Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 12: 645817, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33841427

RESUMEN

T-cell responses to insulin and its precursor proinsulin are central to islet autoimmunity in humans and non-obese diabetic (NOD) mice that spontaneously develop autoimmune diabetes. Mice have two proinsulin genes proinsulin -1 and 2 that are differentially expressed, with predominant proinsulin-2 expression in the thymus and proinsulin-1 in islet beta-cells. In contrast to proinsulin-2, proinsulin-1 knockout NOD mice are protected from autoimmune diabetes. This indicates that proinsulin-1 epitopes in beta-cells maybe preferentially targeted by autoreactive T cells. To study the contribution of proinsulin-1 reactive T cells in autoimmune diabetes, we generated transgenic NOD mice with tetracycline-regulated expression of proinsulin-1 in antigen presenting cells (TIP-1 mice) with an aim to induce immune tolerance. TIP-1 mice displayed a significantly reduced incidence of spontaneous diabetes, which was associated with reduced severity of insulitis and insulin autoantibody development. Antigen experienced proinsulin specific T cells were significantly reduced in in TIP-1 mice indicating immune tolerance. Moreover, T cells from TIP-1 mice expressing proinsulin-1 transferred diabetes at a significantly reduced frequency. However, proinsulin-1 expression in APCs had minimal impact on the immune responses to the downstream antigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) and did not prevent diabetes in NOD 8.3 mice with a pre-existing repertoire of IGRP reactive T cells. Thus, boosting immune tolerance to proinsulin-1 partially prevents islet-autoimmunity. This study further extends the previously established role of proinsulin-1 epitopes in autoimmune diabetes in NOD mice.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Tolerancia Inmunológica , Proinsulina/fisiología , Animales , Autoanticuerpos/análisis , Glucosa-6-Fosfatasa/fisiología , Insulina/inmunología , Ratones , Ratones Endogámicos NOD , Proinsulina/genética , Proinsulina/inmunología
2.
Endocrinology ; 157(8): 3002-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27300767

RESUMEN

The glucose-6-phosphatase catalytic 2 (G6PC2) gene is expressed specifically in pancreatic islet beta cells. Genome-wide association studies have shown that single nucleotide polymorphisms in the G6PC2 gene are associated with variations in fasting blood glucose (FBG) but not fasting plasma insulin. Molecular analyses examining the functional effects of these single nucleotide polymorphisms demonstrate that elevated G6PC2 expression is associated with elevated FBG. Studies in mice complement these genome-wide association data and show that deletion of the G6pc2 gene lowers FBG without affecting fasting plasma insulin. This suggests that, together with glucokinase, G6PC2 forms a substrate cycle that determines the glucose sensitivity of insulin secretion. Because genome-wide association studies and mouse studies demonstrate that elevated G6PC2 expression raises FBG and because chronically elevated FBG is detrimental to human health, increasing the risk of type 2 diabetes, it is unclear why G6PC2 evolved. We show here that the synthetic glucocorticoid dexamethasone strongly induces human G6PC2 promoter activity and endogenous G6PC2 expression in isolated human islets. Acute treatment with dexamethasone selectively induces endogenous G6pc2 expression in 129SvEv but not C57BL/6J mouse pancreas and isolated islets. The difference is due to a single nucleotide polymorphism in the C57BL/6J G6pc2 promoter that abolishes glucocorticoid receptor binding. In 6-hour fasted, nonstressed 129SvEv mice, deletion of G6pc2 lowers FBG. In response to the stress of repeated physical restraint, which is associated with elevated plasma glucocorticoid levels, G6pc2 gene expression is induced and the difference in FBG between wild-type and knockout mice is enhanced. These data suggest that G6PC2 may have evolved to modulate FBG in response to stress.


Asunto(s)
Glucemia/metabolismo , Ayuno/sangre , Glucosa-6-Fosfatasa/fisiología , Estrés Fisiológico , Animales , Células Cultivadas , Dexametasona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa-6-Fosfatasa/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética
3.
FEBS J ; 280(1): 2-27, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23134486

RESUMEN

It is widely accepted that insufficient insulin-stimulated activation of muscle glycogen synthesis is one of the major components of non-insulin-dependent (type 2) diabetes mellitus. Glycogen synthase, a key enzyme in muscle glycogen synthesis, is extensively regulated, both allosterically (by glucose-6-phosphate, ATP, and others) and covalently (by phosphorylation). Although glycogen synthase has been a topic of intense study for more than 50 years, its kinetic characterization has been confounded by its large number of phosphorylation states. Questions remain regarding the function of glycogen synthase regulation and the relative importance of allosteric and covalent modification in fulfilling this function. In this review, we consider both earlier kinetic studies and more recent site-directed mutagenesis and crystal structure studies in a detailed qualitative discussion of the effects of regulation on the kinetics of glycogen synthase. We propose that both allosteric and covalent modification of glycogen synthase may be described by a Monod-Wyman-Changeux model in terms of apparent changes to L, the equilibrium constant for transition between the T and R conformers. As, with the exception of L, all parameters of this model are independent of the glycogen synthase phosphorylation state, the need to determine kinetic parameters for all possible states is eliminated; only the relationship between a particular state and L must be established. We conclude by suggesting that renewed efforts to characterize the relationship between phosphorylation and the kinetics of glycogen synthase are essential in order to obtain a better quantitative understanding of the function of glycogen synthesis regulation. The model we propose may prove useful in this regard.


Asunto(s)
Glucógeno Sintasa/metabolismo , Músculo Esquelético/enzimología , Procesamiento Proteico-Postraduccional , Regulación Alostérica , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Glucosa-6-Fosfatasa/fisiología , Glucógeno Sintasa/química , Humanos , Cinética , Fosforilación , Estructura Cuaternaria de Proteína , Transporte de Proteínas
4.
Blood ; 119(17): 4047-55, 2012 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-22246029

RESUMEN

Glucose-6-phosphatase-ß (G6Pase-ß or G6PC3) deficiency, also known as severe congenital neutropenia syndrome 4, is characterized not only by neutropenia but also by impaired neutrophil energy homeostasis and functionality. We now show the syndrome is also associated with macrophage dysfunction, with murine G6pc3(-/-) macrophages having impairments in their respiratory burst, chemotaxis, calcium flux, and phagocytic activities. Consistent with a glucose-6-phosphate (G6P) metabolism deficiency, G6pc3(-/-) macrophages also have a lower glucose uptake and lower levels of G6P, lactate, and ATP than wild-type macrophages. Furthermore, the expression of NADPH oxidase subunits and membrane translocation of p47(phox) are down-regulated, and G6pc3(-/-) macrophages exhibit repressed trafficking in vivo both during an inflammatory response and in pregnancy. During pregnancy, the absence of G6Pase-ß activity also leads to impaired energy homeostasis in the uterus and reduced fertility of G6pc3(-/-) mothers. Together these results show that immune deficiencies in this congenital neutropenia syndrome extend beyond neutrophil dysfunction.


Asunto(s)
Glucosa-6-Fosfatasa/fisiología , Glucosa-6-Fosfato/metabolismo , Homeostasis/fisiología , Inflamación/patología , Macrófagos/fisiología , Neutropenia/congénito , Animales , Apoptosis , Western Blotting , Calcio/metabolismo , Proliferación Celular , Quimiotaxis , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Citocinas/metabolismo , Femenino , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Técnicas para Inmunoenzimas , Inflamación/genética , Inflamación/metabolismo , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Neutropenia/genética , Neutropenia/metabolismo , Neutropenia/patología , Fagocitosis , Embarazo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Estallido Respiratorio , Transducción de Señal , Síndrome
5.
Diabetes ; 61(2): 425-35, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22190647

RESUMEN

Prevention of autoimmunity requires the elimination of self-reactive T cells during their development in the thymus and maturation in the periphery. Transgenic NOD mice that overexpress islet-specific glucose 6 phosphatase catalytic subunit-related protein (IGRP) in antigen-presenting cells (NOD-IGRP mice) have no IGRP-specific T cells. To study the relative contribution of central and peripheral tolerance mechanisms to deletion of antigen-specific T cells, we crossed NOD-IGRP mice to highly diabetogenic IGRP206-214 T-cell receptor transgenic mice (NOD8.3 mice) and studied the frequency and function of IGRP-specific T cells in the thymus and periphery. Peripheral tolerance was extremely efficient and completely protected NOD-IGRP/NOD8.3 mice from diabetes. Peripheral tolerance was characterized by activation of T cells in peripheral lymphoid tissue where IGRP was expressed followed by activation-induced cell death. Thymectomy showed that thymic output of IGRP-specific transgenic T cells compensated for peripheral deletion to maintain peripheral T-cell numbers. Central tolerance was undetectable until 10 weeks and complete by 15 weeks. These in vivo data indicate that peripheral tolerance alone can protect NOD8.3 mice from autoimmune diabetes and that profound changes in T-cell repertoire can follow subtle changes in thymic antigen presentation.


Asunto(s)
Tolerancia Central , Diabetes Mellitus Tipo 1/prevención & control , Glucosa-6-Fosfatasa/fisiología , Tolerancia Periférica , Proteínas/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T/inmunología , Timo/inmunología , Animales , Femenino , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos
6.
J Pharmacol Exp Ther ; 337(3): 610-20, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21363927

RESUMEN

The effects of a glycogen phosphorylase inhibitor (GPI) and metformin (MT) on hepatic glucose fluxes (µmol · kg(-1) · min(-1)) in the presence of basal and 4-fold basal levels of plasma glucagon were investigated in 18-h fasted conscious dogs. Compared with the vehicle treatment, GPI infusion suppressed net hepatic glucose output (NHGO) completely (-3.8 ± 1.3 versus 9.9 ± 2.8) despite increased glucose 6-phosphate (G-6-P) neogenesis from gluconeogenic precursors (8.1 ± 1.1 versus 5.5 ± 1.1). MT infusion did not alter those parameters. In response to a 4-fold rise in plasma glucagon levels, in the vehicle group, plasma glucose levels were increased 2-fold, and NHGO was increased (43.9 ± 5.7 at 10 min and 22.7 ± 3.4 at steady state) without altering G-6-P neogenesis (3.7 ± 1.5 and 5.5 ± 0.5, respectively). In the GPI group, there was no increase in NHGO due to decreased glucose-6-phosphatase flux associated with reduced G-6-P concentration. A lower G-6-P concentration was the result of increased net glycogenesis without altering G-6-P neogenesis. In the MT group, the increment in NHGO (22.2 ± 4.4 at 10 min and 12.1 ± 3.6 at steady state) was approximately half of that of the vehicle group. The lesser NHGO was associated with reduced glucose-6-phosphatase flux but a rise in G-6-P concentration and only a small incorporation of plasma glucose into glycogen. In conclusion, the inhibition of glycogen phosphorylase a activity decreases basal and glucagon-induced NHGO via redirecting glucose 6-phosphate flux from glucose toward glycogen, and MT decreases glucagon-induced NHGO by inhibiting glucose-6-phosphatase flux and thereby reducing glycogen breakdown.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Glucógeno Fosforilasa de Forma Hepática/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Glucógeno Hepático/metabolismo , Hígado/efectos de los fármacos , Metformina/farmacología , Animales , Glucemia/metabolismo , Perros , Ayuno , Ácidos Grasos no Esterificados/sangre , Ácidos Grasos no Esterificados/metabolismo , Femenino , Glucagón/sangre , Glucagón/metabolismo , Glucagón/farmacología , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/fisiología , Glucosa-6-Fosfatasa/efectos de los fármacos , Glucosa-6-Fosfatasa/fisiología , Glicerol/sangre , Glicerol/metabolismo , Glucógeno Fosforilasa de Forma Hepática/metabolismo , Hematócrito , Indoles/farmacología , Insulina/sangre , Insulina/metabolismo , Ácido Láctico/sangre , Ácido Láctico/metabolismo , Hígado/metabolismo , Masculino , Fenilbutiratos/farmacología
7.
Blood ; 117(14): 3881-92, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21292774

RESUMEN

G6PC3 (or glucose-6-phosphatase-ß) deficiency underlies a congenital neutropenia syndrome in which neutrophils exhibit enhanced endoplasmic reticulum (ER) stress, increased apoptosis, impaired energy homeostasis, and impaired functionality. Here we show that murine G6pc3(-/-) neutrophils undergoing ER stress activate protein kinase-like ER kinase and phosphatidylinositol 3,4,5-trisphosphate/Akt signaling pathways, and that neutrophil apoptosis is mediated in part by the intrinsic mitochondrial pathway. In G6PC3-deficient patients, granulocyte colony-stimulating factor (G-CSF) improves neutropenia, but its impact on neutrophil apoptosis and dysfunction is unknown. We now show that G-CSF delays neutrophil apoptosis in vitro by modulating apoptotic mediators. However, G6pc3(-/-) neutrophils in culture exhibit accelerated apoptosis compared with wild-type neutrophils both in the presence or absence of G-CSF. Limiting glucose (0.6mM) accelerates apoptosis but is more pronounced for wild-type neutrophils, leading to similar survival profiles for both neutrophil populations. In vivo G-CSF therapy completely corrects neutropenia and normalizes levels of p-Akt, phosphatidylinositol 3,4,5-trisphosphate, and active caspase-3. Neutrophils from in vivo G-CSF-treated G6pc3(-/-) mice exhibit increased glucose uptake and elevated intracellular levels of G6P, lactate, and adenosine-5'-triphosphate, leading to improved functionality. Together, the results strongly suggest that G-CSF improves G6pc3(-/-) neutrophil survival by modulating apoptotic mediators and rectifies function by enhancing energy homeostasis.


Asunto(s)
Apoptosis/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Glucosa-6-Fosfatasa/genética , Factor Estimulante de Colonias de Granulocitos/farmacología , Neutrófilos/efectos de los fármacos , Animales , Apoptosis/genética , Células Cultivadas , Evaluación Preclínica de Medicamentos , Metabolismo Energético/genética , Glucosa-6-Fosfatasa/fisiología , Homeostasis/efectos de los fármacos , Homeostasis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Neutrófilos/fisiología , Fosfatos de Fosfatidilinositol/metabolismo , Subunidades de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/fisiología
8.
Life Sci ; 87(11-12): 339-49, 2010 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-20603134

RESUMEN

In a scientific career spanning from 1955 to 2000, my research focused on phosphoenolpyruvate carboxykinase and glucose-6-phosphatase. Grounded in basic enzymology, and initially pursuing the steady-state rate behavior of isolated preparations of these critically important gluconeogenic enzymes, our key findings were confirmed and extended by in situ enzyme rate experiments exploiting isolated liver perfusions. These efforts culminated in the discovery of the liver cytosolic isozyme of carboxykinase, known today as (GTP)PEPCK-C (EC4.1.1.32) and also revealed a biosynthetic function and multicomponent nature of glucose-6-phosphatase (EC3.1.3.9). Discovery that glucose-6-phosphatase possessed an intrinsically biosynthetic activity, now known as carbamyl-P:glucose phosphotransferase - along with a deeper consideration of the enzyme's hydrolytic activity as well as the action of liver glucokinase resulted in the evolution of Tuning/Retuning Hypothesis for blood glucose homeostasis in health and disease. This THEN & NOW review shares with the reader the joy and exhilaration of major scientific discovery and also contrasts the methodologies and approaches on which I relied with those currently in use.


Asunto(s)
Glucemia/metabolismo , Glucosa-6-Fosfatasa/historia , Glucosa-6-Fosfatasa/fisiología , Homeostasis/fisiología , Fisiología/historia , Animales , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Estados Unidos
9.
Mol Ther ; 18(9): 1592-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20571544

RESUMEN

Glycogen storage disease type Ia (GSD-Ia), also known as von Gierke disease, is caused by a deficiency of glucose-6-phosphatase-alpha (G6Pase), a key enzyme in glucose homeostasis. From birth, affected individuals cannot maintain normal blood glucose levels and suffer from a variety of metabolic disorders, leading to life-threatening complications. Gene therapy has been proposed as a possible option for treatment of this illness. Vectors have been constructed from feline immunodeficiency virus (FIV), a nonprimate lentivirus, because the wild-type virus does not cause disease in humans. Previously, we have shown that these vectors are capable of integrating stably into hepatocyte cell lines and adult murine livers and lead to long-term transgene expression. In the current work, we have assessed the ability to attenuate disease symptoms in a murine model of GSD-Ia. Single administration of FIV vectors containing the human G6Pase gene to G6Pase-alpha(-/-) mice did not change the biochemical and pathological phenotype. However, a double neonatal administration protocol led to normalized blood glucose levels, significantly extended survival, improved body weight, and decreased accumulation of liver glycogen associated with the disease. This approach shows a promising paradigm for treating GSD-Ia patients early in life thereby avoiding long-term consequences.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/genética , Glucosa-6-Fosfatasa/fisiología , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/terapia , Virus de la Inmunodeficiencia Felina/genética , Animales , Animales Recién Nacidos , Células COS , Línea Celular , Chlorocebus aethiops , Colesterol/metabolismo , Glucosa-6-Fosfatasa/genética , Humanos , Inmunohistoquímica , Riñón/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Tamaño de los Órganos/genética , Tamaño de los Órganos/fisiología , Reacción en Cadena de la Polimerasa
10.
Am J Physiol Regul Integr Comp Physiol ; 292(4): R1400-7, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17158265

RESUMEN

This paper provides molecular evidence for a liver glyconeogenic pathway, that is, a concomitant activation of hepatic gluconeogenesis and glycogenesis, which could participate in the mechanisms that cope with amino acid excess in high-protein (HP) fed rats. This evidence is based on the concomitant upregulation of phosphoenolpyruvate carboxykinase (PEPCK) gene expression, downregulation of glucose 6-phosphatase catalytic subunit (G6PC1) gene expression, an absence of glucose release from isolated hepatocytes and restored hepatic glycogen stores in the fed state in HP fed rats. These effects are mainly due to the ability of high physiological concentrations of portal blood amino acids to counteract glucagon-induced liver G6PC1 but not PEPCK gene expression. These results agree with the idea that the metabolic pathway involved in glycogen synthesis is dependent upon the pattern of nutrient availability. This nonoxidative glyconeogenic disposal pathway of gluconeogenic substrates copes with amino excess and participates in adjusting both amino acid and glucose homeostasis. In addition, the pattern of PEPCK and G6PC1 gene expression provides evidence that neither the kidney nor the small intestine participated in gluconeogenic glucose production under our experimental conditions. Moreover, the main glucose-6-phosphatase (G6Pase) isoform expressed in the small intestine is the ubiquitous isoform of G6Pase (G6PC3) rather than the G6PC1 isoform expressed in gluconeogenic organs.


Asunto(s)
Proteínas en la Dieta/administración & dosificación , Gluconeogénesis , Glucosa/biosíntesis , Glucógeno Hepático/metabolismo , Hígado/metabolismo , Aminoácidos/sangre , Animales , Glucemia/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ayuno , Regulación de la Expresión Génica , Glucagón/metabolismo , Glucagón/farmacología , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfatasa/metabolismo , Glucosa-6-Fosfatasa/fisiología , Hepatocitos/citología , Hepatocitos/metabolismo , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Insulina/sangre , Insulina/farmacología , Hígado/citología , Hígado/enzimología , Glucógeno Hepático/análisis , Masculino , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/fisiología , Sistema Porta/fisiología , Ratas , Ratas Wistar
11.
J Biol Chem ; 281(52): 39982-9, 2006 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-17023421

RESUMEN

In liver, glucose-6-phosphatase catalyzes the hydrolysis of glucose-6-phosphate (G6P) to glucose and inorganic phosphate, the final step in the gluconeogenic and glycogenolytic pathways. Mutations in the glucose-6-phosphatase catalytic subunit (G6Pase) give rise to glycogen storage disease (GSD) type 1a, which is characterized in part by hypoglycemia, growth retardation, hypertriglyceridemia, hypercholesterolemia, and hepatic glycogen accumulation. Recently, a novel G6Pase isoform was identified, designated UGRP/G6Pase-beta. The activity of UGRP relative to G6Pase in vitro is disputed, raising the question as to whether G6P is a physiologically important substrate for this protein. To address this issue we have characterized the phenotype of UGRP knock-out mice. G6P hydrolytic activity was decreased by approximately 50% in homogenates of UGRP(-/-) mouse brain relative to wild type tissue, consistent with the ability of UGRP to hydrolyze G6P. In addition, female, but not male, UGRP(-/-) mice exhibit growth retardation as do G6Pase(-/-) mice and patients with GSD type 1a. However, in contrast to G6Pase(-/-) mice and patients with GSD type 1a, UGRP(-/-) mice exhibit no change in hepatic glycogen content, blood glucose, or triglyceride levels. Although UGRP(-/-) mice are not hypoglycemic, female UGRP(-/-) mice have elevated ( approximately 60%) plasma glucagon and reduced ( approximately 20%) plasma cholesterol. We hypothesize that the hyperglucagonemia prevents hypoglycemia and that the hypocholesterolemia is secondary to the hyperglucagonemia. As such, the phenotype of UGRP(-/-) mice is mild, indicating that G6Pase is the major glucose-6-phosphatase of physiological importance for glucose homeostasis in vivo.


Asunto(s)
Dominio Catalítico/genética , Colesterol/sangre , Regulación hacia Abajo/genética , Eliminación de Gen , Glucagón/biosíntesis , Glucosa-6-Fosfatasa/genética , Proteínas/genética , Regulación hacia Arriba/genética , Animales , Femenino , Regulación de la Expresión Génica/genética , Glucosa/metabolismo , Glucosa-6-Fosfatasa/biosíntesis , Glucosa-6-Fosfatasa/fisiología , Enfermedad del Almacenamiento de Glucógeno Tipo I/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Homeostasis/genética , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Proteínas/fisiología , Secretoglobinas
12.
Diabetes ; 55(9): 2412-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16936188

RESUMEN

Cytotoxic T-cells are the major mediators of beta-cell destruction in type 1 diabetes, but the molecular mechanisms are not definitively established. We have examined the contribution of perforin and Fas ligand to beta-cell destruction using islet-specific CD8(+) T-cells from T-cell receptor transgenic NOD8.3 mice. NOD8.3 T-cells killed Fas-deficient islets in vitro and in vivo. Perforin-deficient NOD8.3 T-cells were able to destroy wild-type but not Fas-deficient islets in vitro. These results imply that NOD8.3 T-cells use both pathways and that Fas is required for beta-cell killing only when perforin is missing. Consistent with this theory, transgenic NOD8.3 mice with beta-cells that do not respond to Fas ligation were not protected from diabetes. We next investigated the mechanism of protection provided by overexpression of suppressor of cytokine signaling-1 (SOCS-1) in beta-cells of NOD8.3 mice. SOCS-1 islets remained intact when grafted into NOD8.3 mice and were less efficiently killed in vitro. However, addition of exogenous peptide rendered SOCS-1 islets susceptible to 8.3 T-cell-mediated lysis. Therefore, NOD8.3 T-cells use both perforin and Fas pathways to kill beta-cells and the surprising blockade of NOD8.3 T-cell-mediated beta-cell death by SOCS-1 overexpression may be due in part to reduced target cell recognition.


Asunto(s)
Células Secretoras de Insulina/citología , Glicoproteínas de Membrana/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Linfocitos T Citotóxicos/fisiología , Receptor fas/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Linfocitos T CD8-positivos/fisiología , Diabetes Mellitus Tipo 1/fisiopatología , Proteína de Dominio de Muerte Asociada a Fas , Glucosa-6-Fosfatasa/fisiología , Trasplante de Islotes Pancreáticos/fisiología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Perforina , Proteínas Citotóxicas Formadoras de Poros , Proteínas/fisiología , Proteína 1 Supresora de la Señalización de Citocinas
13.
Cell Metab ; 3(4): 267-75, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16581004

RESUMEN

STAT3 regulates glucose homeostasis by suppressing the expression of gluconeogenic genes in the liver. The mechanism by which hepatic STAT3 is regulated by nutritional or hormonal status has remained unknown, however. Here, we show that an increase in the plasma insulin concentration, achieved either by glucose administration or by intravenous insulin infusion, stimulates tyrosine phosphorylation of STAT3 in the liver. This effect of insulin was mediated by the hormone's effects in the brain, and the increase in hepatic IL-6 induced by the brain-insulin action is essential for the activation of STAT3. The inhibition of hepatic glucose production and of expression of gluconeogenic genes induced by intracerebral ventricular insulin infusion was impaired in mice with liver-specific STAT3 deficiency or in mice with IL-6 deficiency. These results thus indicate that IL-6-STAT3 signaling in the liver contributes to insulin action in the brain, leading to the suppression of hepatic glucose production.


Asunto(s)
Encéfalo/metabolismo , Glucosa/metabolismo , Insulina/fisiología , Hígado/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Activación Enzimática , Gluconeogénesis , Glucosa/farmacología , Técnica de Clampeo de la Glucosa , Glucosa-6-Fosfatasa/fisiología , Homeostasis , Insulina/administración & dosificación , Insulina/sangre , Insulina/farmacología , Resistencia a la Insulina , Interleucina-6/análisis , Interleucina-6/fisiología , Macrófagos del Hígado/química , Macrófagos del Hígado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfoenolpiruvato Carboxilasa/fisiología , Fosforilación , Receptor de Insulina/fisiología , Transducción de Señal
14.
Diabetologia ; 48(9): 1851-9, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16012821

RESUMEN

AIMS/HYPOTHESIS: There are three members of the glucose-6-phosphatase (G6Pase) family: (1) the liver/kidney/intestine G6Pase-alpha (encoded by G6PC), which is a key enzyme in glucose homeostasis; (2) the ubiquitous G6Pase-beta (encoded by G6PC3); and (3) the islet-specific G6Pase-related protein (IGRP, encoded by /G6PC2). While G6Pase-alpha and G6Pase-beta are functional glucose-6-phosphate hydrolases, IGRP possesses almost no hydrolase activity. This was unexpected since G6Pase-alpha is more closely related to IGRP than G6Pase-beta. Recently, amino acids 206-214 in IGRP were identified as a beta cell antigen targeted by a prevalent population of pathogenic CD8+ T cells in autoimmune diabetes, suggesting that this peptide confers functional specificity to IGRP. We therefore investigated the molecular events that inactivate IGRP activity and the effects of the beta cell antigen sequence on the stability and enzymatic activity of G6Pase-alpha. METHODS: Studies were performed using site-directed mutagenesis and transient expression assays. Protein stability was evaluated by Western blotting, proteasome inhibitor studies and in vitro transcription-translation. RESULTS: We showed that the residues responsible for G6Pase activity are more extensive than previously recognised. Introducing the IGRP antigenic motif into G6Pase-alpha does not completely destroy activity, although it does destabilise the protein. The low hydrolytic activity in IGRP is due to the combination of multiple independent mutations. CONCLUSIONS/INTERPRETATION: The loss of catalytic activity in IGRP arises from the sum of many sequence differences. G6Pase-alpha mutants containing the beta cell antigen sequence are preferentially degraded in cells, which prevents targeting by pathogenic CD8+ T cells. It is possible that IGRP levels in beta cells could dictate susceptibilities to diabetes.


Asunto(s)
Glucosa-6-Fosfatasa/fisiología , Islotes Pancreáticos/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Clonación Molecular , Secuencia Conservada , Cartilla de ADN , Perros , Glucosa-6-Fosfatasa/química , Glucosa-6-Fosfatasa/genética , Humanos , Islotes Pancreáticos/enzimología , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido
15.
Anal Biochem ; 339(1): 150-6, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15766722

RESUMEN

A high-throughput screening was developed for the detection of phosphatase activity in bacterial colonies. Unlike other methods, the current procedure can be applied to any phosphatase because it uses physiological substrates and detects the compelled product of all phosphatase reactions, that is, orthophosphate. In this method, substrates diffuse from a filter paper across a nitrocellulose membrane to bacterial colonies situated on the opposite face, and then reaction products flow back to the paper. Finally, a colorimetric reagent discloses the presence of orthophosphate in the filter paper. We validated the performance of this assay with several substrates and experimental conditions and with different phosphatases, including a library of randomly mutagenized rapeseed chloroplast fructose-1,6-bisphosphatase. This procedure could be extended to other enzymatic activities provided that an appropriate detection of reaction products is available.


Asunto(s)
Fosfatasa Alcalina/química , Brassica rapa/enzimología , Escherichia coli/enzimología , Fructosa-Bifosfatasa/química , Glucosa-6-Fosfatasa/química , Biblioteca de Péptidos , Fosfatasa Alcalina/fisiología , Cloroplastos/enzimología , Evaluación Preclínica de Medicamentos , Fructosa-Bifosfatasa/fisiología , Fructosafosfatos/metabolismo , Glucosa-6-Fosfatasa/fisiología , Glucofosfatos/metabolismo , Mutagénesis , Especificidad por Sustrato
16.
J Biol Chem ; 280(12): 11114-9, 2005 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-15661744

RESUMEN

Glucose is absolutely essential for the survival and function of the brain. In our current understanding, there is no endogenous glucose production in the brain, and it is totally dependent upon blood glucose. This glucose is generated between meals by the hydrolysis of glucose-6-phosphate (Glc-6-P) in the liver and the kidney. Recently, we reported a ubiquitously expressed Glc-6-P hydrolase, glucose-6-phosphatase-beta (Glc-6-Pase-beta), that can couple with the Glc-6-P transporter to hydrolyze Glc-6-P to glucose in the terminal stages of glycogenolysis and gluconeogenesis. Here we show that astrocytes, the main reservoir of brain glycogen, express both the Glc-6-Pase-beta and Glc-6-P transporter activities and that these activities can couple to form an active Glc-6-Pase complex, suggesting that astrocytes may provide an endogenous source of brain glucose.


Asunto(s)
Encéfalo/metabolismo , Glucosa-6-Fosfatasa/fisiología , Glucosa/biosíntesis , Animales , Antiportadores , Astrocitos/enzimología , Células COS , Glucosa-6-Fosfatasa/genética , Ratones , Proteínas de Transporte de Monosacáridos , Monoéster Fosfórico Hidrolasas/fisiología , Fosfotransferasas/análisis
18.
J Biol Chem ; 278(47): 47098-103, 2003 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-13129915

RESUMEN

A fine control of the blood glucose level is essential to avoid hyper- or hypo-glycemic shocks associated with many metabolic disorders, including diabetes mellitus and type I glycogen storage disease. Between meals, the primary source of blood glucose is gluconeogenesis and glycogenolysis. In the final step of both pathways, glucose-6-phosphate (G6P) is hydrolyzed to glucose by the glucose-6-phosphatase (G6Pase) complex. Because G6Pase (renamed G6Pase-alpha) is primarily expressed only in the liver, kidney, and intestine, it has implied that most other tissues cannot contribute to interprandial blood glucose homeostasis. We demonstrate that a novel, widely expressed G6Pase-related protein, PAP2.8/UGRP, renamed here G6Pase-beta, is an acid-labile, vanadate-sensitive, endoplasmic reticulum-associated phosphohydrolase, like G6Pase-alpha. Both enzymes have the same active site structure, exhibit a similar Km toward G6P, but the Vmax of G6Pase-alpha is approximately 6-fold greater than that of G6Pase-beta. Most importantly, G6Pase-beta couples with the G6P transporter to form an active G6Pase complex that can hydrolyze G6P to glucose. Our findings challenge the current dogma that only liver, kidney, and intestine can contribute to blood glucose homeostasis and explain why type Ia glycogen storage disease patients, lacking a functional liver/kidney/intestine G6Pase complex, are still capable of endogenous glucose production.


Asunto(s)
Glucosa-6-Fosfatasa/clasificación , Glucosa-6-Fosfatasa/fisiología , Enfermedad del Almacenamiento de Glucógeno Tipo I/enzimología , Hipoglucemia/enzimología , Factores de Edad , Secuencia de Aminoácidos , Antiportadores/metabolismo , Glucemia/biosíntesis , ADN Complementario , Glucosa-6-Fosfatasa/genética , Homeostasis , Humanos , Hipoglucemia/etiología , Cinética , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/metabolismo , Unión Proteica , Alineación de Secuencia , Terminología como Asunto , Distribución Tisular
19.
Exp Biol Med (Maywood) ; 227(8): 601-8, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12192101

RESUMEN

Progress has continued to be made over the past 4 years in our understanding of the glucose-6-phosphatase (G6Pase) system. The gene for a second component of the system, the putative glucose-6-P transporter (G6PT), was cloned, and mutations in this gene were found in patients diagnosed with glycogen storage disease type 1b. The functional characterization of this putative G6PT has been initiated, and the relationship between substrate transport via the G6PT and catalysis by the system's catalytic subunit continues to be explored. A lively debate over the feasibility of various aspects of the two proposed models of the G6Pase system persists, and the functional/structural relationships of the individual components of the system remain a hot topic of interest in G6Pase research. New evidence supportive of physiologic roles for the biosynthetic functions of the G6Pase system in vivo also has emerged over the past 4 years.


Asunto(s)
Glucosa-6-Fosfatasa/fisiología , Secuencia de Aminoácidos , Animales , Antiportadores/genética , Antiportadores/fisiología , Catálisis , Dominio Catalítico , Diabetes Mellitus Tipo 2/enzimología , Glucosa/metabolismo , Glucosa-6-Fosfatasa/química , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfato/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Humanos , Ratones , Ratones Noqueados , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/fisiología , Fosfatos/metabolismo , Fosforilación , Conformación Proteica , Subunidades de Proteína , Proteínas Recombinantes de Fusión/fisiología , Relación Estructura-Actividad
20.
Eur J Biochem ; 267(6): 1533-49, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10712583

RESUMEN

The operation of glucose 6-phosphatase (EC 3.1.3.9) (Glc6Pase) stems from the interaction of at least two highly hydrophobic proteins embedded in the ER membrane, a heavily glycosylated catalytic subunit of m 36 kDa (P36) and a 46-kDa putative glucose 6-phosphate (Glc6P) translocase (P46). Topology studies of P36 and P46 predict, respectively, nine and ten transmembrane domains with the N-terminal end of P36 oriented towards the lumen of the ER and both termini of P46 oriented towards the cytoplasm. P36 gene expression is increased by glucose, fructose 2,6-bisphosphate (Fru-2,6-P2) and free fatty acids, as well as by glucocorticoids and cyclic AMP; the latter are counteracted by insulin. P46 gene expression is affected by glucose, insulin and cyclic AMP in a manner similar to P36. Accordingly, several response elements for glucocorticoids, cyclic AMP and insulin regulated by hepatocyte nuclear factors were found in the Glc6Pase promoter. Mutations in P36 and P46 lead to glycogen storage disease (GSD) type-1a and type-1 non a (formerly 1b and 1c), respectively. Adenovirus-mediated overexpression of P36 in hepatocytes and in vivo impairs glycogen metabolism and glycolysis and increases glucose production; P36 overexpression in INS-1 cells results in decreased glycolysis and glucose-induced insulin secretion. The nature of the interaction between P36 and P46 in controling Glc6Pase activity remains to be defined. The latter might also have functions other than Glc6P transport that are related to Glc6P metabolism.


Asunto(s)
Proteínas de Escherichia coli , Glucosa-6-Fosfatasa/fisiología , Glucosa/metabolismo , Proteínas de Transporte de Monosacáridos , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Antiportadores , Proteínas Bacterianas/fisiología , Transporte Biológico , Proteínas Portadoras/fisiología , AMP Cíclico/farmacología , AMP Cíclico/fisiología , Retículo Endoplásmico/enzimología , Inducción Enzimática/efectos de los fármacos , Ácidos Grasos no Esterificados/metabolismo , Ácidos Grasos no Esterificados/farmacología , Fructosadifosfatos/metabolismo , Fructosadifosfatos/farmacología , Regulación de la Expresión Génica , Ingeniería Genética , Vectores Genéticos/genética , Glucocorticoides/farmacología , Glucocorticoides/fisiología , Glucosa/farmacología , Glucosa/fisiología , Glucosa-6-Fosfatasa/química , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/clasificación , Enfermedad del Almacenamiento de Glucógeno Tipo I/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Glucólisis , Humanos , Hidrólisis , Insulina/farmacología , Insulina/fisiología , Hígado/metabolismo , Sustancias Macromoleculares , Microsomas/enzimología , Modelos Moleculares , Datos de Secuencia Molecular , Fosfotransferasas/química , Fosfotransferasas/genética , Fosfotransferasas/fisiología , Sistemas de Mensajero Secundario , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA