191
Views
19
CrossRef citations to date
0
Altmetric
Review

Potential roles of GPR120 and its agonists in the management of diabetes

&
Pages 1013-1027 | Published online: 29 Jul 2014

Abstract

Free fatty acids (FFAs) serve not only as nutrients that provide energy but also as extracellular signaling molecules that manipulate intracellular physiological events through FFA receptors (FFARs) such as FFAR4. FFAR4 is also known as G-protein coupled receptor 120 (GPR120). The main role of GPR120 is to elicit FFA regulation on metabolism homeostasis. GPR120 agonism correlates with prevention of the occurrence and development of metabolic disorders such as obesity and diabetes. GPR120 activation directly or indirectly inhibits inflammation, modulates hormone secretion from the gastrointestinal tract and pancreas, and regulates lipid and/or glucose metabolism in adipose, liver, and muscle tissues, which may help prevent obesity and diabetes. This review summarizes recent advances in physiological roles of GPR120 in preventing insulin resistance and protecting pancreatic islet function, and examines how resident GPR120 in the pancreas may be involved in modulating pancreatic islet function.

Introduction

Metabolic diseases, such as obesity and type 2 diabetes mellitus (T2DM), are ever increasing in prevalence.Citation1,Citation2 Hence, researchers are working to develop pharmaceutical treatments that can alleviate these conditions. In this regard, G-protein coupled receptor 120 (GPR120), which plays important roles in regulating energy metabolism and eliciting anti-inflammatory effects, is an attractive target for diabetes treatments, in particular.

Binding of GPR120 by ω-3 fatty acids (ω-3 FAs) results in various physiological activities that have a stabilizing effect on metabolic homeostasis and may help prevent diabetes.Citation3,Citation4 Patients with T2DM exhibit insufficient pancreatic insulin secretion and insulin resistance in adipose, liver, and muscle; insulin resistance correlates with abnormal energy metabolism, such as a sequela of hepatic steatosis.Citation5 GPR120 activation relieves insulin resistance by enabling adipogenesis in adipose tissue and thereby maintaining a lipid metabolism balance between adipose and liver tissues.Citation3,Citation4,Citation6 Additionally, GPR120 stimulation can also support maintenance of insulin sensitivity through inhibition of inflammation, since GPR120 plays an important role in mediating the anti-inflammatory effects of ω-3 FAs on macrophages.Citation3,Citation4,Citation7Citation10 GPR120 activation may also promote pancreatic β-cell survival and proliferation and stimulate pancreatic insulin secretion indirectly through induction of glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK) secretion from the intestine.Citation11Citation14 Generally speaking, GPR120-mediated modulation on insulin sensitivity, pancreatic insulin secretion, and β-cell mass suggests that GPR120 could be targeted as an anti-diabetic treatment. However, it is not known yet whether pancreatic GPR120 has a role in direct modulatory effects on pancreatic and islet function.

In this review, we first describe the structure, tissue distribution, and agonists of GPR120. We then summarize advances in research on the roles of GPR120 in modulating physiological activities in the gastrointestinal tract and tongue, and in influencing the cellular activities of adipocytes and macrophages. Next, we discuss how GPR120 activation may maintain insulin sensitivity and indirectly promote pancreatic islet function. Finally, in light of evidence suggesting that pancreatic GPR120 stimulation might regulate islet function directly,Citation15,Citation16 we examine the potential mechanism by which local pancreatic GPR120 may modulate islet function.

Structure and distribution of GPR120 Structure

Human GPR120 has two splice variants, a short isoform (NM_181745) and a long isoform (BC101175) with 16 additional amino acids in the third intracellular loop.Citation17 Both isoforms are phosphorylated similarly in the presence of ω-3 FAs, and both recruit β-arrestin-2 readily and undergo internalization,Citation17,Citation18 while the longer intracellular loop of the long isoform prevents G-protein-dependent intracellular calcium mobilization.Citation18 Rat, mouse, and cynomolgus monkey have a GPR120 genetic sequence comprised of a single 1,086 nucleotide sequence encoding a protein that is 361 amino acids long.Citation17,Citation19 Not surprisingly, the cynomolgus monkey’s complementary DNA (cDNA) for GPR120 has higher homology with human GPR120 long isoform than does the mouse or rat genes.Citation19

Similar to other G protein-coupled receptors (GPCRs), GPR120 contains an extracellular N-terminal domain, seven transmembrane domains, and an intracellular C-terminal domain.Citation13,Citation18,Citation20,Citation21 Both N-terminal and transmembrane domains may mediate interaction between GPCRs and their ligands. It has been identified that Arg99 in the transmembrane domain II of GPR120 is an essential residue for binding GPR120 to its ligands.Citation20,Citation22,Citation23 The intracellular C-terminal region of GPR120 is important for its interaction with G-protein or scaffold protein, such as β-arrestin-2. It has been demonstrated that there are several putative β-arrestin-2-binding motifs in the C-terminal domain of GPR120.Citation24 It is noteworthy that the C-terminal fusion of tag or protein (such as hemagglutinin, enhanced yellow fluorescent protein, and Gα16) does not impair the biological function of GPR120.Citation11,Citation20,Citation25

Until now, the spatial structure of GPR120 has not been identified.Citation23 To develop selective GPR120 agonists, researchers have thus constructed GPR120 homology model by utilizing the crystal structure of other GPCRs, such as the β2-adrenoceptor and bovine rhodopsin.Citation20,Citation22,Citation23

Distribution

Expression of GPR120 differs between species (). In the rat, the highest expression of GPR120 was found in the colon, whereas in mouse and human, the highest GPR120 expression is found in the lung.Citation11,Citation13 Beyond the lung, GPR120 is expressed mainly in tissues and cells related to energy metabolism, such as the gastrointestinal tract, tongue, and adipose tissue. Various macrophages also express high levels of GPR120.

Table 1 Tissue distribution of GPR120 in human, mouse, rat, and cynomolgus monkey

GPR120 expression has been confirmed in mouse intestinal endocrine cell lines (STC-1 and GLUTag), mouse intestinal glucose-dependent insulinotropic polypeptide (GIP)-secreting K-cells and human colorectal carcinoma tissue.Citation11,Citation26Citation28 GPR120 colocalizes with GLP-1 in mouse large intestine epithelial cells and human colonic intraepithelial neuroendocrine cells.Citation11,Citation29 GPR120 expression is also detected in mouse gastric ghrelinoma cell lines (MGN3-1 and SG-1), and it colocalizes with ghrelin in stomach ghrelin-secreting cells.Citation30Citation32 It is also expressed in taste bud cells (TBCs) of circumvallate, fungiform, and foliate papillae in rat, mouse, and human.Citation33Citation38 GPR120 has been reported to be co-expressed with phospholipase-Cβ2 (PLCβ2), α-gustducin, transient receptor potential channel type M5 (TRPM5), and GLP-1 in mouse TBCs of circumvallate papillae, indicating that it is mainly in type II TBCs.Citation34Citation36 In addition, GPR120 also colocalizes with cluster of differentiation 36 (CD36) in mouse and human TBCs.Citation36,Citation37 Interestingly, Ozdener et al showed that human GPR120 and CD36 have non-overlapping roles in TBC signaling during oro-gustatory perception of dietary lipids.Citation37

GPR120 messenger RNA (mRNA) has been detected in mouse islets, the murine pancreatic β-cell lines (MIN6, INS-1 and BRIN-BD11), and human pancreas and islets.Citation15,Citation16,Citation39,Citation40 However, others have reported no, or negligible, expression of GPR120 in MIN6 cells and rat pancreas.Citation11,Citation13 Thus, GPR120 expression in murine pancreatic islets and β-cells needs to be further confirmed. Negligible GPR120 expression has been detected in mouse soleus and extensor digitorum longus (EDL) skeletal muscle,Citation3 but GPR120 mRNA levels in cardiac muscle and EDL skeletal muscle of Sprague Dawley rats can be increased by feeding with a high fat diet (HFD).Citation41 GPR120 is also expressed in various mouse white adipose tissues, including subcutaneous, perineal, mesenteric and epididymal adipose, and GPR120 mRNA expression in mouse adipose tissues can also be upregulated with HFD feeding.Citation40 In addition, it has been shown that the differentiated human adipocytes have greater GPR120 mRNA levels than preadipocytes.Citation40 These findings suggest that expression level of GPR120 is associated with lipid metabolism and adipogenesis.

GPR120 expression is also identified in macrophages, such as mouse bone marrow-derived macrophages and intraperitoneal macrophages.Citation3 In liver, GPR120 expression is limited to resident macrophages (Kupffer cells [KCs]) and its expression can be increased significantly in macrophages of adipose and liver by HFD-feeding in mice.Citation3,Citation9 summarizes the tissue distribution of GPR120 in various species.

GPR120 signaling

In gastrointestinal cells, TBCs, and adipocytes, agonist-stimulated GPR120 interacts with Gαq/11, a heterotrimeric G-protein,Citation14,Citation37 leading to PLC activation and subsequent elevation in intracellular Ca2+ concentration ([Ca2+]i). Not only can the Ca2+ mobilization promote GLP-1 and/or CCK secretion from gastrointestinal cells,Citation11,Citation13,Citation14 it can also activate phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), which further induces glucose transporter 4 (GLUT4) translocation to cell membranes in adipocytes,Citation3 provokes tumor progression of colorectal carcinoma (CRC) cells,Citation27 inhibits gastric ghrelin secretion,Citation31,Citation32 and elicits an anti-apoptotic effect in STC-1.Citation42 The mechanism(s) by which GPR120 stimulation regulates these physiological and pathological events is summarized in .

Figure 1 Schematic overview of the potential mechanism by which GPR120-Gαq/11 signaling may affect various physiological and pathological processes.

Notes: Binding of ω-3 FA-activated GPR120 to Gαq/11 stimulates PLC, leading to an increase in intracellular Ca2+ concentration ([Ca2+]i), which may influence diverse physiological processes, such as CCK, GLP-1, and ghrelin secretion, glucose uptake, tumor progression, and apoptosis. aCCK and/or GLP-1 secretion in taste bud cells, intestinal tract and STC-1 cells. bApoptosis of STC-1 cells. cGhrelin secretion of stomach. dGlucose uptake of adipocytes. eTumor progression of colorectal carcinoma.
Abbreviations: ω-3 FAs, ω-3 fatty acids; Akt, protein kinase B; CCK, cholecystokinin; COX-2, cyclooxygenase 2; DAG, diacylglycerol; ERK, extracellular-signal-regulated kinase; FA, fatty acid; Gαq/11, heterotrimeric G-protein; GLP-1, glucagon-like peptide 1; GLUT4, glucose transporter 4; GPR120, G-protein coupled receptor 120; IKK, IκB kinase; IL-8, interleukin 8; NFκB, nuclear factor kappa B; PGE2, prostaglandin E2; PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; PLC, phospholipase-C; PKC, protein kinase C; VEGF, vascular endothelial growth factor; STC-1, mouse intestinal enteroendocrine cell line; Ca2+, intracellular calcium level.
Figure 1 Schematic overview of the potential mechanism by which GPR120-Gαq/11 signaling may affect various physiological and pathological processes.

When stimulated by extracellular ω-3 FA ligands, GPR120 on the cell surface can be phosphorylated by kinases, leading to the increase in affinity of GPR120 intracellular domain to scaffold protein β-arrestin-2. Once GPR120 is bound to β-arrestin-2, the G-protein coupling to GPR120 is prevented, and then GPR120-β-arrestin-2 complex is internalized to the cytoplasm where downstream signaling can proceed.Citation3,Citation18 Thus far, all anti-inflammatory effects of GPR120 ligands on macrophages appear to be mediated by GPR120–β-arrestin-2 signaling.Citation3,Citation7,Citation8 The activation of this signaling can inhibit production of pro-inflammatory factors – such as monocyte chemotactic protein-1 (MCP-1), cyclooxygenase-2 (COX-2), interleukin (IL)-18, and IL-1β – by binding TAB1 (transforming growth factor-β activated kinase 1 [TAK1]-binding protein 1) or an inflammasome known as nucleotide-binding oligomerization leucine-rich repeat and pyrin domain-containing protein (NLRP)3/1b.Citation3,Citation7 summarizes the mechanism by which GPR120 activation elicits anti-inflammatory effects in various macrophages and adipocytes.

Figure 2 Schematic overview of the GPR120-β-arrestin-2 signaling pathway mediating anti-inflammatory effects.

Notes: ω-3 FA-activated GPR120 binds β-arrestin-2, and the resultant complex can interact with TAB1 or NLRP3, leading to inhibition of downstream inflammatory signals, which produce insulin resistance. aHHcy in adipose tissue. bInsulin resistance in liver and adipose tissue.
Abbreviations: ω-3 FAs, ω-3 fatty acids; Akt, protein kinase B; COX-2, cyclooxygenase 2; ER, endoplasmic reticulum; GPR120, G-protein coupled receptor 120; GSIS, glucose-stimulated insulin secretion; HHcy, hyperhomocysteinemia; IKK, IκB kinase; IL, interleukin; iNOS, inducible nitric oxide synthase; IRI, ischemia-reperfusion injury; IRS, insulin receptor substrate; JNK, c-Jun N-terminal kinase; NLRP3, nucleotide-binding oligomerization leucine-rich repeat and pyrin domains-containing protein 3; MCP-1, monocyte chemotactic protein-1; mPGES-1, microsomal prostaglandin E synthase-1; PGE2, prostaglandin E2; TAB1, TAK1-binding protein 1; TAK1, transforming growth factor-β activated kinase 1; TLR2/3/4, toll-like receptor 2/3/4; TNF-α, tumor necrosis factor α.
Figure 2 Schematic overview of the GPR120-β-arrestin-2 signaling pathway mediating anti-inflammatory effects.

GPR120 agonists

Natural agonists

Unsaturated free fatty acids (FFAs) with a chain length of 16–22 carbons and saturated FFAs of 14–18 carbons have been identified as ligands of GPR120, and the carboxylate anion of these FFAs is indispensable for their activity at GPR120.Citation11 Upon binding, these FFAs induce internalization of GPR120 into the cytoplasm (as demonstrated by a fluorescent protein marker), provoke an elevation of [Ca2+]i in HEK293 cells and enhance survival of serum-starved STC-1 cells.Citation11,Citation42

ω-3, ω-6 and ω-9 FAs belong to unsaturated FFAs, some members of these FAs are GPR120 ligands. ω-3 FAs appear to be more potent than ω-6 and ω-9 FAs in stimulating GPR120. The ω-3 FAs docosahexaenoic acid (DHA) and α-linolenic acid (ALA) are the most potent and most common GPR120 agonists,Citation11 but their selectivity is limited in that they interact with GPR40 as well.Citation22 On the other hand, unlike ω-3 FAs, ω-6 and ω-9 FAs have failed to exert anti-inflammatory effects through GPR120 signaling in human macrophage THP-1 cells,Citation7 and ω-6 FAs also have failed to attenuate COX-2 expression in RAW 264.7 cells.Citation8

In terms of pharmaceutical application, it would be desirable to find ligands with selective binding of GPR120 only. Grifolin derivatives (ie, grifolic acid and grifolic acid methyl ether) activate GPR120 selectively, but they are less efficacious than ALA and can inhibit the ALA-induced [Ca2+]I response and extracellular-signal-regulated kinase (ERK) phosphorylation. Thus, they are classified as partial GPR120 agonists with high selectivity.Citation43 Similar to the situation with other natural GPR120 ligands,Citation11 carboxyl groups in grifolin derivatives are necessary for GPR120 stimulation.Citation43

Synthetic agonists

To develop selective agonists of GPR120 with high potency, homology model of GPR120 was constructed by utilizing the crystal structure of other GPCRs;Citation20,Citation22,Citation23 in this regard, the hydrogen bonds between the carboxylic acid residue of ligands and the guanidine of Arg99 in GPR120 have been found to be essential for the activity of ligands at GPR120.

The first synthetic ligand of GPR120, GW9508, was identified by Briscoe et al.Citation44 However, GW9508 is not selective for GPR120, which restricts its application. NCG21, a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist derivative, shows high (albeit incomplete) selectivity for GPR120 over GPR40.Citation22 To investigate the roles of GPR120 in physiological processes, GW9508 and NCG21 can be used with cell lines and tissues that express GPR120 but not GPR40, such as RAW 264.7 cells.Citation3 Knockdown or knockout of GPR120 can be performed to further verify the involvement of this receptor in biological responses induced by its agonists.Citation11 GSK137647A, which can stimulate human, mouse, and rat GPR120, has been used to study the role of GPR120 in regulating GLP-1 secretion from mouse TBCs, although the selectivity of this agonist is not clear.Citation36

Based on structure-activity relationship experiments, Shimpukade et alCitation20 identified TUG-891 [3-(4-((4-fluoro-49-methyl-[1,19-biphenyl]-2-yl)methoxy)phenyl) propanoic acid] as a GPR120 agonist. This compound has high potency on both human and mouse GPR120 and is a more selective and potent agonist for human GPR120 than ALA, GW9508, or NCG21.Citation25 However, the selectivity of TUG-891 for mouse GPR120 is still limited, and the potency and selectivity of TUG-891 for rat GPR120 is unknown. Hence, the potential utility of TUG-891 in animal studies may be restricted.

Differences between natural and synthetic agonists of GPR120

In most cases, natural ligands of FFAs, but not synthetic agonists, are chosen to perform ex vivo and in vivo studies to investigate the role of GPR120 in physiological processes. FFAs can activate GPR120 to exhibit their biological activities in cells, tissues, and animals.Citation3,Citation4 It is unclear whether the potency of natural and synthetic ligands in GPR120 stimulation is different in tissues and/or animals. Nevertheless, it has been reported that the potency of FFAs and synthetic agonists to activate GPR40 is not identical; in MIN6 cells and mouse islets, FFAs can activate GPR40 to enhance glucose-stimulated insulin secretion, while GW9805 only exhibits its stimulatory effect on glucose-stimulated insulin secretion in MIN6 cells and not in mouse islets.Citation44

When using FFAs to study roles of GPR120, it is necessary to manifest the biological responses that are specific for GPR120. As nutrients, FFAs exert their bioactivities through several potential mechanisms. Besides GPCRs, metabolites of FFAs and other FFA-binding proteins, such as long-chain acyl-CoA and PPAR-γ, can also manipulate physiological processes.Citation45Citation47 In contrast, most of the synthetic agonists of GPR120 merely activate GPR120 and GPR40. In view of this, it would be better to select synthetic ligands in order to avoid the interference that could be caused by other factors.

Compared with synthetic ligands, FFAs have other disadvantages. Due to their poor solubility in water, FFAs are bound to the carrier (ie, fatty acid-free bovine serum albumin [BSA]).Citation45,Citation48,Citation49 On the other hand, BSA blunts the potency of FFAs to activate GPCRs, as it can influence interaction between FFAs and GPCRs.Citation11,Citation50,Citation51

Role of GPR120 in regulating cell and tissue functions

Intestinal cells and tissues

Incretin secretion

GLP-1 and CCK are intestinal incretins; that is, proteins that stimulate pancreatic insulin secretion.Citation52Citation54 GPR120 activation can induce GLP-1 and CCK secretion, which may further regulate pancreatic islet function and β-cell proliferation and survival.

The GPR120 agonists ALA or TUG-891 induce GLP-1 secretion in murine enteroendocrine cell lines (STC-1 and GLUTag) via stimulating GPR120.Citation11,Citation13,Citation14,Citation25 ALA administration has also been shown to increase plasma GLP-1 levels in mice and rats, and FFA supplementation or incubation increases mouse plasma CCK levels as well as CCK secretion of STC-1 cells.Citation11Citation14 This stimulatory effect of FFAs on GLP-1 and CCK secretion is dependent on a GPR120-mediated rise in [Ca2+]i.Citation14 FFA-stimulated GPR120 interacts with Gαq/11, leading to activation of PLCβ and generation of inositol trisphosphate, which in turn causes intracellular calcium release from the endoplasmic reticulum (). This calcium release activates TRPM5, which elicits membrane depolarization via cation influx. The resultant depolarization induces opening of voltage-gated calcium channels, allowing calcium influx, which then promotes secretion of GLP-1 and CCK.Citation14

Thus far, there has been no direct evidence confirming whether GPR40, which can also be stimulated by FFAs,Citation11 is involved in FFA-induced CCK and GLP-1 secretion in STC-1 cells. However, GPR40 agonists (ie, AM-1638 and AM-6226) can stimulate GLP-1 secretion from primary rat intestinal cells.Citation55 It may be that FFAs have a preference for GPR120 over GPR40 on STC-1 cells in the modulation of GLP-1 and CCK secretion.

Long-term ALA supplementation has been shown to promote pancreatic insulin secretion and β-cell proliferation in rats.Citation13 This finding is highly encouraging, since it suggests that GPR120 may be harnessed for anti-diabetic activities via not only modulation of insulin secretion but also via an effect on β-cell mass. It has been suggested that ALA effects on insulin secretion and β-cell proliferation may involve an indirect effect through GPR120 stimulation of GLP-1 and CCK secretion.Citation13 However, it is still controversial whether GPR120 is expressed in pancreatic islets/β-cells of mouse and rat.Citation16 If GPR120 is expressed in rodent islets, it would raise the possibility of these effects being realized through a direct mechanism.

High levels of GPR120 mRNA were detected in primary murine K-cells, and incubation of K-cells with linoleic acid (LA) can increase GIP secretion significantly, suggesting that GPR120 may be involved in the induction of GIP secretion.Citation26 GIP is an intestinal K-cell-secreting incretin hormone with anti-apoptotic effects on pancreatic β-cells.Citation26 Thus, it is possible that intestinal GPR120 may mediate an anti-diabetic (anti-apoptotic) effect on β-cells by promoting GIP secretion.

Anti-apoptosis

GPR120 was implicated in LA-induced anti-apoptotic effects in STC-1 cells: LA can enhance cell survival and inhibit caspase-3 activity and DNA fragmentation in nutrient-deprived STC-1 cells. Interestingly, in STC-1 cells, transient transfection with GPR120 cDNA enhances the inhibitory effect of LA on caspase-3 inhibition, but knockdown of GPR120 attenuates LA effect.Citation42 The potential mechanism is that LA binding to GPR120 leads to activation of two independent pathways, ERK and PI3K-Akt (also known as protein kinase B) signaling, which in turn may mediate anti-apoptotic effects ().Citation42

The ERK pathway is associated with growth and differentiation,Citation56,Citation57 whereas Akt signaling is involved in cell survival and proliferation.Citation58 Unsaturated FFAs have been shown to promote proliferation, enhance cell survival, and inhibit apoptosis via activation of ERK and/or PI3K-Akt in two GPR120-expressing human breast cancer cell lines, namely MDA-MB-231 cells and MCF-7 cells.Citation42,Citation59,Citation60 Pancreatic β-cells also undergo apoptosis during the development of diabetes,Citation61,Citation62 and activation of ERK and Akt can protect against β-cell apoptosis and increase insulin secretion.Citation63,Citation64 Therefore, it is possible that activation of pancreatic GPR120 may mediate the anti-apoptotic effects of FFAs in diabetic-islet β-cells via induction of ERK and Akt phosphorylation.

Angiogenesis

In human CRC cells and tissues, GPR120 acts as a tumor-promoting factor that augments angiogenesis and migratory capacity.Citation27 It has been shown that, in human CRC cells and tissues, GPR120 expression is highly inducible and correlates strongly with CRC differentiation and clinical progression.Citation27 Moreover, in human CRC cell lines (HCT116, SW480) and in an HCT116 xenograft mouse model, GPR120 activation upregulates expression and/or secretion of several pro-angiogenic factors, namely vascular endothelial growth factor, IL-8, prostaglandin E2 (PGE2) and COX-2. In addition, GPR120 stimulation can also enhance CRC motility and promote epithelial-mesenchymal transition of CRCs. The role of GPR120 in the pro-angiogenic influence has been attributed to GPR120 binding of Gαq/11, leading to a rise in [Ca2+]i and subsequent activation of the PI3K/Akt pathway, which favors activation of IκB kinase (IKK)/nuclear factor (NF)-κB signaling, and subsequent induction of pro-angiogenic factors ().Citation27

It is noteworthy that, although GPR120 stimulation in human CRCs enhance their migratory ability and increase their expression of COX-2, activation of GPR120 in intraperitoneal macrophages reduce macrophage motility and COX-2 expression.Citation3 These contrasting effects may be due to differential downstream signaling responses being triggered in the different cell lines. In macrophages (RAW 264.7 cell), GPR120 binds β-arrestin-2, leading to inhibition of IKK/NF-κB and c-Jun N-terminal kinase (JNK)/c-Jun pathways. On the other hand, in CRC cells, stimulated GPR120 binds Gαq/11, which consequently activates the IKK/NF-κB pathway. Hence, GPR120 linkage to intracellular signaling mechanism may be cell-type specific.

Stomach

Ghrelin secretion

It has been shown that GPR120, as a receptor of FFAs, is involved in inhibition of secretion of the gastric hormone ghrelin.Citation31,Citation32 FFAs have been shown to inhibit ghrelin secretion both in vitro (transgenic cells in which ghrelin carries a fluorescent marker) and in vivo (serum ghrelin levels of mice with pylorus ligation).Citation31 In addition, Gong et alCitation32 demonstrated that GPR120 stimulation by GW9508 or ALA reduces intracellular acyl-ghrelin content, inhibits acyl-ghrelin secretion in stomach ghrelinoma cell line (SG-1), and decreases fasting-induced plasma acyl-ghrelin levels of mice. These inhibitory effects have been attributed to GPR120 binding of Gαq/11, leading to phosphorylation of ERK via PLC, which may reduce expression of prohormone convertase 1 (the enzyme that converts proghrelin into ghrelin, which upon octanoylation by ghrelin O-acyltransferase [GOAT] becomes acyl-ghrelin) (). However, in another ghrelin-producing cell line (MGN3-1), GPR120 was shown to be uninvolved in the inhibitory effect of ALA on ghrelin secretion.Citation30 Thus, it seems that GPR120 involvement in modulating ghrelin secretion is influenced by cell type, the ligand used, or duration of treatment.

The hormone ghrelin is secreted primarily by stomach cells, and to a far lesser extent by pancreatic cells.Citation65 It stimulates hunger, appetite, and weight gain.Citation66Citation68 Similar to GLP-1, CCK, and insulin, ghrelin can modulate glucose homeostasis levels, and ghrelin levels can be altered dramatically by food intake. Hence, GPR120 activation could regulate metabolic homeostasis through modulation of stomach ghrelin secretion.Citation68 Moreover, it has been reported that blockade of pancreatic ghrelin secretion can increase insulin secretion.Citation69 Given the inhibitory effects of GPR120 activation on stomach ghrelin secretion,Citation31,Citation32 it is possible that GPR120 stimulation may promote insulin secretion by repressing pancreatic ghrelin secretion.

Taste buds

Taste preference and perception

In human subjects, individuals’ overall preference for fatty foods correlates with percentage body fat. That is, relative to lean individuals, obese individuals show, in general, a greater preference for fatty foods.Citation70,Citation71 GPR120 is involved in regulating spontaneous preference for fat content in one’s diet. Compared with wild-type controls, GPR120 knockout mice exhibit a lower oro-gustatory preference for FFAs and reduced taste nerve responses to several FFAs.Citation35 Researchers have speculated that decreased GPR120 expression may account for diminished responsiveness to an FFA-rich diet in ghrelin knockout mice and GOAT knockout mice.Citation72

Taste perception plays a crucial role in modulating food preference and energy homeostasis. GLP-1 signaling can enhance sweet and lipid detection sensitivity.Citation36,Citation73 It has been recently reported that lipids can induce GLP-1 secretion in mouse circumvallate papillae and, consequently, regulate taste perception threshold for sweets and lipids via activation of GPR120.Citation36 FFAs can also induce GLP-1 secretion to modulate fat preference through stimulation of GPR120 in human TBCsCitation37 through a mechanism that is similar to that which occurs in STC-1 cells ().Citation14 Differential perception sensitivity for lipids or sweets may affect interest in fat- or carbohydrate-rich food, which may contribute to fat intake and, consequently, affect risk for obesity.Citation74,Citation75 According to this model, GPR120-mediated modulation of taste perception in TBCs may alter an individual’s susceptibility to obesity. GPR120 activation may increase intake of high-lipid/high-sugar foods by upregulating GLP-1-induced of fat and sweet taste sensitivity in TBCs. However, given the important role of other long-chain fatty acid receptors (ie, GPR40 and CD36) in fat taste preference, it seems that GPR120 activation may not be the primary event in fat taste preference signaling.Citation76,Citation77 Thus, we cannot predict that upregulation or activation of GPR120 in TBCs would produce increased fat/carbohydrate intake.

Adipose tissues

Adipogenesis

GPR120 plays an important role in modulating adipose tissue development and adipocyte differentiation. Liu et alCitation78 and Oh et alCitation3 have shown that differentiation of 3T3-L1 cells is associated with increased levels of GPR120 mRNA. Gotoh et alCitation40 has demonstrated that GPR120 expression in human differentiated adipocytes is much higher than that in human preadipocytes. Ichimura et alCitation4 has shown that adipogenesis is suppressed in mouse embryonic fibroblast-derived adipocytes from GPR120 knockout mice.

During differentiation of 3T3-L1 cells, mRNA levels of GPR120 are influenced by regulation of PPAR-γ2, a crucial transcription factor in adipose tissue differentiation.Citation40 In addition, GPR120 knockdown was found to decrease mRNA levels of PPAR-γ2.Citation40 Moreover, Ichimura et alCitation4 has demonstrated in adipocytes derived from mouse embryonic fibroblasts of GPR120 knockout mice, adipocyte differentiation marker genes (fatty acid binding protein 4, PPAR-γ, and sterol regulatory element binding protein 1) are reduced. It seems GPR120 may interact with these adipogenesis-related proteins, especially PPAR-γ2, during adipocyte differentiation, although, if so, the specific mechanism has not been clarified. It is noteworthy that, besides its role in differentiation, PPAR-γ is also an important modulator of pancreatic β-cell function, glycometabolism, and lipid metabolism.Citation79 Given the interactions between GPR120 and PPAR-γ in adipose, it would be of interest to investigate whether GPR120 can regulate islet function and energy metabolism via interaction with PPAR-γ in other tissues, such as pancreas and liver.

Glucose uptake

GPR120 is involved in glucose metabolism in adipocytes and adipose tissue via regulation of GLUT4.Citation3,Citation78 Oh et alCitation3 has shown in 3T3-L1 adipocytes and primary adipose tissue the stimulated GPR120 binds Gαq/11, leading to activation of the PI3K/Akt pathway, which in turn causes GLUT4 translocation to the cell membrane and, finally, triggers glucose uptake (). Interestingly, GPR120 knockdown can reduce expression of GLUT4 and insulin receptor substrate 1 (IRS1),Citation78 suggesting that GPR120 dysfunction may impair glucose uptake in adipose tissue.

Gαq/11 and IRS1 play crucial, although independent, roles in the activation of the PI3K/Akt/GLUT4 pathway.Citation3 IRS1 transmits extracellular insulin signals to the cell interior, producing intracellular responses that alter glucose metabolism. Based on the observation that DHA did not stimulate IRS1 phosphorylation, Oh et alCitation3 speculated that the stimulatory effect of DHA-induced GPR120 activation on GLUT4 translocation in 3T3-L1 adipocytes must be occurring downstream of IRS1. However, Liu et alCitation78 has shown that IRS1 expression can be decreased by down-regulation of GPR120 expression in differentiated 3T3-L1 adipocytes. Hence, although normal GPR120 stimulation may not activate IRS1 to induce GLUT4 translocation, dysfunctional GPR120 can result in a downregulation of IRS1 expression and, thus, repression of insulin signaling, preventing normal induction of GLUT4-regulated glucose uptake.

Macrophages

Anti-inflammation

GPR120 activation has been shown to produce anti-inflammatory effects in macrophages (RAW 264.7 cells) by repressing activation of IKKβ/NF-κB and JNK/c-Jun signaling, which is induced by toll-like receptor 4 (TLR4) activation or TNF (tumor necrosis factor)-α. This repression can inhibit downstream pro-inflammatory cascades, including induction of MCP-1, TNF-α, IL-6, COX-2, PGE2, and microsomal PGE synthase-1 (mPGES-1).Citation3,Citation8 The mechanism for this anti-inflammatory effect involves DHA-activated GPR120 binding to scaffolding protein β-arrestin-2; GPR120–β-arrestin-2 complex is then internalized into the cytoplasm and binds TAB1, which in turn blocks TAB1/TAK1 association, induction of downstream pro-inflammatory signaling, and generation of inflammatory products ().Citation3

It has been demonstrated, in primary rat adipocytes, that GPR120 activation can reverse hyperhomocysteinemia (HHcy)-induced insulin resistance by inhibiting inflammation signaling.Citation10 HHcy is a condition characterized by abnormally high levels of homocysteine, which correlates with insulin resistance and can elevate risk of death in T2DM.Citation80Citation82 HHcy-induced stress on the endoplasmic reticulum activates JNK phosphorylation, triggers transcription of pro-inflammation markers (eg, MCP-1 and TNF-α) in adipose tissue and plasma, and inhibits Akt activation (shown in primary rat adipocytes); all of which may further exacerbate insulin resistance ().Citation10 GPR120 likely mediates its anti-inflammatory effects on JNK activation through the β-arrestin-2 signaling pathway mentioned above.Citation3 In addition, since homocysteine can repress β-arrestin-2 levels,Citation83 it would be of interest to determine whether GPR120 stimulation might also be involved in anti-inflammatory effects via restoration of β-arrestin-2 expression.

It has been reported that GPR120 expression in liver is limited to resident macrophage KCs, and indeed, GPR120 plays a key role in mediating anti-inflammatory effect in KCs.Citation9 Hepatic ischemia-reperfusion injury can activate inflammatory signaling in KCs, which is mediated by JNK and NF-κB.Citation84Citation86 GPR120 activation in KCs by agonists (GW9508 and Omegaven® [Fresenius Kabi, Bad Homburg, Germany], a clinical ω-3 FA-formulation) can provide protection from such reperfusion injury by inhibiting activation of JNK and NF-κB (), which decreases induction of pro-inflammatory cytokines and upregulates expression of hepatoprotective cytokines.Citation9

GPR120 is also involved in inhibition of inflammasome activation.Citation7 In bone-marrow-derived macrophages and human THP-1 macrophages, ω-3 FAs suppress activation of the NLRP3/1b inflammasome, leading to inhibition of caspase-1 and IL-1β through activation of GPR120 and GPR40.Citation7 In this process, β-arrestin-2 functions as the downstream protein of GPR120 and GPR40 to repress inflammasome activation via binding to NLRP3/1b ().Citation7

Insulin resistance, a characteristic feature of T2DM, is associated with inflammation in adipose, hepatic, and muscle tissues. Moreover, pancreatic islets themselves also undergo inflammation as the disease progresses.Citation87,Citation88 Therefore, given the role of GPR120 in anti-inflammatory effect in macrophages, it seems possible that GPR120 could produce anti-diabetic effects via an anti-inflammatory influence in the above tissues.

GPR120 and its anti-diabetic potential

It has been shown that GPR120 mutations in obese Europeans, named as R270H, are associated with obesity.Citation4 The p.R270H variant cannot functionally transduce signals of FFAs, and thus FFA-induced Ca2+ mobilization and GLP-1 secretion have been found to be significantly suppressed in cells expressing this GPR120 variant.Citation4 This may be one of the reasons why the p.R270H variant of GPR120 is related to obesity in humans. It is noteworthy that the R270H variant is rare in Japanese, suggestive of the ethnic viability in GPR120 variation.Citation89 Indeed, GPR120 dysfunction is also associated with insulin resistance, the other obesity-related symptoms of metabolic disorder.

Insulin sensitivity in adipose, hepatic, and muscle tissues

Consumption of a HFD can induce upregulation of GPR120 expression in white adipose, cardiac, and EDL skeletal muscle tissues of mouse or rat.Citation40,Citation41 Also, GPR120 is expressed in subcutaneous and omental adipose tissues at higher levels in obese individuals than in lean controls.Citation4 Thus, it seems that GPR120 expression becomes upregulated to support maintenance of energy metabolism homeostasis (energy uptake, storage, and utilization). If so, a GPR120 deficiency would impair metabolic balance, which could lead to insulin resistance. Indeed, compared with wild-type mice, GPR120 knockout mice exhibit more severe signs of insulin resistance when fed a HFD. Specifically, they exhibit worse glucose intolerance, larger islets, more Ki67-positive cells in the pancreas, reduced expression of insulin-signaling-related genes (IRS1, IRSβ, or IRS2) in white adipose tissue and liver, and impaired insulin-induced Akt phosphorylation in white adipose tissue, liver, and muscle.Citation4 The notion that GPR120’s role as an FA receptor is critical in regulation of energy metabolism homeostasis is supported strongly by the observation that FA administration can enhance muscle and hepatic insulin sensitivity, increase glucose infusion rate, promote hepatic lipid metabolism, and decrease hepatic steatosis in wild-type mice but not GPR120 knockout mice.Citation3

It has been suggested that a failure to store energy through adipogenesis when excess nutrients are ingested could cause insulin resistance related to an overload of lipids in the liver and muscles.Citation6,Citation90,Citation91 Since FFAs can promote adipogenesis in adipose tissue by activating GPR120, dysfunction of this receptor may lead to a failure of adipocyte differentiation in adipose, resulting in lipid accumulation in the liver.Citation40,Citation78 In the absence of functional GPR120, the decreased lipogenesis in adipose with hepatic steatosis and increased lipogenesis in the liver may contribute to the development of insulin resistance.Citation3,Citation4 If so, then activation of normal GPR120 by FFAs may facilitate normal lipogenesis in adipose and liver, and thereby perhaps prevent hepatic steatosis and maintain insulin sensitivity.

GPR120 might also contribute to metabolic homeostasis via its important role in maintaining normal adipose–liver interactions. GPR120 dysfunction may lead to lipid metabolic disorders due to insufficient production of palmitoleate (C16:1n7), a lipid hormone that modulates metabolic homeostasis and through which adipose interacts with other distant tissues such as liver and muscle.Citation92 In addition, GPR120 may also contribute to maintenance of metabolic balance by way of GLUT4 translocation in adipocytes.Citation3 That is, GPR120 dysfunction may downregulate FA-stimulated glucose transmembrane transport in adipose tissues, leading to decreased glucose uptake and utilization.

Aside from any direct effects of GPR120 on lipid and glucose metabolism, GPR120 activation may temper the pro-inflammatory effects induced by a HFD in mice.Citation3,Citation4 GPR120 expression is increased markedly in macrophages in adipose and liver in obese mice.Citation3 Indeed, ω-3 FA administration decreases the expression of M1 pro-inflammatory genes, decreases inflammatory macrophage infiltration in adipose tissue, and increases transcription of M2 anti-inflammatory genes in adipose tissue of HFD mice, but only if they had a functional GPR120 gene.Citation3 Moreover, ω-3 FA administration inhibits NLRP3 inflammasome activation in HFD mice, leading to reduced caspase-1 activation as well as repressed production of IL-1β, IL-18, TNF-α, and MCP-1, ultimately preventing NLRP3 inflammasome-dependent insulin resistance, which may be due, at least partially, to FA stimulation of GPR120.Citation7 These findings suggest that FA stimulation of GPR120 in macrophages could inhibit HFD-induced inflammation. Moreover, inflammation can interrupt normal energy metabolism, favoring the pathogenesis of insulin resistance through the inflammatory signaling cascades discussed above.Citation93Citation95 Taken together, in macrophages of adipose tissue and liver (KCs), GPR120 activation by FFAs could inhibit these inflammatory signaling pathways and thereby perhaps reverse insulin resistance. In contrast, under conditions of GPR120 dysfunction, deficiency of such protective anti-inflammatory effects may leave tissue vulnerable to the development of insulin resistance. This possibility is supported by findings that mice with a GPR120 deficiency that are fed a HFD lose the benefit of FA stimulation – such as reduction of IL-1β in adipose – leading to a resultant decrease in IRS1 and leaving the mice susceptible to worsening insulin resistance ().Citation4,Citation96

Oh et alCitation3 concluded that FFAs can enhance the insulin sensitivity of skeletal muscle in HFD mice by exerting anti-inflammatory effects. However, GPR120 expression in muscle is negligible, and there has been little evidence to date indicating that GPR120 mediates direct anti-inflammatory effects in muscle macrophages. Since activation of inflammation in adipose macrophages is an inducing factor for insulin resistance in muscle tissue,Citation97 GPR120-mediated anti-inflammation in adipose tissue might also be able to relieve insulin resistance in muscle to some extent.

Although insulin resistance is considered to be one of the T2DM features, some researchers propose that insulin resistance might be also linked with development of type 1 diabetes as well.Citation98Citation100 Given the important roles of GPR120 in the prevention of insulin resistance, it appears that GPR120 activation might also be beneficial for the management of type 1 diabetes.

Pancreatic islet and β-cell function

Intestinal hormone secretion

GPR120 could affect pancreatic islet function through modulation of GLP-1, CCK, and GIP secretion from the intestinal tract. The β-cell dysfunction and loss of β-cell mass that occur as diabetes progresses could cause insufficient pancreatic insulin secretion. GPR120 activation can increase intestinal secretion of CCK and GLP-1, two incretins that promote pancreatic insulin secretion. In addition, since CCK and GLP-1 can also increase β-cell survival, stimulate proliferation of β-cells and improve β-cell function,Citation101Citation104 it seems possible that induction of CCK and GLP-1 secretion by GPR120 activation could expand β-cell mass and support β-cell function. Moreover, the intestinal hormone GIP, which also can promote β-cell survival,Citation105 may be another target for enhancement of β-cell mass through GPR120 activation.Citation26

Resident GPR120 in pancreatic islets

Modulation of gastrointestinal hormone secretion by GPR120 activation is an indirect way of regulating pancreatic islet function. Meanwhile, GPR120 stimulation in pancreatic islets may manipulate islet function directly. Although it remains controversial whether GPR120 is expressed in rodent pancreas, some researchers have detected GPR120 in mouse pancreatic islets, as well as in MIN6 β-cells, and thus have predicted that it may be involved in FFA-induced regulation of pancreatic islet function.Citation16,Citation39

GPR120 activation may affect pancreatic physiology through regulating function of the pancreatic α-cell, which is best known for its glucagon-secreting function. Whalley et al observed GPR120 expression in a murine pancreatic α-cell line (αTC1-6), and they further showed that αTC1-6 cells can secrete GLP-1 and express prohormone convertase 1, which cleaves proglucagon into GLP-1.Citation106 They demonstrated that GPR120 agonism can stimulate GLP-1 secretion and inhibit glucagon secretion in αTC1-6 cells.Citation106 Furthermore, the same group showed that these cells secrete more GLP-1 under diabetic (high-glucose) conditions. To test whether such GLP-1 secretion is induced via GPR120 agonism, further studies are needed to determine whether genetic knockdown or knockout of GPR120 could attenuate or abolish induction of GLP-1 secretion in the presence or absence of a high concentration of glucose.

In human islets, GPR120 expression is associated positively with insulin secretion and insulin content but negatively with hemoglobin A1c percentage, and pancreatic islets from hyperglycemic or diabetic patients have reduced GPR120 expression compared with healthy individuals.Citation15 These observations suggest that human pancreatic insulin secretion correlates with islet GPR120 expression. GPR120 expressed in pancreatic islets may be involved in FA-stimulated insulin secretion from β-cells. Although GPR40 has been demonstrated to mediate FA-induced stimulation on insulin secretion directly,Citation48,Citation50 it appears to account for only about half of FA-stimulated insulin secretion in mice.Citation48 Furthermore, GPR120 can mediate FA-stimulated elevation of [Ca2+]i in intestinal cells, an important event in triggering insulin secretion.Citation14,Citation107 Taken together, these findings suggest that pancreatic GPR120 may have a direct role in regulating insulin secretion from β-cells.

Since GPR120 can mediate inhibition of ghrelin secretion from the stomach, it raises the possibility that GPR120 may also inhibit ghrelin secretion from epsilon cells in pancreatic islets.Citation94,Citation95 And it is encouraging that, Dezaki et alCitation69 reported that blockade of pancreatic islet-derived ghrelin can enhance insulin secretion and thereby prevent glucose intolerance in mice fed a HFD.

In human islets, GPR120 activation by ω-3 FA prevents lipid-induced apoptosis and GPR120 knockdown attenuates ω-3 FA-related anti-apoptotic effects.Citation15 Moreover, in serum-starved STC-1 cells, GPR120 has been implicated in the anti-apoptotic effect of ω-3 FAs via activation of ERK and Akt signaling. Furthermore, induction of ERK and Akt phosphorylation can protect against β-cell apoptosis and increase insulin secretion.Citation64 Hence, there are some clues suggesting that GPR120 may be involved in the anti-apoptosis effects of ω-3 FAs on β-cells in diabetic islets. Interestingly, it has been reported that β-arrestin-2 has important roles in the regulation of insulin-Akt signaling in the liver and pancreatic islets.Citation108,Citation109 The potential mechanism is that β-arrestin-2 is linked to this signaling pathway through scaffolding Akt to insulin receptor.Citation109 If GPR120 stimulation in pancreatic islets could activate Akt signaling so as to protect β-cell function, it may be plausible that β-arrestin-2 also functions as a scaffold protein to recruit Akt.

ω-3 FAs can exert anti-inflammatory and anti-diabetic effects on the pancreas. Using fat-1 transgenic mice, Bellenger et alCitation110 showed that high levels of endogenous ω-3 FAs can prevent streptozotocin-induced diabetes via inhibition of pro-inflammatory factors (eg, TNF-α, NF-κB, IL-1β, MCP-1, and PGE2) in pancreas. Hence, as a ω-3 FA receptor, GPR120 may be involved in ω-3 FA inhibition of inflammation in pancreatic islets.

Diabetic islets have high levels of activated islet macrophages, leading to inflammation (eg, via NLRP3 inflammasome activation) and release of cytokines (eg, IL-8 and MCP-1).Citation97,Citation111 Pancreatic β-cells also ramp up cytokine production under diabetic conditions in a manner that leads to β-cell dysfunction and death.Citation112 For example, IL-1β-induced production of COX-2 and PGE2 can impair glucose-stimulated insulin secretion in human and rodent pancreatic islets.Citation113 Conversely, inhibition of IL-1β-induced NF-κB activation and COX-2/PGE2 generation could enhance pancreatic islet cell function.Citation114 Given that GPR120 plays important roles in mediating anti-inflammation in macrophages,Citation3,Citation7Citation9 it is likely that the anti-diabetic effects of GPR120 may involve anti-inflammatory effects in pancreatic macrophages and β-cells. summarizes the potential mechanism by which GPR120 activation in pancreatic islets exhibits the anti-diabetic effects.

Figure 3 Schematic overview of the potential mechanism by which GPR120 activation in pancreatic islets exerts anti-diabetic effects.

Notes: In pancreatic islets, ω-3 FA-induced GPR120 stimulation may regulate and protect islet function to prevent against diabetes via eliciting anti-apoptotic, anti-inflammatory effects, and/or regulating pancreatic ghrelin, insulin secretion, and/or α-cell GLP-1 secretion.
Abbreviations: Akt, protein kinase B; GPR120, G-protein coupled receptor 120; GLP-1, glucagon-like peptide-1; ERK, extracellular-signal-regulated kinase; FA, fatty acid.
Figure 3 Schematic overview of the potential mechanism by which GPR120 activation in pancreatic islets exerts anti-diabetic effects.

Conclusion and perspective

Although GPR120 acts as a tumor-processing factor in CRC, it may have a positive role in the management of diabetes. GPR120 activation supports metabolic homeostasis by inhibiting inflammation in macrophages and regulating glucose and/or lipid metabolism in adipose, liver, and muscle tissues directly or indirectly. Furthermore, GPR120 stimulation can indirectly promote pancreatic islet function and β-cell proliferation through its modulatory influence on gastrointestinal hormone secretion. Therefore, GPR120 is identified as a potential target for development of anti-diabetic drugs.

Although it is yet unclear whether pancreatic GPR120 regulates islet function, the fact that GPR120 expression is associated with insulin secretion in human islets supports the possibility that GPR120 in the pancreas could be involved in regulating islet function. Given the important roles of GPR120 in the modulation of gastrointestinal hormone secretion, anti-inflammation in macrophages, and anti-apoptosis in enteroendocrine cells and human islets, it is likely that in diabetic islets, GPR120 may promote β-cell survival/proliferation and islet function through the following activities: anti-apoptotic effects in β-cells, anti-inflammatory effects in islet macrophages and/or β-cells, direct stimulation of β-cell insulin secretion, induction of GLP-1 secretion from pancreatic α-cells, and/or inhibition of pancreatic ghrelin secretion.

Acknowledgments

The work was fully supported by the General Research Fund of the Research Grants Council of Hong Kong (Ref No 470413), awarded to PS Leung.

Disclosure

No conflicts of interest relevant to this article are reported.

References

  • Hu FB Globalization of diabetes: the role of diet, lifestyle, and genes Diabetes Care 2011 34 6 1249 1257 21617109
  • Beltran-Sanchez H Harhay MO Harhay MM McElligott S Prevalence and trends of metabolic syndrome in the adult US population, 1999–2010 J Am Coll Cardiol 2013 62 8 697 703 23810877
  • Oh DY Talukdar S Bae EJ GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects Cell 2010 142 5 687 698 20813258
  • Ichimura A Hirasawa A Poulain-Godefroy O Dysfunction of lipid sensor GPR120 leads to obesity in both mouse and human Nature 2012 483 7389 350 354 22343897
  • Matsuzaka T Shimano H Molecular mechanisms involved in hepatic steatosis and insulin resistance J Diabetes Investig 2011 2 3 170 175
  • Danforth EJr Failure of adipocyte differentiation causes type II diabetes mellitus? Nat Genet 2000 26 1 13 10973236
  • Yan Y Jiang W Spinetti T Omega-3 fatty acids prevent inflammation and metabolic disorder through inhibition of NLRP3 inflammasome activation Immunity 2013 38 6 1154 1163 23809162
  • Li X Yu Y Funk CD Cyclooxygenase-2 induction in macrophages is modulated by docosahexaenoic acid via interactions with free fatty acid receptor 4 (FFA4) FASEB J 2013 27 12 4987 4997 24005906
  • Raptis DA Limani P Jang JH Gpr120 on Kupffer cells mediates hepatoprotective effects of omega3-fatty acids J Hepatol 2014 60 3 625 632 24262133
  • Li Y Zhang H Jiang C Hyperhomocysteinemia promotes insulin resistance by inducing endoplasmic reticulum stress in adipose tissue J Biol Chem 2013 288 14 9583 9592 23417716
  • Hirasawa A Tsumaya K Awaji T Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120 Nat Med 2005 11 1 90 94 15619630
  • Tanaka T Katsuma S Adachi T Free fatty acids induce cholecystokinin secretion through GPR120 Naunyn Schmiedebergs Arch Pharmacol 2008 377 4–6 523 527 17972064
  • Tanaka T Yano T Adachi T Cloning and characterization of the rat free fatty acid receptor GPR120: in vivo effect of the natural ligand on GLP-1 secretion and proliferation of pancreatic beta cells Naunyn Schmiedebergs Arch Pharmacol 2008 377 4–6 515 522 18320172
  • Shah BP Liu P Yu T Hansen DR Gilbertson TA TRPM5 is critical for linoleic acid-induced CCK secretion from the enteroendocrine cell line, STC-1 Am J Physiol Cell Physiol 2012 302 1 C210 C219 21998136
  • Taneera J Lang S Sharma A A systems genetics approach identifies genes and pathways for type 2 diabetes in human islets Cell Metab 2012 16 1 122 134 22768844
  • Kebede MA Alquier T Latour MG Poitout V Lipid receptors and islet function: therapeutic implications? Diabetes Obes Metab 2009 11 Suppl 4 10 20 19817784
  • Burns RN Moniri NH Agonism with the omega-3 fatty acids alpha-linolenic acid and docosahexaenoic acid mediates phosphorylation of both the short and long isoforms of the human GPR120 receptor Biochem Biophys Res Commun 2010 396 4 1030 1035 20471368
  • Watson SJ Brown AJ Holliday ND Differential signaling by splice variants of the human free fatty acid receptor GPR120 Mol Pharmacol 2012 81 5 631 642 22282525
  • Moore K Zhang Q Murgolo N Hosted T Duffy R Cloning, expression, and pharmacological characterization of the GPR120 free fatty acid receptor from cynomolgus monkey: comparison with human GPR120 splice variants Comp Biochem Physiol B Biochem Mol Biol 2009 154 4 419 426 19723586
  • Shimpukade B Hudson BD Hovgaard CK Milligan G Ulven T Discovery of a potent and selective GPR120 agonist J Med Chem 2012 55 9 4511 4515 22519963
  • Holliday ND Watson SJ Brown AJ Drug discovery opportunities and challenges at g protein coupled receptors for long chain free fatty acids Front Endocrinol (Lausanne) 2012 2 112 22649399
  • Suzuki T Igari S Hirasawa A Identification of G protein-coupled receptor 120-selective agonists derived from PPARgamma agonists J Med Chem 2008 51 23 7640 7644 19007110
  • Sun Q Hirasawa A Hara T Structure-activity relationships of GPR120 agonists based on a docking simulation Mol Pharmacol 2010 78 5 804 810 20685848
  • Cen B Xiong Y Ma L Pei G Direct and differential interaction of beta-arrestins with the intracellular domains of different opioid receptors Mol Pharmacol 2001 59 4 758 764 11259620
  • Hudson BD Shimpukade B Mackenzie AE 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 5 710 725 23979972
  • Parker HE Habib AM Rogers GJ Gribble FM Reimann F Nutrient-dependent secretion of glucose-dependent insulinotropic polypeptide from primary murine K cells Diabetologia 2009 52 2 289 298 19082577
  • Wu Q Wang H Zhao X Identification of G-protein-coupled receptor 120 as a tumor-promoting receptor that induces angiogenesis and migration in human colorectal carcinoma Oncogene 2013 32 49 5541 5550 23851494
  • Iakoubov R Izzo A Yeung A Whiteside CI Brubaker PL Protein kinase Czeta is required for oleic acid-induced secretion of glucagon-like peptide-1 by intestinal endocrine L cells Endocrinology 2007 148 3 1089 1098 17110421
  • Miyauchi S Hirasawa A Iga T Distribution and regulation of protein expression of the free fatty acid receptor GPR120 Naunyn Schmiedebergs Arch Pharmacol 2009 379 4 427 434 19145429
  • Janssen S Laermans J Iwakura H Tack J Depoortere I Sensing of fatty acids for octanoylation of ghrelin involves a gustatory G-protein PLoS One 2012 7 6 e40168 22768248
  • Lu X Zhao X Feng J Postprandial inhibition of gastric ghrelin secretion by long-chain fatty acid through GPR120 in isolated gastric ghrelin cells and mice Am J Physiol Gastrointest Liver Physiol 2012 303 3 G367 G376 22678998
  • Gong Z Yoshimura M Aizawa S G protein-coupled receptor 120 signaling regulates ghrelin secretion in vivo and in vitro Am J Physiol Endocrinol Metab 2014 306 1 E28 E35 24222669
  • Matsumura S Mizushige T Yoneda T GPR expression in the rat taste bud relating to fatty acid sensing Biomed Res 2007 28 1 49 55 17379957
  • Matsumura S Eguchi A Mizushige T Colocalization of GPR120 with phospholipase-Cbeta2 and alpha-gustducin in the taste bud cells in mice Neurosci Lett 2009 450 2 186 190 19071193
  • Cartoni C Yasumatsu K Ohkuri T Taste preference for fatty acids is mediated by GPR40 and GPR120 J Neurosci 2010 30 25 8376 8382 20573884
  • Martin C Passilly-Degrace P Chevrot M Lipid-mediated release of GLP-1 by mouse taste buds from circumvallate papillae: putative involvement of GPR120 and impact on taste sensitivity J Lipid Res 2012 53 11 2256 2265 22904345
  • Ozdener MH Subramaniam S Sundaresan S CD36- and GPR120-mediated Ca2+ signaling in human taste bud cells mediates differential responses to fatty acids and is altered in obese mice Gastroenterology 2014 146 4 995 1005 24412488
  • Galindo MM Voigt N Stein J G protein-coupled receptors in human fat taste perception Chem Senses 2012 37 2 123 139 21868624
  • Morgan NG Dhayal S G-protein coupled receptors mediating long chain fatty acid signalling in the pancreatic beta-cell Biochem Pharmacol 2009 78 12 1419 1427 19660440
  • Gotoh C Hong YH Iga T The regulation of adipogenesis through GPR120 Biochem Biophys Res Commun 2007 354 2 591 597 17250804
  • Cornall LM Mathai ML Hryciw DH McAinch AJ Diet-induced obesity up-regulates the abundance of GPR43 and GPR120 in a tissue specific manner Cell Physiol Biochem 2011 28 5 949 958 22178946
  • Katsuma S Hatae N Yano T Free fatty acids inhibit serum deprivation-induced apoptosis through GPR120 in a murine enteroendocrine cell line STC-1 J Biol Chem 2005 280 20 19507 19515 15774482
  • Hara T Hirasawa A Sun Q Novel selective ligands for free fatty acid receptors GPR120 and GPR40 Naunyn Schmiedebergs Arch Pharmacol 2009 380 3 247 255 19471906
  • Briscoe CP Peat AJ McKeown SC Pharmacological regulation of insulin secretion in MIN6 cells through the fatty acid receptor GPR40: identification of agonist and antagonist small molecules Br J Pharmacol 2006 148 5 619 628 16702987
  • Roduit R Nolan C Alarcon C A role for the malonyl-CoA/long-chain acyl-CoA pathway of lipid signaling in the regulation of insulin secretion in response to both fuel and nonfuel stimuli Diabetes 2004 53 4 1007 1019 15047616
  • Horia E Watkins BA Complementary actions of docosahexaenoic acid and genistein on COX-2, PGE2 and invasiveness in MDA-MB-231 breast cancer cells Carcinogenesis 2007 28 4 809 815 17052999
  • Zapata-Gonzalez F Rueda F Petriz J Human dendritic cell activities are modulated by the omega-3 fatty acid, docosahexaenoic acid, mainly through PPAR(gamma):RXR heterodimers: comparison with other polyunsaturated fatty acids J Leukoc Biol 2008 84 4 1172 1182 18632990
  • Latour MG Alquier T Oseid E GPR40 is necessary but not sufficient for fatty acid stimulation of insulin secretion in vivo Diabetes 2007 56 4 1087 1094 17395749
  • Zhang Y Xu M Zhang S The role of G protein-coupled receptor 40 in lipoapoptosis in mouse beta-cell line NIT-1 J Mol Endocrinol 2007 38 6 651 661 17556534
  • Itoh Y Kawamata Y Harada M Free fatty acids regulate insulin secretion from pancreatic beta cells through GPR40 Nature 2003 422 6928 173 176 12629551
  • Stoddart LA Brown AJ Milligan G Uncovering the pharmacology of the G protein-coupled receptor GPR40: high apparent constitutive activity in guanosine 5′-O-(3-[35S]thio)triphosphate binding studies reflects binding of an endogenous agonist Mol Pharmacol 2007 71 4 994 1005 17200419
  • Williams RH Champagne J Effects of cholecystokinin, secretin, and pancreatic polypeptide on secretion of gastric inhibitory polypeptide, insulin, and glucagon Life Sci 1979 25 11 947 956 513942
  • Rossetti L Shulman GI Zawalich WS Physiological role of cholecystokinin in meal-induced insulin secretion in conscious rats. Studies with L 364718, a specific inhibitor of CCK-receptor binding Diabetes 1987 36 10 1212 1215 3308589
  • Fried M Schwizer W Beglinger C Physiological role of cholecystokinin on postprandial insulin secretion and gastric meal emptying in man. Studies with the cholecystokinin receptor antagonist loxiglumide Diabetologia 1991 34 10 721 726 1959704
  • Luo J Swaminath G Brown SP A potent class of GPR40 full agonists engages the enteroinsular axis to promote glucose control in rodents PLoS One 2012 7 10 e46300 23056280
  • Woessmann W Mivechi NF Role of ERK activation in growth and erythroid differentiation of K562 cells Exp Cell Res 2001 264 2 193 200 11262176
  • Verheijen MH Wolthuis RM Defize LH den Hertog J Bos JL Interdependent action of RalGEF and Erk in Ras-induced primitive endoderm differentiation of F9 embryonal carcinoma cells Oncogene 1999 18 31 4435 4439 10442634
  • De Vita G Berlingieri MT Visconti R Akt/protein kinase B promotes survival and hormone-independent proliferation of thyroid cells in the absence of dedifferentiating and transforming effects Cancer Res 2000 60 14 3916 3920 10919669
  • Hardy S Langelier Y Prentki M Oleate activates phosphatidylinositol 3-kinase and promotes proliferation and reduces apoptosis of MDA-MB-231 breast cancer cells, whereas palmitate has opposite effects Cancer Res 2000 60 22 6353 6358 11103797
  • Soto-Guzman A Robledo T Lopez-Perez M Salazar EP Oleic acid induces ERK1/2 activation and AP-1 DNA binding activity through a mechanism involving Src kinase and EGFR transactivation in breast cancer cells Mol Cell Endocrinol 2008 294 1–2 81 91 18775472
  • Butler AE Janson J Bonner-Weir S Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes Diabetes 2003 52 1 102 110 12502499
  • Thomas HE McKenzie MD Angstetra E Campbell PD Kay TW Beta cell apoptosis in diabetes Apoptosis 2009 14 12 1389 1404 19322660
  • Wrede CE Dickson LM Lingohr MK Briaud I Rhodes CJ Protein kinase B/Akt prevents fatty acid-induced apoptosis in pancreatic beta-cells (INS-1) J Biol Chem 2002 277 51 49676 49684 12393870
  • Wijesekara N Krishnamurthy M Bhattacharjee A Adiponectin-induced ERK and Akt phosphorylation protects against pancreatic beta cell apoptosis and increases insulin gene expression and secretion J Biol Chem 2010 285 44 33623 33631 20709750
  • Dezaki K Sone H Yada T Ghrelin is a physiological regulator of insulin release in pancreatic islets and glucose homeostasis Pharmacol Ther 2008 118 2 239 249 18433874
  • Vatansever-Ozen S Tiryaki-Sonmez G Bugdayci G Ozen G The effects of exercise on food intake and hunger: relationship with acylated ghrelin and leptin J Sports Sci Med 2011 10 2 283 291 24149873
  • Levin F Edholm T Schmidt PT Ghrelin stimulates gastric emptying and hunger in normal-weight humans J Clin Endocrinol Metab 2006 91 9 3296 3302 16772353
  • Delhanty PJ van der Lely AJ Ghrelin and glucose homeostasis Peptides 2011 32 11 2309 2318 21396419
  • Dezaki K Sone H Koizumi M Blockade of pancreatic islet-derived ghrelin enhances insulin secretion to prevent high-fat diet-induced glucose intolerance Diabetes 2006 55 12 3486 3493 17130496
  • Mela DJ Sacchetti DA Sensory preferences for fats: relationships with diet and body composition Am J Clin Nutr 1991 53 4 908 915 2008871
  • Drewnowski A Brunzell JD Sande K Iverius PH Greenwood MR Sweet tooth reconsidered: taste responsiveness in human obesity Physiol Behav 1985 35 4 617 622 4070436
  • Cai H Cong WN Daimon CM Altered lipid and salt taste responsivity in ghrelin and GOAT null mice PLoS One 2013 8 10 e76553 24124572
  • Shin YK Martin B Golden E Modulation of taste sensitivity by GLP-1 signaling J Neurochem 2008 106 1 455 463 18397368
  • Stewart JE Feinle-Bisset C Golding M Oral sensitivity to fatty acids, food consumption and BMI in human subjects Br J Nutr 2010 104 1 145 152 20196892
  • Chen CS Bench EM Allerton TD Preference for linoleic acid in obesity-prone and obesity-resistant rats is attenuated by the reduction of CD36 on the tongue Am J Physiol Regul Integr Comp Physiol 2013 305 11 R1346 R1355 24154509
  • Godinot N Yasumatsu K Barcos ME Activation of tongue-expressed GPR40 and GPR120 by non caloric agonists is not sufficient to drive preference in mice Neuroscience 2013 25020 25030
  • Martin C Passilly-Degrace P Gaillard D The lipid-sensor candidates CD36 and GPR120 are differentially regulated by dietary lipids in mouse taste buds: impact on spontaneous fat preference PLoS One 2011 6 8 e24014 21901153
  • Liu D Wang L Meng Q Kuang H Liu X G-protein coupled receptor 120 is involved in glucose metabolism in fat cells Cell Mol Biol 2012 Suppl 58 OL1757 OL1762 23046868
  • Auwerx J Cock TA Knouff C PPAR-gamma: a thrifty transcription factor Nucl Recept Signal 2003 1 e006 16604178
  • Hoogeveen EK Kostense PJ Jakobs C Hyperhomocysteinemia increases risk of death, especially in type 2 diabetes: 5-year follow-up of the Hoorn Study Circulation 2000 101 13 1506 1511 10747342
  • Martos R Valle M Morales R Hyperhomocysteinemia correlates with insulin resistance and low-grade systemic inflammation in obese prepubertal children Metabolism 2006 55 1 72 77 16324922
  • Golbahar J Aminzadeh MA Kassab SE Omrani GR Hyperhomocysteinemia induces insulin resistance in male Sprague-Dawley rats Diabetes Res Clin Pract 2007 76 1 1 5 16963146
  • Ozcan U Yilmaz E Ozcan L Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes Science 2006 313 5790 1137 1140 16931765
  • Suetsugu H Iimuro Y Uehara T Nuclear factor {kappa}B inactivation in the rat liver ameliorates short term total warm ischaemia/reperfusion injury Gut 2005 54 6 835 842 15888794
  • Giakoustidis DE Giakoustidis AE Iliadis S Attenuation of liver ischemia/reperfusion induced apoptosis by epigallocatechin-3-gallate via down-regulation of NF-kappaB and c-Jun expression J Surg Res 2010 159 2 720 728 19394642
  • Seki E Brenner DA Karin M A liver full of JNK: signaling in regulation of cell function and disease pathogenesis, and clinical approaches Gastroenterology 2012 143 2 307 320 22705006
  • Donath MY Schumann DM Faulenbach M Islet inflammation in type 2 diabetes: from metabolic stress to therapy Diabetes Care 2008 31 Suppl 2 S161 S164 18227479
  • Imai Y Dobrian AD Morris MA Nadler JL Islet inflammation: a unifying target for diabetes treatment? Trends Endocrinol Metab 2013 24 7 351 360 23484621
  • Waguri T Goda T Kasezawa N Yamakawa-Kobayashi K The combined effects of genetic variations in the GPR120 gene and dietary fat intake on obesity risk Biomed Res 2013 34 2 69 74 23594480
  • McGarry JD Dobbins RL Fatty acids, lipotoxicity and insulin secretion Diabetologia 1999 42 2 128 138 10064091
  • Randle PJ Garland PB Hales CN Newsholme EA The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus Lancet 1963 1 7285 785 789 13990765
  • Cao H Gerhold K Mayers JR Identification of a lipokine, a lipid hormone linking adipose tissue to systemic metabolism Cell 2008 134 6 933 944 18805087
  • Kabayama K Sato T Kitamura F TNFalpha-induced insulin resistance in adipocytes as a membrane microdomain disorder: involvement of ganglioside GM3 Glycobiology 2005 15 1 21 29 15306563
  • Hotamisligil GS The role of TNFalpha and TNF receptors in obesity and insulin resistance J Intern Med 1999 245 6 621 625 10395191
  • Gorgens SW Eckardt K Elsen M Tennagels N Eckel J Chitinase-3-like protein 1 protects skeletal muscle from TNFalpha-induced inflammation and insulin resistance Biochem J 2014 459 3 479 488 24512683
  • Jager J Gremeaux T Cormont M Le Marchand-Brustel Y Tanti JF Interleukin-1beta-induced insulin resistance in adipocytes through down-regulation of insulin receptor substrate-1 expression Endocrinology 2007 148 1 241 251 17038556
  • Wen H Ting JP O’Neill LA A role for the NLRP3 inflammasome in metabolic diseases – did Warburg miss inflammation? Nat Immunol 2012 13 4 352 357 22430788
  • Wilkin TJ The accelerator hypothesis: weight gain as the missing link between Type I and Type II diabetes Diabetologia 2001 44 7 914 922 11508279
  • Sherry NA Tsai EB Herold KC Natural history of beta-cell function in type 1 diabetes Diabetes 2005 54 Suppl 2 S32 S39 16306337
  • Greenbaum CJ Insulin resistance in type 1 diabetes Diabetes Metab Res Rev 2002 18 3 192 200 12112937
  • Buteau J Foisy S Joly E Prentki M Glucagon-like peptide 1 induces pancreatic beta-cell proliferation via transactivation of the epidermal growth factor receptor Diabetes 2003 52 1 124 132 12502502
  • Yusta B Baggio LL Estall JL GLP-1 receptor activation improves beta cell function and survival following induction of endoplasmic reticulum stress Cell Metab 2006 4 5 391 406 17084712
  • Lavine JA Raess PW Stapleton DS Cholecystokinin is up-regulated in obese mouse islets and expands beta-cell mass by increasing beta-cell survival Endocrinology 2010 151 8 3577 3588 20534724
  • Lavine JA Raess PW Davis DB Overexpression of pre-pro-cholecystokinin stimulates beta-cell proliferation in mouse and human islets with retention of islet function Mol Endocrinol 2008 22 12 2716 2728 18845673
  • Kim SJ Winter K Nian C Glucose-dependent insulinotropic polypeptide (GIP) stimulation of pancreatic beta-cell survival is dependent upon phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB) signaling, inactivation of the forkhead transcription factor Foxo1, and down-regulation of bax expression J Biol Chem 2005 280 23 22297 22307 15817464
  • Whalley NM Pritchard LE Smith DM White A Processing of proglucagon to GLP-1 in pancreatic alpha-cells: is this a paracrine mechanism enabling GLP-1 to act on beta-cells? J Endocrinol 2011 211 1 99 106 21795304
  • Ashcroft FM Proks P Smith PA Stimulus-secretion coupling in pancreatic beta cells J Cell Biochem 1994 55 Suppl 54 65 7929618
  • Luan B Zhao J Wu H Deficiency of a beta- arrestin-2 signal complex contributes to insulin resistance Nature 2009 457 7233 1146 1149 19122674
  • Zhang M Zhu Y Mu K Loss of beta-arrestin2 mediates pancreatic-islet dysfunction in mice Biochem Biophys Res Commun 2013 435 3 345 349 23660189
  • Bellenger J Bellenger S Bataille A High pancreatic n-3 fatty acids prevent STZ-induced diabetes in fat-1 mice: inflammatory pathway inhibition Diabetes 2011 60 4 1090 1099 21330635
  • Ehses JA Perren A Eppler E Increased number of islet-associated macrophages in type 2 diabetes Diabetes 2007 56 9 2356 2370 17579207
  • Maedler K Sergeev P Ris F Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets J Clin Invest 2002 110 6 851 860 12235117
  • Parazzoli S Harmon JS Vallerie SN Cyclooxygenase-2, not microsomal prostaglandin E synthase-1, is the mechanism for interleukin-1beta-induced prostaglandin E2 production and inhibition of insulin secretion in pancreatic islets J Biol Chem 2012 287 38 32246 32253 22822059
  • Tran PO Gleason CE Robertson RP Inhibition of interleukin-1beta-induced COX-2 and EP3 gene expression by sodium salicylate enhances pancreatic islet beta-cell function Diabetes 2002 51 6 1772 1778 12031964