Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nat Med ; 23(9): 1086-1094, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28825717

ABSTRACT

Recent large-scale genetic sequencing efforts have identified rare coding variants in genes in the triglyceride-rich lipoprotein (TRL) clearance pathway that are protective against coronary heart disease (CHD), independently of LDL cholesterol (LDL-C) levels. Insight into the mechanisms of protection of these variants may facilitate the development of new therapies for lowering TRL levels. The gene APOC3 encodes apoC-III, a critical inhibitor of triglyceride (TG) lipolysis and remnant TRL clearance. Here we report a detailed interrogation of the mechanism of TRL lowering by the APOC3 Ala43Thr (A43T) variant, the only missense (rather than protein-truncating) variant in APOC3 reported to be TG lowering and protective against CHD. We found that both human APOC3 A43T heterozygotes and mice expressing human APOC3 A43T display markedly reduced circulating apoC-III levels. In mice, this reduction is due to impaired binding of A43T apoC-III to lipoproteins and accelerated renal catabolism of free apoC-III. Moreover, the reduced content of apoC-III in TRLs resulted in accelerated clearance of circulating TRLs. On the basis of this protective mechanism, we developed a monoclonal antibody targeting lipoprotein-bound human apoC-III that promotes circulating apoC-III clearance in mice expressing human APOC3 and enhances TRL catabolism in vivo. These data reveal the molecular mechanism by which a missense variant in APOC3 causes reduced circulating TG levels and, hence, protects from CHD. This protective mechanism has the potential to be exploited as a new therapeutic approach to reduce apoC-III levels and circulating TRL burden.


Subject(s)
Apolipoprotein C-III/genetics , Lipoproteins/metabolism , Mutation, Missense , Triglycerides/metabolism , Aged , Animals , Antibodies, Monoclonal/pharmacology , Apolipoprotein C-III/drug effects , Apolipoproteins B/metabolism , Cholesterol, HDL/metabolism , Chromatography, Liquid , Computer Simulation , Coronary Disease/genetics , Cross-Sectional Studies , Female , Humans , Immunoblotting , Lipid Metabolism/genetics , Lipoproteins/drug effects , Lipoproteins, VLDL/metabolism , Male , Mass Spectrometry , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Protective Factors , Tandem Mass Spectrometry
2.
Bioorg Med Chem ; 21(17): 5081-97, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23871442

ABSTRACT

DGAT-1 is an enzyme that catalyzes the final step in triglyceride synthesis. mRNA knockout experiments in rodent models suggest that inhibitors of this enzyme could be of value in the treatment of obesity and type II diabetes. The carboxylic acid-based DGAT-1 inhibitor 1 was advanced to clinical trials for the treatment of type 2 diabetes, despite of the low passive permeability of 1. Because of questions relating to the potential attenuation of distribution and efficacy of a poorly permeable agent, efforts were initiated to identify compounds with improved permeability. Replacement of the acid moiety in 1 with an oxadiazole led to the discovery of 52, which possesses substantially improved passive permeability. The resulting pharmacodynamic profile of this neutral DGAT-1 inhibitor was found to be similar to 1 at comparable plasma exposures.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Oxazepines/chemistry , Administration, Oral , Animals , Diacylglycerol O-Acyltransferase/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Half-Life , Humans , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/drug therapy , Oxadiazoles/chemistry , Oxazepines/pharmacokinetics , Oxazepines/therapeutic use , Protein Binding , Rats , Structure-Activity Relationship
3.
Am J Physiol Gastrointest Liver Physiol ; 304(11): G958-69, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23558010

ABSTRACT

Alterations in fat metabolism, in particular elevated plasma concentrations of free fatty acids and triglycerides (TG), have been implicated in the pathogenesis of Type 2 diabetes, obesity, and cardiovascular disease. Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1), a member of the large family of membrane-bound O-acyltransferases, catalyzes the final step in triacylglycerol formation. In the intestine, DGAT1 is one of the acyltransferases responsible for the reesterficiation of dietary TG. Following a single dose of a selective pharmacological inhibitor of DGAT1, PF-04620110, a dose-dependent inhibition of TG and vitamin A absorption postprandially was demonstrated in rodents and human subjects. In C57/BL6J mice, acute DGAT1 inhibition alters the temporal and spatial pattern of dietary lipid absorption. To understand the impact of DGAT1 inhibition on enterocyte lipid metabolism, lipomic profiling was performed in rat intestine and plasma as well as human plasma. DGAT1 inhibition causes an enrichment of polyunsaturated fatty acids within the TG class of lipids. This pharmacological intervention gives us insight as to the role of DGAT1 in human dietary lipid absorption.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Intestinal Absorption/drug effects , Oxazepines/pharmacology , Adolescent , Adult , Animals , Case-Control Studies , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Dietary Fats/blood , Dietary Fats/metabolism , Dose-Response Relationship, Drug , Enterocytes/metabolism , Enzyme Inhibitors/pharmacokinetics , Fatty Acids, Unsaturated/blood , Fatty Acids, Unsaturated/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oxazepines/pharmacokinetics , Postprandial Period , Rats , Rats, Sprague-Dawley , Triglycerides/blood , Triglycerides/metabolism , Vitamin A/metabolism
4.
Bioorg Med Chem Lett ; 23(10): 3051-8, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23562063

ABSTRACT

A novel and potent small molecule glucagon receptor antagonist for the treatment of diabetes mellitus is reported. This candidate, (S)-3-[4-(1-{3,5-dimethyl-4-[4-(trifluoromethyl)-1H-pyrazol-1-yl]phenoxy}butyl)benzamido]propanoic acid, has lower molecular weight and lipophilicity than historical glucagon receptor antagonists, resulting in excellent selectivity in broad-panel screening, lower cytotoxicity, and excellent overall in vivo safety in early pre-clinical testing. Additionally, it displays low in vivo clearance and excellent oral bioavailability in both rats and dogs. In a rat glucagon challenge model, it was shown to reduce the glucagon-elicited glucose excursion in a dose-dependent manner and at a concentration consistent with its rat in vitro potency. Its properties make it an excellent candidate for further investigation.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Design , Propionates/pharmacology , Receptors, Glucagon/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Chemistry, Physical , Dogs , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Haplorhini , Humans , Liver/cytology , Mice , Molecular Structure , Propionates/administration & dosage , Propionates/chemical synthesis , Rats , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship
5.
J Med Chem ; 55(3): 1318-33, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22196621

ABSTRACT

Glucokinase is a key regulator of glucose homeostasis, and small molecule allosteric activators of this enzyme represent a promising opportunity for the treatment of type 2 diabetes. Systemically acting glucokinase activators (liver and pancreas) have been reported to be efficacious but in many cases present hypoglycaemia risk due to activation of the enzyme at low glucose levels in the pancreas, leading to inappropriately excessive insulin secretion. It was therefore postulated that a liver selective activator may offer effective glycemic control with reduced hypoglycemia risk. Herein, we report structure-activity studies on a carboxylic acid containing series of glucokinase activators with preferential activity in hepatocytes versus pancreatic ß-cells. These activators were designed to have low passive permeability thereby minimizing distribution into extrahepatic tissues; concurrently, they were also optimized as substrates for active liver uptake via members of the organic anion transporting polypeptide (OATP) family. These studies lead to the identification of 19 as a potent glucokinase activator with a greater than 50-fold liver-to-pancreas ratio of tissue distribution in rodent and non-rodent species. In preclinical diabetic animals, 19 was found to robustly lower fasting and postprandial glucose with no hypoglycemia, leading to its selection as a clinical development candidate for treating type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Enzyme Activators/chemical synthesis , Glucokinase/metabolism , Hepatocytes/metabolism , Hypoglycemic Agents/chemical synthesis , Imidazoles/chemical synthesis , Nicotinic Acids/chemical synthesis , Allosteric Site , Animals , Blood Glucose/metabolism , Dogs , Enzyme Activators/pharmacokinetics , Enzyme Activators/pharmacology , Haplorhini , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , In Vitro Techniques , Insulin-Secreting Cells/metabolism , Male , Models, Molecular , Nicotinic Acids/pharmacokinetics , Nicotinic Acids/pharmacology , Organic Anion Transporters/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Rats, Wistar , Stereoisomerism , Structure-Activity Relationship , Tissue Distribution
6.
PLoS One ; 6(11): e27009, 2011.
Article in English | MEDLINE | ID: mdl-22073239

ABSTRACT

Triglyceride accumulation is associated with obesity and type 2 diabetes. Genetic disruption of diacylglycerol acyltransferase 1 (DGAT1), which catalyzes the final reaction of triglyceride synthesis, confers dramatic resistance to high-fat diet induced obesity. Hence, DGAT1 is considered a potential therapeutic target for treating obesity and related metabolic disorders. However, the molecular events shaping the mechanism of action of DGAT1 pharmacological inhibition have not been fully explored yet. Here, we investigate the metabolic molecular mechanisms induced in response to pharmacological inhibition of DGAT1 using a recently developed computational systems biology approach, the Causal Reasoning Engine (CRE). The CRE algorithm utilizes microarray transcriptomic data and causal statements derived from the biomedical literature to infer upstream molecular events driving these transcriptional changes. The inferred upstream events (also called hypotheses) are aggregated into biological models using a set of analytical tools that allow for evaluation and integration of the hypotheses in context of their supporting evidence. In comparison to gene ontology enrichment analysis which pointed to high-level changes in metabolic processes, the CRE results provide detailed molecular hypotheses to explain the measured transcriptional changes. CRE analysis of gene expression changes in high fat habituated rats treated with a potent and selective DGAT1 inhibitor demonstrate that the majority of transcriptomic changes support a metabolic network indicative of reversal of high fat diet effects that includes a number of molecular hypotheses such as PPARG, HNF4A and SREBPs. Finally, the CRE-generated molecular hypotheses from DGAT1 inhibitor treated rats were found to capture the major molecular characteristics of DGAT1 deficient mice, supporting a phenotype of decreased lipid and increased insulin sensitivity.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Models, Theoretical , Algorithms , Animals , Feeding Behavior , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Triglycerides/blood
7.
BMC Genomics ; 12: 281, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21631939

ABSTRACT

BACKGROUND: Glucagon is an important hormone in the regulation of glucose homeostasis, particularly in the maintenance of euglycemia and prevention of hypoglycemia. In type 2 Diabetes Mellitus (T2DM), glucagon levels are elevated in both the fasted and postprandial states, which contributes to inappropriate hyperglycemia through excessive hepatic glucose production. Efforts to discover and evaluate glucagon receptor antagonists for the treatment of T2DM have been ongoing for approximately two decades, with the challenge being to identify an agent with appropriate pharmaceutical properties and efficacy relative to potential side effects. We sought to determine the hepatic & systemic consequence of full glucagon receptor antagonism through the study of the glucagon receptor knock-out mouse (Gcgr-/-) compared to wild-type littermates. RESULTS: Liver transcriptomics was performed using Affymetric expression array profiling, and liver proteomics was performed by iTRAQ global protein analysis. To complement the transcriptomic and proteomic analyses, we also conducted metabolite profiling (~200 analytes) using mass spectrometry in plasma. Overall, there was excellent concordance (R = 0.88) for changes associated with receptor knock-out between the transcript and protein analysis. Pathway analysis tools were used to map the metabolic processes in liver altered by glucagon receptor ablation, the most notable being significant down-regulation of gluconeogenesis, amino acid catabolism, and fatty acid oxidation processes, with significant up-regulation of glycolysis, fatty acid synthesis, and cholesterol biosynthetic processes. These changes at the level of the liver were manifested through an altered plasma metabolite profile in the receptor knock-out mice, e.g. decreased glucose and glucose-derived metabolites, and increased amino acids, cholesterol, and bile acid levels. CONCLUSIONS: In sum, the results of this study suggest that the complete ablation of hepatic glucagon receptor function results in major metabolic alterations in the liver, which, while promoting improved glycemic control, may be associated with adverse lipid changes.


Subject(s)
Diabetes Mellitus/drug therapy , Gene Expression Profiling , Gene Knockout Techniques , Liver/metabolism , Proteomics , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/genetics , Amino Acids/metabolism , Animals , Carbohydrate Metabolism/genetics , Diabetes Mellitus/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Lipid Metabolism/genetics , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Glucagon/deficiency
8.
ACS Med Chem Lett ; 2(5): 407-12, 2011 May 12.
Article in English | MEDLINE | ID: mdl-24900321

ABSTRACT

Acyl-CoA:diacylglycerol acyltransferase-1 (DGAT-1) catalyzes the final committed step in the biosynthesis of triglycerides. DGAT-1 knockout mice have been shown to be resistant to diet-induced obesity and have increased insulin sensitivity. Thus, inhibition of DGAT-1 may represent an attractive target for the treatment of obesity or type II diabetes. Herein, we report the discovery and characterization of a potent and selective DGAT-1 inhibitor PF-04620110 (3). Compound 3 inhibits DGAT-1 with an IC50 of 19 nM and shows high selectivity versus a broad panel of off-target pharmacologic end points. In vivo DGAT-1 inhibition has been demonstrated through reduction of plasma triglyceride levels in rodents at doses of ≥0.1 mg/kg following a lipid challenge. On the basis of this pharmacologic and pharmacokinetic profile, compound 3 has been advanced to human clinical studies.

9.
J Cereb Blood Flow Metab ; 30(2): 352-60, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19861974

ABSTRACT

Diabetes is an increased risk factor for stroke and results in increased brain damage in experimental animals and humans. The precise mechanisms are unclear, but our earlier studies in the db/db mice suggested that the cerebral inflammatory response initiating recovery was both delayed and diminished in the diabetic mice compared with the nondiabetic db/+ mice. In this study, we investigated the actions of the peroxisome proliferator-activated receptor (PPAR)-gamma agonist darglitazone in treating diabetes and promoting recovery after a hypoxic-ischemic (H/I) insult in the diabetic ob/ob mouse. Male ob/+ and ob/ob mice received darglitazone (1 mg/kg) for 7 days before induction of H/I. Darglitazone restored euglycemia and normalized elevated corticosterone, triglycerides, and very-low-density lipoprotein levels. Darglitazone dramatically reduced the infarct size in the ob/ob mice at 24 h of recovery compared with the untreated group (30+/-13% to 3.3+/-1.6%, n=6 to 8) but did not show any significant effect in the ob/+ mice. Microglial and astrocytic activation monitored by cytokine expression (interleukin-1beta and tumor necrosis factor-alpha) and in situ hybridization studies (bfl1 and glial fibrillary acidic protein) suggest a biphasic inflammatory response, with darglitazone restoring the compromised proinflammatory response(s) in the diabetic mouse at 4 h but suppressing subsequent inflammatory responses at 8 and 24 h in both control and diabetic mice.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Hypoglycemic Agents/pharmacology , Hypoxia-Ischemia, Brain/immunology , Inflammation/immunology , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Blood Glucose/analysis , Corticosterone/blood , Diabetes Mellitus, Experimental/complications , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/metabolism , In Situ Hybridization , Lipoproteins, VLDL/blood , Male , Mice , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , PPAR gamma/drug effects , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood
11.
Bioorg Med Chem Lett ; 15(2): 459-65, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15603973

ABSTRACT

The synthesis, in vitro, and in vivo biological characterization of a series of achiral 5-chloroindoloyl glycine amide inhibitors of human liver glycogen phosphorylase A are described. Improved potency over previously reported compounds in cellular and in vivo assays was observed. The allosteric binding site of these compounds was shown by X-ray crystallography to be the same as that reported previously for 5-chloroindoloyl norstatine amides.


Subject(s)
Amides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Glycogen Phosphorylase/antagonists & inhibitors , Indoles/chemical synthesis , Allosteric Site , Amides/pharmacology , Aminocaproates/chemistry , Aminocaproates/pharmacology , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Glycine/chemistry , Glycine/pharmacology , Glycogen Phosphorylase/metabolism , Humans , Indoles/chemistry , Indoles/pharmacology , Liver/enzymology , Liver/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL