Nitroalkene derivatives of linoleic acid (LNO2) and oleic acid (OA-NO2) are present; however, their biological functions remain to be fully defined. Herein, we report that LNO2 and OA-NO2 inhibit lipopolysaccharide-induced secretion of proinflammatory cytokines in macrophages independent of nitric oxide formation, peroxisome proliferator-activated receptor-γ activation, or induction of heme oxygenase-1 expression. The electrophilic nature of fatty acid nitroalkene derivatives resulted in alkylation of recombinant NF-κB p65 protein in vitro and a similar reaction with p65 in intact macrophages. The nitroalkylation of p65 by fatty acid nitroalkene derivatives inhibited DNA binding activity and repressed NF-κB-dependent target gene expression. Moreover, nitroalkenes inhibited endothelial tumor necrosis factor-α-induced vascular cell adhesion molecule 1 expression and monocyte rolling and adhesion. These observations indicate that nitroalkenes such as LNO2 and OA-NO2, derived from reactions of unsaturated fatty acids and oxides of nitrogen, are a class of endogenous anti-inflammatory mediators. Nitroalkene derivatives of linoleic acid (LNO2) and oleic acid (OA-NO2) are present; however, their biological functions remain to be fully defined. Herein, we report that LNO2 and OA-NO2 inhibit lipopolysaccharide-induced secretion of proinflammatory cytokines in macrophages independent of nitric oxide formation, peroxisome proliferator-activated receptor-γ activation, or induction of heme oxygenase-1 expression. The electrophilic nature of fatty acid nitroalkene derivatives resulted in alkylation of recombinant NF-κB p65 protein in vitro and a similar reaction with p65 in intact macrophages. The nitroalkylation of p65 by fatty acid nitroalkene derivatives inhibited DNA binding activity and repressed NF-κB-dependent target gene expression. Moreover, nitroalkenes inhibited endothelial tumor necrosis factor-α-induced vascular cell adhesion molecule 1 expression and monocyte rolling and adhesion. These observations indicate that nitroalkenes such as LNO2 and OA-NO2, derived from reactions of unsaturated fatty acids and oxides of nitrogen, are a class of endogenous anti-inflammatory mediators. Reactive oxygen species and NO-derived oxidizing, nitrosating and nitrating products mediate diverse cell signaling and pathologic processes in cardiovascular, pulmonary, and neurodegenerative diseases (1Baldus S. Eiserich J.P. Brennan M.L. Jackson R.M. Alexander C.B. Freeman B.A. Free Radic. Biol. Med. 2002; 33: 1010Crossref PubMed Scopus (155) Google Scholar, 2Ischiropoulos H. Beckman J.S. J. Clin. Invest. 2003; 111: 163-169Crossref PubMed Scopus (657) Google Scholar). These reactive inflammatory mediators chemically modify carbohydrates, DNA bases, amino acids, and unsaturated fatty acids to form oxidized, nitrosated and nitrated derivatives. For example, accumulation of inflammatory-induced protein tyrosine nitration products represents a shift from the physiological signal-transducing actions of NO to an oxidative, nitrative, and potentially pathogenic pathway (1Baldus S. Eiserich J.P. Brennan M.L. Jackson R.M. Alexander C.B. Freeman B.A. Free Radic. Biol. Med. 2002; 33: 1010Crossref PubMed Scopus (155) Google Scholar). Recently, it has been reported that nitration products of unsaturated fatty acids (nitroalkenes) are formed via NO-dependent oxidative reactions (3Rubbo H Radi R. Trujillo M. Telleri R. Kalyanaraman B. Barnes S. Kirk M. Freeman B.A. J. Biol. Chem. 1994; 269: 26066-26075Abstract Full Text PDF PubMed Google Scholar, 4Rubbo H. Parthasarathy S. Barnes S. Kirk M. Kalyanaraman B. Freeman B.A. Arch. Biochem. Biophys. 1995; 324: 15-25Crossref PubMed Scopus (239) Google Scholar, 5O'Donnell V.B. Eiserich J.P. Chumley P.H. Jablonsky M.J. Krishna N.R. Kirk M. Barnes S. Darley-Usmar V.M. Freeman B.A. Chem. Res. Toxicol. 1999; 12: 83-92Crossref PubMed Scopus (250) Google Scholar). These derivatives were initially viewed to be, like nitrotyrosine, a “footprint” of NO-dependent redox reactions (3Rubbo H Radi R. Trujillo M. Telleri R. Kalyanaraman B. Barnes S. Kirk M. Freeman B.A. J. Biol. Chem. 1994; 269: 26066-26075Abstract Full Text PDF PubMed Google Scholar, 6Lima E.S. Di Mascio P. Rubbo H. Abdalla D.S. Biochemistry. 2002; 41: 10717-10722Crossref PubMed Scopus (93) Google Scholar). More recently, we have observed that nitrolinoleate (LNO2) 7The abbreviations used are: LNO2, nitrolinoleate (positional isomers 10-nitro-9-cis,12-cis-octadecadienoic acid (C10) and 12-nitro-9-cis,12-cis-octadecadienoic acid (C12)); OA-NO2, esterified nitrooleate (9- and 10-nitro-9-cis-octadecenoic acid (C9 and C10)); LPS, lipopolysaccharide; PMA, phorbol 12-myristate 13-acetate; DETA-NONOate, 3,3-bis(aminoethyl)-1-hydroxy-2-oxo-1-triazene; cPTIO, 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide; PPARγ, peroxisome proliferator-activated receptor; HO-1, heme oxygenase 1; VCAM-1, vascular cell adhesion molecule 1; IL-6, interleukin 6; TNFα, tumor necrosis factor α; MCP-1, monocyte chemoattractant protein 1; ESI, electrospray ionization; MS, mass spectrometry; DTPA, diethylenetriaminepentaacetate; OA, oleic acid; LA, linoleic acid; HUVEC, human umbilical vein endothelial cells; FBS, fetal bovine serum; ELISA, enzyme-linked immunosorbent assay; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; 15d-PGJ2, 15-deoxy-Δ12,14-prostaglandin J2. mediates pluripotent cell signaling actions, since it induces relaxation of phenylephrine-preconstricted rat aortic rings, inhibits thrombin-induced Ca2+ elevations and aggregation of human platelets, and attenuates human neutrophil superoxide generation, degranulation, and integrin expression. These cell responses are mediated by both cGMP- and cAMP-dependent and -independent mechanisms (7Lim D.G. Sweeney S. Bloodsworth A. White C.R. Chumley P.H. Krishna N.R. Schopfer F. O'Donnell V.B. Eiserich J.P. Freeman B.A. Proc. Natl. Acad. Sci. U. S. A. 2002; 99: 15941-15946Crossref PubMed Scopus (107) Google Scholar, 8Coles B. Bloodsworth A. Eiserich J.P. Coffey M.J. McLoughlin R.M. Giddings J.C. Lewis M.J. Haslam R.J. Freeman B.A. O'Donnell V.B. J. Biol. Chem. 2002; 277: 5832-5840Abstract Full Text Full Text PDF PubMed Scopus (92) Google Scholar, 9Coles B. Bloodsworth A. Clark S.R. Lewis M.J. Cross A.R. Freeman B.A. O'Donnell V.B. Circ. Res. 2002; 91: 375-381Crossref PubMed Scopus (147) Google Scholar). LNO2 positional isomers, including 9-, 10-, 12-, and 13-nitro-9,12-cis-octadecadienoic acids, have been identified as free acids in human plasma and red blood cells and as esterified components of plasma lipoproteins and red blood cell membranes (10Baker P.R. Schopfer F.J. Sweeney S. Freeman B.A. Proc. Natl. Acad. Sci. U. S. A. 2004; 101: 11577-11582Crossref PubMed Scopus (182) Google Scholar). In addition, plasma and red cell free and esterified nitrooleate (OA-NO2, isomers 9- and 10-nitro-9-cis-octadecenoic acid) was also identified in healthy human blood (11Baker P.R. Lin Y. Schopfer F.J. Woodcock S.R. Long M.H. Batthyany C. Iles K.E. Baker L.M.S. Branchaud B.P. Chen Y.E. Freeman B.A. J. Biol. Chem. 2005; 280: 42464-42475Abstract Full Text Full Text PDF PubMed Scopus (319) Google Scholar). Current knowledge indicates that enzymatically oxidized unsaturated fatty acid-derived products, such as prostaglandins, thromboxanes, leukotrienes, epoxyeicosatrienoic acids, hydroxyeicosatetraenoic acids, lipoxins, and resolvins serve as lipid mediators or autacoids. These signaling mediators act within a local microenvironment to orchestrate both physiological and pathological events, including platelet aggregation, constriction of vascular smooth muscle, neonatal development, wound healing, and resolution of inflammation (12Serhan C.N. Oliw E. J. Clin. Invest. 2000; 107: 1481-1489Crossref Scopus (135) Google Scholar, 13Smith W.L. Langenbach R. J. Clin. Invest. 2001; 107: 1491-1495Crossref PubMed Scopus (533) Google Scholar). In this context, endogenous nitrated fatty acid derivatives, such as OA-NO2 and LNO2, represent an emerging class of NO and fatty acid-derived signaling molecules (14Kalyanaraman B. Proc. Natl. Acad. Sci. U. S. A. 2004; 101: 11527-11528Crossref PubMed Scopus (64) Google Scholar). At present, the cell signaling mechanisms and detailed structure-function relationships of this family of fatty acid derivatives are an object of interest. Recently, nitrated unsaturated fatty acids were shown to be potent electrophiles that mediate reversible nitroalkylation reactions with both glutathione and the Cys and His residues of proteins. This occurs both in vitro and in vivo and is viewed to transduce redox- and NO-dependent cell signaling by inducing a covalent, thiol-reversible post-translational modification that regulates protein structure, function, and subcellular distribution (15Batthyany C. Schopfer F.J. Baker P.R.S. Durán R. Baker L.M.S. Huang Y. Cerveñansky C. Branchaud B.P. Freeman B.A. J. Biol. Chem. 2006; 281: 20450-20463Abstract Full Text Full Text PDF PubMed Scopus (237) Google Scholar). Herein, we report the effects of LNO2 and OA-NO2 on inflammatory responses in vascular cells, including monocytes/macrophages and endothelial cells. These data indicate that the nitroalkene derivatives of linoleic acid and oleic acid (LNO2 and OA-NO2) are a novel class of endogenous, electrophilic mediators in the vasculature that can exert adaptive anti-inflammatory signaling reactions via the post-translational modification of transcriptional regulatory proteins. LNO2 and OA-NO2 were synthesized, purified, and quantitated as previously described (10Baker P.R. Schopfer F.J. Sweeney S. Freeman B.A. Proc. Natl. Acad. Sci. U. S. A. 2004; 101: 11577-11582Crossref PubMed Scopus (182) Google Scholar, 11Baker P.R. Lin Y. Schopfer F.J. Woodcock S.R. Long M.H. Batthyany C. Iles K.E. Baker L.M.S. Branchaud B.P. Chen Y.E. Freeman B.A. J. Biol. Chem. 2005; 280: 42464-42475Abstract Full Text Full Text PDF PubMed Scopus (319) Google Scholar). Briefly, OA-NO2 and LNO2 were synthesized using a nitrosenylation reaction. OA-NO2 and LNO2 were purified by preparative TLC developed twice using silica HF plates and a solvent system consisting of hexane/ether/acetic acid (70:30:1, v/v/v). The regions corresponding to OA-NO2 and LNO2 were scraped and extracted. Stock concentrations of OA-NO2 and LNO2 were quantitated by chemiluminescent nitrogen analysis, using caffeine as a standard, and confirmed spectrophotometrically using an extinction coefficient (E268) of 8.22 mm–1 cm–1 for OA-NO2 in 100 mm phosphate buffer at pH 7.4. Lipopolysaccharide (LPS), phorbol 12-myristate 13-acetate (PMA), zinc protoporphryin-IX, 3,3-bis(aminoethyl)-1-hydroxy-2-oxo-1-triazene (DETA-NONOate), 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide (cPTIO), sodium nitrite (NaNO2), diethylenetriaminepentaacetate (DTPA), and Na2HPO4 were from Sigma. Rosiglitazone, 15-deoxy-Δ12,14-PGJ2, Wy14643, and GW501516 were from Cayman Chemical (Ann Arbor, MI). Streptavidin-coated plates were from Pierce. Recombinant NF-κB p65 protein and TransAMTM NF-κB p65 Chemi kit were from Active Motif (Carlsbad, CA). The synthesis of biotinylated OA, LA, OA-NO2, and LNO2 was performed as described for the synthesis of biotinylated (15S)-hydroxyeicosatetraenoic acids (16Kang L.T. Vanderhoek J.Y. Anal. Biochem. 1997; 250: 119-122Crossref PubMed Scopus (4) Google Scholar). Briefly, 1-methylpiperidine (5.3 μl, 48μmol) was added to a solution of either OA, LA, OA-NO2, or LNO2 (15 mg in 1.5 ml of methylene chloride). After cooling the mixture to –78 °C, isobutylchloroformate (6.2 μl, 48 μmol) was added. The reaction mixture was maintained at –78 °C for 45 min and then warmed to –20 °C. After 35 min, a heated (80 °C) solution of biotin hydrazide (48.6 mg, 188 μmol) in dimethylformamide (2 ml) was added. The mixture was immediately cooled and maintained at –20 °C for 40 min and then warmed to room temperature. After extraction with methylene chloride, the products were chromatographed and purified by preparative TLC developed twice using silica HF plates, using a solvent system consisting of methylene chloride/methanol (80/20, v/v). The regions corresponding to the biotinylated OA, LA, OA-NO2, and LNO2 were scraped and extracted. Stock concentrations of biotinylated OA, LA, OA-NO2, and LNO2 were quantitated by chemiluminescent nitrogen analysis using caffeine as a standard. The characterization of the biotinylated OA, LA, OA-NO2, and LNO2 was done by direct infusion into a ESI MS (ion trap) at both positive and negative mode or by ESI-liquid chromatography-MS/MS in the negative ion mode, using multiple reaction monitoring and an enhanced product information scan mode on a 4000 hybrid triple quadrupole/linear ion trap mass spectrometer (Q-Trap) (Applied Biosystems, Foster City, CA). The ESI HPLC analysis was performed by C-18 reverse phase chromatography using a MercuryMS chromatography system equipped with a Luna C18 column (Phenomenex, Belmont, CA) equilibrated in 70% A (aqueous formic acid (0.01%)) and 30% B (0.01% formic acid in acetonitrile). One minute after injection, a 2-min gradient was initiated to reach a composition of 60% A and then to 7% A over an additional 4 min, held for 1 min at 7% A, and then back to 70% A and re-equilibrated. The retention times (min) and specific multiple reaction monitorings used for each species were as follows: biotinylated LA 7.50, 519/476; biotinylated OA 7.94, 521/478; biotinylated LNO2 6.90, 564/517; biotinylated OA-NO2 7.30, 566/519. Bone marrow cells were isolated from 8–12-week-old heme oxygenase-1 (HO-1) knockout mice or wild-type mice as previously (17Feng X. Novack D.V. Faccio R. Ory D.S. Aya K. Boyer M.I. McHugh K.P. Ross F.P. Teitelbaum S.L. J. Clin. Invest. 2001; 107: 1137-1144Crossref PubMed Scopus (128) Google Scholar). Briefly, bone marrow macrophages were prepared by culturing isolated bone marrow cells in α-minimal essential medium (Invitrogen) containing 10% heat-inactivated fetal bovine serum (FBS) in the presence of 0.1 volume of culture supernatant from macrophage colony-stimulating factor-producing cells for 2 days. Human umbilical vein endothelial cells (HUVEC) were purchased from BioWhittaker (San Diego, CA). The cells were cultured in endothelial cell growth medium-2 (BioWhittaker), containing 5% fetal calf serum, human basic fibroblast growth factor, insulin-like growth factor, human epithelial growth factor, vascular endothelial growth factor, 50 μg/ml gentamicin, 50 ng/ml amphotericin-B, hydrocortisone, and ascorbic acid. Early passages (passages 3–5) of HUVEC were used for all experiments. THP-1 cells (a human monocyte cell line from ATCC, Manassas, VA; catalog number TIB-202™) were cultured in RPMI 1640 (ATCC, catalog number 30-2001) supplemented with 10% FBS. THP-1 monocytes were differentiated into macrophages with PMA (0.1 μm) for 5 days. RAW264.7 cells (a murine macrophage cell line from ATCC, catalog no. TIB-71™) were cultured in Dulbecco's modified Eagle's medium (Invitrogen) supplemented with 10% FBS. Human peripheral blood mononuclear cells were purchased from Cambrex Bio Science (East Rutherford, NJ; catalog number CC-2702) and cultured with RPMI 1640 supplemented with 10% FBS. The nonadherent cells were removed with PBS before treatment. CV-1 cells (African green monkey kidney fibroblast cell line from ATCC; catalog number CCL-70™) were cultured in Dulbecco's modified Eagle's medium/F-12 supplemented with 10% FBS. Macrophages in fresh 1% FBS or 1% delipidated FBS (Cocalico Biologicals; catalog number CBX4234)-containing culture medium were treated as indicated. Medium was collected from triplicate wells 18–20 h after treatment. The concentrations of human and mouse TNFα, IL-6, and MCP-1 released from cells into medium was measured by enzyme-linked immunosorbent assay (ELISA) kits using protocols supplied by the manufacturer (R&D Systems, Minneapolis, MN). The half-maximal concentration for LNO2 and OA-NO2 inhibition of LPS-induced cytokine synthesis (IC50) was calculated using software obtained from GraphPad Software, Inc. Total RNA from cell pellets was extracted using RNeasy kits (Qiagen Inc., Valencia, CA), and reverse transcription reactions (Advantage RT for PCR kit; Clontech) were performed with 0.5–1 μg of DNase I (Qiagen)-treated RNA. Quantitative real time reverse transcription-PCRs were carried out using the LightCycler thermocycler and the SYBR green I kits (Roche Applied Science) according to the manufacturer's recommendations. Cycle numbers obtained at the log-linear phase of the reaction were plotted against a standard curve prepared with serially diluted control samples. Expression levels of target genes were normalized by concurrent measurement of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA levels. CV-1 cells at ∼85% confluence in 24-well plates were transiently co-transfected using Lipofectamine 2000 (Invitrogen) with a plasmid containing the luciferase gene under the control of three tandem PPAR-response elements (3× PPRE TK-luciferase) in pGL3 basic vector (Promega, Madison, WI) and human PPARγ1 in pcDNA3.1 vector (Invitrogen), respectively. Green fluorescent protein expression plasmid was co-transfected as an internal control for transfection efficiency. Twenty-four hours after transfection, cells were cultured for 4 h in Opti-MEM I (Invitrogen) and then treated with various stimuli as indicated for an additional 16–20 h. RAW264.7 cells were transiently co-transfected with NF-κB-luciferase reporter (NF-κB-luc) (Stratagene, La Jolla, CA) and green fluorescent protein expression plasmids. Twenty-four hours after transfection, cells were pretreated with nitrated fatty acids or control fatty acids in 1% FBS Eagle's minimal essential medium for 16–20 h and then stimulated with LPS (1 μg/ml) for 6 h. Each transfection was performed in triplicate on at least 3 occasions. Reporter luciferase assay kits (Promega) were used to measure the luciferase activity of cells with a luminometer, according to the manufacturer's instructions (Victor II; PerkinElmer Life Sciences). Luciferase activity was normalized by green fluorescent protein units. Partially purified human recombinant p65 protein (470 units/ml) was incubated in phosphate buffer 100 mm containing 100 μm DTPA (pH 7.4) with different concentrations of LNO2, OA-NO2, LA, or OA for 1 h at room temperature. The remaining p65 activity was measured using the TransAM™ NF-κB p65 Chemi kit from Active Motif following the manufacturer's instructions. A modification was introduced into the protocol by avoiding any addition of dithiothreitol to sample and buffer preparations. The amount of p65 added to each well was 10 ng. The p65 activity was not affected by native fatty acids (LA and OA) or vehicle. Thus, the activity of p65 incubated with 1 μm of corresponding native fatty acids was considered maximal (100%). Partially purified human recombinant p65 protein (470 units/ml) was incubated in 100 mm phosphate buffer containing 100 μm DTPA (pH 7.4) with different concentrations of biotinylated LNO2, OA-NO2, LA, or OA for 1 h at room temperature. The reaction mixture (25 ng of p65) was added to streptavidin-coated plates that were previously blocked for 1 h with Tween Tris-buffered saline (TTBS: 0.5% Tween 20, 30 mm Tris, 150 mm NaCl) containing 3% albumin, incubated for 1 h at room temperature, and washed three times using TTBS containing 1% albumin. The plates were then incubated with anti-p65 in 1% albumin containing TTBS for 1 h at room temperature, washed three times, incubated with goat anti-rabbit IgG for 1 h at room temperature, and then washed four times. Color development was followed at 650 nm using tetramethyl benzidine as substrate. Cells treated under different experimental conditions were washed quickly with ice-cold PBS containing 1 mm Na3VO4, frozen in liquid nitrogen, scraped off, and lysed in Nonidet P-40 lysis buffer (1% Nonidet P-40, 25 mm HEPES (pH 7.5), 50 mm NaCl, 50 mm NaF, 5 mm EDTA, 10 nm okadaic acid, 1 mm sodium orthoavanadate, 1 mm phenylmethylsulfonyl fluoride, and 10 μm aprotinin) for 10–15 min on ice. Insoluble material was removed by centrifugation at 14,000 × g for 20 min at 4 °C. Protein concentration was measured in the cleared supernatant via Coomassie dye binding (Bio-Rad). The cell lysates (200 μg) were precipitated overnight with 50 μl of UltraLink Immobilized NeutrAvidin Plus (Pierce), or 50 μl of a 50% slurry of protein G-Sepharose 4 Fast Flow (Sigma) was added to 1 ml of cell lysate with equal amounts of protein and incubated at 4 °C for 1 h with gentle shaking. The precleared lysates (200 μg) were incubated with anti-p65 with constant agitation at 4 °C overnight and then further incubated with protein G-Sepharose 4 Fast Flow for 1 h. These precipitates were washed four times with Nonidet P-40 lysis buffer. The whole cell lysates (20 μg) or precipitates were subjected to SDS-PAGE and electrotransferred onto Hybond-ECL nitrocellulose membrane. Immunoblotting was done using anti-p65 (sc-372), anti-vascular cell adhesion molecule-1 (anti-VCAM-1; sc-13160), anti-actin (sc-1616) (Santa Cruz Biotechnology, Inc., Santa Cruz, CA), and anti-HO-1 (SPA-895) (Stressgen Biotechnologies, Inc., San Diego, CA). HUVEC were grown until 95% confluent in 48-well fibronectin-coated plates (for static adhesion analyses) or 25-mm dishes (for laminar flow analyses) and then incubated for 2 h with or without LNO2. Subsequently, HUVEC were activated with TNFα (2 ng/ml) for 16 h. Static Adhesion of THP-1 Cells on HUVEC—Fluorescently labeled monocytes treated with Cell Tracker Green (Molecular Probes, Eugene, OR) were added at a leukocyte/endothelial ratio of 10:1. After a 30-min incubation at 37 °C, unbound monocytes were removed by washing (nonadherent fraction). Both fractions (adherent and nonadherent cells) were lysed using 1 m NaOH (20 min), and fluorescence was measured at excitation = 480 nm and emission = 515 nm in a PerkinElmer Life Sciences fluorescent plate reader. Adhesion and Rolling of THP-1 Monocyte Cell on HUVEC in Laminar Flow—THP-1 cell adhesion to and rolling on HUVEC during flow were determined using a Glycotech flow chamber system (Rockville, MD) at flow rates corresponding to a wall shear rate of 0.5–1.5 dynes/cm2. Fluorescently labeled THP-1 cells were viewed on a Leica inverted fluorescence microscope equipped with differential interference contrast optics and a Hamamatsu Orca ER digital CCD camera (Compix Inc., Cranberry Township, PA). Real time images of each field were captured at 30 frames/s for 5 min using the automated image capture feature of Simple PCI software (Compix). Resulting time lapse images were analyzed by the motion tracking and analysis feature of Simple PCI software to calculate rolling velocities and the number of firmly bound cells. Cells not moving for 5 s or more were considered firmly bound cells and were counted. The rolling velocities for THP-1 cells were calculated using Simple PCI software. Values are expressed as mean ± S.D. throughout. The data were analyzed using analysis of variance with the Newman-Keuls test unless specified. Values of p < 0.05 were considered to be statistically significant. LNO2 and OA-NO2 Inhibit LPS-induced Secretion of Proinflammatory Cytokines in Macrophages—To explore the physiological actions of LNO2 and OA-NO2, we investigated the impact of synthetic LNO2 and OA-NO2 (Scheme 1), identical to that detected in the healthy human circulation (10Baker P.R. Schopfer F.J. Sweeney S. Freeman B.A. Proc. Natl. Acad. Sci. U. S. A. 2004; 101: 11577-11582Crossref PubMed Scopus (182) Google Scholar, 11Baker P.R. Lin Y. Schopfer F.J. Woodcock S.R. Long M.H. Batthyany C. Iles K.E. Baker L.M.S. Branchaud B.P. Chen Y.E. Freeman B.A. J. Biol. Chem. 2005; 280: 42464-42475Abstract Full Text Full Text PDF PubMed Scopus (319) Google Scholar), on inflammatory responses of monocytes/macrophages. The concentration-dependent effects of LNO2 and OA-NO2 on LPS-induced proinflammatory cytokine secretion by macrophages were first defined. LNO2 and OA-NO2 dose-dependently inhibited the LPS-induced secretion of proinflammatory cytokines, including IL-6, TNFα, and MCP-1, in THP1 and RAW264.7 macrophages in either lipid-rich serum (L-serum) or delipidated serum (D-serum). In THP-1 macrophages, the half-maximal concentration for LNO2 inhibition of LPS-induced proinflammatory cytokine synthesis (LNO2 IC50) was 1.2–1.9 μm (L-serum) and 1.2–1.8 μm (D-serum) for IL-6, 0.4–0.8 μm (L-serum) and 0.5–0.8 μm (D-serum) for TNFα, and 0.5–0.6 μm (L-serum) and 0.5–0.7 μm (D-serum) for MCP-1 (Fig. 1A). The half-maximal concentration for OA-NO2 inhibition of LPS-induced proinflammatory cytokine synthesis (OA-NO2 IC50) was 1.1–1.7 μm (L-serum) and 1.0–1.9 μm (D-serum) for IL-6, 0.3–0.9 μm (L-serum) and 0.5–0.8 μm (D-serum) for TNFα, and 0.4–0.6 μm (L-serum) and 0.5–0.8 μm (D-serum) for MCP-1 (Fig. 1A). In RAW264.7 cells, the LNO2 IC50 was 1.1–1.7 μm (L-serum) and 1.3–1.8 μm (D-serum) for IL-6, 1.4–1.8 μm (L-serum) and 1.2–1.8 μm (D-serum) for TNFα, and 1.1–1.7 μm (L-serum) and 1.1–1.8 μm (D-serum) for MCP-1 (Fig. 1B). The OA-NO2 IC50 was 1.8–2.5 μm (L-serum) and 1.9–2.4 μm (D-serum) for IL-6, 1.7–2.2 μm (L-serum) and 1.4–1.9 μm (D-serum) for TNFα, and 1.2–1.9 μm (L-serum) and 1.3–2.2 μm (D-serum) for MCP-1 (Fig. 1B). Similar responses were observed for the IC50 values of LNO2 and OA-NO2 in the inhibition of LPS-induced proinflammatory cytokine synthesis in primary cultures of human macrophages: LNO2 IC50, 0.5–0.7 μm (d-serum) for IL-6, 0.4–0.6 μm (D-serum) for TNFα, and 0.6–0.8 μm (D-serum) for MCP-1; OA-NO2 IC50, 0.7–0.8 μm (D-serum) for IL-6, 0.5–0.7 μm (D-serum) for TNFα, and 0.6–0.8 μm (D-serum) for MCP-1 (Fig. 1C). In addition, there were no statistically significant differences in the anti-inflammatory effects of these nitroalkenes in the presence or absence of the lipidic components of serum. The native fatty acid precursors of LNO2 and OA-NO2, LA or OA, also displayed no inhibitory effects toward LPS-induced proinflammatory cytokine secretion (Fig. 1). These results affirm that the anti-inflammatory actions of LNO2 and OA-NO2 are direct and not mediated by hydrophobic interactions or via reaction products generated by the interaction of nitroalkenes with the lipophilic components of serum or macrophage membranes. In some control experiments, it was observed that the native fatty acids LA and OA slightly inhibited LPS-induced proinflammatory cytokine synthesis (10–20% inhibition) at concentrations greater than 5 μm (not shown). The subsequent nitration of LA and OA added to cultured cells remains a subject of further investigation. Higher concentrations of LNO2 (∼5 μm) and OA-NO2 (∼5 μm) did not induce cytotoxic effects in our experimental conditions, as evidenced by an absence of alterations in cellular morphology and no detectable release of cellular lactate dehydrogenase (not shown). Collectively, our results indicate that LNO2 and OA-NO2 exert strong anti-inflammatory effects in macrophages. The close correspondence between IC50 values for suppressing LPS-induced cytokine expression and nitroalkene concentration suggests a common mechanism of action.FIGURE 1Nitrated fatty acids inhibit LPS-induced inflammatory cytokine secretion in macrophages. A–C, LNO2 and OA-NO2 inhibited LPS-induced inflammatory cytokine secretion in THP-1 and RAW264.7 macrophages and human peripheral blood mononuclear cells (hPBMC) in either lipid-rich or delipidated serum. Cells were stimulated as indicated overnight in fresh culture medium with 1% lipid-rich normal serum (L-serum) (A and B) or with 1% delipidated serum (D-serum). The secretion of proinflammatory cytokines was assessed by ELISA. Values are expressed as mean ± S.D. (n = 6). *, p < 0.05 versus LPS alone. ND, nondetectable.View Large Image Figure ViewerDownload Hi-res image Download (PPT)FIGURE 1Nitrated fatty acids inhibit LPS-induced inflammatory cytokine secretion in macrophages. A–C, LNO2 and OA-NO2 inhibited LPS-induced inflammatory cytokine secretion in THP-1 and RAW264.7 macrophages and human peripheral blood mononuclear cells (hPBMC) in either lipid-rich or delipidated serum. Cells were stimulated as indicated overnight in fresh culture medium with 1% lipid-rich normal serum (L-serum) (A and B) or with 1% delipidated serum (D-serum). The secretion of proinflammatory cytokines was assessed by ELISA. Values are expressed as mean ± S.D. (n = 6). *, p < 0.05 versus LPS alone. ND, nondetectable.View Large Image Figure ViewerDownload Hi-res image Download (PPT) The Inhibition of Proinflammatory Cytokine Secretion by LNO2 and OA-NO2 in Macrophages Is NO-independent—Nitrated fatty acids can undergo Nef-like decay reactions in aqueous milieu that yield NO (18Schopfer F.J. Baker P.R. Giles G. Chumley P. Batthyany C. Crawford J. Patel R.P. Hogg N. Branchaud B.P. Lancaster Jr., J.R. Freeman B.A. J. Biol. Chem. 2005; 280: 19289-19297Abstract Full Text Full Text PDF PubMed Scopus (157) Google Scholar). The partition coefficient of LNO2 is ∼1,500:1 (hydrophobic versus aqueous compartments). Thus, only 1 in 1,500 molecules of LNO2 are expected to decay to yield NO in serum lipoprotein-containing media and cell models, yielding at the most femtomolar concentrations of NO. OA-NO2 is relatively more stable in aqueous milieu and only minimally decays to release NO (not shown). Furthermore, it has been reported that very high concentrations of the NO donor DETA-NONOate (0.1–1.0 mm) are required to inhibit LPS-induced expression of TNFα