Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1789(5): 403-12, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19217949

RESUMEN

The transcription factor PAX6 plays an important role in transcriptional regulation of the peptide hormone glucagon from pancreatic alpha-cells. PAX6 contains two DNA binding domains, the paired domain (PD) and the homeodomain (HD). While the interaction of the PD with the PAX6 responsive elements G1 and G3 in the rat glucagon gene promoter is well understood, the role of the PAX6 HD for PAX6 binding and function on G1 and G3 remains unclear. In EMSA studies the PAX6 HD was found to be mandatory for PAX6 binding to G1 but not to G3. Transient transfections with luciferase reporter gene constructs revealed the HD to be critical for proper function of PAX6 on both, G1 and G3. Transfection data with variant promoter constructs and limited proteolysis assays demonstrated that the DNA sequence located 5' to the PD binding site plays an important role for PAX6 function and its conformation on the elements G1 and G3. Taken together, our data indicate a PH0-like binding of PAX6 to the glucagon promoter elements G1 and G3 where the HD binding site is abutted directly to the PD binding motif. The data suggest that the PH0-like binding induces a transcriptionally active conformation of PAX6.


Asunto(s)
Proteínas del Ojo/genética , Glucagón/genética , Proteínas de Homeodominio/genética , Factores de Transcripción Paired Box/genética , Proteínas Represoras/genética , Activación Transcripcional , Animales , Secuencia de Bases , Línea Celular , Interpretación Estadística de Datos , Proteínas del Ojo/metabolismo , Glucagón/metabolismo , Células Secretoras de Glucagón/fisiología , Proteínas de Homeodominio/metabolismo , Datos de Secuencia Molecular , Mutación , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Fragmentos de Péptidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Conformación Proteica , Ratas , Proteínas Represoras/metabolismo , Elementos de Respuesta , Alineación de Secuencia , Tripsina/metabolismo
2.
Mol Pharmacol ; 73(2): 509-17, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17962386

RESUMEN

The peptide hormone glucagon stimulates hepatic glucose output, and its levels in the blood are elevated in type 2 diabetes mellitus. The nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARgamma) has essential roles in glucose homeostasis, and thiazolidinedione PPARgamma agonists are clinically important antidiabetic drugs. As part of their antidiabetic effect, thiazolidinediones such as rosiglitazone have been shown to inhibit glucagon gene transcription through binding to PPARgamma and inhibition of the transcriptional activity of PAX6 that is required for cell-specific activation of the glucagon gene. However, how thiazolidinediones and PPARgamma inhibit PAX6 activity at the glucagon promoter remained unknown. After transient transfection of a glucagon promoter-reporter fusion gene into a glucagon-producing pancreatic islet alpha-cell line, ligand-bound PPARgamma was found in the present study to inhibit glucagon gene transcription also after deletion of its DNA-binding domain. Like PPARgamma ligands, also retinoid X receptor (RXR) agonists inhibited glucagon gene transcription in a PPARgamma-dependent manner. In glutathione transferase pull-down assays, the ligand-bound PPARgamma-RXR heterodimer bound to the transactivation domain of PAX6. This interaction depended on the presence of the ligand and RXR, but it was independent of the PPARgamma DNA-binding domain. Chromatin immunoprecipitation experiments showed that PPARgamma is recruited to the PAX6-binding proximal glucagon promoter. Taken together, the results of the present study support a model in which a ligand-bound PPARgamma-RXR heterodimer physically interacts with promoter-bound PAX6 to inhibit glucagon gene transcription. These data define PAX6 as a novel physical target of PPARgamma-RXR.


Asunto(s)
Proteínas del Ojo/metabolismo , Glucagón/antagonistas & inhibidores , Glucagón/genética , Proteínas de Homeodominio/metabolismo , PPAR gamma/metabolismo , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/metabolismo , Receptor gamma X Retinoide/metabolismo , Transcripción Genética/fisiología , Activación Transcripcional/fisiología , Animales , Línea Celular , Dimerización , Proteínas del Ojo/genética , Proteínas del Ojo/fisiología , Glucagón/biosíntesis , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Factor de Transcripción PAX6 , PPAR gamma/genética , PPAR gamma/fisiología , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/fisiología , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Receptor gamma X Retinoide/genética
3.
Biochem J ; 389(Pt 3): 831-41, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15828872

RESUMEN

The pancreatic islet hormone glucagon stimulates hepatic glucose production and thus maintains blood glucose levels in the fasting state. Transcription factors of the Foxa [Fox (forkhead box) subclass A; also known as HNF-3 (hepatocyte nuclear factor-3)] family are required for cell-specific activation of the glucagon gene in pancreatic islet alpha-cells. However, their action on the glucagon gene is poorly understood. In the present study, comparative sequence analysis and molecular characterization using protein-DNA binding and transient transfection assays revealed that the well-characterized Foxa-binding site in the G2 enhancer element of the rat glucagon gene is not conserved in humans and that the human G2 sequence lacks basal enhancer activity. A novel Foxa site was identified that is conserved in rats, mice and humans. It mediates activation of the glucagon gene by Foxa proteins and confers cell-specific promoter activity in glucagon-producing pancreatic islet alpha-cell lines. In contrast with previously identified Foxa-binding sites in the glucagon promoter, which bind nuclear Foxa2, the novel Foxa site was found to bind preferentially Foxa1 in nuclear extracts of a glucagon-producing pancreatic islet alpha-cell line, offering a mechanism that explains the decrease in glucagon gene expression in Foxa1-deficient mice. This site is located just upstream of the TATA box (between -30 and -50), suggesting a role for Foxa proteins in addition to direct transcriptional activation, such as a role in opening the chromatin at the start site of transcription of the glucagon gene.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Glucagón/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Secuencia Conservada , Regulación de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito , Humanos , Ratones , Datos de Secuencia Molecular , Mutación , Regiones Promotoras Genéticas/fisiología , Ratas , Homología de Secuencia de Ácido Nucleico
4.
Elife ; 3: e03640, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25247702

RESUMEN

Translational readthrough gives rise to low abundance proteins with C-terminal extensions beyond the stop codon. To identify functional translational readthrough, we estimated the readthrough propensity (RTP) of all stop codon contexts of the human genome by a new regression model in silico, identified a nucleotide consensus motif for high RTP by using this model, and analyzed all readthrough extensions in silico with a new predictor for peroxisomal targeting signal type 1 (PTS1). Lactate dehydrogenase B (LDHB) showed the highest combined RTP and PTS1 probability. Experimentally we show that at least 1.6% of the total cellular LDHB is targeted to the peroxisome by a conserved hidden PTS1. The readthrough-extended lactate dehydrogenase subunit LDHBx can also co-import LDHA, the other LDH subunit, into peroxisomes. Peroxisomal LDH is conserved in mammals and likely contributes to redox equivalent regeneration in peroxisomes.


Asunto(s)
L-Lactato Deshidrogenasa/metabolismo , Mamíferos/metabolismo , Peroxisomas/enzimología , Biosíntesis de Proteínas , Animales , Secuencia de Bases , Línea Celular , Codón de Terminación/genética , Simulación por Computador , Secuencia Conservada , Genes Reporteros , Genoma , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/genética , Datos de Secuencia Molecular , Señales de Clasificación de Proteína , Transporte de Proteínas , Ratas
5.
Cell Signal ; 26(9): 1792-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24726898

RESUMEN

Insulin biosynthesis is an essential ß-cell function and inappropriate insulin secretion and biosynthesis contribute to the pathogenesis of diabetes mellitus type 2. Previous studies showed that the dual leucine zipper kinase (DLK) induces ß-cell apoptosis. Since ß-cell dysfunction precedes ß-cell loss, in the present study the effect of DLK on insulin gene transcription was investigated in the HIT-T15 ß-cell line. Downregulation of endogenous DLK increased whereas overexpression of DLK decreased human insulin gene transcription. 5'- and 3'-deletion human insulin promoter analyses resulted in the identification of a DLK responsive element that mapped to the DNA binding-site for the ß-cell specific transcription factor MafA. Overexpression of DLK wild-type but not its kinase-dead mutant inhibited MafA transcriptional activity conferred by its transactivation domain. Furthermore, in the non-ß-cell line JEG DLK inhibited MafA overexpression-induced human insulin promoter activity. Overexpression of MafA and DLK or its kinase-dead mutant into JEG cells revealed that DLK but not its mutant reduced MafA protein content. Inhibition of the down-stream DLK kinase c-Jun N-terminal kinase (JNK) by SP600125 attenuated DLK-induced MafA loss. Furthermore, mutation of the serine 65 to alanine, shown to confer MafA protein stability, increased MafA-dependent insulin gene transcription and prevented DLK-induced MafA loss in JEG cells. These data suggest that DLK by activating JNK triggers the phosphorylation and degradation of MafA thereby attenuating insulin gene transcription. Given the importance of MafA for ß-cell function, the inhibition of DLK might preserve ß-cell function and ultimately retard the development of diabetes mellitus type 2.


Asunto(s)
Regulación de la Expresión Génica , Insulina/genética , Insulina/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Antracenos/farmacología , Línea Celular , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/genética , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Interferencia de ARN
6.
Mol Pharmacol ; 63(6): 1289-95, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12761338

RESUMEN

Cyclosporin A and tacrolimus are clinically important immunosuppressive drugs. They share a diabetogenic action as one of their most serious adverse effects. The underlying mechanism is unknown. Previous studies have shown that tacrolimus can inhibit insulin gene transcription at high concentrations in tumor cell lines. To study insulin gene transcription in normal, mature pancreatic islet cells, we used a novel approach in the present study. Transgenic mice that carry a human insulin promoter-reporter gene were generated. The human insulin promoter directed transcription in pancreatic islets and conferred a normal, physiological glucose response to reporter gene expression in isolated islets. After stimulation with glucose, human insulin promoter-mediated gene expression was inhibited in normal, mature islet cells by both tacrolimus and cyclosporin A to a large extent (approximately 70%) and with high potency at concentrations that are known to inhibit calcineurin phosphatase activity (IC50 values of 1 and 35 nM, respectively). Furthermore, glucose stimulated calcineurin phosphatase activity in mouse pancreatic islets, further supporting the view that calcineurin phosphatase activity is an essential part of glucose signaling to the human insulin gene. The high potency of cyclosporin A and tacrolimus in normal islets suggests that inhibition of insulin gene transcription by cyclosporin A and tacrolimus is clinically important and is one mechanism of the diabetogenic effect of these immunosuppressive drugs.


Asunto(s)
Ciclosporina/farmacología , Inmunosupresores/farmacología , Insulina/genética , Islotes Pancreáticos/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Tacrolimus/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Expresión Génica/efectos de los fármacos , Genes Reporteros , Glucosa/farmacología , Humanos , Islotes Pancreáticos/fisiología , Ratones , Ratones Transgénicos , Monoéster Fosfórico Hidrolasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA