Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Biochem J ; 477(5): 1021-1031, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32096546

RESUMEN

Hepatic glucose metabolism signaling downstream of insulin can diverge to multiple pathways including AKT. Genetic studies suggest that AKT is necessary for insulin to suppress gluconeogenesis. To specifically address the role of AKT2, the dominant liver isoform of AKT in the regulation of gluconeogenesis genes, we generated hepatocytes lacking AKT2 (Akt2-/-). We found that, in the absence of insulin signal, AKT2 is required for maintaining the basal level expression of phosphoenolpyruvate carboxyl kinase (PEPCK) and to a lesser extent G6Pase, two key rate-limiting enzymes for gluconeogenesis that support glucose excursion due to pyruvate loading. We further showed that this function of AKT2 is mediated by the phosphorylation of cyclic AMP response element binding (CREB). Phosphorylation of CREB by AKT2 is needed for CREB to induce the expression of PEPCK and likely represents a priming event for unstimulated cells to poise to receive glucagon and other signals. The inhibition of gluconeogenesis by insulin is also dependent on the reduced FOXO1 transcriptional activity at the promoter of PEPCK. When insulin signal is absent, this activity appears to be inhibited by AKT2 in manner that is independent of its phosphorylation by AKT. Together, this action of AKT2 on FOXO1 and CREB to maintain basal gluconeogenesis activity may provide fine-tuning for insulin and glucocorticoid/glucagon to regulate gluconeogenesis in a timely manner to meet metabolic needs.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Glucosa-6-Fosfatasa/biosíntesis , Fosfoenolpiruvato Carboxiquinasa (ATP)/biosíntesis , Proteínas Proto-Oncogénicas c-akt/deficiencia , Animales , Células Cultivadas , Glucosa-6-Fosfatasa/genética , Hepatocitos/enzimología , Ratones , Ratones Noqueados , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Proteínas Proto-Oncogénicas c-akt/genética
2.
J Biol Chem ; 294(48): 18017-18028, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31645433

RESUMEN

Gluconeogenesis (GNG) is de novo production of glucose from endogenous carbon sources. Although it is a commonly studied pathway, particularly in disease, there is a lack of consensus about substrate preference. Moreover, primary hepatocytes are the current gold standard for in vitro liver studies, but no direct comparison of substrate preference at physiological fasting concentrations has been performed. We show that mouse primary hepatocytes prefer glycerol to pyruvate/lactate in glucose production assays and 13C isotope tracing studies at the high concentrations commonly used in the literature, as well as at more relevant fasting, physiological concentrations. In addition, when glycerol, pyruvate/lactate, and glutamine are all present, glycerol is responsible for over 75% of all glucose carbons labeled. We also found that glycerol can induce a rate-limiting enzyme of GNG, glucose-6-phosphatase. Lastly, we suggest that glycerol is a better substrate than pyruvate to test in vivo production of glucose in fasting mice. In conclusion, glycerol is the major carbon source for GNG in vitro and in vivo and should be compared with other substrates when studying GNG in the context of metabolic disease states.


Asunto(s)
Gluconeogénesis/efectos de los fármacos , Glucosa-6-Fosfatasa/biosíntesis , Glicerol/farmacología , Hepatocitos/metabolismo , Animales , Inducción Enzimática/efectos de los fármacos , Hepatocitos/citología , Ácido Láctico/metabolismo , Ratones , Ácido Pirúvico/metabolismo
3.
J Pharmacol Sci ; 138(3): 161-166, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30316692

RESUMEN

As one kind of complications of pregnancy, gestational diabetes mellitus (GDM) can influence the health of maternal-child in clinical practice. The C57 BL/KsJdb/+(db/+) mice, genetic GDM model, and C57 BL/KsJ+/+ (wild-type) mice were purchased and classified into three groups: normal pregnancy (C57 BL/KsJ+/+), GDM (C57 BL/KsJdb/+), and GDM plus Mogroside IIIE (20.0 mg/kg) group. GDM symptoms (maternal body weight, serum glucose, and insulin levels), glucose and insulin tolerance, and reproductive outcome (body weight at birth and litter size of offspring) were investigated. The inflammatory factors such as IL-1ß, IL-6, and TNF-α in the serum and the pancreas were detected by ELISA and qRT-PCR, while the expression of pAMPK, AMPK, pHDAC4, HDAC4, and G6Pase in the livers were analyzed by Western Blot. Mogroside IIIE greatly improved glucose metabolism, insulin tolerance, and reproductive outcome of the GDM mice. Moreover, Mogroside IIIE treatment significantly decreased inflammatory factors expression and relieved GDM symptoms through enhanced AMPK activation, inhibited HDAC4 expression, and reduced production of G6Pase. The alleviation of GDM by Mogroside IIIE was mediated by elevated AMPK activation, which in turn inhibited HDAC4 phosphorylation, and eventually down-regulated G6Pase expression and activity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Diabetes Gestacional/tratamiento farmacológico , Glucósidos/farmacología , Glucósidos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Triterpenos/farmacología , Triterpenos/uso terapéutico , Proteínas Quinasas Activadas por AMP/biosíntesis , Animales , Glucemia/efectos de los fármacos , Femenino , Glucosa-6-Fosfatasa/biosíntesis , Histona Desacetilasas/biosíntesis , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Insulina/sangre , Tamaño de la Camada/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Transgénicos , Páncreas/metabolismo , Fosforilación/efectos de los fármacos , Embarazo
4.
Free Radic Biol Med ; 118: 98-107, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29496617

