Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Genet ; 13(5): e1006819, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28558013

RESUMEN

A deficiency in glucose-6-phosphatase-α (G6Pase-α) in glycogen storage disease type Ia (GSD-Ia) leads to impaired glucose homeostasis and metabolic manifestations including hepatomegaly caused by increased glycogen and neutral fat accumulation. A recent report showed that G6Pase-α deficiency causes impairment in autophagy, a recycling process important for cellular metabolism. However, the molecular mechanism underlying defective autophagy is unclear. Here we show that in mice, liver-specific knockout of G6Pase-α (L-G6pc-/-) leads to downregulation of sirtuin 1 (SIRT1) signaling that activates autophagy via deacetylation of autophagy-related (ATG) proteins and forkhead box O (FoxO) family of transcriptional factors which transactivate autophagy genes. Consistently, defective autophagy in G6Pase-α-deficient liver is characterized by attenuated expressions of autophagy components, increased acetylation of ATG5 and ATG7, decreased conjugation of ATG5 and ATG12, and reduced autophagic flux. We further show that hepatic G6Pase-α deficiency results in activation of carbohydrate response element-binding protein, a lipogenic transcription factor, increased expression of peroxisome proliferator-activated receptor-γ (PPAR-γ), a lipid regulator, and suppressed expression of PPAR-α, a master regulator of fatty acid ß-oxidation, all contributing to hepatic steatosis and downregulation of SIRT1 expression. An adenovirus vector-mediated increase in hepatic SIRT1 expression corrects autophagy defects but does not rectify metabolic abnormalities associated with G6Pase-α deficiency. Importantly, a recombinant adeno-associated virus (rAAV) vector-mediated restoration of hepatic G6Pase-α expression corrects metabolic abnormalities, restores SIRT1-FoxO signaling, and normalizes defective autophagy. Taken together, these data show that hepatic G6Pase-α deficiency-mediated down-regulation of SIRT1 signaling underlies defective hepatic autophagy in GSD-Ia.


Asunto(s)
Autofagia , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Animales , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Factores de Transcripción Forkhead/metabolismo , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfatasa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Hepatocitos/metabolismo , Ratones , PPAR gamma/genética , PPAR gamma/metabolismo , Sirtuina 1/genética
2.
Mol Genet Metab ; 120(3): 229-234, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28096054

RESUMEN

Glycogen storage disease type Ia (GSD-Ia), characterized by impaired glucose homeostasis and chronic risk of hepatocellular adenoma (HCA) and carcinoma (HCC), is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC). We have previously shown that G6pc-/- mice receiving gene transfer mediated by rAAV-G6PC, a recombinant adeno-associated virus (rAAV) vector expressing G6Pase-α, and expressing 3-63% of normal hepatic G6Pase-α activity maintain glucose homeostasis and do not develop HCA/HCC. However, the threshold of hepatic G6Pase-α activity required to prevent tumor formation remained unknown. In this study, we constructed rAAV-co-G6PC, a rAAV vector expressing a codon-optimized (co) G6Pase-α and showed that rAAV-co-G6PC was more efficacious than rAAV-G6PC in directing hepatic G6Pase-α expression. Over an 88-week study, we showed that both rAAV-G6PC- and rAAV-co-G6PC-treated G6pc-/- mice expressing 3-33% of normal hepatic G6Pase-α activity (AAV mice) maintained glucose homeostasis, lacked HCA/HCC, and were protected against age-related obesity and insulin resistance. Of the eleven rAAV-G6PC/rAAV-co-G6PC-treated G6pc-/- mice harboring 0.9-2.4% of normal hepatic G6Pase-α activity (AAV-low mice), 3 expressing 0.9-1.3% of normal hepatic G6Pase-α activity developed HCA/HCC, while 8 did not (AAV-low-NT). Finally, we showed that the AAV-low-NT mice exhibited a phenotype indistinguishable from that of AAV mice expressing ≥3% of normal hepatic G6Pase-α activity. The results establish the threshold of hepatic G6Pase-α activity required to prevent HCA/HCC and show that GSD-Ia mice harboring <2% of normal hepatic G6Pase-α activity are at risk of tumor development.


Asunto(s)
Adenoma de Células Hepáticas/prevención & control , Carcinoma Hepatocelular/prevención & control , Terapia Genética/métodos , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/terapia , Neoplasias Hepáticas/prevención & control , Adenoma de Células Hepáticas/enzimología , Animales , Carcinoma Hepatocelular/enzimología , Dependovirus/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Vectores Genéticos/administración & dosificación , Glucosa/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/complicaciones , Enfermedad del Almacenamiento de Glucógeno Tipo I/enzimología , Homeostasis , Humanos , Hígado/enzimología , Neoplasias Hepáticas/enzimología , Ratones
3.
Hum Mol Genet ; 24(18): 5115-25, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26089201

RESUMEN

Glycogen storage disease type-Ia (GSD-Ia) is caused by a lack of glucose-6-phosphatase-α (G6Pase-α or G6PC) activity. We have shown that gene therapy mediated by a recombinant adeno-associated virus (rAAV) vector expressing human G6Pase-α normalizes blood glucose homeostasis in the global G6pc knockout (G6pc(-/-)) mice for 70-90 weeks. The treated G6pc(-/-) mice expressing 3-63% of normal hepatic G6Pase-α activity (AAV mice) produce endogenous hepatic glucose levels 61-68% of wild-type littermates, have a leaner phenotype and exhibit fasting blood insulin levels more typical of young adult mice. We now show that unlike wild-type mice, the lean AAV mice have increased caloric intake and do not develop age-related obesity or insulin resistance. Pathway analysis shows that signaling by hepatic carbohydrate response element binding protein that improves glucose tolerance and insulin signaling is activated in AAV mice. In addition, several longevity factors in the calorie restriction pathway, including the NADH shuttle systems, NAD(+) concentrations and the AMP-activated protein kinase/sirtuin 1/peroxisome proliferator-activated receptor-γ coactivator 1α pathway are upregulated in the livers of AAV mice. The finding that partial restoration of hepatic G6Pase-α activity in GSD-Ia mice not only attenuates the phenotype of hepatic G6Pase-α deficiency but also prevents the development of age-related obesity and insulin resistance seen in wild-type mice may suggest relevance of the G6Pase-α enzyme to obesity and diabetes.


Asunto(s)
Expresión Génica , Glucosa-6-Fosfatasa/genética , Resistencia a la Insulina/genética , Obesidad/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Dependovirus/genética , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , NAD/metabolismo , Proteínas Nucleares/metabolismo , Obesidad/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Factores de Transcripción/metabolismo
4.
Mol Genet Metab Rep ; 3: 28-32, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26937391

RESUMEN

Glycogen storage disease type Ia (GSD-Ia), characterized by impaired glucose homeostasis and chronic risk of hepatocellular adenoma (HCA), is caused by a deficiency in glucose-6-phosphatase-α (G6Pase-α or G6PC) activity. In a previous 70-90 week-study, we showed that a recombinant adeno-associated virus (rAAV) vector-mediated gene transfer that restores more than 3% of wild-type hepatic G6Pase-α activity in G6pc (-/-) mice corrects hepatic G6Pase-α deficiency with no evidence of HCA. We now examine the minimal hepatic G6Pase-α activity required to confer therapeutic efficacy. We show that rAAV-treated G6pc (-/-) mice expressing 0.2% of wild-type hepatic G6Pase-α activity suffered from frequent hypoglycemic seizures at age 63-65 weeks but mice expressing 0.5-1.3% of wild-type hepatic G6Pase-α activity (AAV-LL mice) sustain 4-6 h of fast and grow normally to age 75-90 weeks. Despite marked increases in hepatic glycogen accumulation, the AAV-LL mice display no evidence of hepatic abnormalities, hepatic steatosis, or HCA. Interprandial glucose homeostasis is maintained by the G6Pase-α/glucose-6-phosphate transporter (G6PT) complex, and G6PT-mediated microsomal G6P uptake is the rate-limiting step in endogenous glucose production. We show that hepatic G6PT activity is increased in AAV-LL mice. These findings are encouraging for clinical studies of G6Pase-α gene-based therapy for GSD-Ia.

5.
Mol Genet Metab ; 114(1): 41-5, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25492228

RESUMEN

Glucose-6-phosphatase-ß (G6Pase-ß or G6PC3) deficiency is characterized by neutropenia and dysfunction in both neutrophils and macrophages. G6Pase-ß is an enzyme embedded in the endoplasmic reticulum membrane that catalyzes the hydrolysis of glucose-6-phosphate (G6P) to glucose and phosphate. To date, 33 separate G6PC3 mutations have been identified in G6Pase-ß-deficient patients but only the p.R253H and p.G260R missense mutations have been characterized functionally for pathogenicity. Here we functionally characterize 16 of the 19 known missense mutations using a sensitive assay, based on a recombinant adenoviral vector-mediated expression system, to demonstrate pathogenicity. Fourteen missense mutations completely abolish G6Pase-ß enzymatic activity while the p.S139I and p.R189Q mutations retain 49% and 45%, respectively of wild type G6Pase-ß activity. A database of residual enzymatic activity retained by the G6Pase-ß mutations will serve as a reference for evaluating genotype-phenotype relationships.


Asunto(s)
Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Mutación Missense , Neutropenia/congénito , Secuencia de Aminoácidos , Animales , Western Blotting , Células COS , Chlorocebus aethiops , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Estudios de Asociación Genética , Vectores Genéticos , Glucosa-6-Fosfatasa/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Neutropenia/genética
6.
Mol Genet Metab ; 110(3): 275-80, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23856420

RESUMEN

Glycogen storage disease type-Ia (GSD-Ia) patients deficient in glucose-6-phosphatase-α (G6Pase-α or G6PC) manifest impaired glucose homeostasis characterized by fasting hypoglycemia, growth retardation, hepatomegaly, nephromegaly, hyperlipidemia, hyperuricemia, and lactic acidemia. Two efficacious recombinant adeno-associated virus pseudotype 2/8 (rAAV8) vectors expressing human G6Pase-α have been independently developed. One is a single-stranded vector containing a 2864-bp of the G6PC promoter/enhancer (rAAV8-GPE) and the other is a double-stranded vector containing a shorter 382-bp minimal G6PC promoter/enhancer (rAAV8-miGPE). To identify the best construct, a direct comparison of the rAAV8-GPE and the rAAV8-miGPE vectors was initiated to determine the best vector to take forward into clinical trials. We show that the rAAV8-GPE vector directed significantly higher levels of hepatic G6Pase-α expression, achieved greater reduction in hepatic glycogen accumulation, and led to a better toleration of fasting in GSD-Ia mice than the rAAV8-miGPE vector. Our results indicated that additional control elements in the rAAV8-GPE vector outweigh the gains from the double-stranded rAAV8-miGPE transduction efficiency, and that the rAAV8-GPE vector is the current choice for clinical translation in human GSD-Ia.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Regiones Promotoras Genéticas , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Glucosa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Humanos , Hígado/metabolismo , Metaboloma , Ratones , Ratones Noqueados , Especificidad de Órganos , Transducción Genética , Transgenes
7.
Hepatology ; 56(5): 1719-29, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22422504

RESUMEN

UNLABELLED: Glycogen storage disease type Ia (GSD-Ia), which is characterized by impaired glucose homeostasis and chronic risk of hepatocellular adenoma (HCA), is caused by deficiencies in the endoplasmic reticulum (ER)-associated glucose-6-phosphatase-α (G6Pase-α or G6PC) that hydrolyzes glucose-6-phosphate (G6P) to glucose. G6Pase-α activity depends on the G6P transporter (G6PT) that translocates G6P from the cytoplasm into the ER lumen. The functional coupling of G6Pase-α and G6PT maintains interprandial glucose homeostasis. We have shown previously that gene therapy mediated by AAV-GPE, an adeno-associated virus (AAV) vector expressing G6Pase-α directed by the human G6PC promoter/enhancer (GPE), completely normalizes hepatic G6Pase-α deficiency in GSD-Ia (G6pc(-/-) ) mice for at least 24 weeks. However, a recent study showed that within 78 weeks of gene deletion, all mice lacking G6Pase-α in the liver develop HCA. We now show that gene therapy mediated by AAV-GPE maintains efficacy for at least 70-90 weeks for mice expressing more than 3% of wild-type hepatic G6Pase-α activity. The treated mice displayed normal hepatic fat storage, had normal blood metabolite and glucose tolerance profiles, had reduced fasting blood insulin levels, maintained normoglycemia over a 24-hour fast, and had no evidence of hepatic abnormalities. After a 24-hour fast, hepatic G6PT messenger RNA levels in G6pc(-/-) mice receiving gene therapy were markedly increased. Because G6PT transport is the rate-limiting step in microsomal G6P metabolism, this may explain why the treated G6pc(-/-) mice could sustain prolonged fasts. The low fasting blood insulin levels and lack of hepatic steatosis may explain the absence of HCA. CONCLUSION: These results confirm that AAV-GPE-mediated gene transfer corrects hepatic G6Pase-α deficiency in murine GSD-Ia and prevents chronic HCA formation.


Asunto(s)
Adenoma/prevención & control , Terapia Genética , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo I/terapia , Neoplasias Hepáticas/prevención & control , Hígado/metabolismo , Animales , Antiportadores/genética , Antiportadores/metabolismo , Glucemia , Índice de Masa Corporal , Peso Corporal , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Terapia Genética/efectos adversos , Vectores Genéticos , Prueba de Tolerancia a la Glucosa , Glucosa-6-Fosfatasa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Homeostasis , Insulina/sangre , Hígado/enzimología , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo
8.
PLoS One ; 6(9): e23157, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21949678

RESUMEN

Blood glucose homeostasis between meals depends upon production of glucose within the endoplasmic reticulum (ER) of the liver and kidney by hydrolysis of glucose-6-phosphate (G6P) into glucose and phosphate (P(i)). This reaction depends on coupling the G6P transporter (G6PT) with glucose-6-phosphatase-α (G6Pase-α). Only one G6PT, also known as SLC37A4, has been characterized, and it acts as a P(i)-linked G6P antiporter. The other three SLC37 family members, predicted to be sugar-phosphate:P(i) exchangers, have not been characterized functionally. Using reconstituted proteoliposomes, we examine the antiporter activity of the other SLC37 members along with their ability to couple with G6Pase-α. G6PT- and mock-proteoliposomes are used as positive and negative controls, respectively. We show that SLC37A1 and SLC37A2 are ER-associated, P(i)-linked antiporters, that can transport G6P. Unlike G6PT, neither is sensitive to chlorogenic acid, a competitive inhibitor of physiological ER G6P transport, and neither couples to G6Pase-α. We conclude that three of the four SLC37 family members are functional sugar-phosphate antiporters. However, only G6PT/SLC37A4 matches the characteristics of the physiological ER G6P transporter, suggesting the other SLC37 proteins have roles independent of blood glucose homeostasis.


Asunto(s)
Antiportadores/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Fosfatos/metabolismo , Animales , Antiportadores/genética , Transporte Biológico , Western Blotting , Células COS , Chlorocebus aethiops , Perfilación de la Expresión Génica , Glucosa/metabolismo , Glucosa-6-Fosfato/metabolismo , Glucosa-6-Fosfato/farmacocinética , Humanos , Mucosa Intestinal/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Proteínas de Transporte de Membrana/genética , Ratones , Microscopía Fluorescente , Proteínas de Transporte de Monosacáridos/genética , Páncreas/metabolismo , Proteolípidos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Mol Ther ; 18(6): 1076-84, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20389290

RESUMEN

Glycogen storage disease type Ia (GSD-Ia) patients deficient in glucose-6-phosphatase-alpha (G6Pase-alpha or G6PC) manifest disturbed glucose homeostasis. We examined the efficacy of liver G6Pase-alpha delivery mediated by AAV-GPE, an adeno-associated virus (AAV) serotype 8 vector expressing human G6Pase-alpha directed by the human G6PC promoter/enhancer (GPE), and compared it to AAV-CBA, that directed murine G6Pase-alpha expression using a hybrid chicken beta-actin (CBA) promoter/cytomegalovirus (CMV) enhancer. The AAV-GPE directed hepatic G6Pase-alpha expression in the infused G6pc(-/-) mice declined 12-fold from age 2 to 6 weeks but stabilized at wild-type levels from age 6 to 24 weeks. In contrast, the expression directed by AAV-CBA declined 95-fold over 24 weeks, demonstrating that the GPE is more effective in directing persistent in vivo hepatic transgene expression. We further show that the rapid decline in transgene expression directed by AAV-CBA results from an inflammatory immune response elicited by the AAV-CBA vector. The AAV-GPE-treated G6pc(-/-) mice exhibit normal levels of blood glucose, blood metabolites, hepatic glycogen, and hepatic fat. Moreover, the mice maintained normal blood glucose levels even after 6 hours of fasting. The complete normalization of hepatic G6Pase-alpha deficiency by the G6PC promoter/enhancer holds promise for the future of gene therapy in human GSD-Ia patients.


Asunto(s)
Terapia Genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/terapia , Hígado/metabolismo , Animales , Dependovirus/genética , Vectores Genéticos , Glucosafosfato Deshidrogenasa/genética , Hígado/enzimología , Ratones , Ratones Noqueados
10.
Genesis ; 47(9): 590-4, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19548314

RESUMEN

Glucose-6-phosphatase-alpha (G6Pase-alpha or G6PC) catalyzes the hydrolysis of glucose-6-phosphate to glucose and is a key enzyme in interprandial glucose homeostasis. Mutations in the human G6PC gene, expressed primarily in the liver, kidney, and intestine, cause glycogen storage disease Type Ia (GSD-Ia), an autosomal recessive disorder characterized by a disturbed glucose homeostasis. For better understanding of the roles of G6Pase-alpha in different tissues and in pathological conditions, we have generated mice harboring a conditional null allele for G6pc by flanking Exon 3 of the G6pc gene with loxP sites. We confirmed the null phenotype by using the EIIa-Cre transgenic approach to generate mice lacking Exon 3 of the G6pc gene. The resulting homozygous Cre-recombined null mice manifest a phenotype mimicking G6Pase-alpha-deficient mice and human GSD-Ia patients. This G6pc conditional null allele will be valuable to examine the consequence of tissue-specific G6Pase-alpha deficiency and the mechanisms of long-term complications in GSD-Ia.


Asunto(s)
Alelos , Modelos Animales de Enfermedad , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Homeostasis/genética , Fenotipo , Animales , Glucemia , Colesterol/sangre , Cartilla de ADN/genética , Componentes del Gen , Ácido Láctico/sangre , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Triglicéridos/sangre , Ácido Úrico/sangre
11.
J Hepatol ; 51(5): 909-17, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19376605

RESUMEN

BACKGROUND/AIMS: Glycogen storage disease type Ib (GSD-Ib) patients deficient in a glucose-6-phosphate transporter (G6PT) manifest disturbed glucose homeostasis, myeloid dysfunctions, and hepatocellular adenoma (HCA). This study was conducted to evaluate whether maintaining normoglycemia in GSD-Ib could prevent HCA. METHODS: We infused neonatal GSD-Ib mice with adeno-associated virus (AAV) carrying G6PT and examined their metabolic and myeloid phenotypes for the 72-week study. RESULTS: The AAV vector delivered the G6PT transgene to the liver and bone marrow. Long-term metabolic correction was achieved alongside a transient myeloid correction. Hepatic G6PT activity was 50% of wild-type levels at 2 weeks post-infusion but declined rapidly thereafter to reach 3% of wild-type levels by age 6 to 72 weeks. Despite this, the infused mice maintained normoglycemia throughout the study, exhibited near normal growth and normalized serum metabolite profiles. However, all five AAV-treated GSD-Ib mice that lived over 50 weeks accumulated excessive hepatic glycogen and fat. Two mice developed steatohepatitis and multiple HCAs with one undergoing malignant transformation. CONCLUSIONS: Normoglycemia alone cannot prevent hepatic steatosis and glycogen accumulation or the development of HCAs in GSD-Ib, providing one explanation why GSD-Ib patients maintaining normoglycemia under intense dietary therapy continue at risk for this long-term complication.


Asunto(s)
Adenoma de Células Hepáticas/prevención & control , Enfermedad del Almacenamiento de Glucógeno Tipo I/sangre , Enfermedad del Almacenamiento de Glucógeno Tipo I/complicaciones , Neoplasias Hepáticas/prevención & control , Adenoma de Células Hepáticas/etiología , Adenoma de Células Hepáticas/metabolismo , Adenoma de Células Hepáticas/patología , Animales , Animales Recién Nacidos , Antiportadores/genética , Antiportadores/metabolismo , Glucemia/metabolismo , Médula Ósea/enzimología , Dependovirus/genética , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/prevención & control , Terapia Genética , Vectores Genéticos , Enfermedad del Almacenamiento de Glucógeno Tipo I/terapia , Humanos , Hígado/enzimología , Hígado/metabolismo , Hígado/patología , Glucógeno Hepático/metabolismo , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
Mol Genet Metab ; 96(1): 32-7, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19008136

RESUMEN

Glycogen storage disease type Ib is caused by deficiencies in the glucose-6-phosphate transporter (G6PT), a phosphate (P(i))-linked antiporter capable of homologous (P(i):P(i)) and heterologous (G6P:P(i)) exchanges similar to the bacterial hexose-6-phosphate transporter, UhpT. Protease protection and glycosylation scanning assays have suggested that G6PT is anchored to the endoplasmic reticulum by 10 transmembrane domains. However, recent homology modeling proposed that G6PT may contain 12 helices and that amino acids essential for the functions of UhpT also play important roles in G6PT. Site-directed mutagenesis and in vitro expression assays demonstrated that only one of the four residues critical for UhpT activity is essential in G6PT. Furthermore, glycosylation scanning and protease sensitivity assays showed that the 10-domain model of G6PT is more probable than the 12-domain UhpT-like model.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Proteínas de Transporte de Monosacáridos/química , Proteínas de Transporte de Monosacáridos/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Células COS , Chlorocebus aethiops , Glucosa-6-Fosfato/metabolismo , Humanos , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/genética , Mutación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad
13.
Mol Genet Metab ; 95(4): 220-3, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18835800

RESUMEN

The glucose-6-phosphate transporter (G6PT) deficient in glycogen storage disease type Ib is a phosphate (P(i))-linked antiporter capable of G6P: P(i) and P(i):P(i) exchanges. We previously characterized G6PT mutations by measuring G6P uptake activities in microsomes co-expressing G6PT and glucose-6-phosphatase-alpha. Here we report a new assay, based on reconstituted proteoliposomes carrying only G6PT, and characterize G6P and P(i) uptake activities of 23 G6PT mutations. We show that co-expression and G6PT-only assays are equivalent in measuring G6PT activity. However, the p.Q133P mutation exhibits differential G6P and P(i) transport activities, suggesting that characterizing G6P and P(i) transport activities of G6PT mutations may yield insights to this genetic disorder.


Asunto(s)
Glucosa-6-Fosfato/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Mutación , Animales , Transporte Biológico , Células COS , Chlorocebus aethiops , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Humanos , Microsomas/metabolismo , Fosfatos/metabolismo
14.
FASEB J ; 22(7): 2206-13, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18337460

RESUMEN

Glycogen storage disease type Ib (GSD-Ib) is caused by deficiencies in the glucose-6-phosphate (G6P) transporter (G6PT) that have been well characterized. Interestingly, deleterious mutations in the G6PT gene were identified in clinical cases of GSD type Ic (GSD-Ic) proposed to be deficient in an inorganic phosphate (P(i)) transporter. We hypothesized that G6PT is both the G6P and P(i) transporter. Using reconstituted proteoliposomes we show that both G6P and P(i) are efficiently taken up into P(i)-loaded G6PT-proteoliposomes. The G6P uptake activity decreases as the internal:external P(i) ratio decreases and the P(i) uptake activity decreases in the presence of external G6P. Moreover, G6P or P(i) uptake activity is not detectable in P(i)-loaded proteoliposomes containing the p.R28H G6PT null mutant. The G6PT-proteoliposome-mediated G6P or P(i) uptake is inhibited by cholorgenic acid and vanadate, both specific G6PT inhibitors. Glucose-6-phosphatase-alpha (G6Pase-alpha), which facilitates microsomal G6P uptake by G6PT, fails to stimulate G6P uptake in P(i)-loaded G6PT-proteoliposomes, suggesting that the G6Pase-alpha-mediated stimulation is caused by decreasing G6P and increasing P(i) concentrations in microsomes. Taken together, our results suggest that G6PT has a dual role as a G6P and a P(i) transporter and that GSD-Ib and GSD-Ic are deficient in the same G6PT gene.


Asunto(s)
Antiportadores/genética , Antiportadores/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Animales , Antiportadores/deficiencia , Transporte Biológico , Células COS , Chlorocebus aethiops , Haplorrinos , Membranas Intracelulares/metabolismo , Proteínas de la Membrana/metabolismo , Microsomas/metabolismo , Proteínas de Transporte de Monosacáridos/deficiencia , Monoéster Fosfórico Hidrolasas/metabolismo , Proteolípidos/metabolismo , Solubilidad
15.
J Clin Invest ; 117(3): 784-93, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17318259

RESUMEN

Neutropenia and neutrophil dysfunction are common in many diseases, although their etiology is often unclear. Previous views held that there was a single ER enzyme, glucose-6-phosphatase-alpha (G6Pase-alpha), whose activity--limited to the liver, kidney, and intestine--was solely responsible for the final stages of gluconeogenesis and glycogenolysis, in which glucose-6-phosphate (G6P) is hydrolyzed to glucose for release to the blood. Recently, we characterized a second G6Pase activity, that of G6Pase-beta (also known as G6PC), which is also capable of hydrolyzing G6P to glucose but is ubiquitously expressed and not implicated in interprandial blood glucose homeostasis. We now report that the absence of G6Pase-beta led to neutropenia; defects in neutrophil respiratory burst, chemotaxis, and calcium flux; and increased susceptibility to bacterial infection. Consistent with this, G6Pase-beta-deficient (G6pc3-/-) mice with experimental peritonitis exhibited increased expression of the glucose-regulated proteins upregulated during ER stress in their neutrophils and bone marrow, and the G6pc3-/- neutrophils exhibited an enhanced rate of apoptosis. Our results define a molecular pathway to neutropenia and neutrophil dysfunction of previously unknown etiology, providing a potential model for the treatment of these conditions.


Asunto(s)
Infecciones Bacterianas/genética , Glucosa-6-Fosfatasa/genética , Neutropenia/genética , Neutrófilos/inmunología , Peritonitis/genética , Subunidades de Proteína/genética , Animales , Infecciones Bacterianas/enzimología , Infecciones Bacterianas/inmunología , Glucemia/análisis , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Glucosa-6-Fosfatasa/análisis , Glucosa-6-Fosfatasa/metabolismo , Glucosa-6-Fosfato/metabolismo , Hematopoyesis/genética , Homeostasis , Ratones , Neutropenia/enzimología , Neutrófilos/enzimología , Peritonitis/enzimología , Peritonitis/microbiología , Subunidades de Proteína/análisis , Subunidades de Proteína/metabolismo
16.
Mol Genet Metab ; 89(3): 233-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16777453

RESUMEN

Glycogen storage disease type Ia (GSD-Ia) is characterized by hypercholesterolemia, hypertriglyceridemia, decreased cholesterol in high density lipoprotein and increased cholesterol in low and very low density lipoprotein fractions. Despite this pro-atherogenic lipid profile, GSD-Ia patients are not at elevated risk for atherosclerosis. Studies have shown that reverse cholesterol transport and antioxidant capacity can be protective against atherosclerosis. In this study, we show that sera from GSD-Ia patients are more efficient than sera from control subjects in promoting the scavenger receptor class B type I (SR-BI)-mediated cellular cholesterol efflux, a key component in reverse cholesterol transport. The major determinants of the SR-BI-mediated cholesterol efflux are serum phospholipid (PL) and HDL-PL. Phospholipid and that ratio of HDL-PL to HDL are increased in GSD-Ia patients. We further show that sera from GSD-Ia patients have increased total antioxidant capacity compared to controls and this increase correlates with elevated levels of uric acid, a powerful plasma antioxidant. Taken together, the results suggest that the increase in SR-BI-mediated cellular cholesterol efflux and antioxidant capacity in the sera of GSD-Ia patients may contribute to protection against premature atherosclerosis.


Asunto(s)
Antioxidantes/metabolismo , Colesterol/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/sangre , Receptores Depuradores de Clase B/metabolismo , Adolescente , Adulto , Aterosclerosis/complicaciones , Niño , Preescolar , Colesterol/sangre , Enfermedad del Almacenamiento de Glucógeno Tipo I/complicaciones , Humanos
17.
FEBS Lett ; 579(21): 4713-8, 2005 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-16098970

RESUMEN

Glycogen storage disease type Ia (GSD-Ia) patients manifest a pro-atherogenic lipid profile but are not at elevated risk for developing atherosclerosis. Serum phospholipid, which correlates positively with the scavenger receptor class B type I (SR-BI)-mediated cholesterol efflux, and apolipoprotein A-IV and E, acceptors for ATP-binding cassette transporter A1 (ABCA1)-mediated cholesterol transport, are increased in GSD-Ia mice. Importantly, sera from GSD-Ia mice are more efficient than sera from control littermates in promoting SR-BI- and ABCA1-mediated cholesterol effluxes. As the first step in reverse cholesterol transport, essential for cholesterol homeostasis, these observations provide one explanation why GSD-Ia patients are apparently protected against premature atherosclerosis.


Asunto(s)
Arteriosclerosis/metabolismo , Colesterol/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Animales , Apolipoproteína A-I/metabolismo , Apolipoproteínas A/genética , Apolipoproteínas A/metabolismo , Arteriosclerosis/prevención & control , Línea Celular , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfatasa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Lípidos/sangre , Macrófagos/metabolismo , Ratones , Ratones Noqueados
18.
Mol Genet Metab ; 83(3): 276-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15542400

RESUMEN

Glycogen storage disease type Ia (GSD-Ia) is caused by deleterious mutations in the glucose-6-phosphatase gene (G6PC). A molecular study of this gene was carried out in 11 Argentinean patients from 8 unrelated families. Four missense (p.Gln54Pro, p.Arg83Cys, p.Thr16Arg, and p.Tyr209Cys) and one deletion (c.79delC) mutations have been identified. Two novel mutations, p.Thr16Arg (c.47C>G) located within the amino-terminal domain and p.Tyr209Cys (c.626A>G) situated in the sixth transmembrane helix, were uncovered in this study. Site-directed mutagenesis and transient expression assays demonstrated that both p.Thr16Arg and p.Tyr209Cys mutations abolished enzymatic activity as well as reduced G6Pase stability.


Asunto(s)
Eliminación de Gen , Expresión Génica , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Mutación Missense/genética , Animales , Argentina , Western Blotting , Células COS , Cricetinae , Cricetulus , Análisis Mutacional de ADN , Cartilla de ADN , Femenino , Glucosa-6-Fosfatasa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Humanos , Masculino , Mutagénesis Sitio-Dirigida , Monoéster Fosfórico Hidrolasas/metabolismo
19.
J Biol Chem ; 279(25): 26215-9, 2004 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-15087461

RESUMEN

The breakdown of tissue glycogen into glucose is critical for blood glucose homeostasis between meals. In the final steps of glycogenolysis, intracellular glucose 6-phosphate (Glc-6-P) is transported into the endoplasmic reticulum where it is hydrolyzed to glucose by glucose-6-phosphatase (Glc-6-Pase). Although the majority of body glycogen is stored in the muscle, the current dogma holds that Glc-6-Pase (now named Glc-6-Pase-alpha) is expressed only in the liver, kidney, and intestine, implying that muscle glycogen cannot contribute to interprandial blood glucose homeostasis. Recently we reported a second Glc-6-P hydrolase, Glc-6-Pase-beta. Glc-6-Pase-beta shares kinetic and structural similarities to Glc-6-Pase-alpha and couples with the Glc-6-P transporter to form an active Glc-6-Pase complex (Shieh, J.-J., Pan, C.-J., Mansfield, B. C., and Chou, J. Y. (2003) J. Biol. Chem. 278, 47098-47103). Here we demonstrate that muscle expresses both Glc-6-Pase-beta and Glc-6-P transporter and that they can couple to form an active Glc-6-Pase complex. Our data suggest that muscle may have a previously unrecognized role in interprandial glucose homeostasis.


Asunto(s)
Glucemia/fisiología , Glucosa-6-Fosfatasa/biosíntesis , Músculos/enzimología , Músculos/fisiología , Adenoviridae/genética , Animales , Northern Blotting , Western Blotting , Células COS , ADN Complementario/metabolismo , Glucosa/metabolismo , Glucosa-6-Fosfatasa/química , Glucosa-6-Fosfato/metabolismo , Glucógeno/metabolismo , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Hígado/metabolismo , Ratones , Músculo Esquelético/metabolismo , Músculos/metabolismo , Unión Proteica , Factores de Tiempo
20.
FEBS Lett ; 562(1-3): 160-4, 2004 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-15044018

RESUMEN

The islet-specific glucose-6-phosphatase-related protein (IGRP) has no known catalytic activity, but is of interest because it is the source of the peptide autoantigen targeted by a prevalent population of pathogenic CD8(+) T cells in non-obese diabetic mice. To better understand the potential roles of this protein in diabetes mellitus, we examine the subcellular localization and membrane topography of human IGRP. We show that IGRP is a glycoprotein, held in the endoplasmic reticulum by nine transmembrane domains, which is degraded in cells predominantly through the proteasome pathway that generates the major histocompatibility complex class I-presented peptides.


Asunto(s)
Acetilcisteína/análogos & derivados , Diabetes Mellitus/metabolismo , Retículo Endoplásmico/metabolismo , Glucosa-6-Fosfatasa , Glicoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas/metabolismo , Acetilcisteína/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Calreticulina/metabolismo , Cisteína Endopeptidasas/metabolismo , Retículo Endoplásmico/química , Inhibidores Enzimáticos/metabolismo , Glicoproteínas/química , Glicoproteínas/genética , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Complejos Multienzimáticos/antagonistas & inhibidores , Complejos Multienzimáticos/metabolismo , Complejo de la Endopetidasa Proteasomal , Estructura Secundaria de Proteína , Proteínas/química , Proteínas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...