Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Redox Biol ; 55: 102429, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35961099

ABSTRACT

Mitochondria-targeted H2S donors are thought to protect against acute ischemia-reperfusion (IR) injury by releasing H2S that decreases oxidative damage. However, the rate of H2S release by current donors is too slow to be effective upon administration following reperfusion. To overcome this limitation here we develop a mitochondria-targeted agent, MitoPerSulf that very rapidly releases H2S within mitochondria. MitoPerSulf is quickly taken up by mitochondria, where it reacts with endogenous thiols to generate a persulfide intermediate that releases H2S. MitoPerSulf is acutely protective against cardiac IR injury in mice, due to the acute generation of H2S that inhibits respiration at cytochrome c oxidase thereby preventing mitochondrial superoxide production by lowering the membrane potential. Mitochondria-targeted agents that rapidly generate H2S are a new class of therapy for the acute treatment of IR injury.

2.
Bioorg Med Chem ; 41: 116208, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34010766

ABSTRACT

We undertook an optimization effort involving propan-2-yl 4-({6-[5-(methanesulfonyl)-2,3-dihydro-1H-indol-1-yl]pyrimidin-4-yl}oxy)piperidine-1-carboxylate 1, which we had previously discovered as a novel G protein-coupled receptor 119 (GPR119) agonist. To occupy a presumed hydrophobic space between the pyrimidine and piperidine rings in interaction with GPR119, we replaced the linker oxygen with nitrogen. Subsequently, the introduction of a substituent at the bridging nitrogen atom was explored. We found that the installation of N-trifluoromethyl group 10 not only enhanced GPR119 agonist activity but also considerably improved the human ether-à-go-go-related gene (hERG) inhibition profile. These improvements were not observed for non-fluorinated substituents, such as ethyl analog 8b. The next optimization effort focused on the exploration of a new surrogate structure for the indoline ring and the isosteric replacements of the piperidine N-Boc group to improve solubility, metabolic stability, and oral bioavailability. As a result, N-{1-[3-(2-fluoropropan-2-yl)-1,2,4-oxadiazol-5-yl]piperidin-4-yl}-6-{[1-(methanesulfonyl)piperidin-4-yl]oxy}-N-(trifluoromethyl)pyrimidin-4-amine (27) was identified as a potent and orally bioavailable GPR119 agonist. This compound augmented insulin secretion and effectively lowered plasma glucose excursion in a diabetic animal model after oral administration. In this study, we discuss the designs, syntheses, and biological activities of a novel series of N-(piperidin-4-yl)-N-(trifluoromethyl)pyrimidin-4-amine derivatives as GPR119 agonists, and to determine the distinctive effect of the N-trifluoromethyl group on hERG inhibition, we also discuss the conformational preference of representative compounds.


Subject(s)
Amines/chemistry , Amines/pharmacology , Gene Expression Regulation/drug effects , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Area Under Curve , Blood Glucose , Diabetes Mellitus, Experimental/drug therapy , Drug Design , Drug Discovery , Insulin/metabolism , Molecular Structure , Rats , Receptors, G-Protein-Coupled/genetics
3.
Bioorg Med Chem ; 34: 116034, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33548803

ABSTRACT

We previously identified a novel series of indolinylpyrimidine derivatives exemplified by 2 in Figure 1, which is an indoline based derivative, as potent GPR119 agonists. Despite the attractive potency of 2, this compound inhibited the human ether-a-go-go-related gene (hERG) K+ channel. We elucidated crucial roles of the methylsulfonyl group of 2 in its interaction with the hERG channel and the GPR119 receptor, presumably as a hydrogen bond acceptor (HBA). To remove the undesirable hERG inhibitory activity, a strategy was implemented to arrange an HBA on a less conformationally flexible framework at the indoline 5-position instead of the methylsulfonyl group. This successfully led to the discovery of a piperidinone ring as a desirable motif at the indoline 5-position, which could minimize hERG liability as shown by 24b. Further optimization focused on the reduction of lipophilicity in terms of more favorable drug-like properties. Consequently, the introduction of a hydroxy group at the 3-position of the piperidinone ring effectively reduced lipophilicity without compromising GPR119 potency, resulting in the identification of (3S)-3-hydroxy-1-{1-[6-({1-[3-(propan-2-yl)-1,2,4-oxadiazol-5-yl]piperidin-4-yl}oxy)pyrimidin-4-yl]- 2,3-dihydro-1H-indol-5-yl}piperidin-2-one ((S)-29) as a novel, potent, and orally bioavailable GPR119 agonist with a well-balanced profile. The pharmacological effects of this compound were also confirmed after single and chronic oral administration in diabetic animal models.


Subject(s)
ERG1 Potassium Channel/antagonists & inhibitors , Gene Expression Regulation/drug effects , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Drug Discovery , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , Humans , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rats
4.
Cell ; 181(5): 1176-1187.e16, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32437660

ABSTRACT

Dysfunctional mitochondria accumulate in many human diseases. Accordingly, mitophagy, which removes these mitochondria through lysosomal degradation, is attracting broad attention. Due to uncertainties in the operational principles of conventional mitophagy probes, however, the specificity and quantitativeness of their readouts are disputable. Thorough investigation of the behaviors and fates of fluorescent proteins inside and outside lysosomes enabled us to develop an indicator for mitophagy, mito-SRAI. Through strict control of its mitochondrial targeting, we were able to monitor mitophagy in fixed biological samples more reproducibly than before. Large-scale image-based high-throughput screening led to the discovery of a hit compound that induces selective mitophagy of damaged mitochondria. In a mouse model of Parkinsons disease, we found that dopaminergic neurons selectively failed to execute mitophagy that promoted their survival within lesions. These results show that mito-SRAI is an essential tool for quantitative studies of mitochondrial quality control.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Lysosomes/metabolism , Mitophagy/physiology , Animals , Autophagy/physiology , Fluorescent Antibody Technique/methods , Fluorescent Dyes/chemistry , Humans , Lysosomes/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitophagy/genetics
5.
PLoS One ; 14(9): e0222653, 2019.
Article in English | MEDLINE | ID: mdl-31525244

ABSTRACT

GPR40/FFAR1 is a Gq protein-coupled receptor expressed in pancreatic ß 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.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Weight Loss/physiology , Animals , Blood Glucose/metabolism , Blood Glucose/physiology , CHO Cells , Calcium/metabolism , Cell Line , Cricetulus , Enteroendocrine Cells/metabolism , Female , Glucagon-Like Peptide 1/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Mice , Obesity/metabolism , Obesity/physiopathology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Rats, Wistar , Signal Transduction , Vagus Nerve/metabolism , Vagus Nerve/physiology
6.
J Pharmacol Exp Ther ; 370(2): 172-181, 2019 08.
Article in English | MEDLINE | ID: mdl-31182471

ABSTRACT

The GPR40/FFA1 receptor is a G-protein-coupled receptor expressed in the pancreatic islets and enteroendocrine cells. Here, we report the pharmacological profiles of (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), a novel full agonist of GPR40. Ca2+ signaling and insulin and glucagon-like peptide-1 (GLP-1) secretion were evaluated in GPR40-expressing CHO, MIN6, and GLUTag cells. Hormone secretions and effects on fasting glucose were tested in rats. Single or repeated dosing effects were evaluated in neonatally streptozotocin-induced diabetic rats (N-STZ-1.5 rats), diet-induced obese (DIO) rats, and GPR40-knockout (Ffar1-/- ) mice. Treatment with SCO-267 activated Gq signaling in both high- and low-FFAR1-expressing CHO cells, stimulated insulin secretion in MIN6 cells, and induced GLP-1 release in GLUTag cells. When administered to normal rats, SCO-267 increased insulin, glucagon, GLP-1, glucose-dependent insulinotropic peptide, and peptide YY (PYY) secretions under nonfasting conditions. These results show the full agonistic property of SCO-267 against GPR40. Hypoglycemia was not induced in SCO-267-treated rats during the fasting condition. In diabetic N-STZ-1.5 rats, SCO-267 was highly effective in improving glucose tolerance in single and 2-week dosing studies. DIO rats treated with SCO-267 for 2 weeks showed elevated plasma GLP-1 and PYY levels, reduced food intake, and decreased body weight. In wild-type mice, SCO-267 induced GLP-1 secretion, food intake inhibition, and body weight reduction; however, these effects were abolished in Ffar1-/- mice, indicating a GPR40-dependent mechanism. In conclusion, SCO-267 stimulated islet and gut hormone secretion, improved glycemic control in diabetic rats, and decreased body weight in obese rats. These data suggest the therapeutic potential of SCO-267 for the treatment of diabetes and obesity.


Subject(s)
Blood Glucose/metabolism , Body Weight/drug effects , Cyclopropanes/pharmacology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Obesity/complications , Piperidines/pharmacology , Propionates/pharmacology , Pyridines/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , CHO Cells , Cricetulus , Cyclopropanes/therapeutic use , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Dogs , Eating/drug effects , Glucagon-Like Peptide 1/metabolism , Humans , Insulin Secretion/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Piperidines/therapeutic use , Propionates/therapeutic use , Pyridines/therapeutic use , Rats
7.
EBioMedicine ; 24: 147-158, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28942281

ABSTRACT

Since impaired mitochondrial ATP production in cardiomyocytes is thought to lead to heart failure, a drug that protects mitochondria and improves ATP production under disease conditions would be an attractive treatment option. In this study, we identified small-molecule drugs, including the anti-parasitic agent, ivermectin, that maintain mitochondrial ATP levels under hypoxia in cardiomyocytes. Mechanistically, transcriptomic analysis and gene silencing experiments revealed that ivermectin increased mitochondrial ATP production by inducing Cox6a2, a subunit of the mitochondrial respiratory chain. Furthermore, ivermectin inhibited the hypertrophic response of human induced pluripotent stem cell-derived cardiomyocytes. Pharmacological inhibition of importin ß, one of the targets of ivermectin, exhibited protection against mitochondrial ATP decline and cardiomyocyte hypertrophy. These findings indicate that maintaining mitochondrial ATP under hypoxia may prevent hypertrophy and improve cardiac function, providing therapeutic options for mitochondrial dysfunction.


Subject(s)
Adenosine Triphosphate/metabolism , Cardiotonic Agents/pharmacology , Cell Hypoxia/drug effects , Mitochondria/drug effects , Myocytes, Cardiac/cytology , Small Molecule Libraries/pharmacology , Animals , Cells, Cultured , Electron Transport Complex IV/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Ivermectin/pharmacology , Mice , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , beta Karyopherins/metabolism
8.
Pharmacol Res Perspect ; 4(3): e00237, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27433346

ABSTRACT

Fasiglifam (TAK-875) is a free fatty acid receptor 1 (FFAR1)/G-protein-coupled receptor 40 (GPR40) agonist that improves glycemic control in type 2 diabetes with minimum risk of hypoglycemia. Fasiglifam potentiates glucose-stimulated insulin secretion (GSIS) from pancreatic ß-cells glucose dependently, although the precise mechanism underlying the glucose dependency still remains unknown. Here, we investigated key cross-talk between the GSIS pathway and FFAR1 signaling, and Ca(2+) dynamics using mouse insulinoma MIN6 cells. We demonstrated that the glucose-dependent insulinotropic effect of fasiglifam required membrane depolarization and that fasiglifam induced a glucose-dependent increase in intracellular Ca(2+) level and amplification of Ca(2+) oscillations. This differed from the sulfonylurea glimepiride that induced changes in Ca(2+) dynamics glucose independently. Stimulation with cell-permeable analogs of IP3 or diacylglycerol (DAG), downstream second messengers of Gαq-FFAR1, augmented GSIS similar to fasiglifam, indicating their individual roles in the potentiation of GSIS pathway. Intriguingly, the IP3 analog triggered similar Ca(2+) dynamics to fasiglifam, whereas the DAG analog had no effect. Despite the lack of an effect on Ca(2+) dynamics, the DAG analog elicited synergistic effects on insulin secretion with Ca(2+) influx evoked by an L-type voltage-dependent calcium channel opener that mimics glucose-dependent Ca(2+) dynamics. These results indicate that the Gαq signaling activated by fasiglifam enhances GSIS pathway via dual potentiating mechanisms in which IP3 amplifies glucose-induced Ca(2+) oscillations and DAG/protein kinase C (PKC) augments downstream secretory mechanisms independent of Ca(2+) oscillations.

9.
J Pharmacol Exp Ther ; 357(1): 217-27, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26813930

ABSTRACT

Sulfonylureas (SUs) are widely used insulin secretagogues, but they have adverse effects including hypoglycemia and secondary failure. Fasiglifam/TAK-875, a selective GPR40 agonist, enhances glucose-stimulated insulin secretion and improves hyperglycemia. In the present study, we compared the in vivo glucose-lowering effects of fasiglifam with SUs. The risk of secondary failure of fasiglifam and the efficacy in rats desensitized to SUs were also evaluated. Moreover, we assessed whether fasiglifam was effective when combined with SUs. In diabetic neonatally streptozotocin-induced rats 1.5 days after birth (N-STZ-1.5), oral administrations of fasiglifam (3-30 mg/kg) dose dependently improved glucose tolerance; the effect was greater than that of glibenclamide at maximal effective doses (glucose AUC: fasiglifam, -37.6%; glibenclamide, -12.3%). Although the glucose-lowering effects of glibenclamide (10 mg/kg/day) were completely diminished in N-STZ-1.5 rats after 4 weeks of treatment, effects were maintained in rats receiving fasiglifam (10 mg/kg/day), even after 15 weeks. Fasiglifam (3-10 mg/kg) was still effective in two models desensitized to SUs: 15-week glibenclamide-treated N-STZ-1.5 rats and aged Zucker diabetic fatty (ZDF) rats. Acute administration of fasiglifam (3 mg/kg) and glimepiride (10 mg/kg) in combination additively decreased glucose AUC (fasiglifam, -25.3%; glimepiride, -20.0%; combination, -43.1%). Although glimepiride (10 mg/kg) decreased plasma glucose below normal in nonfasted control rats, fasiglifam (3 mg/kg) maintained normoglycemia, and no further exaggeration of hypoglycemia was observed with combination treatment. These results indicate that GPR40 agonists could be more effective and durable than SUs. Our results also provide new insights into GPR40 pharmacology and rationale for the use of GPR40 agonists in diabetic patients with SU failure.


Subject(s)
Benzofurans/pharmacology , Hyperglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Receptors, G-Protein-Coupled/agonists , Sulfones/pharmacology , Sulfonylurea Compounds/pharmacology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Drug Synergism , Glucose Tolerance Test , Glyburide/therapeutic use , Hyperglycemia/blood , Male , Rats , Rats, Zucker
11.
Bioorg Med Chem ; 22(5): 1649-66, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24508142

ABSTRACT

GPR119 has emerged as an attractive target for anti-diabetic agents. We identified a structurally novel GPR119 agonist 22c that carries a 5-(methylsulfonyl)indoline motif as an early lead compound. To generate more potent compounds of this series, structural modifications were performed mainly to the central alkylene spacer. Installation of a carbonyl group and a methyl group on this spacer significantly enhanced agonistic activity, resulting in the identification of 2-[1-(5-ethylpyrimidin-2-yl)piperidin-4-yl]propyl 7-fluoro-5-(methylsulfonyl)-2,3-dihydro-1H-indole-1-carboxylate (20). To further expand the chemical series of indoline-based GPR119 agonists, several heterocyclic core systems were introduced as surrogates of the carbamate spacer that mimic the presumed active conformation. This approach successfully produced an indolinylpyrimidine derivative 37, 5-(methylsulfonyl)-1-[6-({1-[3-(propan-2-yl)-1,2,4-oxadiazol-5-yl]piperidin-4-yl}oxy)pyrimidin-4-yl]-2,3-dihydro-1H-indole, which has potent GPR119 agonist activity. In rat oral glucose tolerance tests, these two indoline-based compounds effectively lowered plasma glucose excursion and glucose-dependent insulin secretion after oral administration.


Subject(s)
Hypoglycemic Agents/chemistry , Indoles/chemical synthesis , Receptors, G-Protein-Coupled/chemistry , Animals , Glucose Tolerance Test , Indoles/chemistry , Molecular Structure , Rats , Structure-Activity Relationship
12.
PLoS One ; 8(10): e76280, 2013.
Article in English | MEDLINE | ID: mdl-24130766

ABSTRACT

Selective free fatty acid receptor 1 (FFAR1)/GPR40 agonist fasiglifam (TAK-875), an antidiabetic drug under phase 3 development, potentiates insulin secretion in a glucose-dependent manner by activating FFAR1 expressed in pancreatic ß cells. Although fasiglifam significantly improved glycemic control in type 2 diabetes patients with a minimum risk of hypoglycemia in a phase 2 study, the precise mechanisms of its potent pharmacological effects are not fully understood. Here we demonstrate that fasiglifam acts as an ago-allosteric modulator with a partial agonistic activity for FFAR1. In both Ca(2+) influx and insulin secretion assays using cell lines and mouse islets, fasiglifam showed positive cooperativity with the FFAR1 ligand γ-linolenic acid (γ-LA). Augmentation of glucose-induced insulin secretion by fasiglifam, γ-LA, or their combination was completely abolished in pancreatic islets of FFAR1-knockout mice. In diabetic rats, the insulinotropic effect of fasiglifam was suppressed by pharmacological reduction of plasma free fatty acid (FFA) levels using a lipolysis inhibitor, suggesting that fasiglifam potentiates insulin release in conjunction with plasma FFAs in vivo. Point mutations of FFAR1 differentially affected Ca(2+) influx activities of fasiglifam and γ-LA, further indicating that these agonists may bind to distinct binding sites. Our results strongly suggest that fasiglifam is an ago-allosteric modulator of FFAR1 that exerts its effects by acting cooperatively with endogenous plasma FFAs in human patients as well as diabetic animals. These findings contribute to our understanding of fasiglifam as an attractive antidiabetic drug with a novel mechanism of action.


Subject(s)
Benzofurans/pharmacology , Hypoglycemic Agents/pharmacology , Receptors, G-Protein-Coupled/agonists , Sulfones/pharmacology , Allosteric Regulation/drug effects , Animals , Benzofurans/therapeutic use , Cell Line , Cricetinae , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Drug Partial Agonism , Fatty Acids, Nonesterified/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Insulin Secretion , Male , Mice , Mutation , Rats , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sulfones/therapeutic use , gamma-Linolenic Acid/metabolism
13.
J Med Chem ; 55(8): 3960-74, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22490067

ABSTRACT

G protein-coupled receptor 40 (GPR40)/free fatty acid receptor 1 (FFA1) is a free fatty acid (FFA) receptor that mediates FFA-amplified glucose-stimulated insulin secretion in pancreatic ß-cells. We previously identified (2,3-dihydro-1-benzofuran-3-yl)acetic acid derivative 2 as a candidate, but it had relatively high lipophilicity. Adding a polar functional group on 2 yielded several compounds with lower lipophilicity and little effect on caspase-3/7 activity at 30 µM (a marker of toxicity in human HepG2 hepatocytes). Three optimized compounds showed promising pharmacokinetic profiles with good in vivo effects. Of these, compound 16 had the lowest lipophilicity. Metabolic analysis of 16 showed a long-acting PK profile due to high resistance to ß-oxidation. Oral administration of 16 significantly reduced plasma glucose excursion and increased insulin secretion during an OGTT in type 2 diabetic rats. Compound 16 (TAK-875) is being evaluated in human clinical trials for the treatment of type 2 diabetes.


Subject(s)
Benzofurans/pharmacology , Glucose/pharmacology , Insulin/metabolism , Receptors, G-Protein-Coupled/agonists , Sulfones/pharmacology , Animals , Benzofurans/chemical synthesis , Benzofurans/pharmacokinetics , Diabetes Mellitus, Type 2/drug therapy , Female , Glucose Tolerance Test , Hep G2 Cells , Humans , Insulin Secretion , Male , Rats , Receptors, G-Protein-Coupled/drug effects , Stereoisomerism , Sulfones/chemical synthesis , Sulfones/pharmacokinetics
14.
J Med Chem ; 55(8): 3756-76, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22428944

ABSTRACT

As part of a program to identify potent GPR40 agonists with drug-like properties suitable for clinical development, the incorporation of polar substituents was explored with the intention of decreasing the lipophilicity of our recently disclosed phenylpropanoic acid derivative 1. This incorporation would allow us to mitigate the cytotoxicity issues observed with compound 1 and enable us to move away from the multifunctional free fatty acid-like structure. Substitutions on the 2',6'-dimethylbiphenyl ring were initially undertaken, which revealed the feasibility of introducing polar functionalities at the biphenyl 4'-position. Further optimization of this position and the linker led to the discovery of several 4'-alkoxybiphenyl derivatives, which showed potent GPR40 agonist activities with the best balance in terms of improved cytotoxicity profiles and favorable pharmacokinetic properties. Among them, 3-{2-fluoro-4-[({4'-[(4-hydroxy-1,1-dioxidotetrahydro-2H-thiopyran-4-yl)methoxy]-2',6'-dimethylbiphenyl-3-yl}methyl)amino]phenyl}propanoic acid (35) exhibited a robust plasma glucose-lowering effect and insulinotropic action during an oral glucose tolerance test in rats with impaired glucose tolerance.


Subject(s)
Cyclic S-Oxides/chemical synthesis , Hypoglycemic Agents/chemical synthesis , Phenylpropionates/chemical synthesis , Receptors, G-Protein-Coupled/agonists , Animals , CHO Cells , Calcium/metabolism , Caspases/metabolism , Cell Survival , Cricetinae , Cyclic S-Oxides/pharmacokinetics , Cyclic S-Oxides/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Female , Glucose Intolerance/drug therapy , Hep G2 Cells , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Inhibitory Concentration 50 , Male , Phenylpropionates/pharmacokinetics , Phenylpropionates/therapeutic use , Rats , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
15.
J Med Chem ; 55(4): 1538-52, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22242551

ABSTRACT

The G protein-coupled receptor 40 (GPR40)/free fatty acid receptor 1 (FFA1) has emerged as an attractive target for a novel insulin secretagogue with glucose dependency. We previously identified phenylpropanoic acid derivative 1 (3-{4-[(2',6'-dimethylbiphenyl-3-yl)methoxy]-2-fluorophenyl}propanoic acid) as a potent and orally available GPR40/FFA1 agonist; however, 1 exhibited high clearance and low oral bioavailability, which was likely due to its susceptibility to ß-oxidation at the phenylpropanoic acid moiety. To identify long-acting compounds, we attempted to block the metabolically labile sites at the phenylpropanoic acid moiety by introducing a fused-ring structure. Various fused-ring alkanoic acids with potent GPR40/FFA1 activities and good PK profiles were produced. Further optimizations of the lipophilic portion and the acidic moiety led to the discovery of dihydrobenzofuran derivative 53 ((6-{[4'-(2-ethoxyethoxy)-2',6'-dimethylbiphenyl-3-yl]methoxy}-2,3-dihydro-1-benzofuran-3-yl)acetic acid), which acted as a GPR40/FFA1 agonist with in vivo efficacy during an oral glucose tolerance test (OGTT) in rats with impaired glucose tolerance.


Subject(s)
Acetates/chemical synthesis , Acids, Acyclic/chemical synthesis , Benzofurans/chemical synthesis , Insulin/blood , Receptors, G-Protein-Coupled/agonists , Acetates/pharmacokinetics , Acetates/pharmacology , Acids, Acyclic/pharmacokinetics , Acids, Acyclic/pharmacology , Animals , Benzofurans/pharmacokinetics , Benzofurans/pharmacology , CHO Cells , Cricetinae , Cricetulus , Female , Glucose Intolerance/metabolism , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin Secretion , Models, Molecular , Protein Binding , Rats , Rats, Wistar , Species Specificity
16.
J Pharmacol Exp Ther ; 340(2): 483-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22106100

ABSTRACT

G protein-coupled receptor 40 (GPR40)/free fatty acid 1 (FFA1) is a G protein-coupled receptor involved in free fatty acid-induced insulin secretion. To analyze the effect of our novel GPR40/FFA1-selective agonist, [(3S)-6-({2',6'-dimethyl-4'-[3-(methylsulfonyl)propoxy]biphenyl-3-yl}methoxy)-2,3-dihydro-1-benzofuran-3-yl]acetic acid hemi-hydrate (TAK-875), on insulin and glucagon secretion, we performed hormone secretion assays and measured intracellular Ca²âº concentration ([Ca²âº](i)) in both human and rat islets. Insulin and glucagon secretion were measured in static and dynamic conditions by using groups of isolated rat and human pancreatic islets. [Ca²âº](i) was recorded by using confocal microscopy. GPR40/FFA1 expression was measured by quantitative polymerase chain reaction. In both human and rat islets, TAK-875 enhanced glucose-induced insulin secretion in a glucose-dependent manner. The stimulatory effect of TAK-875 was similar to that produced by glucagon-like peptide-1 and correlated with the elevation of ß-cell [Ca²âº](i). TAK-875 was without effect on glucagon secretion at both 1 and 16 mM glucose in human islets. These data indicate that GPR40/FFA1 influences mainly insulin secretion in a glucose-dependent manner. The ß-cell-specific action of TAK-875 in human islets may represent a therapeutically useful feature that allows plasma glucose control without compromising counter-regulation of glucagon secretion, thus minimizing the risk of hypoglycemia.


Subject(s)
Benzofurans/pharmacology , Glucagon/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Receptors, G-Protein-Coupled/metabolism , Sulfones/pharmacology , ATP-Binding Cassette Transporters/genetics , Animals , Calcium Signaling/drug effects , Drug Synergism , Gene Expression/drug effects , Gene Expression/genetics , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Glucose/pharmacology , Humans , In Vitro Techniques , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Potassium Channels, Inwardly Rectifying/genetics , Rats , Rats, Sprague-Dawley , Receptors, Drug/genetics , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, Glucagon/genetics , Sulfonylurea Receptors
17.
J Pharmacol Exp Ther ; 339(1): 228-37, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21752941

ABSTRACT

G protein-coupled receptor 40/free fatty acid receptor 1 (GPR40/FFA(1)) is highly expressed in pancreatic ß cells and mediates free fatty acid-induced insulin secretion. This study examined the pharmacological effects and potential for avoidance of lipotoxicity of [(3S)-6-({2',6'-dimethyl-4'-[3-(methylsulfonyl)propoxy]biphenyl-3-yl}meth-oxy)-2,3-dihydro-1-benzofuran-3-yl]acetic acid hemi-hydrate) (TAK-875), a novel, orally available, selective GPR40 agonist. Insulinoma cell lines and primary rat islets were used to assess the effects of TAK-875 in vitro. The in vivo effects of TAK-875 on postprandial hyperglycemia, fasting hyperglycemia, and normoglycemia were examined in type 2 diabetic and normal rats. In rat insulinoma INS-1 833/15 cells, TAK-875 increased intracellular inositol monophosphate and calcium concentration, consistent with activation of the Gqα signaling pathway. The insulinotropic action of TAK-875 (10 µM) in INS-1 833/15 and primary rat islets was glucose-dependent. Prolonged exposure of cytokine-sensitive INS-1 832/13 to TAK-875 for 72 h at pharmacologically active concentrations did not alter glucose-stimulated insulin secretion, insulin content, or caspase 3/7 activity, whereas prolonged exposure to palmitic or oleic acid impaired ß cell function and survival. In an oral glucose tolerance test in type 2 diabetic N-STZ-1.5 rats, TAK-875 (1-10 mg/kg p.o.) showed a clear improvement in glucose tolerance and augmented insulin secretion. In addition, TAK-875 (10 mg/kg, p.o.) significantly augmented plasma insulin levels and reduced fasting hyperglycemia in male Zucker diabetic fatty rats, whereas in fasted normal Sprague-Dawley rats, TAK-875 neither enhanced insulin secretion nor caused hypoglycemia even at 30 mg/kg. TAK-875 enhances glucose-dependent insulin secretion and improves both postprandial and fasting hyperglycemia with a low risk of hypoglycemia and no evidence of ß cell toxicity.


Subject(s)
Benzofurans/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Glucose/physiology , Hyperglycemia/drug therapy , Hypoglycemic Agents , Insulin/metabolism , Receptors, G-Protein-Coupled/agonists , Sulfones/pharmacology , Animals , Blood Glucose/metabolism , CHO Cells , Calcium/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cell Survival/drug effects , Cricetinae , Cricetulus , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/blood , Fasting/physiology , Hyperglycemia/blood , Insulin Resistance/physiology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Postprandial Period/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
18.
J Med Chem ; 54(5): 1365-78, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21319751

ABSTRACT

G protein-coupled receptor 40 (GPR40) is being recently considered to be a new potential drug target for the treatment of type 2 diabetes because of its role in the enhancement of free fatty acid-regulated glucose-stimulated insulin secretion in pancreatic ß-cells. We initially identified benzyloxyphenylpropanoic acid (1b) (EC(50) = 510 nM), which was designed based on the structure of free fatty acids, as a promising lead compound with GPR40 agonist activity. Chemical modification of compound 1b led to the discovery of 3-{4-[(2',6'-dimethylbiphenyl-3-yl)methoxy]-2-fluorophenyl}propanoic acid (4p) as a potent GPR40 agonist (EC(50) = 5.7 nM). Compound 4p exhibited acceptable pharmacokinetic profiles and significant glucose-lowering effects during an oral glucose tolerance test in diabetic rats. Moreover, no hypoglycemic event was observed even after administration of a high dose of compound 4p to normal fasted rats. These pharmacological results suggest that GPR40 agonists might be novel glucose-dependent insulin secretagogues with little or no risk of hypoglycemia.


Subject(s)
Biphenyl Compounds/chemical synthesis , Hypoglycemic Agents/chemical synthesis , Phenylpropionates/chemical synthesis , Receptors, G-Protein-Coupled/agonists , Administration, Oral , Animals , Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/pharmacology , Blood Glucose/analysis , CHO Cells , Cricetinae , Cricetulus , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Drug Design , Glucose Tolerance Test , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Male , Phenylpropionates/pharmacokinetics , Phenylpropionates/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship
19.
ACS Med Chem Lett ; 1(6): 290-4, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-24900210

ABSTRACT

GPR40, one of the G protein-coupled receptors predominantly expressed in pancreatic ß-cells, mediates enhancement of glucose-stimulated insulin secretion by free fatty acids. A potent and selective GPR40 agonist is theorized to be a safe and effective antidiabetic drug with little or no risk of hypoglycemia. Cyclization of the phenylpropanoic acid moiety of lead compound 1 produced fused phenylalkanoic acids with favorable in vitro agonist activities and pharmacokinetic profiles. Further optimization led to the discovery of dihydrobenzofuran derivative 9a ([(3S)-6-({2',6'-dimethyl-4'-[3-(methylsulfonyl)propoxy]biphenyl-3-yl}methoxy)-2,3-dihydro-1-benzofuran-3-yl]acetic acid hemi-hydrate, TAK-875) as a potent, selective, and orally bioavailable GPR40 agonist, with a pharmacokinetic profile enabling long-acting drug efficacy. Compound 9a showed potent plasma glucose-lowering action and insulinotropic action during an oral glucose tolerance test in female Wistar fatty rats with impaired glucose tolerance. Compound 9a is currently in clinical trials for the treatment of type 2 diabetes mellitus.

20.
Immunology ; 107(4): 394-402, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12460183

ABSTRACT

We previously found that autoantibodies against mouse lysozyme (ML) were strongly induced in normal BALB/c mice when immunized with mutant ML that has triple mutations rendering the dominant T-cell epitope of hen egg lysozyme (HEL), HEL 107-116. As T cells specific for HEL 107-116 were primed in these mice, the anti-ML immunoglobulin G (IgG) responses would be the result of collaborations between autoreactive B cells specific for ML and T cells specific for HEL 107-116. Serum IgG responses against ML were dominantly focused on the ML 14-69 region, indicating that B cells responding to the epitope escape tolerance. In the present study, we prepared several monoclonal antibodies (mAbs) specific for ML 14-69 and examined their antigen specificities in detail, to characterize the nature of the remaining B-cell repertoire specific for ML. mAbs specific for ML 14-69 interacted weakly with soluble, native ML, but the interactions were strengthened by denaturation of ML. The apparent affinity constants between these mAbs and ML showed an increase, ranging from six- to 80-fold, by denaturation of ML. Therefore, these mAbs were more specific for the denatured determinant than for the determinant in the native structure. These results indicate that a substantial number of autoreactive B cells, specific for the unfolded conformation of ML, escape tolerance and are dominantly involved in the autoantibody response to ML. Our finding provides important information to understand the naturally occurring autoreactive B-cell repertoire in normal mice.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/immunology , Muramidase/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Antibody Affinity/immunology , Antibody Specificity/immunology , Epitopes/immunology , Female , Immune Tolerance/immunology , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred BALB C , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL