Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 29(9): 934-47, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25934505

RESUMEN

MAF1 is a global repressor of RNA polymerase III transcription that regulates the expression of highly abundant noncoding RNAs in response to nutrient availability and cellular stress. Thus, MAF1 function is thought to be important for metabolic economy. Here we show that a whole-body knockout of Maf1 in mice confers resistance to diet-induced obesity and nonalcoholic fatty liver disease by reducing food intake and increasing metabolic inefficiency. Energy expenditure in Maf1(-/-) mice is increased by several mechanisms. Precursor tRNA synthesis was increased in multiple tissues without significant effects on mature tRNA levels, implying increased turnover in a futile tRNA cycle. Elevated futile cycling of hepatic lipids was also observed. Metabolite profiling of the liver and skeletal muscle revealed elevated levels of many amino acids and spermidine, which links the induction of autophagy in Maf1(-/-) mice with their extended life span. The increase in spermidine was accompanied by reduced levels of nicotinamide N-methyltransferase, which promotes polyamine synthesis, enables nicotinamide salvage to regenerate NAD(+), and is associated with obesity resistance. Consistent with this, NAD(+) levels were increased in muscle. The importance of MAF1 for metabolic economy reveals the potential for MAF1 modulators to protect against obesity and its harmful consequences.


Asunto(s)
Proteínas Represoras/genética , Animales , Autofagia/genética , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Metabolismo de los Lípidos/genética , Longevidad/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/genética , ARN de Transferencia/metabolismo , Espermidina/metabolismo
2.
Eur J Nutr ; 58(5): 1829-1845, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29845385

RESUMEN

PURPOSE: High-fructose consumption and chronic stress are both associated with metabolic inflammation and insulin resistance. Recently, disturbed activity of energy sensor AMP-activated protein kinase (AMPK) was recognized as mediator between nutrient-induced stress and inflammation. Thus, we analyzed the effects of high-fructose diet, alone or in combination with chronic stress, on glucose homeostasis, inflammation and expression of energy sensing proteins in the rat liver. METHODS: In male Wistar rats exposed to 9-week 20% fructose diet and/or 4-week chronic unpredictable stress we measured plasma and hepatic corticosterone level, indicators of glucose homeostasis and lipid metabolism, hepatic inflammation (pro- and anti-inflammatory cytokine levels, Toll-like receptor 4, NLRP3, activation of NFκB, JNK and ERK pathways) and levels of energy-sensing proteins AMPK, SIRT1 and peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α). RESULTS: High-fructose diet led to glucose intolerance, activation of NFκB and JNK pathways and increased intrahepatic IL-1ß, TNFα and inhibitory phosphorylation of insulin receptor substrate 1 on Ser307. It also decreased phospho-AMPK/AMPK ratio and increased SIRT1 expression. Stress alone increased plasma and hepatic corticosterone but did not influence glucose tolerance, nor hepatic inflammatory or energy-sensing proteins. After the combined treatment, hepatic corticosterone was increased, glucose tolerance remained preserved, while hepatic inflammation was partially prevented despite decreased AMPK activity. CONCLUSION: High-fructose diet resulted in glucose intolerance, hepatic inflammation, decreased AMPK activity and reduced insulin sensitivity. Chronic stress alone did not exert such effects, but when applied together with high-fructose diet it could partially prevent fructose-induced inflammation, presumably due to increased hepatic glucocorticoids.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Dieta/métodos , Fructosa/administración & dosificación , Inflamación/fisiopatología , Estrés Fisiológico/fisiología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Hígado , Masculino , Ratas , Ratas Wistar
3.
Kidney Int ; 87(5): 940-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25565311

RESUMEN

Urate is the metabolic end point of purines in humans. Although supra-physiological plasma urate levels are associated with obesity, insulin resistance, dyslipidemia, and hypertension, a causative role is debated. We previously established a mouse model of hyperuricemia by liver-specific deletion of Glut9, a urate transporter that provides urate to the hepatocyte enzyme uricase. These LG9 knockout mice show mild hyperuricemia (120 µmol/l), which can be further increased by the urate precursor inosine. Here, we explored the role of progressive hyperuricemia on the cardiovascular function. Arterial blood pressure and heart rate were periodically measured by telemetry over 6 months in LG9 knockout mice supplemented with incremental amounts of inosine in a normal chow diet. This long-term inosine treatment elicited a progressive increase in uricemia up to 300 µmol/l; however, it did not modify heart rate or mean arterial blood pressure in LG9 knockout compared with control mice. Inosine treatment did not alter cardiac morphology or function measured by ultrasound echocardiography. However, it did induce mild renal dysfunction as revealed by higher plasma creatinine levels, lower glomerular filtration rate, and histological signs of chronic inflammation and fibrosis. Thus, in LG9 knockout mice, inosine-induced hyperuricemia was not associated with hypertension despite partial renal deficiency. This does not support a direct role of urate in the control of blood pressure.


Asunto(s)
Presión Sanguínea , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Frecuencia Cardíaca , Hiperuricemia/fisiopatología , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Hiperuricemia/diagnóstico por imagen , Hiperuricemia/etiología , Inosina , Riñón/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Basic Res Cardiol ; 109(1): 399, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24370889

RESUMEN

Sirt3 is a mitochondrial NAD(+)-dependent deacetylase that governs mitochondrial metabolism and reactive oxygen species homeostasis. Sirt3 deficiency has been reported to accelerate the development of the metabolic syndrome. However, the role of Sirt3 in atherosclerosis remains enigmatic. We aimed to investigate whether Sirt3 deficiency affects atherosclerosis, plaque vulnerability, and metabolic homeostasis. Low-density lipoprotein receptor knockout (LDLR(-/-)) and LDLR/Sirt3 double-knockout (Sirt3(-/-)LDLR(-/-)) mice were fed a high-cholesterol diet (1.25 % w/w) for 12 weeks. Atherosclerosis was assessed en face in thoraco-abdominal aortae and in cross sections of aortic roots. Sirt3 deletion led to hepatic mitochondrial protein hyperacetylation. Unexpectedly, though plasma malondialdehyde levels were elevated in Sirt3-deficient mice, Sirt3 deletion affected neither plaque burden nor features of plaque vulnerability (i.e., fibrous cap thickness and necrotic core diameter). Likewise, plaque macrophage and T cell infiltration as well as endothelial activation remained unaltered. Electron microscopy of aortic walls revealed no difference in mitochondrial microarchitecture between both groups. Interestingly, loss of Sirt3 was associated with accelerated weight gain and an impaired capacity to cope with rapid changes in nutrient supply as assessed by indirect calorimetry. Serum lipid levels and glucose tolerance were unaffected by Sirt3 deletion in LDLR(-/-) mice. Sirt3 deficiency does not affect atherosclerosis in LDLR(-/-) mice. However, Sirt3 controls systemic levels of oxidative stress, limits expedited weight gain, and allows rapid metabolic adaptation. Thus, Sirt3 may contribute to postponing cardiovascular risk factor development.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/fisiopatología , Sirtuina 3/deficiencia , Animales , Western Blotting , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Homeostasis , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Receptores de LDL/deficiencia , Receptores de LDL/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Riesgo
5.
Proc Natl Acad Sci U S A ; 108(42): E854-63, 2011 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-21949398

RESUMEN

Obesity is associated with a chronic low-grade inflammation, and specific antiinflammatory interventions may be beneficial for the treatment of type 2 diabetes and other obesity-related diseases. The lipid kinase PI3Kγ is a central proinflammatory signal transducer that plays a major role in leukocyte chemotaxis, mast cell degranulation, and endothelial cell activation. It was also reported that PI3Kγ activity within hematopoietic cells plays an important role in obesity-induced inflammation and insulin resistance. Here, we show that protection from insulin resistance, metabolic inflammation, and fatty liver in mice lacking functional PI3Kγ is largely consequent to their leaner phenotype. We also show that this phenotype is largely based on decreased fat gain, despite normal caloric intake, consequent to increased energy expenditure. Furthermore, our data show that PI3Kγ action on diet-induced obesity depends on PI3Kγ activity within a nonhematopoietic compartment, where it promotes energetic efficiency for fat mass gain. We also show that metabolic modulation by PI3Kγ depends on its lipid kinase activity and might involve kinase-independent signaling. Thus, PI3Kγ is an unexpected but promising drug target for the treatment of obesity and its complications.


Asunto(s)
Tejido Adiposo Blanco/enzimología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Resistencia a la Insulina/fisiología , Obesidad/enzimología , Termogénesis/fisiología , Animales , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Dieta Alta en Grasa/efectos adversos , Hígado Graso/enzimología , Hígado Graso/etiología , Hígado Graso/prevención & control , Inflamación/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Obesidad/etiología , Obesidad/prevención & control , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Esterol Esterasa/metabolismo , Delgadez/enzimología
6.
Am J Physiol Renal Physiol ; 305(5): F786-95, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23804456

RESUMEN

Plasma urate levels are higher in humans than rodents (240-360 vs. ∼30 µM) because humans lack the liver enzyme uricase. High uricemia in humans may protect against oxidative stress, but hyperuricemia also associates with the metabolic syndrome, and urate and uric acid can crystallize to cause gout and renal dysfunctions. Thus, hyperuricemic animal models to study urate-induced pathologies are needed. We recently generated mice with liver-specific ablation of Glut9, a urate transporter providing access of urate to uricase (LG9KO mice). LG9KO mice had moderately high uricemia (∼120 µM). To further increase their uricemia, here we gavaged LG9KO mice for 3 days with inosine, a urate precursor; this treatment was applied in both chow- and high-fat-fed mice. In chow-fed LG9KO mice, uricemia peaked at 300 µM 2 h after the first gavage and normalized 24 h after the last gavage. In contrast, in high-fat-fed LG9KO mice, uricemia further rose to 500 µM. Plasma creatinine strongly increased, indicating acute renal failure. Kidneys showed tubule dilation, macrophage infiltration, and urate and uric acid crystals, associated with a more acidic urine. Six weeks after inosine gavage, plasma urate and creatinine had normalized. However, renal inflammation, fibrosis, and organ remodeling had developed despite the disappearance of urate and uric acid crystals. Thus, hyperuricemia and high-fat diet feeding combined to induce acute renal failure. Furthermore, a sterile inflammation caused by the initial crystal-induced lesions developed despite the disappearance of urate and uric acid crystals.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Proteínas Facilitadoras del Transporte de la Glucosa/deficiencia , Hiperuricemia/sangre , Animales , Cristalización , Dieta Alta en Grasa , Concentración de Iones de Hidrógeno , Hiperuricemia/etiología , Inflamación/inducido químicamente , Inosina/metabolismo , Masculino , Ratones , Ratones Noqueados , Transportadores de Anión Orgánico/deficiencia , Ácido Úrico/sangre , Orina/fisiología
7.
Proc Natl Acad Sci U S A ; 106(36): 15501-6, 2009 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-19706426

RESUMEN

Elevated plasma urate levels are associated with metabolic, cardiovascular, and renal diseases. Urate may also form crystals, which can be deposited in joints causing gout and in kidney tubules inducing nephrolithiasis. In mice, plasma urate levels are controlled by hepatic breakdown, as well as, by incompletely understood renal processes of reabsorption and secretion. Here, we investigated the role of the recently identified urate transporter, Glut9, in the physiological control of urate homeostasis using mice with systemic or liver-specific inactivation of the Glut9 gene. We show that Glut9 is expressed in the basolateral membrane of hepatocytes and in both apical and basolateral membranes of the distal nephron. Mice with systemic knockout of Glut9 display moderate hyperuricemia, massive hyperuricosuria, and an early-onset nephropathy, characterized by obstructive lithiasis, tubulointerstitial inflammation, and progressive inflammatory fibrosis of the cortex, as well as, mild renal insufficiency. In contrast, liver-specific inactivation of the Glut9 gene in adult mice leads to severe hyperuricemia and hyperuricosuria, in the absence of urate nephropathy or any structural abnormality of the kidney. Together, our data show that Glut9 plays a major role in urate homeostasis by its dual role in urate handling in the kidney and uptake in the liver.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Homeostasis/fisiología , Nefritis Intersticial/genética , Ácido Úrico/metabolismo , Ácido Úrico/orina , Análisis de Varianza , Animales , Western Blotting , Hepatocitos/metabolismo , Ratones , Ratones Noqueados , Nefronas/metabolismo
8.
Clocks Sleep ; 4(1): 37-51, 2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35225952

RESUMEN

Both sleep-wake behavior and circadian rhythms are tightly coupled to energy metabolism and food intake. Altered feeding times in mice are known to entrain clock gene rhythms in the brain and liver, and sleep-deprived humans tend to eat more and gain weight. Previous observations in mice showing that sleep deprivation (SD) changes clock gene expression might thus relate to altered food intake, and not to the loss of sleep per se. Whether SD affects food intake in the mouse and how this might affect clock gene expression is, however, unknown. We therefore quantified (i) the cortical expression of the clock genes Per1, Per2, Dbp, and Cry1 in mice that had access to food or not during a 6 h SD, and (ii) food intake during baseline, SD, and recovery sleep. We found that food deprivation did not modify the SD-incurred clock gene changes in the cortex. Moreover, we discovered that although food intake during SD did not differ from the baseline, mice lost weight and increased food intake during subsequent recovery. We conclude that SD is associated with food deprivation and that the resulting energy deficit might contribute to the effects of SD that are commonly interpreted as a response to sleep loss.

9.
FASEB J ; 24(6): 1747-58, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20097878

RESUMEN

The physiological contribution of glucose in thermoregulation is not completely established nor whether this control may involve a regulation of the melanocortin pathway. Here, we assessed thermoregulation and leptin sensitivity of hypothalamic arcuate neurons in mice with inactivation of glucose transporter type 2 (Glut2)-dependent glucose sensing. Mice with inactivation of Glut2-dependent glucose sensors are cold intolerant and show increased susceptibility to food deprivation-induced torpor and abnormal hypothermic response to intracerebroventricular administration of 2-deoxy-d-glucose compared to control mice. This is associated with a defect in regulated expression of brown adipose tissue uncoupling protein I and iodothyronine deiodinase II and with a decreased leptin sensitivity of neuropeptide Y (NPY) and proopiomelanocortin (POMC) neurons, as observed during the unfed-to-refed transition or following i.p. leptin injection. Sites of central Glut-2 expression were identified by a genetic tagging approach and revealed that glucose-sensitive neurons were present in the lateral hypothalamus, the dorsal vagal complex, and the basal medulla but not in the arcuate nucleus. NPY and POMC neurons were, however, connected to nerve terminals from Glut2-expressing neurons. Thus, our data suggest that glucose controls thermoregulation and the leptin sensitivity of NPY and POMC neurons through activation of Glut2-dependent glucose-sensing neurons located outside of the arcuate nucleus.


Asunto(s)
Regulación de la Temperatura Corporal , Transportador de Glucosa de Tipo 2/fisiología , Glucosa/metabolismo , Leptina/farmacología , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Western Blotting , Femenino , Glucosa/análisis , Humanos , Técnicas para Inmunoenzimas , Integrasas , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Neuropéptido Y/genética , Proopiomelanocortina/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Desacopladora 1 , Yodotironina Deyodinasa Tipo II
10.
Nature ; 436(7049): 356-62, 2005 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-16034410

RESUMEN

In obesity and type 2 diabetes, expression of the GLUT4 glucose transporter is decreased selectively in adipocytes. Adipose-specific Glut4 (also known as Slc2a4) knockout (adipose-Glut4(-/-)) mice show insulin resistance secondarily in muscle and liver. Here we show, using DNA arrays, that expression of retinol binding protein-4 (RBP4) is elevated in adipose tissue of adipose-Glut4(-/-) mice. We show that serum RBP4 levels are elevated in insulin-resistant mice and humans with obesity and type 2 diabetes. RBP4 levels are normalized by rosiglitazone, an insulin-sensitizing drug. Transgenic overexpression of human RBP4 or injection of recombinant RBP4 in normal mice causes insulin resistance. Conversely, genetic deletion of Rbp4 enhances insulin sensitivity. Fenretinide, a synthetic retinoid that increases urinary excretion of RBP4, normalizes serum RBP4 levels and improves insulin resistance and glucose intolerance in mice with obesity induced by a high-fat diet. Increasing serum RBP4 induces hepatic expression of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK) and impairs insulin signalling in muscle. Thus, RBP4 is an adipocyte-derived 'signal' that may contribute to the pathogenesis of type 2 diabetes. Lowering RBP4 could be a new strategy for treating type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Resistencia a la Insulina/fisiología , Obesidad/sangre , Proteínas de Unión al Retinol/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4 , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Hepatocitos/metabolismo , Insulina/farmacología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/enzimología , Ratones , Ratones Noqueados , Proteínas de Transporte de Monosacáridos/deficiencia , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculos/efectos de los fármacos , Músculos/metabolismo , Obesidad/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Proteínas de Unión al Retinol/genética , Proteínas Plasmáticas de Unión al Retinol , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología
11.
NMR Biomed ; 23(6): 578-83, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20235335

RESUMEN

The hypothalamus plays an essential role in the central nervous system of mammals by among others regulating glucose homeostasis, food intake, temperature, and to some extent blood pressure. Assessments of hypothalamic metabolism using, e.g. (1)H MRS in mouse models can provide important insights into its function. To date, direct in vivo (1)H MRS measurements of hypothalamus have not been reported. Here, we report that in vivo single voxel measurements of mouse hypothalamus are feasible using (1)H MRS at 14.1T. Localized (1)H MR spectra from hypothalamus were obtained unilaterally (2-2.2 microL, VOI) and bilaterally (4-4.4 microL) with a quality comparable to that of hippocampus (3-3.5 microL). Using LCModel, a neurochemical profile consisting of 21 metabolites was quantified for both hypothalamus and hippocampus with most of the Cramér-Rao lower bounds within 20%. Relative to the hippocampus, the hypothalamus was characterized by high gamma-aminobutryric acid and myo-inositol, and low taurine concentrations. When studying transgenic mice with no glucose transporter isoform 8 expressed, small metabolic changes were observed, yet glucose homeostasis was well maintained. We conclude that a specific neurochemical profile of mouse hypothalamus can be measured by (1)H MRS which will allow identifying and following metabolic alterations longitudinally in the hypothalamus of genetic modified models.


Asunto(s)
Hipotálamo/química , Espectroscopía de Resonancia Magnética/métodos , Animales , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hipotálamo/anatomía & histología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Mol Nutr Food Res ; 64(13): e1901141, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32379936

RESUMEN

SCOPE: Intake of fructose-sweetened beverages and chronic stress (CS) both increase risk of cardiometabolic diseases. The aim is to investigate whether these factors synergistically perturb lipid metabolism in rat liver and kidney. METHODS AND RESULTS: Fractional de novo lipogenesis (fDNL), intrahepatic- and intrarenal-triglycerides (IHTG and IRTG), de novo palmitate (DNPalm) content, FA composition, VLDL-TGs kinetics, and key metabolic gene expression at the end of the feeding and non-feeding phases in rats exposed to standard chow diet, chow diet + CS, 20% liquid high-fructose supplementation (HFr), or HFr+CS are measured. HFr induces hypertriglyceridemia, up-regulates fructose-metabolism and gluconeogenic enzymes, increases IHTG and DNPalm content in IHTG and IRTG, and augments fDNL at the end of the feeding phase. These changes are diminished after the non-feeding phase. CS does not exert such effects, but when combined with HFr, it reduces IHTG and visceral adiposity, enhances lipogenic gene expression and fDNL, and increases VLDL-DNPalm secretion. CONCLUSION: Liquid high-fructose supplementation increases IHTG and VLDL-TG secretion after the feeding phase, the latter being the result of stimulated hepatic and renal DNL. Chronic stress potentiates the effects of high fructose on fDNL and export of newly synthesized VLDL-TGs, and decreases fructose-induced intrahepatic TG accumulation after the feeding phase.


Asunto(s)
Fructosa/efectos adversos , Riñón/efectos de los fármacos , Lipogénesis , Hígado/metabolismo , Estrés Psicológico/fisiopatología , Animales , Composición Corporal , Ingestión de Alimentos , Ingestión de Energía , Enzimas/genética , Enzimas/metabolismo , Regulación de la Expresión Génica , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/fisiología , Riñón/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/fisiología , Lipogénesis/efectos de los fármacos , Lipoproteínas VLDL/metabolismo , Hígado/efectos de los fármacos , Masculino , Palmitatos/metabolismo , Ratas Wistar , Triglicéridos/metabolismo
13.
Am J Physiol Endocrinol Metab ; 297(6): E1420-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19826103

RESUMEN

The synthetic retinoid Fenretinide (FEN) increases insulin sensitivity in obese rodents and is in early clinical trials for treatment of insulin resistance in obese humans with hepatic steatosis (46). We aimed to determine the physiological mechanisms for the insulin-sensitizing effects of FEN. Wild-type mice were fed a high-fat diet (HFD) with or without FEN from 4-5 wk to 36-37 wk of age (preventive study) or following 22 wk of HF diet-induced obesity (12 wk intervention study). Retinol-binding protein-4 (RBP4) knockout mice were also fed the HFD with or without FEN in a preventive study. FEN had minimal effects on HFD-induced body weight gain but markedly reduced HFD-induced adiposity and hyperleptinemia in both studies. FEN-HFD mice gained epididymal fat but not subcutaneous or visceral fat mass in contrast to HFD mice without FEN. FEN did not have a measurable effect on energy expenditure, food intake, physical activity, or stool lipid content. Glucose infusion rate during hyperinsulinemic-euglycemic clamp was reduced 86% in HFD mice compared with controls and was improved 3.6-fold in FEN-HFD compared with HFD mice. FEN improved insulin action on glucose uptake and glycogen levels in muscle, insulin-stimulated suppression of hepatic glucose production, and suppression of serum FFA levels in HFD mice. Remarkably, FEN also reduced hepatic steatosis. In RBP4 knockout mice, FEN reduced the HFD-induced increase in adiposity and hyperleptinemia. In conclusion, long-term therapy with FEN partially prevents or reverses obesity, insulin resistance, and hepatic steatosis in mice on HFD. The anti-adiposity effects are independent of the RBP4 lowering effect.


Asunto(s)
Hígado Graso/metabolismo , Fenretinida/administración & dosificación , Resistencia a la Insulina/fisiología , Obesidad/prevención & control , Retinoides/administración & dosificación , Animales , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Calorimetría Indirecta , Estudios de Cohortes , Esquema de Medicación , Ingestión de Alimentos/efectos de los fármacos , Técnica de Clampeo de la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Proteínas de Unión al Retinol/metabolismo
14.
J Cereb Blood Flow Metab ; 39(9): 1725-1736, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-29561214

RESUMEN

Glucose transporter 2 (Glut2)-positive cells are sparsely distributed in brain and play an important role in the stimulation of glucagon secretion in response to hypoglycemia. We aimed to determine if Glut2-positive cells can influence another response to hypoglycemia, i.e. increased cerebral blood flow (CBF). CBF of adult male mice devoid of Glut2, either globally (ripglut1:glut2-/-) or in the nervous system only (NG2KO), and their respective controls were studied under basal glycemia and insulin-induced hypoglycemia using quantitative perfusion magnetic resonance imaging at 9.4 T. The effect on CBF of optogenetic activation of hypoglycemia responsive Glut2-positive neurons of the paraventricular thalamic area was measured in mice expressing channelrhodopsin2 under the control of the Glut2 promoter. We found that in both ripglut1:glut2-/- mice and NG2KO mice, CBF in basal conditions was higher than in their respective controls and not further activated by hypoglycemia, as measured in the hippocampus, hypothalamus and whole brain. Conversely, optogenetic activation of Glut2-positive cells in the paraventricular thalamic nucleus induced a local increase in CBF similar to that induced by hypoglycemia. Thus, Glut2 expression in the nervous system is required for the control of CBF in response to changes in blood glucose concentrations.


Asunto(s)
Glucemia/metabolismo , Circulación Cerebrovascular , Transportador de Glucosa de Tipo 2/metabolismo , Hipoglucemia/metabolismo , Animales , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Hipoglucemia/sangre , Masculino , Ratones Endogámicos C57BL
15.
Metabolism ; 95: 8-20, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30878493

RESUMEN

BACKGROUND: The peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-dependent transcription factor involved in many aspects of metabolism, immune response and development. Numerous studies relying on tissue-specific invalidation of the Pparg gene have shown distinct facets of its activity, whereas the effects of its systemic inactivation remain unexplored due to embryonic lethality. By maintaining PPARγ expression in the placenta, we recently generated a mouse model carrying Pparg full body deletion (PpargΔ/Δ), which in contrast to a previously published model is totally deprived of any form of adipose tissue. Herein, we propose an in-depth study of the metabolic alterations observed in this new model. METHODS: Young adult mice, both males and females analyzed separately, were first phenotyped for their gross anatomical alterations. Systemic metabolic parameters were analyzed in the blood, in static and in dynamic conditions. A full exploration of energy metabolism was performed in calorimetric cages as well as in metabolic cages. Our study was completed by expression analyses of a set of specific genes. MAIN FINDINGS: PpargΔ/Δ mice show a striking complete absence of any form of adipose tissue, which triggers a complex metabolic phenotype including increased lean mass with organomegaly, hypermetabolism, urinary energy loss, hyperphagia, and increased amino acid metabolism. PpargΔ/Δ mice develop severe type 2 diabetes, characterized by hyperglycemia, hyperinsulinemia, polyuria and polydispsia. They show a remarkable metabolic inflexibility, as indicated by the inability to shift substrate oxidation between glucose and lipids, in both ad libitum fed state and fed/fasted/refed transitions. Moreover, upon fasting PpargΔ/Δ mice enter a severe hypometabolic state. CONCLUSIONS: Our data comprehensively describe the impact of lipoatrophy on metabolic homeostasis. As such, the presented data on PpargΔ/Δ mice gives new clues on what and how to explore severe lipodystrophy and its subsequent metabolic complications in human.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Trastornos del Metabolismo de los Lípidos/genética , Tamaño de los Órganos/genética , PPAR gamma/genética , Tejido Adiposo/anatomía & histología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/genética , Femenino , Eliminación de Gen , Glucosa/metabolismo , Trastornos del Metabolismo de los Lípidos/metabolismo , Lipodistrofia/genética , Lipodistrofia/metabolismo , Ratones , Embarazo
16.
Mol Cell Endocrinol ; 476: 110-118, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29729371

RESUMEN

Both fructose overconsumption and increased glucocorticoids secondary to chronic stress may contribute to overall dyslipidemia. In this study we specifically assessed the effects and interactions of dietary fructose and chronic stress on lipid metabolism in the visceral adipose tissue (VAT) of male Wistar rats. We analyzed the effects of 9-week 20% high fructose diet and 4-week chronic unpredictable stress, separately and in combination, on VAT histology, glucocorticoid prereceptor metabolism, glucocorticoid receptor subcellular redistribution and expression of major metabolic genes. Blood triglycerides and fatty acid composition were also measured to assess hepatic Δ9 desaturase activity. The results showed that fructose diet increased blood triglycerides and Δ9 desaturase activity. On the other hand, stress led to corticosterone elevation, glucocorticoid receptor activation and decrease in adipocyte size, while phosphoenolpyruvate carboxykinase, adipose tissue triglyceride lipase, FAT/CD36 and sterol regulatory element binding protein-1c (SREBP-1c) were increased, pointing to VAT lipolysis and glyceroneogenesis. The combination of stress and fructose diet was associated with marked stimulation of fatty acid synthase and acetyl-CoA carboxylase mRNA level and with increased 11ß-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase protein levels, suggesting a coordinated increase in hexose monophosphate shunt and de novo lipogenesis. It however did not influence the level of peroxisome proliferator-activated receptor-gamma, SREBP-1c and carbohydrate responsive element-binding protein. In conclusion, our results showed that only combination of dietary fructose and stress increase glucocorticoid prereceptor metabolism and stimulates lipogenic enzyme expression suggesting that interaction between stress and fructose may be instrumental in promoting VAT expansion and dysfunction.


Asunto(s)
Dieta , Grasa Intraabdominal/metabolismo , Metabolismo de los Lípidos , Receptores de Glucocorticoides/metabolismo , Transducción de Señal , Estrés Psicológico/metabolismo , Animales , Corticosterona/sangre , Ácidos Grasos/sangre , Fructosa , Regulación de la Expresión Génica , Insulina/sangre , Metabolismo de los Lípidos/genética , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Estearoil-CoA Desaturasa/metabolismo , Estrés Psicológico/sangre , Transcripción Genética , Triglicéridos/sangre
17.
J Clin Invest ; 113(4): 635-45, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14966573

RESUMEN

The role of the gluco-incretin hormones GIP and GLP-1 in the control of beta cell function was studied by analyzing mice with inactivation of each of these hormone receptor genes, or both. Our results demonstrate that glucose intolerance was additively increased during oral glucose absorption when both receptors were inactivated. After intraperitoneal injections, glucose intolerance was more severe in double- as compared to single-receptor KO mice, and euglycemic clamps revealed normal insulin sensitivity, suggesting a defect in insulin secretion. When assessed in vivo or in perfused pancreas, insulin secretion showed a lack of first phase in Glp-1R(-/-) but not in Gipr(-/-) mice. In perifusion experiments, however, first-phase insulin secretion was present in both types of islets. In double-KO islets, kinetics of insulin secretion was normal, but its amplitude was reduced by about 50% because of a defect distal to plasma membrane depolarization. Thus, gluco-incretin hormones control insulin secretion (a) by an acute insulinotropic effect on beta cells after oral glucose absorption (b) through the regulation, by GLP-1, of in vivo first-phase insulin secretion, probably by an action on extra-islet glucose sensors, and (c) by preserving the function of the secretory pathway, as evidenced by a beta cell autonomous secretion defect when both receptors are inactivated.


Asunto(s)
Polipéptido Inhibidor Gástrico/metabolismo , Glucagón/metabolismo , Insulina/metabolismo , Fragmentos de Péptidos/metabolismo , Precursores de Proteínas/metabolismo , Receptores de la Hormona Gastrointestinal/metabolismo , Receptores de Glucagón/metabolismo , Animales , Glucemia/metabolismo , Carbacol/metabolismo , AMP Cíclico/metabolismo , Femenino , Péptido 1 Similar al Glucagón , Prueba de Tolerancia a la Glucosa , Secreción de Insulina , Masculino , Ratones , Ratones Noqueados , Páncreas/metabolismo , Receptores de la Hormona Gastrointestinal/genética , Receptores de Glucagón/genética
18.
PLoS One ; 11(2): e0150266, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26918849

RESUMEN

Glucose is the most important metabolic substrate of the retina and maintenance of normoglycemia is an essential challenge for diabetic patients. Chronic, exaggerated, glycemic excursions could lead to cardiovascular diseases, nephropathy, neuropathy and retinopathy. We recently showed that hypoglycemia induced retinal cell death in mouse via caspase 3 activation and glutathione (GSH) decrease. Ex vivo experiments in 661W photoreceptor cells confirmed the low-glucose induction of death via superoxide production and activation of caspase 3, which was concomitant with a decrease of GSH content. We evaluate herein retinal gene expression 4 h and 48 h after insulin-induced hypoglycemia. Microarray analysis demonstrated clusters of genes whose expression was modified by hypoglycemia and we discuss the potential implication of those genes in retinal cell death. In addition, we identify by gene set enrichment analysis, three important pathways, including lysosomal function, GSH metabolism and apoptotic pathways. Then we tested the effect of recurrent hypoglycemia (three successive 4h periods of hypoglycemia spaced by 48 h recovery) on retinal cell death. Interestingly, exposure to multiple hypoglycemic events prevented GSH decrease and retinal cell death, or adapted the retina to external stress by restoring GSH level comparable to control situation. We hypothesize that scavenger GSH is a key compound in this apoptotic process, and maintaining "normal" GSH level, as well as a strict glycemic control, represents a therapeutic challenge in order to avoid side effects of diabetes, especially diabetic retinopathy.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hipoglucemia/inducido químicamente , Insulina/toxicidad , Familia de Multigenes/genética , Retina/metabolismo , Enfermedad Aguda , Adaptación Fisiológica , Animales , Apoptosis/genética , Fragmentación del ADN , Retinopatía Diabética/prevención & control , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Femenino , Depuradores de Radicales Libres , Regulación de la Expresión Génica/fisiología , Técnica de Clampeo de la Glucosa , Glutatión/metabolismo , Hipoglucemia/genética , Hipoglucemia/metabolismo , Insulina/farmacología , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Oxidación-Reducción , Distribución Aleatoria , Retina/patología , Factores de Tiempo
19.
Front Physiol ; 7: 142, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27148080

RESUMEN

Glutathione (GSH) deficits have been observed in several mental or degenerative illness, and so has the metabolic syndrome. The impact of a decreased glucose metabolism on the GSH system is well-known, but the effect of decreased GSH levels on the energy metabolism is unclear. The aim of the present study was to investigate the sensitivity to insulin in the mouse knockout (KO) for the modulatory subunit of the glutamate cysteine ligase (GCLM), the rate-limiting enzyme of GSH synthesis. Compared to wildtype (WT) mice, GCLM-KO mice presented with reduced basal plasma glucose and insulin levels. During an insulin tolerance test, GCLM-KO mice showed a normal fall in glycemia, indicating normal insulin secretion. However, during the recovery phase, plasma glucose levels remained lower for longer in KO mice despite normal plasma glucagon levels. This is consistent with a normal counterregulatory hormonal response but impaired mobilization of glucose from endogenous stores. Following a resident-intruder stress, during which stress hormones mobilize glucose from hepatic glycogen stores, KO mice showed a lower hyperglycemic level despite higher plasma cortisol levels when compared to WT mice. The lower hepatic glycogen levels observed in GCLM-KO mice could explain the impaired glycogen mobilization following induced hypoglycemia. Altogether, our results indicate that reduced liver glycogen availability, as observed in GCLM-KO mice, could be at the origin of their lower basal and challenged glycemia. Further studies will be necessary to understand how a GSH deficit, typically observed in GCLM-KO mice, leads to a deficit in liver glycogen storage.

20.
PLoS One ; 10(1): e0116760, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25629159

RESUMEN

MicroRNAs (miRNAs) are small, non-coding RNA molecules that regulate gene expression post-transcriptionally. MiRNAs are implicated in various biological processes associated with obesity, including adipocyte differentiation and lipid metabolism. We used a neuronal-specific inhibition of miRNA maturation in adult mice to study the consequences of miRNA loss on obesity development. Camk2a-CreERT2 (Cre+) and floxed Dicer (Dicerlox/lox) mice were crossed to generate tamoxifen-inducible conditional Dicer knockouts (cKO). Vehicle- and/or tamoxifen-injected Cre+;Dicerlox/lox and Cre+;Dicer+/+ served as controls. Four cohorts were used to a) measure body composition, b) follow food intake and body weight dynamics, c) evaluate basal metabolism and effects of food deprivation, and d) assess the brain transcriptome consequences of miRNA loss. cKO mice developed severe obesity and gained 18 g extra weight over the 5 weeks following tamoxifen injection, mainly due to increased fat mass. This phenotype was highly reproducible and observed in all 38 cKO mice recorded and in none of the controls, excluding possible effects of tamoxifen or the non-induced transgene. Development of obesity was concomitant with hyperphagia, increased food efficiency, and decreased activity. Surprisingly, after reaching maximum body weight, obese cKO mice spontaneously started losing weight as rapidly as it was gained. Weight loss was accompanied by lowered O2-consumption and respiratory-exchange ratio. Brain transcriptome analyses in obese mice identified several obesity-related pathways (e.g. leptin, somatostatin, and nemo-like kinase signaling), as well as genes involved in feeding and appetite (e.g. Pmch, Neurotensin) and in metabolism (e.g. Bmp4, Bmp7, Ptger1, Cox7a1). A gene cluster with anti-correlated expression in the cerebral cortex of post-obese compared to obese mice was enriched for synaptic plasticity pathways. While other studies have identified a role for miRNAs in obesity, we here present a unique model that allows for the study of processes involved in reversing obesity. Moreover, our study identified the cortex as a brain area important for body weight homeostasis.


Asunto(s)
Eliminación de Gen , MicroARNs/genética , MicroARNs/metabolismo , Neuronas/metabolismo , Obesidad/genética , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Animales , Metabolismo Basal/genética , Encéfalo/metabolismo , Exones , Ayuno , Técnicas de Silenciamiento del Gen , Estudios de Asociación Genética , Genotipo , Hiperfagia/genética , Locomoción , Ratones , Ratones Noqueados , Obesidad/metabolismo , Especificidad de Órganos/genética , Fenotipo , Tamoxifeno/administración & dosificación , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA