51
|
Expression of the candidate fat taste receptors in human fungiform papillae and the association with fat taste function. Br J Nutr 2018; 120:64-73. [DOI: 10.1017/s0007114518001265] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractSignificant experimental evidence supports fat as a taste modality; however, the associated peripheral mechanisms are not well established. Several candidate taste receptors have been identified, but their expression pattern and potential functions in human fungiform papillae remain unknown. The aim of this study is to identify the fat taste candidate receptors and ion channels that were expressed in human fungiform taste buds and their association with oral sensory of fatty acids. For the expression analysis, quantitative RT-PCR (qRT-PCR) from RNA extracted from human fungiform papillae samples was used to determine the expression of candidate fatty acid receptors and ion channels. Western blotting analysis was used to confirm the presence of the proteins in fungiform papillae. Immunohistochemistry analysis was used to localise the expressed receptors or ion channels in the taste buds of fungiform papillae. The correlation study was analysed between the expression level of the expressed fat taste receptors or ion channels indicated by qRT-PCR and fat taste threshold, liking of fatty food and fat intake. As a result, qRT-PCR and western blotting indicated that mRNA and protein of CD36, FFAR4, FFAR2, GPR84 and delayed rectifying K+ channels are expressed in human fungiform taste buds. The expression level of CD36 was associated with the liking difference score (R −0·567, β=−0·04, P=0·04) between high-fat and low-fat food and FFAR2 was associated with total fat intake (ρ=−0·535, β=−0·01, P=0·003) and saturated fat intake (ρ=−0·641, β=−0·02, P=0·008).
Collapse
|
52
|
Eleazu C, Charles A, Eleazu K, Achi N. Free fatty acid receptor 1 as a novel therapeutic target for type 2 diabetes mellitus-current status. Chem Biol Interact 2018; 289:32-39. [PMID: 29704509 DOI: 10.1016/j.cbi.2018.04.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/21/2018] [Accepted: 04/24/2018] [Indexed: 02/07/2023]
Abstract
The incidence of type 2 diabetes mellitus (T2DM) has been on the increase in recent times. Although several oral treatments for T2DM are available, some of them have been found to elicit undesirable side effects. This therefore underscores the need for new treatment options with lesser side effects than the existing ones for people with T2DM. Free fatty acid receptor 1 (FFAR1), also known as GPR40, belongs to a class of G-protein coupled receptors that are encoded by FFAR1 genes in humans. It is expressed in the pancreatic β-cells and it is activated by medium- and long-chain saturated and unsaturated fatty acids. Thus it responds to endogenous medium and long chain unsaturated fatty acids, resulting in enhancement of insulin secretion during increased glucose levels. The glucose dependency of insulin secretion has made this receptor a very good target for developing therapies that could be efficacious with fewer side effects than the existing therapies for the treatment of T2DM. Given that tremendous efforts have been made in recent times in developing novel FFAR1 agonists with antidiabetic potentials, this article provides a current status of knowledge on the efforts made so far in developing novel FFAR1 agonists that would be of relevance in the management of T2DM.
Collapse
Affiliation(s)
- Chinedum Eleazu
- Federal University, Ndufu-Alike, Ikwo, Ebonyi State, Nigeria.
| | - Ayogu Charles
- Federal University, Ndufu-Alike, Ikwo, Ebonyi State, Nigeria
| | - Kate Eleazu
- Ebonyi State University Abakaliki, Ebonyi State, Nigeria
| | - Ngozi Achi
- Michael Okpara University of Agriculture, Umudike, Nigeria
| |
Collapse
|
53
|
A Newly Discovered Antifibrotic Pathway Regulated by Two Fatty Acid Receptors. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1132-1148. [DOI: 10.1016/j.ajpath.2018.01.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/20/2017] [Accepted: 01/16/2018] [Indexed: 01/12/2023]
|
54
|
Yamaguchi M, Samuchiwal SK, Quehenberger O, Boyce JA, Balestrieri B. Macrophages regulate lung ILC2 activation via Pla2g5-dependent mechanisms. Mucosal Immunol 2018; 11:615-626. [PMID: 29346348 PMCID: PMC5976507 DOI: 10.1038/mi.2017.99] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/17/2017] [Indexed: 02/04/2023]
Abstract
Group V phospholipase A2 (Pla2g5) is a lipid-generating enzyme necessary for macrophage effector functions in pulmonary inflammation. However, the lipid mediators involved and their cellular targets have not been identified. Mice lacking Pla2g5 showed markedly reduced lung ILC2 activation and eosinophilia following repetitive Alternaria Alternata inhalation. While Pla2g5-null mice had Wt levels of immediate IL-33 release after one Alternaria dose, they failed to upregulate IL-33 in macrophages following repeated Alternaria administration. Unexpectedly, while adoptive transfer of bone marrow-derived (BM)-macrophages restored ILC2 activation and eosinophilia in Alternaria-exposed Pla2g5-null mice, exogenous IL-33 did not. Conversely, transfers of Pla2g5-null BM-macrophages reduced inflammation in Alternaria-exposed Wt mice. Mass spectrometry analysis of free fatty acids (FFAs) demonstrated significantly reduced FFAs (including linoleic acid (LA) and oleic acid (OA)) in lung and BM-macrophages lacking Pla2g5. Exogenous administration of LA or LA+OA to Wt mice sharply potentiated IL-33-induced lung eosinophilia and ILC2 expansion in vitro and in vivo. In contrast, OA potentiated IL-33-induced inflammation and ILC2 expansion in Pla2g5-null mice, but LA was inactive both in vivo and in vitro. Notably, Pla2g5-null ILC2s showed significantly reduced expression of the FFA-receptor-1 compared to Wt ILC2s. Thus, macrophage-associated Pla2g5 contributes significantly to type-2 immunity through regulation of IL-33 induction and FFA-driven ILC2 activation.
Collapse
Affiliation(s)
- Munehiro Yamaguchi
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Sachin K. Samuchiwal
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Oswald Quehenberger
- Department of Medicine, Department of Pharmacology, University of California, San Diego, CA 92093, USA
| | - Joshua A. Boyce
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Barbara Balestrieri
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
55
|
New thiazolidinedione LPSF/GQ-2 inhibits NFκB and MAPK activation in LPS-induced acute lung inflammation. Int Immunopharmacol 2018; 57:91-101. [PMID: 29475100 DOI: 10.1016/j.intimp.2018.02.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/26/2018] [Accepted: 02/14/2018] [Indexed: 12/22/2022]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are responsible for high mortality rates in critical patients. Despite >50 years of intensive research, there is no pharmacologically effective treatment to treat ALI. PPARs agonists, chemically named thiazolidinediones (TZDs) have emerged as potential drugs for the treatment of ALI and ARDS due to their anti-inflammatory efficacy. The present study aims to evaluate the potential anti-inflammatory effects of new TZDs derivatives, LPSF/GQ-2 and LPSF/RA-4, on ALI induced by LPS. BALB/c mice were divided into five groups: 1) Control; 2) LPS intranasal 25 μg; 3) LPSF/GQ-2 30 mg/kg + LPS; 4) LPSF/RA-4 20 mg/kg + LPS; and 5) DEXA 1 mg/Kg + LPS. BALF analyses revealed that LPSF/GQ-2 and LPSF/RA-4 reduced NO levels in BALF and inflammatory cell infiltration induced by LPS. MPO levels were also reduced by the LPSF/GQ-2 and LPSF/RA-4 pre-treatments. In contrast, histopathological analyses showed better tissue protection with LPSF/GQ-2 than DEXA and LPSF/RA-4 groups. Similarly, LPSF/GQ-2 reduced inflammatory markers (IL-1, iNOS, TNFα, IL-1β, IL-6) better than LPSF/RA-4. The LPSF/GQ-2 anti-inflammatory action could be attributed to the inhibition of NFκB, ERK, p38, and PARP pathways. In contrast, LPSF/RA-4 had no effect on the expression of p38, JNK, NFκB. The present study indicates that LPSF/GQ-2 presents a potential therapeutic role as an anti-inflammatory drug for ALI.
Collapse
|
56
|
Discovery of a novel potent GPR40 full agonist. Bioorg Med Chem Lett 2018; 28:720-726. [DOI: 10.1016/j.bmcl.2018.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/10/2018] [Accepted: 01/11/2018] [Indexed: 11/18/2022]
|
57
|
Meza-Cuenca F, Medina-Contreras JML, Mailloux-Salinas P, Bautista-Hernández LA, Buentello-Volante B, Domínguez-López A, Garfias Y, Correa-López PV, Bautista de Lucio VM, Bravo G. Characterization of free fatty acid receptors expression in an obesity rat model with high sucrose diet. J Recept Signal Transduct Res 2018; 38:76-82. [PMID: 29369009 DOI: 10.1080/10799893.2018.1426609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION/AIMS In recent years, it has been shown that free fatty acids receptors (FFAR) of whose function in the cell surface plays a significant role in the regulation of cell function and nutrition as well are activated by various endogenous ligands, but mainly by fatty acids. Within FFAR of our interest are GPR 41, 43 and 120. The functions of these receptors are varied and dependent on the tissue where they are. The activation and signaling of these receptors, FFAR, are involved in many physiological processes, and currently the target of many drugs in metabolic disorders like obesity, diabetes and atherosclerosis. MATERIAL AND METHODS Obesity was induced with hypercaloric diet (HD) in male Wistar rats for 20 weeks (n = 10). At the end, adipose tissue (abdominal and subcutaneous) was taken to perform assays for relative quantification mRNA expression by end-point RT-PCR and protein level expression by Western blot. RESULTS These present data have shown for the first time that total mRNA isolation and protein expression from both adipose tissues (abdominal and subcutaneous) of rat in obesity condition yield significative statistical difference among the control versus obese groups, showing that the diet high in carbohydrates modifies the total presence of mRNA and protein level expression of the receptors GPR41, 43 and 120. CONCLUSIONS Further comparative methods are in process to clarify whether or not the obesity changes the functional receptors in these two tissues for new pharmacological approaches.
Collapse
Affiliation(s)
- Fabián Meza-Cuenca
- a Pharmacobiology Department , Cinvestav-IPN Unidad Coapa , Mexico City , Mexico.,b Microbiology and Ocular Proteomics, Research Unit, Institute of Ophthalmology "Fundación de Asistencia Privada Conde de Valenciana" , Mexico City , Mexico
| | | | | | - Luis A Bautista-Hernández
- b Microbiology and Ocular Proteomics, Research Unit, Institute of Ophthalmology "Fundación de Asistencia Privada Conde de Valenciana" , Mexico City , Mexico
| | - Beatríz Buentello-Volante
- b Microbiology and Ocular Proteomics, Research Unit, Institute of Ophthalmology "Fundación de Asistencia Privada Conde de Valenciana" , Mexico City , Mexico
| | - Alfredo Domínguez-López
- b Microbiology and Ocular Proteomics, Research Unit, Institute of Ophthalmology "Fundación de Asistencia Privada Conde de Valenciana" , Mexico City , Mexico
| | - Yonathan Garfias
- b Microbiology and Ocular Proteomics, Research Unit, Institute of Ophthalmology "Fundación de Asistencia Privada Conde de Valenciana" , Mexico City , Mexico.,c Department of Biochemistry, Faculty of Medicine , Universidad Nacional Autónoma de México , Mexico City , Mexico
| | | | - Víctor M Bautista de Lucio
- b Microbiology and Ocular Proteomics, Research Unit, Institute of Ophthalmology "Fundación de Asistencia Privada Conde de Valenciana" , Mexico City , Mexico
| | - Guadalupe Bravo
- a Pharmacobiology Department , Cinvestav-IPN Unidad Coapa , Mexico City , Mexico
| |
Collapse
|
58
|
20-HETE promotes glucose-stimulated insulin secretion in an autocrine manner through FFAR1. Nat Commun 2018; 9:177. [PMID: 29330456 PMCID: PMC5766607 DOI: 10.1038/s41467-017-02539-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 12/07/2017] [Indexed: 01/02/2023] Open
Abstract
The long-chain fatty acid receptor FFAR1 is highly expressed in pancreatic β-cells. Synthetic FFAR1 agonists can be used as antidiabetic drugs to promote glucose-stimulated insulin secretion (GSIS). However, the physiological role of FFAR1 in β-cells remains poorly understood. Here we show that 20-HETE activates FFAR1 and promotes GSIS via FFAR1 with higher potency and efficacy than dietary fatty acids such as palmitic, linoleic, and α-linolenic acid. Murine and human β-cells produce 20-HETE, and the ω-hydroxylase-mediated formation and release of 20-HETE is strongly stimulated by glucose. Pharmacological inhibition of 20-HETE formation and blockade of FFAR1 in islets inhibits GSIS. In islets from type-2 diabetic humans and mice, glucose-stimulated 20-HETE formation and 20-HETE-dependent stimulation of GSIS are strongly reduced. We show that 20-HETE is an FFAR1 agonist, which functions as an autocrine positive feed-forward regulator of GSIS, and that a reduced glucose-induced 20-HETE formation contributes to inefficient GSIS in type-2 diabetes. FFAR1 receptor is highly expressed in beta cells and its activation has been suggested as therapy against type-2 diabetes. Here, Tunaru et al. show that 20-hydroxyeicosatetraenoic acid, produced within the islets upon glucose stimulation, acts in an autocrine manner to stimulate insulin secretion via FFAR1 activation.
Collapse
|
59
|
Rorsman P, Ashcroft FM. Pancreatic β-Cell Electrical Activity and Insulin Secretion: Of Mice and Men. Physiol Rev 2018; 98:117-214. [PMID: 29212789 PMCID: PMC5866358 DOI: 10.1152/physrev.00008.2017] [Citation(s) in RCA: 456] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/30/2017] [Accepted: 06/18/2017] [Indexed: 12/14/2022] Open
Abstract
The pancreatic β-cell plays a key role in glucose homeostasis by secreting insulin, the only hormone capable of lowering the blood glucose concentration. Impaired insulin secretion results in the chronic hyperglycemia that characterizes type 2 diabetes (T2DM), which currently afflicts >450 million people worldwide. The healthy β-cell acts as a glucose sensor matching its output to the circulating glucose concentration. It does so via metabolically induced changes in electrical activity, which culminate in an increase in the cytoplasmic Ca2+ concentration and initiation of Ca2+-dependent exocytosis of insulin-containing secretory granules. Here, we review recent advances in our understanding of the β-cell transcriptome, electrical activity, and insulin exocytosis. We highlight salient differences between mouse and human β-cells, provide models of how the different ion channels contribute to their electrical activity and insulin secretion, and conclude by discussing how these processes become perturbed in T2DM.
Collapse
Affiliation(s)
- Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Frances M Ashcroft
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
60
|
Cooper A, Singh S, Hook S, Tyndall JDA, Vernall AJ. Chemical Tools for Studying Lipid-Binding Class A G Protein-Coupled Receptors. Pharmacol Rev 2017; 69:316-353. [PMID: 28655732 DOI: 10.1124/pr.116.013243] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 05/15/2017] [Indexed: 12/16/2022] Open
Abstract
Cannabinoid, free fatty acid, lysophosphatidic acid, sphingosine 1-phosphate, prostanoid, leukotriene, bile acid, and platelet-activating factor receptor families are class A G protein-coupled receptors with endogenous lipid ligands. Pharmacological tools are crucial for studying these receptors and addressing the many unanswered questions surrounding expression of these receptors in normal and diseased tissues. An inherent challenge for developing tools for these lipid receptors is balancing the often lipophilic requirements of the receptor-binding pharmacophore with favorable physicochemical properties to optimize highly specific binding. In this study, we review the radioligands, fluorescent ligands, covalent ligands, and antibodies that have been used to study these lipid-binding receptors. For each tool type, the characteristics and design rationale along with in vitro and in vivo applications are detailed.
Collapse
Affiliation(s)
- Anna Cooper
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sameek Singh
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | | | | |
Collapse
|
61
|
Tsuda N, Kawaji A, Takagi M, Higashi C, Nakamura M, Hosaka Y, Sakaki J. Free fatty acid receptor 1 agonist, MR1704, lowers blood glucose levels in rats unresponsive to the sulfonylurea, glibenclamide. Drug Dev Res 2017; 79:16-21. [PMID: 29080222 DOI: 10.1002/ddr.21416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 10/12/2017] [Indexed: 12/22/2022]
Abstract
Preclinical Research & Development MR1704 is a selective G protein-coupled receptor 40/free fatty acid receptor 1 agonist, which exhibited favorable pharmacokinetic profiles and glucose-lowering effects in animal models. We studied the effects of MR1704 in a sulfonylurea-desensitized Sprague-Dawley rat model and evaluated the risk of pancreatic β-cell exhaustion compared to that of glibenclamide in Zucker fatty rats. Rats fed ad libitum a diet containing 0.03% glibenclamide exhibited lower non-fasting blood glucose levels compared to those in rats fed a control diet during the first 6 days. However, the response to glibenclamide disappeared on day 9. In a rat oral glucose tolerance test (OGTT), MR1704 reduced the plasma glucose excursion, whereas glibenclamide did not show this effect. In Zucker fatty rats, oral administration of MR1704 reduced glucose excursion during the OGTT, and the effects of MR1704 were maintained after 2-week treatment. In contrast, the glucose-lowering effects of glibenclamide were diminished, and glucose tolerance was aggravated after 2-week treatment. These results indicated that MR1704 provided more sustainable effects compared to those of the sulfonylurea, glibenclamide suggesting that MR1704 may be an attractive therapeutic option for diabetic patients who are unresponsive to sulfonylurea treatment.
Collapse
Affiliation(s)
- Naoto Tsuda
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Atsuko Kawaji
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Mitsuhiro Takagi
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Chika Higashi
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Masaki Nakamura
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Yoshitaka Hosaka
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Junichi Sakaki
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| |
Collapse
|
62
|
Tsuda N, Kawaji A, Sato T, Takagi M, Higashi C, Kato Y, Ogawa K, Naba H, Ohkouchi M, Nakamura M, Hosaka Y, Sakaki J. A novel free fatty acid receptor 1 (GPR40/FFAR1) agonist, MR1704, enhances glucose-dependent insulin secretion and improves glucose homeostasis in rats. Pharmacol Res Perspect 2017; 5. [PMID: 28805970 PMCID: PMC5684856 DOI: 10.1002/prp2.340] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/09/2017] [Accepted: 06/13/2017] [Indexed: 11/17/2022] Open
Abstract
Activation of G protein‐coupled receptor 40/Free fatty acid receptor 1 (GPR40/FFAR1), which is highly expressed in pancreatic β cells, is considered an important pharmacologic target for the treatment of type 2 diabetes mellitus. The aim of this study was to determine the effect of MR1704, a novel GPR40/FFAR1 agonist, on glucose homeostasis in rats. MR1704 is a highly potent and selective, orally bioavailable agonist with similar in vitro potencies among humans, mice, and rats. Treatment of rat islets with MR1704 increased glucose‐dependent insulin secretion. Augmentation of glucose‐dependent insulin secretion was abolished by adding a GPR40/FFAR1 antagonist. In mouse, insulinoma MIN6 cells, palmitic acid induced the activity of caspase 3/7 after a 72‐h exposure, while pharmacologically active concentrations of MR1704 did not. In an oral glucose tolerance test in normal Sprague‐Dawley rats, orally administered MR1704 (1–10 mg·kg−1) reduced plasma glucose excursion and enhanced insulin secretion, but MR1704 did not induce hypoglycemia, even at 300 mg·kg−1, in fasted Sprague‐Dawley rats. In addition, orally administered MR1704 reduced plasma glucose excursion and enhanced insulin secretion in diabetic Goto‐Kakizaki rats. Oral administration of MR1704 once daily to Goto‐Kakizaki rats reduced their blood glucose levels during a 5‐week treatment period without reducing pancreatic insulin content; as a result, hemoglobin A1C levels significantly decreased. These results suggest that MR1704 improves glucose homeostasis through glucose‐dependent insulin secretion with a low risk of hypoglycemia and pancreatic toxicity. MR1704 shows promise as a new, glucose‐lowering drug to treat type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Naoto Tsuda
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Atsuko Kawaji
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Toshihiro Sato
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Mitsuhiro Takagi
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Chika Higashi
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Yutaka Kato
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Kumiko Ogawa
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Hiroyasu Naba
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Munetaka Ohkouchi
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Masaki Nakamura
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Yoshitaka Hosaka
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| | - Junichi Sakaki
- Research Center, Mochida Pharmaceutical Company Limited, Shizuoka, Japan
| |
Collapse
|
63
|
Targeting Peroxisome Proliferator-Activated Receptors Using Thiazolidinediones: Strategy for Design of Novel Antidiabetic Drugs. INTERNATIONAL JOURNAL OF MEDICINAL CHEMISTRY 2017; 2017:1069718. [PMID: 28656106 PMCID: PMC5474549 DOI: 10.1155/2017/1069718] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/23/2017] [Accepted: 05/07/2017] [Indexed: 11/18/2022]
Abstract
Thiazolidinediones are a class of well-established antidiabetic drugs, also named as glitazones. Thiazolidinedione structure has been an important structural domain of research, involving design and development of new drugs for the treatment of type 2 diabetes. Extensive research on the mechanism of action and the structural requirements has revealed that the intended antidiabetic activity in type 2 diabetes is due to their agonistic effect on peroxisome proliferator-activated receptor (PPAR) belonging to the nuclear receptor super family. Glitazones have specific affinity to PPARγ, one of the subtypes of PPARs. Certain compounds under development have dual PPARα/γ agonistic activity which might be beneficial in obesity and diabetic cardiomyopathy. Interesting array of hybrid compounds of thiazolidinedione PPARγ agonists exhibited therapeutic potential beyond antidiabetic activity. Pharmacology and chemistry of thiazolidinediones as PPARγ agonists and the potential of newer analogues as dual agonists of PPARs and other emerging targets for the therapy of type 2 diabetes are presented. This review highlights the possible modifications of the structural components in the general frame work of thiazolidinediones with respect to their binding efficacy, potency, and selectivity which would guide the future research in design of novel thiazolidinedione derivatives for the management of type 2 diabetes.
Collapse
|
64
|
Lu J, Byrne N, Wang J, Bricogne G, Brown FK, Chobanian HR, Colletti SL, Di Salvo J, Thomas-Fowlkes B, Guo Y, Hall DL, Hadix J, Hastings NB, Hermes JD, Ho T, Howard AD, Josien H, Kornienko M, Lumb KJ, Miller MW, Patel SB, Pio B, Plummer CW, Sherborne BS, Sheth P, Souza S, Tummala S, Vonrhein C, Webb M, Allen SJ, Johnston JM, Weinglass AB, Sharma S, Soisson SM. Structural basis for the cooperative allosteric activation of the free fatty acid receptor GPR40. Nat Struct Mol Biol 2017; 24:570-577. [PMID: 28581512 DOI: 10.1038/nsmb.3417] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 05/08/2017] [Indexed: 12/19/2022]
Abstract
Clinical studies indicate that partial agonists of the G-protein-coupled, free fatty acid receptor 1 GPR40 enhance glucose-dependent insulin secretion and represent a potential mechanism for the treatment of type 2 diabetes mellitus. Full allosteric agonists (AgoPAMs) of GPR40 bind to a site distinct from partial agonists and can provide additional efficacy. We report the 3.2-Å crystal structure of human GPR40 (hGPR40) in complex with both the partial agonist MK-8666 and an AgoPAM, which exposes a novel lipid-facing AgoPAM-binding pocket outside the transmembrane helical bundle. Comparison with an additional 2.2-Å structure of the hGPR40-MK-8666 binary complex reveals an induced-fit conformational coupling between the partial agonist and AgoPAM binding sites, involving rearrangements of the transmembrane helices 4 and 5 (TM4 and TM5) and transition of the intracellular loop 2 (ICL2) into a short helix. These conformational changes likely prime GPR40 to a more active-like state and explain the binding cooperativity between these ligands.
Collapse
Affiliation(s)
- Jun Lu
- Department of Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Noel Byrne
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - John Wang
- Department of In vitro Pharmacology, Merck Research Laboratories, West Point, Pennsylvania, USA
| | | | - Frank K Brown
- Department of Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Harry R Chobanian
- Department of Medicinal Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Steven L Colletti
- Department of Medicinal Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Jerry Di Salvo
- Department of In vitro Pharmacology, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Brande Thomas-Fowlkes
- Department of In vitro Pharmacology, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Yan Guo
- Department of Medicinal Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Dawn L Hall
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Jennifer Hadix
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Nicholas B Hastings
- Department of In vitro Pharmacology, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Jeffrey D Hermes
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Thu Ho
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Andrew D Howard
- Department of Cardiometabolic Disease, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Hubert Josien
- Department of Medicinal Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Maria Kornienko
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Kevin J Lumb
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Michael W Miller
- Department of Medicinal Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Sangita B Patel
- Department of Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Barbara Pio
- Department of Medicinal Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Christopher W Plummer
- Department of Medicinal Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Bradley S Sherborne
- Department of Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Payal Sheth
- Department of In vitro Pharmacology, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Sarah Souza
- Department of In vitro Pharmacology, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Srivanya Tummala
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | | | - Maria Webb
- Department of In vitro Pharmacology, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Samantha J Allen
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Jennifer M Johnston
- Department of Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Adam B Weinglass
- Department of In vitro Pharmacology, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Sujata Sharma
- Department of Screening and Protein Science, Merck Research Laboratories, West Point, Pennsylvania, USA
| | - Stephen M Soisson
- Department of Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania, USA
| |
Collapse
|
65
|
Neuman JC, Schaid MD, Brill AL, Fenske RJ, Kibbe CR, Fontaine DA, Sdao SM, Brar HK, Connors KM, Wienkes HN, Eliceiri KW, Merrins MJ, Davis DB, Kimple ME. Enriching Islet Phospholipids With Eicosapentaenoic Acid Reduces Prostaglandin E 2 Signaling and Enhances Diabetic β-Cell Function. Diabetes 2017; 66:1572-1585. [PMID: 28193789 PMCID: PMC5440023 DOI: 10.2337/db16-1362] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 02/09/2017] [Indexed: 12/26/2022]
Abstract
Prostaglandin E2 (PGE2) is derived from arachidonic acid, whereas PGE3 is derived from eicosapentaenoic acid (EPA) using the same downstream metabolic enzymes. Little is known about the impact of EPA and PGE3 on β-cell function, particularly in the diabetic state. In this work, we determined that PGE3 elicits a 10-fold weaker reduction in glucose-stimulated insulin secretion through the EP3 receptor as compared with PGE2 We tested the hypothesis that enriching pancreatic islet cell membranes with EPA, thereby reducing arachidonic acid abundance, would positively impact β-cell function in the diabetic state. EPA-enriched islets isolated from diabetic BTBR Leptinob/ob mice produced significantly less PGE2 and more PGE3 than controls, correlating with improved glucose-stimulated insulin secretion. NAD(P)H fluorescence lifetime imaging showed that EPA acts downstream and independently of mitochondrial function. EPA treatment also reduced islet interleukin-1β expression, a proinflammatory cytokine known to stimulate prostaglandin production and EP3 expression. Finally, EPA feeding improved glucose tolerance and β-cell function in a mouse model of diabetes that incorporates a strong immune phenotype: the NOD mouse. In sum, increasing pancreatic islet EPA abundance improves diabetic β-cell function through both direct and indirect mechanisms that converge on reduced EP3 signaling.
Collapse
Affiliation(s)
- Joshua C Neuman
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Michael D Schaid
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Allison L Brill
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Rachel J Fenske
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Carly R Kibbe
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Danielle A Fontaine
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Sophia M Sdao
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Integrated Program in Biochemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Harpreet K Brar
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Kelsey M Connors
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Haley N Wienkes
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Kevin W Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Matthew J Merrins
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Integrated Program in Biochemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Dawn B Davis
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Michelle E Kimple
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| |
Collapse
|
66
|
Pillaiyar T, Köse M, Sylvester K, Weighardt H, Thimm D, Borges G, Förster I, von Kügelgen I, Müller CE. Diindolylmethane Derivatives: Potent Agonists of the Immunostimulatory Orphan G Protein-Coupled Receptor GPR84. J Med Chem 2017; 60:3636-3655. [PMID: 28406627 DOI: 10.1021/acs.jmedchem.6b01593] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Gi protein-coupled receptor GPR84, which is activated by (hydroxy)fatty acids, is highly expressed on immune cells. Recently, 3,3'-diindolylmethane was identified as a heterocyclic, nonlipid-like GPR84 agonist. We synthesized a broad range of diindolylmethane derivatives by condensation of indoles with formaldehyde in water under microwave irradiation. The products were evaluated at the human GPR84 in cAMP and β-arrestin assays. Structure-activity relationships (SARs) were steep. 3,3'-Diindolylmethanes bearing small lipophilic residues at the 5- and/or 7-position of the indole rings displayed the highest activity in cAMP assays, the most potent agonists being di(5-fluoro-1H-indole-3-yl)methane (38, PSB-15160, EC50 80.0 nM) and di(5,7-difluoro-1H-indole-3-yl)methane (57, PSB-16671, EC50 41.3 nM). In β-arrestin assays, SARs were different, indicating biased agonism. The new compounds were selective versus related fatty acid receptors and the arylhydrocarbon receptor. Selected compounds were further investigated and found to display an ago-allosteric mechanism of action and increased stability in comparison to the lead structure.
Collapse
Affiliation(s)
- Thanigaimalai Pillaiyar
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn , An der Immenburg 4, D-53121 Bonn, Germany
| | - Meryem Köse
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn , An der Immenburg 4, D-53121 Bonn, Germany
| | - Katharina Sylvester
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn , An der Immenburg 4, D-53121 Bonn, Germany
| | - Heike Weighardt
- Life and Medical Sciences (LIMES) Institute, Immunology and Environment, University of Bonn , Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Dominik Thimm
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn , An der Immenburg 4, D-53121 Bonn, Germany
| | - Gleice Borges
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn , An der Immenburg 4, D-53121 Bonn, Germany
| | - Irmgard Förster
- Life and Medical Sciences (LIMES) Institute, Immunology and Environment, University of Bonn , Carl-Troll-Straße 31, 53115 Bonn, Germany
| | - Ivar von Kügelgen
- Department of Pharmacology and Toxicology, University of Bonn , 53105 Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn , An der Immenburg 4, D-53121 Bonn, Germany
| |
Collapse
|
67
|
Yang Y, Gong L. Palmitoleate inhibits insulin transcription by activating the ERK1/2 pathway in rat pancreatic β-cells. Exp Ther Med 2017; 13:2805-2811. [PMID: 28587345 PMCID: PMC5450653 DOI: 10.3892/etm.2017.4344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/04/2016] [Indexed: 02/05/2023] Open
Abstract
The aim of the present study was to evaluate the effects of palmitoleate on insulin secretion and insulin mRNA levels, and to investigate the transcriptional regulation of insulin. INS-1 rat insulinoma cells were treated with palmitoleate in the presence of high glucose, and the amount of secreted insulin was measured via radioimmunoassay. Reverse transcription-quantitative polymerase chain reaction was performed to evaluate the mRNA levels of insulin and pancreatic and duodenal homeobox 1 (PDX1) under palmitoleate treatment. The levels of PDX1, peroxisome proliferator-activated receptor gamma (PPARG), extracellular signal-regulated kinase (ERK)1/2 and phosphorylated ERK1/2 were measured using western blot analysis. Low concentrations of palmitoleate significantly induced insulin secretion (P=0.024), whereas the mRNA levels of insulin and PDX1 were markedly reduced. However, the inhibitory effects were reversed with the addition of U0126, suggesting that the ERK1/2-mediated pathway may be the underlying mechanism responsible for palmitoleate-induced downregulation of insulin mRNA. Exposure of INS-1 cells to high glucose significantly increased the phosphorylation of ERK1/2 (P=0.039), which was further enhanced by palmitoleate (P=0.025). Exposure of INS-1 cells to high glucose significantly decreased PPARG (P=0.001), which was further decreased by the addition of palmitoleate. U0126 was able to reverse the palmitoleate-induced effects. In conclusion, the present study suggested that palmitoleate may induce insulin secretion and inhibit insulin mRNA expression in pancreatic β-cells.
Collapse
Affiliation(s)
- Yumei Yang
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Liangliang Gong
- Department of Rheumatology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| |
Collapse
|
68
|
Neuman JC, Fenske RJ, Kimple ME. Dietary polyunsaturated fatty acids and their metabolites: Implications for diabetes pathophysiology, prevention, and treatment. NUTRITION AND HEALTHY AGING 2017; 4:127-140. [PMID: 28447067 PMCID: PMC5391679 DOI: 10.3233/nha-160004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Affiliation(s)
- Joshua C. Neuman
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Rachel J. Fenske
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michelle E. Kimple
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| |
Collapse
|
69
|
Li Z, Xu X, Huang W, Qian H. Free Fatty Acid Receptor 1 (FFAR1) as an Emerging Therapeutic Target for Type 2 Diabetes Mellitus: Recent Progress and Prevailing Challenges. Med Res Rev 2017; 38:381-425. [DOI: 10.1002/med.21441] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/23/2017] [Accepted: 02/14/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Zheng Li
- Center of Drug Discovery, State Key Laboratory of Natural Medicines; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 P.R. China
| | - Xue Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 P.R. China
| | - Wenlong Huang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 P.R. China
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 P.R. China
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 P.R. China
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 P.R. China
| |
Collapse
|
70
|
Hidalgo-Figueroa S, Navarrete-Vázquez G, Estrada-Soto S, Giles-Rivas D, Alarcón-Aguilar FJ, León-Rivera I, Giacoman-Martínez A, Miranda Pérez E, Almanza-Pérez JC. Discovery of new dual PPARγ-GPR40 agonists with robust antidiabetic activity: Design, synthesis and in combo drug evaluation. Biomed Pharmacother 2017; 90:53-61. [PMID: 28342366 DOI: 10.1016/j.biopha.2017.03.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/09/2017] [Accepted: 03/10/2017] [Indexed: 01/17/2023] Open
Abstract
The design of compounds 1 and 2 was based on the similar scaffold of pharmacophoric groups for PPARγ and GPR40 agonists. In order to find new compounds with improved biological activity, the current manuscript describes a new dual PPARγ-GPR40 agonist. We synthesized two compounds, which were prepared following a multistep synthetic route, and the relative mRNA expression levels of PPARγ, GLUT4, and GPR40 were quantified in cell culture, as well as insulin secretion and [Ca2+] intracellular levels. Compound 1 showed a 7-times increase in the mRNA expression of PPARγ, which in turn enhanced the expression levels of GLUT4 respect to control and pioglitazone. It also showed an increase of 2-fold in the [Ca2+]i level allowing an increment on insulin release, being as active as the positive control (glibenclamide), causing also an increase of 2-fold in mRNA expression of GPR40. Furthermore, the compound 2 showed lower activity than the compound 1. The ester of 1 showed antidiabetic activity at a 50mg/kg single dose in streptozotocin-nicotinamide-induced diabetic mice model. In addition, we achieved a molecular docking study of compound 1 on PPARγ and GPR40 receptors, showing a great affinity for both targets. We observed important polar interactions between the carboxylic group and main residues into the binding pocket. Therefore, the compound 1 has a potential for the development of antidiabetic agents with newfangled dual action.
Collapse
Affiliation(s)
- Sergio Hidalgo-Figueroa
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana- Iztapalapa, Apdo.-Postal 55-535, CP 09340, México, D.F., Mexico.
| | - Gabriel Navarrete-Vázquez
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001 Col. Chamilpa, C.P. 62209, Cuernavaca, Morelos, Mexico
| | - Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001 Col. Chamilpa, C.P. 62209, Cuernavaca, Morelos, Mexico
| | - Diana Giles-Rivas
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001 Col. Chamilpa, C.P. 62209, Cuernavaca, Morelos, Mexico
| | - Francisco J Alarcón-Aguilar
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana- Iztapalapa, Apdo.-Postal 55-535, CP 09340, México, D.F., Mexico
| | - Ismael León-Rivera
- Centro de Investigaciones Químicas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mor. 62209, Mexico
| | - Abraham Giacoman-Martínez
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana- Iztapalapa, Apdo.-Postal 55-535, CP 09340, México, D.F., Mexico
| | - Elizabeth Miranda Pérez
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana- Iztapalapa, Apdo.-Postal 55-535, CP 09340, México, D.F., Mexico
| | - Julio C Almanza-Pérez
- Laboratorio de Farmacología, Depto. Ciencias de la Salud, D.C.B.S., Universidad Autónoma Metropolitana- Iztapalapa, Apdo.-Postal 55-535, CP 09340, México, D.F., Mexico.
| |
Collapse
|
71
|
Abstract
Overfeeding of fat can cause various metabolic disorders including obesity and type 2 diabetes (T2D). Diet provided free fatty acids (FFAs) are not only essential nutrients, but they are also recognized as signaling molecules, which stimulate various important biological functions. Recently, several G protein-coupled receptors (GPCRs), including FFA1-4, have been identified as receptors of FFAs by various physiological and pharmacological studies. FFAs exert physiological functions through these FFA receptors (FFARs) depending on carbon chain length and degree of unsaturation. Functional analyses have revealed that several important metabolic processes, such as peptide hormone secretion, cell maturation and nerve activities, are regulated by FFARs and thereby FFARs contribute to the energy homeostasis through these physiological functions. Hence, FFARs are expected to be promising pharmacological targets for metabolic disorders since imbalances in energy homeostasis lead to metabolic disorders. In human, it is established that different responses of individuals to endogenous ligands and chemical drugs may be due to differences in the ability of such ligands to activate nucleotide polymorphic variants of receptors. However, the clear links between genetic variations that are involved in metabolic disorders and polymorphisms receptors have been relatively difficult to assess. In this review, I summarize current literature describing physiological functions of FFARs and genetic variations of those receptors to discuss the potential of FFARs as drug targets for metabolic disorders.
Collapse
Affiliation(s)
- Atsuhiko Ichimura
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29, Sakyo-ku, yoshidashimoadachi-cho, Kyoto, 606-8501, Japan.
| |
Collapse
|
72
|
Abstract
Of the 415 million people suffering from diabetes worldwide, 90% have type 2 diabetes. Type 2 diabetes is characterized by hyperglycemia and occurs in obese individuals as a result of insulin resistance and inadequate insulin levels. Accordingly, diabetes drugs are tailored to enhance glucose disposal or target the pancreatic islet β cell to increase insulin secretion. The majority of the present-day insulin secretagogues, however, increase the risk of iatrogenic hypoglycemia, and hence alternatives are actively sought. The long-chain fatty acid, G protein-coupled receptor FFA1/Gpr40, is expressed in β cells, and its activation potentiates insulin secretion in a glucose-dependent manner. Preclinical data indicate that FFA1 agonism is an effective treatment to restore glucose homeostasis in rodent models of diabetes. This initial success prompted clinical trials in type 2 diabetes patients, the results of which were promising; however, the field suffered a significant setback when the lead compound TAK-875/fasiglifam was withdrawn from clinical development due to liver safety concerns. Nevertheless, recent developments have brought to light a surprising complexity of FFA1 agonist action, signaling diversity, and biological outcomes, raising hopes that with a greater understanding of the mechanisms at play the second round will be more successful.
Collapse
Affiliation(s)
- Julien Ghislain
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- CRCHUM, University of Montreal, 900 rue St Denis, Montreal, QC, Canada, H2X 0A9
| | - Vincent Poitout
- Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada.
- CRCHUM, University of Montreal, 900 rue St Denis, Montreal, QC, Canada, H2X 0A9.
- Department of Medicine, University of Montreal, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
73
|
Miśta D, Króliczewska B, Pecka-Kiełb E, Kapuśniak V, Zawadzki W, Graczyk S, Kowalczyk A, Łukaszewicz E, Bednarczyk M. Effect of in ovo injected prebiotics and synbiotics on the caecal fermentation and intestinal morphology of broiler chickens. ANIMAL PRODUCTION SCIENCE 2017. [DOI: 10.1071/an16257] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Manipulations of the intestinal microbiota composition may improve the health and performance of chickens. In ovo technology allows the administration of a bioactive substance to enter directly into the incubating egg. The objective of the present study was to investigate the effect of in ovo administered prebiotics or synbiotics on microbial activity products in the caeca and the development of the small intestine of broiler chickens. Ross 308 male chickens hatched from eggs injected in ovo with prebiotics or synbiotics were used in this study. Five experimental groups were formed: C (Control) – injected with 0.9% NaCl, the Pre-1 and Pre-2 groups – injected with prebiotics: inulin or Bi2tos, respectively, and the Syn-1 and Syn-2 groups – injected with synbiotics: inulin with Lactococcus lactis subsp. lactis IBB SL1 or Bi2tos with Lactococcus lactis subsp. cremoris IBB SC1, respectively. At the age of 7, 21 and 35 days, 10 chicks of each group were randomly selected, weighed and slaughtered, and the jejunal samples were collected for histological examinations, whereas caecal samples were collected to analyse the end products of microbial fermentation. Synbiotic treatment increased bodyweight, as observed in the Syn-1 group (P < 0.05). The propionate molar proportion was highest in the groups treated with synbiotics, especially in the Syn-1 group (P < 0.01). Furthermore, the molar proportion of acetate was also lowest in the Syn-1 group (P < 0.05). In ovo synbiotics treatment increased the villus length : crypt depth ratio in the jejunal mucosa, which might improve nutrient absorption and contribute to the increased weight of chickens. These effects suggest that the in ovo administration of synbiotics may be an effective method to increase bodyweight, improve the short-chain fatty acid caecal profile and increase the villus length : crypt depth ratio in the jejunal mucosa. These effects were more pronounced in the Syn-1 group than the Syn-2 group.
Collapse
|
74
|
Hamdouchi C, Kahl SD, Patel Lewis A, Cardona GR, Zink RW, Chen K, Eessalu TE, Ficorilli JV, Marcelo MC, Otto KA, Wilbur KL, Lineswala JP, Piper JL, Coffey DS, Sweetana SA, Haas JV, Brooks DA, Pratt EJ, Belin RM, Deeg MA, Ma X, Cannady EA, Johnson JT, Yumibe NP, Chen Q, Maiti P, Montrose-Rafizadeh C, Chen Y, Reifel Miller A. The Discovery, Preclinical, and Early Clinical Development of Potent and Selective GPR40 Agonists for the Treatment of Type 2 Diabetes Mellitus (LY2881835, LY2922083, and LY2922470). J Med Chem 2016; 59:10891-10916. [DOI: 10.1021/acs.jmedchem.6b00892] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Chafiq Hamdouchi
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Steven D. Kahl
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Anjana Patel Lewis
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Guemalli R. Cardona
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Richard W. Zink
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Keyue Chen
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Thomas E. Eessalu
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - James V. Ficorilli
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Marialuisa C. Marcelo
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Keith A. Otto
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Kelly L. Wilbur
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Jayana P. Lineswala
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Jared L. Piper
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - D. Scott Coffey
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Stephanie A. Sweetana
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Joseph V. Haas
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Dawn A. Brooks
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | | | - Ruth M. Belin
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Mark A. Deeg
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Xiaosu Ma
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Ellen A. Cannady
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Jason T. Johnson
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Nathan P. Yumibe
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Qi Chen
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Pranab Maiti
- Jubilant Biosys Research Center, 560 022 Bangalore, India
| | - Chahrzad Montrose-Rafizadeh
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Yanyun Chen
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| | - Anne Reifel Miller
- Lilly
Research Laboratories, A division of Eli Lilly and Company, Lilly
Corporate Center, DC: 0540, Indianapolis, Indiana 46285, United States
| |
Collapse
|
75
|
Priyadarshini M, Wicksteed B, Schiltz GE, Gilchrist A, Layden BT. SCFA Receptors in Pancreatic β Cells: Novel Diabetes Targets? Trends Endocrinol Metab 2016; 27:653-664. [PMID: 27091493 PMCID: PMC4992600 DOI: 10.1016/j.tem.2016.03.011] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/17/2016] [Accepted: 03/20/2016] [Indexed: 01/07/2023]
Abstract
Nutrient sensing receptors are key metabolic mediators of responses to dietary and endogenously derived nutrients. These receptors are largely G-protein-coupled receptors (GPCRs) and many are gaining significant interest as drug targets with a potential therapeutic role in metabolic diseases. A distinct subclass of nutrient sensing GPCRs, two short chain fatty acid (SCFA) receptors (FFA2 and FFA3) are uniquely responsive to gut microbiota derived nutrients (such as acetate, propionate, and butyrate). Pharmacological, molecular, and genetic studies have investigated their role in organismal glucose metabolism and recently in pancreatic β cell biology. Here, we summarize the present knowledge on the role of these receptors as metabolic sensors in β cell function and physiology, revealing new therapeutic opportunities for type 2 diabetes.
Collapse
Affiliation(s)
- Medha Priyadarshini
- Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Barton Wicksteed
- Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Gary E Schiltz
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL 60208, USA; Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Annette Gilchrist
- Midwestern University Department of Pharmaceutical Sciences, Downers Grove, IL 60515, USA
| | - Brian T Layden
- Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
76
|
Alvarez-Curto E, Milligan G. Metabolism meets immunity: The role of free fatty acid receptors in the immune system. Biochem Pharmacol 2016; 114:3-13. [DOI: 10.1016/j.bcp.2016.03.017] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/17/2016] [Indexed: 12/11/2022]
|
77
|
Liu D, Archer N, Duesing K, Hannan G, Keast R. Mechanism of fat taste perception: Association with diet and obesity. Prog Lipid Res 2016; 63:41-9. [DOI: 10.1016/j.plipres.2016.03.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/22/2016] [Accepted: 03/09/2016] [Indexed: 12/11/2022]
|
78
|
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
|
79
|
Milligan G, Shimpukade B, Ulven T, Hudson BD. Complex Pharmacology of Free Fatty Acid Receptors. Chem Rev 2016; 117:67-110. [PMID: 27299848 DOI: 10.1021/acs.chemrev.6b00056] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are historically the most successful family of drug targets. In recent times it has become clear that the pharmacology of these receptors is far more complex than previously imagined. Understanding of the pharmacological regulation of GPCRs now extends beyond simple competitive agonism or antagonism by ligands interacting with the orthosteric binding site of the receptor to incorporate concepts of allosteric agonism, allosteric modulation, signaling bias, constitutive activity, and inverse agonism. Herein, we consider how evolving concepts of GPCR pharmacology have shaped understanding of the complex pharmacology of receptors that recognize and are activated by nonesterified or "free" fatty acids (FFAs). The FFA family of receptors is a recently deorphanized set of GPCRs, the members of which are now receiving substantial interest as novel targets for the treatment of metabolic and inflammatory diseases. Further understanding of the complex pharmacology of these receptors will be critical to unlocking their ultimate therapeutic potential.
Collapse
Affiliation(s)
- Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow G12 8QQ, Scotland, United Kingdom
| | - Bharat Shimpukade
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark , Campusvej 55, DK-5230 Odense M, Denmark
| | - Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark , Campusvej 55, DK-5230 Odense M, Denmark
| | - Brian D Hudson
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow G12 8QQ, Scotland, United Kingdom
| |
Collapse
|
80
|
Bertrand R, Hamp I, Brönstrup M, Weck R, Lukacevic M, Polyak A, Ross TL, Gotthardt M, Plettenburg O, Derdau V. Synthesis of GPR40 targeting 3 H- and 18 F-probes towards selective beta cell imaging. J Labelled Comp Radiopharm 2016; 59:604-610. [PMID: 27282912 DOI: 10.1002/jlcr.3412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/11/2016] [Accepted: 05/09/2016] [Indexed: 11/07/2022]
Abstract
Diabetes affects an increasing number of patients worldwide and is responsible for a significant rise in healthcare expenses. Imaging of β-cells in vivo is expected to contribute to an improved understanding of the underlying pathophysiology, improved diagnosis, and development of new treatment options for diabetes. Here, we describe the first radiosyntheses of [3 H]-TAK875 and [18 F]-TAK875 derivatives to be used as β-cell imaging probes addressing the free fatty acid receptor 1 (FFAR1/GPR40). The fluorine-labeled derivative showed similar agonistic activity as TAK875 in a functional assay. The radiosynthesis of the 18 F-labelled tracer 2a was achieved with 16.7 ± 5.7% radiochemical yield in a total synthesis time of 60-70 min.
Collapse
Affiliation(s)
- Romain Bertrand
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main, Germany
- Department of Nuclear Medicine, Radboud UMC, Nijmegen, The Netherlands
| | - Isabel Hamp
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main, Germany
| | - Mark Brönstrup
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Remo Weck
- DSAR/Drug Disposition, Sanofi GmbH, Frankfurt am Main, Germany
| | - Mario Lukacevic
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Andras Polyak
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Martin Gotthardt
- Department of Nuclear Medicine, Radboud UMC, Nijmegen, The Netherlands
| | - Oliver Plettenburg
- Diabetes Division, Research & Translational Medicine, Sanofi GmbH, Frankfurt am Main, Germany
- Institute of Medicinal Chemistry, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, Neuherberg DE-85764, Germany
- Leibniz Universität Hannover, Schneiderberg 1b, 30167, Hannover, Germany
| | - Volker Derdau
- DSAR/Drug Disposition, Sanofi GmbH, Frankfurt am Main, Germany
| |
Collapse
|
81
|
Wang S, Dougherty EJ, Danner RL. PPARγ signaling and emerging opportunities for improved therapeutics. Pharmacol Res 2016; 111:76-85. [PMID: 27268145 DOI: 10.1016/j.phrs.2016.02.028] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 02/29/2016] [Indexed: 01/23/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated nuclear receptor that regulates glucose and lipid metabolism, endothelial function and inflammation. Rosiglitazone (RGZ) and other thiazolidinedione (TZD) synthetic ligands of PPARγ are insulin sensitizers that have been used for the treatment of type 2 diabetes. However, undesirable side effects including weight gain, fluid retention, bone loss, congestive heart failure, and a possible increased risk of myocardial infarction and bladder cancer, have limited the use of TZDs. Therefore, there is a need to better understand PPARγ signaling and to develop safer and more effective PPARγ-directed therapeutics. In addition to PPARγ itself, many PPARγ ligands including TZDs bind to and activate G protein-coupled receptor 40 (GPR40), also known as free fatty acid receptor 1. GPR40 signaling activates stress kinase pathways that ultimately regulate downstream PPARγ responses. Recent studies in human endothelial cells have demonstrated that RGZ activation of GPR40 is essential to the optimal propagation of PPARγ genomic signaling. RGZ/GPR40/p38 MAPK signaling induces and activates PPARγ co-activator-1α, and recruits E1A binding protein p300 to the promoters of target genes, markedly enhancing PPARγ-dependent transcription. Therefore in endothelium, GPR40 and PPARγ function as an integrated signaling pathway. However, GPR40 can also activate ERK1/2, a proinflammatory kinase that directly phosphorylates and inactivates PPARγ. Thus the role of GPR40 in PPARγ signaling may have important implications for drug development. Ligands that strongly activate PPARγ, but do not bind to or activate GPR40 may be safer than currently approved PPARγ agonists. Alternatively, biased GPR40 agonists might be sought that activate both p38 MAPK and PPARγ, but not ERK1/2, avoiding its harmful effects on PPARγ signaling, insulin resistance and inflammation. Such next generation drugs might be useful in treating not only type 2 diabetes, but also diverse chronic and acute forms of vascular inflammation such as atherosclerosis and septic shock.
Collapse
Affiliation(s)
- Shuibang Wang
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Edward J Dougherty
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert L Danner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
82
|
Synthesis and biological evaluation of GPR40/FFAR1 agonists containing 3,5-dimethylisoxazole. Eur J Med Chem 2016; 116:46-58. [DOI: 10.1016/j.ejmech.2016.03.054] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 01/09/2023]
|
83
|
Christiansen E, Hudson BD, Hansen AH, Milligan G, Ulven T. Development and Characterization of a Potent Free Fatty Acid Receptor 1 (FFA1) Fluorescent Tracer. J Med Chem 2016; 59:4849-58. [DOI: 10.1021/acs.jmedchem.6b00202] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Elisabeth Christiansen
- Department
of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Brian D. Hudson
- Molecular
Pharmacology Group, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Anders Højgaard Hansen
- Department
of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Graeme Milligan
- Molecular
Pharmacology Group, Institute of Molecular, Cell and Systems Biology,
College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Trond Ulven
- Department
of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| |
Collapse
|
84
|
Ulven T, Christiansen E. Dietary Fatty Acids and Their Potential for Controlling Metabolic Diseases Through Activation of FFA4/GPR120. Annu Rev Nutr 2016; 35:239-63. [PMID: 26185978 DOI: 10.1146/annurev-nutr-071714-034410] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It is well known that the amount and type of ingested fat impacts the development of obesity and metabolic diseases, but the potential for beneficial effects from fat has received less attention. It is becoming clear that the composition of the individual fatty acids in diet is important. Besides acting as precursors of potent signaling molecules, dietary fatty acids act directly on intracellular and cell surface receptors. The free fatty acid receptor 4 (FFA4, previously GPR120) is linked to the regulation of body weight, inflammation, and insulin resistance and represents a potential target for the treatment of metabolic disorders, including type 2 diabetes and obesity. In this review, we discuss the various types of dietary fatty acids, the link between FFA4 and metabolic diseases, the potential effects of the individual fatty acids on health, and the ability of fatty acids to activate FFA4. We also discuss the possibility of dietary schemes that implement activation of FFA4.
Collapse
Affiliation(s)
- Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark;
| | | |
Collapse
|
85
|
Nutritional Signaling via Free Fatty Acid Receptors. Int J Mol Sci 2016; 17:450. [PMID: 27023530 PMCID: PMC4848906 DOI: 10.3390/ijms17040450] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/07/2016] [Accepted: 03/17/2016] [Indexed: 12/30/2022] Open
Abstract
Excess energy is stored primarily as triglycerides, which are mobilized when demand for energy arises. Dysfunction of energy balance by excess food intake leads to metabolic diseases, such as obesity and diabetes. Free fatty acids (FFAs) provided by dietary fat are not only important nutrients, but also contribute key physiological functions via FFA receptor (FFAR)-mediated signaling molecules, which depend on FFAs' carbon chain length and the ligand specificity of the receptors. Functional analyses have revealed that FFARs are critical for metabolic functions, such as peptide hormone secretion and inflammation, and contribute to energy homeostasis. In particular, recent studies have shown that the administration of selective agonists of G protein-coupled receptor (GPR) 40 and GPR120 improved glucose metabolism and systemic metabolic disorders. Furthermore, the anti-inflammation and energy metabolism effects of short chain FAs have been linked to the activation of GPR41 and GPR43. In this review, we summarize recent progress in research on FFAs and their physiological roles in the regulation of energy metabolism.
Collapse
|
86
|
Ren XM, Cao LY, Zhang J, Qin WP, Yang Y, Wan B, Guo LH. Investigation of the Binding Interaction of Fatty Acids with Human G Protein-Coupled Receptor 40 Using a Site-Specific Fluorescence Probe by Flow Cytometry. Biochemistry 2016; 55:1989-96. [DOI: 10.1021/acs.biochem.6b00079] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xiao-Min Ren
- State Key Laboratory of Environmental
Chemistry and Eco-toxicology, Research Center for Eco-environmental
Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, P.O. Box 2871, Beijing 100085, China
| | - Lin-Ying Cao
- State Key Laboratory of Environmental
Chemistry and Eco-toxicology, Research Center for Eco-environmental
Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, P.O. Box 2871, Beijing 100085, China
| | - Jing Zhang
- State Key Laboratory of Environmental
Chemistry and Eco-toxicology, Research Center for Eco-environmental
Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, P.O. Box 2871, Beijing 100085, China
| | - Wei-Ping Qin
- College of Life and Environmental
Science, Minzu University of China, Beijing 100081, China
| | - Yu Yang
- State Key Laboratory of Environmental
Chemistry and Eco-toxicology, Research Center for Eco-environmental
Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, P.O. Box 2871, Beijing 100085, China
| | - Bin Wan
- State Key Laboratory of Environmental
Chemistry and Eco-toxicology, Research Center for Eco-environmental
Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, P.O. Box 2871, Beijing 100085, China
| | - Liang-Hong Guo
- State Key Laboratory of Environmental
Chemistry and Eco-toxicology, Research Center for Eco-environmental
Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, P.O. Box 2871, Beijing 100085, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| |
Collapse
|
87
|
Hansen SVF, Christiansen E, Urban C, Hudson BD, Stocker CJ, Due-Hansen ME, Wargent ET, Shimpukade B, Almeida R, Ejsing CS, Cawthorne MA, Kassack MU, Milligan G, Ulven T. Discovery of a Potent Free Fatty Acid 1 Receptor Agonist with Low Lipophilicity, Low Polar Surface Area, and Robust in Vivo Efficacy. J Med Chem 2016; 59:2841-6. [DOI: 10.1021/acs.jmedchem.5b01962] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Steffen V. F. Hansen
- Department
of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Elisabeth Christiansen
- Department
of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Christian Urban
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Düsseldorf, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| | - Brian D. Hudson
- Institute
of Molecular, Cell and Systems Biology, College of Medical, Veterinary
and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K
| | - Claire J. Stocker
- Clore
Laboratory, University of Buckingham, Hunter Street, Buckingham MK18 1EG, U.K
| | - Maria E. Due-Hansen
- Department
of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Ed T. Wargent
- Clore
Laboratory, University of Buckingham, Hunter Street, Buckingham MK18 1EG, U.K
| | - Bharat Shimpukade
- Department
of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Reinaldo Almeida
- Department
of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical
Sciences, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Christer S. Ejsing
- Department
of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical
Sciences, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Michael A. Cawthorne
- Clore
Laboratory, University of Buckingham, Hunter Street, Buckingham MK18 1EG, U.K
| | - Matthias U. Kassack
- Institute
of Pharmaceutical and Medicinal Chemistry, University of Düsseldorf, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| | - Graeme Milligan
- Institute
of Molecular, Cell and Systems Biology, College of Medical, Veterinary
and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, U.K
| | - Trond Ulven
- Department
of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| |
Collapse
|
88
|
Loaiza A, Carretta MD, Taubert A, Hermosilla C, Hidalgo MA, Burgos RA. Differential intracellular calcium influx, nitric oxide production, ICAM-1 and IL8 expression in primary bovine endothelial cells exposed to nonesterified fatty acids. BMC Vet Res 2016; 12:38. [PMID: 26916791 PMCID: PMC4766702 DOI: 10.1186/s12917-016-0654-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 02/15/2016] [Indexed: 11/16/2022] Open
Abstract
Background Nonesterified fatty acids (NEFAs) are involved in proinflammatory processes in cattle, including in the increased expression of adhesion molecules in endothelial cells. However, the mechanisms underlying these effects are still unknown. The aim of this study was to assess the effects of NEFAs on the intracellular calcium (Ca2+i) influx, nitric oxide production, and ICAM-1 and IL-8 expression in primary bovine umbilical vein endothelial cells (BUVECs). Results Myristic (MA), palmitic (PA), stearic (SA), oleic (OA) and linoleic acid (LA) rapidly increased Ca2+i. The calcium response to all tested NEFAs showed an extracellular calcium dependence and only the LA response was significantly inhibited until the intracellular calcium was chelated. The EC50 values for MA and LA were 125 μM and 37 μM, respectively, and the MA and LA effects were dependent on calcium release from the endoplasmic reticulum stores and on the L-type calcium channels. Only the calcium response to MA was significantly reduced by GW1100, a selective G-protein-coupled free fatty acid receptor (GPR40) antagonist. We also detected a functional FFAR1/GPR40 protein in BUVECs by using western blotting and the FFAR1/GPR40 agonist TAK-875. Only LA increased the cellular nitric oxide levels in a calcium-dependent manner. LA stimulation but not MA stimulation increased ICAM-1 and IL-8-expression in BUVECs. This effect was inhibited by GW1100, an antagonist of FFAR1/GPR40, but not by U-73122, a phospholipase C inhibitor. Conclusions These findings strongly suggest that each individual NEFA stimulates endothelial cells in a different way, with clearly different effects on intracellular calcium mobilization, NO production, and IL-8 and ICAM-1 expression in primary BUVECs. These findings not only extend our understanding of NEFA-mediated diseases in ruminants, but also provide new insight into the different molecular mechanisms involved during endothelial cell activation by NEFAs. Electronic supplementary material The online version of this article (doi:10.1186/s12917-016-0654-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anitsi Loaiza
- Instituto de Farmacología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - María D Carretta
- Instituto de Farmacología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Anja Taubert
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - María A Hidalgo
- Instituto de Farmacología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael A Burgos
- Instituto de Farmacología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
89
|
Design, synthesis, and biological evaluation of novel thiazolidinediones as PPARγ/FFAR1 dual agonists. Eur J Med Chem 2016; 109:157-72. [DOI: 10.1016/j.ejmech.2015.12.049] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/27/2015] [Accepted: 12/28/2015] [Indexed: 01/29/2023]
|
90
|
Ito R, Tsujihata Y, Suzuki M, Miyawaki K, Matsuda K, Takeuchi K. Fasiglifam/TAK-875, a Selective GPR40 Agonist, Improves Hyperglycemia in Rats Unresponsive to Sulfonylureas and Acts Additively with Sulfonylureas. ACTA ACUST UNITED AC 2016; 357:217-27. [DOI: 10.1124/jpet.115.230730] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/21/2016] [Indexed: 02/01/2023]
|
91
|
Suzuki K, Kaneko-Kawano T. Biological roles and therapeutic potential of G protein-coupled receptors for free fatty acids and metabolic intermediates. ACTA ACUST UNITED AC 2016. [DOI: 10.7600/jpfsm.5.213] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Kenji Suzuki
- College of Pharmaceutical Sciences, Ritsumeikan University
| | | |
Collapse
|
92
|
Ma Z, Lin DCH, Sharma R, Liu J, Zhu L, Li AR, Kohn T, Wang Y, Liu J(J, Bartberger MD, Medina JC, Zhuang R, Li F, Zhang J, Luo J, Wong S, Tonn GR, Houze JB. Discovery of the imidazole-derived GPR40 agonist AM-3189. Bioorg Med Chem Lett 2016; 26:15-20. [DOI: 10.1016/j.bmcl.2015.11.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/09/2015] [Accepted: 11/16/2015] [Indexed: 11/30/2022]
|
93
|
Abstract
FFA1 is a G protein-coupled receptor activated by medium- to long-chain fatty acids. FFA1 plays important roles in various physiological processes such as insulin secretion and energy metabolism. FFA1 expressed on pancreatic β-cells and intestine contributes to insulin and incretin secretion, respectively. These physiological functions of FFA1 are interesting as an attractive drug target for type II diabetes and metabolic disorders. A number of synthetic FFA1 ligands have been developed and they have contributed to our current understanding of the physiological and pathophysiological functions of FFA1 both in in vitro and in vivo studies. In addition, these synthetic ligands also provided information on the structure-activity relationships of FFA1 ligands. Further, FFA1 protein crystallized with one of the high affinity agonist leads provided useful insights for the development of more effective ligands. Among FFA1 ligands, several compounds have been further investigated in the clinical trials. Thus, FFA1 ligands have great potential as drug candidates. In this section, recent progress about FFA1 ligands and the possibility of their clinical use are described.
Collapse
|
94
|
Anti-Inflammatory and Insulin-Sensitizing Effects of Free Fatty Acid Receptors. Handb Exp Pharmacol 2016; 236:221-231. [PMID: 27873088 DOI: 10.1007/164_2016_47] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic low-grade inflammation in macrophages and adipose tissues can promote the development of obesity and type 2 diabetes. Free fatty acids (FFAs) have important roles in various tissues, acting as both essential energy sources and signaling molecules. FFA receptors (FFARs) can modulate inflammation in various types of cells and tissues; however the underlying mechanisms mediating these effects are unclear. FFARs are activated by specific FFAs; for example, GPR40 and GPR120 are activated by medium and long chain FFAs, GPR41 and GPR43 are activated by short chain FFAs, and GPR84 is activated by medium-chain FFAs. To date, a number of studies associated with the physiological functions of G protein-coupled receptors (GPCRs) have reported that these GPCRs are expressed in various tissues and involved in inflammatory and metabolic responses. Thus, the development of selective agonists or antagonists for various GPCRs may facilitate the establishment of novel therapies for the treatment of various diseases. In this review, we summarize current literature describing the potential of GPCRs as therapeutic targets for inflammatory and metabolic disorders.
Collapse
|
95
|
Molecular mechanisms of target recognition by lipid GPCRs: relevance for cancer. Oncogene 2015; 35:4021-35. [DOI: 10.1038/onc.2015.467] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/02/2015] [Accepted: 11/02/2015] [Indexed: 12/18/2022]
|
96
|
Kurihara T, Miyata A. [Involvement of free fatty acid receptor GPR40/FFAR1 in the regulation of spinal nociceptive transmission]. Nihon Yakurigaku Zasshi 2015; 146:309-314. [PMID: 26657121 DOI: 10.1254/fpj.146.309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
|
97
|
Guo S, Zhang J, Zhang S, Li J. A Single Amino Acid Mutation (R104P) in the E/DRY Motif of GPR40 Impairs Receptor Function. PLoS One 2015; 10:e0141303. [PMID: 26505901 PMCID: PMC4624708 DOI: 10.1371/journal.pone.0141303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/06/2015] [Indexed: 11/18/2022] Open
Abstract
Type 2 Diabetes Mellitus with insulin resistance, pancreatic β cell dysfunction, and hepatic glucose overproduction is increasing in epidemic proportions worldwide. G protein-coupled receptor 40 (GPR40), a clinically proven anti-diabetic drug target, is mainly expressed in pancreatic β cells and insulin-secreting cell lines. Long chain fatty acids (LCFA) increase intracellular calcium concentration and amplify glucose-stimulated insulin secretion by activating GPR40. Here we report that the arginine 104 (R104) is critical for the normal function of GPR40. Mutation of R104 to Proline (R104P) results in complete loss of the receptor function. Linoleic acid, ligand of GPR40, could not elicit calcium increase and ERK phosphorylation in cells expressing this mutant receptor. Further study indicated the R104P mutation reduces cell surface localization of GPR40 without affecting the expression of the protein. The small portion of GPR40 R104P mutant that is still located on the membrane has no physiological function, and does not internalize in response to linoleic acid stimulation. These data demonstrate that R104 in GPR40 is critically involved in the normal receptor functions. Interestingly, R104P is a registered single-nucleotide polymorphism of GPR40. The relationship of this GPR40 variant and type 2 diabetes warrants further investigation.
Collapse
Affiliation(s)
- Shimeng Guo
- School of life sciences, Shanghai University, Shanghai, China
- Chinese Academy of Sciences Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiandong Zhang
- College of Pharmacy, Nanchang University, Nanchang, China
- Chinese Academy of Sciences Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shuyong Zhang
- Chinese Academy of Sciences Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- * E-mail: (SZ); (JL)
| | - Jing Li
- Chinese Academy of Sciences Key Laboratory of Receptor Research, National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- * E-mail: (SZ); (JL)
| |
Collapse
|
98
|
Christensen LW, Kuhre RE, Janus C, Svendsen B, Holst JJ. Vascular, but not luminal, activation of FFAR1 (GPR40) stimulates GLP-1 secretion from isolated perfused rat small intestine. Physiol Rep 2015; 3:e12551. [PMID: 26381015 PMCID: PMC4600392 DOI: 10.14814/phy2.12551] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/25/2015] [Accepted: 08/29/2015] [Indexed: 12/23/2022] Open
Abstract
Glucagon-like peptide 1 (GLP-1) plays a central role in modern treatment of type 2 diabetes (T2DM) in the form of GLP-1 enhancers and GLP-1 mimetics. An alternative treatment strategy is to stimulate endogenous GLP-1 secretion from enteroendocrine L cells using a targeted approach. The G-protein-coupled receptor, FFAR1 (previously GPR40), expressed on L cells and activated by long-chain fatty acids (LCFAs) is a potential target. A link between FFAR1 activation and GLP-1 secretion has been demonstrated in cellular models and small-molecule FFAR1 agonists have been developed. In this study, we examined the effect of FFAR1 activation on GLP-1 secretion using isolated, perfused small intestines from rats, a physiologically relevant model allowing distinction between direct and indirect effects of FFAR1 activation. The endogenous FFAR1 ligand, linoleic acid (LA), and four synthetic FFAR1 agonists (TAK-875, AMG 837, AM-1638, and AM-5262) were administered through intraluminal and intra-arterial routes, respectively, and dynamic changes in GLP-1 secretion were evaluated. Vascular administration of 10 μmol/L TAK-875, 10 μmol/L AMG 837, 1 μmol/L and 0.1 μmol/L AM-1638, 1 μmol/L AM-6252, and 1 mmol/L LA, all significantly increased GLP-1 secretion compared to basal levels (P < 0.05), whereas luminal administration of LA and FFAR1 agonists was ineffective. Thus, both natural and small-molecule agonists of the FFAR1 receptor appear to require absorption prior to stimulating GLP-1 secretion, indicating that therapies based on activation of nutrient sensing may be more complex than hitherto expected.
Collapse
Affiliation(s)
- Louise W Christensen
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| | - Rune E Kuhre
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Janus
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| | - Berit Svendsen
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
99
|
Wang S, Awad KS, Elinoff JM, Dougherty EJ, Ferreyra GA, Wang JY, Cai R, Sun J, Ptasinska A, Danner RL. G Protein-coupled Receptor 40 (GPR40) and Peroxisome Proliferator-activated Receptor γ (PPARγ): AN INTEGRATED TWO-RECEPTOR SIGNALING PATHWAY. J Biol Chem 2015; 290:19544-57. [PMID: 26105050 DOI: 10.1074/jbc.m115.638924] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) ligands have been widely used to treat type 2 diabetes mellitus. However, knowledge of PPARγ signaling remains incomplete. In addition to PPARγ, these drugs also activate G protein-coupled receptor 40 (GPR40), a Gαq-coupled free fatty acid receptor linked to MAPK networks and glucose homeostasis. Notably, p38 MAPK activation has been implicated in PPARγ signaling. Here, rosiglitazone (RGZ) activation of GPR40 and p38 MAPK was found to boost PPARγ-induced gene transcription in human endothelium. Inhibition or knockdown of p38 MAPK or expression of a dominant negative (DN) p38 MAPK mutant blunted RGZ-induced PPARγ DNA binding and reporter activity in EA.hy926 human endothelial cells. GPR40 inhibition or knockdown, or expression of a DN-Gαq mutant likewise blocked activation of both p38 MAPK and PPARγ reporters. Importantly, RGZ induction of PPARγ target genes in primary human pulmonary artery endothelial cells (PAECs) was suppressed by knockdown of either p38 MAPK or GPR40. GPR40/PPARγ signal transduction was dependent on p38 MAPK activation and induction of PPARγ co-activator-1 (PGC1α). Silencing of p38 MAPK or GPR40 abolished the ability of RGZ to induce phosphorylation and expression of PGC1α in PAECs. Knockdown of PGC1α, its essential activator SIRT1, or its binding partner/co-activator EP300 inhibited RGZ induction of PPARγ-regulated genes in PAECs. RGZ/GPR40/p38 MAPK signaling also led to EP300 phosphorylation, an event that enhances PPARγ target gene transcription. Thus, GPR40 and PPARγ can function as an integrated two-receptor signal transduction pathway, a finding with implications for rational drug development.
Collapse
Affiliation(s)
- Shuibang Wang
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Keytam S Awad
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Jason M Elinoff
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Edward J Dougherty
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Gabriela A Ferreyra
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Jennifer Y Wang
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Rongman Cai
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Junfeng Sun
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Anetta Ptasinska
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| | - Robert L Danner
- From the Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
100
|
Hirasawa A. [Free Fatty Acid Receptor Family: A New Therapeutic Target for Metabolic Diseases]. YAKUGAKU ZASSHI 2015; 135:769-77. [PMID: 26028412 DOI: 10.1248/yakushi.14-00250-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Free fatty acids (FFAs) are not only essential nutritional components but they also act as signaling molecules in various physiological processes. A strategy to deorphanize G-protein-coupled receptors (GPCRs) identified a series of receptors for FFAs that play significant roles in nutrition regulation. In this free fatty acid receptor family, FFAR1 (GPR40) and FFAR4 (GPR120) are activated by long-chain FFAs. FFAR1 regulates insulin secretion in pancreatic β-cells, whereas FFAR4 promotes the secretion of glucagon-like peptide-1 (GLP-1) in the intestine, and also acts as a lipid sensor in adipose tissue to sense dietary fat and control energy balance. In this review, we discuss recent advances in the pharmacological characterization of FFAR1 and FFAR4, and we present a summary of current understandings of their physiological roles and their potential as drug targets.
Collapse
Affiliation(s)
- Akira Hirasawa
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Science, Kyoto University; 46-29 Yoshida-Shimo-Adachi-cho, Sakyo-ku, Kyoto 606-8501; 2) Inistitute for Integrated Medical Scinces, Tokyo Women's Medical University; 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|