Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 127(8): 3167-3176, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28737505

ABSTRACT

Proinflammatory leukotrienes (LTs) are produced by 5-lipoxygenase (5-LO) aided by 5-LO-activating protein (FLAP). LT biosynthesis inhibitors are currently under clinical investigation as treatments for respiratory and cardiovascular diseases. Here, we have revealed a sex bias in the efficiency of clinically relevant LT biosynthesis inhibitors, showing that their effects are superior in females. We found that androgens cause these sex differences by impeding the LT-biosynthetic 5-LO/FLAP complex assembly. Lower doses of the FLAP inhibitor MK886 were required to reduce LTB4 levels in exudates of female versus male mice and rats. Following platelet-activating factor-induced shock, MK886 increased survival exclusively in female mice, and this effect was abolished by testosterone administration. FLAP inhibitors and the novel-type 5-LO inhibitors licofelone and sulindac sulfide exhibited higher potencies in human blood from females, and bioactive 5-LO/FLAP complexes were formed in female, but not male, human and murine leukocytes. Supplementation of female blood or leukocytes with 5α-dihydrotestosterone abolished the observed sex differences. Our data suggest that females may benefit from anti-LT therapy to a greater extent than males, prompting consideration of sex issues in LT modifier development.


Subject(s)
Androgens/metabolism , Leukotrienes/biosynthesis , Sex Factors , Testosterone/administration & dosage , 5-Lipoxygenase-Activating Proteins/metabolism , Animals , Arachidonate 5-Lipoxygenase/metabolism , Dihydrotestosterone/metabolism , Female , Humans , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Leukocytes/metabolism , Lipoxygenase Inhibitors/pharmacology , Male , Mice , Pyrroles/administration & dosage , Rats , Rats, Wistar , Sulindac/administration & dosage , Sulindac/analogs & derivatives , Testosterone/metabolism
2.
Biochem Pharmacol ; 112: 60-71, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27157409

ABSTRACT

5-Lipoxygenase (5-LO) catalyzes the first two steps in leukotriene (LT) biosynthesis. Because LTs play pivotal roles in allergy and inflammation, 5-LO represents a valuable target for anti-inflammatory drugs. Here, we investigated the molecular mechanism, the pharmacological profile, and the in vivo effectiveness of the novel 1,2-benzoquinone-featured 5-LO inhibitor RF-22c. Compound RF-22c potently inhibited 5-LO product synthesis in neutrophils and monocytes (IC50⩾22nM) and in cell-free assays (IC50⩾140nM) without affecting 12/15-LOs, cyclooxygenase (COX)-1/2, or arachidonic acid release, in a specific and reversible manner, supported by molecular docking data. Antioxidant or iron-chelating properties were not evident for RF-22c and 5-LO-regulatory cofactors like Ca(2+) mobilization, ERK-1/2 activation, and 5-LO nuclear membrane translocation and interaction with 5-LO-activating protein (FLAP) were unaffected. RF-22c (0.1mg/kg; i.p.) impaired (I) bronchoconstriction in ovalbumin-sensitized mice challenged with acetylcholine, (II) exudate formation in carrageenan-induced paw edema, and (III) zymosan-induced leukocyte infiltration in air pouches. Taken together, RF-22c is a highly selective and potent 5-LO inhibitor in intact human leukocytes with pronounced effectiveness in different models of inflammation that warrants further preclinical analysis of this agent as anti-inflammatory drug.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arachidonate 5-Lipoxygenase/metabolism , Benzoquinones/pharmacology , Bronchoconstriction/drug effects , Leukotrienes/biosynthesis , Lipoxygenase Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Benzoquinones/administration & dosage , Benzoquinones/therapeutic use , Blood Platelets/drug effects , Blood Platelets/enzymology , Blood Platelets/immunology , Bronchoconstriction/immunology , Cells, Cultured , Edema/drug therapy , Edema/enzymology , Edema/immunology , Escherichia coli/drug effects , Escherichia coli/genetics , Female , Humans , Lipoxygenase Inhibitors/administration & dosage , Lipoxygenase Inhibitors/therapeutic use , Mice, Inbred BALB C , Molecular Docking Simulation , Monocytes/drug effects , Monocytes/enzymology , Monocytes/immunology , Neutrophils/drug effects , Neutrophils/enzymology , Neutrophils/immunology
3.
Eur J Med Chem ; 94: 132-9, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25765759

ABSTRACT

5-Lipoxygenase (5-LO) is a potential target for pharmacological intervention with various inflammatory and allergic diseases. Starting from the natural dual 5-LO/microsomal prostaglandin E2 synthase (mPGES)-1 inhibitor embelin (2,5-dihydroxy-3-undecyl-1,4-benzoquinone, 2) that suppresses 5-LO activity in human primary leukocytes with IC50 = 0.8-2 µM, we synthesized 48 systematically modified derivatives of 2. We modified the 1,4-quinone to 1,2-quinone, mono- or bimethylated the hydroxyl groups, and varied the C11-n-alkyl residue (C4- to C16-n-alkyl or prenyl) of 2. Biological evaluation yields potent analogues being superior over 2 and obvious structure-activity relationships (SAR) for inhibition of 5-LO. Interestingly, conversion to 1,2-benzoquinone and bimethylation of the hydroxyl moieties strongly improves 5-LO inhibition in polymorphonuclear leukocytes versus 2 up to 60-fold, exemplified by the C12-n-alkyl derivative 22c (4,5-dimethoxy-3-dodecyl-1,2-benzoquinone) with IC50 = 29 nM. Regarding inhibition of mPGES-1, none of the novel benzoquinones could outperform the parental compound 2 (IC50 = 0.21 µM), and only modest suppressive effects on 12- and 15-LOs were evident. Together, our detailed SAR study reveals 22c as highly potent 5-LO-selective lead compound in intact cells that warrants further preclinical evaluation as anti-inflammatory agent.


Subject(s)
Benzoquinones/chemistry , Lipoxygenase Inhibitors/chemistry , Lipoxygenase Inhibitors/pharmacology , Neutrophils/drug effects , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Chemistry Techniques, Synthetic , Drug Evaluation, Preclinical/methods , Humans , Inhibitory Concentration 50 , Intramolecular Oxidoreductases/metabolism , Lipoxygenase Inhibitors/chemical synthesis , Neutrophils/enzymology , Prostaglandin-E Synthases , Structure-Activity Relationship
4.
Pharmacol Res ; 94: 42-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25681061

ABSTRACT

5-Lipoxygenase (5-LO), the key enzyme in the biosynthesis of pro-inflammatory leukotrienes (LTs) from arachidonic acid, is regulated by androgens in human neutrophils and monocytes accounting for sex differences in LT formation. Here we show that progesterone suppresses the synthesis of 5-LO metabolites in human primary monocytes. 5-LO product formation in monocytes stimulated with Ca(2+)-ionophore A23187 or with lipopolysaccharide/formyl peptide was suppressed by progesterone at concentrations of 10-100 nM in cells from females and at 1 µM in cells from males. Progesterone down-regulated 5-LO product formation in a rapid and reversible manner, but did not significantly inhibit 5-LO activity in cell-free assays using monocyte homogenates. Also, arachidonic acid release and its metabolism to other eicosanoids in monocytes were not significantly reduced by progesterone. The inhibitory effect of progesterone on LTs was still observed when mitogen-activated protein kinases were pharmacologically blocked, stimulatory 1-oleoyl-2-acetyl-sn-glycerol was exogenously supplied, or extracellular Ca(2+) was removed by chelation. Instead, suppression of PKA by means of two different pharmacological approaches (i.e. H89 and a cell-permeable PKA inhibitor peptide) prevented inhibition of 5-LO product generation by progesterone, to a similar extent as observed for the PKA activators prostaglandin E2 and 8-Br-cAMP, suggesting the involvement of PKA. In summary, progesterone affects the capacity of human primary monocytes to generate 5-LO products and, in addition to androgens, may account for sex-specific effects on pro-inflammatory LTs.


Subject(s)
Arachidonate 5-Lipoxygenase/biosynthesis , Monocytes/metabolism , Progesterone/pharmacology , Arachidonic Acid/metabolism , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation/drug effects , Humans , Monocytes/drug effects , Monocytes/enzymology , Primary Cell Culture , Signal Transduction
5.
J Med Chem ; 57(9): 3715-23, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24697244

ABSTRACT

The enzymes 5-lipoxygenase (5-LO) and glycogen synthase kinase (GSK)-3 represent promising drug targets in inflammation. We made use of the bisindole core of indirubin, present in GSK-3 inhibitors, to innovatively target 5-LO at the ATP-binding site for the design of dual 5-LO/GSK-3 inhibitors. Evaluation of substituted indirubin derivatives led to the identification of (3Z)-6-bromo-3-[(3E)-3-hydroxyiminoindolin-2-ylidene]indolin-2-one (15) as a potent, direct, and reversible 5-LO inhibitor (IC50 = 1.5 µM), with comparable cellular effectiveness on 5-LO and GSK-3. Together, we present indirubins as novel chemotypes for the development of 5-LO inhibitors, the interference with the ATP-binding site as a novel strategy for 5-LO targeting, and dual 5-LO/GSK-3 inhibition as an unconventional and promising concept for anti-inflammatory intervention.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Cytokines/biosynthesis , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Indoles/chemistry , Inhibitory Concentration 50 , Molecular Structure , Monocytes/drug effects , Monocytes/metabolism
6.
Cardiovasc Res ; 101(3): 522-32, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24368834

ABSTRACT

AIMS: The small molecule indirubin-3'-monoxime (I3MO) has been shown to inhibit vascular smooth muscle cell (VSMC) proliferation and neointima formation in vivo. The influence of I3MO on VSMC migration and vascular inflammation, two additional key players during the onset of atherosclerosis and restenosis, should be investigated. METHODS AND RESULTS: We examined the influence of I3MO on VSMC migration, with focus on monocyte-derived leukotrienes (LTs) and platelet-derived growth factors (PDGFs) as elicitors. Exogenous LTB4 and cysteinyl leukotrienes as well as LT-enriched conditioned medium of activated primary human monocytes induced VSMC migration, which was inhibited by I3MO. I3MO also blunted migration of VSMC stimulated with the PDGF, the strongest motogen tested in this study. Induction of haem oxygenase 1 accounted for this anti-migratory activity of I3MO in VSMC. Notably, I3MO not only interfered with the migratory response in VSMC, but also suppressed the production of pro-migratory LT in monocytes. Conditioned media from monocytes that were activated in the presence of I3MO failed to induce VSMC migration. In cell-based and cell-free assays, I3MO selectively inhibited 5-lipoxygenase (5-LO), the key enzyme in LT biosynthesis, with an IC50 in the low micromolar range. CONCLUSION: Our study reveals a novel dual inhibitory mode of I3MO on LT-mediated VSMC migration: (i) I3MO interferes with pro-migratory signalling in VSMC and (ii) I3MO suppresses LT biosynthesis in monocytes by direct inhibition of 5-LO. These inhibitory actions on both migratory stimulus and response complement the previously demonstrated anti-proliferative properties of I3MO and may further promote I3MO as promising vasoprotective compound.


Subject(s)
Cell Movement/drug effects , Cysteine/metabolism , Indoles/pharmacology , Leukotrienes/metabolism , Myocytes, Smooth Muscle/drug effects , Oximes/pharmacology , Signal Transduction/drug effects , Arachidonate 5-Lipoxygenase/metabolism , Cell Movement/physiology , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neointima/drug therapy , Platelet-Derived Growth Factor/metabolism
7.
J Biol Chem ; 288(49): 35592-603, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24165129

ABSTRACT

We showed previously that the small molecule indirubin-3'-monoxime (I3MO) prevents vascular smooth muscle cell (VSMC) proliferation by selectively inhibiting signal transducer and activator of transcription 3 (STAT3). Looking for the underlying upstream molecular mechanism, we here reveal the important role of reactive oxygen species (ROS) for PDGF-induced STAT3 activation in VSMC. We show that neither NADPH-dependent oxidases (Noxes) nor mitochondria, but rather 12/15-lipoxygenase (12/15-LO) are pivotal ROS sources involved in the redox-regulated signal transduction from PDGFR to STAT3. Accordingly, pharmacological and genetic interference with 12/15-LO activity selectively inhibited PDGF-induced Src activation and STAT3 phosphorylation. I3MO is able to blunt PDGF-induced ROS and 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) production, indicating an inhibitory action of I3MO on 12/15-LO and consequently on STAT3. We identify 12/15-LO as a hitherto unrecognized signaling hub in PDGF-triggered STAT3 activation and show for the first time a negative impact of I3MO on 12/15-LO.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Proto-Oncogene Proteins c-sis/metabolism , STAT3 Transcription Factor/metabolism , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Becaplermin , Cell Proliferation/drug effects , Cells, Cultured , Humans , Indoles/pharmacology , Mice , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Oximes/pharmacology , Phosphorylation , RNA, Small Interfering/genetics , Rats , Reactive Oxygen Species/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , src-Family Kinases/metabolism
8.
Eur J Med Chem ; 67: 269-79, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23871907

ABSTRACT

5-Lipoxygenase (5-LO), an enzyme that catalyzes the initial steps in the biosynthesis of pro-inflammatory leukotrienes, is an attractive drug target for the pharmacotherapy of inflammatory and allergic diseases. Here, we present the discovery and biological evaluation of novel series of 1,4-benzoquinones and respective resorcinol derivatives that efficiently inhibit human 5-LO, with little effects on other human lipoxygenases. SAR analysis revealed that the potency of the compounds strongly depends on structural features of the lipophilic residues, where bulky naphthyl or dibenzofuran moieties favor 5-LO inhibition. Among the 1,4-benzoquinones, compound Ig 5-[(2-naphthyl)methyl]-2-hydroxy-2,5-cyclohexadiene-1,4-dione potently blocked 5-LO activity in cell-free assays with IC50 = 0.78 µM, and suppressed 5-LO product synthesis in polymorphonuclear leukocytes with IC50 = 2.3 µM. Molecular docking studies suggest a concrete binding site for Ig in 5-LO where select π-π interactions along with hydrogen bond interactions accomplish binding to the active site of the enzyme. Together, our study reveals novel valuable 5-LO inhibitors with potential for further preclinical assessment as anti-inflammatory compounds.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Benzoquinones/pharmacology , Drug Discovery , Lipoxygenase Inhibitors/pharmacology , Resorcinols/pharmacology , Benzoquinones/chemical synthesis , Benzoquinones/chemistry , Dose-Response Relationship, Drug , Humans , Lipoxygenase Inhibitors/chemical synthesis , Lipoxygenase Inhibitors/chemistry , Models, Molecular , Molecular Structure , Recombinant Proteins/metabolism , Resorcinols/chemical synthesis , Resorcinols/chemistry , Structure-Activity Relationship
9.
Biochem Pharmacol ; 86(4): 476-86, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23623753

ABSTRACT

Embelin (2,5-dihydroxy-3-undecyl-1,4-benzoquinone) possesses anti-inflammatory and anti-carcinogenic properties in vivo, and these features have been related to interference with multiple targets including XIAPs, NFκB, STAT-3, Akt and mTOR. However, interference with these proteins requires relatively high concentrations of embelin (IC50>4 µM) and cannot fully explain its bioactivity observed in several functional studies. Here we reveal human 5-lipoxygenase (5-LO) and microsomal prostaglandin E2 synthase (mPGES)-1 as direct molecular targets of embelin. Thus, embelin potently suppressed the biosynthesis of eicosanoids by selective inhibition of 5-LO and mPGES-1 with IC50=0.06 and 0.2 µM, respectively. In intact human polymorphonuclear leukocytes and monocytes, embelin consistently blocked the biosynthesis of various 5-LO products regardless of the stimulus (fMLP or A23187) with IC50=0.8-2 µM. Neither the related human 12- and 15-LO nor the cyclooxygenases-1 and -2 or cytosolic phospholipase A2 were significantly affected by 10 µM embelin. Inhibition of 5-LO and mPGES-1 by embelin was (I) essentially reversible after wash-out, (II) not impaired at higher substrate concentrations, (III) unaffected by inclusion of Triton X-100, and (IV) did not correlate to its proposed antioxidant properties. Docking simulations suggest concrete binding poses in the active sites of both 5-LO and mPGES-1. Because 5-LO- and mPGES-1-derived eicosanoids play roles in inflammation and cancer, the interference of embelin with these enzymes may contribute to its biological effects and suggests embelin as novel chemotype for development of dual 5-LO/mPGES-1 inhibitors.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anticarcinogenic Agents/pharmacology , Arachidonate 5-Lipoxygenase/metabolism , Benzoquinones/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Lipoxygenase Inhibitors/pharmacology , Microsomes/enzymology , Antioxidants/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Eicosanoids/antagonists & inhibitors , Free Radical Scavengers/pharmacology , Humans , Leukocytes/drug effects , Leukocytes/enzymology , Molecular Docking Simulation , Prostaglandin-E Synthases , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors
10.
J Med Chem ; 55(20): 8958-62, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22992107

ABSTRACT

mPGES-1 is a promising target for development of new anti-inflammatory drugs. We aimed to create mPGES-1 inhibitors by modifying the structure of NSAIDs by replacing the carboxylic acid functionality by sulfonamide moieties. Compounds were also tested for 5-LOX inhibition. The most potent mPGES-1 inhibitor was lonazolac derivative 22 (IC50 = 0.16 µM), while the best 5-LOX inhibition was attained by indomethacin derivative 17 (IC50 = 0.9 µM). Inhibition of COX-1 activity was completely removed.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Arachidonate 5-Lipoxygenase/metabolism , Intramolecular Oxidoreductases/antagonists & inhibitors , Lipoxygenase Inhibitors/chemical synthesis , Sulfonamides/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Line , Humans , Indomethacin/analogs & derivatives , Indomethacin/chemical synthesis , Indomethacin/chemistry , Indomethacin/pharmacology , Lipoxygenase Inhibitors/chemistry , Lipoxygenase Inhibitors/pharmacology , Microsomes/drug effects , Microsomes/metabolism , Prostaglandin-E Synthases , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacology , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology
11.
J Chromatogr A ; 1267: 217-23, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-22909891

ABSTRACT

A new sensitive method using α-cyclodextrin-modified micellar electrokinetic chromatography has been developed to separate and quantify arachidonic acid metabolites of the lipoxygenase pathways in human polymorphonuclear leukocytes, i.e. leukotriene B(4), 6-trans-leukotriene B(4), 6-trans-12-epi-leukotriene B(4), 5(S)-hydroxy-6-trans-8,11,14-cis-eicosatetraenoic acid, 12(S)-hydroxy-6-trans-8,11,14-cis-eicosatetraenoic acid, and 15(S)-hydroxy-6-trans-8,11,14-cis-eicosatetraenoic acid. The electrophoresis system was optimized with regard to the pH, boric acid, SDS and α-cyclodextrin concentration as well as separation voltage and temperature using a three level resolution IV fractional factorial design and a five level circumscribed central composite design. The resulting optimized conditions included 80 mM sodium borate buffer, pH 10.07, containing 16.6mM sodium dodecyl sulfate, and 15 mM α-cyclodextrin, using a separation voltage of 12.5 kV at 23°C. Sensitivity was enhanced employing head-column field amplified sample stacking which resulted in limits of quantification between 30 and 50 ng/mL and limits of detection between 10 and 17 ng/mL after solid phase extraction of the lipoxygenase products. The method was validated according to the recommendations of the International Conference on Harmonization and applied to the determination of the lipoxygenase metabolites in polymorphonuclear leukocytes upon stimulation with Ca(2+)-ionophore A23187 and arachidonic acid. Robustness was confirmed using a three level resolution IV fractional factorial design. The novel method is suitable for the analysis of various arachidonic acid metabolites produced by cells and may be used for evaluation of lipoxygenase inhibitors.


Subject(s)
Arachidonate 5-Lipoxygenase/chemistry , Chromatography, Micellar Electrokinetic Capillary/methods , Leukocytes/enzymology , Arachidonate 5-Lipoxygenase/metabolism , Cells, Cultured , Chromatography, Micellar Electrokinetic Capillary/instrumentation , Cyclodextrins/administration & dosage , Cyclodextrins/chemistry , Humans , Leukocytes/chemistry
12.
Bioorg Med Chem ; 20(16): 5012-6, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22795900

ABSTRACT

Microsomal prostaglandin E(2) synthase-1 (mPGES-1) has been recognized as novel, promising drug target for anti-inflammatory and anticancer drugs. mPGES-1 catalyzes the synthesis of the inducible prostaglandin E(2) in response to pro-inflammatory stimuli, rendering this enzyme extremely interesting in drug discovery process owing to the drastic reduction of the severe side effects typical for traditional non-steroidal anti-inflammatory drugs. In the course of our investigations focused on this topic, we identified two interesting molecules bearing the γ-hydroxybutenolide scaffold which potently inhibit the activity of mPGES-1. Notably, the lead compound 2c that inhibited mPGES-1 with IC(50) = 0.9 µM, did not affect other related enzymes within the arachidonic acid cascade.


Subject(s)
4-Butyrolactone/pharmacology , Enzyme Inhibitors/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , 4-Butyrolactone/chemical synthesis , 4-Butyrolactone/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Intramolecular Oxidoreductases/metabolism , Molecular Structure , Prostaglandin-E Synthases , Structure-Activity Relationship
13.
Biochem Pharmacol ; 84(5): 681-91, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22732453

ABSTRACT

Eicosanoids are potent lipid mediators derived from phospholipase (PL)-released arachidonic acid (AA) coupled to subsequent metabolism by cyclooxygenase (COX)-1/2 or lipoxygenases (LO) which are involved in a variety of homeostatic biological functions and inflammation. We have investigated three lupeolic acids (LA) from the gum resin of Boswellia carterii for their ability to interfere with eicosanoid biosynthesis in human blood cells. A novel, yet unknown C(28)-hydroxylated LA, that is, 3α-acetoxy-28-hydroxylup-20(29)-en-4ß-oic acid (Ac-OH-LA) was found to inhibit the biosynthesis of COX-, 5-LO- and 12-LO-derived eicosanoids from endogenous AA in activated platelets, neutrophils, and monocytes from human blood with consistent IC(50) values of 2.3-6.9 µM. In contrast, two other LAs lacking the C(28)-OH moiety were essentially inactive in this respect. Inhibition of eicosanoids by Ac-OH-LA correlated with reduced release of AA in intact cells. When AA was exogenously provided as substrate for cellular eicosanoid biosynthesis the inhibitory effects of Ac-OH-LA were essentially reversed, even though some inhibition of 5-LO and COX-1 product formation still remained. Finally, by means of a cell-free phospholipid hydrolysis assay using human recombinant cytosolic PLA(2)α, we show that Ac-OH-LA may directly interfere with cPLA(2)α activity (IC(50) = 3.6 µM). Together, we identified a novel, naturally occuring C(28)-hydroxylated LA which acts as efficient inhibitor of cPLA(2)α and consequently suppresses eicosanoid biosynthesis in intact cells.


Subject(s)
Cytosol/enzymology , Eicosanoids/antagonists & inhibitors , Phospholipase A2 Inhibitors , Triterpenes/pharmacology , Arachidonic Acid/blood , Cells, Cultured , Eicosanoids/biosynthesis , Humans , Hydroxylation , Magnetic Resonance Spectroscopy , Monocytes/drug effects , Monocytes/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Triterpenes/chemistry
14.
Bioorg Med Chem Lett ; 22(2): 1202-7, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22192589

ABSTRACT

The release of arachidonic acid, a precursor in the production of prostaglandins and leukotrienes, is achieved by activity of the cytosolic phospholipase A(2)α (cPLA(2)α). Signaling mediated by this class of bioactive lipids, which are collectively referred to as eicosanoids, has numerous effects in physiological and pathological processes. Herein, we report the development of a ligand-based pharmacophore model and pharmacophore-based virtual screening of the National Cancer Institute (NCI) database, leading to the identification of 4-(hexadecyloxy)-3-(2-(hydroxyimino)-3-oxobutanamido)benzoic acid (NSC 119957) as cPLA(2)α inhibitor in cell-free and cell-based in vitro assays.


Subject(s)
Aminobenzoates/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Group IV Phospholipases A2/antagonists & inhibitors , Oximes/pharmacology , Aminobenzoates/chemical synthesis , Aminobenzoates/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Group IV Phospholipases A2/metabolism , Humans , Models, Molecular , Molecular Structure , Oximes/chemical synthesis , Oximes/chemistry , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...