RESUMEN

Hepatic production and release of metabolites, nutrients and micronutrient transporters is tightly regulated at the level of gene expression. In this regard, transcription factor FOXO1 modulates the expression of genes such as G6PC and SELENOP, encoding the catalytic subunit of glucose 6-phosphatase and the plasma selenium transporter selenoprotein P, respectively. Here, we analyzed the role of cysteine residues in FOXO1 in controlling its activity with respect to regulation of G6PC and SELENOP in HepG2 human hepatoma cells. None of the seven FOXO1 cysteines affected FOXO1 binding to DNA or its basal subcellular distribution. Whereas overexpression of wildtype FOXO1 caused a strong induction of both G6PC and SELENOP promoter activities and mRNA levels, the induction was lowered by approx. 50% if cysteine-deficient FOXO1 was overexpressed instead. Only the most C-terminal of the seven FOXO1 cysteines, Cys612, was required and sufficient to ensure full FOXO1 transactivation activity. Coexpression of FOXO1 coregulators, CBP or PGC1α, had a strong synergistic effect in stimulating G6PC promoter activity and expression, fully relying on the presence of FOXO1 Cys612. Similarly, a synergistic effect of FOXO1 and CBP was observed for SELENOP. In contrast, stimulation of SELENOP by PGC1α was independent of FOXO1-Cys612, due to the close proximity of a hepatocyte nuclear factor-4α binding site to the FOXO1 binding site within the SELENOP promoter, as demonstrated using mutant SELENOP promoter constructs. In summary, full basal FOXO1 transactivation activity relies on Cys612, which mediates synergistic effects of coregulators, CBP or PGC1α, on FOXO1 transcriptional activity. The extent of Cys612 contribution depends on the promoter context of FOXO1 target genes.


Asunto(s)
Proteína Forkhead Box O1/química , Proteína Forkhead Box O1/metabolismo , Fragmentos de Péptidos/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Sialoglicoproteínas/metabolismo , Activación Transcripcional/fisiología , Cisteína/química , Glucosa-6-Fosfatasa/biosíntesis , Células HEK293 , Células Hep G2 , Humanos , Selenoproteína P/biosíntesis
5.
Biochimie ; 140: 106-116, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28711683

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) presents with growing prevalence worldwide, though its pharmacological treatment remains to be established. This study aimed to evaluate the effects of a PPAR-alpha agonist on liver tissue structure, ultrastructure, and metabolism, focusing on gene and protein expression of de novo lipogenesis and gluconeogenesis pathways, in diet-induced obese mice. Male C57BL/6 mice (three months old) received a control diet (C, 10% of lipids, n = 10) or a high-fat diet (HFD, 50% of lipids, n = 10) for ten weeks. These groups were subdivided to receive the treatment (n = 5 per group): C, C-alpha (PPAR-alpha agonist, 2.5 mg/kg/day mixed in the control diet), HFD and HFD-alpha group (PPAR-alpha agonist, 2.5 mg/kg/day mixed in the HFD). The effects were compared with biometrical, biochemical, molecular biology and transmission electron microscopy (TEM) analyses. HFD showed greater body mass (BM) and insulinemia than C, both of which were tackled by the treatment in the HFD-alpha group. Increased hepatic protein expression of glucose-6-phosphatase, CHREBP and gene expression of PEPCK in HFD points to increased gluconeogenesis. Treatment rescued these parameters in the HFD-alpha group, eliciting a reduced hepatic glucose output, confirmed by the smaller GLUT2 expression in HFD-alpha than in HFD. Conversely, favored de novo lipogenesis was found in the HFD group by the increased expression of PPAR-gamma, and its target gene SREBP-1, FAS and GK when compared to C. The treatment yielded a marked reduction in the expression of all lipogenic factors. TEM analyses showed a greater numerical density of mitochondria per area of tissue in treated than in untreated groups, suggesting an increase in beta-oxidation and the consequent NAFLD control. PPAR-alpha activation reduced BM and treated insulin resistance (IR) and NAFLD by increasing the number of mitochondria and reducing hepatic gluconeogenesis and de novo lipogenesis protein and gene expressions in a murine obesity model.


Asunto(s)
Grasas de la Dieta/efectos adversos , Hígado/metabolismo , Mitocondrias Hepáticas/metabolismo , Obesidad/tratamiento farmacológico , PPAR alfa/agonistas , Pirimidinas/farmacología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Grasas de la Dieta/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa-6-Fosfatasa/biosíntesis , Resistencia a la Insulina , Lipogénesis/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Mitocondrias Hepáticas/patología , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Proteínas Nucleares/biosíntesis , Obesidad/inducido químicamente , Obesidad/metabolismo , Obesidad/patología , PPAR alfa/metabolismo , PPAR gamma/biosíntesis , Fosfoenolpiruvato Carboxiquinasa (ATP)/biosíntesis , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/biosíntesis , Factores de Transcripción/biosíntesis , Receptor fas/biosíntesis
6.
J Biol Chem ; 292(25): 10444-10454, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28473467

RESUMEN

Maintenance of whole-body glucose homeostasis is critical to glycemic function. Genetic variants mapping to chromosome 8p23.1 in genome-wide association studies have been linked to glycemic traits in humans. The gene of known function closest to the mapped region, PPP1R3B (protein phosphatase 1 regulatory subunit 3B), encodes a protein (GL) that regulates glycogen metabolism in the liver. We therefore sought to test the hypothesis that hepatic PPP1R3B is associated with glycemic traits. We generated mice with either liver-specific deletion (Ppp1r3bΔhep ) or liver-specific overexpression of Ppp1r3b The Ppp1r3b deletion significantly reduced glycogen synthase protein abundance, and the remaining protein was predominantly phosphorylated and inactive. As a consequence, glucose incorporation into hepatic glycogen was significantly impaired, total hepatic glycogen content was substantially decreased, and mice lacking hepatic Ppp1r3b had lower fasting plasma glucose than controls. The concomitant loss of liver glycogen impaired whole-body glucose homeostasis and increased hepatic expression of glycolytic enzymes in Ppp1r3bΔhep mice relative to controls in the postprandial state. Eight hours of fasting significantly increased the expression of two critical gluconeogenic enzymes, phosphoenolpyruvate carboxykinase and glucose-6-phosphatase, above the levels in control livers. Conversely, the liver-specific overexpression of Ppp1r3b enhanced hepatic glycogen storage above that of controls and, as a result, delayed the onset of fasting-induced hypoglycemia. Moreover, mice overexpressing hepatic Ppp1r3b upon long-term fasting (12-36 h) were protected from blood ketone-body accumulation, unlike control and Ppp1r3bΔhep mice. These findings indicate a major role for Ppp1r3b in regulating hepatic glycogen stores and whole-body glucose/energy homeostasis.


Asunto(s)
Glucemia/metabolismo , Metabolismo Energético/fisiología , Gluconeogénesis/fisiología , Glucógeno/biosíntesis , Hígado/metabolismo , Proteína Fosfatasa 1/biosíntesis , Animales , Glucemia/genética , Ayuno/sangre , Regulación Enzimológica de la Expresión Génica/fisiología , Glucosa-6-Fosfatasa/biosíntesis , Glucosa-6-Fosfatasa/genética , Glucógeno/genética , Ratones , Ratones Noqueados , Especificidad de Órganos , Fosfoenolpiruvato Carboxiquinasa (ATP)/biosíntesis , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Proteína Fosfatasa 1/genética
7.
PLoS One ; 10(7): e0132113, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26148093

RESUMEN

Increased maternal androgen exposure during pregnancy programmes a polycystic ovary syndrome (PCOS)-like condition, with metabolic dysfunction, in adult female offspring. Other in utero exposures associated with the development of insulin resistance, such as intrauterine growth restriction and exposure to prenatal glucocorticoids, are associated with altered fetal gluconeogenesis. We therefore aimed to assess the effect of maternal androgenisation on the expression of PEPCK and G6PC in the ovine fetus. Pregnant Scottish Greyface sheep were treated with twice weekly testosterone propionate (TP; 100mg) or vehicle control from day 62 to day 102 of gestation. At day 90 and day 112 fetal plasma and liver and kidney tissue was collected for analysis. PEPCK and G6PC expression were analysed by quantitative RT-PCR, immunohistochemistry and western blotting. PEPCK and G6PC were localised to fetal hepatocytes but maternal androgens had no effect on female or male fetuses. PEPCK and G6PC were also localised to the renal tubules and renal PEPCK (P<0.01) and G6PC (P = 0.057) were lower in females after prenatal androgenisation with no change in male fetuses. These tissue and sex specific observations could not be explained by alterations in fetal insulin or cortisol. The sexual dimorphism may be related to the increase in circulating estrogen (P<0.01) and testosterone (P<0.001) in females but not males. The tissue specific effects may be related to the increased expression of ESR1 (P<0.01) and AR (P<0.05) in the kidney when compared to the fetal liver. After discontinuation of maternal androgenisation female fetal kidney PEPCK expression normalised. These data further highlight the fetal and sexual dimorphic effects of maternal androgenisation, an antecedent to adult disease and the plasticity of fetal development.


Asunto(s)
Andrógenos/efectos adversos , Gluconeogénesis/efectos de los fármacos , Riñón/embriología , Síndrome del Ovario Poliquístico/embriología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Propionato de Testosterona/efectos adversos , Andrógenos/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucosa-6-Fosfatasa/biosíntesis , Hepatocitos/metabolismo , Humanos , Riñón/patología , Masculino , Fosfoenolpiruvato Carboxiquinasa (ATP)/biosíntesis , Síndrome del Ovario Poliquístico/inducido químicamente , Síndrome del Ovario Poliquístico/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/patología , Ovinos , Propionato de Testosterona/farmacología
8.
PLoS One ; 10(4): e0122987, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25849936

RESUMEN

Thyroid hormone receptor (TR) agonists have been proposed as therapeutic agents to treat non-alcoholic fatty liver disease (NAFLD) and insulin resistance. We investigated the ability of the TR agonists GC-1 and KB2115 to reduce hepatic steatosis in ob/ob mice. Both compounds markedly reduced hepatic triglyceride levels and ameliorated hepatic steatosis. However, the amelioration of fatty liver was not sufficient to improve insulin sensitivity in these mice and reductions in hepatic triglycerides did not correlate with improvements in insulin sensitivity or glycemic control. Instead, the effects of TR activation on glycemia varied widely and were found to depend upon the time of treatment as well as the compound and dosage used. Lower doses of GC-1 were found to further impair glycemic control, while a higher dose of the same compound resulted in substantially improved glucose tolerance and insulin sensitivity, despite all doses being equally effective at reducing hepatic triglyceride levels. Improvements in glycemic control and insulin sensitivity were observed only in treatments that also increased body temperature, suggesting that the induction of thermogenesis may play a role in mediating these beneficial effects. These data illustrate that the relationship between TR activation and insulin sensitivity is complex and suggests that although TR agonists may have value in treating NAFLD, their effect on insulin sensitivity must also be considered.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Receptores de Hormona Tiroidea/agonistas , Acetatos/farmacología , Acetatos/uso terapéutico , Anilidas/farmacología , Anilidas/uso terapéutico , Animales , Glucemia/metabolismo , Temperatura Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inducción Enzimática/efectos de los fármacos , Glucosa-6-Fosfatasa/biosíntesis , Masculino , Ratones , Ratones Obesos , Fenoles/farmacología , Fenoles/uso terapéutico , Factores de Tiempo
9.
Mol Biochem Parasitol ; 194(1-2): 64-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24798031

RESUMEN

The liver fluke, Opisthorchis felineus of the Opisthorchiidae family, is a well-known causative agent of opisthorchiasis in Russia and Europe. The aim of this work was to identify genes encoding thyroid hormone receptors in O. felineus, and to analyze the expression of possible target genes in response to treatment with exogenous thyroid hormones. We identified two genes encoding thyroid hormone receptors in the O. felineus genome, THRA and THRB. The genes were differentially expressed through the life cycle. The maximal level of mRNA expression of THRA1 and THRB was observed in adult worms. Treatment of the worms with triiodothyronine and thyroxine resulted in an increase in glucose 6-phosphatase mRNA expression and a decrease in malate dehydrogenase mRNA expression, potential gene targets of thyroid hormones. These data indicate that thyroid hormone receptors may perform essential roles in physiological processes in adult O. felineus.


Asunto(s)
Opisthorchis/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa-6-Fosfatasa/biosíntesis , Malato Deshidrogenasa/biosíntesis , Opisthorchis/efectos de los fármacos , Opisthorchis/genética , Receptores de Hormona Tiroidea/genética , Tiroxina/metabolismo , Triyodotironina/metabolismo
10.
FASEB J ; 28(8): 3339-50, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24755741

RESUMEN

Reduced AMP kinase (AMPK) activity has been shown to play a key deleterious role in increased hepatic gluconeogenesis in diabetes, but the mechanism whereby this occurs remains unclear. In this article, we document that another AMP-dependent enzyme, AMP deaminase (AMPD) is activated in the liver of diabetic mice, which parallels with a significant reduction in AMPK activity and a significant increase in intracellular glucose accumulation in human HepG2 cells. AMPD activation is induced by a reduction in intracellular phosphate levels, which is characteristic of insulin resistance and diabetic states. Increased gluconeogenesis is mediated by reduced TORC2 phosphorylation at Ser171 by AMPK in these cells, as well as by the up-regulation of the rate-limiting enzymes PEPCK and G6Pc. The mechanism whereby AMPD controls AMPK activation depends on the production of a specific AMP downstream metabolite through AMPD, uric acid. In this regard, humans have higher uric acid levels than most mammals due to a mutation in uricase, the enzyme involved in uric acid degradation in most mammals, that developed during a period of famine in Europe 1.5 × 10(7) yr ago. Here, working with resurrected ancestral uricases obtained from early hominids, we show that their expression on HepG2 cells is enough to blunt gluconeogenesis in parallel with an up-regulation of AMPK activity. These studies identify a key role AMPD and uric acid in mediating hepatic gluconeogenesis in the diabetic state, via a mechanism involving AMPK down-regulation and overexpression of PEPCK and G6Pc. The uricase mutation in the Miocene likely provided a survival advantage to help maintain glucose levels under conditions of near starvation, but today likely has a role in the pathogenesis of diabetes.


Asunto(s)
AMP Desaminasa/fisiología , Gluconeogénesis/fisiología , Hígado/metabolismo , Inanición/fisiopatología , Ácido Úrico/metabolismo , AMP Desaminasa/antagonistas & inhibidores , AMP Desaminasa/genética , Proteínas Quinasas Activadas por AMP/fisiología , Animales , Diabetes Mellitus Experimental/metabolismo , Europa (Continente) , Regulación Enzimológica de la Expresión Génica , Gluconeogénesis/efectos de los fármacos , Glucosa-6-Fosfatasa/biosíntesis , Células Hep G2 , Historia Antigua , Hominidae/fisiología , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Hígado/enzimología , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Complejos Multiproteicos/fisiología , Fosfatos/metabolismo , Fosfatos/farmacología , Fosfoenolpiruvato Carboxiquinasa (ATP)/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Selección Genética , Organismos Libres de Patógenos Específicos , Inanición/historia , Serina-Treonina Quinasas TOR/fisiología , Transducción Genética , Urato Oxidasa/genética , Urato Oxidasa/historia , Urato Oxidasa/metabolismo , Ácido Úrico/farmacología
11.
J Biol Chem ; 289(4): 2396-404, 2014 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-24311788

RESUMEN

We showed recently that apoA-IV improves glucose homeostasis by enhancing pancreatic insulin secretion in the presence of elevated levels of glucose. Therefore, examined whether apolipoprotein A-IV (apoA-IV) also regulates glucose metabolism through the suppression of hepatic gluconeogenesis. The ability of apoA-IV to lower gluconeogenic gene expression and glucose production was measured in apoA-IV(-/-) and wild-type mice and primary mouse hepatocytes. The transcriptional regulation of Glc-6-Pase and phosphoenolpyruvate carboxykinase (PEPCK) by apoA-IV was determined by luciferase activity assay. Using bacterial two-hybrid library screening, NR1D1 was identified as a putative apoA-IV-binding protein. The colocalization and interaction between apoA-IV and NR1D1 were confirmed by immunofluorescence, in situ proximity ligation assay, and coimmunoprecipitation. Enhanced recruitment of NR1D1 and activity by apoA-IV to Glc-6-Pase promoter was verified with ChIP and a luciferase assay. Down-regulation of apoA-IV on gluconeogenic genes is mediated through NR1D1, as illustrated in cells with NR1D1 knockdown by siRNA. We found that apoA-IV suppresses the expression of PEPCK and Glc-6-Pase in hepatocytes; decreases hepatic glucose production; binds and activates nuclear receptor NR1D1 and stimulates NR1D1 expression; in cells lacking NR1D1, fails to inhibit PEPCK and Glc-6-Pase gene expression; and stimulates higher hepatic glucose production and higher gluconeogenic gene expression in apoA-IV(-/-) mice. We conclude that apoA-IV inhibits hepatic gluconeogenesis by decreasing Glc-6-Pase and PEPCK gene expression through NR1D1. This novel regulatory pathway connects an influx of energy as fat from the gut (and subsequent apoA-IV secretion) with inhibition of hepatic glucose production.


Asunto(s)
Apolipoproteínas A/metabolismo , Gluconeogénesis/fisiología , Glucosa/biosíntesis , Hepatocitos/metabolismo , Hígado/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Animales , Apolipoproteínas A/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Glucosa/genética , Glucosa-6-Fosfatasa/biosíntesis , Glucosa-6-Fosfatasa/genética , Células HEK293 , Células Hep G2 , Hepatocitos/citología , Humanos , Hígado/citología , Ratones , Ratones Noqueados , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/biosíntesis , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Regiones Promotoras Genéticas/fisiología
12.
J Biol Chem ; 288(42): 30365-30372, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-23995837

RESUMEN

Hepatic gluconeogenesis is a concerted process that integrates transcriptional regulation with hormonal signals. A major regulator is thyroid hormone (TH), which acts through its nuclear receptor (TR) to induce the expression of the hepatic gluconeogenic genes, phosphoenolpyruvate carboxykinase (PCK1) and glucose-6-phosphatase (G6PC). Forkhead transcription factor FoxO1 also is an important regulator of these genes; however, its functional interactions with TR are not known. Here, we report that TR-mediated transcriptional activation of PCK1 and G6PC in human hepatic cells and mouse liver was FoxO1-dependent and furthermore required FoxO1 deacetylation by the NAD(+)-dependent deacetylase, SirT1. siRNA knockdown of FoxO1 decreased, whereas overexpression of FoxO1 increased, TH-dependent transcriptional activation of PCK1 and G6PC in cultured hepatic cells. FoxO1 siRNA knockdown also decreased TH-mediated transcription in vivo. Additionally, TH was unable to induce FoxO1 deacetylation or hepatic PCK1 gene expression in TH receptor ß-null (TRß(-/-)) mice. Moreover, TH stimulated FoxO1 recruitment to the PCK1 and G6PC gene promoters in a SirT1-dependent manner. In summary, our results show that TH-dependent deacetylation of a second metabolically regulated transcription factor represents a novel mechanism for transcriptional integration of nuclear hormone action with cellular energy status.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Gluconeogénesis/fisiología , Hígado/metabolismo , Hormonas Tiroideas/metabolismo , Transcripción Genética/fisiología , Activación Transcripcional/fisiología , Acetilación , Animales , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Glucosa-6-Fosfatasa/biosíntesis , Glucosa-6-Fosfatasa/genética , Células Hep G2 , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Noqueados , Fosfoenolpiruvato Carboxiquinasa (GTP)/biosíntesis , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Regiones Promotoras Genéticas/fisiología , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Hormonas Tiroideas/genética
13.
Endocr J ; 60(8): 941-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23665701

RESUMEN

Our previous study reported that thyroid-stimulating hormone (TSH) promotes cholesterol synthesis via the cyclic adenosine monophosphate/protein kinase A/cAMP regulatory element-binding protein (cAMP/PKA/CREB) pathway after binding to TSH receptors (TSHR) in the liver. The hepatic cAMP/PKA/CREB pathway also plays an important role in maintaining fasting glucose homeostasis. These findings implied a possible role for TSH in hepatic glucose metabolism. In this study, we used TSH receptor knockout mice (Tshr-ko mice) to clarify the effect of Tshr deletion on hepatic glucose metabolism, and investigated whether the effects of TSH directly regulate hepatic gluconeogenesis in HepG2 cells. Tshr-ko mice exhibited decreased fasting blood glucose levels, increased insulin sensitivity but normal level of fasting plasma insulin. Tshr deletion impaired hepatic glucose production by down-regulating the expression of glucose-6-phosphatase (G6P) and phosphoenolpyruvate pyruvate carboxylase (PEPCK) mRNA, two rate-limiting enzymes in hepatic gluconeogenesis, and enhancing the abundance of hepatic glucokinase (GK), the first enzyme regulating glycogen synthesis. Moreover, Tshr deletion inhibited the protein expression of hepatic phospho-CREB and increased the protein expression of hepatic phospho-AMP-activated protein kinase (p-AMPK), two up-stream regulators of PEPCK and G6P mRNA. In HepG2 cells, TSH increased the expression of G6P and PEPCK at mRNA level. These results indicated the simulative effects of TSH on hepatic glucose production in vivo and in vitro, suggesting a novel role for TSH in hepatic glucose metabolism.


Asunto(s)
Glucemia/metabolismo , Gluconeogénesis/efectos de los fármacos , Hígado/metabolismo , Receptores de Tirotropina/deficiencia , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación hacia Abajo , Ayuno , Gluconeogénesis/genética , Glucosa/metabolismo , Glucosa-6-Fosfatasa/biosíntesis , Células Hep G2 , Humanos , Insulina/sangre , Resistencia a la Insulina , Glucógeno Hepático/metabolismo , Ratones , Ratones Noqueados , Fosfoenolpiruvato Carboxiquinasa (GTP)/biosíntesis , Tirotropina/farmacología
14.
Asian Pac J Trop Med ; 5(11): 875-81, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23146801

RESUMEN

OBJECTIVE: To investigate the hypoglycemic effect of the aqueous extract of Octomeles sumatrana (O. sumatrana) (OS) in streptozotocin-induced diabetic rats (STZ) and its molecular mechanisms. METHODS: Diabetes was induced by intraperitoneal (i.p.) injection of streptozotocin (55 mg/kg) in to male Sprague-Dawley rats. Rats were divided into six different groups; normal control rats were not induced with STZ and served as reference, STZ diabetic control rats were given normal saline. Three groups were treated with OS aqueous extract at 0.2, 0.3 and 0.5 g/kg, orally twice daily continuously for 21 d. The fifth group was treated with glibenclamide (6 mg/kg) in aqueous solution orally continuously for 21 d. After completion of the treatment period, biochemical parameters and expression levels of glucose transporter 2 (Slc2a2), glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PCK1) were determined in liver by quantitative real time PCR. RESULTS: Administration of OS at different doses to STZ induced diabetic rats, resulted in significant decrease (P<0.05) in blood glucose level in a dose dependent manner by 36%, 48%, and 64% at doses of 0.2, 0.3 and 0.5 g/kg, respectively, in comparison to the STZ control values. Treatment with OS elicited an increase in the expression level of Slc2a2 gene but reduced the expression of G6Pase and PCK1 genes. Morefore, OS treated rats, showed significantly lower levels of serum alanine transaminase (ALT), aspartate aminotransferase (AST) and urea levels compared to STZ untreated rats. The extract at different doses elicited signs of recovery in body weight gain when compared to STZ diabetic controls although food and water consumption were significantly lower in treated groups compared to STZ diabetic control group. CONCLUSIONS: O. sumatrana aqueous extract is beneficial for improvement of hyperglycemia by increasing gene expression of liver Slc2a2 and reducing expression of G6Pase and PCK1 genes in streptozotocin-induced diabetic rats.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Helechos/química , Hipoglucemiantes/administración & dosificación , Extractos Vegetales/administración & dosificación , Administración Oral , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 2/biosíntesis , Glucosa-6-Fosfatasa/biosíntesis , Hipoglucemiantes/aislamiento & purificación , Masculino , Fosfoenolpiruvato Carboxiquinasa (GTP)/biosíntesis , Extractos Vegetales/aislamiento & purificación , Ratas , Ratas Sprague-Dawley
15.
J Biol Chem ; 287(50): 41875-87, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23074219

RESUMEN

Cyclic AMP (cAMP) induces steroidogenic enzyme gene expression and stimulates testosterone production in Leydig cells. Phosphoenolpyruvate carboxykinase (PEPCK) is expressed in Leydig cells, but its role has not been defined. In this study, we found that PEPCK and glucose-6-phosphatase (Glc-6-Pase) are increased significantly following cAMP treatment of mouse Leydig cells. Moreover, cAMP treatment increased recruitment of the cAMP-response element-binding transcription factor and decreased recruitment of the corepressor DAX-1 on the pepck promoter. Furthermore, cAMP induced an increase in ATP that correlated with a decrease in phospho-AMP-activated protein kinase (AMPK). In contrast, knockdown or inhibition of PEPCK decreased ATP and increased phospho-AMPK. Treatment with an AMPK activator or overexpression of the constitutively active form of AMPK inhibited cAMP-induced steroidogenic enzyme promoter activities and gene expression. Liver receptor homolog-1 (LRH-1) was involved in cAMP-induced steroidogenic enzyme gene expression but was inhibited by AMPK activation in Leydig cells. Additionally, inhibition or knockdown of PEPCK and Glc-6-Pase decreased cAMP-mediated induction of steroidogenic enzyme gene expression and steroidogenesis. Finally, pubertal mouse (8-week-old) testes and human chorionic gonadotropin-induced prepubertal mouse testes showed increased PEPCK and Glc-6-Pase gene expression. Taken together, these results suggest that induction of PEPCK and Glc-6-Pase by cAMP plays an important role in Leydig cell steroidogenesis.


Asunto(s)
Glucosa-6-Fosfatasa/biosíntesis , Células Intersticiales del Testículo/enzimología , Fosfoenolpiruvato Carboxiquinasa (ATP)/biosíntesis , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , AMP Cíclico/genética , AMP Cíclico/metabolismo , Receptor Nuclear Huérfano DAX-1/genética , Receptor Nuclear Huérfano DAX-1/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Glucosa-6-Fosfatasa/genética , Células HeLa , Humanos , Células Intersticiales del Testículo/citología , Masculino , Ratones , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Regiones Promotoras Genéticas/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo
16.
PLoS One ; 7(7): e41168, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22848439

RESUMEN

BACKGROUND: MAP kinase phosphatase 3 (MKP-3) is known to attenuate the ERK signaling pathway. It has been recently demonstrated that MKP-3 is also a player in promoting hepatic glucose output in obese state by interacting and activating FOXO1. Reduction of hepatic MKP-3 expression is sufficient to reduce blood glucose levels in both diet-induced and genetically obese mice. METHODOLOGY/PRINCIPAL FINDINGS: In current study, the mechanism of MKP-3/FOXO1 interaction and the effects on transcription of gluconeogenic gene and glucose output was investigated in Fao hepatoma cells by using mutated MKP-3 and FOXO1 adenoviral constructs. The results indicate that MKP-3 phosphatase activity is not required for MKP-3/FOXO1 interaction but is essential for FOXO1 nuclear translocation and MKP-3 promoted gluconeogenesis. Compared to GFP control (1±0.38), MKP-3 increased G6Pase gene expression by 242% (3.42±0.62) while inactive MKP-3 does not change G6Pase expression (0.98±0.17). The residues 200-260 of MKP-3 and the residues 360-456 of FOXO1 are essential for mediating MKP-3/FOXO1 interaction. Interestingly, ERK phosphorylation deficient but not Akt phosphorylation deficient FOXO1 mutant lost interaction with MKP-3. Furthermore, in vivo experiments showed that Akt phosphorylation resistant FOXO1 3A mutant is sufficient to rescue the hypoglycemia caused by MKP-3 knock down in the liver of lean mice (from 141±6.78 to 209±14.64 mg/dL). CONCLUSIONS/SIGNIFICANCE: 1) Critical residues mediating MKP-3/FOXO1 interaction have been identified; 2) ERK phosphorylation deficient FOXO1 mutant is as potent as Akt phosphorylation deficient FOXO1 mutant in activating transcription of gluconeogenic genes; 3) Constitutively active FOXO1 can rescue the hypoglycemic effect caused by reduced hepatic MKP-3 expression in vivo.


Asunto(s)
Núcleo Celular/metabolismo , Fosfatasa 6 de Especificidad Dual/biosíntesis , Factores de Transcripción Forkhead/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Gluconeogénesis/fisiología , Hígado/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Línea Celular , Núcleo Celular/genética , Fosfatasa 6 de Especificidad Dual/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Técnicas de Silenciamiento del Gen , Glucosa-6-Fosfatasa/biosíntesis , Glucosa-6-Fosfatasa/genética , Humanos , Hígado/citología , Ratones , Mutación , Mapeo Peptídico/métodos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
17.
Artículo en Inglés | MEDLINE | ID: mdl-22676927

RESUMEN

OBJECTIVE: The objective of this study was to analyze the relationship between the uptake of (18)F-2-fluoro-2-deoxy-d-glucose (FDG) by positron emission tomography-computerized tomography (PET-CT) and glucose metabolism/hypoxia markers in oral squamous cell carcinoma (OSCC). STUDY DESIGN: Thirty-six patients with OSCC (tongue [n = 23], buccal mucosa [n = 7], and floor of the mouth [n = 6]) were assessed and underwent incisional biopsy and subsequently received FDG-PET-CT. Expressions of hypoxia-inducible factor 1α (HIF-1α), glucose transporter protein 1 (GLUT-1), hexokinase-II (HK-II), and glucose-6-phosphatase (G6Pase) were immunohistochemically quantified, and FDG uptake was evaluated by the maximum standardized uptake values (SUV(max)) at the primary tumor site. RESULTS: FDG uptake was found to be significantly correlated with the T classification of OSCC but not with other clinicopathologic characteristics, such as the N classification, clinical type, and histologic grade of malignancy. In the early-stage (T1 and T2) tumor, FDG uptake was significantly associated with the expression levels of GLUT-1, HK II, and HIF-1α, and the expression levels of GLUT-1 and HK-II significantly correlated with HIF-1α expression levels. However, there were no correlations between the expression levels of these molecules and SUV(max) in the late-stage (T3 and T4) tumor. CONCLUSIONS: FDG uptake was significantly associated with the expression levels of glucose metabolism-related molecules, such as GLUT-1, HK II, and HIF-1α, especially in early-stage tumors.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Glucosa/metabolismo , Hipoxia/metabolismo , Neoplasias de la Boca/metabolismo , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/patología , Femenino , Fluorodesoxiglucosa F18 , Transportador de Glucosa de Tipo 1/biosíntesis , Transportador de Glucosa de Tipo 1/genética , Glucosa-6-Fosfatasa/biosíntesis , Glucosa-6-Fosfatasa/genética , Hexoquinasa/biosíntesis , Hexoquinasa/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Persona de Mediana Edad , Suelo de la Boca/metabolismo , Mucosa Bucal/metabolismo , Neoplasias de la Boca/patología , Clasificación del Tumor , Estadificación de Neoplasias , Tomografía de Emisión de Positrones , Estadísticas no Paramétricas , Lengua/metabolismo
18.
Eur J Pharmacol ; 679(1-3): 139-43, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22296760

RESUMEN

The constitutive androstane receptor (CAR, NR1I3) has a central role in detoxification processes, regulating the expression of a set of genes involved in metabolism. The dual role of NR1I3 as both a xenosensor and as a regulator of endogenous energy metabolism has recently been accepted. Here, we investigated the mechanism of transcriptional regulation of the glucose metabolising genes phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase) by the cis isomer of 2,4,6-triphenyldioxane-1,3 (cisTPD), a highly effective NR1I3 activator in rat liver. It was shown that expression of the gluconeogenic genes PEPCK and G6Pase was repressed by cisTPD treatment under fasting conditions. Western-blot analysis demonstrated a clear reduction in the intensity of PEPCK and G6Pase immunobands from the livers of cisTPD-treated animals relative to bands from the livers of control animals. Chromatin immunoprecipitation assays demonstrated that cisTPD prevents the binding of FOXO1 to the insulin response sequences in the PEPCK and G6Pase gene promoters in rat liver. Moreover, cisTPD-activated NR1I3 inhibited NR2A1 (HNF-4) transactivation by competing with NR2A1 for binding to the NR2A1-binding element (DR1-site) in the gluconeogenic gene promoters. Thus, our results are consistent with the hypothesis that the cisTPD-activated NR1I3 participates in the regulation of the gluconeogenic genes PEPCK and G6Pase.


Asunto(s)
Dioxanos/farmacología , Regulación de la Expresión Génica/fisiología , Gluconeogénesis/fisiología , Glucosa-6-Fosfatasa/biosíntesis , Fosfoenolpiruvato Carboxiquinasa (GTP)/biosíntesis , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Receptor de Androstano Constitutivo , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Gluconeogénesis/efectos de los fármacos , Factor Nuclear 4 del Hepatocito/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Ratas , Ratas Wistar , Factores de Transcripción/antagonistas & inhibidores
19.
Am J Physiol Endocrinol Metab ; 302(4): E409-16, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22127228

RESUMEN

The aim of the present study was to examine the effects of pyrrolidine dithiocarbamate (PDTC) on hepatic glycogen synthesis and FoxO1 transcriptional activity in type 2 diabetic rats and the mechanism underlying these effects. Fasting blood glucose and glycogen deposition, together with expressions of two key genes related to gluconeogenesis, were studied in the liver of rats fed a normal diet (NC), high-fat diet (HFD)-induced insulin-resistant rats made type 2 diabetic by a single intraperitoneal injection of streptozotocin (DM), and a DM with intervention of PDTC (DM + PDTC) for 1 wk. The phosphorylation of Akt, GSK-3ß, and FoxO1 was assessed in liver extracts of fasted rats by Western blot, whereas indirect immunofluorescence staining was performed to determine the cellular distribution of FoxO1. The DM rats exhibited obvious increases in fasting blood glucose as well as decreased hepatic glycogen content compared with the NC group. Activation of the Akt/GSK-3ß pathway and inactivating phosphorylation of FoxO1 were reduced greatly in DM rat livers (P < 0.01). By contrast, PDTC treatment protected DM rats against high fasting blood glucose and hepatic glycogen deposition loss. PDTC also elicited an increase in Akt/GSK-3ß signaling and subsequent inactivation and nuclear export of FoxO1 in DM rat livers, which translated into a significant reduction in the expression of two FoxO1 target genes, phosphoenolpyruvate carboxykinase and glucose-6-phosphatase. This study suggests that PDTC enhances hepatic glycogen synthesis, whereas it reduces FoxO1 transcriptional activity in DM rats.


Asunto(s)
Antioxidantes/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Factores de Transcripción Forkhead/metabolismo , Glucógeno Hepático/biosíntesis , Hígado/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Pirrolidinas/farmacología , Tiocarbamatos/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Glucemia/efectos de los fármacos , Dieta Alta en Grasa , Ayuno/sangre , Gluconeogénesis/efectos de los fármacos , Glucosa-6-Fosfatasa/biosíntesis , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Resistencia a la Insulina , Hígado/química , Hígado/metabolismo , Masculino , Fosfoenolpiruvato Carboxiquinasa (ATP)/biosíntesis , Fosforilación , Ratas , Ratas Wistar
20.
J Clin Invest ; 121(12): 4916-20, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22056385

RESUMEN

Increased endogenous glucose production (EGP) is a hallmark of type 2 diabetes mellitus. While there is evidence for central regulation of EGP by activation of hypothalamic ATP-sensitive potassium (K(ATP)) channels in rodents, whether these central pathways contribute to regulation of EGP in humans remains to be determined. Here we present evidence for central nervous system regulation of EGP in humans that is consistent with complementary rodent studies. Oral administration of the K(ATP) channel activator diazoxide under fixed hormonal conditions substantially decreased EGP in nondiabetic humans and Sprague Dawley rats. In rats, comparable doses of oral diazoxide attained appreciable concentrations in the cerebrospinal fluid, and the effects of oral diazoxide were abolished by i.c.v. administration of the K(ATP) channel blocker glibenclamide. These results suggest that activation of hypothalamic K(ATP) channels may be an important regulator of EGP in humans and that this pathway could be a target for treatment of hyperglycemia in type 2 diabetes mellitus.


Asunto(s)
Diazóxido/farmacología , Gluconeogénesis/efectos de los fármacos , Hipotálamo/metabolismo , Canales de Potasio/fisiología , Adulto , Animales , Glucemia/análisis , Barrera Hematoencefálica , Bloqueadores de los Canales de Calcio/administración & dosificación , Bloqueadores de los Canales de Calcio/farmacología , Diazóxido/administración & dosificación , Diazóxido/líquido cefalorraquídeo , Diazóxido/farmacocinética , Diazóxido/uso terapéutico , Método Doble Ciego , Inducción Enzimática/efectos de los fármacos , Femenino , Gluconeogénesis/fisiología , Técnica de Clampeo de la Glucosa , Glucosa-6-Fosfatasa/antagonistas & inhibidores , Glucosa-6-Fosfatasa/biosíntesis , Glucosa-6-Fosfatasa/genética , Gliburida/administración & dosificación , Gliburida/farmacología , Humanos , Hipotálamo/fisiopatología , Inyecciones Intraventriculares , Insulina/sangre , Activación del Canal Iónico/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Fosforilación/efectos de los fármacos , Canales de Potasio/agonistas , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA