Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Hum Nutr Diet ; 35(5): 791-803, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-34967062

RESUMEN

BACKGROUND: Lifestyle intervention studies to treat type 2 diabetes (T2D) are on the rise. However, in-depth research is lacking into the dietary changes that participants make. METHODS: The present study aimed to observe the dietary intake of participants following the group program 'Reverse Diabetes2 Now' (RD2N) over 12 months. The web-based 24-h dietary recall-tool Compl-eat was used to collect dietary intake data. RESULTS: In total, 147 T2D patients were included in a cross-sectional study (n = 37 at baseline, n = 58 at 6 months, n = 52 at 12 months). A lower intake of total energy, carbohydrates and iodine was found for the groups at 6 and 12 months compared to the baseline group. The absolute consumption of total fat and saturated fat did not differ between the groups; only the percentage as total calorie consumption decreased. Consumption of vegetables and full-fat yoghurt was higher in groups at 6 and 12 months compared to the group at baseline. Consumption of bread, cakes and sweet biscuits, pasta/rice/tortillas, artificially sweetened soft drinks, and crisps were lower in the groups at 6 and 12 months compared to the group at baseline. Similar results were observed in a separate prospective study in 22 participants over 12 months following the same lifestyle-intervention. CONCLUSIONS: Overall, participants shifted their dietary intake somewhat towards a healthier dietary pattern with overall lower energy and carbohydrates and more vegetables. Moreover, participants largely maintained this healthier pattern over 12 months. There were some concerns regarding iodine intake. These promising results need to be confirmed in a fully-scaled study, as well in a comparison with controls.


Asunto(s)
Diabetes Mellitus Tipo 2 , Yodo , Estudios Transversales , Diabetes Mellitus Tipo 2/terapia , Carbohidratos de la Dieta , Grasas de la Dieta , Ingestión de Energía , Humanos , Estilo de Vida , Estudios Prospectivos , Edulcorantes , Verduras
2.
BMJ Nutr Prev Health ; 2(1): 43-50, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-33235957

RESUMEN

INTRODUCTION: Prevalence of type 2 diabetes (T2D) is increasing rapidly and lifestyle interventions to reverse diabetes are seen as a possible solution to stop this trend. New practice-based evidence is needed to gain more insight in the actual, and above all scientific, basis for these claims. METHODS: This observational study with a pretest post-test design aimed to pilot a 6-month multicomponent outpatient group-based nutrition and lifestyle intervention programme on glycaemic control and use of glucose lowering medication in motivated T2D patients with a body mass index (BMI) >25 kg/m2 in the Netherlands (February 2015-March 2016). RESULTS: 74 T2D patients (56% female) aged 57.4±8.0 years with mean BMI 31.2±4.2 kg/m2 and mean waist circumference 105.4±10.2 cm were included in the study. Compared with baseline, mean HbA1c levels at 6 months were 5 mmol/mol lower (SD=10, p<0.001) and the number of participants with HbA1c levels ≤53 mmol/mol after intervention had increased (from 36% (n=26/72) to 60% (n=43/72)). At baseline, 90% of participants were taking at least one type of glucose lowering medication. At 6 months, 49% (n=35/72) of the participants had reduced their medication or eliminated it completely (13%). Secondary outcomes were significantly lower fasting glucose levels (- 1.2±2.6 mmol/L), body weight (-4.9±5.1 kg), BMI (-1.70±1.69 kg/m2) and waist circumference (-9.4±5.0 cm). Plasma lipids remained unchanged except for a decrease in triglyceride levels. Furthermore, self-reported quality of life was significantly higher while experienced fatigue and sleep problems were significantly lower. CONCLUSION: This pilot study showed that a 6-month multicomponent group-based program in a routine care setting could improve glycaemic control and reduce the use of glucose lowering medication in motivated T2D diabetics. A fully scaled study is needed to confirm these results.

3.
Nat Commun ; 8(1): 1804, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-29180704

RESUMEN

Vps34 PI3K is thought to be the main producer of phosphatidylinositol-3-monophosphate, a lipid that controls intracellular vesicular trafficking. The organismal impact of systemic inhibition of Vps34 kinase activity is not completely understood. Here we show that heterozygous Vps34 kinase-dead mice are healthy and display a robustly enhanced insulin sensitivity and glucose tolerance, phenotypes mimicked by a selective Vps34 inhibitor in wild-type mice. The underlying mechanism of insulin sensitization is multifactorial and not through the canonical insulin/Akt pathway. Vps34 inhibition alters cellular energy metabolism, activating the AMPK pathway in liver and muscle. In liver, Vps34 inactivation mildly dampens autophagy, limiting substrate availability for mitochondrial respiration and reducing gluconeogenesis. In muscle, Vps34 inactivation triggers a metabolic switch from oxidative phosphorylation towards glycolysis and enhanced glucose uptake. Our study identifies Vps34 as a new drug target for insulin resistance in Type-2 diabetes, in which the unmet therapeutic need remains substantial.


Asunto(s)
Resistencia a la Insulina , Mitocondrias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia/fisiología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasas Clase III , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Técnicas de Sustitución del Gen , Glucosa/análisis , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Glucólisis/fisiología , Hepatocitos , Heterocigoto , Humanos , Insulina/metabolismo , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Cultivo Primario de Células
4.
Cell Metab ; 26(1): 212-229.e12, 2017 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-28683288

RESUMEN

Thyroid hormones (THs) act in the brain to modulate energy balance. We show that central triiodothyronine (T3) regulates de novo lipogenesis in liver and lipid oxidation in brown adipose tissue (BAT) through the parasympathetic (PSNS) and sympathetic nervous system (SNS), respectively. Central T3 promotes hepatic lipogenesis with parallel stimulation of the thermogenic program in BAT. The action of T3 depends on AMP-activated protein kinase (AMPK)-induced regulation of two signaling pathways in the ventromedial nucleus of the hypothalamus (VMH): decreased ceramide-induced endoplasmic reticulum (ER) stress, which promotes BAT thermogenesis, and increased c-Jun N-terminal kinase (JNK) activation, which controls hepatic lipid metabolism. Of note, ablation of AMPKα1 in steroidogenic factor 1 (SF1) neurons of the VMH fully recapitulated the effect of central T3, pointing to this population in mediating the effect of central THs on metabolism. Overall, these findings uncover the underlying pathways through which central T3 modulates peripheral metabolism.


Asunto(s)
Metabolismo Energético , Hipotálamo/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Transducción de Señal , Hormonas Tiroideas/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Termogénesis , Triyodotironina/metabolismo
5.
FASEB J ; 31(3): 989-997, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27895108

RESUMEN

We investigated the independent and interactive impact of the common APOE genotype and marine n-3 polyunsaturated fatty acids (PUFAs) on the development of obesity and associated cardiometabolic dysfunction in a murine model. Human APOE3 and APOE4 targeted replacement mice were fed either a control high-fat diet (HFD) or an HFD supplemented with 3% n-3 PUFAs from fish oil (HFD + FO) for 8 wk. We established the impact of intervention on food intake, body weight, and visceral adipose tissue (VAT) mass; plasma, lipids (cholesterol and triglycerides), liver enzymes, and adipokines; glucose and insulin during an intraperitoneal glucose tolerance test; and Glut4 and ApoE expression in VAT. HFD feeding induced more weight gain and higher plasma lipids in APOE3 compared to APOE4 mice (P < 0.05), along with a 2-fold higher insulin and impaired glucose tolerance. Supplementing APOE3, but not APOE4, animals with dietary n-3 PUFAs decreased body-weight gain, plasma lipids, and insulin (P < 0.05) and improved glucose tolerance, which was associated with increased VAT Glut4 mRNA levels (P < 0.05). Our findings demonstrate that an APOE3 genotype predisposes mice to develop obesity and its metabolic complications, which was attenuated by n-3 PUFA supplementation.-Slim, K. E., Vauzour, D., Tejera, N., Voshol, P. J., Cassidy, A., Minihane, A. M. The effect of dietary fish oil on weight gain and insulin sensitivity is dependent on APOE genotype in humanized targeted replacement mice.


Asunto(s)
Apolipoproteínas E/genética , Aceites de Pescado/farmacología , Genotipo , Resistencia a la Insulina , Obesidad/prevención & control , Aumento de Peso/efectos de los fármacos , Alelos , Animales , Apolipoproteínas E/metabolismo , Dieta Alta en Grasa/efectos adversos , Aceites de Pescado/uso terapéutico , Humanos , Grasa Intraabdominal/efectos de los fármacos , Masculino , Ratones , Obesidad/etiología , Obesidad/genética , Aumento de Peso/genética
6.
Proc Natl Acad Sci U S A ; 112(45): 13850-5, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26508640

RESUMEN

Adipose triglyceride lipase (ATGL) initiates intracellular triglyceride (TG) catabolism. In humans, ATGL deficiency causes neutral lipid storage disease with myopathy (NLSDM) characterized by a systemic TG accumulation. Mice with a genetic deletion of ATGL (AKO) also accumulate TG in many tissues. However, neither NLSDM patients nor AKO mice are exceedingly obese. This phenotype is unexpected considering the importance of the enzyme for TG catabolism in white adipose tissue (WAT). In this study, we identified the counteracting mechanisms that prevent excessive obesity in the absence of ATGL. We used "healthy" AKO mice expressing ATGL exclusively in cardiomyocytes (AKO/cTg) to circumvent the cardiomyopathy and premature lethality observed in AKO mice. AKO/cTg mice were protected from high-fat diet (HFD)-induced obesity despite complete ATGL deficiency in WAT and normal adipocyte differentiation. AKO/cTg mice were highly insulin sensitive under hyperinsulinemic-euglycemic clamp conditions, eliminating insulin insensitivity as a possible protective mechanism. Instead, reduced food intake and altered signaling by peroxisome proliferator-activated receptor-gamma (PPAR-γ) and sterol regulatory element binding protein-1c in WAT accounted for the phenotype. These adaptations led to reduced lipid synthesis and storage in WAT of HFD-fed AKO/cTg mice. Treatment with the PPAR-γ agonist rosiglitazone reversed the phenotype. These results argue for the existence of an adaptive interdependence between lipolysis and lipid synthesis. Pharmacological inhibition of ATGL may prove useful to prevent HFD-induced obesity and insulin resistance.


Asunto(s)
Adaptación Fisiológica , Dieta Alta en Grasa , Conducta Alimentaria , Lipasa/fisiología , Lipólisis , Obesidad/prevención & control , Animales , Lipasa/genética , Ratones , Ratones Noqueados , Obesidad/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Fenotipo
7.
PLoS One ; 10(7): e0131859, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26176620

RESUMEN

BACKGROUND: Obesity is associated with a hypercoagulable state and increased risk for thrombotic cardiovascular events. OBJECTIVE: Establish the onset and reversibility of the hypercoagulable state during the development and regression of nutritionally-induced obesity in mice, and its relation to transcriptional changes and clearance rates of coagulation factors as well as its relation to changes in metabolic and inflammatory parameters. METHODS: Male C57BL/6J mice were fed a low fat (10% kcal as fat; LFD) or high fat diet (45% kcal as fat; HFD) for 2, 4, 8 or 16 weeks. To study the effects of weight loss, mice were fed the HFD for 16 weeks and switched to the LFD for 1, 2 or 4 weeks. For each time point analyses of plasma and hepatic mRNA levels of coagulation factors were performed after overnight fasting, as well as measurements of circulating metabolic and inflammatory parameters. Furthermore, in vivo clearance rates of human factor (F) VII, FVIII and FIX proteins were determined after 2 weeks of HFD-feeding. RESULTS: HFD feeding gradually increased the body and liver weight, which was accompanied by a significant increase in plasma glucose levels from 8 weeks onwards, while insulin levels were affected after 16 weeks. Besides a transient rise in cytokine levels at 2 weeks after starting the HFD, no significant effect on inflammation markers was present. Increased plasma levels of fibrinogen, FII, FVII, FVIII, FIX, FXI and FXII were observed in mice on a HFD for 2 weeks, which in general persisted throughout the 16 weeks of HFD-feeding. Interestingly, with the exception of FXI the effects on plasma coagulation levels were not paralleled by changes in relative transcript levels in the liver, nor by decreased clearance rates. Switching from HFD to LFD reversed the HFD-induced procoagulant shift in plasma, again not coinciding with transcriptional modulation. CONCLUSIONS: Changes in dietary fat content rapidly alter the mouse plasma coagulation profile, thereby preceding plasma metabolic changes, which cannot be explained by changes in relative expression of coagulation factors or decreased clearance rates.


Asunto(s)
Factores de Coagulación Sanguínea/análisis , Dieta con Restricción de Grasas , Grasas de la Dieta/sangre , Animales , Citocinas/sangre , Dieta Alta en Grasa , Grasas de la Dieta/metabolismo , Factor IX/análisis , Factor VII/análisis , Factor VIII/análisis , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/etiología , Tiempo de Tromboplastina Parcial , Tiempo de Protrombina , ARN/aislamiento & purificación , ARN/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Cell Rep ; 11(3): 335-43, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25865886

RESUMEN

Hypothalamic ribosomal S6K1 has been suggested as a point of convergence for hormonal and nutrient signals in the regulation of feeding behavior, bodyweight, and glucose metabolism. However, the long-term effects of manipulating hypothalamic S6K1 signaling on energy homeostasis and the cellular mechanisms underlying these roles are unclear. We therefore inactivated S6K1 in pro-opiomelanocortin (POMC) and agouti-related protein (AgRP) neurons, key regulators of energy homeostasis, but in contrast to the current view, we found no evidence that S6K1 regulates food intake and bodyweight. In contrast, S6K1 signaling in POMC neurons regulated hepatic glucose production and peripheral lipid metabolism and modulated neuronal excitability. S6K1 signaling in AgRP neurons regulated skeletal muscle insulin sensitivity and was required for glucose sensing by these neurons. Our findings suggest that S6K1 signaling is not a general integrator of energy homeostasis in the mediobasal hypothalamus but has distinct roles in the regulation of glucose homeostasis by POMC and AgRP neurons.


Asunto(s)
Metabolismo Energético/fisiología , Conducta Alimentaria/fisiología , Glucosa/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Peso Corporal/fisiología , Homeostasis/fisiología , Resistencia a la Insulina/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proopiomelanocortina/metabolismo , Transducción de Señal/fisiología
9.
Diabetes ; 63(9): 2984-95, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24812426

RESUMEN

Dysregulated glucagon secretion accompanies islet inflammation in type 2 diabetes. We recently discovered that interleukin (IL)-6 stimulates glucagon secretion from human and rodent islets. IL-6 family cytokines require the glycoprotein 130 (gp130) receptor to signal. In this study, we elucidated the effects of α-cell gp130 receptor signaling on glycemic control in type 2 diabetes. IL-6 family cytokines were elevated in islets in rodent models of this disease. gp130 receptor activation increased STAT3 phosphorylation in primary α-cells and stimulated glucagon secretion. Pancreatic α-cell gp130 knockout (αgp130KO) mice showed no differences in glycemic control, α-cell function, or α-cell mass. However, when subjected to streptozotocin plus high-fat diet to induce islet inflammation and pathophysiology modeling type 2 diabetes, αgp130KO mice had reduced fasting glycemia, improved glucose tolerance, reduced fasting insulin, and improved α-cell function. Hyperinsulinemic-euglycemic clamps revealed no differences in insulin sensitivity. We conclude that in a setting of islet inflammation and pathophysiology modeling type 2 diabetes, activation of α-cell gp130 receptor signaling has deleterious effects on α-cell function, promoting hyperglycemia. Antagonism of α-cell gp130 receptor signaling may be useful for the treatment of type 2 diabetes.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/fisiopatología , Células Secretoras de Glucagón/metabolismo , Animales , Receptor gp130 de Citocinas/antagonistas & inhibidores , Dieta Alta en Grasa , Glucagón/metabolismo , Interleucina-6/metabolismo , Interleucina-6/farmacología , Masculino , Ratones , Ratones Noqueados , Fosforilación , Ratas , Factor de Transcripción STAT3/metabolismo
10.
J Lipid Res ; 55(7): 1434-47, 2014 07.
Artículo en Inglés | MEDLINE | ID: mdl-24837748

RESUMEN

Here, we investigated how LDL receptor deficiency (Ldlr(-/-)) modulates the effects of testosterone on obesity and related metabolic dysfunctions. Though sham-operated Ldlr(-/-) mice fed Western-type diet for 12 weeks became obese and showed disturbed plasma glucose metabolism and plasma cholesterol and TG profiles, castrated mice were resistant to diet-induced obesity and had improved glucose metabolism and reduced plasma TG levels, despite a further deterioration in their plasma cholesterol profile. The effect of hypogonadism on diet-induced weight gain of Ldlr(-/-) mice was independent of ApoE and Lrp1. Indirect calorimetry analysis indicated that hypogonadism in Ldlr(-/-) mice was associated with increased metabolic rate. Indeed, mitochondrial cytochrome c and uncoupling protein 1 expression were elevated, primarily in white adipose tissue, confirming increased mitochondrial metabolic activity due to thermogenesis. Testosterone replacement in castrated Ldlr(-/-) mice for a period of 8 weeks promoted diet-induced obesity, indicating a direct role of testosterone in the observed phenotype. Treatment of sham-operated Ldlr(-/-) mice with the aromatase inhibitor exemestane for 8 weeks showed that the obesity of castrated Ldlr(-/-) mice is independent of estrogens. Overall, our data reveal a novel role of Ldlr as functional modulator of metabolic alterations associated with hypogonadism.


Asunto(s)
Grasas de la Dieta/efectos adversos , Hipogonadismo/metabolismo , Obesidad/metabolismo , Receptores de LDL/metabolismo , Testosterona/metabolismo , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Grasas de la Dieta/farmacología , Hipogonadismo/inducido químicamente , Hipogonadismo/genética , Hipogonadismo/patología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Receptores de LDL/genética , Testosterona/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 34(6): 1187-92, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24675658

RESUMEN

OBJECTIVE: Platelets abundantly express the membrane receptor CD36 and store its ligand thrombospondin-1 (TSP1) in the α-granules. We investigated whether released TSP1 can support platelet adhesion and thrombus formation via interaction with CD36. APPROACH AND RESULTS: Mouse platelets deficient in CD36 showed reduced adhesion to TSP1 and subsequent phosphatidylserine expression. Deficiency in either CD36 or TSP1 resulted in markedly increased dissolution of thrombi formed on collagen, although thrombus buildup was unchanged. In mesenteric vessels in vivo, deficiency in CD36 prolonged the time to occlusion and enhanced embolization, which was in agreement with earlier observations in TSP1-deficient mice. Thrombi formed using wild-type blood stained positively for secreted TSP1. Releasate from wild-type but not from TSP1-deficient platelets enhanced platelet activation, phosphatidylserine expression, and thrombus formation on collagen. The enhancement was dependent on CD36 because it was without effect on thrombus formation by CD36-deficient platelets. CONCLUSIONS: These results demonstrate an anchoring role of platelet-released TSP1 via CD36 in platelet adhesion and collagen-dependent thrombus stabilization. Thus, the TSP1-CD36 tandem is another platelet ligand-receptor axis contributing to the maintenance of a stable thrombus.


Asunto(s)
Antígenos CD36/fisiología , Colágeno/metabolismo , Trombosis/etiología , Trombospondina 1/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Activación Plaquetaria , Adhesividad Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología
12.
Proc Natl Acad Sci U S A ; 111(11): E1043-52, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591600

RESUMEN

Physical activity increases energy metabolism in exercising muscle. Whether acute exercise elicits metabolic changes in nonexercising muscles remains unclear. We show that one of the few genes that is more highly induced in nonexercising muscle than in exercising human muscle during acute exercise encodes angiopoietin-like 4 (ANGPTL4), an inhibitor of lipoprotein lipase-mediated plasma triglyceride clearance. Using a combination of human, animal, and in vitro data, we show that induction of ANGPTL4 in nonexercising muscle is mediated by elevated plasma free fatty acids via peroxisome proliferator-activated receptor-δ, presumably leading to reduced local uptake of plasma triglyceride-derived fatty acids and their sparing for use by exercising muscle. In contrast, the induction of ANGPTL4 in exercising muscle likely is counteracted via AMP-activated protein kinase (AMPK)-mediated down-regulation, promoting the use of plasma triglycerides as fuel for active muscles. Our data suggest that nonexercising muscle and the local regulation of ANGPTL4 via AMPK and free fatty acids have key roles in governing lipid homeostasis during exercise.


Asunto(s)
Angiopoyetinas/metabolismo , Ejercicio Físico/fisiología , Homeostasis/fisiología , Metabolismo de los Lípidos/fisiología , Músculo Esquelético/fisiología , Adulto , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/sangre , Angiopoyetinas/fisiología , Ácidos Grasos/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Análisis por Micromatrices , Persona de Mediana Edad
13.
Obesity (Silver Spring) ; 21(10): 2037-45, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23512311

RESUMEN

OBJECTIVE: The scavenger receptor CD36 facilitates the cellular uptake of long-chain fatty acids. As CD36-deficiency attenuates the development of high fat diet (HFD)-induced obesity, the role of CD36-deficiency in preadipocyte recruitment and adipocyte function was set out to characterize. DESIGN AND METHODS: Fat cell size and number were determined in gonadal, visceral, and subcutaneous adipose tissue of CD36(-/-) and WT mice after 6 weeks on HFD. Basal lipolysis and insulin-inhibited lipolysis were investigated in gonadal adipose tissue. RESULTS: CD36(-/-) mice showed a reduction in adipocyte size in all fat pads. Gonadal adipose tissue also showed a lower total number of adipocytes because of a lower number of very small adipocytes (diameter <50 µm). This was accompanied by an increased pool of preadipocytes, which suggests that CD36-deficiency reduces the capacity of preadipocytes to become adipocytes. Regarding lipolysis, in adipose tissue from CD36(-/-) mice, cAMP levels were increased and both basal and 8-bromo-cAMP stimulated lipolysis were higher. However, insulin-mediated inhibition of lipolysis was more potent in CD36(-/-) mice. CONCLUSIONS: These results indicate that during fat depot expansion, CD36-deficiency negatively affects preadipocyte recruitment and that in mature adipocytes, CD36-deficiency is associated with increased basal lipolysis and insulin responsiveness.


Asunto(s)
Adipocitos/fisiología , Antígenos CD36/fisiología , Lipólisis/fisiología , 8-Bromo Monofosfato de Adenosina Cíclica/metabolismo , Tejido Adiposo/metabolismo , Animales , Antígenos CD36/deficiencia , Diferenciación Celular , Tamaño de la Célula , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos/metabolismo , Insulina/metabolismo , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Triglicéridos/metabolismo
14.
PLoS One ; 8(2): e56922, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23441226

RESUMEN

AIMS: Beta cells adapt to an increased insulin demand by enhancing insulin secretion via increased beta cell function and/or increased beta cell number. While morphological and functional heterogeneity between individual islets exists, it is unknown whether regional differences in beta cell adaptation occur. Therefore we investigated beta cell adaptation throughout the pancreas in a model of high-fat diet (HFD)-induced insulin resistance in mice. METHODS: C57BL/6J mice were fed a HFD to induce insulin resistance, or control diet for 6 weeks. The pancreas was divided in a duodenal (DR), gastric (GR) and splenic (SR) region and taken for either histology or islet isolation. The capacity of untreated islets from the three regions to adapt in an extrapancreatic location was assessed by transplantation under the kidney capsule of streptozotocin-treated mice. RESULTS: SR islets showed 70% increased beta cell proliferation after HFD, whereas no significant increase was found in DR and GR islets. Furthermore, isolated SR islets showed twofold enhanced glucose-induced insulin secretion after HFD, as compared with DR and GR islets. In contrast, transplantation of islets isolated from the three regions to an extrapancreatic location in diabetic mice led to a similar decrease in hyperglycemia and no difference in beta cell proliferation. CONCLUSIONS: HFD-induced insulin resistance leads to topologically heterogeneous beta cell adaptation and is most prominent in the splenic region of the pancreas. This topological heterogeneity in beta cell adaptation appears to result from extrinsic factors present in the islet microenvironment.


Asunto(s)
Adaptación Fisiológica , Dieta Alta en Grasa , Células Secretoras de Insulina/metabolismo , Animales , Proliferación Celular , Glucosa/metabolismo , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/trasplante , Masculino , Ratones , Páncreas/metabolismo
15.
Nutrition ; 29(1): 276-83, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22985971

RESUMEN

OBJECTIVE: We investigated whether a reduced iso-α acid derived from an extract of Humulus lupulus L., META060, had an effect on weight gain, body composition, and metabolism in a high-fat-diet (HFD) fed mouse model. METHODS: Weight gain was monitored for up to 20 wk in mice receiving a low-fat diet, an HFD, or an HFD supplemented with META060 or rosiglitazone. Body composition was determined using dual-energy x-ray absorptiometric analysis. Indirect calorimetric measurements were performed to investigate the energy balance in the mice, and oral glucose tolerance tests were administered to examine the effect of META060 on the glycemic response. RESULTS: The HFD-fed mice administered META060 for 14 wk had a significantly lower mean weight than HFD-fed mice (30.58 ± 0.5 versus 37.88 ± 0.7 g, P < 0.05). Indirect calorimetric measurements showed an increased metabolic flexibility in mice supplemented with META060. In addition, glucose tolerance was improved, comparable to the effects of rosiglitazone treatment. CONCLUSIONS: META060 has potential therapeutic value for managing obesity and insulin resistance, and further research into the mechanism of action is warranted.


Asunto(s)
Humulus , Resistencia a la Insulina , Obesidad/prevención & control , Animales , Composición Corporal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Prueba de Tolerancia a la Glucosa , Humulus/química , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Fitoterapia , Extractos Vegetales/química , Extractos Vegetales/farmacología , Rosiglitazona , Tiazolidinedionas/farmacología
16.
Proc Natl Acad Sci U S A ; 109(51): 20943-8, 2012 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-23213245

RESUMEN

Synucleins are a family of homologous proteins principally known for their involvement in neurodegeneration. γ-Synuclein is highly expressed in human white adipose tissue and increased in obesity. Here we show that γ-synuclein is nutritionally regulated in white adipose tissue whereas its loss partially protects mice from high-fat diet (HFD)-induced obesity and ameliorates some of the associated metabolic complications. Compared with HFD-fed WT mice, HFD-fed γ-synuclein-null mutant mice display increased lipolysis, lipid oxidation, and energy expenditure, and reduced adipocyte hypertrophy. Knockdown of γ-synuclein in adipocytes causes redistribution of the key lipolytic enzyme ATGL to lipid droplets and increases lipolysis. γ-Synuclein-deficient adipocytes also contain fewer SNARE complexes of a type involved in lipid droplet fusion. We hypothesize that γ-synuclein may deliver SNAP-23 to the SNARE complexes under lipogenic conditions. Via these independent but complementary roles, γ-synuclein may coordinately modulate lipid storage by influencing lipolysis and lipid droplet formation. Our data reveal γ-synuclein as a regulator of lipid handling in adipocytes, the function of which is particularly important in conditions of nutrient excess.


Asunto(s)
Tejido Adiposo/metabolismo , Lipólisis , Obesidad/metabolismo , Células 3T3 , Adipocitos/citología , Animales , Dieta , Genotipo , Lípidos/química , Lipogénesis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , gamma-Sinucleína
17.
Atherosclerosis ; 220(2): 362-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21798539

RESUMEN

OBJECTIVE: The liver is the key organ involved in systemic inflammation, but the relation between hepatic inflammation and atherogenesis is poorly understood. Since nuclear factor-κB (NF-κB) is a central regulator of inflammatory processes, we hypothesized that chronically enhanced hepatic NF-κB activation, through hepatocyte-specific expression of IκB kinase-ß (IKKß) (LIKK), will aggravate atherosclerosis development in APOE*3-Leiden (E3L) mice. METHODS AND RESULTS: E3L.LIKK and E3L control littermates were fed a Western-type diet for 24 weeks. E3L.LIKK mice showed a 2.3-fold increased atherosclerotic lesion area and more advanced atherosclerosis in the aortic root with less segments without atherosclerotic lesions (11% vs. 42%), and more segments with mild (63% vs. 44%) and severe (26% vs. 14%) lesions. Expression of LIKK did not affect basal levels of inflammatory parameters, but plasma cytokine levels tended to be higher in E3L.LIKK mice after lipopolysaccharide (LPS) administration. E3L.LIKK mice showed transiently increased plasma cholesterol levels, confined to (V)LDL. This transient character resulted in a mild (+17%) increased cumulative plasma cholesterol exposure. CONCLUSION: We conclude that selective activation of NF-κB in hepatocytes considerably promotes atherosclerosis development which is (at least partly) explained by an increased sensitivity to proinflammatory triggers and transiently increased plasma cholesterol levels.


Asunto(s)
Enfermedades de la Aorta/enzimología , Apolipoproteína E3/metabolismo , Aterosclerosis/enzimología , Hepatocitos/enzimología , Quinasa I-kappa B/metabolismo , Inflamación/enzimología , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Apolipoproteína E3/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Biomarcadores/sangre , VLDL-Colesterol/sangre , Citocinas/sangre , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Humanos , Quinasa I-kappa B/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/sangre , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Factores de Tiempo , Regulación hacia Arriba
18.
Metabolism ; 61(1): 99-107, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21816446

RESUMEN

Obesity and its associated conditions such as type 2 diabetes mellitus are major causes of morbidity and mortality. The iminosugar N-(5-adamantane-1-yl-methoxy-pentyl)-deoxynojirimycin (AMP-DNM) improves insulin sensitivity in rodent models of insulin resistance and type 2 diabetes mellitus. In the current study, we characterized the impact of AMP-DNM on substrate oxidation patterns, food intake, and body weight gain in obese mice. Eight ob/ob mice treated with 100 mg/(kg d) AMP-DNM mixed in the food and 8 control ob/ob mice were placed in metabolic cages during the first, third, and fifth week of the experiment for measurement of substrate oxidation rates, energy expenditure, activity, and food intake. Mice were killed after 6 weeks of treatment. Initiation of treatment with AMP-DNM resulted in a rapid increase in fat oxidation by 129% (P = .05), a decrease in carbohydrate oxidation by 35% (P = .01), and a reduction in food intake by approximately 26% (P < .01) compared with control mice. Treatment with AMP-DNM decreased hepatic triglyceride content by 66% (P < .01) and, in line with the elevated fat oxidation rates, increased hepatic carnitine palmitoyl transferase 1a expression. Treatment with AMP-DNM increased plasma levels of the appetite-regulating peptide YY compared with control mice. Treatment with AMP-DNM rapidly reduces food intake and increases fat oxidation, resulting in improvement of the obese phenotype. These features of AMP-DNM, together with its insulin-sensitizing capacity, make it an attractive candidate drug for the treatment of obesity and its associated metabolic derangements.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Adamantano/análogos & derivados , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , 1-Desoxinojirimicina/farmacología , Adamantano/farmacología , Tejido Adiposo/metabolismo , Animales , Metabolismo de los Hidratos de Carbono/efectos de los fármacos , Carnitina O-Palmitoiltransferasa/metabolismo , Ghrelina/metabolismo , Glucosa/metabolismo , Iminoazúcares/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/genética , Oxidación-Reducción , Péptido YY/metabolismo , Triglicéridos/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
19.
Am J Physiol Endocrinol Metab ; 301(6): E1099-107, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21862721

RESUMEN

Systemic inflammation is strongly involved in the pathophysiology of the metabolic syndrome, a cluster of metabolic risk factors that includes hypertriglyceridemia. Aspirin treatment lowers inflammation via inhibition of NF-κB activity but also reduces hypertriglyceridemia in humans. The aim of this study was to investigate the mechanism by which aspirin improves hypertriglyceridemia. Human apolipoprotein CI (apoCI)-expressing mice (APOC1 mice), an animal model with elevated plasma triglyceride (TG) levels, as well as normolipidemic wild-type (WT) mice were fed a high-fat diet (HFD) and treated with aspirin. Aspirin treatment reduced hepatic NF-κB activity in HFD-fed APOC1 and WT mice, and in addition, aspirin decreased plasma TG levels (-32%, P < 0.05) in hypertriglyceridemic APOC1 mice. This TG-lowering effect could not be explained by enhanced VLDL-TG clearance, but aspirin selectively reduced hepatic production of VLDL-TG in both APOC1 (-28%, P < 0.05) and WT mice (-33%, P < 0.05) without affecting VLDL-apoB production. Aspirin did not alter hepatic expression of genes involved in FA oxidation, lipogenesis, and VLDL production but decreased the incorporation of plasma-derived FA by the liver into VLDL-TG (-24%, P < 0.05), which was independent of hepatic expression of genes involved in FA uptake and transport. We conclude that aspirin improves hypertriglyceridemia by decreasing VLDL-TG production without affecting VLDL particle production. Therefore, the inhibition of inflammatory pathways by aspirin could be an interesting target for the treatment of hypertriglyceridemia.


Asunto(s)
Aspirina/farmacología , Dieta Alta en Grasa , Hipertrigliceridemia/prevención & control , Lipoproteínas VLDL/metabolismo , Triglicéridos/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Apolipoproteína C-I/genética , Aspirina/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Hipertrigliceridemia/sangre , Hipertrigliceridemia/etiología , Hipertrigliceridemia/metabolismo , Lipoproteínas VLDL/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Triglicéridos/sangre
20.
J Lipid Res ; 52(9): 1712-22, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21700834

RESUMEN

Insulin signaling in the central nervous system (CNS) is required for the inhibitory effect of insulin on glucose production. Our aim was to determine whether the CNS is also involved in the stimulatory effect of circulating insulin on the tissue-specific retention of fatty acid (FA) from plasma. In wild-type mice, hyperinsulinemic-euglycemic clamp conditions stimulated the retention of both plasma triglyceride-derived FA and plasma albumin-bound FA in the various white adipose tissues (WAT) but not in other tissues, including brown adipose tissue (BAT). Intracerebroventricular (ICV) administration of insulin induced a similar pattern of tissue-specific FA partitioning. This effect of ICV insulin administration was not associated with activation of the insulin signaling pathway in adipose tissue. ICV administration of tolbutamide, a K(ATP) channel blocker, considerably reduced (during hyperinsulinemic-euglycemic clamp conditions) and even completely blocked (during ICV administration of insulin) WAT-specific retention of FA from plasma. This central effect of insulin was absent in CD36-deficient mice, indicating that CD36 is the predominant FA transporter in insulin-stimulated FA retention by WAT. In diet-induced insulin-resistant mice, these stimulating effects of insulin (circulating or ICV administered) on FA retention in WAT were lost. In conclusion, in insulin-sensitive mice, circulating insulin stimulates tissue-specific partitioning of plasma-derived FA in WAT in part through activation of K(ATP) channels in the CNS. Apparently, circulating insulin stimulates fatty acid uptake in WAT but not in BAT, directly and indirectly through the CNS.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Sistema Nervioso Central/metabolismo , Ácidos Grasos/metabolismo , Insulina/sangre , Canales KATP/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Dieta , Insulina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/fisiopatología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...