Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 12(10): 1069-76, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21836635

RESUMEN

The nicotinamide adenine dinucleotide (NAD(+))-dependent deacetylase SIRT1 is a major metabolic regulator activated by energy stresses such as fasting or calorie restriction. SIRT1 activation during fasting not only relies on the increase in the NAD(+)/NADH ratio caused by energy deprivation but also involves an upregulation of SIRT1 mRNA and protein levels in various metabolic tissues. We demonstrate that SIRT1 expression is controlled systemically by the activation of the cyclic AMP response-element-binding protein upon low nutrient availability. Conversely, in the absence of energetic stress, the carbohydrate response-element-binding protein represses the expression of SIRT1. Altogether, these results demonstrate that SIRT1 expression is tightly controlled at the transcriptional level by nutrient availability and further underscore that SIRT1 is a crucial metabolic checkpoint connecting the energetic status with transcriptional programmes.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Sirtuina 1/genética , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Células CHO , Línea Celular Tumoral , Cricetinae , Ayuno , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Sirtuina 1/metabolismo , Activación Transcripcional
2.
J Biol Chem ; 286(30): 26913-20, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21632533

RESUMEN

We evaluated the metabolic impact of farnesoid X receptor (FXR) activation by administering a synthetic FXR agonist (GW4064) to mice in which obesity was induced by a high fat diet. Administration of GW4064 accentuated body weight gain and glucose intolerance induced by the high fat diet and led to a pronounced worsening of the changes in liver and adipose tissue. Mechanistically, treatment with GW4064 decreased bile acid (BA) biosynthesis, BA pool size, and energy expenditure, whereas reconstitution of the BA pool in these GW4064-treated animals by BA administration dose-dependently reverted the metabolic abnormalities. Our data therefore suggest that activation of FXR with synthetic agonists is not useful for long term management of the metabolic syndrome, as it reduces the BA pool size and subsequently decreases energy expenditure, translating as weight gain and insulin resistance. In contrast, expansion of the BA pool size, which can be achieved by BA administration, could be an interesting strategy to manage the metabolic syndrome.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Diabetes Mellitus/inducido químicamente , Grasas de la Dieta/efectos adversos , Metabolismo Energético/efectos de los fármacos , Isoxazoles/efectos adversos , Obesidad/inducido químicamente , Receptores Citoplasmáticos y Nucleares/agonistas , Células 3T3-L1 , Animales , Peso Corporal/efectos de los fármacos , Diabetes Mellitus/metabolismo , Grasas de la Dieta/farmacología , Isoxazoles/farmacología , Síndrome Metabólico/tratamiento farmacológico , Ratones , Obesidad/metabolismo
3.
Nature ; 439(7075): 484-9, 2006 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-16400329

RESUMEN

While bile acids (BAs) have long been known to be essential in dietary lipid absorption and cholesterol catabolism, in recent years an important role for BAs as signalling molecules has emerged. BAs activate mitogen-activated protein kinase pathways, are ligands for the G-protein-coupled receptor (GPCR) TGR5 and activate nuclear hormone receptors such as farnesoid X receptor alpha (FXR-alpha; NR1H4). FXR-alpha regulates the enterohepatic recycling and biosynthesis of BAs by controlling the expression of genes such as the short heterodimer partner (SHP; NR0B2) that inhibits the activity of other nuclear receptors. The FXR-alpha-mediated SHP induction also underlies the downregulation of the hepatic fatty acid and triglyceride biosynthesis and very-low-density lipoprotein production mediated by sterol-regulatory-element-binding protein 1c. This indicates that BAs might be able to function beyond the control of BA homeostasis as general metabolic integrators. Here we show that the administration of BAs to mice increases energy expenditure in brown adipose tissue, preventing obesity and resistance to insulin. This novel metabolic effect of BAs is critically dependent on induction of the cyclic-AMP-dependent thyroid hormone activating enzyme type 2 iodothyronine deiodinase (D2) because it is lost in D2-/- mice. Treatment of brown adipocytes and human skeletal myocytes with BA increases D2 activity and oxygen consumption. These effects are independent of FXR-alpha, and instead are mediated by increased cAMP production that stems from the binding of BAs with the G-protein-coupled receptor TGR5. In both rodents and humans, the most thermogenically important tissues are specifically targeted by this mechanism because they coexpress D2 and TGR5. The BA-TGR5-cAMP-D2 signalling pathway is therefore a crucial mechanism for fine-tuning energy homeostasis that can be targeted to improve metabolic control.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Metabolismo Energético/efectos de los fármacos , Hormonas Tiroideas/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/enzimología , Tejido Adiposo Pardo/metabolismo , Adiposidad/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Dióxido de Carbono/metabolismo , Ácido Cólico/farmacología , AMP Cíclico/biosíntesis , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Eliminación de Gen , Homeostasis/efectos de los fármacos , Humanos , Yoduro Peroxidasa/deficiencia , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Musculares/efectos de los fármacos , Células Musculares/enzimología , Células Musculares/metabolismo , Músculo Esquelético/citología , Consumo de Oxígeno/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Yodotironina Deyodinasa Tipo II
4.
Proc Natl Acad Sci U S A ; 106(43): 18357-61, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19826084

RESUMEN

Genome-wide studies reveal that transcription by RNA polymerase II (Pol II) is dynamically regulated. To obtain a comprehensive view of a single transcription cycle, we switched on transcription of five long human genes (>100 kbp) with tumor necrosis factor-alpha (TNFalpha) and monitored (using microarrays, RNA fluorescence in situ hybridization, and chromatin immunoprecipitation) the appearance of nascent RNA, changes in binding of Pol II and two insulators (the cohesin subunit RAD21 and the CCCTC-binding factor CTCF), and modifications of histone H3. Activation triggers a wave of transcription that sweeps along the genes at approximately 3.1 kbp/min; splicing occurs cotranscriptionally, a major checkpoint acts several kilobases downstream of the transcription start site to regulate polymerase transit, and Pol II tends to stall at cohesin/CTCF binding sites.


Asunto(s)
Transcripción Genética , Sitios de Unión , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN , Humanos , Hibridación in Situ , Intrones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , ARN Polimerasa II/metabolismo , Empalme del ARN , ARN Mensajero/biosíntesis , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Activación Transcripcional , Factor de Necrosis Tumoral alfa/genética , Cohesinas
5.
FASEB J ; 24(11): 4613-26, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20647546

RESUMEN

Glucose-evoked mitochondrial signals augment ATP synthesis in the pancreatic ß cell. This activation of energy metabolism increases the cytosolic ATP/ADP ratio, which stimulates plasma membrane electrical activity and insulin granule exocytosis. We have recently demonstrated that matrix pH increases during nutrient stimulation of the pancreatic ß cell. Here, we have tested whether mitochondrial matrix pH controls oxidative phosphorylation and metabolism-secretion coupling in the rat ß-cell line INS-1E. Acidification of the mitochondrial matrix pH by nigericin blunted nutrient-dependent respiratory and ATP responses (continuously monitored in intact cells). Using electrophysiology and single cell imaging, we find that the associated defects in energy metabolism suppress glucose-stimulated plasma membrane electrical activity and cytosolic calcium transients. The same parameters were unaffected after direct stimulation of electrical activity with tolbutamide, which bypasses mitochondrial function. Furthermore, lowered matrix pH strongly inhibited sustained, but not first-phase, insulin secretion. Our results demonstrate that the matrix pH exerts a control function on oxidative phosphorylation in intact cells and that this mode of regulation is of physiological relevance for the generation of downstream signals leading to insulin granule exocytosis. We propose that matrix pH serves a novel signaling role in sustained cell activation.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Fosforilación Oxidativa , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Glucosa/farmacología , Células HeLa , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Células Secretoras de Insulina/efectos de los fármacos , Ionóforos/farmacología , Nigericina/farmacología , Consumo de Oxígeno/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos
6.
Am J Physiol Gastrointest Liver Physiol ; 298(2): G151-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19815629

RESUMEN

Rodents undergo gestational hepatomegaly to meet the increased metabolic demands on the maternal liver during pregnancy. This is an important physiological process, but the mechanisms and signals driving pregnancy-induced liver growth are not known. Here, we show that liver growth during pregnancy precedes maternal body weight gain, is proportional to fetal number, and is a result of hepatocyte hypertrophy associated with cell-cycle progression, polyploidy, and altered expression of cell-cycle regulators p53, Cyclin-D1, and p27. Because circulating reproductive hormones and bile acids are raised in normal pregnant women and can cause liver growth in rodents, these compounds are candidates for the signal driving gestational liver enlargement in rodents. Administration of pregnancy levels of reproductive hormones was not sufficient to cause liver growth, but mouse pregnancy was associated with increased serum bile acid levels. It is known that the bile acid sensor Fxr is required for normal recovery from partial hepatectomy, and we demonstrate that Fxr(-/-) mice undergo gestational liver growth by adaptive hepatocyte hyperplasia. This is the first identification of any component that is required to maintain the normal mechanisms of gestational hepatomegaly and also implicates Fxr in a physiologically normal process that involves control of the hepatocyte cell cycle. Understanding pregnancy-induced hepatocyte hypertrophy in mice could suggest mechanisms for safely increasing functional liver capacity in women during increased metabolic demand.


Asunto(s)
Regeneración Hepática/fisiología , Hígado/fisiología , Preñez/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Colatos/metabolismo , Colatos/farmacología , Receptor de Androstano Constitutivo , Ciclina D1/genética , Ciclina D1/metabolismo , Metabolismo Energético/fisiología , Femenino , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/fisiología , Hígado/citología , Hígado/crecimiento & desarrollo , Ratones , Ratones Mutantes , Ovariectomía , Poliploidía , Embarazo , Fase S/efectos de los fármacos , Fase S/fisiología
7.
Arterioscler Thromb Vasc Biol ; 26(12): 2652-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17008592

RESUMEN

OBJECTIVE: Tumor necrosis factor (TNF)-alpha initiates numerous changes in endothelial cell (EC) gene expression that contributes to the pathology of various diseases including inflammation. We hypothesized that TNF-alpha-mediated gene induction involves multiple signaling pathways, and that inhibition of one or more of these pathways may selectively target subsets of TNF-alpha-responsive genes and functions. METHODS AND RESULTS: Human umbilical vein endothelial cells (ECs) were preincubated with inhibitors of PI3 kinase (LY294002), histone deacetylases (HDAC) (trichostatin A [TSA]), de novo protein synthesis (CHX), proteasome (MG-132), and GATA factors (K-11430) before exposure to TNF-alpha at 4 hours and analyzed by microarray. TNF-alpha-mediated induction of vascular cell adhesion molecule-1 (VCAM-1) was attenuated by all of these inhibitors, whereas in contrast, stimulation of intercellular adhesion molecule-1 (ICAM-1) was blocked by MG-132 alone. Moreover TSA blocked TNF-alpha-mediated induction of monocyte adhesion both in vitro and in vivo through the suppression of VCAM-1. Further analysis demonstrated that HDAC3 plays a significant role in the regulation of TNF-alpha-mediated VCAM-1 expression. CONCLUSIONS: TNF-alpha activates ECs via multiple signaling pathways, and these pathways may be selectively targeted to modulate EC function. Moreover, TSA treatment reduced monocyte adhesion via VCAM-1 suppression in vitro and in vivo, suggesting that TSA might be useful for the attenuation of the inflammatory response in EC.


Asunto(s)
Endotelio Vascular/metabolismo , Inhibidores de Histona Desacetilasas , Monocitos/citología , Monocitos/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Azepinas/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Cromonas/farmacología , Selectina E/genética , Selectina E/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histona Desacetilasas/fisiología , Humanos , Ácidos Hidroxámicos/farmacología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Leupeptinas/farmacología , Ratones , Ratones Transgénicos , Monocitos/efectos de los fármacos , Morfolinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Activación Transcripcional , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/genética
8.
Physiol Genomics ; 15(3): 199-208, 2003 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-12966135

RESUMEN

The induction of specific tolerance would be the ultimate achievement in transplant immunology, but the precise mechanisms of immunologic tolerance remain largely unknown. Here, we investigated global gene expression analysis in tolerizing murine cardiac allografts by means of oligonucleotide microarrays. Tolerance induction was achieved in cardiac allografts from BALB/c to C57BL/6 mice by daily intraperitoneal injection of anti-CD80 and anti-CD86 monoclonal antibodies (mAbs). Comparative analysis revealed 64 genes to be induced more extensively in the tolerizing than in the syngeneic isografts, and 16 genes than in the rejecting allografts. Two genes were specifically upregulated in the tolerizing allografts. In the tolerizing allografts there were induced marked expressions of a number of genes for pro-inflammatory factors, including interferon-gamma-inducible cytokines and chemokines, as well as apoptosis-related genes, which were also upregulated in the rejecting allografts. Moreover, these gene expression patterns continued to be upregulated more than 70 days posttransplant. These results provide evidence that immunologic tolerance can be induced and maintained in the presence of prominent pro-inflammatory gene expression in vivo.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Antígeno B7-1/inmunología , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Trasplante de Corazón/métodos , Tolerancia Inmunológica/genética , Glicoproteínas de Membrana/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD/fisiología , Antígeno B7-1/fisiología , Antígeno B7-2 , Análisis por Conglomerados , Perfilación de la Expresión Génica/estadística & datos numéricos , Regulación de la Expresión Génica/inmunología , Regulación de la Expresión Génica/fisiología , Rechazo de Injerto/genética , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Tolerancia Inmunológica/inmunología , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Miocardio/patología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/estadística & datos numéricos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
9.
J Atheroscler Thromb ; 11(2): 62-72, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15153665

RESUMEN

In addition to a lipid-lowering effect, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) have an effect on the expression levels of many genes. In order to elucidate the range of this effect as comprehensively as possible, we investigated the changes in gene expression profiles brought about by atorvastatin or pitavastatin in cultured human umbilical vein endothelial cells (HUVEC), cultured human coronary artery smooth muscle cells (HCASMC) and cultured human hepatocarcinoma Hep G2 cells by means of DNA microarrays. Among the 6146 genes in the array, statins affected the expression levels of genes involved in coagulation, vascular constriction and cell growth in a cell-type specific manner. In HUVEC, they induced integrin beta4 and thrombomodulin profoundly, and profoundly suppressed pentraxin 3 both at 8 and 24 hours. In HCASMC, the statins induced thrombomodulin and urokinase inhibitor, and potently suppressed the cysteine-rich angiogenic inducer 61 and cyclin B. Many genes related to the cell cycle and/or growth were also regulated in HUVEC and HCASMC by the statins. These results indicate that many aspects of the pleiotropic effect can be mediated by transcriptional control by statins. Genes newly identified by this study may be useful in statin therapy.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Perfilación de la Expresión Génica , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , ARN/genética , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Atheroscler Thromb ; 9(4): 178-83, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12226549

RESUMEN

Large-scale clinical trials have demonstrated significant reductions in cardiovascular events following statin therapy. The observed benefit of statin therapy, however, may be greater in these trials than is to be expected from lowering lipid levels alone. In order to clarify the mechanism by which statins prevent cardiovascular events in vascular wall cells, we investigated the changes in gene expression profiles after incubation with atorvastatin or pitavastatin in cultured human umbilical vein endothelial cells using DNA microarrays. Statins affected the expression levels of genes involved in inflammation, coagulation, and vascular constriction. The mRNA levels for interleukin-8 (IL-8) and monocyte chemoattractant protein-1 (MCP-1) decreased after statin treatment. Statins reduced mRNA levels of plasminogen activator inhibitor-1 (PAI-1) and increased the mRNA levels of thrombomodulin. Statins reduced the mRNA levels of endothelin-1 and increased the mRNA levels of nitric oxide synthase-3 (eNOS). These results show that, statins are clinically effective because of their ability to change the gene expression profile of endothelial cells thereby preventing vascular events.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Pirroles/farmacología , Quinolinas/farmacología , ARN Mensajero/metabolismo , Atorvastatina , Coagulación Sanguínea/genética , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Inflamación/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Receptores de LDL/genética , Vasoconstricción/genética
11.
Cell Metab ; 20(4): 603-13, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25176150

RESUMEN

Reverse cholesterol transport (RCT) is an antiatherogenic process in which excessive cholesterol from peripheral tissues is transported to the liver and finally excreted from the body via the bile. The nuclear receptor liver receptor homolog 1 (LRH-1) drives expression of genes regulating RCT, and its activity can be modified by different posttranslational modifications. Here, we show that atherosclerosis-prone mice carrying a mutation that abolishes SUMOylation of LRH-1 on K289R develop less aortic plaques than control littermates when exposed to a high-cholesterol diet. The mechanism underlying this atheroprotection involves an increase in RCT and its associated hepatic genes and is secondary to a compromised interaction of LRH-1 K289R with the corepressor prospero homeobox protein 1 (PROX1). Our study reveals that the SUMOylation status of a single nuclear receptor lysine residue can impact the development of a complex metabolic disease such as atherosclerosis.


Asunto(s)
Colesterol/metabolismo , Proteínas de Homeodominio/metabolismo , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Transporte Biológico , Células Cultivadas , Células HEK293 , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Humanos , Hígado/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores Citoplasmáticos y Nucleares/genética , Receptores de LDL/deficiencia , Receptores de LDL/genética , Alineación de Secuencia , Sumoilación , Activación Transcripcional , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
12.
Mol Metab ; 2(3): 314-23, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24049743

RESUMEN

Altered adipose tissue formation is a well-known effectors of obesity and T2D. Here, we describe the role of Lrh1 and its co-repressor Shp in the control of adipocyte formation. Expression of Lrh1 in the pre-adipocyte containing SVF is induced in obese mice models and humans while Shp expression is reduced. We demonstrate, that Lrh1 is an inhibitor of adipogenesis while Shp acts functions as an activator through repression of Lrh1 activity. This regulation is at least in part modulated by estradiol conversion through the regulation of Cyp19a1 gene expression. In vivo, loss of Lrh1 leads to induced adipogenesis, while loss of Shp causes uncontrolled activation of Lrh1 and reduced adipogenesis. As Shp expression has been linked to the development of obesity and metabolic disorders, it is possible that alterations of the Shp/Lrh1 network lead to changes in adipocyte formation, which might contribute to the development of obesity associated T2D.

13.
J Clin Invest ; 122(8): 2817-26, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22772466

RESUMEN

Liver receptor homolog 1 (LRH-1), an established regulator of cholesterol and bile acid homeostasis, has recently emerged as a potential drug target for liver disease. Although LRH-1 activation may protect the liver against diet-induced steatosis and insulin resistance, little is known about how LRH-1 controls hepatic glucose and fatty acid metabolism under physiological conditions. We therefore assessed the role of LRH-1 in hepatic intermediary metabolism. In mice with conditional deletion of Lrh1 in liver, analysis of hepatic glucose fluxes revealed reduced glucokinase (GCK) and glycogen synthase fluxes as compared with those of wild-type littermates. These changes were attributed to direct transcriptional regulation of Gck by LRH-1. Impaired glucokinase-mediated glucose phosphorylation in LRH-1-deficient livers was also associated with reduced glycogen synthesis, glycolysis, and de novo lipogenesis in response to acute and prolonged glucose exposure. Accordingly, hepatic carbohydrate response element-binding protein activity was reduced in these animals. Cumulatively, these data identify LRH-1 as a key regulatory component of the hepatic glucose-sensing system required for proper integration of postprandial glucose and lipid metabolism.


Asunto(s)
Glucosa/metabolismo , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Glucoquinasa/genética , Glucoquinasa/metabolismo , Glucógeno Sintasa/metabolismo , Glucólisis , Metabolismo de los Lípidos , Glucógeno Hepático/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Proteínas Nucleares/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/metabolismo , Transcripción Genética
14.
PLoS One ; 7(8): e38286, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22952571

RESUMEN

BACKGROUND: Besides well-established roles of bile acids (BA) in dietary lipid absorption and cholesterol homeostasis, it has recently become clear that BA is also a biological signaling molecule. We have shown that strategies aimed at activating TGR5 by increasing the BA pool size with BA administration may constitute a significant therapeutic advance to combat the metabolic syndrome and suggest that such strategies are worth testing in a clinical setting. Bile acid binding resin (BABR) is known not only to reduce serum cholesterol levels but also to improve glucose tolerance and insulin resistance in animal models and humans. However, the mechanisms by which BABR affects glucose homeostasis have not been established. We investigated how BABR affects glycemic control in diet-induced obesity models. METHODS AND FINDINGS: We evaluated the metabolic effect of BABR by administrating colestimide to animal models for the metabolic syndrome. Administration of BABR increased energy expenditure, translating into significant weight reduction and insulin sensitization. The metabolic effects of BABR coincide with activation of cholesterol and BA synthesis in liver and thermogenesis in brown adipose tissue. Interestingly, these effects of BABR occur despite normal food intake and triglyceride absorption. Administration of BABR and BA had similar effects on BA composition and thermogenesis, suggesting that they both are mediated via TGR5 activation. CONCLUSION: Our data hence suggest that BABR could be useful for the management of the impaired glucose tolerance of the metabolic syndrome, since they not only lower cholesterol levels, but also reduce obesity and improve insulin resistance.


Asunto(s)
Ácidos y Sales Biliares/química , Colesterol/metabolismo , Resina de Colestiramina/química , Epiclorhidrina/química , Imidazoles/química , Receptores Acoplados a Proteínas G/metabolismo , Resinas Sintéticas/química , Animales , Colesterol/sangre , Metabolismo Energético , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Masculino , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Unión Proteica , Transducción de Señal
15.
Cell Metab ; 13(5): 601-11, 2011 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-21531342

RESUMEN

Mitochondrial Ca(2+) signals have been proposed to accelerate oxidative metabolism and ATP production to match Ca(2+)-activated energy-consuming processes. Efforts to understand the signaling role of mitochondrial Ca(2+) have been hampered by the inability to manipulate matrix Ca(2+) without directly altering cytosolic Ca(2+). We were able to selectively buffer mitochondrial Ca(2+) rises by targeting the Ca(2+)-binding protein S100G to the matrix. We find that matrix Ca(2+) controls signal-dependent NAD(P)H formation, respiration, and ATP changes in intact cells. Furthermore, we demonstrate that matrix Ca(2+) increases are necessary for the amplification of sustained glucose-dependent insulin secretion in ß cells. Through the regulation of NAD(P)H in adrenal glomerulosa cells, matrix Ca(2+) also acts as a positive signal in reductive biosynthesis, which stimulates aldosterone secretion. Our dissection of cytosolic and mitochondrial Ca(2+) signals reveals the physiological importance of matrix Ca(2+) in energy metabolism required for signal-dependent hormone secretion.


Asunto(s)
Aldosterona/metabolismo , Calcio/metabolismo , Citosol/metabolismo , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Animales , Calbindinas , Células Cultivadas , Glucosa/metabolismo , Técnicas para Inmunoenzimas , Insulina/metabolismo , Potencial de la Membrana Mitocondrial , NADP/metabolismo , Consumo de Oxígeno , Ratas , Zona Glomerular/citología , Zona Glomerular/metabolismo
16.
Cell Metab ; 11(3): 213-9, 2010 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-20197054

RESUMEN

During fasting and after exercise, skeletal muscle efficiently switches from carbohydrate to lipid as the main energy source to preserve glycogen stores and blood glucose levels for glucose-dependent tissues. Skeletal muscle cells sense this limitation in glucose availability and transform this information into transcriptional and metabolic adaptations. Here we demonstrate that AMPK acts as the prime initial sensor that translates this information into SIRT1-dependent deacetylation of the transcriptional regulators PGC-1alpha and FOXO1, culminating in the transcriptional modulation of mitochondrial and lipid utilization genes. Deficient AMPK activity compromises SIRT1-dependent responses to exercise and fasting, resulting in impaired PGC-1alpha deacetylation and blunted induction of mitochondrial gene expression. Thus, we conclude that AMPK acts as the primordial trigger for fasting- and exercise-induced adaptations in skeletal muscle and that activation of SIRT1 and its downstream signaling pathways are improperly triggered in AMPK-deficient states.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Ayuno/metabolismo , Factores de Transcripción Forkhead/metabolismo , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Sirtuina 1/metabolismo , Transactivadores/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Células Cultivadas , Metabolismo Energético , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Genes Mitocondriales/genética , Glucosa/metabolismo , Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transducción de Señal , Sirtuina 1/genética , Transactivadores/genética , Factores de Transcripción , Regulación hacia Arriba/genética
17.
Cell Metab ; 10(3): 167-77, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19723493

RESUMEN

TGR5 is a G protein-coupled receptor expressed in brown adipose tissue and muscle, where its activation by bile acids triggers an increase in energy expenditure and attenuates diet-induced obesity. Using a combination of pharmacological and genetic gain- and loss-of-function studies in vivo, we show here that TGR5 signaling induces intestinal glucagon-like peptide-1 (GLP-1) release, leading to improved liver and pancreatic function and enhanced glucose tolerance in obese mice. In addition, we show that the induction of GLP-1 release in enteroendocrine cells by 6alpha-ethyl-23(S)-methyl-cholic acid (EMCA, INT-777), a specific TGR5 agonist, is linked to an increase of the intracellular ATP/ADP ratio and a subsequent rise in intracellular calcium mobilization. Altogether, these data show that the TGR5 signaling pathway is critical in regulating intestinal GLP-1 secretion in vivo, and suggest that pharmacological targeting of TGR5 may constitute a promising incretin-based strategy for the treatment of diabesity and associated metabolic disorders.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Ácidos Cólicos/farmacología , Glucosa/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Células CHO , Calcio/metabolismo , Línea Celular , Ácidos Cólicos/química , Cricetinae , Cricetulus , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Homeostasis , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Fosforilación Oxidativa , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/deficiencia , Transducción de Señal
18.
Cell Metab ; 9(4): 339-49, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19356715

RESUMEN

We show here high levels of expression and secretion of the chemokine CXC ligand 5 (CXCL5) in the macrophage fraction of white adipose tissue (WAT). Moreover, we find that CXCL5 is dramatically increased in serum of human obese compared to lean subjects. Conversely, CXCL5 concentration is decreased in obese subjects after a weight reduction program, or in obese non-insulin-resistant, compared to insulin-resistant, subjects. Most importantly we demonstrate that treatment with recombinant CXCL5 blocks insulin-stimulated glucose uptake in muscle in mice. CXCL5 blocks insulin signaling by activating the Jak2/STAT5/SOCS2 pathway. Finally, by treating obese, insulin-resistant mice with either anti-CXCL5 neutralizing antibodies or antagonists of CXCR2, which is the CXCL5 receptor, we demonstrate that CXCL5 mediates insulin resistance. Furthermore CXCR2-/- mice are protected against obesity-induced insulin resistance. Taken together, these results show that secretion of CXCL5 by WAT resident macrophages represents a link between obesity, inflammation, and insulin resistance.


Asunto(s)
Tejido Adiposo/metabolismo , Quimiocina CXCL5/metabolismo , Resistencia a la Insulina , Obesidad/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Animales , Quimiocina CXCL5/deficiencia , Quimiocina CXCL5/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología , Factor de Necrosis Tumoral alfa/farmacología
19.
Genes Dev ; 22(14): 1871-6, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18628394

RESUMEN

Female fertility requires normal ovarian follicular growth and ovulation. The nuclear receptor liver receptor homolog 1 has been implicated in processes as diverse as bile acid metabolism, steroidogenesis, and cell proliferation. In the ovary, Lrh1 is expressed exclusively in granulosa and luteal cells. Using somatic targeted mutagenesis, we show that mice lacking Lrh1 in granulosa cells are sterile, due to anovulation. The preovulatory stimulus fails to elicit cumulus expansion, luteinization, and follicular rupture in these mice. Multiple defects, including severely reduced transactivation of the Lrh1 target gene, nitric oxide synthase 3, leads to increased intrafollicular estradiol levels in the absence of Lrh1. This further causes dysfunction of prostaglandin and hyaluronic acid cascades and interrupts cumulus expansion. Lack of Lrh1 also interferes with progesterone synthesis because of failure of normal expression of the Lrh1 targets, steroidogenic acute regulatory protein and cytochrome P450 side-chain cleavage. In addition, expression of extracellular matrix proteases essential for ovulation is compromised. These results demonstrate that Lrh1 is a regulator of multiple mechanisms essential for maturation of ovarian follicles and for ovulation. Lrh1 is therefore a key modulator of female fertility and a potential target for contraception.


Asunto(s)
Folículo Ovárico/fisiología , Ovulación/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Inmunoprecipitación de Cromatina , Sistema Enzimático del Citocromo P-450/metabolismo , Estradiol/metabolismo , Femenino , Fertilidad/fisiología , Células de la Granulosa/metabolismo , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Folículo Ovárico/citología , Fenotipo , Fosfoproteínas/metabolismo , Progesterona/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Radioinmunoensayo
20.
Cell Metab ; 8(5): 347-58, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19046567

RESUMEN

The NAD(+)-dependent deacetylase SIRT1 controls metabolic processes in response to low nutrient availability. We report the metabolic phenotype of mice treated with SRT1720, a specific and potent synthetic activator of SIRT1 that is devoid of direct action on AMPK. SRT1720 administration robustly enhances endurance running performance and strongly protects from diet-induced obesity and insulin resistance by enhancing oxidative metabolism in skeletal muscle, liver, and brown adipose tissue. These metabolic effects of SRT1720 are mediated by the induction of a genetic network controlling fatty acid oxidation through a multifaceted mechanism that involves the direct deacetylation of PGC-1alpha, FOXO1, and p53 and the indirect stimulation of AMPK signaling through a global metabolic adaptation mimicking low energy levels. Combined with our previous work on resveratrol, the current study further validates SIRT1 as a target for the treatment of metabolic disorders and characterizes the mechanisms underlying the therapeutic potential of SIRT1 activation.


Asunto(s)
Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Enfermedades Metabólicas/metabolismo , Obesidad/metabolismo , Sirtuinas/metabolismo , Acetilación , Animales , Dieta , Grasas de la Dieta/administración & dosificación , Metabolismo Energético , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Resistencia a la Insulina/fisiología , Masculino , Enfermedades Metabólicas/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Obesidad/tratamiento farmacológico , Oxidación-Reducción , Condicionamiento Físico Animal , Sirtuina 1 , Sirtuinas/agonistas , Sirtuinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA