Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Lipid Res ; 58(8): 1561-1578, 2017 08.
Article in English | MEDLINE | ID: mdl-28583918

ABSTRACT

GPR40 and GPR120 are fatty acid sensors that play important roles in glucose and energy homeostasis. GPR40 potentiates glucose-dependent insulin secretion and demonstrated in clinical studies robust glucose lowering in type 2 diabetes. GPR120 improves insulin sensitivity in rodents, albeit its mechanism of action is not fully understood. Here, we postulated that the antidiabetic efficacy of GPR40 could be enhanced by coactivating GPR120. A combination of GPR40 and GPR120 agonists in db/db mice, as well as a single molecule with dual agonist activities, achieved superior glycemic control compared with either monotherapy. Compared with a GPR40 selective agonist, the dual agonist improved insulin sensitivity in ob/ob mice measured by hyperinsulinemic-euglycemic clamp, preserved islet morphology, and increased expression of several key lipolytic genes in adipose tissue of Zucker diabetic fatty rats. Novel insights into the mechanism of action for GPR120 were obtained. Selective GPR120 activation suppressed lipolysis in primary white adipocytes, although this effect was attenuated in adipocytes from obese rats and obese rhesus, and sensitized the antilipolytic effect of insulin in rat and rhesus primary adipocytes. In conclusion, GPR120 agonism enhances insulin action in adipose tissue and yields a synergistic efficacy when combined with GPR40 agonism.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Experimental/metabolism , Lipolysis , Receptors, G-Protein-Coupled/metabolism , Adipose Tissue/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation/drug effects , Insulin Resistance , Islets of Langerhans/drug effects , Islets of Langerhans/physiopathology , Lipolysis/drug effects , Male , Mice , Rats , Receptors, G-Protein-Coupled/agonists
2.
PLoS One ; 7(11): e49572, 2012.
Article in English | MEDLINE | ID: mdl-23185367

ABSTRACT

Hyperglucagonemia is implicated in the pathophysiology of hyperglycemia. Antagonism of the glucagon receptor (GCGR) thus represents a potential approach to diabetes treatment. Herein we report the characterization of GRA1, a novel small-molecule GCGR antagonist that blocks glucagon binding to the human GCGR (hGCGR) and antagonizes glucagon-induced intracellular accumulation of cAMP with nanomolar potency. GRA1 inhibited glycogenolysis dose-dependently in primary human hepatocytes and in perfused liver from hGCGR mice, a transgenic line of mouse that expresses the hGCGR instead of the murine GCGR. When administered orally to hGCGR mice and rhesus monkeys, GRA1 blocked hyperglycemic responses to exogenous glucagon. In several murine models of diabetes, acute and chronic dosing with GRA1 significantly reduced blood glucose concentrations and moderately increased plasma glucagon and glucagon-like peptide-1. Combination of GRA1 with a dipeptidyl peptidase-4 inhibitor had an additive antihyperglycemic effect in diabetic mice. Hepatic gene-expression profiling in monkeys treated with GRA1 revealed down-regulation of numerous genes involved in amino acid catabolism, an effect that was paralleled by increased amino acid levels in the circulation. In summary, GRA1 is a potent glucagon receptor antagonist with strong antihyperglycemic efficacy in preclinical models and prominent effects on hepatic gene-expression related to amino acid metabolism.


Subject(s)
Gene Expression Regulation , Hypoglycemic Agents/pharmacology , Pyrazoles/pharmacology , Receptors, Glucagon/antagonists & inhibitors , beta-Alanine/analogs & derivatives , Administration, Oral , Animals , Dose-Response Relationship, Drug , Gene Expression Profiling , Glucagon/blood , Glucagon/chemistry , Glycogen/metabolism , Glycogenolysis , Hepatocytes/drug effects , Hormones/blood , Humans , Iodine Radioisotopes/chemistry , Liver/metabolism , Macaca mulatta , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Chemical , beta-Alanine/pharmacology
3.
Bioorg Med Chem Lett ; 20(20): 6088-92, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20832306
4.
Eur J Pharmacol ; 584(1): 192-201, 2008 Apr 14.
Article in English | MEDLINE | ID: mdl-18346728

ABSTRACT

The use of the thiazolidinedione insulin sensitizers rosiglitazone and pioglitazone for the treatment of type 2 diabetes mellitus in recent years has proven to be effective in helping patients resume normal glycemic control. However, their use is often associated with undesirable side effects including peripheral edema, congestive heart failure and weight gain. Here, we report the identification and characterization of a novel selective PPARgamma modulator, SPPARgammaM5 ((2S)-2-(2-chloro-5-{[3-(4-chlorophenoxy)-2-methyl-6-(trifluoromethoxy)-1H-indol-1-yl]methyl} phenoxy)propionic acid), which has notable insulin sensitizing properties and a superior tolerability profile to that of rosiglitazone. SPPARgammaM5 is a potent ligand of human PPARgamma with high selectivity versus PPARalpha or PPARdelta in receptor competitive binding assays. In cell-based transcriptional activation assays, SPPARgammaM5 was a potent partial agonist of human PPARgamma in comparison to the PPARgamma full agonist rosiglitazone. Compared to rosiglitazone or the PPARgamma full agonist COOH (2-(2-(4-phenoxy-2-propylphenoxy)ethyl)indole-5-acetic acid), SPPARgammaM5 induced an attenuated PPARgamma-regulated gene expression profile in fully differentiated 3T3-L1 adipocytes and white adipose tissue of chronically treated db/db mice. SPPARgammaM5 treatment also reduced the insulin resistance index by homeostasis model assessment (HOMA), suggesting an improvement in insulin resistance in these db/db mice. Treatment of obese Zucker rats with either rosiglitazone or SPPARgammaM5 resulted in an improvement in selected parameters that serve as surrogate indicators of insulin resistance and hyperlipidemia. However, unlike rosiglitazone, SPPARgammaM5 did not cause significant fluid retention or cardiac hypertrophy in these rats. Thus, compounds such as SPPARgammaM5 may offer beneficial effects on glycemic control with significantly attenuated adverse effects.


Subject(s)
Acetates/pharmacology , Cardiovascular Diseases/chemically induced , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Indoles/pharmacology , Insulin Resistance , PPAR gamma/drug effects , Propionates/pharmacology , Thiazolidinediones/pharmacology , 3T3-L1 Cells , Acetates/adverse effects , Acetates/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , COS Cells , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Chlorocebus aethiops , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Partial Agonism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hemodilution , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/metabolism , Indoles/adverse effects , Indoles/metabolism , Insulin Resistance/genetics , Male , Mice , Mice, Inbred Strains , PPAR alpha/drug effects , PPAR alpha/metabolism , PPAR delta/drug effects , PPAR delta/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Propionates/adverse effects , Propionates/metabolism , Protein Binding , Rats , Rats, Zucker , Rosiglitazone , Thiazolidinediones/adverse effects , Thiazolidinediones/metabolism , Transcriptional Activation/drug effects , Transfection , Water-Electrolyte Balance/drug effects
5.
Proteomics ; 6(7): 2101-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16518871

ABSTRACT

Here we describe the use of SELDI-MS to detect dose-dependent peptide changes in plasma from mice treated with vehicle or rosiglitazone at one of two doses (10 and 30 mg/kg). SELDI features differentiating spectra from the three conditions were found and used to train classifiers. Samples treated with vehicle could be reliably distinguished from samples treated with either dose, but samples treated with the different doses could not be reliably distinguished from one another. We conclude that while SELDI-TOF mass spectra can be used to distinguish treated from untreated samples, the reproducibility and information content of SELDI-TOF are currently not sufficient as a pharmacodynamic readout to distinguish between mice treated with 10 or 30 mg/kg of rosiglitazone. This raises more general questions about whether SELDI's sensitivity is sufficient for detecting dose-dependent changes in plasma.


Subject(s)
Peptides/blood , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Thiazolidinediones/administration & dosage , Animals , Biomarkers/blood , Dose-Response Relationship, Drug , Male , Mice , Models, Biological , Peptides/analysis , Peptides/chemistry , Reproducibility of Results , Rosiglitazone , Thiazolidinediones/pharmacology
6.
Biochem Biophys Res Commun ; 318(2): 323-8, 2004 May 28.
Article in English | MEDLINE | ID: mdl-15120604

ABSTRACT

Here, we characterize the actions of MK-0767, a dual ligand of the nuclear receptors peroxisome proliferator-activated receptor (PPAR)alpha and PPARgamma. In cell-based assays, MK-0767 produced potent activation of human PPARgamma and PPARalpha with a gamma:alpha potency ratio of approximately 2. The dual agonist induced high affinity interactions of PPARalpha and PPARgamma with the transcriptional coactivator CBP in vitro. In ob/ob mice, MK-0767 normalized hyperglycemia and hyperinsulinemia with equal or greater potency and efficacy than pioglitazone. Treatment of hamsters with MK-0767 produced substantial reductions in blood cholesterol and triglycerides. In dogs, MK-0767 reduced serum cholesterol levels with a potency more than 10-fold greater than simvastatin. The efficacies of MK-0767 and simvastatin were additive when given together. We conclude that MK-0767 is a potent dual PPARalpha/gamma agonist with robust insulin sensitizing and hypolipidemic activities.


Subject(s)
Benzamides/pharmacology , Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Thiazoles/pharmacology , Transcription Factors/agonists , Animals , Benzamides/chemistry , Blood Glucose/analysis , COS Cells , Chlorocebus aethiops , Cholesterol/blood , Cricetinae , Dogs , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Hypoglycemic Agents/chemistry , Hypolipidemic Agents/chemistry , Insulin/blood , Male , Mesocricetus , Mice , Mice, Obese , Pioglitazone , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Simvastatin/pharmacology , Thiazolidinediones/pharmacology , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/drug effects , Triglycerides/blood
7.
J Med Chem ; 47(12): 3255-63, 2004 Jun 03.
Article in English | MEDLINE | ID: mdl-15163205

ABSTRACT

A series of chromane-2-carboxylic acid derivatives was synthesized and evaluated for PPAR agonist activities. A structure-activity relationship was developed toward PPARalpha/gamma dual agonism. As a result, (2R)-7-(3-[2-chloro-4-(4-fluorophenoxy)phenoxy]propoxy)-2-ethylchromane-2-carboxylic acid (48) was identified as a potent, structurally novel, selective PPARalpha/gamma dual agonist. Compound 48 exhibited substantial antihyperglycemic and hypolipidemic activities when orally administered in three different animal models: the db/db mouse type 2 diabetes model, a Syrian hamster lipid model, and a dog lipid model.


Subject(s)
Benzopyrans/chemical synthesis , Chromans/chemical synthesis , Hypoglycemic Agents/chemical synthesis , Hypolipidemic Agents/chemical synthesis , Phenyl Ethers/chemical synthesis , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Animals , Benzopyrans/chemistry , Benzopyrans/pharmacokinetics , Benzopyrans/pharmacology , Chromans/chemistry , Chromans/pharmacokinetics , Chromans/pharmacology , Cricetinae , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Dogs , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/chemistry , Hypolipidemic Agents/pharmacokinetics , Hypolipidemic Agents/pharmacology , Macaca mulatta , Male , Mesocricetus , Mice , Phenyl Ethers/chemistry , Phenyl Ethers/pharmacokinetics , Phenyl Ethers/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Stereoisomerism , Structure-Activity Relationship , Trans-Activators/chemical synthesis , Trans-Activators/chemistry , Trans-Activators/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Endocrinology ; 145(4): 1640-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14701675

ABSTRACT

Patients with type 2 diabetes mellitus exhibit hyperglycemia and dyslipidemia as well as a markedly increased incidence of atherosclerotic cardiovascular disease. Here we report the characterization of a novel arylthiazolidinedione capable of lowering both glucose and lipid levels in animal models. This compound, designated TZD18, is a potent agonist with dual human peroxisome proliferator-activated receptor (PPAR)-alpha/gamma activities. In keeping with its PPARgamma activity, TZD18 caused complete normalization of the elevated glucose in db/db mice and Zucker diabetic fatty rats. TZD18 lowered both cholesterol and triglycerides in hamsters and dogs. TZD18 inhibited cholesterol biosynthesis at steps before mevalonate and reduced hepatic levels of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity. Moreover, TZD18 significantly suppressed gene expression of fatty acid synthesis and induced expression of genes for fatty acid degradation and triglyceride clearance. Studies on 17 additional PPARalpha or PPARalpha/gamma agonists showed that lipid lowering in hamsters correlated with the magnitude of hepatic gene expression changes. Importantly, the presence of PPARgamma agonism did not affect the relationship between hepatic gene expression and lipid lowering. Taken together, these data suggest that PPARalpha/gamma agonists, such as TZD18, affect lipid homeostasis, leading to an antiatherogenic plasma lipid profile. Agents with these properties may provide favorable means for treatment of type 2 diabetes and dyslipidemia and the prevention of atherosclerotic cardiovascular disease.


Subject(s)
Homeostasis/drug effects , Hypolipidemic Agents/pharmacology , Lipid Metabolism , Phenyl Ethers/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Thiazolidinediones/pharmacology , Transcription Factors/agonists , Animals , Blood Glucose/metabolism , COS Cells , Cholesterol/biosynthesis , Cholesterol/blood , Cricetinae , Diabetes Mellitus/blood , Dogs , Gene Expression/drug effects , Humans , Hydroxymethylglutaryl CoA Reductases/drug effects , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipids/blood , Liver/metabolism , Male , Mesocricetus , Mice , Obesity/blood , Phenyl Ethers/chemistry , RNA, Messenger/metabolism , Rats , Rats, Zucker , Thiazolidinediones/chemistry , Triglycerides/antagonists & inhibitors , Triglycerides/blood , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...