Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Bioorg Med Chem ; 25(15): 4175-4193, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28642028

ABSTRACT

Somatostatin receptor subtype 5 (SSTR5) has emerged as a novel attractive drug target for type 2 diabetes mellitus. Starting from N-benzyl azetidine derivatives 1 and 2 as in-house hit compounds, we explored the introduction of a carboxyl group into the terminal benzene of 1 to enhance SSTR5 antagonistic activity by the combination of the substituents at the 3-position of the isoxazoline. Incorporation of a carboxyl group at the 4-position of the benzene ring resulted in a significant enhancement in potency, however, the 4-benzoic acid derivative 10c exhibited moderate human ether-a-go-go related gene (hERG) inhibitory activity. A subsequent optimization study revealed that replacement of the 4-benzoic acid with an isonipecotic acid dramatically reduced hERG inhibition (5.6% inhibition at 30µM) by eliminating π-related interaction with hERG K+ channel, which resulted in the identification of 1-(2-((2,6-diethoxy-4'-fluorobiphenyl-4-yl)methyl)-5-oxa-2,6-diazaspiro[3.4]oct-6-en-7-yl)piperidin-4-carboxylic acid 25a (hSSTR5/mSSTR5 IC50=9.6/57nM). Oral administration of 25a in high-fat diet fed C57BL/6J mice augmented insulin secretion in a glucose-dependent manner and lowered blood glucose concentration.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Receptors, Somatostatin/antagonists & inhibitors , Animals , CHO Cells , Carbon-13 Magnetic Resonance Spectroscopy , Cricetulus , Drug Discovery , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Proton Magnetic Resonance Spectroscopy
2.
Bioorg Med Chem ; 25(15): 4153-4162, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28622905

ABSTRACT

Somatostatin (SST) is a peptide hormone comprising 14 or 28 amino acids that inhibits endocrine and exocrine secretion via five distinct G-protein-coupled receptors (SSTR1-5). SSTR5 has an important role in inhibiting the secretion of pancreatic and gastrointestinal hormones (e.g., insulin, GLP-1, PYY) through the binding of SSTs; hence, SSTR5 antagonists are expected to be novel anti-diabetic drugs. In the course of our lead generation program of SSTR5 antagonists, we have discovered a novel spiroazetidine derivative 3a. However, pharmacological evaluation of 3a revealed that it had to be administered at a high dose (100mg/kg) to show a persistent glucose-lowering effect in an oral glucose tolerance test (OGTT). We therefore initiated an optimization study based on 3a aimed at improving the antagonistic activity and mean residence time (MRT), resulting in the identification of 2-cyclopropyl-5-methoxybiphenyl derivative 3k. However, 3k did not show a sufficient persistent glucose-lowering effect in an OGTT; moreover, hERG inhibition was observed. Hence, further optimization study of the biphenyl moiety of compound 3k, focused on improving the pharmacokinetic (PK) profile and hERG inhibition, was conducted. Consequently, the introduction of a chlorine atom at the 6-position on the biphenyl moiety addressed a putative metabolic soft spot and increased the dihedral angle of the biphenyl moiety, leading to the discovery of 3p with an improved PK profile and hERG inhibition. Furthermore, 3p successfully exhibited a persistent glucose-lowering effect in an OGTT at a dose of 3mg/kg.


Subject(s)
Ether-A-Go-Go Potassium Channels/genetics , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Receptors, Somatostatin/antagonists & inhibitors , Drug Design , Drug Discovery , Glucose Tolerance Test , Humans , Hypoglycemic Agents/chemistry
3.
Bioorg Med Chem Lett ; 22(14): 4769-72, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22677317

ABSTRACT

Spiro-pyrazolidinedione derivatives without quaternary chiral center were discovered by structure-based drug design and characterized as potent acetyl-CoA carboxylase (ACC) inhibitors. The high metabolic stability of the spiro-pyrazolo[1,2-a]pyridazine scaffold and enhancement of the activity by incorporation of a 7-methoxy group on the benzothiophene core successfully led to the identification of compound 4c as an orally bioavailable and highly potent ACC inhibitor. Oral administration of 4c significantly decreased the values of the respiratory quotient in rats, indicating the stimulation of fatty acid oxidation.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Pyrazoles/chemistry , Spiro Compounds/chemistry , Animals , Enzyme Inhibitors/pharmacology , Humans , Microsomes/drug effects , Microsomes/enzymology , Models, Molecular , Molecular Structure , Pyrazoles/pharmacology , Rats , Spiro Compounds/pharmacology , Stereoisomerism , Structure-Activity Relationship
4.
Bioorg Med Chem ; 19(21): 6261-73, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21975069

ABSTRACT

Melanin-concentrating hormone receptor 1 (MCHR1) antagonists have been studied as potential agents for the treatment of obesity. Initial structure-activity relationship studies of in-house hit compound 1a and subsequent optimization studies resulted in the identification of tetrahydroisoquinoline derivative 23, 1-(2-acetyl-1,2,3,4-tetrahydroisoquinolin-7-yl)-4-[4-(4-chlorophenyl)piperidin-1-yl]butan-1-one, as a potent hMCHR1 antagonist. A homology model of hMCHR1 suggests that these compounds interact with Asn 294 and Asp 123 in the binding site of hMCHR1 to enhance binding affinity. Oral administration of compound 23 dose-dependently reduced food intake in diet-induced obesity (DIO)-F344 rats.


Subject(s)
Anti-Obesity Agents/chemistry , Anti-Obesity Agents/pharmacology , Benzazepines/chemistry , Benzazepines/pharmacology , Obesity/drug therapy , Receptors, Pituitary Hormone/antagonists & inhibitors , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Benzazepines/chemical synthesis , Benzazepines/pharmacokinetics , CHO Cells , Cricetinae , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Dynamics Simulation , Obesity/metabolism , Protein Binding , Rats , Rats, Inbred F344 , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
5.
Bioorg Med Chem ; 19(18): 5539-52, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21856163

ABSTRACT

Human melanin-concentrating hormone receptor 1 (hMCHR1) antagonists are promising targets for obesity treatment. We identified the tetrahydronaphthalene derivative 1a with modest binding affinity for hMCHR1 by screening an in-house G protein-coupled receptor (GPCR) ligand library. We synthesized a series of 6-aminomethyl-5,6,7,8-tetrahydronaphthalenes and evaluated their activity as hMCHR1 antagonists. Modification of the biphenylcarbonylamino group revealed that the biphenyl moiety played a crucial role in the interaction of the antagonist with the receptor. The stereoselective effect of the chiral center on binding affinity generated the novel 6-aminomethyl-7,8-dihydronaphthalene scaffold without a chiral center. Optimization of the amino group led to the identification of a potent antagonist 2s (4'-fluoro-N-[6-(1-pyrrolidinylmethyl)-7,8-dihydro-2-naphthalenyl][1,1'-biphenyl]-4-carboxamide), which significantly inhibited the nocturnal food intake in rats after oral administration. Pharmacokinetic analysis confirmed that 2s had good oral bioavailability and brain penetrance. This antagonist appears to be a viable lead compound that can be used to develop a promising therapy for obesity.


Subject(s)
Drug Discovery , Receptors, Somatostatin/antagonists & inhibitors , Tetrahydronaphthalenes/chemical synthesis , Tetrahydronaphthalenes/pharmacology , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Female , Humans , Ligands , Male , Mice , Mice, Inbred Strains , Mice, Obese , Models, Molecular , Molecular Structure , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Tetrahydronaphthalenes/chemistry
6.
J Clin Invest ; 115(12): 3564-72, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322794

ABSTRACT

Ghrelin is the endogenous ligand for the growth hormone secretagogue receptor (GHSR; ghrelin receptor). Since its discovery, accumulating evidence has suggested that ghrelin may play a role in signaling and reversing states of energy insufficiency. For example, ghrelin levels rise following food deprivation, and ghrelin administration stimulates feeding and increases body weight and adiposity. However, recent loss-of-function studies have raised questions regarding the physiological significance of ghrelin in regulating these processes. Here, we present results of a study using a novel GHSR-null mouse model, in which ghrelin administration fails to acutely stimulate food intake or activate arcuate nucleus neurons. We show that when fed a high-fat diet, both female and male GHSR-null mice eat less food, store less of their consumed calories, preferentially utilize fat as an energy substrate, and accumulate less body weight and adiposity than control mice. Similar effects on body weight and adiposity were also observed in female, but not male, GHSR-null mice fed standard chow. GHSR deletion also affected locomotor activity and levels of glycemia. These findings support the hypothesis that ghrelin-responsive pathways are an important component of coordinated body weight control. Moreover, our data suggest that ghrelin signaling is required for development of the full phenotype of diet-induced obesity.


Subject(s)
Diet , Obesity/genetics , Peptide Hormones/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Adipose Tissue/metabolism , Alleles , Analysis of Variance , Animal Feed , Animals , Blood Glucose/metabolism , Blotting, Southern , Blotting, Western , Body Composition , Body Weight , Crosses, Genetic , DNA/metabolism , Female , Gene Deletion , Genetic Predisposition to Disease , Genotype , Ghrelin , Heterozygote , Homeostasis , Hyperglycemia/metabolism , Insulin-Like Growth Factor I/metabolism , Leptin/metabolism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Models, Genetic , Neurons/metabolism , Obesity/metabolism , Peptide Hormones/chemistry , Phenotype , RNA, Messenger/metabolism , Receptors, Ghrelin , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombination, Genetic , Signal Transduction , Silver Staining , Time Factors
7.
SLAS Discov ; 23(2): 154-163, 2018 02.
Article in English | MEDLINE | ID: mdl-28846466

ABSTRACT

Ghrelin O-acyl transferase (GOAT; MBOAT4) catalyzes O-acylation at serine-3 of des-acyl ghrelin. Acyl ghrelin is secreted by stomach X/A-like cells and plays a role in appetite and metabolism. Therefore, GOAT has been expected to be a novel antiobesity target because it is responsible for acyl ghrelin production. Here, we report homogeneous time-resolved fluorescence (HTRF) and enzyme-linked immunosorbent assay (ELISA) methods utilizing human GOAT-expressing microsomes as a novel high-throughput assay system for the discovery of hit compounds and optimization of lead compounds. Hit compounds exemplified by compound A (2-[(2,4-dichlorobenzyl)sulfanyl]-1,3-benzoxazole-5-carboxylic acid) were identified by high-throughput screening using the HTRF assay and confirmed to have GOAT inhibitory activity using the ELISA. Based on the hit compound information, the novel lead compound (compound B, (4-chloro-6-{[2-methyl-6-(trifluoromethyl)pyridin-3-yl]methoxy}-1-benzothiophen-3-yl)acetic acid) was synthesized and exhibited potent GOAT inhibition with oral bioavailability. Both the hit compound and lead compound showed octanoyl-CoA competitive inhibitory activity. Moreover, these two compounds decreased acyl ghrelin production in the stomach of mice after their oral administration. These novel findings demonstrate that GOAT is a druggable target, and its inhibitors are promising antiobesity drugs.


Subject(s)
Acyltransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Ghrelin/metabolism , Acyl Coenzyme A/metabolism , Acylation/drug effects , Administration, Oral , Animals , Biological Availability , Drug Discovery/methods , Enzyme Inhibitors/pharmacokinetics , High-Throughput Screening Assays/methods , Humans , Male , Mice , Mice, Inbred C57BL , Microsomes/drug effects , Microsomes/metabolism , Stomach/drug effects
8.
Magn Reson Insights ; 7: 33-40, 2014.
Article in English | MEDLINE | ID: mdl-25574137

ABSTRACT

The focus of current treatments for obesity is to reduce the body weight or visceral fat, which requires longer duration to show effect. In this study, we investigated the short-term changes in fat metabolism in liver, abdomen, and skeletal muscle during antiobesity interventions including Sibutra mine treatment and diet restriction in obese rats using magnetic resonance imaging, magnetic resonance spectroscopy, and blood chemistry. Sibutramine is an antiobesity drug that results in weight loss by increasing satiety and energy expenditure. The Sibutramine-treated rats showed reduction of liver fat and intramyocellular lipids on day 3. The triglycerides (TG) decreased on day 1 and 3 compared to baseline (day 0). The early response/nonresponse in different fat depots will permit optimization of treatment for better clinical outcome rather than staying with a drug for longer periods.

9.
J Med Chem ; 55(5): 2353-66, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22309223

ABSTRACT

It was found that 3-(aminomethyl)quinoline derivatives showed high binding affinities for melanin-concentrating hormone receptor 1 (MCHR1) with reduced affinity for serotonin receptor 2c (5-HT2c) when the dihydronaphthalene nucleus of compound 1 (human MCHR1, IC(50) = 1.9 nM; human 5-HT2c receptor, IC(50) = 0.53 nM) was replaced by other bicyclic core scaffolds. Among the synthesized compounds, 8-methylquinoline derivative 5v especially showed high binding affinity (IC(50) = 0.54 nM), potent in vitro antagonistic activity (IC(50) = 2.8 nM) for MCHR1, and negligible affinity for 5-HT2c receptor (IC(50) > 1000 nM). Oral administration of 5v significantly and dose-dependently suppressed nocturnal food intake in diet-induced obese rats and did not affect food intake in MCHR1-deficient mice. These results and rat pharmacokinetic study findings suggested that compound 5v is a highly potent, orally bioavailable, and centrally acting nonpeptide MCHR1 antagonist.


Subject(s)
Anti-Obesity Agents/chemical synthesis , Benzamides/chemical synthesis , Quinolines/chemical synthesis , Receptors, Somatostatin/antagonists & inhibitors , Administration, Oral , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Benzamides/pharmacokinetics , Benzamides/pharmacology , Biological Availability , Eating/drug effects , Humans , Mice , Mice, Knockout , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Receptor, Serotonin, 5-HT2C/metabolism , Receptors, Somatostatin/genetics , Structure-Activity Relationship
10.
J Med Chem ; 55(9): 4336-51, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22490048

ABSTRACT

Recently, we discovered 3-aminomethylquinoline derivative 1, a selective, highly potent, centrally acting, and orally bioavailable human MCH receptor 1 (hMCHR1) antagonist, that inhibited food intake in F344 rats with diet-induced obesity (DIO). Subsequent investigation of 1 was discontinued because 1 showed potent hERG K(+) channel inhibition in a patch-clamp study. To decrease hERG K(+) channel inhibition, experiments with ligand-based drug designs based on 1 and a docking study were conducted. Replacement of the terminal p-fluorophenyl group with a cyclopropylmethoxy group, methyl group introduction on the benzylic carbon at the 3-position of the quinoline core, and employment of a [2-(acetylamino)ethyl]amino group as the amine portion eliminated hERG K(+) channel inhibitory activity in a patch-clamp study, leading to the discovery of N-{3-[(1R)-1-{[2-(acetylamino)ethyl]amino}ethyl]-8-methylquinolin-7-yl}-4-(cyclopropylmethoxy)benzamide (R)-10h. The compound (R)-10h showed potent inhibitory activity against hMCHR1 and dose-dependently suppressed food intake in a 2-day study on DIO-F344 rats. Furthermore, practical chiral synthesis of (R)-10h was performed to determine the molecule's absolute configuration.


Subject(s)
Anti-Obesity Agents/chemistry , Anti-Obesity Agents/pharmacology , Benzamides/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Obesity/drug therapy , Quinolines/pharmacology , Receptors, Pituitary Hormone/antagonists & inhibitors , Animals , Anti-Obesity Agents/chemical synthesis , Benzamides/chemical synthesis , Benzamides/chemistry , CHO Cells , Cricetinae , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Inhibitory Concentration 50 , Ligands , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Obesity/genetics , Obesity/metabolism , Quinolines/chemical synthesis , Quinolines/chemistry , Rats , Rats, Inbred F344 , Receptors, Pituitary Hormone/metabolism , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism , Structure-Activity Relationship
11.
Regul Pept ; 155(1-3): 91-8, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19361544

ABSTRACT

Ghrelin is a hormone that influences many physiological processes and behaviors, such as food intake, insulin and growth hormone release, and a coordinated response to chronic stress. However, little is known about the molecular pathways governing ghrelin release and ghrelin cell function. To better study ghrelin cell physiology, we have generated several transgenic mouse lines expressing humanized Renilla reniformis green fluorescent protein (hrGFP) under the control of the mouse ghrelin promoter. hrGFP expression was especially abundant in the gastric oxyntic mucosa, in a pattern mirroring that of ghrelin immunoreactivity and ghrelin mRNA. hrGFP expression also was observed in the duodenum, but not in the brain, pancreatic islet, or testis. In addition, we used fluorescent activated cell sorting (FACS) to collect and partially characterize highly enriched populations of gastric ghrelin cells. We suggest that these novel ghrelin-hrGFP transgenic mice will serve as useful tools to better understand ghrelin cell physiology.


Subject(s)
Ghrelin/metabolism , Animals , Brain/metabolism , Duodenum/metabolism , Flow Cytometry , Gastric Mucosa/metabolism , Ghrelin/blood , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Islets of Langerhans/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL