Skip to main content

Exploring G Protein-Coupled Receptor Signaling in Primary Pancreatic Islets

Abstract

Background

Targeting G protein-coupled receptors (GPCRs) in pancreatic cells is feasible to modulate glucose-induced insulin secretion. Because pancreatic islets consist of several cell types and GPCRs can couple to more than one G-protein family, results obtained in pancreatic cell lines do not always match the response in primary cells or intact islets. Therefore, we set out to establish a protocol to analyze second messenger activation in mouse pancreatic islets.

Results

Activation of Gq/11-coupled receptor expressed in primary β cells increased the second messenger IP1 in an accumulation assay. Applying a Gq/11 protein inhibitor completely abolished this signal. Activation of the V1 vasopressin and ghrelin receptors, predominantly expressed in the less abundant alpha and delta cells, was not sufficient to induce a significant IP1 increase in this assay. However, fura-2-based fluorescence imaging showed calcium signals upon application of arginine vasopressin or ghrelin within intact pancreatic islets. Using the here established protocol we were also able to determine changes in intracellular cAMP levels induced by receptors coupling to Gs and Gi/o proteins.

Conclusions

Detection of the second messengers IP1, cAMP, and calcium, can be used to reliably analyze GPCR activation in intact islets.

Background

Diabetes mellitus is a major metabolic disorder characterized by high glucose levels in the blood which can lead to secondary diseases. Because more than 425 million people world-wide are affected by diabetes, development of novel therapeutics is one of the major current health care challenges. Type 2 diabetes (T2D), accounting for more than 90% of the patients [1], results from insulin resistance of peripheral tissues and impaired β-cell function including altered hormone secretion [2]. The main signal for secretion of insulin, glucagon, and somatostatin is the changing blood glucose level. It is well-established that secretion of all pancreatic islet hormones can be modified by intracellular cyclic AMP (cAMP) and calcium ion levels [3, 4]. G protein-coupled receptors (GPCRs), a superfamily of transmembrane receptors, are the main modulators of these signal molecules. They are considered as major drug targets as they transduce extracellular signals into intracellular responses [5]. Agonists of GPCRs are already used in T2D therapy or are currently in clinical trials [6,7,8,9].

Almost 300 GPCRs expressed in pancreatic islets offer multiple opportunities to modulate insulin, glucagon, and somatostatin secretion [10, 11]. However, most of these receptors are still considered orphans because their agonists and/or their signaling properties are currently unknown [12]. Further complexity is added by the fact that most GPCRs couple to multiple G proteins depending on the cell type and cellular environment where they are expressed in [13]. To evaluate the coupling properties of GPCRs in pancreatic islets, islet-derived cell lines are usually analyzed to determine second messenger responses of GPCRs [14,15,16]. However, results obtained from cell lines suggest aberrant properties, such as a different glucose-induced insulin secretion (GSIS) [17]. Further studies show differences in β cell-derived cell lines and β cells isolated from islets regarding their response to nutrients [18, 19] and their GPCR expression patterns. For example, the β cell-derived cell lines RINm5F and INS-1 express greatly reduced amounts of muscarinic acetylcholine receptor type 3 (M3R) compared to pancreatic islets, and thus, no carbachol (CCh)-induced inositol phosphate accumulation and insulin secretion was detectable in these cell lines [20]. Growth hormone secretagogue receptor/ghrelin receptor (GHS-R) expression has been shown in the β cell-derived cell lines INS-1 and MIN6 [21, 22], while transcriptome analysis of primary islets revealed no GHS-R expression in β cells but only in δ cells [23, 24]. These transcriptome data finally explained the paradox why the Gq/11-coupled GHS-R [25] induced a significant reduction in insulin secretion [26, 27]. The UDP receptor P2Y6 triggered a reduction of GSIS in the β-TC6 cell line while P2Y6 activation in β cells isolated from primary islets increases GSIS [28, 29]. Activation of GPR40 by the agonistic compound GW9508 led to increased GSIS in MIN6 cells but not primary islets [30]. Furthermore, variable coupling properties of GPCRs have been found in different β cell lines. For example, activation of the free fatty acid receptor 2 (FFAR2) by acetate induced a Gq/11 protein-mediated IP1 increase in MIN6 cells but a Gi protein-mediated reduction of cAMP in INS-1 cells [31].

In view of these challenges, methods are required to characterize G protein-signaling cascades in primary pancreatic islets. In particular, the function of a large number of less characterized or orphan GPCRs remains to be clarified in pancreatic islets. Progress was made in monitoring cAMP in primary islets by using genetically modified mouse models expressing FRET-based cAMP sensors [32]. However, this method relies on a transgenic mouse model expressing the cAMP sensor. Therefore, we set out to determine second messenger accumulation in isolated wild-type mouse islets. Thus, we adapted established second messenger protocols routinely performed in cell lines for determining similar responses ex vivo, i.e. in whole or dispersed pancreatic islets. We studied well-characterized GPCRs known to be expressed in pancreatic islets and evaluated incubation procedures as well as islet handling.

Results

RNAseq-based expression analysis of mouse pancreatic islets [33] revealed highly expressed GPCRs with well-studied signal transduction properties (Table 1). These receptors were used as proof-of-principle to establish protocols for determination of their second messengers. Thus, islets (150 ± 12 per preparation) from mouse pancreata were isolated and disintegrated using trypsin in Ca2+-free conditions. After passaging through a 35 μm filter, we obtained 67,000 ± 9000 viable cells per preparation which were sufficient to load 13 wells of a 384-well plate (Fig. 1).

Table 1 GPCRs tested in this study. Receptors are given in order from high to low expression [33]
Fig. 1
figure 1

Schematic depiction of the experimental procedures necessary for second messenger analysis in primary islet cells. Islets are isolated by collagenase digestion and disintegrated using Trypsin/EDTA solution. Second messenger determination was carried out in multi-well format

IP1 Measurement

In a first approach, we used carbachol (CCh) to stimulate M3R, which is highly expressed in the β cell-derived cell line MIN6 and in pancreatic islets, to evaluate optimal conditions for determining the second messenger IP1. For control purposes, MIN6 cells were incubated with CCh to stimulate endogenously expressed receptors and accumulation of IP1 was determined. CCh significantly increased IP1 concentration after 1 h of stimulation to 2.1-fold of the basal level (Fig. 2a). Application of a specific inhibitor for Gq/11 proteins, FR900359 [34], completely abolished basal and CCh-stimulated receptor activity (Fig. 2a). To transfer this established method to primary pancreatic islet cells, dispersed islets were seeded into poly-L-lysine-coated 96-well plates to become adherent overnight. Stimulation with CCh for 4 h at 37 °C did not lead to an increase in IP1 concentration (Fig. 2b). Therefore, an IP1 accumulation protocol for suspension cells was used and CCh was applied for 1 or 4 h. Whereas incubation for 4 h did not lead to an increase in accumulated IP1 (Fig. 2c), stimulation with CCh for 1 h induced a significant increase in IP1 concentration to 210% of the basal level (Fig. 2d). FR900359, the specific inhibitor of Gq/11 proteins completely abolished CCh-induced IP1 accumulation, indicating Gq/11-dependent signal transduction in dispersed islets (Fig. 2d).

Fig. 2
figure 2

Optimization of conditions to measure IP1 accumulation in dispersed islets. a MIN6 cells were seeded into 96-well plates and incubated with 100 μM CCh with or without the Gq/11 inhibitor FR900359. CCh induced a significant increase in IP1 concentration which was completely abolished by FR900359. b Dispersed islets were seeded into coated 96-well plates to become adherent overnight and incubated with 100 μM CCh for 4 h, however, CCh failed to induce IP1 accumulation. c Using a suspension-based protocol for IP1 determination, dispersed islets were exposed immediately after dispersion to 100 μM CCh for 4 h. CCh also failed to raise IP1 levels in primary islet cells. d Reducing the incubation time to 1 h led to a significant increase of IP1 in CCh treated cells. This effect was dependent on Gq/11 activity because application of the Gq/11 inhibitor FR900359 completely abolished IP1 accumulation. Data are given as means ± SEM of three to five independent experiments each performed in triplicates. Statistical significance was tested with the two-tailed paired t test (**p ≤ 0.01; ***p ≤ 0.001)

We then analyzed IP1 responses of other well-characterized Gq/11-coupled GPCRs (Table 1) displaying high expression in pancreatic islets [33] in suspended cells under the above optimized conditions (Fig. 3). We stimulated disintegrated islets with the P2Y6 receptor agonist UDP (Fig. 3a) and the FFAR2 agonist acetate (Fig. 3b) and observed a significant increase in IP1 concentration by 1.5- and 2.5-fold, respectively. This data indicates that activation of receptors highly expressed in pancreatic β cells induce a measurable IP1 accumulation. However, stimulation of receptors predominantly expressed in α and δ cells was not sufficient to induce a significant IP1 response. Neither activation of growth hormone secretagogue receptor/ghrelin receptor (GHS-R) in δ cells (Fig. 3c) nor stimulation of vasopressin receptor type 1 (V1R) by arginine vasopressin (AVP) in α and δ cells (Fig. 3d) resulted in an increase of intracellular IP1 concentration.

Fig. 3
figure 3

Analyzing IP1 response of different Gq/11 protein-coupled receptors expressed in pancreatic islets. a, b Stimulation of receptors highly expressed in pancreatic β cells resulted in a significant increase in IP1 concentration. P2Y6 (a) was stimulated with 10 μM UDP and FFAR2 (b) with 1 mM acetate. c, d Activation of GHS-R expressed in δ cells by 300 nM ghrelin (c) and V1R expressed in α and δ cells by 100 nM AVP (d) did not lead to a significant increase of IP1 concentration. Data are given as means ± SEM of three experiments each performed in triplicates. Statistical significance was tested with a two-tailed paired t test (*p ≤ 0.05; **p ≤ 0.01)

cAMP Measurement

Activation of GPCRs coupling to either Gs- or Gi/o proteins changes the intracellular cAMP concentration which in turn modulates pancreatic hormone secretion. To optimize conditions for intracellular cAMP determination we initially stimulated adenylyl cyclases nonspecifically with forskolin (Fig. 4a). The most stable cAMP accumulation was achieved by simultaneous incubation of islet cells with forskolin and the beads necessary for cAMP detection. We next targeted glucagon-like peptide 1 receptor (GLP-1R), the most abundant GPCR in pancreatic islets [33]. Incubation of pancreatic islet cells with 100 nM GLP-1 yielded a significant increase of cAMP concentration to 690% of the basal level (Fig. 4b). For analysis of Gi/o protein-coupled receptors, a preceding stimulation with forskolin is required to achieve reducible cAMP concentrations. We stimulated ubiquitously expressed somatostatin receptors (SstR) with somatostatin (Sst-14) and observed a potent reduction of the forskolin-induced cAMP levels to 13% compared to forskolin-stimulated islet cells (Fig. 4c).

Fig. 4
figure 4

Analyzing cAMP levels in response to activation of Gs- or Gi/o protein-coupled receptors in dispersed islets. a The unspecific activation of adenylyl cyclases by 10 μM forskolin resulted in an almost 8-fold increase of cAMP. b Activation of the highly expressed GLP-1R by its agonist induced a significant increase in cAMP (> 6-fold). c Incubation with 1 μM forskolin increased intracellular cAMP concentration which can be reduced by 1 μM Sst-14 due to activation of Gi/o protein-coupled SstR. Given are the means ± SEM of four to five experiments each performed in triplicates. Statistical significance was tested with the two-tailed paired t test (*p ≤ 0.05; **p ≤ 0.01)

Ca2+ Imaging of Pancreatic Islets

Measurement of the second messengers cAMP and IP1 seems feasible for receptors which couple to Gq/11, Gs, or Gi proteins and show expression in the abundant pancreatic β cells. Because the Gq/11 protein-coupled V1R and GHS-R are mainly expressed in α and δ cells, we tested the ability of AVP and ghrelin, respectively, to induce Ca2+ signals in these less abundant islet cell types. Ca2+ imaging of dispersed primary islet cells and intact pancreatic islets using the fluorescent dye fura-2 has been previously described [35, 36]. One disadvantage of Ca2+ measurement in intact islets is the slow adhering process to coverslips which lasts for several days [36]. Therefore, we coated glass coverslips with poly-L-lysine and observed reliable adherence of islets within 48 h enabling perfusion of extracellular solutions. To induce Ca2+ signals we loaded intact islets with fura-2 AM and applied the GHS-R agonist ghrelin or the V1R agonist AVP via the bath solution. For control purposes, CCh was perfused subsequently to activate Gq/11 protein-coupled M3R which are mainly expressed in β cells. Ghrelin induced a detectable Ca2+ signal in some regions of the islet whereas CCh evoked a large response in all parts of the islet (Fig. 5a, b). Application of AVP evoked robust Ca2+ elevations in several regions of the islet and CCh again induced a widespread Ca2+ signal (Fig. 5c, d). These results indicate the functionality of both receptors in pancreatic islets.

Fig. 5
figure 5

Ca2+ imaging of pancreatic islets. a Ca2+ imaging experiments were performed in single pancreatic islets loaded with fura-2 AM. Images show the fluorescence ratio (F340/F380) of one islet before and after addition of 300 nM ghrelin and 100 μM CCh. b Time course of Ca2+ responses in three regions within the islet is shown and corresponds to (a). c Images show the fluorescence ratio (F340/F380) of another islet before and after addition of 100 nM AVP and 100 μM CCh. d Time course of Ca2+ responses in three regions within the islet is shown and corresponds to (c)

Second Messenger Analysis in Islets Displaying Reduced Insulin Secretion

T2D is not only characterized by insulin insensitivity of the peripheral tissue but also by inadequate insulin secretion upon glucose treatment which finally results in an impaired glucose tolerance. Increasing insulin secretion by modulating second messenger response is one strategy to improve glucose tolerance [6]. Therefore, an evaluation of the second messenger response in dysfunctional pancreatic islets can help to identify receptor targets to increase insulin secretion. We, therefore, used a P2Y14 knock-out mouse model with reduced glucose-induced insulin release [33] to assess islet responsiveness to GPCR agonists. Islets from knock-out animals and wild-type littermates were stimulated with the previously tested agonists CCh for IP1 measurements, GLP-1 for cAMP determination, as well as CCh and AVP to assess Ca2+ signals. All tested substances induced a significant increase in second messenger concentrations. However, neither IP1, cAMP, nor Ca2+ signals differed between wild-type and knock-out mice (Fig. 6a-c), suggesting that second messenger responsiveness in knock-out islets is not impaired and that the reduced glucose-responsiveness does not depend on changed second messenger amounts.

Fig. 6
figure 6

Determining second messenger responses in islets of a P2Y14-deficient mouse model. a Activation of the Gq protein signaling pathway by the muscarinic receptor agonist CCh (100 μM) induces significant increases in IP1 concentration in wild-type (light grey) and P2Y14 knock-out (dark grey) islets. b Activation of GLP-1R using 100 nM GLP-1 induced a significant increase in cAMP concentration in wild-type and P2Y14 knock-out islets. Shown are the means ± SEM of two independent experiments each performed in duplicates. Statistical significance (w/o vs. stimulus) was tested with the two-tailed unpaired t test (*p ≤ 0.05). c Ca2+ imaging experiments were performed in the presence of CCh (100 μM) and AVP (100 nM) in single pancreatic islets from wild-type and P2Y14 knock-out mice loaded with fura-2 AM. Stimulus-evoked changes in the fluorescence ratio (F340/F380) were obtained by subtracting basal from peak value of 10 regions within each islet. Mean values of ΔRatio (F340/F380) ± SEM were calculated from mean values of regions of each wild-type and P2Y14 knockout islet (n = 3)

Discussion

In pancreatic islets, extracellular signals modulating hormone secretion are often mediated by GPCRs and subsequent changes of intracellular second messengers [37]. Because GPCRs may represent novel targets in diabetes therapy [38, 39], evaluating the impact of receptor activation is necessary. Several cell lines representing different pancreatic cell types [15] have been used to study the effect of GPCR activation.

M3R is a well-characterized GPCR with numerous agonists, inverse agonists and allosteric modulators. Pharmacological studies showed that M3R activation results in generation of inositol (1,4,5)-trisphosphate via coupling to Gq/11 proteins [40]. As MIN6 cells have been shown to functionally express M3R [41], we used this cell line to measure a CCh-induced IP1 signal. As expected, receptor activation by CCh raised intracellular IP1 by approximately 2-fold, an effect depending on Gq/11-protein activity (Fig. 2a). Applying the same protocol, we determined IP1 accumulation in dispersed islets, but no signal was detectable in adherent islet cells most probably due to the low number of cells attached in 96-well plates. Using dispersed islets directly to determine agonist-induced IP1 accumulation we observed a 2-fold increase upon CCh application similar to the response in MIN6 cells (Fig. 2a, d). In addition, CCh induced a strong Ca2+ signal in pancreatic islets (Fig. 5) as reported previously [41].

The same protocol was applied to other Gq/11 protein-coupled receptors expressed in pancreatic islets [33]. Of the tested agonists, UDP and acetate were able to induce a significant increase in IP1 accumulation in primary islet cells by about 1.5-fold and 2.5-fold, respectively. Previous results reporting UDP stimulation of P2Y6 are contradicting. While UDP reduced GSIS in β-TC6 cells [28], other groups observed an increased GSIS in β-TC6 cells and primary islets induced by UDP [29, 42]. The latter reports are in line with pharmacological characterization of P2Y6 using UDP stimulation [43] and our data showing an increased IP1 concentration, indicating Gq/11 protein activation. Acetate has been shown to induce a Gq/11 protein-mediated Ca2+ elevation in the pancreatic β cell line β-TC3 [44] and to increase IP1 concentration in MIN6 cells and primary mouse islets by about 1.5-fold [31], indicating that the here described method provides robust signals.

We could not detect IP1 signals for GHS-R or V1R in pancreatic islets stimulated with ghrelin and AVP, respectively. Recent RNAseq analysis data revealed the repertoire of GPCRs specifically expressed in the different pancreatic islet cell types [23, 24]. GHS-R and V1R are predominantly expressed in α and/or δ cells [23, 24], cell types with a markedly lower abundancy (about 10–15% and 5–10% of total cell number, respectively) in pancreatic islets [45]. It can, therefore, be assumed that the number of cells expressing these receptors limits signal detection. However, for Gq/11 and Gi/o protein-coupled receptors, imaging of intracellular Ca2+ with a resolution suitable for single cells might be one strategy to circumvent this problem. As showed for GHS-R, ghrelin-induced receptor activation is detectable by an increase of intracellular Ca2+ within regions of whole pancreatic islets (Fig. 5a). Although previous work showed effects of ghrelin on HIT-T15 and INS-1E β cells [46, 47], recent work demonstrated a cell-type restricted expression of GHS-R to pancreatic δ cells [23, 24] which is compatible with our Ca2+ imaging experiments. AVP has been shown to increase intracellular Ca2+ in In-R1-G9, an α cell-derived cell line, resulting in secretion of glucagon [48, 49]. V1R expression has also been claimed in pancreatic β cells with insulin secretion upon AVP stimulation being dependent on phospholipase C and adenylyl cyclase [49,50,51]. However, current transcriptome data does not support expression of AVP receptors in pancreatic β cells [23, 24]. Here, we show that similarly to ghrelin AVP does not induce a measurable increase in intracellular IP1 concentration but receptor activation can be monitored by Ca2+ imaging experiments within regions of the islet (Fig. 5c).

We finally applied the protocol established for islet dispersion for measurement of receptor-stimulated cAMP accumulation. Determination of cAMP in pancreatic islets has been performed previously using a transgenic mouse model containing a cAMP reporter [52]. This model allows for imaging of cAMP fluctuations. However, it requires tetracycline transactivator to achieve cell-type specific activation which is more time- and resource-consuming. The here established protocol can be used for wild-type islets as well as for islets isolated from diverse knock-out mouse models to evaluate second messenger responses without requiring cross-breeding with reporter mice. Another approach used isolated islets to determine cAMP upon glucose administration [53], however, this protocol recommends the usage of typically 25 to 50 islets per sample for optimal cAMP determination. Because one preparation typically yields 150 islets per mouse, it is insufficient to analyze different receptors. We have used forskolin, GLP-1, and SSt to evaluate changes in cAMP (Fig. 4). Forskolin, an unspecific activator of adenylyl cyclases, and the incretin GLP-1 significantly raised intracellular cAMP by 8-fold and 6.5-fold, respectively. GLP-1R activation has been previously shown to increase cAMP concentration in the β cell-derived cell lines INS-1E and MIN6 by about 3- and 2-fold, respectively [54, 55]. Somatostatin was shown to inhibit the cAMP response in INS-1 cells pretreated with 1 μM forskolin [56]. We found a clear reduction of forskolin-induced cAMP accumulation by 87%. Thus, our data demonstrate that the here presented protocol for measurement of cAMP in primary islets shows adequate sensitivity, is resource-sparing, and does not require transgenic cAMP reporter mice.

Because modulating second messenger responses is one option to overcome reduced insulin secretion, we finally evaluated this protocol on pancreatic islets with impaired function [33]. Islets from P2Y14 knock-out animals display stimulus-dependent increases in IP1 and cAMP concentrations as well as intracellular Ca2+ signals. These responses are comparable to those induced in wild-type islets suggesting that the impaired insulin secretion is not a result of an overall changed second messenger responsiveness in this mouse model. These data further indicate that our method can be used to evaluate the effect of receptor activation also in mouse models of pancreatic islet dysfunction. Since comparable islet cell number and viability is essential for the here described method, models of islet dysfunction induced by streptozotocin treatment might not be suitable for our analysis because streptozotocin drastically reduces islet cell number [57].

Furthermore, the control of cell vitality within individual preparations is necessary to obtain reliable results. We, therefore, suggest stimuli like CCh and forskolin, which induce strong and highly reproducible responses as control compounds to eliminate false-negative results.

Conclusion

With the ongoing discussion about the suitability of pancreatic cell lines for studying functions of pancreatic islets [58, 59], the usage of primary islets can help to clarify contradictory results. Here, we present optimized protocols to determine receptor-mediated second messenger levels of IP1 and cAMP in primary islets. Compared with existing protocols using transgenic reporter animals, islets obtained from wild-type animals are sufficient for the here described methods. Further, due to improved sensitivity, less islet cells are required and, therefore, several different conditions can be tested from one preparation.

Methods

Materials

If not mentioned otherwise, reagents and standard chemicals purchased from Sigma-Aldrich or Carl Roth. Cell culture media and reagents were obtained from Gibco (ThermoFisher Scientific) and poly-L-lysine (MW > 300,000) from Biochrom. Cell culture material was purchased from Sarstedt.

Cell Culture

MIN6 cells were grown in Dulbecco’s minimum essential medium supplemented with 15% fetal bovine serum (FBS), 100 U/ml penicillin, 100 μg/ml streptomycin, and 142 μM mercaptoethanol in a humidified incubator with 5% CO2/ 95% air at 37 °C.

IP1 measurements were performed using the IP-One Tb kit (Cisbio). Thus, 50,000 MIN6 cells were seeded per well of a 96-well plate. Two days after seeding, cells were washed with stimulation buffer containing 10 mM LiCl and incubated with 100 μM CCh for 60 min. Cells were lysed using the provided lysis buffer and IP1 levels were determined (see below).

Animals

C57BL6/N mice were bred under specific pathogen-free conditions on 12 h light/ 12 h dark cycle, 21 °C, and 55% humidity with ad libitum access to food and water. All experiments were conducted in accordance with European Directive 2010/63/EU on the protection of animals used for scientific purposes and were performed with permission from the Animal Care and Use Committee (ACUC# T24/16; ACUC# T19/18) and the Government of the State of Saxony, Germany. Mice were between 10 and 14 weeks of age and were matched to age and gender.

Isolation and Culture of Murine Islets

Pancreatic islets were isolated from mice sacrificed by cervical dislocation. Collagenase P was dissolved (0.5 mg/ml) in ice-cold Krebs-Ringer buffer (KRB: 115 mM NaCl, 4.7 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, 2.56 mM CaCl2, 20 mM NaHCO3, 10 mM Hepes, 0.1% BSA, 5 mM glucose, pH 7.3) and was injected via the common bile duct into pancreas. Afterwards, the distended pancreas was digested in shaking water bath at 37 °C for 12–15 min. Vital islets were washed twice in warm KRB and hand-picked under the stereomicroscope. Isolated islets were cultured in RPMI containing 10% FBS, 100 U/ml penicillin, and 100 μg/ml streptomycin overnight at 37 °C and 5% CO2/ 95% air in a humidified atmosphere.

Islet Dispersion

All reagents were warmed in a 37 °C water bath. Islets were washed in Ca2+-free KRB (115 mM NaCl, 4.7 mM KCl, 1.2 mM KH2PO4, 1.2 mM MgSO4, 20 mM NaHCO3, 10 mM Hepes, 0.1% BSA, 5 mM glucose, pH 7.3) containing 1 mM EGTA and transferred into a petri dish. Vital islets were hand-picked into a 1.5 ml reagent tube and centrifuged at 200 rpm for 1 min. The buffer was removed and islets were resuspended in 200 μl Ca2+-free KRB containing 1 mM EGTA and 0.05% Trypsin/EDTA by gently pipetting. After 60 s of incubation, 1 ml RPMI media with 10% FBS was carefully added. After centrifugation (1 min, 2000 rpm) islets were suspended in 500 μl RPMI media and transferred through a 35 μm filter (Falcon). After washing with 500 μl RPMI media, cells were transferred to the respective assay buffer. Cell number was determined by counting using a Neubauer chamber.

IP1 Accumulation Measurement

For analysis of adherent islet cells, 10,000 cells/well were seeded into poly-L-lysine-coated 96-wells plates (0.1 mg poly-L-lysine/ml) to become adherent overnight. Stimulation was performed in 35 μl of assay buffer (Cisbio) containing 10 mM LiCl and the indicated agonists. After 60 min incubation at 37 °C in a humidified incubator, cells were lysed with 30 μl lysis buffer provided by the manufacturer. Lysats were kept frozen until IP1 measurement using the IP-One Tb kit (Cisbio). Thus, 14 μl of lysed stimulated cells were transferred to ProxiPlate-384 Plus microplates (PerkinElmer Life Sciences) and incubated with 3 μl acceptor-bead solution and 3 μl donor-bead solution (bead stock solutions were diluted 1:20 in lysis buffer). After 60 min incubation at room temperature in the dark, fluorescence was determined using the EnVision Multilabel Reader (PerkinElmer Life Sciences) by excitation at 330 nm and emission at 620 nm.

To analyze IP1 accumulation in suspension cells, 5000 islet cells/well were incubated in ProxiPlate-384 Plus microplates (PerkinElmer Life Sciences) with the indicated substances to stimulate GPCR in 14 μl assay buffer (Cisbio) containing 10 mM LiCl. After the indicated time points, 3 μl acceptor-bead solution and 3 μl donor-bead solution (1:20 dilution in lysis buffer) were added. After 60 min incubation at room temperature in the dark, fluorescence was determined using the EnVision Multilabel Reader (PerkinElmer Life Sciences) by excitation at 330 nm and emission at 620 nm.

cAMP Accumulation Measurement

The cAMP accumulation was determined in 384-well white OptiPlate microplates (PerkinElmer Life Sciences) using the AlphaScreen cAMP assay kit (PerkinElmer Life Sciences). Thereto, dispersed islets (5000 cells/well) were resuspended in cAMP stimulation buffer (HBSS containing 0.5 mM IBMX, 5 mM Hepes, 0.1% BSA, pH 7.4) with the indicated substances in a final volume of 10 μl containing 0.1 μl of cAMP acceptor beads. After 30 min 15 μl of donor-bead solution containing 0.1 μl streptavidin donor beads and 0.3 μl biotinylated cAMP in lysis buffer (5 mM Hepes, 0.1% BSA, 0.3% Tween-20, pH 7.4) was added and incubated for further 60 min at room temperature. Values were determined by excitation at 640 nm and emission at 570 nm using the EnVision Multilabel Reader (PerkinElmer Life Sciences).

Ca2+ Imaging

Pancreatic mouse islets were transferred into 12-well plates 1 day after preparation on poly-L-lysine coated glass coverslips (12 mm diameter, 3–4 islets per coverslip). Two days after seeding, coverslips were transferred into a new 12-well plate, where islets were loaded with 5 μM fura-2 AM dissolved in standard bath solution for 60 min at 20–22 °C. The standard bath solution contained 140 mM NaCl, 10 mM Hepes, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, and 3 mM glucose (pH 7.4). After loading, the coverslip was transferred to a perfusion chamber (Warner Instruments) mounted on an upright microscope (Olympus BX51WI). Islets were visualized using a 20x water immersion objective (UMPlanFl, Olympus) and perfused with standard bath solution and stimulated by adding 300 nM ghrelin, 100 nM AVP, or 100 μM CCh to standard bath solution. Fura-2-based Ca2+ imaging was performed in intact pancreatic islets using a monochromator-based imaging system and the imaging software TILLvisION 4.0 (both T.I.L.L. Photonics). Emitted fluorescence at 510 nm (excited at 340 nm and 380 nm) was collected with a CCD camera (PCO Imaging), acquired at intervals of 2 s and corrected for background fluorescence.

Statistical Analysis

If not stated otherwise, data is presented as means ± SEM. Statistical analysis was performed using GraphPad Prism version 6.0. As stated in the figure legends, data was analyzed using a two-tailed paired student’s t test. P-values with p ≤ 0.05 were considered statistically significant.

Availability of Data and Materials

Data used and analyzed in this study are included in this article. Transcriptome data were performed by [33] and GPCR expression analyzed from the given FPKM value.

Abbreviations

AVP:

arginine vasopressin

cAMP:

3′,5′-cyclic adenosine monophosphate

CCh:

carbachol

FFAR2:

Free fatty acid receptor type 2

GHS-R:

growth hormone secretagogue receptor/ghrelin receptor

GLP-1R:

glucagon-like peptide-1 receptor

GPCR:

G protein-coupled receptor

GSIS:

glucose-induced insulin secretion

M3R:

muscarinic acetylcholine receptor type 3

Sst-R:

somatostatin receptor

T2D:

type 2 diabetes

UDP:

uridine 5′-diphosphate

References

  1. International Diabetes Federation. IDF Diabetes Atlas, 8th edn. Brussels: International Diabetes Federation; 2017.

  2. DeFronzo RA, Ferrannini E, Groop L, Henry RR, Herman WH, Holst JJ, et al. Type 2 diabetes mellitus. Nat Rev Dis Primers. 2015;1:15019.

    Article  PubMed  Google Scholar 

  3. Malaisse WJ, Malaisse-Lagae F, Mayhew D. A possible role for the adenylcyclase system in insulin secretion. J Clin Invest. 1967;46:1724–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wollheim CB, Sharp GW. Regulation of insulin release by calcium. Physiol Rev. 1981;61:914–73.

    Article  CAS  PubMed  Google Scholar 

  5. Sriram K, Insel PA. G protein-coupled receptors as targets for approved drugs: how many targets and how many drugs? Mol Pharmacol. 2018;93:251–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Frias JP, Nauck MA, Van J, Kutner ME, Cui X, Benson C, et al. Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet. 2018;392:2180–93.

    Article  CAS  PubMed  Google Scholar 

  7. Ritter K, Buning C, Halland N, Pöverlein C, Schwink L. G protein-coupled receptor 119 (GPR119) agonists for the treatment of diabetes: recent Progress and prevailing challenges. J Med Chem. 2016;59:3579–92.

    Article  CAS  PubMed  Google Scholar 

  8. Trujillo JM, Nuffer W. GLP-1 receptor agonists for type 2 diabetes mellitus: recent developments and emerging agents. Pharmacotherapy. 2014;34:1174–86.

    Article  CAS  PubMed  Google Scholar 

  9. Watterson KR, Hudson BD, Ulven T, Milligan G. Treatment of type 2 diabetes by free fatty acid receptor agonists. Front Endocrinol (Lausanne). 2014;5:137.

    Article  Google Scholar 

  10. 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. Pharmacol Ther. 2013;139:359–91.

    Article  CAS  PubMed  Google Scholar 

  11. Regard JB, Sato IT, Coughlin SR. Anatomical profiling of G protein-coupled receptor expression. Cell. 2008;135:561–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Al-Amily IM, Dunér P, Groop L, Salehi A. The functional impact of G protein-coupled receptor 142 (Gpr142) on pancreatic β-cell in rodent. Pflugers Arch. 2019;471:633–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kobilka BK. G protein coupled receptor structure and activation. Biochim Biophys Acta. 2007;1768:794–807.

    Article  CAS  PubMed  Google Scholar 

  14. Ishihara H, Asano T, Tsukuda K, Katagiri H, Inukai K, Anai M, et al. Pancreatic beta cell line MIN6 exhibits characteristics of glucose metabolism and glucose-stimulated insulin secretion similar to those of normal islets. Diabetologia. 1993;36:1139–45.

    Article  CAS  PubMed  Google Scholar 

  15. Skelin M, Rupnik M, Cencic A. Pancreatic beta cell lines and their applications in diabetes mellitus research. Altex. 2010;27:105–13.

    Article  PubMed  Google Scholar 

  16. Santerre RF, Cook RA, Crisel RM, Sharp JD, Schmidt RJ, Williams DC, Wilson CP. Insulin synthesis in a clonal cell line of simian virus 40-transformed hamster pancreatic beta cells. Proc Natl Acad Sci U S A. 1981;78:4339–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Efrat S. Regulation of insulin secretion: insights from engineered beta-cell lines. Ann N Y Acad Sci. 2004;1014:88–96.

    Article  CAS  PubMed  Google Scholar 

  18. Orecná M, Hafko R, Bacová Z, Podskocová J, Chorvát D, Strbák V. Different secretory response of pancreatic islets and insulin secreting cell lines INS-1 and INS-1E to osmotic stimuli. Physiol Res. 2008;57:935–45.

    PubMed  Google Scholar 

  19. Schulze T, Morsi M, Brüning D, Schumacher K, Rustenbeck I. Different responses of mouse islets and MIN6 pseudo-islets to metabolic stimulation: a note of caution. Endocrine. 2016;51:440–7.

    Article  CAS  PubMed  Google Scholar 

  20. Iismaa TP, Kerr EA, Wilson JR, Carpenter L, Sims N, Biden TJ. Quantitative and functional characterization of muscarinic receptor subtypes in insulin-secreting cell lines and rat pancreatic islets. Diabetes. 2000;49:392–8.

    Article  CAS  PubMed  Google Scholar 

  21. Colombo M, Gregersen S, Xiao J, Hermansen K. Effects of ghrelin and other neuropeptides (CART, MCH, orexin a and B, and GLP-1) on the release of insulin from isolated rat islets. Pancreas. 2003;27:161–6.

    Article  CAS  PubMed  Google Scholar 

  22. Wierup N, Yang S, McEvilly RJ, Mulder H, Sundler F. Ghrelin is expressed in a novel endocrine cell type in developing rat islets and inhibits insulin secretion from INS-1 (832/13) cells. J Histochem Cytochem. 2004;52:301–10.

    Article  CAS  PubMed  Google Scholar 

  23. Adriaenssens AE, Svendsen B, Lam BYH, Yeo GSH, Holst JJ, Reimann F, Gribble FM. Transcriptomic profiling of pancreatic alpha, beta and delta cell populations identifies delta cells as a principal target for ghrelin in mouse islets. Diabetologia. 2016;59:2156–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. DiGruccio MR, Mawla AM, Donaldson CJ, Noguchi GM, Vaughan J, Cowing-Zitron C, et al. Comprehensive alpha, beta and delta cell transcriptomes reveal that ghrelin selectively activates delta cells and promotes somatostatin release from pancreatic islets. Mol Metab. 2016;5:449–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Holst B, Cygankiewicz A, Jensen TH, Ankersen M, Schwartz TW. High constitutive signaling of the ghrelin receptor--identification of a potent inverse agonist. Mol Endocrinol. 2003;17:2201–10.

    Article  CAS  PubMed  Google Scholar 

  26. Salehi A, La Dornonville de Cour C, Håkanson R, Lundquist I. Effects of ghrelin on insulin and glucagon secretion: a study of isolated pancreatic islets and intact mice. Regul Pept. 2004;118:143–50.

    Article  CAS  PubMed  Google Scholar 

  27. Reimer MK, Pacini G, Ahrén B. Dose-dependent inhibition by ghrelin of insulin secretion in the mouse. Endocrinology. 2003;144:916–21.

    Article  CAS  PubMed  Google Scholar 

  28. Ohtani M, Suzuki J-I, Jacobson KA, Oka T. Evidence for the possible involvement of the P2Y(6) receptor in Ca (2+) mobilization and insulin secretion in mouse pancreatic islets. Purinergic Signal. 2008;4:365–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Parandeh F, Abaraviciene SM, Amisten S, Erlinge D, Salehi A. Uridine diphosphate (UDP) stimulates insulin secretion by activation of P2Y6 receptors. Biochem Biophys Res Commun. 2008;370:499–503.

    Article  CAS  PubMed  Google Scholar 

  30. Briscoe CP, Peat AJ, McKeown SC, Corbett DF, Goetz AS, Littleton TR, et al. Pharmacological regulation of insulin secretion in MIN6 cells through the fatty acid receptor GPR40: identification of agonist and antagonist small molecules. Br J Pharmacol. 2006;148:619–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. McNelis JC, Lee YS, Mayoral R, van der Kant R, Johnson AMF, Wollam J, Olefsky JM. GPR43 potentiates β-cell function in obesity. Diabetes. 2015;64:3203–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tengholm A. Cyclic AMP dynamics in the pancreatic β-cell. Ups J Med Sci. 2012;117:355–69.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Meister J, Le Duc D, Ricken A, Burkhardt R, Thiery J, Pfannkuche H, et al. The G protein-coupled receptor P2Y14 influences insulin release and smooth muscle function in mice. J Biol Chem. 2014;289:23353–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Schrage R, Schmitz A-L, Gaffal E, Annala S, Kehraus S, Wenzel D, et al. The experimental power of FR900359 to study Gq-regulated biological processes. Nat Commun. 2015;6:10156.

    Article  CAS  PubMed  Google Scholar 

  35. Sussman KE, Leitner JW, Draznin B. Cytosolic free-calcium concentrations in normal pancreatic islet cells. Effect of secretagogues and somatostatin. Diabetes. 1987;36:571–7.

    Article  CAS  PubMed  Google Scholar 

  36. Roe MW, Lancaster ME, Mertz RJ, Worley JF, Dukes ID. Voltage-dependent intracellular calcium release from mouse islets stimulated by glucose. J Biol Chem. 1993;268:9953–6.

    CAS  PubMed  Google Scholar 

  37. Howell SL, Jones PM, Persaud SJ. Regulation of insulin secretion: the role of second messengers. Diabetologia. 1994;37(Suppl 2):S30–5.

    Article  CAS  PubMed  Google Scholar 

  38. Ahrén B. Islet G protein-coupled receptors as potential targets for treatment of type 2 diabetes. Nat Rev Drug Discov. 2009;8:369–85.

    Article  PubMed  CAS  Google Scholar 

  39. Reimann F, Gribble FM. G protein-coupled receptors as new therapeutic targets for type 2 diabetes. Diabetologia. 2016;59:229–33.

    Article  CAS  PubMed  Google Scholar 

  40. Wess J, Blin N, Mutschler E, Blüml K. Muscarinic acetylcholine receptors: structural basis of ligand binding and G protein coupling. Life Sci. 1995;56:915–22.

    Article  CAS  PubMed  Google Scholar 

  41. Tamarina NA, Kuznetsov A, Rhodes CJ, Bindokas VP, Philipson LH. Inositol (1,4,5)-trisphosphate dynamics and intracellular calcium oscillations in pancreatic beta-cells. Diabetes. 2005;54:3073–81.

    Article  CAS  PubMed  Google Scholar 

  42. Sassmann A, Gier B, Gröne H-J, Drews G, Offermanns S, Wettschureck N. The Gq/G11-mediated signaling pathway is critical for autocrine potentiation of insulin secretion in mice. J Clin Invest. 2010;120:2184–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Communi D, Parmentier M, Boeynaems JM. Cloning, functional expression and tissue distribution of the human P2Y6 receptor. Biochem Biophys Res Commun. 1996;222:303–8.

    Article  CAS  PubMed  Google Scholar 

  44. Priyadarshini M, Villa SR, Fuller M, Wicksteed B, Mackay CR, Alquier T, et al. An acetate-specific GPCR, FFAR2, regulates insulin secretion. Mol Endocrinol. 2015;29:1055–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Orci L, Unger RH. Functional subdivision of islets of Langerhans and possible role of D cells. Lancet. 1975;2:1243–4.

    Article  CAS  PubMed  Google Scholar 

  46. Granata R, Settanni F, Biancone L, Trovato L, Nano R, Bertuzzi F, et al. Acylated and unacylated ghrelin promote proliferation and inhibit apoptosis of pancreatic beta-cells and human islets: involvement of 3′,5′-cyclic adenosine monophosphate/protein kinase a, extracellular signal-regulated kinase 1/2, and phosphatidyl inositol 3-kinase/Akt signaling. Endocrinology. 2007;148:512–29.

    Article  CAS  PubMed  Google Scholar 

  47. Gauna C, Delhanty PJD, van Aken MO, Janssen JAMJL, Themmen APN, Hofland LJ, et al. Unacylated ghrelin is active on the INS-1E rat insulinoma cell line independently of the growth hormone secretagogue receptor type 1a and the corticotropin releasing factor 2 receptor. Mol Cell Endocrinol. 2006;251:103–11.

    Article  CAS  PubMed  Google Scholar 

  48. Yibchok-Anun S, Cheng H, Chen TH, Hsu WH. Mechanisms of AVP-induced glucagon release in clonal alpha-cells in-R1-G9: involvement of Ca(2+)-dependent and -independent pathways. Br J Pharmacol. 2000;129:257–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Folny V, Raufaste D, Lukovic L, Pouzet B, Rochard P, Pascal M, Serradeil-Le GC. Pancreatic vasopressin V1b receptors: characterization in in-R1-G9 cells and localization in human pancreas. Am J Physiol Endocrinol Metab. 2003;285:E566–76.

    Article  CAS  PubMed  Google Scholar 

  50. Oshikawa S, Tanoue A, Koshimizu T-A, Kitagawa Y, Tsujimoto G. Vasopressin stimulates insulin release from islet cells through V1b receptors: a combined pharmacological/knockout approach. Mol Pharmacol. 2004;65:623–9.

    Article  CAS  PubMed  Google Scholar 

  51. Mohan S, Moffett RC, Thomas KG, Irwin N, Flatt PR. Vasopressin receptors in islets enhance glucose tolerance, pancreatic beta-cell secretory function, proliferation and survival. Biochimie. 2019;158:191–8.

    Article  CAS  PubMed  Google Scholar 

  52. Kim JW, Roberts CD, Berg SA, Caicedo A, Roper SD, Chaudhari N. Imaging cyclic AMP changes in pancreatic islets of transgenic reporter mice. PLoS One. 2008;3:e2127.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Neuman JC, Truchan NA, Joseph JW, Kimple ME. A method for mouse pancreatic islet isolation and intracellular cAMP determination. J Vis Exp. 2014;(88):e50374.

  54. Ramos LS, Zippin JH, Kamenetsky M, Buck J, Levin LR. Glucose and GLP-1 stimulate cAMP production via distinct adenylyl cyclases in INS-1E insulinoma cells. J Gen Physiol. 2008;132:329–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Rajan S, Dickson LM, Mathew E, Orr CMO, Ellenbroek JH, Philipson LH, Wicksteed B. Chronic hyperglycemia downregulates GLP-1 receptor signaling in pancreatic β-cells via protein kinase A. Mol Metab. 2015;4:265–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Burns SM, Vetere A, Walpita D, Dančík V, Khodier C, Perez J, et al. High-throughput luminescent reporter of insulin secretion for discovering regulators of pancreatic Beta-cell function. Cell Metab. 2015;21:126–37.

    Article  CAS  PubMed  Google Scholar 

  57. Li Z, Karlsson FA, Sandler S. Islet loss and alpha cell expansion in type 1 diabetes induced by multiple low-dose streptozotocin administration in mice. J Endocrinol. 2000;165:93–9.

    Article  CAS  PubMed  Google Scholar 

  58. Nakashima K, Kanda Y, Hirokawa Y, Kawasaki F, Matsuki M, Kaku K. MIN6 is not a pure beta cell line but a mixed cell line with other pancreatic endocrine hormones. Endocr J. 2009;56:45–53.

    Article  CAS  PubMed  Google Scholar 

  59. Merglen A, Theander S, Rubi B, Chaffard G, Wollheim CB, Maechler P. Glucose sensitivity and metabolism-secretion coupling studied during two-year continuous culture in INS-1E insulinoma cells. Endocrinology. 2004;145:667–78.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Jessica Michelle Jannicke for outstanding animal care and Angela Schulz for providing P2Y14 knock-out mice and wild-type littermates.

Funding

This work was supported by the German Research Foundation CRC 1052/B6 (project number 209933838) and the European Social Fund.

Author information

Authors and Affiliations

Authors

Contributions

JR, TS, and DT designed the experiments. JR, RK, and DT performed the experiments, analyzed, and interpreted the data. All authors contributed to the manuscript, read and approved the final manuscript.

Corresponding author

Correspondence to Doreen Thor.

Ethics declarations

Ethics Approval

All experiments were conducted in accordance with European Directive 2010/63/EU on the protection of animals used for scientific purposes and were performed with permission from the Animal Care and Use Committee (ACUC# T24/16; ACUC# T19/18) and the Government of the State of Saxony, Germany.

Consent for Publication

Not applicable.

Competing Interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Röthe, J., Kraft, R., Schöneberg, T. et al. Exploring G Protein-Coupled Receptor Signaling in Primary Pancreatic Islets. Biol Proced Online 22, 4 (2020). https://doi.org/10.1186/s12575-019-0116-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12575-019-0116-y

Keywords