Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Hepatol ; 69(5): 1099-1109, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29981427

RESUMEN

BACKGROUND & AIMS: Embedded into a complex signaling network that coordinates glucose uptake, usage and production, the nuclear bile acid receptor FXR is expressed in several glucose-processing organs including the liver. Hepatic gluconeogenesis is controlled through allosteric regulation of gluconeogenic enzymes and by glucagon/cAMP-dependent transcriptional regulatory pathways. We aimed to elucidate the role of FXR in the regulation of fasting hepatic gluconeogenesis. METHODS: The role of FXR in hepatic gluconeogenesis was assessed in vivo and in mouse primary hepatocytes. Gene expression patterns in response to glucagon and FXR agonists were characterized by quantitative reverse transcription PCR and microarray analysis. FXR phosphorylation by protein kinase A was determined by mass spectrometry. The interaction of FOXA2 with FXR was identified by cistromic approaches and in vitro protein-protein interaction assays. The functional impact of the crosstalk between FXR, the PKA and FOXA2 signaling pathways was assessed by site-directed mutagenesis, transactivation assays and restoration of FXR expression in FXR-deficient hepatocytes in which gene expression and glucose production were assessed. RESULTS: FXR positively regulates hepatic glucose production through two regulatory arms, the first one involving protein kinase A-mediated phosphorylation of FXR, which allowed for the synergistic activation of gluconeogenic genes by glucagon, agonist-activated FXR and CREB. The second arm involves the inhibition of FXR's ability to induce the anti-gluconeogenic nuclear receptor SHP by the glucagon-activated FOXA2 transcription factor, which physically interacts with FXR. Additionally, knockdown of Foxa2 did not alter glucagon-induced and FXR agonist enhanced expression of gluconeogenic genes, suggesting that the PKA and FOXA2 pathways regulate distinct subsets of FXR responsive genes. CONCLUSIONS: Thus, hepatic glucose production is regulated during physiological fasting by FXR, which integrates the glucagon/cAMP signal and the FOXA2 signal, by being post-translationally modified, and by engaging in protein-protein interactions, respectively. LAY SUMMARY: Activation of the nuclear bile acid receptor FXR regulates gene expression networks, controlling lipid, cholesterol and glucose metabolism, which are mostly effective after eating. Whether FXR exerts critical functions during fasting is unknown. The results of this study show that FXR transcriptional activity is regulated by the glucagon/protein kinase A and the FOXA2 signaling pathways, which act on FXR through phosphorylation and protein-protein interactions, respectively, to increase hepatic glucose synthesis.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Ayuno/metabolismo , Gluconeogénesis , Factor Nuclear 3-beta del Hepatocito/fisiología , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Regulación de la Expresión Génica , Glucagón/fisiología , Glucosa/metabolismo , Hepatocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación
2.
FEBS Lett ; 587(23): 3883-90, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24396871

RESUMEN

Nur77 is a stress sensor in pancreatic ß-cells, which negatively regulates glucose-stimulated insulin secretion. We recently showed that a lipotoxic shock caused by exposure of ß-cells to the saturated fatty acid palmitate strongly increases Nur77 expression. Here, using dual luciferase reporter assays and Nur77 promoter deletion constructs, we identified a regulatory cassette between -1534 and -1512 bp upstream from the translational start site mediating Nur77 promoter activation in response to palmitate exposure. Chromatin immunoprecipitation, transient transfection and siRNA-mediated knockdown assays revealed that palmitate induced Nur77 promoter activation involves Sp1 recruitment and ZBP89 release from the gene promoter.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Secretoras de Insulina/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Ácido Palmítico/farmacología , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Ratones , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño , Factor de Transcripción Sp1/genética , Factores de Transcripción/genética
3.
FEBS Lett ; 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24512852

RESUMEN

Nur77 is a stress sensor in pancreatic ß-cells, which negatively regulates glucose-stimulated insulin secretion. We recently showed that a lipotoxic shock caused by exposure of ß-cells to the saturated fatty acid palmitate strongly increases Nur77 expression. Here, using dual luciferase reporter assays and Nur77 promoter deletion constructs, we identified a regulatory cassette between -1534 and -1512 bp upstream from the translational start site mediating Nur77 promoter activation in response to palmitate exposure. Chromatin immunoprecipitation, transient transfection and siRNA-mediated knockdown assays revealed that palmitate induced Nur77 promoter activation involves Sp1 recruitment and ZBP89 release from the gene promoter.

4.
Mol Endocrinol ; 26(3): 399-413, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22301783

RESUMEN

The NR4A orphan nuclear receptors Nur77, Nurr1, and Nor1 exert multiple cellular and metabolic functions. These transcriptional regulators are activated in response to extracellular stresses, including lipotoxic fatty acids (FA) and proinflammatory cytokines. The contribution of NR4As to ß-cell pathophysiology is, however, unknown. We have therefore examined the role of NR4As as downstream contributors to FA-induced ß-cell dysfunctions. Human pancreatic islets and insulinoma ß-cells were used to determine transcriptional programs elicited by NR4A, which were compared to those triggered by palmitate treatment. Functional studies evaluated the consequence of an increased NR4A expression on insulin biosynthesis and secretion and cell viability in insulinoma ß-cells. FA and cytokine treatment increased NR4A expression in pancreatic ß-cells, with Nur77 being most highly inducible in murine ß-cells. Nur77, Nurr1, or Nor1 modulated common and distinct clusters of genes involved notably in cation homeostasis and insulin gene transcription. By altering zinc homeostasis, insulin gene transcription, and secretion, Nur77 was found to be a major transcriptional mediator of part of FA-induced ß-cell dysfunctions. The repressive role of Nur77 in insulin gene regulation was tracked down to protein-protein interaction with FoxO1, a pivotal integrator of the insulin gene regulatory network. The present study identifies a member of the NR4A nuclear receptor subclass, Nur77/NR4A1, as a modulator of pancreatic ß-cell biology. Together with its previously documented role in liver and muscle, its role in ß-cells establishes Nur77 as an important integrator of glucose metabolism.


Asunto(s)
Glucosa/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Animales , Línea Celular , Cromogranina A/metabolismo , Ácidos Grasos/fisiología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucosa/metabolismo , Humanos , Insulina/genética , Secreción de Insulina , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Páncreas/citología , Páncreas/metabolismo , Páncreas/fisiología , Regiones Promotoras Genéticas , Unión Proteica , Estrés Fisiológico
5.
FEBS Lett ; 584(13): 2845-51, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20447400

RESUMEN

Farnesoid X receptor (FXR) is highly expressed in liver and intestine where it controls bile acid (BA), lipid and glucose homeostasis. Here we show that FXR is expressed and functional, as assessed by target gene expression analysis, in human islets and beta-cell lines. FXR is predominantly cytosolic-localized in the islets of lean mice, but nuclear in obese mice. Compared to FXR+/+ mice, FXR-/- mice display a normal architecture and beta-cell mass but the expression of certain islet-specific genes is altered. Moreover, glucose-stimulated insulin secretion (GSIS) is impaired in the islets of FXR-/- mice. Finally, FXR activation protects human islets from lipotoxicity and ameliorates their secretory index.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ácido Palmítico/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Western Blotting , Células Cultivadas , Humanos , Técnicas In Vitro , Isoxazoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Obesidad/metabolismo , Ratas , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética
6.
Cell Physiol Biochem ; 24(5-6): 451-60, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19910685

RESUMEN

Apolipoprotein A-V is an important determinant of plasma triglyceride level in both humans and mice. This study showed the physiological impact of apoA-V on insulin secretion in rat pancreatic beta-cells (INS-1 cells). In order to precise the mechanism of action, binding experiments coupled to mass spectrometry were performed to identify a potential membrane receptor. Results showed an interaction between apoA-V and midkine protein. Confocal microscopy confirmed the plasma membrane co-localisation of this two-proteins after the treatment of INS-1 cells with the apo-AV recombinant protein and indicated that the cell surface midkine could be involved in apoA-V endocytosis, since these two proteins were co-translocated at the plasma membrane or in the cytosol compartment. This co-localisation is correlated with an increase in insulin secretion in a dose dependant manner during short incubation period. Reduction of midkine expression by small interfering RNA duplexes revealed a decrease in the ability of these transfected cells to secrete insulin in presence of apoA-V. Competition experiments for the apoA-V-midkine binding at the cell surface using antibody directed against midkine is able to influence INS-1 cell function as insulin secretion. Our results showed apoA-V ability to enhance insulin secretion in beta-cells and provide evidence of an internalization pathway involving the midkine as partner.


Asunto(s)
Apolipoproteínas/metabolismo , Citocinas/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Secuencia de Aminoácidos , Animales , Apolipoproteína A-V , Apolipoproteínas/análisis , Línea Celular Tumoral , Citocinas/análisis , Citocinas/genética , Endocitosis , Inmunoprecipitación , Secreción de Insulina , Midkina , Datos de Secuencia Molecular , Unión Proteica , ARN Interferente Pequeño/metabolismo , Ratas , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
7.
J Mol Biol ; 380(5): 789-98, 2008 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-18572192

RESUMEN

The apolipoprotein A5 gene (APOA5) is a key player in determining triglyceride concentrations in humans and mice. Since diabetes is often associated with hypertriglyceridemia, this study explores whether APOA5 gene expression is regulated by alteration in glucose homeostasis and the related pathways. d-Glucose activates APOA5 gene expression in a time- and dose-dependent manner in hepatocytes, and the glycolytic pathway involved was determined using d-glucose analogues and metabolites. Together, transient transfections, electrophoretic mobility shift assays and chromatin immunoprecipitation assays show that this regulation occurs at the transcriptional level through an increase of USF1/2 binding to an E-box in the APOA5 promoter. We show that this phenomenon is not due to an increase of mRNA or protein expression levels of USF. Using protein phosphatases 1 and 2A inhibitor, we demonstrate that d-glucose regulates the APOA5 gene via a dephosphorylation mechanism, resulting in an enhanced USF1/2-promoter binding. Last, subsequent suppressions of USF1/2 and phosphatases mRNA through siRNA gene silencing abolished the regulation. We demonstrate that the APOA5 gene is up regulated by d-glucose and USF through phosphatase activation. These findings may provide a new cross-talk between glucose and lipid metabolism.


Asunto(s)
Apolipoproteínas A/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Animales , Apolipoproteína A-V , Apolipoproteínas A/genética , Línea Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucólisis , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Proteína Fosfatasa 1/farmacología , Proteína Fosfatasa 2/farmacología , ARN Mensajero/metabolismo , Ratas , Factores de Tiempo , Transcripción Genética , Factores Estimuladores hacia 5'/metabolismo
8.
Cell Res ; 16(7): 661-70, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16773041

RESUMEN

Liver X receptors (LXRs) are members of the nuclear receptor superfamily, which have been implicated in lipid homeostasis and more recently in glucose metabolism. Here, we show that glucose does not change LXRalpha protein level, but affects its localization in pancreatic beta-cells. LXRalpha is found in the nucleus at 8 mM glucose and in the cytoplasm at 4.2 mM. Addition of glucose translocates LXRalpha from the cytoplasm into the nucleus. Moreover, after the activation of LXR by its synthetic non-steroidal agonist (T0901317), insulin secretion and glucose uptake are increased at 8 mM and decreased at 4.2 mM glucose in a dose-dependent manner. Furthermore, at low glucose condition, okadaic acid reversed LXRalpha effect on insulin secretion, suggesting the involvement of glucose signaling through a phosphorylation-dependent mechanism.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Inhibidores Enzimáticos/metabolismo , Hidrocarburos Fluorados , Insulina/metabolismo , Células Secretoras de Insulina/citología , Receptores X del Hígado , Ácido Ocadaico/metabolismo , Receptores Nucleares Huérfanos , Péptidos/genética , Péptidos/metabolismo , Ratas , Receptores Citoplasmáticos y Nucleares/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Sulfonamidas/metabolismo , Receptor fas/metabolismo
9.
Ann Med ; 38(1): 2-10, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16448983

RESUMEN

Hypertriglyceridemia is an independent risk factor for the development of cardiovascular disease and is often associated with diabetes, inflammation and the metabolic syndrome. Recently, apolipoprotein A5 (APOA5) was identified as a novel member of the APOA1/C3/A4 gene cluster. Data from mice over-expressing or lacking APOA5 provide direct evidence that this apolipoprotein plays a role in triglyceride metabolism. Moreover, plasma triglyceride levels were found to be strongly associated with APOA5 polymorphisms. The human APOA5 gene is regulated by transcription factors known to affect triglyceride metabolism such as PPARa, RORa, LXR and SREBP-1c and this supports its function. Insulin and interleukins regulate APOA5 gene expression and provide novel clues for the role of this apolipoprotein. To date, the triglyceride lowering action of apoA-V is attributed to the activation of lipoprotein lipase and an acceleration of very low density lipoprotein catabolism. Recent findings indicate that APOA5 could also influence cholesterol homeostasis and probably play a role in hypertriglyceridemia associated with diabetes and inflammation. This review aims to give a comprehensive summary of the current literature and supports the view that APOA5 plays a relevant role in lipid metabolism.


Asunto(s)
Apolipoproteínas/genética , Regulación de la Expresión Génica , Hipertrigliceridemia/genética , Lipoproteínas VLDL/sangre , Triglicéridos/sangre , Animales , Apolipoproteína A-V , Apolipoproteínas A , Humanos , Hipertrigliceridemia/sangre , Polimorfismo Genético
10.
Arterioscler Thromb Vasc Biol ; 25(6): 1186-92, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15790933

RESUMEN

OBJECTIVE: The newly identified apolipoprotein A5 (APOA5), selectively expressed in the liver, is a crucial determinant of plasma triglyceride levels. Because elevated plasma triglyceride concentrations constitute an independent risk factor for cardiovascular diseases, it is important to understand how the expression of this gene is regulated. In the present study, we identified the retinoic acid receptor-related orphan receptor-alpha (RORalpha) as a regulator of human APOA5 gene expression. METHODS AND RESULTS: Using electromobility shift assays, we first demonstrated that RORalpha1 and RORalpha4 proteins can bind specifically to a direct repeat 1 site present at the position -272/-260 in the APOA5 gene promoter. In addition, using transient cotransfection experiments in HepG2 and HuH7 cells, we demonstrated that both RORalpha1 and RORalpha4 strongly increase APOA5 promoter transcriptional activity in a dose-dependent manner. Finally, adenoviral overexpression of hRORalpha in HepG2 cells led to enhanced hAPOA5 mRNA accumulation. We show that the homologous region in mouse apoa5 promoter is not functional. Moreover, we show that in staggerer mice, apoa5 gene is not affected by RORalpha. CONCLUSIONS: These findings identify RORalpha1 and RORalpha4 as transcriptional activators of human APOA5 gene expression. These data suggest an additional important physiological role for RORalpha in the regulation of genes involved in lipid homeostasis and probably in the development of atherosclerosis.


Asunto(s)
Apolipoproteínas/genética , Aterosclerosis/fisiopatología , Receptores de Superficie Celular/genética , Receptores Citoplasmáticos y Nucleares/genética , Transactivadores/genética , Activación Transcripcional/fisiología , Adenoviridae/genética , Animales , Apolipoproteína A-V , Apolipoproteínas A , Aterosclerosis/genética , Carcinoma Hepatocelular , Línea Celular Tumoral , Homeostasis/fisiología , Humanos , Neoplasias Hepáticas , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras , Receptores Huérfanos Similares al Receptor Tirosina Quinasa , Receptores de Superficie Celular/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transactivadores/metabolismo , Triglicéridos/sangre
11.
Mol Cell Biol ; 25(4): 1537-48, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15684402

RESUMEN

The apolipoprotein A5 gene (APOA5) has been repeatedly implicated in lowering plasma triglyceride levels. Since several studies have demonstrated that hyperinsulinemia is associated with hypertriglyceridemia, we sought to determine whether APOA5 is regulated by insulin. Here, we show that cell lines and mice treated with insulin down-regulate APOA5 expression in a dose-dependent manner. Furthermore, we found that insulin decreases human APOA5 promoter activity, and subsequent deletion and mutation analyses uncovered a functional E box in the promoter. Electrophoretic mobility shift and chromatin immunoprecipitation assays demonstrated that this APOA5 E box binds upstream stimulatory factors (USFs). Moreover, in transfection studies, USF1 stimulates APOA5 promoter activity, and the treatment with insulin reduced the binding of USF1/USF2 to the APOA5 promoter. The inhibition of the phosphatidylinositol 3-kinase (PI3K) pathway abolished insulin's effect on APOA5 gene expression, while the inhibition of the P70 S6 kinase pathway with rapamycin reversed its effect and increased APOA5 gene expression. Using an oligonucleotide precipitation assay for USF from nuclear extracts, we demonstrate that phosphorylated USF1 fails to bind to the APOA5 promoter. Taken together, these data indicate that insulin-mediated APOA5 gene transrepression could involve a phosphorylation of USFs through the PI3K and P70 S6 kinase pathways that modulate their binding to the APOA5 E box and results in APOA5 down-regulation. The effect of exogenous hyperinsulinemia in men showed a decrease in the plasma ApoAV level. These results suggest a potential contribution of the APOA5 gene in hypertriglyceridemia associated with hyperinsulinemia.


Asunto(s)
Apolipoproteínas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Insulina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Adulto , Animales , Apolipoproteína A-V , Apolipoproteínas A , Células Cultivadas , Cartilla de ADN/genética , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Proteínas Quinasas S6 Ribosómicas 70-kDa/antagonistas & inhibidores , Sirolimus/farmacología , Factores de Transcripción/metabolismo , Factores Estimuladores hacia 5'
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...