Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
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.
TH Open ; 7(2): e97-e104, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37101592

ABSTRACT

Background Factor XIa (FXIa) is an emerging therapeutic target, and FXIa inhibition is a promising mechanism to improve therapeutic index over current anticoagulants. Milvexian (BMS-986177/JNJ-70033093) is an oral small-molecule FXIa inhibitor. Objective Milvexian's antithrombotic efficacy was characterized in a rabbit arteriovenous (AV) shunt model of venous thrombosis and compared with the factor Xa inhibitor apixaban and the direct thrombin inhibitor dabigatran. Methods The AV shunt model of thrombosis was conducted in anesthetized rabbits. Vehicle or drugs were administered as intravenous bolus plus a continuous infusion. Thrombus weight was the primary efficacy endpoint. Ex vivo activated partial thromboplastin time (aPTT), prothrombin time (PT), and thrombin time (TT) were measured as the pharmacodynamic responses. Results Milvexian dose dependently reduced thrombus weights by 34.3 ± 7.9, 51.6 ± 6.8 ( p < 0.01; n = 5), and 66.9 ± 4.8% ( p < 0.001; n = 6) versus vehicle at 0.25 + 0.17, 1.0 + 0.67, and 4.0 ± 2.68 mg/kg bolus + mg/kg/h infusion, respectively. Ex vivo clotting data supported a dose-dependent prolongation of aPTT (with 1.54-, 2.23-, and 3.12-fold increases from baseline upon the AV shunt start), but no changes in PT and TT. Dose-dependent inhibition in thrombus weight and clotting assays was also demonstrated for both apixaban and dabigatran as the references for the model validation. Conclusion Results demonstrate that milvexian is an effective anticoagulant for prevention of venous thrombosis in the rabbit model, which supports the utility of milvexian in venous thrombosis, as seen in the phase 2 clinical study.

3.
J Med Chem ; 65(15): 10419-10440, 2022 08 11.
Article in English | MEDLINE | ID: mdl-35862732

ABSTRACT

Activated factor XI (FXIa) inhibitors are promising novel anticoagulants with low bleeding risk compared with current anticoagulants. The discovery of potent FXIa inhibitors with good oral bioavailability has been challenging. Herein, we describe our discovery effort, utilizing nonclassical interactions to improve potency, cellular permeability, and oral bioavailability by enhancing the binding while reducing polar atoms. Beginning with literature-inspired pyridine N-oxide-based FXIa inhibitor 1, the imidazole linker was first replaced with a pyrazole moiety to establish a polar C-H···water hydrogen-bonding interaction. Then, structure-based drug design was employed to modify lead molecule 2d in the P1' and P2' regions, with substituents interacting with key residues through various nonclassical interactions. As a result, a potent FXIa inhibitor 3f (Ki = 0.17 nM) was discovered. This compound demonstrated oral bioavailability in preclinical species (rat 36.4%, dog 80.5%, and monkey 43.0%) and displayed a dose-dependent antithrombotic effect in a rabbit arteriovenous shunt model of thrombosis.


Subject(s)
Factor XIa , Pyridines , Animals , Anticoagulants/chemistry , Anticoagulants/pharmacology , Dogs , Drug Design , Factor XIa/metabolism , Pyridines/pharmacology , Rabbits , Rats
4.
Bioorg Med Chem Lett ; 30(17): 127387, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32738984

ABSTRACT

(2S,3R,4R,5S,6R)-2-Aryl-5,5-difluoro-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4-diols and (2S,3R,4R,5S,6R)-2-aryl-5-fluoro-5-methyl-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4-diols were discovered as dual inhibitors of sodium glucose co-transporter proteins (e.g. SGLT1 and SGLT2) through rational drug design, efficient synthesis, and in vitro and in vivo evaluation. Compound 6g demonstrated potent dual inhibitory activities (IC50 = 96 nM for SGLT1 and IC50 = 1.3 nM for SGLT2). It showed robust inhibition of blood glucose excursion in an oral glucose tolerance test (OGTT) in Sprague Dawley (SD) rats when dosed at both 1 mg/kg and 10 mg/kg orally. It also demonstrated postprandial glucose control in db/db mice when dosed orally at 10 mg/kg.


Subject(s)
Glucosides/chemistry , Hypoglycemic Agents/chemistry , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2/chemistry , Sodium-Glucose Transporter 2/metabolism , Animals , Blood Glucose/analysis , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Disease Models, Animal , Drug Design , Glucose Tolerance Test , Glucosides/metabolism , Glucosides/pharmacology , Glucosides/therapeutic use , Half-Life , Halogenation , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Microsomes/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/metabolism , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 29(20): 126668, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31519374

ABSTRACT

Type 2 diabetes mellitus (T2DM) is characterized by chronically elevated plasma glucose levels. The inhibition of glucagon-induced hepatic glucose output via antagonism of the glucagon receptor (GCGR) using a small-molecule antagonist is a promising mechanism for improving glycemic control in the diabetic state. The present work discloses the discovery of indazole-based ß-alanine derivatives as potent GCGR antagonists through an efficient enantioselective synthesis and structure-activity relationship (SAR) exploration and optimization. Compounds within this class exhibited excellent pharmacokinetic properties in multiple preclinical species. In an acute dog glucagon challenge test, compound 13K significantly inhibited glucagon-mediated blood glucose increase when dosed orally at 10 mg/kg.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/chemical synthesis , Indazoles/chemistry , Receptors, Glucagon/antagonists & inhibitors , beta-Alanine/chemical synthesis , Amino Acid Sequence , Animals , Blood Glucose/drug effects , Carbohydrate Metabolism , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Hypoglycemic Agents/pharmacokinetics , Liver/metabolism , Mice , Models, Molecular , Molecular Structure , Protein Binding , Rats , Structure-Activity Relationship , beta-Alanine/pharmacokinetics
6.
J Pharmacol Exp Ther ; 371(2): 375-384, 2019 11.
Article in English | MEDLINE | ID: mdl-31451508

ABSTRACT

JNJ-64179375 (JNJ-9375) is a recombinant human IgG4 monoclonal antibody engineered to mimic an IgA antibody that was identified in a patient who exhibited markedly prolonged clotting times but without spontaneous bleeding episodes over several years of follow-up. The crystal structure of the JNJ-9375 antigen-binding fragment/thrombin complex showed an almost identical binding mode to that of the patient IgA. In the current study, we characterized the in vitro and in vivo properties of JNJ-9375. Surface plasmon resonance studies demonstrated that JNJ-9375 binds to α-thrombin with high affinity and specificity (K D: 0.8 nM for human thrombin). JNJ-9375 produced concentration-dependent prolongation of in vitro clotting assays in human plasma, including thrombin time (TT), ecarin clotting time, prothrombin time, and activated partial thromboplastin time, with EC2X values of 4.4, 12.4, 172.6, and 202.7 µg/ml, respectively. JNJ-9375 inhibited thrombin-induced platelet aggregation in human plasma with an IC50 value of 52.6 nM (7.8 µg/ml) and produced concentration-dependent prolongation of reaction time tested by thromboelastography. JNJ-9375 pretreatment resulted in dose-dependent reduction in thrombus formation in the rat arteriovenous (AV) shunt model of thrombosis. Robust efficacy was observed at 0.3 mg/kg accompanied by 1.5× of TT. Bleeding was increased at 3 mg/kg in a rat tail transection bleeding model demonstrating a therapeutic index of 10× compared with 1× for apixaban in the same models. Our data suggest that thrombin exosite I inhibition is efficacious against thrombosis in a pretreatment prevention animal model. SIGNIFICANCE STATEMENT: JNJ-9375 is a novel, fully human monoclonal antibody that binds to the exosite I region of thrombin with high affinity and specificity. JNJ-9375 concentration dependently prolonged clotting times and inhibited thrombin-induced platelet aggregation in in vitro assays based on its mechanism of action. In an in vivo rat AV shunt model, JNJ-9375 prevented thrombus formation in a dose-dependent fashion while demonstrating reduced bleeding risk. The present study demonstrated the antithrombotic effects of inhibiting the exosite I region of thrombin when given in a prevention mode in preclinical animal models.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antithrombins/pharmacology , Immunoglobulin G/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Recombinant Proteins/pharmacology , Animals , Antibodies, Monoclonal/metabolism , Antithrombins/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Dose-Response Relationship, Drug , Humans , Immunoglobulin G/metabolism , Macaca fascicularis , Male , Mice , Platelet Aggregation Inhibitors/metabolism , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism
7.
Bioorg Med Chem Lett ; 28(21): 3446-3453, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30268701

ABSTRACT

A new series of (2S,3R,4R,5S,6R)-5-fluoro-6-(hydroxymethyl)-2-aryltetrahydro-2H-pyran-3,4-diols as dual inhibitors of sodium glucose co-transporter proteins (SGLTs) were disclosed. Two methods were developed to efficiently synthesize C5-fluoro-lactones 3 and 4, which are key intermediates to the C5-fluoro-hexose based C-aryl glucosides. Compound 2b demonstrated potent hSGLT1 and hSGLT2 inhibition (IC50 = 43 nM for SGLT1 and IC50 = 9 nM for SGLT2). It showed robust inhibition of blood glucose excursion in oral glucose tolerance test (OGTT) in Sprague Dawley (SD) rats and exerted pronounced antihyperglycemic effects in db/db mice and high-fat diet-fed ZDF rats when dosed orally at 10 mg/kg.


Subject(s)
Deoxyglucose/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/therapeutic use , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Administration, Oral , Animals , Blood Glucose/drug effects , Deoxyglucose/administration & dosage , Deoxyglucose/analogs & derivatives , Deoxyglucose/chemical synthesis , Drug Design , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Macaca fascicularis , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Rats, Sprague-Dawley , Rats, Zucker , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors/chemical synthesis , Sodium-Glucose Transporter 2 Inhibitors/chemistry , Structure-Activity Relationship
8.
J Pharmacol Exp Ther ; 365(3): 676-687, 2018 06.
Article in English | MEDLINE | ID: mdl-29674332

ABSTRACT

The sodium/glucose cotransporters (SGLT1 and SGLT2) transport glucose across the intestinal brush border and kidney tubule. Dual SGLT1/2 inhibition could reduce hyperglycemia more than SGLT2-selective inhibition in patients with type 2 diabetes. However, questions remain about altered gastrointestinal (GI) luminal glucose and tolerability, and this was evaluated in slc5a1-/- mice or with a potent dual inhibitor (compound 8; SGLT1 Ki = 1.5 ± 0.5 nM 100-fold greater potency than phlorizin; SGLT2 Ki = 0.4 ± 0.2 nM). 13C6-glucose uptake was quantified in slc5a1-/- mice and in isolated rat jejunum. Urinary glucose excretion (UGE), blood glucose (Sprague-Dawley rats), glucagon-like peptide 1 (GLP-1), and hemoglobin A1c (HbA1c) levels (Zucker diabetic fatty rats) were measured. Intestinal adaptation and rRNA gene sequencing was analyzed in C57Bl/6 mice. The blood 13C6-glucose area under the curve (AUC) was reduced in the absence of SGLT1 by 75% (245 ± 6 vs. 64 ± 6 mg/dl⋅h in wild-type vs. slc5a1-/- mice) and compound 8 inhibited its transport up to 50% in isolated rat jejunum. Compound 8 reduced glucose excursion more than SGLT2-selective inhibition (e.g., AUC = 129 ± 3 vs. 249 ± 5 mg/dl⋅h for 1 mg/kg compound 8 vs. dapagliflozin) with similar UGE but a lower renal glucose excretion threshold. In Zucker diabetic fatty rats, compound 8 decreased HbA1c and increased total GLP-1 without changes in jejunum SGLT1 expression, mucosal weight, or villus length. Overall, compound 8 (1 mg/kg for 6 days) did not increase cecal glucose concentrations or bacterial diversity in C57BL/6 mice. In conclusion, potent dual SGLT1/2 inhibition lowers blood glucose by reducing intestinal glucose absorption and the renal glucose threshold but minimally impacts the intestinal mucosa or luminal microbiota in chow-fed rodents.


Subject(s)
Blood Glucose/metabolism , Colon/drug effects , Colon/microbiology , Microbiota/drug effects , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Animals , Biodiversity , Colon/metabolism , Male , Mice , Rats , Sodium-Glucose Transporter 2 Inhibitors/metabolism
9.
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
10.
ACS Med Chem Lett ; 8(5): 560-565, 2017 May 11.
Article in English | MEDLINE | ID: mdl-28523111

ABSTRACT

We have discovered a novel series of tetrahydrobenzimidazoles 3 as TGR5 agonists. Initial structure-activity relationship studies with an assay that measured cAMP levels in murine enteroendocrine cells (STC-1 cells) led to the discovery of potent agonists with submicromolar EC50 values for mTGR5. Subsequent optimization through methylation of the 7-position of the core tetrahydrobenzimidazole ring resulted in the identification of potent agonists for both mTGR5 and hTGR5 (human enteroendocrine NCI-H716 cells). While the lead compounds displayed low to moderate exposure after oral dosing, they significantly reduced blood glucose levels in C57 BL/6 mice at 30 mg/kg and induced a 13-22% reduction in the area under the blood glucose curve (AUC)0-120 min in oral glucose tolerance tests (OGTT).

12.
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
13.
Arterioscler Thromb Vasc Biol ; 36(6): 1090-100, 2016 06.
Article in English | MEDLINE | ID: mdl-27127201

ABSTRACT

OBJECTIVE: Hyperlipidemia-induced endothelial cell (EC) activation is considered as an initial event responsible for monocyte recruitment in atherogenesis. However, it remains poorly defined what is the mechanism underlying hyperlipidemia-induced EC activation. Here, we tested a novel hypothesis that mitochondrial reactive oxygen species (mtROS) serve as signaling mediators for EC activation in early atherosclerosis. APPROACH AND RESULTS: Metabolomics and transcriptomics analyses revealed that several lysophosphatidylcholine (LPC) species, such as 16:0, 18:0, and 18:1, and their processing enzymes, including Pla2g7 and Pla2g4c, were significantly induced in the aortas of apolipoprotein E knockout mice during early atherosclerosis. Using electron spin resonance and flow cytometry, we found that LPC 16:0, 18:0, and 18:1 induced mtROS in primary human aortic ECs, independently of the activities of nicotinamide adenine dinucleotide phosphate oxidase. Mechanistically, using confocal microscopy and Seahorse XF mitochondrial analyzer, we showed that LPC induced mtROS via unique calcium entry-mediated increase of proton leak and mitochondrial O2 reduction. In addition, we found that mtROS contributed to LPC-induced EC activation by regulating nuclear binding of activator protein-1 and inducing intercellular adhesion molecule-1 gene expression in vitro. Furthermore, we showed that mtROS inhibitor MitoTEMPO suppressed EC activation and aortic monocyte recruitment in apolipoprotein E knockout mice using intravital microscopy and flow cytometry methods. CONCLUSIONS: ATP synthesis-uncoupled, but proton leak-coupled, mtROS increase mediates LPC-induced EC activation during early atherosclerosis. These results indicate that mitochondrial antioxidants are promising therapies for vascular inflammation and cardiovascular diseases.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Lysophosphatidylcholines/metabolism , Mitochondria/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Aorta/drug effects , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Calcium Signaling , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Gene Expression Profiling/methods , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lysophosphatidylcholines/pharmacology , Membrane Potential, Mitochondrial , Metabolomics/methods , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Oxidation-Reduction , Oxidative Stress/drug effects , Phenotype , Time Factors , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
14.
Biomark Res ; 1(1): 17, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-24252331

ABSTRACT

Insulin resistance associated with type 2 diabetes mellitus (T2DM), obesity, and atherosclerosis is a global health problem. A portfolio of abnormalities of metabolic and vascular homeostasis accompanies T2DM and obesity, which are believed to conspire to lead to accelerated atherosclerosis and premature death. The complexity of metabolic changes in the diseases presents challenges for a full understanding of the molecular pathways contributing to the development of these diseases. The recent advent of new technologies in this area termed "Metabolomics" may aid in comprehensive metabolic analysis of these diseases. Therefore, metabolomics has been extensively applied to the metabolites of T2DM, obesity, and atherosclerosis not only for the assessment of disease development and prognosis, but also for the biomarker discovery of disease diagnosis. Herein, we summarize the recent applications of metabolomics technology and the generated datasets in the metabolic profiling of these diseases, in particular, the applications of these technologies to these diseases at the cellular, animal models, and human disease levels. In addition, we also extensively discuss the mechanisms linking the metabolic profiling in insulin resistance, T2DM, obesity, and atherosclerosis, with a particular emphasis on potential roles of increased production of reactive oxygen species (ROS) and mitochondria dysfunctions.

15.
Front Biosci (Landmark Ed) ; 17(6): 2327-49, 2012 06 01.
Article in English | MEDLINE | ID: mdl-22652782

ABSTRACT

Endothelial progenitor cells (EPCs) are involved in the maintenance of endothelial homoeostasis and in the process of new vessel formation. Experimental and clinical studies have shown that atherosclerosis is associated with reduced numbers and dysfunction of EPCs; and that medications alone are able to partially reverse the impairment of EPCs in patients with atherosclerosis. Therefore, novel EPC-based therapies may provide enhancement in restoring EPCs' population and improvement of vascular function. Here, for a better understanding of the molecular mechanisms underlying EPC impairment in atherosclerosis, we provide a comprehensive overview on EPC characteristics, phenotypes, and the signaling pathways underlying EPC impairment in atherosclerosis.


Subject(s)
Adult Stem Cells/pathology , Atherosclerosis/pathology , Endothelial Cells/pathology , Adult Stem Cells/physiology , Animals , Atherosclerosis/physiopathology , Atherosclerosis/therapy , Cell Differentiation , Endothelial Cells/physiology , Hematopoietic Stem Cell Mobilization , Humans , Inflammation/pathology , Inflammation/physiopathology , Macrophages/pathology , Macrophages/physiology , Mice , Models, Cardiovascular , Monocytes/pathology , Monocytes/physiology , Neovascularization, Physiologic , Regeneration , Signal Transduction , Stem Cell Transplantation
16.
PLoS One ; 7(2): e30555, 2012.
Article in English | MEDLINE | ID: mdl-22355316

ABSTRACT

BACKGROUND: Canagliflozin is a sodium glucose co-transporter (SGLT) 2 inhibitor in clinical development for the treatment of type 2 diabetes mellitus (T2DM). METHODS: (14)C-alpha-methylglucoside uptake in Chinese hamster ovary-K cells expressing human, rat, or mouse SGLT2 or SGLT1; (3)H-2-deoxy-d-glucose uptake in L6 myoblasts; and 2-electrode voltage clamp recording of oocytes expressing human SGLT3 were analyzed. Graded glucose infusions were performed to determine rate of urinary glucose excretion (UGE) at different blood glucose (BG) concentrations and the renal threshold for glucose excretion (RT(G)) in vehicle or canagliflozin-treated Zucker diabetic fatty (ZDF) rats. This study aimed to characterize the pharmacodynamic effects of canagliflozin in vitro and in preclinical models of T2DM and obesity. RESULTS: Treatment with canagliflozin 1 mg/kg lowered RT(G) from 415±12 mg/dl to 94±10 mg/dl in ZDF rats while maintaining a threshold relationship between BG and UGE with virtually no UGE observed when BG was below RT(G). Canagliflozin dose-dependently decreased BG concentrations in db/db mice treated acutely. In ZDF rats treated for 4 weeks, canagliflozin decreased glycated hemoglobin (HbA1c) and improved measures of insulin secretion. In obese animal models, canagliflozin increased UGE and decreased BG, body weight gain, epididymal fat, liver weight, and the respiratory exchange ratio. CONCLUSIONS: Canagliflozin lowered RT(G) and increased UGE, improved glycemic control and beta-cell function in rodent models of T2DM, and reduced body weight gain in rodent models of obesity.


Subject(s)
Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Experimental/drug therapy , Glucosides/therapeutic use , Hyperglycemia/drug therapy , Kidney/physiopathology , Thiophenes/therapeutic use , Animals , CHO Cells , Canagliflozin , Cells, Cultured , Cricetinae , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Glucose Tolerance Test , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Kidney/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Rats , Rats, Zucker , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2/genetics , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2 Inhibitors , Weight Gain/drug effects
17.
Bioorg Med Chem Lett ; 22(4): 1750-5, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22264481

ABSTRACT

The design and synthesis of a second generation GPR119-agonist clinical candidate for the treatment of diabetes is described. Compound 16 (APD597, JNJ-38431055) was selected for preclinical development based on a good balance between agonist potency, intrinsic activity and in particular on its good solubility and reduced drug-drug interaction potential. In addition, extensive in vivo studies showed a more favorable metabolic profile that may avoid the generation of long lasting metabolites with the potential to accumulate in clinical studies.


Subject(s)
Drug Discovery , Hypoglycemic Agents/chemistry , Piperidines/chemistry , Piperidines/pharmacokinetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Receptors, G-Protein-Coupled/agonists , Animals , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Humans , Hypoglycemic Agents/pharmacokinetics , Mice , Mice, Inbred C57BL , Molecular Structure , Rats , Rats, Sprague-Dawley
18.
J Med Chem ; 52(23): 7544-69, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19366247

ABSTRACT

As part of a program aimed at the development of selective estrogen receptor modulators (SERMs), novel chromene scaffolds, benzopyranobenzoxapanes, were discovered. Many compounds showed binding affinity as low as 1.6-200 nM, displayed antagonist behaviors in the MCF-7 human breast adenocarcinoma cell line as well in Ishikawa cell line with IC(50) values in the range 0.2-360 nM. On the basis of the side chain substitution, various compounds demonstrated strong inhibitory activity in anti-uterotropic assay. Compound 7-(R) and its major metabolites 5-(R) and 6-(R) were evaluated in several in vivo models of estrogen action. Relative to a full estrogen agonist (ethynyl estradiol) and the SERM raloxifene, 7-(R) was found to be a potent SERM that behaved as antagonist in the uterus and exhibited estrogen agonistic activity on bone, plasma lipids, hot flush, and vagina. The overall pharmacokinetic profile and stability were significantly improved compared to those of the phase 2 development compound 9-(R).


Subject(s)
Benzopyrans/chemistry , Benzopyrans/pharmacology , Postmenopause/drug effects , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacology , Animals , Benzopyrans/chemical synthesis , Benzopyrans/therapeutic use , Bone Resorption/drug therapy , Cell Line, Tumor , Cholesterol/blood , Drug Evaluation, Preclinical , Drug Stability , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Hot Flashes/drug therapy , Humans , Organ Size/drug effects , Ovariectomy , Postmenopause/blood , Rats , Selective Estrogen Receptor Modulators/chemical synthesis , Selective Estrogen Receptor Modulators/therapeutic use , Structure-Activity Relationship , Substrate Specificity , Uterus/pathology , Vagina/drug effects , Vagina/metabolism
19.
Exp Physiol ; 92(1): 161-73, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17053038

ABSTRACT

The action potential of Purkinje fibres is markedly shortened by tetrodotoxin, suggesting the possibility that a slowly inactivating sodium current might flow during the plateau. The aim of the present experiments was to investigate, in canine cardiac Purkinje single cells by means of a whole cell patch clamp technique, whether a sodium current slowly inactivates at less negative potentials and (if so) some of its distinctive characteristics. The results showed that a 500 ms depolarizing step from a holding potential of -90 mV to -50 mV induced the fast inward current I(Na) (labelled here I(Na1)). With steps to -40 mV or less negative values, a slowly decaying component (tentatively labelled here I(Na2)) appeared, which peaked at -30 to -20 mV and decayed slowly and incompletely during the 500 ms steps. The I(Na2) was present also during steps to -10 mV, but then the transient outward current (I(to)) appeared. When the holding potential (V(h)) was decreased to -60 to -50 mV, I(Na2) disappeared even if a small I(Na1) might still be present. Tetrodotoxin (30 mum), lignocaine (100 mum) and cadmium (0.2 mm; but not manganese, 1 mm) blocked I(Na2). During fast depolarizing ramps, the rapid inactivation of I(Na1) was followed by a negative slope region. During repolarizing ramps, a region of positive slope was present, whereas I(Na1) was absent. At less negative values of V(h), the amplitude of the negative and positive slopes became gradually smaller. Gradually faster ramps increased the magnitude of the negative slope, and tetrodotoxin (30 mum) reduced or abolished it. Thus, Purkinje cells have a slowly decaying inward current owing to Na(+) entry (I(Na2)) that is different in several ways from the fast I(Na1) and that appears important for the duration of the plateau.


Subject(s)
Ion Channel Gating , Purkinje Fibers/metabolism , Sodium Channels/metabolism , Sodium/metabolism , Action Potentials , Animals , Cadmium/metabolism , Dogs , Heart Conduction System/metabolism , In Vitro Techniques , Ion Channel Gating/drug effects , Kinetics , Lidocaine/pharmacology , Manganese/metabolism , Patch-Clamp Techniques , Purkinje Fibers/drug effects , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Tetrodotoxin/pharmacology
20.
J Med Chem ; 49(11): 3056-9, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16722623

ABSTRACT

A novel SERM (selective estrogen receptor modulators), 1-(R), a chromene-derived bisbenzopyran, was discovered to alleviate hot flushes and effectively increase vaginal fluidity in rats. Moreover, 1-(R) was found to have beneficial effects on plasma cholesterol and bone metabolism while maintaining antiestrogenic activity in the uterus. The biological profile of its enantiomer 1-(S) was also evaluated.


Subject(s)
Benzopyrans/chemical synthesis , Body Fluids/drug effects , Hot Flashes/drug therapy , Selective Estrogen Receptor Modulators/chemical synthesis , Vagina/drug effects , Vaginal Diseases/drug therapy , Animals , Benzopyrans/chemistry , Benzopyrans/pharmacology , Bone Density/drug effects , Cell Line , Cholesterol/blood , Female , Humans , Organ Size/drug effects , Ovariectomy , Rats , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacology , Stereoisomerism , Uterus/drug effects , Vagina/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL