1
|
Li Y, Li W, Zhou D, Zeng Z, Han Y, Chen Q, Wang Z, Wang G, Feng S, Cao W. Microcin Y utilizes its stable structure and biological activity to regulate the metabolism of intestinal probiotics and effectively clear gut Salmonella. Int J Biol Macromol 2024; 274:133290. [PMID: 38908631 DOI: 10.1016/j.ijbiomac.2024.133290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/09/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
MccY is a novel, structurally stable microcin with antibacterial activity against Enterobacteriaceae. However, the bioavailability of orally administrated MccY is unknown. This study evaluated the effects of MccY as a antimicrobial on pre-digestion in vitro and its intake, digestion and gut metabolism in vivo. The result of pre-digestion results that MccY maintained its biological activity and was resistant to decomposition. The study established a safe threshold of 4.46-9.92 mg/kg for the MccY dosage-body weight relationship in BALB/c mice. Mice fed with MccY demonstrated improved body weight and intestinal barrier function, accompanied with increased IgM immunogenicity and decreased levels of TNF-α, IL-6, and IL-10 in the intestine. MccY significantly facilitates the growth and activity of probiotics including Lactobacillus, Prevotella, and Bacteroides, and leading to the production of SCFAs and MCFAs during bacterial interactions. Furthermore, MccY effectively protects against the inflammatory response caused by Salmonella Typhimurium infection and effectively clears the Salmonella bacteria from the gut. In conclusion, MccY is seen as a promising new therapeutic target drug for enhancing the intestinal microbe-barrier axis and preventing enteritis.
Collapse
Affiliation(s)
- Yu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wenjing Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Di Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhiwei Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yu Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qinxi Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zepeng Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Guyao Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Saixiang Feng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, Guangzhou, China; Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China.
| | - Weisheng Cao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, China; Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, Guangzhou, China; Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, China.
| |
Collapse
|
2
|
Wu D, Weng S, Xu S, Li Y, Zhou J. Yiqi Huazhuo decoction increases insulin secretion in type 2 diabetic rats by regulating the pancreatic GPR40-IP3R-1 signaling pathway. Front Pharmacol 2023; 14:1136778. [PMID: 36998612 PMCID: PMC10043368 DOI: 10.3389/fphar.2023.1136778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
Objective: Yiqi Huazhuo Decoction (YD) reduces blood glucose, glycated hemoglobin, body weight, and insulin resistance in patients with type 2 diabetes mellitus (T2DM), but its exact mechanisms are unknown. This study investigated the therapeutic effects and mechanisms of YD on impaired insulin secretion in T2DM rats.Methods: T2DM rats were randomized to the model, YD-lo (15 mg/kg/d YD, 10 weeks), YD-hi (30 mg/kg/d YD, 10 weeks), positive drug (TAK-875), and healthy control groups. The rats underwent an oral glucose tolerance test (OGTT), glucose-stimulated insulin secretion (GSIS) test, and serum lipid measurements. High-fat and high-glucose-injured RIN-m5f cells were treated with YD (30 or 150 mg/mL) for 48 h. GPR40 and IP3R-1 expression levels were determined by immunofluorescence, qRT-PCR, and western blot.Results: Compared with the model group, the OGTT area under the curve (AUC) in the YD-hi group was decreased by 26.7%, the insulin release test (IRT) AUC in the YD-hi group was increased by 45.9%, and the GSIS AUC was increased by 33.9% (p < 0.05). Compared with the model cells, the insulin secretion after glucose stimulation in the YD-hi group was increased by 24.5%, similar to the TAK-875 group (23.1%) (p > 0.05). GPR40 and IP3R-1 mRNA in the model cells were decreased by 49.5% and 51.2% compared with the control cells (p < 0.05). In the YD-hi group, GPR40 and IP3R-1 mRNA levels were increased by 58.1% and 39.3% (p < 0.05), similar to the TAK-875 group. The changes in protein expression were similar to mRNA.Conclusion: YD promotes insulin secretion from pancreatic islet β-cell in T2DM rats by regulating the GPR40-IP3R-1 pathway, thereby reducing blood glucose.
Collapse
Affiliation(s)
- Dongjiao Wu
- Department of Rheumatology, Ningbo Municipal Hospital of TCM, Affliated Hospital of Zhejiang Chinese Medical University, Ningbo, China
| | - Siying Weng
- Department of Endocrinology, Ningbo Municipal Hospital of TCM, Affliated Hospital of Zhejiang Chinese Medical University, Ningbo, China
- *Correspondence: Siying Weng,
| | - Shuyi Xu
- College of the Third Clinical Medical, Ningbo Municipal Hospital of TCM, Affliated Hospital of Zhejiang Chinese Medical University, Ningbo, China
| | - Yan Li
- College of the Third Clinical Medical, Ningbo Municipal Hospital of TCM, Affliated Hospital of Zhejiang Chinese Medical University, Ningbo, China
| | - Jianyang Zhou
- Department of Endocrinology, Ningbo Municipal Hospital of TCM, Affliated Hospital of Zhejiang Chinese Medical University, Ningbo, China
| |
Collapse
|
3
|
Ježek P, Holendová B, Jabůrek M, Dlasková A, Plecitá-Hlavatá L. Contribution of Mitochondria to Insulin Secretion by Various Secretagogues. Antioxid Redox Signal 2022; 36:920-952. [PMID: 34180254 PMCID: PMC9125579 DOI: 10.1089/ars.2021.0113] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Significance: Mitochondria determine glucose-stimulated insulin secretion (GSIS) in pancreatic β-cells by elevating ATP synthesis. As the metabolic and redox hub, mitochondria provide numerous links to the plasma membrane channels, insulin granule vesicles (IGVs), cell redox, NADH, NADPH, and Ca2+ homeostasis, all affecting insulin secretion. Recent Advances: Mitochondrial redox signaling was implicated in several modes of insulin secretion (branched-chain ketoacid [BCKA]-, fatty acid [FA]-stimulated). Mitochondrial Ca2+ influx was found to enhance GSIS, reflecting cytosolic Ca2+ oscillations induced by action potential spikes (intermittent opening of voltage-dependent Ca2+ and K+ channels) or the superimposed Ca2+ release from the endoplasmic reticulum (ER). The ATPase inhibitory factor 1 (IF1) was reported to tune the glucose sensitivity range for GSIS. Mitochondrial protein kinase A was implicated in preventing the IF1-mediated inhibition of the ATP synthase. Critical Issues: It is unknown how the redox signal spreads up to the plasma membrane and what its targets are, what the differences in metabolic, redox, NADH/NADPH, and Ca2+ signaling, and homeostasis are between the first and second GSIS phase, and whether mitochondria can replace ER in the amplification of IGV exocytosis. Future Directions: Metabolomics studies performed to distinguish between the mitochondrial matrix and cytosolic metabolites will elucidate further details. Identifying the targets of cell signaling into mitochondria and of mitochondrial retrograde metabolic and redox signals to the cell will uncover further molecular mechanisms for insulin secretion stimulated by glucose, BCKAs, and FAs, and the amplification of secretion by glucagon-like peptide (GLP-1) and metabotropic receptors. They will identify the distinction between the hub β-cells and their followers in intact and diabetic states. Antioxid. Redox Signal. 36, 920-952.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Blanka Holendová
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Jabůrek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Dlasková
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lydie Plecitá-Hlavatá
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
4
|
Zhong YL, Ji Y, Wang H, Wang X, Gauthier DR. Highly Enantioselective Rhodium-Catalyzed Transfer Hydrogenation of Tetrasubstituted Olefins: Application toward the Synthesis of GPR40 Agonist MK-2305. Org Lett 2022; 24:3254-3258. [PMID: 35467884 DOI: 10.1021/acs.orglett.2c01021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A highly efficient enantioselective synthesis for the potent G-protein-coupled receptor 40 agonist MK-2305 was developed. The key tetrasubstituted olefin was prepared via a stereoselective Mukaiyama aldol reaction/elimination sequence. The highly enantioselective rhodium-catalyzed transfer hydrogenation of the tetrasubstituted olefin afforded the target compound MK-2305 in excellent optical and chemical purity. The key asymmetric transfer hydrogenation proceeds in excellent yields and enantioselectivities for a variety of substrates. The superior reactivity of the tethered catalysts was revealed by NMR studies.
Collapse
Affiliation(s)
- Yong-Li Zhong
- Department of Process Research and Development, Merck & Co., Inc., P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Yining Ji
- Department of Process Research and Development, Merck & Co., Inc., P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Heather Wang
- Department of Process Research and Development, Merck & Co., Inc., P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Xiao Wang
- Department of Process Research and Development, Merck & Co., Inc., P.O. Box 2000, Rahway, New Jersey 07065, United States
| | - Donald R Gauthier
- Department of Process Research and Development, Merck & Co., Inc., P.O. Box 2000, Rahway, New Jersey 07065, United States
| |
Collapse
|
5
|
Free fatty acid receptor 1: a ray of hope in the therapy of type 2 diabetes mellitus. Inflammopharmacology 2021; 29:1625-1639. [PMID: 34669065 DOI: 10.1007/s10787-021-00879-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022]
Abstract
Free fatty acid receptor 1 (FFAR1) is a G-protein coupled receptor with prominent expression on pancreatic beta cells, bones, intestinal cells as well as the nerve cells. This receptor mediates a multitude of functions in the body including release of incretins, secretion of insulin as well as sensation of pain. Since FFAR1 causes secretion of insulin and regulates glucose metabolism, efforts were made to unfold its structure followed by discovering agonists for the receptor and the utilization of these agonists in the therapy of type 2 diabetes mellitus. Development of such functional FFAR1 agonists is a necessity because the currently available therapy for type 2 diabetes mellitus has numerous drawbacks, of which, the major one is hypoglycemia. Since the most prominent effect of the FFAR1 agonists is on glucose concentration in the body, so the major research is focused on treating type 2 diabetes mellitus, though the agonists could benefit other metabolic disorders and neurological disorders as well. The agonists developed so far had one major limitation, i.e., hepatotoxicity. Although, the only agonist that could reach phase 3 clinical trials was TAK-875 developed by Takeda Pharmaceuticals but it was also withdrawn due to toxic effects on the liver. Thus, there are numerous agonists for the varied binding sites of the receptor but no drug available yet. There does seem to be a ray of hope in the drugs that target FFAR1 but a lot more efforts towards drug discovery would result in the successful management of type 2 diabetes mellitus.
Collapse
|
6
|
Henquin JC. Non-glucose modulators of insulin secretion in healthy humans: (dis)similarities between islet and in vivo studies. Metabolism 2021; 122:154821. [PMID: 34174327 DOI: 10.1016/j.metabol.2021.154821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022]
Abstract
Optimal metabolic homeostasis requires precise temporal and quantitative control of insulin secretion. Both in vivo and in vitro studies have often focused on the regulation by glucose although many additional factors including other nutrients, neurotransmitters, hormones and drugs, modulate the secretory function of pancreatic β-cells. This review is based on the analysis of clinical investigations characterizing the effects of non-glucose modulators of insulin secretion in healthy subjects, and of experimental studies testing the same modulators in islets isolated from normal human donors. The aim was to determine whether the information gathered in vitro can reliably be translated to the in vivo situation. The comparison evidenced both convincing similarities and areas of discordance. The lack of coherence generally stems from the use of exceedingly high concentrations of test agents at too high or too low glucose concentrations in vitro, which casts doubts on the physiological relevance of a number of observations made in isolated islets. Future projects resorting to human islets should avoid extreme experimental conditions, such as oversized stimulations or inhibitions of β-cells, which are unlikely to throw light on normal insulin secretion and contribute to the elucidation of its defects.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium.
| |
Collapse
|
7
|
Rani L, Grewal AS, Sharma N, Singh S. Recent Updates on Free Fatty Acid Receptor 1 (GPR-40) Agonists for the Treatment of Type 2 Diabetes Mellitus. Mini Rev Med Chem 2021; 21:426-470. [PMID: 33100202 DOI: 10.2174/1389557520666201023141326] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The global incidence of type 2 diabetes mellitus (T2DM) has enthused the development of new antidiabetic targets with low toxicity and long-term stability. In this respect, free fatty acid receptor 1 (FFAR1), which is also recognized as a G protein-coupled receptor 40 (GPR40), is a novel target for the treatment of T2DM. FFAR1/GPR40 has a high level of expression in β-cells of the pancreas, and the requirement of glucose for stimulating insulin release results in immense stimulation to utilise this target in the medication of T2DM. METHODS The data used for this review is based on the search of several scienctific databases as well as various patent databases. The main search terms used were free fatty acid receptor 1, FFAR1, FFAR1 agonists, diabetes mellitus, G protein-coupled receptor 40 (GPR40), GPR40 agonists, GPR40 ligands, type 2 diabetes mellitus and T2DM. RESULTS The present review article gives a brief overview of FFAR1, its role in T2DM, recent developments in small molecule FFAR1 (GPR40) agonists reported till now, compounds of natural/plant origin, recent patents published in the last few years, mechanism of FFAR1 activation by the agonists, and clinical status of the FFAR1/GPR40 agonists. CONCLUSION The agonists of FFAR1/GRP40 showed considerable potential for the therapeutic control of T2DM. Most of the small molecule FFAR1/GPR40 agonists developed were aryl alkanoic acid derivatives (such as phenylpropionic acids, phenylacetic acids, phenoxyacetic acids, and benzofuran acetic acid derivatives) and thiazolidinediones. Some natural/plant-derived compounds, including fatty acids, sesquiterpenes, phenolic compounds, anthocyanins, isoquinoline, and indole alkaloids, were also reported as potent FFAR1 agonists. The clinical investigations of the FFAR1 agonists demonstrated their probable role in the improvement of glucose control. Though, there are some problems still to be resolved in this field as some FFAR1 agonists terminated in the late phase of clinical studies due to "hepatotoxicity." Currently, PBI-4050 is under clinical investigation by Prometic. Further investigation of pharmacophore scaffolds for FFAR1 full agonists as well as multitargeted modulators and corresponding clinical investigations will be anticipated, which can open up new directions in this area.
Collapse
Affiliation(s)
- Lata Rani
- Chitkara University School of Basic Sciences, Chitkara University, Himachal Pradesh, India
| | - Ajmer Singh Grewal
- Chitkara University School of Basic Sciences, Chitkara University, Himachal Pradesh, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
8
|
In silico design of bioisosteric modifications of drugs for the treatment of diabetes. Future Med Chem 2021; 13:691-700. [PMID: 33715419 DOI: 10.4155/fmc-2020-0374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Aim: To identify virtual bioisosteric replacements of two GPR40 agonists. Materials & methods: Bioinformatic docking of candidate molecules featuring a wide range of carboxylic acid bioisosteres into complex with GPR40 was performed using TAK-875 and GW9508 templates. Results: This study suggests that 2,6-difluorophenol and squaric acid motifs are the preferred bioisosteric groups for conferring GPR40 affinity. Conclusion: This study suggests that compounds 10 and 20 are worthy synthetic targets.
Collapse
|
9
|
Ghislain J, Poitout V. Targeting lipid GPCRs to treat type 2 diabetes mellitus - progress and challenges. Nat Rev Endocrinol 2021; 17:162-175. [PMID: 33495605 DOI: 10.1038/s41574-020-00459-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/04/2020] [Indexed: 02/07/2023]
Abstract
Therapeutic approaches to the treatment of type 2 diabetes mellitus that are designed to increase insulin secretion either directly target β-cells or indirectly target gastrointestinal enteroendocrine cells (EECs), which release hormones that modulate insulin secretion (for example, incretins). Given that β-cells and EECs both express a large array of G protein-coupled receptors (GPCRs) that modulate insulin secretion, considerable research and development efforts have been undertaken to design therapeutic drugs targeting these GPCRs. Among them are GPCRs specific for free fatty acid ligands (lipid GPCRs), including free fatty acid receptor 1 (FFA1, otherwise known as GPR40), FFA2 (GPR43), FFA3 (GPR41) and FFA4 (GPR120), as well as the lipid metabolite binding glucose-dependent insulinotropic receptor (GPR119). These lipid GPCRs have demonstrated important roles in the control of islet and gut hormone secretion. Advances in lipid GPCR pharmacology have led to the identification of a number of synthetic agonists that exert beneficial effects on glucose homeostasis in preclinical studies. Yet, translation of these promising results to the clinic has so far been disappointing. In this Review, we present the physiological roles, pharmacology and clinical studies of these lipid receptors and discuss the challenges associated with their clinical development for the treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Julien Ghislain
- Montreal Diabetes Research Center, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada.
- Department of Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
10
|
Grundmann M, Bender E, Schamberger J, Eitner F. Pharmacology of Free Fatty Acid Receptors and Their Allosteric Modulators. Int J Mol Sci 2021; 22:ijms22041763. [PMID: 33578942 PMCID: PMC7916689 DOI: 10.3390/ijms22041763] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 12/19/2022] Open
Abstract
The physiological function of free fatty acids (FFAs) has long been regarded as indirect in terms of their activities as educts and products in metabolic pathways. The observation that FFAs can also act as signaling molecules at FFA receptors (FFARs), a family of G protein-coupled receptors (GPCRs), has changed the understanding of the interplay of metabolites and host responses. Free fatty acids of different chain lengths and saturation statuses activate FFARs as endogenous agonists via binding at the orthosteric receptor site. After FFAR deorphanization, researchers from the pharmaceutical industry as well as academia have identified several ligands targeting allosteric sites of FFARs with the aim of developing drugs to treat various diseases such as metabolic, (auto)inflammatory, infectious, endocrinological, cardiovascular, and renal disorders. GPCRs are the largest group of transmembrane proteins and constitute the most successful drug targets in medical history. To leverage the rich biology of this target class, the drug industry seeks alternative approaches to address GPCR signaling. Allosteric GPCR ligands are recognized as attractive modalities because of their auspicious pharmacological profiles compared to orthosteric ligands. While the majority of marketed GPCR drugs interact exclusively with the orthosteric binding site, allosteric mechanisms in GPCR biology stay medically underexploited, with only several allosteric ligands currently approved. This review summarizes the current knowledge on the biology of FFAR1 (GPR40), FFAR2 (GPR43), FFAR3 (GPR41), FFAR4 (GPR120), and GPR84, including structural aspects of FFAR1, and discusses the molecular pharmacology of FFAR allosteric ligands as well as the opportunities and challenges in research from the perspective of drug discovery.
Collapse
Affiliation(s)
- Manuel Grundmann
- Research and Early Development, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany;
- Correspondence:
| | - Eckhard Bender
- Drug Discovery Sciences, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany; (E.B.); (J.S.)
| | - Jens Schamberger
- Drug Discovery Sciences, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany; (E.B.); (J.S.)
| | - Frank Eitner
- Research and Early Development, Bayer Pharmaceuticals, Bayer AG, 42096 Wuppertal, Germany;
| |
Collapse
|
11
|
Zhao YF, Li XC, Liang XY, Zhao YY, Xie R, Zhang LJ, Zhang XC, Chen C. GPR120 Regulates Pancreatic Polypeptide Secretion From Male Mouse Islets via PLC-Mediated Calcium Mobilization. Endocrinology 2020; 161:5900686. [PMID: 32877513 DOI: 10.1210/endocr/bqaa157] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 02/07/2023]
Abstract
The free fatty acid receptor G protein-coupled receptor 120 (GPR120) is expressed in pancreatic islets, but its specific cell distribution and function have not been fully established. In this study, a GPR120-IRES-EGFP knockin (KI) mouse was generated to identify GPR120-expressing cells with enhanced green fluorescence proteins (EGFP). EGFP-positive cells collected from KI mouse islets by flow cytometry had a significantly higher expression of pancreatic polypeptide (PP) evidenced by reverse transcriptase (RT)-quantitative polymerase chain reaction (qPCR). Single-cell RT-PCR and immunocytochemical double staining also demonstrated the coexpression of GPR120 with PP in mouse islets. The GPR120-specific agonist TUG-891 significantly increased plasma PP levels in mice. TUG-891 significantly increased PP levels in islet medium in vitro, which was markedly attenuated by GPR120 small interfering RNA treatment. TUG-891-stimulated PP secretion in islets was fully blocked by pretreatment with YM-254890 (a Gq protein inhibitor), U73122 (a phospholipase C inhibitor), or thapsigargin (an inducer of endoplasmic reticulum Ca2+ depletion), respectively. TUG-891 triggered the increase in intracellular free Ca2+ concentrations ([Ca2+]i) in PP cells, which was also eliminated by YM-254890, U73122, or thapsigargin. GPR120 gene expression was significantly reduced in islets of high-fat diet (HFD)-induced obese mice. TUG-891-stimulated PP secretion was also significantly diminished in vivo and in vitro in HFD-induced obese mice compared with that in normal-chow diet control mice. In summary, this study demonstrated that GPR120 is expressed in mouse islet PP cells and GPR120 activation stimulated PP secretion via the Gq/PLC-Ca2+ signaling pathway in normal-chow diet mice but with diminished effects in HFD-induced obese mice.
Collapse
Affiliation(s)
- Yu-Feng Zhao
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Xiao-Cheng Li
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Xiang-Yan Liang
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Yan-Yan Zhao
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Rong Xie
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Li-Jun Zhang
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Xiao-Chun Zhang
- Institute of Basic Medical Sciences, Xi'an Medical University, Xi'an, China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| |
Collapse
|
12
|
Li Z, Liu C, Zhou Z, Hu L, Deng L, Ren Q, Qian H. A novel FFA1 agonist, CPU025, improves glucose-lipid metabolism and alleviates fatty liver in obese-diabetic (ob/ob) mice. Pharmacol Res 2020; 153:104679. [DOI: 10.1016/j.phrs.2020.104679] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/20/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022]
|
13
|
Bartoszek A, Moo EV, Binienda A, Fabisiak A, Krajewska JB, Mosińska P, Niewinna K, Tarasiuk A, Martemyanov K, Salaga M, Fichna J. Free Fatty Acid Receptors as new potential therapeutic target in inflammatory bowel diseases. Pharmacol Res 2019; 152:104604. [PMID: 31846762 DOI: 10.1016/j.phrs.2019.104604] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/19/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023]
Abstract
Family of Free Fatty Acid Receptors (FFARs), specific G protein-coupled receptors comprises of four members: FFAR1-4, where each responds to different chain length of fatty acids (FAs). Over the years, FFARs have become attractive pharmacological targets in the treatment of type 2 diabetes, metabolic syndrome, cardiovascular diseases and asthma; recent studies also point to their role in inflammation. It is now well-established that activation of FFAR1 and FFAR4 by long and medium chain FAs may lead to reduction of inflammatory state; FFAR2 and FFAR3 are activated by short chain FAs, but only FFAR2 was shown to alleviate inflammation, mostly by neutrophil inhibition. All FFARs have thus been proposed as targets in inflammatory bowel diseases (IBD). Here we discuss current knowledge and future directions in FFAR research related to IBD.
Collapse
Affiliation(s)
- Adrian Bartoszek
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Ee Von Moo
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Agata Binienda
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Adam Fabisiak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland; Department of Digestive Tract Diseases, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Julia B Krajewska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Paula Mosińska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Karolina Niewinna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Aleksandra Tarasiuk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Kirill Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Maciej Salaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
14
|
GPR40 activation initiates store-operated Ca 2+ entry and potentiates insulin secretion via the IP3R1/STIM1/Orai1 pathway in pancreatic β-cells. Sci Rep 2019; 9:15562. [PMID: 31664108 PMCID: PMC6820554 DOI: 10.1038/s41598-019-52048-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/12/2019] [Indexed: 12/17/2022] Open
Abstract
The long-chain fatty acid receptor GPR40 plays an important role in potentiation of glucose-induced insulin secretion (GIIS) from pancreatic β-cells. Previous studies demonstrated that GPR40 activation enhances Ca2+ release from the endoplasmic reticulum (ER) by activating inositol 1,4,5-triphosphate (IP3) receptors. However, it remains unknown how ER Ca2+ release via the IP3 receptor is linked to GIIS potentiation. Recently, stromal interaction molecule (STIM) 1 was identified as a key regulator of store-operated Ca2+ entry (SOCE), but little is known about its contribution in GPR40 signaling. We show that GPR40-mediated potentiation of GIIS is abolished by knockdown of IP3 receptor 1 (IP3R1), STIM1 or Ca2+-channel Orai1 in insulin-secreting MIN6 cells. STIM1 and Orai1 knockdown significantly impaired SOCE and the increase of intracellular Ca2+ by the GPR40 agonist, fasiglifam. Furthermore, β-cell-specific STIM1 knockout mice showed impaired fasiglifam-mediated GIIS potentiation not only in isolated islets but also in vivo. These results indicate that the IP3R1/STIM1/Orai1 pathway plays an important role in GPR40-mediated SOCE initiation and GIIS potentiation in pancreatic β-cells.
Collapse
|
15
|
Kimura I, Ichimura A, Ohue-Kitano R, Igarashi M. Free Fatty Acid Receptors in Health and Disease. Physiol Rev 2019; 100:171-210. [PMID: 31487233 DOI: 10.1152/physrev.00041.2018] [Citation(s) in RCA: 488] [Impact Index Per Article: 97.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fatty acids are metabolized and synthesized as energy substrates during biological responses. Long- and medium-chain fatty acids derived mainly from dietary triglycerides, and short-chain fatty acids (SCFAs) produced by gut microbial fermentation of the otherwise indigestible dietary fiber, constitute the major sources of free fatty acids (FFAs) in the metabolic network. Recently, increasing evidence indicates that FFAs serve not only as energy sources but also as natural ligands for a group of orphan G protein-coupled receptors (GPCRs) termed free fatty acid receptors (FFARs), essentially intertwining metabolism and immunity in multiple ways, such as via inflammation regulation and secretion of peptide hormones. To date, several FFARs that are activated by the FFAs of various chain lengths have been identified and characterized. In particular, FFAR1 (GPR40) and FFAR4 (GPR120) are activated by long-chain saturated and unsaturated fatty acids, while FFAR3 (GPR41) and FFAR2 (GPR43) are activated by SCFAs, mainly acetate, butyrate, and propionate. In this review, we discuss the recent reports on the key physiological functions of the FFAR-mediated signaling transduction pathways in the regulation of metabolism and immune responses. We also attempt to reveal future research opportunities for developing therapeutics for metabolic and immune disorders.
Collapse
Affiliation(s)
- Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Atsuhiko Ichimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Ryuji Ohue-Kitano
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Miki Igarashi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| |
Collapse
|
16
|
Ackerson T, Amberg A, Atzrodt J, Arabeyre C, Defossa E, Dorau M, Dudda A, Dwyer J, Holla W, Kissner T, Kohlmann M, Kürzel U, Pánczél J, Rajanna S, Riedel J, Schmidt F, Wäse K, Weitz D, Derdau V. Mechanistic investigations of the liver toxicity of the free fatty acid receptor 1 agonist fasiglifam (TAK875) and its primary metabolites. J Biochem Mol Toxicol 2019; 33:e22345. [DOI: 10.1002/jbt.22345] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/23/2019] [Accepted: 04/04/2019] [Indexed: 12/21/2022]
Affiliation(s)
| | | | - Jens Atzrodt
- Integrated Drug Discovery, Sanofi Frankfurt Germany
| | | | | | | | - Angela Dudda
- Global Project Management Unit, DCV, Sanofi Frankfurt Germany
| | | | | | | | - Markus Kohlmann
- Global Project Management Unit, DCV, Sanofi Frankfurt Germany
| | - Ulrich Kürzel
- Drug Metabolism and Pharmacokinetics, Sanofi Frankfurt Germany
| | - József Pánczél
- Drug Metabolism and Pharmacokinetics, Sanofi Frankfurt Germany
| | | | - Jens Riedel
- Drug Metabolism and Pharmacokinetics, Sanofi Frankfurt Germany
| | | | | | - Dietmar Weitz
- Drug Metabolism and Pharmacokinetics, Sanofi Frankfurt Germany
| | | |
Collapse
|
17
|
Freitas RDS, Campos MM. Protective Effects of Omega-3 Fatty Acids in Cancer-Related Complications. Nutrients 2019; 11:nu11050945. [PMID: 31035457 PMCID: PMC6566772 DOI: 10.3390/nu11050945] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/24/2022] Open
Abstract
Omega-3 polyunsaturated fatty acids (PUFAs) are considered immunonutrients and are commonly used in the nutritional therapy of cancer patients due to their ample biological effects. Omega-3 PUFAs play essential roles in cell signaling and in the cell structure and fluidity of membranes. They participate in the resolution of inflammation and have anti-inflammatory and antinociceptive effects. Additionally, they can act as agonists of G protein-coupled receptors, namely, GPR40/FFA1 and GPR120/FFA4. Cancer patients undergo complications, such as anorexia-cachexia syndrome, pain, depression, and paraneoplastic syndromes. Interestingly, the 2017 European Society for Clinical Nutrition and Metabolism (ESPEN) guidelines for cancer patients only discuss the use of omega-3 PUFAs for cancer-cachexia treatment, leaving aside other cancer-related complications that could potentially be managed by omega-3 PUFA supplementation. This critical review aimed to discuss the effects and the possible underlying mechanisms of omega-3 PUFA supplementation in cancer-related complications. Data compilation in this critical review indicates that further investigation is still required to assess the factual benefits of omega-3 PUFA supplementation in cancer-associated illnesses. Nevertheless, preclinical evidence reveals that omega-3 PUFAs and their metabolites might modulate pivotal pathways underlying complications secondary to cancer, indicating that this is a promising field of knowledge to be explored.
Collapse
Affiliation(s)
- Raquel D S Freitas
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre 90619-900, RS, Brazil.
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, PUCRS, Porto Alegre 90619-900, RS, Brazil.
| | - Maria M Campos
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre 90619-900, RS, Brazil.
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, PUCRS, Porto Alegre 90619-900, RS, Brazil.
- Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre 90619-900, RS, Brazil.
| |
Collapse
|
18
|
Nath V, Ahuja R, Kumar V. Identification of novel G-protein-coupled receptor 40 (GPR40) agonists by hybrid in silico-screening techniques and molecular dynamics simulations thereof. J Biomol Struct Dyn 2018; 37:3764-3787. [DOI: 10.1080/07391102.2018.1527255] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Virendra Nath
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Rohini Ahuja
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Vipin Kumar
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
19
|
Marcinak JF, Munsaka MS, Watkins PB, Ohira T, Smith N. Liver Safety of Fasiglifam (TAK-875) in Patients with Type 2 Diabetes: Review of the Global Clinical Trial Experience. Drug Saf 2018; 41:625-640. [PMID: 29492878 DOI: 10.1007/s40264-018-0642-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Fasiglifam (TAK-875) is a G protein-coupled receptor 40 agonist that was being investigated for treatment of type 2 diabetes mellitus (T2DM). A development program was terminated late in phase III clinical trials due to liver safety concerns. METHODS The liver safety of fasiglifam was assessed from data based on six phase II and nine phase III double-blind studies and two open-label studies with emphasis on pooled data from 15 double-blind studies from both global and Japanese development programs. Taking into consideration different daily doses of fasiglifam administered in clinical studies, the primary comparisons were between all patients exposed to fasiglifam (any dose) versus placebo, and, where applicable, versus the two active comparators, sitagliptin or glimepiride. A Liver Safety Evaluation Committee consisting of hepatologists blinded to treatment assignments evaluated hepatic adverse events (AEs) and serious AEs (SAEs) for causal relationship to study drug. RESULTS The analysis included data from 9139 patients with T2DM in 15 double-blind controlled studies who received either fasiglifam (n = 5359, fasiglifam group), fasiglifam and sitagliptin (n = 123), or a comparator agent (n = 3657, non-exposed group consisting of placebo and other antidiabetic agents). Exposure to treatment for more than 1 year ranged from 249 patients in the placebo arm, to 370 patients in the glimepiride arm and 617 patients in the fasiglifam 50 mg arm. The primary focus of the analysis was on the hepatic safety of fasiglifam. The overall safety profile based on treatment-emergent AEs (TEAEs), SAEs, deaths, and withdrawal due to AEs was similar between fasiglifam and placebo (excluding liver test abnormalities). However, there was an increased incidence rate of serum alanine aminotransferase (ALT) elevations > 3 × upper limit of normal (ULN), 5 × ULN, and 10 × ULN in fasiglifam-treated patients compared with those treated with placebo or active comparators. ALT elevations > 3 × ULN for fasiglifam were 2.7% compared with 0.8 and 0.5% for the active comparators and placebo. There did not appear to be a clear dose response in incidence of ALT elevations between patients receiving 25 or 50 mg daily. The cumulative incidence of elevations in serum ALT > 3 × ULN was higher in the first 6 months of treatment with fasiglifam compared with both placebo and the active comparators, but the rate of new ALT elevations appeared to be similar across all treatment groups thereafter. No demographic or baseline patient characteristics were identified to predict elevations exceeding ALT > 3 × ULN in fasiglifam-treated patients. The pattern of liver injury with fasiglifam was hepatocellular, and there were no reports of liver-related deaths, liver failure or life-threatening liver injury. Most fasiglifam-associated ALT elevations were asymptomatic and resolved promptly upon discontinuing treatment, but in two patients the recovery was prolonged. Importantly, three important serious liver injury cases were identified among fasiglifam-treated patients; one case was adjudicated to be a clear Hy's Law case and the two remaining cases were considered to closely approximate Hy's Law cases. CONCLUSIONS Although the incidence of overall AEs, SAEs, and deaths was similar between fasiglifam and placebo, a liver signal was identified based primarily on the difference in liver chemistry values in the fasiglifam group compared with the placebo and active comparator groups. Three serious liver injuries were attributed to fasiglifam treatment. Clinical development of fasiglifam was halted due to these liver safety concerns.
Collapse
Affiliation(s)
- John F Marcinak
- Takeda Pharmaceuticals, Takeda Development Center Americas, Inc., 1 Takeda Pkwy, Deerfield, IL, 60015, USA.
| | - Melvin S Munsaka
- Takeda Pharmaceuticals, Takeda Development Center Americas, Inc., 1 Takeda Pkwy, Deerfield, IL, 60015, USA
| | - Paul B Watkins
- Eshelman School of Pharmacy, Institute for Drug Safety Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Takashi Ohira
- Takeda Pharmaceutical Company Limited, 1-1, Doshomachi 4-chome, Chuo-ku, Osaka, Japan
| | - Neila Smith
- Takeda Pharmaceuticals, Takeda Development Center Americas, Inc., 1 Takeda Pkwy, Deerfield, IL, 60015, USA
| |
Collapse
|
20
|
Sloop KW, Emmerson PJ, Statnick MA, Willard FS. The current state of GPCR-based drug discovery to treat metabolic disease. Br J Pharmacol 2018; 175:4060-4071. [PMID: 29394497 DOI: 10.1111/bph.14157] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/14/2017] [Accepted: 01/19/2018] [Indexed: 02/06/2023] Open
Abstract
One approach of modern drug discovery is to identify agents that enhance or diminish signal transduction cascades in various cell types and tissues by modulating the activity of GPCRs. This strategy has resulted in the development of new medicines to treat many conditions, including cardiovascular disease, psychiatric disorders, HIV/AIDS, certain forms of cancer and Type 2 diabetes mellitus (T2DM). These successes justify further pursuit of GPCRs as disease targets and provide key learning that should help guide identifying future therapeutic agents. This report reviews the current landscape of GPCR drug discovery with emphasis on efforts aimed at developing new molecules for treating T2DM and obesity. We analyse historical efforts to generate GPCR-based drugs to treat metabolic disease in terms of causal factors leading to success and failure in this endeavour. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Kyle W Sloop
- Diabetes and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Paul J Emmerson
- Diabetes and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Michael A Statnick
- Diabetes and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Francis S Willard
- Quantitative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| |
Collapse
|
21
|
Marcinak J, Vakilynejad M, Kogame A, Tagawa Y. Evaluation of the Pharmacokinetics and Safety of a Single Oral Dose of Fasiglifam in Subjects with Mild or Moderate Hepatic Impairment. Drugs R D 2018; 18:109-118. [PMID: 29488154 PMCID: PMC5995786 DOI: 10.1007/s40268-018-0229-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND AIMS Fasiglifam, a potent, selective novel agonist of G protein-coupled receptor 40, stimulates insulin secretion at elevated blood glucose levels in a glucose-dependent manner. This study evaluated the potential effect of hepatic impairment on the pharmacokinetics and safety of a single dose of fasiglifam and its metabolite M-I. Fasiglifam's clinical development was halted due to liver safety concerns. METHODS In this phase I, open-label study, subjects with mild or moderate hepatic impairment, along with matched controls (gender, weight, age, and smoking status), received a single, 25-mg oral dose of fasiglifam. Blood samples were collected through 336 h post-dose for pharmacokinetic evaluation. RESULTS Overall, 73% of subjects were male with a mean age of 54 years. Compared with normal hepatic function subjects (n = 14), mean systemic fasiglifam exposure (Cmax and AUC∞) was reduced in mild (n = 8) and moderate (n = 8) hepatic impairment subjects by approximately 20-40%. However, the observed percent unbound drug plasma concentration appeared comparable across all groups. Mean oral clearance was higher and terminal half-life lower in subjects with mild or moderate hepatic impairment compared with normal hepatic function subjects. Fasiglifam M-I systemic exposure increased by approximately twofold in subjects with mild or moderate hepatic impairment compared with those with normal hepatic function. Fasiglifam was well tolerated, and there were no reports of hypoglycemia. CONCLUSION Hepatic status did not significantly impact systemic exposure of fasiglifam in this study, in fact, a decrease was observed, suggesting no dose reduction would be required for patients with hepatic impairment.
Collapse
Affiliation(s)
- John Marcinak
- Pharmacovigilence, Takeda Development Center Americas Inc., 1 Takeda Parkway, Deerfield, IL, 60015, USA.
| | - Majid Vakilynejad
- Quantitative Clinical Pharmacology, Takeda Development Center Americas Inc., Deerfield, IL, USA
| | | | | |
Collapse
|
22
|
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
|
23
|
Marcinak J, Cao C, Lee D, Ye Z. Fasiglifam for glycaemic control in people with type 2 diabetes: A phase III, placebo-controlled study. Diabetes Obes Metab 2017; 19:1714-1721. [PMID: 28493502 DOI: 10.1111/dom.13004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/03/2017] [Accepted: 05/07/2017] [Indexed: 01/20/2023]
Abstract
AIM To investigate the effect of fasiglifam on glycaemic control in people with type 2 diabetes mellitus (T2DM). METHODS In total, 421 people with T2DM and glycated haemoglobin (HbA1c) ≥7.0% and ≤10.5% who had received only diet and exercise treatment for ≥12 weeks prior to screening were randomized to receive fasiglifam 25 or 50 mg or placebo. The primary efficacy endpoint was change from baseline in HbA1c at week 24. RESULTS The mean participant age was 53.5 years, mean baseline body mass index 32.3 kg/m2 , and mean baseline HbA1c level 8.05%. Least squares mean changes in HbA1c from baseline to week 24 were: -0.93% (fasiglifam 50 mg), -0.65% (fasiglifam 25 mg) and -0.17% (placebo). Treatment-emergent adverse events (TEAEs) occurred in 53.3%, 48.2% and 39.9% of participants receiving fasiglifam 25 mg, fasiglifam 50 mg, and placebo, respectively. Three participants in each group experienced a serious adverse event (AE). Nine participants had alanine aminotransferase (ALT) elevations >3× upper limit of normal: 5 (3.6%) in the fasiglifam 25-mg group, 4 (2.8%) in the fasiglifam 50-mg group, and none in the placebo group. CONCLUSIONS The data indicate that fasiglifam effectively reduced HbA1c from baseline for 24 weeks in participants with T2DM. The incidence of TEAEs was higher in the fasiglifam groups; however, the incidence of serious AEs was low overall and similar between groups. ALT elevations were observed only in the fasiglifam groups, which contributed to the decision to terminate the fasiglifam programme after completion of the present study.
Collapse
Affiliation(s)
- John Marcinak
- Takeda Development Center Americas, Inc., Deerfield, Illinois
| | - Charles Cao
- Takeda Development Center Americas, Inc., Deerfield, Illinois
| | - Douglas Lee
- Takeda Development Centre Europe, Ltd., London, UK
| | - Zhan Ye
- Takeda Development Center Americas, Inc., Deerfield, Illinois
| |
Collapse
|
24
|
Joyal JS, Gantner ML, Smith LEH. Retinal energy demands control vascular supply of the retina in development and disease: The role of neuronal lipid and glucose metabolism. Prog Retin Eye Res 2017; 64:131-156. [PMID: 29175509 DOI: 10.1016/j.preteyeres.2017.11.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/11/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Jean-Sébastien Joyal
- Department of Pediatrics, Pharmacology and Ophthalmology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Canada.
| | - Marin L Gantner
- The Lowy Medical Research Institute, La Jolla, United States
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston MA 02115, United States.
| |
Collapse
|
25
|
Quercetin/oleic acid-based G-protein-coupled receptor 40 ligands as new insulin secretion modulators. Future Med Chem 2017; 9:1873-1885. [DOI: 10.4155/fmc-2017-0113] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Aim: Management of Type 2 diabetes mellitus by diet is achievable at the early stage of the disease; patients usually underestimate this approach and an appropriate drug therapy is required. Results: Starting from quercetin and oleic acid, that have effect on insulin secretion, a small set of hybrid molecules was synthesized. Insulin secretion was evaluated in both in vitro and ex vivo models. AV1 was able to enhance insulin secretion dose dependently, behaving as a conceivable agonist of G-protein-coupled receptor 40. Conclusion: AV1 represents an interesting tool that interacts with G-protein-coupled receptor 40. Further studies will be carried out to evaluate the exact binding mode, and also to enlarge the library of these antidiabetic agents. [Formula: see text]
Collapse
|
26
|
Peng XV, Marcinak JF, Raanan MG, Cao C. Combining the G-protein-coupled receptor 40 agonist fasiglifam with sitagliptin improves glycaemic control in patients with type 2 diabetes with or without metformin: A randomized, 12-week trial. Diabetes Obes Metab 2017; 19:1127-1134. [PMID: 28239939 DOI: 10.1111/dom.12921] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 02/15/2017] [Indexed: 12/23/2022]
Abstract
AIMS To evaluate the efficacy and safety of fasiglifam, an orally active G-protein-coupled receptor 40 agonist, in combination with the dipeptidyl peptidase-4 inhibitor sitagliptin, in patients with type 2 diabetes inadequately controlled with diet/exercise (± metformin). MATERIALS AND METHODS In this randomized, double-blind, phase II study, 368 patients received once-daily placebo, sitagliptin 100 mg, fasiglifam 25 or 50 mg, or the combination of sitagliptin 100 mg plus fasiglifam 25 or 50 mg. The primary endpoint was change from baseline glycated haemoglobin (HbA1c) at 12 weeks; a key secondary endpoint was change in fasting plasma glucose (FPG). RESULTS The fasiglifam 25 and 50 mg combination regimens produced significantly greater HbA1c reductions than sitagliptin (treatment differences of -0.45% and -0.61%; P < .01, respectively) or respective doses of fasiglifam monotherapy (-0.43% and -0.48%; P < .01) and significantly greater FPG reductions than sitagliptin (-1.1 mmol/L for both combination regimens; P < .01). Improved glycaemic control occurred by week 1 for FPG and week 4 for HbA1c in all groups. Hypoglycaemia rates were low (≤3.3%) and similar across treatments. Liver enzymes >3 × upper limit of normal occurred in four patients (fasiglifam 25 mg, n = 1; fasiglifam 50 mg, n = 2; 1 fasiglifam/sitagliptin 50/100 mg, n = 1). CONCLUSIONS Combination of fasiglifam and sitagliptin provided significant additional effects on glycaemic control, with hypoglycaemia rates similar to placebo with or without metformin. This study provides supportive clinical evidence for the complementary mechanism of actions of this GPR40 agonist and DPP-4 inhibitor.
Collapse
Affiliation(s)
- Xuejun V Peng
- Takeda Development Center Americas, Inc., Deerfield, Illinois
| | - John F Marcinak
- Takeda Development Center Americas, Inc., Deerfield, Illinois
| | - Marsha G Raanan
- Takeda Development Center Americas, Inc., Deerfield, Illinois
| | - Charlie Cao
- Takeda Development Center Americas, Inc., Deerfield, Illinois
| |
Collapse
|
27
|
Sabrautzki S, Kaiser G, Przemeck GKH, Gerst F, Lorza-Gil E, Panse M, Sartorius T, Hoene M, Marschall S, Häring HU, Hrabě de Angelis M, Ullrich S. Point mutation of Ffar1 abrogates fatty acid-dependent insulin secretion, but protects against HFD-induced glucose intolerance. Mol Metab 2017; 6:1304-1312. [PMID: 29031729 PMCID: PMC5641630 DOI: 10.1016/j.molmet.2017.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 07/05/2017] [Accepted: 07/11/2017] [Indexed: 01/06/2023] Open
Abstract
Objective The fatty acid receptor 1 (FFAR1/GPR40) mediates fatty acid-dependent augmentation of glucose-induced insulin secretion (GIIS) in pancreatic β-cells. Genetically engineered Ffar1-knockout/congenic mice univocally displayed impaired fatty acid-mediated insulin secretion, but in vivo experiments delivered controversial results regarding the function of FFAR1 in glucose homeostasis and liver steatosis. This study presents a new coisogenic mouse model carrying a point mutation in Ffar1 with functional consequence. These mice reflect the situations in humans in which point mutations can lead to protein malfunction and disease development. Methods The Munich N-ethyl-N-nitrosourea (ENU) mutagenesis-derived F1 archive containing over 16,800 sperms and corresponding DNA samples was screened for mutations in the coding region of Ffar1. Two missense mutations (R258W and T146S) in the extracellular domain of the protein were chosen and homozygote mice were generated. The functional consequence of these mutations was examined in vitro in isolated islets and in vivo in chow diet and high fat diet fed mice. Results Palmitate, 50 μM, and the FFAR1 agonist TUG-469, 3 μM, stimulated insulin secretion in islets of Ffar1T146S/T146S mutant mice and of wild-type littermates, while in islets of Ffar1R258W/R258W mutant mice, these stimulatory effects were abolished. Insulin content and mRNA levels of Ffar1, Glp1r, Ins2, Slc2a2, Ppara, and Ppard were not significantly different between wild-type and Ffar1R258W/R258W mouse islets. Palmitate exposure, 600 μM, significantly increased Ppara mRNA levels in wild-type but not in Ffar1R258W/R258W mouse islets. On the contrary, Slc2a2 mRNA levels were significantly reduced in both wild-type and Ffar1R258W/R258W mouse islets after palmitate treatment. HFD feeding induced glucose intolerance in wild-type mice. Ffar1R258W/R258W mutant mice remained glucose tolerant although their body weight gain, liver steatosis, insulin resistance, and plasma insulin levels were not different from those of wild-type littermates. Worth mentioning, fasting plasma insulin levels were lower in Ffar1R258W/R258W mice. Conclusion A point mutation in Ffar1 abrogates the stimulatory effect of palmitate on GIIS, an effect that does not necessarily translate to HFD-induced glucose intolerance. Generation of mice carrying point mutations in Ffar1 using ENU. FFAR1 point mutation R258W abrogates fatty acid-induced insulin secretion. Dysfunctional FFAR1 inhibits the development of diet-induced glucose intolerance.
Collapse
Affiliation(s)
- Sibylle Sabrautzki
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and the German Mouse Clinic, 85764 Neuherberg, Germany; Research Unit Comparative Medicine, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | - Gabriele Kaiser
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany
| | - Gerhard K H Przemeck
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and the German Mouse Clinic, 85764 Neuherberg, Germany
| | - Felicia Gerst
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany
| | - Estela Lorza-Gil
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany
| | - Madhura Panse
- University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Tina Sartorius
- University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Miriam Hoene
- University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Susan Marschall
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and the German Mouse Clinic, 85764 Neuherberg, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany; University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Martin Hrabě de Angelis
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Experimental Genetics and the German Mouse Clinic, 85764 Neuherberg, Germany; Chair of Experimental Genetics, School of Life Sciences Weihenstephan, Technische Universität München, Alte Akademie 8, 85354 München, Germany
| | - Susanne Ullrich
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen (IDM), 72076 Tübingen, Germany; University Hospital Tübingen, Department of Internal Medicine, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany.
| |
Collapse
|
28
|
Feng XT, Duan HM, Li SL. Protective role of Pollen Typhae total flavone against the palmitic acid-induced impairment of glucose-stimulated insulin secretion involving GPR40 signaling in INS-1 cells. Int J Mol Med 2017; 40:922-930. [DOI: 10.3892/ijmm.2017.3070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/05/2017] [Indexed: 11/05/2022] Open
|
29
|
Kristinsson H, Sargsyan E, Manell H, Smith DM, Göpel SO, Bergsten P. Basal hypersecretion of glucagon and insulin from palmitate-exposed human islets depends on FFAR1 but not decreased somatostatin secretion. Sci Rep 2017; 7:4657. [PMID: 28680093 PMCID: PMC5498543 DOI: 10.1038/s41598-017-04730-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 06/01/2017] [Indexed: 12/19/2022] Open
Abstract
In obesity fasting levels of both glucagon and insulin are elevated. In these subjects fasting levels of the free fatty acid palmitate are raised. We have demonstrated that palmitate enhances glucose-stimulated insulin secretion from isolated human islets via free fatty acid receptor 1 (FFAR1/GPR40). Since FFAR1 is also present on glucagon-secreting alpha-cells, we hypothesized that palmitate simultaneously stimulates secretion of glucagon and insulin at fasting glucose concentrations. In addition, we hypothesized that concomitant hypersecretion of glucagon and insulin was also contributed by reduced somatostatin secretion. We found basal glucagon, insulin and somatostatin secretion and respiration from human islets, to be enhanced during palmitate treatment at normoglycemia. Secretion of all hormones and mitochondrial respiration were lowered when FFAR1 or fatty acid β-oxidation was inhibited. The findings were confirmed in the human beta-cell line EndoC-βH1. We conclude that fatty acids enhance both glucagon and insulin secretion at fasting glucose concentrations and that FFAR1 and enhanced mitochondrial metabolism but not lowered somatostatin secretion are crucial in this effect. The ability of chronically elevated palmitate levels to simultaneously increase basal secretion of glucagon and insulin positions elevated levels of fatty acids as potential triggering factors for the development of obesity and impaired glucose control.
Collapse
Affiliation(s)
- H Kristinsson
- Department of Medical Cell Biology, Uppsala University, BMC, Husargatan 3, Uppsala, Sweden.
| | - E Sargsyan
- Department of Medical Cell Biology, Uppsala University, BMC, Husargatan 3, Uppsala, Sweden
| | - H Manell
- Department of Medical Cell Biology, Uppsala University, BMC, Husargatan 3, Uppsala, Sweden
| | - D M Smith
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, UK
| | - S O Göpel
- AstraZeneca R&D Gothenburg, CVMD Bioscience, Gothenburg, Sweden
| | - P Bergsten
- Department of Medical Cell Biology, Uppsala University, BMC, Husargatan 3, Uppsala, Sweden
| |
Collapse
|
30
|
Hellström-Lindahl E, Åberg O, Ericsson C, O'Mahony G, Johnström P, Skrtic S, Eriksson O. Toward molecular imaging of the free fatty acid receptor 1. Acta Diabetol 2017; 54:663-668. [PMID: 28409274 PMCID: PMC5486603 DOI: 10.1007/s00592-017-0989-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/30/2017] [Indexed: 12/16/2022]
Abstract
AIMS Molecular imaging of the free fatty acid receptor 1 (FFAR1) would be a valuable tool for drug development by enabling in vivo target engagement studies in human. It has also been suggested as a putative target for beta cell imaging, but the inherent lipophilicity of most FFAR1 binders produces high off-target binding, which has hampered progress in this area. The aim of this study was to generate a suitable lead compound for further PET labeling. METHODS In order to identify a lead compound for future PET labeling for quantitative imaging of FFAR1 in human, we evaluated tritiated small molecule FFAR1 binding probes ([3H]AZ1, [3H]AZ2 and [3H]TAK-875) for their off-target binding, receptor density and affinity in human pancreatic tissue (islets and exocrine) and rodent insulinoma. RESULTS [3H]AZ1 showed improved specificity to FFAR1, with decreased off-target binding compared to [3H]AZ2 and [3H]TAK-875, while retaining high affinity in the nanomolar range. FFAR1 density in human islets was approximately 50% higher than in exocrine tissue. CONCLUSIONS AZ1 is a suitable lead compound for PET labeling for molecular imaging of FFAR1 in humans, due to high affinity and reduced off-target binding.
Collapse
Affiliation(s)
- Ewa Hellström-Lindahl
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, SE-751 83, Uppsala, Sweden
| | - Ola Åberg
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, SE-751 83, Uppsala, Sweden
| | | | | | - Peter Johnström
- Personalised Healthcare and Biomarkers, AstraZeneca PET Science Centre, Karolinska Institutet, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Stanko Skrtic
- AstraZeneca R&D, SE-431 50, Mölndal, Sweden
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45, Gothenburg, Sweden
| | - Olof Eriksson
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, SE-751 83, Uppsala, Sweden.
| |
Collapse
|
31
|
Satapati S, Qian Y, Wu MS, Petrov A, Dai G, Wang SP, Zhu Y, Shen X, Muise ES, Chen Y, Zycband E, Weinglass A, Di Salvo J, Debenham JS, Cox JM, Lan P, Shah V, Previs SF, Erion M, Kelley DE, Wang L, Howard AD, Shang J. GPR120 suppresses adipose tissue lipolysis and synergizes with GPR40 in antidiabetic efficacy. J Lipid Res 2017; 58:1561-1578. [PMID: 28583918 DOI: 10.1194/jlr.m075044] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/02/2017] [Indexed: 12/28/2022] Open
Abstract
GPR40 and GPR120 are fatty acid sensors that play important roles in glucose and energy homeostasis. GPR40 potentiates glucose-dependent insulin secretion and demonstrated in clinical studies robust glucose lowering in type 2 diabetes. GPR120 improves insulin sensitivity in rodents, albeit its mechanism of action is not fully understood. Here, we postulated that the antidiabetic efficacy of GPR40 could be enhanced by coactivating GPR120. A combination of GPR40 and GPR120 agonists in db/db mice, as well as a single molecule with dual agonist activities, achieved superior glycemic control compared with either monotherapy. Compared with a GPR40 selective agonist, the dual agonist improved insulin sensitivity in ob/ob mice measured by hyperinsulinemic-euglycemic clamp, preserved islet morphology, and increased expression of several key lipolytic genes in adipose tissue of Zucker diabetic fatty rats. Novel insights into the mechanism of action for GPR120 were obtained. Selective GPR120 activation suppressed lipolysis in primary white adipocytes, although this effect was attenuated in adipocytes from obese rats and obese rhesus, and sensitized the antilipolytic effect of insulin in rat and rhesus primary adipocytes. In conclusion, GPR120 agonism enhances insulin action in adipose tissue and yields a synergistic efficacy when combined with GPR40 agonism.
Collapse
Affiliation(s)
| | - Ying Qian
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Margaret S Wu
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Aleksandr Petrov
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Ge Dai
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Sheng-Ping Wang
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Yonghua Zhu
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Xiaolan Shen
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Eric S Muise
- Genetics and Pharmacogenomics, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Ying Chen
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Emanuel Zycband
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Adam Weinglass
- Genetics and Pharmacology, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Jerry Di Salvo
- Genetics and Pharmacology, Merck & Co., Inc., Kenilworth, NJ 07033
| | - John S Debenham
- Genetics and Chemistry, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Jason M Cox
- Genetics and Chemistry, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Ping Lan
- Genetics and Chemistry, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Vinit Shah
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Stephen F Previs
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Mark Erion
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - David E Kelley
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Liangsu Wang
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Andrew D Howard
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033
| | - Jin Shang
- Cardiometabolic Disease, Merck & Co., Inc., Kenilworth, NJ 07033.
| |
Collapse
|
32
|
Miller C, Pachanski MJ, Kirkland ME, Kosinski DT, Mane J, Bunzel M, Cao J, Souza S, Thomas-Fowlkes B, Di Salvo J, Weinglass AB, Li X, Myers RW, Knagge K, Carrington PE, Hagmann WK, Trujillo ME. GPR40 partial agonist MK-2305 lower fasting glucose in the Goto Kakizaki rat via suppression of endogenous glucose production. PLoS One 2017; 12:e0176182. [PMID: 28542610 PMCID: PMC5441580 DOI: 10.1371/journal.pone.0176182] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 04/06/2017] [Indexed: 11/19/2022] Open
Abstract
GPR40 (FFA1) is a fatty acid receptor whose activation results in potent glucose lowering and insulinotropic effects in vivo. Several reports illustrate that GPR40 agonists exert glucose lowering in diabetic humans. To assess the mechanisms by which GPR40 partial agonists improve glucose homeostasis, we evaluated the effects of MK-2305, a potent and selective partial GPR40 agonist, in diabetic Goto Kakizaki rats. MK-2305 decreased fasting glucose after acute and chronic treatment. MK-2305-mediated changes in glucose were coupled with increases in plasma insulin during hyperglycemia and glucose challenges but not during fasting, when glucose was normalized. To determine the mechanism(s) mediating these changes in glucose metabolism, we measured the absolute contribution of precursors to glucose production in the presence or absence of MK-2305. MK-2305 treatment resulted in decreased endogenous glucose production (EGP) driven primarily through changes in gluconeogenesis from substrates entering at the TCA cycle. The decrease in EGP was not likely due to a direct effect on the liver, as isolated perfused liver studies showed no effect of MK-2305 ex vivo and GPR40 is not expressed in the liver. Taken together, our results suggest MK-2305 treatment increases glucose stimulated insulin secretion (GSIS), resulting in changes to hepatic substrate handling that improve glucose homeostasis in the diabetic state. Importantly, these data extend our understanding of the underlying mechanisms by which GPR40 partial agonists reduce hyperglycemia.
Collapse
Affiliation(s)
- Corin Miller
- Departments of Translational Imaging Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Michele J. Pachanski
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Melissa E. Kirkland
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Daniel T. Kosinski
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Joel Mane
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Michelle Bunzel
- Departments of Translational Imaging Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Jin Cao
- Departments of Translational Imaging Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Sarah Souza
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Brande Thomas-Fowlkes
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Jerry Di Salvo
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Adam B. Weinglass
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Xiaoyan Li
- Cardio-Metabolic Diseases, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Robert W. Myers
- In Vitro Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Kevin Knagge
- David H Murdock Research Institute, Kannapolis, North Carolina, United States of America
| | - Paul E. Carrington
- Cardio-Metabolic Diseases, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - William K. Hagmann
- Chemistry, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Maria E. Trujillo
- In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
33
|
Kumar A, Bharti SK, Kumar A. Therapeutic molecules against type 2 diabetes: What we have and what are we expecting? Pharmacol Rep 2017; 69:959-970. [PMID: 28822958 DOI: 10.1016/j.pharep.2017.04.003] [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: 01/05/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/29/2022]
Abstract
World Health Organization (WHO) has identified diabetes as one of the fastest growing non-communicable diseases with 422 million patients around the world in 2014. Diabetes, a metabolic disease, is characterized primarily by hyperglycemia which results in various macrovascular and microvascular complications like cardiovascular disease and neuropathies which can significantly deteriorate the quality of life. The body either does not manufactures enough insulin (type 1 diabetes or T1DM) or becomes insensitive to physiologically secreted insulin or both (type 2 diabetes or T2DM). The majority of the diabetic population is affected by type 2 diabetes. Currently, hyperglycemia is treated by a broad range of molecules such as biguanides, sulfonylurea, insulin, thiazolidinediones, incretin mimetics, and DPP-4 inhibitors exerting different mechanisms. However, new drug classes have indeed come in the market such as SGLT-2 inhibitors and other are in the experimental stages such as GPR 40 agonists, GSK-3 inhibitors, GK activators and GPR21 inhibitors which definitely could be anticipated as safe and effective for diabetes therapy. This article reviews the general approach to currently approved therapies for type 2 diabetes and focusing on novel approaches that could be a panacea and might be useful in the future for diabetes patients.
Collapse
Affiliation(s)
- Ashwini Kumar
- Department of Biotechnology, National Institute of Technology Raipur, Raipur, Chhattisgarh, India
| | | | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology Raipur, Raipur, Chhattisgarh, India.
| |
Collapse
|
34
|
Delphinidin Reduces Glucose Uptake in Mice Jejunal Tissue and Human Intestinal Cells Lines through FFA1/GPR40. Int J Mol Sci 2017; 18:ijms18040750. [PMID: 28379159 PMCID: PMC5412335 DOI: 10.3390/ijms18040750] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/21/2017] [Accepted: 03/27/2017] [Indexed: 11/23/2022] Open
Abstract
Anthocyanins are pigments with antihyperglycemic properties, and they are potential candidates for developing functional foods for the therapy or prevention of Diabetes mellitus type 2 (DM2). The mechanism of these beneficial effects of anthocyanins are, however, hard to explain, given their very low bioavailability due to poor intestinal absorption. We propose that free fatty acid receptor 1 (FFA1, also named GPR40), is involved in an inhibitory effect of the anthocyanidin delphinidin over intestinal glucose absorption. We show the direct effects of delphinidin on the intestine using jejunum samples from RF/J mice, and the human intestinal cell lines HT-29, Caco-2, and NCM460. By the use of specific pharmacological antagonists, we determined that delphinidin inhibits glucose absorption in both mouse jejunum and a human enterocytic cell line in a FFA1-dependent manner. Delphinidin also affects the function of sodium-glucose cotransporter 1 (SGLT1). Intracellular signaling after FFA1 activation involved cAMP increase and cytosolic Ca2+ oscillations originated from intracellular Ca2+ stores and were followed by store-operated Ca2+ entry. Taken together, our results suggest a new GPR-40 mediated local mechanism of action for delphinidin over intestinal cells that may in part explain its antidiabetic effect. These findings are promising for the search for new prevention and pharmacological treatment strategies for DM2 management.
Collapse
|
35
|
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
|
36
|
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
|
37
|
Li AJ, Wiater MF, Wang Q, Wank S, Ritter S. Deletion of GPR40 fatty acid receptor gene in mice blocks mercaptoacetate-induced feeding. Am J Physiol Regul Integr Comp Physiol 2016; 310:R968-74. [PMID: 26984894 DOI: 10.1152/ajpregu.00548.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/10/2016] [Indexed: 12/12/2022]
Abstract
Both increased and decreased fatty acid (FA) availability contribute to control of food intake. For example, it is well documented that intestinal FA reduces feeding by triggering enterondocrine secretion of satietogenic peptides, such as cholecystokinin (CCK) and glucagon-like peptide-1 (GLP-1). In contrast, mechanisms by which decreased FA availability increase feeding are not well understood. Over the past three decades substantial research related to FA availability and increased feeding has involved use of the orexigenic compound mercaptoacetate (MA). Because MA reportedly inhibits FA oxidation, it has been assumed that reduced FA oxidation accounts for the orexigenic action of MA. Recently, however, we demonstrated that MA antagonizes G protein-coupled receptor 40 (GPR40), a membrane receptor for long and medium chain FA. We also demonstrated that, by antagonizing GPR40, MA inhibits GLP-1 secretion and attenuates vagal afferent activation by FA. Because both vagal afferent activation and GLP-1 inhibit food intake, we postulated that inhibition of GPR40 by MA might underlie the orexigenic action of MA. We tested this hypothesis using male and female GPR40 knockout (KO) and wild-type (WT) mice. Using several testing protocols, we found that MA increased feeding in WT, but not GPR40 KO mice, and that GPR40 KO mice gained more weight than WT on a high-fat diet. Metabolic monitoring after MA or saline injection in the absence of food did not reveal significant differences in respiratory quotient or energy expenditure between treatment groups or genotypes. These results support the hypothesis that MA stimulates food intake by blocking FA effects on GPR40.
Collapse
Affiliation(s)
- Ai-Jun Li
- Programs in Neuroscience, Washington State University, Pullman, Washington; and
| | - Michael F Wiater
- Programs in Neuroscience, Washington State University, Pullman, Washington; and
| | - Qing Wang
- Programs in Neuroscience, Washington State University, Pullman, Washington; and
| | - Stephen Wank
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Sue Ritter
- Programs in Neuroscience, Washington State University, Pullman, Washington; and
| |
Collapse
|
38
|
Mayer M, Nudurupati S, Peng X, Marcinak J. Evaluation of the pharmacokinetics and safety of a single oral dose of fasiglifam in subjects with normal or varying degrees of impaired renal function. Drugs R D 2016; 14:273-82. [PMID: 25374042 PMCID: PMC4269813 DOI: 10.1007/s40268-014-0066-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
INTRODUCTION Approximately one-third of patients with type 2 diabetes mellitus (T2DM) have concurrent renal impairment. There are limited therapeutic options for these patients. Fasiglifam is a G protein-coupled receptor 40 agonist that was under investigation for the treatment of T2DM. The objective of this study was to evaluate the potential effect of renal impairment on the pharmacokinetics and safety of a single dose of fasiglifam and its metabolite M-1. METHODS This was a phase I, open-label, parallel-group study. Subjects with varying degrees of renal function received a single oral dose of fasiglifam 50 mg. Blood and urine samples were collected through 168 h postdose. Study endpoints were pharmacokinetic and safety variables. RESULTS Fifty-three subjects were enrolled. Mean fasiglifam plasma concentrations were higher in subjects with mild renal impairment compared with other groups, but within each renal function cohort, plasma concentrations tended to decrease with decreasing renal function. Regression analyses indicated that fasiglifam exposure decreased and M-1 exposure increased with decreasing renal function. Predicted exposure values at about the midpoint of creatinine clearance for each renal impairment group differed by up to 21% (fasiglifam) and 87% (M-1) from that of the normal renal function group. Hemodialysis had no effect on fasiglifam or M-1 exposure. Fasiglifam renal clearance (CLR) was not affected, but M-1 CLR decreased with increasing impairment. No incidences of hypoglycemia were reported during the study. CONCLUSION Varying renal function status did not have a significant impact on the clearance of fasiglifam in this study.
Collapse
Affiliation(s)
- Michael Mayer
- Takeda Development Center Americas, Inc., One Takeda Parkway, Deerfield, IL, USA,
| | | | | | | |
Collapse
|
39
|
Oh DY, Olefsky JM. G protein-coupled receptors as targets for anti-diabetic therapeutics. Nat Rev Drug Discov 2016; 15:161-72. [DOI: 10.1038/nrd.2015.4] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
40
|
Li AJ, Wang Q, Dinh TT, Simasko SM, Ritter S. Mercaptoacetate blocks fatty acid-induced GLP-1 secretion in male rats by directly antagonizing GPR40 fatty acid receptors. Am J Physiol Regul Integr Comp Physiol 2016; 310:R724-32. [PMID: 26791830 DOI: 10.1152/ajpregu.00387.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/19/2016] [Indexed: 12/13/2022]
Abstract
Mercaptoacetate (MA) is an orexigenic agent reported to block fatty acid (FA) oxidation. Recently, however, we reported evidence from isolated nodose ganglion neurons that MA antagonizes the G protein-coupled long- and medium-chain FA receptor GPR40. GPR40 mediates FA-induced secretion of the satietogenic incretin peptide glucagon-like peptide 1 (GLP-1), by enteroendocrine L cells, as well as FA-induced enhancement of glucose-stimulated insulin secretion. Our results in cultured nodose neurons suggest that MA would also block GPR40 in enteroendocrine cells controlling GLP-1 secretion. If so, this would suggest an alternative mechanism by which MA increases food intake. We tested the hypothesis that MA blocks FA-induced GLP-1 secretion in vitro using cultured STC-1 cells (a murine enteroendocrine cell line) and in vivo in adult male rats. In vitro, MA blocked the increase in both cytosolic Ca(2+)and GLP-1 release stimulated by FAs and also reduced (but less effectively) the response of STC-1 cells to grifolic acid, a partial agonist of the GPR120 FA receptor. In vivo, MA reduced GLP-1 secretion following olive oil gavage while also increasing glucose and decreasing insulin levels. The carnitine palmatoyltransferase 1 antagonist etomoxir did not alter these responses. Results indicate that MA's actions, including its orexigenic effect, are mediated by GPR40 (and possibly GPR120) receptor antagonism and not by blockade of fat oxidation, as previously believed. Analysis of MA's interaction with GPR40 may facilitate understanding of the multiple functions of this receptor and the manner in which FAs participate in the control of hunger and satiety.
Collapse
Affiliation(s)
- Ai-Jun Li
- Programs in Neuroscience, Washington State University, Pullman, Washington
| | - Qing Wang
- Programs in Neuroscience, Washington State University, Pullman, Washington
| | - Thu T Dinh
- Programs in Neuroscience, Washington State University, Pullman, Washington
| | - Steve M Simasko
- Programs in Neuroscience, Washington State University, Pullman, Washington
| | - Sue Ritter
- Programs in Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
41
|
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
|
42
|
Kristinsson H, Bergsten P, Sargsyan E. Free fatty acid receptor 1 (FFAR1/GPR40) signaling affects insulin secretion by enhancing mitochondrial respiration during palmitate exposure. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3248-57. [DOI: 10.1016/j.bbamcr.2015.09.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 09/04/2015] [Accepted: 09/21/2015] [Indexed: 12/23/2022]
|
43
|
Tang XL, Wang CN, Zhu XY, Ni X. Rosiglitazone inhibition of calvaria-derived osteoblast differentiation is through both of PPARγ and GPR40 and GSK3β-dependent pathway. Mol Cell Endocrinol 2015; 413:78-89. [PMID: 26116229 DOI: 10.1016/j.mce.2015.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 05/20/2015] [Accepted: 06/10/2015] [Indexed: 01/17/2023]
Abstract
Rosiglitazone (RSG) can cause bone loss, however the mechanisms remain largely unknown. This study aims to investigate the effects of RSG on differentiation and mineralization of osteoblasts using primary cultured mouse fetal calvaria-derived osteoblasts as a model, and elucidate the receptor and signaling pathways responsible for these effects. We found that RSG suppressed the differentiation and mineralization of calvaria-derived osteoblasts. Peroxisome proliferators-activated receptor γ (PPARγ) siRNA significantly reversed the inhibitory effect of RSG on osteogenic differentiation. The expression of G protein-coupled receptor (GPR) 40 was suppressed during differentiation, but was increased by RSG treatment. GPR40 siRNA significantly reversed the inhibitory effect of RSG on osteogenesis. RSG activated glycogen synthase kinase (GSK)-3β, which in turn decreased β-catenin expression. RSG-induced GSK3β activation was mediated through both PPARγ and GPR40. These results suggest that both PPARγ and GRP40 are required for RSG-induced inhibition of mouse calvaria osteoblast differentiation, which is mediated through GSK3β-dependent pathway.
Collapse
Affiliation(s)
- Xiao-Lu Tang
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Second Military Medical University, Shanghai 200433, China
| | - Chang-Nan Wang
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Second Military Medical University, Shanghai 200433, China
| | - Xiao-Yan Zhu
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Second Military Medical University, Shanghai 200433, China.
| | - Xin Ni
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
44
|
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
|
45
|
Mancini AD, Poitout V. GPR40 agonists for the treatment of type 2 diabetes: life after 'TAKing' a hit. Diabetes Obes Metab 2015; 17:622-9. [PMID: 25604916 DOI: 10.1111/dom.12442] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/13/2015] [Accepted: 01/17/2015] [Indexed: 12/17/2022]
Abstract
The free fatty acid receptor GPR40 has been proposed as a potential target for type 2 diabetes (T2D) pharmacotherapy. This idea has been validated in both preclinical and clinical studies, in which activation of GPR40 was shown to improve glycaemic control by stimulating glucose-dependent insulin secretion; however, the recent termination of phase III clinical trials using the GPR40 agonist TAK-875 (fasiglifam) has raised important questions regarding the long-term safety and viability of targeting GPR40 and, more specifically, about our understanding of this receptor's basic biology. In the present review, we provide a summary of established and novel concepts related to GPR40's pharmacobiology and discuss the current status and future outlook for GPR40-based drug development for the treatment of T2D.
Collapse
Affiliation(s)
- A D Mancini
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - V Poitout
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
46
|
Woolcott OO, Richey JM, Kabir M, Chow RH, Iyer MS, Kirkman EL, Stefanovski D, Lottati M, Kim SP, Harrison LN, Ionut V, Zheng D, Hsu IR, Catalano KJ, Chiu JD, Bradshaw H, Wu Q, Bergman RN. High-fat diet-induced insulin resistance does not increase plasma anandamide levels or potentiate anandamide insulinotropic effect in isolated canine islets. PLoS One 2015; 10:e0123558. [PMID: 25855974 PMCID: PMC4391925 DOI: 10.1371/journal.pone.0123558] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/05/2015] [Indexed: 01/09/2023] Open
Abstract
Background Obesity has been associated with elevated plasma anandamide levels. In addition, anandamide has been shown to stimulate insulin secretion in vitro, suggesting that anandamide might be linked to hyperinsulinemia. Objective To determine whether high-fat diet-induced insulin resistance increases anandamide levels and potentiates the insulinotropic effect of anandamide in isolated pancreatic islets. Design and Methods Dogs were fed a high-fat diet (n = 9) for 22 weeks. Abdominal fat depot was quantified by MRI. Insulin sensitivity was assessed by the euglycemic-hyperinsulinemic clamp. Fasting plasma endocannabinoid levels were analyzed by liquid chromatography-mass spectrometry. All metabolic assessments were performed before and after fat diet regimen. At the end of the study, pancreatic islets were isolated prior to euthanasia to test the in vitro effect of anandamide on islet hormones. mRNA expression of cannabinoid receptors was determined in intact islets. The findings in vitro were compared with those from animals fed a control diet (n = 7). Results Prolonged fat feeding increased abdominal fat content by 81.3±21.6% (mean±S.E.M, P<0.01). In vivo insulin sensitivity decreased by 31.3±12.1% (P<0.05), concomitant with a decrease in plasma 2-arachidonoyl glycerol (from 39.1±5.2 to 15.7±2.0 nmol/L) but not anandamide, oleoyl ethanolamide, linoleoyl ethanolamide, or palmitoyl ethanolamide. In control-diet animals (body weight: 28.8±1.0 kg), islets incubated with anandamide had a higher basal and glucose-stimulated insulin secretion as compared with no treatment. Islets from fat-fed animals (34.5±1.3 kg; P<0.05 versus control) did not exhibit further potentiation of anandamide-induced insulin secretion as compared with control-diet animals. Glucagon but not somatostatin secretion in vitro was also increased in response to anandamide, but there was no difference between groups (P = 0.705). No differences in gene expression of CB1R or CB2R between groups were found. Conclusions In canines, high-fat diet-induced insulin resistance does not alter plasma anandamide levels or further potentiate the insulinotropic effect of anandamide in vitro.
Collapse
Affiliation(s)
- Orison O. Woolcott
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| | - Joyce M. Richey
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Morvarid Kabir
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Robert H. Chow
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Malini S. Iyer
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Erlinda L. Kirkman
- Department of Animal Resources, University of Southern California, Los Angeles, California, United States of America
| | - Darko Stefanovski
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Maya Lottati
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Stella P. Kim
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - L. Nicole Harrison
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Viorica Ionut
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Dan Zheng
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Isabel R. Hsu
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Karyn J. Catalano
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Jenny D. Chiu
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Heather Bradshaw
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, United States of America
| | - Qiang Wu
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| |
Collapse
|
47
|
Hirozane Y, Motoyaji T, Maru T, Okada K, Tarui N. Generating thermostabilized agonist-bound GPR40/FFAR1 using virus-like particles and a label-free binding assay. Mol Membr Biol 2015; 31:168-75. [PMID: 25068810 DOI: 10.3109/09687688.2014.923588] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Elucidating the detailed mechanism of activation of membrane protein receptors and their ligand binding is essential for structure-based drug design. Membrane protein crystal structure analysis successfully aids in understanding these fundamental molecular interactions. However, protein crystal structure analysis of the G-protein-coupled receptor (GPCR) remains challenging, even for the class of GPCRs which have been included in the majority of structure analysis reports among membrane proteins, due to the substantial instability of these receptors when extracted from lipid bilayer membranes. It is known that increased thermostability tends to decrease conformational flexibility, which contributes to the generation of diffraction quality crystals. However, this is still not straightforward, and significant effort is required to identify thermostabilized mutants that are optimal for crystallography. To address this issue, a versatile screening platform based on a label-free ligand binding assay combined with transient overexpression in virus-like particles was developed. This platform was used to generate thermostabilized GPR40 [also known as free fatty acid receptor 1 (FFAR1)] for fasiglifam (TAK-875). This demonstrated that the thermostabilized mutant GPR40 (L42A/F88A/G103A/Y202F) was successfully used for crystal structure analysis.
Collapse
Affiliation(s)
- Yoshihiko Hirozane
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Ltd , Fujisawa, Kanagawa , Japan
| | | | | | | | | |
Collapse
|
48
|
Kim IS, Yang SY, Han JH, Jung SH, Park HS, Myung CS. Differential Gene Expression in GPR40-Overexpressing Pancreatic β-cells Treated with Linoleic Acid. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:141-9. [PMID: 25729276 PMCID: PMC4342734 DOI: 10.4196/kjpp.2015.19.2.141] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/05/2015] [Accepted: 01/07/2015] [Indexed: 01/20/2023]
Abstract
"G protein-coupled receptor 40" (GPR40), a receptor for long-chain fatty acids, mediates the stimulation of glucose-induced insulin secretion. We examined the profiles of differential gene expression in GPR40-activated cells treated with linoleic acid, and finally predicted the integral pathways of the cellular mechanism of GPR40-mediated insulinotropic effects. After constructing a GPR40-overexpressing stable cell line (RIN-40) from the rat pancreatic β-cell line RIN-5f, we determined the gene expression profiles of RIN-5f and RIN-40. In total, 1004 genes, the expression of which was altered at least twofold, were selected in RIN-5f versus RIN-40. Moreover, the differential genetic profiles were investigated in RIN-40 cells treated with 30 µM linoleic acid, which resulted in selection of 93 genes in RIN-40 versus RIN-40 treated with linoleic acid. Based on the Kyoto Encyclopedia of Genes and Genomes Pathway (KEGG, http://www.genome.jp/kegg/), sets of genes induced differentially by treatment with linoleic acid in RIN-40 cells were found to be related to mitogen-activated protein (MAP) kinase- and neuroactive ligand-receptor interaction pathways. A gene ontology (GO) study revealed that more than 30% of the genes were associated with signal transduction and cell proliferation. Thus, this study elucidated a gene expression pattern relevant to the signal pathways that are regulated by GPR40 activation during the acute period. Together, these findings increase our mechanistic understanding of endogenous molecules associated with GPR40 function, and provide information useful for identification of a target for the management of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- In-Su Kim
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - So-Young Yang
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea. ; Institute of Drug Research & Development, Chungnam National University, Daejeon 305-764, Korea
| | - Joo-Hui Han
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Sang-Hyuk Jung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Hyun-Soo Park
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea
| | - Chang-Seon Myung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon 305-764, Korea. ; Institute of Drug Research & Development, Chungnam National University, Daejeon 305-764, Korea
| |
Collapse
|
49
|
Kaiser D, Oetjen E. Something old, something new and something very old: drugs for treating type 2 diabetes. Br J Pharmacol 2015; 171:2940-50. [PMID: 24641580 DOI: 10.1111/bph.12624] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/13/2014] [Accepted: 01/30/2014] [Indexed: 12/28/2022] Open
Abstract
Diabetes mellitus belongs to the most rapidly increasing diseases worldwide. Approximately 90-95% of these patients suffer from type 2 diabetes mellitus, which is characterized by peripheral insulin resistance and the progressive loss of beta-cell function and mass. Considering the complications of this chronic disease, a reliable anti-diabetic treatment is indispensable. An ideal oral anti-diabetic drug should not only correct glucose homeostasis but also preserve or even augment beta-cell function and mass, ameliorate the subclinical inflammation present under insulin-resistant conditions and prevent the macro- and microvascular consequences of diabetes in order to reduce the mortality. Despite the many anti-diabetic drugs already in use, there is an ongoing research for additional drugs, guided by different concepts of the pathogenesis of type 2 diabetes. This review will briefly summarize current oral anti-diabetic drugs. In addition, emerging strategies for the treatment of diabetes will be described, among them the inhibition of glucagon action and anti-inflammatory drugs. Their suitability as 'ideal anti-diabetic drugs' will be discussed.
Collapse
Affiliation(s)
- D Kaiser
- Department of Clinical Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
50
|
Abstract
The goal for the treatment of patients with diabetes has today shifted from merely reducing glucose concentrations to preventing the natural decline in β-cell function and delay the progression of disease. Pancreatic β-cell dysfunction and decreased β-cell mass are crucial in the development of diabetes. The β-cell defects are the main pathogenesis in patients with type 1 diabetes and are associated with type 2 diabetes as the disease progresses. Recent studies suggest that human pancreatic β-cells have a capacity for increased proliferation according to increased demands for insulin. In humans, β-cell mass has been shown to increase in patients showing insulin-resistance states such as obesity or in pregnancy. This capacity might be useful for identifying new therapeutic strategies to reestablish a functional β-cell mass. In this context, therapeutic approaches designed to increase β-cell mass might prove a significant way to manage diabetes and prevent its progression. This review describes the various β-cell defects that appear in patients with diabetes and outline the mechanisms of β-cell failure. We also review common methods for assessing β-cell function and mass and methodological limitations in vivo. Finally, we discuss the current therapeutic approaches to improve β-cell function and increase β-cell mass.
Collapse
Affiliation(s)
- Kyong Yeun Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Kyoung Min Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|