Exploring the insulin secretory properties of the PGD2-GPR44/DP2 axis in vitro and in a randomized phase-1 trial of type 2 diabetes patients

Stanko Skrtic, Björn Tyrberg, Malin Broberg, Hans Ericsson, Volker Schnecke, Magnus Kjaer, Marcus Hompesch, Eva-Marie Andersson, Erik Ryberg, Alexander Aivazidis, Charlotte Wennberg Huldt, Lars Löfgren, Linda Morrow, Joanna Parkinson, Tina Rydén-Bergsten, Elaine Watkins, Maria Sörhede Winzell, Stanko Skrtic, Björn Tyrberg, Malin Broberg, Hans Ericsson, Volker Schnecke, Magnus Kjaer, Marcus Hompesch, Eva-Marie Andersson, Erik Ryberg, Alexander Aivazidis, Charlotte Wennberg Huldt, Lars Löfgren, Linda Morrow, Joanna Parkinson, Tina Rydén-Bergsten, Elaine Watkins, Maria Sörhede Winzell

Abstract

Aims/hypothesis: GPR44 (DP2, PTGDR2, CRTh2) is the receptor for the pro-inflammatory mediator prostaglandin D2 (PGD2) and it is enriched in human islets. In rodent islets, PGD2 is produced in response to glucose, suggesting that the PGD2-GPR44/DP2 axis may play a role in human islet function during hyperglycemia. Consequently, the aim of this work was to elucidate the insulinotropic role of GPR44 antagonism in vitro in human beta-cells and in type 2 diabetes (T2DM) patients.

Methods: We determined the drive on PGD2 secretion by glucose and IL-1beta, as well as, the impact on insulin secretion by pharmacological GPR44/DP2 antagonism (AZD1981) in human islets and beta-cells in vitro. To test if metabolic control would be improved by antagonizing a hyperglycemia-driven increased PGD2 tone, we performed a proof-of-mechanism study in 20 T2DM patients (average 54 years, HbA1c 9.4%, BMI 31.6 kg/m2). The randomized, double-blind, placebo-controlled cross-over study consisted of two three-day treatment periods (AZD1981 or placebo) separated by a three-day wash-out period. Mixed meal tolerance test (MMTT) and intravenous graded glucose infusion (GGI) was performed at start and end of each treatment period. Assessment of AZD1981 pharmacokinetics, glucose, insulin, C-peptide, glucagon, GLP-1, and PGD2 pathway biomarkers were performed.

Results: We found (1) that PGD2 is produced in human islet in response to high glucose or IL-1beta, but likely by stellate cells rather than endocrine cells; (2) that PGD2 suppresses both glucose and GLP-1 induced insulin secretion in vitro; and (3) that the GPR44/DP2 antagonist (AZD1981) in human beta-cells normalizes insulin secretion. However, AZD1981 had no impact on neither glucose nor incretin dependent insulin secretion in humans (GGI AUC C-peptide 1-2h and MMTT AUC Glucose 0-4h LS mean ratios vs placebo of 0.94 (80% CI of 0.90-0.98, p = 0.12) and 0.99 (90% CI of 0.94-1.05, p = 0.45), despite reaching the expected antagonist exposure.

Conclusion/interpretation: Pharmacological inhibition of the PGD2-GPR44/DP2 axis has no major impact on the modulation of acute insulin secretion in T2DM patients.

Trial registration: ClinicalTrials.gov NCT02367066.

Conflict of interest statement

All authors apart from MH, LM and EW are employees or shareholders of AstraZeneca AB. MH, LM, and EW received funding as a contract research organization by AstraZeneca for the clinical study. EW is affiliated with ProSciento Inc. There are no patents, products in development, or marketed products to declare. This does not alter our adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Study flow chart according to…
Fig 1. Study flow chart according to CONSORT 2010 guidelines.
The design of a human Proof of Mechanism study to test the Mechanism of Action hypothesis. The phase 1 study was designed as a randomised, double-blind, placebo-controlled, multiple-dose, cross-over study, consisting of a run-in, two treatment periods separated by a wash-out period and a follow-up for each subject. The subjects were T2DM patients on metformin (n = 20).
Fig 2. Tissue expression of GPR44/DP2 and…
Fig 2. Tissue expression of GPR44/DP2 and PGD2 related genes and characterization of pancreatic stellate cells.
PTGDR2 (GPR44/DP2) mRNA expression in human beta-cells (Healthy, n = 197; T2DM, n = 112) (a) and pancreatic stellate cells (Healthy, n = 23; T2DM, n = 31) (b). Heat map of stellate cell signature gene expression sub-classified as quiescent, immune activated (imm) and standard activated (stand) stellate cells according to the description in the main text (c). Quantification of stellate cell subpopulations in healthy and T2D donor human islets (d). Expression of GPR44/DP2 and PGD2 related genes in subclasses of stellate cells (e). Data are presented as mean ± SEM (a-b), total number of cells (d), and individual cells (e). *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001.
Fig 3. Human islet gene expression and…
Fig 3. Human islet gene expression and regulation by high glucose with and without IL-1beta.
Expression of PTGDR2 (GPR44/DP2) (a), PTGDR1 (DP1) (b), PTGDS (L-PGDS) (c), PTGS1 (COX-1) (d) and PTGS2 (COX-2) (e). In vitro human islet PGD2 secretion (f). Data are presented as mean ± SEM from 5 different human islet donors, where each condition was evaluated in quadruplicates. Glc = glucose. *p<0.05, **p<0.01, ****p<0.0001.
Fig 4. Effect of PGD 2 on…
Fig 4. Effect of PGD2 on insulin secretion and in vitro pharmacology of AZD1981 in the human beta-cell line EndoC- betaH1.
Dose response curve for the PGD2 analogue (15(R)-15-methyl-PGD2) on insulin secretion, from which the EC50 was determined (a). The in vitro potency of the GPR44/DP2 antagonist AZD1981 was determined using DMR (IC50 = 4.9±1nM) (b), cAMP (IC50 = 21±8nM) (c) and by glucose-stimulated insulin secretion (IC50 = 13±2nM) (d). The assays were performed in the human beta cell line EndoC-betaH1 with addition of 150 pM 15(R)-15-methyl-PGD2. Insulin secretion in EndoC-betaH1 cells was determined with and without addition of 150pM 15(R)-15-methyl-PGD2 at 11.1 mM glucose and 100nM exendin-4 (GLP-1 receptor agonist). 100nM of AZD1981 was added to restore GSIS. The effect of PGD2 on human islet insulin secretion was studied in islets from three donors where each incubation condition was run with 5 islets in 6 replicates (f). Data are presented as mean ± SEM.*p<0.05, **p<0.01***p<0.001.
Fig 5. Hypothesis for the endogenous PGD…
Fig 5. Hypothesis for the endogenous PGD2-GPR44/DP2 axis mechanism of action in islets.
Glucose and IL-1beta induced PTGS2 (COX-2) expression in stellate cells which stimulates the production of PGD2 synthesis. PGD2 is secreted and may act in a paracrine fashion on GPR44/DP2 on beta-cells, inhibiting cAMP and thereby reducing GSIS.
Fig 6. Mixed Meal Tolerance Test in…
Fig 6. Mixed Meal Tolerance Test in T2DM patients.
Glucose assessments at baseline (a), and after 3 days treatment (b) or Insulin Secretion Rate (ISR) at baseline (c) and after 3 days treatment (d). Circles connected with dotted lines represent placebo and squares connected with full lines represent AZD1981. Data are presented as mean ± SD.

References

    1. White MG, Shaw JA, Taylor R. Type 2 Diabetes: The Pathologic Basis of Reversible beta-Cell Dysfunction. Diabetes Care. 2016;39(11):2080–8.
    1. Song I, Muller C, Louw J, Bouwens L. Regulating the beta cell mass as a strategy for type-2 diabetes treatment. Current drug targets. 2015;16(5):516–24.
    1. O'Byrne PM, Naji N, Gauvreau GM. Severe asthma: future treatments. Clinical and experimental allergy: journal of the British Society for Allergy and Clinical Immunology. 2012;42(5):706–11.
    1. Norman P. Update on the status of DP2 receptor antagonists; from proof of concept through clinical failures to promising new drugs. Expert Opin Investig Drugs. 2013;23(1):55–66.
    1. Kuna P, Bjermer L, Tornling G. Two Phase II randomized trials on the CRTh2 antagonist AZD1981 in adults with asthma. Drug design, development and therapy. 2016;10:2759–70.
    1. Luker T, Bonnert R, Brough S, Cook AR, Dickinson MR, Dougall I, et al. Substituted indole-1-acetic acids as potent and selective CRTh2 antagonists-discovery of AZD1981. Bioorg Med Chem Lett. 2011;21(21):6288–92.
    1. Hellström-Lindahl E, Danielsson A, Ponten F, Czernichow P, Korsgren O, Johansson L, et al. GPR44 is a pancreatic protein restricted to the human beta cell. Acta Diabetol. 2016;53(3):413–21.
    1. Lindskog C, Korsgren O, Ponten F, Eriksson JW, Johansson L, Danielsson A. Novel pancreatic beta cell-specific proteins: antibody-based proteomics for identification of new biomarker candidates. Journal of proteomics. 2012;75(9):2611–20.
    1. Amisten S, Salehi A, Rorsman P, Jones PM, Persaud SJ. An atlas and functional analysis of G-protein coupled receptors in human islets of Langerhans. Pharmacology & therapeutics. 2013;139(3):359–91.
    1. Hata AN, Zent R, Breyer MD, Breyer RM. Expression and molecular pharmacology of the mouse CRTH2 receptor. The Journal of pharmacology and experimental therapeutics. 2003;306(2):463–70.
    1. Evans MH, Pace CS, Clements RS Jr. Endogenous prostaglandin synthesis and glucose-induced insulin secretion from the adult rat pancreatic islet. Diabetes. 1983;32(6):509–15.
    1. Robertson RP. Eicosanoids as pluripotential modulators of pancreatic islet function. Diabetes. 1988;37(4):367–70.
    1. Kimple ME, Keller MP, Rabaglia MR, Pasker RL, Neuman JC, Truchan NA, et al. Prostaglandin E2 receptor, EP3, is induced in diabetic islets and negatively regulates glucose- and hormone-stimulated insulin secretion. Diabetes. 2013;62(6):1904–12.
    1. Schmidt JA, Bell FM, Akam E, Marshall C, Dainty IA, Heinemann A, et al. Biochemical and pharmacological characterization of AZD1981, an orally available selective DP2 antagonist in clinical development for asthma. Br J Pharmacol. 2013;168(7):1626–38.
    1. Ravassard P, Hazhouz Y, Pechberty S, Bricout-Neveu E, Armanet M, Czernichow P, et al. A genetically engineered human pancreatic β cell line exhibiting glucose-inducible insulin secretion. J Clin Invest. 2011;121(9):3589–97.
    1. Segerstolpe A, Palasantza A, Eliasson P, Andersson EM, Andreasson AC, Sun X, et al. Single-Cell Transcriptome Profiling of Human Pancreatic Islets in Health and Type 2 Diabetes. Cell Metab. 2016;24(4):593–607.
    1. Zaccaria M, De Palo E, Zago E, Sicolo N, Erle G, Federspil G. Metabolic and endocrine responses to a standard mixed meal. A physiologic study. Acta Diabetol Lat. 1979;16(1):45–53.
    1. Byrne MM, Sturis J, Polonsky KS. Insulin secretion and clearance during low-dose graded glucose infusion. Am J Physiol. 1995;268(1 Pt 1):E21–7.
    1. Solis-Herrera C, Triplitt C, Garduno-Garcia JeJ, Adams J, DeFronzo RA, Cersosimo E. Mechanisms of glucose lowering of dipeptidyl peptidase-4 inhibitor sitagliptin when used alone or with metformin in type 2 diabetes: a double-tracer study. Diabetes Care. 2013;36(9):2756–62.
    1. Hovorka R, Chassin L, Luzio SD, Playle R, Owens DR. Pancreatic beta-cell responsiveness during meal tolerance test: model assessment in normal subjects and subjects with newly diagnosed noninsulin-dependent diabetes mellitus. J Clin Endocrinol Metab. 1998;83(3):744–50.
    1. Baron M, Veres A, Wolock SL, Faust AL, Gaujoux R, Vetere A, et al. A Single-Cell Transcriptomic Map of the Human and Mouse Pancreas Reveals Inter- and Intra-cell Population Structure. Cell systems. 2016;3(4):346–60 e4.
    1. Schröder R, Janssen N, Schmidt J, Kebig A, Merten N, Hennen S, et al. Deconvolution of complex G protein-coupled receptor signaling in live cells using dynamic mass redistribution measurements. Nat Biotechnol. 2010;28(9):943–9.
    1. Carboneau BA, Breyer RM, Gannon M. Regulation of pancreatic β-cell function and mass dynamics by prostaglandin signaling. J Cell Commun Signal. 2017.
    1. Monnier L, Colette C, Dunseath GJ, Owens DR. The loss of postprandial glycemic control precedes stepwise deterioration of fasting with worsening diabetes. Diabetes Care. 2007;30(2):263–9.
    1. Kjems LL, Holst JJ, Vølund A, Madsbad S. The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes. 2003;52(2):380–6.
    1. Schuligoi R, Schmidt R, Geisslinger G, Kollroser M, Peskar BA, Heinemann A. PGD2 metabolism in plasma: kinetics and relationship with bioactivity on DP1 and CRTH2 receptors. Biochem Pharmacol. 2007;74(1):107–17.
    1. Murata T, Maehara T. Discovery of anti-inflammatory role of prostaglandin D2. The Journal of veterinary medical science. 2016;78(11):1643–7.
    1. Parazzoli S, Harmon JS, Vallerie SN, Zhang T, Zhou H, Robertson RP. Cyclooxygenase-2, not microsomal prostaglandin E synthase-1, is the mechanism for interleukin-1beta-induced prostaglandin E2 production and inhibition of insulin secretion in pancreatic islets. J Biol Chem. 2012;287(38):32246–53.
    1. Shanmugam N, Todorov IT, Nair I, Omori K, Reddy MA, Natarajan R. Increased expression of cyclooxygenase-2 in human pancreatic islets treated with high glucose or ligands of the advanced glycation endproduct-specific receptor (AGER), and in islets from diabetic mice. Diabetologia. 2006;49(1):100–7.
    1. Omary MB, Lugea A, Lowe AW, Pandol SJ. The pancreatic stellate cell: a star on the rise in pancreatic diseases. J Clin Invest. 2007;117(1):50–9.
    1. Apte MV, Wilson JS. Dangerous liaisons: pancreatic stellate cells and pancreatic cancer cells. J Gastroenterol Hepatol. 2012;27 Suppl 2:69–74.
    1. Pandol S, Gukovskaya A, Edderkaoui M, Edderkoui M, Dawson D, Eibl G, et al. Epidemiology, risk factors, and the promotion of pancreatic cancer: role of the stellate cell. J Gastroenterol Hepatol. 2012;27 Suppl 2:127–34.
    1. Ryall CL, Viloria K, Lhaf F, Walker AJ, King A, Jones P, et al. Novel role for matricellular proteins in the regulation of islet β cell survival: the effect of SPARC on survival, proliferation, and signaling. J Biol Chem. 2014;289(44):30614–24.
    1. Li FF, Chen BJ, Li W, Li L, Zha M, Zhou S, et al. Islet Stellate Cells Isolated from Fibrotic Islet of Goto-Kakizaki Rats Affect Biological Behavior of Beta-Cell. J Diabetes Res. 2016;2016:6924593

Source: PubMed

3
订阅