Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
bioRxiv ; 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37333249

ABSTRACT

Idiopathic Pulmonary Fibrosis (IPF) is a chronic parenchymal lung disease characterized by repetitive alveolar cell injury, myofibroblast proliferation, and excessive extracellular matrix deposition for which unmet need persists for effective therapeutics. The bioactive eicosanoid, prostaglandin F2α, and its cognate receptor FPr (Ptfgr) are implicated as a TGFß1 independent signaling hub for IPF. To assess this, we leveraged our published murine PF model (IER - SftpcI73T) expressing a disease-associated missense mutation in the surfactant protein C (Sftpc) gene. Tamoxifen treated IER-SftpcI73T mice develop an early multiphasic alveolitis and transition to spontaneous fibrotic remodeling by 28 days. IER-SftpcI73T mice crossed to a Ptgfr null (FPr-/-) line showed attenuated weight loss and gene dosage dependent rescue of mortality compared to FPr+/+ cohorts. IER-SftpcI73T/FPr-/- mice also showed reductions in multiple fibrotic endpoints for which administration of nintedanib was not additive. Single cell RNA sequencing, pseudotime analysis, and in vitro assays demonstrated Ptgfr expression predominantly within adventitial fibroblasts which were reprogrammed to an "inflammatory/transitional" cell state in a PGF2α/FPr dependent manner. Collectively, the findings provide evidence for a role for PGF2α signaling in IPF, mechanistically identify a susceptible fibroblast subpopulation, and establish a benchmark effect size for disruption of this pathway in mitigating fibrotic lung remodeling.

2.
JCI Insight ; 8(24)2023 Dec 22.
Article in English | MEDLINE | ID: mdl-37934604

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic parenchymal lung disease characterized by repetitive alveolar cell injury, myofibroblast proliferation, and excessive extracellular matrix deposition for which unmet need persists for effective therapeutics. The bioactive eicosanoid, prostaglandin F2α, and its cognate receptor FPr (Ptgfr) are implicated as a TGF-ß1-independent signaling hub for IPF. To assess this, we leveraged our published murine PF model (IER-SftpcI73T) expressing a disease-associated missense mutation in the surfactant protein C (Sftpc) gene. Tamoxifen-treated IER-SftpcI73T mice developed an early multiphasic alveolitis and transition to spontaneous fibrotic remodeling by 28 days. IER-SftpcI73T mice crossed to a Ptgfr-null (FPr-/-) line showed attenuated weight loss and gene dosage-dependent rescue of mortality compared with FPr+/+ cohorts. IER-SftpcI73T/FPr-/- mice also showed reductions in multiple fibrotic endpoints for which administration of nintedanib was not additive. Single-cell RNA-Seq, pseudotime analysis, and in vitro assays demonstrated Ptgfr expression predominantly within adventitial fibroblasts, which were reprogrammed to an "inflammatory/transitional" cell state in a PGF2α /FPr-dependent manner. Collectively, the findings provide evidence for a role for PGF2α signaling in IPF, mechanistically identify a susceptible fibroblast subpopulation, and establish a benchmark effect size for disruption of this pathway in mitigating fibrotic lung remodeling.


Subject(s)
Dinoprost , Idiopathic Pulmonary Fibrosis , Mice , Animals , Dinoprost/metabolism , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Fibrosis , Population Dynamics
3.
Bioorg Med Chem Lett ; 22(13): 4341-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22658862

ABSTRACT

Long chain L-2-hydroxy acid oxidase 2 (Hao2) is a peroxisomal enzyme expressed in the kidney and the liver. Hao2 was identified as a candidate gene for blood pressure (BP) quantitative trait locus (QTL) but the identity of its physiological substrate and its role in vivo remains largely unknown. To define a pharmacological role of this gene product, we report the development of selective inhibitors of Hao2. We identified pyrazole carboxylic acid hits 1 and 2 from screening of a compound library. Lead optimization of these hits led to the discovery of 15-XV and 15-XXXII as potent and selective inhibitors of rat Hao2. This report details the structure activity relationship of the pyrazole carboxylic acids as specific inhibitors of Hao2.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Carboxylic Acids/chemistry , Enzyme Inhibitors/chemistry , Pyrazoles/chemistry , Thiophenes/chemistry , Alcohol Oxidoreductases/metabolism , Animals , Binding Sites , Carboxylic Acids/chemical synthesis , Carboxylic Acids/pharmacokinetics , Computer Simulation , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Humans , Kidney/enzymology , Kidney/metabolism , Liver/enzymology , Liver/metabolism , Protein Structure, Tertiary , Pyrazoles/chemical synthesis , Pyrazoles/therapeutic use , Rats , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/therapeutic use
4.
Bioorg Med Chem Lett ; 21(12): 3596-602, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21571530

ABSTRACT

GPR91, a 7TM G-Protein-Coupled Receptor, has been recently deorphanized with succinic acid as its endogenous ligand. Current literature indicates that GPR91 plays role in various pathophysiology including renal hypertension, autoimmune disease and retinal angiogenesis. Starting from a small molecule high-throughput screening hit 1 (hGPR91 IC(50): 0.8 µM)-originally synthesized in Merck for Bradykinin B(1) Receptor (BK(1)R) program, systematic structure-activity relationship study led us to discover potent and selective hGPR91 antagonists e.g. 2c, 4c, and 5 g (IC(50): 7-35 nM; >1000 fold selective against hGPR99, a closest related GPCR; >100 fold selective in Drug Matrix screening). This initial work also led to identification of two structurally distinct and orally bio-available lead compounds: 5g (%F: 26) and 7e (IC(50): 180 nM; >100 fold selective against hGPR99; %F: 87). A rat pharmacodynamic assay was developed to characterize the antagonists in vivo using succinate induced increase in blood pressure. Using two representative antagonists, 2c and 4c, the GPR91 target engagement was subsequently demonstrated using the designed pharmacodynamic assay.


Subject(s)
Drug Discovery , Receptors, G-Protein-Coupled/antagonists & inhibitors , Small Molecule Libraries/chemical synthesis , Administration, Oral , Animals , Inhibitory Concentration 50 , Male , Molecular Structure , Rats , Rats, Wistar , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
5.
Sci Rep ; 11(1): 2118, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483531

ABSTRACT

Lung fibrosis, or the scarring of the lung, is a devastating disease with huge unmet medical need. There are limited treatment options and its prognosis is worse than most types of cancer. We previously discovered that MK-0429 is an equipotent pan-inhibitor of αv integrins that reduces proteinuria and kidney fibrosis in a preclinical model. In the present study, we further demonstrated that MK-0429 significantly inhibits fibrosis progression in a bleomycin-induced lung injury model. In search of newer integrin inhibitors for fibrosis, we characterized monoclonal antibodies discovered using Adimab's yeast display platform. We identified several potent neutralizing integrin antibodies with unique human and mouse cross-reactivity. Among these, Ab-31 blocked the binding of multiple αv integrins to their ligands with IC50s comparable to those of MK-0429. Furthermore, both MK-0429 and Ab-31 suppressed integrin-mediated cell adhesion and latent TGFß activation. In IPF patient lung fibroblasts, TGFß treatment induced profound αSMA expression in phenotypic imaging assays and Ab-31 demonstrated potent in vitro activity at inhibiting αSMA expression, suggesting that the integrin antibody is able to modulate TGFß action though mechanisms beyond the inhibition of latent TGFß activation. Together, our results highlight the potential to develop newer integrin therapeutics for the treatment of fibrotic lung diseases.


Subject(s)
Antibodies/metabolism , Fibroblasts/metabolism , Integrin alphaV/metabolism , Pulmonary Fibrosis/metabolism , Animals , Antibodies/immunology , Bleomycin , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Fibroblasts/cytology , Humans , Integrin alphaV/immunology , Male , Mice, Inbred C57BL , Naphthyridines/pharmacology , Propionates/pharmacology , Protein Binding , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/prevention & control
6.
Physiol Genomics ; 42A(1): 24-32, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20587620

ABSTRACT

Hypertension is a condition with major cardiovascular and renal complications, affecting nearly a billion patients worldwide. Few validated gene targets are available for pharmacological intervention, so there is a need to identify new biological pathways regulating blood pressure and containing novel targets for treatment. The genetically hypertensive "blood pressure high" (BPH), normotensive "blood pressure normal" (BPN), and hypotensive "blood pressure low" (BPL) inbred mouse strains are an ideal system to study differences in gene expression patterns that may represent such biological pathways. We profiled gene expression in liver, heart, kidney, and aorta from BPH, BPN, and BPL mice and determined which biological processes are enriched in observed organ-specific signatures. As a result, we identified multiple biological pathways linked to blood pressure phenotype that could serve as a source of candidate genes causal for hypertension. To distinguish in the kidney signature genes whose differential expression pattern may cause changes in blood pressure from those genes whose differential expression pattern results from changes in blood pressure, we integrated phenotype-associated genes into Genetic Bayesian networks. The integration of data from gene expression profiling and genetics networks is a valuable approach to identify novel potential targets for the pharmacological treatment of hypertension.


Subject(s)
Gene Expression Profiling , Hypertension/genetics , Myocardium/metabolism , Animals , Aorta/metabolism , Blood Pressure/genetics , Disease Models, Animal , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred Strains , Models, Biological , Oligonucleotide Array Sequence Analysis
7.
Bioorg Med Chem Lett ; 20(11): 3372-5, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20452209

ABSTRACT

A series of pyrazolyl propionyl cyclohexenamides were discovered as full agonists for the high affinity niacin receptor GPR109A. The structure-activity relationship (SAR) studies were aimed to improve activity on GPR109A, reduce Cytochrome P450 2C8 (CYP2C8) and Cytochrome P450 2C9 (CYP2C9) inhibition, reduce serum shift and improve pharmacokinetic (PK) profiles.


Subject(s)
Amides/chemistry , Cyclohexenes/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Cyclohexenes/chemistry , Cyclohexenes/pharmacokinetics , Mice , Rats , Receptors, Nicotinic , Structure-Activity Relationship
8.
Cell Rep Med ; 1(4): 100056, 2020 07 21.
Article in English | MEDLINE | ID: mdl-33205063

ABSTRACT

Fibrosis, or the accumulation of extracellular matrix, is a common feature of many chronic diseases. To interrogate core molecular pathways underlying fibrosis, we cross-examine human primary cells from various tissues treated with TGF-ß, as well as kidney and liver fibrosis models. Transcriptome analyses reveal that genes involved in fatty acid oxidation are significantly perturbed. Furthermore, mitochondrial dysfunction and acylcarnitine accumulation are found in fibrotic tissues. Substantial downregulation of the PGC1α gene is evident in both in vitro and in vivo fibrosis models, suggesting a common node of metabolic signature for tissue fibrosis. In order to identify suppressors of fibrosis, we carry out a compound library phenotypic screen and identify AMPK and PPAR as highly enriched targets. We further show that pharmacological treatment of MK-8722 (AMPK activator) and MK-4074 (ACC inhibitor) reduce fibrosis in vivo. Altogether, our work demonstrate that metabolic defect is integral to TGF-ß signaling and fibrosis.


Subject(s)
Fibrosis/genetics , Fibrosis/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Adenylate Kinase/metabolism , Animals , Benzimidazoles/pharmacology , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Gene Expression/genetics , Gene Expression Profiling/methods , Humans , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Specificity/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Transcriptome/genetics , Transforming Growth Factor beta/metabolism
9.
Biochem Biophys Res Commun ; 377(3): 987-91, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-18952058

ABSTRACT

Heavy exercise or oxygen deficit often links with higher levels of arterial lactate and lower levels of plasma free fatty acids (FFA). Treatment with lactate reduces circulating levels of FFA in vivo and lipolysis in adipose tissues in vitro. However, the underlying mechanism has remained unclear. Here we employ pharmacological and genetic approaches to show that GPR81, an orphan G-protein-coupled receptor with relatively restricted expression in the adipose tissues, functions as a receptor for lactate and can mediate an anti-lipolytic effect of lactate. GPR81 may thus function as a sensor of lactate that can modulate the FFA pool under exercise or conditions of oxygen deficit.


Subject(s)
Adipose Tissue/metabolism , Fatty Acids, Nonesterified/metabolism , Lactic Acid/metabolism , Lipolysis , Receptors, G-Protein-Coupled/physiology , Adipose Tissue/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Lactic Acid/pharmacology , Ligands , Lipolysis/drug effects , Lipolysis/genetics , Mice , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, Nicotinic/genetics , Receptors, Nicotinic/physiology , Transfection
10.
Bioorg Med Chem Lett ; 18(18): 4963-7, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18760600

ABSTRACT

A homology model of the nicotinic acid receptor GPR109A was constructed based on the X-ray crystal structure of bovine rhodopsin. An HTS hit was docked into the homology model. Characterization of the binding pocket by a grid-based surface calculation of the docking model suggested that a larger hydrophobic body plus a polar tail would improve interaction between the ligand and the receptor. The designed compounds were synthesized, and showed significantly improved binding affinity and activation of GPR109A.


Subject(s)
Models, Molecular , Nicotinic Agonists/chemical synthesis , Nicotinic Agonists/pharmacology , Receptors, G-Protein-Coupled/agonists , ortho-Aminobenzoates/chemical synthesis , ortho-Aminobenzoates/pharmacology , Binding Sites , Combinatorial Chemistry Techniques , Humans , Molecular Structure , Niacin/metabolism , Nicotinic Agonists/chemistry , Receptors, Nicotinic , Structure-Activity Relationship , ortho-Aminobenzoates/chemistry
11.
J Med Chem ; 50(25): 6303-6, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-17994679

ABSTRACT

Biaryl anthranilides are reported as potent and selective full agonists for the high affinity niacin receptor GPR109A. The SAR presented outlines approaches to reduce serum shift and both CYPCYP2C8 and CYP2C9 liabilities, while improving PK and maintaining excellent receptor activity. Compound 2i exhibited good in vivo antilipolytic efficacy while providing a significantly improved therapeutic index over vasodilation (flushing) with respect to niacin in the mouse model.


Subject(s)
Receptors, G-Protein-Coupled/agonists , ortho-Aminobenzoates/chemical synthesis , Amides/chemical synthesis , Amides/pharmacokinetics , Amides/pharmacology , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP2C9 , Humans , In Vitro Techniques , Mice , Microsomes, Liver/metabolism , Radioligand Assay , Receptors, Nicotinic , Structure-Activity Relationship , ortho-Aminobenzoates/pharmacokinetics , ortho-Aminobenzoates/pharmacology
13.
Thromb Res ; 145: 133-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27318768

ABSTRACT

Platelet activation plays a crucial role in hemostasis and thrombosis. Thrombin, the most potent stimulus of platelet activation, mediates platelet activation via the protease activated receptors (PARs). The platelet PAR repertoire in mediating thrombin's action differs across species. Only nonhuman primate (NHP) platelet activation is known to be similar to humans, mediated by PAR1 and PAR4, hence limiting translational in vivo studies of PAR's role in thrombosis and hemostasis to NHPs. Earlier studies have demonstrated a range of distinct in vitro activities of PAR1 and 4 in platelet activation yet the implications of these events in vivo is unclear. The objective of this study is to investigate and compare the roles of PAR1 and PAR4 in hemostasis and thrombosis in a relevant animal species. NHP models for pharmacokinetic, ex vivo platelet aggregation responses, FeCI3 injury-mediated arterial thrombosis and template bleeding were developed in Cynomolgus Macaques. Potent and selective small molecule antagonists of PAR1 and PAR4 were characterized in an array of in vitro assays, and subsequently examined head-to-head in the NHP models. Treatment of NHPs with antagonists of PAR1 or PAR4 both resulted in strong inhibition of ex vivo platelet aggregation. At doses that led to similar inhibition of platelet aggregation, animals treated with the PAR4 antagonist showed similar levels of anti-thrombotic efficacy, but longer bleeding times, compared to animals treated with the PAR1 antagonist. These findings suggest that PAR1 antagonism will likely produce a larger therapeutic index (ie. a larger anti-thrombotic efficacy over bleeding risk margin) than PAR4 antagonism.


Subject(s)
Hemorrhage/drug therapy , Platelet Activation/drug effects , Receptors, Thrombin/antagonists & inhibitors , Thrombosis/drug therapy , Animals , Hemorrhage/etiology , Macaca fascicularis
14.
Biochim Biophys Acta ; 1581(3): 100-8, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-12020637

ABSTRACT

A rapid in vitro assay was developed for monitoring protein-mediated cholesterol monomerization from bile acid aggregates. This assay uses a fluorescent cholesterol analog, 22-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-23,24-bisnor-5-cholen-3 beta-ol (NBD-cholesterol), which was shown to be absorbed by hamster in a fashion similar to cholesterol. The fluorescence of aggregates of NBD-cholesterol was strongly quenched in 2.5 mM of taurocholic acid. Addition of proteins from enterocytes of hamster small intestine led to a time- and dose-dependent dequenching of NBD-cholesterol fluorescence. Comparable dequenching can be detected with SDS and appears to involve monomerization of the NBD-cholesterol. Purification of enterocyte extract by sequential chromatography revealed an approximately 140-kDa protein complex (p140) able to mediate the monomerization of NBD-cholesterol. Each p140 complex mediated monomerization of 2.7 NBD-cholesterol molecules. The p140 complex appeared to be formed by dimerization of two approximately 58-kDa molecules since SDS-PAGE revealed a single dominant band at 58 kDa (p58). Protein sequence analysis suggested that p58 is protein-disulfide isomerase (PDI), and this conclusion was confirmed by cloning of hamster PDI, and detection of PDI enzyme activity in the purified fraction. Additional studies with either pure PDI or lysates of cells transfected with hamster PDI showed that PDI by itself was not sufficient for monomerizing cholesterol. Further, despite a similar mobility on SDS-PAGE (approximately 58 kDa), the p140 complex appeared approximately 45-kDa larger than pure PDI (approximately 95 kDa) when analyzed by a gel-filtration chromatography. The p140 complex may thus contain an unidentified molecule(s) in addition to PDI that may contribute importantly to cholesterol monomerization.


Subject(s)
4-Chloro-7-nitrobenzofurazan/analogs & derivatives , 4-Chloro-7-nitrobenzofurazan/chemistry , 4-Chloro-7-nitrobenzofurazan/metabolism , Cholesterol/analogs & derivatives , Cholesterol/chemistry , Cholesterol/metabolism , Intestine, Small/enzymology , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Amino Acid Sequence , Animals , Bile Acids and Salts/metabolism , Cricetinae , Humans , Macromolecular Substances , Male , Mesocricetus , Models, Chemical , Molecular Sequence Data , Molecular Weight , Polymers/metabolism , Spectrometry, Fluorescence , Time Factors
15.
J Comp Neurol ; 481(4): 403-15, 2005 Jan 24.
Article in English | MEDLINE | ID: mdl-15593342

ABSTRACT

The development of the cerebellar cortex depends on intrinsic genetic programs and orchestrated cell-cell/cell-matrix interactions. Matrix metalloproteinases (MMPs) are proteolytic enzymes that play an important role in these interactions. MMP-2 and MMP-9 are involved in diverse neuronal functions including migration, process extension, and synaptic plasticity. We investigated the spatiotemporal pattern of expression/activity of MMP-2/MMP-9 in the developing cerebellum and their role in the histogenesis of the cerebellar cortex. The levels of transcripts of MMP-2/MMP-9 were measured with real-time quantitative polymerase chain reaction. An initial decrease in MMP-2/MMP-9 transcripts was observed between postnatal days 3 (PD3) and PD6, and the mRNA levels remained relatively constant thereafter. Zymographic analysis revealed that the expression/activity of MMP-2/MMP-9 persisted longer than their transcripts; the downregulation occurred around PD9, suggesting a mechanism of translational or post-translational regulation. The gelatinase activity was localized in the external granule layer (EGL) and the internal granule layer during PD3-PD12. The immunoreactivity of MMP-2 was mainly localized in the EGL, the Bergmann glial fibers, and the Purkinje cell layer (PCL), whereas MMP-9 immunoreactivity was detected intensively in the PCL and the extracellular space of the molecular layer. Expression of MMP-9 was relatively weak in the EGL. The immunoreactivity of MMP-2/MMP-9 became undetectable after PD21. A similar expression pattern of MMP-2/MMP-9 was observed in organotypic cerebellar slice cultures. Exposure of organotypic slices to a specific MMP-2/MMP-9 inhibitor significantly increased the thickness of the EGL and concurrently decreased the number of migrating granule neurons in the molecular layer. Thus, MMP-2 and MMP-9 play a role in the postnatal cerebellar morphogenesis.


Subject(s)
Cerebellar Cortex/enzymology , Extracellular Matrix/enzymology , Gene Expression Regulation, Developmental/physiology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Cell Differentiation/physiology , Cerebellar Cortex/cytology , Cerebellar Cortex/embryology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Organ Culture Techniques , Organogenesis/physiology , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
16.
Atherosclerosis ; 181(2): 251-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16039278

ABSTRACT

Diabetes mellitus is one of the major risk factors associated with atherosclerosis and coronary heart disease but the mechanistic links between the disease and atherosclerosis are not well understood. In this study, we investigated the effect of the deletion of the long-form leptin receptor on the progression of atherosclerosis in ApoE-/- mouse. ApoE-/-;db/db double knockout mice as well as ApoE-/-;db/+ and ApoE-/- littermates were generated by crossing ApoE-/- and db/+ mice. On a regular chow diet, ApoE-/-;db/db mice at 20 weeks of age exhibited features typical of type 2 diabetes: obesity, hyperglycemia, hyperinsulinemia and dyslipidemia and had significantly accelerated atherosclerosis compared with their age-matched ApoE-/- littermates as assessed by either the percentage of the aorta bearing lesion (5.3+/-0.9% for ApoE-/-;db/db versus 1.5+/-0.5% for ApoE-/-) or by aortic lipid content ( approximately 1.5-2-fold increase in free cholesterol and approximately 3-4-fold increase in cholesteryl ester). The atherosclerosis in these ApoE-/-;db/db mice was further accelerated by feeding mice with a Western diet and markedly inhibited by fenofibrate with a 2.5- and 5.3-fold reduction of the lesion in male and female mice, respectively. The results from this study demonstrate that type 2 diabetes can accelerate atherogenesis in mice. This mouse model may provide insight into the mechanistic link between type 2 diabetes and atherosclerosis as well as serve as a valuable tool for evaluating therapeutics.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/genetics , Disease Models, Animal , Hypercholesterolemia/genetics , Mice, Knockout , Receptors, Cell Surface/genetics , Animals , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cholesterol/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Female , Fenofibrate/pharmacology , Hypercholesterolemia/drug therapy , Hypercholesterolemia/physiopathology , Hypolipidemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Receptors, Leptin , Severity of Illness Index
17.
J Med Chem ; 48(17): 5589-99, 2005 Aug 25.
Article in English | MEDLINE | ID: mdl-16107159

ABSTRACT

The design and synthesis of a novel class of 2,3-dihydrobenzofuran-2-carboxylic acids as highly potent and subtype-selective PPARalpha agonists are reported. Systematic study of structure-activity relationships has identified several key structural elements within this class for maintaining the potency and subtype selectivity. Select compounds were evaluated in animal models of dyslipidemia using Syrian hamsters and male Beagle dogs, and all these compounds displayed excellent cholesterol- and triglyceride-lowering activity at dose levels that were much lower than the marketed weak PPARalpha agonist fenofibrate.


Subject(s)
Benzofurans/chemical synthesis , Carboxylic Acids/chemical synthesis , Hypolipidemic Agents/chemical synthesis , PPAR alpha/agonists , Animals , Benzofurans/chemistry , Benzofurans/pharmacology , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Cholesterol/blood , Cricetinae , Dogs , Humans , Hyperlipidemias/drug therapy , Hypolipidemic Agents/chemistry , Hypolipidemic Agents/pharmacology , In Vitro Techniques , Male , Mesocricetus , Molecular Conformation , PPAR alpha/genetics , Radioligand Assay , Stereoisomerism , Structure-Activity Relationship , Transcriptional Activation , Triglycerides/blood
18.
J Pharmacol Toxicol Methods ; 71: 137-46, 2015.
Article in English | MEDLINE | ID: mdl-25304940

ABSTRACT

INTRODUCTION: In vivo profiles of aldosterone synthase inhibitors (ASIs) have been investigated utilizing various rodent models. Due to lack of CYP17 activity, rodents produce corticosterone rather than cortisol as that of humans, which raised concern to their effectiveness in translational pharmacological characterization of ASI. METHODS: A rhesus monkey model that combines a low sodium diet with adrenocorticotropin (ACTH) treatment was developed. Plasma concentrations of steroid metabolites associated with reactions catalyzed by CYP11B2 and CYP11B1 were measured concurrently by a UPLC/MS method. RESULTS: Plasma concentration of aldosterone in regular diet fed rhesus monkeys was low at 109pg/mL. Aldosterone concentrations were increased to 252pg/mL when animals were maintained on a low sodium diet for 3weeks, and to 300pg/mL with ACTH treatment at 0.3mg/kg. The combination of low sodium diet with ACTH treatment further increased plasma concentration of aldosterone to 730pg/mL and other steroid metabolites at various levels. Intravenous administration of ASI, fadrozole (0.001-1mg/kg) or LCI699 (0.003-3mg/kg), led to dose-dependent reductions in aldosterone and 18-hydroxycorticosterone, increases in 11-deoxycorticosterone and 11-deoxycortisol, and bell-shaped changes in cortisol and corticosterone. In vivo selectivity of CYP11B2/CYP11B1 for fadrazole was 26-fold and LCI-699 was 27-fold, which was consistent with relative selectivity using in vitro values from recombinant cells transfected with rhesus monkey CYP11B2 and CYP11B1. DISCUSSION: This model enables concurrent characterization of pharmacokinetics, pharmacodynamics and selectivity of CYP11B2 over CYP11B1 inhibition in the same animal. It may be used as a translational model for pharmacological characterization of ASI.


Subject(s)
Cytochrome P-450 CYP11B2/antagonists & inhibitors , Enzyme Inhibitors/pharmacokinetics , Models, Animal , Adrenocorticotropic Hormone/administration & dosage , Adrenocorticotropic Hormone/pharmacokinetics , Animals , Cytochrome P-450 CYP11B2/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Macaca mulatta , Male , Sodium, Dietary/administration & dosage , Sodium, Dietary/pharmacokinetics , Steroid 11-beta-Hydroxylase/antagonists & inhibitors , Steroid 11-beta-Hydroxylase/metabolism , Steroids/blood , Steroids/metabolism
19.
Eur J Pharmacol ; 758: 107-14, 2015 Jul 05.
Article in English | MEDLINE | ID: mdl-25857224

ABSTRACT

Vorapaxar is a novel protease-activated receptor-1 (PAR-1) antagonist recently approved for the reduction of thrombotic cardiovascular events in patients with a history of myocardial infarction or with peripheral arterial disease. Patients who received vorapaxar in addition to standard of care antiplatelet therapy had an increased incidence of major bleeding events compared with placebo. To assess whether platelet transfusion can restore hemostasis in primates on triple antiplatelet therapy, template bleeding times were assessed concurrently in the buccal mucosa, finger pad, and distolateral tail of anesthetized cynomolgus macaques to evaluate bleeding with vorapaxar as either monotherapy or in combination with aspirin or aspirin and clopidogrel. Aspirin (5mg/kg, IV) or vorapaxar (1mg/kg, PO) alone had no significant effect on bleeding times in the three vascular beds examined. A modest (<2-fold) increase in bleeding time was achieved in the three beds with the dual combination of aspirin and vorapaxar. Major increases in bleeding time were achieved in the three beds with the triple combination of aspirin (5mg/kg, IV), vorapaxar (1mg/kg, PO), and clopidogrel (1mg/kg, PO). Transfusion of fresh human platelet rich plasma, but not platelet poor plasma, reversed the increase in bleeding time in the triple therapy group. Transfusion of human platelets may be a viable approach in situations requiring a rapid reversal of platelet function in individuals treated with triple anti-platelet therapy that includes vorapaxar.


Subject(s)
Aspirin/adverse effects , Drug Therapy, Combination/adverse effects , Hemorrhage/therapy , Lactones/adverse effects , Platelet Aggregation Inhibitors/adverse effects , Platelet Transfusion , Pyridines/adverse effects , Receptors, Thrombin/antagonists & inhibitors , Ticlopidine/analogs & derivatives , Animals , Aspirin/administration & dosage , Bleeding Time , Clopidogrel , Hemorrhage/chemically induced , Humans , Lactones/administration & dosage , Macaca fascicularis , Platelet Aggregation Inhibitors/administration & dosage , Pyridines/administration & dosage , Ticlopidine/administration & dosage , Ticlopidine/adverse effects
20.
Blood Coagul Fibrinolysis ; 26(8): 893-902, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26192114

ABSTRACT

This report aims at exploring quantitatively the relationship between FXII inhibition and thromboprotection. FXII full and partial null in rats were established via zinc finger nuclease-mediated knockout and siRNA-mediated knockdown, respectively. The rats were subsequently characterized in thrombosis and hemostasis models. Knockout rats exhibited complete thromboprotection in both the arteriovenous shunt model (∼100% clot weight reduction) and the FeCl3-induced arterial thrombosis model (no reduction in blood flow), without any increase in cuticle bleeding time compared with wild-type control rats. Ex-vivo aPTT and the ellagic acid-triggered thrombin generation assay (TGA) exhibited anticoagulant changes. In contrast, ex-vivo PT or high tissue factor-triggered TGA was indistinguishable from control. Rats receiving single doses (0, 0.01, 0.03, 0.1, 0.3, 1 mg/kg) of FXII siRNA exhibited dose-dependent knockdown in liver FXII mRNA and plasma FXII protein (95 and 99%, respectively, at 1 mg/kg) at day 7 post dosing. FXII knockdown was associated with dose-dependent thromboprotection (maximal efficacy achieved with 1 mg/kg in both models) and negligible change in cuticle bleeding times. Ex-vivo TGA triggered with low-level (0.5 µmol/l) ellagic acid tracked best with the knockdown levels and efficacy. Our findings confirm and extend literature reports of an attractive benefit-to-risk profile of targeting FXII for antithrombotic therapies. Titrating of FXII is instructive for its pharmacological inhibition. The knockout rat is valuable for evaluating both mechanism-based safety concerns and off-target effects of FXII(a) inhibitors. Detailed TGA analyses will inform on optimal trigger conditions in studying pharmacodynamic effects of FXII(a) inhibition.


Subject(s)
Endodeoxyribonucleases/genetics , Factor XII/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Thrombolytic Therapy/methods , Thrombosis/therapy , Animals , Arteriovenous Shunt, Surgical , Chlorides/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Ellagic Acid/pharmacology , Endodeoxyribonucleases/metabolism , Factor XII/genetics , Factor XII/metabolism , Ferric Compounds/pharmacology , Gene Knockout Techniques , Hemorrhage/prevention & control , Humans , Liver/metabolism , Male , Partial Thromboplastin Time , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Thrombin/metabolism , Thrombosis/chemically induced , Thrombosis/genetics , Thrombosis/pathology , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL