Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
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
2.
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
3.
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
4.
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.

5.
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
6.
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.

7.
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
8.
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
9.
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
10.
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
11.
Circ Res ; 96(2): 189-96, 2005 Feb 04.
Article in English | MEDLINE | ID: mdl-15604418

ABSTRACT

Red blood cells may regulate tissue circulation and O2 delivery by releasing the vasodilator ATP in response to hypoxia. When released extracellularly, ATP is rapidly degraded to ADP in the circulation by ectonucleotidases. In this study, we show that ADP acting on P2Y13 receptors on red blood cells serves as a negative feedback pathway for the inhibition of ATP release. mRNA of the ADP receptor P2Y13 was highly expressed in human red blood cells and reticulocytes. The stable ADP analogue 2-MeSADP decreased ATP release from red blood cells by inhibition of cAMP. The P2Y12 and P2Y13 receptor antagonist AR-C67085 (30 micromol/L), but not the P2Y1 blocker MRS2179, inhibited the effects of 2-MeSADP. At doses where AR-C67085 only blocks P2Y12 (100 nmol/L), it had no effect. AR-C67085 and the nucleotidase apyrase increased cAMP per se, indicating a constant cAMP inhibitory effect of endogenous extracellular ADP. 2-MeSADP reduced plasma ATP concentrations in an in vivo pig model. Our results indicate that the ATP degradation product ADP inhibits ATP release by acting on the red blood cell P2Y13 receptor. This negative feedback system could be important in the control of plasma ATP levels and tissue circulation.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/physiology , Adenosine Monophosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Erythrocytes/metabolism , Feedback, Physiological/physiology , Receptors, Purinergic P2/physiology , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Apyrase/biosynthesis , Cyclic AMP/biosynthesis , Epinephrine/pharmacology , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Female , Humans , Male , Microdialysis , Phentolamine/pharmacology , Propranolol/pharmacology , RNA, Messenger/biosynthesis , Receptors, Purinergic P2/biosynthesis , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/genetics , Reticulocytes/metabolism , Sus scrofa , Thionucleotides/pharmacology
12.
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
13.
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
14.
Endocrinology ; 147(7): 3173-80, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16627575

ABSTRACT

Monounsaturated fatty acids, such as oleic acid (OA), and certain milk proteins, especially whey protein (WP), have insulinotropic effects and can reduce postprandial glycemia. This effect may involve the incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1). To explore this, we examined the release and inactivation of GIP and GLP-1 after administration of glucose with or without OA or WP through gastric gavage in anesthetized C57BL/6J mice. Insulin responses to glucose (75 mg) were 3-fold augmented by addition of WP (75 mg; P < 0.01), which was associated with enhanced oral glucose tolerance (P < 0.01). The insulin response to glucose was also augmented by addition of OA (34 mg; P < 0.05) although only 1.5-fold and with no associated increase in glucose elimination. The slope of the glucose-insulin curve was increased by OA (1.7-fold; P < 0.05) and by WP (4-fold; P < 0.01) compared with glucose alone, suggesting potentiation of glucose-stimulated insulin release. WP increased GLP-1 secretion (P < 0.01), whereas GIP secretion was unaffected. OA did not affect GIP or GLP-1 secretion. Nevertheless, WP increased the levels of both intact GIP and intact GLP-1 (both P < 0.01), and OA increased the levels of intact GLP-1 (P < 0.05). WP inhibited dipeptidyl peptidase IV activity in the proximal small intestine by 50% (P < 0.05), suggesting that luminal degradation of WP generates small fragments, which are substrates for dipeptidyl peptidase IV and act as competitive inhibitors. We therefore conclude that fat and protein may serve as exogenous regulators of secretion and inactivation of the incretin hormones with beneficial influences on glucose metabolism.


Subject(s)
Gastrointestinal Hormones/metabolism , Glucose/metabolism , Animals , Area Under Curve , Dietary Fats/metabolism , Dipeptidyl Peptidase 4/metabolism , Female , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , Insulin/metabolism , Kinetics , Mice , Mice, Inbred C57BL
15.
Diabetes ; 53 Suppl 3: S215-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15561913

ABSTRACT

This study characterizes the high-fat diet-fed mouse as a model for impaired glucose tolerance (IGT) and type 2 diabetes. Female C57BL/6J mice were fed a high-fat diet (58% energy by fat) or a normal diet (11% fat). Body weight was higher in mice fed the high-fat diet already after the first week, due to higher dietary intake in combination with lower metabolic efficiency. Circulating glucose increased after 1 week on high-fat diet and remained elevated at a level of approximately 1 mmol/l throughout the 12-month study period. In contrast, circulating insulin increased progressively by time. Intravenous glucose challenge revealed a severely compromised insulin response in association with marked glucose intolerance already after 1 week. To illustrate the usefulness of this model for the development of new treatment, mice were fed an orally active inhibitor of dipeptidyl peptidase-IV (LAF237) in the drinking water (0.3 mg/ml) for 4 weeks. This normalized glucose tolerance, as judged by an oral glucose tolerance test, in association with augmented insulin secretion. We conclude that the high-fat diet-fed C57BL/6J mouse model is a robust model for IGT and early type 2 diabetes, which may be used for studies on pathophysiology and development of new treatment.


Subject(s)
Diabetes Mellitus, Type 2/therapy , Dietary Fats , Glucose Intolerance/therapy , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Dipeptidyl Peptidase 4/metabolism , Disease Models, Animal , Female , Glucose Intolerance/blood , Glucose Tolerance Test , Mice , Mice, Inbred C57BL , Protease Inhibitors/pharmacology
16.
Diabetes ; 53 Suppl 3: S92-6, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15561929

ABSTRACT

We studied islet function in mice with beta-cell-targeted expression of a dominant-negative mutant of hepatocyte nuclear factor (HNF)-1alpha. At age 2-3 months, anesthetized transgenic and wild-type male mice underwent an intravenous glucose (1 g/kg) tolerance test (IVGTT). It was found that transgenic mice had an abolished insulin response in association with severe glucose intolerance. In other tests, the 5-min insulin response to intravenous arginine was impaired by 79% (P=0.032) and the 15-min insulin response to gastric glucose was suppressed by 97% (P=0.006). In islets incubated for 60 min, the insulin response to glucose (3.3-22.2 mmol/l) was impaired by >80% in transgenic mice. In contrast, insulin responses to nonglucose secretagogues were only partially suppressed (to GLP-1 [100 nmol/l] by 40%, to carbachol [1 micromol/l] by 20%, and to palmitate [0.5 mmol/l] by 15%), whereas the response to depolarization by KCl (50 mmol/l) was not reduced. Finally, the IVGTT data insulin sensitivity in transgenic mice was not significantly different from that of wild-type mice. Thus, mice with targeted suppression of beta-cell HNF-1alpha represent a good diabetes model exhibiting severely impaired insulin secretion after glucose with marked glucose intolerance. In contrast, the insulin responses to nonglucose stimuli are not suppressed when the islet insulin content is taken into account.


Subject(s)
DNA-Binding Proteins/genetics , Islets of Langerhans/physiology , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Arginine/pharmacology , Blood Glucose/metabolism , Carbachol/pharmacology , Genes, Dominant , Glucose/pharmacology , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Insulin/analysis , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Transgenic , Mutation , Palmitic Acid/pharmacology , Potassium Chloride/pharmacology , Rats
17.
Diabetes ; 53(1): 122-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14693706

ABSTRACT

Lipids may serve as coupling factors in K(ATP)-independent glucose sensing in beta-cells. We have previously demonstrated that beta-cells harbor lipase activities, one of which is the hormone-sensitive lipase. Whether beta-cell lipases are critical for glucose-stimulated insulin secretion (GSIS) by providing lipid-derived signals from endogenous lipids is unknown. Therefore, using a lipase inhibitor (orlistat), we examined whether lipase inhibition reduces insulin secretion. Islet lipolysis stimulated by glucose and diglyceride lipase activity was abolished by orlistat. Incubation of rat islets with orlistat dose dependently inhibited GSIS; this inhibition was reversed by 1 mmol/l palmitate, suggesting that orlistat acts via impaired formation of an acylglyceride-derived coupling signal. Orlistat inhibited the potentiating effect of forskolin on GSIS, an effect proposed to be due to activation of a lipase. In perifused islets, orlistat attenuated mainly the second phase of insulin secretion. Because the rise in islet ATP/ADP levels in response to glucose and oxidation of the sugar were unaffected by orlistat whereas the second phase of insulin secretion was reduced, it seems likely that a lipid coupling factor involved in K(ATP)-independent glucose sensing has been perturbed. Thus, beta-cell lipase activity is involved in GSIS, emphasizing the important role of beta-cell lipid metabolism for insulin secretion.


Subject(s)
Insulin/metabolism , Islets of Langerhans/physiology , Lipase/metabolism , Lipolysis/physiology , Adipocytes/drug effects , Adipocytes/enzymology , Adipocytes/physiology , Animals , Cells, Cultured , Colforsin/pharmacology , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Kinetics , Lactones/pharmacology , Lipase/antagonists & inhibitors , Lipoprotein Lipase/metabolism , Orlistat , Perfusion , Rats
18.
Diabetes ; 52(8): 2057-65, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12882923

ABSTRACT

Lipid perturbations associated with triglyceride overstorage in beta-cells impair insulin secretion, a process termed lipotoxicity. To assess the role of hormone-sensitive lipase, which is expressed and enzymatically active in beta-cells, in the development of lipotoxicity, we generated transgenic mice overexpressing hormone-sensitive lipase specifically in beta-cells. Transgenic mice developed glucose intolerance and severely blunted glucose-stimulated insulin secretion when challenged with a high-fat diet. As expected, both lipase activity and forskolin-stimulated lipolysis was increased in transgenic compared with wild-type islets. This was reflected in significantly lower triglycerides levels in transgenic compared with wild-type islets in mice receiving the high-fat diet, whereas no difference in islet triglycerides was found between the two genotypes under low-fat diet conditions. Our results highlight the importance of mobilization of the islet triglyceride pool in the development of beta-cell lipotoxicity. We propose that hormone-sensitive lipase is involved in mediating beta-cell lipotoxicity by providing ligands for peroxisome proliferator-activated receptors and other lipid-activated transcription factors, which in turn alter the expression of critical genes. One such gene might be uncoupling protein-2, which was found to be upregulated in transgenic islets, a change that was accompanied by decreased ATP levels.


Subject(s)
Islets of Langerhans/enzymology , Membrane Transport Proteins , Mitochondrial Proteins , Sterol Esterase/genetics , Sterol Esterase/metabolism , Triglycerides/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Fatty Acids/metabolism , Female , Gene Expression Regulation, Enzymologic , Glucose Transporter Type 2 , Immunohistochemistry , Ion Channels , Islets of Langerhans/chemistry , Islets of Langerhans/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Monosaccharide Transport Proteins/analysis , Oxidation-Reduction , Phenotype , Proteins/metabolism , Rats , Uncoupling Protein 2 , Up-Regulation
19.
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
20.
Physiol Behav ; 83(4): 623-30, 2004 Dec 30.
Article in English | MEDLINE | ID: mdl-15621068

ABSTRACT

A high-fat diet easily promotes hyperphagia giving an impression of an uncontrolled process. Fat digestion itself however provides control of fat intake through the digestion itself, carried out by pancreatic lipase and its protein cofactor colipase, and through enterostatin, a peptide released from procolipase during fat digestion. Procolipase (-/-) knockout mice have a severely reduced fat digestion and fat uptake, pointing to a major role of the digestive process itself. With a normal fat digestion, enterostatin basically restricts fat intake by preventing the overconsumption of fat. The mechanism for enterostatin might be an inhibition of a mu-opioid-mediated pathway, demonstrated through binding studies on SK-N-MC-cells and crude brain membranes. Another target protein of enterostatin is the beta-subunit of F1F0-ATPase, displaying a distinct binding of enterostatin, established through an aqueous two-phase partition system. The binding of enterostatin to F1-ATPase was partially displaced by beta-casomorphin, a peptide stimulating fat intake and acting competitively to enterostatin. We frame a hypothesis that regulation of fat intake through enterostatin contains a reward component, which is an F1-ATPase-mediated pathway, possibly complemented with an opioidergic pathway.


Subject(s)
Colipases/physiology , Eating/physiology , Fats , Protein Precursors/physiology , Analgesics, Opioid/pharmacology , Animals , Binding Sites , Carrier Proteins/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Eating/drug effects , Endorphins/pharmacology , Enzyme Precursors , Fats/administration & dosage , Humans , Ion Channels , Membrane Proteins/metabolism , Mitochondrial Proteins , Mitochondrial Proton-Translocating ATPases/metabolism , Models, Biological , Neural Pathways/physiology , Uncoupling Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL