Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Diabetes Investig ; 15(4): 429-436, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38243676

ABSTRACT

AIMS/INTRODUCTION: Glucagon plays an essential role in hepatic glucogenesis by enhancing glycogen breakdown, inducing gluconeogenesis, and suppressing glycogenesis. Moreover, glucagon increases cyclic adenosine monophosphate (cAMP) levels, thereby activating protein kinase A (PKA) and cAMP guanine nucleotide exchange factor (also known as Epac). Although the function of PKA in the liver has been studied extensively, the function of hepatic Epac is poorly understood. The aim of this study was to elucidate the role of Epac in mediating the action of glucagon on the hepatocytes. MATERIALS AND METHODS: Epac mRNA and protein expression, localization, and activity in the hepatocytes were analyzed by reverse transcription polymerase chain reaction, western blotting, immunofluorescence staining, and Rap1 activity assay, respectively. Additionally, we investigated the effects of an Epac-specific activator, 8-CPT, and an Epac-specific inhibitor, ESI-05, on glycogen metabolism in isolated rat hepatocytes. Further mechanisms of glycogen metabolism were evaluated by examining glucokinase (GK) translocation and mRNA expression of gluconeogenic enzymes. RESULTS: Epac2, but not Epac1, was predominantly expressed in the liver. Moreover, 8-CPT inhibited glycogen accumulation and GK translocation and enhanced the mRNA expression of gluconeogenic enzymes. ESI-05 failed to reverse glucagon-induced suppression of glycogen storage and partially inhibited glucagon-induced GK translocation and the mRNA expression of gluconeogenic enzymes. CONCLUSIONS: Epac signaling plays a role in mediating the glucogenic action of glucagon in the hepatocytes.


Subject(s)
Benzene Derivatives , Glucagon , Hepatocytes , Sulfones , Rats , Animals , Glucagon/metabolism , Hepatocytes/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , RNA, Messenger/metabolism , Glycogen/metabolism
2.
Br J Nutr ; 130(11): 1852-1858, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38713062

ABSTRACT

D-allulose, D-sorbose and D-tagatose are D-fructose isomers that are called rare sugars. These rare sugars have been studied intensively in terms of biological production and food application as well as physiological effects. There are limited papers with regard to the transporters mediating the intestinal absorption of these rare sugars. We examined whether these rare sugars are absorbed via sodium-dependent glucose cotransporter 1 (SGLT1) as well as via GLUT type 5 (GLUT5) using rats. High-fructose diet fed rats, which express more intestinal GLUT5, exhibited significantly higher peripheral concentrations, Cmax and AUC0­180 min when D-allulose, D-sorbose and D-tagatose were orally administrated. KGA-2727, a selective SGLT1 inhibitor, did not affect the peripheral and portal vein concentrations and pharmacokinetic parameters of these rare sugars. The results suggest that D-allulose, D-sorbose and D-tagatose are likely transported via GLUT5 but not SGLT1 in rat small intestine.


Subject(s)
Fructose , Glucose Transporter Type 5 , Glycosides , Hexoses , Intestinal Absorption , Sodium-Glucose Transporter 1 , Sorbose , Animals , Sodium-Glucose Transporter 1/metabolism , Male , Rats , Glucose Transporter Type 5/metabolism , Sorbose/metabolism , Rats, Sprague-Dawley , Rats, Wistar
3.
Physiol Rep ; 10(9): e15297, 2022 05.
Article in English | MEDLINE | ID: mdl-35546434

ABSTRACT

This study investigated the combined effects of exercise training and D-allulose intake on endurance capacity in mice. Male C57BL/6J mice were fed either a control diet (Con) or a 3% D-allulose diet (Allu) and further divided into the sedentary (Sed) or exercise training (Ex) groups (Con-Sed, Con-Ex, Allu-Sed, Allu-Ex, respectively; n = 6-7/group). The mice in the Ex groups were trained on a motor-driven treadmill 5 days/week for 4 weeks (15-18 m/min, 60 min). After the exercise training period, all mice underwent an exhaustive running test to assess their endurance capacity. At 48 h after the running test, the mice in the Ex groups were subjected to run at 18 m/min for 60 min again. Then the gastrocnemius muscle and liver were sampled immediately after the exercise bout. The running time until exhaustion tended to be higher in the Allu-Ex than in the Con-Ex group (p = 0.08). The muscle glycogen content was significantly lower in the Con-Ex than in the Con-Sed group and was significantly higher in the Allu-Ex than in the Con-Ex group (p < 0.05). Moreover, exercise training increased the phosphorylation levels of adenosine monophosphate-activated protein kinase (AMPK) in the muscle and liver. The phosphorylation levels of acetyl coenzyme A carboxylase (ACC), a downstream of AMPK, in the muscle and liver were significantly higher in the Allu-Ex than in the Con-Sed group (p < 0.05), suggesting that the combination of exercise training and D-allulose might have activated the AMPK-ACC signaling pathway, which is associated with fatty acid oxidation in the muscle and liver. Taken together, our data suggested the combination of exercise training and D-allulose intake as an effective strategy to upregulate endurance capacity in mice. This may be associated with sparing glycogen content and enhancing activation of AMPK-ACC signaling in the skeletal muscle.


Subject(s)
AMP-Activated Protein Kinases , Physical Conditioning, Animal , AMP-Activated Protein Kinases/metabolism , Animals , Fructose/metabolism , Glycogen/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Physical Conditioning, Animal/physiology
4.
Nutrients ; 14(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35276765

ABSTRACT

d-Allulose, a rare sugar, improves glucose metabolism and has been proposed as a candidate calorie restriction mimetic. This study aimed to investigate the effects of d-allulose on aerobic performance and recovery from exhaustion and compared them with the effects of exercise training. Male C57BL/6J mice were subjected to exercise and allowed to run freely on a wheel. Aerobic performance was evaluated using a treadmill. Glucose metabolism was analyzed by an intraperitoneal glucose tolerance test (ipGTT). Skeletal muscle intracellular signaling was analyzed by Western blotting. Four weeks of daily oral administration of 3% d-allulose increased running distance and shortened recovery time as assessed by an endurance test. d-Allulose administration also increased the maximal aerobic speed (MAS), which was observed following treatment for >3 or 7 days. The improved performance was associated with lower blood lactate levels and increased liver glycogen levels. Although d-allulose did not change the overall glucose levels as determined by ipGTT, it decreased plasma insulin levels, indicating enhanced insulin sensitivity. Finally, d-allulose enhanced the phosphorylation of AMP-activated protein kinase and acetyl-CoA carboxylase and the expression of peroxisome proliferator-activated receptor γ coactivator 1α. Our results indicate that d-allulose administration enhances endurance ability, reduces fatigue, and improves insulin sensitivity similarly to exercise training. d-Allulose administration may be a potential treatment option to alleviate obesity and enhance aerobic exercise performance.


Subject(s)
Fructose , Insulin Resistance , Animals , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL
5.
Metabol Open ; 11: 100112, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34381987

ABSTRACT

d-Allose is the C3 epimer of d-glucose and has been reported to have beneficial health effects. The transporter mediating intestinal transport of d-allose is unknown. We examined whether d-allose is absorbed via sodium-dependent glucose cotransporter 1 (SGLT1) as well as via glucose transporter type 5 (GLUT5) using rats. For examination of absorption via SGLT1, KGA-2727, an SGLT1-specific inhibitor, and d-allose were orally administered. KGA-2727 blocked the increase of plasma d-allose levels and suppressed them throughout the experiment (0-180 min), whereas without KGA-2727, the plasma d-allose levels peaked at around 60-90 min. For examination of absorption via GLUT5, rats were fed a high-fructose diet for 3weeks to increase the abundance and activity of GLUT5 in the small intestine. High-fructose diet-fed rats did not exhibit significant changes in the plasma d-allose levels compared to control rats fed a high-glucose diet. These results indicate that SGLT1 but not GLUT5 mediates the intestinal absorption of d-allose.

6.
Food Chem ; 277: 604-608, 2019 Mar 30.
Article in English | MEDLINE | ID: mdl-30502192

ABSTRACT

d-Allulose has been reported to have beneficial health effects. However, the transport system(s) mediating intestinal d-allulose transport has not yet been clearly identified. The aim of this study was to investigate whether intestinal d-allulose transport is mediated by glucose transporter type 5 (GLUT5). When d-allulose alone was gavaged, plasma d-allulose levels were dramatically higher in rats previously fed fructose. This suggests enhanced intestinal d-allulose absorption paralleled increases in GLUT5 expression observed only in fructose-fed rats. When d-allulose was gavaged with d-fructose, previously observed increases in plasma d-allulose levels were dampened and delayed, indicating d-fructose inhibited transepithelial d-allulose transport into plasma. Tracer D-[14C]-fructose uptake rate was reduced to 54.8% in 50 mM d-allulose and to 16.4% in 50 mM d-fructose, suggesting d-allulose competed with D-[14C]-fructose and the affinity of d-allulose for GLUT5 was lower than that of d-fructose. GLUT5 clearly mediates, likely at lower affinity relative to d-fructose, intestinal d-allulose transport.


Subject(s)
Fructose/metabolism , Glucose Transporter Type 5/metabolism , Intestine, Small/enzymology , Animals , Biological Transport , Blood Glucose , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/metabolism , Fructose/blood , Glucose/analysis , Glucose/metabolism , Glucose Transporter Type 5/genetics , Male , Rats , Rats, Sprague-Dawley , Substrate Specificity
7.
J Agric Food Chem ; 65(13): 2888-2894, 2017 Apr 05.
Article in English | MEDLINE | ID: mdl-28209058

ABSTRACT

Ingestion of high-fructose corn syrup (HFCS) is associated with the risk of both diabetes and obesity. Rare sugar syrup (RSS) has been developed by alkaline isomerization of HFCS and has anti-obesity and anti-diabetic effects. However, the influence of RSS on glucose metabolism has not been explored. We investigated whether long-term administration of RSS maintains glucose tolerance and whether the underlying mechanism involves hepatic glucokinase translocation. Wistar rats were administered water, RSS, or HFCS in drinking water for 10 weeks and then evaluated for glucose tolerance, insulin tolerance, liver glycogen content, and subcellular distribution of liver glucokinase. RSS significantly suppressed body weight gain and abdominal fat mass (p < 0.05). The glucose tolerance test revealed significantly higher blood glucose levels in the HFCS group compared to the water group, whereas the RSS group had significantly lower blood glucose levels from 90 to 180 min (p < 0.05). At 30, 60, and 90 min, the levels of insulin in the RSS group were significantly lower than those in the water group (p < 0.05). The amount of hepatic glycogen was more than 3 times higher in the RSS group than that in the other groups. After glucose loading, the nuclear export of glucokinase was significantly increased in the RSS group compared to the water group. These results imply that RSS maintains glucose tolerance and insulin sensitivity, at least partly, by enhancing nuclear export of hepatic glucokinase.


Subject(s)
Blood Glucose/metabolism , Fructose/analysis , Glucokinase/metabolism , High Fructose Corn Syrup/analysis , Insulin Resistance , Liver/enzymology , Animals , Biological Transport , Fructose/metabolism , Glucose Tolerance Test , High Fructose Corn Syrup/metabolism , Insulin/metabolism , Liver/metabolism , Male , Rats , Rats, Wistar
8.
Pharmacol Ther ; 155: 49-59, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26297965

ABSTRACT

Obesity and type 2 diabetes mellitus (T2DM) are the leading worldwide risk factors for mortality. The inextricably interlinked pathological progression from excessive weight gain, obesity, and hyperglycemia to T2DM, usually commencing from obesity, typically originates from overconsumption of sugar and high-fat diets. Although most patients require medications, T2DM is manageable or even preventable with consumption of low-calorie diet and maintaining body weight. Medicines like insulin, metformin, and thiazolidinediones that improve glycemic control; however, these are associated with weight gain, high blood pressure, and dyslipidemia. These situations warrant the attentive consideration of the role of balanced foods. Recently, we have discovered advantages of a rare sugar, D-allulose, a zero-calorie functional sweetener having strong anti-hyperlipidemic and anti-hyperglycemic effects. Study revealed that after oral administration in rats D-allulose readily entered the blood stream and was eliminated into urine within 24h. Cell culture study showed that D-allulose enters into and leaves the intestinal enterocytes via glucose transporters GLUT5 and GLUT2, respectively. In addition to D-allulose's short-term effects, the characterization of long-term effects has been focused on preventing commencement and progression of T2DM in diabetic rats. Human trials showed that D-allulose attenuates postprandial glucose levels in healthy subjects and in borderline diabetic subjects. The anti-hyperlipidemic effect of D-allulose, combined with its anti-inflammatory actions on adipocytes, is beneficial for the prevention of both obesity and atherosclerosis and is accompanied by improvements in insulin resistance and impaired glucose tolerance. Therefore, this review presents brief discussions focusing on physiological functions and potential benefits of D-allulose on obesity and T2DM.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Fructose/therapeutic use , Hypoglycemic Agents/therapeutic use , Hypolipidemic Agents/therapeutic use , Obesity/drug therapy , Animals , Anti-Inflammatory Agents/pharmacokinetics , Diabetes Mellitus, Type 2/metabolism , Drug Monitoring , Fructose/pharmacokinetics , Glucose/metabolism , Humans , Hypoglycemic Agents/pharmacokinetics , Hypolipidemic Agents/pharmacokinetics , Liver/metabolism , Obesity/metabolism
9.
Article in English | MEDLINE | ID: mdl-24106476

ABSTRACT

We previously reported that the type 2 diabetic Goto-Kakizaki (GK) rats at young adult ages (6-12 weeks) exhibited increased visceral fat mass and hyperleptinemia, due to hyperphagia caused primarily by neuropeptide Y (NPY) overexpression in the hypothalamic arcuate nucleus. Later, we found that GK rats continued to exhibit mesenteric fat accumulation and hyperleptinemia at least until 26 weeks of age, while hyperphagia and NPY overexpression ceased at 15 weeks of age. Therefore, we hypothesized that the long-lasting fat accumulation and hyperleptinemia are due to unidentified brain dysfunction other than NPY overexpression. In GK rats aged 26 weeks, glucose transporter-2 (GLUT2) mRNA expression in ventromedial hypothalamus (VMH) was markedly reduced in parallel with significant decreases in brain-derived neurotrophic factor (BDNF) mRNA level and BDNF-expressing cell numbers in the VMH. Pharmacologic inhibition of glucose utilization reduced BDNF mRNA expression in VMH in vivo and in vitro. The results suggested that impaired glucose utilization caused the reduction of BDNF. On the other hand, intracerebroventricular injection of BDNF for 6 days ameliorated hyperleptinemia in a long-lasting manner concurrently with feeding suppression in GK rats. Restricted feeding paired to BDNF-treated rats reduced plasma leptin level only transiently. BDNF treatment also reduced mesenteric fat mass in GK rats. These results reveal a novel action mode of BDNF to long-lastingly counteract visceral adiposity and hyperleptinemia in addition to and independently of its anorexigenic action. These results suggest that visceral fat accumulation and hyperleptinemia are at least partly due to the reduction of BDNF in VMH primarily caused by impaired glucose utilization in GK rats. The BDNF supplementation could provide an effective treatment of visceral obesity, hyperleptinemia and leptin resistance in type 2 diabetes.

10.
J Diabetes Investig ; 3(5): 432-40, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-24843603

ABSTRACT

UNLABELLED: Aims/Introduction: Excessive intake of sucrose can cause severe health issues, such as diabetes mellitus. In animal studies, consumption of a high-sucrose diet (SUC) has been shown to cause obesity, insulin resistance and glucose intolerance. However, several in vivo experiments have been carried out using diets with much higher sucrose contents (50-70% of the total calories) than are typically ingested by humans. In the present study, we examined the effects of a moderate SUC on glucose metabolism and the underlying mechanism. MATERIALS AND METHODS: C57BL/6J mice received a SUC (38.5% sucrose), a high-starch diet (ST) or a control diet for 5 weeks. We assessed glucose tolerance, incretin secretion and liver glucose metabolism. RESULTS: An oral glucose tolerance test (OGTT) showed that plasma glucose levels in the early phase were significantly higher in SUC-fed mice than in ST-fed or control mice, with no change in plasma insulin levels at any stage. SUC-fed mice showed a significant improvement in insulin sensitivity. Glucagon-like peptide-1 (GLP-1) secretion 15 min after oral glucose administration was significantly lower in SUC-fed mice than in ST-fed or control mice. Hepatic glucokinase (GCK) activity was significantly reduced in SUC-fed mice. During the OGTT, the accumulation of glycogen in the liver was suppressed in SUC-fed mice in a time-dependent manner. CONCLUSIONS: These results indicate that mice that consume a moderate SUC show glucose intolerance with a reduction in hepatic GCK activity and impairment in GLP-1 secretion. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2012.00208.x, 2012).

11.
Biochem Biophys Res Commun ; 405(1): 7-12, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21187061

ABSTRACT

A rare sugar, D-psicose has progressively been evaluated as a unique metabolic regulator of glucose and lipid metabolism, and thus represents a promising compound for the treatment of type 2 diabetes mellitus (T2DM). The present study was undertaken to examine the underlying effector organs of D-psicose in lowering blood glucose and abdominal fat by exploiting a T2DM rat model, Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Rats were fed 5% D-psicose or 5% D-glucose supplemented in drinking water, and only water in the control for 13 weeks and the protective effects were compared. A non-diabetic Long-Evans Tokushima Otsuka (LETO), fed with water served as a counter control of OLETF. After 13 weeks feeding, D-psicose treatment significantly reduced the increase in body weight and abdominal fat mass. Oral glucose tolerance test (OGTT) showed the reduced blood glucose and insulin levels suggesting the improvement of insulin resistance in OLETF rats. Oil-red-O staining elucidated that D-psicose significantly reduced lipid accumulation in the liver. Immunohistochemical analysis showed D-psicose induced glucokinase translocation from nucleus to cytoplasm of the liver which enhances glucokinase activity and subsequent synthesis of glycogen in the liver. D-psicose also protected the pathological change of the ß-cells of pancreatic islets. These data demonstrate that D-psicose controls blood glucose levels by reducing lipotoxicity in liver and by preserving pancreatic ß-cell function.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Fructose/therapeutic use , Insulin Resistance , Abdominal Fat/drug effects , Abdominal Fat/pathology , Animals , Body Weight/drug effects , Cell Nucleus/enzymology , Cytoplasm/enzymology , Cytoprotection , Eating/drug effects , Fatty Liver/drug therapy , Fatty Liver/pathology , Glucokinase/metabolism , Glucose Tolerance Test , Homeostasis/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Lipid Metabolism/drug effects , Male , Pancreas/drug effects , Pancreas/pathology , Protein Transport/drug effects , Rats , Rats, Inbred OLETF
12.
J Clin Invest ; 117(1): 246-57, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200721

ABSTRACT

Glucokinase (Gck) functions as a glucose sensor for insulin secretion, and in mice fed standard chow, haploinsufficiency of beta cell-specific Gck (Gck(+/-)) causes impaired insulin secretion to glucose, although the animals have a normal beta cell mass. When fed a high-fat (HF) diet, wild-type mice showed marked beta cell hyperplasia, whereas Gck(+/-) mice demonstrated decreased beta cell replication and insufficient beta cell hyperplasia despite showing a similar degree of insulin resistance. DNA chip analysis revealed decreased insulin receptor substrate 2 (Irs2) expression in HF diet-fed Gck(+/-) mouse islets compared with wild-type islets. Western blot analyses confirmed upregulated Irs2 expression in the islets of HF diet-fed wild-type mice compared with those fed standard chow and reduced expression in HF diet-fed Gck(+/-) mice compared with those of HF diet-fed wild-type mice. HF diet-fed Irs2(+/-) mice failed to show a sufficient increase in beta cell mass, and overexpression of Irs2 in beta cells of HF diet-fed Gck(+/-) mice partially prevented diabetes by increasing beta cell mass. These results suggest that Gck and Irs2 are critical requirements for beta cell hyperplasia to occur in response to HF diet-induced insulin resistance.


Subject(s)
Dietary Fats/pharmacology , Glucokinase/physiology , Insulin Resistance/physiology , Insulin-Secreting Cells/pathology , Insulin/physiology , Intracellular Signaling Peptides and Proteins/physiology , Phosphoproteins/physiology , Animals , Glucokinase/deficiency , Glucokinase/genetics , Humans , Hyperplasia , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphoproteins/deficiency , Phosphoproteins/genetics , Signal Transduction
13.
Biol Pharm Bull ; 29(2): 216-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16462021

ABSTRACT

Glucokinase (GK) is known to be the critical glucose sensor of pancreatic B-cells. However, the localization and functional role of GK in the brain remains to be elucidated. In this study, we measured both the activity and mRNA level of GK in the hypothalamic nuclei and the cortex of rats injected intraperitoneally with streptozotocin or vehicle. GK activity was measured by a fluorometric assay; and the GK mRNA level, by use of the real-time reverse transcription polymerase chain reaction. GK activity in vehicle-treated rats was high in the arcuate nucleus, moderate or low in the ventromedial nucleus, lateral hypothalamic area, and paraventricular nucleus, and very low in the cortex. The order of GK mRNA level was almost the same as that of GK activity. GK activity and GK mRNA level only in the arcuate nucleus of streptozotocin-treated rats at 7 d, but not at 2 d, after treatment were lower than those of vehicle-treated rats. The results suggest that prolonged hyperglycemia induced by diabetes decreased the activity of GK in the arcuate nucleus.


Subject(s)
Arcuate Nucleus of Hypothalamus/enzymology , Cerebral Cortex/enzymology , Diabetes Mellitus, Experimental/enzymology , Glucokinase/metabolism , Animals , Blood Glucose/analysis , Blotting, Western , Glucokinase/biosynthesis , Insulin/blood , Male , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Streptozocin
14.
Hum Mol Genet ; 13(11): 1147-57, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15102714

ABSTRACT

Mutant mouse models are indispensable tools for clarifying the functions of genes and for elucidating the underlying pathogenic mechanisms of human diseases. Currently, several large-scale mutagenesis projects that employ the chemical mutagen N-ethyl-N-nitrosourea (ENU) are underway worldwide. One specific aim of our ENU mutagenesis project is to generate diabetic mouse models. We screened 9375 animals for dominant traits using a clinical biochemical test and thereby identified 11 mutations in the glucokinase (Gk) gene that were associated with hyperglycemia. GK is a key regulator of insulin secretion in the pancreatic beta-cell. Approximately 190 heterozygous mutations in the human GK gene have been reported to cause maturity onset diabetes of the young, type 2 (MODY2). In addition, five mutations have been reported to cause permanent neonatal diabetes mellitus (PNDM) when present on both alleles. The mutations in our 11 hyperglycemic mutants are located at different positions in Gk. Four have also been found in human MODY2 patients, and another mutant bears its mutation at the same location that is mutated in a PNDM patient. Thus, ENU mutagenesis is effective for developing mouse models for various human genetic diseases, including diabetes mellitus. Some of our Gk mutant lines displayed impaired glucose-responsive insulin secretion and the mutations had different effects on Gk mRNA levels and/or the stability of the GK protein. This collection of Gk mutants will be valuable for understanding GK gene function, for dissecting the function of the enzyme and as models of human MODY2 and PNDM.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Glucokinase/genetics , Mice, Mutant Strains , Amino Acid Sequence , Animals , Blood Glucose/analysis , Ethylnitrosourea , Female , Gene Expression , Glucose Tolerance Test , Homozygote , Insulin/administration & dosage , Insulin/metabolism , Insulin Resistance , Liver/pathology , Male , Mice , Molecular Sequence Data , Mutagenesis , Phenotype , Point Mutation , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...