Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Mol Immunol ; 18(6): 1463-1475, 2021 06.
Article in English | MEDLINE | ID: mdl-31797905

ABSTRACT

His-tRNA synthetase (HARS) is targeted by autoantibodies in chronic and acute inflammatory anti-Jo-1-positive antisynthetase syndrome. The extensive activation and migration of immune cells into lung and muscle are associated with interstitial lung disease, myositis, and morbidity. It is unknown whether the sequestration of HARS is an epiphenomenon or plays a causal role in the disease. Here, we show that HARS circulates in healthy individuals, but it is largely undetectable in the serum of anti-Jo-1-positive antisynthetase syndrome patients. In cultured primary human skeletal muscle myoblasts (HSkMC), HARS is released in increasing amounts during their differentiation into myotubes. We further show that HARS regulates immune cell engagement and inhibits CD4+ and CD8+ T-cell activation. In mouse and rodent models of acute inflammatory diseases, HARS administration downregulates immune activation. In contrast, neutralization of extracellular HARS by high-titer antibody responses during tissue injury increases susceptibility to immune attack, similar to what is seen in humans with anti-Jo-1-positive disease. Collectively, these data suggest that extracellular HARS is homeostatic in normal subjects, and its sequestration contributes to the morbidity of the anti-Jo-1-positive antisynthetase syndrome.


Subject(s)
Histidine-tRNA Ligase/blood , Immunity , Organ Specificity , Animals , Autoantibodies/blood , Case-Control Studies , Cell Differentiation/drug effects , Disease Models, Animal , Female , Histidine-tRNA Ligase/immunology , Humans , Immunity/drug effects , Immunomodulation/drug effects , Insulin-Like Growth Factor I/pharmacology , Lung/drug effects , Lung/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice, Inbred C57BL , Middle Aged , Muscle Cells/drug effects , Muscle Cells/enzymology , Muscles/drug effects , Muscles/pathology , Myositis/blood , Myositis/diagnostic imaging , Myositis/immunology , Organ Specificity/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tomography, X-Ray Computed
2.
Nucleic Acids Res ; 43(19): 9123-32, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26446989

ABSTRACT

The in vivo potency of antisense oligonucleotides (ASO) has been significantly increased by reducing their length to 8-15 nucleotides and by the incorporation of high affinity RNA binders such as 2', 4'-bridged nucleic acids (also known as locked nucleic acid or LNA, and 2',4'-constrained ethyl [cET]). We now report the development of a novel ASO design in which such short ASO monomers to one or more targets are co-synthesized as homo- or heterodimers or multimers via phosphodiester linkers that are stable in plasma, but cleaved inside cells, releasing the active ASO monomers. Compared to current ASOs, these multimers and multi-targeting oligonucleotides (MTOs) provide increased plasma protein binding and biodistribution to liver, and increased in vivo efficacy against single or multiple targets with a single construct. In vivo, MTOs synthesized in both RNase H-activating and steric-blocking oligonucleotide designs provide ≈4-5-fold increased potency and ≈2-fold increased efficacy, suggesting broad therapeutic applications.


Subject(s)
Oligonucleotides, Antisense/chemistry , Animals , Apolipoprotein C-III/genetics , Apolipoprotein C-III/metabolism , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Dimerization , Female , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , Oligonucleotides, Antisense/pharmacokinetics , Oligonucleotides, Antisense/pharmacology , Tissue Distribution
3.
J Pharmacokinet Pharmacodyn ; 41(2): 127-39, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24578187

ABSTRACT

Human Hexokinase IV, or glucokinase (GK), is a regulator of glucose concentrations in the body. It plays a key role in pancreatic insulin secretion as well as glucose biotransformation in the liver, making it a potentially viable target for treatment of Type 2 diabetes. Allosteric activators of GK have been shown to decrease blood glucose concentrations in both animals and humans. Here, the development of a mathematical model is presented that describes glucose modulation in an ob/ob mouse model via administration of a potent GK activator, with the goal of projecting a human efficacious dose and plasma exposure. The model accounts for the allosteric interaction between GK, the activator, and glucose using a modified Hill function. Based on model simulations using data from the ob/ob mouse and in vitro studies, human projections of glucose response to the GK activator are presented, along with dose and regimen predictions to maintain clinically significant decreases in blood glucose in a Type 2 diabetic patient. This effort serves as a basis to build a detailed mechanistic understanding of GK and its role as a therapeutic target for Type 2 diabetes, and it highlights the benefits of using such an approach in a drug discovery setting.


Subject(s)
Azetidines/pharmacology , Benzofurans/pharmacology , Diabetes Mellitus, Type 2/metabolism , Enzyme Activators/pharmacokinetics , Glucokinase/metabolism , Hypoglycemic Agents/pharmacokinetics , Models, Biological , Animals , Azetidines/pharmacokinetics , Azetidines/therapeutic use , Benzofurans/pharmacokinetics , Benzofurans/therapeutic use , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Enzyme Activators/therapeutic use , Humans , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Obese
4.
J Pharmacol Exp Ther ; 346(2): 270-80, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23720456

ABSTRACT

Fibroblast growth factor (FGF)21 improves insulin sensitivity, reduces body weight, and reverses hepatic steatosis in preclinical species. We generated long-acting FGF21 mimetics by site-specific conjugation of the protein to a scaffold antibody. Linking FGF21 through the C terminus decreased bioactivity, whereas bioactivity was maintained by linkage to selected internal positions. In mice, these CovX-Bodies retain efficacy while increasing half-life up to 70-fold compared with wild-type FGF21. A preferred midlinked CovX-Body, CVX-343, demonstrated enhanced in vivo stability in preclinical species, and a single injection improved glucose tolerance for 6 days in ob/ob mice. In diet-induced obese mice, weekly doses of CVX-343 reduced body weight, blood glucose, and lipids levels. In db/db mice, CVX-343 increased glucose tolerance, pancreatic ß-cell mass, and proliferation. CVX-343, created by linkage of the CovX scaffold antibody to the engineered residue A129C of FGF21 protein, demonstrated superior preclinical pharmacodynamics by extending serum half-life of FGF21 while preserving full therapeutic functionality.


Subject(s)
Antibodies/chemistry , Fibroblast Growth Factors/chemistry , Hypoglycemic Agents/chemistry , 3T3-L1 Cells , Animals , Body Weight/drug effects , Cysteine/chemistry , Delayed-Action Preparations , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Energy Metabolism/drug effects , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Immunoglobulin Fab Fragments/chemistry , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Lysine/chemistry , Macaca fascicularis , Male , Mice , Mice, Obese , Molecular Mimicry , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry
5.
Bioorg Med Chem Lett ; 21(22): 6773-7, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21974949

ABSTRACT

A novel series of potent inhibitors of glucosylceramide synthase are described. The optimization of biochemical and cellular potency as well as ADME properties led to compound 23c. Broad tissue distribution was obtained following oral administration to mice. Thus 23c could be another useful tool compound for studying the effects of GCS inhibition in vitro and in vivo.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Administration, Oral , Animals , Drug Discovery , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Humans , Mice , Mice, Inbred C57BL , Structure-Activity Relationship
6.
Am J Physiol Endocrinol Metab ; 295(5): E1142-51, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18728225

ABSTRACT

c-Jun NH(2)-terminal kinase (JNK) plays an important role in insulin resistance; however, identification of pharmacologically potent and selective small molecule JNK inhibitors has been limited. Compound A has a cell IC(50) of 102 nM and is at least 100-fold selective against related kinases and 27-fold selective against glycogen synthase kinase-3beta and cyclin-dependent kinase-2. In C57BL/6 mice, compound A reduced LPS-mediated increases in both plasma cytokine levels and phosphorylated c-Jun in adipose tissue. Treatment of mice fed a high-fat diet with compound A for 3 wk resulted in a 13.1 +/- 1% decrease in body weight and a 9.3 +/- 1.5% decrease in body fat, compared with a 6.6 +/- 2.1% increase in body weight and a 6.7 +/- 2.1% increase in body fat in vehicle-treated mice. Mice pair fed to those that received compound A exhibited a body weight decrease of 7 +/- 1% and a decrease in body fat of 1.6 +/- 1.3%, suggesting that reductions in food intake could not account solely for the reductions in adiposity observed. Compound A dosed at 30 mg/kg for 13 days in high-fat fed mice resulted in a significant decrease in phosphorylated c-Jun in adipose tissue accompanied by a decrease in weight and reductions in glucose and triglycerides and increases in insulin sensitivity to levels comparable with those in lean control mice. The ability of compound A to reduce the insulin-stimulated phosphorylation of insulin receptor substrate-1 (IRS-1) von Ser307 and partially reverse the free fatty acid inhibition of glucose uptake in 3T3L1 adipocytes, suggests that enhancement of insulin signaling in addition to weight loss may contribute to the effects of compound A on insulin sensitization in vivo. Pharmacological inhibition of JNK using compound A may therefore offer an effective therapy for type 2 diabetes mediated at least in part via weight reduction.


Subject(s)
Aminopyridines/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , 3T3-L1 Cells , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Aminopyridines/pharmacokinetics , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Cytokines/blood , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Eating/drug effects , Humans , Insulin/blood , Insulin/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/metabolism , Obesity/drug therapy , Obesity/etiology , Obesity/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-jun/metabolism , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
7.
Biochem Biophys Res Commun ; 365(4): 740-5, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-18029262

ABSTRACT

The inhibition of 11betahydroxysteroid dehydrogenase 1 (11betaHSD1), an enzyme that catalyzes the conversion of inactive cortisone to active cortisol, is an attractive target to treat diabetes by suppressing hepatic gluconeogenesis. To test this hypothesis, we developed a novel glucocorticoid-induced diabetic KK mouse model and used 11betaHSD1 antisense oligonucleotide (ASO) as an inhibitory tool. KK mice were treated with 25 or 50mg/kg/day of 11betaHSD1 ASO for 28 days. On day 25, cortisone pellets were surgically implanted to induce diabetes. In the ASO-treated mice, plasma blood glucose levels were significantly reduced by up to 54%. In parallel, cortisol and other diabetes endpoints were also significantly reduced. Hepatic 11betaHSD1 mRNA was suppressed by up to 84% with a concomitant respective decrease of up to 49% in the expression of PEPCK. The results suggest that inhibition of 11betaHSD1 activity reduces the availability of cortisol to activate the glucocorticoid receptor, down regulates gluconeogenesis and thus reduces plasma glucose levels in cortisone-induced diabetic KK mice.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Cortisone , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Disease Models, Animal , Gene Silencing , Genetic Therapy/methods , Oligonucleotides, Antisense/administration & dosage , Animals , Diabetes Mellitus/chemically induced , Diabetes Mellitus/pathology , Gene Targeting , Male , Mice , Mice, Inbred C57BL , Treatment Outcome
8.
Nat Rev Drug Discov ; 6(10): 793-810, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17906642

ABSTRACT

Retinoic acid receptors (RARs) are ligand-controlled transcription factors that function as heterodimers with retinoid X receptors (RXRs) to regulate cell growth and survival. The success of RAR modulation in the treatment of acute promyelocytic leukaemia (APL) has stimulated considerable interest in the development of RAR and RXR modulators. This has been aided by recent advances in the understanding of the biological role of RARs and RXRs and in the design of selective receptor modulators that might overcome the limitations of current drugs. Here, we discuss the challenges and opportunities for therapeutic strategies based on RXR and RAR modulators, with a focus on cancer and metabolic diseases such as diabetes and obesity.


Subject(s)
Diabetes Mellitus/drug therapy , Neoplasms/drug therapy , Obesity/drug therapy , Receptors, Retinoic Acid/metabolism , Retinoids , Clinical Trials as Topic , Diabetes Mellitus/metabolism , Humans , Ligands , Neoplasms/metabolism , Obesity/metabolism , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Retinoid X Receptors/agonists , Retinoid X Receptors/antagonists & inhibitors , Retinoid X Receptors/metabolism , Retinoids/chemical synthesis , Retinoids/chemistry , Retinoids/therapeutic use
9.
Bioorg Med Chem Lett ; 16(23): 6116-9, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16979341

ABSTRACT

A series of novel pyridine-2-propanoic acids was synthesized. A structure-activity relationship study of these compounds led to the identification of potent dual PPARalpha/gamma agonists with varied isoform selectivity. Based on the results of efficacy studies in diabetic (db/db) mice, and the desired pharmacokinetic parameters, compound (S)-13 was selected for further profiling.


Subject(s)
Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacology , PPAR alpha/agonists , PPAR gamma/agonists , Pyridines/chemistry , Pyridines/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Cell Line, Tumor , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Male , Mice , Molecular Structure , PPAR alpha/metabolism , PPAR gamma/metabolism , Pyridines/chemical synthesis , Pyridines/therapeutic use , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 16(23): 6120-3, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16973358

ABSTRACT

A series of novel pyridine-3-propanoic acids was synthesized. A structure-activity relationship study of these compounds led to the identification of potent dual PPARalpha/gamma agonists with varied isoform selectivity. Based on the results of efficacy studies in diabetic (db/db) mice, and the desired pharmacokinetic parameters, compounds (S)-14 and (S)-19 were selected for further profiling.


Subject(s)
Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacology , PPAR alpha/agonists , PPAR gamma/agonists , Pyridines/blood , Pyridines/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Cell Line, Tumor , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Ether/chemistry , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Mice , Molecular Structure , PPAR alpha/metabolism , PPAR gamma/metabolism , Pyridines/chemical synthesis , Pyridines/therapeutic use , Structure-Activity Relationship , Thiazolidinediones/chemistry
11.
Endocrinology ; 147(2): 1044-53, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16269450

ABSTRACT

Specific retinoid X receptor (RXR) agonists, such as LG100268 (LG268), and the thiazolidinedione (TZD) PPARgamma agonists, such as rosiglitazone, produce insulin sensitization in rodent models of insulin resistance and type 2 diabetes. In sharp contrast to the TZDs that produce significant increases in body weight gain, RXR agonists reduce body weight gain and food consumption. Unfortunately, RXR agonists also suppress the thyroid hormone axis and generally produce hypertriglyceridemia. Heterodimer-selective RXR modulators have been identified that, in rodents, retain the metabolic benefits of RXR agonists with reduced side effects. These modulators bind specifically to RXR with high affinity and are RXR homodimer partial agonists. Although RXR agonists activate many heterodimer partners, these modulators selectively activate RXR:PPARalpha and RXR:PPARgamma, but not RXR:RARalpha, RXR:LXRalpha, RXR:LXRbeta, or RXR:FXRalpha. We report the in vivo characterization of one RXR modulator, LG101506 (LG1506). In Zucker fatty (fa/fa) rats, LG1506 is a potent insulin sensitizer that also enhances the insulin-sensitizing activities of rosiglitazone. Administration of LG1506 reduces both body weight gain and food consumption and blocks the TZD-induced weight gain when coadministered with rosiglitazone. LG1506 does not significantly suppress the thyroid hormone axis in rats, nor does it elevate triglycerides in Sprague Dawley rats. However, LG1506 produces a unique pattern of triglycerides elevation in Zucker rats. LG1506 elevates high-density lipoprotein cholesterol in humanized apolipoprotein A-1-transgenic mice. Therefore, selective RXR modulators are a promising approach for developing improved therapies for type 2 diabetes, although additional studies are needed to understand the strain-specific effects on triglycerides.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Fatty Acids, Unsaturated/administration & dosage , Hypoglycemic Agents/administration & dosage , Obesity/drug therapy , Phenyl Ethers/administration & dosage , Retinoid X Receptors/agonists , Thiazolidinediones/administration & dosage , Analysis of Variance , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/physiology , Area Under Curve , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Dose-Response Relationship, Drug , Drug Interactions , Female , Hypoglycemic Agents/therapeutic use , Mice , Mice, Transgenic , Obesity/blood , Obesity/complications , PPAR gamma/agonists , PPAR gamma/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Retinoid X Receptors/metabolism , Rosiglitazone , Statistics, Nonparametric , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Thyroid Gland/drug effects , Triglycerides/blood
12.
Bioorg Med Chem Lett ; 14(24): 6113-6, 2004 Dec 20.
Article in English | MEDLINE | ID: mdl-15546740

ABSTRACT

To understand the species selectivity in a series of alpha-methyl-alpha-phenoxy carboxylic acid PPARalpha/gamma dual agonists (1-11), structure-based molecular modeling was carried out in the ligand binding pockets of both human and mouse PPARalpha. This study suggested that interaction of both 4-phenoxy and phenyloxazole substituents of these ligands with F272 and M279 in mouse PPARalpha leads to the species-specific divergence in ligand binding. Insights obtained in the molecular modeling studies of these key interactions resulted in the ability to convert a human-selective PPARalpha agonist to a human and mouse dual agonist within the same platform.


Subject(s)
Cinnamates/chemical synthesis , Models, Molecular , PPAR alpha/agonists , Animals , Cinnamates/chemistry , Cinnamates/pharmacology , Drug Design , Humans , Ligands , Mice , Molecular Structure , Species Specificity , Structure-Activity Relationship
13.
Endocrinology ; 145(2): 565-73, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14605005

ABSTRACT

The retinoid X receptor (RXR), a ubiquitously expressed intracellular receptor, regulates pathways controlling glucose, triglycerides, cholesterol, and bile acid metabolism. In addition to its role in those metabolic pathways, we reported that RXR activation with a pan agonist [e.g. LG100268 (LG268)] decreases both body weight gain (BWG) and food consumption (FC) in obese, insulin-resistant rodents. In parallel with those changes in energy balance, we show here that activation of RXR pathways results in adipose tissue remodeling, particularly within sc fat where the rate of apoptosis is increased 5-fold. This change may underlie the selective decrease in fat mass observed in Zucker fatty rats treated with LG268 for 6 wk. Because FC is strongly correlated with BWG in treated animals, we hypothesized that regulation of FC might be the primary mechanism underlying reduced BWG during RXR agonist administration. Importantly, decreased FC is due to decreased meal size, suggestive of induced satiety rather than malaise and/or aversion to food. Furthermore, administration of LG268 directly into the brain via intracerebroventricular injection also reduces FC, BWG, and insulin, whereas the elevation in triglycerides observed after oral administration is absent. The latter observation suggests that RXR actions on energy balance and lipid homeostasis are separable. Therefore, ligand-mediated activation of either an RXR homodimer or an unidentified heterodimeric complex regulates pathways controlling energy balance at least in part via a central nervous system-mediated mechanism.


Subject(s)
Appetite Regulation/physiology , Receptors, Retinoic Acid/physiology , Transcription Factors/physiology , Adipose Tissue/cytology , Adipose Tissue/physiopathology , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/pharmacology , Apoptosis , Brain/drug effects , Brain/physiopathology , Eating , Energy Metabolism , Female , Food , Homeostasis , Injections, Intraventricular , Insulin/blood , Lipids/blood , Obesity/drug therapy , Obesity/physiopathology , Organic Chemicals , Rats , Rats, Zucker , Receptors, Retinoic Acid/agonists , Retinoid X Receptors , Satiation , Transcription Factors/agonists , Triglycerides/blood , Weight Gain
14.
Endocrinology ; 143(8): 2880-5, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12130551

ABSTRACT

The retinoid X receptor (RXR) agonist bexarotene can cause clinically significant hypothyroidism in cutaneous T cell lymphoma patients. The mechanism by which the RXR agonist produces this effect is unclear. We have studied the impact of a selective RXR agonist (rexinoid), LG100268, on rat thyroid axis hormones and show that the acute phase of hypothyroidism is associated with reduced pituitary TSH secretion. A single oral administration of LG100268 to naive Sprague Dawley rats causes a rapid and statistically significant decline in TSH levels (apparent in 0.5-1 h). Total T(4) and T(3) levels decline more gradually, reaching statistical significance 24 h after treatment. Increasing doses of LG100268 produce greater suppression of thyroid axis hormones. To investigate the mechanism(s) mediating this suppression, we determined pituitary TSHbeta mRNA, TSH protein levels, and TRH-stimulated TSH secretion. Two hours after treatment, neither TSHbeta mRNA nor TSH protein levels were altered by LG100268. However, LG100268 treatment reduced the area under the curve for TRH-stimulated TSH secretion by 54%. We have identified an unexpected mechanism by which rexinoids induce hypothyroidism by acutely reducing TSH secretion from the anterior pituitary. This mechanism is independent of the rexinoid's previously demonstrated inhibition of TSHbeta gene transcription.


Subject(s)
Hypothyroidism/chemically induced , Nicotinic Acids/pharmacology , Receptors, Retinoic Acid/physiology , Tetrahydronaphthalenes/pharmacology , Transcription Factors/physiology , Animals , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Retinoic Acid/agonists , Retinoid X Receptors , Thyroid Hormones/blood , Thyrotropin/blood , Thyrotropin/genetics , Transcription Factors/agonists
15.
Diabetes ; 51(4): 1083-7, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11916929

ABSTRACT

A novel nonthiazolidinedione dual peroxisome proliferator- activated receptor (PPAR)-alpha/gamma agonist, LY465608, was designed to address the major metabolic disturbances of type 2 diabetes. LY465608 altered PPAR-responsive genes in liver and fat of db/db mice and dose-dependently lowered plasma glucose in hyperglycemic male Zucker diabetic fatty (ZDF) rats, with an ED(50) for glucose normalization of 3.8 mg small middle dot kg(-1) small middle dot day(-1). Metabolic improvements were associated with enhanced insulin sensitivity, as demonstrated in female obese Zucker (fa/fa) rats using both oral glucose tolerance tests and hyperinsulinemic-euglycemic clamps. Further characterization of LY465608 revealed metabolic changes distinct from a selective PPAR-gamma agonist, which were presumably due to the concomitant PPAR-alpha agonism, lower respiratory quotient, and less fat accumulation, despite a similar impact on glycemia in male ZDF rats. In addition to these alterations in diabetic and insulin-resistant animals, LY465608 dose-dependently elevated HDL cholesterol and lowered plasma triglycerides in human apolipoprotein A-I transgenic mice, demonstrating that this compound significantly improves primary cardiovascular risk factors. Overall, these studies demonstrate that LY465608 beneficially impacts multiple facets of type 2 diabetes and associated cardiovascular risk, including those facets involved in the development of micro- and macrovascular complications, which are the major sources for morbidity and mortality in these patients.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Hyperglycemia/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Metabolic Syndrome/physiology , Organic Chemicals , Receptors, Cytoplasmic and Nuclear/agonists , Thiazolidinediones , Transcription Factors/agonists , Animals , Blood Glucose/drug effects , DNA-Binding Proteins/agonists , Diabetes Mellitus, Type 2/blood , Dose-Response Relationship, Drug , Energy Intake/drug effects , Energy Metabolism/drug effects , Glucose Tolerance Test , Male , Mice , Mice, Mutant Strains , Rats , Rats, Zucker , Rosiglitazone , Thiazoles/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...