Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Toxicol Sci ; 46(1): 11-24, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33408297

RESUMEN

Fibrates and statins have been widely used to reduce triglyceride and cholesterol levels, respectively. Besides its lipid-lowering effect, the side effect of muscle atrophy after fibrate administration to humans has been demonstrated in some studies. Combination therapy with fibrates and statins also increases the risk of rhabdomyolysis. FoxO1, a member of the FoxO forkhead type transcription factor family, is markedly upregulated in skeletal muscle in energy-deprived states and induces muscle atrophy via the expression of E3-ubiquitine ligases. In this study, we investigated the changes in FoxO1 and its targets in murine skeletal muscle with fenofibrate treatment. High doses of fenofibrate (greater than 0.5% (wt/wt)) over one week increased the expression of FoxO1 and its targets in the skeletal muscles of mice and decreased skeletal muscle weight. These fenofibrate-induced changes were diminished in the PPARα knockout mice. When the effect of combination treatment with fenofibrate and lovastatin was investigated, a significant increase in FoxO1 protein levels was observed despite the lack of deterioration of muscle atrophy. Collectively, our findings suggest that a high dose of fenofibrate over one week causes skeletal muscle atrophy via enhancement of FoxO1, and combination treatment with fenofibrate and lovastatin may further increase FoxO1 protein level.


Asunto(s)
Fenofibrato/efectos adversos , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Expresión Génica/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Lovastatina/efectos adversos , Músculo Esquelético/patología , Atrofia Muscular/inducido químicamente , Atrofia Muscular/genética , Animales , Atrofia , Quimioterapia Combinada/efectos adversos , Fenofibrato/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Lovastatina/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Rabdomiólisis/inducido químicamente , Rabdomiólisis/genética
2.
Am J Physiol Endocrinol Metab ; 305(2): E213-29, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23695215

RESUMEN

LKB1 phosphorylates members of the AMP-activated protein kinase (AMPK) family. LKB1 and AMPK in the skeletal muscle are believed to regulate not only fuel oxidation during exercise but also exercise capacity. LKB1 was also required to prevent diaphragm fatigue, which was shown to affect exercise performance. Using mice expressing dominant negative (DN) mutants of LKB1 and AMPK, specifically in the skeletal muscle but not in the heart, we investigated the roles of LKB1 and AMPK activity in exercise performance and the effects of these kinases on the characteristics of respiratory and locomotive muscles. In the diaphragm and gastrocnemius, both AMPK-DN and LKB1-DN mice showed complete loss of AMPKα2 activity, and LKB1-DN mice showed a reduction in LKB1 activity. Exercise capacity was significantly reduced in LKB1-DN mice, with a marked reduction in oxygen consumption and carbon dioxide production during exercise. The diaphragm from LKB1-DN mice showed an increase in myosin heavy chain IIB and glycolytic enzyme expression. Normal respiratory chain function and CPT I activity were shown in the isolated mitochondria from LKB1-DN locomotive muscle, and the expression of genes related to fiber type, mitochondria function, glucose and lipid metabolism, and capillarization in locomotive muscle was not different between LKB1-DN and AMPK-DN mice. We concluded that LKB1 in the skeletal muscle contributes significantly to exercise capacity and oxygen uptake during exercise. LKB1 mediated the change of fiber-type distribution in the diaphragm independently of AMPK and might be responsible for the phenotypes we observed.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Metabolismo Energético/fisiología , Músculo Esquelético/metabolismo , Consumo de Oxígeno/fisiología , Resistencia Física/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Nucleótidos de Adenina/metabolismo , Animales , Western Blotting , Peso Corporal/fisiología , Dióxido de Carbono/metabolismo , Cartilla de ADN , Diafragma/anatomía & histología , Diafragma/metabolismo , Locomoción/fisiología , Malonil Coenzima A/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Microtúbulos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Esquelético/anatomía & histología , Tamaño de los Órganos/fisiología , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
Med Sci Sports Exerc ; 45(7): 1410-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23377836

RESUMEN

PURPOSE: We examined whether continuous and intermittent physical activity (PA) differentially influence fat utilization. METHODS: This was a randomized crossover study. Nine healthy young male participants performed two 39-h (two nights, three days) PA sessions (continuous and intermittent exercise) in a respiratory chamber to measure energy expenditure (EE) and substrate oxidation. Participants used a stationary cycling ergometer continuously for 40 min and then 45 min in the continuous PA trial and for 5 min every 30 min 17 times in the intermittent PA trial. They consumed high-carbohydrate meals corresponding to predicted daily total EE for 3 d before entering the respiratory chamber and four high-fat meals corresponding to predicted total EE in the chamber. RESULTS: Twenty-three-hour RER adjusted for sleeping RER on the preceding day was significantly lower in the intermittent PA trial than that in the continuous PA trial (P = 0.021). Twenty-three-hour RER adjusted for sleeping RER on the preceding day was correlated with accumulated consecutive minutes of METs ≤ 1.5 (3 min or more, r = 0.477; 5 min or more, r = 0.510; 10 min or more, r = 0.605). CONCLUSIONS: The intermittent PA trial induced greater fat utilization than the continuous PA trial. The present study, therefore, suggests that intermittent PA has a beneficial effect on 24-h fat oxidation after consumption of a high-fat meal, which may help prevent weight gain over time.


Asunto(s)
Grasas de la Dieta/metabolismo , Metabolismo Energético , Ejercicio Físico/fisiología , Acelerometría , Adulto , Biomarcadores/sangre , Calorimetría Indirecta , Estudios Cruzados , Carbohidratos de la Dieta/metabolismo , Prueba de Esfuerzo , Humanos , Modelos Lineales , Masculino , Comidas , Consumo de Oxígeno , Factores de Tiempo
4.
Methods Mol Biol ; 952: 99-116, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23100227

RESUMEN

A high saturated fat diet induces fatty liver in mice. The fatty liver is triggered initially by an increase in PPARγ2 protein in the liver, which leads to increased expression of lipogenic genes. Inactivation of PPARγ2 may improve fatty liver induced by a high saturated fat diet. RNA interference of PPARγ2 should be a powerful tool to elucidate the role of PPARγ2 in the development of fatty liver. Here, we describe our method for constructing the adenovirus vector containing short hairpin RNA targeted for PPARγ2. Adenovirus vector-mediated RNA interference for PPARγ2 should be useful for clarifying the biological role of the PPARγ2 pathway in various tissues and for therapeutic application in a variety of diseases.


Asunto(s)
Técnicas de Silenciamiento del Gen/métodos , Hígado/metabolismo , PPAR gamma/deficiencia , PPAR gamma/genética , Adenoviridae/genética , Adenoviridae/fisiología , Animales , Secuencia de Bases , ADN Complementario/genética , Regulación de la Expresión Génica/genética , Vectores Genéticos/genética , Ratones , Oligonucleótidos/genética , Plásmidos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Transformación Genética , Carga Viral
5.
PLoS One ; 6(12): e28290, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22174785

RESUMEN

BACKGROUND: Maximal oxygen uptake (VO(2max)) predicts mortality and is associated with endurance performance. Trained subjects have a high VO(2max) due to a high cardiac output and high metabolic capacity of skeletal muscles. Peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a nuclear receptor coactivator, promotes mitochondrial biogenesis, a fiber-type switch to oxidative fibers, and angiogenesis in skeletal muscle. Because exercise training increases PGC-1α in skeletal muscle, PGC-1α-mediated changes may contribute to the improvement of exercise capacity and VO(2max). There are three isoforms of PGC-1α mRNA. PGC-1α-b protein, whose amino terminus is different from PGC-1α-a protein, is a predominant PGC-1α isoform in response to exercise. We investigated whether alterations of skeletal muscle metabolism by overexpression of PGC-1α-b in skeletal muscle, but not heart, would increase VO(2max) and exercise capacity. METHODOLOGY/PRINCIPAL FINDINGS: Transgenic mice showed overexpression of PGC-1α-b protein in skeletal muscle but not in heart. Overexpression of PGC-1α-b promoted mitochondrial biogenesis 4-fold, increased the expression of fatty acid transporters, enhanced angiogenesis in skeletal muscle 1.4 to 2.7-fold, and promoted exercise capacity (expressed by maximum speed) by 35% and peak oxygen uptake by 20%. Across a broad range of either the absolute exercise intensity, or the same relative exercise intensities, lipid oxidation was always higher in the transgenic mice than wild-type littermates, suggesting that lipid is the predominant fuel source for exercise in the transgenic mice. However, muscle glycogen usage during exercise was absent in the transgenic mice. CONCLUSIONS/SIGNIFICANCE: Increased mitochondrial biogenesis, capillaries, and fatty acid transporters in skeletal muscles may contribute to improved exercise capacity via an increase in fatty acid utilization. Increases in PGC-1α-b protein or function might be a useful strategy for sedentary subjects to perform exercise efficiently, which would lead to prevention of life-style related diseases and increased lifespan.


Asunto(s)
Músculo Esquelético/metabolismo , Consumo de Oxígeno , Transactivadores/metabolismo , Animales , Composición Corporal/fisiología , Metabolismo de los Hidratos de Carbono , Ayuno/fisiología , Regulación de la Expresión Génica , Glucógeno/metabolismo , Humanos , Ácido Láctico/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Miocardio/metabolismo , Especificidad de Órganos , Tamaño de los Orgánulos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Condicionamiento Físico Animal , Isoformas de Proteínas/metabolismo , Factores de Transcripción
6.
J Nutr Biochem ; 22(6): 543-53, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20801631

RESUMEN

The effects of a diet rich in saturated fat on fatty liver formation and the related mechanisms that induce fatty liver were examined. C57BL/6J mice were fed butter or safflower oil as a high-fat (HF) diet (40% fat calories) for 2, 4, 10, or 17 weeks. Although both HF diets induced similar levels of obesity, HF butter-fed mice showed a two to threefold increase in liver triacylglycerol (TG) concentration compared to HF safflower oil-fed mice at 4 or 10 weeks without hyperinsulinemia. At 4 weeks, increases in peroxisome proliferator-activated receptor γ2 (PPARγ2), CD36, and adipose differentiation-related protein (ADRP) mRNAs were observed in HF butter-fed mice; at 10 weeks, an increase in sterol regulatory element-binding protein-1c (SREBP-1c) was observed; at 17 weeks, these increases were attenuated. At 4 weeks, a single injection of adenoviral vector-based short hairpin interfering RNA against PPARγ2 in HF butter-fed mice reduced PPARγ protein and mRNA of its target genes (CD36 and ADRP) by 43%, 43%, and 39%, respectively, with a reduction in liver TG concentration by 38% in 5 days. PPARγ2 knockdown also reduced mRNAs in lipogenic genes (fatty-acid-synthase, stearoyl-CoA desaturase 1, acetyl-CoA carboxylase 1) without alteration of SREBP-1c mRNA. PPARγ2 knockdown reduced mRNAs in genes related to inflammation (CD68, interleukin-1ß, tumor necrosis factor-α, and monocyte chemoattractant protein-1). In conclusion, saturated fatty acid-rich oil induced fatty liver in mice, and this was triggered initially by an increase in PPARγ2 protein in the liver, which led to increased expression of lipogenic genes. Inactivation of PPARγ2 may improve fatty liver induced by HF saturated fat.


Asunto(s)
Mantequilla , Hígado Graso/metabolismo , Hígado/metabolismo , PPAR gamma/metabolismo , Animales , Ácidos Grasos/administración & dosificación , Hígado Graso/etiología , Hígado Graso/genética , Técnicas de Silenciamiento del Gen , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , PPAR gamma/genética , Perilipina-2 , ARN Mensajero/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
7.
Biochem Biophys Res Commun ; 399(1): 98-103, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20643106

RESUMEN

Subjects with high blood levels of inflammatory markers and patients with chronic inflammatory disorders are at high risk for stroke. Dietary restriction (DR) suppresses systemic inflammation to deter age-related chronic diseases. To examine whether DR delays the onset of stroke, 10-week-old stroke-prone spontaneously hypertensive rats (SHRSP) were assigned to either a control (ad libitum) or DR (50% diet of control) group, and day of stroke onset and lifespan were observed. DR markedly delayed the onset of stroke in SHRSP compared to control without affecting blood pressure. Day of stroke onset (median) in the control group was 34days, whereas it was 70days in the DR group. After 2weeks of DR and before the onset of stroke, plasma levels of interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha), and monocyte chemoattractant protein-1 (MCP-1) and their mRNA expression levels in adipose tissue were significantly lower in the DR rats than in the control rats. Intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) mRNA expression levels in cerebrovascular endothelial cells (CVECs), and macrophage infiltration into brain were lower in the DR rats than in the control rats. IL-1beta and TNF-alpha treatment in CVECs increased MCP-1, C-reactive protein, ICAM-1, and VCAM-1 mRNA and their protein levels in vitro. In conclusion, suppression of inflammation in response to DR may lead to a delay in the onset of stroke independent of any effect on blood pressure in SHRSP.


Asunto(s)
Tejido Adiposo/metabolismo , Dieta , Inflamación/dietoterapia , Accidente Cerebrovascular/prevención & control , Animales , Presión Sanguínea , Quimiocina CCL2/metabolismo , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/metabolismo , Masculino , Ratas , Ratas Endogámicas SHR , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
8.
Biochem J ; 427(1): 171-8, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20088826

RESUMEN

FOXO1 (forkhead box O1), a forkhead-type transcription factor whose gene expression is up-regulated in the skeletal muscle during starvation, appears to be a key molecule of energy metabolism and skeletal muscle atrophy. Cathepsin L, a lysosomal proteinase whose expression is also up-regulated in the skeletal muscle during starvation, is induced in transgenic mice overexpressing FOXO1 relative to wild-type littermates. In the present study, we conducted in vivo and in vitro experiments focusing on FOXO1 regulation of Ctsl (cathepsin L gene; CTSL1 in humans) expression in the skeletal muscle. During fasting and refeeding of C57BL/6 mice, Ctsl was regulated in parallel with FOXO1 in the skeletal muscle. Fasting-induced Ctsl expression was attenuated in transgenic mice overexpressing a dominant-negative form of FOXO1 or in skeletal-muscle-specific Foxo1-knockout mice relative to respective wild-type controls. Using C2C12 mouse myoblasts overexpressing a constitutively active form of FOXO1, we showed that FOXO1 induces Ctsl expression. Moreover, we found FOXO1-binding sites in both the mouse Ctsl and human CTSL1 promoters. The luciferase reporter analysis revealed that the mouse Ctsl and human CTSL1 promoters are activated by FOXO1, which is abolished by mutations in the consensus FOXO1-binding sites. Gel mobility-shift and chromatin immunoprecipiation assays showed that FOXO1 is recruited and binds to the Ctsl promoter. The present study provides in vivo and in vitro evidence that Ctsl is a direct target of FOXO1 in the skeletal muscle, thereby suggesting a role for the FOXO1/cathepsin L pathway in fasting-induced skeletal muscle metabolic change and atrophy.


Asunto(s)
Catepsina L/fisiología , Factores de Transcripción Forkhead/fisiología , Regulación de la Expresión Génica , Músculo Esquelético/metabolismo , Animales , Sitios de Unión , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Femenino , Proteína Forkhead Box O1 , Humanos , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/citología , Mioblastos/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Obesity (Silver Spring) ; 18(2): 314-21, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19680236

RESUMEN

Epigenetic mechanisms are likely to be involved in the development of obesity. This study was designed to examine the role of a DNA methyltransferase (Dnmt3a), in obese adipose tissue. The gene expression of Dnmts was examined by quantitative real-time PCR analysis. Transgenic mice overexpressing Dnmt3a in the adipose tissue driven by the aP2 promoter were created (Dnmt3a mice). DNA methylation of downregulated genes was examined using bisulfite DNA methylation analysis. Dnmt3a mice were fed a methyl-supplemented or high-fat diet, and subjected to body weight measurement and gene expression analysis of the adipose tissue. Expression of Dnmt3a was markedly upregulated in the adipose tissue of obese mice. The complementary DNA (cDNA) microarray analysis of Dnmt3a mice revealed a slight decrease in the gene expression of secreted frizzled-related protein 1 (SFRP1) and marked increase in that of interferon responsive factor 9 (IRF9). In the SFRP1 promoter, DNA methylation was not markedly increased in Dnmt3a mice relative to wild-type mice. In experiments with a high-fat diet or methyl-supplemented diet, body weight did not differ significantly with the genotypes. Gene expression levels of inflammatory cytokines such as tumor necrosis factor-alpha (TNF-alpha) and monocyte chemoattractant protein-1 (MCP-1) were higher in Dnmt3a mice than in wild-type mice on a high-fat diet. This study suggests that increased expression of Dnmt3a in the adipose tissue may contribute to obesity-related inflammation. The data highlight the potential role of Dnmt3a in the adult tissue as well as in the developing embryo and cancer.


Asunto(s)
Tejido Adiposo/enzimología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Obesidad/enzimología , Tejido Adiposo/patología , Animales , Células Cultivadas , Quimiocina CCL2/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , ADN Metiltransferasa 3A , Grasas de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/genética , Perfilación de la Expresión Génica/métodos , Genotipo , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética , Obesidad/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba
10.
Mech Ageing Dev ; 130(7): 420-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19426753

RESUMEN

Caloric restriction (CR) is the most robust and reproducible intervention that can extend lifespan in rodents. Studies in invertebrates have led to the identification of genes that regulate lifespan, some of which encode components of the insulin or insulin-like signaling pathway, including DAF-16 (C. elegans) and dFOXO (Drosophila). Mice subjected to CR for 8 weeks showed an increase in FOXO1 mRNA and other longevity-related genes: Gadd 45alpha, glutamine synthase, and catalase in skeletal muscle. To investigate whether FOXO1 expression affects longevity in mammals, transgenic mice were studied that over-express FOXO1 in their skeletal muscle (FOXO1 mice), and in which muscle atrophy occurs. FOXO1 mice showed increases in Gadd 45alpha, and glutamine synthase proteins in skeletal muscle. In FOXO1 mice, the phosphorylation/dephosphorylation state of the p70 S6K and 4E-BP1 proteins were not altered, suggesting that translation initiation of protein synthesis might not be suppressed. The lifespan of FOXO1 mice was similar to their wild-type littermates. FOXO1 overexpression could not prevent aging-induced reduction in catalase, CuZu-SOD, and Mn-SOD mRNA in skeletal muscle. These data suggest that an increase in FOXO1 protein and its activation in skeletal muscle does not extend lifespan in mice.


Asunto(s)
Factores de Transcripción Forkhead/biosíntesis , Regulación de la Expresión Génica , Longevidad/fisiología , Proteínas Musculares/biosíntesis , Músculo Esquelético/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Restricción Calórica , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Catalasa/genética , Catalasa/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Factores Eucarióticos de Iniciación , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Glutamato Sintasa/genética , Glutamato Sintasa/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Iniciación de la Cadena Peptídica Traduccional/fisiología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/fisiología , ARN Mensajero/biosíntesis , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
11.
Cell Metab ; 9(2): 191-202, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19187775

RESUMEN

Acetate is activated to acetyl-CoA by acetyl-CoA synthetase 2 (AceCS2), a mitochondrial enzyme. Here, we report that the activation of acetate by AceCS2 has a specific and unique role in thermogenesis during fasting. In the skeletal muscle of fasted AceCS2(-/-) mice, ATP levels were reduced by 50% compared to AceCS2(+/+) mice. Fasted AceCS2(-/-) mice were significantly hypothermic and had reduced exercise capacity. Furthermore, when fed a low-carbohydrate diet, 4-week-old weaned AceCS2(-/-) mice also exhibited hypothermia accompanied by sustained hypoglycemia that led to a 50% mortality. Therefore, AceCS2 plays a significant role in acetate oxidation needed to generate ATP and heat. Furthermore, AceCS2(-/-) mice exhibited increased oxygen consumption and reduced weight gain on a low-carbohydrate diet. Our findings demonstrate that activation of acetate by AceCS2 plays a pivotal role in thermogenesis, especially under low-glucose or ketogenic conditions, and is crucially required for survival.


Asunto(s)
Acetato CoA Ligasa/fisiología , Metabolismo Energético , Termogénesis/fisiología , Acetato CoA Ligasa/genética , Adenosina Trifosfato/metabolismo , Animales , Ayuno , Hipoglucemia/etiología , Hipotermia Inducida , Ratones , Ratones Noqueados , Consumo de Oxígeno
12.
J Hepatol ; 49(3): 441-50, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18620774

RESUMEN

BACKGROUND/AIMS: We examined whether dietary fish oil can prevent acute ethanol (alcohol)-induced fatty liver. METHODS: Mice were fed safflower oil, fish oil, or safflower oil plus a PPAR alpha activator on the day prior to ethanol administration. Oil red O staining, serum analysis, and RT-PCR were used to analyze ethanol-induced fatty liver. RESULTS: In mice fed safflower oil, ethanol increased liver TG 3-fold, with activation of SREBP-1c and ChREBP, which promote de novo lipogenesis, and increases in expression of mRNAs for PPAR gamma and DGATs mRNAs, which promote TG synthesis. When mice were fed fish oil, ethanol-induced fatty liver was reduced by 73%. Fish oil decreased SREBP-1c activity and increased PPAR alpha activity. However, levels of DGAT1, DGAT2, ChREBP, LPK, and PPAR gamma mRNAs were increased in response to ethanol in mice fed fish oil. Prior administration of Wy14643, PPAR alpha activator, did not inhibit ethanol-induced fatty liver, suggesting that PPAR alpha played little role in prevention of ethanol-induced fatty liver by fish oil. CONCLUSIONS: A single dose of ethanol increases the liver TG level via several mechanisms; however, prior ingestion of fish oil effectively prevents ethanol-induced fatty liver, at least in part, by decreasing basal SREBP-1c activity, especially a marked reduction in SCD1.


Asunto(s)
Grasas Insaturadas en la Dieta/uso terapéutico , Etanol/efectos adversos , Hígado Graso/inducido químicamente , Hígado Graso/prevención & control , Aceites de Pescado/uso terapéutico , Animales , Anticolesterolemiantes/farmacología , Glucemia/metabolismo , Grasas Insaturadas en la Dieta/administración & dosificación , Grasas Insaturadas en la Dieta/farmacología , Relación Dosis-Respuesta a Droga , Ácidos Grasos no Esterificados/sangre , Hígado Graso/metabolismo , Aceites de Pescado/administración & dosificación , Aceites de Pescado/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/metabolismo , Pirimidinas/farmacología , Aceite de Cártamo/uso terapéutico , Estearoil-CoA Desaturasa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo
13.
Endocrinology ; 149(5): 2293-305, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18202130

RESUMEN

Sterol regulatory element binding protein 1c (SREBP1c) is a master regulator of lipogenic gene expression in liver and adipose tissue, where its expression is regulated by a heterodimer of nuclear receptor-type transcription factors retinoid X receptor-alpha (RXRalpha) and liver X receptor-alpha (LXRalpha). Despite the potential importance of SREBP1c in skeletal muscle, little is known about the regulation of SREBP1c in that setting. Here we report that gene expression of RXRgamma is markedly decreased by fasting and is restored by refeeding in mouse skeletal muscle, in parallel with changes in gene expression of SREBP1c. RXRgamma or RXRalpha, together with LXRalpha, activate the SREBP1c promoter in vitro. Moreover, transgenic mice overexpressing RXRgamma specifically in skeletal muscle showed increased gene expression of SREBP1c with increased triglyceride content in their skeletal muscles. In contrast, transgenic mice overexpressing the dominant-negative form of RXRgamma showed decreased SREBP1c gene expression. The expression of Forkhead-O1 transcription factor (FOXO1), which can suppress the function of multiple nuclear receptors, is negatively correlated to that of SREBP1c in skeletal muscle during nutritional change. Moreover, transgenic mice overexpressing FOXO1 specifically in skeletal muscle exhibited decreased gene expression of both RXRgamma and SREBP1c. In addition, FOXO1 suppressed RXRalpha/LXRalpha-mediated SREBP1c promoter activity in vitro. These findings provide in vivo and in vitro evidence that RXR/LXR up-regulates SREBP1c gene expression and that FOXO1 antagonizes this effect of RXR/LXR in skeletal muscle.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Factores de Transcripción Forkhead/fisiología , Músculo Esquelético/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Receptor gamma X Retinoide/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Animales , Secuencia de Bases , Células Cultivadas , Ingestión de Alimentos/fisiología , Ayuno/metabolismo , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Humanos , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Especificidad de Órganos , Receptores Nucleares Huérfanos , Regiones Promotoras Genéticas , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
14.
Am J Physiol Endocrinol Metab ; 294(3): E600-6, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18089762

RESUMEN

Uncoupling protein 2 (UCP2) is a possible target molecule for energy dissipation. Many dietary fats, including safflower oil and lard, induce obesity in C57BL/6 mice, whereas fish oil does not. Fish oil increases UCP2 expression in hepatocytes and may enhance UCP2 activity by activating the UCP2 molecule or altering the lipid bilayer environment. To examine the role of liver UCP2 in obesity, we created transgenic mice that overexpressed human UCP2 in hepatocytes and examined whether UCP2 transgenic mice showed less obesity when fed a high-fat diet (safflower oil or lard). In addition, we examined whether fish oil had antiobesity effects in UCP2 knockout mice. UCP2 transgenic and wild-type mice fed a high-fat diet (safflower oil or lard) developed obesity to a similar degree. UCP2 knockout and wild-type mice fed fish oil had lower rates of obesity than mice fed safflower oil. Remarkably, safflower oil did not induce obesity in female UCP2 knockout mice, an unexpected phenotype for which we presently have no explanation. However, this unexpected effect was not observed in male UCP2 knockout mice or in UCP2 knockout mice fed a high-lard diet. These data indicate that liver UCP2 is not essential for fish oil-induced decreases in body fat.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Aceites de Pescado/uso terapéutico , Canales Iónicos/fisiología , Proteínas Mitocondriales/fisiología , Obesidad/terapia , Animales , Grasas de la Dieta/administración & dosificación , Femenino , Expresión Génica , Canales Iónicos/deficiencia , Canales Iónicos/genética , Hígado/química , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Obesidad/etiología , ARN Mensajero/análisis , Aceite de Cártamo/administración & dosificación , Caracteres Sexuales , Proteína Desacopladora 2
15.
Hepatology ; 46(6): 1779-90, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17935225

RESUMEN

UNLABELLED: Diets high in sucrose/fructose or fat can result in hepatic steatosis (fatty liver). We analyzed the effects of dietary fish oil on fatty liver induced by sucrose, safflower oil, and butter in ddY mice. In experiment I, mice were fed a high-starch diet [70 energy% (en%) starch] plus 20% (wt/wt) sucrose in the drinking water or fed a high-safflower oil diet (60 en%) for 11 weeks. As a control, mice were fed a high-starch diet with drinking water. Fish oil (10 en%) was either supplemented or not. Mice supplemented with sucrose or fed safflower oil showed a 1.7-fold or 2.2-fold increased liver triglyceride content, respectively, compared with that of control mice. Fish oil completely prevented sucrose-induced fatty liver, whereas it exacerbated safflower oil-induced fatty liver. Sucrose increased SREBP-1c and target gene messenger RNAs (mRNAs), and fish oil completely inhibited these increases. In experiment II, mice were fed a high-safflower oil or a high-butter diet, with or without fish oil supplementation. Fish oil exacerbated safflower oil-induced fatty liver but did not affect butter-induced fatty liver. Fish oil increased expression of peroxisome proliferator-activated receptor gamma (PPARgamma) and target CD36 mRNA in safflower oil-fed mice. These increases were not observed in sucrose-supplemented or butter-fed mice. CONCLUSION: The effects of dietary fish oil on fatty liver differ according to the cause of fatty liver; fish oil prevents sucrose-induced fatty liver but exacerbates safflower oil-induced fatty liver. The exacerbation of fatty liver may be due, at least in part, to increased expression of liver PPARgamma.


Asunto(s)
Hígado Graso/prevención & control , Aceites de Pescado/uso terapéutico , Sacarosa/efectos adversos , Edulcorantes/efectos adversos , Animales , Mantequilla/efectos adversos , Hígado Graso/etiología , Masculino , Ratones , Aceite de Cártamo/efectos adversos
16.
Cell Metab ; 6(1): 55-68, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17618856

RESUMEN

Adiponectin has been shown to stimulate fatty acid oxidation and enhance insulin sensitivity through the activation of AMP-activated protein kinase (AMPK) in the peripheral tissues. The effects of adiponectin in the central nervous system, however, are still poorly understood. Here, we show that adiponectin enhances AMPK activity in the arcuate hypothalamus (ARH) via its receptor AdipoR1 to stimulate food intake; this stimulation of food intake by adiponectin was attenuated by dominant-negative AMPK expression in the ARH. Moreover, adiponectin also decreased energy expenditure. Adiponectin-deficient mice showed decreased AMPK phosphorylation in the ARH, decreased food intake, and increased energy expenditure, exhibiting resistance to high-fat-diet-induced obesity. Serum and cerebrospinal fluid levels of adiponectin and expression of AdipoR1 in the ARH were increased during fasting and decreased after refeeding. We conclude that adiponectin stimulates food intake and decreases energy expenditure during fasting through its effects in the central nervous system.


Asunto(s)
Adiponectina/fisiología , Ingestión de Alimentos , Hipotálamo/enzimología , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP , Adenoviridae/genética , Adiponectina/líquido cefalorraquídeo , Adiponectina/genética , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético , Femenino , Hipotálamo/patología , Técnicas para Inmunoenzimas , Hibridación in Situ , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multienzimáticos/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Sondas ARN , Receptores de Adiponectina , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de Leptina
17.
J Biol Chem ; 282(29): 21176-86, 2007 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-17510058

RESUMEN

The mammalian target of rapamycin (mTOR) is regulated by growth factors to promote protein synthesis. In mammalian skeletal muscle, the Forkhead-O1 transcription factor (FOXO1) promotes catabolism by activating ubiquitin-protein ligases. Using C2C12 mouse myoblasts that stably express inducible FOXO1-ER fusion proteins and transgenic mice that specifically overexpress constitutively active FOXO1 in skeletal muscle (FOXO(++/+)), we show that FOXO1 inhibits mTOR signaling and protein synthesis. Activation of constitutively active FOXO1 induced the expression of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) mRNA via binding to the promoter. This resulted in an increased total 4E-BP1 abundance and a reduced 4E-BP1 (Thr-37/46) phosphorylation. The reduction in 4E-BP1 phosphorylation was associated with a reduction in the abundance of Raptor and mTOR proteins, Raptor-associated mTOR, reduced phosphorylation of the downstream protein p70S6 kinase, and attenuated incorporation of [(14)C]phenylalanine into protein. The FOXO(++/+) mice, characterized by severe skeletal muscle atrophy, displayed similar patterns of mRNA expression and protein abundance to those observed in the constitutively active FOXO1 C2C12 myotubes. These data suggest that FOXO1 may be an important therapeutic target for human diseases where anabolism is impaired.


Asunto(s)
Proteínas Portadoras/química , Factores de Transcripción Forkhead/fisiología , Regulación de la Expresión Génica , Músculo Esquelético/metabolismo , Fosfoproteínas/química , Proteínas Quinasas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Sitios de Unión , Proteínas de Ciclo Celular , Factores Eucarióticos de Iniciación , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/química , Ratones , Ratones Transgénicos , Modelos Biológicos , Fenotipo , Fosforilación , Unión Proteica , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR
18.
FEBS Lett ; 581(1): 91-6, 2007 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-17182038

RESUMEN

Paternally expressed 1 (Peg1)/mesoderm specific transcript (Mest) is an imprinted gene, which is only transcribed from the paternal (father's) allele. In some human cancer tissues, an alternatively spliced variant of PEG1/MEST mRNA using a different promoter of a distinct first exon is expressed from both paternal and maternal alleles. We previously reported that Peg1/Mest expression was markedly up-regulated in obese adipose tissue in mice. Moreover, transgenic overexpression of Peg1/Mest in the adipose tissue resulted in the enlargement of adipocytes in size. Given the potential pathophysiologic relevance in obesity, we examined the nature of increased expression of Peg1/Mest in obese adipose tissue. In obese adipose tissue, expression of Peg1/Mest was increased, but not that of other imprinted genes tested. The transcription rate of Peg1/Mest was increased in obese adipose tissue. We found at least four isoforms of mouse Peg1/Mest generated by use of the alternative first exons. We also demonstrated that the abundantly expressed Peg1/Mest in obese adipose tissue retained monoallelic expression. This is the first report of monoallelic induction of Peg1/Mest in adult tissues.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Alelos , Regulación de la Expresión Génica , Impresión Genómica , Obesidad/metabolismo , Proteínas/metabolismo , Adipocitos/patología , Tejido Adiposo/patología , Empalme Alternativo , Animales , Línea Celular , Tamaño de la Célula , Exones , Humanos , Ratones , Ratones Obesos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Obesidad/genética , Obesidad/patología , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Proteínas/genética , Transgenes
19.
Nihon Rinsho ; 64(11): 2083-8, 2006 Nov.
Artículo en Japonés | MEDLINE | ID: mdl-17087300

RESUMEN

Major risk factors for cardiovascular diseases (myocardial infarction, angina pectoris, brain infarction) are age, male, smoking, high LDL-cholesterol, low HDL-cholesterol, high blood pressure, and diabetes mellitus. In Japanese population at large, healthy life-style to prevent cardiovascular diseases are; quit smoking, walking faster, saturated fat intake ranging 4.5-7 en%, lesser intake of trans fatty acids, cholesterol intake less 750 mg/day (male) and 600 mg/day (female), eat fish everyday, eat more folic acid, B6, and B12, eat grain, eat soybean products. However, it is not known whether this recommendation is also applied to NIDDM to prevent cardiovascular diseases. Based on reported evidences, to prevent cardiovascular diseases, NIDDM should quit smoking, eat fish everyday, and increase physical activity.


Asunto(s)
Arteriosclerosis/etiología , Arteriosclerosis/prevención & control , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/prevención & control , Diabetes Mellitus Tipo 2/complicaciones , Estilo de Vida , Dieta , Grasas de la Dieta/administración & dosificación , Medicina Basada en la Evidencia , Ejercicio Físico/fisiología , Femenino , Productos Pesqueros , Humanos , Masculino , Factores de Riesgo , Cese del Hábito de Fumar
20.
Am J Pathol ; 169(4): 1129-39, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17003473

RESUMEN

Peroxisome proliferator-activated receptor-gamma co-activator-1alpha (PGC-1alpha) is a key nuclear receptor co-activator for mitochondrial biogenesis. Here we report that overexpression of PGC-1alpha in skeletal muscles increased mitochondrial number and caused atrophy of skeletal muscle, especially type 2B fiber-rich muscles (gastrocnemius, quadriceps, and plantaris). Muscle atrophy became evident at 25 weeks of age, and a portion of the muscle was replaced by adipocytes. Mice showed increased energy expenditure and reduced body weight; thyroid hormone levels were normal. Mitochondria exhibited normal respiratory chain activity per mitochondrion; however, mitochondrial respiration was not inhibited by an ATP synthase inhibitor, oligomycin, clearly indicating that oxidative phosphorylation was uncoupled. Accordingly, ATP content in gastrocnemius was markedly reduced. A similar phenotype is observed in Luft's disease, a mitochondrial disorder that involves increased uncoupling of respiration and muscle atrophy. Our results indicate that overexpression of PGC-1alpha in skeletal muscle increases not only mitochondrial biogenesis but also uncoupling of respiration, resulting in muscle atrophy.


Asunto(s)
Adenosina Trifosfato/deficiencia , Atrofia Muscular/genética , Transactivadores/genética , Activación Transcripcional , Adipocitos/patología , Animales , Peso Corporal/genética , Proliferación Celular , Respiración de la Célula , Femenino , Masculino , Ratones , Ratones Transgénicos , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/ultraestructura , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Atrofia Muscular/patología , Fosforilación Oxidativa , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA