Reviews
G-protein–coupled receptors controlling pancreatic β-cell functional mass for the treatment of type 2 diabetes

https://doi.org/10.1016/j.coemr.2020.09.010Get rights and content

Abstract

The first phase of insulin secretion is altered during the early stages of diabetes and is followed by progressive loss of the ability of glucose to stimulate insulin secretion. The primary causes of insulin secretory defects are still unknown. Better understanding of the signaling pathways linked to the activation of G-protein–coupled receptors and their mutual interactions in the β-cells remains crucial. This represents a prerequisite for any strategy to develop therapeutic tools that aim to modulate the insulin secretory function and/or mass of β-cells. This review summarizes knowledge over the last two years on G-protein–coupled receptors functionally expressed in β-cells, which present key features for the development of therapy for type 2 diabetes.

Introduction

Type 2 diabetes (T2D) is a serious health problem that affects million people worldwide [1]. The pathophysiology of T2D is characterized by insulin resistance and progressive impairment of β-cell function. The insulin resistance of T2D, although important for its pathophysiology, is not sufficient to establish the disease unless major deficiency of β-cell function coexists [2]. Reduced β-cell mass has also been proposed to be associated with T2D [3]. Therefore, innovative therapeutic strategies aiming at preserving β-cell function and survival are of great interest to slow the progression or even to prevent T2D [4,5]. β-Cell dysfunction and death are caused by multiple stressors including chronic hyperglycemia (glucotoxicity), lipotoxicity, oxidative stress, endoplasmic reticulum (ER) stress, the formation of amyloid deposits, and proinflammatory cytokines [6]. Unfortunately, none of the currently and widely prescribed antidiabetic drugs favor the maintenance of endogenous functional β-cell mass, revealing an unmet medical need.

In vivo, the insulin secretion is triggered by the circulating nutrients, mainly by the glucose [7,8]. Glucose triggers insulin secretion when it is taken up into the β-cell through the glucose transporter 2. When glucose is metabolized, the ATP/ADP ratio is increased. This results in the closure of the ATP-sensitive potassium channels and membrane depolarization which favors the opening of the voltage-gated calcium channels increasing the calcium influx within the β-cell. Elevation of the calcium concentration in the cytosol triggers insulin secretion (Figure 1) [8]. The glucose-induced insulin secretion is radically modulated in terms of amplitude by many factors such as hormones, growth factors, or neurotransmitters, most which act by activation or inhibition of intracellular signaling pathways engaged by seven membrane-spanning G-protein–coupled receptors (GPCRs) or tyrosine kinase receptors. Activation of GPCRs results in different β-cell signaling, involving the cAMP/protein kinase A (PKA)/Epac, and the inositol triphosphate (IP3)/diacylglycerol (DAG) pathways, as well as changes in protein phosphorylation and protein acylation. The cAMP/PKA and the IP3/DAG pathways engaged during the activation of GPCRs are among the most important signaling pathways for the β-cell biology (Figure 1). Gαs mediates increases in intracellular cAMP associated with increased insulin secretion, while Gαi mediates decreases in intracellular cAMP and inhibition of insulin secretion. Gαq mediates increases in IP3 and DAG production through the activation of phospholipase C (PLC) associated with increased release of calcium (Ca2+) from the ER and enhanced insulin secretion [9,10]. GPCRs control the dynamics of the exocytosis of insulin granules to maintain the state of differentiation and to regulate the β-cell survival programs [9]. Table 1 presents the GPCRs expressed in β-cells (in bold the GPCRs covered in this review).

Section snippets

Glucagon-like peptide-1 receptor

Specific glucagon-like peptide-1 (GLP-1) receptors (GLP-1Rs) are coupled to Gαs with subsequent activation of adenylate cyclase and elevation of cAMP levels in β-cells. When activated, GLP-1R potentiates glucose-induced insulin secretion through cAMP production. GLP-1R is a key target for T2D treatment [11].

Beyond Gαs coupling to cAMP production, GLP-1R undergoes agonist-mediated endocytosis. Over the last two years, the consequences of modulating GLP-1R endocytic trafficking were investigated.

Glucagon and somatostatin receptors

The islet hormones glucagon and somatostatin affect β-cell function through paracrine effects within the islets: glucagon released from α-cells stimulates insulin secretion and somatostatin released from δ-cells inhibits insulin secretion. Somatostatin from δ-cells acts on SSTR2 receptor coupled to Gi/Go proteins, which results in the inhibition of adenylate cyclase activity and a decrease in cAMP production. δ-Cells and the paracrine interaction between the δ-cells and β-cells were shown to be

Lipid-binding GPCRs

Activation of GPR40 is known to potentiate glucose-induced insulin secretion from β-cells and enhances incretin hormone release from enteroendocrine cells of the small intestine. When activated by long-chain free fatty acids such as palmitate and oleic acid, GPR40 stimulates PLC through Gαq coupling. The IP3 pathway was found to play an important role in GPR40-mediated potentiation of glucose-induced insulin release in β-cells [18]. Full agonist and synthetic small-molecule superagonists of

Neurotransmitter receptor

Activation of the parasympathetic nerves stimulates insulin secretion. The major parasympathetic neurotransmitters are acetylcholine, vasoactive intestinal polypeptide, pituitary, adenylate cyclase–activating polypeptide, and gastrin-releasing peptide [28]. M3 muscarinic ACh receptors (M3Rs) are functionally expressed in β-cells and enhance insulin secretion via coupling to Gαq activating PLC. A positive allosteric modulator of M3R function was found to improve glucose homeostasis in mice by

Novel GPCRs expressed in β-cells

GPR55 is a cannabinoid receptor coupled to Gαq signaling. GPR55 activation was shown to increase glucose-induced insulin secretion, to decrease ER stress-mediated apoptosis, to upregulate antiapoptotic genes such as Bcl-2 and Bcl-xL, and to induce the phosphorylation/activation of the transcription factor cAMP response element binding protein crucial for β-cell survival [30,31].

A class A of GPCR family, GPR142, has been recently reported to be highly expressed in β-cells and to play a role for

New technologies to investigate GPCR expression and signaling

A microfluidic system allowing the registration of intracellular signaling dynamics and hormone secretion within the pancreatic islets was developed and confirmed differences in GPCR signaling pathways between human β- and α-cells [37].

Using quantification of mRNAs encoding all peptide ligands of GPCRs in isolated human and mouse pancreatic islets, Atanes et al [38] proposed detailed islet GPCR peptide ligand atlases favoring the identification of GPCR/peptide signaling pathways relevant for

Toward the development of new therapy for T2D: dual agonist and triagonist strategy to activate GPCRs

Current pharmacological therapies that target single GPCR were reported to exert limited efficacy for T2D treatment. It is important to note that novel approaches with hybrid peptides designed that activate more than one GPCR at once to generate beneficial effects through synergistic actions have shown great promising results for T2D treatment. Unimolecular dual and triagonists, mainly associated with GPCRs such as GLP-1/glucagon/GIP receptors, have shown great efficacy in preclinical models to

Signaling cross-talks between GPCRs, tyrosine kinase, and sulfonylurea receptors

We and others reported signaling cross-talks between GPCRs and tyrosine kinase receptors controlling β-cell function and survival. Regulation of FFAR4 (GPR120) by activation of receptor tyrosine kinases was recently reported. Insulin, insulin-like growth factor-1, epidermal growth factor, and, to a lesser extent, platelet-derived growth factor were found to induce GPR120 internalization. Whether this new evidenced signaling cross-talk between FFAR4/GPR120 and tyrosine kinase receptors is

Conclusion

GPCRs expressed in β-cells receive considerable attention because of their potential as targets in drug development for T2D treatment. Over the next few years, the complex interdependencies between the different messengers and signaling pathways engaged by the activation of GPCRs in β-cells that regulate and shape the insulin secretory response have to be considered. In addition, GPCRs with known ligands need to be further studied in terms of ligand specificity, intracellular signaling, and

Conflict of interest statement

Nothing declared.

Acknowledgements

Morgane Delobel is supported by the CIFRE grant Phd fellowhip from MedBioMed.

References (47)

  • O. Rudenko et al.

    The aromatic amino acid sensor GPR142 controls metabolism through balanced regulation of pancreatic and gut hormones

    Mol Metab

    (2019)
  • J.T. Walker et al.

    Integrated human pseudoislet system and microfluidic platform demonstrate differences in GPCR signaling in islet cells

    JCI Insight

    (2020)
  • P. Atanes et al.

    Defining G protein-coupled receptor peptide ligand expressomes and signalomes in human and mouse islets

    Cell Mol Life Sci

    (2018)
  • P. Ambery et al.

    MEDI0382, a GLP-1 and glucagon receptor dual agonist, in obese or overweight patients with type 2 diabetes: a randomised, controlled, double-blind, ascending dose and phase 2a study

    Lancet

    (2018)
  • C.J. Nolan et al.

    Insulin resistance and insulin hypersecretion in the metabolic syndrome and type 2 diabetes: time for a conceptual framework shift

    Diabetes Vasc Dis Res

    (2019)
  • A.E. Butler et al.

    Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes

    Diabetes

    (2003)
  • A. Yehya et al.

    New therapeutic strategies for type 2 diabetes (CME)

    Methodist Debakey Cardiovasc J

    (2018)
  • E. Burgos-Moron et al.

    Relationship between oxidative stress, ER stress, and inflammation in type 2 diabetes: the battle continues

    J Clin Med

    (2019)
  • J.C. Henquin

    Triggering and amplifying pathways of regulation of insulin secretion by glucose

    Diabetes

    (2000)
  • T. Buenaventura et al.

    A targeted RNAi screen identifies endocytic trafficking factors that control GLP-1 receptor signaling in pancreatic beta-cells

    Diabetes

    (2018)
  • B. Jones et al.

    Targeting GLP-1 receptor trafficking to improve agonist efficacy

    Nat Commun

    (2018)
  • T. Buenaventura et al.

    Agonist-induced membrane nanodomain clustering drives GLP-1 receptor responses in pancreatic beta cells

    PLoS Biol

    (2019)
  • Y. Manchanda et al.

    Spatiotemporal control of GLP-1 receptor activity

    Curr Opin Endocr Metabol Res

    (February 2021)
  • Cited by (0)

    View full text