1
|
Yoon J, Lee DG, Song H, Hong D, Park JS, Hong C, An KM, Lee JW, Park JT, Yoon H, Tak J, Kim SG. Xelaglifam, a novel GPR40/FFAR1 agonist, exhibits enhanced β-arrestin recruitment and sustained glycemic control for type 2 diabetes. Biomed Pharmacother 2024; 177:117044. [PMID: 38941892 DOI: 10.1016/j.biopha.2024.117044] [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: 04/23/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024] Open
Abstract
Xelaglifam, developed as a GPR40/FFAR1 agonist, induces glucose-dependent insulin secretion and reduces circulating glucose levels for Type 2 diabetes treatment. This study investigated the effects of Xelaglifam in comparison with Fasiglifam on the in vitro/in vivo anti-diabetic efficacy and selectivity, and the mechanistic basis. In vitro studies on downstream targets of Xelaglifam were performed in GPR40-expressing cells. Xelaglifam treatment exhibited dose-dependent effects, increasing inositol phosphate-1, Ca2+ mobilization, and β-arrestin recruitment (EC50: 0.76 nM, 20 nM, 68 nM), supporting its role in Gq protein-dependent and G-protein-independent mechanisms. Despite a lack of change in the cAMP pathway, the Xelaglifam-treated group demonstrated increased insulin secretion compared to Fasiglifam in HIT-T15 β cells under high glucose conditions. High doses of Xelaglifam (<30 mg/kg) did not induce hypoglycemia in Sprague-Dawley rats. In addition, Xelaglifam lowered glucose and increased insulin levels in diabetic rat models (GK, ZDF, OLETF). In GK rats, 1 mg/kg of Xelaglifam improved glucose tolerance (33.4 % and 15.6 % for the 1 and 5 h) after consecutive glucose challenges. Moreover, repeated dosing in ZDF and OLETF rats resulted in superior glucose tolerance (34 % and 35.1 % in ZDF and OLETF), reducing fasting hyperglycemia (18.3 % and 30 % in ZDF and OLETF) at lower doses; Xelaglifam demonstrated a longer-lasting effect with a greater effect on β-cells including 3.8-fold enhanced insulin secretion. Co-treatment of Xelaglifam with SGLT-2 inhibitors showed additive or synergistic effects. Collectively, these results demonstrate the therapeutic efficacy and selectivity of Xelaglifam on GPR40, supportive of its potential for the treatment of Type 2 diabetes.
Collapse
Affiliation(s)
- Jongmin Yoon
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea; College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Don-Gil Lee
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Haengjin Song
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Dahae Hong
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Ji Soo Park
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Changhee Hong
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Kyung Mi An
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Jung Woo Lee
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Joon-Tae Park
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Hongchul Yoon
- YUNOVIA Co., Ltd., 20, Samsung 1-ro 1-gil, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do 10326, Republic of Korea.
| |
Collapse
|
2
|
Wargent ET, Kępczyńska MA, Kaspersen MH, Ulven ER, Arch JRS, Ulven T, Stocker CJ. Chronic administration of hydrolysed pine nut oil to mice improves insulin sensitivity and glucose tolerance and increases energy expenditure via a free fatty acid receptor 4-dependent mechanism. Br J Nutr 2024; 132:13-20. [PMID: 38751244 DOI: 10.1017/s0007114524000965] [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] [Indexed: 05/28/2024]
Abstract
A healthy diet is at the forefront of measures to prevent type 2 diabetes. Certain vegetable and fish oils, such as pine nut oil (PNO), have been demonstrated to ameliorate the adverse metabolic effects of a high-fat diet. The present study investigates the involvement of the free fatty acid receptors 1 (FFAR1) and 4 (FFAR4) in the chronic activity of hydrolysed PNO (hPNO) on high-fat diet-induced obesity and insulin resistance. Male C57BL/6J wild-type, FFAR1 knockout (-/-) and FFAR4-/- mice were placed on 60 % high-fat diet for 3 months. Mice were then dosed hPNO for 24 d, during which time body composition, energy intake and expenditure, glucose tolerance and fasting plasma insulin, leptin and adiponectin were measured. hPNO improved glucose tolerance and decreased plasma insulin in the wild-type and FFAR1-/- mice, but not the FFAR4-/- mice. hPNO also decreased high-fat diet-induced body weight gain and fat mass, whilst increasing energy expenditure and plasma adiponectin. None of these effects on energy balance were statistically significant in FFAR4-/- mice, but it was not shown that they were significantly less than in wild-type mice. In conclusion, chronic hPNO supplementation reduces the metabolically detrimental effects of high-fat diet on obesity and insulin resistance in a manner that is dependent on the presence of FFAR4.
Collapse
Affiliation(s)
- Edward Taynton Wargent
- Institute of Translational Medicine, Clore Laboratory, University of Buckingham, Buckingham, MK18 1EG, UK
| | - Małgorzata A Kępczyńska
- Institute of Translational Medicine, Clore Laboratory, University of Buckingham, Buckingham, MK18 1EG, UK
| | - Mads H Kaspersen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense, Denmark
| | - Elisabeth Rexen Ulven
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100Copenhagen, Denmark
| | - Jonathan R S Arch
- Institute of Translational Medicine, Clore Laboratory, University of Buckingham, Buckingham, MK18 1EG, UK
| | - Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100Copenhagen, Denmark
| | | |
Collapse
|
3
|
Ahmed R, de Souza RJ, Li V, Banfield L, Anand SS. Twenty years of participation of racialised groups in type 2 diabetes randomised clinical trials: a meta-epidemiological review. Diabetologia 2024; 67:443-458. [PMID: 38177564 PMCID: PMC10844363 DOI: 10.1007/s00125-023-06052-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/16/2023] [Indexed: 01/06/2024]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes mellitus prevalence is increasing globally and the greatest burden is borne by racialised people. However, there are concerns that the enrolment of racialised people into RCTs is limited, resulting in a lack of ethnic and racial diversity. This may differ depending whether an RCT is government funded or industry funded. The aim of this study was to review the proportions of racialised and white participants included in large RCTs of type 2 diabetes pharmacotherapies relative to the disease burden of type 2 diabetes in these groups. METHODS The Ovid MEDLINE database was searched from 1 January 2000 to 31 December 2020. English language reports of RCTs of type 2 diabetes pharmacotherapies published in select medical journals were included. Studies were included in this review if they had a sample size of at least 100 participants and all participants were adults with type 2 diabetes. Industry-funded trials must have recruited participants from at least two countries. Government-funded trials were not held to the same standard because they are typically conducted in a single country. Data including the numbers and proportions of participants by ethnicity and race were extracted from trial reports. The participation-to-prevalence ratio (PPR) was calculated for each trial by dividing the percentage of white and racialised participants in each trial by the percentage of white and racialised participants with type 2 diabetes, respectively, for the regions of recruitment. A random-effects meta-analysis was used to generate the pooled PPRs and 95% CIs across study types. A PPR <0.80 indicates under-representation and a PPR >1.20 indicates over-representation. Risk of bias assessments were not conducted for this study as the objective was to examine recruitment of racialised and white participants rather than evaluate the trustworthiness of clinical trial outcomes. RESULTS A total of 83 trials were included, involving 283,122 participants, of which 15 were government-funded and 68 were industry-funded trials. In government-funded trials, the PPR for white participants was 1.11 (95% CI 0.99, 1.24) and the PPR for racialised participants was 0.72 (95% CI 0.60, 0.86). In industry-funded trials, the PPR for white participants was 1.95 (95% CI 1.74, 2.18) and the PPR for racialised participants was 0.36 (95% CI 0.32, 0.42). The limitations of this study include the reliance on investigator-reported ethnicity and race to classify participants as 'white' or 'racialised', the use of estimates for type 2 diabetes prevalence and demographic data, and the high levels of heterogeneity of pooled estimates. However, despite these limitations, the results were consistent with respect to direction. CONCLUSIONS/INTERPRETATION Racialised participants are under-represented in government- and industry-funded type 2 diabetes trials. Strategies to improve recruitment and enrolment of racialised participants into RCTs should be developed. REGISTRATION Open Science Framework registration no. f59mk ( https://osf.io/f59mk ) FUNDING: The authors received no financial support for this research or authorship of the article.
Collapse
Affiliation(s)
- Rabeeyah Ahmed
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Chanchlani Research Centre, McMaster University, Hamilton, ON, Canada
| | - Russell J de Souza
- Chanchlani Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Vincent Li
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Laura Banfield
- Health Sciences Library, McMaster University, Hamilton, ON, Canada
| | - Sonia S Anand
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.
- Chanchlani Research Centre, McMaster University, Hamilton, ON, Canada.
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
4
|
El-Seedi HR, Salama S, El-Wahed AAA, Guo Z, Di Minno A, Daglia M, Li C, Guan X, Buccato DG, Khalifa SAM, Wang K. Exploring the Therapeutic Potential of Royal Jelly in Metabolic Disorders and Gastrointestinal Diseases. Nutrients 2024; 16:393. [PMID: 38337678 PMCID: PMC10856930 DOI: 10.3390/nu16030393] [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: 11/30/2023] [Revised: 01/07/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Metabolic disorders, encompassing diabetes mellitus, cardiovascular diseases, gastrointestinal disorders, etc., pose a substantial global health threat, with rising morbidity and mortality rates. Addressing these disorders is crucial, as conventional drugs often come with high costs and adverse effects. This review explores the potential of royal jelly (RJ), a natural bee product rich in bioactive components, as an alternative strategy for managing metabolic diseases. RJ exhibits diverse therapeutic properties, including antimicrobial, estrogen-like, anti-inflammatory, hypotensive, anticancer, and antioxidant effects. This review's focus is on investigating how RJ and its components impact conditions like diabetes mellitus, cardiovascular disease, and gastrointestinal illnesses. Evidence suggests that RJ serves as a complementary treatment for various health issues, notably demonstrating cholesterol- and glucose-lowering effects in diabetic rats. Specific RJ-derived metabolites, such as 10-hydroxy-2-decenoic acid (10-HDA), also known as the "Queen bee acid," show promise in reducing insulin resistance and hyperglycemia. Recent research highlights RJ's role in modulating immune responses, enhancing anti-inflammatory cytokines, and suppressing key inflammatory mediators. Despite these promising findings, further research is needed to comprehensively understand the mechanisms underlying RJ's therapeutic effects.
Collapse
Affiliation(s)
- Hesham R. El-Seedi
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, Uppsala University, Biomedical Centre, P.O. Box 591, SE-751 24 Uppsala, Sweden
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China;
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang 210024, China
| | - Suzy Salama
- Indigenous Knowledge and Heritage Center, Ghibaish College of Science and Technology, Ghibaish 51111, Sudan;
| | - Aida A. Abd El-Wahed
- Department of Bee Research, Plant Protection Research Institute, Agricultural Research Centre, Giza 12627, Egypt;
| | - Zhiming Guo
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China;
| | - Alessandro Di Minno
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy; (A.D.M.); (M.D.); (D.G.B.)
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy; (A.D.M.); (M.D.); (D.G.B.)
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Chuan Li
- School of Food Science and Engineering, Hainan University, Haikou 570228, China;
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Daniele Giuseppe Buccato
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy; (A.D.M.); (M.D.); (D.G.B.)
| | - Shaden A. M. Khalifa
- Psychiatry and Neurology Department, Capio Saint Göran’s Hospital, Sankt Göransplan 1, 112 19 Stockholm, Sweden
| | - Kai Wang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| |
Collapse
|
5
|
Guan HP, Xiong Y. Learn from failures and stay hopeful to GPR40, a GPCR target with robust efficacy, for therapy of metabolic disorders. Front Pharmacol 2022; 13:1043828. [PMID: 36386134 PMCID: PMC9640913 DOI: 10.3389/fphar.2022.1043828] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/13/2022] [Indexed: 09/10/2023] Open
Abstract
GPR40 is a class A G-protein coupled receptor (GPCR) mainly expressed in pancreas, intestine, and brain. Its endogenous ligand is long-chain fatty acids, which activate GPR40 after meal ingestion to induce secretion of incretins in the gut, including GLP-1, GIP, and PYY, the latter control appetite and glucose metabolism. For its involvement in satiety regulation and metabolic homeostasis, partial and AgoPAM (Positive Allosteric Modulation agonist) GPR40 agonists had been developed for type 2 diabetes (T2D) by many pharmaceutical companies. The proof-of-concept of GPR40 for control of hyperglycemia was achieved by clinical trials of partial GPR40 agonist, TAK-875, demonstrating a robust decrease in HbA1c (-1.12%) after chronic treatment in T2D. The development of TAK-875, however, was terminated due to liver toxicity in 2.7% patients with more than 3-fold increase of ALT in phase II and III clinical trials. Different mechanisms had since been proposed to explain the drug-induced liver injury, including acyl glucuronidation, inhibition of mitochondrial respiration and hepatobiliary transporters, ROS generation, etc. In addition, activation of GPR40 by AgoPAM agonists in pancreas was also linked to β-cell damage in rats. Notwithstanding the multiple safety concerns on the development of small-molecule GPR40 agonists for T2D, some partial and AgoPAM GPR40 agonists are still under clinical development. Here we review the most recent progress of GPR40 agonists development and the possible mechanisms of the side effects in different organs, and discuss the possibility of developing novel strategies that retain the robust efficacy of GPR40 agonists for metabolic disorders while avoid toxicities caused by off-target and on-target mechanisms.
Collapse
|
6
|
Free Fatty Acid Receptors (FFARs) in Adipose: Physiological Role and Therapeutic Outlook. Cells 2022; 11:cells11040750. [PMID: 35203397 PMCID: PMC8870169 DOI: 10.3390/cells11040750] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 12/11/2022] Open
Abstract
Fatty acids (FFAs) are important biological molecules that serve as a major energy source and are key components of biological membranes. In addition, FFAs play important roles in metabolic regulation and contribute to the development and progression of metabolic disorders like diabetes. Recent studies have shown that FFAs can act as important ligands of G-protein-coupled receptors (GPCRs) on the surface of cells and impact key physiological processes. Free fatty acid-activated receptors include FFAR1 (GPR40), FFAR2 (GPR43), FFAR3 (GPR41), and FFAR4 (GPR120). FFAR2 and FFAR3 are activated by short-chain fatty acids like acetate, propionate, and butyrate, whereas FFAR1 and FFAR4 are activated by medium- and long-chain fatty acids like palmitate, oleate, linoleate, and others. FFARs have attracted considerable attention over the last few years and have become attractive pharmacological targets in the treatment of type 2 diabetes and metabolic syndrome. Several lines of evidence point to their importance in the regulation of whole-body metabolic homeostasis including adipose metabolism. Here, we summarize our current understanding of the physiological functions of FFAR isoforms in adipose biology and explore the prospect of FFAR-based therapies to treat patients with obesity and Type 2 diabetes.
Collapse
|
7
|
Patti AM, Giglio RV, Papanas N, Serban D, Stoian AP, Pafili K, Al Rasadi K, Rajagopalan K, Rizvi AA, Ciaccio M, Rizzo M. Experimental and Emerging Free Fatty Acid Receptor Agonists for the Treatment of Type 2 Diabetes. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58010109. [PMID: 35056417 PMCID: PMC8779029 DOI: 10.3390/medicina58010109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 04/11/2023]
Abstract
The current management of Type 2 Diabetes Mellitus (T2DM) includes incretin-based treatments able to enhance insulin secretion and peripheral insulin sensitivity as well as improve body mass, inflammation, plasma lipids, blood pressure, and cardiovascular outcomes. Dietary Free Fatty Acids (FFA) regulate metabolic and anti-inflammatory processes through their action on incretins. Selective synthetic ligands for FFA1-4 receptors have been developed as potential treatments for T2DM. To comprehensively review the available evidence for the potential role of FFA receptor agonists in the treatment of T2DM, we performed an electronic database search assessing the association between FFAs, T2DM, inflammation, and incretins. Evidence indicates that FFA1-4 agonism increases insulin sensitivity, induces body mass loss, reduces inflammation, and has beneficial metabolic effects. There is a strong inter-relationship between FFAs and incretins. FFA receptor agonism represents a potential target for the treatment of T2DM and may provide an avenue for the management of cardiometabolic risk in susceptible individuals. Further research promises to shed more light on this emerging topic.
Collapse
Affiliation(s)
- Angelo Maria Patti
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, 90133 Palermo, Italy; (A.M.P.); (M.R.)
| | - Rosaria Vincenza Giglio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (R.V.G.); (M.C.)
| | - Nikolaos Papanas
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, 68132 Alexandroupolis, Greece; (N.P.); (K.P.)
| | - Dragos Serban
- Forth Surgery Department, Faculty of Medicine, Carol Davila University, 050098 Bucharest, Romania;
| | - Anca Pantea Stoian
- Department of Diabetes, Faculty of Medicine, Nutrition and Metabolic Diseases, Carol Davila University, 050474 Bucharest, Romania;
| | - Kalliopi Pafili
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, 68132 Alexandroupolis, Greece; (N.P.); (K.P.)
| | - Khalid Al Rasadi
- Medical Research Center, Sultan Qaboos University, Muscat 123, Oman;
| | - Kanya Rajagopalan
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA;
| | - Ali A. Rizvi
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA;
- Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, SC 29208, USA
- Correspondence:
| | - Marcello Ciaccio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Laboratory Medicine, University of Palermo, 90127 Palermo, Italy; (R.V.G.); (M.C.)
- Department of Laboratory Medicine, University Hospital, 90127 Palermo, Italy
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, 90133 Palermo, Italy; (A.M.P.); (M.R.)
- Department of Diabetes, Faculty of Medicine, Nutrition and Metabolic Diseases, Carol Davila University, 050474 Bucharest, Romania;
- Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| |
Collapse
|
8
|
Miedzybrodzka EL, Gribble FM, Reimann F. Targeting the Enteroendocrine System for Treatment of Obesity. Handb Exp Pharmacol 2022; 274:487-513. [PMID: 35419620 DOI: 10.1007/164_2022_583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mimetics of the anorexigenic gut hormone glucagon-like peptide 1 (GLP-1) were originally developed as insulinotropic anti-diabetic drugs but also evoke significant weight loss, leading to their recent approval as obesity therapeutics. Co-activation of receptors for GLP-1 and other gut hormones which reduce food intake - peptide YY (PYY3-36), cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP) - is now being explored clinically to enhance efficacy. An alternative approach involves pharmacologically stimulating endogenous secretion of these hormones from enteroendocrine cells (EECs) to recapitulate the metabolic consequences of bariatric surgery, where highly elevated postprandial levels of GLP-1 and PYY3-36 are thought to contribute to improved glycaemia and weight loss.
Collapse
Affiliation(s)
- Emily L Miedzybrodzka
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
9
|
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
|
10
|
Nishizaki H, Matsuoka O, Kagawa T, Kobayashi A, Watanabe M, Moritoh Y. SCO-267, a GPR40 Full Agonist, Stimulates Islet and Gut Hormone Secretion and Improves Glycemic Control in Humans. Diabetes 2021; 70:2364-2376. [PMID: 34321316 PMCID: PMC8571351 DOI: 10.2337/db21-0451] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/20/2021] [Indexed: 11/21/2022]
Abstract
SCO-267 is a full agonist of the free fatty acid receptor 1 (GPR40), which regulates the secretion of islet and gut hormones. In this phase 1 study, we aimed to evaluate the clinical profile of single and multiple once-daily oral administration of SCO-267 in healthy adults and patients with diabetes. Plasma SCO-267 concentration was seen to increase in a dose-dependent manner after administration, and its plasma exposure was maintained for 24 h. Repeated dose did not alter the pharmacokinetic profile of SCO-267 in healthy adults. SCO-267 was generally safe and well tolerated at all evaluated single and multiple doses. Single and repeated doses of SCO-267 stimulated the secretion of insulin, glucagon, glucagon-like peptide 1, glucose-dependent insulinotropic polypeptide, and peptide YY in healthy adults. Furthermore, a single dose of SCO-267 stimulated the secretion of these hormones, decreased fasting hyperglycemia, and improved glycemic control during an oral glucose tolerance test in patients with diabetes, without inducing hypoglycemia. This study is the first to demonstrate the clinical effects of a GPR40 full agonist. SCO-267 is safe and well tolerated and exhibits once-daily oral dosing potential. Its robust therapeutic effects on hormonal secretion and glycemic control make SCO-267 an attractive drug candidate for the treatment of diabetes.
Collapse
Affiliation(s)
| | - Osamu Matsuoka
- Medical Corporation Heishinkai ToCROM Clinic, Tokyo, Japan
| | | | | | | | | |
Collapse
|
11
|
Shen YJ, Shen YC, Lee WS, Yang KT. Methyl palmitate protects heart against ischemia/reperfusion-induced injury through G-protein coupled receptor 40-mediated activation of the PI3K/AKT pathway. Eur J Pharmacol 2021; 905:174183. [PMID: 34015318 DOI: 10.1016/j.ejphar.2021.174183] [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: 11/23/2020] [Revised: 05/08/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022]
Abstract
This study aimed to investigate whether methyl palmitate (MP) exerts cardioprotective effect against the ischemia/reperfusion (I/R) injury and its mechanisms underlying. The cultured adult cardiomyocytes were treated with vehicle or lactic acid ischemic buffer (pH 6.8) during hypoxia/reoxygenation. In addition, the cardioprotective effect of MP was evaluated using the ex vivo heart model of I/R injury. Here, we found that MP significantly reduced the I/R-induced cardiomyocyte death. Treatment with GW1100 (a GPR40-antagonist) or wortmannin (a phosphatidylinositol 3-kinase, PI3K, specific inhibitor) significantly attenuated the level of phospho-AKT (p-AKT) and abolished the MP-induced cardioprotection against the I/R-induced injury. Using the ex vivo I/R model, we also demonstrated that pretreatment with MP significantly reduced the size of myocardial infarction and the levels of cleaved-caspase 3 and MDA, and increased the protein levels of GPR40 and p-AKT induced by I/R. The cardioprotective effect of MP was evaluated also using the in vivo heart model of I/R injury. We demonstrated that post-ischemic treatment with MP significantly attenuated the size of myocardial infarction and the serum level of CK-MB induced by in vivo I/R model. Taken together, our data suggest that MP could provide significant cardioprotection against the I/R injury, and the underlying mechanisms by which MP prevented the cardiomyocyte death might be mediated through the GPR40-activated PI3K/AKT signaling pathways. These findings suggest the potential applications of MP in the treatment of I/R-induced heart injury.
Collapse
Affiliation(s)
- Yan-Jhih Shen
- Ph.D. Program in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Yan-Cheng Shen
- Ph.D. Program in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Sen Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kun-Ta Yang
- Department of Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
12
|
Bazydlo-Guzenda K, Buda P, Matloka M, Mach M, Stelmach F, Dzida R, Smuga D, Hucz-Kalitowska J, Teska-Kaminska M, Vialichka V, Dubiel K, Kaminska B, Wieczorek M, Pieczykolan J. CPL207280 - a novel GPR40/FFA1-specific agonist shows a favorable safety profile and exerts anti-diabetic effects in type 2 diabetic animals.. Mol Pharmacol 2021; 100:335-347. [PMID: 34349026 DOI: 10.1124/molpharm.121.000260] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/14/2021] [Indexed: 11/22/2022] Open
Abstract
G protein-coupled receptor 40 (GPR40) is a free fatty acid receptor mainly expressed in pancreatic β-cells activated by medium- and long-chain fatty acids and regulating insulin secretion via an increase in cytosolic free calcium ([Ca2+]i). Activation of GPR40 in pancreatic β-cells may improve glycemic control in type 2 diabetes through enhancement of glucose-stimulated insulin secretion. However, the most clinically advanced GPR40 agonist - TAK-875 (fasiglifam) - was withdrawn from phase III due to its hepatotoxicity resulting from the inhibition of pivotal bile acid transporters. Here, we present a new, potent CPL207280 agonist and compare it with fasiglifam in numerous in vitro and in vivo studies. CPL207280 showed greater potency than fasiglifam in a Ca2+ influx assay with a hGPR40 protein (EC50=80 vs. 270 nM, respectively). At the 10 µM concentration, it showed 3.9 times greater enhancement of GSIS in mouse MIN6 pancreatic β cells. In Wistar Han rats and C57BL6 mice challenged with glucose, CPL207280 stimulated 2.5-times greater insulin secretion without causing hypoglycemia at 10 mg/kg compared with fasiglifam. In three diabetic rat models, CPL207280 improved glucose tolerance and increased insulin area under the curve by 212%, 142%, and 347%, respectively. Evaluation of potential off-target activity (Safety47{trade mark, serif}) and selectivity of CPL207280 (at 10 μM) did not show any significant off-target activity. We conclude that CPL207280 is a potent enhancer of glucose-stimulated insulin secretion in animal disease models with no risk of hypoglycemia at therapeutic doses. Therefore, we propose the CPL207280 compound as a compelling candidate for type 2 diabetes treatment. Significance Statement GPR40 is a well-known and promising target for diabetes. This study is the first to show the safety and effects of CPL207280, a novel GPR40/FFA1 agonist, on glucose homeostasis both in vitro and in vivo in different diabetic animal models. Therefore, we propose the CPL207280 compound as a novel, glucose-lowering agent, overcoming T2D patients' unmet medical needs.
Collapse
Affiliation(s)
| | - Pawel Buda
- Research and Development Centre, Celon Pharma SA, Poland
| | | | - Mateusz Mach
- Research and Development Centre, Celon Pharma SA, Poland
| | - Filip Stelmach
- Research and Development Centre, Celon Pharma SA, Poland
| | - Radoslaw Dzida
- Research and Development Centre, Celon Pharma SA, Poland
| | - Damian Smuga
- Research and Development Centre, Celon Pharma, Poland
| | | | | | | | | | - Bozena Kaminska
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Poland
| | | | | |
Collapse
|
13
|
Nauck MA, Wefers J, Meier JJ. Treatment of type 2 diabetes: challenges, hopes, and anticipated successes. Lancet Diabetes Endocrinol 2021; 9:525-544. [PMID: 34181914 DOI: 10.1016/s2213-8587(21)00113-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
Despite the successful development of new therapies for the treatment of type 2 diabetes, such as glucagon-like peptide-1 (GLP-1) receptor agonists and sodium-glucose cotransporter-2 inhibitors, the search for novel treatment options that can provide better glycaemic control and at reduce complications is a continuous effort. The present Review aims to present an overview of novel targets and mechanisms and focuses on glucose-lowering effects guiding this search and developments. We discuss not only novel developments of insulin therapy (eg, so-called smart insulin preparation with a glucose-dependent mode of action), but also a group of drug classes for which extensive research efforts have not been rewarded with obvious clinical impact. We discuss the potential clinical use of the salutary adipokine adiponectin and the hepatokine fibroblast growth factor (FGF) 21, among others. A GLP-1 peptide receptor agonist (semaglutide) is now available for oral absorption, and small molecules activating GLP-1 receptors appear on the horizon. Bariatric surgery and its accompanying changes in the gut hormonal milieu offer a background for unimolecular peptides interacting with two or more receptors (for GLP-1, glucose-dependent insulinotropic polypeptide, glucagon, and peptide YY) and provide more substantial glycaemic control and bodyweight reduction compared with selective GLP-1 receptor agonists. These and additional approaches will help expand the toolbox of effective medications needed for optimising the treatment of well delineated subgroups of type 2 diabetes or help develop personalised approaches for glucose-lowering drugs based on individual characteristics of our patients.
Collapse
Affiliation(s)
- Michael A Nauck
- Diabetes Division, Katholisches Klinikum Bochum, St Josef Hospital, Ruhr University Bochum, Bochum, Germany.
| | - Jakob Wefers
- Diabetes Division, Katholisches Klinikum Bochum, St Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Juris J Meier
- Diabetes Division, Katholisches Klinikum Bochum, St Josef Hospital, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
14
|
Nunes Marsiglio-Librais G, Aparecida Vilas-Boas E, Carlein C, Hoffmann MDA, Roma LP, Carpinelli AR. Evidence for NADPH oxidase activation by GPR40 in pancreatic β-cells. Redox Rep 2021; 25:41-50. [PMID: 32354273 PMCID: PMC7241480 DOI: 10.1080/13510002.2020.1757877] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective: Investigate the involvement of the fatty acids receptor GPR40 in the assembly and activation of NADPH oxidase and the implications on pancreatic β-cell function. Methods: BRIN-BD11 β-cells were exposed to GPR40 agonist (GW9508) or linoleic acid in different glucose concentrations. Superoxide and H2O2 were analyzed, respectively, by DHE fluorescence and by fluorescence of the H2O2 sensor, roGFP2-Orp1. Protein contents of p47phox in plasma membrane and cytosol were analyzed by western blot. NADPH oxidase role was evaluated by p22phox siRNA or by pharmacological inhibition with VAS2870. NOX2 KO islets were used to measure total cytosolic calcium and insulin secretion. Results: GW9508 and linoleic acid increased superoxide and H2O2 contents at 5.6 and 8.3 mM of glucose. In addition, in 5.6 mM, but not at 16.7 mM of glucose, activation of GPR40 led to the translocation of p47phox to the plasma membrane. Knockdown of p22phox abolished the increase in superoxide after GW9508 and linoleic acid. No differences in insulin secretion were found between wild type and NOX2 KO islets treated with GW9508 or linoleic acid. Discussion: We report for the first time that acute activation of GPR40 leads to NADPH oxidase activation in pancreatic β-cells, without impact on insulin secretion.
Collapse
Affiliation(s)
| | - Eloisa Aparecida Vilas-Boas
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil.,Department of Biophysics, Center for Human and Molecular Biology, CIPMM, Saarland University, Homburg/Saar, Germany
| | - Christopher Carlein
- Department of Biophysics, Center for Human and Molecular Biology, CIPMM, Saarland University, Homburg/Saar, Germany
| | | | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology, CIPMM, Saarland University, Homburg/Saar, Germany
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
15
|
Yang M, Reimann F, Gribble FM. Chemosensing in enteroendocrine cells: mechanisms and therapeutic opportunities. Curr Opin Endocrinol Diabetes Obes 2021; 28:222-231. [PMID: 33449572 DOI: 10.1097/med.0000000000000614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Enteroendocrine cells (EECs) are scattered chemosensory cells in the intestinal epithelium that release hormones with a wide range of actions on intestinal function, food intake and glucose homeostasis. The mechanisms by which gut hormones are secreted postprandially, or altered by antidiabetic agents and surgical interventions are of considerable interest for future therapeutic development. RECENT FINDINGS EECs are electrically excitable and express a repertoire of G-protein coupled receptors that sense nutrient and nonnutrient stimuli, coupled to intracellular Ca2+ and cyclic adenosine monophosphate. Our knowledge of EEC function, previously developed using mouse models, has recently been extended to human cells. Gut hormone release in humans is enhanced by bariatric surgery, as well as by some antidiabetic agents including sodium-coupled glucose transporter inhibitors and metformin. SUMMARY EECs are important potential therapeutic targets. A better understanding of their chemosensory mechanisms will enhance the development of new therapeutic strategies to treat metabolic and gastrointestinal diseases.
Collapse
Affiliation(s)
- Ming Yang
- University of Cambridge, Institute of Metabolic Science and MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Cambridge, UK
| | | | | |
Collapse
|
16
|
Screening of a novel free fatty acid receptor 1 (FFAR1) agonist peptide by phage display and machine learning based-amino acid substitution. Biochem Biophys Res Commun 2021; 550:177-183. [PMID: 33706101 DOI: 10.1016/j.bbrc.2021.02.142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 02/28/2021] [Indexed: 11/27/2022]
Abstract
Free fatty acid receptor 1 (FFAR1 or GPR40) has attracted attention for the treatment of type 2 diabetes mellitus, and various small-molecule agonists have been developed. However, most FFAR1 agonists as well as endogenous ligands, such as linoleic acids, have high lipophilicity, and their high lipophilicity is related to off-target toxicity. Therefore, we need to focus on new ligand candidates with less toxicity. In this study, we screened peptides with FFAR1 agonist activity as new ligand candidates. First, we used phage display to identify peptides with high affinity to FFAR1. Next, the agonist activities of peptides determined by the phage display were evaluated by the TGF-α shedding assay. Finally, to improve the FFAR1 agonist activity of the peptide, we performed an inclusive single amino acid substitution and sequence analysis. Logistic regression (LR) analysis using 120 physiochemical properties was performed to predict peptides with high FFAR1 agonist activity. STTGTQY determined by phage display promoted glucose-stimulated insulin secretion in pancreatic MIN6 cells. Furthermore, STKGTF predicted by the LR analysis showed high insulin secretion at low concentrations compared to STTGTQY. The results of this study suggest that peptides could be new candidates as FFAR1 agonists.
Collapse
|
17
|
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
|
18
|
Choi SH, Lee R, Nam SM, Kim DG, Cho IH, Kim HC, Cho Y, Rhim H, Nah SY. Ginseng gintonin, aging societies, and geriatric brain diseases. Integr Med Res 2021; 10:100450. [PMID: 32817818 PMCID: PMC7426447 DOI: 10.1016/j.imr.2020.100450] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A dramatic increase in aging populations and low birth rates rapidly drive aging societies and increase aging-associated neurodegenerative diseases. However, functional food or medicinal formulations to prevent geriatric brain disorders are not readily available. Panax ginseng is a candidate, since ginseng has long-been consumed as a rejuvenating agent. However, the underlying molecular mechanisms and the components of ginseng that are responsible for brain rejuvenation and human longevity are unknown. Accumulating evidence shows that gintonin is a candidate for the anti-aging ingredient of ginseng, especially in brain senescence. METHODS Gintonin, a glycolipoprotein complex, contains three lipid-derived G protein-coupled receptor ligands: lysophosphatidic acids (LPAs), lysophosphatidylinositols (LPIs), and linoleic acid (LA). LPA, LPI, and LA act on six LPA receptor subtypes, GPR55, and GPR40, respectively. These G protein-coupled receptors are distributed within the nervous and non-nervous systems of the human body. RESULTS Gintonin-enriched fraction (GEF) exhibits anti-brain senescence and effects against disorders such as Alzheimer's disease (AD), Huntington's disease (HD), and Parkinson's disease (PD). Oral administration of gintonin in animal models of d-galactose-induced brain aging, AD, HD, and PD restored cognitive and motor functions. The underlying molecular mechanisms of gintonin-mediated anti-brain aging and anti-neurodegenerative diseases include neurogenesis, autophagy stimulation, anti-apoptosis, anti-oxidative stress, and anti-inflammatory activities. This review describes the characteristics of gintonin and GEF, and how gintonin exerts its effects on brain aging and brain associated-neurodegenerative diseases. CONCLUSION Finally, we describe how GEF can be applied to improve the quality of life of senior citizens in aging societies.
Collapse
Affiliation(s)
- Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Rami Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sung Min Nam
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Do-Geun Kim
- Neurovascular Biology Laboratory, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Yoonjeong Cho
- Center for Neuroscience Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Yang M, Zhang CY. G protein-coupled receptors as potential targets for nonalcoholic fatty liver disease treatment. World J Gastroenterol 2021; 27:677-691. [PMID: 33716447 PMCID: PMC7934005 DOI: 10.3748/wjg.v27.i8.677] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/24/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a broad-spectrum disease, ranging from simple hepatic steatosis to nonalcoholic steatohepatitis, which can progress to cirrhosis and liver cancer. Abnormal hepatic lipid accumulation is the major manifestation of this disease, and lipotoxicity promotes NAFLD progression. In addition, intermediate metabolites such as succinate can stimulate the activation of hepatic stellate cells to produce extracellular matrix proteins, resulting in progression of NAFLD to fibrosis and even cirrhosis. G protein-coupled receptors (GPCRs) have been shown to play essential roles in metabolic disorders, such as NAFLD and obesity, through their function as receptors for bile acids and free fatty acids. In addition, GPCRs link gut microbiota-mediated connections in a variety of diseases, such as intestinal diseases, hepatic steatosis, diabetes, and cardiovascular diseases. The latest findings show that gut microbiota-derived acetate contributes to liver lipogenesis by converting dietary fructose into hepatic acetyl-CoA and fatty acids. GPCR agonists, including peptides and natural products like docosahexaenoic acid, have been applied to investigate their role in liver diseases. Therapies such as probiotics and GPCR agonists may be applied to modulate GPCR function to ameliorate liver metabolism syndrome. This review summarizes the current findings regarding the role of GPCRs in the development and progression of NAFLD and describes some preclinical and clinical studies of GPCR-mediated treatment. Overall, understanding GPCR-mediated signaling in liver disease may provide new therapeutic options for NAFLD.
Collapse
Affiliation(s)
- Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
| | - Chun-Ye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65212, United States
| |
Collapse
|
20
|
Koyama R, Ookawara M, Watanabe M, Moritoh Y. Chronic Exposure to SCO-267, an Allosteric GPR40 Full Agonist, Is Effective in Improving Glycemic Control in Rats. Mol Pharmacol 2021; 99:286-293. [PMID: 33547250 DOI: 10.1124/molpharm.120.000168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/15/2021] [Indexed: 11/22/2022] Open
Abstract
Full agonist-mediated activation of free fatty acid receptor 1 (FFAR1/GPR40) alleviates diabetes in rodents. Considering that diabetes is a chronic disease, assessment of treatment durability of chronic exposure to a GPR40 full agonist is pivotal for treating patients with diabetes. However, the physiologic significance of chronic in vitro and in vivo exposure to GPR40 full agonists is largely unclear. Here, we evaluated the in vitro and in vivo effects of chronic treatment with SCO-267, a GPR40 full agonist, on signal transduction and glucose control. In vitro experiments showed that SCO-267 is an allosteric full agonist for GPR40, which activates the Gα q, Gα s, and Gα 12/13 pathways and β-arrestin recruitment. The calcium signal response was largely sustained in GPR40-overexpressing CHO cells even after prolonged incubation with SCO-267. To evaluate the in vivo relevance of chronic exposure to GPR40 full agonists, SCO-267 (1 and 10 mg/kg) was administered once daily to neonatally streptozotocin-induced diabetic rats for 15-33 days, and glucose control was evaluated. After 15 days of dosing followed by the drug washout period, SCO-267 improved glucose tolerance, most likely by increasing insulin sensitivity in rats. After 33 days, repeated exposure to SCO-267 was highly effective in improving glucose tolerance in rats. Furthermore, chronic exposure to SCO-267 increased pancreatic insulin content. These results demonstrated that even after chronic exposure, SCO-267 effectively activates GPR40 in cells and rats, suggesting the clinical application of SCO-267 in treating chronic diseases including diabetes. SIGNIFICANCE STATEMENT: GPR40 is a validated therapeutic target for diabetes. This study showed that even after chronic exposure, SCO-267, an allosteric GPR40 full agonist, effectively activates GPR40 in cells and rats; these results suggest a durable efficacy of SCO-267 in patients.
Collapse
Affiliation(s)
- Ryokichi Koyama
- Research Division, SCOHIA PHARMA, Inc., Fujisawa, Kanagawa, Japan
| | - Mitsugi Ookawara
- Research Division, SCOHIA PHARMA, Inc., Fujisawa, Kanagawa, Japan
| | | | - Yusuke Moritoh
- Research Division, SCOHIA PHARMA, Inc., Fujisawa, Kanagawa, Japan
| |
Collapse
|
21
|
Taylor SI, Yazdi ZS, Beitelshees AL. Pharmacological treatment of hyperglycemia in type 2 diabetes. J Clin Invest 2021; 131:142243. [PMID: 33463546 PMCID: PMC7810496 DOI: 10.1172/jci142243] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is a major public health problem, affecting about 10% of the population. Pharmacotherapy aims to protect against microvascular complications, including blindness, end-stage kidney disease, and amputations. Landmark clinical trials have demonstrated that intensive glycemic control slows progression of microvascular complications (retinopathy, nephropathy, and neuropathy). Long-term follow-up has demonstrated that intensive glycemic control also decreases risk of macrovascular disease, albeit rigorous evidence of macrovascular benefit did not emerge for over a decade. The US FDA's recent requirement for dedicated cardiovascular outcome trials ushered in a golden age for understanding the clinical profiles of new type 2 diabetes drugs. Some clinical trials with sodium-glucose cotransporter-2 (SGLT2) inhibitors and glucagon-like peptide 1 (GLP1) receptor agonists reported data demonstrating cardiovascular benefit (decreased risk of major adverse cardiovascular events and hospitalization for heart failure) and slower progression of diabetic kidney disease. This Review discusses current guidelines for use of the 12 classes of drugs approved to promote glycemic control in patients with type 2 diabetes. The Review also anticipates future developments with potential to improve the standard of care: availability of generic dipeptidylpeptidase-4 (DPP4) inhibitors and SGLT2 inhibitors; precision medicine to identify the best drugs for individual patients; and new therapies to protect against chronic complications of diabetes.
Collapse
|
22
|
Lin C, Cai X, Yang W, Lv F, Nie L, Ji L. Age, sex, disease severity, and disease duration difference in placebo response: implications from a meta-analysis of diabetes mellitus. BMC Med 2020; 18:322. [PMID: 33190640 PMCID: PMC7667845 DOI: 10.1186/s12916-020-01787-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/17/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The placebo response in patients with diabetes mellitus is very common. A systematic evaluation needs to be updated with the current evidence about the placebo response in diabetes mellitus and the associated factors in clinical trials of anti-diabetic medicine. METHODS Literature research was conducted in Medline, Embase, the Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov for studies published between the date of inception and June 2019. Randomized placebo-controlled trials conducted in type 1and type 2 diabetes mellitus (T1DM/T2DM) were included. Random-effects model and meta-regression analysis were accordingly used. This meta-analysis was registered in PROSPERO as CRD42014009373. RESULTS Significantly weight elevation (effect size (ES) = 0.33 kg, 95% CI, 0.03 to 0.61 kg) was observed in patients with placebo treatments in T1DM subgroup while significantly HbA1c reduction (ES = - 0.12%, 95% CI, - 0.16 to - 0.07%) and weight reduction (ES = - 0.40 kg, 95% CI, - 0.50 to - 0.29 kg) were observed in patients with placebo treatments in T2DM subgroup. Greater HbA1c reduction was observed in patients with injectable placebo treatments (ES = - 0.22%, 95% CI, - 0.32 to - 0.11%) versus oral types (ES = - 0.09%, 95% CI, - 0.14 to - 0.04%) in T2DM (P = 0.03). Older age (β = - 0.01, 95% CI, - 0.02 to - 0.01, P < 0.01) and longer diabetes duration (β = - 0.02, 95% CI, - 0.03 to - 0.21 × 10-2, P = 0.03) was significantly associated with more HbA1c reduction by placebo in T1DM. However, younger age (β = 0.02, 95% CI, 0.01 to 0.03, P = 0.01), lower male percentage (β = 0.01, 95% CI, 0.22 × 10-2, 0.01, P < 0.01), higher baseline BMI (β = - 0.02, 95% CI, - 0.04 to - 0.26 × 10-2, P = 0.02), and higher baseline HbA1c (β = - 0.09, 95% CI, - 0.16 to - 0.01, P = 0.02) were significantly associated with more HbA1c reduction by placebo in T2DM. Shorter diabetes duration (β = 0.06, 95% CI, 0.06 to 0.10, P < 0.01) was significantly associated with more weight reduction by placebo in T2DM. However, the associations between baseline BMI, baseline HbA1c, and placebo response were insignificant after the adjusted analyses. CONCLUSION The placebo response in diabetes mellitus was systematically outlined. Age, sex, disease severity (indirectly reflected by baseline BMI and baseline HbA1c), and disease duration were associated with placebo response in diabetes mellitus. The association between baseline BMI, baseline HbA1c, and placebo response may be the result of regression to the mean.
Collapse
Affiliation(s)
- Chu Lin
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| | - Wenjia Yang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Fang Lv
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Lin Nie
- Department of Endocrinology and Metabolism, Beijing Airport Hospital, Beijing, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| |
Collapse
|
23
|
Mizuno H, Kihara Y. Druggable Lipid GPCRs: Past, Present, and Prospects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:223-258. [PMID: 32894513 DOI: 10.1007/978-3-030-50621-6_10] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) have seven transmembrane spanning domains and comprise the largest superfamily with ~800 receptors in humans. GPCRs are attractive targets for drug discovery because they transduce intracellular signaling in response to endogenous ligands via heterotrimeric G proteins or arrestins, resulting in a wide variety of physiological and pathophysiological responses. The endogenous ligands for GPCRs are highly chemically diverse and include ions, biogenic amines, nucleotides, peptides, and lipids. In this review, we follow the KonMari method to better understand druggable lipid GPCRs. First, we have a comprehensive tidying up of lipid GPCRs including receptors for prostanoids, leukotrienes, specialized pro-resolving mediators (SPMs), lysophospholipids, sphingosine 1-phosphate (S1P), cannabinoids, platelet-activating factor (PAF), free fatty acids (FFAs), and sterols. This tidying up consolidates 46 lipid GPCRs and declutters several perplexing lipid GPCRs. Then, we further tidy up the lipid GPCR-directed drugs from the literature and databases, which identified 24 clinical drugs targeting 16 unique lipid GPCRs available in the market and 44 drugs under evaluation in more than 100 clinical trials as of 2019. Finally, we introduce drug designs for GPCRs that spark joy, such as positive or negative allosteric modulators (PAM or NAM), biased agonism, functional antagonism like fingolimod, and monoclonal antibodies (MAbs). These strategic drug designs may increase the efficacy and specificity of drugs and reduce side effects. Technological advances will help to discover more endogenous lipid ligands from the vast number of remaining orphan GPCRs and will also lead to the development novel lipid GPCR drugs to treat various diseases.
Collapse
Affiliation(s)
| | - Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
24
|
Souza PR, Walker ME, Goulding NJ, Dalli J, Perretti M, Norling LV. The GPR40 Agonist GW9508 Enhances Neutrophil Function to Aid Bacterial Clearance During E. coli Infections. Front Immunol 2020; 11:573019. [PMID: 33133087 PMCID: PMC7550532 DOI: 10.3389/fimmu.2020.573019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
G-protein-coupled receptor 40 (GPR40) is known to play a role in the regulation of fatty acids, insulin secretion, and inflammation. However, the function of this receptor in human neutrophils, one of the first leukocytes to arrive at the site of infection, remains to be fully elucidated. In the present study, we demonstrate that GPR40 is upregulated on activated human neutrophils and investigated the functional effects upon treatment with a selective agonist; GW9508. Interestingly, GPR40 expression was up-regulated after neutrophil stimulation with platelet-activating factor (10 nM) or leukotriene B4 (LTB4, 10 nM) suggesting potential regulatory roles for this receptor during inflammation. Indeed, GW9508 (1 and 10 μM) increased neutrophil chemotaxis in response to the chemokine IL-8 (30 ng/ml) and enhanced phagocytosis of Escherichia coli by approximately 50% when tested at 0.1 and 1 μM. These results were translated in vivo whereby administration of GW9508 (10 mg/kg, i.p.) during E. coli infections resulted in elevated peritoneal leukocyte infiltration with a higher phagocytic capacity. Importantly, GW9508 administration also modulated the lipid mediator profile, with increased levels of the pro-resolving mediators resolvin D3 and lipoxins. In conclusion, GPR40 is expressed by activated neutrophils and plays an important host protective role to aid clearance of bacterial infections.
Collapse
Affiliation(s)
- Patricia R Souza
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mary E Walker
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Nicolas J Goulding
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jesmond Dalli
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Lucy V Norling
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
25
|
Furukawa H, Miyamoto Y, Hirata Y, Watanabe K, Hitomi Y, Yoshitomi Y, Aida J, Noguchi N, Takakura N, Takami K, Miwatashi S, Hirozane Y, Hamada T, Ito R, Ookawara M, Moritoh Y, Watanabe M, Maekawa T. Design and Identification of a GPR40 Full Agonist ( SCO-267) Possessing a 2-Carbamoylphenyl Piperidine Moiety. J Med Chem 2020; 63:10352-10379. [PMID: 32900194 DOI: 10.1021/acs.jmedchem.0c00843] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
GPR40/FFAR1 is a G-protein-coupled receptor expressed in pancreatic β-cells and enteroendocrine cells. GPR40 activation stimulates secretions of insulin and incretin, both of which are the pivotal regulators of glycemic control. Therefore, a GPR40 agonist is an attractive target for the treatment of type 2 diabetes mellitus. Using the reported biaryl derivative 1, we shifted the hydrophobic moiety to the terminal aryl ring and replaced the central aryl ring with piperidine, generating 2-(4,4-dimethylpentyl)phenyl piperidine 4a, which had improved potency for GPR40 and high lipophilicity. We replaced the hydrophobic moiety with N-alkyl-N-aryl benzamides to lower the lipophilicity and restrict the N-alkyl moieties to the presumed lipophilic pocket using the intramolecular π-π stacking of cis-preferential N-alkyl-N-aryl benzamide. Among these, orally available (3S)-3-cyclopropyl-3-(2-((1-(2-((2,2-dimethylpropyl)(6-methylpyridin-2-yl)carbamoyl)-5-methoxyphenyl)piperidin-4-yl)methoxy)pyridin-4-yl)propanoic acid (SCO-267) effectively stimulated insulin secretion and GLP-1 release and ameliorated glucose tolerance in diabetic rats via GPR40 full agonism.
Collapse
Affiliation(s)
- Hideki Furukawa
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yasufumi Miyamoto
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yasuhiro Hirata
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Koji Watanabe
- Research Division, SCOHIA PHARMA Inc., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yuko Hitomi
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yayoi Yoshitomi
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Jumpei Aida
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Naoyoshi Noguchi
- Research Division, SCOHIA PHARMA Inc., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Nobuyuki Takakura
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kazuaki Takami
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Seiji Miwatashi
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yoshihiko Hirozane
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Teruki Hamada
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Ryo Ito
- Research, Takeda Pharmaceutical Company, Ltd., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Mitsugi Ookawara
- Research Division, SCOHIA PHARMA Inc., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yusuke Moritoh
- Research Division, SCOHIA PHARMA Inc., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masanori Watanabe
- Research Division, SCOHIA PHARMA Inc., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tsuyoshi Maekawa
- Research Division, SCOHIA PHARMA Inc., Shonan Health Innovation Park, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
26
|
Koido M, Kawakami E, Fukumura J, Noguchi Y, Ohori M, Nio Y, Nicoletti P, Aithal GP, Daly AK, Watkins PB, Anayama H, Dragan Y, Shinozawa T, Takebe T. Polygenic architecture informs potential vulnerability to drug-induced liver injury. Nat Med 2020; 26:1541-1548. [DOI: 10.1038/s41591-020-1023-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/20/2020] [Indexed: 12/12/2022]
|
27
|
Atanasio S, Deganutti G, Reynolds CA. Addressing free fatty acid receptor 1 (FFAR1) activation using supervised molecular dynamics. J Comput Aided Mol Des 2020; 34:1181-1193. [PMID: 32851580 DOI: 10.1007/s10822-020-00338-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/18/2020] [Indexed: 01/12/2023]
Abstract
The free fatty acid receptor 1 (FFAR1, formerly GPR40), is a potential G protein-coupled receptor (GPCR) target for the treatment of type 2 diabetes mellitus (T2DM), as it enhances glucose-dependent insulin secretion upon activation by endogenous long-chain free fatty acids. The presence of two allosterically communicating binding sites and the lack of the conserved GPCR structural motifs challenge the general knowledge of its activation mechanism. To date, four X-ray crystal structures are available for computer-aided drug design. In this study, we employed molecular dynamics (MD) and supervised molecular dynamics (SuMD) to deliver insights into the (un)binding mechanism of the agonist MK-8666, and the allosteric communications between the two experimentally determined FFAR1 binding sites. We found that FFAR1 extracellular loop 2 (ECL2) mediates the binding of the partial agonist MK-8666. Moreover, simulations showed that the agonists MK-8666 and AP8 are reciprocally stabilized and that AP8 influences MK-8666 unbinding from FFAR1.
Collapse
Affiliation(s)
- Silvia Atanasio
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Giuseppe Deganutti
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK. .,Centre for Sport, Exercise and Life Sciences, Coventry University, Alison Gingell Building, Coventry, CV1 5FB, UK.
| | - Christopher A Reynolds
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK.,Centre for Sport, Exercise and Life Sciences, Coventry University, Alison Gingell Building, Coventry, CV1 5FB, UK
| |
Collapse
|
28
|
Drzazga A, Okulus M, Rychlicka M, Biegała Ł, Gliszczyńska A, Gendaszewska-Darmach E. Lysophosphatidylcholine Containing Anisic Acid Is Able to Stimulate Insulin Secretion Targeting G Protein Coupled Receptors. Nutrients 2020; 12:E1173. [PMID: 32331428 PMCID: PMC7230207 DOI: 10.3390/nu12041173] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/31/2022] Open
Abstract
Diabetes mellitus is a worldwide health problem with high rates of mortality and morbidity. Management of diabetes mellitus by dietary components is achievable especially at the initial stage of the disease. Several studies confirmed the antidiabetic activities of simple phenolic acids and lysophosphatidylcholine (LPC). The main goal of this study was to identify new potential insulin secretion modulators obtained by combining the structures of two natural compounds, namely O-methyl derivatives of phenolic acids and phospholipids. LPC and phosphatidylcholine bearing methoxylated aromatic carboxylic acids were tested as potential agents able to improve glucose-stimulated insulin secretion (GSIS) and intracellular calcium mobilization in MIN6 β pancreatic cell line. Our results show that LPC with covalently bonded molecule of p-anisic acid at the sn-1 position was able to induce GSIS and intracellular calcium flux. Notably, 1-anisoyl-2-hydroxy-sn-glycero-3-phosphocholine did not affect the viability of MIN6 cells, suggesting its potential safe use. Furthermore, we have shown that three G protein coupled receptors, namely GPR40, GPR55, and GPR119, are targeted by this LPC derivative.
Collapse
Affiliation(s)
- Anna Drzazga
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924 Lodz, Poland; (A.D.); (Ł.B.)
| | - Marta Okulus
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland; (M.O.); (M.R.)
| | - Magdalena Rychlicka
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland; (M.O.); (M.R.)
| | - Łukasz Biegała
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924 Lodz, Poland; (A.D.); (Ł.B.)
| | - Anna Gliszczyńska
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland; (M.O.); (M.R.)
| | - Edyta Gendaszewska-Darmach
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924 Lodz, Poland; (A.D.); (Ł.B.)
| |
Collapse
|
29
|
Vilas-Boas EA, Karabacz N, Marsiglio-Librais GN, Valle MMR, Nalbach L, Ampofo E, Morgan B, Carpinelli AR, Roma LP. Chronic activation of GPR40 does not negatively impact upon BRIN-BD11 pancreatic β-cell physiology and function. Pharmacol Rep 2020; 72:1725-1737. [PMID: 32274767 PMCID: PMC7704488 DOI: 10.1007/s43440-020-00101-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/18/2020] [Accepted: 03/21/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Free fatty acids (FFAs) are known for their dual effects on insulin secretion and pancreatic β-cell survival. Short-term exposure to FFAs, such as palmitate, increases insulin secretion. On the contrary, long-term exposure to saturated FFAs results in decreased insulin secretion, as well as triggering oxidative stress and endoplasmic reticulum (ER) stress, culminating in cell death. The effects of FFAs can be mediated either via their intracellular oxidation and consequent effects on cellular metabolism or via activation of the membrane receptor GPR40. Both pathways are likely to be activated upon both short- and long-term exposure to FFAs. However, the precise role of GPR40 in β-cell physiology, especially upon chronic exposure to FFAs, remains unclear. METHODS We used the GPR40 agonist (GW9508) and antagonist (GW1100) to investigate the impact of chronically modulating GPR40 activity on BRIN-BD11 pancreatic β-cells physiology and function. RESULTS We observed that chronic activation of GPR40 did not lead to increased apoptosis, and both proliferation and glucose-induced calcium entry were unchanged compared to control conditions. We also observed no increase in H2O2 or superoxide levels and no increase in the ER stress markers p-eIF2α, CHOP and BIP. As expected, palmitate led to increased H2O2 levels, decreased cell viability and proliferation, as well as decreased metabolism and calcium entry. These changes were not counteracted by the co-treatment of palmitate-exposed cells with the GPR40 antagonist GW1100. CONCLUSIONS Chronic activation of GPR40 using GW9508 does not negatively impact upon BRIN-BD11 pancreatic β-cells physiology and function. The GPR40 antagonist GW1100 does not protect against the deleterious effects of chronic palmitate exposure. We conclude that GPR40 is probably not involved in mediating the toxicity associated with chronic palmitate exposure.
Collapse
Affiliation(s)
- Eloisa Aparecida Vilas-Boas
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil.,Department of Biophysics, Center for Human and Molecular Biology, Saarland University, Universität Des Saarlandes, CIPMM, Geb. 48, 66421, Homburg/Saar, Germany
| | - Noémie Karabacz
- Department of Biophysics, Center for Human and Molecular Biology, Saarland University, Universität Des Saarlandes, CIPMM, Geb. 48, 66421, Homburg/Saar, Germany
| | | | - Maíra Melo Rezende Valle
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Lisa Nalbach
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg/Saar, Germany
| | - Bruce Morgan
- Institute of Biochemistry, Center for Human and Molecular Biology (ZHMB), Saarland University, 66123, Saarbrücken, Germany
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo (USP), Sao Paulo, SP, Brazil
| | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology, Saarland University, Universität Des Saarlandes, CIPMM, Geb. 48, 66421, Homburg/Saar, Germany.
| |
Collapse
|
30
|
Cho YJ, Choi SH, Lee R, Hwang H, Rhim H, Cho IH, Kim HC, Lee JI, Hwang SH, Nah SY. Ginseng Gintonin Contains Ligands for GPR40 and GPR55. Molecules 2020; 25:molecules25051102. [PMID: 32121640 PMCID: PMC7179172 DOI: 10.3390/molecules25051102] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/17/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
Gintonin, a novel ginseng-derived glycolipoprotein complex, has an exogenous ligand for lysophosphatidic acid (LPA) receptors. However, recent lipid analysis of gintonin has shown that gintonin also contains other bioactive lipids besides LPAs, including linoleic acid and lysophosphatidylinositol (LPI). Linoleic acid, a free fatty acid, and LPI are known as ligands for the G-protein coupled receptors (GPCR), GPR40, and GPR55, respectively. We, herein, investigated whether gintonin could serve as a ligand for GPR40 and GPR55, using the insulin-secreting beta cell-derived cell line INS-1 and the human prostate cancer cell line PC-3, respectively. Gintonin dose-dependently enhanced insulin secretion from INS-1 cells. Gintonin-stimulated insulin secretion was partially inhibited by a GPR40 receptor antagonist but not an LPA1/3 receptor antagonist and was down-regulated by small interfering RNA (siRNA) against GPR40. Gintonin dose-dependently induced [Ca2+]i transients and Ca2+-dependent cell migration in PC-3 cells. Gintonin actions in PC-3 cells were attenuated by pretreatment with a GPR55 antagonist and an LPA1/3 receptor antagonist or by down-regulating GPR55 with siRNA. Taken together, these results demonstrated that gintonin-mediated insulin secretion by INS-1 cells and PC-3 cell migration were regulated by the respective activation of GPR40 and GPR55 receptors. These findings indicated that gintonin could function as a ligand for both receptors. Finally, we demonstrated that gintonin contained two more GPCR ligands, in addition to that for LPA receptors. Gintonin, with its multiple GPCR ligands, might provide the molecular basis for the multiple pharmacological actions of ginseng.
Collapse
Affiliation(s)
- Yeon-Jin Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea; (Y.-J.C.); (S.-H.C.); (R.L.)
| | - Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea; (Y.-J.C.); (S.-H.C.); (R.L.)
| | - Rami Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea; (Y.-J.C.); (S.-H.C.); (R.L.)
| | - Hongik Hwang
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea; (H.H.); (H.R.)
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea; (H.H.); (H.R.)
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, Department of Science in Korean Medicine and Brain Korea 21 Plus Program, Graduate School, Kyung Hee University, Seoul 02447, Korea;
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology program, College of Pharmacy, Kangwon National University, Chunchon 24341, Korea;
| | - Jeong-Ik Lee
- Department of Veterinary Obstetrics and Theriogenology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea;
| | - Sung-Hee Hwang
- Department of Pharmaceutical Engineering, College of Health Sciences, Sangji University, Wonju 26339, Korea
- Correspondence: (S.-H.H.); (S.-Y.N.); Tel.: +82-33-738-7922 (S.-H.H.); +82-2-450-4154 (S.-Y.N.)
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea; (Y.-J.C.); (S.-H.C.); (R.L.)
- Correspondence: (S.-H.H.); (S.-Y.N.); Tel.: +82-33-738-7922 (S.-H.H.); +82-2-450-4154 (S.-Y.N.)
| |
Collapse
|
31
|
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]
|
32
|
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
|
33
|
The Role of Long-Chain Fatty Acids in Inflammatory Bowel Disease. Mediators Inflamm 2019; 2019:8495913. [PMID: 31780872 PMCID: PMC6874876 DOI: 10.1155/2019/8495913] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/03/2019] [Indexed: 02/05/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a complicated disease involving multiple pathogenic factors. The complex relationships between long-chain fatty acids (LCFAs) and the morbidity of IBD drive numerous studies to unravel the underlying mechanisms. A better understanding of the role of LCFAs in IBD will substitute or boost the current IBD therapies, thereby obtaining mucosal healing. In this review, we focused on the roles of LCFAs on the important links of inflammatory regulation in IBD, including in the pathogen recognition phase and in the inflammatory resolving phase, and the effects of LCFAs on immune cells in IBD.
Collapse
|
34
|
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
|
35
|
Gupta MK, Vasudevan NT. GPCRs and Insulin Receptor Signaling in Conversation: Novel Avenues for Drug Discovery. Curr Top Med Chem 2019; 19:1436-1444. [PMID: 31512997 DOI: 10.2174/1568026619666190712211642] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/17/2019] [Accepted: 01/24/2019] [Indexed: 01/02/2023]
Abstract
Type 2 diabetes is a major health issue worldwide with complex metabolic and endocrine abnormalities. Hyperglycemia, defects in insulin secretion and insulin resistance are classic features of type 2 diabetes. Insulin signaling regulates metabolic homeostasis by regulating glucose and lipid turnover in the liver, skeletal muscle and adipose tissue. Major treatment modalities for diabetes include the drugs from the class of sulfonyl urea, Insulin, GLP-1 agonists, SGLT2 inhibitors, DPP-IV inhibitors and Thiazolidinediones. Emerging antidiabetic therapeutics also include classes of drugs targeting GPCRs in the liver, adipose tissue and skeletal muscle. Interestingly, recent research highlights several shared intermediates between insulin and GPCR signaling cascades opening potential novel avenues for diabetic drug discovery.
Collapse
Affiliation(s)
- Manveen K Gupta
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, Ohio 44106, United States
| | | |
Collapse
|
36
|
Lucy D, Purvis GSD, Zeboudj L, Chatzopoulou M, Recio C, Bataille CJR, Wynne GM, Greaves DR, Russell AJ. A Biased Agonist at Immunometabolic Receptor GPR84 Causes Distinct Functional Effects in Macrophages. ACS Chem Biol 2019; 14:2055-2064. [PMID: 31465201 DOI: 10.1021/acschembio.9b00533] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
GPR84 is an orphan G-protein-coupled receptor that is expressed on immune cells and implicated in several inflammatory diseases. The validation of GPR84 as a therapeutic target is hindered by the narrow range of available chemical tools and consequent poor understanding of GPR84 pathophysiology. Here we describe the discovery and characterization of DL-175, a potent, selective, and structurally novel GPR84 agonist and the first to display significantly biased signaling across GPR84-overexpressing cells, primary murine macrophages, and human U937 cells. By comparing DL-175 with reported GPR84 ligands, we show for the first time that biased GPR84 agonists have markedly different abilities to induce chemotaxis in human myeloid cells, while causing similar levels of phagocytosis enhancement. This work demonstrates that biased agonism at GPR84 enables the selective activation of functional responses in immune cells and delivers a high-quality chemical probe for further investigation.
Collapse
Affiliation(s)
- Daniel Lucy
- Department of Chemistry, University of Oxford, Mansfield Road Oxford OX1 3TA, U.K
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, U.K
| | - Gareth S. D. Purvis
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, U.K
| | - Lynda Zeboudj
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, U.K
| | - Maria Chatzopoulou
- Department of Chemistry, University of Oxford, Mansfield Road Oxford OX1 3TA, U.K
| | - Carlota Recio
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, U.K
| | | | - Graham M. Wynne
- Department of Chemistry, University of Oxford, Mansfield Road Oxford OX1 3TA, U.K
| | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, U.K
| | - Angela J. Russell
- Department of Chemistry, University of Oxford, Mansfield Road Oxford OX1 3TA, U.K
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K
| |
Collapse
|
37
|
GPR40 full agonism exerts feeding suppression and weight loss through afferent vagal nerve. PLoS One 2019; 14:e0222653. [PMID: 31525244 PMCID: PMC6746387 DOI: 10.1371/journal.pone.0222653] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/03/2019] [Indexed: 12/18/2022] Open
Abstract
GPR40/FFAR1 is a Gq protein-coupled receptor expressed in pancreatic β cells and enteroendocrine cells, and mediates insulin and incretin secretion to regulate feeding behavior. Several GPR40 full agonists have been reported to reduce food intake in rodents by regulating gut hormone secretion in addition to their potent glucose-lowering effects; however, detailed mechanisms of feeding suppression are still unknown. In the present study, we characterized T-3601386, a novel compound with potent full agonistic activity for GPR40, by using in vitro Ca2+ mobilization assay in Chinese hamster ovary (CHO) cells expressing FFAR1 and in vivo hormone secretion assay. We also evaluated feeding suppression and weight loss after the administration of T-3601386 and investigated the involvement of the vagal nerve in these effects. T-3601386, but not a partial agonist fasiglifam, increased intracellular Ca2+ levels in CHO cells with low FFAR1 expression, and single dosing of T-3601386 in diet-induced obese (DIO) rats elevated plasma incretin levels, suggesting full agonistic properties of T-3601386 against GPR40. Multiple doses of T-3601386, but not fasiglifam, in DIO rats showed dose-dependent weight loss accompanied by feeding suppression and durable glucagon-like peptide-1 elevation, all of which were completely abolished in Ffar1-/- mice. Immunohistochemical analysis in the nuclei of the solitary tract demonstrated that T-3601386 increased the number of c-Fos positive cells, which also disappeared in Ffar1-/- mice. Surgical vagotomy and drug-induced deafferentation counteracted the feeding suppression and weight loss induced by the administration of T-3601386. These results suggest that T-3601386 exerts incretin release and weight loss in a GPR40-dependent manner, and that afferent vagal nerves are important for the feeding suppression induced by GPR40 full agonism. Our novel findings raise the possibility that GPR40 full agonist can induce periphery-derived weight reduction, which may provide benefits such as less adverse effects in central nervous system compared to centrally-acting anti-obesity drugs.
Collapse
|
38
|
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
|
39
|
Ueno H, Ito R, Abe SI, Ookawara M, Miyashita H, Ogino H, Miyamoto Y, Yoshihara T, Kobayashi A, Tsujihata Y, Takeuchi K, Watanabe M, Yamada Y, Maekawa T, Nishigaki N, Moritoh Y. SCO-267, a GPR40 Full Agonist, Improves Glycemic and Body Weight Control in Rat Models of Diabetes and Obesity. J Pharmacol Exp Ther 2019; 370:172-181. [DOI: 10.1124/jpet.118.255885] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 04/01/2019] [Indexed: 12/14/2022] Open
|
40
|
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
|
41
|
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
|
42
|
Kong D, Guo S, Yang Y, Guo B, Xie X, Hu W. Synthesis and biological evaluation of novel potent FFA1 agonists containing 2,3-dihydrobenzo[b][1,4]dioxine. Bioorg Med Chem Lett 2019; 29:848-852. [PMID: 30685095 DOI: 10.1016/j.bmcl.2019.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/07/2018] [Accepted: 01/15/2019] [Indexed: 10/27/2022]
Abstract
FFA1 (free fatty acid receptor 1) has emerged as an attractive antidiabetic target due to its role in mediating the enhancement of glucose-stimulated insulin secretion in pancreatic β cells with a low risk of hypoglycemia. Many reported FFA1 agonists possessed somewhat pharmacokinetic and/or safety issues. Herein, we describe the identification of 2,3-dihydrobenzo[b][1,4]dioxine as a novel scaffold for FFA1 agonists. Comprehensive structure-activity relationship study based on this scaffold led to the discovery of (S)-3-(4-(((S)-7-(4-methoxyphenyl)-2,3-dihydrobenzo [b][1,4]dioxin-2-yl)methoxy) phenyl)hex-4-ynoic acid (26k), which displayed a potent FFA1 agonistic activity and good pharmacokinetic profiles. Subsequent in vivo studies demonstrated that compound 26k significantly improved the glucose tolerance in ICR mice. In summary, compound 26k is a promising drug candidate for further investigation.
Collapse
Affiliation(s)
- Deyu Kong
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Shimeng Guo
- CAS Key Laboratory of Receptor Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Yushe Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Bin Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China.
| | - Xin Xie
- CAS Key Laboratory of Receptor Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Wenhao Hu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
43
|
Huang H, Meegalla SK, Lanter JC, Winters MP, Zhao S, Littrell J, Qi J, Rady B, Lee PS, Liu J, Martin T, Lam WW, Xu F, Lim HK, Wilde T, Silva J, Otieno M, Pocai A, Player MR. Discovery of a GPR40 Superagonist: The Impact of Aryl Propionic Acid α-Fluorination. ACS Med Chem Lett 2019; 10:16-21. [PMID: 30655940 PMCID: PMC6331191 DOI: 10.1021/acsmedchemlett.8b00444] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/03/2018] [Indexed: 12/11/2022] Open
Abstract
GPR40 is a G-protein-coupled receptor which mediates fatty acid-induced glucose-stimulated insulin secretion from pancreatic beta cells and incretion release from enteroendocrine cells of the small intestine. GPR40 full agonists exhibit superior glucose lowering compared to partial agonists in preclinical species due to increased insulin and GLP-1 secretion, with the added benefit of promoting weight loss. In our search for potent GPR40 full agonists, we discovered a superagonist which displayed excellent in vitro potency and superior efficacy in the Gαs-mediated signaling pathway. Most synthetic GPR40 agonists have a carboxylic acid headgroup, which may cause idiosyncratic toxicities, including drug-induced-liver-injury (DILI). With a methyl group and a fluorine atom substituted at the α-C of the carboxylic acid group, 19 is not only highly efficacious in lowering glucose and body weight in rodent models but also has a low DILI risk due to its stable acylglucuronide metabolite.
Collapse
Affiliation(s)
- Hui Huang
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Sanath K. Meegalla
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - James C. Lanter
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Michael P. Winters
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Shuyuan Zhao
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - James Littrell
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Jenson Qi
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Brian Rady
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Paul S. Lee
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Jianying Liu
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Tonya Martin
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Wing W. Lam
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Fran Xu
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Heng Keang Lim
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Thomas Wilde
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Jose Silva
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Monicah Otieno
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Alessandro Pocai
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| | - Mark R. Player
- Departments
of Medicinal Chemistry, Cardiovascular & Metabolism in Vitro Biology, Cardiovascular &
Metabolism in Vivo Pharmacology, andPreclinical Drug Safety, Janssen Research & Development, Welsh and McKean Roads,Spring House, Pennsylvania 19477-0776, United States
| |
Collapse
|
44
|
Xu F, Zhou H, Liu X, Zhang X, Wang Z, Hou T, Wang J, Qu L, Zhang P, Piao H, Liang X. Label-free cell phenotypic study of FFA4 and FFA1 and discovery of novel agonists of FFA4 from natural products. RSC Adv 2019; 9:15073-15083. [PMID: 35516320 PMCID: PMC9064241 DOI: 10.1039/c9ra02142f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/08/2019] [Indexed: 01/23/2023] Open
Abstract
In this article, pharmacological studies of the free fatty acid receptor (FFA) 4 and FFA1 were conducted in transfected CHO cells (FFA4&FFA1) and HT29 cells with application of a label-free dynamic mass redistribution (DMR) assay. Commercially available compounds including α-linolenic acid (ALA), GW9508, TUG891, GSK137647A, TAK875, MEDICA16, AH7614 and GW1100, were used to validate the assay; real-time tracing of ligand-induced cell responses elucidated pharmacological properties of ligand–receptor interactions. A pool of 140 natural compounds was screened using the CHO-FFA4 cells. Three new FFA4 agonists with novel skeletons were discovered and they were dihydrotanshinone, emodin and acetylshikonin (EC50 values were 32.88, 38.18 and 10.17 μM, respectively). Ligand selectivity was compared between FFA4 and FFA1; dihydrotanshinone and emodin displayed FFA4 selectivity, while acetylshikonin shared FFA1 and FFA4 agonist activities with EC50 values comparable to the endogenous ligand ALA. The three novel FFA4 agonists provide a promising chemical starting point for identification and optimization of drugs used for treating metabolic and inflammatory diseases. Besides, this work will help to explain the mechanism of actions of natural products. Pharmacological studies of the FFA4 and FFA1 and discovery of three novel agonists was conducted using a label-free DMR assay.![]()
Collapse
|
45
|
Wang X, Xu Y, Feng S, Huang X, Meng X, Chen J, Guo L, Ge J, Zhang J, Chen J, Cheng L, Gu K, Zhang Y, Jiang Q, Ning X. A potent free fatty acid receptor 1 agonist with a glucose-dependent antihyperglycemic effect. Chem Commun (Camb) 2019; 55:8975-8978. [DOI: 10.1039/c9cc04040d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PAFA is a promising free fatty acid receptor 1 agonist with a glucose-dependent antihyperglycemic effect, allowing for treating type-2 diabetes.
Collapse
|
46
|
Qiang Y, Zhang X. Pharmacokinetics of TAK‐875 and its toxic metabolite TAK‐875‐ acylglucuronide in rat plasma by liquid chromatography tandem mass spectrometry. Biomed Chromatogr 2018; 33:e4441. [PMID: 30456906 DOI: 10.1002/bmc.4441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/01/2018] [Accepted: 11/12/2018] [Indexed: 01/23/2023]
Affiliation(s)
- Yongzai Qiang
- Department of PharmacyThe Affiliated Hospital of Inner Mongolia Medical University Hohot Inner Mongolia China
| | - Xiaohui Zhang
- Department of PharmacyThe Affiliated Hospital of Inner Mongolia Medical University Hohot Inner Mongolia China
| |
Collapse
|
47
|
Design and optimization of 2,3-dihydrobenzo[b][1,4]dioxine propanoic acids as novel GPR40 agonists with improved pharmacokinetic and safety profiles. Bioorg Med Chem 2018; 26:5780-5791. [DOI: 10.1016/j.bmc.2018.10.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023]
|
48
|
Menon V, Lincoff AM, Nicholls SJ, Jasper S, Wolski K, McGuire DK, Mehta CR, Rosenstock J, Lopez C, Marcinak J, Cao C, Nissen SE. Fasiglifam-Induced Liver Injury in Patients With Type 2 Diabetes: Results of a Randomized Controlled Cardiovascular Outcomes Safety Trial. Diabetes Care 2018; 41:2603-2609. [PMID: 30459247 DOI: 10.2337/dc18-0755] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 08/21/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To evaluate the cardiovascular (CV) safety of fasiglifam, a first-in-man G-protein-coupled receptor 40 (GPR40) agonist, in patients with type 2 diabetes. RESEARCH DESIGN AND METHODS A phase 3 multicenter randomized double-blind placebo-controlled two-arm trial was intended to randomize 5,000 participants with type 2 diabetes at high CV risk to fasiglifam or placebo. The primary objective of the trial was to rule out an upper noninferiority bound >1.3 for a one-sided 97.5% confidence limit of the hazard ratio (HR) for CV composite events during treatment with fasiglifam compared with placebo. The primary outcome was the time to first occurrence of any component of the major adverse CV event composite of CV death, nonfatal myocardial infarction, nonfatal stroke, and hospitalization for unstable angina. RESULTS The study enrolled 3,207 participants but was terminated because of liver safety concerns. Increased rates of liver enzyme elevation (AST/ALT ≥3-5 × upper limit of normal [ULN]) with fasiglifam were observed. The incidence of ALT or AST ≥3 × ULN with fasiglifam compared with placebo was 2.1% vs. 0.5%, P < 0.001, and the incidence for ≥10 × ULN was 0.31% vs. 0.06%, P < 0.001. A primary CV composite outcome occurred in 40 participants, 2.5% each in the fasiglifam and placebo arms at 12 months (HR 1.05; 95% CI 0.67, 1.63). CONCLUSIONS Development of fasiglifam was terminated due to concerns of drug-induced liver injury. Performance of a U.S. Food and Drug Administration-mandated CV outcomes trial supported the termination of the fasiglifam clinical program.
Collapse
Affiliation(s)
- Venu Menon
- Cleveland Clinic Coordinating Center for Clinical Research (C5Research), Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - A Michael Lincoff
- Cleveland Clinic Coordinating Center for Clinical Research (C5Research), Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Stephen J Nicholls
- South Australian Heart and Medical Research Institute, The University of Adelaide, Adelaide, Australia
| | - Susan Jasper
- Cleveland Clinic Coordinating Center for Clinical Research (C5Research), Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Kathy Wolski
- Cleveland Clinic Coordinating Center for Clinical Research (C5Research), Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Darren K McGuire
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX
| | | | | | - Claudia Lopez
- Takeda Development Center Americas, Inc., Deerfield, IL
| | - John Marcinak
- Takeda Development Center Americas, Inc., Deerfield, IL
| | - Charlie Cao
- Takeda Development Center Americas, Inc., Deerfield, IL
| | | | | |
Collapse
|
49
|
Skelin M, Javor E, Lucijanić M, Lucijanić T, Jakupović L, Rahelić D. The role of glucagon in the possible mechanism of cardiovascular mortality reduction in type 2 diabetes patients. Int J Clin Pract 2018; 72:e13274. [PMID: 30295381 DOI: 10.1111/ijcp.13274] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 09/15/2018] [Indexed: 01/10/2023] Open
Abstract
AIM Type 2 diabetes (T2D) is one of the major public health issues worldwide. The main cause of mortality and morbidity among T2D patients are cardiovascular (CV) causes. Various antidiabetics are used in T2D treatment, but until recently they lacked clear evidence of the reduction in CV mortality and all-cause mortality as independent study end-points. The aim of this article was to present and critically evaluate potential mechanisms behind the remarkable results documented in trials with new antidiabetics for the treatment of T2D. METHODS Relevant data were collected using the MEDLINE, PubMed, EMBASE, Web of Science, Science Direct, and Scopus databases with the key words: "type 2 diabetes," "mortality," "glucagon," "empagliflozin," "liraglutide," "insulin" and "QTc." Searches were not limited to specific publication types or study designs. RESULTS The EMPA-REG OUTCOME trial with empagliflozin and LEADER trial with liraglutide presented remarkable results regarding the reduction in mortality in T2D treatment. However, the potential mechanism for those beneficial effects is difficult to determine. It is not likely that improvements in classic CV risk factors are responsible for the observed effect. A potential mechanism may be caused by the elevation of postprandial (PP) glucagon concentrations that can be seen with an empagliflozin and liraglutide therapy, which could have beneficial effects considering the myocardial electrical stability in T2D patients. CONCLUSION This hypothesis throws new light upon possible mechanisms of reduction in mortality in T2D patients.
Collapse
Affiliation(s)
- Marko Skelin
- Department of Pharmacy, General Hospital Šibenik, Šibenik, Croatia
| | - Eugen Javor
- Department of Pharmacy, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Marko Lucijanić
- Department of Hematology, Dubrava University Hospital, Zagreb, Croatia
| | - Tomo Lucijanić
- Department of Endocrinology, Diabetes and Clinical Pharmacology, Dubrava University Hospital, Zagreb, Croatia
| | - Lejsa Jakupović
- Healthcare Institution of Community Pharmacy Slavonski Brod, Slavonski Brod, Croatia
| | - Dario Rahelić
- Department of Endocrinology, Diabetes and Clinical Pharmacology, Dubrava University Hospital, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
50
|
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
|