Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Traffic ; 13(1): 54-69, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21985333

RESUMO

The brain-spliced isoform of Myosin Va (BR-MyoVa) plays an important role in the transport of dense core secretory granules (SGs) to the plasma membrane in hormone and neuropeptide-producing cells. The molecular composition of the protein complex that recruits BR-MyoVa to SGs and regulates its function has not been identified to date. We have identified interaction between SG-associated proteins granuphilin-a/b (Gran-a/b), BR-MyoVa and Rab27a, a member of the Rab family of GTPases. Gran-a/b-BR-MyoVa interaction is direct, involves regions downstream of the Rab27-binding domain, and the C-terminal part of Gran-a determines exon specificity. MyoVa and Gran-a/b are partially colocalised on SGs and disruption of Gran-a/b-BR-MyoVa binding results in a perinuclear accumulation of SGs which augments nutrient-stimulated hormone secretion in pancreatic beta-cells. These results indicate the existence of at least another binding partner of BR-MyoVa that was identified as rabphilin-3A (Rph-3A). BR-MyoVa-Rph-3A interaction is also direct and enhanced when secretion is activated. The BR-MyoVa-Rph-3A and BR-MyoVa-Gran-a/b complexes are linked to a different subset of SGs, and simultaneous inhibition of these complexes nearly completely blocks stimulated hormone release. This study demonstrates that multiple binding partners of BR-MyoVa regulate SG transport, and this molecular mechanism is universally used by neuronal, endocrine and neuroendocrine cells.


Assuntos
Membrana Celular/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Vesículas Secretórias/metabolismo , Animais , Encéfalo/metabolismo , Hormônios/metabolismo , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Insulinoma/patologia , Cadeias Pesadas de Miosina/genética , Miosina Tipo V/genética , Células PC12 , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Ligação Proteica , Isoformas de Proteínas , Transporte Proteico , Ratos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP
2.
J Biol Chem ; 286(51): 44005-44014, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22065581

RESUMO

PAS kinase (PASK) is a glucose-regulated protein kinase involved in the control of pancreatic islet hormone release and insulin sensitivity. We aimed here to identify mutations in the PASK gene that may be associated with young-onset diabetes in humans. We screened 18 diabetic probands with unelucidated maturity-onset diabetes of the young (MODY). We identified two rare nonsynonymous mutations in the PASK gene (p.L1051V and p.G1117E), each of which was found in a single MODY family. Wild type or mutant PASKs were expressed in HEK 293 cells. Kinase activity of the affinity-purified proteins was assayed as autophosphorylation at amino acid Thr307 or against an Ugp1p-derived peptide. Whereas the PASK p.G1117E mutant displayed a ∼25% increase with respect to wild type PASK in the extent of autophosphorylation, and a ∼2-fold increase in kinase activity toward exogenous substrates, the activity of the p.L1051V mutant was unchanged. Amino acid Gly1117 is located in an α helical region opposing the active site of PASK and may elicit either: (a) a conformational change that increases catalytic efficiency or (b) a diminished inhibitory interaction with the PAS domain. Mouse islets were therefore infected with adenoviruses expressing wild type or mutant PASK and the regulation of insulin secretion was examined. PASK p.G1117E-infected islets displayed a 4-fold decrease in glucose-stimulated (16.7 versus 3 mM) insulin secretion, chiefly reflecting a 4.5-fold increase in insulin release at low glucose. In summary, we have characterized a rare mutation (p.G1117E) in the PASK gene from a young-onset diabetes family, which modulates glucose-stimulated insulin secretion.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Adulto , Animais , Linhagem Celular , Diabetes Mellitus/metabolismo , Genômica , Glucagon/metabolismo , Células HEK293 , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Secreção de Insulina , Masculino , Proteínas de Membrana/metabolismo , Modelos Genéticos , Mutagênese , Fosforilação , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo
3.
Diabetes ; 53 Suppl 3: S84-91, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15561928

RESUMO

Accumulation of triglyceride in islets may contribute to the loss of glucose-stimulated insulin secretion (GSIS) in some forms of type 2 diabetes (Diraison et al., Biochem J 373:769-778, 2004). Here, we use adenoviral vectors and oligonucleotide microarrays to determine the effects of the forced expression of SREBP1c on the gene expression profile of rat islets. Sterol regulatory element binding protein-1c (SREBP1c) overexpression led to highly significant (P <0.1 with respect to null adenovirus) changes in the expression of 1,238 genes or expressed sequence tags, of which 1,180 (95.3%) were upregulated. By contrast, overexpression of constitutively active AMP-activated protein kinase (AMPK), expected to promote lipolysis, altered the expression of 752 genes, of which 702 (93%) were upregulated. To identify specific targets for SREBP1c or AMPK, we eliminated messages that were 1) affected in the same direction by the expression of either protein, 2) changed by less than twofold, or 3) failed a positive false discovery test; 206 SREBP1c-regulated genes (195; 95% upregulated) and 48 AMPK-regulated genes (33; 69% upregulated) remained. As expected, SREBP1c-induced genes included those involved in cholesterol (6), fatty acid (3), and eicosanoid synthesis. Interestingly, somatostatin receptor (sstr1) expression was increased by SREBP1c, whereas AMPK induced the expression of peptide YY, the early endocrine pancreas marker.


Assuntos
Adenilato Quinase/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Ilhotas Pancreáticas/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Transcrição/metabolismo , Adenoviridae , Adenilato Quinase/genética , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Etiquetas de Sequências Expressas , Regulação da Expressão Gênica , Zíper de Leucina , Masculino , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína de Ligação a Elemento Regulador de Esterol 1 , Fatores de Transcrição/genética , Transcrição Gênica , Transfecção
4.
Diabetes ; 51(8): 2536-45, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12145168

RESUMO

Acetyl-CoA carboxylase I (ACCI) is a key lipogenic enzyme whose induction in islet beta-cells may contribute to glucolipotoxicity. Here, we provide evidence that enhanced insulin release plays an important role in the activation of this gene by glucose. Glucose (30 vs. 3 mmol/l) increased ACCI mRNA levels approximately 4-fold and stimulated ACCI (pII) promoter activity >30-fold in MIN6 cells. The latter effect was completely suppressed by blockade of insulin release or of insulin receptor signaling. However, added insulin substantially, but not completely, mimicked the effects of glucose, suggesting that intracellular metabolites of glucose may also contribute to transcriptional stimulation. Mutational analysis of the ACCI promoter, and antibody microinjection, revealed that the effect of glucose required sterol response element binding protein (SREBP)-1c. Moreover, adenoviral transduction with dominant-negative-acting SREBP1c blocked ACCI gene induction, whereas constitutively active SREBP1c increased ACCI mRNA levels. Finally, glucose also stimulated SREBP1c transcription, although this effect was independent of insulin release. These data suggest that glucose regulates ACCI gene expression in the beta-cell by complex mechanisms that may involve the covalent modification of SREBP1c. However, overexpression of SREBP1c also decreased glucose-stimulated insulin release, implicating SREBP1c induction in beta-cell lipotoxicity in some forms of type 2 diabetes.


Assuntos
Acetil-CoA Carboxilase/genética , Acetiltransferases/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Insulina/metabolismo , Animais , Linhagem Celular , Vetores Genéticos , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Cloreto de Potássio/farmacologia , Regiões Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1 , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção , Verapamil/farmacologia
5.
Biochem J ; 378(Pt 3): 769-78, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-14690455

RESUMO

Accumulation of intracellular lipid by pancreatic islet beta-cells has been proposed to inhibit normal glucose-regulated insulin secretion ('glucolipotoxicity'). In the present study, we determine whether over-expression in rat islets of the lipogenic transcription factor SREBP1c (sterol-regulatory-element-binding protein-1c) affects insulin release, and whether changes in islet lipid content may be reversed by activation of AMPK (AMP-activated protein kinase). Infection with an adenovirus encoding the constitutively active nuclear fragment of SREBP1c resulted in expression of the protein in approx. 20% of islet cell nuclei, with a preference for beta-cells at the islet periphery. Real-time PCR (TaqMan) analysis showed that SREBP1c up-regulated the expression of FAS (fatty acid synthase; 6-fold), acetyl-CoA carboxylase-1 (2-fold), as well as peroxisomal-proliferator-activated receptor-gamma (7-fold), uncoupling protein-2 (1.4-fold) and Bcl2 (B-cell lymphocytic-leukaemia proto-oncogene 2; 1.3-fold). By contrast, levels of pre-proinsulin, pancreatic duodenal homeobox-1, glucokinase and GLUT2 (glucose transporter isoform-2) mRNAs were unaltered. SREBP1c-transduced islets displayed a 3-fold increase in triacylglycerol content, decreased glucose oxidation and ATP levels, and a profound inhibition of glucose-, but not depolarisation-, induced insulin secretion. Culture of islets with the AMPK activator 5-amino-4-imidazolecarboxamide riboside decreased the expression of the endogenous SREBP1c and FAS genes, and reversed the effect of over-expressing active SREBP1c on FAS mRNA levels and cellular triacylglycerol content. We conclude that SREBP1c over-expression, even when confined to a subset of beta-cells, leads to defective insulin secretion from islets and may contribute to some forms of Type II diabetes.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Ligação a DNA/metabolismo , Glucose/antagonistas & inibidores , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Lipídeos/biossíntese , Ribonucleotídeos/farmacologia , Fatores de Transcrição , Acetiltransferases/biossíntese , Acetiltransferases/genética , Adenoviridae/genética , Animais , Apoptose , Proteínas Estimuladoras de Ligação a CCAAT/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Ácido Graxo Sintases/biossíntese , Ácido Graxo Sintases/genética , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Proteína de Ligação a Elemento Regulador de Esterol 1 , Transdução Genética , Triglicerídeos/metabolismo
6.
Am J Clin Nutr ; 77(3): 559-64, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12600843

RESUMO

BACKGROUND: A high-carbohydrate, low-fat diet is recommended for the prevention of atherosclerosis, because it reduces plasma cholesterol concentrations. However, such a diet can increase plasma triacylglycerol concentrations--an undesirable side effect. The addition of nondigestible carbohydrate could reduce the risk of elevated triacylglycerol concentrations. OBJECTIVE: The objective was to determine whether the addition of a moderate dose of inulin to a moderately high-carbohydrate diet would decrease hepatic lipogenesis and plasma triacylglycerol concentrations and have a cholesterol-lowering action. DESIGN: Eight healthy subjects were studied twice in a double-blind, randomized, placebo-controlled crossover study after consuming for 3 wk a moderately high-carbohydrate, low-fat diet (55% of total energy) plus an oral placebo or 10 g high-performance inulin/d. Hepatic lipogenesis and cholesterol synthesis (deuterated water method), plasma lipid concentrations, fatty acid synthase, acetyl-CoA carboxylase 1, and sterol responsive element binding protein 1c messenger RNA concentrations were measured in adipose tissue at the end of the 2 diet periods. RESULTS: Plasma triacylglycerol concentrations and hepatic lipogenesis were lower after inulin than after placebo ingestion (P < 0.05), but cholesterol synthesis and plasma cholesterol concentrations were not significantly different between the 2 groups. None of the adipose tissue messenger RNA concentrations changed significantly after inulin ingestion. CONCLUSIONS: The addition of high-performance inulin to a moderately high-carbohydrate, low-fat diet has a beneficial effect on plasma lipids by decreasing hepatic lipogenesis and plasma triacylglycerol concentrations. These results support the use of nondigestible carbohydrate for reducing risk factors for atherosclerosis.


Assuntos
Colesterol/sangue , Carboidratos da Dieta/administração & dosagem , Inulina/administração & dosagem , Fígado/metabolismo , Triglicerídeos/sangue , Adulto , Anticolesterolemiantes/administração & dosagem , Arteriosclerose/etiologia , Arteriosclerose/prevenção & controle , Água Corporal/metabolismo , Colesterol/biossíntese , Estudos Cross-Over , Deutério , Inquéritos sobre Dietas , Método Duplo-Cego , Feminino , Humanos , Lipídeos/biossíntese , Masculino , Placebos , RNA Mensageiro , Fatores de Risco
7.
Mol Biol Cell ; 23(22): 4444-55, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22993210

RESUMO

Myosin- and Rab-interacting protein (MyRIP), which belongs to the protein kinase A (PKA)-anchoring family, is implicated in hormone secretion. However, its mechanism of action is not fully elucidated. Here we investigate the role of MyRIP in myosin Va (MyoVa)-dependent secretory granule (SG) transport and secretion in pancreatic beta cells. These cells solely express the brain isoform of MyoVa (BR-MyoVa), which is a key motor protein in SG transport. In vitro pull-down, coimmunoprecipitation, and colocalization studies revealed that MyRIP does not interact with BR-MyoVa in glucose-stimulated pancreatic beta cells, suggesting that, contrary to previous notions, MyRIP does not link this motor protein to SGs. Glucose-stimulated insulin secretion is augmented by incretin hormones, which increase cAMP levels and leads to MyRIP phosphorylation, its interaction with BR-MyoVa, and phosphorylation of the BR-MyoVa receptor rabphilin-3A (Rph-3A). Rph-3A phosphorylation on Ser-234 was inhibited by small interfering RNA knockdown of MyRIP, which also reduced cAMP-mediated hormone secretion. Demonstrating the importance of this phosphorylation, nonphosphorylatable and phosphomimic Rph-3A mutants significantly altered hormone release when PKA was activated. These data suggest that MyRIP only forms a functional protein complex with BR-MyoVa on SGs when cAMP is elevated and under this condition facilitates phosphorylation of SG-associated proteins, which in turn can enhance secretion.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Células Secretoras de Insulina/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Vesículas Secretórias/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Células Cultivadas , Glucose/farmacologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Modelos Biológicos , Fosforilação , Ratos
8.
J Lipid Res ; 49(4): 814-22, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18178930

RESUMO

Previous studies have reported both positive and negative effects of culture of islets at high glucose concentrations on regulated insulin secretion. Here, we have reexamined this question in mouse islets and determined the role of changes in lipid synthesis in the effects of glucose. Glucose-stimulated insulin secretion (GSIS) and gene expression were examined in islets from C57BL/6 mice or littermates deleted for sterol-regulatory element binding protein-1 (SREBP1) after 4 days of culture at high glucose concentrations. Culture of control islets at 30 versus 8 mmol/l glucose led to enhanced secretion at both basal (3 mmol/l) and stimulatory (17 mmol/l) glucose concentrations and to enhanced triacylglycerol accumulation. These changes were associated with increases in the expression of genes involved in glucose sensing (glucose transporter 2, glucokinase, sulfonylurea receptor 1, inwardly rectifying K(+) channel 6.2), differentiation (pancreatic duodenal homeobox 1), and lipogenesis (Srebp1, fatty acid synthase, acetyl-coenzyme A carboxylase 1, stearoyl-coenzyme A desaturase 1). When cultured at either 8 or 30 mmol/l glucose, SREBP1-deficient (SREBP1(-/-)) islets displayed reduced GSIS and triacylglycerol content compared with normal islets. Correspondingly, glucose induction of the above genes in control islets was no longer observed in SREBP1(-/-) mouse islets. We conclude that enhanced lipid synthesis mediated by SREBP1c-dependent genes is required for the adaptive changes in islet gene expression and insulin secretion at high glucose concentrations.


Assuntos
Regulação da Expressão Gênica , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína de Ligação a Elemento Regulador de Esterol 1/deficiência , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Triazenos/farmacologia , Triglicerídeos/metabolismo
9.
Diabetes ; 57(2): 415-23, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18003756

RESUMO

OBJECTIVE: Apoptotic destruction of insulin-producing pancreatic beta-cells is involved in the etiology of both type 1 and type 2 diabetes. AMP-activated protein kinase (AMPK) is a sensor of cellular energy charge whose sustained activation has recently been implicated in pancreatic beta-cell apoptosis and in islet cell death posttransplantation. Here, we examine the importance of beta-cell AMPK in cytokine-induced apoptosis and in the cytotoxic action of CD8(+) T-cells. RESEARCH DESIGN AND METHODS: Clonal MIN6 beta-cells or CD1 mouse pancreatic islets were infected with recombinant adenoviruses encoding enhanced green fluorescent protein (eGFP/null), constitutively active AMPK (AMPK-CA), or dominant-negative AMPK (AMPK-DN) and exposed or not to tumor necrosis factor-alpha, interleukin-1beta, and interferon-gamma. Apoptosis was detected by monitoring the cleavage of caspase-3 and DNA fragmentation. The cytotoxic effect of CD8(+) purified T-cells was examined against pancreatic islets from NOD mice infected with either null or the AMPK-DN-expressing adenoviruses. RESULTS: Exposure to cytokines, or expression of AMPK-CA, induced apoptosis in clonal MIN6 beta-cells and CD1 mouse pancreatic islets. By contrast, overexpression of AMPK-DN protected against the proapoptotic effect of these agents, in part by preventing decreases in cellular ATP, and lowered the cytotoxic effect of CD8(+) T-cells toward NOD mouse islets. CONCLUSIONS: Inhibition of AMPK activity enhances islet survival in the face of assault by either cytokines or T-cells. AMPK may therefore represent an interesting therapeutic target to suppress immune-mediated beta-cell destruction and may increase the efficacy of islet allografts in type 1 diabetes.


Assuntos
Apoptose/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Células Secretoras de Insulina/fisiologia , Complexos Multienzimáticos/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/imunologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocinas/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo
10.
Am J Physiol Endocrinol Metab ; 292(5): E1340-7, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17227960

RESUMO

H(2)O administration has recently been proposed as a simple and convenient method to measure protein synthesis rates. (2)H(2)O administration results in deuterium labeling of free amino acids such as alanine, and incorporation into proteins of labeled alanine can then be used to measure protein synthesis rates. We examined first whether during (2)H(2)O administration plasma free alanine enrichment is a correct estimate of the enrichment in the tissue amino acid pools used for protein synthesis. We found that, after (2)H(2)O administration, deuterium labeling in plasma free alanine equilibrated rapidly with body water, and stable enrichment values were obtained within 20 min. Importantly, oral administration of (2)H(2)O induced no difference of labeling between portal and peripheral circulation except for the initial 10 min after a loading dose. The kinetics of free alanine labeling were comparable in various tissues (liver, skeletal muscle, heart) and in plasma with identical plateau values. We show next that increased glycolytic rate or absorption of unlabeled amino acids from ingested meals do not modify alanine labeling. Calculated synthesis rates of mixed proteins were much higher (20- to 70-fold) in plasma and liver than in muscle and heart. Last, comparable replacement rates of apoB100-VLDL were obtained in humans by using the kinetics of incorporation into apoB100 of infused labeled leucine or of alanine labeled by (2)H(2)O administration. All of these results support (2)H(2)O as a safe, reliable, useful, and convenient tracer for studies of protein synthesis, including proteins with slow turnover rate.


Assuntos
Alanina/metabolismo , Apolipoproteína B-100/metabolismo , Óxido de Deutério/metabolismo , Receptores de LDL/metabolismo , Adulto , Alanina/química , Animais , Apolipoproteína B-100/química , Óxido de Deutério/química , Feminino , Humanos , Cinética , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Biossíntese Peptídica , Ratos , Ratos Wistar , Receptores de LDL/química
11.
J Lipid Res ; 47(11): 2482-91, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16891625

RESUMO

Pancreatic beta-cell dysfunction is central to the pathogenesis of type 2 diabetes and may involve secretory failure through glucolipotoxity. The relative importance of the transcription factors carbohydrate-responsive element binding protein (ChREBP), sterol-responsive element binding protein-1c (SREBP-1c), and upstream stimulatory factor (USF) in the induction of lipogenic genes by glucose remains unclear. By confocal imaging, we show that ChREBP translocates to the nucleus in MIN6 beta cells in response to glucose. Both ChREBP and SREBP-1c were required for the induction of the fatty acid synthase (FAS) promoter by glucose, and chromatin immunoprecipitation (ChIP) assay revealed that glucose induced the binding of both ChREBP and SREBP-1c to the FAS promoter without affecting USF2 binding. By contrast, ChIP assay revealed that high glucose prompted direct binding of ChREBP, but not SREBP-1c or USF2, to the liver-type pyruvate kinase (L-PK) promoter. This event was indispensable for the induction of the L-PK gene by glucose, as demonstrated by RNA silencing, single-cell promoter analysis, and quantitative real-time PCR. We conclude that ChREBP is a critical regulator of lipogenic genes in the beta cell and may play a role in the development of glucolipotoxicity and beta cell failure through alteration of gene expression in type 2 diabetes.


Assuntos
Ácido Graxo Sintases/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Nucleares/fisiologia , Piruvato Quinase/metabolismo , Fatores de Transcrição/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Apoptose , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Inativação Gênica , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição/metabolismo , Triglicerídeos/metabolismo
12.
Am J Physiol Endocrinol Metab ; 291(5): E982-94, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16772326

RESUMO

Accumulation of intracellular lipid may contribute to defective insulin secretion in type 2 diabetes. Although Zucker diabetic fatty (ZDF; fa/fa) rat islets are fat-laden and overexpress the lipogenic master gene, sterol regulatory element binding protein 1c (SREBP-1c), the contribution of SREBP-1c to the secretory defects observed in this model remains unclear. Here we compare the gene expression profile of lean control (fa/+) and ZDF rat islets in the absence or presence of dominant-negative SREBP-1c (SREBP-1c DN). ZDF islets displayed elevated basal insulin secretion at 3 mmol/l glucose but a severely depressed response to 17 mmol/l glucose. While SREBP-1c DN reduced basal insulin secretion from ZDF islets, glucose-stimulated insulin secretion was not improved. Of 57 genes differentially regulated in ZDF islets and implicated in glucose metabolism, vesicle trafficking, ion fluxes, and/or exocytosis, 21 were upregulated and 5 were suppressed by SREBP-1c DN. Genes underrepresented in ZDF islets were either unaffected (Glut-2, Kir6.2, Rab3), stimulated (voltage-dependent Ca(2+) channel subunit alpha1D, CPT2, SUR2, rab9, syt13), or inhibited (syntaxin 7, secretogranin-2) by SREBP-1c inhibition. Correspondingly, SREBP-1c DN largely corrected decreases in the expression of the transcription factors Pdx-1 and MafA but did not affect the abnormalities in Pax6, Arx, hepatic nuclear factor-1alpha (HNF1alpha), HNF3beta/Forkhead box-a2 (Foxa2), inducible cyclic AMP early repressor (ICER), or transcription factor 7-like 2 (TCF7L2) expression observed in ZDF islets. We conclude that upregulation of SREBP-1c and mild increases in triglyceride content do not explain defective glucose-stimulated insulin secretion from ZDF rats. However, overexpression of SREBP-1c may contribute to enhanced basal insulin secretion in this model.


Assuntos
Perfilação da Expressão Gênica , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Obesidade/genética , Obesidade/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal/fisiologia , Cálcio/metabolismo , Insulina/metabolismo , Secreção de Insulina , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Zucker , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
13.
J Lipid Res ; 44(4): 846-53, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12562844

RESUMO

We assessed the contributions of human liver and adipose tissue de novo lipogenesis (DNL) to triacylglycerol (TAG) synthesis. Volunteers were fed a high-energy, high-carbohydrate diet (HC, n = 5) or a normocaloric diet (NC, n = 10). NC subjects remained in the fasting state (Study 1, n = 5) or received oral glucose (Study 2, n = 5) throughout the test (12 h). HC subjects remained in the fasting state (Study 3). They ingested deuterated water and [U-13C]acetate to trace lipogenesis. Adipose tissue fatty-acid (FA) synthase (FAS), acetyl-CoA carboxylase 1 (ACC1), and SREBP-1c mRNA were measured. Plasma TAG-FA was labeled by 13C and deuterium showing active liver lipogenesis, which was stimulated (P < 0.05) by oral glucose and HC diet. Adipose tissue TAG had no detectable 13C enrichment in any test, showing no significant incorporation of TAG-FA provided by liver lipogenesis, but were labeled by deuterium in all tests, showing active DNL in situ; however, rough quantitative estimates showed that adipose DNL was minimal (<1 g), and poorly stimulated by oral glucose or HC diet. mRNA levels were not increased by the HC diet. Adipose DNL is active in humans, but contributes little to TAG stores and is less responsive than liver DNL to stimulation by carbohydrates.


Assuntos
Tecido Adiposo/metabolismo , Carboidratos/farmacologia , Regulação da Expressão Gênica , Lipídeos/biossíntese , Fígado/metabolismo , Acetil-CoA Carboxilase/genética , Adolescente , Adulto , Proteínas Estimuladoras de Ligação a CCAAT/genética , Carboidratos/administração & dosagem , Proteínas de Ligação a DNA/genética , Ácido Graxo Sintases/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/análise , Proteína de Ligação a Elemento Regulador de Esterol 1 , Fatores de Transcrição/genética
14.
Am J Physiol Endocrinol Metab ; 282(1): E46-51, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11739082

RESUMO

To determine whether increased lipogenesis contributes to human obesity, we measured (postabsorptive state), in lean and obese subjects, lipid synthesis (deuterated water method) and the mRNA concentration (RT-competitive PCR) in subcutaneous adipose tissue of fatty acid synthase (FAS) and sterol regulatory element-binding protein (SREBP)-1c. Before energy restriction, obese subjects had an increased contribution of hepatic lipogenesis to the circulating triglyceride pool (14.5 +/- 1.3 vs. 7.5 +/- 1.9%, P < 0.01) without enhancement of cholesterol synthesis. This increased hepatic lipogenesis represented an excess of 2-5 g/day of triglycerides, which would represent 0.7-1.8 kg on a yearly basis. The lipogenic capacity of adipose tissue appeared, on the contrary, decreased with lower FAS mRNA levels (P < 0.01) and a trend for decreased SREBP-1c mRNA (P = 0.06). Energy restriction in obese patients decreased plasma insulin (P < 0.05) and leptin (P < 0.05) and normalized hepatic lipogenesis. FAS mRNA levels were unchanged, whereas SREBP-1c increased. In conclusion, subjects with established obesity have an increased hepatic lipogenesis that could contribute to their excessive fat mass but no evidence for an increased lipogenic capacity of adipose tissue.


Assuntos
Tecido Adiposo/fisiopatologia , Expressão Gênica , Lipídeos/biossíntese , Fígado/metabolismo , Obesidade/genética , Obesidade/metabolismo , Fatores de Transcrição , Tecido Adiposo/metabolismo , Adulto , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas de Ligação a DNA/genética , Ácido Graxo Sintases/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Valores de Referência , Proteína de Ligação a Elemento Regulador de Esterol 1 , Magreza
15.
Cell Biochem Biophys ; 40(3 Suppl): 179-90, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15289653

RESUMO

The mechanisms by which changes in glucose concentration regulate gene expression and insulin secretion in pancreatic islet beta-cells are only partly understood. Here we describe the development of new technologies for examining these processes at the level of single living beta-cells. We also present recent findings, made using these and other techniques, which implicate a role for adenosine 5'-monophosphate-activated protein kinase in glucose signaling in these cells.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Perfilação da Expressão Gênica , Glucose/metabolismo , Células Secretoras de Insulina/citologia , Insulina/metabolismo , Animais , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Humanos , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos , Microscopia de Fluorescência/métodos , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Fatores de Tempo
16.
Am J Physiol Endocrinol Metab ; 287(3): E390-404, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15126236

RESUMO

Peroxisome proliferator-activated receptor-gamma (PPARgamma) serves as a target for the thiazolidinedione class of antidiabetic drugs and is an important regulator of adipose tissue differentiation. By contrast, the principal target genes for PPARgamma in the pancreatic islet and the impact of their induction on insulin secretion are largely undefined. Here, we show that mRNAs encoding both isoforms of rodent PPARgamma, gamma1 and gamma2, are expressed in primary rat islets and are upregulated by overexpresssion of the lipogenic transcription factor sterol response element-binding protein 1c. Unexpectedly, however, oligonucleotide microarray analysis demonstrates that graded activation of PPARgamma achieved with 1) the thiazolidinedione GW-347845, 2) transduction with adenoviral PPARgamma1, or 3) a combination of both treatments progressively enhances the expression of genes involved in fatty acid oxidation and transport. Moreover, maximal activation of PPARgamma1 reduces islet triglyceride levels and enhances the oxidation of exogenous palmitate while decreasing glucose oxidation, cellular ATP content, and glucose-, but not depolarization-stimulated, insulin secretion. We conclude that, in the context of the pancreatic islet, the principal response to PPARgamma expression and activation is the activation of genes involved in the disposal, rather than the synthesis, of fatty acids. Although fatty acid oxidation may have beneficial effects on beta-cell function in the longer term by countering beta-cell "lipotoxicity," the acute response to this metabolic shift is a marked inhibition of insulin secretion.


Assuntos
Perfilação da Expressão Gênica , Expressão Gênica/fisiologia , Ilhotas Pancreáticas/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ácidos Graxos/metabolismo , Glucose/metabolismo , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Cloreto de Potássio/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1 , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA