed in which 55 obese, pre-diabetic men received oral amoxicillin (broad-spectrum antibiotic), vancomycin (antibiotic directed against Gram-positive bacteria) or placebo (microcrystalline cellulose) capsules for 7days (1500mg/day). Plasma lipid and lipoprotein, non-cholesterol sterol, bile acid and oxy(phyto)sterol concentrations were determined at baseline and after 1-week intervention. RESULTS: Plasma secondary bile acids correlated negatively with cholestanol (marker for cholesterol absorption, r=-0.367; P<0.05) and positively with lathosterol concentrations (marker for cholesterol synthesis, r=0.430; P<0.05). Fasting plasma secondary bile acid concentrations were reduced after vancomycin treatment as compared to placebo treatment (-0.24±0.22µmol/L vs. -0.08±0.29µmol/L; P<0.01). Vancomycin and amoxicillin treatment did not affect markers for cholesterol metabolism, plasma TAG, total cholesterol, LDL-C or HDL-C concentrations as compared to placebo. In addition, both antibiotic treatments did not affect individual isoforms or total plasma oxyphytosterol or oxycholesterol concentrations. CONCLUSION: Despite strong correlations between plasma bile acid concentrations and cholesterol metabolism (synthesis and absorption), amoxicillin and vancomycin treatment for 7days did not affect plasma lipid and lipoprotein, plasma non-cholesterol sterol and oxy(phyto)sterol concentrations in obese, pre-diabetic men.


Asunto(s)
Amoxicilina/farmacología , Antibacterianos/farmacología , Colesterol/metabolismo , Vancomicina/farmacología , Administración Oral , Amoxicilina/administración & dosificación , Antibacterianos/administración & dosificación , Ácidos y Sales Biliares/sangre , Ácidos y Sales Biliares/metabolismo , Colesterol/sangre , Método Doble Ciego , Humanos , Masculino , Persona de Mediana Edad , Obesidad/sangre , Obesidad/metabolismo , Estado Prediabético/sangre , Estado Prediabético/metabolismo , Vancomicina/administración & dosificación
17.
Clin Pharmacol Ther ; 101(2): 254-263, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27542885

RESUMEN

Natriuretic peptide (NP) deficiency and sustained renin-angiotensin system activation are associated with impaired oxidative metabolism and predispose to type-2 diabetes. We hypothesized that sacubitril/valsartan (LCZ696), which augments NP through neprilysin inhibition while blocking angiotensin II type-1 (AT1 )-receptors, improves insulin sensitivity, lipid mobilization, and oxidation. After 8 weeks of treatment of obese patients with hypertension, sacubitril/valsartan 400 mg q.d., but not amlodipine 10 mg q.d., was associated with a significant increase from baseline in the insulin sensitivity index (hyperinsulinemic-euglycemic clamp), and tended to be higher in patients treated with sacubitril/valsartan compared to amlodipine. Abdominal adipose tissue interstitial glycerol concentrations increased with sacubitril/valsartan, but decreased with amlodipine. Whole-body lipolysis and substrate oxidation did not change with either treatment. Results confirm that sacubitril/valsartan treatment leads to a metabolic benefit in the study population and supports the relevance of neprilysin inhibition along with AT1 -receptor blockade in the regulation of human glucose and lipid metabolism.


Asunto(s)
Aminobutiratos/farmacología , Antihipertensivos/farmacología , Resistencia a la Insulina/fisiología , Neprilisina/antagonistas & inhibidores , Obesidad/metabolismo , Tetrazoles/farmacología , Tejido Adiposo/efectos de los fármacos , Adulto , Aminobutiratos/uso terapéutico , Amlodipino/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Compuestos de Bifenilo , Combinación de Medicamentos , Metabolismo Energético/efectos de los fármacos , Femenino , Glicerol/análisis , Humanos , Hipertensión/tratamiento farmacológico , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Péptidos Natriuréticos/genética , Péptidos Natriuréticos/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Tetrazoles/uso terapéutico , Valsartán
18.
Obes Rev ; 16(9): 715-57, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26179344

RESUMEN

Disturbances in fatty acid metabolism in adipose tissue, liver, skeletal muscle, gut and pancreas play an important role in the development of insulin resistance, impaired glucose metabolism and type 2 diabetes mellitus. Alterations in diet composition may contribute to prevent and/or reverse these disturbances through modulation of fatty acid metabolism. Besides an increased fat mass, adipose tissue dysfunction, characterized by an altered capacity to store lipids and an altered secretion of adipokines, may result in lipid overflow, systemic inflammation and excessive lipid accumulation in non-adipose tissues like liver, skeletal muscle and the pancreas. These impairments together promote the development of impaired glucose metabolism, insulin resistance and type 2 diabetes mellitus. Furthermore, intrinsic functional impairments in either of these organs may contribute to lipotoxicity and insulin resistance. The present review provides an overview of fatty acid metabolism-related pathways in adipose tissue, liver, skeletal muscle, pancreas and gut, which can be targeted by diet or food components, thereby improving glucose metabolism.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Páncreas/metabolismo , Diabetes Mellitus Tipo 2/etiología , Dieta , Humanos , Obesidad/complicaciones , Receptor Cross-Talk
19.
J Clin Endocrinol Metab ; 89(6): 2690-6, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15181043

RESUMEN

The present study was designed to investigate the effects of angiotensin II (Ang II) on adipose and skeletal muscle tissue blood flow and lipolysis in normal-weight and obese subjects using the microdialysis technique. Microdialysis probes were placed in the abdominal sc adipose tissue left and right from the umbilicus and in the gastrocnemius muscle of both legs in eight normal-weight and eight obese men. Probes were consecutively perfused with 1.0 nM Ang II, 1.0 microM Ang II, and 1.0 microM Ang II + 48 microM hydralazine or with Ringer solution (control). Ethanol and glycerol concentrations in the dialysate were measured as an indicator of local blood flow and lipolysis, respectively. Ang II caused an increase in ethanol outflow/inflow ratio, compared with baseline values both in adipose tissue (average of both groups, Ang 1.0 nM: 0.03 +/- 0.01, P = 0.02; Ang 1.0 microM: 0.05 +/- 0.01, P < 0.01) and muscle (average of both groups, Ang 1.0 nM: 0.02 +/- 0.01, P = 0.09; Ang 1.0 microM: 0.04 +/- 0.01, P = 0.01), indicating a decrease in local blood flow. These effects were not significantly different in obese and normal-weight subjects. The decrease in local blood flow was accompanied by unchanged interstitial glycerol concentrations in adipose tissue (except during the supraphysiological dose) and skeletal muscle, suggesting that Ang II inhibits lipolysis in both tissues. Thus, the present data suggest that Ang II decreases local blood flow in a dose-dependent manner and inhibits lipolysis both in adipose and skeletal muscle tissue. These effects were not significantly different in obese and normal-weight subjects in both tissues.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Angiotensina II/administración & dosificación , Lipólisis/efectos de los fármacos , Músculo Esquelético/irrigación sanguínea , Obesidad/fisiopatología , Vasoconstrictores/administración & dosificación , Tejido Adiposo/metabolismo , Peso Corporal , Depresores del Sistema Nervioso Central/administración & dosificación , Interacciones Farmacológicas , Etanol/administración & dosificación , Glicerol/metabolismo , Humanos , Hidralazina/administración & dosificación , Masculino , Microdiálisis , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Vasodilatadores/administración & dosificación
20.
Obes Rev ; 4(1): 43-55, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12608526

RESUMEN

Angiotensin II (Ang II), acting on the AT1 and AT2 receptors in mammalian cells, is the vasoactive component of the renin-angiotensin system (RAS). Several components of the RAS have been demonstrated in different tissues, including adipose tissue. Although the effects of Ang II on metabolism have not been studied widely, it is intriguing to assume that components of the RAS produced by adipocytes may play an autocrine, a paracrine and/or an endocrine role in the pathophysiology of obesity and provide a potential pathway through which obesity leads to hypertension and type 2 diabetes mellitus. In the first part of this review, we will describe the production of Ang II, the different receptors through which Ang II exerts its effects and summarize the concomitant intracellular signalling cascades. Thereafter, potential Ang II-induced mechanisms, which may be associated with obesity and obesity-related disorders, will be considered. Finally, we will focus on the different pharmaceutical agents that interfere with the RAS and highlight the possible implications of these drugs in the treatment of obesity-related disorders.


Asunto(s)
Tejido Adiposo/fisiología , Tejido Adiposo/fisiopatología , Obesidad/fisiopatología , Receptores de Angiotensina/metabolismo , Sistema Renina-Angiotensina/fisiología , Angiotensina II/metabolismo , Animales , Humanos , Receptor de Angiotensina Tipo 1 , Receptor de Angiotensina Tipo 2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA