Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
ScientificWorldJournal ; 2021: 1583154, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34531707

RESUMEN

Ayu-narezushi, a traditional Japanese fermented food, comprises abundant levels of lactic acid bacteria (LAB) and free amino acids. This study aimed to examine the potential beneficial effects of ayu-narezushi and investigated whether ayu-narezushi led to improvements in the Tsumura Suzuki obese diabetes (TSOD) mice model of spontaneous metabolic syndrome because useful LAB are known as probiotics that regulate intestinal function. In the present study, the increased body weight of the TSOD mice was attenuated in those fed the ayu-narezushi-comprised chow (ayu-narezushi group) compared with those fed the normal rodent chow (control group). Serum triglyceride and cholesterol levels were significantly lower in the Ayu-narezushi group than in the control group at 24 weeks of age. Furthermore, hepatic mRNA levels of carnitine-palmitoyl transferase 1 and acyl-CoA oxidase, which related to fatty acid oxidation, were significantly increased in the ayu-narezushi group than in the control group at 24 weeks of age. In conclusion, these results suggested that continuous feeding with ayu-narezushi improved obesity and dyslipidemia in the TSOD mice and that the activation of fatty acid oxidation in the liver might contribute to these improvements.


Asunto(s)
Modelos Animales de Enfermedad , Alimentos Fermentados , Metabolismo de los Lípidos , Síndrome Metabólico/dietoterapia , Osmeriformes , Acil-CoA Oxidasa/biosíntesis , Acil-CoA Oxidasa/genética , Animales , Peso Corporal , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Colesterol/sangre , Dislipidemias/dietoterapia , Dislipidemias/genética , Inducción Enzimática , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Grasa Intraabdominal/química , Grasa Intraabdominal/patología , Japón , Hígado/metabolismo , Síndrome Metabólico/sangre , Síndrome Metabólico/genética , Ratones , Ratones Obesos , Obesidad/dietoterapia , Obesidad/genética , Oryza , Oxidación-Reducción , PPAR alfa/biosíntesis , PPAR alfa/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Cloruro de Sodio , Triglicéridos/sangre
2.
Am J Physiol Endocrinol Metab ; 311(3): E649-60, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27507552

RESUMEN

Impaired skeletal muscle mitochondrial fatty acid oxidation (mFAO) has been implicated in the etiology of insulin resistance. Carnitine palmitoyltransferase-1 (CPT1) is a key regulatory enzyme of mFAO whose activity is inhibited by malonyl-CoA, a lipogenic intermediate. Whereas increasing CPT1 activity in vitro has been shown to exert a protective effect against lipid-induced insulin resistance in skeletal muscle cells, only a few studies have addressed this issue in vivo. We thus examined whether a direct modulation of muscle CPT1/malonyl-CoA partnership is detrimental or beneficial for insulin sensitivity in the context of diet-induced obesity. By using a Cre-LoxP recombination approach, we generated mice with skeletal muscle-specific and inducible expression of a mutated CPT1 form (CPT1mt) that is active but insensitive to malonyl-CoA inhibition. When fed control chow, homozygous CPT1mt transgenic (dbTg) mice exhibited decreased CPT1 sensitivity to malonyl-CoA inhibition in isolated muscle mitochondria, which was sufficient to substantially increase ex vivo muscle mFAO capacity and whole body fatty acid utilization in vivo. Moreover, dbTg mice were less prone to high-fat/high-sucrose (HFHS) diet-induced insulin resistance and muscle lipotoxicity despite similar body weight gain, adiposity, and muscle malonyl-CoA content. Interestingly, these CPT1mt-protective effects in dbTg-HFHS mice were associated with preserved muscle insulin signaling, increased muscle glycogen content, and upregulation of key genes involved in muscle glucose metabolism. These beneficial effects of muscle CPT1mt expression suggest that a direct modulation of the malonyl-CoA/CPT1 partnership in skeletal muscle could represent a potential strategy to prevent obesity-induced insulin resistance.


Asunto(s)
Carnitina O-Palmitoiltransferasa/biosíntesis , Dieta Alta en Grasa/efectos adversos , Sacarosa en la Dieta/efectos adversos , Resistencia a la Insulina , Malonil Coenzima A/metabolismo , Músculo Esquelético/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/genética , Metabolismo Energético/efectos de los fármacos , Glucosa/metabolismo , Masculino , Malonil Coenzima A/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/metabolismo , Mutación/genética , Obesidad/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
3.
Biosci Biotechnol Biochem ; 78(9): 1584-91, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25209508

RESUMEN

Many animal studies on improvement of lipid metabolism, using dietary components, fast the animals on the final day of the feeding. Although fasting has a significant impact on lipid metabolism, its time-dependent influence is not fully understood. We examined the effects of several fasting times on lipid metabolism. Rats fed with a semisynthetic diet for 2 wk were killed after 0 (9:00 am), 6 (7:00 am-1:00 pm), 9 (0:00 am-9:00 am), and 13 h (8:00 pm-9:00 am) of fasting. Compared to the 0 h group, marked reduction of liver weight and hepatic triacylglycerol content was observed in the 9 and 13 h groups. Activities of hepatic enzymes involved in fatty acid synthesis gradually decreased during fasting. In contrast, drastic time-dependent reduction of gene expression, of the enzymes, was observed. Expression of carnitine palmitoyltransferase mRNA was higher in the fasting groups than in the 0 h group. Our study showed that fasting has a significant impact on several parameters related to lipid metabolism in rat liver.


Asunto(s)
Carnitina O-Palmitoiltransferasa/biosíntesis , Ayuno/fisiología , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Animales , Ayuno/metabolismo , Regulación Enzimológica de la Expresión Génica , Hígado/enzimología , ARN Mensajero/biosíntesis , Ratas
4.
J Biol Chem ; 287(25): 21224-32, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22539351

RESUMEN

The brain-specific isoform carnitine palmitoyltransferase 1C (CPT1C) has been implicated in the hypothalamic regulation of food intake and energy homeostasis. Nevertheless, its molecular function is not completely understood, and its role in other brain areas is unknown. We demonstrate that CPT1C is expressed in pyramidal neurons of the hippocampus and is located in the endoplasmic reticulum throughout the neuron, even inside dendritic spines. We used molecular, cellular, and behavioral approaches to determine CPT1C function. First, we analyzed the implication of CPT1C in ceramide metabolism. CPT1C overexpression in primary hippocampal cultured neurons increased ceramide levels, whereas in CPT1C-deficient neurons, ceramide levels were diminished. Correspondingly, CPT1C knock-out (KO) mice showed reduced ceramide levels in the hippocampus. At the cellular level, CPT1C deficiency altered dendritic spine morphology by increasing immature filopodia and reducing mature mushroom and stubby spines. Total protrusion density and spine head area in mature spines were unaffected. Treatment of cultured neurons with exogenous ceramide reverted the KO phenotype, as did ectopic overexpression of CPT1C, indicating that CPT1C regulation of spine maturation is mediated by ceramide. To study the repercussions of the KO phenotype on cognition, we performed the hippocampus-dependent Morris water maze test on mice. Results show that CPT1C deficiency strongly impairs spatial learning. All of these results demonstrate that CPT1C regulates the levels of ceramide in the endoplasmic reticulum of hippocampal neurons, and this is a relevant mechanism for the correct maturation of dendritic spines and for proper spatial learning.


Asunto(s)
Carnitina O-Palmitoiltransferasa/biosíntesis , Ceramidas/metabolismo , Dendritas/enzimología , Metabolismo Energético/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Metabolismo de los Lípidos/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Células Piramidales/enzimología , Animales , Conducta Animal/fisiología , Carnitina O-Palmitoiltransferasa/genética , Células Cultivadas , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/genética , Errores Innatos del Metabolismo Lipídico/enzimología , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/patología , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Células Piramidales/citología
5.
Br J Nutr ; 110(11): 2114-26, 2013 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23731955

RESUMEN

The intake of whey protein isolate (WPI) is known to reduce high-fat diet (HFD)-induced body-weight gain and adiposity. However, the molecular mechanisms are not fully understood. To this end, we fed C57BL/6J mice for 8 weeks with diets containing 10 % energy as fat (low-fat diet, LFD) or 45 % energy as fat (HFD) enriched with either 20 % energy as casein (LFD and HFD) or WPI (high-fat WPI). Metabolic parameters and the hypothalamic and epididymal adipose tissue expression of energy balance-related genes were investigated. The HFD increased fat mass and plasma leptin levels and decreased the dark-phase energy intake, meal number, RER, and metabolic (VO2 and heat) and locomotor activities compared with the LFD. The HFD increased the hypothalamic tissue mRNA expression of the leptin receptor, insulin receptor (INSR) and carnitine palmitoyltransferase 1b (CPT1b). The HFD also reduced the adipose tissue mRNA expression of GLUT4 and INSR. In contrast, WPI reduced fat mass, normalised dark-phase energy intake and increased meal size in HFD-fed mice. The dietary protein did not have an impact on plasma leptin, insulin, glucose or glucagon-like peptide 1 levels, but increased plasma TAG levels in HFD-fed mice. At a cellular level, WPI significantly reduced the HFD-associated increase in the hypothalamic tissue mRNA expression of the leptin receptor, INSR and CPT1b. Also, WPI prevented the HFD-induced reduction in the adipose tissue mRNA expression of INSR and GLUT4. In comparison with casein, the effects of WPI on energy intake and hypothalamic and adipose tissue gene expression may thus represent a state of reduced susceptibility to weight gain on a HFD.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Dieta Alta en Grasa , Ingestión de Energía , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Proteínas de la Leche/uso terapéutico , Sobrepeso/dietoterapia , Adiposidad , Animales , Conducta Animal , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Dieta Alta en Grasa/efectos adversos , Susceptibilidad a Enfermedades , Epidídimo , Conducta Alimentaria , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Hipotálamo/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Sobrepeso/etiología , Receptor de Insulina/biosíntesis , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de Leptina/biosíntesis , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Proteína de Suero de Leche
6.
Br J Nutr ; 110(11): 1968-77, 2013 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23656796

RESUMEN

In the present study, quadruplicate groups of juvenile Atlantic salmon (Salmo salar) were fed plant protein-based diets with increasing arginine inclusions (range 28·8-37·4 g/kg DM) to investigate whether arginine supplementation affects growth and lipid accumulation through an elevated polyamine turnover. Dietary lysine was held at a constant concentration, just below the requirement. All other amino acids were balanced and equal in the diets. Arginine supplementation increased protein and fat accretion, without affecting the hepatosomatic or visceralsomatic indices. Dietary arginine correlated with putrescine in the liver (R 0·78, P= 0·01) and with ornithine in the muscle, liver and plasma (P= 0·0002, 0·003 and 0·0002, respectively). The mRNA of ornithine decarboxylase, the enzyme producing putrescine, was up-regulated in the white adipose tissue of fish fed the high-arginine inclusion compared with those fed the low-arginine diet. Concomitantly, spermidine/spermine-(N1)-acetyltransferase, the rate-limiting enzyme for polyamine turnover that consumes acetyl-CoA, showed an increased activity in the liver of fish fed the arginine-supplemented diets. In addition, lower acetyl-CoA concentrations were observed in the liver of fish fed the high-arginine diet, while ATP, which is used in the process of synthesising spermidine and spermine, did not show a similar trend. Gene expression of the rate-limiting enzyme for ß-oxidation of long-chain fatty acids, carnitine palmitoyl transferase-1, was up-regulated in the liver of fish fed the high-arginine diet. Taken together, the data support that increased dietary arginine activates polyamine turnover and ß-oxidation in the liver of juvenile Atlantic salmon and may act to improve the metabolic status of the fish.


Asunto(s)
Arginina/metabolismo , Dieta/veterinaria , Suplementos Dietéticos , Metabolismo Energético , Poliaminas/metabolismo , Salmo salar/metabolismo , Acetiltransferasas/biosíntesis , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/crecimiento & desarrollo , Tejido Adiposo Blanco/metabolismo , Animales , Acuicultura , Arginina/administración & dosificación , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Dieta/efectos adversos , Proteínas en la Dieta/efectos adversos , Proteínas en la Dieta/metabolismo , Inducción Enzimática , Proteínas de Peces/biosíntesis , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Isoenzimas/biosíntesis , Isoenzimas/genética , Isoenzimas/metabolismo , Metabolismo de los Lípidos , Hígado/enzimología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Ornitina/sangre , Ornitina/metabolismo , Ornitina Descarboxilasa/biosíntesis , Ornitina Descarboxilasa/genética , Ornitina Descarboxilasa/metabolismo , Proteínas de Plantas/efectos adversos , Proteínas de Plantas/metabolismo , Putrescina/metabolismo , Salmo salar/sangre , Salmo salar/crecimiento & desarrollo
7.
J Biol Chem ; 286(27): 23799-807, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21586575

RESUMEN

The conversion of pyruvate to acetyl-CoA in mitochondria is catalyzed by the pyruvate dehydrogenase complex (PDC). Activity of PDC is inhibited by phosphorylation via the pyruvate dehydrogenase kinases (PDKs). Here, we examined the regulation of Pdk4 gene expression by the CCAAT/enhancer-binding protein ß (C/EBPß). C/EBPß modulates the expression of multiple hepatic genes including those involved in metabolism, development, and inflammation. We found that C/EBPß induced Pdk4 gene expression and decreased PDC activity. This transcriptional induction was mediated through two C/EBPß binding sites in the Pdk4 promoter. C/EBPß participates in the hormonal regulation of gluconeogenic genes. Previously, we reported that Pdk4 was induced by thyroid hormone (T(3)). Therefore, we investigated the role of C/EBPß in the T(3) regulation of Pdk4. T(3) increased C/EBPß abundance in primary rat hepatocytes. Knockdown of C/EBPß with siRNA diminished the T(3) induction of the Pdk4 and carnitine palmitoyltransferase (Cpt1a) genes. CPT1a is an initiating step in the mitochondrial oxidation of long chain fatty acids. Our results indicate that C/EBPß stimulates Pdk4 expression and participates in the T(3) induction of the Cpt1a and Pdk4 genes.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Hepatocitos/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Elementos de Respuesta/fisiología , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Gluconeogénesis/fisiología , Células Hep G2 , Hepatocitos/citología , Humanos , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Complejo Piruvato Deshidrogenasa/biosíntesis , Complejo Piruvato Deshidrogenasa/genética , Ratas , Triyodotironina/genética , Triyodotironina/metabolismo
8.
Can J Physiol Pharmacol ; 90(1): 99-111, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22188509

RESUMEN

Diabetes mellitus (DM) is commonly associated with metabolic and cardiac dysfunctions. The aim of this study was to examine the effect of ghrelin on metabolic and cardiac dysfunctions in a type-2 diabetes mellitus (T2DM) rat model. For this, 48 male adult Sprague-Dawley rats were divided equally into 4 groups: Group I, fed normal chow, served as normal control group; Groups II-IV, were fed a high-fat diet for 2 weeks followed by injection of streptozotocin (STZ) (35 mg/kg body mass) to create a model of T2DM; Group II, were not treated; Group III, were treated with the vehicle (saline); Group IV, were treated with ghrelin (40 µg/kg body mass) twice daily for 10 days. The untreated diabetic rats showed a significant increase in serum fasting blood glucose, insulin homeostasis model assessment (HOMA) index, triglycerides (TGs), low-density lipoprotein cholesterol (LDL-C), total serum cholesterol (TC), and body mass, with a decrease in high-density lipoprotein cholesterol (HDL-C) (p < 0.05). Hearts isolated from diabetic rats showed a significant increase in myocardial fat content, a significant decrease in GLUT4, and an increase in acyl-CoA oxidase enzyme mRNA (p < 0.05). Ghrelin administration for 10 days caused a significant improvement in lipid profile, HOMA index, and body mass, and significantly corrected the myocardial mass, significantly reduced the fat content of the myocardium, significantly increased GLUT4, and decreased acyl CoA oxidase mRNA (p < 0.05). Thus, ghrelin improves both the metabolic functions and the disturbed energy metabolism in the cardiac muscle of obese diabetic rats.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Ghrelina/uso terapéutico , Hipoglucemiantes/uso terapéutico , Miocardio/metabolismo , Acil-CoA Oxidasa/biosíntesis , Animales , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Carnitina O-Palmitoiltransferasa/biosíntesis , Colesterol/sangre , Diabetes Mellitus Experimental/patología , Dieta Alta en Grasa/efectos adversos , Ghrelina/farmacología , Transportador de Glucosa de Tipo 4/biosíntesis , Corazón/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Hipertrofia/complicaciones , Hipertrofia/tratamiento farmacológico , Hipoglucemiantes/farmacología , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Miocardio/enzimología , Miocardio/patología , Ratas , Ratas Sprague-Dawley , Triglicéridos/sangre
9.
BMC Cancer ; 11: 56, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21294903

RESUMEN

BACKGROUND: Mammary carcinomas have been associated with a high-fat diet, and the rate of breast cancer in overweight post-menopausal women is up to 50% higher than in their normal-weight counterparts. Epidemiological studies suggest that prolactin (PRL) plays a role in the progression of breast cancer. The current study examined breast cancer as a metabolic disease in the context of altered fatty acid catabolism by examining the effect of PRL on carnitine palmitoyl transferase 1 (CPT1), an enzyme that shuttles long-chain fatty acids into the mitochondrial matrix for ß-oxidation. The effect of PRL on the adenosine 5'-monophosphate-activated protein kinase (AMPK) energy sensing pathway was also investigated. METHODS: MCF-7 and MDA-MB-231 breast cancer cells and 184B5 normal breast epithelial cells treated with 100 ng/ml of PRL for 24 hr were used as in vitro models. Real-time PCR was employed to quantify changes in mRNA levels and Western blotting was carried out to evaluate changes at the protein level. A non-radioactive CPT1 enzyme activity assay was established and siRNA transfections were performed to transiently knock down specific targets in the AMPK pathway. RESULTS: PRL stimulation increased the expression of CPT1A (liver isoform) at the mRNA and protein levels in both breast cancer cell lines, but not in 184B5 cells. In response to PRL, a 20% increase in CPT1 enzyme activity was observed in MDA-MB-231 cells. PRL treatment resulted in increased phosphorylation of the α catalytic subunit of AMPK at Thr172, as well as phosphorylation of acetyl-CoA carboxylase (ACC) at Ser79. A siRNA against liver kinase B1 (LKB1) reversed these effects in breast cancer cells. PRL partially restored CPT1 activity in breast cancer cells in which CPT1A, LKB1, or AMPKα-1 were knocked down. CONCLUSIONS: PRL enhances fatty acid ß-oxidation by stimulating CPT1 expression and/or activity in MCF-7 and MDA-MB-231 breast cancer cells. These PRL-mediated effects are partially dependent on the LKB1-AMPK pathway, although the regulation of CPT1 is also likely to be influenced by other mechanisms. Ultimately, increased CPT1 enzyme activity may contribute to fueling the high energy demands of cancer cells. Targeting metabolic pathways that are governed by PRL, which has already been implicated in the progression of breast cancer, may be of therapeutic benefit.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Prolactina/farmacología , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Oxidación-Reducción , Fosforilación/efectos de los fármacos , Prolactina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteínas Recombinantes/farmacología , Transfección
10.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33465052

RESUMEN

Chronic kidney disease (CKD) remains a major epidemiological, clinical, and biomedical challenge. During CKD, renal tubular epithelial cells (TECs) present a persistent inflammatory and profibrotic response. Fatty acid oxidation (FAO), the main source of energy for TECs, is reduced in kidney fibrosis and contributes to its pathogenesis. To determine whether gain of function in FAO (FAO-GOF) could protect from fibrosis, we generated a conditional transgenic mouse model with overexpression of the fatty acid shuttling enzyme carnitine palmitoyl-transferase 1A (CPT1A) in TECs. Cpt1a-knockin (CPT1A-KI) mice subjected to 3 models of renal fibrosis (unilateral ureteral obstruction, folic acid nephropathy [FAN], and adenine-induced nephrotoxicity) exhibited decreased expression of fibrotic markers, a blunted proinflammatory response, and reduced epithelial cell damage and macrophage influx. Protection from fibrosis was also observed when Cpt1a overexpression was induced after FAN. FAO-GOF restored oxidative metabolism and mitochondrial number and enhanced bioenergetics, increasing palmitate oxidation and ATP levels, changes that were also recapitulated in TECs exposed to profibrotic stimuli. Studies in patients showed decreased CPT1 levels and increased accumulation of short- and middle-chain acylcarnitines, reflecting impaired FAO in human CKD. We propose that strategies based on FAO-GOF may constitute powerful alternatives to combat fibrosis inherent to CKD.


Asunto(s)
Carnitina O-Palmitoiltransferasa/biosíntesis , Regulación Enzimológica de la Expresión Génica , Túbulos Renales/enzimología , Insuficiencia Renal Crónica/prevención & control , Animales , Carnitina O-Palmitoiltransferasa/genética , Modelos Animales de Enfermedad , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Fibrosis , Túbulos Renales/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/genética
11.
Biochem Biophys Res Commun ; 393(1): 89-94, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20102700

RESUMEN

OBJECTIVE: The accumulation of lipids in macrophages contributes to the development of atherosclerosis. Strategies to reduce lipid accumulation in macrophages may have therapeutic potential for preventing and treating atherosclerosis and cardiovascular complications. The antidiabetic drug metformin has been reported to reduce lipid accumulation in adipocytes. In this study, we examined the effects of metformin on lipid accumulation in macrophages and investigated the mechanisms involved. METHODS AND RESULTS: We observed that metformin significantly reduced palmitic acid (PA)-induced intracellular lipid accumulation in macrophages. Metformin promoted the expression of carnitine palmitoyltransferase I (CPT-1), while reduced the expression of fatty acid-binding protein 4 (FABP4) which was involved in PA-induced lipid accumulation. Quantitative real-time PCR showed that metformin regulates FABP4 expression at the transcriptional level. We identified forkhead transcription factor FOXO1 as a positive regulator of FABP4 expression. Inhibiting FOXO1 expression with FOXO1 siRNA significantly reduced basal and PA-induced FABP4 expression. Overexpression of wild-type FOXO1 and constitutively active FOXO1 significantly increased FABP4 expression, whereas dominant negative FOXO1 dramatically decreased FABP4 expression. Metformin reduced FABP4 expression by promoting FOXO1 nuclear exclusion and subsequently inhibiting its activity. CONCLUSIONS: Taken together, these results suggest that metformin reduces lipid accumulation in macrophages by repressing FOXO1-mediated FABP4 transcription. Thus, metformin may have a protective effect against lipid accumulation in macrophages and may serve as a therapeutic agent for preventing and treating atherosclerosis in metabolic syndrome.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/genética , Factores de Transcripción Forkhead/metabolismo , Hipoglucemiantes/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Metformina/farmacología , Transcripción Genética/efectos de los fármacos , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Carnitina O-Palmitoiltransferasa/biosíntesis , Línea Celular Tumoral , Regulación hacia Abajo , Proteína Forkhead Box O1 , Humanos , Hipoglucemiantes/uso terapéutico , Metabolismo de los Lípidos/genética , Macrófagos/metabolismo , Metformina/uso terapéutico , Ácido Palmítico/farmacología , Regulación hacia Arriba
12.
Biochim Biophys Acta ; 1778(6): 1382-9, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18359285

RESUMEN

Carnitine palmitoyltransferase (CPT) I is regulated by several genetic and non-genetic factors including allosteric inhibition, mitochondrial membrane composition and/or fluidity and transcriptional regulation of enzyme content. To determine the intrinsic differences in these regulating factors that may result in differences between tissues in fatty acid oxidation ability, mitochondria were isolated from red, white and heart muscles and liver tissue from rainbow trout. Maximal activity (V(max)) for beta-oxidation enzymes and citrate synthase per mg tissue protein as well as CPT I in isolated mitochondria followed a pattern across tissues of red muscle>heart>white muscle>liver suggesting both quantitative and qualitative differences in mitochondria. CPT I inhibition showed a similar pattern with the highest malonyl-CoA concentration to inhibit activity by 50% (IC(50)) found in red muscle while liver had the lowest. Tissue malonyl-CoA content was highest in white muscle with no differences between the other tissues. Interestingly, the gene expression profiles did not follow the same pattern as the tissue enzyme activity. CPT I mRNA expression was greatest in heart>red muscle>white muscle>liver. In contrast, PPARalpha mRNA was greatest in the liver>red muscle>heart>white muscle. There were no significant differences in the mRNA expression of PPARbeta between tissues. As well, no significant differences were found in the mitochondrial membrane composition between tissues, however, there was a tendency for red muscle to exhibit higher proportions of PUFAs as well as a decreased PC:PE ratio, both of which would indicate increased membrane fluidity. In fact, there were significant correlations between IC(50) of CPT I for malonyl-CoA and indicators of membrane fluidity across tissues. This supports the notion that sensitivity of CPT I to its allosteric regulator could be modulated by changes in mitochondrial membrane composition and/or fluidity.


Asunto(s)
Carnitina O-Palmitoiltransferasa/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Fluidez de la Membrana/fisiología , Membranas Mitocondriales/enzimología , Proteínas Mitocondriales/biosíntesis , Oncorhynchus mykiss/metabolismo , Regulación Alostérica/fisiología , Animales , Carnitina O-Palmitoiltransferasa/aislamiento & purificación , Ácidos Grasos Insaturados/metabolismo , Malonil Coenzima A/metabolismo , Membranas Mitocondriales/química , Especificidad de Órganos/fisiología , Oxidación-Reducción , PPAR alfa/biosíntesis , PPAR-beta/biosíntesis , ARN Mensajero/biosíntesis , Transcripción Genética/fisiología
13.
Biochem Biophys Res Commun ; 378(1): 68-72, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-19013135

RESUMEN

We previously demonstrated that adiponectin has anti-fibrogenic and anti-inflammatory effects in the liver of mouse models of various liver diseases. However, its role in liver regeneration remains unclear. The aim of this study was to determine the role of adiponectin in liver regeneration. We assessed liver regeneration after partial hepatectomy in wild-type (WT) and adiponectin knockout (KO) mice. We analyzed DNA replication and various signaling pathways involved in cell proliferation and metabolism. Adiponectin KO mice exhibited delayed DNA replication and increased lipid accumulation in the regenerating liver. The expression levels of peroxisome proliferator-activated receptor (PPAR) alpha and carnitine palmitoyltransferase-1 (CPT-1), a key enzyme in mitochondrial fatty acid oxidation, were decreased in adiponectin KO mice, suggesting possible contribution of altered fat metabolism to these phenomena. Collectively, the present results highlight a new role for adiponectin in the process of liver regeneration.


Asunto(s)
Regeneración Hepática , Hígado/fisiología , Adiponectina/genética , Adiponectina/fisiología , Animales , Carnitina O-Palmitoiltransferasa/biosíntesis , Ciclo Celular , Ácidos Grasos/metabolismo , Expresión Génica , Hepatectomía , Hepatocitos/citología , Hepatocitos/metabolismo , Hígado/citología , Hígado/metabolismo , Regeneración Hepática/genética , Ratones , Ratones Noqueados , Oxidación-Reducción , PPAR alfa/biosíntesis , ARN Mensajero/biosíntesis , Triglicéridos/metabolismo
14.
Br J Nutr ; 102(6): 803-15, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19298684

RESUMEN

Conjugated linoleic acid (CLA) isomers have been reported to reduce body weight and beneficially affect glucose metabolism in animals, but the results are inconsistent and seem to depend on animal model and type of CLA isomer. In the present study, feeding male Zucker fa/fa rats diets supplemented with 1% trans-10, cis-12-CLA for 10 d reduced the liver TAG content without improving the overall adiposity, and enhanced hepatic mitochondrial and peroxisomal beta-oxidation. The increased carnitine palmitoyltransferase (CPT)-I activity and mRNA level as well as the increased n-3:n-6 PUFA ratio in liver suggest that trans-10, cis-12-CLA increased the hepatic beta-oxidation by stimulation of PPARalpha. The reduced hepatic TAG content may be partly due to lower activity of stearoyl-CoA desaturase, as the ratios of 18 : 1n-9:18 : 0 and 16 : 1n-7:16 : 0 were reduced in liver. Trans-10, cis-12-CLA increased the CPT-I mRNA in retroperitoneal white adipose tissue (WAT), and increased uncoupling protein-2 mRNA in epididymal and inguinal WAT depots. Leptin mRNA level was decreased in all examined WAT depots, implying reduced insulin sensitivity. The resistin mRNA level was increased in all WAT depots, whereas adiponectin mRNA was reduced in inguinal and retroperitoneal WAT. The present results suggest that dietary supplementation with trans-10, cis-12-CLA may increase the catabolism of lipids in liver and adipose tissue. Moreover, we provide new data suggesting that trans-10, cis-12-CLA modulates the expression of resistin and adiponectin inversely in adipose tissue. Hence, the present results suggest that trans-10, cis-12-CLA may have some beneficial effects on lipid metabolism and adiposity but possibly reduces insulin sensitivity.


Asunto(s)
Leptina/biosíntesis , Ácidos Linoleicos Conjugados/farmacología , Hígado/metabolismo , Obesidad/metabolismo , Triglicéridos/metabolismo , Adipoquinas/biosíntesis , Adipoquinas/genética , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Crecimiento/efectos de los fármacos , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Leptina/genética , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/patología , Masculino , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/genética , Obesidad/patología , Obesidad/fisiopatología , Tamaño de los Órganos/efectos de los fármacos , ARN Mensajero/genética , Ratas , Ratas Zucker , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Triglicéridos/sangre , Proteína Desacopladora 2
15.
Biochem Biophys Res Commun ; 375(4): 517-21, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18706397

RESUMEN

Effects of non-esterified fatty acids (FAs) are accentuated when applied together with elevated glucose through preferential use of glucose as fuel, which leads to decreased oxidation of FAs. We examined how over-expression of the mitochondrial FA transporter carnitine palmitoyltransferase 1 (CPT1) affects glucose-stimulated insulin secretion (GSIS), apoptosis and ER stress in INS-1E cells cultured in the presence of elevated levels of glucose and palmitate. INS-1E cells were infected with Tet-ON regulated adenovirus containing CPT1 and cultured for 48h in the presence of 0.5mM palmitate and 20mM glucose. Over-expressing CPT1 lowered basal insulin secretion in a dose-dependent manner thereby improving GSIS from INS-1E cells. Also, apoptosis was alleviated and ER-stress markers p-eIF2alpha and CHOP were decreased in cells over-expressing CPT1. We conclude that regulated over-expression of CPT1 is beneficial for glucolipotoxic beta-cells.


Asunto(s)
Carnitina O-Palmitoiltransferasa/biosíntesis , Ácidos Grasos no Esterificados/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Palmitatos/metabolismo , Adenoviridae , Animales , Apoptosis , Carnitina O-Palmitoiltransferasa/genética , Línea Celular , Retículo Endoplásmico/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Ácidos Grasos no Esterificados/toxicidad , Glucosa/toxicidad , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Palmitatos/farmacología , Ratas , Factor de Transcripción CHOP/metabolismo
16.
Biochem J ; 403(3): 511-8, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17284167

RESUMEN

The PGC-1s (peroxisome-proliferator-activated receptor gamma co-activators) are a family of transcriptional regulators that induce the expression of various metabolic genes. PGC-1 proteins stimulate genes involved in mitochondrial biogenesis, fatty acid oxidation and hepatic gluconeogenesis. Previous studies have demonstrated that the PGC-1alpha and beta isoforms interact with nuclear receptors through the conserved LXXLL (leucine-X-X-leucine-leucine) motifs. In the present study, we have investigated the mechanisms by which these PGC-1 isoforms stimulate gene expression. We have determined that the N-terminus of PGC-1 is responsible for transcriptional activation. Two conserved peptide motifs were identified in the N-terminus of PGC-1alpha and beta isoforms. These domains were named AD1 and AD2 (activation domain 1 and 2). Deletion of both of these motifs decreased the induction of various PGC-1-regulated genes including the PEPCK (phosphoenolpyruvate carboxykinase) and CPT-I (carnitine palmitoyltransferase-I) genes. It was determined that amino acids containing a negative charge in AD1 and the leucine residues in AD2 were important for the transcriptional induction of the PEPCK and CPT-I genes. Disruption of the AD motifs did not diminish the ability of the PGC-1alpha protein to associate with the PEPCK or CPT-I genes. In addition, deletion of the AD domains did not eliminate the ability of PGC-1alpha to interact with the thyroid hormone receptor. The data indicate that the AD1 and AD2 motifs mediate the induction of many PGC-1- responsive genes, but they do not contribute to the recruitment of PGC-1 to target genes.


Asunto(s)
Estructura Terciaria de Proteína , Factores de Transcripción/química , Secuencia de Aminoácidos , Carnitina O-Palmitoiltransferasa/biosíntesis , Células Cultivadas , Vectores Genéticos/fisiología , Humanos , Regiones Promotoras Genéticas/fisiología , Factores de Transcripción/genética , Activación Transcripcional , Triyodotironina/farmacología
17.
Theranostics ; 8(19): 5452-5468, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555557

RESUMEN

Omental metastasis occurs frequently in gastric cancer (GC) and is considered one of the major causes of gastric cancer-related mortality. Recent research indicated that omental adipocytes might mediate this metastatic predilection. Phosphatidylinositol transfer protein, cytoplasmic 1 (PITPNC1) was identified to have a crucial role in metastasis. However, whether PITPNC1 participates in the interaction between adipocytes and GC omental metastasis is unclear. Methods: We profiled and analyzed the expression of PITPNC1 through analysis of the TCGA database as well as immunohistochemistry staining using matched GC tissues, adjacent normal gastric mucosa tissues (ANTs), and omental metastatic tissues. The regulation of PITPNC1 by adipocytes was explored by co-culture systems. By using both PITPNC1 overexpression and silencing methods, the role of PITPNC1 in anoikis resistance and metastasis was determined through in vitro and in vivo experiments. Results: PITPNC1 was expressed at higher rates in GC tissues than in ANTs; notably, it was higher in omental metastatic lesions. Elevated expression of PITPNC1 predicted higher rates of omental metastasis and a poor prognosis. PITPNC1 promoted anoikis resistance through fatty acid metabolism by upregulating CD36 and CPT1B expression. Further, PITPNC1 was elevated by adipocytes and facilitated GC omental metastasis. Lastly, in vivo studies showed that PITPNC1 was a therapeutic indicator of fatty acid oxidation (FAO) inhibition. Conclusion: Elevated expression of PITPNC1 in GC is correlated with an advanced clinical stage and a poor prognosis. PITPNC1 promotes anoikis resistance through enhanced FAO, which is regulated by omental adipocytes and consequently facilitates GC omental metastasis. Targeting PITPNC1 might present a promising strategy to treat omental metastasis.


Asunto(s)
Adipocitos/patología , Ácidos Grasos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Neoplasias Peritoneales/fisiopatología , Neoplasias Peritoneales/secundario , Neoplasias Gástricas/patología , Neoplasias Gástricas/fisiopatología , Adenocarcinoma/patología , Adenocarcinoma/fisiopatología , Animales , Anoicis , Antígenos CD36/biosíntesis , Carnitina O-Palmitoiltransferasa/biosíntesis , Línea Celular Tumoral , Supervivencia Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Expresión Génica , Silenciador del Gen , Humanos , Inmunohistoquímica , Proteínas de Transporte de Membrana/análisis , Proteínas de Transporte de Membrana/genética , Ratones Desnudos , Modelos Teóricos , Regulación hacia Arriba
18.
Mol Med Rep ; 17(5): 6840-6846, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29512720

RESUMEN

Pioglitazone (PIO) has been reported to be effective for nonalcoholic fatty liver disease (NAFLD) and alogliptin (ALO) may have efficacy against NAFLD progression in patients with type 2 diabetes mellitus (T2DM). The present study examined the effectiveness of ALO in a rodent model of NAFLD and diabetes mellitus. KK­Ay mice were used to produce an NAFLD model via administration of a choline­deficient (CD) diet. To examine the effects of alogliptin, KK­Ay mice were provided with a CD diet with 0.03% ALO and/or 0.02% PIO orally for 8 weeks. Biochemical parameters, pathological alterations and hepatic mRNA levels associated with fatty acid metabolism were assessed. Severe hepatic steatosis was observed in KK­Ay mice fed with a CD diet, which was alleviated by the administration of ALO and/or PIO. ALO administration increased the hepatic carnitine palmitoyltransferase 1a (CPT1a) mRNA expression level and enhanced the Thr172 phosphorylation of AMP­activated protein kinase α (AMPKα) in the liver. PIO administration tended to decrease the hepatic fatty acid synthase mRNA expression level and increase the serum adiponectin level. Homeostasis model of assessment­insulin resistance values tended to improve with ALO and PIO administration. ALO and PIO alleviated hepatic steatosis in KK­Ay mice fed with a CD diet. ALO increased hepatic mRNA expression levels associated with fatty acid oxidation. In addition, the results of the present study suggested that ALO promotes CPT1a expression via Thr172 phosphorylation of AMPKα.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Carnitina O-Palmitoiltransferasa/biosíntesis , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Piperidinas/farmacología , Uracilo/análogos & derivados , Proteínas Quinasas Activadas por AMP/genética , Animales , Carnitina O-Palmitoiltransferasa/genética , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Fosforilación/efectos de los fármacos , Uracilo/farmacología
19.
Cell Death Dis ; 8(10): e3110, 2017 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-29022907

RESUMEN

The p38α mitogen-activated protein kinase (MAPK) has been related to gluconeogenesis and lipid metabolism. However, the roles and related mechanisms of p38α MAPK in intestinal failure (IF)-associated liver steatosis remained poor understood. Here, our experimental evidence suggested that p38α MAPK significantly suppressed the fat accumulation in livers of IF patients mainly through two mechanisms. On the one hand, p38α MAPK increased hepatic bile acid (BA) synthesis by upregulating the expression of the rate-limiting enzyme cholesterol 7-α-hydroxylase (CYP7A1) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), which in turn activated the transcription of the CYP7A1. On the other hand, p38α MAPK promoted fatty acid (FA) ß-oxidation via upregulating peroxisome proliferator-activated receptor alpha (PPARα) and its transcriptional target genes carnitine palmitoyltransferase 1A (CPT1A) and peroxisomal acyl-coenzyme aoxidase 1 (ACOX1). Dual luciferase assays indicated that p38α MAPK increased the transcription of PPARα, PGC-1α and CYP7A1 by upregulating their promoters' activities. In addition, in vitro and in vivo assays indicated p38α MAPK negatively regulates the hepatic steatosis by controlling JNK activation. In conculsion, our findings demonstrate that hepatic p38α MAPK functions as a negative regulator of liver steatosis in maintaining BA synthesis and FAO by antagonizing the c-Jun N-terminal kinase (JNK).


Asunto(s)
Ácidos Grasos/metabolismo , Hígado Graso/patología , Intestinos/patología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Acil-CoA Oxidasa/biosíntesis , Animales , Ácidos y Sales Biliares/biosíntesis , Carnitina O-Palmitoiltransferasa/biosíntesis , Células Cultivadas , Colesterol 7-alfa-Hidroxilasa/biosíntesis , Colesterol 7-alfa-Hidroxilasa/genética , Modelos Animales de Enfermedad , Humanos , Lactante , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metabolismo de los Lípidos , Hígado/patología , PPAR alfa/biosíntesis , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/biosíntesis , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas Sprague-Dawley , Transcripción Genética/genética , Activación Transcripcional
20.
Physiol Genomics ; 27(2): 131-40, 2006 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-16849632

RESUMEN

Tub is a member of a small gene family, the tubby-like proteins (TULPs), with predominant expression in neurons. Mice carrying a mutation in Tub develop retinal and cochlear degeneration as well as late-onset obesity with insulin resistance. During behavioral and metabolic testing, we found that homozygous C57BL/6J-Tub(tub) mice have a lower respiratory quotient than C57BL/6J controls before the onset of obesity, indicating that tubby homozygotes fail to activate carbohydrate metabolism and instead rely on fat metabolism for energy needs. In concordance with this, tubby mice show higher excretion of ketone bodies and accumulation of glycogen in the liver. Quantitation of liver mRNA levels shows that, during the transition from light to dark period, tubby mice fail to induce glucose-6-phosphate dehydrogenase (G6pdh), the rate-limiting enzyme in the pentose phosphate pathway that normally supplies NADPH for de novo fatty acid synthesis and glutathione reduction. Reduced G6PDH protein levels and enzymatic activity in tubby mice lead accordingly to lower levels of NADPH and reduced glutathione (GSH), respectively. mRNA levels for the lipolytic enzymes acetyl-CoA synthetase and carnitine palmitoyltransferase are increased during the dark cycle and decreased during the light period, and several citric acid cycle genes are dysregulated in tubby mice. Examination of hypothalamic gene expression showed high levels of preproorexin mRNA leading to accumulation of orexin peptide in the lateral hypothalamus. We hypothesize that abnormal hypothalamic orexin expression leads to changes in liver carbohydrate metabolism and may contribute to the moderate obesity observed in tubby mice.


Asunto(s)
Metabolismo de los Hidratos de Carbono/genética , Metabolismo Energético/genética , Ratones Mutantes/metabolismo , Proteínas/genética , Acetato CoA Ligasa/biosíntesis , Acetato CoA Ligasa/genética , Proteínas Adaptadoras Transductoras de Señales , Proteína Relacionada con Agouti , Animales , Química Encefálica , Dióxido de Carbono/metabolismo , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Ritmo Circadiano , Ciclo del Ácido Cítrico/genética , Enfermedades Cocleares/genética , Ingestión de Alimentos , Inducción Enzimática/genética , Genes Recesivos , Glucosafosfato Deshidrogenasa/biosíntesis , Glucosafosfato Deshidrogenasa/genética , Glutatión/deficiencia , Homocigoto , Hipotálamo/metabolismo , Resistencia a la Insulina/genética , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Metabolismo de los Lípidos , Lipólisis/genética , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes/genética , Actividad Motora , NADP/deficiencia , Neuropéptido Y/biosíntesis , Neuropéptido Y/genética , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Obesidad/genética , Orexinas , Oxígeno/metabolismo , Consumo de Oxígeno/genética , Vía de Pentosa Fosfato/genética , Proteínas/fisiología , Degeneración Retiniana/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA