Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 28: 500-513, 2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35592498

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a promising therapeutic agent for treatment of type 2 diabetes (T2D) and non-alcoholic steatohepatitis (NASH). We show that therapeutic levels of FGF21 were achieved following subcutaneous (s.c.) administration of mRNA encoding human FGF21 proteins. The efficacy of mRNA was assessed following 2-weeks repeated s.c. dosing in diet-induced obese (DIO), mice which resulted in marked decreases in body weight, plasma insulin levels, and hepatic steatosis. Pharmacokinetic/pharmacodynamic (PK/PD) modelling of several studies in both lean and DIO mice showed that mRNA encoding human proteins provided improved therapeutic coverage over recombinant dosed proteins in vivo. This study is the first example of s.c. mRNA therapy showing pre-clinical efficacy in a disease-relevant model, thus, showing the potential for this modality in the treatment of chronic diseases, including T2D and NASH.

2.
Diabetologia ; 63(7): 1355-1367, 2020 07.
Article in English | MEDLINE | ID: mdl-32350565

ABSTRACT

AIMS/HYPOTHESIS: Inflammatory signals and increased prostaglandin synthesis play a role during the development of diabetes. The prostaglandin D2 (PGD2) receptor, GPR44/DP2, is highly expressed in human islets and activation of the pathway results in impaired insulin secretion. The role of GPR44 activation on islet function and survival rate during chronic hyperglycaemic conditions is not known. In this study, we investigate GPR44 inhibition by using a selective GPR44 antagonist (AZ8154) in human islets both in vitro and in vivo in diabetic mice transplanted with human islets. METHODS: Human islets were exposed to PGD2 or proinflammatory cytokines in vitro to investigate the effect of GPR44 inhibition on islet survival rate. In addition, the molecular mechanisms of GPR44 inhibition were investigated in human islets exposed to high concentrations of glucose (HG) and to IL-1ß. For the in vivo part of the study, human islets were transplanted under the kidney capsule of immunodeficient diabetic mice and treated with 6, 60 or 100 mg/kg per day of a GPR44 antagonist starting from the transplantation day until day 4 (short-term study) or day 17 (long-term study) post transplantation. IVGTT was performed on mice at day 10 and day 15 post transplantation. After termination of the study, metabolic variables, circulating human proinflammatory cytokines, and hepatocyte growth factor (HGF) were analysed in the grafted human islets. RESULTS: PGD2 or proinflammatory cytokines induced apoptosis in human islets whereas GPR44 inhibition reversed this effect. GPR44 inhibition antagonised the reduction in glucose-stimulated insulin secretion induced by HG and IL-1ß in human islets. This was accompanied by activation of the Akt-glycogen synthase kinase 3ß signalling pathway together with phosphorylation and inactivation of forkhead box O-1and upregulation of pancreatic and duodenal homeobox-1 and HGF. Administration of the GPR44 antagonist for up to 17 days to diabetic mice transplanted with a marginal number of human islets resulted in reduced fasting blood glucose and lower glucose excursions during IVGTT. Improved glucose regulation was supported by increased human C-peptide levels compared with the vehicle group at day 4 and throughout the treatment period. GPR44 inhibition reduced plasma levels of TNF-α and growth-regulated oncogene-α/chemokine (C-X-C motif) ligand 1 and increased the levels of HGF in human islets. CONCLUSIONS/INTERPRETATION: Inhibition of GPR44 in human islets has the potential to improve islet function and survival rate under inflammatory and hyperglycaemic stress. This may have implications for better survival rate of islets following transplantation.


Subject(s)
DNA-Binding Proteins/metabolism , Islets of Langerhans/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/metabolism , Transcription Factors/metabolism , Apoptosis/physiology , Blotting, Western , Cell Death/physiology , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion/physiology , Prostaglandin D2 , Real-Time Polymerase Chain Reaction
3.
EJNMMI Res ; 8(1): 113, 2018 Dec 27.
Article in English | MEDLINE | ID: mdl-30588560

ABSTRACT

BACKGROUND: The G-protein-coupled receptor 44 (GPR44) is a beta cell-restricted target that may serve as a marker for beta cell mass (BCM) given the development of a suitable PET ligand. METHODS: The binding characteristics of the selected candidate, AZ12204657, at human GPR44 were determined using in vitro ligand binding assays. AZ12204657 was radiolabeled using 11C- or 3H-labeled methyl iodide ([11C/3H]CH3I) in one step, and the conversion of [11C/3H]CH3I to the radiolabeled product [11C/3H]AZ12204657 was quantitative. The specificity of radioligand binding to GPR44 and the selectivity for beta cells were evaluated by in vitro binding studies on pancreatic sections from human and non-human primates as well as on homogenates from endocrine and exocrine pancreatic compartments. RESULTS: The radiochemical purity of the resulting radioligand [11C]AZ12204657 was > 98%, with high molar activity (MA), 1351 ± 575 GBq/µmol (n = 18). The radiochemical purity of [3H]AZ12204657 was > 99% with MA of 2 GBq/µmol. Pancreatic binding of [11C/3H]AZ12204657 was co-localized with insulin-positive islets of Langerhans in non-diabetic individuals and individuals with type 2 diabetes (T2D). The binding of [11C]AZ12204657 to GPR44 was > 10 times higher in islet homogenates compared to exocrine homogenates. In human islets of Langerhans GPR44 was co-expressed with insulin, but not glucagon as assessed by co-staining and confocal microscopy. CONCLUSION: We radiolabeled [11C]AZ12204657, a potential PET radioligand for the beta cell-restricted protein GPR44. In vitro evaluation demonstrated that [3H]AZ12204657 and [11C]AZ12204657 selectively target pancreatic beta cells. [11C]AZ12204657 has promising properties as a marker for human BCM.

4.
J Biol Chem ; 293(15): 5492-5508, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29414779

ABSTRACT

Neurturin (NRTN) provides trophic support to neurons and is considered a therapeutic agent for neurodegenerative diseases, such as Parkinson's disease. It binds to its co-receptor GFRa2, and the resulting NRTN-GFRa2 complex activates the transmembrane receptors rearranged during transfection (RET) or the neural cell adhesion molecule (NCAM). We report the crystal structure of NRTN, alone and in complex with GFRa2. This is the first crystal structure of a GFRa with all three domains and shows that domain 1 does not interact directly with NRTN, but it may support an interaction with RET and/or NCAM, via a highly conserved surface. In addition, biophysical results show that the relative concentration of GFRa2 on cell surfaces can affect the functional affinity of NRTN through avidity effects. We have identified a heparan sulfate-binding site on NRTN and a putative binding site in GFRa2, suggesting that heparan sulfate has a role in the assembly of the signaling complex. We further show that mutant NRTN with reduced affinity for heparan sulfate may provide a route forward for delivery of NRTN with increased exposure in preclinical in vivo models and ultimately to Parkinson's patients.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/chemistry , Heparitin Sulfate/chemistry , Multiprotein Complexes/chemistry , Neurturin/chemistry , Signal Transduction , Crystallography, X-Ray , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Heparitin Sulfate/metabolism , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurturin/genetics , Neurturin/metabolism , Protein Domains , Protein Structure, Quaternary
5.
PLoS One ; 12(12): e0189060, 2017.
Article in English | MEDLINE | ID: mdl-29206860

ABSTRACT

The mechanism behind the glucose lowering effect occurring after specific activation of GPR120 is not completely understood. In this study, a potent and selective GPR120 agonist was developed and its pharmacological properties were compared with the previously described GPR120 agonist Metabolex-36. Effects of both compounds on signaling pathways and GLP-1 secretion were investigated in vitro. The acute glucose lowering effect was studied in lean wild-type and GPR120 null mice following oral or intravenous glucose tolerance tests. In vitro, in GPR120 overexpressing cells, both agonists signaled through Gαq, Gαs and the ß-arrestin pathway. However, in mouse islets the signaling pathway was different since the agonists reduced cAMP production. The GPR120 agonists stimulated GLP-1 secretion both in vitro in STC-1 cells and in vivo following oral administration. In vivo GPR120 activation induced significant glucose lowering and increased insulin secretion after intravenous glucose administration in lean mice, while the agonists had no effect in GPR120 null mice. Exendin 9-39, a GLP-1 receptor antagonist, abolished the GPR120 induced effects on glucose and insulin following an intravenous glucose challenge. In conclusion, GLP-1 secretion is an important mechanism behind the acute glucose lowering effect following specific GPR120 activation.


Subject(s)
Blood Glucose/metabolism , Glucagon-Like Peptide 1/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , CHO Cells , Cell Line , Cricetulus , Cyclic AMP/biosynthesis , Female , GTP-Binding Proteins/metabolism , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , beta-Arrestins/metabolism
6.
Diabetes ; 66(7): 2007-2018, 2017 07.
Article in English | MEDLINE | ID: mdl-28408435

ABSTRACT

Neurturin (NRTN), a member of the glial-derived neurotrophic factor family, was identified from an embryonic chicken pancreatic cDNA library in a screen for secreted factors. In this study, we assessed the potential antidiabetic activities of NRTN relative to liraglutide, a glucagon-like peptide 1 receptor agonist, in Zucker diabetic fatty (ZDF) rats. Subcutaneous administration of NRTN to 8-week-old male ZDF rats prevented the development of hyperglycemia and improved metabolic parameters similar to liraglutide. NRTN treatment increased pancreatic insulin content and ß-cell mass and prevented deterioration of islet organization. However, unlike liraglutide-treated rats, NRTN-mediated improvements were not associated with reduced body weight or food intake. Acute NRTN treatment did not activate c-Fos expression in key feeding behavior and metabolic centers in ZDF rat brain or directly enhance glucose-stimulated insulin secretion from pancreatic ß-cells. Treating 10-week-old ZDF rats with sustained hyperglycemia with liraglutide resulted in some alleviation of hyperglycemia, whereas NRTN was not as effective despite improving plasma lipids and fasting glucose levels. Interestingly, coadministration of NRTN and liraglutide normalized hyperglycemia and other metabolic parameters, demonstrating that combining therapies with distinct mechanism(s) can alleviate advanced diabetes. This emphasizes that therapeutic combinations can be more effective to manage diabetes in individuals with uncontrolled hyperglycemia.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Liraglutide/pharmacology , Neurturin/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Disease Models, Animal , Eating/drug effects , Feeding Behavior/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Organ Size , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Zucker
7.
PLoS One ; 10(3): e0121204, 2015.
Article in English | MEDLINE | ID: mdl-25793295

ABSTRACT

Islet transplantation has become a viable clinical treatment, but is still compromised by long-term graft failure. Exendin-4, a glucagon-like peptide 1 receptor agonist, has in clinical studies been shown to improve insulin secretion in islet transplanted patients. However, little is known about the effect of exendin-4 on other metabolic parameters. We therefore aimed to determine what influence exendin-4 would have on revascularized minimal human islet grafts in a state of graft failure in terms of glucose metabolism, body weight, lipid levels and graft survival. Introducing the bilateral, subcapsular islet transplantation model, we first transplanted diabetic mice with a murine graft under the left kidney capsule sufficient to restore normoglycemia. After a convalescent period, we performed a second transplantation under the right kidney capsule with a minimal human islet graft and allowed for a second recovery. We then performed a left-sided nephrectomy, and immediately started treatment with exendin-4 with a low (20µg/kg/day) or high (200µg/kg/day) dose, or saline subcutaneously twice daily for 15 days. Blood was sampled, blood glucose and body weight monitored. The transplanted human islet grafts were collected at study end point and analyzed. We found that exendin-4 exerts its effect on failing human islet grafts in a bell-shaped dose-response curve. Both doses of exendin-4 equally and significantly reduced blood glucose. Glucagon-like peptide 1 (GLP-1), C-peptide and pro-insulin were conversely increased. In the course of the treatment, body weight and cholesterol levels were not affected. However, immunohistochemistry revealed an increase in beta cell nuclei count and reduced TUNEL staining only in the group treated with a low dose of exendin-4 compared to the high dose and control. Collectively, these results suggest that exendin-4 has a potential rescue effect on failing, revascularized human islets in terms of lowering blood glucose, maintaining beta cell numbers, and improving metabolic parameters during hyperglycemic stress.


Subject(s)
Graft Survival/drug effects , Islets of Langerhans Transplantation , Islets of Langerhans/blood supply , Peptides/pharmacology , Venoms/pharmacology , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , C-Peptide/metabolism , Cell Count , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/therapy , Exenatide , Fasting/blood , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , Humans , Hyperglycemia/blood , Hyperglycemia/complications , Hyperglycemia/drug therapy , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Male , Mice, Inbred BALB C , Models, Animal , Peptides/administration & dosage , Peptides/therapeutic use , Venoms/administration & dosage , Venoms/therapeutic use
8.
Am J Physiol Endocrinol Metab ; 303(11): E1287-95, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23032688

ABSTRACT

Changes in adipose tissue distribution and ectopic fat storage in, liver and skeletal muscle tissue impact whole body insulin sensitivity in both humans and experimental animals. Numerous mouse models of obesity, insulin resistance, and diabetes exist; however, current methods to assess mouse phenotypes commonly involve direct harvesting of the tissues of interest, precluding the possibility of repeated measurements in the same animal. In this study, we demonstrate that whole body 3-D imaging of body fat composition can be used to analyze distribution as well as redistribution of fat after intervention by repeated assessment of intrahepatocellular lipids (IHCL), intra-abdominal, subcutaneous, and total adipose tissue (IAT, SAT, and TAT) and brown adipose tissue (BAT). C57BL/6J mice fed a cafeteria diet for 16 wk were compared with mice fed standard chow for 16 wk and mice switched from café diet to standard chow after 12 wk. MRI determinations were made at 9 and 15 wk, and autopsy was performed at 16 wk. There was a strong correlation between MRI-calculated weights in vivo at 15 wk and measured weights at 16 wk ex vivo for IAT (r = 0.99), BAT (r = 0.93), and IHCL (r = 0.97). IHCL and plasma insulin increased steeply relative to body weight at body weights above 45 g. This study demonstrates that the use of 3-D imaging to assess body fat composition may allow substantial reductions in animal usage. The dietary interventions indicated that a marked metabolic deterioration occurred when the mice had gained a certain fat mass.


Subject(s)
Adipose Tissue/diagnostic imaging , Body Fat Distribution/instrumentation , Disease Models, Animal , Liver/diagnostic imaging , Obesity/diagnostic imaging , Adipose Tissue/metabolism , Animal Feed , Animals , Body Composition , Cross-Sectional Studies , Energy Metabolism/physiology , Female , Imaging, Three-Dimensional/veterinary , Insulin Resistance/physiology , Liver/metabolism , Longitudinal Studies , Magnetic Resonance Imaging/veterinary , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/metabolism , Phenotype , Radiography , Random Allocation , Triglycerides/blood
9.
Eur J Pharmacol ; 663(1-3): 80-6, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21586282

ABSTRACT

Glucokinase (GK) plays a key role in maintaining glucose homeostasis by promoting insulin secretion from pancreatic beta cells and increasing hepatic glucose uptake. Here we investigate the effects of acute and chronic GK activation on glucose tolerance and insulin secretion in mice with diet-induced insulin resistance. In the acute study, a small molecule GK activator (GKA71) was administered to mice fed a high-fat diet for 8 weeks. In the long-term study, GKA71 was provided in the diet for 4 weeks to high-fat diet-fed mice. Glucose tolerance was measured after intravenous glucose administration, and insulin secretion was measured both in vivo and in vitro. Acute GK activation efficiently improved glucose tolerance in association with increased insulin secretion after intravenous glucose both in control and high-fat fed mice. Chronic GK activation significantly reduced basal plasma glucose and insulin, and improved glucose tolerance despite reduced insulin secretion after intravenous glucose, suggesting improved insulin sensitivity. Isolated islets from chronically GKA71-treated mice displayed augmented insulin secretion at 8.3 mmol/l glucose, without affecting glucose oxidation. High-fat diet fed mice had reduced glycogen and increased triglyceride in liver compared to control mice, and these parameters were not altered by long-term GK activation. We conclude that GK activation in high-fat diet-fed mice potently reduces glycaemia and improves glucose tolerance, with combined effect both to stimulate insulin secretion from islets and improve insulin sensitivity.


Subject(s)
Blood Glucose/metabolism , Dietary Fats/adverse effects , Glucokinase/metabolism , Sulfones/pharmacology , Thiadiazoles/pharmacology , Administration, Oral , Animals , Enzyme Activation/drug effects , Female , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/physiopathology , Liver/drug effects , Liver/metabolism , Mice , Rats , Sulfones/administration & dosage , Thiadiazoles/administration & dosage , Time Factors
10.
Eur J Pharmacol ; 626(2-3): 297-305, 2010 Jan 25.
Article in English | MEDLINE | ID: mdl-19818749

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) are transcription factors belonging to the nuclear receptor superfamily. Several reports have shown that PPARdelta is involved in lipid metabolism, increasing fat oxidation and depleting lipid accumulation. Whether PPARdelta is involved in the regulation of glucose metabolism is not completely understood. In this study, we examined effects of long-term PPARdelta activation on glycemic control, islet function and insulin sensitivity in diabetic db/db mice. Male db/db mice were administered orally once daily with a selective and partial PPARdelta agonist (NNC 61-5920, 30 mg/kg) for eight weeks; control mice received vehicle. Fasting and non-fasting plasma glucose were reduced, reflected in reduced hemoglobinA(1c) (3.6+/-1.6% vs. 5.4+/-1.8 in db/db controls, P<0.05) and furthermore, the AUC(glucose) after oral glucose (3g/kg) was reduced by 67% (P<0.05) after long-term PPARdelta activation. Following intravenous glucose (1g/kg), glucose tolerance was improved after PPARdelta activation (K(G) 1.3+/-0.6 vs. -0.05+/-0.7 %/min, P=0.048). Insulin sensitivity, measured as the glucose clearance after intravenous injection of glucose (1g/kg) and insulin (0.75 or 1.0 U/kg), during inhibition of endogenous insulin secretion by diazoxide (25mg/kg), was improved (K(G) 2.9+/-0.6 vs. 1.3+/-0.3 %/min in controls, P<0.05) despite lower insulin levels. Furthermore, islets isolated from PPARdelta agonist treated mice demonstrated improved glucose responsiveness as well as improved cellular topography. In conclusion, PPARdelta agonism alleviates insulin resistance and improves islet function and topography, resulting in improved glycemia in diabetic db/db mice. This suggests that activation of PPARdelta improves glucose metabolism and may therefore potentially be target for treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus/physiopathology , Insulin/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/physiopathology , PPAR delta/metabolism , Animals , Body Weight/drug effects , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Fatty Acids, Nonesterified/metabolism , Glucose/metabolism , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Oxidation-Reduction/drug effects , PPAR delta/agonists , Palmitates/metabolism , Time Factors
11.
J Clin Endocrinol Metab ; 94(8): 2887-92, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19435824

ABSTRACT

CONTEXT: The insulin response to meal ingestion is more rapid in the morning than in the afternoon. Whether this is explained by a corresponding variation in the incretin hormones is not known. OBJECTIVE: Our objective was to assess islet and incretin hormones after meal ingestion in the morning vs. afternoon. DESIGN, SETTINGS, AND PARTICIPANTS: Ingestion at 0800 and 1700 h of a standardized meal (524 kcal) in healthy lean males (n = 12) at a University Clinical Research Unit. MAIN OUTCOME MEASURES: We assessed early (30-min) area under the curve (AUC30) of plasma levels of insulin and intact (i) and total (t) glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) after meal ingestion and made an estimation of beta-cell function by model analysis of glucose and C-peptide. RESULTS: Peak glucose was lower in the morning than in the afternoon (6.1 +/- 0.2 vs. 7.4 +/- 0.3 mmol/liter, P = 0.001). AUC30(insulin) (4.9 +/- 0.6 vs. 2.8 +/- 0.4 nmol/liter . 30 min; P = 0.012), AUC30(tGLP-1) (300 +/- 40 vs. 160 +/- 30 pmol/liter . 30 min, P = 0.002), AUC30(iGIP) (0.7 +/- 0.1 vs. 0.3 +/- 0.1 nmol/liter . 30 min, P = 0.002), and AUC30(tGIP) (1.1 +/- 0.1 vs. 0.6 +/- 0.1 nmol/liter . min, P = 0.007) were all higher in the morning. AUC30(iGLP-1) (r = 0.68; P = 0.021) and AUC30(iGIP) (r = 0.78; P = 0.001) both correlated to AUC30(insulin). Model analysis of beta-cell function showed a higher first-hour potentiation factor in the morning (P = 0.009). This correlated negatively with the 60-min glucose level (r = -0.63; P < 0.001). CONCLUSIONS: The early release of GLP-1 and GIP are more pronounced in the morning than in the afternoon. This may contribute to the more rapid early insulin response, more pronounced potentiation of beta-cell function, and lower glucose after the morning meal.


Subject(s)
Incretins/blood , Islets of Langerhans/physiology , Adult , Blood Glucose/analysis , C-Peptide/analysis , Fatty Acids, Nonesterified/blood , Gastric Inhibitory Polypeptide/blood , Glucagon/blood , Humans , Insulin/blood , Male , Postprandial Period , Time Factors
12.
Am J Physiol Endocrinol Metab ; 295(4): E779-84, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18612044

ABSTRACT

Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) regulate islet function after carbohydrate ingestion. Whether incretin hormones are of importance for islet function after ingestion of noncarbohydrate macronutrients is not known. This study therefore examined integrated incretin and islet hormone responses to ingestion of pure fat (oleic acid; 0.88 g/kg) or protein (milk and egg protein; 2 g/kg) over 5 h in healthy men, aged 20-25 yr (n=12); plain water ingestion served as control. Both intact (active) and total GLP-1 and GIP levels were determined as was plasma activity of dipeptidyl peptidase-4 (DPP-4). Following water ingestion, glucose, insulin, glucagon, GLP-1, and GIP levels and DPP-4 activity were stable during the 5-h study period. Both fat and protein ingestion increased insulin, glucagon, GIP, and GLP-1 levels without affecting glucose levels or DPP-4 activity. The GLP-1 responses were similar after protein and fat, whereas the early (30 min) GIP response was higher after protein than after fat ingestion (P<0.001). This was associated with sevenfold higher insulin and glucagon responses compared with fat ingestion (both P<0.001). After protein, the early GIP, but not GLP-1, responses correlated to insulin (r(2)=0.86; P=0.0001) but not glucagon responses. In contrast, after fat ingestion, GLP-1 and GIP did not correlate to islet hormones. We conclude that, whereas protein and fat release both incretin and islet hormones, the early GIP secretion after protein ingestion may be of primary importance to islet hormone secretion.


Subject(s)
Dietary Fats/pharmacology , Dietary Proteins/pharmacology , Incretins/biosynthesis , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Pancreatic Hormones/metabolism , Adult , Blood Glucose/metabolism , Body Water/metabolism , Diet , Dipeptidyl Peptidase 4/blood , Fatty Acids, Nonesterified/blood , Gastric Inhibitory Polypeptide/biosynthesis , Glucagon/blood , Glucagon-Like Peptide 1/biosynthesis , Humans , Insulin/blood , Male , Triglycerides/blood
13.
J Endocrinol ; 197(1): 181-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18372244

ABSTRACT

To study whether the incretin effect is involved in adaptively increased insulin secretion in insulin resistance, glucose was infused at a variable rate to match glucose levels after oral glucose (25 mg) in normal anesthetized C57BL/6J female mice or in mice rendered insulin resistant by 8 weeks of high-fat feeding. Insulin response was markedly higher after oral than i.v. glucose in both groups, and this augmentation was even higher in high-fat fed than normal mice. In normal mice, the area under the curve (AUC(insulin)) was augmented from 4.0+/-0.8 to 8.0+/-1.8 nmol/lx60 min by the oral glucose, i.e. by a factor of 2 (P=0.023), whereas in the high-fat fed mice, AUC(insulin) was augmented from 0.70+/-0.4 to 12.4+/-2.5 nmol/lx60 min, i.e. by a factor of 17 (P<0.001). To examine whether the incretin hormone glucagon-like peptide-1 (GLP-1) is responsible for this difference, the effect of i.v. GLP-1 was compared in normal and high-fat fed mice. The sensitivity to i.v. GLP-1 in stimulating insulin secretion was increased in the high-fat diet fed mice: the lowest effective dose of GLP-1 was 650 pmol/kg in normal mice and 13 pmol/kg in the high-fat diet fed mice. We conclude that 1) the incretin effect contributes by approximately 50% to insulin secretion by the oral glucose in normal mice, 2) this effect is markedly exaggerated in insulin-resistant mice fed a high-fat diet, and 3) this augmented incretin contribution in the high-fat fed mice may partially be explained by GLP-1.


Subject(s)
Dietary Fats/administration & dosage , Glucose/administration & dosage , Insulin/metabolism , Administration, Oral , Animals , Area Under Curve , Female , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/blood , Injections, Intravenous , Insulin Secretion , Mice , Mice, Inbred C57BL
14.
J Mol Endocrinol ; 40(2): 93-100, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18234911

ABSTRACT

Glucagon-like peptide 1 (GLP-1) augments glucose-stimulated insulin secretion (GSIS) through cAMP-induced activation of protein kinase A (PKA), and stimulates beta-cell proliferation and reduces beta-cell apoptosis in rodent islets. This study explored islet GSIS, PKA expression, and markers of apoptosis (caspase 3/7 activity) and proliferation (PKBalpha and pancreatic and duodenal homeobox gene 1, Pdx-1) after 2 weeks of treatment with the GLP-1 receptor agonist exendin-4 (2 nmol/kg once daily) in female mice with high-fat diet-induced insulin resistance (HFD; 58% fat by energy). Islets were isolated 20 h after the last exendin-4 injection, when effects of circulating exendin-4 had vanished. The glucose responsiveness in islets from HFD-fed mice at 8.3 mM glucose was reduced compared with islets from control mice fed a normal diet due to increased basal insulin secretion. However, GSIS increased in islets from HFD-fed exendin-4-treated animals (0.124+/-0.012 ng/h per islet in HFD-Ex-4 versus 0.062+/-0.010 in HFD, P=0.006). Furthermore, the insulin response to forskolin was increased (2.7+/-0.3 in HFD-Ex-4 versus 2.0+/-0.2 ng/h per islet in HFD, P=0.011) and PKAcat expression was increased, while PKAreg was reduced in islets from exendin-4-treated mice. In contrast, protein expression of PKBalpha, Pdx-1, and caspase 3/7 activity was not affected by exendin-4 treatment. We conclude that GLP-1 receptor activation in HFD-fed mice has durable effects on GSIS, in association with augmented signaling through the PKA pathway. These effects are seen beyond those induced by circulating exendin-4 already after 2 weeks of once-daily treatment in mice, whereas markers for islet proliferation and apoptosis were unaffected by this treatment.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Diet , Fatty Acids/administration & dosage , Insulin/metabolism , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Peptides/pharmacology , Venoms/pharmacology , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Blood Glucose , Body Weight/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Proliferation/drug effects , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Exenatide , Fatty Acids/pharmacology , Female , In Vitro Techniques , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Mice , Mice, Inbred C57BL , Peptides/administration & dosage , Perfusion , Venoms/administration & dosage
15.
Pharmacol Ther ; 116(3): 437-48, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17900700

ABSTRACT

Islet function is regulated by a number of different signals. A main signal is generated by glucose, which stimulates insulin secretion and inhibits glucagon secretion. The glucose effects are modulated by many factors, including hormones, neurotransmitters and nutrients. Several of these factors signal through guanine nucleotide-binding protein (G protein)-coupled receptors (GPCR). Examples of islet GPCR are GPR40 and GPR119, which are GPCR with fatty acids as ligands, the receptors for the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), the receptors for the islet hormones glucagon and somatostatin, the receptors for the classical neurotransmittors acetylcholine (ACh; M(3) muscarinic receptors) and noradrenaline (beta(2)- and alpha(2)-adrenoceptors) and for the neuropeptides pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP; PAC(1) and VPAC(2) receptors), cholecystokinin (CCK(A) receptors) and neuropeptide Y (NPY Y1 receptors). Other islet GPCR are the cannabinoid receptor (CB(1) receptors), the vasopressin receptors (V1(B) receptors) and the purinergic receptors (P(2Y) receptors). The islet GPCR couple mainly to adenylate cyclase and to phospholipase C (PLC). Since important pharmacological strategies for treatment of type 2 diabetes are stimulation of insulin secretion and inhibition of glucagon secretion, islet GPCR are potential drug targets. This review summarizes knowledge on islet GPCR.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Islets of Langerhans/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Glucagon-Like Peptide-1 Receptor , Humans , Insulin/metabolism , Insulin Secretion , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, Gastrointestinal Hormone/physiology , Receptors, Glucagon/physiology , Receptors, Neurotransmitter/physiology , Signal Transduction
16.
Peptides ; 28(9): 1805-13, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17559974

ABSTRACT

Vasoactive intestinal polypeptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are two closely related neuropeptides that are expressed in islets and in islet parasympathetic nerves. Both peptides bind to their common G-protein-coupled receptors, VPAC1 and VPAC2, and PACAP, in addition to the specific receptor PAC1, all three of which are expressed in islets. VIP and PACAP stimulate insulin secretion in a glucose-dependent manner and they both also stimulate glucagon secretion. This action is achieved through increased formation of cAMP after activation of adenylate cyclase and stimulation of extracellular calcium uptake. Deletion of PAC1 receptors or VPAC2 receptors results in glucose intolerance. These peptides may be of importance in mediating prandial insulin secretion and the glucagon response to hypoglycemia. Animal studies have also suggested that activation of the receptors, in particular VPAC2 receptors, may be used as a therapeutic approach for the treatment of type 2 diabetes. This review summarizes the current knowledge of the potential role of VIP and PACAP in islet function.


Subject(s)
Islets of Langerhans/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Vasoactive Intestinal Peptide/pharmacology , Animals , Humans , Insulin/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Models, Biological , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/antagonists & inhibitors , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Signal Transduction/drug effects , Vasoactive Intestinal Peptide/physiology
17.
Regul Pept ; 143(1-3): 97-103, 2007 Oct 04.
Article in English | MEDLINE | ID: mdl-17482289

ABSTRACT

Inhibition of dipeptidyl peptidase-4 (DPP-4) is currently explored as a novel therapy of type 2 diabetes. The strategy has been shown to improve glycemia in most, but not all, rodent forms of glucose intolerance. In this study, we explored the effects of DPP-4 inhibition in mice with beta-cell overexpression of human islet amyloid polypeptide (IAPP). We therefore administered the orally active and highly selective DPP-4 inhibitor, vildagliptin (3 micromol/mouse daily) to female mice with beta-cell overexpression of human IAPP. Controls were given plain water, and a series of untreated wildtype mice was also included. After five weeks, an intravenous glucose tolerance test showed improved glucose disposal and a markedly enhanced insulin response in mice treated with vildagliptin. After eight weeks, a gastric tolerance test showed that vildagliptin improved glucose tolerance and markedly (approximately ten-fold) augmented the insulin response in association with augmented (approximately five-fold) levels of intact glucagon-like peptide-1 (GLP-1). Furthermore, after nine weeks, islets were isolated. Islets from vildagliptin-treated mice showed augmented glucose-stimulated insulin response and a normalization of the islet insulin content, which was reduced by approximately 50% in transgenic controls versus wildtype animals. Double immunostaining of pancreatic islets for insulin and glucagon revealed that transgenic islets displayed severely disturbed intra-islet topography with frequently observed centrally located alpha-cells. Treatment with vildagliptin restored the islet topography. We therefore conclude that DPP-4 inhibition improves islet function and islet topography in mice with beta-cell specific transgenic overexpression of human IAPP.


Subject(s)
Amyloid/metabolism , Dipeptidyl-Peptidase IV Inhibitors , Glucagon-Like Peptide 1/pharmacology , Insulin/pharmacology , Islets of Langerhans/drug effects , Adamantane/analogs & derivatives , Adamantane/pharmacology , Amyloid/genetics , Animals , Dipeptidyl Peptidase 4/metabolism , Female , Glucose/metabolism , Glucose Tolerance Test , Humans , Immunohistochemistry , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Mice, Transgenic , Nitriles/pharmacology , Pyrrolidines/pharmacology , Vildagliptin
18.
Biochem Biophys Res Commun ; 354(1): 240-5, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17214971

ABSTRACT

The free fatty acid receptor, GPR40, has been coupled with insulin secretion via its expression in pancreatic beta-cells. However, the role of GPR40 in the release of glucagon has not been studied and previous attempts to identify the receptor in alpha-cells have been unfruitful. Using double-staining for glucagon and GPR40 expression, we demonstrate that the two are expressed in the same cells in the periphery of mouse islets. In-R1-G9 hamster glucagonoma cells respond dose-dependently to linoleic acid stimulation by elevated phosphatidyl inositol hydrolysis and glucagon release and the cells become increasingly responsive to fatty acid stimulation when overexpressing GPR40. Isolated mouse islets also secrete glucagon in response to linoleic acid, a response that was abolished by antisense treatment against GPR40. This study demonstrates that GPR40 is present and active in pancreatic alpha-cells and puts further emphasis on the importance of this nutrient sensing receptor in islet function.


Subject(s)
Glucagon-Secreting Cells/physiology , Glucagon/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cells, Cultured , Cricetinae , Female , Mice
19.
Metabolism ; 56(1): 122-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17161234

ABSTRACT

The high fat-fed mouse is an experimental model for studies of islet dysfunction as a mechanism for glucose intolerance and for evaluation of therapeutic targets. This model is, however, dynamic with a temporal and dietary fat content-dependent impact on islet function and glucose tolerance, the details of which are unknown. This study therefore examined the time course of changes in the insulin response to intravenous glucose (1 g/kg) in relation to glucose tolerance in female mice after 1, 3, 8, or 16 weeks of feeding with diets containing 11% fat (normal diet [ND]), 30% fat (medium-fat diet [MFD]), or 58% fat (high-fat diet [HFD]; by energy). High-fat diet increased body weight and body fat content, whereas MFD did not. The insulin response (postglucose suprabasal mean 1- and 5-minute insulin) was impaired after 1 week on MFD (481+/- 33 pmol/L) or HFD (223 +/- 31 pmol/L) compared with ND (713 +/- 46 pmol/L, both P < .001). This was accompanied by impaired glucose elimination compared with ND (both P < .001). Over the 16-week study period, the insulin response adaptively increased in the groups fed with HFD and MFD, to be not significantly different from ND after 16 weeks. This compensation normalized glucose tolerance in MFD, whereas the glucose tolerance was still below normal in HFD. Insulin clearance, as judged by elimination of intravenous human insulin, was not altered in HFD, suggesting that the observed changes in insulin responses to glucose are due to changes in insulin secretion rather than to changes in insulin clearance. We conclude that time- and dietary fat-dependent dynamic adaptive islet compensation evolves after introducing HFD in mice and that MFD-fed mice is a novel nonobese model of glucose intolerance.


Subject(s)
Blood Glucose/metabolism , Dietary Fats/administration & dosage , Glucose Intolerance/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Adipose Tissue/metabolism , Animals , Biopsy , Body Composition/physiology , Body Weight/physiology , Dietary Fats/metabolism , Energy Metabolism , Female , Glucose/administration & dosage , Glucose Tolerance Test , Insulin/blood , Liver/metabolism , Mice , Mice, Inbred C57BL , Triglycerides/metabolism
20.
Diabetes ; 55(12): 3463-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17130493

ABSTRACT

In previous studies, glucagon receptor knockout mice (Gcgr(-/-)) display reduced blood glucose and increased glucose tolerance, with hyperglucagonemia and increased levels of glucagon-like peptide (GLP)-1. However, the role of glucagon receptor signaling for the regulation of islet function and insulin sensitivity is unknown. We therefore explored beta-cell function and insulin sensitivity in Gcgr(-/-) and wild-type mice. The steady-state glucose infusion rate during hyperinsulinemic-euglycemic clamp was elevated in Gcgr(-/-) mice, indicating enhanced insulin sensitivity. Furthermore, the acute insulin response (AIR) to intravenous glucose was higher in Gcgr(-/-) mice. The augmented AIR to glucose was blunted by the GLP-1 receptor antagonist, exendin-3. In contrast, AIR to intravenous administration of other secretagogues was either not affected (carbachol) or significantly reduced (arginine, cholecystokinin octapeptide) in Gcgr(-/-) mice. In islets isolated from Gcgr(-/-) mice, the insulin responses to glucose and several insulin secretagogues were all significantly blunted compared with wild-type mice. Furthermore, glucose oxidation was reduced in islets from Gcgr(-/-) mice. In conclusion, the present study shows that glucagon signaling is required for normal beta-cell function and that insulin action is improved when disrupting the signal. In vivo, augmented GLP-1 levels compensate for the impaired beta-cell function in Gcgr(-/-) mice.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/pharmacology , Receptors, Glucagon/deficiency , Receptors, Glucagon/genetics , Animals , Arginine/pharmacology , Blood Glucose/metabolism , Carbachol/pharmacology , Glucagon/physiology , Glucose/metabolism , Glucose Clamp Technique , Glucose Tolerance Test , Hyperinsulinism , Insulin/metabolism , Insulin Secretion , Kinetics , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...