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










Publication year range
1.
J Pharmacol Exp Ther ; 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849142

ABSTRACT

Acute Kidney Injury (AKI) is characterized by an abrupt decline in kidney function and has been associated with excess risks of death, kidney disease progression, and cardiovascular events. The kidney has a high energetic demand with mitochondrial health being essential to renal function and damaged mitochondria has been reported across AKI subtypes. 5' adenosine monophosphate-activated protein kinase (AMPK) activation preserves cellular energetics through improvement of mitochondrial function and biogenesis when ATP levels are low such as under ischemia-induced AKI. We developed a selective potent small molecule pan AMPK activator, compound 1, and tested its ability to increase AMPK activity and preserve kidney function during ischemia/reperfusion injury in rats. A single administration of 1 caused sustained activation of AMPK for at least 24 hours, protected against acute tubular necrosis, and reduced clinical markers of tubular injury such as NephroCheck and Fractional Excretion of Sodium (FENa). Reduction in plasma creatinine and increased Glomerular Filtration Rate (GFR) indicated preservation of kidney function. Surprisingly, we observed a strong diuretic effect of AMPK activation associated with natriuresis both with and without AKI. Our findings demonstrate that activation of AMPK leads to protection of tubular function under hypoxic/ischemic conditions which holds promise as a potential novel therapeutic approach for AKI. Significance Statement No approved pharmacological therapies currently exist for acute kidney injury. We developed Compound 1 which dose-dependently activated AMPK in the kidney and protected kidney function and tubules after ischemic renal injury in the rat. This was accompanied by natriuresis in injured as well as uninjured rats.

2.
Front Endocrinol (Lausanne) ; 13: 1061688, 2022.
Article in English | MEDLINE | ID: mdl-36482991

ABSTRACT

The free fatty acid receptor 1 (FFAR1/GPR40) mediates fatty acid-induced insulin secretion from pancreatic ß-cells. At least 3 distinct binding sites exist on the FFAR1 receptor and numerous synthetic ligands have been investigated for their anti-diabetic actions. Fasiglifam, binds to site-1 and stimulates intra-cellular calcium release and improves glycemic control in diabetic patients. Recently, small molecule FFAR1 agonists were discovered which bind to site-3, stimulating both intra-cellular calcium and cAMP, resulting in insulin and glucagon-like peptide-1 (GLP-1) secretion. The ability of our site-3 FFAR1 agonist (compound A) to control blood glucose was evaluated in spontaneously diabetic cynomolgus monkeys during an oral glucose tolerance test. In type-2 diabetic (T2D) animals, significant reductions in blood glucose and insulin were noted. To better understand the mechanism of these in vivo findings, we evaluated the effect of compound A in islets under several conditions of dysfunction. First, healthy human and non-human primate islets were treated with compound A and showed potentiation of insulin and glucagon secretion from both species. Next, we determined glucose-responsive insulin secretion under gluco-lipotoxic conditions and from islets isolated from type-2 diabetic humans. Despite a dysfunctional phenotype that failed to secrete insulin in response to glucose, site-3 FFAR1 agonism not only enhanced insulin secretion, but restored glucose responsiveness across a range of glucose concentrations. Lastly, we treated ex vivo human islets chronically with a sulfonylurea to induce secondary beta-cell failure. Again, this model showed reduced glucose-responsive insulin secretion that was restored and potentiated by site-3 FFAR1 agonism. Together these data suggest a mechanism for FFAR1 where agonists have direct effects on islet hormone secretion that can overcome a dysfunctional T2D phenotype. These unique characteristics of FFAR1 site-3 agonists make them an appealing potential therapy to treat type-2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Secretion , Receptors, G-Protein-Coupled , Blood Glucose , Calcium , Diabetes Mellitus, Type 2/drug therapy , Glucose/pharmacology , Insulin , Receptors, G-Protein-Coupled/agonists , Macaca fascicularis , Animals
3.
Front Endocrinol (Lausanne) ; 12: 652628, 2021.
Article in English | MEDLINE | ID: mdl-34054727

ABSTRACT

The glucagon-like peptide-1 receptor (GLP-1R) is a G-protein-coupled receptor (GPCR) whose activation results in suppression of food intake and improvement of glucose metabolism. Several receptor interacting proteins regulate the signaling of GLP-1R such as G protein-coupled receptor kinases (GRK) and ß-arrestins. Here we evaluated the physiological and pharmacological impact of GRK inhibition on GLP-1R activity leveraging small molecule inhibitors of GRK2 and GRK3. We demonstrated that inhibition of GRK: i) inhibited GLP-1-mediated ß-arrestin recruitment, ii) enhanced GLP-1-induced insulin secretion in isolated islets and iii) has additive effect with dipeptidyl peptidase 4 in mediating suppression of glucose excursion in mice. These findings highlight the importance of GRK to modulate GLP-1R function in vitro and in vivo. GRK inhibition is a potential therapeutic approach to enhance endogenous and pharmacologically stimulated GLP-1R signaling.


Subject(s)
G-Protein-Coupled Receptor Kinase 1/antagonists & inhibitors , Glucagon-Like Peptide 1/metabolism , Amides/chemistry , Animals , CHO Cells , Calcium/metabolism , Cricetulus , Diabetes Mellitus/metabolism , Dipeptidyl Peptidase 4/metabolism , Eating , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Mice , Obesity/metabolism , Phosphorylation , Receptors, Glucagon/metabolism , Renal Insufficiency, Chronic/metabolism , Signal Transduction , beta-Arrestins/metabolism
4.
Bioorg Med Chem Lett ; 30(23): 127602, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33038544

ABSTRACT

G-protein coupled receptor kinase 2 (GRK2), which is upregulated in the failing heart, appears to play a critical role in heart failure (HF) progression in part because enhanced GRK2 activity promotes dysfunction of ß-adrenergic signaling and myocyte death. An orally bioavailable GRK2 inhibitor could offer unique therapeutic outcomes that cannot be attained by current heart failure treatments that directly target GPCRs or angiotensin-converting enzyme. Herein, we describe the discovery of a potent, selective, and orally bioavailable GRK2 inhibitor, 8h, through high-throughput screening, hit-to-lead optimization, structure-based design, molecular modelling, synthesis, and biological evaluation. In the cellular target engagement assays, 8h enhances isoproterenol-mediated cyclic adenosine 3',5'-monophosphate (cAMP) production in HEK293 cells overexpressing GRK2. Compound 8h was further evaluated in a human stem cell-derived cardiomyocyte (HSC-CM) contractility assay and potentiated isoproterenol-induced beating rate in HSC-CMs.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , Enzyme Assays , G-Protein-Coupled Receptor Kinase 2/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Molecular Docking Simulation , Molecular Structure , Myocytes, Cardiac/drug effects , Phthalazines/chemical synthesis , Phthalazines/pharmacokinetics , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/chemical synthesis , Quinazolines/metabolism , Quinazolines/pharmacokinetics , Structure-Activity Relationship
5.
ACS Med Chem Lett ; 10(1): 16-21, 2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30655940

ABSTRACT

GPR40 is a G-protein-coupled receptor which mediates fatty acid-induced glucose-stimulated insulin secretion from pancreatic beta cells and incretion release from enteroendocrine cells of the small intestine. GPR40 full agonists exhibit superior glucose lowering compared to partial agonists in preclinical species due to increased insulin and GLP-1 secretion, with the added benefit of promoting weight loss. In our search for potent GPR40 full agonists, we discovered a superagonist which displayed excellent in vitro potency and superior efficacy in the Gαs-mediated signaling pathway. Most synthetic GPR40 agonists have a carboxylic acid headgroup, which may cause idiosyncratic toxicities, including drug-induced-liver-injury (DILI). With a methyl group and a fluorine atom substituted at the α-C of the carboxylic acid group, 19 is not only highly efficacious in lowering glucose and body weight in rodent models but also has a low DILI risk due to its stable acylglucuronide metabolite.

6.
J Med Chem ; 61(22): 10276-10298, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30339387

ABSTRACT

A novel series of 6-benzhydryl-4-amino-quinolin-2-ones was discovered as cannabinoid type 1 receptor (CB1R) inverse agonists based on the high-throughput screening hit, compound 1a. Structure-activity relationships were studied to improve in vitro/in vivo pharmacology and restrict distribution to the peripheral circulation. We adopted several strategies such as increasing topological polar surface area, incorporating discrete polyethylene glycol side chains, and targeting P-glycoprotein (P-gp) to minimize access to the brain. Compound 6a is a P-gp substrate and a potent and highly selective CB1R inverse agonist, demonstrating excellent in vivo metabolic stability and a low brain to plasma ratio. However, brain receptor occupancy studies showed that compound 6a may accumulate in brain with repeat dosing. This was evidenced by compound 6a inhibiting food intake and inducing weight loss in diet-induced obese mice. Thus, a strategy based on P-gp efflux may not be adequate for peripheral restriction of the disclosed quinolinone series.


Subject(s)
Drug Inverse Agonism , Quinolones/chemistry , Quinolones/pharmacology , Receptor, Cannabinoid, CB1/agonists , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Conformation , Quinolones/metabolism , Quinolones/pharmacokinetics , Rats , Receptor, Cannabinoid, CB1/chemistry , Receptor, Cannabinoid, CB1/metabolism , Structure-Activity Relationship , Tissue Distribution
7.
Bioorg Med Chem Lett ; 28(7): 1182-1187, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29523385

ABSTRACT

Synthesis and biological evaluation of benzocyclobutane-C-glycosides as potent and orally active SGLT1/SGLT2 dual inhibitors are described. Compound 19 showed high inhibitory potency at SGLT1 (IC50 = 45 nM), and excellent potency at SGLT2 (IC50 = 1 nM). It also displayed excellent PK profiles in mice, rats, dogs and monkeys (F = 78-107%). In SD rats, compound 19 treatments significantly reduced blood glucose levels in a dose-dependent manner. In ZDF rats, compound 19 displayed anti-hyperglycemic effect up to 24 h. Therefore, compound 19 may serve as valuable pharmacological tool, and potential use as a treatment for metabolic syndrome.


Subject(s)
Benzene Derivatives/pharmacology , Cyclobutanes/pharmacology , Glycosides/pharmacology , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors , Administration, Oral , Animals , Benzene Derivatives/administration & dosage , Benzene Derivatives/chemistry , Cyclobutanes/administration & dosage , Cyclobutanes/chemistry , Dogs , Dose-Response Relationship, Drug , Glycosides/administration & dosage , Glycosides/chemistry , Haplorhini , Humans , Mice , Molecular Structure , Rats , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2/metabolism , Structure-Activity Relationship
8.
Mol Pharmacol ; 93(6): 581-591, 2018 06.
Article in English | MEDLINE | ID: mdl-29572336

ABSTRACT

GPR40 is a clinically validated molecular target for the treatment of diabetes. Many GPR40 agonists have been identified to date, with the partial agonist fasiglifam (TAK-875) reaching phase III clinical trials before its development was terminated due to off-target liver toxicity. Since then, attention has shifted toward the development of full agonists that exhibit superior efficacy in preclinical models. Full agonists bind to a distinct binding site, suggesting conformational plasticity and a potential for biased agonism. Indeed, it has been suggested that alternative pharmacology may be required for meaningful efficacy. In this study, we described the discovery and characterization of Compound A, a newly identified GPR40 allosteric full agonist highly efficacious in human islets at potentiating glucose-stimulated insulin secretion. We compared Compound A-induced GPR40 activity to that induced by both fasiglifam and AM-1638, another allosteric full agonist previously reported to be highly efficacious in preclinical models, at a panel of G proteins. Compound A was a full agonist at both the Gαq and Gαi2 pathways, and in contrast to fasiglifam Compound A also induced Gα12 coupling. Compound A and AM-1638 displayed similar activity at all pathways tested. The Gα12/Gα13-mediated signaling pathway has been linked to protein kinase D activation as well as actin remodeling, well known to contribute to the release of insulin vesicles. Our data suggest that the pharmacology of GPR40 is complex and that Gα12/Gα13-mediated signaling, which may contribute to GPR40 agonists therapeutic efficacy, is a specific property of GPR40 allosteric full agonists.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Glucose/metabolism , Insulin Secretion/physiology , Insulin/metabolism , Islets of Langerhans/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Benzofurans/pharmacology , CHO Cells , Cell Line , Cricetulus , HEK293 Cells , Humans , Hypoglycemic Agents/pharmacology , Insulin Secretion/drug effects , Islets of Langerhans/drug effects , Protein Kinase C/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfones/pharmacology
9.
SLAS Discov ; 22(4): 433-439, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28328322

ABSTRACT

Monoacylglycerol acyltransferase enzymes (MGAT1, MGAT2, and MGAT3) convert monoacylglycerol to diacylglycerol (DAG). MGAT1 and MGAT2 are both implicated in obesity-related metabolic diseases. Conventional MGAT enzyme assays use radioactive substrates, wherein the product of the MGAT-catalyzed reaction is usually resolved by time-consuming thin layer chromatography (TLC) analysis. Furthermore, microsomal membrane preparations typically contain endogenous diacylglycerol acyltransferase (DGAT) from the host cells, and these DGAT activities can further acylate DAG to form triglyceride (TG). Our mass spectrometry (liquid chromatography-tandem mass spectrometry, or LC/MS/MS) MGAT2 assay measures human recombinant MGAT2-catalyzed formation of didecanoyl-glycerol from 1-decanoyl-rac-glycerol and decanoyl-CoA, to produce predominantly 1,3-didecanoyl-glycerol. Unlike 1,2-DAG, 1,3-didecanoyl-glycerol is proved to be not susceptible to further acylation to TG. 1,3-Didecanoyl-glycerol product can be readily solubilized and directly subjected to high-throughput mass spectrometry (HTMS) without further extraction in a 384-well format. We also have established the LC/MS/MS MGAT activity assay in the intestinal microsomes from various species. Our assay is proved to be highly sensitive, and thus it allows measurement of endogenous MGAT activity in cell lysates and tissue preparations. The implementation of the HTMS MGAT activity assay has facilitated the robust screening and evaluation of MGAT inhibitors for the treatment of metabolic diseases.


Subject(s)
Diglycerides/analysis , Enzyme Inhibitors/analysis , High-Throughput Screening Assays , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Acyl Coenzyme A/metabolism , Animals , Chlorocebus aethiops , Chromatography, Liquid/methods , Diglycerides/antagonists & inhibitors , Diglycerides/biosynthesis , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Haplorhini , Humans , Intestines/drug effects , Intestines/enzymology , Kinetics , Mice , Microsomes/drug effects , Microsomes/enzymology , N-Acetylglucosaminyltransferases/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Tandem Mass Spectrometry/methods
10.
Anal Biochem ; 524: 68-75, 2017 05 01.
Article in English | MEDLINE | ID: mdl-27665677

ABSTRACT

Monoacylglycerol acyltransferase 2 (MGAT2) catalyzes the synthesis of diacylglycerol (DAG) from free fatty acids (FFA) and sn-monoacylglycerol (MG), the two major hydrolysis products of dietary fat. To demonstrate MGAT2-mediated cellular activity of triglyceride (TG) synthesis, we utilized 1-oleoyl-glycerol-d5 as a substrate to trace MGAT2-driven 1-oleoyl-glycerol-d5 incorporation into TG in HEK293 cells stably expressing human MGAT2. The oleoyl-glycerol-d5 incorporated major TG species were then quantified by liquid chromatography electrospray ionization tandem mass spectrometry (LC/ESI/MS/MS) in a 96-well format. Conventional MGAT2 target-engagement in vivo assays measure the elevation of total plasma TG by orally dosing a bolus of TG oil. We developed a novel LC/ESI/MS/MS-based fat absorption assay to assess the ability of MGAT2 inhibitors to inhibit fat absorption in CD1 mice by a meal tolerance test consisting of a mixture of liquid Boost plus® and 0.59 g/kg U13C-TG oil. The newly resynthesized plasma heavy TGs containing three 13C in the glycerol backbone and two U13C-acyl-chains, which represented the digested, absorbed and resynthesized TGs, were then quantitated by LC/ESI/MS/MS. With this assay, we identified a potent MGAT2 inhibitor that blocked MGAT2-mediated activity in vitro and in vivo. The use of 1-oleoyl-glycerol-d5 and U13C-TG oil followed by LC/ESI/MS/MS detection of stable-isotopic labeled DAG, TG, or glycerol provides a wide range of applications to study pathophysiological regulation of the monoacylglycerol pathway and MGAT2 activity.


Subject(s)
Glycerides/metabolism , Glycerol/metabolism , Lipid Metabolism , N-Acetylglucosaminyltransferases/metabolism , Tandem Mass Spectrometry/methods , Animals , Chromatography, Liquid/methods , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Mice
11.
J Pharmacol Exp Ther ; 351(2): 423-31, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25216746

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) plays a major role in renal glucose reabsorption. To analyze the potential of insulin-independent blood glucose control, the effects of the novel SGLT2 inhibitor canagliflozin on renal glucose reabsorption and the progression of hyperglycemia were analyzed in Zucker diabetic fatty (ZDF) rats. The transporter activity of recombinant human and rat SGLT2 was inhibited by canagliflozin, with 150- to 12,000-fold selectivity over other glucose transporters. Moreover, in vivo treatment with canagliflozin induced glucosuria in mice, rats, and dogs in a dose-dependent manner. It inhibited apparent glucose reabsorption by 55% in normoglycemic rats and by 94% in hyperglycemic rats. The inhibition of glucose reabsorption markedly reduced hyperglycemia in ZDF rats but did not induce hypoglycemia in normoglycemic animals. The change in urinary glucose excretion should not be used as a marker to predict the glycemic effects of this SGLT2 inhibitor. In ZDF rats, plasma glucose and HbA1c levels progressively increased with age, and pancreatic ß-cell failure developed at 13 weeks of age. Treatment with canagliflozin for 8 weeks from the prediabetic stage suppressed the progression of hyperglycemia, prevented the decrease in plasma insulin levels, increased pancreatic insulin contents, and minimized the deterioration of islet structure. These results indicate that selective inhibition of SGLT2 with canagliflozin controls the progression of hyperglycemia by inhibiting renal glucose reabsorption in ZDF rats. In addition, the preservation of ß-cell function suggests that canagliflozin treatment reduces glucose toxicity via an insulin-independent mechanism.


Subject(s)
Glucose/metabolism , Glucosides/pharmacology , Hyperglycemia/drug therapy , Hyperglycemia/pathology , Kidney/drug effects , Thiophenes/pharmacology , Animals , Blood Glucose/drug effects , Canagliflozin , Diabetes Mellitus, Experimental , Dogs , Glycated Hemoglobin/metabolism , Humans , Hyperglycemia/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Kidney/metabolism , Kidney/pathology , Mice , Rats , Rats, Zucker , Sodium-Glucose Transporter 2/metabolism
12.
J Histochem Cytochem ; 60(9): 694-705, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22740344

ABSTRACT

Endothelial lipase (EL), a member of the triglyceride lipase gene family, has been shown to be a key player in HDL metabolism. Northern blots revealed that EL was highly expressed in endothelium, thyroid, lung, placenta, liver, and testis. In liver and adrenal gland, EL protein was localized with vascular endothelial cells but not parenchymal cells. EL was shown to be upregulated in tissues such as atherosclerotic plaque where it was located in macrophages, endothelial cells, and medial smooth muscle cells. The purpose of this study was to investigate the cellular localization of EL in thyroid and other tissues where EL is known to be expressed. Besides its presence in vascular endothelial and smooth muscle cells, EL protein was detected in the epithelial cells that line the follicles within the thyroid gland. EL-specific immunostaining was also found near the cell surface as well as in the cytoplasm of adipocytes. Using immunoblots, EL expression was confirmed in cultured human omental and subcutaneous adipocytes. EL expression, however, was not found in preadipocytes. These findings suggest that EL plays a role in thyroid and adipocyte biology in addition to its well-known role in endothelial function and HDL metabolism.


Subject(s)
Adipocytes/enzymology , Epithelial Cells/enzymology , Lipase/metabolism , Thyroid Gland/enzymology , Animals , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Humans , Immunohistochemistry , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Omentum/cytology , Omentum/enzymology , Organ Specificity , Species Specificity , Subcutaneous Fat/cytology , Subcutaneous Fat/enzymology
13.
J Lipid Res ; 53(6): 1106-16, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22493088

ABSTRACT

Diacylglycerol acyltransferase (DGAT) catalyzes the final step in triglyceride (TG) synthesis. There are two isoforms, DGAT1 and DGAT2, with distinct protein sequences and potentially different physiological functions. To date, the ability to determine clear functional differences between DGAT1 and DGAT2, especially with respect to hepatic TG synthesis, has been elusive. To dissect the roles of these two key enzymes, we pretreated HepG2 hepatoma cells with (13)C(3)-D(5)-glycerol or (13)C(18)-oleic acid, and profiled the major isotope-labeled TG species by liquid chromatography tandem mass spectrometry. Selective DGAT1 and DGAT2 inhibitors demonstrated that (13)C(3)-D(5)-glycerol-incorporated TG synthesis was mediated by DGAT2, not DGAT1. Conversely, (13)C(18)-oleoyl-incorporated TG synthesis was predominantly mediated by DGAT1. To trace hepatic TG synthesis and VLDL triglyceride (VLDL-TG) secretion in vivo, we administered D(5)-glycerol to mice and measured plasma levels of D(5)-glycerol-incorporated TG. Treatment with an antisense oligonucleotide (ASO) to DGAT2 led to a significant reduction in D(5)-glycerol incorporation into VLDL-TG. In contrast, the DGAT2 ASO had no effect on the incorporation of exogenously administered (13)C(18)-oleic acid into VLDL-TG. Thus, our results indicate that DGAT1 and DGAT2 mediate distinct hepatic functions: DGAT2 is primarily responsible for incorporating endogenously synthesized FAs into TG, whereas DGAT1 plays a greater role in esterifying exogenous FAs to glycerol.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Enzyme Assays/methods , Glycerol/metabolism , Liver/enzymology , Oleic Acid/metabolism , Animals , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/genetics , Enzyme Inhibitors/pharmacology , Esterification/drug effects , Fatty Acids/biosynthesis , Fatty Acids/metabolism , Hep G2 Cells , Humans , Isotope Labeling , Lipoproteins, VLDL/metabolism , Male , Mice , Oligonucleotides, Antisense/genetics , Triglycerides/biosynthesis
14.
J Lipid Res ; 51(12): 3559-67, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20805092

ABSTRACT

Acyl-CoA:diacylglycerol acyltransferase (DGAT) catalyzes the terminal step in triglyceride (TG) synthesis using diacylglycerol (DAG) and fatty acyl-CoA as substrates. In the liver, the production of VLDL permits the delivery of hydrophobic TG from the liver to peripheral tissues for energy metabolism. We describe here a novel high-content, high-throughput LC/MS/MS-based cellular assay for determining DGAT activity. We treated endogenous DGAT-expressing cells with stable isotope-labeled [¹³C18]oleic acid. The [¹³C18]oleoyl-incorporated TG and DAG lipid species were profiled. The TG synthesis pathway assay was optimized to a one-step extraction, followed by LC/MS/MS quantification. Further, we report a novel LC/MS/MS method for tracing hepatic TG synthesis and VLDL-TG secretion in vivo by administering [¹³C18]oleic acid to rats. The [¹³C18]oleic acid-incorporated VLDL-TG was detected after one-step extraction without conventional separation of TG and recovery by derivatizing [¹³C18]oleic acid for detection. Using potent and selective DGAT1 inhibitors as pharmacological tools, we measured changes in [¹³C18]oleoyl-incorporated TG and DAG and demonstrated that DGAT1 inhibition significantly reduced [¹³C18]oleoyl-incorporated VLDL-TG. This DGAT1-selective assay will enable researchers to discern differences between the roles of DGAT1 and DGAT2 in TG synthesis in vitro and in vivo.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Enzyme Assays/methods , Liver/enzymology , Animals , Carbon Radioisotopes/metabolism , Cells, Cultured , Chromatography, Liquid , Hepatocytes/cytology , Hepatocytes/enzymology , Humans , Insecta/cytology , Insecta/enzymology , Insecta/virology , Kidney/cytology , Kidney/embryology , Kidney/enzymology , Lipoproteins, VLDL/chemistry , Lipoproteins, VLDL/metabolism , Male , Oleic Acid/metabolism , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Triglycerides/chemistry , Triglycerides/metabolism
15.
J Med Chem ; 52(23): 7432-45, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19731961

ABSTRACT

We have discovered two related chemical series of nonpeptide urotensin-II (U-II) receptor antagonists based on piperazino-phthalimide (5 and 6) and piperazino-isoindolinone (7) scaffolds. These structure types are distinctive from those of U-II receptor antagonist series reported in the literature. Antagonist 7a exhibited single-digit nanomolar potency in rat and human cell-based functional assays, as well as strong binding to the human U-II receptor. In advanced pharmacological testing, 7a blocked the effects of U-II in vitro in a rat aortic ring assay and in vivo in a rat ear-flush model. A discussion of U-II receptor antagonist pharmacophores is presented, and a specifically defined model is suggested from tricycle 13, which has a high degree of conformational constraint.


Subject(s)
Isoindoles/chemistry , Isoindoles/pharmacology , Phthalimides/chemistry , Phthalimides/pharmacology , Piperazines/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Aorta/drug effects , Aorta/physiology , CHO Cells , Cricetinae , Cricetulus , High-Throughput Screening Assays , Humans , Isoindoles/chemical synthesis , Male , Phthalimides/chemical synthesis , Piperazine , Rats , Rats, Wistar
16.
Bioorg Med Chem Lett ; 17(23): 6489-92, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17933531

ABSTRACT

Various 4-phenylpiperidine-benzoxazin-3-ones were synthesized and biologically evaluated as urotensin-II (U-II) receptor antagonists. Compound 12i was identified from in vitro evaluation as a low nanomolar antagonist against both rat and human U-II receptors. This compound showed in vivo efficacy in reversing the ear-flush response induced by U-II in rats.


Subject(s)
Benzoxazines/chemical synthesis , Piperidines/chemical synthesis , Receptors, G-Protein-Coupled/antagonists & inhibitors , Urotensins/metabolism , Animals , Benzoxazines/pharmacology , CHO Cells , Cricetinae , Cricetulus , Humans , Piperidines/pharmacology , Rats , Receptors, G-Protein-Coupled/physiology , Structure-Activity Relationship , Urotensins/antagonists & inhibitors , Urotensins/physiology
17.
J Biomol Screen ; 8(1): 65-71, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12854999

ABSTRACT

Target validation is one of rate-limiting steps in the modern drug discovery. The authors developed a strategy of combining adenovirus-mediated gene transfer for efficient target functionality validation, both in vivo and in vitro, with baculovirus expression to produce sufficient quantities of protein for high-throughput screening (HTS). The incorporation of green fluorescent protein (GFP) in the adenovirus vectors accelerates recombinant adenovirus plaque purification, whereas the use of epitope and affinity tags facilitates the identification and purification of recombinant protein. In this generalized scheme, the flexible modular design of viral vectors facilitates the transition between target validation and HTS. In the example presented, functional target validation in vivo was achieved by overexpressing the target gene in cell-based models and in the mouse cortex following adenovirus-mediated gene delivery. In this context, target overexpression resulted in the accumulation of a disease-related biomarker both in vitro and in vivo. A baculovirus-based expressional system was then generated to produce enough target protein for HTS. Thus, the use of these viral expression systems represents a generalized method for rapid target functionality validation and HTS assay development, which could be applied to numerous target candidates being elucidated in gene discovery programs.


Subject(s)
Adenoviridae , Genetic Vectors , Recombinant Proteins/analysis , Computational Biology
18.
Am J Physiol Renal Physiol ; 285(4): F792-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12783779

ABSTRACT

Recent studies have indicated that urotensin II (UII), a cyclic peptide, is vasoactive and may be involved in cardiovascular dysfunctions. It remains unknown, however, whether UII plays a role in the control of renal vascular tone and tubular function. In the present study, a continuous infusion of synthetic human UII (hUII) into the renal artery (RA) in anesthetized rats was found to increase renal blood flow (RBF) and urinary water and sodium excretion (UV and UNaV) in a dose-dependent manner. At a dose of 20 ng. kg-1. min-1, it increased RBF by 20% and UV and UNaV by 94 and 109%, respectively. Nitric oxide (NO) synthase inhibitor NG-nitro-l-arginine methyl ester (l-NAME) completely abolished hUII-induced increases in RBF and water/sodium excretion. In isolated, pressurized, and phenylephrine-precontracted small RA with internal diameter of approximately 200 microm, hUII produced a concentration-dependent vasodilation with a maximal response of 55% at 1.5 microM. l-NAME significantly blocked this hUII-induced vasodilation by 60%. In denuded RA, hUII had neither vasodilator nor vasoconstrictor effect. With the use of 4,5-diaminofluorescein diacetate-based fluorescence imaging analysis of NO levels, hUII (1 microM) was shown to double the NO levels within the endothelium of freshly dissected small RA, and l-NAME blocked this UII-induced production of endothelial NO. These results indicate that UII produces vasodilator and natriuretic effects in the kidney and that UII-induced vasodilation is associated with increased endothelial NO in the RA.


Subject(s)
Kidney/drug effects , Natriuretic Agents/pharmacology , Nitric Oxide/physiology , Urotensins/pharmacology , Vasodilator Agents/pharmacology , Animals , Blood Pressure/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Glomerular Filtration Rate/drug effects , Humans , In Vitro Techniques , Kidney/physiology , Male , Nitric Oxide/biosynthesis , Rats , Rats, Sprague-Dawley , Renal Artery/drug effects , Renal Artery/metabolism , Renal Circulation/drug effects , Vasodilation
19.
J Biol Chem ; 277(51): 49545-53, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12393916

ABSTRACT

Sphingosine kinase catalyzes the formation of sphingosine 1-phosphate, a lipid second messenger that has been implicated in a number of agonist-driven cellular responses including mitogenesis, anti-apoptosis, and expression of inflammatory molecules. Despite the importance of sphingosine kinase, very little is known regarding its structure or mechanism of catalysis. Moreover, sphingosine kinase does not contain recognizable catalytic or substrate-binding sites, based on sequence motifs found in other kinases. Here we have elucidated the nucleotide-binding site of human sphingosine kinase 1 (hSK1) through a combination of site-directed mutagenesis and affinity labeling with the ATP analogue, FSBA. We have shown that Gly(82) of hSK1 is involved in ATP binding since mutation of this residue to alanine resulted in an enzyme with an approximately 45-fold higher K(m)((ATP)). We have also shown that Lys(103) is important in catalysis since an alanine substitution of this residue ablates catalytic activity. Furthermore, we have shown that this residue is covalently modified by FSBA. Our data, combined with amino acid sequence comparison, suggest a motif of SGDGX(17-21)K is involved in nucleotide binding in the sphingosine kinases. This motif differs in primary sequence from all previously identified nucleotide-binding sites. It does, however, share some sequence and likely structural similarity with the highly conserved glycine-rich loop, which is known to be involved in anchoring and positioning the nucleotide in the catalytic site of many protein kinases.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine/analogs & derivatives , Nucleotides/chemistry , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Adenosine/pharmacology , Adenosine Triphosphate/pharmacology , Affinity Labels/pharmacology , Alanine/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Azides/pharmacology , Baculoviridae/metabolism , Binding Sites , Blotting, Western , Calmodulin/metabolism , Catalysis , Catalytic Domain , Cell Line , DNA, Complementary/metabolism , Gene Deletion , Humans , Insecta , Kinetics , Lysine/chemistry , Mass Spectrometry , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Photoaffinity Labels/pharmacology , Protein Binding , Protein Folding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...