Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 238
1.
Cells ; 10(9)2021 09 21.
Article En | MEDLINE | ID: mdl-34572144

Incretin-potentiated glucose-stimulated insulin secretion (GSIS) is critical to maintaining euglycemia, of which GLP-1 receptor (GLP-1R) on ß-cells plays an indispensable role. Recently, α-cell-derived glucagon but not intestine-derived GLP-1 has been proposed as the critical hormone that potentiates GSIS via GLP-1R. However, the function of glucagon receptors (GCGR) on ß-cells remains elusive. Here, using GCGR or GLP-1R antagonists, in combination with glucagon, to treat single ß-cells, α-ß cell clusters and isolated islets, we found that glucagon potentiates insulin secretion via ß-cell GCGR at physiological but not high concentrations of glucose. Furthermore, we transfected primary mouse ß-cells with RAB-ICUE (a genetically encoded cAMP fluorescence indicator) to monitor cAMP level after glucose stimulation and GCGR activation. Using specific inhibitors of different adenylyl cyclase (AC) family members, we revealed that high glucose concentration or GCGR activation independently evoked cAMP elevation via AC5 in ß-cells, thus high glucose stimulation bypassed GCGR in promoting insulin secretion. Additionally, we generated ß-cell-specific GCGR knockout mice which glucose intolerance was more severe when fed a high-fat diet (HFD). We further found that ß-cell GCGR activation promoted GSIS more than GLP-1R in HFD, indicating the critical role of GCGR in maintaining glucose homeostasis during nutrient overload.


Glucagon/pharmacology , Insulin Secretion/physiology , Receptors, Glucagon/physiology , Animals , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/physiology , Glucose/metabolism , Glucose Intolerance/metabolism , Insulin/metabolism , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glucagon/antagonists & inhibitors , Signal Transduction
2.
Front Endocrinol (Lausanne) ; 12: 792912, 2021.
Article En | MEDLINE | ID: mdl-35095763

The first intracellular loop (ICL1) of G protein-coupled receptors (GPCRs) has received little attention, although there is evidence that, with the 8th helix (H8), it is involved in early conformational changes following receptor activation as well as contacting the G protein ß subunit. In class B1 GPCRs, the distal part of ICL1 contains a conserved R12.48KLRCxR2.46b motif that extends into the base of the second transmembrane helix; this is weakly conserved as a [R/H]12.48KL[R/H] motif in class A GPCRs. In the current study, the role of ICL1 and H8 in signaling through cAMP, iCa2+ and ERK1/2 has been examined in two class B1 GPCRs, using mutagenesis and molecular dynamics. Mutations throughout ICL1 can either enhance or disrupt cAMP production by CGRP at the CGRP receptor. Alanine mutagenesis identified subtle differences with regard elevation of iCa2+, with the distal end of the loop being particularly sensitive. ERK1/2 activation displayed little sensitivity to ICL1 mutation. A broadly similar pattern was observed with the glucagon receptor, although there were differences in significance of individual residues. Extending the study revealed that at the CRF1 receptor, an insertion in ICL1 switched signaling bias between iCa2+ and cAMP. Molecular dynamics suggested that changes in ICL1 altered the conformation of ICL2 and the H8/TM7 junction (ICL4). For H8, alanine mutagenesis showed the importance of E3908.49b for all three signal transduction pathways, for the CGRP receptor, but mutations of other residues largely just altered ERK1/2 activation. Thus, ICL1 may modulate GPCR bias via interactions with ICL2, ICL4 and the Gß subunit.


Amino Acid Motifs/physiology , Receptors, Calcitonin Gene-Related Peptide/ultrastructure , Receptors, Corticotropin-Releasing Hormone/ultrastructure , Receptors, Glucagon/ultrastructure , Calcitonin Receptor-Like Protein/metabolism , Calcitonin Receptor-Like Protein/physiology , Calcitonin Receptor-Like Protein/ultrastructure , Calcium Signaling , Cyclic AMP/metabolism , HEK293 Cells , Humans , MAP Kinase Signaling System , Molecular Dynamics Simulation , Protein Domains , Protein Structure, Tertiary , Receptor Activity-Modifying Protein 1/metabolism , Receptor Activity-Modifying Protein 1/physiology , Receptor Activity-Modifying Protein 1/ultrastructure , Receptors, Calcitonin Gene-Related Peptide/metabolism , Receptors, Calcitonin Gene-Related Peptide/physiology , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/physiology , Receptors, G-Protein-Coupled , Receptors, Glucagon/metabolism , Receptors, Glucagon/physiology
3.
Diabetologia ; 64(1): 142-151, 2021 01.
Article En | MEDLINE | ID: mdl-33043402

AIMS/HYPOTHESIS: The endocrine pancreas comprises the islets of Langerhans, primarily consisting of beta cells, alpha cells and delta cells responsible for secretion of insulin, glucagon and somatostatin, respectively. A certain level of intra-islet communication is thought to exist, where the individual hormones may reach the other islet cells and regulate their secretion. Glucagon has been demonstrated to importantly regulate insulin secretion, while somatostatin powerfully inhibits both insulin and glucagon secretion. In this study we investigated how secretion of somatostatin is regulated by paracrine signalling from glucagon and insulin. METHODS: Somatostatin secretion was measured from perfused mouse pancreases isolated from wild-type as well as diphtheria toxin-induced alpha cell knockdown, and global glucagon receptor knockout (Gcgr-/-) mice. We studied the effects of varying glucose concentrations together with infusions of arginine, glucagon, insulin and somatostatin, as well as infusions of antagonists of insulin, somatostatin and glucagon-like peptide 1 (GLP-1) receptors. RESULTS: A tonic inhibitory role of somatostatin was demonstrated with infusion of somatostatin receptor antagonists, which significantly increased glucagon secretion at low and high glucose, whereas insulin secretion was only increased at high glucose levels. Infusion of glucagon dose-dependently increased somatostatin secretion approximately twofold in control mice. Exogenous glucagon had no effect on somatostatin secretion in Gcgr-/- mice, and a reduced effect when combined with the GLP-1 receptor antagonist exendin 9-39. Diphtheria toxin-induced knockdown of glucagon producing cells led to reduced somatostatin secretion in response to 12 mmol/l glucose and arginine infusions. In Gcgr-/- mice (where glucagon levels are dramatically increased) overall somatostatin secretion was increased. However, infusion of exendin 9-39 in Gcgr-/- mice completely abolished somatostatin secretion in response to glucose and arginine. Neither insulin nor an insulin receptor antagonist (S961) had any effect on somatostatin secretion. CONCLUSIONS/INTERPRETATION: Our findings demonstrate that somatostatin and glucagon secretion are linked in a reciprocal feedback cycle with somatostatin inhibiting glucagon secretion at low and high glucose levels, and glucagon stimulating somatostatin secretion via the glucagon and GLP-1 receptors. Graphical abstract.


Glucagon/physiology , Insulin/physiology , Somatostatin/metabolism , Animals , Arginine/administration & dosage , Cell Communication , Diphtheria Toxin/pharmacology , Gene Knockdown Techniques , Glucagon/administration & dosage , Glucagon-Like Peptide-1 Receptor/drug effects , Glucagon-Like Peptide-1 Receptor/physiology , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/physiology , Glucose/administration & dosage , Insulin/administration & dosage , Insulin Secretion/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glucagon/deficiency , Receptors, Glucagon/genetics , Receptors, Glucagon/physiology , Receptors, Somatostatin/antagonists & inhibitors , Signal Transduction/physiology , Somatostatin/administration & dosage
4.
Neuropharmacology ; 170: 108042, 2020 06 15.
Article En | MEDLINE | ID: mdl-32147454

Alzheimer's disease (AD) is a progressively neurodegenerative disorder, which seriously affects human health and cannot be stopped by current treatments. Type 2 diabetes mellitus (T2DM) is a risk factor for AD. Our recent studies reported the neuroprotective effects of a GLP-1/GIP/Glucagon receptor triagonist (Triagonist), a novel unimolecular anti-diabetic drug, in cognitive and pathological improvements of 3xTg-AD mice. However, the detailed electrophysiological and molecular mechanisms underlying neuroprotection remain unexplored. The present study investigated the underlying electrophysiological and molecular mechanisms further by using whole-cell patch clamp techniques. Our results revealed that chronic Triagonist treatment effectively reduced working memory and reference memory errors of 3xTg-AD mice in a radial maze test. In addition, the Triagonist increased spontaneous excitatory synaptic activities, differentially modulated voltage- and chemically-gated Ca2+ flux, and reduced the over-excitation of pyramidal neurons in hippocampal slices of 3xTg-AD mice. In addition, chronic Triagonist treatment also up-regulated the expression levels of synaptophysin and PSD-95 in the hippocampus of 3xTg-AD mice. These results indicate that the Triagonist could improve memory formation, as well as synaptic transmission, Ca2+ balance, and neuronal excitability in 3xTg-AD mice. These neuroprotective effects of Triagonist may be involved in the up-regulation of synaptophysin and PSD-95. Therefore, the study suggests that multi-receptor agonists might be a novel therapeutic strategy for the treatment of AD.


Calcium Signaling/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Memory, Short-Term/drug effects , Receptors, Gastrointestinal Hormone/agonists , Receptors, Glucagon/agonists , Synaptic Transmission/drug effects , Amyloid beta-Protein Precursor/genetics , Animals , Calcium Signaling/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Glucagon-Like Peptide-1 Receptor/physiology , Hippocampus/drug effects , Hippocampus/physiology , Homeostasis/drug effects , Homeostasis/physiology , Humans , Hypoglycemic Agents/administration & dosage , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory, Short-Term/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/physiology , Organ Culture Techniques , Presenilin-1/genetics , Receptors, Gastrointestinal Hormone/physiology , Receptors, Glucagon/physiology , Synaptic Transmission/physiology , tau Proteins/genetics
5.
Gen Comp Endocrinol ; 261: 148-165, 2018 05 15.
Article En | MEDLINE | ID: mdl-29510149

The teleost fish-specific genome duplication gave rise to a great number of species inhabiting diverse environments with different access to nutrients and life histories. This event produced duplicated gcg genes, gcga and gcgb, for proglucagon-derived peptides, glucagon and GLP-1 and duplicated gcgr receptor genes, gcgra and gcgrb, which play key roles connecting the consumption of nutrients with glucose metabolism. We conducted a systematic survey of the genomes from 28 species of fish (24 bony (Superclass Osteichthyes), 1 lobe-finned (Class Sarcoperygii), 1 cartilaginous (Superclass Chondrichthyes), and 2 jawless (Superclass Agnatha)) and find that almost all surveyed ray-finned fish contain gcga and gcgb genes with different coding potential and duplicated gcgr genes, gcgra and gcgrb that form two separate clades in the phylogenetic tree consistent with the accepted species phylogeny. All gcgb genes encoded only glucagon and GLP-1 and gcga genes encoded glucagon, GLP-1, and GLP-2, indicating that gcga was subfunctionalized to produce GLP-2. We find a single glp2r, but no glp1r suggesting that duplicated gcgrb was neofunctionalized to bind GLP-1, as demonstrated for the zebrafish gcgrb (Oren et al., 2016). In functional experiments with zebrafish gcgrb and GLP-1 from diverse fish we find that anglerfish GLP-1a, encoded by gcga, is less biologically active than the gcgb anglerfish GLP-1b paralog. But some other fish (zebrafish, salmon, and catfish) gcga GLP-1a display similar biological activities, indicating that the regulation of glucose metabolism by GLP-1 in ray-finned fish is species-specific. Searches of genomes in cartilaginous fish identified a proglucagon gene that encodes a novel GLP-3 peptide in addition to glucagon, GLP-1, and GLP-2, as well as a single gcgr, glp2r, and a new glucagon receptor-like receptor whose identity still needs to be confirmed. The sequence of the shark GLP-1 contained an N-terminal mammalian-like extension that in mammals undergoes a proteolytic cleavage to release biologically active GLP-1. Our results indicate that early in vertebrate evolution diverse regulatory mechanisms emerged for the control of glucose metabolism by proglucagon-derived peptides and their receptors and that in ray-finned fish they included subfunctionalization and neofunctionalization of these genes.


Fishes/genetics , Proglucagon/physiology , Receptors, Glucagon/physiology , Animals , Carbohydrate Metabolism/genetics , Catfishes/genetics , Energy Metabolism/genetics , Evolution, Molecular , Fishes/classification , Fishes/metabolism , Glucose/metabolism , Phylogeny , Proglucagon/genetics , Receptors, Glucagon/genetics , Salmon/genetics , Zebrafish/genetics
6.
Curr Opin Clin Nutr Metab Care ; 20(4): 300-309, 2017 Jul.
Article En | MEDLINE | ID: mdl-28463898

PURPOSE OF REVIEW: Glucagon and GLP-1 share the same origin (i.e., proglucagon); primarily GLP-1 is generated from intestinal L-cells and glucagon from pancreatic α-cell, but intestinal glucagon and pancreatic GLP-1 secretion is likely. Glucose kinetics are tightly regulated by pancreatic hormones insulin and glucagon, but other hormones, including glucagon-like peptide-1 (GLP-1), also play an important role. The purpose of this review is to describe the recent findings on the mechanisms by which these two hormones regulate glucose kinetics. RECENT FINDINGS: Recent findings showed new important mechanisms of action of glucagon and GLP-1 in the regulation of glucose metabolism. Knock out of glucagon receptors protects against hyperglycemia without causing hypoglycemia. GLP-1 not only stimulates insulin secretion, but it has also an independent effect on the liver and inhibits glucose production. Moreover, when coinfused with glucagon, GLP-1 limits the hyperglycemic effects. Both hormones have also central effects on gastric emptying (delayed), intestinal motility (reduced), and satiety (increased). SUMMARY: The implications of these findings are very important for the management of type 2 diabetes given that GLP-1 receptor agonist are currently approved for the treatment of hyperglycemia and glucagon receptor antagonists and GLP-1/glucagon dual agonists are under development.


Glucagon-Like Peptide 1/physiology , Glucagon/physiology , Glucose/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Fasting , Gastric Emptying/physiology , Gastrointestinal Motility/physiology , Glucagon/blood , Glucagon/pharmacology , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/physiology , Gluconeogenesis/drug effects , Glucose/biosynthesis , Homeostasis , Humans , Hyperglycemia/drug therapy , Kinetics , Liver/drug effects , Liver/metabolism , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/physiology , Satiation/physiology
7.
J Endocrinol ; 228(3): 171-8, 2016 Mar.
Article En | MEDLINE | ID: mdl-26698567

Stimulation of insulin secretion by short-term glucagon receptor (GCGR) activation is well characterized; however, the effect of long-term GCGR activation on ß-cell function is not known, but of interest, since hyperglucagonemia occurs early during development of type 2 diabetes. Therefore, we examined whether chronic GCGR activation affects insulin secretion in glucose intolerant mice. To induce chronic GCGR activation, high-fat diet fed mice were continuously (2 weeks) infused with the stable glucagon analog ZP-GA-1 and challenged with oral glucose and intravenous glucose±glucagon-like peptide 1 (GLP1). Islets were isolated to evaluate the insulin secretory response to glucose±GLP1 and their pancreas were collected for immunohistochemical analysis. Two weeks of ZP-GA-1 infusion reduced insulin secretion both after oral and intravenous glucose challenges in vivo and in isolated islets. These inhibitory effects were corrected for by GLP1. Also, we observed increased ß-cell area and islet size. We conclude that induction of chronic ZP-GA-1 levels in glucose intolerant mice markedly reduces insulin secretion, and thus, we suggest that chronic activation of the GCGR may contribute to the failure of ß-cell function during development of type 2 diabetes.


Glucose Intolerance/physiopathology , Insulin/metabolism , Receptors, Glucagon/physiology , Animals , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat , Female , Glucagon/administration & dosage , Glucagon/analogs & derivatives , Glucagon-Like Peptide 1/administration & dosage , Glucose/administration & dosage , Glucose Intolerance/etiology , Glucose Tolerance Test/methods , Insulin Secretion , Insulin-Secreting Cells/physiology , Mice , Mice, Inbred C57BL , Receptors, Glucagon/drug effects
9.
J Vasc Res ; 52(1): 41-52, 2015.
Article En | MEDLINE | ID: mdl-25966620

UNLABELLED: Diabetic patients have an increased risk of restenosis and late stent thrombosis after angioplasty, i.e. complications that are related to a defective re-endothelialization. Exendin-4, a stable glucagon-like peptide (GLP)-1 receptor agonist, has been suggested to influence the formation of intimal hyperplasia and to increase endothelial cell proliferation in vitro. Thus, the aim of this study was to investigate the mechanisms by which treatment with exendin-4 could influence re-endothelialization and intimal hyperplasia after vascular injury. METHODS: Sprague-Dawley rats were subjected to balloon injury of the left common carotid artery and treated for 4 weeks with exendin-4 or vehicle. Intimal hyperplasia and vessel wall elasticity were monitored noninvasively by high-frequency ultrasound, and re-endothelialization was evaluated upon sacrifice using Evans blue dye. RESULTS AND CONCLUSION: Exendin-4 selectively reduced the proliferation of smooth muscle cells (SMCs) and intimal hyperplasia in vivo without affecting the re-endothelialization process, but treatment with exendin-4 improved arterial wall elasticity. Our data also show that exendin-4 significantly decreased the proliferation and increased the apoptosis of SMCs in vitro, effects that appear to be mediated through cAMP signaling and endothelial nitric oxide synthase following GLP-1 receptor activation. Together, these effects of exendin-4 are highly desirable and may lead to an improved outcome for patients undergoing vascular interventions.


Carotid Artery Injuries/drug therapy , Carotid Artery, Common/pathology , Endothelium, Vascular/drug effects , Peptides/therapeutic use , Receptors, Glucagon/agonists , Venoms/therapeutic use , Animals , Blood Glucose/analysis , Carotid Artery Injuries/pathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/metabolism , Cell Division/drug effects , Cells, Cultured , Cyclic AMP/physiology , Disease Models, Animal , Endothelium, Vascular/physiology , Exenatide , Fatty Acids, Nonesterified/blood , Glucagon-Like Peptide-1 Receptor , Hyperplasia , Insulin/blood , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Nitric Oxide Synthase Type III/metabolism , Peptides/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/physiology , Regeneration/drug effects , Tunica Intima/drug effects , Tunica Intima/pathology , Vascular Stiffness/drug effects , Venoms/pharmacology
10.
Diabetes ; 64(7): 2537-49, 2015 Jul.
Article En | MEDLINE | ID: mdl-25735732

Obesity and diabetes are characterized by increased inflammation reflecting disordered control of innate immunity. We reveal a local intestinal intraepithelial lymphocyte (IEL)-GLP-1 receptor (GLP-1R) signaling network that controls mucosal immune responses. Glp1r expression was enriched in intestinal IEL preparations and copurified with markers of Tαß and Tγδ IELs, the two main subsets of intestinal IELs. Exendin-4 increased cAMP accumulation in purified IELs and reduced the production of cytokines from activated IELs but not from splenocytes ex vivo. These actions were mimicked by forskolin, absent in IELs from Glp1r(-/-) mice, and attenuated by the GLP-1R agonist exendin (9-39) consistent with a GLP-1R-dependent mechanism of action. Furthermore, Glp1r(-/-) mice exhibited dysregulated intestinal gene expression, an abnormal representation of microbial species in feces, and enhanced sensitivity to intestinal injury following administration of dextran sodium sulfate. Bone marrow transplantation using wild-type C57BL/6 donors normalized expression of multiple genes regulating immune function and epithelial integrity in Glp1r(-/-) recipient mice, whereas acute exendin-4 administration robustly induced the expression of genes encoding cytokines and chemokines in normal and injured intestine. Taken together, these findings define a local enteroendocrine-IEL axis linking energy availability, host microbial responses, and mucosal integrity to the control of innate immunity.


Intestinal Mucosa/immunology , Lymphocytes/immunology , Receptors, Glucagon/agonists , Receptors, Glucagon/physiology , Animals , Cytokines/analysis , Exenatide , Female , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Peptides/pharmacology , Signal Transduction , Venoms/pharmacology
11.
Diabetes ; 64(7): 2624-35, 2015 Jul.
Article En | MEDLINE | ID: mdl-25720388

GLP-1 receptor (GLP-1R) agonists may improve endothelial function (EF) via metabolic improvement and direct vascular action. The current study determined the effect of GLP-1R agonist exenatide on postprandial EF in type 2 diabetes and the mechanisms underlying GLP-1R agonist-mediated vasodilation. Two crossover studies were conducted: 36 participants with type 2 diabetes received subcutaneous exenatide or placebo for 11 days and EF, and glucose and lipid responses to breakfast and lunch were determined; and 32 participants with impaired glucose tolerance (IGT) or diet-controlled type 2 diabetes had EF measured before and after intravenous exenatide, with or without the GLP-1R antagonist exendin-9. Mechanisms of GLP-1R agonist action were studied ex vivo on human subcutaneous adipose tissue arterioles and endothelial cells. Subcutaneous exenatide increased postprandial EF independent of reductions in plasma glucose and triglycerides. Intravenous exenatide increased fasting EF, and exendin-9 abolished this effect. Exenatide elicited eNOS activation and NO production in endothelial cells, and induced dose-dependent vasorelaxation and reduced high-glucose or lipid-induced endothelial dysfunction in arterioles ex vivo. These effects were reduced with AMPK inhibition. In conclusion, exenatide augmented postprandial EF in subjects with diabetes and prevented high-glucose and lipid-induced endothelial dysfunction in human arterioles. These effects were largely direct, via GLP-1R and AMPK activation.


Diabetes Mellitus, Type 2/drug therapy , Endothelial Cells/drug effects , Peptides/pharmacology , Receptors, Glucagon/agonists , Vasodilation/drug effects , Venoms/pharmacology , AMP-Activated Protein Kinases/physiology , Blood Glucose/analysis , Cells, Cultured , Cross-Over Studies , Diabetes Mellitus, Type 2/physiopathology , Double-Blind Method , Endothelial Cells/physiology , Exenatide , Female , Glucagon-Like Peptide-1 Receptor , Humans , Male , Receptors, Glucagon/physiology , Triglycerides/blood
12.
Diabetes ; 64(7): 2376-87, 2015 Jul.
Article En | MEDLINE | ID: mdl-25633417

Several lines of evidence implicate excess glucagon secretion in the elevated rates of hepatic glucose production (HGP), hyperglycemia, and ketosis characteristic of uncontrolled insulin-deficient diabetes (uDM), but whether hyperglucagonemia is required for hyperglycemia in this setting is unknown. To address this question, adult male Wistar rats received either streptozotocin (STZ) to induce uDM (STZ-DM) or vehicle and remained nondiabetic. Four days later, animals received daily subcutaneous injections of either the synthetic GLP-1 receptor agonist liraglutide in a dose-escalating regimen to reverse hyperglucagonemia or its vehicle for 10 days. As expected, plasma glucagon levels were elevated in STZ-DM rats, and although liraglutide treatment lowered glucagon levels to those of nondiabetic controls, it failed to attenuate diabetic hyperglycemia, elevated rates of glucose appearance (Ra), or increased hepatic gluconeogenic gene expression. In contrast, it markedly reduced levels of both plasma ketone bodies and hepatic expression of the rate-limiting enzyme involved in ketone body production. To independently confirm this finding, in a separate study, treatment of STZ-DM rats with a glucagon-neutralizing antibody was sufficient to potently lower plasma ketone bodies but failed to normalize elevated levels of either blood glucose or Ra. These data suggest that in rats with uDM, hyperglucagonemia is required for ketosis but not for increased HGP or hyperglycemia.


Diabetes Mellitus, Experimental/complications , Glucagon/blood , Glucose/metabolism , Hyperglycemia/etiology , Ketosis/etiology , Liver/metabolism , Animals , Forkhead Box Protein O1 , Forkhead Transcription Factors/physiology , Glucagon/physiology , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Hyperglycemia/blood , Insulin/pharmacology , Ketone Bodies/blood , Ketosis/blood , Liraglutide , Male , Rats, Wistar , Receptors, Glucagon/physiology , Streptozocin
13.
Diabetes ; 64(1): 79-89, 2015 Jan.
Article En | MEDLINE | ID: mdl-25114295

Glucagon-like peptide-1 (GLP-1) is a hormone that stimulates insulin secretion. Receptors for GLP-1 are also found in the brain, including the hippocampus, the center for memory and learning. Diabetes is a risk factor for decreased memory functions. We studied effects of GLP-1 and exendin-4, a GLP-1 receptor agonist, on γ-aminobutyric acid (GABA) signaling in hippocampal CA3 pyramidal neurons. GABA is the main inhibitory neurotransmitter and decreases neuronal excitability. GLP-1 (0.01-1 nmol/L) transiently enhanced synaptic and tonic currents, and the effects were blocked by exendin (9-39). Ten pmol/L GLP-1 increased both the spontaneous inhibitory postsynaptic current (sIPSC) amplitudes and frequency by a factor of 1.8. In 0.1, 1 nmol/L GLP-1 or 10, 50, or 100 nmol/L exendin-4, only the sIPSC frequency increased. The tonic current was enhanced by 0.01-1 nmol/L GLP-1 and by 0.5-100 nmol/L exendin-4. When action potentials were inhibited by tetrodotoxin (TTX), inhibitory postsynaptic currents decreased and currents were no longer potentiated by GLP-1 or exendin-4. In contrast, although the tonic current decreased in TTX, it was still enhanced by GLP-1 or exendin-4. The results demonstrate GLP-1 receptor regulation of hippocampal function and are consistent with GLP-1 receptor agonists enhancing GABAA signaling by pre- and postsynaptic mechanisms.


CA3 Region, Hippocampal/physiology , Glucagon-Like Peptide 1/physiology , Inhibitory Postsynaptic Potentials/physiology , Peptides/pharmacology , Receptors, GABA-A/physiology , Receptors, Glucagon/physiology , Venoms/pharmacology , Animals , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , Exenatide , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Inhibitory Postsynaptic Potentials/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Memory/physiology , Organ Culture Techniques , Patch-Clamp Techniques , Peptides/physiology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats, Wistar , Receptors, Glucagon/agonists , Receptors, Glucagon/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium Channel Blockers/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tetrodotoxin/pharmacology , gamma-Aminobutyric Acid/metabolism
14.
Biochem Biophys Res Commun ; 456(3): 727-32, 2015 Jan 16.
Article En | MEDLINE | ID: mdl-25511693

Glucagon is released from the pancreatic islets postprandially and under hypoglycemic and cold conditions, and regulates glucose metabolism, feeding, energy expenditure and heat production, the functions partly controlled by the brain. Peripheral glucagon could signal to the brain via passing through the blood-brain barrier and/or acting on the vagal afferent. However, the latter remains to be determined. The present study aimed to clarify whether glucagon directly interacts with the nodose ganglion (NG) neurons of vagal afferent nerves in mice. In vivo study showed that intraperitoneal injection of glucagon induced phosphorylation of extracellular signal regulated kinase 1 and 2 (ERK1/2), cellular activation makers, in NG neurons. In fura-2 microfluorometric studies, glucagon increased cytosolic Ca(2+) concentration ([Ca(2+)]i) in single NG neurons. The glucagon-induced [Ca(2+)]i increases were suppressed by a glucagon receptor antagonist, des-His(1)-[Glu(9)]-Glucagon (1-29) amide, and the glucagon receptor mRNA was expressed in NG neurons. The majority of glucagon-responsive NG neurons exhibited [Ca(2+)]i responses to insulin and cholecystokinin-8, the hormones that are secreted postprandially and implicated in satiety. These results demonstrate that glucagon, by interacting with the glucagon receptor, directly activates vagal afferent nerves, possibly being relayed to the signaling to the brain and formation of satiety.


Calcium Signaling/physiology , Glucagon/physiology , Neurons, Afferent/physiology , Nodose Ganglion/physiology , Receptors, Glucagon/physiology , Satiety Response/physiology , Vagus Nerve/physiology , Animals , Calcium Signaling/drug effects , Glucagon/administration & dosage , Glucagon/analogs & derivatives , Glucagon/pharmacology , Humans , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Neurons, Afferent/drug effects , Neurons, Afferent/enzymology , Nodose Ganglion/drug effects , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptors, Glucagon/antagonists & inhibitors , Satiety Response/drug effects , Vagus Nerve/drug effects
16.
Med Clin (Barc) ; 143 Suppl 2: 2-7, 2014 Sep.
Article Es | MEDLINE | ID: mdl-25437458

The hormone glucagon-like peptide-1 (GLP-1) is synthesized and secreted by L cells in the small intestine in response to food ingestion. After reaching the general circulation it has a half-life of 2-3 minutes due to degradation by the enzyme dipeptidyl peptidase-4. Its physiological role is directed to control plasma glucose concentration, though GLP-1 also plays other different metabolic functions following nutrient absorption. Biological activities of GLP-1 include stimulation of insulin biosynthesis and glucose-dependent insulin secretion by pancreatic beta cell, inhibition of glucagon secretion, delay of gastric emptying and inhibition of food intake. GLP-1 is able to reduce plasma glucose levels in patients with type 2 diabetes and also can restore beta cell sensitivity to exogenous secretagogues, suggesting that the increasing GLP-1 concentration may be an useful therapeutic strategy for the treatment of patients with type 2 diabetes.


Diabetes Mellitus, Type 2/physiopathology , Glucagon-Like Peptide 1/physiology , Glucose/metabolism , Animals , Blood Glucose/analysis , Carbohydrate Metabolism, Inborn Errors/physiopathology , Clinical Trials as Topic , Diabetes Mellitus, Type 2/metabolism , Eating/physiology , Gastric Emptying/physiology , Gastric Inhibitory Polypeptide/physiology , Glucagon/metabolism , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide-1 Receptor , Homeostasis , Humans , Hyperglycemia/physiopathology , Incretins/physiology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Malabsorption Syndromes/physiopathology , Mice , Mice, Knockout , Models, Biological , Obesity/physiopathology , Receptors, Glucagon/deficiency , Receptors, Glucagon/physiology
17.
Med Clin (Barc) ; 143 Suppl 2: 28-34, 2014 Sep.
Article Es | MEDLINE | ID: mdl-25437463

The wide ubiquity of GLP-1 receptors in the body has stimulated the search for different extrapancreatic actions of GLP-1 and its receptor agonists. Thus, severe cardioprotective effects directed on myocardial ischaemia and dysfunction as well as diverse antiaterogenic actions have been reported. Also, native and GLP-1 receptor agonists have demonstrated significant beneficial effects on liver steatosis and fibrosis and on neuronal protection in experimental models of Alzheimer, and Parkinson's disease as well as on cerebral ischaemia. Recent evidences suggest that these drugs may also be useful for prevention and treatment of diabetic retinopathy, nephropathy and peripheral neuropathy. Good results have also been reported in psoriasis. Despite we still need confirmation that these promising effects can be applied to clinical practice, they offer new interesting perspectives for treatment of type 2 diabetes associated complications and give to GLP-1 receptor agonists an even more integral position in diabetes therapy.


Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/physiology , Receptors, Glucagon/agonists , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Obesity Agents/therapeutic use , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cardiovascular Diseases/prevention & control , Clinical Trials as Topic , Disease Models, Animal , Endothelium, Vascular/drug effects , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide-1 Receptor , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Kidney Diseases/prevention & control , Lipid Metabolism/drug effects , Liver Diseases/prevention & control , Multicenter Studies as Topic , Nervous System Diseases/prevention & control , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Organ Specificity , Receptors, Glucagon/physiology , Recombinant Proteins/therapeutic use
18.
Rev Neurol ; 59(11): 517-24, 2014 Dec 01.
Article Es | MEDLINE | ID: mdl-25418147

INTRODUCTION: The glucagon-like peptide-1 (GLP-1) mimetics are an established therapeutic option for patients with type 2 diabetes. However, the properties of the GLP-1 mimetics go beyond the strict metabolic control of the patients with diabetes. The neuroprotective effects of GLP-1 have been shown in recent studies opening new areas of research in neurodegenerative diseases such as Alzheimer's disease (AD), among others. AIM. Systematic review including experimental studies and human clinical trials demonstrating the neuroprotective properties of GLP-1 mimetics in AD. DEVELOPMENT: The experimental studies that have been conducted in rodent models of AD have demonstrated the neuroprotective properties of GLP-1 in the central nervous system reducing beta-amyloid plaques, the oxidative stress and the inflammatory brain response. Clinical trials in patients with cognitive impairment and AD testing the effects of GLP-1 analogs have recently started. CONCLUSION: The GLP-1 analogs have neuroprotective properties. Considering that type 2 diabetes is a risk factor for cognitive impairment and dementia, the benefits of GLP-1 mimetics on cognition must be considered. Likewise, the GLP-1 mimetics represent a promising treatment for neurodegenerative diseases such as AD.


TITLE: Analogos del glucagon-like peptide-1 (GLP-1): una nueva estrategia de tratamiento para la enfermedad de Alzheimer?Introduccion. Los analogos del glucagon-like peptide-1 (GLP-1) son una opcion terapeutica establecida en los pacientes con diabetes tipo 2. Sin embargo, las propiedades de los analogos del GLP-1 van mas alla del control estrictamente metabolico del paciente diabetico. Los efectos neuroprotectores de los analogos del GLP-1 se han puesto de manifiesto en estudios recientes y han abierto nuevos campos de investigacion en trastornos neurodegenerativos como la enfermedad de Alzheimer (EA), entre otros. Objetivo. Revision sistematica de los estudios experimentales y ensayos clinicos en humanos que demuestran las propiedades neuroprotectoras de los analogos del GLP-1 en la EA. Desarrollo. Los estudios experimentales que se han llevado a cabo en modelos de roedores con EA demuestran las propiedades neuroprotectoras de los analogos del GLP-1 sobre el sistema nervioso central que reducen las placas de beta-amiloide, el estres oxidativo y la respuesta inflamatoria cerebral. Recientemente se han puesto en marcha estudios con analogos del GLP-1 en humanos con deterioro cognitivo y EA. Conclusiones. Los analogos del GLP-1 presentan propiedades neuroprotectoras. Al considerarse la diabetes tipo 2 un factor de riesgo para el deterioro cognitivo y la demencia, deben considerarse los beneficios de los analogos del GLP-1 sobre la cognicion. Del mismo modo, los analogos del GLP-1 suponen un tratamiento prometedor en la EA.


Alzheimer Disease/drug therapy , Glucagon-Like Peptide 1/agonists , Neuroprotective Agents/therapeutic use , Alzheimer Disease/metabolism , Animals , Blood-Brain Barrier , Brain Chemistry , Clinical Trials as Topic , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/psychology , Drug Evaluation, Preclinical , Exenatide , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide-1 Receptor , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Incretins/physiology , Insulin Resistance , Liraglutide , Models, Neurological , Neuroprotective Agents/pharmacology , Peptides/pharmacology , Peptides/therapeutic use , Randomized Controlled Trials as Topic , Receptors, Glucagon/drug effects , Receptors, Glucagon/physiology , Risk Factors , Venoms/pharmacology , Venoms/therapeutic use
20.
Thromb Haemost ; 112(5): 1051-64, 2014 Nov.
Article En | MEDLINE | ID: mdl-25030617

High shear stress that develops in the arteriovenous fistula of chronic kidney diseases (CKD) may increase H2O2 and thromboxane A2 (TXA2) release, thereby exacerbating endothelial dysfunction, thrombosis, and neointimal hyperplasia. We investigated whether glucagon-like peptide-1 receptor agonist/exendin-4, a potentially cardiovascular protective agent, could improve TXA2-induced arteriovenous fistula injury in CKD. TXA2 administration to H2O2-exposed human umbilical vein endothelial cells increased apoptosis, senescence, and detachment; these phenotypes were associated with the downregulation of phosphorylated endothelial nitric oxide synthase/heme oxygenase-1 (eNOS/HO-1) signalling. Exendin-4 reduced H2O2/TXA2-induced endothelial injury via inhibition of apoptosis-related mechanisms and restoration of phosphorylated eNOS/HO-1 signalling. Male Wistar rats subjected to right common carotid artery-external jugular vein anastomosis were treated with exendin-4 via cervical implant osmotic pumps for 16-42 days. High shear stress induced by the arteriovenous fistula significantly increased venous haemodynamics, blood and tissue H2O2 and TXB2 levels, macrophage/monocyte infiltration, fibrosis, proliferation, and adhesion molecule-1 expression. Apoptosis was also increased due to NADPH oxidase gp91 activation and mitochondrial Bax translocation in the proximal end of the jugular vein of CKD rats. Exendin-4-treatment of rats with CKD led to the restoration of normal endothelial morphology and correction of arteriovenous fistula function. Exendin-4 treatment or thromboxane synthase gene deletion in CKD mice markedly reduced ADP-stimulated platelet adhesion to venous endothelium, and prevented venous occlusion in FeCl3-injured vessels by upregulation of HO-1. Together, these data reveal that the use of glucagon-like peptide-1 receptor agonists is an effective strategy for treatment of CKD-induced arteriovenous fistula failure.


Arteriovenous Shunt, Surgical/adverse effects , Peptides/therapeutic use , Receptors, Glucagon/agonists , Renal Insufficiency, Chronic/therapy , Venoms/therapeutic use , Venous Thrombosis/prevention & control , Anastomosis, Surgical , Animals , Apoptosis/drug effects , Carotid Artery, Common/pathology , Carotid Artery, Common/surgery , Cell Adhesion/drug effects , Cellular Senescence/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Exenatide , Glucagon-Like Peptide-1 Receptor , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/toxicity , Jugular Veins/pathology , Jugular Veins/surgery , Male , Mice , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Peptides/pharmacology , Rats , Rats, Wistar , Receptors, Glucagon/physiology , Thromboxane A2/toxicity , Venoms/pharmacology , Venous Thrombosis/etiology
...