Long-chain fatty acids (LCFAs) are not only energy sources but also serve as signaling molecules. GPR120, an LCFA receptor, plays key roles in maintaining metabolic homeostasis. However, whether endogenous ligand-GPR120 circuits exist and how such circuits function in pancreatic islets are unclear. Here, we found that endogenous GPR120 activity in pancreatic δ-cells modulated islet functions. At least two unsaturated LCFAs, oleic acid (OA) and linoleic acid (LA), were identified as GPR120 agonists within pancreatic islets. These two LCFAs promoted insulin secretion by inhibiting somatostatin secretion and showed bias activation of GPR120 in a model system. Compared with OA, LA exerted higher potency in promoting insulin secretion, which is dependent on β-arrestin2 function. Moreover, GPR120 signaling was impaired in the diabetic db/db model, and replenishing OA and LA improved islet function in both the db/db and streptozotocin-treated diabetic models. Consistently, the administration of LA improved glucose metabolism in db/db mice. Collectively, our results reveal that endogenous LCFA-GPR120 circuits exist and modulate homeostasis in pancreatic islets. The contributions of phenotype differences caused by different LCFA-GPR120 circuits within islets highlight the roles of fine-tuned ligand–receptor signaling networks in maintaining islet homeostasis.

G protein-coupled receptors (GPCRs) sense diverse extracellular stimuli and play key roles in cell–cell communication. Several ligand-GPCR circuits have been discovered in pancreatic islets. For instance, the GLP-1–GLP-1R circuit in Langerhans islets regulates both insulin secretion and islet regeneration (13). Acetylcholine produced by human α-cells modulates insulin secretion via muscarinic acetylcholine receptors (4,5). Moreover, epigenetic regulation of somatostatin (SST) secretion by endogenous urocortin3 (UCN3) via CRHR2 serve as key mechanisms for islet homeostasis (6,7). However, the signaling circuits involved with fatty acid (FA) GPCRs in pancreatic islets are not fully understood.

GPR120 is one of the top 20 genes determined to be related to type 2 diabetes by systems genetic analysis (8). GPR120 deficiency in mice leads to obesity (9). In pancreatic islets, GPR120 is expressed mainly in α- and δ-cells, and activation of GPR120 in δ-cells is reported to inhibit SST release (1014). Although GPR120 is the receptor for exogenous n-3 FAs and several endogenous lipids, such as palmitic acid-hydroxy stearic acid (PAHSA) (1519), the characteristics of the ligand-GPR120 circuits in islets are unclear. Moreover, at least Gq, Gi, and β-arrestins are known to couple with GPR120 in a ligand-dependent manner, thus generating signaling diversity (10,15,2023). Notably, attenuation of harmful effects by amplification of the particular downstream signals of a single GPCR may have new therapeutic potential for diseases (2428), such as oliceridine, a recently U.S. Food and Drug Administration-approved pain killer. In this regard, it is important to determine whether endogenous GPR120 ligands are produced in pancreatic islets and, if so, to elucidate their identities and signaling biases. In the current study, we set out to delineate potential LCFA-GPR120 circuit within islets.

Mice

Sst-Cre mice (stock no. 013044), Ins2-Cre mice (stock no. 003573), db/+ mice (stock no. 000697), and green fluorescent protein (GFP)-reporter mice (stock no. 007676) with a C57BL/6 background were obtained from The Jackson Laboratory and were used in our previous studies (7,25,29). β-arrestin1−/− and β-arrestin2−/− mice on a C57BL/6 background were obtained from Dr R.J. Lefkowitz (Duke University, Durham, NC) and G. Pei (Tongji University, Shanghai, China) and used in our previous study (7,25,29). Gcg-Cre mice (stock no. C001052) with a C57BL/6 background were purchased from Cyagen (Suzhou, China). All mouse work was performed with the approval of the Institutional Animal Care and Use Committee of Peking University Health Science Center and Shandong University Cheeloo College of Medicine.

Human Samples

Human pancreatic islet samples were provided by the Department of General Surgery, Qilu Hospital of Shandong University, with informed written consent from patients and approval by the Clinical Research Ethics Committee of Shandong University (ECSBMSSDU2018-1-023). Human pancreatic islets came from 6 male donors undergoing pancreaticoduodenectomy, aged 45 ± 3, and their BMI was 25.19 ± 0.56 kg/m2.

Constructs

Mouse GPR120, β-arrestin1, β-arrestin2, and G protein subunits (Gαi, Gαq, Gβ1, and Gγ2) genes were subcloned into the pcDNA3.1 expression vectors. GPR120-YFP, GFP10-Gγ2 Gαq-RlucII, and Gαi-RlucII were created by fusion or insertion of different sequences to constructs. The human GPR120 mutation R270H was generated using the QuikChange Mutagenesis Kit (Stratagene). All of the constructs and mutations were verified by DNA sequencing.

RNA Extraction and Quantitative RT-PCR

Total RNA was extracted from isolated mouse islets or single cells derived from the islets using TRIzol reagent (Invitrogen). cDNA synthesis was performed using a commercial quantitative (q)RT-PCR kit (Toyobo, FSQ-101). qRT-PCR was performed on a LightCycler qPCR apparatus (Bio-Rad) using FastStart SYBR Green Master Mix (Roche). All primer sequences used for qRT-PCR assays are listed in Supplementary Table 1.

In Vitro Mouse and Human Islet Culture

Mouse pancreatic islets were isolated from adult mice and cultured as previously described (30,31). Human islets were isolated from human pancreas obtained from the Department of General Surgery, Qilu Hospital of Shandong University, through digestion by collagenase P at 37°C for 30–50 min. After stop digestion and sedimentation, the islets were hand-picked for collection using a stereoscopic microscope and were cultured overnight in DMEM containing 5.56 mmol/L glucose (Biological Industries), 10% FBS (Gibco), and 0.1% penicillin/streptomycin (Biological Industries).

Insulin, SST, and Glucagon Measurement

The hormone secretion measurement, including that of insulin and SST, was performed as previously described by our group (7,29,31,32). Stock free FAs were dissolved in DMSO and then emulsified in modified Krebs-Ringer bicarbonate buffer with an Autotune series high-intensity ultrasonic processor until homogenous (19,33).

Glucose Tolerance Test

Mice were fasted for 16 h before the glucose tolerance test. The plasma glucose levels were measured in tail blood by using a FreeStyle Lite Glucose Meter (Roche). After measurement of the baseline glucose level (0 min), the mice were intraperitoneally injected with 2 mg/g body wt glucose, and the glucose levels at 30, 60, 90, 120 min after injection were detected.

Bioluminescence Resonance Energy Transfer Measurement

The β-arrestin recruitment and Gβγ-dissociation Bioluminescence Resonance Energy Transfer (BRET) assay were performed as previously described (25,34). The BRET signal was examined by using a Mithras LB 940 Multimode Microplate Reader (Berthold Technologies). The ratio of luminescence at 530 nm–to–485 nm and 510 nm–to–400 nm represented the β-arrestin recruitment and Gβγ-dissociation BRET signal intensity, respectively.

GloSensor cAMP Assay

The GloSensor cAMP assay was performed as previously described (30,35). The cAMP signal was examined using a Multimode Plate Reader (EnVision, PerkinElmer).

Bias Calculation

We used the operation model to calculate the signal bias of the ligand, choosing one signal pathway as a reference, generally compared with one endogenous ligand of the GPCR (36). The β-value as a factor to quantify the bias of the ligand is calculated as follows:
β = log where P1 denotes the reference pathway and P2 denotes the other pathway; β > 0 indicates ligand bias to the reference pathway, and β < 0 indicates ligand bias to the other pathway.

Immunofluorescence Staining and Coimmunoprecipitation

The islets or cells were fixed and subjected to fluorescence microscopic analysis using an LSM 780 laser scanning confocal microscope system (ZEISS, Oberkochen, Germany), according to our previous publications (7,31). The recruitment of β-arrestin1 and β-arrestin2 to pancreatic islets by GPR120 was examined by coimmunoprecipitation experiments using a method similar to that in our previous publications (24,25,37). Antibodies used are listed in Supplementary Table 2.

Metabolomics Analysis

Islets isolated from db/+ and db/db mice or the supernatant (100 μL) of islets isolated from wild-type mice after treatment were obtained for metabolomics analysis as previously described (38). The ΔOA or ΔLA concentration values were calculated by measuring the OA or LA concentration in the islet supernatant under 11.1 mmol/L glucose conditions and then subtracting the concentration in the islet supernatant under 2.8 mmol/L glucose conditions.

Statistics

All results in this study are presented as the mean ± SEM. An unpaired two-tailed Student t test was used to compare two groups. One-way ANOVA, followed by the Dunnett test, was used for comparisons between multiple groups. Statistical analysis and graph generation were performed using GraphPad Prism 8 software. P values <0.05 were considered statistically significant.

Data and Resource Availability

Data sets generated in this study are available from the corresponding author upon reasonable request.

Endogenous GPR120 Activity Regulates Paracrine Interactions In Islets

Preincubation of mouse pancreatic islets with 30 μmol/L AH7614, a selective GPR120 antagonist, significantly promoted SST secretion in response to 11.1 mmol/L glucose stimulation (Fig. 1B–D and Supplementary Fig. 1F). Simultaneously, glucose-stimulated insulin secretion was significantly decreased after AH7614 incubation (Fig. 1D). In contrast, glucose-stimulated glucagon secretion was not significantly changed (Fig. 1E). The effects of AH7614 on mouse islet hormone secretion were recapitulated by adenovirus-mediated knockdown of GPR120 expression in islets, with no effect on the expression of GRP40 and other relevant FA receptors (Fig. 1F–H and Supplementary Fig. 1H and I). Previous findings and our results both indicated that GPR120 is mostly enriched in mouse pancreatic δ-cells (1013) (Fig. 1A and Supplementary Fig. 1AE). Using mouse pancreatic islets derived from Sst-cre+/− mice and infected with GPR120-shRNAf/f lentivirus in vitro, we were able to specifically knock down GPR120 expression in mouse pancreatic δ-cells and examine the contribution of GPR120 residing in δ-cells to islet hormone secretion (Supplementary Fig. 2AD). Importantly, compared with islets from Sst-cre+/− littermates infected with the shNCf/f (negative control shRNA) virus, Sst-cre+/− GPR120-shRNAf/f islets exhibited higher SST secretion and lower insulin secretion in response to high-glucose stimulation (Fig. 1I and J). However, glucose-induced glucagon secretion did not significantly differ (Fig. 1K).

Figure 1

Endogenous GPR120 activity regulates paracrine interactions in islets isolated from wild-type mice and Sst-cre+/− mice. A: Representative images of colorimetric RNAscope in situ hybridization for GPR120 and immunostaining of SST+ pancreatic δ-cells in pancreatic sections of wild-type mice. Scale bar: 50 μm. (n = 6 mice per group; three to five random areas were selected from each islet section, and six to eight sections were randomly selected from each mouse.) B: Schematic of the experiment conducted to test whether endogenous GPR120 activity participates in hormone secretion in islets. AH7614, a selective GPR120 antagonist, was used at 30 μmol/L to inhibit endogenous GPR120 activity. Glucose-induced SST secretion (C), glucose-induced insulin secretion (D), and glucose regulation of glucagon secretion (E) in islets pretreated with or without 30 μmol/L AH7614. Glucose-induced SST secretion (F), glucose-induced insulin secretion (G), and glucose regulation of glucagon secretion (H) in islets pretreated with adenoviruses expressing negative control shRNA or GPR120 shRNA. Glucose-induced SST secretion (I), glucose-induced insulin secretion (J), and glucose regulation of glucagon secretion (K) in islets isolated from Sst-cre+/− mice pretreated with lentiviruses expressing a control sequence or GPR120 shRNAf/f. In CK, a total of 200 islets from two to four mice of the indicated genotypes were grouped (50 islets/group) for the indicated hormone secretion measurement in one experiment. The data are from six independent experiments (n = 6). In C–E, ***P < 0.001 and ns for islets treated with AH7614 compared with those treated with vehicle. In FH, ***P < 0.001 and ns for islets treated with adenoviruses expressing GPR120 shRNA compared with those treated with adenoviruses expressing a control sequence. In IK, ***P < 0.001 and ns for islets derived from Sst-cre+/− mice treated with lentiviruses expressing GPR120 shRNAf/f compared with those treated with lentiviruses expressing a control sequence. The bars represent mean ± SEM. All data statistics were analyzed using two-way ANOVA with the Dunnett post hoc test.

Figure 1

Endogenous GPR120 activity regulates paracrine interactions in islets isolated from wild-type mice and Sst-cre+/− mice. A: Representative images of colorimetric RNAscope in situ hybridization for GPR120 and immunostaining of SST+ pancreatic δ-cells in pancreatic sections of wild-type mice. Scale bar: 50 μm. (n = 6 mice per group; three to five random areas were selected from each islet section, and six to eight sections were randomly selected from each mouse.) B: Schematic of the experiment conducted to test whether endogenous GPR120 activity participates in hormone secretion in islets. AH7614, a selective GPR120 antagonist, was used at 30 μmol/L to inhibit endogenous GPR120 activity. Glucose-induced SST secretion (C), glucose-induced insulin secretion (D), and glucose regulation of glucagon secretion (E) in islets pretreated with or without 30 μmol/L AH7614. Glucose-induced SST secretion (F), glucose-induced insulin secretion (G), and glucose regulation of glucagon secretion (H) in islets pretreated with adenoviruses expressing negative control shRNA or GPR120 shRNA. Glucose-induced SST secretion (I), glucose-induced insulin secretion (J), and glucose regulation of glucagon secretion (K) in islets isolated from Sst-cre+/− mice pretreated with lentiviruses expressing a control sequence or GPR120 shRNAf/f. In CK, a total of 200 islets from two to four mice of the indicated genotypes were grouped (50 islets/group) for the indicated hormone secretion measurement in one experiment. The data are from six independent experiments (n = 6). In C–E, ***P < 0.001 and ns for islets treated with AH7614 compared with those treated with vehicle. In FH, ***P < 0.001 and ns for islets treated with adenoviruses expressing GPR120 shRNA compared with those treated with adenoviruses expressing a control sequence. In IK, ***P < 0.001 and ns for islets derived from Sst-cre+/− mice treated with lentiviruses expressing GPR120 shRNAf/f compared with those treated with lentiviruses expressing a control sequence. The bars represent mean ± SEM. All data statistics were analyzed using two-way ANOVA with the Dunnett post hoc test.

Close modal

We also characterized endogenous GPR120 activity in human pancreatic islets by application of AH7614, which recapitulated the phenotype identified in mouse islets by significantly enhancing SST secretion in response to 11.1 mmol/L glucose stimulation (Supplementary Fig. 1G). These data suggest that the activity of endogenous GPR120, especially that residing in pancreatic δ-cells, regulates hormone release and paracrine interactions in islets.

Potential Endogenous Ligands of GPR120 in Pancreatic Islets

We collected the mouse islet supernatant under 2.8 mmol/L glucose and 11.1 mmol/L glucose conditions and then examined the effects of these supernatants on GPR120 activation using in vitro recruitment assays for arrestin, a pivotal transducer of GPCR signaling. In HEK293 cells overexpressing GPR120-GFP and Rluc–β-arrestin2, the 11.1 mmol/L glucose-treated islet supernatant promoted more β-arrestin2 recruitment to GPR120 than the low glucose–treated islet supernatant (Fig. 2A).

Figure 2

Identification of endogenous ligands of GPR120 in pancreatic islets isolated from wild-type (WT) mice. A: Recruitment of β-arrestin2 to GPR120 in GPR120-overexpressing HEK293 cells in response to incubation with supernatants of islets in response to treatment with 2.8 mmol/L or 11.1 mmol/L glucose for 1 h. A total of 400 islets from six to eight WT mice were grouped (200 islets/group) in one experiment. The data are from six independent experiments (n = 6). B: FA composition in the supernatant of islets upon treatment with 2.8 mmol/L or 11.1 mmol/L glucose for 1 h. A total of 400 islets from six to eight WT mice were grouped (200 islets/group) for metabolomics analysis in one experiment. The data are from six independent experiments (n = 6). We experimentally determined that 1,000 islets approximately have 10 μL volume and estimated that the average islet has a volume of 10 nL. Using these estimations, we predicted that the extracellular OA and LA concentrations in each islet are 209.99 ± 13.92 μmol/L and 182.01 ± 7.87 μmol/L, respectively, at low glucose, and are 322.86 ± 10.87 μmol/L (OA) and 440.8 ± 11.47 μmol/L (LA) at high glucose. C: Recruitment of β-arrestin2 to GPR120 in GPR120-overexpressing HEK293 cells in response to incubation with ΔOA (2.3 μmol/L), ΔLA (5.2 μmol/L), or control vehicle. The data are from three independent experiments (n = 3). D: Recruitment of β-arrestin2 to GPR120 in GPR120-overexpressing HEK293 cells in response to incubation with supernatants of islets treated with 2.8 mmol/L glucose or 11.1 mmol/L glucose pretreated with vehicle or 1 μmol/L MK8245 for 8 h. E: Recruitment of β-arrestin2 to GPR120 in GPR120-overexpressing HEK293 cells in response to incubation with supernatants of islets treated with 2.8 mmol/L glucose or 11.1 mmol/L glucose pretreated with or without 20 μmol/L SC26196 for 8 h. F: FA composition in the supernatant of islets upon treatment with 2.8 mmol/L or 11.1 mmol/L glucose pretreated with vehicle or 1 μmol/L MK8245 for 8 h. G: FA composition in the supernatant of islets upon treatment with 2.8 mmol/L or 11.1 mmol/L glucose pretreated with vehicle or 20 μmol/L SC26196 for 8 h. In D–G, a total of 800 islets from 12 to 16 mice were grouped (200 islets/group) for the indicated measurement in one experiment. The data are from six independent experiments (n = 6). H: GPR120 internalization in HEK293 cells treated with 200 μmol/L LA or OA as assessed by immunofluorescence at 0 min and 20 min. The arrows indicate the colocalization of GPR120 (GFP) and β-arrestin2 (RFP). Scale bar, 10 μm. In A, ***P < 0.001 for GPR120-overexpressing HEK293 cells incubated with the supernatant of islets exposed to 1 mmol/L glucose compared with those incubated with the supernatant of islets exposed to 11.1 mmol/L glucose. In B, ***P < 0.001 for islet supernatants treated with 1 mmol/L glucose compared with those treated with 11.1 mmol/L glucose. In C, ***P < 0.001 for GPR120-overexpressing HEK293 cells incubated with ΔOA (2.3 μmol/L) or ΔLA (5.2 μmol/L) compared with those treated with vehicle. In D and E, *P < 0.05, **P < 0.01, and ***P < 0.001 for GPR120-overexpressing HEK293 cells pretreated with MK8245 or SC26196 compared with those with vehicle. ###P < 0.001 for GPR120-overexpressing HEK293 cells incubated with 11.1 mmol/L glucose compared with treatment with 2.8 mmol/L glucose. In F and G, *P < 0.05, ***P < 0.001, and ns for islet supernatants pretreated with MK8245 or SC26196 compared with those with vehicle; ###P < 0.001 for islet supernatants treated with 2.8 mmol/L glucose compared with those treated with 11.1 mmol/L glucose. The bars represent mean ± SEM. Data statistics were analyzed using one-way (AC) or two-way (DG) ANOVA with the Dunnett post hoc test.

Figure 2

Identification of endogenous ligands of GPR120 in pancreatic islets isolated from wild-type (WT) mice. A: Recruitment of β-arrestin2 to GPR120 in GPR120-overexpressing HEK293 cells in response to incubation with supernatants of islets in response to treatment with 2.8 mmol/L or 11.1 mmol/L glucose for 1 h. A total of 400 islets from six to eight WT mice were grouped (200 islets/group) in one experiment. The data are from six independent experiments (n = 6). B: FA composition in the supernatant of islets upon treatment with 2.8 mmol/L or 11.1 mmol/L glucose for 1 h. A total of 400 islets from six to eight WT mice were grouped (200 islets/group) for metabolomics analysis in one experiment. The data are from six independent experiments (n = 6). We experimentally determined that 1,000 islets approximately have 10 μL volume and estimated that the average islet has a volume of 10 nL. Using these estimations, we predicted that the extracellular OA and LA concentrations in each islet are 209.99 ± 13.92 μmol/L and 182.01 ± 7.87 μmol/L, respectively, at low glucose, and are 322.86 ± 10.87 μmol/L (OA) and 440.8 ± 11.47 μmol/L (LA) at high glucose. C: Recruitment of β-arrestin2 to GPR120 in GPR120-overexpressing HEK293 cells in response to incubation with ΔOA (2.3 μmol/L), ΔLA (5.2 μmol/L), or control vehicle. The data are from three independent experiments (n = 3). D: Recruitment of β-arrestin2 to GPR120 in GPR120-overexpressing HEK293 cells in response to incubation with supernatants of islets treated with 2.8 mmol/L glucose or 11.1 mmol/L glucose pretreated with vehicle or 1 μmol/L MK8245 for 8 h. E: Recruitment of β-arrestin2 to GPR120 in GPR120-overexpressing HEK293 cells in response to incubation with supernatants of islets treated with 2.8 mmol/L glucose or 11.1 mmol/L glucose pretreated with or without 20 μmol/L SC26196 for 8 h. F: FA composition in the supernatant of islets upon treatment with 2.8 mmol/L or 11.1 mmol/L glucose pretreated with vehicle or 1 μmol/L MK8245 for 8 h. G: FA composition in the supernatant of islets upon treatment with 2.8 mmol/L or 11.1 mmol/L glucose pretreated with vehicle or 20 μmol/L SC26196 for 8 h. In D–G, a total of 800 islets from 12 to 16 mice were grouped (200 islets/group) for the indicated measurement in one experiment. The data are from six independent experiments (n = 6). H: GPR120 internalization in HEK293 cells treated with 200 μmol/L LA or OA as assessed by immunofluorescence at 0 min and 20 min. The arrows indicate the colocalization of GPR120 (GFP) and β-arrestin2 (RFP). Scale bar, 10 μm. In A, ***P < 0.001 for GPR120-overexpressing HEK293 cells incubated with the supernatant of islets exposed to 1 mmol/L glucose compared with those incubated with the supernatant of islets exposed to 11.1 mmol/L glucose. In B, ***P < 0.001 for islet supernatants treated with 1 mmol/L glucose compared with those treated with 11.1 mmol/L glucose. In C, ***P < 0.001 for GPR120-overexpressing HEK293 cells incubated with ΔOA (2.3 μmol/L) or ΔLA (5.2 μmol/L) compared with those treated with vehicle. In D and E, *P < 0.05, **P < 0.01, and ***P < 0.001 for GPR120-overexpressing HEK293 cells pretreated with MK8245 or SC26196 compared with those with vehicle. ###P < 0.001 for GPR120-overexpressing HEK293 cells incubated with 11.1 mmol/L glucose compared with treatment with 2.8 mmol/L glucose. In F and G, *P < 0.05, ***P < 0.001, and ns for islet supernatants pretreated with MK8245 or SC26196 compared with those with vehicle; ###P < 0.001 for islet supernatants treated with 2.8 mmol/L glucose compared with those treated with 11.1 mmol/L glucose. The bars represent mean ± SEM. Data statistics were analyzed using one-way (AC) or two-way (DG) ANOVA with the Dunnett post hoc test.

Close modal

We then used lipidomics to analyze the islet supernatants, focusing on free FAs and PAHSA, which have been reported to be endogenous GPR120 ligands in other tissues (16). In particular, unsaturated FAs were investigated because saturated FAs were reported to be much weaker agonists than unsaturated FAs to induce GPR120 activation (15,19). PAHSA levels were lower, while OA (C18:1) and LA (C18:2) levels were higher, in the 11.1 mmol/L glucose supernatant than in the low-glucose supernatant (Fig. 2B and Supplementary Fig. 2E). Because PAHSA has been reported to be a potent GPR120 agonist (16), the decreased PAHSA level is unlikely to contribute to the increased GPR120 endogenous activity under high-glucose conditions. To further investigate whether OA and LA are potential GPR120 ligands in pancreatic islets that respond to high-glucose stimulation, we calculated the absolute values of ΔOA and ΔLA concentrations by comparing the islet supernatants from the 11.1 mmol/L glucose condition and 2.8 mmol/L glucose condition, and the results revealed concentrations of 2.26 ± 0.02 μmol/L and 5.18 ± 0.02 μmol/L, respectively (Fig. 2B). Our data indicated that GPR120 was able to sense ΔOA or ΔLA under these conditions by a β-arrestin2 recruitment assay (Fig. 2C). Moreover, we examined the effect of MK8245 and SC26196 on GPR120 activity induced by islet supernatant at the 11.1 mmol/L glucose condition (Fig. 2D and E). Whereas MK8245 is an OA synthetase (stearoyl CoA desaturase 1 [SCD1]) inhibitor (39), SC26196 is reported as an LA desaturase (FA desaturase 2 [FADS2]) inhibitor (40) (Supplementary Fig. 2F). Preincubation with MK8245 for 8 h reduced the corresponding GPR120 activity and OA level, and preincubation with SC26196 increased the corresponding GPR120 activity and LA level (Fig. 2D–G). Internalization of GPR120 and colocalization of GPR120 with β-arrestin2 in response to OA or LA treatment further supported that OA and LA are potential GPR120 ligands in islets (Fig. 2H). Considering the broad roles of SCD1 and FADS2, it did not exclude other FA metabolites might also participate in endogenous GPR120 signaling within islets. Collectively, these data supported that the increase in OA and LA concentrations in pancreatic islets might contribute to GPR120 activation in our assays. Further experiments using quantitative metabolomics, pharmacological characterization of every component of individual FAs, and knockout models of SCD1 and FADS2 may help to investigate the detailed roles of LA and FA in endogenous GPR120 signaling circuit within pancreatic isles.

Biased Activation of GPR120 by OA and LA in HEK293 Cells

G protein and β-arrestin are both common signal transducers downstream of GPCRs (24,25,30,37,4146). Understanding the G protein or arrestin signaling biases of GPR120 in response to different endogenous ligands may facilitate the delineation of GPR120 functions in different physiological contexts (20,21). In general, the concentration-dependent curves suggested that OA and LA exhibited Gi activities similar to that of the reference endogenous GPR120 ligand 9-PAHSA (16,47) (Fig. 3D). Although LA showed stronger Gq activity than 9-PAHSA, both OA and LA recruited less arrestin than 9-PAHSA, the reference molecule. In particular, LA showed significantly stronger Gq and β-arrestin2 activity than OA (Fig. 3A–D and Supplementary Fig. 3A). We then used the biased β-value, which can be derived through an operational model (43), to quantify the relative activity of one pathway over another compared with that of the pathway induced by the reference agonist (41) (Fig. 3E). The β-value showed that LA was more biased toward β-arrestin2 and Gq than OA (Fig. 3E and Supplementary Fig. 3C). These data indicate that OA and LA show different bias properties in a model system.

Figure 3

Biases of endogenous GPR120 ligands using HEK293 model. Dose-dependent β-arrestin1 (A) or β-arrestin2 (B) recruitment to GPR120 in GPR120-transfected HEK293 cells in response to stimulation with TUG891, PAHSA, OA, or LA. The data are from three independent experiments (n = 3) and are expressed as the means ± SEMs. C: Dose-dependent inhibition of FSK-induced cAMP accumulation in GPR120-transfected HEK293 cells in response to stimulation with TUG891, PAHSA, OA, or LA. The data are from three independent experiments (n = 3) and are expressed as the means ± SEMs. D: Dose-dependent Gβγ release from Gq in GPR120-transfected HEK293 cells in response to stimulation with TUG891, PAHSA, OA, or LA. RLU, relative luciferase unit. The data are from three independent experiments (n = 3) and are expressed as the means ± SEMs. E: Biases of TUG891, OA, and LA calculated for GPR120 using PAHSA as the reference ligand.

Figure 3

Biases of endogenous GPR120 ligands using HEK293 model. Dose-dependent β-arrestin1 (A) or β-arrestin2 (B) recruitment to GPR120 in GPR120-transfected HEK293 cells in response to stimulation with TUG891, PAHSA, OA, or LA. The data are from three independent experiments (n = 3) and are expressed as the means ± SEMs. C: Dose-dependent inhibition of FSK-induced cAMP accumulation in GPR120-transfected HEK293 cells in response to stimulation with TUG891, PAHSA, OA, or LA. The data are from three independent experiments (n = 3) and are expressed as the means ± SEMs. D: Dose-dependent Gβγ release from Gq in GPR120-transfected HEK293 cells in response to stimulation with TUG891, PAHSA, OA, or LA. RLU, relative luciferase unit. The data are from three independent experiments (n = 3) and are expressed as the means ± SEMs. E: Biases of TUG891, OA, and LA calculated for GPR120 using PAHSA as the reference ligand.

Close modal

Islet Functions Are Regulated by GPR120 to Different Extents in Response to Stimulation by OA and LA

Both OA and LA inhibited glucose-stimulated SST secretion and increased insulin and glucagon secretion (Fig. 4A–C). Notably, the same concentration of LA (200 μmol/L) exerted greater effects on promoting insulin and glucagon secretion and inhibiting SST secretion than OA (200 μmol/L) (Fig. 4A–C and Supplementary Fig. 3D). Importantly, the effects of OA and LA on SST and insulin secretion were significantly reversed by δ-cell–specific GPR120 knockdown (Fig. 4D–G). Although application of the selective GPR40 antagonist GW1100 showed no significant effects on SST secretion modulated by OA or LA, it partially decreased insulin secretion in response to OA or LA stimulation (Supplementary Fig. 3E and F). Therefore, both GPR120 and GPR40 participated in insulin secretion in response to OA or LA stimulation. These results demonstrate the important role of pancreatic δ-cell GPR120 in contributing to OA and LA functions to regulate SST and insulin secretion from islets (Fig. 4D and E).

Figure 4

Effects of the endogenous GPR120 ligands OA and LA on hormone secretion in islets isolated from wild-type mice and Sst-cre+/− mice. Effects of OA (200 μmol/L) or LA (200 μmol/L) treatment on glucose-induced SST (A), insulin (B), or GCG (C) secretion in islets. A total of 200 islets from two to four wild-type mice were grouped (50 islets/group) for SST, insulin, or GCG secretion measurement in one experiment. The data are from six independent experiments (n = 6). Effects of OA or LA treatment on glucose-induced SST (D), insulin (E), or GCG (F) secretion in islets isolated from Sst-Cre+/− mice pretreated with lentiviruses expressing a control sequence or GPR120 shRNAf/f. A total of 300 islets from four to six Sst-Cre+/− mice were grouped (50 islets/group) for SST, insulin, or GCG secretion measurement in one experiment. The data are from six independent experiments (n = 6). G: Schematic of the roles of the endogenous ligand-GPR120 circuits in islets in glucose-induced SST and insulin secretion. In AC, ***P < 0.001 for islets treated with OA or LA compared with those treated with vehicle; ###P < 0.001 for islets treated with OA compared with those treated with LA. In DF, ***P < 0.001 for islets derived from Sst-Cre+/− mice treated with OA or LA compared with those treated with vehicle; ###P < 0.001 and ns for islets derived from Sst-Cre+/− mice treated with lentiviruses expressing GPR120 shRNAf/f compared with those treated with lentiviruses expressing a control sequence upon treatment with OA or LA. The bars represent mean ± SEM. Data statistics were analyzed using two-way ANOVA with the Dunnett post hoc test.

Figure 4

Effects of the endogenous GPR120 ligands OA and LA on hormone secretion in islets isolated from wild-type mice and Sst-cre+/− mice. Effects of OA (200 μmol/L) or LA (200 μmol/L) treatment on glucose-induced SST (A), insulin (B), or GCG (C) secretion in islets. A total of 200 islets from two to four wild-type mice were grouped (50 islets/group) for SST, insulin, or GCG secretion measurement in one experiment. The data are from six independent experiments (n = 6). Effects of OA or LA treatment on glucose-induced SST (D), insulin (E), or GCG (F) secretion in islets isolated from Sst-Cre+/− mice pretreated with lentiviruses expressing a control sequence or GPR120 shRNAf/f. A total of 300 islets from four to six Sst-Cre+/− mice were grouped (50 islets/group) for SST, insulin, or GCG secretion measurement in one experiment. The data are from six independent experiments (n = 6). G: Schematic of the roles of the endogenous ligand-GPR120 circuits in islets in glucose-induced SST and insulin secretion. In AC, ***P < 0.001 for islets treated with OA or LA compared with those treated with vehicle; ###P < 0.001 for islets treated with OA compared with those treated with LA. In DF, ***P < 0.001 for islets derived from Sst-Cre+/− mice treated with OA or LA compared with those treated with vehicle; ###P < 0.001 and ns for islets derived from Sst-Cre+/− mice treated with lentiviruses expressing GPR120 shRNAf/f compared with those treated with lentiviruses expressing a control sequence upon treatment with OA or LA. The bars represent mean ± SEM. Data statistics were analyzed using two-way ANOVA with the Dunnett post hoc test.

Close modal

We next examined the modulation of glucagon secretion by OA and LA via GPR120 in pancreatic δ-cells. GPR120 knockdown did not have significant effects on glucagon secretion, which is consistent with previous research (48) (Fig. 1H). Upon stimulation with OA or LA, the glucagon release level was significantly increased, which was not reversed by knockdown of GPR120 in δ-cells (Fig. 4C and F). We speculated that the increased glucagon secretion in response to OA or LA stimulation may be due to the expression of GPR120 in pancreatic α-cells. The involvement of GPR120 in pancreatic δ-cells with glucagon secretion could be complicated because glucagon secretion is modulated by both SST and insulin (14,4952). Further investigations should be conducted to clarify the detailed underlying mechanism of this phenotype in the future.

Inflammation and oxidative stress are two important pathways involved in islet dysfunction and diabetes development (53,54). We observed that both OA and LA were able to reduce the expression of the inflammation-related genes Ccl2, Ccl5, Cxcl10, and Cxcl12 and stress response markers Cyba, Sod2, Fth1, and Nos2 induced by combined tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and interferon (IFN)-γ treatment in islets isolated from wild-type mice (55,56) (Fig. 5A and B). Notably, under conditions of specific knockdown of GPR120 expression in δ-cells, the anti-inflammatory and antistress response effects of the two ligands were significantly weakened, suggesting that GPR120 mediated these effects (Fig. 5C and D and Supplementary Fig. 4A and B). In particular, LA showed greater anti-inflammatory effects than OA at 200 μmol/L (Fig. 5A and C). To further explore the mechanism of the anti-inflammatory effects of GPR120 signaling in δ-cells, we isolated primary δ-cells from islets of Sst-cre+/− mice pretreated with lentiviruses expressing a control sequence or GPR120 shRNAf/f. We found that the expression and secretion of inflammatory factors in primary δ-cells increased significantly in response to proinflammatory cytokines, which were downregulated by stimulation with OA or LA (Supplementary Fig. 4C and D). Knockdown of GPR120 in δ-cells reversed the effects of OA and LA, providing evidence to support that GPR120 plays key roles in the anti-inflammatory effects of LA and OA on δ-cells (Supplementary Fig. 4CE). These results provide a better understanding of the function of pancreatic δ-cells.

Figure 5

Effects of endogenous GPR120 ligands on inflammation and oxidative stress. A: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the mRNA levels of inflammatory markers, including Ccl2, Ccl5, Cxcl10 and Cxcl12, stimulated by proinflammatory cytokines (30 units/mL IL-1β, 1,000 units/mL TNF-α, and 1,000 units/mL IFN-γ) in islets. B: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the mRNA levels of stress response genes, including Cyba, Sod2, Fth1, and Nos2, stimulated by proinflammatory cytokines in islets. In A and B, a total of 400 islets from six to eight wild-type (WT) mice were grouped (100 islets/group) for RNA extraction in one experiment. The data are from six independent experiments (n = 6). C: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the secretion of several inflammatory marker proteins, including CCL2, CCL5, CXCL10, and CXCL12, stimulated by proinflammatory cytokines (30 units/mL IL-1β, 1000 units/mL TNF-α, and 1000 units/mL IFN-γ) in islets isolated from Sst-Cre+/− mice pretreated with lentiviruses expressing a control sequence or GPR120 shRNAf/f. A total of 600 islets from 10 to 12 Sst-Cre+/− mice were grouped (100 islets/group) for secretion measurement in one experiment. The data are from three independent experiments (n = 3). D: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the fluorescence levels of ROS and NO in islets isolated from Sst-Cre+/− mice pretreated with lentiviruses expressing a control sequence or GPR120 shRNAf/f. A total of 900 islets from 14 to 16 Sst-Cre+/− mice were grouped (150 islets/group) for ROS and NO measurement in one experiment. The ROS and NO levels were quantitated using an assay kit following the manufacturer’s instructions. The ROS levels were measured with the Reactive Oxygen Species Assay Kit (cat. E004-1-1, Nanjing Jiancheng Bio) and NO with Nitric Oxide Assay Kit (cat. A012-1-2, Nanjing Jiancheng Bio). The data are from three independent experiments (n = 3). Dose-dependent insulin (E) or SST (F) secretion of islets isolated from WT mice in response to stimulation of OA or LA. A total of 250 islets from three to five WT mice were grouped (50 islets/group) for insulin or SST secretion measurement. The data are from six independent experiments (n = 6). G and H: Dose-dependent ccl5 and cxcl10 expression of islets isolated from WT mice in response to OA or LA. A total of 500 islets from 8 to 10 WT mice were grouped (100 islets/group) for RNA extraction in one experiment. The data are from six independent experiments (n = 6). I and J: Function bias of LA calculated through operational model using OA ligand as reference. In A and B, ***P < 0.001 for islets treated with OA or LA compared with those treated with vehicle upon stimulation with cytokines; ##P < 0.01, ###P < 0.001, and ns for islets treated with OA compared with those treated with LA upon stimulation with cytokines. In C and D, **P < 0.01 and ***P < 0.001 for islets induced by proinflammatory cytokines treated with OA or LA compared with islets induced by proinflammatory cytokines treated with vehicle; #P < 0.05 and ###P < 0.001 for isolated from Sst-Cre+/− mice pretreated with lentiviruses expressing GPR120 shRNAf/f compared with those treated with lentiviruses expressing negative control sequence. The bars represent mean ± SEM. Data statistics were analyzed using two-way ANOVA with the Dunnett post hoc test.

Figure 5

Effects of endogenous GPR120 ligands on inflammation and oxidative stress. A: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the mRNA levels of inflammatory markers, including Ccl2, Ccl5, Cxcl10 and Cxcl12, stimulated by proinflammatory cytokines (30 units/mL IL-1β, 1,000 units/mL TNF-α, and 1,000 units/mL IFN-γ) in islets. B: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the mRNA levels of stress response genes, including Cyba, Sod2, Fth1, and Nos2, stimulated by proinflammatory cytokines in islets. In A and B, a total of 400 islets from six to eight wild-type (WT) mice were grouped (100 islets/group) for RNA extraction in one experiment. The data are from six independent experiments (n = 6). C: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the secretion of several inflammatory marker proteins, including CCL2, CCL5, CXCL10, and CXCL12, stimulated by proinflammatory cytokines (30 units/mL IL-1β, 1000 units/mL TNF-α, and 1000 units/mL IFN-γ) in islets isolated from Sst-Cre+/− mice pretreated with lentiviruses expressing a control sequence or GPR120 shRNAf/f. A total of 600 islets from 10 to 12 Sst-Cre+/− mice were grouped (100 islets/group) for secretion measurement in one experiment. The data are from three independent experiments (n = 3). D: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the fluorescence levels of ROS and NO in islets isolated from Sst-Cre+/− mice pretreated with lentiviruses expressing a control sequence or GPR120 shRNAf/f. A total of 900 islets from 14 to 16 Sst-Cre+/− mice were grouped (150 islets/group) for ROS and NO measurement in one experiment. The ROS and NO levels were quantitated using an assay kit following the manufacturer’s instructions. The ROS levels were measured with the Reactive Oxygen Species Assay Kit (cat. E004-1-1, Nanjing Jiancheng Bio) and NO with Nitric Oxide Assay Kit (cat. A012-1-2, Nanjing Jiancheng Bio). The data are from three independent experiments (n = 3). Dose-dependent insulin (E) or SST (F) secretion of islets isolated from WT mice in response to stimulation of OA or LA. A total of 250 islets from three to five WT mice were grouped (50 islets/group) for insulin or SST secretion measurement. The data are from six independent experiments (n = 6). G and H: Dose-dependent ccl5 and cxcl10 expression of islets isolated from WT mice in response to OA or LA. A total of 500 islets from 8 to 10 WT mice were grouped (100 islets/group) for RNA extraction in one experiment. The data are from six independent experiments (n = 6). I and J: Function bias of LA calculated through operational model using OA ligand as reference. In A and B, ***P < 0.001 for islets treated with OA or LA compared with those treated with vehicle upon stimulation with cytokines; ##P < 0.01, ###P < 0.001, and ns for islets treated with OA compared with those treated with LA upon stimulation with cytokines. In C and D, **P < 0.01 and ***P < 0.001 for islets induced by proinflammatory cytokines treated with OA or LA compared with islets induced by proinflammatory cytokines treated with vehicle; #P < 0.05 and ###P < 0.001 for isolated from Sst-Cre+/− mice pretreated with lentiviruses expressing GPR120 shRNAf/f compared with those treated with lentiviruses expressing negative control sequence. The bars represent mean ± SEM. Data statistics were analyzed using two-way ANOVA with the Dunnett post hoc test.

Close modal

Separation Window of the Functional Biases of OA and LA

We investigated the concentration-dependent effects of OA and LA on hormone secretion and anti-inflammation in pancreatic islets (Fig. 5E–J and Supplementary Fig. 5AF). As the concentration of OA and LA increased, insulin secretion showed a dose-dependent increase using mouse islets (Fig. 5E and F and Supplementary Fig. 5A and B). The half-maximal effective concentration (EC50) values for insulin secretion in response to LA and OA were 55.97 ± 4.38 μmol/L and 343.7 ± 27.41 μmol/L, respectively, which are separated by one magnitude. The Emax of LA-induced insulin secretion is also ∼1.5-fold that of OA. In contrast, SST secretion decreased with increasing concentrations of LA and OA, with higher potency found for LA (EC50 = 55.4 ± 5.63 μmol/L) than OA (EC50 = 218.2 ± 28.65 μmol/L). These results indicated that LA showed higher potency or greater efficacy on insulin and SST secretion than OA.

However, in response to combined TNF-α, IL-1β, and IFN-γ treatment, a much smaller dose-dependent separation between OA and LA was observed in terms of their abilities to inhibit the expression of two inflammation-related genes, Ccl5 and Cxcl10 (Fig. 5G and H). The separation window for the potency between LA and OA in terms of the effects on Ccl5 and Cxcl10 expression was less than twofold, and their efficacies were comparable. Knockdown of GPR120 offset the anti-inflammatory effect, confirming the dependence on GPR120 for these effects (Supplementary Fig. 5C and D). We then used the operational model to derive the biased parameter β-value, which indicated that LA exhibited insulin/SST secretion functional bias relative to OA (Fig. 5I and J and Supplementary Fig. 5E and F). Taken together, these results demonstrate that OA and LA exhibit biased islet functions in a dose-dependent manner via interaction with GPR120. LA and OA showed a larger functional separation window for insulin/SST secretion compared with that for the effects on anti-inflammatory gene expression.

Contribution of β-arrestin2 Mediated Biased GPR120 Signaling to Islet Function

Notably, the difference of the biased factor in arrestin signaling between OA and LA was similar to their biased factor β in insulin/SST secretion (Fig. 3E and Fig. 5I and J). We therefore examined whether the functional differences between OA and LA are mediated by β-arrestin2. When the synthetic GPR120 agonist TUG891 was used as the control, both OA and LA were found to promote the direct interaction between native GPR120 and β-arrestin1 or β-arrestin2 in mouse pancreatic islets by coimmunoprecipitation assays (Fig. 6A and B and Supplementary Fig. 6AE). Deletion of β-arrestin2 partly eliminated the differences between OA and LA in promoting insulin secretion and inhibiting SST secretion (Fig. 6C and D and Supplementary Fig. 6F and G). However, there was no significant difference between the islets derived from β-arrestin1−/− mice or their wild-type littermates in terms of insulin secretion under the presence or absence of GPR120 agonist stimulation (Fig. 6E and F). Previous studies have shown that GPR120 internalization with β-arrestin2 mediates anti-inflammatory effects in RAW 264.7 cells (15). We next used β-arrestin2−/− islets to explore whether the anti-inflammatory and antistress response effects of both GPR120 ligands were mediated by β-arrestin2. We observed that the addition of OA and LA did not obviously decrease the secretion of inflammatory factors or reactive oxygen species (ROS) and nitric oxide (NO) production after β-arrestin2 deletion in pancreatic islets, which is consistent with the mRNA examination data (Fig. 6G–I and Supplementary Fig. 6H and I). In islets derived from β-arrestin1−/− mice or Gq knockdown by transduction of the shRNA-Gqf/f lentivirus in islets isolated from Sst-Cre+/− mice, the anti-inflammatory effects of OA and LA showed no significant difference compared with islets derived from wild-type littermates or islet transduction with control shRNA, respectively (Supplementary Figs. 7A–D, and 8AD and G). Pentoxifylline (PTX) treatment showed no significant effects on anti-inflammatory functions, but could partially reverse the effects of OA or LA on SST secretion (Supplementary Fig. 8E and F). Therefore, β-arrestin2–mediated signaling might serve as the major contributor to the greater effect of LA on islet hormone and anti-inflammatory effects than OA at the same concentration (200 μmol/L).

Figure 6

Contribution of β-arrestin2-mediated biased GPR120 signaling to islet function. A: Representative Western blots showing the coimmunoprecipitation of β-arrestin1 and β-arrestin2 with GPR120 in pancreatic islets. Islets from wild-type (WT) mice were grouped (500 islets/group) and stimulated with 10 μmol/L TUG891, 200 μmol/L LA, and 200 μmol/L OA for 10 min. β-arrestin1 and β-arrestin2 were immunoprecipitated by GPR120 antibody and protein A/G agarose, the formation of GPR120/β-arrestin1 and GPR120/β-arrestin2 complex were detected by specific antibodies. IB, immunoblotting; IP, immunoprecipitation. B: Quantification of β-arrestin1 (left) and β-arrestin2 levels (right) after coimmunoprecipitation by protein A/G agarose from the lysates of islets lysates cultured with GPR120 antibody overnight at 4°C. Islets were stimulated with 10 μmol/L TUG891, 200 μmol/L LA, and 200 μmol/L OA for 10 min. The data are correlated with Fig. 7A and are from three independent experiments (n = 3). Glucose-induced insulin (C) or SST (D) secretion in islets isolated from WT and β-arrestin2−/− mice upon stimulation with OA or LA. A total of 200 islets from two to four WT or β-arrestin2−/− mice were grouped (50 islets/group) for SST or insulin secretion measurement in one experiment. The data are from six independent experiments (n = 6). Glucose-induced insulin (E) or SST (F) secretion in islets isolated from WT and β-arrestin1−/− mice upon stimulation with OA or LA. A total of 200 islets from two to four WT or β-arrestin1−/− mice were grouped (50 islets/group) for SST or insulin secretion measurement in one experiment. The data are from six independent experiments (n = 6). G: Effects of OA and LA treatment on the secretion of several inflammatory marker proteins, including CCL2, CCL5, CXCL10, and CXCL12, stimulated by proinflammatory cytokines (30 units/mL IL-1β, 1,000 units/mL TNF-α, and 1,000 units/mL IFN-γ) in islets isolated from β-arrestin2−/− or WT mice. A total 400 islets from six to eight WT or β-arrestin2−/− mice were grouped (100 islets/group) for secretion measurement in one experiment. The data are from three independent experiments (n = 3). Effects of OA and LA treatment on the fluorescence levels of ROS (H) or NO (I) stimulated by proinflammatory cytokines (30 units/mL IL-1β, 1,000 units/mL TNF-α, and 1,000 units/mL IFN-γ) in islets isolated from WT or β-arrestin2−/− mice. A total of 600 islets from 10 to 12 WT or β-arrestin2−/− mice were grouped (150 islets/group) for ROS or NO measurement in one experiment. The ROS and NO levels were quantitated using an assay kit following the manufacturer’s instructions. The ROS levels were measured with the Reactive Oxygen Species Assay Kit (cat. E004-1-1, Nanjing Jiancheng Bio) and NO with Nitric Oxide Assay Kit (cat. A012-1-2, Nanjing Jiancheng Bio). The data are from three independent experiments (n = 3). In C and D and GI, ***P < 0.001 and ns for islets isolated from WT mice compared with islets isolated from β-arrestin2−/− mice. In E and F, ns for islets isolated from WT mice compared with islets isolated from β-arrestin1−/− mice. The bars represent mean ± SEM. Data statistics were analyzed using two-way ANOVA with the Dunnett post hoc test.

Figure 6

Contribution of β-arrestin2-mediated biased GPR120 signaling to islet function. A: Representative Western blots showing the coimmunoprecipitation of β-arrestin1 and β-arrestin2 with GPR120 in pancreatic islets. Islets from wild-type (WT) mice were grouped (500 islets/group) and stimulated with 10 μmol/L TUG891, 200 μmol/L LA, and 200 μmol/L OA for 10 min. β-arrestin1 and β-arrestin2 were immunoprecipitated by GPR120 antibody and protein A/G agarose, the formation of GPR120/β-arrestin1 and GPR120/β-arrestin2 complex were detected by specific antibodies. IB, immunoblotting; IP, immunoprecipitation. B: Quantification of β-arrestin1 (left) and β-arrestin2 levels (right) after coimmunoprecipitation by protein A/G agarose from the lysates of islets lysates cultured with GPR120 antibody overnight at 4°C. Islets were stimulated with 10 μmol/L TUG891, 200 μmol/L LA, and 200 μmol/L OA for 10 min. The data are correlated with Fig. 7A and are from three independent experiments (n = 3). Glucose-induced insulin (C) or SST (D) secretion in islets isolated from WT and β-arrestin2−/− mice upon stimulation with OA or LA. A total of 200 islets from two to four WT or β-arrestin2−/− mice were grouped (50 islets/group) for SST or insulin secretion measurement in one experiment. The data are from six independent experiments (n = 6). Glucose-induced insulin (E) or SST (F) secretion in islets isolated from WT and β-arrestin1−/− mice upon stimulation with OA or LA. A total of 200 islets from two to four WT or β-arrestin1−/− mice were grouped (50 islets/group) for SST or insulin secretion measurement in one experiment. The data are from six independent experiments (n = 6). G: Effects of OA and LA treatment on the secretion of several inflammatory marker proteins, including CCL2, CCL5, CXCL10, and CXCL12, stimulated by proinflammatory cytokines (30 units/mL IL-1β, 1,000 units/mL TNF-α, and 1,000 units/mL IFN-γ) in islets isolated from β-arrestin2−/− or WT mice. A total 400 islets from six to eight WT or β-arrestin2−/− mice were grouped (100 islets/group) for secretion measurement in one experiment. The data are from three independent experiments (n = 3). Effects of OA and LA treatment on the fluorescence levels of ROS (H) or NO (I) stimulated by proinflammatory cytokines (30 units/mL IL-1β, 1,000 units/mL TNF-α, and 1,000 units/mL IFN-γ) in islets isolated from WT or β-arrestin2−/− mice. A total of 600 islets from 10 to 12 WT or β-arrestin2−/− mice were grouped (150 islets/group) for ROS or NO measurement in one experiment. The ROS and NO levels were quantitated using an assay kit following the manufacturer’s instructions. The ROS levels were measured with the Reactive Oxygen Species Assay Kit (cat. E004-1-1, Nanjing Jiancheng Bio) and NO with Nitric Oxide Assay Kit (cat. A012-1-2, Nanjing Jiancheng Bio). The data are from three independent experiments (n = 3). In C and D and GI, ***P < 0.001 and ns for islets isolated from WT mice compared with islets isolated from β-arrestin2−/− mice. In E and F, ns for islets isolated from WT mice compared with islets isolated from β-arrestin1−/− mice. The bars represent mean ± SEM. Data statistics were analyzed using two-way ANOVA with the Dunnett post hoc test.

Close modal

Notably, the cellular context of islet cell types could alter the signaling bias, especially due to the differential expression of G protein or arrestin subtypes between different cell types. Difficulties remain in measuring the G protein or arrestin activity directly in vivo. Therefore, we examined the expression profiles of different G protein or arrestin subtypes in primary pancreatic islet δ-cells to provide preliminary insight into how the expression profiles of pancreatic islet δ-cells may affect the signaling bias of GPR120 properties. Our data indicated that β-arrestin2, Gs, G11, Gi2, and Gi3 were more relatively enriched in pancreatic δ-cells compared with the brain (Supplementary Fig. 3B). This observation is consistent with the contribution of β-arrestin2 signaling to GPR120 function in δ-cells.

Impairment of Endogenous Islet GPR120 Circuits in a Diabetic Model

Not only was GPR120 expression decreased, but the levels of the islet GPR120 ligands OA and LA were also significantly reduced in islets derived from db/db mice (Fig. 7A and B). These data suggested that the endogenous islet ligand-GPR120 circuits were impaired in the diabetic db/db models. In contrast, there was little difference in PAHSA content in islets from db/db mice compared with those from db/+ mice (Supplementary Fig. 9A). Treating the islets isolated from db/db mice with 200 μmol/L OA and 200 μmol/L LA increased GPR120 receptor expression and glucose-stimulated insulin secretion and reduced SST secretion, with a greater effect mediated by LA than by OA at the same concentration (Fig. 7C and D and Supplementary Fig. 9B). In addition, the administration of OA and LA ameliorated inflammation and oxidative stress in the islets derived from db/db mice (Fig. 7E–G and Supplementary Fig. 9C and D). Moreover, LA significantly ameliorated the glucose tolerance impairment in db/db mice, while OA showed no significant effects (Fig. 7H). In streptozotocin (STZ)-induced diabetic mice, we also found that OA and LA could restore insulin secretion and antagonize inflammation in isolated islets (Fig. 7I–K and Supplementary Fig. 9F and G).

Figure 7

Functions of endogenous ligands (OA and LA) via GPR120 in diabetes mice. A: qRT-PCR analysis of the mRNA expression levels of GPR120 in islets isolated from db/+ and db/db mice. A total of 200 islets from two to four db/+ or db/db mice were grouped (100 islets/group) for RNA extraction in one experiment. The data are from six independent experiments (n = 6). B: FA composition in islets isolated from db/+ and db/db mice. A total of 400 islets from six to eight db/+ or db/db mice were grouped (200 islets/group) for metabolomics analysis in one experiment. The data are from six independent experiments (n = 6). The OA and LA concentrations in 266 μL lysate of 200 isolated db/+ islets are 2.2 ± 0.2 μmol/L (OA) and 2.0 ± 0.1 μmol/L (LA), and in that of db/db islets are 1.0 ± 0.1 μmol/L (OA) and 0.6 ± 0.05 μmol/L (LA). We experimentally determined that 1,000 islets approximately have 10 μL volume and estimated that the average islet has a volume of 10 nL. Using this estimation, we predicted that the OA and LA concentrations are 311.11 ± 24.68 μmol/L and 269.13 ± 16.93 μmol/L, respectively, in each db/+ islet; and 137.89 ± 13.98 μmol/L (OA) and 83.68 ± 6.61 μmol/L (LA) in each db/db islet. Effects of OA and LA treatment on glucose-induced insulin (C) and SST (D) secretion in islets isolated from db/+ or db/db mice pretreated with adenoviruses expressing control or GPR120 shRNA. A total of 350 islets from four to six db/+ or db/db mice were grouped (50 islets/group) for insulin or SST secretion measurement in one experiment. The data are from six independent experiments (n = 6). Effects of OA and LA treatment on the fluorescence levels of ROS (E) and NO (F) in islets isolated from db/+ or db/db mice pretreated with adenoviruses expressing control or GPR120 shRNA. A total of 900 islets from 16 to 18 db/+ or db/db mice were grouped (150 islets/group) for ROS or NO measurement in one experiment. The ROS and NO levels were quantitated using an assay kit following the manufacturer’s instructions. The ROS levels were measured with the Reactive Oxygen Species Assay Kit (cat. E004-1-1, Nanjing Jiancheng Bio) and NO with Nitric Oxide Assay Kit (cat. A012-1-2, Nanjing Jiancheng Bio). The data are from three independent experiments (n = 3). G: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the secretion of several inflammatory marker proteins, including CCL2, CCL5, CXCL10, and CXCL12 in islets isolated from db/+ or db/db mice pretreated with adenoviruses expressing control or GPR120 shRNA. There were 600 islets from 8 to 12 db/+ or db/db mice grouped (100 islets/group) for secretion measurement in one experiment. The data are from three independent experiments (n = 3). H: Plasma glucose levels during the glucose tolerance test in db/+ mice and their db/db littermates treated with vehicle control, OA, or LA every other day (10 μg/g, i.p.) for 10 weeks. Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on glucose-induced insulin (I) and SST (J) secretion in islets isolated from wild-type (WT) or STZ mice pretreated with adenoviruses expressing control or GPR120 shRNA. A total of 350 islets from six to eight control or STZ mice were grouped (50 islets/group) for insulin or SST secretion measurement in one experiment. The data are from six independent experiments (n = 6). K: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the secretion of several inflammatory marker proteins, including CCL2, CCL5, CXCL10, and CXCL12 in islets isolated from WT or STZ mice pretreated with adenoviruses expressing control or GPR120 shRNA. A total of 600 islets from 10 to 12 control or STZ mice were grouped (100 islets/group) for secretion measurement in one experiment. The data are from three independent experiments (n = 3). In A and B, *P < 0.05, ***P < 0.001, and ns for islets isolated from db/db mice compared with islets isolated from db/+ mice. In C and D, ***P < 0.001 for islets isolated from db/db mice compared with islets isolated from db/+ mice; ###P < 0.001 for islets isolated from db/db mice treated with OA or LA compared with those treated with vehicle; $$$P < 0.001 for islets isolated from db/db mice treated with OA or LA pretreated with adenoviruses expressing GPR120 shRNA compared with those expressing a control sequence. In EG, *P < 0.05, **P < 0.01, ***P < 0.001, and ns for islets isolated from db/db mice treated with OA or LA compared with those treated with vehicle; #P < 0.05, ##P < 0.01, ###P < 0.001, and ns for islets isolated from db/db mice treated with OA or LA pretreated with adenoviruses expressing GPR120 shRNA compared with those expressing a control sequence. In H, **P < 0.01, ***P < 0.001, and ns for OA- or LA-treated db/db mice compared with vehicle-treated mice. In I and J, ***P < 0.001 for islets isolated from STZ mice compared with islets isolated from WT mice; ###P < 0.001 for islets isolated from STZ mice treated with OA or LA compared with those treated with vehicle; $$$P < 0.001 for islets isolated from STZ mice treated with OA or LA pretreated with adenoviruses expressing GPR120 shRNA compared with those expressing a control sequence. In K, *P < 0.05, **P < 0.01, ***P < 0.001, and ns for islets isolated from STZ mice treated with OA or LA compared with those treated with vehicle; #P < 0.05, ##P < 0.01, ###P < 0.001, and ns for islets isolated from STZ mice treated with OA or LA pretreated with adenoviruses expressing GPR120 shRNA compared with those expressing a control sequence. The bars represent mean ± SEM. Data statistics were analyzed using one-way ANOVA (A and B) or two-way ANOVA (CK) with the Dunnett post hoc test.

Figure 7

Functions of endogenous ligands (OA and LA) via GPR120 in diabetes mice. A: qRT-PCR analysis of the mRNA expression levels of GPR120 in islets isolated from db/+ and db/db mice. A total of 200 islets from two to four db/+ or db/db mice were grouped (100 islets/group) for RNA extraction in one experiment. The data are from six independent experiments (n = 6). B: FA composition in islets isolated from db/+ and db/db mice. A total of 400 islets from six to eight db/+ or db/db mice were grouped (200 islets/group) for metabolomics analysis in one experiment. The data are from six independent experiments (n = 6). The OA and LA concentrations in 266 μL lysate of 200 isolated db/+ islets are 2.2 ± 0.2 μmol/L (OA) and 2.0 ± 0.1 μmol/L (LA), and in that of db/db islets are 1.0 ± 0.1 μmol/L (OA) and 0.6 ± 0.05 μmol/L (LA). We experimentally determined that 1,000 islets approximately have 10 μL volume and estimated that the average islet has a volume of 10 nL. Using this estimation, we predicted that the OA and LA concentrations are 311.11 ± 24.68 μmol/L and 269.13 ± 16.93 μmol/L, respectively, in each db/+ islet; and 137.89 ± 13.98 μmol/L (OA) and 83.68 ± 6.61 μmol/L (LA) in each db/db islet. Effects of OA and LA treatment on glucose-induced insulin (C) and SST (D) secretion in islets isolated from db/+ or db/db mice pretreated with adenoviruses expressing control or GPR120 shRNA. A total of 350 islets from four to six db/+ or db/db mice were grouped (50 islets/group) for insulin or SST secretion measurement in one experiment. The data are from six independent experiments (n = 6). Effects of OA and LA treatment on the fluorescence levels of ROS (E) and NO (F) in islets isolated from db/+ or db/db mice pretreated with adenoviruses expressing control or GPR120 shRNA. A total of 900 islets from 16 to 18 db/+ or db/db mice were grouped (150 islets/group) for ROS or NO measurement in one experiment. The ROS and NO levels were quantitated using an assay kit following the manufacturer’s instructions. The ROS levels were measured with the Reactive Oxygen Species Assay Kit (cat. E004-1-1, Nanjing Jiancheng Bio) and NO with Nitric Oxide Assay Kit (cat. A012-1-2, Nanjing Jiancheng Bio). The data are from three independent experiments (n = 3). G: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the secretion of several inflammatory marker proteins, including CCL2, CCL5, CXCL10, and CXCL12 in islets isolated from db/+ or db/db mice pretreated with adenoviruses expressing control or GPR120 shRNA. There were 600 islets from 8 to 12 db/+ or db/db mice grouped (100 islets/group) for secretion measurement in one experiment. The data are from three independent experiments (n = 3). H: Plasma glucose levels during the glucose tolerance test in db/+ mice and their db/db littermates treated with vehicle control, OA, or LA every other day (10 μg/g, i.p.) for 10 weeks. Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on glucose-induced insulin (I) and SST (J) secretion in islets isolated from wild-type (WT) or STZ mice pretreated with adenoviruses expressing control or GPR120 shRNA. A total of 350 islets from six to eight control or STZ mice were grouped (50 islets/group) for insulin or SST secretion measurement in one experiment. The data are from six independent experiments (n = 6). K: Effects of OA (200 μmol/L) and LA (200 μmol/L) treatment on the secretion of several inflammatory marker proteins, including CCL2, CCL5, CXCL10, and CXCL12 in islets isolated from WT or STZ mice pretreated with adenoviruses expressing control or GPR120 shRNA. A total of 600 islets from 10 to 12 control or STZ mice were grouped (100 islets/group) for secretion measurement in one experiment. The data are from three independent experiments (n = 3). In A and B, *P < 0.05, ***P < 0.001, and ns for islets isolated from db/db mice compared with islets isolated from db/+ mice. In C and D, ***P < 0.001 for islets isolated from db/db mice compared with islets isolated from db/+ mice; ###P < 0.001 for islets isolated from db/db mice treated with OA or LA compared with those treated with vehicle; $$$P < 0.001 for islets isolated from db/db mice treated with OA or LA pretreated with adenoviruses expressing GPR120 shRNA compared with those expressing a control sequence. In EG, *P < 0.05, **P < 0.01, ***P < 0.001, and ns for islets isolated from db/db mice treated with OA or LA compared with those treated with vehicle; #P < 0.05, ##P < 0.01, ###P < 0.001, and ns for islets isolated from db/db mice treated with OA or LA pretreated with adenoviruses expressing GPR120 shRNA compared with those expressing a control sequence. In H, **P < 0.01, ***P < 0.001, and ns for OA- or LA-treated db/db mice compared with vehicle-treated mice. In I and J, ***P < 0.001 for islets isolated from STZ mice compared with islets isolated from WT mice; ###P < 0.001 for islets isolated from STZ mice treated with OA or LA compared with those treated with vehicle; $$$P < 0.001 for islets isolated from STZ mice treated with OA or LA pretreated with adenoviruses expressing GPR120 shRNA compared with those expressing a control sequence. In K, *P < 0.05, **P < 0.01, ***P < 0.001, and ns for islets isolated from STZ mice treated with OA or LA compared with those treated with vehicle; #P < 0.05, ##P < 0.01, ###P < 0.001, and ns for islets isolated from STZ mice treated with OA or LA pretreated with adenoviruses expressing GPR120 shRNA compared with those expressing a control sequence. The bars represent mean ± SEM. Data statistics were analyzed using one-way ANOVA (A and B) or two-way ANOVA (CK) with the Dunnett post hoc test.

Close modal

Decreased GPR120 Activity With R270H Mutation in Response to LA and OA Stimulation

The single nucleotide polymorphism p.R270H mutation of GPR120 is a deleterious nonsynonymous mutation and is reported to be associated with human obesity (9,57). Importantly, both β-arrestin recruitment and G protein dissociation in p.R270H of GPR120 was decreased compared with that of wild-type GPR120 in response to stimulation with OA or LA (Fig. 8A–I). The Gi activity of GPR120 p.R270H in response to LA was particularly impaired (Fig. 8F). Notably, only the EC50 of OA stimulated the activity of the Gi pathway of GPR120 p.R270H, and the EC50 of LA-stimulated activity of β-arrestin2 of GPR120 p.R270H was lower than 200 μmol/L. Notably, a previous study has suggested that the p.R270H variant led to increased fasting glucose levels (57). We therefore speculated that the p.R270H variant of GPR120 may cause impaired pancreatic islet δ-cell function, which contributes to the fasting glucose level and could be investigated in human samples or in GPR120 p.R270H knock-in mice in the future.

Figure 8

The human GPR120 variant (R270H) reduces the ability to recruit β-arrestins under the stimulation of agonists. Dose-dependent β-arrestin1 recruitment to GPR120 in human GPR120 R270H variant and wild-type (WT) GPR120-transfected HEK293 cells in response to stimulation with OA (A) or LA (B). Dose-dependent β-arrestin2 recruitment to GPR120 in human GPR120 R270H variant and wild type (WT) GPR120-transfected HEK293 cells in response to stimulation with OA (C) or LA (D). Dose-dependent Gi heterotrimers dissociation in human GPR120 R270H variant and WT GPR120-transfected HEK293 cells in response to stimulation with OA (E) or LA (F). Dose-dependent Gq heterotrimers dissociation in human GPR120 R270H variant and WT GPR120-transfected HEK293 cells in response to stimulation with OA (G) or LA (H). I: Bias property of GPR120 R270H variant that induced by OA or LA calculated for WT GPR120 as the reference construct. In AI, the data are from three independent experiments (n = 3).

Figure 8

The human GPR120 variant (R270H) reduces the ability to recruit β-arrestins under the stimulation of agonists. Dose-dependent β-arrestin1 recruitment to GPR120 in human GPR120 R270H variant and wild-type (WT) GPR120-transfected HEK293 cells in response to stimulation with OA (A) or LA (B). Dose-dependent β-arrestin2 recruitment to GPR120 in human GPR120 R270H variant and wild type (WT) GPR120-transfected HEK293 cells in response to stimulation with OA (C) or LA (D). Dose-dependent Gi heterotrimers dissociation in human GPR120 R270H variant and WT GPR120-transfected HEK293 cells in response to stimulation with OA (E) or LA (F). Dose-dependent Gq heterotrimers dissociation in human GPR120 R270H variant and WT GPR120-transfected HEK293 cells in response to stimulation with OA (G) or LA (H). I: Bias property of GPR120 R270H variant that induced by OA or LA calculated for WT GPR120 as the reference construct. In AI, the data are from three independent experiments (n = 3).

Close modal

Paralleling our work, a recent GPR120 study (11) found that activation of GPR120 expressed in pancreatic δ-cells by synthetic agonists was able to modulate hormone secretion of islets. In the current study, we found that OA and LA may play a role as potential endogenous GPR120 ligands in response to high-glucose stimulation. Our measured OA and LA concentrations of 100 μL supernatant from 200 isolated islets were 4.2 ± 0.3 μmol/L (OA) and 3.6 ± 0.2 μmol/L (LA) under low-glucose conditions and 6.5 ± 0.2 μmol/L (OA) and 8.8 ± 0.2 μmol/L (LA) under high-glucose conditions. If we assumed that each islet had a 10 nL volume, the estimated intraislet concentrations of OA and LA could reach 322.9 μmol/L and 440.8 μmol/L, respectively. Therefore, we estimated that the intraislet concentrations of OA and LA could range from 3.6 μmol/L to 440.8 μmol/L. FAs in vivo are presented in various forms, including the aqueous-phase form that is bound to proteins, such as albumin, as well as the free fraction. Whereas lipidomics could provide useful information for the free fraction of FAs, the fraction of FAs bound with proteins is still difficult to quantify. Importantly, recent research reported a genetically encoded GPCR activation-based fluorescent sensor that enables rapid and specific detection of endogenous ligands, such as dompamine and cannabinoid, in vivo. Thus, this newly developed technology could facilitate the analysis of GPCR ligands in vivo (5860). We are currently working with Yulong Li’s group to develop a genetic sensor that will enable the real-time detection of concentrations of free FAs in vivo. Notably, the metabolic pathways of LA and OA production and degradation are mediated by a panel of enzymes, including, but not limited to, FA synthase (FASN), SCD1, adipose triglyceride lipase (ATGL), and hormone-sensitive lipase (HSL) (61). In addition, the transportation of OA and LA contribute to extracellular concentrations of LA and OA, which requires the functions of specific transporters, such as FA translocase (FAT, CD36), FA transport protein (FATP), and the membrane FA-binding protein (FABP) families, as well as caveolins (62). Under high-glucose conditions for 1 h, the mRNA levels of the abovementioned proteins were not significantly changed. We therefore speculated that the enhanced extracellular OA and LA concentrations are related to the posttranslational modification-regulated functions of these proteins, which awaits further investigation.

Importantly, the two potentially endogenous ligands of GPR120 within islets, OA and LA, exhibited different signaling biases toward G protein subtypes and arrestin in a modeled HEK293 system. In HEK293 cells, LA was more biased toward the Gq and β-arrestin2 pathways than OA (Fig. 3B, C, and E). Because pancreatic δ-cells represent only a small proportion of islet cells, the measurement of second messengers or determination of receptor-arrestin interactions for biased characterization of GPCRs in pancreatic δ-cells is still very challenging and requires the development of new technology. In the current study, we used islets derived from β-arrestin1−/− or β-arrestin2−/− mice or specifically knocked-down Gq in pancreatic δ-cells to study the contribution of the functions of these effectors. We found that β-arrestin2 mediated SST/insulin secretion, the antistress response and the anti-inflammatory effects of several potentially endogenous GPR120 ligands in islets. The Gi signaling downstream of GPR120 may contribute to the SST secretion modulated by OA or LA. The combination of the application of GPR120-biased ligands, specific antagonists, and genetic knockdown all suggested that these phenotypes are very likely to be modulated directly by the GPR120–β-arrestin2 complex, which is similar to previous findings that the GPR120–β-arrestin2 complex mediated anti-inflammatory effects (15). However, our results do not exclude the possibility that other GPCR-arrestin interactions downstream of GPR120 may also be involved. Therefore, specific approaches, such as GPR120-arrestin chimeras or GPR120 ligands with exclusive bias properties, may help to address such questions in future research.

β-Arrestins are important downstream effectors that mediate a variety of GPCR functions (2426,37,6365). It has been reported that GPR120-β-arrestin2 signaling mediated the anti-inflammatory effects in macrophages by disrupting the TAB1-TAK1 complex and inhibiting the NLRP3 inflammasome (15,66). In adipocytes, GPR120 stimulation modulates peroxisome proliferator–activated receptor-γ phosphorylation via the association of β-arrestin2 with extracellular signal–regulated kinase (67). In addition, acceleration of wound healing by engagement of docosahexaenoic acid with GPR120 is dependent on β-arrestin2 signaling (68). In islets, it has been reported that β-arrestin1 is an important pancreatic δ-cell regulator in response to activation of the adrenergic system (32). β-Arrestin2 is reported to be required for β-cell mass plasticity and unlikely to be involved in the regulation of insulin secretion (69). However, another study showed reduced insulin secretion in β-arrestin2–deficient β-cells (70). We speculated that arrestin functions are highly dependent on the cellular context and specific agonist-receptor engagement because recent studies have suggested that receptor phospho-barcode and seven transmembrane bundle-mediated interactions are highly relevant to diverse arrestin functions (24,7173). In the current study, we found that β-arrestin2 mediated the hormone secretion, anti-inflammation, and antistress response effects of OA and LA using β-arrestin2–deficient mice. These studies collectively suggested that activation of GPR120–β-arrestin2 signaling is generally beneficial for metabolism. The functions and phosphoproteomics downstream of each GPCR-arrestin complex are different. Further in-depth studies of GPR120–β-arrestin2 signaling in pancreatic islets could facilitate the development of therapeutics targeting GPR120.

Notably, recent studies have reported that the GPR120 agonist Metabolex 36 could decrease SST secretion of δ-cells in a PTX-sensitive manner (10). However, another study showed that PTX preincubation had no effects on SST secretion inhibited by the GPR120 agonist cpdA (11). PTX concentrations were similar in these two works, but the GPR120 agonists were different. It is possible that Metabolex 36 and cpdA have different abilities to activate the Gi and arrestin pathways. In particular, our data suggest that β-arrestin2 and Gi signaling contribute to changes in SST secretion downstream of GPR120 activation using pharmacological characterization combined with β-arrestin2−/− mice. Future experiments characterizing the biased property between CpdA and Metabolex 36 and using Gi-specific knockout mouse models will help to clarify these discrepancies.

In addition, LA exhibited functional bias to modulate insulin/SST secretion compared with anti-inflammatory function, using OA as a reference ligand. There is an obvious functional separation window between OA and LA for insulin/SST secretion but not for the regulation of inflammatory gene expression. These results suggest that the different functions of GPR120 are connected to the bias of endogenous ligands.

Another significant finding was that endogenous GPR120 signaling circuits were downregulated within islets in the context of diabetes. In the human obese population, p.R270H nonsynonymous variants of GPR120 have been identified as a risk factor for obesity (9). In this study, we found that p.R270H in humans decreased its ability to sense the GPR120 ligands OA and LA. In particular, p.R270H was still able to respond to LA stimulation, despite requiring a higher concentration, suggesting therapeutic potential for LA or its synthetic analog. We further investigated whether the local GPR120 pathway in pancreatic islets is damaged in other diabetic models. We found that GPR120 circuits in islets were impaired in db/db diabetic model mice, with decreased GPR120 expression and decreased levels of the endogenous islet GPR120 ligands OA and LA. Treating islets isolated from db/db mice with LA or OA significantly improved insulin secretion and ameliorated inflammation as well as in the STZ diabetic model mice. Moreover, LA administration at a dose of 10 μg/g LA i.p. for 10 weeks was found to improve glucose metabolism in db/db mice. These results suggest that the endogenous GPR120 circuits are damaged in this diabetic model and that restoring the endogenous GPR120 pathways alleviates diabetic phenotypes in pancreatic islets.

In summary, we have revealed endogenous ligand-GPR120 circuits in pancreatic islets. The two identified potentially endogenous GPR120 ligands, OA and LA, exhibit different biases toward the Gq and β-arrestin2 pathways in modeled HEK293 cells, and β-arrestin2 signaling contributes to improved islet hormone secretion and islet homeostasis. The GPR120 circuits are downregulated in diabetic models, and restoring these circuits via exogenous lipid administration improves islet function. Our research revealed important endogenous FA-GPCR circuits that harbor different signaling biases that are connected to their functions in pancreatic islets.

Y.-Q.D., X.-Y.S, J.C., and J.W. contributed equally to this work.

This article contains supplementary material online at https://doi.org/10.2337/figshare.19566082.

Funding. Support was received from theNational Natural Science Foundation of China Grant (92057121 to X.Y., 32130055 to J.-P.S., and 31701230 to Z.Y.), the National Science Fund for Excellent Young Scholars Grant (81822008 to X.Y.), the National Key Basic Research Program of China Grant (2019YFA0904200 to J.-P.S., and 2018YFC1003600 to X.Y. and J.-P.S.), the Major Fundamental Research Program of Natural Science Foundation of Shandong Province, China (ZR2021ZD18 to X.Y. and ZR2020ZD39 to J.-P.S.), the National Science Fund for Distinguished Young Scholars Grant (81773704 to J.-P.S.), Innovative Research Team in University Grant (IRT_17R68 to X.Y.), and the Key Research Project of the Natural Science Foundation of Beijing, China (Z20J00124 to J.-P.S.).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. Y.-Q.D. contributed to identify the GPR120 agonists, OA and LA, and examine the function and signal transduction. Y.-Q.D., X.-Y.S., J.C., J.W., J.-P.S., and X.Y., participated in data analysis and interpretation. Y.-Q.D., X.-Y.S., J.C., J.W., J.-P.S., and X.Y. wrote the manuscript. Y.-Q.D., X.-Y.S., J.W., and W.P. isolated mice islets and performed ex vivo experiments. Y.-Q.D., X.-Y.S., and J.-Y.L. performed immunofluorescence studies. Y.-Q.D., J.-P.S., and X.Y. designed the islet culture and all other experiments. J.C. contributed to provide the db/db and db/+ mice and perform the RNAscope experiments. J.C. and L.-J.Z. tested the β-arrestins recruitment of p.R270H variants. J.W. contributed to provide the STZ-induced mice model. W.-T.A. contributed to provide the β-arrestin2−/− mice. X.-N.T. and P.X. performed preparation of GPR120 protein. X.-N.T. and P.X. performed preparation of GPR120 protein. Y.-F.X. provided the human islets. Y.-L.J. and M.L. provided insightful ideas. Z.Y. guided the BRET assay of β-arrestins recruitment and Gβγ-dissociation. J.-P.S. and X.Y. conceived and initiated the study of the GPR120 in islet circuits, as well as the biased signaling of GPR120 in pancreatic islets. J.-P.S. and X.Y. and supervised the overall project design and execution. All of the authors saw and commented on the manuscript. J.-P.S. and X.Y. are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

1.
De León
DD
,
Deng
S
,
Madani
R
,
Ahima
RS
,
Drucker
DJ
,
Stoffers
DA
.
Role of endogenous glucagon-like peptide-1 in islet regeneration after partial pancreatectomy
.
Diabetes
2003
;
52
:
365
371
2.
Salehi
M
,
Aulinger
B
,
Prigeon
RL
,
D’Alessio
DA
.
Effect of endogenous GLP-1 on insulin secretion in type 2 diabetes
.
Diabetes
2010
;
59
:
1330
1337
3.
Smith
EP
,
An
Z
,
Wagner
C
, et al
.
The role of β cell glucagon-like peptide-1 signaling in glucose regulation and response to diabetes drugs
.
Cell Metab
2014
;
19
:
1050
1057
4.
Molina
J
,
Rodriguez-Diaz
R
,
Fachado
A
,
Jacques-Silva
MC
,
Berggren
P-O
,
Caicedo
A
.
Control of insulin secretion by cholinergic signaling in the human pancreatic islet
.
Diabetes
2014
;
63
:
2714
2726
5.
Rodriguez-Diaz
R
,
Dando
R
,
Jacques-Silva
MC
, et al
.
Alpha cells secrete acetylcholine as a non-neuronal paracrine signal priming beta cell function in humans
.
Nat Med
2011
;
17
:
888
892
6.
van der Meulen
T
,
Donaldson
CJ
,
Cáceres
E
, et al
.
Urocortin3 mediates somatostatin-dependent negative feedback control of insulin secretion
.
Nat Med
2015
;
21
:
769
776
7.
Li
Q
,
Cui
M
,
Yang
F
, et al
.
A cullin 4B-RING E3 ligase complex fine-tunes pancreatic δ cell paracrine interactions
.
J Clin Invest
2017
;
127
:
2631
2646
8.
Taneera
J
,
Lang
S
,
Sharma
A
, et al
.
A systems genetics approach identifies genes and pathways for type 2 diabetes in human islets
.
Cell Metab
2012
;
16
:
122
134
9.
Ichimura
A
,
Hirasawa
A
,
Poulain-Godefroy
O
, et al
.
Dysfunction of lipid sensor GPR120 leads to obesity in both mouse and human
.
Nature
2012
;
483
:
350
354
10.
Stone
VM
,
Dhayal
S
,
Brocklehurst
KJ
, et al
.
GPR120 (FFAR4) is preferentially expressed in pancreatic delta cells and regulates somatostatin secretion from murine islets of Langerhans
.
Diabetologia
2014
;
57
:
1182
1191
11.
Croze
ML
,
Flisher
MF
,
Guillaume
A
, et al
.
Free fatty acid receptor 4 inhibitory signaling in delta cells regulates islet hormone secretion in mice
.
Mol Metab
2021
;
45
:
101166
12.
DiGruccio
MR
,
Mawla
AM
,
Donaldson
CJ
, 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
458
13.
Adriaenssens
AE
,
Svendsen
B
,
Lam
BY
, et al
.
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
2165
14.
Rorsman
P
,
Huising
MO
.
The somatostatin-secreting pancreatic δ-cell in health and disease
.
Nat Rev Endocrinol
2018
;
14
:
404
414
15.
Oh
DY
,
Talukdar
S
,
Bae
EJ
, et al
.
GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects
.
Cell
2010
;
142
:
687
698
16.
Yore
MM
,
Syed
I
,
Moraes-Vieira
PM
, et al
.
Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects
.
Cell
2014
;
159
:
318
332
17.
Ulven
T
,
Christiansen
E
.
Dietary fatty acids and their potential for controlling metabolic diseases through activation of FFA4/GPR120
.
Annu Rev Nutr
2015
;
35
:
239
263
18.
Christiansen
E
,
Watterson
KR
,
Stocker
CJ
, et al
.
Activity of dietary fatty acids on FFA1 and FFA4 and characterisation of pinolenic acid as a dual FFA1/FFA4 agonist with potential effect against metabolic diseases
.
Br J Nutr
2015
;
113
:
1677
1688
19.
Hirasawa
A
,
Tsumaya
K
,
Awaji
T
, et al
.
Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120
.
Nat Med
2005
;
11
:
90
94
20.
Milligan
G
,
Alvarez-Curto
E
,
Hudson
BD
,
Prihandoko
R
,
Tobin
AB
.
FFA4/GPR120: pharmacology and therapeutic opportunities
.
Trends Pharmacol Sci
2017
;
38
:
809
821
21.
Liu
HD
,
Wang
WB
,
Xu
ZG
, et al
.
FFA4 receptor (GPR120): a hot target for the development of anti-diabetic therapies
.
Eur J Pharmacol
2015
;
763
:
160
168
22.
Engelstoft
MS
,
Park
WM
,
Sakata
I
, et al
.
Seven transmembrane G protein-coupled receptor repertoire of gastric ghrelin cells
.
Mol Metab
2013
;
2
:
376
392
23.
Milligan
G
,
Shimpukade
B
,
Ulven
T
,
Hudson
BD
.
Complex Pharmacology of Free Fatty Acid Receptors
.
Chem Rev
2017
;
117
:
67
110
24.
Yang
F
,
Xiao
P
,
Qu
CX
, et al
.
Allosteric mechanisms underlie GPCR signaling to SH3-domain proteins through arrestin
.
Nat Chem Biol
2018
;
14
:
876
886
25.
Liu
C-H
,
Gong
Z
,
Liang
Z-L
, et al
.
Arrestin-biased AT1R agonism induces acute catecholamine secretion through TRPC3 coupling
.
Nat Commun
2017
;
8
:
14335
26.
Smith
JS
,
Lefkowitz
RJ
,
Rajagopal
S
.
Biased signalling: from simple switches to allosteric microprocessors
.
Nat Rev Drug Discov
2018
;
17
:
243
260
27.
Schmid
CL
,
Kennedy
NM
,
Ross
NC
, et al
.
Bias Factor and Therapeutic Window Correlate to Predict Safer Opioid Analgesics
.
Cell
2017
;
171
:
1165
1175.e13
28.
Manglik
A
,
Lin
H
,
Aryal
DK
, et al
.
Structure-based discovery of opioid analgesics with reduced side effects
.
Nature
2016
;
537
:
185
190
29.
Li
N
,
Yang
Z
,
Li
Q
, et al
.
Ablation of somatostatin cells leads to impaired pancreatic islet function and neonatal death in rodents
.
Cell Death Dis
2018
;
9
:
682
30.
Ning
SL
,
Zheng
WS
,
Su
J
, et al
.
Different downstream signalling of CCK1 receptors regulates distinct functions of CCK in pancreatic beta cells
.
Br J Pharmacol
2015
;
172
:
5050
5067
31.
Lin
J-Y
,
Cheng
J
,
Du
Y-Q
, et al
.
In vitro expansion of pancreatic islet clusters facilitated by hormones and chemicals
.
Cell Discov
2020
;
6
:
20
32.
Wang
H-M
,
Dong
J-H
,
Li
Q
, et al
.
A stress response pathway in mice upregulates somatostatin level and transcription in pancreatic delta cells through Gs and β-arrestin 1
.
Diabetologia
2014
;
57
:
1899
1910
33.
Bauer
PV
,
Duca
FA
,
Waise
TMZ
, et al
.
Lactobacillus gasseri in the upper small intestine impacts an ACSL3-dependent fatty acid-sensing pathway regulating whole-body glucose homeostasis
.
Cell Metab
2018
;
27
:
572
587.e6
34.
Olsen
RHJ
,
DiBerto
JF
,
English
JG
, et al
.
TRUPATH, an open-source biosensor platform for interrogating the GPCR transducerome
.
Nat Chem Biol
2020
;
16
:
841
849
35.
Hu
Q-X
,
Dong
J-H
,
Du
H-B
, et al
.
Constitutive Gαi coupling activity of very large G protein-coupled receptor 1 (VLGR1) and its regulation by PDZD7 protein
.
J Biol Chem
2014
;
289
:
24215
24225
36.
Rajagopal
S
,
Ahn
S
,
Rominger
DH
, et al
.
Quantifying ligand bias at seven-transmembrane receptors
.
Mol Pharmacol
2011
;
80
:
367
377
37.
Dong
JH
,
Wang
YJ
,
Cui
M
, et al
.
Adaptive activation of a stress response pathway improves learning and memory through Gs and β-arrestin-1-regulated lactate metabolism
.
Biol Psychiatry
2017
;
81
:
654
670
38.
Zhang
X
,
Zhang
Y
,
Wang
P
, et al
.
Adipocyte hypoxia-inducible factor 2α suppresses atherosclerosis by promoting adipose ceramide catabolism
.
Cell Metab
2019
;
30
:
937
951.e5
39.
Oballa
RM
,
Belair
L
,
Black
WC
, et al
.
Development of a liver-targeted stearoyl-CoA desaturase (SCD) inhibitor (MK-8245) to establish a therapeutic window for the treatment of diabetes and dyslipidemia
.
J Med Chem
2011
;
54
:
5082
5096
40.
Obukowicz
MG
,
Welsch
DJ
,
Salsgiver
WJ
, et al
.
Novel, selective delta6 or delta5 fatty acid desaturase inhibitors as antiinflammatory agents in mice
.
J Pharmacol Exp Ther
1998
;
287
:
157
166
41.
Yang
F
,
Yu
X
,
Liu
C
, et al
.
Phospho-selective mechanisms of arrestin conformations and functions revealed by unnatural amino acid incorporation and F-19-NMR
.
Nat Commun
2015
;
6
:
8202
42.
Li
T
,
Yu
B
,
Liu
Z
, et al
.
Homocysteine directly interacts and activates the angiotensin II type I receptor to aggravate vascular injury
.
Nat Commun
2018
;
9
:
11
43.
Liu
Q
,
He
Q-T
,
Lyu
X
, et al
.
DeSiphering receptor core-induced and ligand-dependent conformational changes in arrestin via genetic encoded trimethylsilyl 1H-NMR probe
.
Nat Commun
2020
;
11
:
4857
44.
Dou
H
,
Wang
C
,
Wu
X
, et al
.
Calcium influx activates adenylyl cyclase 8 for sustained insulin secretion in rat pancreatic beta cells
.
Diabetologia
2015
;
58
:
324
333
45.
Yang
F
,
Mao
C
,
Guo
L
, et al
.
Structural basis of GPBAR activation and bile acid recognition
.
Nature
2020
;
587
:
499
504
46.
Dohlman
HG
,
Campbell
SL
.
Regulation of large and small G proteins by ubiquitination
.
J Biol Chem
2019
;
294
:
18613
18623
47.
Hudson
BD
,
Shimpukade
B
,
Mackenzie
AE
, et al
.
The pharmacology of TUG-891, a potent and selective agonist of the free fatty acid receptor 4 (FFA4/GPR120), demonstrates both potential opportunity and possible challenges to therapeutic agonism
.
Mol Pharmacol
2013
;
84
:
710
725
48.
Suckow
AT
,
Polidori
D
,
Yan
W
, et al
.
Alteration of the glucagon axis in GPR120 (FFAR4) knockout mice: a role for GPR120 in glucagon secretion
.
J Biol Chem
2014
;
289
:
15751
15763
49.
Svendsen
B
,
Holst
JJ
.
Paracrine regulation of somatostatin secretion by insulin and glucagon in mouse pancreatic islets
.
Diabetologia
2021
;
64
:
142
151
50.
Bansal
P
,
Wang
Q
.
Insulin as a physiological modulator of glucagon secretion
.
Am J Physiol Endocrinol Metab
2008
;
295
:
E751
E761
51.
Cooperberg
BA
,
Cryer
PE
.
Insulin reciprocally regulates glucagon secretion in humans
.
Diabetes
2010
;
59
:
2936
2940
52.
Kawamori
D
,
Kurpad
AJ
,
Hu
J
, et al
.
Insulin signaling in alpha cells modulates glucagon secretion in vivo
.
Cell Metab
2009
;
9
:
350
361
53.
Donath
MY
,
Shoelson
SE
.
Type 2 diabetes as an inflammatory disease
.
Nat Rev Immunol
2011
;
11
:
98
107
54.
Robertson
RP
,
Harmon
J
,
Tran
POT
,
Poitout
V
.
Beta-cell glucose toxicity, lipotoxicity, and chronic oxidative stress in type 2 diabetes
.
Diabetes
2004
;
53
(
Suppl. 1
):
S119
S124
55.
Xie
K
,
Xu
B
,
Zhang
Y
, et al
.
A multi-method evaluation of the effects of Inflammatory cytokines (IL-1β, IFN-γ, TNF-α) on pancreatic β-cells
.
J Cell Physiol
2018
;
233
:
9375
9382
56.
Lakey
JRT
,
Suarez-Pinzon
WL
,
Strynadka
K
, et al
.
Peroxynitrite is a mediator of cytokine-induced destruction of human pancreatic islet beta cells
.
Lab Invest
2001
;
81
:
1683
1692
57.
Bonnefond
A
,
Lamri
A
,
Leloire
A
, et al
.
Contribution of the low-frequency, loss-of-function p.R270H mutation in FFAR4 (GPR120) to increased fasting plasma glucose levels
.
J Med Genet
2015
;
52
:
595
598
58.
Sun
F
,
Zeng
J
,
Jing
M
, et al
.
A genetically encoded fluorescent sensor enables rapid and specific detection of dopamine in flies, fish, and mice
.
Cell
2018
;
174
:
481
496.e19
59.
Wu
Z
,
He
K
,
Chen
Y
, et al
.
A sensitive GRAB sensor for detecting extracellular ATP in vitro and in vivo
.
Neuron
2022
;
110
:
770
782.e5
60.
Dong
A
,
He
K
,
Dudok
B
, et al
.
A fluorescent sensor for spatiotemporally resolved imaging of endocannabinoid dynamics in vivo
.
Nat Biotechnol
11 November 2021 [E-pub ahead of print] DOI: 10.1038/s41587-021-01074-4
61.
Saponaro
C
,
Gaggini
M
,
Carli
F
,
Gastaldelli
A
.
The subtle balance between lipolysis and lipogenesis: a critical point in metabolic homeostasis
.
Nutrients
2015
;
7
:
9453
9474
62.
Schwenk
RW
,
Holloway
GP
,
Luiken
JJ
,
Bonen
A
,
Glatz
JF
.
Fatty acid transport across the cell membrane: regulation by fatty acid transporters
.
Prostaglandins Leukot Essent Fatty Acids
2010
;
82
:
149
154
63.
Zhang
DL
,
Sun
YJ
,
Ma
ML
, et al
.
Gq activity- and β-arrestin-1 scaffolding-mediated ADGRG2/CFTR coupling are required for male fertility
.
eLife
2018
;
7
:
e33432
64.
Park
JY
,
Qu
CX
,
Li
RR
, et al
.
Structural mechanism of the arrestin-3/JNK3 interaction
.
Structure
2019
;
27
:
1162
1170.e3
65.
Liu
J
,
Li
QX
,
Wang
XJ
, et al
.
β-Arrestins promote podocyte injury by inhibition of autophagy in diabetic nephropathy
.
Cell Death Dis
2016
;
7
:
e2183
66.
Yan
Y
,
Jiang
W
,
Spinetti
T
, et al
.
Omega-3 fatty acids prevent inflammation and metabolic disorder through inhibition of NLRP3 inflammasome activation
.
Immunity
2013
;
38
:
1154
1163
67.
Paschoal
VA
,
Walenta
E
,
Talukdar
S
, et al
.
Positive reinforcing mechanisms between GPR120 and PPARγ modulate insulin sensitivity
.
Cell Metab
2020
;
31
:
1173
1188.e5
68.
Arantes
EL
,
Dragano
N
,
Ramalho
A
, et al
.
Topical docosahexaenoic acid (DHA) accelerates skin wound healing in rats and activates GPR120
.
Biol Res Nurs
2016
;
18
:
411
419
69.
Ravier
MA
,
Leduc
M
,
Richard
J
, et al
.
β-Arrestin2 plays a key role in the modulation of the pancreatic beta cell mass in mice
.
Diabetologia
2014
;
57
:
532
541
70.
Zhu
L
,
Almaça
J
,
Dadi
PK
, et al
.
β-arrestin-2 is an essential regulator of pancreatic β-cell function under physiological and pathophysiological conditions
.
Nat Commun
2017
;
8
:
14295
71.
Yang
F
,
Yu
X
,
Liu
C
, et al
.
Phospho-selective mechanisms of arrestin conformations and functions revealed by unnatural amino acid incorporation and (19)F-NMR
.
Nat Commun
2015
;
6
:
8202
72.
He
QT
,
Xiao
P
,
Huang
SM
, et al
.
Structural studies of phosphorylation-dependent interactions between the V2R receptor and arrestin-2
.
Nat Commun
2021
;
12
:
2396
73.
Nobles
KN
,
Xiao
K
,
Ahn
S
, et al
.
Distinct phosphorylation sites on the β(2)-adrenergic receptor establish a barcode that encodes differential functions of β-arrestin
.
Sci Signal
2011
;
4
:
ra51
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.