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










Publication year range
1.
Cell Metab ; 21(3): 379-91, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25738454

ABSTRACT

Glucagon-like peptide-1 (GLP-1) secreted from enteroendocrine L cells promotes nutrient disposal via the incretin effect. However, the majority of L cells are localized to the distal gut, suggesting additional biological roles for GLP-1. Here, we demonstrate that GLP-1 receptor (GLP-1R) signaling controls mucosal expansion of the small bowel (SB) and colon. These actions did not require the epidermal growth factor (EGF) or intestinal epithelial insulin-like growth factor (IGF1) receptors but were absent in Glp1r(-/-) mice. Polyp number and size were increased in SB of exendin-4-treated Apc(Min/+) mice, whereas polyp number was reduced in SB and colon of Glp1r(-/-):Apc(Min/+) mice. Exendin-4 increased fibroblast growth factor 7 (Fgf7) expression in colonic polyps of Apc(Min/+) mice and failed to increase intestinal growth in mice lacking Fgf7. Exogenous exendin-4 and Fgf7 regulated an overlapping set of genes important for intestinal growth. Thus, gain and loss of GLP-1R signaling regulates gut growth and intestinal tumorigenesis.


Subject(s)
Colon/metabolism , Fibroblast Growth Factor 7/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Animals , Carcinogenesis/metabolism , Cell Proliferation/physiology , Colon/physiology , Colon/physiopathology , Epidermal Growth Factor/metabolism , Exenatide , Female , Incretins/metabolism , Intestinal Mucosa/physiology , Intestinal Mucosa/physiopathology , Intestine, Small/pathology , Intestine, Small/physiology , Male , Mice , Mice, Inbred C57BL , Peptides/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Venoms/metabolism
2.
J Biol Chem ; 289(23): 15927-41, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24742675

ABSTRACT

Glucose homeostasis in mammals is dependent on the opposing actions of insulin and glucagon. The Golgi N-acetylglucosaminyltransferases encoded by Mgat1, Mgat2, Mgat4a/b/c, and Mgat5 modify the N-glycans on receptors and solute transporter, possibly adapting activities in response to the metabolic environment. Herein we report that Mgat5(-/-) mice display diminished glycemic response to exogenous glucagon, together with increased insulin sensitivity. Glucagon receptor signaling and gluconeogenesis in Mgat5(-/-) cultured hepatocytes was impaired. In HEK293 cells, signaling by ectopically expressed glucagon receptor was increased by Mgat5 expression and GlcNAc supplementation to UDP-GlcNAc, the donor substrate shared by Mgat branching enzymes. The mobility of glucagon receptor in primary hepatocytes was reduced by galectin-9 binding, and the strength of the interaction was dependent on Mgat5 and UDP-GlcNAc levels. Finally, oral GlcNAc supplementation rescued the glucagon response in Mgat5(-/-) hepatocytes and mice, as well as glycolytic metabolites and UDP-GlcNAc levels in liver. Our results reveal that the hexosamine biosynthesis pathway and GlcNAc salvage contribute to glucose homeostasis through N-glycan branching on glucagon receptor.


Subject(s)
Hexosamines/biosynthesis , Polysaccharides/metabolism , Receptors, Glucagon/metabolism , Animals , Chromatography, Liquid , Glucagon/pharmacology , HEK293 Cells , Humans , Hypoglycemia/metabolism , Hypoglycemia/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Tandem Mass Spectrometry
3.
Diabetes ; 62(4): 1196-205, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23160527

ABSTRACT

Glucagon is a critical regulator of glucose homeostasis; however, mechanisms regulating glucagon action and α-cell function and number are incompletely understood. To elucidate the role of the hepatic glucagon receptor (Gcgr) in glucagon action, we generated mice with hepatocyte-specific deletion of the glucagon receptor. Gcgr(Hep)(-/-) mice exhibited reductions in fasting blood glucose and improvements in insulin sensitivity and glucose tolerance compared with wild-type controls, similar in magnitude to changes observed in Gcgr(-/-) mice. Despite preservation of islet Gcgr signaling, Gcgr(Hep)(-/-) mice developed hyperglucagonemia and α-cell hyperplasia. To investigate mechanisms by which signaling through the Gcgr regulates α-cell mass, wild-type islets were transplanted into Gcgr(-/-) or Gcgr(Hep)(-/-) mice. Wild-type islets beneath the renal capsule of Gcgr(-/-) or Gcgr(Hep)(-/-) mice exhibited an increased rate of α-cell proliferation and expansion of α-cell area, consistent with changes exhibited by endogenous α-cells in Gcgr(-/-) and Gcgr(Hep)(-/-) pancreata. These results suggest that a circulating factor generated after disruption of hepatic Gcgr signaling can increase α-cell proliferation independent of direct pancreatic input. Identification of novel factors regulating α-cell proliferation and mass may facilitate the generation and expansion of α-cells for transdifferentiation into ß-cells and the treatment of diabetes.


Subject(s)
Glucagon-Secreting Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Liver/metabolism , Receptors, Glucagon/metabolism , Animals , Blood Glucose , Female , Glucagon/administration & dosage , Glucagon/blood , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/pathology , Glucose/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hyperplasia , Insulin Resistance , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glucagon/genetics , Signal Transduction
4.
Endocrinology ; 154(1): 127-39, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23183176

ABSTRACT

Glucagon-like peptide-1 receptor (GLP-1R) agonists reduce lipid accumulation in peripheral tissues, attenuating atherosclerosis and hepatic steatosis in preclinical studies. We examined whether GLP-1R activation decreases atherosclerosis progression in high-fat diet-fed male ApoE(-/-) mice after administration of streptozotocin and treatment with the long-acting GLP-1R agonist taspoglutide administered once monthly vs. metformin in the drinking water for 12 wk. Taspoglutide did not reduce plaque area or lipid content in the aortic arch or abdominal aorta, and no significant change in aortic macrophage accumulation was detected after taspoglutide or metformin. In contrast, hepatic triglyceride levels were significantly reduced in livers from taspoglutide-treated mice. Both peripheral and intracerebroventricular administration of exendin-4 rapidly decreased plasma triglyceride levels in fasted mice, and taspoglutide therapy in ApoE(-/-) mice modulated the expression of hepatic genes controlling fatty acid uptake and oxidation. We were unable to detect expression of the entire Glp1r coding sequence in macrophages isolated from ApoE(-/-), C57BL/6, and IL10(-/-) mice. Similarly, Glp1r mRNA transcripts were not detected in RNA from isolated murine hepatocytes. Using Western blotting and tissue extracts from Glp1r(+/+) and Glp1r(-/-) mice, and cells transfected with a tagged murine GLP-1R cDNA, we could not validate the sensitivity and specificity of three different GLP-1R antisera commonly used for the detection of GLP-1R protein. Taken together, these findings illustrate divergent actions of GLP-1R agonists on atherosclerosis progression and accumulation of ectopic lipid in ApoE(-/-) mice and highlight the importance of indirect GLP-1R actions for the control of hepatic lipid accumulation.


Subject(s)
Atherosclerosis/metabolism , Liver/metabolism , Receptors, Glucagon/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Blotting, Western , Body Composition/drug effects , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Hepatocytes/drug effects , Hepatocytes/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Liver/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Knockout , Peptides/pharmacology , Receptors, Glucagon/agonists , Receptors, Glucagon/genetics
5.
Gastroenterology ; 139(3): 857-68, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20546737

ABSTRACT

BACKGROUND & AIMS: Glucagon-like peptide-2 (GLP-2) is a gut hormone that increases gut growth, reduces mucosal cell death, and augments mesenteric blood flow and nutrient absorption. Exogenous GLP-2(1-33) also stimulates glucagon secretion and enhances gut barrier function with implications for susceptibility to systemic inflammation and subsequent metabolic dysregulation. We examined the importance of GLP-2 receptor (GLP-2R) signaling for glucose homeostasis in multiple models of metabolic stress, diabetes, and obesity. METHODS: Body weight, islet function, glucose tolerance, and islet histology were studied in wild-type, high-fat fed, lean diabetic, Glp2r(-/-) and ob/ob:Glp2r(-/-) mice. RESULTS: GLP-2 did not stimulate glucagon secretion from isolated pancreatic islets in vitro, and exogenous GLP-2 had no effect on the glucagon response to insulin-induced hypoglycemia in vivo. Glp2r(-/-) mice exhibit no change in glycemia, and plasma glucagon levels were similar in Glp2r(-/-) and Glp2r(+/+) mice after hypoglycemia or after oral or intraperitoneal glucose challenge. Moreover, glucose homeostasis was comparable in Glp2r(-/-) and Glp2r(+/+) mice fed a high-fat diet for 5 months or after induction of streptozotocin-induced diabetes. In contrast, loss of the GLP-2R leads to increased glucagon secretion and alpha-cell mass, impaired intraperitoneal glucose tolerance and hyperglycemia, reduced beta-cell mass, and decreased islet proliferation in ob/ob:Glp2r(-/-) mice. CONCLUSIONS: Our results show that, although the GLP-2R is not critical for the stimulation or suppression of glucagon secretion or glucose homeostasis in normal or lean diabetic mice, elimination of GLP-2R signaling in obese mice impairs the normal islet adaptive response required to maintain glucose homeostasis.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Glucagon-Like Peptide 2/metabolism , Islets of Langerhans/metabolism , Obesity/metabolism , Receptors, Glucagon/metabolism , Stress, Physiological , Adaptation, Physiological , Animals , Blood Glucose/metabolism , Body Weight , Cell Proliferation , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Disease Models, Animal , Glucagon/blood , Glucagon-Like Peptide-2 Receptor , Glucose Intolerance/metabolism , Glucose Intolerance/physiopathology , Glucose Tolerance Test , Hypoglycemia/metabolism , Hypoglycemia/physiopathology , Insulin/blood , Islets of Langerhans/pathology , Islets of Langerhans/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/pathology , Obesity/physiopathology , Receptors, Glucagon/deficiency , Receptors, Glucagon/genetics , Signal Transduction , Time Factors
6.
J Biol Chem ; 285(3): 1989-2002, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19915011

ABSTRACT

Strategies based on activating GLP-1 receptor (GLP-1R) are intensively developed for the treatment of type 2 diabetes. The exhaustive knowledge of the signaling pathways linked to activated GLP-1R within the beta-cells is of major importance. In beta-cells, GLP-1 activates the ERK1/2 cascade by diverse pathways dependent on either Galpha(s)/cAMP/cAMP-dependent protein kinase (PKA) or beta-arrestin 1, a scaffold protein. Using pharmacological inhibitors, beta-arrestin 1 small interfering RNA, and islets isolated from beta-arrestin 1 knock-out mice, we demonstrate that GLP-1 stimulates ERK1/2 by two temporally distinct pathways. The PKA-dependent pathway mediates rapid and transient ERK1/2 phosphorylation that leads to nuclear translocation of the activated kinases. In contrast, the beta-arrestin 1-dependent pathway produces a late ERK1/2 activity that is restricted to the beta-cell cytoplasm. We further observe that GLP-1 phosphorylates the cytoplasmic proapoptotic protein Bad at Ser-112 but not at Ser-155. We find that the beta-arrestin 1-dependent ERK1/2 activation engaged by GLP-1 mediates the Ser-112 phosphorylation of Bad, through p90RSK activation, allowing the association of Bad with the scaffold protein 14-3-3, leading to its inactivation. beta-Arrestin 1 is further found to mediate the antiapoptotic effect of GLP-1 in beta-cells through the ERK1/2-p90RSK-phosphorylation of Bad. This new regulatory mechanism engaged by activated GLP-1R involving a beta-arrestin 1-dependent spatiotemporal regulation of the ERK1/2-p90RSK activity is now suspected to participate in the protection of beta-cells against apoptosis. Such signaling mechanism may serve as a prototype to generate new therapeutic GLP-1R ligands.


Subject(s)
Apoptosis/drug effects , Arrestins/metabolism , Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , bcl-Associated Death Protein/metabolism , 14-3-3 Proteins/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation/drug effects , Insulin-Secreting Cells/drug effects , Mice , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Serine , Signal Transduction/drug effects , Time Factors , bcl-Associated Death Protein/chemistry , beta-Arrestin 1 , beta-Arrestins
7.
Gastroenterology ; 137(6): 2146-57, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19766644

ABSTRACT

BACKGROUND & AIMS: Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) activate pathways involved in beta cell survival and proliferation in vitro; we compared the relative importance of exogenous and endogenous GIP receptor (GIPR) and GLP-1 receptor (GLP-1R) activation for beta cell cytoprotection in mice. METHODS: The effects of incretin hormone receptor signaling on beta cell regeneration and survival were assessed in mice following administration of streptozotocin in the absence or presence of the GIPR agonist [D-Ala(2)]-GIP (D-GIP), the GLP-1R agonist exendin-4, or the dipeptidyl peptidase-4 inhibitor sitagliptin. Beta cell survival was assessed in Gipr(-/-) mice given streptozotocin and by gene expression profiling of RNA from islets isolated from Glp1r(-/-) and Gipr(-/-) mice. The antiapoptotic actions of sitagliptin were assessed in wild-type and dual incretin receptor knockout (DIRKO) mice. RESULTS: Administration of exendin-4 for 7 or 60 days improved blood glucose and insulin levels, reduced islet cell apoptosis, and increased pancreatic insulin content and beta cell mass. In contrast, D-GIP was less effective at improving these parameters under identical experimental conditions. Furthermore, Gipr(-/-) mice did not exhibit increased sensitivity to streptozotocin-induced diabetes. Sitagliptin reduced hemoglobin A(1c) levels and increased plasma and pancreatic levels of insulin after streptozotocin administration to wild-type mice. Sitagliptin reduced the levels of activated caspase-3 in wild-type islets but not in beta cells from DIRKO mice. CONCLUSIONS: There are functionally important differences in the pharmacologic and physiologic roles of incretin receptors in beta cells. GLP-1R signaling exerts more robust control of beta cell survival, relative to GIPR activation or dipeptidylpeptidase-4 inhibition in mice in vivo.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Insulin-Secreting Cells/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Signal Transduction , Animals , Apoptosis , Blood Glucose/metabolism , Caspase 3/metabolism , Cell Line , Cell Proliferation , Cell Survival , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Exenatide , Gastric Inhibitory Polypeptide/analogs & derivatives , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide-1 Receptor , Glycated Hemoglobin/metabolism , Hypoglycemic Agents/pharmacology , Incretins/pharmacology , Insulin/blood , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptides/pharmacology , Pyrazines/pharmacology , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/deficiency , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/agonists , Receptors, Glucagon/deficiency , Receptors, Glucagon/genetics , Signal Transduction/drug effects , Sitagliptin Phosphate , Time Factors , Triazoles/pharmacology , Venoms/pharmacology
8.
Gastroenterology ; 137(3): 997-1005, 1005.e1-4, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19482026

ABSTRACT

BACKGROUND & AIMS: Excessive postprandial lipemia is a prevalent condition that results from intestinal oversecretion of apolipoprotein B48 (apoB48)-containing lipoproteins. Glucagon-like peptide-2 (GLP-2) is a gastrointestinal-derived intestinotropic hormone that links nutrient absorption to intestinal structure and function. We investigated the effects of GLP-2 on intestinal lipid absorption and lipoprotein production. METHODS: Intestinal lipid absorption and chylomicron production were quantified in hamsters, wild-type mice, and Cd36(-/-) mice infused with exogenous GLP-2. Newly synthesized apoB48 was metabolically labelled in primary hamster jejunal fragments. Fatty acid absorption was measured, and putative fatty acid transporters were assessed by immunoblotting. RESULTS: Human GLP-2 increased secretion of the triglyceride (TG)-rich lipoprotein (TRL)-apoB48 following oral administration of olive oil to hamsters; TRL and cholesterol mass each increased 3-fold. Fast protein liquid chromatography profiling indicated that GLP-2 stimulated secretion of chylomicron/very low-density lipoprotein-sized particles. Moreover, GLP-2 directly stimulated apoB48 secretion in jejunal fragments cultured ex vivo, increased expression of fully glycosylated cluster of differentiation 36/fatty acid translocase (CD36), and induced intestinal absorption of [(3)H]triolein. The ability of GLP-2 to increase intestinal lipoprotein production was lost in Cd36(-/-) mice. CONCLUSIONS: GLP-2 stimulates intestinal apoB48-containing lipoprotein secretion, possibly through increased lipid uptake, via a pathway that requires CD36. These findings suggest that GLP-2 represents a nutrient-dependent signal that regulates intestinal lipid absorption and the assembly and secretion of TRLs from intestinal enterocytes.


Subject(s)
Chylomicrons/metabolism , Glucagon-Like Peptide 2/physiology , Intestinal Absorption/physiology , Jejunum/metabolism , Lipid Metabolism , Lipoproteins/metabolism , Triglycerides/metabolism , Animals , Apolipoprotein B-48/blood , Apolipoprotein B-48/metabolism , CD36 Antigens/metabolism , Cricetinae , Dietary Fats, Unsaturated/administration & dosage , Fatty Acid Transport Proteins/metabolism , Fatty Acids/metabolism , Glucagon-Like Peptide 2/pharmacology , Intestinal Absorption/drug effects , Lipoproteins/chemistry , Male , Mesocricetus , Mice , Mice, Inbred C57BL , Mice, Knockout , Triglycerides/chemistry , Triolein/metabolism
9.
Cell Metab ; 8(5): 359-71, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19046568

ABSTRACT

Glucagon receptor (Gcgr) signaling maintains hepatic glucose production during the fasting state; however, the importance of the Gcgr for lipid metabolism is unclear. We show here that fasted Gcgr-/- mice exhibit a significant increase in hepatic triglyceride secretion and fasting increases fatty acid oxidation (FAO) in wild-type (WT) but not in Gcgr-/- mice. Moreover fasting upregulated the expression of FAO-related hepatic mRNA transcripts in Gcgr+/+ but not in Gcgr-/- mice. Exogenous glucagon administration reduced plasma triglycerides in WT mice, inhibited TG synthesis and secretion, and stimulated FA beta oxidation in Gcgr+/+ hepatocytes. The actions of glucagon on TG synthesis and FAO were abolished in PPARalpha-/- hepatocytes. These findings demonstrate that the Gcgr receptor is required for control of lipid metabolism during the adaptive metabolic response to fasting.


Subject(s)
Fasting/physiology , Glucagon/physiology , Hepatocytes/metabolism , Receptors, Glucagon/physiology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adaptation, Biological , Animals , Cells, Cultured , Fatty Acids/metabolism , Female , Glucagon/pharmacology , Hepatocytes/drug effects , Lipid Metabolism , Male , Mice , Mice, Knockout , Oxidation-Reduction , PPAR alpha/genetics , PPAR alpha/metabolism , RNA, Messenger/biosynthesis , Receptors, Glucagon/genetics , Triglycerides/blood , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Diabetes ; 56(12): 3006-13, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17717280

ABSTRACT

OBJECTIVE: Dipeptidyl peptidase-4 (DPP4) inhibitors lower blood glucose in diabetic subjects; however, the mechanism of action through which these agents improve glucose homeostasis remains incompletely understood. Although glucagon-like peptide (GLP)-1 and glucose-dependent insulinotropic polypeptide (GIP) represent important targets for DPP4 activity, whether additional substrates are important for the glucose-lowering actions of DPP4 inhibitors remains uncertain. RESEARCH DESIGN AND METHODS: We examined the efficacy of continuous vildagliptin administration in wild-type (WT) and dual incretin receptor knockout (DIRKO) mice after 8 weeks of a high-fat diet. RESULTS: Vildagliptin had no significant effect on food intake, energy expenditure, body composition, body weight gain, or insulin sensitivity in WT or DIRKO mice. However, glycemic excursion after oral glucose challenge was significantly reduced in WT but not in DIRKO mice after vildagliptin treatment. Moreover, vildagliptin increased levels of glucose-stimulated plasma insulin and reduced levels of cholesterol and triglycerides in WT but not in DIRKO mice. Vildagliptin treatment reduced the hepatic expression of genes important for cholesterol synthesis and fatty acid oxidation, including phospho-mevalonate kinase (Mvk), acyl-coenzyme dehydrogenase medium chain (Acadm), mevalonate (diphospho)decarboxylase (Mvd), and Acyl-CoA synthetase (Acsl1), in WT but not in DIRKO mice. However, vildagliptin also reduced levels of hepatic mRNA transcripts for farnesyl di-phosphate transferase (Fdft1), acetyl coenzyme A acyltransferase 1 (Acaa1), and carnitine palmitoyl transferase 1 (Cpt 1) in DIRKO mice. No direct effect of GLP-1 receptor agonists was detected on cholesterol or triglyceride synthesis and secretion in WT hepatocytes. CONCLUSIONS: These findings illustrate that although GLP-1 and GIP receptors represent the dominant molecular mechanisms for transducing the glucoregulatory actions of DPP4 inhibitors, prolonged DPP4 inhibition modulates the expression of genes important for lipid metabolism independent of incretin receptor action in vivo.


Subject(s)
Adamantane/analogs & derivatives , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Hypoglycemic Agents/therapeutic use , Incretins/physiology , Nitriles/therapeutic use , Pyrrolidines/therapeutic use , Receptors, Gastrointestinal Hormone/physiology , Receptors, Glucagon/physiology , Receptors, Peptide/physiology , Adamantane/therapeutic use , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cell Division/drug effects , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Mice , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Glucagon/drug effects , Receptors, Peptide/deficiency , Receptors, Peptide/genetics , Vildagliptin
11.
Ann N Y Acad Sci ; 1070: 161-6, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16888159

ABSTRACT

Miniglucagon (glucagon 19-29) is the ultimate processing product of proglucagon, present in the glucagon-secreting granules of the alpha cells, at a close vicinity of the insulin-secreting beta cells. Co-released with glucagon and thanks to its original mode of action and its huge potency, it suppresses, inside the islet of Langerhans, the detrimental effect of glucagon on insulin secretion, while it leaves untouched the beneficial effect of glucagon on glucose competence of the beta cell. At the periphery, miniglucagon is processed at the surface of glucagon- and insulin-sensitive cells from circulating glucagon. At that level, it acts via a cellular pathway which uses initial molecular steps distinct from that of insulin which, when impaired, are involved in insulin resistence. This bypass allows miniglucagon to act as an insulin-like component, a characteristic which makes this peptide of particular interest from a pathophysiological and pharmacological point of views in understanding and treating metabolic diseases, such as the type 2 diabetes.


Subject(s)
Glucagon/metabolism , Animals , Biological Transport , Glucagon/chemistry , Glucose/metabolism , Metalloendopeptidases/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism
12.
Endocrinology ; 146(2): 643-54, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15498890

ABSTRACT

The p44/p42 MAPKs (ERK1/2) cascade regulates beta-cell nuclear events, which modulates cell differentiation and gene transcription, whereas its implication in processes occurring in the cytoplasm, such as activation of the exocytotic machinery, is still unclear. Using the MIN6 beta-cell line and isolated rat islets of Langerhans, we investigated whether glucose, by activating the ERK1/2 cascade, induces phosphorylation of cytoplasmic proteins implicated in exocytosis of insulin granules such as synapsin I. We observed that the majority of ERK1/2 activity induced by glucose remains in the cytoplasm and physically interacts with synapsin I, allowing phosphorylation of the substrate. Therefore, we reexamined the potential requirement of ERK1/2 for insulin secretion. Blocking activation of ERK1/2 using MEK1/2, the MAPK kinase inhibitor PD98059 or using small interfering RNA-mediated silencing of ERK1 and ERK2 expressions resulted in partial inhibition of glucose-induced insulin release, indicating that ERK1/2 pathway participates also in the regulation of insulin secretion. Moreover, using the pancreatic islet perifusion model, we found that the ERK1/2 activity participates in the first and second phases of insulin release induced by glucose. Taken together, our results demonstrate new aspects of the glucose-dependent actions of ERK1/2 in beta-cells exerted on cytoplasmic proteins, including synapsin I, and participating in the overall glucose-induced insulin secretion.


Subject(s)
Insulin/metabolism , Islets of Langerhans/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Synapsins/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/enzymology , Cytoplasm/enzymology , Glucose/pharmacology , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Pancreatic Neoplasms , Phosphorylation/drug effects , RNA, Small Interfering , Rats , Rats, Wistar
13.
J Biol Chem ; 279(19): 20345-55, 2004 May 07.
Article in English | MEDLINE | ID: mdl-14988413

ABSTRACT

By using the MIN6 cell line and pancreatic islets, we show that in the presence of a low glucose concentration, corresponding to physiological glucagon release from alpha cells, glucagon treatment of the beta cell caused a rapid, time-dependent phosphorylation and activation of p44/p42 mitogen-activated protein kinase (ERK1/2) independently from extracellular calcium influx. Inhibition of either cAMP-dependent protein kinase (PKA) or MEK completely blocked ERK1/2 activation by glucagon. However, no significant activation of several upstream activators of MEK, including Shc-p21(Ras) and phosphatidylinositol 3-kinase, was observed in response to glucagon treatment. Chelation of intracellular calcium (intracellular [Ca(2+)]) reduced glucagon-mediated ERK1/2 activation. In addition, internalization of glucagon receptors through clathrin-coated pits formation is required for ERK1/2 activation. Remarkably, glucagon promotes the nuclear translocation of ERK1/2 and induces the phosphorylation of cAMP-response element-binding protein (CREB). Miniglucagon, produced from glucagon and released together with the mother hormone from the alpha cells in low glucose situations, blocks the insulinotropic effect of glucagon, whereas it does not inhibit the glucagon-induced PKA/ERK1/2/CREB pathway. We conclude that glucagon-induced ERK1/2 activation is mediated by PKA and that an increase in [Ca(2+)](i) is required for maximal ERK activation. Our results uncover a novel mechanism by which the PKA/ERK1/2 signaling network engaged by glucagon, in situation of low glucose concentration, regulates phosphorylation of CREB, a transcription factor crucial for normal beta cell function and survival.


Subject(s)
Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Glucagon/metabolism , Islets of Langerhans/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cell Line , Cell Survival , Cells, Cultured , Clathrin/chemistry , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Glucagon/chemistry , Islets of Langerhans/cytology , Male , Mice , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Precipitin Tests , Protein Structure, Tertiary , Rats , Rats, Wistar , Signal Transduction , Time Factors , Transcription, Genetic
14.
Ann N Y Acad Sci ; 1030: 230-42, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15659802

ABSTRACT

Long-term hyperglycemia, a major characteristic of the diabetic state, contributes to the deterioration of the beta cell function, a concept known as beta cell glucotoxicity. We used the MIN6 beta cell line and isolated rat islets to clarify the signaling mechanism(s) used by glucose to activate cAMP-responsive element binding protein (CREB), a transcription factor crucial for beta cell biology, and to evaluate the possible downregulation of this mechanism mediated by long-term hyperglycemia. We report that glucose (10 mM) induces an increase in cytosolic calcium concentration that leads to cAMP-induced protein kinase A (PKA) activation, promoting nuclear translocation of activated ERK1/2. The observation that glucose-induced CREB phosphorylation was totally inhibited by the PKA inhibitor H89 (2 microM) and reduced by 50% with the ERK1/2 inhibitor PD98059 (20 microM) indicates that ERK1/2, located downstream of PKA, cooperates with PKA and is responsible for half of the PKA-mediated CREB phosphorylation elicited by glucose in MIN6 beta cells. We also found that exposure of mu cells for 24 h to high glucose (25 mM) induced a 70% decrease in cellular ERK1/2 and a 50% decrease in CREB content. In high-glucose-treated, ERK1/2- and CREB-downregulated beta cells, there was a loss of glucose (10 mM, 5 min)-stimulated ERK1/2 and CREB phosphorylation that was associated with nuclear apoptotic characteristics. Since we have shown that activation of ERK1/2 is crucial for CREB phosphorylation, loss of the ERK1/2-CREB signaling pathway in beta cells due to long-term hyperglycemia is likely to exacerbate beta cell failure in diabetic states by affecting physiologically relevant gene expression and by inducing apoptosis.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Glucose/toxicity , Islets of Langerhans/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , CREB-Binding Protein , Calcium/metabolism , Cell Line , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Ion Transport , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Phosphorylation , Protein Binding , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...