Intra-islet glucagon signaling is critical for maintaining glucose homeostasis.

Glucagon, a hormone released from pancreatic alpha-cells, plays a key role in maintaining proper glucose homeostasis and has been implicated in the pathophysiology of diabetes. In vitro studies suggest that intra-islet glucagon can modulate the function of pancreatic beta-cells. However, because of the lack of suitable experimental tools, the in vivo physiological role of this intra-islet cross-talk has remained elusive. To address this issue, we generated a novel mouse model that selectively expressed an inhibitory designer G protein-coupled receptor (Gi DREADD) in α-cells only. Drug-induced activation of this inhibitory designer receptor almost completely shut off glucagon secretion in vivo, resulting in significantly impaired insulin secretion, hyperglycemia, and glucose intolerance. Additional studies with mouse and human islets indicated that intra-islet glucagon stimulates insulin release primarily by activating β-cell GLP-1 receptors. These new findings strongly suggest that intra-islet glucagon signaling is essential for maintaining proper glucose homeostasis in vivo. Our work may pave the way toward the development of novel classes of antidiabetic drugs that act by modulating intra-islet cross-talk between α- and β-cells.

[1]  D. Kätzel,et al.  Pharmacokinetic and pharmacodynamic actions of clozapine-N-oxide, clozapine, and compound 21 in DREADD-based chemogenetics in mice , 2019, Scientific Reports.

[2]  P. MacDonald,et al.  β Cell tone is defined by proglucagon peptides through cAMP signaling. , 2019, JCI insight.

[3]  J. Holst,et al.  Insulin Secretion Depends on Intra-islet Glucagon Signaling. , 2018, Cell reports.

[4]  D. Drucker Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. , 2018, Cell metabolism.

[5]  J. Wess,et al.  β-arrestin-2 is an essential regulator of pancreatic β-cell function under physiological and pathophysiological conditions , 2017, Nature Communications.

[6]  K. Kaestner,et al.  High-fidelity Glucagon-CreER mouse line generated by CRISPR-Cas9 assisted gene targeting , 2017, Molecular metabolism.

[7]  Reid H. J. Olsen,et al.  Cre‐dependent DREADD (Designer Receptors Exclusively Activated by Designer Drugs) mice , 2016, Genesis.

[8]  R. Unger,et al.  Glucagon is the key factor in the development of diabetes , 2016, Diabetologia.

[9]  U. Krus,et al.  Altered serotonin (5-HT) 1D and 2A receptor expression may contribute to defective insulin and glucagon secretion in human type 2 diabetes , 2015, Peptides.

[10]  B. Ahrén Glucagon – Early breakthroughs and recent discoveries , 2015, Peptides.

[11]  H. Fu,et al.  Short‐term administration of the glucagon receptor antagonist LY2409021 lowers blood glucose in healthy people and in those with type 2 diabetes , 2015, Diabetes, obesity & metabolism.

[12]  B. Roth,et al.  DREADDs (designer receptors exclusively activated by designer drugs): chemogenetic tools with therapeutic utility. , 2015, Annual review of pharmacology and toxicology.

[13]  Sukriti Goyal,et al.  Computational identification of novel natural inhibitors of glucagon receptor for checking type II diabetes mellitus , 2014, BMC Bioinformatics.

[14]  Shalini Jain,et al.  Novel designer receptors to probe GPCR signaling and physiology. , 2013, Trends in pharmacological sciences.

[15]  M. Ravier,et al.  Tolbutamide Controls Glucagon Release From Mouse Islets Differently Than Glucose , 2013, Diabetes.

[16]  P. MacDonald,et al.  SSTR2 is the functionally dominant somatostatin receptor in human pancreatic β- and α-cells. , 2012, American journal of physiology. Endocrinology and metabolism.

[17]  A. Cherrington,et al.  Glucagonocentric restructuring of diabetes: a pathophysiologic and therapeutic makeover. , 2012, The Journal of clinical investigation.

[18]  D. Accili,et al.  Hormonal regulation of hepatic glucose production in health and disease. , 2011, Cell metabolism.

[19]  B. Roth,et al.  Remote Control of Neuronal Signaling , 2011, Pharmacological Reviews.

[20]  C. Ricordi,et al.  Alpha cells secrete acetylcholine as a non-neuronal paracrine signal priming human beta cell function , 2011, Nature Medicine.

[21]  Jian Hua Li,et al.  A chemical-genetic approach to study G protein regulation of β cell function in vivo , 2009, Proceedings of the National Academy of Sciences.

[22]  Y. Hayashi,et al.  Mice deficient for glucagon gene-derived peptides display normoglycemia and hyperplasia of islet {alpha}-cells but not of intestinal L-cells. , 2009, Molecular endocrinology.

[23]  M. Nicolelis,et al.  Remote Control of Neuronal Activity in Transgenic Mice Expressing Evolved G Protein-Coupled Receptors , 2009, Neuron.

[24]  S. Coughlin,et al.  Probing cell type-specific functions of Gi in vivo identifies GPCR regulators of insulin secretion. , 2007, The Journal of clinical investigation.

[25]  P. Gilon,et al.  The GluCre‐ROSA26EYFP mouse: A new model for easy identification of living pancreatic α‐cells , 2007 .

[26]  B. Roth,et al.  Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand , 2007, Proceedings of the National Academy of Sciences.

[27]  T. Schwartz,et al.  Characterization of glucagon-like peptide-1 receptor beta-arrestin 2 interaction: a high-affinity receptor phenotype. , 2005, Molecular endocrinology.

[28]  B. Wolf,et al.  Structural and functional abnormalities in the islets isolated from type 2 diabetic subjects. , 2004, Diabetes.

[29]  B. Wolf,et al.  Insulin gene transfer enhances the function of human islet grafts , 2003, Diabetologia.

[30]  C. Chu,et al.  The direct effects of catecholamines on hepatic glucose production occur via α1- and β2-receptors in the dog , 2000 .

[31]  J. Reubi,et al.  Somatostatin Receptors , 1997, Trends in Endocrinology & Metabolism.

[32]  J. Habener,et al.  Insulinotropic Glucagon-Like Peptide I Receptor Expression in Glucagon-Producing α-Cells of the Rat Endocrine Pancreas , 1997, Diabetes.

[33]  Shaoping Deng,et al.  Cloning and Functional Expression of the Human Islet GLP-1 Receptor: Demonstration That Exendin-4 Is an Agonist and Exendin-(9–39) an Antagonist of the Receptor , 1993, Diabetes.

[34]  C. Strader,et al.  Cloning and functional expression of a human glucagon-like peptide-1 receptor. , 1993, Biochemical and biophysical research communications.

[35]  D. Drucker,et al.  Glucagon gene expression in vertebrate brain. , 1988, The Journal of biological chemistry.

[36]  J. Holst,et al.  Disruption of glucagon receptor signaling causes hyperaminoacidemia exposing a possible liver-alpha-cell axis. , 2018, American journal of physiology. Endocrinology and metabolism.

[37]  T. Schwartz,et al.  Glucose metabolism is altered after loss of L cells and α-cells but not influenced by loss of K cells. , 2013, American journal of physiology. Endocrinology and metabolism.

[38]  C. Stanley,et al.  Glucokinase activation repairs defective bioenergetics of islets of Langerhans isolated from type 2 diabetics. , 2012, American journal of physiology. Endocrinology and metabolism.

[39]  B. Ahrén Islet G protein-coupled receptors as potential targets for treatment of type 2 diabetes , 2009, Nature Reviews Drug Discovery.

[40]  I. Quesada,et al.  REVIEW Physiology of the pancreatic a -cell and glucagon secretion: role in glucose homeostasis and diabetes , 2008 .

[41]  P. Gilon,et al.  The GluCre-ROSA26EYFP mouse: a new model for easy identification of living pancreatic alpha-cells. , 2007, FEBS letters.

[42]  A. Kelly High Fidelity-What Is It? , 1954 .