Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 296
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 293(9): 3028-3038, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29326169

ABSTRACT

The cyclooxygenases COX-1 and COX-2 oxygenate arachidonic acid (AA) to prostaglandin H2 (PGH2). COX-2 also oxygenates the endocannabinoids 2-arachidonoylglycerol (2-AG) and arachidonoylethanolamide (AEA) to the corresponding PGH2 analogs. Both enzymes are targets of nonsteroidal anti-inflammatory drugs (NSAIDs), but NSAID-mediated COX inhibition is associated with gastrointestinal toxicity. One potential strategy to counter this toxicity is to also inhibit fatty acid amide hydrolase (FAAH), which hydrolyzes bioactive fatty acid ethanolamides (FAEs) into fatty acids and ethanolamine. Here, we investigated the mechanism of COX inhibition by ARN2508, an NSAID that inhibits both COXs and FAAH with high potency, target selectivity, and decreased gastrointestinal toxicity in mouse models, presumably due to its ability to increase levels of FAEs. A 2.27-Å-resolution X-ray crystal structure of the COX-2·(S)-ARN2508 complex reveals that ARN2508 adopts a binding pose similar to that of its parent NSAID flurbiprofen. However, ARN2508's alkyl tail is inserted deep into the top channel, an active site region not exploited by any previously reported NSAID. As for flurbiprofen, ARN2508's potency is highly dependent on the configuration of the α-methyl group. Thus, (S)-ARN2508 is more potent than (R)-ARN2508 for inhibition of AA oxygenation by both COXs and 2-AG oxygenation by COX-2. Also, similarly to (R)-flurbiprofen, (R)-ARN2508 exhibits substrate selectivity for inhibition of 2-AG oxygenation. Site-directed mutagenesis confirms the importance of insertion of the alkyl tail into the top channel for (S)-ARN2508's potency and suggests a role for Ser-530 as a determinant of the inhibitor's slow rate of inhibition compared with that of (S)-flurbiprofen.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Catalytic Domain , Cyclooxygenase Inhibitors/metabolism , Cyclooxygenase Inhibitors/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Cyclooxygenase Inhibitors/chemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Phenylcarbamates/chemistry , Phenylcarbamates/metabolism , Phenylcarbamates/pharmacology , Phenylpropionates/chemistry , Phenylpropionates/metabolism , Phenylpropionates/pharmacology , Prostaglandin-Endoperoxide Synthases/chemistry , Protein Binding , Stereoisomerism , Substrate Specificity
2.
Bioorg Chem ; 80: 164-173, 2018 10.
Article in English | MEDLINE | ID: mdl-29929077

ABSTRACT

Two new series of pyrazolo[3,4-d]pyrimidine bearing thiazolidinone moiety were designed and synthesized. The newly synthesized compounds were evaluated for their in vitro (COX-1 and COX-2) inhibitory assay. Compounds that showed promising COX-2 selectivity were further subjected to in vivo anti-inflammatory screening applying formalin induced paw edema (acute model) and cotton-pellet induced granuloma (chronic model) assays using celecoxib and diclofenac sodium as reference drugs. The histopathological and ulcerogenic potential were also determined. In vivo anti-inflammatory data showed that compounds 2, 6, 7d displayed anti-inflammatory activity higher than both references in the formalin induced paw edema model. On the other hand, compounds 2, 3d, 3e, 7b and 7d displayed anti-inflammatory activity greater than or nearly equivalent to diclofenac sodium in the cotton pellet-induced granuloma assay. Moreover, most of the tested compounds revealed good gastrointestinal safety profile. Collectively, compounds 2 and 7d were considered as promising candidates in managing both acute and chronic inflammation with safe gastrointestinal margin.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Drug Design , Edema/drug therapy , Pyrazoles/chemistry , Pyrimidines/chemistry , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/therapeutic use , Celecoxib/therapeutic use , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/metabolism , Cyclooxygenase Inhibitors/therapeutic use , Diclofenac/therapeutic use , Edema/chemically induced , Edema/veterinary , Female , Granuloma/chemically induced , Granuloma/drug therapy , Granuloma/veterinary , Pyrazoles/metabolism , Pyrazoles/therapeutic use , Pyrimidines/metabolism , Pyrimidines/therapeutic use , Rats , Rats, Wistar , Structure-Activity Relationship , Thiazolidines/chemistry
3.
AAPS PharmSciTech ; 18(8): 2898-2909, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28429293

ABSTRACT

The aim of this study was to investigate the potential of surfactant-based nanovesicular system (spanlastics) for topical delivery of fenoprofen calcium (FPCa) to eliminate its oral gastrointestinal adverse effects. FPCa-loaded spanlastics were prepared by thin film hydration (TFH) technique according to a full factorial design to investigate the influence of formulation variables on the drug entrapment efficiency (%EE), particle size (PS), deformability index (DI), and the % drug released after 24 h through the cellulose membrane (Q24h) using Design-Expert® software. The optimized formula (composed of Span 60 and Tween 60 as an edge activator at weight ratio of 8: 2 in presence of Transcutol P as a cosolvent in the hydration media) exhibited the highest %EE (49.91 ± 2.60%), PS of 536.1 ± 17.14 nm, DI of 5.07 ± 0.06 g, and Q24h of 61.11 ± 2.70%; it was also characterized for morphology and physical stability. In vitro release study of FPCa-loaded spanlastic gel and conventional FPCa gel through a synthetic membrane and hairless rat skin were evaluated. The skin permeation study revealed that spanlastic gel exhibited both consistent and prolonged action. Finally, the % inhibition of carrageenan-induced rat paw edema of spanlastic gel was three times higher than the conventional FPCa gel after 24 h. In conclusion, spanlastic-based gel could be a great approach for improving topical delivery of fenoprofen calcium, providing both prolonged and enhanced anti-inflammatory activity in the treatment of arthritis.


Subject(s)
Drug Delivery Systems/methods , Fenoprofen/administration & dosage , Fenoprofen/metabolism , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Skin/metabolism , Administration, Topical , Animals , Cyclooxygenase Inhibitors/administration & dosage , Cyclooxygenase Inhibitors/metabolism , Drug Carriers/administration & dosage , Drug Carriers/metabolism , Drug Evaluation, Preclinical/methods , Drug Liberation/drug effects , Drug Liberation/physiology , Edema/drug therapy , Edema/metabolism , Elasticity , Male , Particle Size , Rats , Rats, Hairless , Skin/drug effects , Skin Absorption/drug effects , Skin Absorption/physiology , Surface-Active Agents/administration & dosage , Surface-Active Agents/metabolism
4.
Prog Drug Res ; 71: 181-3, 2016.
Article in English | MEDLINE | ID: mdl-26939289
5.
J Biol Chem ; 289(10): 6799-6808, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24425867

ABSTRACT

Oxicams are widely used nonsteroidal anti-inflammatory drugs (NSAIDs), but little is known about the molecular basis of the interaction with their target enzymes, the cyclooxygenases (COX). Isoxicam is a nonselective inhibitor of COX-1 and COX-2 whereas meloxicam displays some selectivity for COX-2. Here we report crystal complexes of COX-2 with isoxicam and meloxicam at 2.0 and 2.45 angstroms, respectively, and a crystal complex of COX-1 with meloxicam at 2.4 angstroms. These structures reveal that the oxicams bind to the active site of COX-2 using a binding pose not seen with other NSAIDs through two highly coordinated water molecules. The 4-hydroxyl group on the thiazine ring partners with Ser-530 via hydrogen bonding, and the heteroatom of the carboxamide ring of the oxicam scaffold interacts with Tyr-385 and Ser-530 through a highly coordinated water molecule. The nitrogen atom of the thiazine and the oxygen atom of the carboxamide bind to Arg-120 and Tyr-355 via another highly ordered water molecule. The rotation of Leu-531 in the structure opens a novel binding pocket, which is not utilized for the binding of other NSAIDs. In addition, a detailed study of meloxicam·COX-2 interactions revealed that mutation of Val-434 to Ile significantly reduces inhibition by meloxicam due to subtle changes around Phe-518, giving rise to the preferential inhibition of COX-2 over COX-1.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/metabolism , Piroxicam/analogs & derivatives , Thiazines/metabolism , Thiazoles/metabolism , Amino Acid Substitution , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Arginine/chemistry , Arginine/genetics , Arginine/metabolism , Binding Sites , Catalytic Domain , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/genetics , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/genetics , Cyclooxygenase Inhibitors/chemistry , Hydrogen Bonding , Leucine/chemistry , Leucine/genetics , Leucine/metabolism , Meloxicam , Mice , Mutation , Piroxicam/chemistry , Piroxicam/metabolism , Protein Structure, Secondary , Serine/chemistry , Serine/genetics , Serine/metabolism , Thiazines/chemistry , Thiazoles/chemistry , Tyrosine/chemistry , Tyrosine/genetics , Tyrosine/metabolism , Water
6.
Clin Exp Pharmacol Physiol ; 42(2): 162-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25399887

ABSTRACT

No reflow after reperfusion therapy for myocardial infarction is a strong predictor of clinical outcome. Increased levels of inflammatory factors, including C-reactive protein (CRP), in patients with acute myocardial infarction (AMI) undergoing primary percutaneous coronary intervention (PCI) may affect myocardial perfusion. However, why the no-reflow phenomenon increases in inflammation stress after PCI is not clear. The aim of the present study was to determine the effects and molecular mechanisms underlying the effects of CRP on the expression of cyclo-oxygenase (COX) on the development of the no-reflow phenomenon. There was a significant increase in plasma levels of CRP and interleukin (IL)-6 in no-reflow patients, suggesting that inflammatory factors play an important role in the development of the no-reflow phenomenon. The mechanisms involved were further evaluated after reperfusion in a rat model mimicking the no-reflow phenomenon. Compared with normal reflow rats, there were significant increases in both COX-1 and COX-2 in cardiac tissue from no-reflow rats. The COX inhibitor indomethacin (5 mg/kg, i.p.) significantly reduced the no-reflow area. In another series of experiments, human coronary artery endothelial cells (HCAEC) were treated with CRP at clinically relevant concentrations (5-25 µg/mL). C-Reactive protein significantly increased COX-1 and COX-2 levels in a time- and concentration-dependent manner. In addition, extracellular signal-regulated kinase (ERK) and Jun N-terminal kinase (JNK) were activated in CRP (5, 10, 25 µg/mL)-treated HCAEC cultures. Furthermore, the ERK inhibitor pd98059 (30 µmol/L) and the JNK inhibitor sp600125 (10 µmol/L) blocked CRP-induced COX-1 and COX-2 expression for 12 h. Together, the findings of the present study suggest that CRP can promote the development of the no-reflow phenomenon by increasing COX-1 and COX-2 expression, which is regulated, in part, via ERK and JNK activity.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Inflammation/pathology , Myocardial Infarction/pathology , No-Reflow Phenomenon/pathology , Acute Disease , Animals , C-Reactive Protein/metabolism , Cyclooxygenase Inhibitors/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Indomethacin/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , Male , Middle Aged , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , No-Reflow Phenomenon/drug therapy , No-Reflow Phenomenon/metabolism , Rats , Rats, Sprague-Dawley
7.
Mem Inst Oswaldo Cruz ; 110(2): 181-91, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25946241

ABSTRACT

Chagas disease, caused by the intracellular protozoan Trypanosoma cruzi, is a serious health problem in Latin America. During this parasitic infection, the heart is one of the major organs affected. The pathogenesis of tissue remodelling, particularly regarding cardiomyocyte behaviour after parasite infection and the molecular mechanisms that occur immediately following parasite entry into host cells are not yet completely understood. When cells are infected with T. cruzi, they develop an inflammatory response, in which cyclooxygenase-2 (COX-2) catalyses rate-limiting steps in the arachidonic acid pathway. However, how the parasite interaction modulates COX-2 activity is poorly understood. In this study, the H9c2 cell line was used as our model and we investigated cellular and biochemical aspects during the initial 48 h of parasitic infection. Oscillatory activity of COX-2 was observed, which correlated with the control of the pro-inflammatory environment in infected cells. Interestingly, subcellular trafficking was also verified, correlated with the control of Cox-2 mRNA or the activated COX-2 protein in cells, which is directly connected with the assemble of stress granules structures. Our collective findings suggest that in the very early stage of the T. cruzi-host cell interaction, the parasite is able to modulate the cellular metabolism in order to survives.


Subject(s)
Cell Nucleus/metabolism , Cyclooxygenase 2/metabolism , Host-Parasite Interactions/physiology , Myocytes, Cardiac/parasitology , RNA, Protozoan/metabolism , Trypanosoma cruzi/enzymology , Active Transport, Cell Nucleus/physiology , Animals , CELF Proteins/metabolism , Cell Line , Cyclooxygenase Inhibitors/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/analysis , Fluorescent Dyes , Immunoprecipitation , In Situ Hybridization , Interleukin-1beta/analysis , Nitrobenzenes/pharmacology , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Time Factors , Trypanosoma cruzi/physiology
8.
J Chem Inf Model ; 54(5): 1488-99, 2014 05 27.
Article in English | MEDLINE | ID: mdl-24786949

ABSTRACT

Cyclooxygenase-1 (COX-1) is one of the main targets of most pain-relieving pharmaceuticals. Although the enzyme is well characterized, it is known to be a difficult target for automated molecular docking and scoring. We collected from the literature a structurally diverse set of 45 nonsteroidal anti-inflammatory drugs (NSAIDs) and COX-2-selective inhibitors (coxibs) with a wide range of binding affinities for COX-1. The binding of this data set to a homology model of human COX-1 was analyzed with different combinations of molecular docking algorithms, scoring functions, and the linear interaction energy (LIE) method for estimating binding affinities. It is found that the computational protocols for estimation of binding affinities are extremely sensitive to the initial orientations of the ligands in the binding pocket. To overcome this limitation, we propose a systematic exploration of docking poses using the LIE calculations as a postscoring function. This scheme yields predictions in excellent agreement with experiment, with a mean unsigned error of 0.9 kcal/mol for binding free energies and structures of high quality. A significant improvement of the results is also seen when averaging over experimental data from several independent measurements.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase Inhibitors/metabolism , Cyclooxygenase Inhibitors/pharmacology , Drug Design , Molecular Docking Simulation , Algorithms , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 1/chemistry , Cyclooxygenase Inhibitors/chemistry , Humans , Ligands , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Structure-Activity Relationship , Thermodynamics
9.
Bioorg Med Chem ; 22(8): 2529-34, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24650702

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) achieve their anti-inflammatory effect by inhibiting cyclooxygenase activity. We previously suggested that in addition to cyclooxygenase-inhibition at the gastric mucosa, NSAID-induced gastric mucosal cell death is required for the formation of NSAID-induced gastric lesions in vivo. We showed that celecoxib exhibited the most potent membrane permeabilizing activity among the NSAIDs tested. In contrast, we have found that the NSAID rofecoxib has very weak membrane permeabilizing activity. To understand the membrane permeabilizing activity of coxibs in terms of their structure-activity relationship, we separated the structures of celecoxib and rofecoxib into three parts, synthesized hybrid compounds by substitution of each of the parts, and examined the membrane permeabilizing activities of these hybrids. The results suggest that the sulfonamidophenyl subgroup of celecoxib or the methanesulfonylphenyl subgroup of rofecoxib is important for their potent or weak membrane permeabilizing activity, respectively. These findings provide important information for design and synthesis of new coxibs with lower membrane permeabilizing activity.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cyclooxygenase Inhibitors/chemistry , Lactones/chemistry , Lipid Bilayers/chemistry , Pyrazoles/chemistry , Sulfonamides/chemistry , Sulfones/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Celecoxib , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/metabolism , Lactones/metabolism , Lipid Bilayers/metabolism , Liposomes/chemistry , Liposomes/metabolism , Protein Binding , Pyrazoles/metabolism , Spectrometry, Fluorescence , Structure-Activity Relationship , Sulfonamides/metabolism , Sulfones/metabolism
10.
J Clin Pharm Ther ; 39(5): 455-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25070498

ABSTRACT

The ongoing important debate about the relative benefits/risks of COX-1 or COX-2 NSAIDs is hampered by the use of a measure of 'selectivity' that is inherently flawed. An alternative measure provides more meaningful and clinically relevant information.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Confounding Factors, Epidemiologic , Cyclooxygenase 1/metabolism , Cyclooxygenase 2 Inhibitors/metabolism , Cyclooxygenase Inhibitors/metabolism , Humans , Inhibitory Concentration 50
11.
J Pharmacol Exp Ther ; 346(1): 96-104, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23596058

ABSTRACT

Liver first-pass metabolism differs considerably among organic nitrates, but little information exists on the mechanism of denitration of these compounds in hepatic tissue. The metabolism of nitrooxybutyl-esters of flurbiprofen and ferulic-acid, a class of organic nitrates with potential therapeutic implication in variety of different conditions, was investigated in comparison with glyceryl trinitrate (GTN) in human liver by a multiple approach, using a spontaneous metabolism-independent nitric oxide (NO) donor [3-(aminopropyl)-1-hydroxy-3-isopropyl-2-oxo-1-triazene (NOC-5)] as a reference tool. Nitrooxybutyl-esters were rapidly and quantitatively metabolized to their respective parent compounds and the organic nitrate moiety nitrooxybutyl-alcohol (NOBA). Differently from GTN, which was rapidly and completely metabolized to nitrite, NOBA was slowly metabolized to nitrate. In contrast to the spontaneous NO donor NOC-5, NOBA and GTN did not generate detectable NO and failed to suppress the activity of cytochrome P450, an enzyme known to be inhibited by NO. The direct identification of NOBA after liver metabolism targets this compound as the functional organic nitrate metabolite of nitrooxybutyl-esters. Moreover, the investigation of the pathways for denitration of NOBA and GTN suggests that organic nitrates are not primarily metabolized to NO in the liver but to different extents of nitrite or nitrate depending in their different chemical structure. Therefore, cytochrome P450-dependent metabolism of concomitant drugs is not likely to be affected by oral coadministration of organic nitrates. However, the first pass may differently affect the pharmacological profile of organic nitrates in connection with the different extent of denitration and the distinct bioactive species generated and exported from the liver (nitrate or nitrite).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cytochrome P-450 CYP1A2/metabolism , Liver/metabolism , Microsomes, Liver/metabolism , Mitochondria, Liver/metabolism , Nitric Oxide/metabolism , Nitro Compounds/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Butanes/metabolism , Butanes/pharmacology , Cyclooxygenase Inhibitors/metabolism , Cyclooxygenase Inhibitors/pharmacology , Cytochrome P-450 CYP1A2 Inhibitors , Cytosol/drug effects , Cytosol/enzymology , Cytosol/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Female , Flurbiprofen/analogs & derivatives , Flurbiprofen/metabolism , Flurbiprofen/pharmacology , Free Radical Scavengers/metabolism , Free Radical Scavengers/pharmacology , Humans , Kinetics , Liver/drug effects , Liver/enzymology , Male , Metabolic Detoxication, Phase I , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Mitochondria, Liver/drug effects , Mitochondria, Liver/enzymology , Nitro Compounds/pharmacology , Nitroglycerin/metabolism , Nitroglycerin/pharmacology , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology
12.
Curr Pain Headache Rep ; 17(4): 327, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23423598

ABSTRACT

Indomethacin, as a member of the non-steroidal anti-inflammatory drug class, plays a special role in the treatment of headaches. By definition, it is completely efficacious in the treatment of the primary headache disorders paroxysmal hemicrania and hemicrania continua. Therefore, indomethacin is also used as a tool for differential diagnosis in headache clinics. Indomethacin has a clear action as a cyclooxygenase inhibitor. Additional mechanisms and interactions with cell signaling pathways and inflammatory pathways are considered in this article. However, it is not known what mechanism or interaction with pathophysiological mechanisms is the key to indomethacin's specific pharmacology in headache therapy. Focusing on headache therapy, we summarize the current knowledge of pharmacology, treatment options, and recommendations for the use of indomethacin in primary headaches. New findings from the field of headache research, as well as from Alzheimer's disease and cancer research on the pharmacological actions of indomethacin and their potential implications on the pathophysiology of indomethacin sensitive headaches, are discussed.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Headache/drug therapy , Indomethacin/pharmacology , Indomethacin/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cyclooxygenase Inhibitors/metabolism , Headache/diagnosis , Headache/metabolism , Humans , Indomethacin/metabolism , Treatment Outcome
13.
Proc Natl Acad Sci U S A ; 107(1): 28-33, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19955429

ABSTRACT

Pain associated with inflammation involves prostaglandins synthesized from arachidonic acid (AA) through cyclooxygenase-2 (COX-2) pathways while thromboxane A(2) formed by platelets from AA via cyclooxygenase-1 (COX-1) mediates thrombosis. COX-1 and COX-2 are both targets of nonselective nonsteroidal antiinflammatory drugs (nsNSAIDs) including aspirin whereas COX-2 activity is preferentially blocked by COX-2 inhibitors called coxibs. COXs are homodimers composed of identical subunits, but we have shown that only one subunit is active at a time during catalysis; moreover, many nsNSAIDS bind to a single subunit of a COX dimer to inhibit the COX activity of the entire dimer. Here, we report the surprising observation that celecoxib and other coxibs bind tightly to a subunit of COX-1. Although celecoxib binding to one monomer of COX-1 does not affect the normal catalytic processing of AA by the second, partner subunit, celecoxib does interfere with the inhibition of COX-1 by aspirin in vitro. X-ray crystallographic results obtained with a celecoxib/COX-1 complex show how celecoxib can bind to one of the two available COX sites of the COX-1 dimer. Finally, we find that administration of celecoxib to dogs interferes with the ability of a low dose of aspirin to inhibit AA-induced ex vivo platelet aggregation. COX-2 inhibitors such as celecoxib are widely used for pain relief. Because coxibs exhibit cardiovascular side effects, they are often prescribed in combination with low-dose aspirin to prevent thrombosis. Our studies predict that the cardioprotective effect of low-dose aspirin on COX-1 may be blunted when taken with coxibs.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase 2 Inhibitors/metabolism , Cyclooxygenase Inhibitors/metabolism , Isoenzymes/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/chemistry , Aspirin/metabolism , Aspirin/pharmacology , Catalytic Domain , Crystallography, X-Ray , Cyclooxygenase 1/chemistry , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacology , Dogs , Humans , Isoenzymes/chemistry , Models, Molecular , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Protein Binding , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism
14.
FASEB J ; 25(1): 382-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20833872

ABSTRACT

Docosahexaenoic acid (DHA), an important component of marine lipids, exhibits anti-inflammatory activity related to some of its oxygenated metabolites, such as neuroprotectin/protectin D1 [NPD1/PD1; 10(R),17(S)-dihydroxy-docosa-4Z,7Z, 11E,13E,15Z,19Z-hexaenoic acid] produced through the 15-lipoxygenase pathway. However, other metabolites from DHA can be produced through this pathway, and other polyunsaturated fatty acids (PUFAs) of nutritional value may be oxygenated as well. Their biological activities remain unknown. Isomers of protectin D1 were synthesized using soybean lipoxygenase and tested for their ability to inhibit human blood platelet aggregation. A geometric isomer called PDX, previously described with the 11E,13Z,15E geometry, instead of 11E,13E,15Z in PD1, inhibited platelet aggregation at submicromolar concentrations when induced by either collagen, arachidonic acid, or thromboxane. The inhibition occurred at the level of both the cyclooxygenase activity and thromboxane receptor site. Interestingly, all the metabolites tested exhibiting the E,Z,E-conjugated triene were active, whereas E,E,Z trienes (as in PD1) or all-trans (E,E,E) trienes were inactive. We conclude that PDX and other oxygenated products from PUFAs of nutritional interest, having the E,Z,E-conjugated triene motif and collectively named poxytrins (PUFA oxygenated trienes), might have antithrombotic potential.


Subject(s)
Dicarboxylic Acids/pharmacology , Docosahexaenoic Acids/pharmacology , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Arachidonic Acid/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen/pharmacology , Cyclic AMP/metabolism , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dicarboxylic Acids/chemistry , Dicarboxylic Acids/metabolism , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/metabolism , Fatty Acids, Unsaturated/chemistry , Humans , Isomerism , Oxygen/metabolism , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Thromboxane/antagonists & inhibitors , Receptors, Thromboxane/metabolism , Thromboxanes/pharmacology
16.
Arch Pharm (Weinheim) ; 345(11): 878-83, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22907715

ABSTRACT

A novel series of 3,6,6-trimethyl-4-oxo-4,5,6,7-tetrahydroindazole-1-acetic acid derivatives was designed and synthesized by a new one-step pathway. Structure elucidation of the synthesized compounds was confirmed by various spectral and elemental analyses. The prepared compounds were evaluated for their ability to inhibit cyclooxygenase-2 (COX-2) and cyclooxygenase-1 (COX-1) enzymes in vitro. Among the synthesized compounds, the 2-(3,6,6-trimethyl-4-oxo-4,5,6,7-tetrahydroindazol-1-yl)acetic acid 4 emerged as the most potent COX-2 inhibitor (IC(50) value: 150 nM) with the highest selectivity index (COX-1/COX-2 inhibition ratio: 570.6). Docking studies of compound 4 in the active site of COX-2 recognized its potential binding mode to the enzyme. Based on the preliminary results, compound 4 was considered as a lead compound for further optimization.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Indazoles/pharmacology , Molecular Docking Simulation , Animals , Cyclooxygenase 1/drug effects , Cyclooxygenase 1/metabolism , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/metabolism , Cyclooxygenase Inhibitors/pharmacology , Drug Design , Indazoles/chemical synthesis , Indazoles/chemistry , Inhibitory Concentration 50 , Sheep , Structure-Activity Relationship
17.
Biochemistry ; 50(35): 7684-93, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21805999

ABSTRACT

The inducible microsomal prostaglandin E(2) synthase 1 (MPGES1) is an integral membrane protein coexpressed with and functionally coupled to cyclooxygenase 2 (COX-2) generating the pro-inflammatory molecule PGE(2). The development of effective inhibitors of MPGES1 holds promise as a highly selective route for controlling inflammation. In this paper, we describe the use of backbone amide H/D exchange mass spectrometry to map the binding sites of different types of inhibitors of MPGES1. The results reveal the locations of specific inhibitor binding sites that include the GSH binding site and a hydrophobic cleft in the protein thought to accommodate the prostaglandin H(2) substrate. In the absence of three-dimensional crystal structures of the enzyme-bound inhibitors, the results provide clear physical evidence that three pharmacologically active inhibitors bind in a hydrophobic cleft composed of sections of transmembrane helices Ia, IIb, IIIb, and IVb at the interface of subunits in the trimer. In principle, the H/D exchange behavior of the protein can be used as a preliminary guide for optimization of inhibitor efficacy. Finally, a comparison of the structures and H/D exchange behavior of MPGES1 and the related enzyme MGST1 in the presence of glutathione and the inhibitor glutathione sulfonate confirms the unusual observation that two proteins from the same superfamily harbor GSH binding sites in different locations.


Subject(s)
Cyclooxygenase Inhibitors/metabolism , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/chemistry , Binding Sites , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacology , Glutathione/chemistry , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Intramolecular Oxidoreductases/metabolism , Prostaglandin-E Synthases , Protein Binding , Protein Structure, Secondary , Substrate Specificity/drug effects
18.
Eur J Neurosci ; 33(4): 632-41, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21219477

ABSTRACT

In many retinal diseases, it is the death of photoreceptors that leads to blindness. In previous in vitro and in vivo studies, basic fibroblast growth factor (bFGF) has been shown to increase retinal cell survival. More recently, reactive oxygen species (ROS) have also been shown to promote cell survival, contrary to the traditional view that they are solely destructive molecules. Due to this possible link, we hypothesised that bFGF could stimulate the production of ROS, which in turn stimulates the protein kinase B (Akt) survival pathway. Flow cytometry was used to measure the fluorescence of oxidised dihydrorhodamine, a ROS indicator, in the murine 661W photoreceptor cell line under several different conditions. Expression of cyclooxygenase (Cox) enzymes was evaluated by immunohistochemistry, and the response of photoreceptor cells to exogenous bFGF in the explanted mouse retina was studied by confocal microscopy. Exogenous addition of bFGF to 661W cells resulted in an increase in ROS production that lasted for 24 h. When this ROS production was inhibited, bFGF-induced phosphorylation of Akt was prevented. Through the use of inhibitors and small interfering RNA in the cell line, the source of this production was shown to be Cox and to involve the activation of phospholipases A(2) + C. This pathway may also occur in the mouse retina, as we showed that the retina expressed Cox1&2, and that photoreceptors in explanted retina respond to bFGF by increasing their ROS levels. These results demonstrate that exogenous bFGF can stimulate ROS production through the activation of Cox, and activate the Akt pathway.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Cell Line , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/metabolism , Diclofenac/metabolism , Membrane Proteins/metabolism , Mice , Oxidation-Reduction , Phosphatidylinositol 3-Kinases/genetics , Phospholipases A/metabolism , Photoreceptor Cells, Vertebrate/cytology , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Type C Phospholipases/metabolism
19.
Drug Metab Dispos ; 39(5): 838-46, 2011 May.
Article in English | MEDLINE | ID: mdl-21321060

ABSTRACT

Drug-induced hepatotoxicity is a major problem in drug development, and reactive metabolites generated by cytochrome P450s are suggested to be one of the causes. CYP2C9 is one of the major enzymes in hepatic drug metabolism. In the present study, we developed a highly sensitive cell-based screening system for CYP2C9-mediated metabolic activation using an adenovirus vector expressing CYP2C9 (AdCYP2C9). Human hepatocarcinoma HepG2 cells infected with our constructed AdCYP2C9 for 2 days at multiplicity of infection 10 showed significantly higher diclofenac 4'-hydroxylase activity than human hepatocytes. AdCYP2C9-infected cells were treated with several hepatotoxic drugs, resulting in a significant increase in cytotoxicity by treatment with losartan, benzbromarone, and tienilic acid. Metabolic activation of losartan by CYP2C9 has never been reported, although the metabolic activations of benzbromarone and tienilic acid have been reported. To identify the reactive metabolites of losartan, the semicarbazide adducts of losartan were investigated by liquid chromatography-tandem mass spectrometry. Two CYP2C9-specific semicarbazide adducts of losartan (S1 and S2) were detected. S2 adduct formation suggested that a reactive metabolite was produced from the aldehyde metabolite E3179, but a possible metabolite from S1 adduct formation was not produced via E3179. In conclusion, a highly sensitive cell-based assay to evaluate CYP2C9-mediated metabolic activation was established, and we found for the first time that CYP2C9 is involved in the metabolic activation of losartan. This cell-based assay system would be useful for evaluating drug-induced cytotoxicity caused by human CYP2C9.


Subject(s)
Antihypertensive Agents/metabolism , Aryl Hydrocarbon Hydroxylases/metabolism , Hepatocytes/metabolism , Losartan/metabolism , Antihypertensive Agents/pharmacology , Antihypertensive Agents/toxicity , Benzbromarone/metabolism , Biotransformation , Cyclooxygenase Inhibitors/metabolism , Cytochrome P-450 CYP2C9 , Diclofenac/metabolism , HEK293 Cells , Hep G2 Cells , Hepatocytes/drug effects , Humans , Losartan/pharmacology , Losartan/toxicity , NF-E2-Related Factor 2/metabolism , Semicarbazides/metabolism , Sensitivity and Specificity , Time Factors , Uricosuric Agents/metabolism
20.
Biochem J ; 429(2): 273-82, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20450491

ABSTRACT

Resveratrol has demonstrated cancer chemopreventive activity in animal models and some clinical trials are underway. In addition, resveratrol was shown to promote cell survival, increase lifespan and mimic caloric restriction, thereby improving health and survival of mice on high-calorie diet. All of these effects are potentially mediated by the pleiotropic interactions of resveratrol with different enzyme targets including COX-1 (cyclo-oxygenase-1) and COX-2, NAD+-dependent histone deacetylase SIRT1 (sirtuin 1) and QR2 (quinone reductase 2). Nonetheless, the health benefits elicited by resveratrol as a direct result of these interactions with molecular targets have been questioned, since it is rapidly and extensively metabolized to sulfate and glucuronide conjugates, resulting in low plasma concentrations. To help resolve these issues, we tested the ability of resveratrol and its metabolites to modulate the function of some known targets in vitro. In the present study, we have shown that COX-1, COX-2 and QR2 are potently inhibited by resveratrol, and that COX-1 and COX-2 are also inhibited by the resveratrol 4'-O-sulfate metabolite. We determined the X-ray structure of resveratrol bound to COX-1 and demonstrate that it occupies the COX active site similar to other NSAIDs (non-steroidal anti-inflammatory drugs). Finally, we have observed that resveratrol 3- and 4'-O-sulfate metabolites activate SIRT1 equipotently to resveratrol, but that activation is probably a substrate-dependent phenomenon with little in vivo relevance. Overall, the results of this study suggest that in vivo an interplay between resveratrol and its metabolites with different molecular targets may be responsible for the overall beneficial health effects previously attributed only to resveratrol itself.


Subject(s)
Stilbenes/metabolism , Stilbenes/pharmacology , Animals , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/metabolism , Anticarcinogenic Agents/pharmacology , Catalytic Domain , Crystallography, X-Ray , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/metabolism , Cyclooxygenase Inhibitors/pharmacology , Humans , In Vitro Techniques , Mice , Models, Molecular , Quinone Reductases/antagonists & inhibitors , Resveratrol , Sirtuin 1/metabolism , Stilbenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL