Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Endocrinol ; 228(1): 61-73, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26493453

RESUMO

Type 2 diabetes (T2D) is associated with defective insulin secretion, which in turn contributes to worsening glycaemic control and disease progression. The genetic cause(s) associated with impaired insulin secretion in T2D are not well elucidated. Here we used the polygenic New Zealand Obese (NZO) mouse model, which displays all the cardinal features of T2D including hyperglycaemia to identify genes associated with ß-cell dysfunction. A genome-wide scan identified a major quantitative trait locus (QTL) on chromosome 7 associated with defective glucose-mediated insulin secretion. Using congenic strains, the locus was narrowed to two candidate genes encoding the components of the KATP channel: Abcc8 (SUR1) and Kcnj11 (Kir6.2). The NZO Abcc8 allele was associated with a ∼211 bp deletion in its transcript and reduced expression of SUR1. Transgenic NZO mice were generated that expressed the WT Abcc8/Kcnj11 genes and displayed significant improvements in early-phase glucose-mediated insulin secretion and glucose tolerance, confirming Abcc8 as a causative gene. Importantly, we showed that despite improving ß-cell function in the NZO transgenic mice, there was no enhancement of insulin sensitivity or body weight. This study provides evidence for a role of Abcc8 in early-phase glucose-mediated insulin secretion and validates this gene as a contributor to ß-cell dysfunction in T2D.


Assuntos
Diabetes Mellitus Tipo 2/genética , Insulina/metabolismo , Receptores de Sulfonilureias/genética , Animais , Glicemia/análise , Diabetes Mellitus Tipo 2/fisiopatologia , Feminino , Deleção de Genes , Genótipo , Glucose/farmacologia , Intolerância à Glucose , Teste de Tolerância a Glucose , Insulina/sangue , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Camundongos Transgênicos , Obesidade/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Locos de Características Quantitativas/genética
2.
Diabetes ; 61(5): 1122-32, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22517657

RESUMO

Liver fructose-1,6-bisphosphatase (FBPase) is a regulatory enzyme in gluconeogenesis that is elevated by obesity and dietary fat intake. Whether FBPase functions only to regulate glucose or has other metabolic consequences is not clear; therefore, the aim of this study was to determine the importance of liver FBPase in body weight regulation. To this end we performed comprehensive physiologic and biochemical assessments of energy balance in liver-specific transgenic FBPase mice and negative control littermates of both sexes. In addition, hepatic branch vagotomies and pharmacologic inhibition studies were performed to confirm the role of FBPase. Compared with negative littermates, liver-specific FBPase transgenic mice had 50% less adiposity and ate 15% less food but did not have altered energy expenditure. The reduced food consumption was associated with increased circulating leptin and cholecystokinin, elevated fatty acid oxidation, and 3-ß-hydroxybutyrate ketone levels, and reduced appetite-stimulating neuropeptides, neuropeptide Y and Agouti-related peptide. Hepatic branch vagotomy and direct pharmacologic inhibition of FBPase in transgenic mice both returned food intake and body weight to the negative littermates. This is the first study to identify liver FBPase as a previously unknown regulator of appetite and adiposity and describes a novel process by which the liver participates in body weight regulation.


Assuntos
Adiposidade/fisiologia , Apetite/fisiologia , Frutose-Bifosfatase/metabolismo , Fígado/enzimologia , Adiposidade/genética , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Apetite/genética , Colecistocinina/metabolismo , Relação Dose-Resposta a Droga , Ingestão de Alimentos , Metabolismo Energético , Ácidos Graxos/metabolismo , Feminino , Frutose-Bifosfatase/antagonistas & inibidores , Frutose-Bifosfatase/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Hidroxibutiratos , Corpos Cetônicos , Leptina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Oxirredução , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo
3.
Am J Physiol Endocrinol Metab ; 295(5): E1132-41, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18780768

RESUMO

Increased endogenous glucose production (EGP) predominantly from the liver is a characteristic feature of type 2 diabetes, which positively correlates with fasting hyperglycemia. Gluconeogenesis is the biochemical pathway shown to significantly contribute to increased EGP in diabetes. Fructose-1,6-bisphosphatase (FBPase) is a regulated enzyme in gluconeogenesis that is increased in animal models of obesity and insulin resistance. However, whether a specific increase in liver FBPase can result in increased EGP has not been shown. The objective of this study was to determine the role of upregulated liver FBPase in glucose homeostasis. To achieve this goal, we generated human liver FBPase transgenic mice under the control of the transthyretin promoter, using insulator sequences to flank the transgene and protect it from site-of-integration effects. This resulted in a liver-specific model, as transgene expression was not detected in other tissues. Mice were studied under the following conditions: 1) at two ages (24 wk and 1 yr old), 2) after a 60% high-fat diet, and 3) when bred to homozygosity. Hemizygous transgenic mice had an approximately threefold increase in total liver FBPase mRNA with concomitant increases in FBPase protein and enzyme activity levels. After high-fat feeding, hemizygous transgenics were glucose intolerant compared with negative littermates (P < 0.02). Furthermore, when bred to homozygosity, chow-fed transgenic mice showed a 5.5-fold increase in liver FBPase levels and were glucose intolerant compared with negative littermates, with a significantly higher rate of EGP (P < 0.006). This is the first study to show that FBPase regulates EGP and whole body glucose homeostasis in a liver-specific transgenic model. Our homozygous transgenic model may be useful for testing human FBPase inhibitor compounds with the potential to treat patients with type 2 diabetes.


Assuntos
Glicemia/metabolismo , Frutose-Bifosfatase/metabolismo , Glucose/metabolismo , Fígado/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Frutose-Bifosfatase/genética , Expressão Gênica , Intolerância à Glucose , Glucose-6-Fosfatase/metabolismo , Homozigoto , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Insulina/sangue , Resistência à Insulina , Fígado/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Ácido Pirúvico/metabolismo
4.
Endocrinology ; 147(6): 2764-72, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16497803

RESUMO

In type 2 diabetes, increased endogenous glucose production (EGP) as a result of elevated gluconeogenesis contributes to hyperglycemia. An increase in glycerol gluconeogenesis has led to the suggestion that, in obese human subjects with type 2 diabetes, there may be an increase in the activity of the gluconeogenic enzyme fructose-1,6-bisphosphatase (FBPase). The aim of this study was to generate transgenic mice that overexpress human liver FBPase in the liver and assess the consequences to whole-body and hepatic glucose metabolism. FBPase transgenic mice had significantly higher levels of transgene expression in the liver and, as a result, had increased FBPase protein and enzyme activity levels in the liver. This resulted in an increase in the rate of glycerol conversion to glucose but not in EGP. The increased expression of FBPase in the liver did not result in any significant differences compared with littermate control mice in insulin or glucose tolerance. Therefore, it appears that, on its own, an increase in FBPase does not lead to impaired regulation of EGP and hence does not affect whole-body glucose metabolism. This suggests that, for EGP to be increased, other factors associated with obesity are also required.


Assuntos
Frutose-Bifosfatase/fisiologia , Gluconeogênese , Glicerol/metabolismo , Fígado/enzimologia , Animais , Glicemia/análise , Feminino , Frutose-Bifosfatase/genética , Humanos , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Fosfoenolpiruvato Carboxiquinase (GTP)/fisiologia
5.
J Mol Endocrinol ; 35(1): 39-48, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16087720

RESUMO

Type 2 diabetes is characterized by islet dysfunction resulting in hyperglycemia, which can then lead to further deterioration in islet function. A possible mechanism for hyperglycemia-induced islet dysfunction is the accumulation of advanced glycation end products (AGE). The DBA/2 mouse develops pancreatic islet dysfunction when exposed to a high glucose environment and/or obesity-induced insulin resistance. To determine the biochemical cause of dysfunction, DBA/2 and C57BL/6 control islets were incubated in 11.1 mM or 40 mM glucose in the absence or presence of the AGE inhibitor aminoguanidine (AG) for 10 days. Basal (2.8 mM glucose) insulin release was increased in both DBA/2 and C57BL/6 islets incubated with 40 mM vs 11.1 mM glucose for 10 days. Chronic exposure to hyperglycemia decreased glucose (20 mM)-stimulated insulin secretion in DBA/2 but not in C57BL/6 islets. AG significantly increased fold-induced insulin release in high glucose cultured DBA/2 mouse islets, but did not affect C57BL/6 islet function. DBA/2 islet glucokinase was significantly reduced following 40 mM glucose culture, compared with 11.1 mM glucose cultured DBA/2 islets and 40 mM glucose cultured C57BL/6 islets. Incubation of islets with AG resulted in a normalization of DBA/2 islet glucokinase levels. In conclusion, chronic high glucose-induced increases in AGE can result in islet dysfunction and this is associated with reduced glucokinase levels in a mouse model with susceptibility to islet failure.


Assuntos
Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Glucoquinase/metabolismo , Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiopatologia , Animais , Sequência de Bases , Sobrevivência Celular , DNA/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Glucose/administração & dosagem , Transportador de Glucose Tipo 2 , Produtos Finais de Glicação Avançada/metabolismo , Hexoquinase/metabolismo , Técnicas In Vitro , Insulina/genética , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Proteínas de Transporte de Monossacarídeos/metabolismo , Proinsulina/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...