Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Nutr ; 152(8): 1862-1871, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35511216

RESUMEN

BACKGROUND: The role of hepatoportal glucose sensors is poorly understood in the context of insulin resistance. OBJECTIVES: We assessed the effects of glucose infusion in the portal vein on insulin tolerance in 2 rat models of insulin resistance, and the role of capsaicin sensitive nerves in this signal. METHODS: Male Wistar rats, 8 weeks old, weighing 250-275 g, were used. Insulin and glucose tolerance were assessed following a 4-hour infusion of either glucose or saline through catheterization in the portal vein in 3 paradigms. In experiment 1, for diet-induced insulin resistance, rats were fed either a control diet (energy content: proteins = 22.5%, carbohydrates = 64.1%, and lipids = 13.4%) or a high-fat diet (energy content: proteins = 15.3%, carbohydrates = 40.3%, and lipids =44.4%) for 4 months. In experiment 2, for centrally induced peripheral insulin resistance, catheters were inserted in the carotid artery to deliver either an emulsion of triglycerides [intralipid (IL)] or saline towards the brain for 24 hours. In experiment 3, for testing the role of capsaicin-sensitive nerves, experiment 2 was repeated following a periportal treatment with capsaicin or vehicle. RESULTS: In experiment 1, when compared to rats fed the control diet, rats fed the high-fat diet exhibited decreased insulin and glucose tolerance (P ≤ 0.05) that was restored with a glucose infusion in the portal vein (P ≤ 0.05). In experiment 2, infusion of a triglyceride emulsion towards the brain (IL rats) decreased insulin and glucose tolerance and increased hepatic endogenous production when compared to saline-infused rats (P ≤ 0.05). Glucose infusion in the portal vein in IL rats restored insulin and glucose tolerance, as well as hepatic glucose production, to controls levels (P ≤ 0.05). In experiment 3, portal infusion of glucose did not increase insulin tolerance in IL rats that received a periportal pretreatment with capsaicin. CONCLUSIONS: Stimulation of hepatoportal glucose sensors increases insulin tolerance in rat models of insulin resistance and requires the presence of capsaicin-sensitive nerves.


Asunto(s)
Resistencia a la Insulina , Insulina , Animales , Glucemia/metabolismo , Capsaicina/metabolismo , Capsaicina/farmacología , Emulsiones/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Insulina Regular Humana/farmacología , Hígado/metabolismo , Masculino , Fibras Nerviosas/metabolismo , Vena Porta/metabolismo , Ratas , Ratas Wistar , Triglicéridos/metabolismo
2.
Diabetes ; 70(9): 2067-2080, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34078628

RESUMEN

Excess chronic contact between microbial motifs and intestinal immune cells is known to trigger a low-grade inflammation involved in many pathologies such as obesity and diabetes. The important skewing of intestinal adaptive immunity in the context of diet-induced obesity (DIO) is well described, but how dendritic cells (DCs) participate in these changes is still poorly documented. To address this question, we challenged transgenic mice with enhanced DC life span and immunogenicity (DChBcl-2 mice) with a high-fat diet. Those mice display resistance to DIO and metabolic alterations. The DIO-resistant phenotype is associated with healthier parameters of intestinal barrier function and lower intestinal inflammation. DChBcl-2 DIO-resistant mice demonstrate a particular increase in tolerogenic DC numbers and function, which is associated with strong intestinal IgA, T helper 17, and regulatory T-cell immune responses. Microbiota composition and function analyses reveal that the DChBcl-2 mice microbiota is characterized by lower immunogenicity and an enhanced butyrate production. Cohousing experiments and fecal microbial transplantations are sufficient to transfer the DIO resistance status to wild-type mice, demonstrating that maintenance of DCs' tolerogenic ability sustains a microbiota able to drive DIO resistance. The tolerogenic function of DCs is revealed as a new potent target in metabolic disease management.


Asunto(s)
Células Dendríticas/metabolismo , Microbioma Gastrointestinal/fisiología , Inflamación/metabolismo , Enfermedades Metabólicas/metabolismo , Obesidad/metabolismo , Animales , Células Dendríticas/patología , Dieta Alta en Grasa , Inflamación/patología , Masculino , Enfermedades Metabólicas/patología , Ratones , Ratones Transgénicos , Obesidad/patología
3.
Diabetes ; 70(1): 76-90, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33139329

RESUMEN

Besides cytoplasmic lipase-dependent adipocyte fat mobilization, the metabolic role of lysosomal acid lipase (LAL), highly expressed in adipocytes, is unclear. We show that the isolated adipocyte fraction, but not the total undigested adipose tissue (ATs), from obese patients has decreased LAL expression compared with that from nonobese people. Lentiviral-mediated LAL knockdown in the 3T3L1 mouse cell line to mimic the obese adipocytes condition did not affect lysosome density or autophagic flux, but it did increase triglyceride storage and disrupt endoplasmic reticulum cholesterol, as indicated by activated SREBP. Conversely, mice with adipose-specific LAL overexpression (Adpn-rtTA x TetO-hLAL) gained less weight and body fat than did control mice fed a high-fat diet, resulting in ameliorated glucose tolerance. Blood cholesterol level in the former was lower than that of control mice, although triglyceridemia in the two groups of mice was similar. The adipose-specific LAL-overexpressing mouse phenotype depends on the housing temperature and develops only under mild hypothermic stress (e.g., room temperature) but not at thermoneutrality (30°C), demonstrating the prominent contribution of brown AT (BAT) thermogenesis. LAL overexpression increased levels of BAT free cholesterol, decreased SREBP targets, and induced the expression of genes involved in initial steps of mitochondrial steroidogenesis, suggesting conversion of lysosome-derived cholesterol to pregnenolone. In conclusion, our study demonstrates that adipose LAL drives tissue-cholesterol homeostasis and affects BAT metabolism, suggesting beneficial LAL activation in anti-obesity approaches aimed at reactivating thermogenic energy expenditure.


Asunto(s)
Adipocitos/metabolismo , Autofagia/fisiología , Colesterol/metabolismo , Metabolismo de los Lípidos/fisiología , Obesidad/metabolismo , Esterol Esterasa/metabolismo , Células 3T3 , Adulto , Animales , Retículo Endoplásmico/metabolismo , Humanos , Lipólisis/fisiología , Ratones , Persona de Mediana Edad , Termogénesis/fisiología , Triglicéridos/metabolismo
4.
EBioMedicine ; 58: 102895, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32739864

RESUMEN

BACKGROUND: Bariatric surgery is an effective treatment for type 2 diabetes. Early post-surgical enhancement of insulin secretion is key for diabetes remission. The full complement of mechanisms responsible for improved pancreatic beta cell functionality after bariatric surgery is still unclear. Our aim was to identify pathways, evident in the islet transcriptome, that characterize the adaptive response to bariatric surgery independently of body weight changes. METHODS: We performed entero-gastro-anastomosis (EGA) with pyloric ligature in leptin-deficient ob/ob mice as a surrogate of Roux-en-Y gastric bypass (RYGB) in humans. Multiple approaches such as determination of glucose tolerance, GLP-1 and insulin secretion, whole body insulin sensitivity, ex vivo glucose-stimulated insulin secretion (GSIS) and functional multicellular Ca2+-imaging, profiling of mRNA and of miRNA expression were utilized to identify significant biological processes involved in pancreatic islet recovery. FINDINGS: EGA resolved diabetes, increased pancreatic insulin content and GSIS despite a persistent increase in fat mass, systemic and intra-islet inflammation, and lipotoxicity. Surgery differentially regulated 193 genes in the islet, most of which were involved in the regulation of glucose metabolism, insulin secretion, calcium signaling or beta cell viability, and these were normalized alongside changes in glucose metabolism, intracellular Ca2+ dynamics and the threshold for GSIS. Furthermore, 27 islet miRNAs were differentially regulated, four of them hubs in a miRNA-gene interaction network and four others part of a blood signature of diabetes resolution in ob/ob mice and in humans. INTERPRETATION: Taken together, our data highlight novel miRNA-gene interactions in the pancreatic islet during the resolution of diabetes after bariatric surgery that form part of a blood signature of diabetes reversal. FUNDING: European Union's Horizon 2020 research and innovation programme via the Innovative Medicines Initiative 2 Joint Undertaking (RHAPSODY), INSERM, Société Francophone du Diabète, Institut Benjamin Delessert, Wellcome Trust Investigator Award (212625/Z/18/Z), MRC Programme grants (MR/R022259/1, MR/J0003042/1, MR/L020149/1), Diabetes UK (BDA/11/0004210, BDA/15/0005275, BDA 16/0005485) project grants, National Science Foundation (310030-188447), Fondation de l'Avenir.


Asunto(s)
Diabetes Mellitus Tipo 2/cirugía , Redes Reguladoras de Genes , Células Secretoras de Insulina/química , MicroARNs/genética , Obesidad/cirugía , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Derivación Gástrica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Obesos , Obesidad/genética , Obesidad/metabolismo
5.
Autophagy ; 16(12): 2156-2166, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-31992125

RESUMEN

Adipose tissue (AT) fibrosis in obesity compromises adipocyte functions and responses to intervention-induced weight loss. It is driven by AT progenitors with dual fibro/adipogenic potential, but pro-fibrogenic pathways activated in obesity remain to be deciphered. To investigate the role of macroautophagy/autophagy in AT fibrogenesis, we used Pdgfra-CreErt2 transgenic mice to create conditional deletion of Atg7 alleles in AT progenitor cells (atg7 cKO) and examined sex-dependent, depot-specific AT remodeling in high-fat diet (HFD)-fed mice. Mice with atg7 cKO had markedly decreased extracellular matrix (ECM) gene expression in visceral, subcutaneous, and epicardial adipose depots compared to Atg7lox/lox littermates. ECM gene program regulation by autophagy inhibition occurred independently of changes in the mass of fat tissues or adipocyte numbers of specific depots, and cultured preadipocytes treated with pharmacological or siRNA-mediated autophagy disruptors could mimic these effects. We found that autophagy inhibition promotes global cell-autonomous remodeling of the paracrine TGF-BMP family landscape, whereas ECM gene modulation was independent of the autophagic regulation of GTF2IRD1. The progenitor-specific mouse model of ATG7 inhibition confirms the requirement of autophagy for white/beige adipocyte turnover, and combined to in vitro experiments, reveal progenitor autophagy dependence for AT fibrogenic response to HFD, through the paracrine remodeling of TGF-BMP factors balance. Abbreviations: CQ: chloroquine; ECM: extracellular matrix; EpiAT: epididymal adipose tissue; GTF2IRD1: general transcription factor II I repeat domain-containing 1; HFD: high-fat diet; KO: knockout; OvAT: ovarian adipose tissue; PDGFR: platelet derived growth factor receptor; ScAT: subcutaneous adipose tissue; TGF-BMP: transforming growth factor-bone morphogenic protein.


Asunto(s)
Tejido Adiposo/patología , Autofagia , Dieta Alta en Grasa , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Autofagia/genética , Proteína 7 Relacionada con la Autofagia/deficiencia , Proteína 7 Relacionada con la Autofagia/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Femenino , Fibrosis , Atrios Cardíacos/metabolismo , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Musculares/metabolismo , Regiones Promotoras Genéticas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Caracteres Sexuales , Transducción de Señal , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
6.
Diabetologia ; 60(7): 1314-1324, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28456865

RESUMEN

AIMS/HYPOTHESIS: Regulation of energy balance involves the participation of many factors, including nutrients, among which are circulating lipids, acting as peripheral signals informing the central nervous system of the energy status of the organism. It has been shown that neuronal lipoprotein lipase (LPL) participates in the control of energy balance by hydrolysing lipid particles enriched in triacylglycerols. Here, we tested the hypothesis that LPL in the mediobasal hypothalamus (MBH), a well-known nucleus implicated in the regulation of metabolic homeostasis, could also contribute to the regulation of body weight and glucose homeostasis. METHODS: We injected an adeno-associated virus (AAV) expressing Cre-green fluorescent protein into the MBH of Lpl-floxed mice (and wild-type mice) to specifically decrease LPL activity in the MBH. In parallel, we injected an AAV overexpressing Lpl into the MBH of wild-type mice. We then studied energy homeostasis and hypothalamic ceramide content. RESULTS: The partial deletion of Lpl in the MBH in mice led to an increase in body weight compared with controls (37.72 ± 0.7 g vs 28.46 ± 0.12, p < 0.001) associated with a decrease in locomotor activity. These mice developed hyperinsulinaemia and glucose intolerance. This phenotype also displayed reduced expression of Cers1 in the hypothalamus as well as decreased concentration of several C18 species of ceramides and a 3-fold decrease in total ceramide intensity. Conversely, overexpression of Lpl specifically in the MBH induced a decrease in body weight. CONCLUSIONS/INTERPRETATION: Our study shows that LPL in the MBH is an important regulator of body weight and glucose homeostasis.


Asunto(s)
Glucosa/metabolismo , Hipotálamo/metabolismo , Lipoproteína Lipasa/metabolismo , Aumento de Peso , Animales , Composición Corporal , Peso Corporal , Calorimetría , Ceramidas/metabolismo , Dependovirus , Eliminación de Gen , Prueba de Tolerancia a la Glucosa , Proteínas Fluorescentes Verdes/metabolismo , Homeostasis , Hidrólisis , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Factores de Tiempo , Triglicéridos/sangre
8.
Cell Metab ; 22(4): 646-57, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26278050

RESUMEN

Feeding behavior is exquisitely regulated by homeostatic and hedonic neural substrates that integrate energy demand as well as the reinforcing and rewarding aspects of food. Understanding the net contribution of homeostatic and reward-driven feeding has become critical because of the ubiquitous source of energy-dense foods and the consequent obesity epidemic. Hypothalamic agouti-related peptide-secreting neurons (AgRP neurons) provide the primary orexigenic drive of homeostatic feeding. Using models of neuronal inhibition or ablation, we demonstrate that the feeding response to a fast ghrelin or serotonin receptor agonist relies on AgRP neurons. However, when palatable food is provided, AgRP neurons are dispensable for an appropriate feeding response. In addition, AgRP-ablated mice present exacerbated stress-induced anorexia and palatable food intake--a hallmark of comfort feeding. These results suggest that, when AgRP neuron activity is impaired, neural circuits sensitive to emotion and stress are engaged and modulated by food palatability and dopamine signaling.


Asunto(s)
Proteína Relacionada con Agouti/genética , Neuronas/metabolismo , Proteína Relacionada con Agouti/deficiencia , Animales , Dopamina/metabolismo , Ingestión de Alimentos , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Transducción de Señal
9.
Nat Med ; 21(6): 610-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25939064

RESUMEN

Accumulation of visceral adipose tissue correlates with elevated inflammation and increased risk of metabolic diseases. However, little is known about the molecular mechanisms that control its pathological expansion. Transcription factor interferon regulatory factor 5 (IRF5) has been implicated in polarizing macrophages towards an inflammatory phenotype. Here we demonstrate that mice lacking Irf5, when placed on a high-fat diet, show no difference in the growth of their epididymal white adipose tissue (epiWAT) but they show expansion of their subcutaneous white adipose tissue, as compared to wild-type (WT) mice on the same diet. EpiWAT from Irf5-deficient mice is marked by accumulation of alternatively activated macrophages, higher collagen deposition that restricts adipocyte size, and enhanced insulin sensitivity compared to epiWAT from WT mice. In obese individuals, IRF5 expression is negatively associated with insulin sensitivity and collagen deposition in visceral adipose tissue. Genome-wide analysis of gene expression in adipose tissue macrophages highlights the transforming growth factor ß1 (TGFB1) gene itself as a direct target of IRF5-mediated inhibition. This study uncovers a new function for IRF5 in controlling the relative mass of different adipose tissue depots and thus insulin sensitivity in obesity, and it suggests that inhibition of IRF5 may promote a healthy metabolic state during this condition.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Inflamación/genética , Factores Reguladores del Interferón/genética , Obesidad/genética , Animales , Dieta Alta en Grasa , Regulación de la Expresión Génica , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Resistencia a la Insulina/genética , Macrófagos , Ratones , Obesidad/tratamiento farmacológico , Obesidad/patología , Factor de Crecimiento Transformador beta1/biosíntesis
10.
Cell Metab ; 17(4): 607-17, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23562080

RESUMEN

The delivery of blood-borne molecules conveying metabolic information to neural networks that regulate energy homeostasis is restricted by brain barriers. The fenestrated endothelium of median eminence microvessels and tight junctions between tanycytes together compose one of these. Here, we show that the decrease in blood glucose levels during fasting alters the structural organization of this blood-hypothalamus barrier, resulting in the improved access of metabolic substrates to the arcuate nucleus. These changes are mimicked by 2-deoxyglucose-induced glucoprivation and reversed by raising blood glucose levels after fasting. Furthermore, we show that VEGF-A expression in tanycytes modulates these barrier properties. The neutralization of VEGF signaling blocks fasting-induced barrier remodeling and significantly impairs the physiological response to refeeding. These results implicate glucose in the control of blood-hypothalamus exchanges through a VEGF-dependent mechanism and demonstrate a hitherto unappreciated role for tanycytes and the permeable microvessels associated with them in the adaptive metabolic response to fasting.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Barrera Hematoencefálica/metabolismo , Epéndimo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Desoxiglucosa/farmacología , Epéndimo/citología , Ayuno , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Uniones Estrechas/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
11.
EMBO J ; 31(22): 4276-88, 2012 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-22990237

RESUMEN

Obesity-related diseases such as diabetes and dyslipidemia result from metabolic alterations including the defective conversion, storage and utilization of nutrients, but the central mechanisms that regulate this process of nutrient partitioning remain elusive. As positive regulators of feeding behaviour, agouti-related protein (AgRP) producing neurons are indispensible for the hypothalamic integration of energy balance. Here, we demonstrate a role for AgRP-neurons in the control of nutrient partitioning. We report that ablation of AgRP-neurons leads to a change in autonomic output onto liver, muscle and pancreas affecting the relative balance between lipids and carbohydrates metabolism. As a consequence, mice lacking AgRP-neurons become obese and hyperinsulinemic on regular chow but display reduced body weight gain and paradoxical improvement in glucose tolerance on high-fat diet. These results provide a direct demonstration of a role for AgRP-neurons in the coordination of efferent organ activity and nutrient partitioning, providing a mechanistic link between obesity and obesity-related disorders.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Hipotálamo/metabolismo , Animales , Metabolismo de los Hidratos de Carbono/fisiología , Ingestión de Alimentos/fisiología , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Masculino , Ratones , Músculo Esquelético/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Páncreas/metabolismo , Aumento de Peso/fisiología
12.
Diabetes ; 60(10): 2598-607, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21852673

RESUMEN

OBJECTIVE: In healthy rodents, intestinal sugar absorption in response to sugar-rich meals and insulin is regulated by GLUT2 in enterocyte plasma membranes. Loss of insulin action maintains apical GLUT2 location. In human enterocytes, apical GLUT2 location has not been reported but may be revealed under conditions of insulin resistance. RESEARCH DESIGN AND METHODS: Subcellular location of GLUT2 in jejunal enterocytes was analyzed by confocal and electron microscopy imaging and Western blot in 62 well-phenotyped morbidly obese subjects and 7 lean human subjects. GLUT2 locations were assayed in ob/ob and ob/+ mice receiving oral metformin or in high-fat low-carbohydrate diet-fed C57Bl/6 mice. Glucose absorption and secretion were respectively estimated by oral glucose tolerance test and secretion of [U-(14)C]-3-O-methyl glucose into lumen. RESULTS: In human enterocytes, GLUT2 was consistently located in basolateral membranes. Apical GLUT2 location was absent in lean subjects but was observed in 76% of obese subjects and correlated with insulin resistance and glycemia. In addition, intracellular accumulation of GLUT2 with early endosome antigen 1 (EEA1) was associated with reduced MGAT4a activity (glycosylation) in 39% of obese subjects on a low-carbohydrate/high-fat diet. Mice on a low-carbohydrate/high-fat diet for 12 months also exhibited endosomal GLUT2 accumulation and reduced glucose absorption. In ob/ob mice, metformin promoted apical GLUT2 and improved glucose homeostasis. Apical GLUT2 in fasting hyperglycemic ob/ob mice tripled glucose release into intestinal lumen. CONCLUSIONS: In morbidly obese insulin-resistant subjects, GLUT2 was accumulated in apical and/or endosomal membranes of enterocytes. Functionally, apical GLUT2 favored and endosomal GLUT2 reduced glucose transepithelial exchanges. Thus, altered GLUT2 locations in enterocytes are a sign of intestinal adaptations to human metabolic pathology.


Asunto(s)
Membrana Celular/metabolismo , Grasas de la Dieta/administración & dosificación , Enterocitos/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Obesidad Mórbida/metabolismo , Adulto , Animales , Diabetes Mellitus Tipo 2/metabolismo , Carbohidratos de la Dieta/administración & dosificación , Carbohidratos de la Dieta/farmacología , Transportador de Glucosa de Tipo 2/genética , Humanos , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Persona de Mediana Edad , Adulto Joven
13.
PLoS One ; 6(6): e21184, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21698161

RESUMEN

BACKGROUND: Daily variations in lipid concentrations in both gut lumen and blood are detected by specific sensors located in the gastrointestinal tract and in specialized central areas. Deregulation of the lipid sensors could be partly involved in the dysfunction of glucose homeostasis. The study aimed at comparing the effect of Medialipid (ML) overload on insulin secretion and sensitivity when administered either through the intestine or the carotid artery in mice. METHODOLOGY/PRINCIPAL FINDINGS: An indwelling intragastric or intracarotid catheter was installed in mice and ML or an isocaloric solution was infused over 24 hours. Glucose and insulin tolerance and vagus nerve activity were assessed. Some mice were treated daily for one week with the anti-lipid peroxidation agent aminoguanidine prior to the infusions and tests. The intestinal but not the intracarotid infusion of ML led to glucose and insulin intolerance when compared with controls. The intestinal ML overload induced lipid accumulation and increased lipid peroxidation as assessed by increased malondialdehyde production within both jejunum and duodenum. These effects were associated with the concomitant deregulation of vagus nerve. Administration of aminoguanidine protected against the effects of lipid overload and normalized glucose homeostasis and vagus nerve activity. CONCLUSIONS/SIGNIFICANCE: Lipid overload within the intestine led to deregulation of gastrointestinal lipid sensing that in turn impaired glucose homeostasis through changes in autonomic nervous system activity.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Intestinos/efectos de los fármacos , Peroxidación de Lípido , Animales , Guanidinas/farmacología , Resistencia a la Insulina , Mucosa Intestinal/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Ratones , Estrés Oxidativo
14.
PLoS One ; 4(8): e6649, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19680547

RESUMEN

BACKGROUND: Deregulation of hypothalamic fatty acid sensing lead to hepatic insulin-resistance which may partly contribute to further impairment of glucose homeostasis. METHODOLOGY: We investigated here whether hypothalamic nitric oxide (NO) could mediate deleterious peripheral effect of central lipid overload. Thus we infused rats for 24 hours into carotid artery towards brain, either with heparinized triglyceride emulsion (Intralipid, IL) or heparinized saline (control rats). PRINCIPAL FINDINGS: Lipids infusion led to hepatic insulin-resistance partly related to a decreased parasympathetic activity in the liver assessed by an increased acetylcholinesterase activity. Hypothalamic nitric oxide synthases (NOS) activities were significantly increased in IL rats, as the catalytically active neuronal NOS (nNOS) dimers compared to controls. This was related to a decrease in expression of protein inhibitor of nNOS (PIN). Effect of IL infusion on deregulated hepatic insulin-sensitivity was reversed by carotid injection of non selective NOS inhibitor NG-monomethyl-L-arginine (L-NMMA) and also by a selective inhibitor of the nNOS isoform, 7-Nitro-Indazole (7-Ni). In addition, NO donor injection (L-arginine and SNP) within carotid in control rats mimicked lipid effects onto impaired hepatic insulin sensitivity. In parallel we showed that cultured VMH neurons produce NO in response to fatty acid (oleic acid). CONCLUSIONS/SIGNIFICANCE: We conclude that cerebral fatty acid overload induces an enhancement of nNOS activity within hypothalamus which is, at least in part, responsible fatty acid increased hepatic glucose production.


Asunto(s)
Resistencia a la Insulina , Hígado/fisiología , Óxido Nítrico/fisiología , Ácido Oléico/administración & dosificación , Animales , Inhibidores Enzimáticos/farmacología , Conducta Alimentaria , Glucosa/metabolismo , Hipotálamo/enzimología , Óxido Nítrico/biosíntesis , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Ratas
15.
Am J Physiol Regul Integr Comp Physiol ; 296(4): R929-35, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19211723

RESUMEN

The increase in adiponectin levels in obese patients with untreated dyslipidemia and its mRNA expression in adipose tissue of obese animals are one of the most interesting consequences of rimonabant treatment. Thus, part of rimonabant's metabolic effects could be related to an enhancement of adiponectin secretion and its consequence on the modulation of insulin action, as well as energy homeostasis. The present study investigated the effects of rimonabant in adiponectin knockout mice (Ad(-/-)) exposed to diet-induced obesity conditions. Six-week-old Ad(-/-) male mice and their wild-type littermate controls (Ad(+/+)) were fed a high-fat diet for 7 mo. During the last month, animals were administered daily either with vehicle or rimonabant by mouth (10 mg/kg). High-fat feeding induced weight gain by about 130% in both wild-type and Ad(-/-) mice. Obesity was associated with hyperinsulinemia and insulin resistance. Treatment with rimonabant led to a significant and similar decrease in body weight in both Ad(+/+) and Ad(-/-) mice compared with vehicle-treated animals. In addition, rimonabant significantly improved insulin sensitivity in Ad(+/+) mice compared with Ad(+/+) vehicle-treated mice by decreasing hepatic glucose production and increasing glucose utilization index in both visceral and subcutaneous adipose tissue. In contrast, rimonabant failed to improve insulin sensitivity in Ad(-/-) mice, despite the loss in body weight. Rimonabant's effect on body weight appeared independent of the adiponectin pathway, whereas adiponectin seems required to mediate rimonabant-induced improvement of insulin sensitivity in rodents.


Asunto(s)
Fármacos Antiobesidad/farmacología , Hiperinsulinismo/prevención & control , Resistencia a la Insulina , Obesidad/tratamiento farmacológico , Piperidinas/farmacología , Pirazoles/farmacología , Pérdida de Peso/efectos de los fármacos , Adiponectina/deficiencia , Adiponectina/genética , Adiponectina/metabolismo , Animales , Grasas de la Dieta , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Hiperinsulinismo/etiología , Hiperinsulinismo/metabolismo , Hiperinsulinismo/fisiopatología , Insulina/sangre , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/metabolismo , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Obesidad/fisiopatología , Rimonabant , Grasa Subcutánea/efectos de los fármacos , Grasa Subcutánea/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...