Elsevier

Pharmacology & Therapeutics

Volume 139, Issue 3, September 2013, Pages 359-391
Pharmacology & Therapeutics

An atlas and functional analysis of G-protein coupled receptors in human islets of Langerhans

https://doi.org/10.1016/j.pharmthera.2013.05.004Get rights and content

Abstract

G-protein coupled receptors (GPCRs) regulate hormone secretion from islets of Langerhans, and recently developed therapies for type-2 diabetes target islet GLP-1 receptors. However, the total number of GPCRs expressed by human islets, as well as their function and interactions with drugs, is poorly understood. In this review we have constructed an atlas of all GPCRs expressed by human islets: the ‘islet GPCRome’. We have used this atlas to describe how islet GPCRs interact with their endogenous ligands, regulate islet hormone secretion, and interact with drugs known to target GPCRs, with a focus on drug/receptor interactions that may affect insulin secretion. The islet GPCRome consists of 293 GPCRs, a majority of which have unknown effects on insulin, glucagon and somatostatin secretion. The islet GPCRs are activated by 271 different endogenous ligands, at least 131 of which are present in islet cells. A large signalling redundancy was also found, with 119 ligands activating more than one islet receptor. Islet GPCRs are also the targets of a large number of clinically used drugs, and based on their coupling characteristics and effects on receptor signalling we identified 107 drugs predicted to stimulate and 184 drugs predicted to inhibit insulin secretion. The islet GPCRome highlights knowledge gaps in the current understanding of islet GPCR function, and identifies GPCR/ligand/drug interactions that might affect insulin secretion, which are important for understanding the metabolic side effects of drugs. This approach may aid in the design of new safer therapeutic agents with fewer detrimental effects on islet hormone secretion.

Introduction

Islets of Langerhans are clusters of endocrine cells distributed throughout the pancreas that account for 1–2% of the pancreas mass. Islets from all mammalian species are similar in size and contain approximately 1000 endocrine cells. The insulin-secreting β-cell comprises 80–90% of all cells in rodent islets but only 50–60% of the human islet (Cabrera et al., 2006, Kim et al., 2009). Islets also contain glucagon-secreting α-cells and somatostatin-secreting δ-cells, and the balance of islet hormone secretion is critically important in the maintenance of fuel homeostasis. Thus, elevations in circulating glucose concentrations are sensed by β-cells through GLUT1/2 transporters that allow glucose to enter β-cells, where it is metabolised to stimulate insulin secretion when the glucose concentration exceeds 3 mM (in man) and 5 mM (mice). Insulin circulates in the blood and acts at skeletal myocytes and adipocytes to facilitate glucose uptake via membrane insertion of the insulin-sensitive glucose transporter GLUT4, and it also stimulates fuel storage in liver, fat and skeletal muscle. When glucose levels fall in the post-absorptive state glucagon is secreted from islet α-cells and this counter-regulatory hormone restores blood glucose concentrations by stimulating glycogenolysis, gluconeogenesis and lipolysis. Like insulin, somatostatin secretion also increases in response to elevations in glucose and this islet hormone can act in a paracrine manner at β-cells to inhibit insulin release, thus adding a level of fine tuning to minimise the possibility of hypoglycaemia. Collectively, the reciprocal stimulation and inhibition of insulin/somatostatin and glucagon maintains plasma glucose levels within the healthy range of 3.5 mM (during fasting) and 7 mM (post-prandially).

Islets are highly innervated, obtaining input from parasympathetic and sympathetic neurotransmitters and biologically active neuropeptides, and they also have a robust vascular supply that allows circulating agents to regulate islet secretory function (Cabrera et al., 2006, Kim et al., 2009). This permits close regulation of islet hormone secretion, with neurotransmitters such as noradrenaline inhibiting insulin secretion and stimulating glucagon release to increase glucose availability during times of stress. In addition, the incretin GLP-1, released from gastrointestinal L-cells in response to food intake, reaches islet capillary beds via the circulation and reduces blood glucose concentrations through stimulation of insulin secretion and inhibition of glucagon release.

A diverse range of neurotransmitters, neuropeptides and blood-borne regulatory agents exert their effects on islet cells by binding to specific cell surface receptors, the majority of which are seven transmembrane spanning G-protein coupled receptors (GPCRs) that sense changes in the local cellular environment. The roles of some islet GPCRs, such as those responding to the neurotransmitters acetylcholine and noradrenaline with stimulation and inhibition of insulin secretion, respectively, or to the incretin hormones GLP-1 and GIP with potentiation of glucose-stimulated insulin secretion, are well known (Ahren, 2000, Cataland et al., 1974, Kreymann et al., 1987). However, the roles of many other islet GPCRs are poorly understood and very little is known about islet GPCR expression or how the integrated signalling via these GPCRs contributes to deliver the final fine tuning of islet hormone secretion. Moreover, a majority of studies are focused on a limited population of well-known GPCRs, with functional studies carried out predominantly using rodent islets or rodent islet cell lines, which may not always be translatable to human physiology. This review focuses on the expression and function of all known non-olfactory GPCRs in human islets of Langerhans.

The human GPCR superfamily is the largest membrane receptor family in the human genome (Fredriksson et al., 2003). GPCRs regulate a large number of functions in the human body, and they are also important targets of modern medicines. Signalling downstream of GPCR activation following ligand binding is complex, and the net signalling output from individual GPCRs is determined by several different factors such as presence and nature of agonists, constitutive signalling and receptor internalisation, receptor heteromerisation and availability of guanine nucleotide-binding proteins (G-proteins) (Baker & Hill, 2007). GPCR signalling pathways in islet cells, as in all other cell types, involve the interaction of ligand-bound GPCRs with G-proteins, which results in the enzymatic regulation of second messenger generation. The nature of the second messenger signalling depends on which type of G-protein is activated by the GPCR. The major G-protein-mediated signalling pathways are transduced through GPCR interactions with the α subunits of Gs (activates adenylate cyclase to stimulate cAMP production), Gi/Go (inhibits adenylate cyclase to decrease cAMP production), Gq/G11 (activates phospholipase C to generate diacylglycerol and inositol 1,4,5 trisphosphate, which mobilises intracellular Ca2+) and G12/13 (activates the small GTPase Rho to regulate actin cytoskeleton remodelling). GPCRs can also signal via the modulation of ion channel activity, and many Gi/Go coupled GPCRs elevate Ca2+ by activating Ca2+ flux channels (Billington & Penn, 2003), or by direct modulation of exocytosis (Rosengren et al., 2010). GPCR signalling is very complex, and often involves the simultaneous activation of several second messenger systems (Gs and Gq/G11, Gq/G11 and G12/13 or Gi/Go and Gq/G11), with the net effect of activation or inhibition of cAMP production and simultaneous mobilisation of Ca2+ and/or effects on the actin cytoskeleton. In addition, some GPCRs signal via other pathways, such as Wnt signalling by Frizzled receptors (Komiya & Habas, 2008).

In this review data from the IUPHAR GPCR database (Sharman et al., 2011), GeneCards.org, ingenuity.com and PubMed.gov have been used to define an up-to-date functional human GPCRome. The large olfactory receptor subfamily of GPCRs, as well as vomeronasal and pseudogene GPCRs, were not included in this study. Although we could detect human islet mRNAs encoding the receptors BAI1, GPR137, P2RY11 and SSTR5 using non-quantitative PCR, we were not able to identify any qPCR primers suitable for the relative quantification of expression of these receptors (as indicated in Fig. 3). In addition, mRNAs encoding the hydroxycarboxylic acid GPCRs HCAR2 and HCAR3 were not quantified due to the extreme sequence homology between the two genes, which makes it impossible to determine the expression of each of the individual receptors.

Most GPCRs are expressed at relatively low levels, making accurate expression profiling using microarrays challenging (Nagalakshmi et al., 2008), so alternative technologies including quantitative RT-PCR (qPCR) and RNA-sequencing are often used. The relative expression of GPCR mRNAs in mouse islets has been profiled previously by qPCR (Regard et al., 2007, Regard et al., 2008) and we have now used a similar approach to create this atlas of GPCR mRNA expression by human islets. Quantification of the 384 non-odorant human GPCRs currently known was performed using Qiagen's QuantiTect qPCR primers and QuantiFast kits as described elsewhere (Amisten, 2012) with cDNA templates obtained from human islets isolated at the Islet Transplantation Units at King's College London and at the Oxford Centre for Diabetes, Endocrinology and Metabolism. All islet donors were non-diabetic (50% male, 50% female), with an age range of 43–59 years (mean 49.5 ± 3.5) and BMI of 22–33 kg/m2 (mean 28.2 ± 2.3). Human islet GPCR mRNA expression data were normalised against GAPDH mRNA expression in the same samples using the ΔΔCt method (Pfaffl, 2001). All GPCR primers were also used to quantify the GPCRomes in eight unrelated human tissues and cell lines, and the obtained expression profiles from all tissues (data not shown) were matched with published expression and functional data to minimise false negative results due to non-functional qPCR primers.

Of the 384 GPCRs screened, we detected and quantified 293 GPCR mRNAs in human islets by qPCR, which accounts for 76.3% of all known functional, non-odorant GPCRs. Data were manually extracted from PubMed, Ingenuity Pathways Analysis (www.ingenuity.com), the IUPHAR GPCR database (Sharman et al., 2011) and Drug-Bank (Knox et al., 2011) to identify endogenous ligands with established agonist or antagonist effects at these GPCRs. As described in Section 2, at least one endogenous ligand is known for 210 (71.7%) of the GPCRs that we detected in human islets, while 83 (28%) are classified as orphans (including orphan taste receptors) for which no ligands have currently been identified.

Manual PubMed data mining was used to identify publications in which the detected human islet GPCRs have been implicated in the regulation of insulin, glucagon or somatostatin secretion. Most of the published functional studies on islet GPCRs have been performed using isolated mouse or rat islets, and in some cases rodent immortalised cell lines such as MIN6, INS-1, HIT-T15, RINm5F or RIN-14B cells. When data on islet GPCR function were available from multiple sources, the following experimental model preference was used: human islets and human in vivo experiments > mouse, rat islets and rodent in vivo experiments > islet and in vivo experiments from other species (dog, rabbit etc.) > rodent cell lines. It was found that for a majority of the GPCRs identified there is currently no information on their roles in the regulation of islet hormone secretion, as shown in Fig. 1. The GPCRs were grouped based on the IUPHAR classification system into 69 subfamilies based on their ligand usage. Published information on their effects on insulin, glucagon and somatostatin secretion is summarised in Section 3.

Section 4 describes the predicted effects on islet hormone secretion of clinically used drugs that target GPCRs, based on published data on the reported pharmacological effects of therapeutic agents acting as agonists and antagonists at particular GPCRs. Special emphasis has been given to drugs that act as agonists of receptors that inhibit as well as antagonists of receptors that stimulate insulin secretion, as they might have adverse effects on glucose homeostasis and therefore be unsuitable for use in individuals with diabetes.

Section snippets

Human islet G-protein coupled receptor ligand usage

Human islets display a complex network of receptor/ligand interactions, with a large proportion of GPCRs sharing ligands. Endogenous GPCR ligands can be classified according to their molecular structures into four main groups: peptides/proteins, small organic molecules (nucleotides, free fatty acids, amino acids, etc.), monatomic ions (H+, Zn2+, Ca2+) and large biological macromolecules (e.g. glycosaminoglycans). Ligands for the GPCRs expressed in human islets are described below and the

Regulation of islet hormone secretion by G-protein coupled receptors identified in human islets

Agonists that regulate the secretion of islet hormones through GPCR activation do so largely through changes in concentrations of the second messengers cAMP, diacylglycerol, inositol 1,4,5-trisphosphate and intracellular Ca2+. Fig. 3 details the expression levels of all GPCR family mRNAs that we have detected in human islets relative to GAPDH mRNA, together with the known roles of these GPCRs in the regulation of insulin, glucagon and somatostatin secretion, and this information is summarised

Human islet G-protein coupled receptors as drug targets

Surprisingly, only a handful of the 293 human islet GPCRs that we have identified in human islets are approved drug targets for the treatment of type 2 diabetes (T2D): the GLP-1 receptor agonists exenatide and liraglutide (Russell, 2013), as well as the dopamine, serotonin and alpha2-adrenergic receptor agonist bromocriptine. While the mechanisms of action of GLP-1 receptor analogues are well known, the exact mechanism of action of bromocriptine, which lowers Hb1Ac by 0.4–0.8% (Via et al., 2010

Conclusions and perspectives

It has long been known that GPCRs are important regulators of islet function, but very little is known about how a majority of GPCRs participate in this regulation. The way different GPCRs interact as an ‘islet GPCRome’ at the systemic level is also unknown. Perhaps the most surprising observation emanating from this compilation is the large number of GPCR mRNAs present in human islets. In fact, 293 GPCRs, close to 80% of all the 384 functional, non-odorant GPCRs encoded in the human genome,

Acknowledgments

This project was supported by funding from the MRC, Diabetes UK (11/0004172) and the Wellcome Trust. The initial stages of the project were funded by the Crafoord Foundation (Sweden), a Swedish Research Council international postdoctoral fellowship and an EFSD/Lilly Research Fellowship. The funding sources had no involvement in study design, the collection, analysis and interpretation of data, in the writing of the report, and in the decision to submit the paper for publication. We are very

References (284)

  • N. Dufton et al.

    Therapeutic anti-inflammatory potential of formyl-peptide receptor agonists

    Pharmacol Ther

    (2010)
  • G. Fagherazzi et al.

    Consumption of artificially and sugar-sweetened beverages and incident type 2 diabetes in the Etude Epidemiologique aupres des femmes de la Mutuelle Generale de l'Education Nationale-European Prospective Investigation into Cancer and Nutrition cohort

    Am J Clin Nutr

    (2013)
  • H.C. Fehmann et al.

    The effects of two FMRFamide related peptides (A-18-F-amide and F-8-F-amide; ‘morphine modulating peptides’) on the endocrine and exocrine rat pancreas

    Neuropeptides

    (1990)
  • E. Flodgren et al.

    GPR40 is expressed in glucagon producing cells and affects glucagon secretion

    Biochem Biophys Res Commun

    (2007)
  • R. Fujii et al.

    Identification of a neuropeptide modified with bromine as an endogenous ligand for GPR7

    J Biol Chem

    (2002)
  • B.R. Gedulin et al.

    Role of endogenous amylin in glucagon secretion and gastric emptying in rats demonstrated with the selective antagonist, AC187

    Regul Pept

    (2006)
  • C. Gouarderes et al.

    Functional differences between NPFF1 and NPFF2 receptor coupling: high intrinsic activities of RFamide-related peptides on stimulation of [35S]GTPgammaS binding

    Neuropharmacology

    (2007)
  • S. Gregersen et al.

    Endothelin-1 (ET-1)-potentiated insulin secretion: involvement of protein kinase C and the ET(A) receptor subtype

    Metabolism

    (2000)
  • M.R. Heitmeier et al.

    Role of cyclooxygenase-2 in cytokine-induced beta-cell dysfunction and damage by isolated rat and human islets

    J Biol Chem

    (2004)
  • M. Hirano et al.

    Novel reciprocal regulation of cAMP signaling and apoptosis by orphan G-protein-coupled receptor GPRC5A gene expression

    Biochem Biophys Res Commun

    (2006)
  • B. Holst et al.

    Common structural basis for constitutive activity of the ghrelin receptor family

    J Biol Chem

    (2004)
  • Z.A. Abdel-Malek

    Melanocortin receptors: their functions and regulation by physiological agonists and antagonists

    Cell Mol Life Sci

    (2001)
  • J.W. Adams et al.

    Myocardial expression, signaling, and function of GPR22: a protective role for an orphan G protein-coupled receptor

    Am J Physiol Heart Circ Physiol

    (2008)
  • E. Adeghate et al.

    Mechanism of orexin B-stimulated insulin and glucagon release from the pancreas of normal and diabetic rats

    Pancreas

    (2011)
  • E. Adeghate et al.

    Diabetes mellitus influences the degree of colocalization of calcitonin gene-related peptide with insulin and somatostatin in the rat pancreas

    Pancreas

    (2004)
  • B. Ahren

    Autonomic regulation of islet hormone secretion—implications for health and disease

    Diabetologia

    (2000)
  • B. Ahren et al.

    Peptide YY does not inhibit glucose-stimulated insulin secretion in humans

    Eur J Endocrinol

    (1996)
  • B. Ahren et al.

    Effects of vasoactive intestinal polypeptide (VIP), secretin and gastrin on insulin secretion in the mouse

    Diabetologia

    (1981)
  • B. Ahren et al.

    Interaction of vasoactive intestinal peptide (VIP) with cholinergic stimulation of glucagon secretion

    Experientia

    (1982)
  • J.O. Akpan et al.

    The effects of prostaglandins on secretion of glucagon and insulin by the perfused rat pancreas

    Can J Biochem

    (1979)
  • S. Ali et al.

    Dual elimination of the glucagon and GLP-1 receptors in mice reveals plasticity in the incretin axis

    J Clin Invest

    (2011)
  • H.T. Al-Majed et al.

    ACTH stimulates insulin secretion from MIN6 cells and primary mouse and human islets of Langerhans

    J Endocrinol

    (2004)
  • H.T. Al-Majed et al.

    Effect of 17beta-estradiol on insulin secretion and cytosolic calcium in Min6 mouse insulinoma cells and human islets of Langerhans

    Pancreas

    (2005)
  • S. Al-Salam et al.

    Diabetes mellitus decreases the expression of calcitonin-gene related peptide, gamma-amino butyric acid and glutamic acid decarboxylase in human pancreatic islet cells

    Neuro Endocrinol Lett

    (2009)
  • R.S. Ames et al.

    Human urotensin-II is a potent vasoconstrictor and agonist for the orphan receptor GPR14

    Nature

    (1999)
  • S. Amisten

    A rapid and efficient platelet purification protocol for platelet gene expression studies

    Methods Mol Biol

    (2012)
  • S. Amisten et al.

    ADP mediates inhibition of insulin secretion by activation of P2Y13 receptors in mice

    Diabetologia

    (2010)
  • T. Angelone et al.

    Endocrine orchestration of cardiovascular, gastrointestinal and hypothalamic control

    Curr Med Chem

    (2011)
  • R.M. Barbosa et al.

    Control of pulsatile 5-HT/insulin secretion from single mouse pancreatic islets by intracellular calcium dynamics

    J Physiol

    (1998)
  • W.M. Bennet et al.

    Presence of neuropeptide Y and its messenger ribonucleic acid in human islets: evidence for a possible paracrine role

    J Clin Endocrinol Metab

    (1996)
  • F.J. Bermudez-Silva et al.

    Presence of functional cannabinoid receptors in human endocrine pancreas

    Diabetologia

    (2008)
  • C.K. Billington et al.

    Signaling and regulation of G protein-coupled receptors in airway smooth muscle

    Respir Res

    (2003)
  • M.F. Bolliger et al.

    The cell-adhesion G protein-coupled receptor BAI3 is a high-affinity receptor for C1q-like proteins

    Proc Natl Acad Sci U S A

    (2011)
  • M.M. Bonaventura et al.

    GABAB receptors and glucose homeostasis: evaluation in GABAB receptor knockout mice

    Am J Physiol Endocrinol Metab

    (2008)
  • W. Born et al.

    Receptors for calcitonin gene-related peptide, adrenomedullin, and amylin: the contributions of novel receptor-activity-modifying proteins

    Receptors Channels

    (2002)
  • G. Bottcher et al.

    Peptide YY: intrapancreatic localization and effects on insulin and glucagon secretion in the mouse

    Pancreas

    (1989)
  • J.E. Bowe et al.

    GPR54 peptide agonists stimulate insulin secretion from murine, porcine and human islets

    Islets

    (2012)
  • J.E. Bowe et al.

    Kisspeptin stimulation of insulin secretion: mechanisms of action in mouse islets and rats

    Diabetologia

    (2009)
  • M. Braun et al.

    Regulated exocytosis of GABA-containing synaptic-like microvesicles in pancreatic beta-cells

    J Gen Physiol

    (2004)
  • M. Braun et al.

    GABAB receptor activation inhibits exocytosis in rat pancreatic beta-cells by G-protein-dependent activation of calcineurin

    J Physiol

    (2004)
  • Cited by (163)

    View all citing articles on Scopus
    1

    These authors contributed equally.

    View full text