1
|
Yamamoto Y, Narumi K, Yamagishi N, Nishi T, Ito T, Iseki K, Kobayashi M, Kanai Y. Oral administration of linoleic acid immediately before glucose load ameliorates postprandial hyperglycemia. Front Pharmacol 2023; 14:1197743. [PMID: 37583904 PMCID: PMC10424117 DOI: 10.3389/fphar.2023.1197743] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
Introduction: Fatty acids are a major nutrient in dietary fat, some of which are ligands of long-chain fatty acid receptors, including G-protein-coupled receptor (GPR) 40 and GPR120. Pretreatment with GPR40 agonists enhanced the secretion of insulin in response to elevating blood glucose levels after glucose load in a diabetes model, but pretreatment with GPR120 agonist did not ameliorate postprandial hyperglycemia. This study examined whether oral administration of linoleic acid (LA), a GPR40 and GPR120 agonist, immediately before glucose load would affect the elevation of postprandial blood glucose levels in rats. Methods: Male rats and rats with type 1 diabetes administered streptozocin were orally administered LA, trilinolein, α-linolenic acid (α-LA), oleic acid, TAK-875, or TUG-891 immediately before glucose load. Blood glucose levels were measured before, then 15, 30, 60 and 120 min after glucose load. CACO-2 cells were used to measure the uptake of [14C] α-MDG for 30 min with or without LA. Gastric content from rats administered LA was collected 15 and 30 min after glucose load, and blood samples were collected for measurement of glucagon-like peptide 1 (GLP-1) and cholecystokinin concentrations. Results: The elevation of postprandial blood glucose levels was slowed by LA but not by trilinolein in rats without promotion of insulin secretion, and this effect was also observed in rats with type 1 diabetes. The uptake of α-MDG, an SGLT-specific substrate, was, however, not inhibited by LA. Gastric emptying was slowed by LA 15 min after glucose load, and GLP-1, but not cholecystokinin, level was elevated by LA 15 min after glucose load. TUG-891, a GPR120 agonist, ameliorated postprandial hyperglycemia but TAK-875, a GPR40 agonist, did not. Pretreatment with AH7614, a GPR120 antagonist, partially canceled the improvement of postprandial hyperglycemia induced by LA. α-LA, which has high affinity with GPR120 as well as LA, slowed the elevation of postprandial blood glucose levels, but oleic acid, which has lower affinity with GPR120 than LA, did not. Conclusion: Oral administration of LA immediately after glucose load ameliorated postprandial hyperglycemia due to slowing of gastric emptying via promotion of GLP-1 secretion. The mechanisms may be associated with GPR120 pathway.
Collapse
Affiliation(s)
- Yuta Yamamoto
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Katsuya Narumi
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Naoko Yamagishi
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Toshio Nishi
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takao Ito
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Ken Iseki
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yoshimitsu Kanai
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
2
|
Kurtz R, Anderman MF, Shepard BD. GPCRs get fatty: the role of G protein-coupled receptor signaling in the development and progression of nonalcoholic fatty liver disease. Am J Physiol Gastrointest Liver Physiol 2021; 320:G304-G318. [PMID: 33205999 PMCID: PMC8202238 DOI: 10.1152/ajpgi.00275.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), characterized by the abnormal deposition of lipids within the liver not due to alcohol consumption, is a growing epidemic affecting over 30% of the United States population. Both simple fatty liver and its more severe counterpart, nonalcoholic steatohepatitis, represent one of the most common forms of liver disease. Recently, several G protein-coupled receptors have emerged as targets for therapeutic intervention for these disorders. These include those with known hepatic function as well as those involved in global metabolic regulation. In this review, we highlight these emerging therapeutic targets, focusing on several common themes including their activation by microbial metabolites, stimulatory effect on insulin and incretin secretion, and contribution to glucose tolerance. The overlap in ligands, localization, and downstream effects of activation indicate the interdependent nature of these receptors and highlight the importance of this signaling family in the development and prevention of NAFLD.
Collapse
Affiliation(s)
- Ryan Kurtz
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Meghan F. Anderman
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Blythe D. Shepard
- Department of Human Science, Georgetown University, Washington, District of Columbia
| |
Collapse
|
3
|
Cosín-Roger J, Ortiz-Masia D, Barrachina MD, Calatayud S. Metabolite Sensing GPCRs: Promising Therapeutic Targets for Cancer Treatment? Cells 2020; 9:cells9112345. [PMID: 33113952 PMCID: PMC7690732 DOI: 10.3390/cells9112345] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
G-protein-coupled receptors constitute the most diverse and largest receptor family in the human genome, with approximately 800 different members identified. Given the well-known metabolic alterations in cancer development, we will focus specifically in the 19 G-protein-coupled receptors (GPCRs), which can be selectively activated by metabolites. These metabolite sensing GPCRs control crucial processes, such as cell proliferation, differentiation, migration, and survival after their activation. In the present review, we will describe the main functions of these metabolite sensing GPCRs and shed light on the benefits of their potential use as possible pharmacological targets for cancer treatment.
Collapse
Affiliation(s)
- Jesús Cosín-Roger
- Hospital Dr. Peset, Fundación para la Investigación Sanitaria y Biomédica de la Comunitat Valenciana, FISABIO, 46017 Valencia, Spain
- Correspondence: ; Tel.: +34-963851234
| | - Dolores Ortiz-Masia
- Departament of Medicine, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Maria Dolores Barrachina
- Departament of Pharmacology and CIBER, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (M.D.B.); (S.C.)
| | - Sara Calatayud
- Departament of Pharmacology and CIBER, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (M.D.B.); (S.C.)
| |
Collapse
|
4
|
Hashimoto T, Mogami H, Tsuriya D, Morita H, Sasaki S, Kumada T, Suzuki Y, Urano T, Oki Y, Suda T. G-protein-coupled receptor 40 agonist GW9508 potentiates glucose-stimulated insulin secretion through activation of protein kinase Cα and ε in INS-1 cells. PLoS One 2019; 14:e0222179. [PMID: 31498851 PMCID: PMC6733457 DOI: 10.1371/journal.pone.0222179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 08/23/2019] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE The mechanism by which G-protein-coupled receptor 40 (GPR40) signaling amplifies glucose-stimulated insulin secretion through activation of protein kinase C (PKC) is unknown. We examined whether a GPR40 agonist, GW9508, could stimulate conventional and novel isoforms of PKC at two glucose concentrations (3 mM and 20 mM) in INS-1D cells. METHODS Using epifluorescence microscopy, we monitored relative changes in the cytosolic fluorescence intensity of Fura2 as a marker of change in intracellular Ca2+ ([Ca2+]i) and relative increases in green fluorescent protein (GFP)-tagged myristoylated alanine-rich C kinase substrate (MARCKS-GFP) as a marker of PKC activation in response to GW9508 at 3 mM and 20 mM glucose. To assess the activation of the two PKC isoforms, relative increases in membrane fluorescence intensity of PKCα-GFP and PKCε-GFP were measured by total internal reflection fluorescence microscopy. Specific inhibitors of each PKC isotype were constructed and synthesized as peptide fusions with the third α-helix of the homeodomain of Antennapedia. RESULTS At 3 mM glucose, GW9508 induced sustained MARCKS-GFP translocation to the cytosol, irrespective of changes in [Ca2+]i. At 20 mM glucose, GW9508 induced sustained MARCKS-GFP translocation but also transient translocation that followed sharp increases in [Ca2+]i. Although PKCα translocation was rarely observed, PKCε translocation to the plasma membrane was sustained by GW9508 at 3 mM glucose. At 20 mM glucose, GW9508 induced transient translocation of PKCα and sustained translocation as well as transient translocation of PKCε. While the inhibitors (75 μM) of each PKC isotype reduced GW9508-potentiated, glucose-stimulated insulin secretion in INS-1D cells, the PKCε inhibitor had a more potent effect. CONCLUSION GW9508 activated PKCε but not PKCα at a substimulatory concentration of glucose. Both PKC isotypes were activated at a stimulatory concentration of glucose and contributed to glucose-stimulated insulin secretion in insulin-producing cells.
Collapse
Affiliation(s)
- Takuya Hashimoto
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
- * E-mail:
| | - Hideo Mogami
- Department of Health and Nutrition, Tokoha University, Shizuoka, Japan
| | - Daisuke Tsuriya
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Hiroshi Morita
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Shigekazu Sasaki
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Tatsuro Kumada
- Department of Occupational Therapy, Tokoha University, Shizuoka, Japan
| | - Yuko Suzuki
- Department of Medical Physiology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Tetsumei Urano
- Department of Medical Physiology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Yutaka Oki
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
- Department of Family and Community Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Takafumi Suda
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| |
Collapse
|
5
|
Wei W, Ehlerding EB, Lan X, Luo QY, Cai W. Molecular imaging of β-cells: diabetes and beyond. Adv Drug Deliv Rev 2019; 139:16-31. [PMID: 31378283 DOI: 10.1016/j.addr.2018.06.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/27/2018] [Accepted: 06/26/2018] [Indexed: 02/09/2023]
Abstract
Since diabetes is becoming a global epidemic, there is a great need to develop early β-cell specific diagnostic techniques for this disorder. There are two types of diabetes (i.e., type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM)). In T1DM, the destruction of pancreatic β-cells leads to reduced insulin production or even absolute insulin deficiency, which consequently results in hyperglycemia. Actually, a central issue in the pathophysiology of all types of diabetes is the relative reduction of β-cell mass (BCM) and/or impairment of the function of individual β-cells. In the past two decades, scientists have been trying to develop imaging techniques for noninvasive measurement of the viability and mass of pancreatic β-cells. Despite intense scientific efforts, only two tracers for positron emission tomography (PET) and one contrast agent for magnetic resonance (MR) imaging are currently under clinical evaluation. β-cell specific imaging probes may also allow us to precisely and specifically visualize transplanted β-cells and to improve transplantation outcomes, as transplantation of pancreatic islets has shown promise in treating T1DM. In addition, some of these probes can be applied to the preoperative detection of hidden insulinomas as well. In the present review, we primarily summarize potential tracers under development for imaging β-cells with a focus on tracers for PET, SPECT, MRI, and optical imaging. We will discuss the advantages and limitations of the various imaging probes and extend an outlook on future developments in the field.
Collapse
|
6
|
Chen T, Ning M, Ye Y, Wang K, Leng Y, Shen J. Design, synthesis and structure-activity relationship studies of GPR40 agonists containing amide linker. Eur J Med Chem 2018; 152:175-194. [PMID: 29705709 DOI: 10.1016/j.ejmech.2018.04.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/22/2018] [Accepted: 04/10/2018] [Indexed: 01/19/2023]
Abstract
Free fatty acid receptor 1 (FFAR1/GPR40) attracted significant attention as a potential target for developing novel antidiabetic drugs because of its unique mechanism in glucose homeostasis. Several reports have expressed concerns about central nervous system (CNS) penetration of GPR40 agonists, which is possibly attributed to their high lipophilicity and low total polar surface area. Herein, we report our efforts to improve the physicochemical properties and pharmacokinetic profiles of LY2881835, a GPR40 agonist that had undergone Phase I clinical trial, through a series of structural optimizations. We identified an orally efficacious compound, 15k, which possessed increased plasma exposure, prolonged half-life and reduced CNS exposure and liver to plasma distribution ratio compared with LY2881835. 15k is a potentially valuable lead compound in the development of safe and efficacious GPR40-targeted drugs to treat type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Tingting Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Mengmeng Ning
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Yangliang Ye
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Kai Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Ying Leng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
| | - Jianhua Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
| |
Collapse
|
7
|
Rives ML, Rady B, Swanson N, Zhao S, Qi J, Arnoult E, Bakaj I, Mancini A, Breton B, Lee SP, Player MR, Pocai A. GPR40-Mediated G α12 Activation by Allosteric Full Agonists Highly Efficacious at Potentiating Glucose-Stimulated Insulin Secretion in Human Islets. Mol Pharmacol 2018; 93:581-591. [PMID: 29572336 DOI: 10.1124/mol.117.111369] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/20/2018] [Indexed: 12/25/2022] Open
Abstract
GPR40 is a clinically validated molecular target for the treatment of diabetes. Many GPR40 agonists have been identified to date, with the partial agonist fasiglifam (TAK-875) reaching phase III clinical trials before its development was terminated due to off-target liver toxicity. Since then, attention has shifted toward the development of full agonists that exhibit superior efficacy in preclinical models. Full agonists bind to a distinct binding site, suggesting conformational plasticity and a potential for biased agonism. Indeed, it has been suggested that alternative pharmacology may be required for meaningful efficacy. In this study, we described the discovery and characterization of Compound A, a newly identified GPR40 allosteric full agonist highly efficacious in human islets at potentiating glucose-stimulated insulin secretion. We compared Compound A-induced GPR40 activity to that induced by both fasiglifam and AM-1638, another allosteric full agonist previously reported to be highly efficacious in preclinical models, at a panel of G proteins. Compound A was a full agonist at both the Gαq and Gαi2 pathways, and in contrast to fasiglifam Compound A also induced Gα12 coupling. Compound A and AM-1638 displayed similar activity at all pathways tested. The Gα12/Gα13-mediated signaling pathway has been linked to protein kinase D activation as well as actin remodeling, well known to contribute to the release of insulin vesicles. Our data suggest that the pharmacology of GPR40 is complex and that Gα12/Gα13-mediated signaling, which may contribute to GPR40 agonists therapeutic efficacy, is a specific property of GPR40 allosteric full agonists.
Collapse
Affiliation(s)
- Marie-Laure Rives
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Brian Rady
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Nadia Swanson
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Shuyuan Zhao
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Jenson Qi
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Eric Arnoult
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Ivona Bakaj
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Arturo Mancini
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Billy Breton
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - S Paul Lee
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Mark R Player
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| | - Alessandro Pocai
- Molecular and Cellular Pharmacology, Janssen Research & Development, LLC, La Jolla, California (M.-L.R., N.S.); Cardiovascular and Metabolism (B.R., S.Z., J.Q., I.B., S.P.L., M.R.P., A.P.), and Computational Chemistry (E.A.), Janssen Research & Development, LLC, Spring House, Pennsylvania; and Domain Therapeutics NA Inc., Montreal, Quebec, Canada (A.M., B.B.)
| |
Collapse
|
8
|
Sabrautzki S, Kaiser G, Przemeck GKH, Gerst F, Lorza-Gil E, Panse M, Sartorius T, Hoene M, Marschall S, Häring HU, Hrabě de Angelis M, Ullrich S. Point mutation of Ffar1 abrogates fatty acid-dependent insulin secretion, but protects against HFD-induced glucose intolerance. Mol Metab 2017; 6:1304-1312. [PMID: 29031729 PMCID: PMC5641630 DOI: 10.1016/j.molmet.2017.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 07/05/2017] [Accepted: 07/11/2017] [Indexed: 01/06/2023] Open
Abstract
Objective The fatty acid receptor 1 (FFAR1/GPR40) mediates fatty acid-dependent augmentation of glucose-induced insulin secretion (GIIS) in pancreatic β-cells. Genetically engineered Ffar1-knockout/congenic mice univocally displayed impaired fatty acid-mediated insulin secretion, but in vivo experiments delivered controversial results regarding the function of FFAR1 in glucose homeostasis and liver steatosis. This study presents a new coisogenic mouse model carrying a point mutation in Ffar1 with functional consequence. These mice reflect the situations in humans in which point mutations can lead to protein malfunction and disease development. Methods The Munich N-ethyl-N-nitrosourea (ENU) mutagenesis-derived F1 archive containing over 16,800 sperms and corresponding DNA samples was screened for mutations in the coding region of Ffar1. Two missense mutations (R258W and T146S) in the extracellular domain of the protein were chosen and homozygote mice were generated. The functional consequence of these mutations was examined in vitro in isolated islets and in vivo in chow diet and high fat diet fed mice. Results Palmitate, 50 μM, and the FFAR1 agonist TUG-469, 3 μM, stimulated insulin secretion in islets of Ffar1T146S/T146S mutant mice and of wild-type littermates, while in islets of Ffar1R258W/R258W mutant mice, these stimulatory effects were abolished. Insulin content and mRNA levels of Ffar1, Glp1r, Ins2, Slc2a2, Ppara, and Ppard were not significantly different between wild-type and Ffar1R258W/R258W mouse islets. Palmitate exposure, 600 μM, significantly increased Ppara mRNA levels in wild-type but not in Ffar1R258W/R258W mouse islets. On the contrary, Slc2a2 mRNA levels were significantly reduced in both wild-type and Ffar1R258W/R258W mouse islets after palmitate treatment. HFD feeding induced glucose intolerance in wild-type mice. Ffar1R258W/R258W mutant mice remained glucose tolerant although their body weight gain, liver steatosis, insulin resistance, and plasma insulin levels were not different from those of wild-type littermates. Worth mentioning, fasting plasma insulin levels were lower in Ffar1R258W/R258W mice. Conclusion A point mutation in Ffar1 abrogates the stimulatory effect of palmitate on GIIS, an effect that does not necessarily translate to HFD-induced glucose intolerance. Generation of mice carrying point mutations in Ffar1 using ENU. FFAR1 point mutation R258W abrogates fatty acid-induced insulin secretion. Dysfunctional FFAR1 inhibits the development of diet-induced glucose intolerance.
Collapse
Affiliation(s)
- Sibylle Sabrautzki
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and the German Mouse Clinic, 85764 Neuherberg, Germany; Research Unit Comparative Medicine, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | - Gabriele Kaiser
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany
| | - Gerhard K H Przemeck
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and the German Mouse Clinic, 85764 Neuherberg, Germany
| | - Felicia Gerst
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany
| | - Estela Lorza-Gil
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany
| | - Madhura Panse
- University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Tina Sartorius
- University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Miriam Hoene
- University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Susan Marschall
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and the German Mouse Clinic, 85764 Neuherberg, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany; University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Martin Hrabě de Angelis
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and the German Mouse Clinic, 85764 Neuherberg, Germany; Chair of Experimental Genetics, School of Life Sciences Weihenstephan, Technische Universität München, Alte Akademie 8, 85354 München, Germany
| | - Susanne Ullrich
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany; University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany.
| |
Collapse
|
9
|
Bertrand R, Wolf A, Ivashchenko Y, Löhn M, Schäfer M, Brönstrup M, Gotthardt M, Derdau V, Plettenburg O. Synthesis and Characterization of a Promising Novel FFAR1/GPR40 Targeting Fluorescent Probe for β-Cell Imaging. ACS Chem Biol 2016; 11:1745-54. [PMID: 27115176 DOI: 10.1021/acschembio.5b00791] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Diabetes affects an increasing number of patients worldwide and is responsible for a significant rise in healthcare expenses. Imaging of β-cells bears the potential to contribute to an improved understanding, diagnosis, and development of new treatment options for diabetes. Here, we describe the first small molecule fluorescent probe targeting the free fatty acid receptor 1 (FFAR1/GPR40). This receptor is highly expressed on β-cells, and was up to now unexplored for imaging purposes. We designed a novel probe by facile modification of the selective and potent FFAR1 agonist TAK-875. Effective and specific binding of the probe was demonstrated using FFAR1 overexpressing cells. We also successfully labeled FFAR1 on MIN6 and INS1E cells, two widely used β-cell models, by applying an effective amplification protocol. Finally, we showed that the probe is capable of inducing insulin secretion in a glucose-dependent manner, thus demonstrating that functional activity of the probe was maintained. These results suggest that our probe represents a first important step to successful β-cell imaging by targeting FFAR1. The developed probe may prove to be particularly useful for in vitro and ex vivo studies of diabetic cellular and animal models to gain new insights into disease pathogenesis.
Collapse
Affiliation(s)
- Romain Bertrand
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main 65926, Germany
- Department
of Nuclear Medicine, Radboud UMC, Nijmegen 6525, The Netherlands
| | - Andrea Wolf
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main 65926, Germany
| | - Yuri Ivashchenko
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main 65926, Germany
| | - Matthias Löhn
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main 65926, Germany
| | - Matthias Schäfer
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main 65926, Germany
| | - Mark Brönstrup
- DSAR/Drug
Disposition, Sanofi GmbH, Frankfurt am Main 65926, Germany
- Helmholtz Centre
for Infection Research, Braunschweig 38124, Germany
| | - Martin Gotthardt
- Department
of Nuclear Medicine, Radboud UMC, Nijmegen 6525, The Netherlands
| | - Volker Derdau
- DSAR/Drug
Disposition, Sanofi GmbH, Frankfurt am Main 65926, Germany
| | - Oliver Plettenburg
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main 65926, Germany
- Institute
of Medicinal Chemistry, Helmholtz Zentrum München, Ingolstaedter
Landstr. 1, Neuherberg 85764, Germany
- Leibniz University
Hannover, Schneiderberg 1 B, Hannover 30167, Germany
| |
Collapse
|
10
|
Tucker RM, Mattes RD, Running CA. Mechanisms and effects of "fat taste" in humans. Biofactors 2014; 40:313-26. [PMID: 24591077 DOI: 10.1002/biof.1162] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/18/2014] [Indexed: 12/11/2022]
Abstract
Evidence supporting a "taste" cue from fat in the oral cavity continues to accrue. The proposed stimuli for fat taste, non-esterified fatty acids (NEFA), are released from food through hydrolytic rancidity and lipase activity derived from foods or saliva. NEFA must then be released from the food matrix, negotiate the aqueous environment to reach taste cell surfaces, and interact with receptors such as CD36 and GPR120 or diffuse across cell membranes to initiate a taste signal. Knowledge of these processes in non-gustatory tissues should inform understanding of taste responses to NEFA. Additionally, downstream effects of oral triglyceride exposure have been observed in numerous studies. Data specific to effects of NEFA versus triglyceride are scarce, but modified sham feeding trials with triglyceride document cephalic phase responses including elevations in serum lipids and insulin as well as potential, but debated, effects on gut peptides, appetite, and thermogenesis. In this review, we highlight the mechanisms by which NEFA migrate to and interact with taste cells, and then we examine physiological responses to oral fat exposure.
Collapse
Affiliation(s)
- Robin M Tucker
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| | | | | |
Collapse
|
11
|
Tomita T, Hosoda K, Fujikura J, Inagaki N, Nakao K. The G-Protein-Coupled Long-Chain Fatty Acid Receptor GPR40 and Glucose Metabolism. Front Endocrinol (Lausanne) 2014; 5:152. [PMID: 25309513 PMCID: PMC4176464 DOI: 10.3389/fendo.2014.00152] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/12/2014] [Indexed: 11/23/2022] Open
Abstract
Free fatty acids (FFAs) play a pivotal role in metabolic control and cell signaling processes in various tissues. In particular, FFAs are known to augment glucose-stimulated insulin secretion by pancreatic beta cells, where fatty acid-derived metabolites, such as long-chain fatty acyl-CoAs, are believed to act as crucial effectors. Recently, G-protein-coupled receptor 40 (GPR40), a receptor for long-chain fatty acids, was reported to be highly expressed in pancreatic beta cells and involved in the regulation of insulin secretion. Hence, GPR40 is considered to be a potential therapeutic target for the treatment of diabetes. In this review, we summarize the identification and gene expression patterns of GPR40 and its role in glucose metabolism. We also discuss the potential application of GPR40 as a therapeutic target.
Collapse
Affiliation(s)
- Tsutomu Tomita
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
- *Correspondence: Tsutomu Tomita, Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo, Kyoto 606-8507, Japan e-mail:
| | - Kiminori Hosoda
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junji Fujikura
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuwa Nakao
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
12
|
Huang H, Dai MH, Tao YX. Physiology and Therapeutics of the Free Fatty Acid Receptor GPR40. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:67-94. [DOI: 10.1016/b978-0-12-800101-1.00003-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Matsuda-Nagasumi K, Takami-Esaki R, Iwachidow K, Yasuhara Y, Tanaka H, Ogi K, Nakata M, Yano T, Hinuma S, Taketomi S, Odaka H, Kaisho Y. Lack of GPR40/FFAR1 does not induce diabetes even under insulin resistance condition. Diabetes Obes Metab 2013; 15:538-45. [PMID: 23331570 DOI: 10.1111/dom.12065] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/13/2012] [Accepted: 01/12/2013] [Indexed: 11/28/2022]
Abstract
AIMS G protein-coupled receptor/free fatty acid receptor 1 (GPR40/FFAR1 ) regulates free fatty acid-induced insulin secretion. This study has been performed to clarify whether or not loss of GPR40/FFAR1 function exacerbates diabetes, that is, whether GPR40 has an essential physiological role in the development of diabetes or not. METHODS We generated GPR40/FFAR1 knockout (KO) mice and analysed their phenotypes in vitro and in vivo under the condition of dietary or genetically induced insulin resistance. RESULTS GPR40/FFAR1 KO mice kept on a high-fat diet became obese, developed glucose intolerance to a similar degree as GPR40/FFAR1 wild-type (WT) mice. In addition, the phenotype of KO mice harbouring diabetogenic KK background genes showed glucose intolerance at a level similar to level for control KK mice. In both mouse models with insulin resistance, insulin secretion after oral glucose load and homeostasis model assessment-insulin resistance (HOMA-IR) did not change between GPR40/FFAR1 KO and WT mice. Although glucose-induced insulin secretion under high palmitate concentration was significantly lower in KO than in WT islets, pancreatic insulin content and insulin secretion stimulated with glucose alone were not different between KO and WT mice. CONCLUSIONS GPR40/FFAR1 has a major role in regulating fatty-acid-mediated insulin secretion, but the lack of GPR40/FFAR1 does not exacerbate glucose intolerance and insulin resistance induced by high-fat diet or diabetogenic KK gene. Our findings indicate that loss of GPR40/FFAR1 function does not play an important role in inducing or exacerbating diabetes.
Collapse
Affiliation(s)
- K Matsuda-Nagasumi
- Metabolic Disease Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Odori S, Hosoda K, Tomita T, Fujikura J, Kusakabe T, Kawaguchi Y, Doi R, Takaori K, Ebihara K, Sakai Y, Uemoto S, Nakao K. GPR119 expression in normal human tissues and islet cell tumors: evidence for its islet-gastrointestinal distribution, expression in pancreatic beta and alpha cells, and involvement in islet function. Metabolism 2013; 62:70-8. [PMID: 22883930 DOI: 10.1016/j.metabol.2012.06.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 06/09/2012] [Accepted: 06/27/2012] [Indexed: 12/25/2022]
Abstract
OBJECTIVE GPR119 is reportedly involved in regulating glucose metabolism and food intake in rodents, but little is known about its expression and functional significance in humans. To begin to assess the potential clinical importance of GPR119, the distribution of GPR119 gene expression in humans was examined. MATERIALS/METHODS Expression of GPR119 mRNA in fresh samples of normal human pancreas (n=19) and pancreatic islets (n=3) and in insulinomas (n=2) and glucagonomas (n=2), all collected at surgery, was compared with the mRNA expression of various receptors highly expressed and operative in human pancreatic islets. RESULTS GPR119 mRNA was most abundant in the pancreas, followed by the duodenum, stomach, jejunum, ileum and colon. Pancreatic levels of GPR119 mRNA were similar to those of GPR40 mRNA and were higher than those of GLP1R and SUR1 mRNA, which are strongly expressed in human pancreatic islets. Moreover, levels of GPR119 mRNA in pancreatic islets were more than 10 times higher than in adjacent pancreatic tissue, as were levels of GPR40 mRNA. GPR119 mRNA was also abundant in two cases of insulinoma and two cases of glucagonoma, but was undetectable in a pancreatic acinar cell tumor. Similar results were obtained with mouse pancreatic islets, MIN6 insulinoma cells and alpha-TC glucagonoma cells. CONCLUSIONS The results provide evidence of an islet-gastrointestinal distribution of GPR119, its expression in pancreatic beta and alpha cells, and its possible involvement in islet function. They also provide the basis for a better understanding of the potential clinical importance of GPR119.
Collapse
Affiliation(s)
- Shinji Odori
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Cuny T, Guerci B, Cariou B. New avenues for the pharmacological management of type 2 diabetes: An update. ANNALES D'ENDOCRINOLOGIE 2012; 73:459-68. [DOI: 10.1016/j.ando.2012.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
16
|
Luo J, Swaminath G, Brown SP, Zhang J, Guo Q, Chen M, Nguyen K, Tran T, Miao L, Dransfield PJ, Vimolratana M, Houze JB, Wong S, Toteva M, Shan B, Li F, Zhuang R, Lin DCH. A potent class of GPR40 full agonists engages the enteroinsular axis to promote glucose control in rodents. PLoS One 2012; 7:e46300. [PMID: 23056280 PMCID: PMC3467217 DOI: 10.1371/journal.pone.0046300] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 08/29/2012] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes is characterized by impaired glucose homeostasis due to defects in insulin secretion, insulin resistance and the incretin response. GPR40 (FFAR1 or FFA1) is a G-protein-coupled receptor (GPCR), primarily expressed in insulin-producing pancreatic β-cells and incretin-producing enteroendocrine cells of the small intestine. Several GPR40 agonists, including AMG 837 and TAK-875, have been disclosed, but no GPR40 synthetic agonists have been reported that engage both the insulinogenic and incretinogenic axes. In this report we provide a molecular explanation and describe the discovery of a unique and potent class of GPR40 full agonists that engages the enteroinsular axis to promote dramatic improvement in glucose control in rodents. GPR40 full agonists AM-1638 and AM-6226 stimulate GLP-1 and GIP secretion from intestinal enteroendocrine cells and increase GSIS from pancreatic islets, leading to enhanced glucose control in the high fat fed, streptozotocin treated and NONcNZO10/LtJ mouse models of type 2 diabetes. The improvement in hyperglycemia by AM-1638 was reduced in the presence of the GLP-1 receptor antagonist Ex(9–39)NH2.
Collapse
Affiliation(s)
- Jian Luo
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Gayathri Swaminath
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Sean P. Brown
- Department of Therapeutic Discovery, Amgen Inc., South San Francisco, California, United States of America
| | - Jane Zhang
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Qi Guo
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Michael Chen
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Kathy Nguyen
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Thanhvien Tran
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Lynn Miao
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Paul J. Dransfield
- Department of Therapeutic Discovery, Amgen Inc., South San Francisco, California, United States of America
| | - Marc Vimolratana
- Department of Therapeutic Discovery, Amgen Inc., South San Francisco, California, United States of America
| | - Jonathan B. Houze
- Department of Therapeutic Discovery, Amgen Inc., South San Francisco, California, United States of America
| | - Simon Wong
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California, United States of America
| | - Maria Toteva
- Department of Pharmaceutics, Amgen Inc., South San Francisco, California, United States of America
| | - Bei Shan
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Frank Li
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Run Zhuang
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
| | - Daniel C.-H. Lin
- Department of Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
17
|
Feng XT, Leng J, Xie Z, Li SL, Zhao W, Tang QL. GPR40: a therapeutic target for mediating insulin secretion (review). Int J Mol Med 2012; 30:1261-6. [PMID: 23023155 DOI: 10.3892/ijmm.2012.1142] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 08/26/2012] [Indexed: 11/05/2022] Open
Abstract
G-protein-coupled receptor 40 (GPR40), known as free fatty acid receptor 1, is mainly expressed in pancreatic β-cells and activated by medium- and long-chain fatty acids. Increasing evidence indicates that the activation of GPR40 in cells causes insulin secretion, and GPR40 has become an attractive therapeutic target for type 2 diabetes. Recently, certain novel GPR40 agonists have been identified that regulate glucose-stimulated insulin secretion, leading to the development of new drugs for the treatment of type 2 diabetes. In this review, we focus on progress in the physiological role of GPR40 and potential drugs targeting GPR40 over the past decade.
Collapse
Affiliation(s)
- Xiao-Tao Feng
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, P.R. China
| | | | | | | | | | | |
Collapse
|
18
|
Talukdar S, Olefsky JM, Osborn O. Targeting GPR120 and other fatty acid-sensing GPCRs ameliorates insulin resistance and inflammatory diseases. Trends Pharmacol Sci 2011; 32:543-50. [PMID: 21663979 DOI: 10.1016/j.tips.2011.04.004] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 04/25/2011] [Accepted: 04/29/2011] [Indexed: 01/08/2023]
Abstract
The past decade has seen great progress in the understanding of the molecular pharmacology, physiological function and therapeutic potential of G-protein-coupled receptors (GPCRs). Free fatty acids (FFAs) have been demonstrated to act as ligands of several GPCRs including GPR40, GPR43, GPR84, GPR119 and GPR120. We have recently shown that GPR120 acts as a physiological receptor of ω3 fatty acids in macrophages and adipocytes, which mediate potent anti-inflammatory and insulin sensitizing effects. The important role GPR120 plays in the control of inflammation raises the possibility that targeting this receptor could have therapeutic potential in many inflammatory diseases including obesity and type 2 diabetes. In this review paper, we discuss lipid-sensing GPCRs and highlight potential outcomes of targeting such receptors in ameliorating disease.
Collapse
Affiliation(s)
- Saswata Talukdar
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA 92093, USA.
| | | | | |
Collapse
|
19
|
DC260126, a small-molecule antagonist of GPR40, improves insulin tolerance but not glucose tolerance in obese Zucker rats. Biomed Pharmacother 2011; 64:647-51. [PMID: 20888730 DOI: 10.1016/j.biopha.2010.06.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Accepted: 06/19/2010] [Indexed: 01/21/2023] Open
Abstract
GPR40 is a G-protein-coupled receptor specifically expressed in pancreatic islets, which maybe mediate both acute and chronic effects of free fatty acids (FFAs) on β-cell function. However, it is still a matter of debate whether GPR40 represents a novel therapeutic target for type 2 diabetes. To this aim, we evaluated the effect of DC260126, a small-molecule antagonist of GPR40, on glucose and lipid metabolism in obese Zucker rats. Rats were treated intraperitoneally with 6 mg/kg of DC260126 for 8 weeks. DC260126 could significantly decrease serum insulin levels, improve insulin tolerance and increase Akt phosphorylation levels in liver, suggesting improved insulin sensitivity in DC260126-treated rats. However, DC260126 did not affect food intake, body weight, blood glucose and lipids. Throughout the experimental period, no significant difference in glucose tolerance was observed between the vehicle and DC260126-treated rats. These results indicate that GPR40 antagonists may not be beneficial for the treatment of type 2 diabetes, although GPR40 antagonists could improve insulin tolerance and increase insulin signaling in vivo.
Collapse
|
20
|
Holliday ND, Watson SJ, Brown AJH. Drug discovery opportunities and challenges at g protein coupled receptors for long chain free Fatty acids. Front Endocrinol (Lausanne) 2011; 2:112. [PMID: 22649399 PMCID: PMC3355945 DOI: 10.3389/fendo.2011.00112] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 12/15/2011] [Indexed: 01/13/2023] Open
Abstract
Discovery of G protein coupled receptors for long chain free fatty acids (FFAs), FFA1 (GPR40) and GPR120, has expanded our understanding of these nutrients as signaling molecules. These receptors have emerged as important sensors for FFA levels in the circulation or the gut lumen, based on evidence from in vitro and rodent models, and an increasing number of human studies. Here we consider their promise as therapeutic targets for metabolic disease, including type 2 diabetes and obesity. FFA1 directly mediates acute FFA-induced glucose-stimulated insulin secretion in pancreatic beta-cells, while GPR120 and FFA1 trigger release of incretins from intestinal endocrine cells, and so indirectly enhance insulin secretion and promote satiety. GPR120 signaling in adipocytes and macrophages also results in insulin sensitizing and beneficial anti-inflammatory effects. Drug discovery has focused on agonists to replicate acute benefits of FFA receptor signaling, with promising early results for FFA1 agonists in man. Controversy surrounding chronic effects of FFA1 on beta-cells illustrates that long term benefits of antagonists also need exploring. It has proved challenging to generate highly selective potent ligands for FFA1 or GPR120 subtypes, given that both receptors have hydrophobic orthosteric binding sites, which are not completely defined and have modest ligand affinity. Structure activity relationships are also reliant on functional read outs, in the absence of robust binding assays to provide direct affinity estimates. Nevertheless synthetic ligands have already helped dissect specific contributions of FFA1 and GPR120 signaling from the many possible cellular effects of FFAs. Approaches including use of fluorescent ligand binding assays, and targeting allosteric receptor sites, may improve further pre-clinical ligand development at these receptors, to exploit their unique potential to target multiple facets of diabetes.
Collapse
Affiliation(s)
- Nicholas D. Holliday
- Cell Signalling Research Group, School of Biomedical Sciences, The Medical School, Queen’s Medical Centre, University of NottinghamNottingham, UK
- *Correspondence: Nicholas D. Holliday, Cell Signalling Research Group, School of Biomedical Sciences, The Medical School, Queen’s Medical Centre, University of Nottingham, Floor C, Nottingham NG7 2UH, UK. e-mail:
| | - Sarah-Jane Watson
- Cell Signalling Research Group, School of Biomedical Sciences, The Medical School, Queen’s Medical Centre, University of NottinghamNottingham, UK
| | | |
Collapse
|
21
|
Hudson B, Smith NJ, Milligan G. Experimental Challenges to Targeting Poorly Characterized GPCRs: Uncovering the Therapeutic Potential for Free Fatty Acid Receptors. PHARMACOLOGY OF G PROTEIN COUPLED RECEPTORS 2011; 62:175-218. [DOI: 10.1016/b978-0-12-385952-5.00006-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
22
|
Lin DCH, Zhang J, Zhuang R, Li F, Nguyen K, Chen M, Tran T, Lopez E, Lu JYL, Li XN, Tang L, Tonn GR, Swaminath G, Reagan JD, Chen JL, Tian H, Lin YJ, Houze JB, Luo J. AMG 837: a novel GPR40/FFA1 agonist that enhances insulin secretion and lowers glucose levels in rodents. PLoS One 2011; 6:e27270. [PMID: 22087278 PMCID: PMC3210765 DOI: 10.1371/journal.pone.0027270] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 10/13/2011] [Indexed: 01/25/2023] Open
Abstract
Agonists of GPR40 (FFA1) have been proposed as a means to treat type 2 diabetes. Through lead optimization of a high throughput screening hit, we have identified a novel GPR40 agonist called AMG 837. The objective of these studies was to understand the preclinical pharmacological properties of AMG 837. The activity of AMG 837 on GPR40 was characterized through GTPγS binding, inositol phosphate accumulation and Ca(2+) flux assays. Activity of AMG 837 on insulin release was assessed on isolated primary mouse islets. To determine the anti-diabetic activity of AMG 837 in vivo, we tested AMG 837 using a glucose tolerance test in normal Sprague-Dawley rats and obese Zucker fatty rats. AMG 837 was a potent partial agonist in the calcium flux assay on the GPR40 receptor and potentiated glucose stimulated insulin secretion in vitro and in vivo. Acute administration of AMG 837 lowered glucose excursions and increased glucose stimulated insulin secretion during glucose tolerance tests in both normal and Zucker fatty rats. The improvement in glucose excursions persisted following daily dosing of AMG 837 for 21-days in Zucker fatty rats. Preclinical studies demonstrated that AMG 837 was a potent GPR40 partial agonist which lowered post-prandial glucose levels. These studies support the potential utility of AMG 837 for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Daniel C-H Lin
- Metabolic Disorders, Amgen Inc., South San Francisco, California, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Yang M, Chisholm JW, Soelaiman S, Shryock JC. Sulfonylureas uncouple glucose-dependence for GPR40-mediated enhancement of insulin secretion from INS-1E cells. Mol Cell Endocrinol 2010; 315:308-13. [PMID: 19815053 DOI: 10.1016/j.mce.2009.09.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Accepted: 09/28/2009] [Indexed: 10/20/2022]
Abstract
Activation of GPR40 is reported to enhance insulin secretion in the presence of glucose. We determined whether sulfonylureas could replace glucose for GPR40-mediated enhancement of insulin secretion and investigated underlying mechanisms using INS-1E cells. GW9508, a specific agonist of GPR40, significantly enhanced insulin secretion in the presence of high concentrations of glucose. In contrast, sulfonylureas increased insulin secretion in the absence of glucose. In the presence of sulfonylureas, activation of GPR40 significantly enhanced insulin secretion. The L-type calcium channel (LTCC) activator S-(-)-Bay K8644 also concentration-dependently increased insulin secretion in the absence of glucose. In the presence of 10 micromol/L S-(-)-Bay K8644, GW9508 significantly increased insulin secretion. On the other hand, the LTCC blocker nifedipine significantly inhibited insulin secretion mediated by either glucose, glipizide or glucose plus GW9508. Thus, sulfonylureas could replace glucose to support GPR40-mediated enhancement of insulin secretion, whereas blockage of LTCC reduced both glucose and sulfonylurea-mediated insulin secretion.
Collapse
Affiliation(s)
- Ming Yang
- Biology, Gilead Sciences, Inc., 3172 Porter Drive, Palo Alto, CA 94304, USA.
| | | | | | | |
Collapse
|
24
|
|
25
|
Karaki SI, Kuwahara A. Roles of Short-Chain Fatty Acids and their Receptors in Colonic Motility. Biosci Microflora 2010. [DOI: 10.12938/bifidus.29.31] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Shin-ichiro Karaki
- Laboratory of Physiology, Institute for Environmental Sciences, University of Shizuoka
| | - Atsukazu Kuwahara
- Laboratory of Physiology, Institute for Environmental Sciences, University of Shizuoka
| |
Collapse
|
26
|
Abstract
G-protein coupled receptors (GPCRs) are targets of approximately 30% of currently marketed drugs. Over the last few years, a number of GPCRs expressed in pancreatic beta-cells and activated by lipids have been discovered. GPR40 was shown to be activated by medium- to long-chain fatty acids (FAs). It has since been shown that GPR40 contributes to FA amplification of glucose-induced insulin secretion. Although some controversy still exists as to whether GPR40 agonists or antagonists should be designed as novel type 2 diabetes drugs, data obtained in our laboratory and others strongly suggest that GPR40 agonism might represent a valuable therapeutic approach. GPR119 is expressed in pancreatic beta-cells and enteroendocrine L-cells, and augments circulating insulin levels both through its direct insulinotropic action on beta-cells and through FA stimulation of glucagon-like peptide 1 (GLP-1) secretion. GPR120 is expressed in L-cells and was also shown to mediate FA-stimulated GLP-1 release. Finally, GPR41 and GPR43 are receptors for short-chain FAs and may indirectly regulate beta-cell function via adipokine secretion. Although the discovery of these various lipid receptors opens new and exciting avenues of research for drug development, a number of questions regarding their mechanisms of action and physiological roles remain to be answered.
Collapse
Affiliation(s)
- M A Kebede
- Montréal Diabetes Research Center, CRCHUM, QC, Canada
| | | | | | | |
Collapse
|
27
|
Gilbertson T, Yu T, Shah B. Gustatory Mechanisms for Fat Detection. Front Neurosci 2009. [DOI: 10.1201/9781420067767-c3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
28
|
Morgan NG, Dhayal S. G-protein coupled receptors mediating long chain fatty acid signalling in the pancreatic beta-cell. Biochem Pharmacol 2009; 78:1419-27. [PMID: 19660440 DOI: 10.1016/j.bcp.2009.07.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 07/27/2009] [Accepted: 07/27/2009] [Indexed: 11/16/2022]
Abstract
It is increasingly clear that some of the effects of both free and derivatised long chain fatty acids in pancreatic beta-cells are mediated by a group of G-protein coupled receptors. Some of these display close structural homology while others are more divergent. This Commentary reviews the expression and functional roles of three such molecules, GPR40, GPR119 and GPR120. GPR40 is the best characterised of this group and appears to mediate the acute stimulatory effects of long chain fatty acids on insulin secretion. GPR40 has also been proposed as a potential mediator of fatty acid toxicity but this is more controversial. GPR119 is also involved in stimulation of insulin secretion and responds primarily to ethanolamide derivatives of long chain fatty acids and also to some lysophospholipids rather than to free fatty acids. It may represent a useful target for the development of new insulin secretagogues aimed to enhance insulin release in patients with type 2 diabetes. GPR120 is the most enigmatic of the lipid-responsive cell-surface receptors and its function remains to be established. It has been proposed to play a cytoprotective role in certain other cell types but it is unclear whether it fulfils a similar function in beta-cells.
Collapse
Affiliation(s)
- Noel G Morgan
- Institute of Biomedical & Clinical Science, John Bull Building, Peninsula Medical School, Plymouth PL6 8BU, UK.
| | | |
Collapse
|
29
|
Nagasumi K, Esaki R, Iwachidow K, Yasuhara Y, Ogi K, Tanaka H, Nakata M, Yano T, Shimakawa K, Taketomi S, Takeuchi K, Odaka H, Kaisho Y. Overexpression of GPR40 in pancreatic beta-cells augments glucose-stimulated insulin secretion and improves glucose tolerance in normal and diabetic mice. Diabetes 2009; 58:1067-76. [PMID: 19401434 PMCID: PMC2671040 DOI: 10.2337/db08-1233] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE GPR40 is a G protein-coupled receptor regulating free fatty acid-induced insulin secretion. We generated transgenic mice overexpressing the hGPR40 gene under control of the mouse insulin II promoter and used them to examine the role of GPR40 in the regulation of insulin secretion and glucose homeostasis. RESEARCH DESIGN AND METHODS Normal (C57BL/6J) and diabetic (KK) mice overexpressing the hGPR40 gene under control of the insulin II promoter were generated, and their glucose metabolism and islet function were analyzed. RESULTS In comparison with nontransgenic littermates, hGPR40 transgenic mice exhibited improved oral glucose tolerance with an increase in insulin secretion. Although islet morphologic analysis showed no obvious differences between hGPR40 transgenic and nontransgenic mice, isolated islets from hGPR40 transgenic mice had enhanced insulin secretion in response to high glucose (16 mmol/l) compared with those from nontransgenic mice, and they both had similar low glucose (3 mmol/l)-stimulated insulin secretion. In addition, hGPR40 transgenic islets significantly increased insulin secretion against a naturally occurring agonist palmitate in the presence of 11 mmol/l glucose. hGPR40 transgenic mice were also found to be resistant to high-fat diet-induced glucose intolerance, and hGPR40 transgenic mice harboring KK background showed augmented insulin secretion and improved oral glucose tolerance compared with nontransgenic littermates. CONCLUSIONS Our results suggest that GPR40 may have a role in regulating glucose-stimulated insulin secretion and plasma glucose levels in vivo and that pharmacological activation of GPR40 may provide a novel insulin secretagogue beneficial for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Kae Nagasumi
- Pharmaceutical Research Division, Pharmacology [corrected] Research Laboratories I, Takeda Pharmaceutical Company Limited, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Stoddart LA, Smith NJ, Milligan G. International Union of Pharmacology. LXXI. Free fatty acid receptors FFA1, -2, and -3: pharmacology and pathophysiological functions. Pharmacol Rev 2008; 60:405-17. [PMID: 19047536 DOI: 10.1124/pr.108.00802] [Citation(s) in RCA: 252] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Identification of G protein-coupled receptors that are activated by free fatty acids has led to considerable interest in their pharmacology and function because of the wide range of normal physiology and disease states in which fatty acids have been implicated. Free fatty acid receptor (FFA) 1 is activated by medium- to long-chain fatty acids and is expressed in the insulin-producing beta-cells of the pancreas. Activation of FFA1 has been proposed to mediate fatty acid augmentation of glucose-stimulated insulin secretion although it is unclear whether the known long-term detrimental effects of beta-cell exposure to high levels of fatty acids are also mediated through this receptor. The related receptors FFA2 and FFA3 are both activated by short-chain fatty acids although they have key differences in the signaling pathways they activate and tissue expression pattern. The aim of this review is to provide a comprehensive overview of the current understanding of the pharmacology and physiological role of these fatty acid receptors.
Collapse
Affiliation(s)
- Leigh A Stoddart
- Molecular Pharmacology Group, Neuroscience and Molecular Pharmacology, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom.
| | | | | |
Collapse
|
31
|
Vettor R, Granzotto M, De Stefani D, Trevellin E, Rossato M, Farina MG, Milan G, Pilon C, Nigro A, Federspil G, Vigneri R, Vitiello L, Rizzuto R, Baratta R, Frittitta L. Loss-of-function mutation of the GPR40 gene associates with abnormal stimulated insulin secretion by acting on intracellular calcium mobilization. J Clin Endocrinol Metab 2008; 93:3541-50. [PMID: 18583466 DOI: 10.1210/jc.2007-2680] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Free fatty acids (FFAs) acutely stimulate but chronically impair glucose-stimulated insulin secretion from beta-cells. The G protein-coupled transmembrane receptor 40 (GPR40) mediates both acute and chronic effects of FFAs on insulin secretion and plays a role in glucose homeostasis. Limited information is available on the effect of GPR40 genetic abnormalities on insulin secretion and metabolic regulation in human subjects. STUDY DESIGN AND RESULTS For in vivo studies, we screened 734 subjects for the coding region of GPR40 and identified a new single-nucleotide mutation (Gly180Ser). The mean allele frequency was 0.75%, which progressively increased (P < 0.05) from nonobese subjects (0.42%) to moderately obese (body mass index = 30-39.9 kg/m2, 1.07%) and severely obese patients (body mass index > or = 40 kg/m2, 2.60%). The relationship between the GPR40 mutation, insulin secretion, and metabolic alterations was studied in 11 Gly/Ser mutation carriers. In these subjects, insulin secretion (insulinogenic index derived from oral glucose tolerance test) was significantly lower than in 692 Gly/Gly carriers (86.0 +/- 48.2 vs. 183.7 +/- 134.4, P < 0.005). Moreover, a case-control study indicated that plasma insulin and C-peptide responses to a lipid load were significantly (P < 0.05) lower in six Gly/Ser than in 12 Gly/Gly carriers. In vitro experiments in HeLa cells cotransfected with aequorin and the mutated Gly/Ser GPR40 indicated that intracellular Ca2+ concentration increase after oleic acid was significantly lower than in Gly/Gly GPR40-transfected cells. This fact was confirmed using fura-2 acetoxymethyl ester. CONCLUSIONS This newly identified GPR40 variant results in a loss of function that prevents the beta-cell ability to adequately sense lipids as an insulin secretory stimulus because of impaired intracellular Ca2+ concentration increase.
Collapse
Affiliation(s)
- Roberto Vettor
- Endocrine-Metabolic Laboratory, Internal Medicine, Department of Medical and Surgical Sciences, University of Padova, via Ospedale, 105, I-35128 Padova, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Rayasam GV, Tulasi VK, Davis JA, Bansal VS. Fatty acid receptors as new therapeutic targets for diabetes. Expert Opin Ther Targets 2007; 11:661-71. [PMID: 17465724 DOI: 10.1517/14728222.11.5.661] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
G-protein-coupled receptors (GPCRs) are key regulators of several physiological functions. Their roles in cellular signal transduction have made them the target for majority of all currently prescribed drugs. Additionally, there are many orphan GPCRs that provide potential novel therapeutic targets. Several GPCRs are involved in metabolic regulation and glucose homeostasis such as GLP-1 receptor, glucagon receptor, adiponectin receptor and so on. Recently, free fatty acids (FFAs) have been demonstrated as ligands for orphan GPCRs and have been proposed to play a critical role in physiological glucose homeostasis. GPR40 and GPR120 are activated by medium and long-chain FFAs, whereas GPR41 and GPR43 can be activated by short-chain FFAs. GPR40, which is preferentially expressed in pancreatic beta-cells, mediates the majority of the effects of FFAs on insulin secretion. In this review, these findings and also critical analysis of these GPCRs as novel targets for diabetes are discussed.
Collapse
MESH Headings
- Animals
- Bacteria/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Dietary Carbohydrates/metabolism
- Drug Delivery Systems
- Drug Design
- Fatty Acids, Nonesterified/metabolism
- Fermentation
- Humans
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin/metabolism
- Insulin Resistance
- Insulin Secretion
- Intestines/microbiology
- Liver/metabolism
- Mice
- Muscle, Skeletal/metabolism
- Obesity/complications
- Obesity/drug therapy
- Obesity/metabolism
- Rats
- Receptors, Cell Surface/drug effects
- Receptors, Cell Surface/physiology
- Receptors, G-Protein-Coupled/administration & dosage
- Receptors, G-Protein-Coupled/drug effects
- Receptors, G-Protein-Coupled/physiology
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Geetha Vani Rayasam
- Metabolic Disorders, Department of Pharmacology, Research & Development (R&D III), Ranbaxy, Sector 18, Gurgaon, Haryana, India.
| | | | | | | |
Collapse
|
33
|
Flodgren E, Olde B, Meidute-Abaraviciene S, Winzell MS, Ahrén B, Salehi A. GPR40 is expressed in glucagon producing cells and affects glucagon secretion. Biochem Biophys Res Commun 2007; 354:240-5. [PMID: 17214971 DOI: 10.1016/j.bbrc.2006.12.193] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Accepted: 12/23/2006] [Indexed: 10/23/2022]
Abstract
The free fatty acid receptor, GPR40, has been coupled with insulin secretion via its expression in pancreatic beta-cells. However, the role of GPR40 in the release of glucagon has not been studied and previous attempts to identify the receptor in alpha-cells have been unfruitful. Using double-staining for glucagon and GPR40 expression, we demonstrate that the two are expressed in the same cells in the periphery of mouse islets. In-R1-G9 hamster glucagonoma cells respond dose-dependently to linoleic acid stimulation by elevated phosphatidyl inositol hydrolysis and glucagon release and the cells become increasingly responsive to fatty acid stimulation when overexpressing GPR40. Isolated mouse islets also secrete glucagon in response to linoleic acid, a response that was abolished by antisense treatment against GPR40. This study demonstrates that GPR40 is present and active in pancreatic alpha-cells and puts further emphasis on the importance of this nutrient sensing receptor in islet function.
Collapse
Affiliation(s)
- Erik Flodgren
- Department of Clinical Sciences, Biomedical Center, Lund University, Lund, Sweden.
| | | | | | | | | | | |
Collapse
|