OXIDIZED LOW DENSITY LIPOPROTEIN REGULATES APOPTOSIS AND GROWTH FACTOR PRODUCTION IN MACROPHAGES by MAZIAR RIAZY M.D. Tehran University of Medical Sciences, 2000 A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY in THE FACULTY OF GRADUATE STUDIES (Experimental Medicine) THE UNIVERSITY OF BRITISH COLUMBIA (Vancouver) December 2009 © Maziar Riazy 2009 ii Abstract Macrophages and oxidized LDL (oxLDL) both play key roles in the pathogenesis of atherogenesis. This thesis is focused on the effects of oxLDL on macrophage cytokine secretion and macrophage survival. The first project defines some of the mechanisms by which oxLDL increases secretion of vascular endothelial growth factor (VEGF), a pro-inflammatory growth factor known to be involved in atherogenesis. We show that both the protein and lipid components of oxLDL contribute to induction of VEGF secretion. Disruption of the genes for CD36, SR-A, or LOX-1 scavenger receptors had no effect. The atypical protein kinase c (PKCδ) was activated by oxLDL, and this activation was essential for the induction of VEGF. OxLDL could be atherogenic through increasing macrophage number within the plaque. Our group has previously shown that oxLDL induces growth and inhibits apoptosis in macrophages. In the second project, we sought to determine if members of the scavenger receptor family were required for the prosurvival effect of oxLDL in macrophages. We used mouse strains lacking different scavenger receptors and found that oxLDL-mediated survival is not dependent on CD36, SR-A, LOX-1, TLR4, its signaling partner CD14, or FcγRIIb. Significant inhibition of oxLDL uptake by a combined inactivation of CD36 and SR-A did not reduce the prosurvival effect of oxLDL. In the third project, we sought to characterize the oxidative modification of LDL that is responsible for the prosurvival effect. We found that both protein and lipid components of oxLDL can induce growth in macrophages. This seems to be mediated by modification of amino groups in apoB or in phosphatidylethanolamine by lipid peroxidation products. Further characterization of these oxidation products suggested that unfragmented hydroperoxide or endoperoxide-containing oxidation products of linoleic acid and arachidonic acid derivatize amino groups. When LDL or other proteins are modified in this fashion, they acquire the ability to induce pro- survival signaling pathways in macrophages. HPLC-MSMS studies showed that some of the arachidonic acid-derived lysine adducts are isolevuglandin (isoLG)-derived adducts that contain lactam and hydroxylactam rings. MSMS analysis of linoleic acid autoxidation adducts was consistent with 5 or 6 membered nitrogen-containing heterocycles derived from unfragmented oxidation products. iii Table of contents Abstract ............................................................................................................................... ii Table of contents ................................................................................................................ iii List of tables ..................................................................................................................... viii List of figures ..................................................................................................................... ix List of abbreviations ......................................................................................................... xii Acknowledgements ......................................................................................................... xvii Dedication ...................................................................................................................... xviii Co-authorship statement .................................................................................................. xix 1. Introduction ....................................................................................................................1 1.1 Atherosclerosis .............................................................................................................1 1.1.1 Definition and epidemiology .................................................................................1 1.1.2 Structure of the atherosclerotic plaque…………... ...............................................2 1.1.3 Lipid theory of atherogenesis: cholesterol and low-density lipoprotein ...............3 1.1.4 Response-to-injury hypothesis: shear stress and endothelial dysfunction ............4 1.1.5 Response-to-retention hypothesis .........................................................................6 1.1.6 Modified low-density lipoprotein and foam cell formation ..................................6 1.1.7 Oxidative modification hypothesis of atherosclerosis ..........................................8 1.1.8 Immune system and atherosclerosis ......................................................................9 1.2 Monocytes, macrophages, and atherosclerosis ..........................................................13 1.2.1 Monocyte/macrophage function and heterogeneity ............................................13 1.2.2 Pattern recognition receptors in macrophages.....................................................14 1.2.3 Role of monocytes/macrophages in atherosclerosis ............................................16 1.2.4 OxLDL and macrophages ...................................................................................17 1.2.5 Oxidized LDL and macrophage toxicity and apoptosis ......................................18 iv 1.2.6 Oxidized LDL and macrophage growth and survival .........................................19 1.2.7 Oxidized LDL-mediated macrophage survival vs apoptosis ..............................22 1.2.8 Macrophage survival vs apoptosis in atherosclerosis .........................................24 1.3 Vascular endothelial growth factor ............................................................................26 1.3.1 Discovery of VEGF and its functions .................................................................26 1.3.2 VEGF expression ................................................................................................27 1.3.3 VEGF and atherosclerosis ...................................................................................29 1.4 Lipid oxidation ...........................................................................................................31 1.4.1 Oxidation of polyunsaturated fatty acids ............................................................31 1.4.2 Oxidation of cholesterol and cholesterol esters ...................................................36 1.4.3 Oxidation of phospholipids .................................................................................38 1.4.4 Studying lipid oxidation ......................................................................................41 1.4.5 Modification of proteins by fatty acid oxidation products ..................................42 1.5 Objectives ..................................................................................................................48 2. Materials and methods ................................................................................................49 2.1 Materials ...................................................................................................................49 2.2 Animals ....................................................................................................................50 2.3 Cell culture ...............................................................................................................50 2.4 Lipoprotein isolation, modification, and labeling ....................................................51 2.5 Lipoprotein characterization and analytical procedures ...........................................52 2.6 Modification of lysine methyl ester with PUFA oxidation products .......................52 2.7 Lipid extraction, liposome preparation and modification ........................................53 2.8 apoB solubilization ...................................................................................................53 2.9 Cell viability assay ...................................................................................................54 2.10 Apoptosis assay .......................................................................................................54 v 2.11 ELISA ......................................................................................................................55 2.12 Immunoblotting .......................................................................................................55 2.13 PKCδ immunoprecipitation and in vitro kinase assay .............................................56 2.14 Lipoprotein uptake assay .........................................................................................56 2.15 Quantitative reverse transcription polymerase chain reaction ................................57 2.16 Hydroperoxide measurement ..................................................................................57 2.17 Mass spectrometry and tandem mass spectrometry ...............................................58 2.18 High performance liquid chromatography-coupled mass spectrometry and tandem mass spectrometry ..................................................................................................58 2.19 Statistical analysis ...................................................................................................59 3. OxLDL-mediated VEGF secretion in macrophages.................................................61 3.1 Introduction/rationale .................................................................................................61 3.2 Results ........................................................................................................................62 3.2.1 OxLDL induces VEGF expression and secretion in macrophages .....................62 3.2.2 Type and degree of modification influences oxLDL-mediated VEGF secretion .............................................................................................................................62 3.2.3 OxLDL and ceramide phosphate both increase macrophage secretion through different mechanisms ..........................................................................................63 3.2.4 Scavenger receptors and oxLDL-mediated VEGF secretion ..............................64 3.2.5 VEGF secretion is independent of oxLDL uptake ..............................................64 3.2.6 OxLDL-mediated VEGF secretion is independent of MAPKK .........................65 3.2.7 PI3K is important in oxLDL-induced VEGF secretion ......................................65 3.2.8 OxLDL-induced VEGF secretion is independent of classical and novel PKC isoforms ..............................................................................................................65 3.2.9 PKCδ mediates oxLDL-induced VEGF secretion ...............................................66 vi 3.3 Summary/discussion ..................................................................................................66 4. Receptors for oxLDL and its prosurvival effect in macrophages ...........................86 4.1 Introduction/rationale ................................................................................................86 4.2 Results .......................................................................................................................86 4.2.1 LOX-1 signaling is not required for oxLDL prosurvival activity .....................86 4.2.2 OxLDL-mediated macrophage survival is independent of CD36 or SR-A receptors ...........................................................................................................87 4.2.3 OxLDL-mediated macrophage survival is independent of oxLDL uptake .......87 4.2.4 Toll-like receptor 4 (TLR4) is not essential for oxLDL-mediated survival ......88 4.2.5 CD14 is not required for oxLDL prosurvival activity .......................................88 4.2.6 Fc gamma receptor IIb (FcγRIIb) and oxLDL-mediated survival ....................88 4.3 Summary/discussion ..................................................................................................89 5. Fluorescent adducts formed by reaction of oxidized unsaturated fatty acids with amines induce macrophage growth ..........................................................................108 5.1 Introduction/rationale ...............................................................................................108 5.2 Results ......................................................................................................................108 5.2.1 Both lipid and protein moiety of oxLDL promote macrophage growth ...........108 5.2.2 OPs react with lysine residues ...........................................................................109 5.2.3 Amine modification by OPs is sufficient for growth induction ........................109 5.2.4 Borohydride-reducible functional groups in OPs are essential for the growth- promoting effect ................................................................................................109 5.2.5 Alkenals do not account for the growth-inducing activity of OPs ....................110 5.2.6 Hydroperoxides and/or endoperoxides contribute to the induction of growth by OPs ...................................................................................................................110 vii 5.2.7 Autoxidation of arachidonic and linoleic acids produces mainly unfragmented oxygenated species ............................................................................................112 5.2.8 OP modification of lysine and apoB generate fluorescent adducts ...................115 5.2.9 Unfragmented linoleic acid autoxidation products can modify lysine methyl ester ...........................................................................................................................115 5.2.10 Putative structures of LOP-LME adducts .......................................................116 5.2.11 Unfragmented arachidonic acid autoxidation products can modify lysine methyl ester .................................................................................................................119 5.2.12 AOP-LME adducts include isolevuglandin-modified LME ...........................122 5.2.13 Fluorescent peaks of AOP-LME correspond to isolevuglandin-derived adducts ........................................................................................................................122 5.3 Summary/discussion ................................................................................................123 6. Summary.....................................................................................................................149 7. References ...................................................................................................................152 viii List of tables 1. Introduction Table 1.1 Cell survival and apoptosis with various methods of LDL oxidation ............23 5. Fluorescent adducts formed by reaction of oxidized unsaturated fatty acids with amines induce macrophage growth Table 5.1 Hydroperoxides and/or endoperoxides contribute to the induction of growth by OPs ..........................................................................................................111 Table 5.2 Time course analysis of polyunsaturated fatty acid autoxidation products .....................................................................................................................114 Table 5.3 ESI-MSMS analysis and possible assignments of adducts of lysine methyl ester with autoxidized linoleic acid. ............................................................118 Table 5.4 Elemental composition analysis of AOP-LME adducts...............................121 ix List of figures 1. Introduction Figure 1.1 Postulated role of oxLDL in the formation of atherosclerotic lesions .............12 Figure 1.2 Working model of oxLDL-mediated inhibition of apoptosis ...........................21 Figure 1.3 Autoxidation of linoleic acid ............................................................................35 Figure 1.4 Autoxidation of phospholipids .........................................................................39 Figure 1.5 Protein modification by reactive products of PUFA oxidation ........................45 Figure 1.6 Protein modification by levuglandins and isolevuglandins ..............................46 Figure 1.7 Concerted reaction between linoleic acid hydroperoxides and lysine ..............47 3. OxLDL-mediated VEGF secretion in macrophages Figure 3.1 OxLDL increases VEGF expression and secretion in RAW264.7 ...................71 Figure 3.2 OxLDL increases VEGF secretion in primary macrophages ...........................73 Figure 3.3 Effect of degree of LDL oxidation on VEGF secretion. ..................................74 Figure 3.4 Effect of type of apoB modification on VEGF secretion .................................75 Figure 3.5 Ceramide-1-phosphate increases VEGF secretion in RAW267.4 cells ...........76 Figure 3.6 Ceramide inhibits Cer-P-mediated, but not oxLDL-mediated VEGF secretion .........................................................................................................................77 Figure 3.7 LOX-1, SR-A, and CD36 are not essential for oxLDL-mediated VEGF secretion .........................................................................................................................78 Figure 3.8 OxLDL uptake is significantly reduced in SR-A/CD36 double knockout MPMs .........................................................................................................................79 Figure 3.9 MAPKK is not essential for oxLDL-mediated VEGF secretion ......................80 Figure 3.10 Role of PI3K in oxLDL mediated VEGF secretion in RAW264.7 cells ........81 Figure 3.11 Role of PI3K in oxLDL mediated VEGF secretion in hMDMs .....................82 x Figure 3.12 OxLDL-induced VEGF secretion is independent of classical and novel PKC isoforms............................................................................................................83 Figure 3.13 PKCδ mediates induction of VEGF secretion by oxLDL ..............................84 Figure 3.14 OxLDL induces PKCδ activation ...................................................................85 4. Receptors for oxLDL and its prosurvival effect in macrophages Figure 4.1 LOX-1 is not essential for oxLDL-mediated survival ......................................92 Figure 4.2 LOX-1 is not essential for the anti-apoptotic effect of oxLDL ........................93 Figure 4.3 SR-A and CD36 are not required for oxLDL-mediated survival .....................95 Figure 4.4 OxLDL-mediated inhibition of apoptosis is independent of SR-A and CD36 ..........................................................................................................................96 Figure 4.5 OxLDL uptake in BMDMs depends on SR-A/CD36 but not LOX-1 ..............98 Figure 4.6 TLR4 does not contribute to the prosurvival effect of oxLDL ........................99 Figure 4.7 TLR4 is not essential for the anti-apoptotic effect of oxLDL ........................100 Figure 4.8 CD14 is not essential for the prosurvival effect of oxLDL ............................102 Figure 4.9 CD14 does not contribute to the anti-apoptotic effect of oxLDL ..................103 Figure 4.10 Fc gamma receptor IIb (FcγRIIb) is not required for the prosurvival effect of oxLDL ..........................................................................................................105 Figure 4.11 Anti-apoptotic effect of oxLDL and Fc gamma receptor IIb (FcγRIIb) ......106 5. Fluorescent adducts formed by reaction of oxidized unsaturated fatty acids with amines induce macrophage growth Figure 5.1 Lipid and protein fractions of oxLDL promote macrophage growth ............129 Figure 5.2 OPs react with lysine residues ........................................................................130 Figure 5.3 Amine modification by OPs is sufficient for growth induction .....................131 xi Figure 5.4 Protein modification by reactive products of PUFA oxidation ......................132 Figure 5.5 Borohydride-reducible functional groups in OPs are essential for the growth- promoting effect .............................................................................................133 Figure 5.6 Alkenals do not account for growth-inducing activity of OPs .......................134 Figure 5.7 ESI-MS analysis of neat and thermally oxidized linoleic and arachidonic acids ........................................................................................................................135 Figure 5.8 ESI-MS analysis of adducts formed by modification of lysine methyl ester with oxidized linoleic acid .....................................................................................136 Figure 5.9 MSMS analysis and putative structures of m/z = 455 in LOP-LME .............137 Figure 5.10 HPLC-MS study of AOP-LME adducts .......................................................139 Figure 5.11 m/z of 475, 491, 493, and 509 are unique to AOP-LME reaction mixture ........................................................................................................................141 Figure 5.12 AOP-LME adducts are singly charged and protonated species ...................142 Figure 5.13 m/z = 509 is an isolevuglandin-derived hydroxylactam-containing adduct ........................................................................................................................143 Figure 5.14 m/z = 493 is an isolevuglandin-derived lactam-containing adduct ..............145 Figure 5.15 HPLC-MSMS analysis of m/z = 493 ...........................................................147 Figure 5.16 Fluorescent peaks of AOP-LME correspond to m/z of 491 .........................148 xii List of abbreviations 4-HHE 4-hydroxyhexenal 4-HNE 4-hydroxynonenal AA arachidonic acid ACAT acyl-coenzyme A:cholesterol acyltransferase Ac-LDL acetylated LDL AOP-LDL arachidonate oxidation product-modified LDL APCI atmospheric-pressure chemical ionization apo apolipoprotein aSMase acid sphingomyelinase BCA bicinchoninic acid Bcl B-cell lymphoma BHT butylated hydroxytoluene BMDM bone-marrow derived macrophage CCR2 chemokine receptor 2 Cer-P ceramide-1-phosphate CHD coronary heart disease CIS-MS ion-spray mass spectrometry CM chylomicron COP conjugatable oxidation products CRP C-reactive protein CX3CR1 receptor for fractalkine DiI 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate DiI-LDL DiI labeled LDL DiI-oxLDL DiI labeled oxLDL EC endothelial cell EDTA ethylenediaminetetraacetic acid eEF2K eukaryotic elongation factor 2 kinase eIF4E eukaryotic initiation factor 4E E-LDL multiple-enzyme modified LDL eNOS endothelial nitric oxide synthase EPOX-chol epoxy cholesterol Erk extracellular signal-regulated kinase ES- negative ion scan mode ES+ positive ion scan mode xiii ESI electrospray ionization FAK focal adhesion kinase FBS fetal bovine serum FcγRIIb Fc gamma receptor IIb FeLDL ferrous-ion modified LDL GC gas chromatoghraphy GM-CSF granulocyte/macrophage colony stimulating factor HDL high-density lipoprotein HIF hypoxia-inducible factor hMDM human monocyte-derived macrophage HMG-CoA 3-hydroxy-3-methyl-glutaryl-CoA HO-1 heme oxygenase 1 HODE hydroxy octadecadienoic acid HPLC high performance liquid chromatography HPODE hydroperoxy octadecadienoic acid HRE hypoxia-responsive element ICAM-1 intercellular cell adhesion molecule 1 IFNγ interferon γ IHD ischemic heart disease IL interleukin iNOS inducible nitric oxide synthase IRES internal ribosome entry site isoLG isolevuglandin isoP isoprostane JAM junctional adhesion molecule K-chol keto cholesterol LA linoleic acid LDL low-density lipoprotein LDLR LDL receptor LEE lysine ethyl ester LG levuglandin LME L-lysine-methyl ester LOP-LDL linoleate oxidation product-modified LDL LOX-1 lectin-like oxidized LDL receptor 1 LP lipoprotein xiv LPC lysophosphatidylcholine LPL lipoprotein lipase LPO lipoxygenase LPS lipopolysaccharide LRR leucine-rich repeat LXR liver X receptor MALDI matrix-assisted laser desorption ionization MAPK mitogen-activated protein kinase MCP-1 monocyte chemotactic protein 1 M-CSF macrophage colony stimulating factor MDA malondialdehyde Mertk mer tyrosine kinase MIF migration inhibitory factor mM millimolar mmol 10 -3 moles MMP matrix metalloproteinase MPM mouse peritoneal macrophage MPO myeloperoxidase MS mass spectrometry MSMS tandem mass spectrometry mTOR mammalian target of rapamycin MTS 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)- 2H-tetrazolium NaBH4 sodium borohydride NAD(P)H nicotinamide adenine dinucleotide (phosphate) NF-κB nuclear factor-κB NO nitric oxide NOD nucleotide-binding oligomerization domain NRP neuropilin OH-chol hydroxy cholesterol OO·-chol hydroperoxy cholesterol radical OOH-chol hydroperoxyl cholesterol OP oxidation product OP-LDL oxidation product-modified LDL oxLDL oxidized LDL xv oxPL oxidized phospholipid p70S6K ribosomal protein S6 kinase I PAD peripheral arterial disease PAF platelet activating factor PAMP pathogen-associated molecular patterns PAPC 1-palmitoyl, 2-arachidonyl, phosphatidylcholine PC phosphatidylcholine PDK-1 phosphoinositide dependent protein kinase 1 PE phosphatidylethanolamine p-HA p-hydroxyphenylacetaldehyde PI3K phosphotidylinositol-3 kinase PI propidium iodide PKB protein kinase B PKC protein kinase C PL phospholipid PLA2 phospholipase A2 PLGF placental growth factor PLPC 1-palmitoyl, 2-linoloyl, phosphatidylcholine PMA phorbol 12-myristate 13-acetate PMS phenazine methosulfate PON paraoxonase PPAR peroxisome proliferator-activated receptor PRR pattern recognition receptor PSGL-1 P-selectin glycoprotein ligand-1 PUFA polyunsaturated fatty acid RAGE receptor for advanced glycation products RCT reverse cholesterol transport ROS reactive oxygen species RTK receptor tyrosine kinase SD standard deviation SMase sphingomyelinase SMC smooth muscle cell SR scavenger receptor SR-A scavenger receptor A SR-B scavenger receptor B xvi SRE sterol regulatory element SREBP sterol regulatory element-binding protein SR-PSOX scavenger receptor for phosphatidylserine and oxidized LDL sSMase secretory sphingomyelinase STAT signal transducer and activator of transcription TBA 2-thiobarbituric acid TBARS TBA-reactive substances TF tissue factor TGF transforming growth factor Th T helper lymphocyte TLC thin-layer chromatoghraphy TLR toll-like receptor TNF tumor necrosis factor TPP triphenylphosphine UTR untranslated region VCAM vascular cell adhesion molecule VEC vascular endothelial cell VEGF vascular endothelial growth factor VEGFR vascular endothelial growth factor receptor VLDL very low-density lipoprotein VPF vascular permeability factor VSMC vascular smooth muscle cell WT wild-type XO xanthine oxidase XTT 2,3-bis[2-methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxanilide ε-DNA etheno-DNA xvii Acknowledgments This thesis would not have been possible without the help and support of many great individuals to whom I am greatly indebted. First and foremost I would like to thank my supervisor, Dr. Urs P. Steinbrecher, for his mentorship, for providing me the opportunity to learn and grow and for offering guidance yet allowing me to exercise independence. I would also like to thank Dr. Vincent Duronio who has always been approachable and provided great scientific insight. In addition, I would like to extend my gratitude to my supervisory committee Drs. Megan K. Levings, David Granville, and Cheryl Wellington. Dr. Antonio Gómez-Muñoz, a collaborator from the University of the Basque Country, provided valuable discussions and advice. Chapter 5 of this dissertation was made possible by collaborators with expertise in mass spectrometry. In particular, I express my sincere gratitude to Dr. Guenter Eigendorf from the UBC Department of Chemistry and Dr. Emma S. Guns from the UBC Department of Urologic Sciences. An invaluable benefit of my PhD journey has been the friendships I have acquired which made the completion of this thesis much easier and enjoyable. Dr. Sarwat Jamil, Dr. Joseph Anthony, Dr. Shih Wei Wang, Payman Hojabrpour, Stephanie Cheah, Rupinder Dhesi and Andrew Ming-Lum have all provided me with intellectual, scientific, and collective support within the laboratory environment and beyond. In particular, I would like to acknowledge Dr. Johnny H. Chen, a great colleague and friend whose scientific approach to research and humanitarian approach to life has been inspirational. During my PhD program, I had the privilege of working with and learning from skilled technicians and research associates. Special thanks are owed to Payman Hojabrpour and Marilee Lougheed for their contributions to this work. I am particularly grateful to Hans Adomat from the Laboratory of Dr. Emma S Guns for coaching me on HPLC-MS and HPLC-MSMS methodologies and interpretation. And last but certainly not least, I wish to thank Mr. Patrick Carew, for diligently providing all graduate students in the UBC Experimental Medicine program with administrative support. xviii Dedication This work is dedicated to my mother, father, and brother. Thank you for your unconditional love and support throughout my life. xix Co-authorship statement I would like to acknowledge that Ms. Marilee Lougheed performed the experiments that are shown in Figures 5.1, 5.2, 5.3, 5.4, panel C of Figure 5.7, and Table 5.1. She was also responsible for the initial mass spectrometry experiments that were carried out in collaboration with Dr. Guenther Eigendorf in the UBC Department of Chemistry. I was responsible for the analysis and interpretation of these spectra as well as of the later HPLC-MSMS studies that I performed with the assistance of Mr. Hans Adomat. Although chapter 5 is based on collaborative experimental work, as acknowledged here, I was solely responsible for the writing of the chapter. 1 1. Introduction 1.1 Atherosclerosis 1.1.1 Definition and epidemiology Atherosclerosis is a chronic inflammatory disease of large and medium-sized arteries characterized by the accumulation of lipids, cells, and connective tissue within the arterial wall. Plaques may progressively increase in size so that they eventually compromise blood flow, or the plaque can ulcerate or rupture, leading to clot formation. Either event can cause ischemia in distal regions supplied by that artery. Atherosclerosis is the most common cause of coronary heart disease or CHD (which includes myocardial infarction and angina pectoris), ischemic cerebrovascular accidents (strokes), and peripheral arterial disease (PAD) [1]. The World Health Organization Global Burden of Disease project reported in 2008 that ischemic heart disease followed by cerebrovascular diseases are respectively the top two leading causes of death worldwide. It was also projected in that report that they will remain among the top 4 leading causes of death and that the disease burden for both will continually rise by 2030 [2]. In North America, lifestyle choices have made atherosclerosis a particularly prevalent health issue. Increased attention to smoking cessation and control of risk factors has resulted in a decline in the number of cardiovascular deaths in the past 20 years, but the burden for society remains very high. In Canada, one out of every three deaths is caused by cardiovascular disease with CHD accounting for more than 50% [3]. In the United States of America, one out of every 5 deaths is due to CHD. It is estimated that every minute four Americans will have a coronary episode, of which one will be fatal. In the USA, the estimated 2009 health care cost attributed to CHD and stroke is $165.4 billion [4]. 1.1.2 Structure of the atherosclerotic plaque Even though the clinical presentation varies depending on the site of plaque formation, the pathophysiological mechanisms and the structure of the plaque remains similar. The development of the atherosclerotic plaque has been classified into 6 stages according to the Stary classification [5-7]. Type I lesions or “initial lesions” are generally visible only with a microscope, and show thickening of the 2 intima and an increased number of macrophages in the intima, some of which contain lipid droplets (macrophage foam cells - Figure 1) [6]. As the lesion further progresses, foam cells increase in number and group together to produce type II lesions. In addition to macrophages, other inflammatory cells such as T lymphocytes have been reported in type II lesions though in small numbers [8]. Quite often, type II lesions become elevated and grossly visible as “fatty streaks”. Type III lesions represent a transitional stage which bridges between early lesions (the first two types) and more advanced types and therefore are considered “intermediate lesions”. Type III lesions are marked by appearance of lipid droplets outside the cells [8]. Expansion and coalition of these free lipid droplets form a distinct “lipid core” which completes the transition from early to a more advanced stage of atherosclerotic plaque (“atheroma” - type IV) [5, 6]. A common theme in all advanced lesions is that they are formed by repeated cycles of inflammation, perhaps driven by modified lipids and the influx of inflammatory cells. The inflammatory response leads to disorganization of normal artery wall architecture [5, 9]. As the atheroma progresses, a distinct fibrous layer (fibrous cap) separates the lipid core from the endothelium, and this is a hallmark of type V lesions. Type VI lesions are those that have undergone complications such as fissuring, ulceration, or dissection - all of which are often associated with thrombosis (Figure 1.1 and [5]). The mechanisms responsible for plaque initiation and progression are not entirely understood. However, a large amount of information is available and will be reviewed briefly in the following section. 1.1.3 Lipid theory of atherogenesis: cholesterol and low-density lipoprotein A strikingly early and permanent constituent of all stages of an atherosclerotic plaque is the lipid - mainly cholesterol delivered by apolipoprotein B (apoB) containing lipoproteins. The lipid theory of atherosclerosis proposes a causal role for lipid transudation into the intimal layer and essentially states that increased cholesterol level (mainly low-density lipoprotein (LDL) -associated cholesterol) is atherogenic. Early in the 20 th century, Anitschkow showed experimentally that diet-induced hypercholesterolemia by itself is sufficient to induce atherosclerosis [10]. As hydrophobic compounds, 3 lipids in free form cannot be found in aqueous phases. In plasma, lipids can be found in association with specialized proteins (apolipoproteins) in water-soluble complexes known as lipoproteins (LPs). Two widely used methods for classification of LPs are based on hydrated density or mobility on agarose gel electrophoresis. Goffman first described different classes of lipoproteins based on analytical ultracentrifugation [11] and the principle of separation of lipoproteins by density was extended to preparative ultracentrifugation by Havel et al [12]. Based on density, lipoproteins are classified as chylomicron (CM) with a density <0.94 g/mL, very low-density lipoprotein (VLDL, 0.94 250 μm, the frequency of neovascularization is increased nine-fold relative to smaller plaques. Furthermore, inhibition of angiogenesis can slow plaque growth [411]. Neoangiogenesis also seems to be important in plaque instability and rupture (reviewed in [412]). VEGF can promote plaque growth by way of its angiogenic properties. In addition, it has pro- inflammatory properties such as recruitment of macrophages and induction of vascular permeability. As mentioned, VEGF can promote monocyte migration through VEGFR1 on the surface of monocytes [383]. A more recent study however, showed VEGFR1 signaling is not essential for monocyte recruitment to the plaques [413]. In this study, it was shown that VEGF-induced MCP-1 production in VSMCs was responsible for monocyte recruitment. Lucerna et al. recently showed that neointimal VEGF upregulation advances plaque growth and promotes plaque instability possibly through recruitment of inflammatory cells (as opposed to promoting angiogenesis) [414]. VEGF is also capable of inducing MCP-1 in ECs [415]. Increased expression of VEGF has been documented in atherosclerotic plaques of humans and animals and this correlated with the plaque stage [416-419]. Increased VEGF delivery in cholesterol-fed apoB100/apoE double knockout mice and in cholesterol-fed rabbits increased the extent of 30 atherosclerosis [420, 421]. On the other hand, inhibitors of VEGF-dependent angiogenesis inhibited intimal neovascularization and reduced atherosclerosis in animal models of atherosclerosis [422, 423]. It was recently shown that oral vaccination against VEGFR2 (flk-1) reduced its expression in endothelial cells of LDLR -/- mouse and also reduced neoangiogenesis with an overall atheroprotective effect [424]. The clinical significance of VEGF in atherosclerosis is still under debate [425]. VEGF could also be beneficial as it can promote arteriogenesis and establish collateral perfusion. Clinical trials such as VIVA [426] and others (reviewed in [427] ) have suggested a potential therapeutic role for this cytokine in management of coronary artery disease . A gene polymorphisim study suggested a protective role for VEGF [428]. Asahara et al. have shown that VEGF promotes endothelialization after interventions such as stenting and reduces the risk of stent re-stenosis [429]. However, other groups have shown that increased VEGF correlates with increased hyperplasia and re-stenosis [430, 431]. The Kuipo Angiogenesis Trial (KAT) showed no effect of VEGF in preventing re-stenosis [432]. Overall, VEGF may be a double-edged sword. Within the plaque environment, it is implicated as an important factor in lesion neovascularization, which may lead to plaque growth, instability, and rupture. On the other hand, VEGF can act to reestablish perfusion within an area by promoting angiogenesis mediated collateral formation. 31 1.4 Lipid oxidation Lipid peroxidation can be initiated by exposure to strong oxidants such as singlet O2 or ozone (O3), or through reactions catalyzed by light (photoxidation), radical-producing agents, or enzymes (enzymatic oxidation). In the following sections, oxidation of different lipids will be discussed with a focus on oxidation of polyunsaturated fatty acids (PUFA). 1.4.1 Oxidation of polyunsaturated fatty acids The main polyunsaturated fatty acids (PUFAs) in LDL are linoleic acid (LA; 18:2, n-6) and arachidonic acid (AA; 24:4, n-6). Each molecule of LDL contains 1100 molecules of LA and 153 molecules of AA, mostly esterified to phospholipids or cholesterol [316]. LA and AA are also released during lipolysis of triglyceride-rich lipoproteins. Arachidonic acid can also be released from cell membrane phospholipids by PLA2 as the initial step in the arachidonic acid cascade, which involves three major branches: cyclooxygenase, lipoxygenase, and epoxidase [433-435]. Non-enzymatic oxidation of PUFAs generates products some of which structurally differ from their enzymatically-generated cousins. At first, as a simpler model, I will explain the general mechanism for autoxidation of LA (with two unsaturated bonds) and at each relevant section expand that to explain more complicated products of AA autoxidation. LA has one active bis-allylic methylene group (carbon 11, in the middle of: –C=C–C–C=C– (Figure 1.3)). The resonance structure makes carbon 11 electron-rich and prone to hydrogen abstraction, which yields an unstable radical intermediate with a hybrid pentadienyl configuration (Figure 1.3). This structure can react with oxygen either at carbon 9 or 13 to produce the corresponding 9- or 13- hydroperoxy radical. This also results in rearrangement of the double bond to form a conjugated diene (conjugated denotes the two double bonds are separated by one saturated bond). These hydroperoxy radicals can now propagate the oxidation process as they are avid electron acceptors. After hydrogen abstraction, they will form more stable hydroperoxides (OOH). In non-enzymatic peroxidation systems the ratio of 9- to 13-hydroperoxide is almost one. Considering cis and trans stereoisomers, there will be 4 different combinations possible as there are two double bonds. The relative concentrations of final 32 products depend on many factors such as the temperature or presence of antioxidants. The generation of 11-hydroperoxy-LA (a non-conjugated diene) has been under debate for years, however, Brash et al. have shown that in the presence of 5% α-tocopherol, this isomer can account for up to 10% of initial products [436]. A schematic presentation of these steps is depicted in Figure 1.3. A hydroperoxide can attack a double bond of another fatty acid to generate an epoxide. Additionally, hydroperoxides are relatively labile. This can lead to formation of alkoxyl (RO·) and hydroxyl (·OH) radicals which can propagate the oxidation and also produce ketones, epoxide, or alcohols. Example of ketone and epoxide formation from hydroperoxide can be found in Figure 1.3. Hydroperoxides can also undergo fragmentation to produce aldehydes [437] or oligomerize with other OPs. AA autoxidation follows a similar pattern, however, since AA has three bis-allylic methylene groups, three different pentadienyl radicals can be formed. Therefore the number of products greatly increases. The primary products of AA peroxidation are the 5-, 8-, 9-, 11-, 12-, and 15-hydroperoxide isomers. LA and AA hydroperoxides can undergo a second hydrogen abstraction to form dihydroperoxides. When there are more than two unsaturated bonds such as in linolenic acid (18:3) or AA, the hydroperoxide isomers are not produced in equal amounts. For example, the “outer” hydroperoxides of arachidonic (5- and 12-) are far more abundant than “inner” (8-, 9-, 11-, and 12-) hydroperoxides. This is due to rapid 1,3 cyclization of the “inner” hydroperoxides [438, 439] to produce epidioxides. A 1,5 cyclization will produce a bicyclic endoperoxide which, in AA autoxidation, is the pathway for production of prostaglandin-like OPs [440] , the precursor structures for isoprostanes (isoPs) and isolevuglandins [441, 442]. Generation of isoPs is accomplished by rearrangement of the bicyclic endoperoxide precursor with an intramolecular attack by a carbon-centered radical on a double bond resulting in formation of a cyclopentane ring (Figure 1.4). They can be biologically active and some of them are useful markers of non-enzymatic lipid peroxidation in vivo [443]. The cyclization is inhibited by antioxidants, which result in equal concentrations of outer and inner hydroperoxides [439]. Levuglandins (LGs) are reactive dicarbanoyl ketoaldehydes that are generated during enzymatic oxidation of arachidonic acid by cleavage of C-C and O-O bond of the bicyclic peroxide nucleus of a bicyclic endoperoxides (e.g. prostaglandin H) [444]. Isolevuglandins (isoLGs) are generated via similar 33 rearrangements during non-enzymatic oxidation of AA (Figure 1.4 and [442]). As aldehydes, LGs and isoLGs can form adducts with amino groups such as lysine residues and covalently modify proteins [445] which is discussed in more detail in section 1.4.5. Recently Fessel et al. discovered a new class of AA OPs - called isofuranes (isoFs) [446]. They showed that, with increased oxygen tension, the carbon- centered radical is attacked by oxygen to form a hydroperoxide radical (instead of intramolecular attack by a double-bond to form isoPs). Subsequent rearrangements produce a tetrahydrofuran ring (hence the name isofurane). Yin et al. recently described another new class of arachidonic acid oxidation products termed dioxolane-isoprostanes [447]. These oxidation products have both a bicyclic endoperoxide and a cyclic peroxide functionality on the same molecule. LA and AA oxidation products can also undergo oligomerization. The simplest such reaction would be the reaction of two lipid oxyradical (e.g. LOO·) with each other, to yield a dimer radical. In biological systems lipid peroxidation often occurs in bilayer membranes or lipoproteins, and in these systems dimers are usually composed of fatty esters (containing hydroperoxy, hydroxyl, or keto functional groups) linked through their peroxide groups [448]. These dimers may undergo thermal cleavage (fragmentation) to produce volatile compounds (next paragraph). Fragmentation (or decomposition) is another mechanism for formation of secondary products from primary products of PUFA oxidation. Two major mechanisms explaining fragmentation products are: homolytic β-scission [449] and Hock cleavage [437]. In homolytic scission, both products are radicals (each retaining an unpaired electron) instead of ions (one fragment losing and the other gaining an electron). The results will be aldehyde-, alkyl- (if saturated), and/or alkene- (if unsaturated) radicals. Of course, these radicals seek stabilization by reacting with electron donors to produce alkanes or alkenes, or react with hydroxyl groups to make alcohols and aldehydes. Hock cleavage involves breaking of C-C bond with an insertion of oxygen in between which will in turn rearrange to form oxo or keto groups. Based on these mechanisms, 9 and 13 hydroperoxy LA fragmentation products include (but are not limited to) 2,4 decadienal, 3-nonenal, hexenal, 4-hydroxy nonenal (4-HNE), and pentane. It is important to remember that unsaturated secondary oxidation products can themselves undergo the same processes (dimerization or decomposition) to give more products. This leads to accumulation of 34 unsaturated aldehydes and dialdehydes (such as malondialdehyde or MDA) in advanced stages of oxidation. Figure 1.3 on next page illustrates some of the primary and secondary OPs of LA autoxidation. Figure 1.4 illustrates products of phospholipid oxidation (1-palmitoyl 2-arachidonyl phosphatidylcholine or PAPC in particular). 35 Figure 1.3 Autoxidation of linoleic acid (LA). In the initiation step, a hydrogen atom is abstracted from bis-allylic methylene group (carbon 11) which leads to formation of pentadienyl radical. Addition of oxygen will produce 9- or 13-hydroxy conjugated diene radicals which themselves are capable of propagating the oxidation chain reaction. In the presence of α-tochopherol, another product, 11 hydroperoxy LA, comprises about 10% of primary oxidation products. Boxed areas present two examples of monomeric unfragmented oxidation products of LA. Further oxidation and rearrangements (RA) can produce epoxy alcohol (right) or epoxy keto compounds (left). 36 1.4.2 Oxidation of cholesterol and cholesterol esters Cholesterol oxidation products (oxysterols) can be generated in vivo by enzymatic or non- enzymatic reactions. Different members of the cytochrome P450 family of oxidases can enzymatically oxidize (hydroxylate) carbons - in the cholesterol ring or in its side-chain. Mitochondrial CYP27A1 hydroxylases attack carbon 27 to produce 27-hydroxy-cholesterol (27OH-chol - formerly known as 26OH-chol). Other side-chain hydroxylases produce 24 and 25OH-chol [450] which carry important functions in cholesterol and bile acid homeostasis. For example, 27OH-chol formation is the first step in the alternative pathway of bile acid synthesis [451]. The classical bile acid pathway is regulated by CYP7A1 hydroxylase which inserts a hydroxyl in the 7α-position of the sterol ring, the first product in the classical pathway. CYP3A4 oxidizes cholesterol to form 4βOH-chol [452] . 24, 25-epoxy cholesterol (24,25 epox-chol), an activator of liver X receptor (LXR), is another important oxysterol formed enzymatically by a shunt pathway during cholesterol biosynthesis [453]. This oxysterol has recently received some attention as it seems, in macrophages, increasing this oxysterol can promote cholesterol efflux through LXRα-activated transcription [454, 455]. Cholesterol makes up to half of the lipids within cell membranes and under some conditions it may be more susceptible to oxidation than PUFAs [456]. Non-enzymatic cholesterol oxidation gives rise to four major products: hydroperoxy-, hydroxy-, keto, or epoxy- cholesterol. The bond dissociation energy between hydrogen and carbon 7 (in the B ring) is relatively low (88Kcal/M) [457] which explains why this site is frequently attacked during non-enzymatic cholesterol oxidation. After the initial hydrogen abstraction, addition of another oxygen atom produces 7α or 7β hydroperoxy radical (7α/7βOO·-chol). Similar to PUFA oxidation, this hydroperoxy radical by itself can propagate the oxidation process. By abstraction of a hydrogen atom, the radical is reduced to the corresponding hydroperoxide (7α/7βOOH-chol). Further hydrogen abstraction and loss of water will produce the stable product hydroxyl or keto cholesterol (7α/7βOH-chol and 7α/βK-chol). Another pathway for formation of keto-cholesterol is through Russell mechanism where two cholesterol hydroperoxy radicals form keto- and hydroxy-cholesterol and release one singlet oxygen (O2) [458]. The third pathway to keto-cholesterol involves enzymatic (dehydrogenase) or non-enzymatic loss of hydrogen from the hydroxyl group [459]. 37 The non-enzymatic reaction occurs via hydrogen abstraction by a hydroperoxy radical – a reaction that promotes termination of the chain reaction [460]. Another major product of cholesterol oxidation is 5,6 epoxy cholesterol - either α or β epimers - (5α,6α or 5β,6β epox-chol, respectively). A mechanism for cholesterol epoxide formation is hydroperoxy radical attack at the 5 and 6 carbon (the double bond of the B ring) with formation of a hydroperoxy intermediate. Dissociation of an alkoxyl radical thereafter leaves an epoxide behind [461]. Another pathway of epoxy cholesterol formation is unique to cholesterol esters such as cholesterol linoleate. Fatty acyl peroxidation (R-OOH formation) and intramolecular rearrangement leads to formation of epox-chol esterified to an alkoxyl (R-OH) group [462]. Exposure to non-radical ROS (such as O2, O3, and HOCl) can also oxidize cholesterol. The main products are 5OOH-, 6OOH-, or 7OOH-chol for O2 and products such as 5β,6β epox-chol for ozone (reviewed in [458]). HOCl oxidation of cholesterol can commonly happen during phagocytosis where local concentration of this ROS increases. Signature products (like 5,6 dichlorinated sterols) of such oxidation were described by Hazen et al. [463]. Of course, HOCl can produce other cholesterol oxidation products such as (α or β) 5,6epox-chol and 4OH-chol [464]. In plasma the top four oxysterols, in decreasing order of abundance, are: 27OH-, 24OH-, 7αOH-, and 5,6β epox-chol [465]. Side chain oxidation is not generally observed with non-enzymatic pathways indicating that most plasma oxysterols are enzymatically generated. Oxysterols found in copper oxidized LDL, in decreasing order of abundance, are: 7K-, 7OOH-, 7OH-, 5,6 epox-chol [466] which reflects the non-enzymatic (free-radical induced) nature of mechanisms involved. Other forms of oxidatively modified LDL more or less exhibit the same pattern [467, 468] but it seems that 7K-chol is a more abundant in the LDL that has been extensively oxidized by copper while 7OOH-chol is found more in enzymatic or metal-free oxidation systems [468]. Studies of animal and human atherosclerotic lesions have shown that the concentration of oxysterols is significantly higher within plaques compared to normal intima. For example one study showed increased levels of 7OH-, 7OOH-, 5,6 epox-, and 7K-chol [469]. In this study, 27OH-chol was also relatively abundant which hints both enzymatic and non- enzymatic processes could be relevant within atherosclerotic plaques. Other studies have shown that 38 27OH-chol is the main oxysterol in the plaques followed by 7K- and 7OH-chol [468]. The abundance of 27OH-chol in plaques could be due to the activity of mitochondrial CYP27A1 within macrophages. 1.4.3 Oxidation of phospholipids Glycerophospholipids (PLs) contain two fatty acyl groups esterified to a glycerol backbone with a polar head group (such as phosphocholine) at the sn-3 position. Oxidized PLs (oxPLs) are biologically active and can even serve as markers for cardiovascular diseases [470]. Commonly the fatty acyl esterified to the sn-2 position of glycerol backbone is AA or (even more frequently) LA. PLPC (1- palmitoyl, 2-linoloyl, phosphatidylcholine) and PAPC are in fact among the most abundant PLs (and sources of AA and LA) in vivo. As such, PL oxidation is more-or-less governed by the same mechanisms as in PUFA oxidation (described in section 1.4.1). PLs can undergo enzymatic (e.g. lipoxygenases) or non-enzymatic oxidation. PAPC, for example can generate primary or secondary OPs, some of which are shown in Figure 4. OxPLs might play a role in atherogenesis mainly due to their pro- inflammatory properties. They have shown to promote macrophage recruitment, induce binding of monocytes to ECs [471, 472], increase expression of inflammatory cytokines [473-475], increase thrombogenicity by increasing TF production [476] or by mimicking platelet activating factor (PAF) activity [477]. In addition, oxPLs act as PAMPs and these can be recognized by CD36 [112, 478]. 39 Figure 1.4 Autoxidation of phospholipids. Conjugated dienes are formed through oxidation of arachidonic acid on sn-2 position of 1-palmitoyl,2-arachidonyl, glycerophosphocholine (PAPC) (1). Further oxidation will generate a hydroperoxy (OO·) radicals. Through rearrangements (RA) it can generate prostaglandin-like compounds (3, 4). Further rearrangements can generate isoprostanes, isolevuglandins, or isothromboxanes (5). PEIPC can undergo dehydration to produce PECPC. On the other hand, hydroperoxy radical can propagate oxidation by hydrogen abstraction from another PUFA and convert into hydroperoxide (2). Arachidonic acid-derived hydroperoxides in PAPC can decompose to produce proinflammatory truncated oxPLs such as POVPC and PGPC and ligands of CD36 (HOdiPC for example) (7) or free reactive aldehydes such as HNE. (HOOA-PC: 5-hydroxy-8-oxo-6-octenoic acid esters of LPC, HOdiA-PC: 5-hydroxy-8-oxo-6-octenedioic acid esters of LPC. KOOA-PC: 5-keto-8-oxo- 6-octenoic acid esters of LPC, PEC-PC 1-hexadecanoyl-2-(5,6-epoxyisoprostane A2 oyl)-sn-glycero-3- phosphocholine, PEI-PC 1-hexadecanoyl-2-(5,6-epoxyisoprostane E2 oyl)-sn-glycero-3-phosphocholine, 40 POVPC 1-palmitoyl, 2- (9-oxononanoyl)-sn-glycero-3-phosphocholine, PGPC: 1-palmitoyl, 2-glutaroyl- sn-glycero-3-phosphocholine, HNE 4-hydroxy-trans-2-nonenal). Reprinted from Biochimica et Biophysica Acta, Vol(1772), Fruhwirth GO, Loidl A, Hermetter A, Oxidized phospholipids: from molecular properties to disease, 718-736, Copyright (2007), with permission from Elsevier [479]. 41 1.4.4 Studying lipid oxidation Simple methods of studying lipid peroxidation include colorimetric and spectrophotometric techniques. Conjugated dienes are produced during peroxidation of PUFA, and can conveniently be monitored by their UV absorption at 234 nm. 2-Thiobarbituric acid (TBA) can react with a few secondary products of PUFA oxidation such as malondialdehyde (MDA). These TBA-reactive substances (TBARS) bind to two TBA molecules and generate a pigment with red/pink color and maximum absorbance at 532-535 nm [480]. Lysine derivatization by PUFA oxidation products generates fluorescent products with a maximum excitation/emission at 350/430 nm. The fluorescence spectrum for MDA-modified proteins is slightly different (excitation/emission of 390/475) [481]. DNA modification by OPs of AA produces fluorophors with maximum excitation/emission of 315/420 nm [482]. The above methods are simple and convenient, but are semiquantitative at best and provide no structural information. A variety of chromatographic methods can be used to resolve OPs. Thin-layer chromatography (TLC) has been used to separate secondary products of LA oxidation. However the resolution of this method is usually inadequate to separate all products even in simple systems [483]. For that reason, TLC is mainly limited to qualitative studies of OPs. More powerful chromatographic methods include gas chromatography (GC) [484, 485] and high performance liquid chromatography (HPLC) which with chiral columns allows separation of cis and trans stereoisomers [486, 487]. GC is mostly used for volatile oxidation products such as aldehydes, ketones, and hydrocarbons. HPLC is useful in studying thermally unstable OPs such as hydropexides. High performance size exclusion columns have also been successfully used to resolve monomeric, dimeric, and oligomeric secondary products [488]. Mass spectrometry (MS) is a technique that has revolutionized the research in this field. MS can be coupled with HPLC or GC and used in tandem with another mass spectrometer (MSMS). In MSMS, the “parent” ion is fragmented and the daughter ions separated and further monitored to give more information regarding the structure of the parent ion. In the setting of GC-MS, since higher temperatures and volatile compounds are studied, the compounds in vapor state are bombarded with electron impact ionization (EI) to charge and fragment the compound. With HPLC-MS, however, there is the problem of 42 compounds being in the solvent phase and labile with harsher methods of GC-MS. Thermospray, electrospray ionization (ESI) [489], matrix-assisted laser desorption ionization (MALDI) [490], and atmospheric-pressure chemical ionization (APCI) [491] are examples of softer ionization techniques. With ESI and MALDI, there will be practically no fragmentation of compounds which allows for studying the non-volatile and thermally unstable OPs. A special variety of ESI, uses injection of a central complexing ion (such as silver ion) to generate coordinated charged ions. This technique, called coordinated ion-spray mass spectrometry (CIS-MS), greatly increases the sensitivity [492]. Harvilla et al. applied CIS-MS to the study of OPs and showed that Ag+ adducts of (hydro)peroxides, with a mass of M+107 or M+109, behave exactly the same way (in terms of fragmentation) as free OPs - with an added advantage of superior sensitivity [493]. Lipidomics has lagged behind proteomics at least in part because of the difficulties associated with analysis of this group of compounds. Complexities of reactions during oxidation, lack of standards, and instability of products have further complicated research in the field of lipid oxidation. The major technological breakthroughs have occurred mostly within the past 20 years and this field is still in its formative years. 1.4.5 Modification of proteins by fatty acid oxidation products A major mechanism by which oxidative stress exerts its deleterious effects is modification of macromolecules. For example, bifunctional aldehydes generated through decomposition of OPs such as 4-HNE and MDA are capable of modifying DNA or proteins. DNA modification by these OPs produces etheno-DNA (ε-DNA) which is potentially genotoxic. Excision repair will release modified bases that can be traced in the urine [494]. Modification of proteins by oxidized lipids is believed to create a fluorescent lipofuscin-like product [495]. Lipofuscin is a pigment granule that accumulates intracellularly. Its abundance increases with age and represents repeated cycles of oxidative damage. When accumulated in macrophages [496] or atherosclerotic lesions [497], some authorities refer to this pigment as “ceroid” which is principally made up of OPs covalently linked to proteins. The best studied OPs responsible for modification of 43 lysine residues are reactive aldehydes such as MDA. The proposed reaction mechanism for MDA involves Schiff-base formation by reaction of an aldehyde group with primary amino groups of proteins (Figure 1.5A). Decomposition of OPs can also create γ-hydroxy aldehydes (such as 4-HNE) which are more reactive than MDA (Figure 1.5B). After the initial reaction through their aldehyde group as described for MDA, the intra-molecular cyclization and dehydration generates a pyrrole ring (Figure 1.5B). Another mechanism for addition of such reactive aldehydes involves Michael-adduct formation. The aldehyde group can then form a Schiff-base with another lysine residue to cross-link the protein or interact with the hydroxyl group on the same adduct to make a hemi-acetal group (Figure 1.5B) [479, 498, 499]. IsoLGs, as γ-ketoaldehydes, are extremely reactive. In fact assaying them in biological systems them has proved to be challenging as they form Schiff-base adducts within seconds [500]. Subsequent reactions will generate pyrroles, hydroxyl lactams, and lactams as described above for γ- hydroxy aldehydes (Figure 1.6 and [442]). Except for isoLGs, all the OP-induced modifications described above require fragmentation of oxidation products to allow reactivity. Fruebis et al. have suggested that reactive aldehydes derived from fragmentation cannot account for the majority of fluorescence generated by modification of protein by OP [501]. They proposed a mechanism by which hydroperoxides modify proteins without the need for decomposition (Figure 1.7). To support this hypothesis, the same group developed a polyclonal antibody that recognized hydroperoxide-modified proteins but not native or aldehyde-modified proteins. Using this polyclonal antibody, they showed that such epitopes exist in vivo [502]. Modification of proteins by oxidized lipids is of interest as it can alter their function. As well, OP modification can be used as a marker of oxidative damage. For example, oxidatively modified LDL has been detected in atherosclerotic lesions, and oxidized LDL has been found to have numerous biologic properties that could accelerate atherogenesis. Haberland et al. showed that modification of 15% of lysine residues within apoB by MDA renders it a ligand for scavenger receptors. Interestingly, to create SR ligands, non-oxidative modifications (acetylation or succinylation) had to modify at least 60% of lysines [503, 504]. Steinbrecher showed that extensive oxidation of LDL with copper ion modifies nearly 30% of apoB lysine residues and generates the typical fluorescence pattern discussed for OP- 44 modified proteins [505]. This type of oxidation is not limited to protein modification but also results in extensive oxidation of LDL lipids and hydrolysis of PC to LPC [505]. To determine if a given effect of oxLDL was due to protein modification by OPs, our lab developed a simple model for LDL modification in which OPs of AA or LA are used to modify LDL under conditions where oxidation of LDL lipids itself is inhibited [506]. This oxidation product-modified LDL (OP-LDL) was shown to share many biological properties of copper-oxidized LDL. For example OP-LDL is taken up avidly by macrophages [274] and is able to compete with oxLDL for uptake even in SR-A knockout mice [268]. OP-LDL also induces macrophage growth [307]. It has been shown that autoantibodies against epitopes created by OP- protein modification exists in apoE knockout mice which supports the notion that such modifications occur in vivo [87]. Immunohistochemical studies have also shown the existence of OP-modified epitopes in hypercholesterolemic rats [507]. However the structure of these eiptopes in OP modified LDL has not been elucidated. A different group later developed another metal-ion independent, peroxidation- dependent method to modify LDL [508]. One importance of such models is that they provide a more physiological alternative to copper-oxidized LDL. 45 Figure 1.5 Protein modification by reactive products of PUFA oxidation. A) Aldehydes can form Schiff bases by reacting with epsilon amine groups of lysine residues on proteins. Schiff bases are potentially reversible, but bifunctional aldehydes such as malondialdehyde (MDA) can form stable cross- link proteins by this mechanism. B) γ-hydroxy alkenals are significantly more reactive than MDA. Similar to A, they can form a Schiff base with primary amines of lysine residues. However they can then form a pyrrole ring through reaction of amide with hydroxyl group. Alternatively they can react, through Michael addition, with amino acids containing primary amines (R‟-NH2) or thiol (R‟SH) groups. Dehydration of the adduct can then form a hemi-acetal ring. Reprinted from Biochimica et Biophysica Acta, Vol(1772), Fruhwirth GO, Loidl A, Hermetter A, Oxidized phospholipids: from molecular properties to disease, 718-736, Copyright (2007), with permission from Elsevier [479]. 46 Figure 1.6 Protein modification by levuglandins and isolevuglandins Levuglandins (LG) and their non-enzymatically-generated counterparts, isolevuglandins (isoLGs), are extremely reactive. Within seconds of generation they form Schiff base adducts with lysine residues of proteins which later form nitrogen-containing heterocycles such as pyrrole by cyclization and loss of water. Continued oxidative processes will lead to generation of hydroxylactams and lactams. Reprinted from Molecular Nutrition & Food Research, Nov 2005; Volume (49), Pages:1050-62, Zhang W, Salomon RG, Oxidized phospholipids, isolevuglandins, and atherosclerosis, Copyright Wiley-VCH Verlag GmbH & Co. KGaA. Reproduced with permission [509]. 47 Figure 1.7 Concerted reaction between linoleic acid hydroperoxides and lysine. As proposed by Fruebis et al., unfragmented hydroperoxides may modify the primary amine of lysine residues. This involves the nucleophilic displacement of hydroperoxy group onto an unsaturated bond and consequent formation of an epoxy group. The secondary amine (imino group) can then react with either carbon of the epoxy ring to generate nitrogen containing 5- or 6-membered heterocyclic adducts ((1) and (2), respectively). Adapted from Figure 3B in Fruebis J, Parthasarathy S, Steinberg D, Evidence for a concerted reaction between lipid hydroperoxides and polypeptides Proceedings of the National Academy of Sciences of the United States of America, 1992; Volume (89), Pages: 10588-10592, [501]. Used with permission. 48 1.5 Objectives Previous studies reported that oxLDL can induce VEGF secretion in macrophages. However, there is neither agreement nor complete understanding of the signaling mechanisms underlying this effect. As well, there is very little information about the possible requirement for plasma membrane receptors for oxLDL to induce VEGF. The objectives of studies in chapter 3 were to shed light on the signaling pathways and determine the possible contribution of major scavenger receptors in oxLDL- mediated VEGF secretion. Several groups have also reported that oxLDL promotes survival and growth in macrophages. However, very little information has been published regarding the involvement of scavenger receptors in mediating growth and survival. The studies in chapter 4 were designed to test if an endocytic or signaling pattern recognition receptor(s) might be required for oxLDL-mediated macrophage survival. The 5 th chapter of this dissertation is a study to try to define the molecular structure that was required for growth and survival signaling in macrophages. To make this analytically feasible, we chose to characterize our model of modified LDL generated by oxidation products of polyunsaturated fatty acids. 49 2. Materials and methods 2.1 Materials Phenazine methosulfate (PMS), arachidonic acid, linoleic acid, L-lysine-methyl ester dihydrochloride, phosphatidyl-L-serine, 1-stearoyl lysophosphatidylcholine, acetic anhydride, butylated hydroxytoluene (BHT), cholesterol, pyrrole, sphingomyelinase (SMase), myeloperoxidase (MPO), catalase, octyl glucoside, 2,3-bis[2-methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxanilide (XTT), N-methyldibenzopyrazine methyl sulfate, and trinitrobenzenesulfonic acid were obtained from Sigma-Aldrich (Mississauga, ON, Canada). Egg phosphatidylcholine (PC) and 1,2-dipalmitoyl-sn- glycero-3-phosphoethanolamine (DPPE) were obtained from Northern Lipids (Vancouver, BC, Canada). Cayman Chemical (Ann Arbor, MI) supplied 4-hydroxyhexenal and 4-hydroxy nonenal. Ceramide-1- phosphate was from Avaniti Polar lipids (Alabaster, AL). Fetal bovine serum (FBS), DMEM and RPMI 1640 medium, sodium pyruvate and L-glutamine were from Gibco Invitrogen. Fisher Scientific (Vancouver, British Columbia, Canada) supplied 3-(4,5-dimethylthiazol-2-yl)-5-(3- carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS), glucose, L-tyrosine, sodium borohydride and sodium hypochlorite. Glucose oxidase was from Roche Diagnostics (Laval, PQ). Centriplus 20 ultrafilters were obtained from Amicon (Beverly, MA). Antibodies to PKB, Erk2, phospho-PKB (Ser-473), and phospho-Erk1/2 (Thr-202/Tyr-204) were purchased from Cell Signaling Technologies. Protein G Sepharose beads and anti-PKCδ (C-20) and isotope control antibodies were obtained from Santa Cruz (Santa Cruz, CA). Nitrocellulose membranes, protein standards, and Bradford protein assay kits were purchased from Bio-Rad (Hercules, CA). Ficoll-paque and reagents for enhanced chemiluminescence were from Amersham Biosciences. BCA protein assay reagents were from Pierce (Milwaukee, WI). Recombinant human M-CSF and mouse and human VEGF ELISA kits were purchased from R&D Systems (Minneapolis, MN). The fluorescent dye 1,1'-dioctadecyl-3,3,3',3'- tetramethylindocarbocyanine perchlorate (DiI) was from Molecular Probes. [γ-32P] labeled ATP was purchased from PerkinElmer (Waltham, MA). Propidium iodide, RNase A, 1,1'-dioctadecyl-3,3,3',3'- tetramethylindocarbocyanine perchlorate (DiI) were obtained from Invitrogen (Burlington, ON, Canada). Titan gel lipoprotein kit was from Helena Laboratories (distributed by Inter Medico, Markham, Ontario). 50 Defined fetal bovine serum (FBS) was from HyClone (Logan, UT). All other reagents including inhibitors were Calbiochem products (San Diego, CA). 2.2 Animals CD-1 mice were from the UBC Animal Care Unit. Wild-type C57/bl6, TLR4, CD14, and FCγRIIb knockout mice were all purchased from The Jackson Laboratory (Bar Harbor, MA). CD36/SR- A double knockout mice were generously provided by Dr. M. W. Freeman [269]. LOX-1 -/- mice were generated in the laboratory of Dr. T. Sawamura, and were kindly sent to us by Dr. Osamu Cynshi, Chugai Pharmaceutical Corp, Tokyo. Genotypes of offspring of knockout mice were verified by PCR. All animal procedures were in accordance with the guidelines of the Canadian Council on Animal Care and were approved by the Animal Care Committee of the University of British Columbia. 2.3 Cell Culture RAW 264.7 cells were purchased from American Type Culture Collection (ATCC, Manassas, VA) and maintained in DMEM supplemented with FBS 10%, L-glutamine and sodium pyruvate (1 mM final concentration). Cells were harvested using a Teflon cell lifter. For experiments, FBS concentration was 1% (v/v). Resident mouse peritoneal macrophages (MPMs) were isolated by peritoneal lavage with PBS [510]. 500,000 cells were plated in each well of a 12-well plate in 1 mL RPMI, 10% FBS, L-glutamine, sodium pyruvate. For human monocyte-derived macrophages (hMDMs), peripheral blood mononuclear cells were isolated by Ficoll-paque centrifugation. Monocytes were isolated from mononuclear cells by adherence and were differentiated to macrophages in the presence of M-CSF for 7 days, changing the media every 48 h. All experiments using human macrophages started at the 7 th day post isolation. Bone marrow derived macrophages (BMDMs) were isolated from femurs of 6-8 weeks old wild- type or knockout animals as described [310]. Cells were plated in 10-cm diameter tissue culture plates and incubated overnight in DMEM containing L-glutamine, sodium pyruvate, 10% FBS, and 10% L-cell conditioned media as a source of macrophage colony stimulating factor (M-CSF). Non-adherent cells 51 (monocytes) were then collected, and differentiated to macrophages by a further 5-day incubation in the same medium. Macrophages were scraped off the plate using a Teflon cell lifter, counted and re-plated at specified densities and incubated overnight in the same medium. In all experiments using BMDMs, cell treatment started at the 7 th day post isolation. 2.4 Lipoprotein isolation, modification, and labeling LDL (d = 1.019 to 1.063 g/mL) was isolated by sequential ultracentrifugation of EDTA- anticoagulated fasting plasma obtained from healthy normolipidemic volunteers as described [310]. Prior to oxidation, LDL was dialyzed to reduce EDTA concentration to 10 µM. Different degrees of oxidation were achieved by incubating 200 µg/mL LDL for 2, 5, or 24 h at 37 °C with 5 μM copper sulfate. Oxidation was stopped by addition of 40 µM butylated hydroxytoluene (BHT) and 300 µM EDTA [310]. The modified LDL was then washed and concentrated to about 1 mg/mL using Amicon Centriplus 20 ultrafilters. Ferrous-ion-modified LDL (FeLDL) was prepared by dialyzing 200 µg/mL LDL against PBS containing 1 µM FeSO4 until LDL-associated carotenoids were consumed as judged by color change [511]. Similar to copper-oxidized LDL, FeLDL was washed and concentrated using ultrafilters. Acetylation of LDL was performed by sequential addition of aliquots of acetic anhydride as described by Goldstein, Brown, and colleagues [512]. Hydroxyl alkenal-modified LDL was prepared by addition of 4- hydroxyhexenal (0.5 mg) or 4-hydroxynonenal (1.5 mg) to 1 mg of LDL dissolved in PBS containing 10 µM EDTA. These modified LDL preparations were extensively dialyzed to remove unbound reactive material. For modification of LDL by fatty acid oxidation-products, 10 mg neat arachidonic acid or linoleic acid was autoxidized in a glass test tube by exposure to air at 37ºC for 72 h and the water-soluble component was incubated overnight at room temperature with LDL in the presence of 300 µM EDTA and 50 µM BHT [506]. p-hydroxyphenylacetaldehyde (p-HA)- modified LDL was generated by addition of 1 mL of 2 mM pHA to 1 mg of LDL and incubated for 24 h at 37°C. As a source of pHA, NaOCl (0.9 M) was 52 sequentially added to L-tyrosine (2 mM) in ice-cold phosphate (20 mM) buffer containing 100 µM DTPA (pH 7.0) until a final 1:1 molar ratio was achieved. The solution was then warmed for 1h at 37°C and filter sterilized. Efficient conversion of L-tyrosine to pHA occurs under these conditions [513]. Fluorescence labeling of LDL and oxLDL was done by addition of 75 µL of 3 mg/mL DiI solution to 4 mg (protein) of native or oxidized LDL in 8 mL of lipoprotein-deficient serum ( d>1.21 g/mL plasma fraction). After incubation for 8 h at 37ºC, labeled lipoproteins were reisolated by ultracentrifugation. DiI was extracted from lipoproteins with isopropanol and fluorescence was measured using a microplate fluorometer (Fluoroskan Ascent FL, Thermo LabSystems) with excitation/emission wavelengths set at 530/590 nm. Within the range of 0 to 1000 ng, a linear correlation existed between DiI concentration and fluorescence intensity with a lower detection limit of 5 ng diI. In preparations used in these experiments, 15-18 and 9-12 ng of DiI was incorporated into each μg (protein) of nLDL and oxLDL, respectively. 2.5 Lipoprotein characterization and analytical procedures Purity of LDL isolation and extent of modification was confirmed by agarose gel electrophoresis using a Titan gel lipoprotein kit. Lipoprotein bands were visualized by Fat Red staining. Electrophoretic migration was determined by measuring the distance between the point of origin and the leading edge of lipoprotein. apoB modification was quantified and expressed as the migration of the modified LDL relative to that of native LDL. Lipoprotein concentration was calculated based on the amount of protein measured by bicinchoninic acid (BCA) assay and expressed as milligrams per milliliter. Due to reactivity of OP products with BCA reagent, the Lowry protein assay [514] was used for protein measurements in OP-, 4- HHE-, 4-HNE-, and pHA-modified LDLs. 2.6 Modification of lysine-methyl-ester by PUFA oxidation products 3 mg of oxidation products of AA or LA (as described in section 2.4) dissolved in 300 μL of PBS were added to 200 μL of lysine-methyl-ester or LME (from a freshly prepared aqueous solution of 53 10 mg/mL, pH 8.0) and total volume was adjusted to 1 mL with PBS. The mixture was incubated at room temperature overnight in airtight glass tubes. 2.7 Lipid extraction, liposome preparation and modification Lipid extraction from lipoproteins was carried out according to the Bligh and Dyer method [515] with Hajar‟s modification [516] to increase the yield. One mg of native or oxidized LDL in 0.5 ml PBS containing 10 mM HCl were mixed with ice-cold methanol:chloroform (2:1) and vortexed. Then a solution containing 2 M KCl, 0.2 M H3PO4 (in water) mixed 1:1 with chloroform was added and vortexed again. Centrifugation at 2000 rpm for 10 minutes yields an upper aqueous layer and a lower organic (lipid) layer with denatured protein at the interface. For liposome preparation, the organic phase from the lipid extraction was dried under nitrogen and the lipids were resuspended by vortexing in 1.5 mL of liposome buffer (150 mM NaCl containing 0.1 mM EDTA and 10 mM HEPES, pH 7.5). The mixture was passed 10 times through a 0.1 µM polycarbonate membrane at 37°C under nitrogen flow using a Lipex mini-extruder. Concentrations of liposomes are expressed according to the initial amount of LDL protein used for extraction. Phospholipid liposomes were prepared by mixing 10µM of DPPE (for PE liposomes) or egg PC (for PC liposomes) with equimolar cholesterol and vortexing in 2 mL of liposome buffer and extruded as described above. Liposome concentrations are expressed in terms of initial concentrations of phospholipid assuming 95% recovery after extrusion. The size of liposomes was determined by a submicron particle sizer (Nicomp Model 270). The mean diameter of PC was 136±20 nm, and that of PE liposomes was 126±53 nm. Liposomes were modified by PUFA oxidation products as follows: OPs of AA or LA (described in section 2.4) were resuspended in 1 mL of PBS and incubated with liposomes (2 mg total lipid) for 8 h at room temperature. 2.8 apoB solubilization After the extraction as described in previous section, the middle (protein) layer was washed with 2 mL of ice-cold H2O and 2 ml of acetone. After a final wash with water the apoB was solubilized in 0.5 54 mL of a 15 mg/mL aqueous solution of octyl glucoside containing 10 mM NaOH for 30 minutes at room temperature. Excess detergent was then removed by dialysis against PBS. 2.9 Cell viability assay Macrophage cell number in MPMs was estimated by the XTT formazan method, which measures the rate of reduction of the XTT dye by mitochondria. We have previously documented a close correlation between macrophage number and formazan dye reduction [308]. Peritoneal macrophages were isolated as described in 2.3, and plated at 1*10 4 cells/well with 100 µL RPMI 1640 containing 10% FBS. After overnight incubation, each well was supplemented with 50 µL of RPMI medium containing 25µM PMS and 1 mg/mL XTT. After 4 h of incubation in at 37°C, absorbance was measured at 450 nm. For later experiments involving survival in BMDMs, MTS was substituted for XTT as it permits a 1-step assay procedure [310]. BMDMs from wild-type or knockout mice were harvested and expanded in culture for 5 days as described in 2.3. Cells were harvested with a Teflon scraper, counted, and each well was seeded with 3*10 4 cells in 100 µL RPMI 1640 containing 10% FBS and 10% L-cell conditioned media. Cells were incubated overnight to allow them to adhere. At the start of each experiment, wells were washed twice with PBS and cells incubated in fresh media with or without M- CSF and indicated concentrations of native or modified lipoproteins. At the end of the experiment, 50 µL RPMI 1640 containing 333 μg/mL MTS and 25 ng/mL PMS was added to each well. After 2 h incubation at 37ºC, absorbance at 490 nm was measured. Where applicable, inhibitors were added in fresh media, 45 minutes prior to addition of native of modified LDL. All cell viability experiments were done at least in triplicate. 2.10 Apoptosis assay Propidium iodide (PI) staining and FACS analysis was used to quantitate the sub-diploid population. At the end of each time point, BMDMs from wild-type or knockout mice were permeabilized using ethanol 70% (v/v) for 1 h at 4ºC and washed twice in PBS containing 0.01% glucose. Cells were then resuspended in the same buffer plus RNase A (final concentration 0.1 mg/mL) and PI (0.12 55 mg/mL). Fluorescence was measured with a BD FACS Canto. The data were analyzed with FCS Express Pro Software Version 3 (De Novo Software, Thornhill, Canada). 2.11 ELISA Conditioned medium was collected and centrifuged at 14,000 rpm for 10 min. The VEGF concentration in the supernatant was determined using the Quantikine human and mouse VEGF ELISA kits (R&D Systems) according to the manufacturer‟s instructions. The result was normalized to total cell protein content. Where applicable, inhibitors were added 45 min before adding the native or modified LDL. 2.12 Immunoblotting Cells were lysed by ice-cold solubilization buffer (containing 50 mM Tris-HCl PH 7.7, 1% Triton X-100, 10% glycerol, 100 mM NaCl, 2.5 nM EDTA, 10 mM Na plus freshly added sodium orthovanadate (0.2 mM), sodium molybdate (1 mM), and protease inhibitor cocktail containing aprotinin (10 µg/mL) and leupeptin (10 µg/mL). Thereafter, lysates were centrifuged at 14,000 rpm for 10 min and protein content of the supernatant was determined by BCA assay. Sample concentrations were adjusted to 1 mg/mL and sample buffer was then added to each lysate and heated for 5 minutes at 90 ºC. Equal amounts of protein were loaded onto an SDS-PAGE separating gel (10%) with low molecular weight protein ladder (Biorad) for calibration. Protein was then transferred to nitrocellulose paper, blocked for 1 h in Tris-buffered saline (TBS) containing 5% skim milk with 0.1% Tween 20, and then incubated overnight with the primary antibody anti-phospho PKB or Erk2 in TBS and 0.1% Tween 20, membranes were incubated with horseradish peroxidase-conjugated secondary antibody at 1:1000 dilution for 1 h. Bands were visualized using enhanced chemiluminescence. As a loading control, phospho-antibodies were stripped with Tris buffer containing SDS (2%) and β-2-mercaptoethanol (100 mM) at 50°C for 20 minutes, washed, and reprobed with antibody to unphosphorylated PKB or Erk2. 56 2.13 PKCζ immunoprecipitation and in vitro kinase assay Cell lysates were prepared as for western blots. For each condition, equal amounts (500 µg/mL) of protein were incubated overnight with 2 µg of anti-PKCδ (C-20) or isotope control antibodies at 4ºC. Antigen/antibody complexes were pulled down using protein G Sepharose beads and washed twice in lysis buffer and then twice in kinase buffer (described below). Kinase buffer composition was β-glycerophosphate (25 mM), MOPS (20 mM), EGTA (5 mM), EDTA (2 mM), and MgCl2 (20 mM), sodium orthovanadate, and sodium molybdate. Each kinase reaction also contained bead/protein complex, myelin basic protein (5 µg), phosphatidylserine (4 µg) and [γ-32P] ATP (0.166µCi). The reaction was carried out at 37ºC for 20 minutes, and was stopped by addition of hydrochloric acid. The mixture was then spotted on P81 filter papers and washed with o- phosphoric acid. Radioactivity on filters was measured using a liquid scintillation analyzer. The specificity of the reaction for PKCδ was verified using an in vitro kinase inhibitor (PKCδ pseudosubstrate) according to the manufacturer‟s instructions. This inhibitor completely blocked the oxLDL-induced increase in kinase activity. 2.14 Lipoprotein uptake assay Uptake of DiI-labeled LDL was compared in BMDMs from wild type, LOX-1, and CD36/SR-A double knockout (DKO). BMDMs were seeded at 10 6 cells/well of 6-well plates and incubated with 25 µg/mL of either unlabeled lipoprotein (autofluorescence control) or DiI labeled native LDL or oxLDL (DiI-LDL and DiI-oxLDL, respectively). After 24 hours, cells were washed with PBS and harvested using a Teflon cell scraper. Equal aliquots were taken for protein measurements. The remaining cells were centrifuged and the pellet was dissolved in isopropanol. DiI fluorescence of each condition was measured (excitation: 530 nm, emission: 590 nm ) in triplicates with unstained control as background. A serial dilution of DiI was used to create a standard curve. Results were normalized to protein content and expressed as percentage of DiI-oxLDL uptake in wild-type macrophages. In MPMs, uptake of fluorescently labeled lipoproteins was compared in macrophages from wild- type and CD36/SR-A double knockout (DKO) mice using flow cytometry. MPMs from WT or DKO 57 mice were incubated with 25 µg/mL of DiI-LDL, DiI-oxLDL, or unlabeled lipoprotein (unstained control). After 24 hours cells were washed twice with PBS and harvested using Teflon cell-scraper and then resuspended in PBS containing 1% BSA. Up to 20,000 events were acquired on a BD FACS Canto. The data were analyzed with FCS Express Pro Software Version 3 (De Novo Software, Thornhill, Canada). DiI uptake for each condition was calculated by determining the geometric mean of the corresponding fluorescence profile after correction for fluorescence in the unstained control (expressed as Δ mean fluorescent intensity or ΔMFI). 2.15 Quantitative reverse transcription polymerase chain reaction RNA was isolated from macrophages using an RNeasy mini kit (Qiagen, Mississauga, ON, Canada). Nucleic acid concentration and purity were tested by measuring absorbance at 260 and 280 nm, respectively with an Ultrospec 3000 spectrophotometer (LKB Pharmacia Biotech). Total RNA was utilized as a template for first strand cDNA synthesis using Supertranciptase II and OligodT primers (Invitrogen, Burlington, ON, Canada) according to the manufacturer‟s instructions. The resulting cDNA was amplified using the VEGF forward primer 5'- AGTCCCATGAAGTGATCAAGTTCA-3‟ and the reverse primer 5‟- ATCCGCATGATCTGCATGG- 3‟. β-actin gene expression was determined as a reference with forward primer 5‟- AGGCCCAGAGCAAGAGAGG-3‟ and reverse primer 5‟-TACATGGCTGGGGTGTTGAA-3‟. These sequences were obtained from previously published work [517]. qPCR was performed with an ABI Prism 7300 Sequence Detector (Applied Biosystems) using a QuantiTect SYBR Green PCR Kit (Qiagen, Mississauga, ON). Amplification was performed according to the ABI 7300 standard protocol with data collection at stage 3, step 2 (60ºC 1 min). All assays were performed in triplicate and data were expressed as fold increase relative to no-treatment control. 2.16 Hydroperoxide measurement Hydoperoxide content was measured by a lipid hydroperoxide assay kit (Cayman Chemical Co., Ann Arbor, MI) according to manufacturer‟s protocol. This assay measures the hydroperoxide content 58 directly by redox reactions relying on hydroperoxide-dependent ferric ion generation which then reacts with a chromogen giving color with maximum absorbance at 500 nm. Background was determined by measuring the absorbance after treatment with triphenylphosphine multiplied by 1.28 to correct for the effect of triphenylphosphine on the chromogen. 2.17 Mass spectrometry and tandem mass spectrometry In early experiments, unit mass and MSMS data were obtained on an Esquire Ion Trap instrument (Bruker Daltonics, Billirica, USA). A Micromass LCT time of flight (TOF) system (Micromass, Manchester, UK) was used to perform accurate mass measurements. Samples were dissolved in methanol at a concentration of 1 mg/mL and injected into the electrospray ionization source at a flow rate of 10 μL/min. Mass spectrometers used for HPLC-MS and HPLC-MSMS are described below. 2.18 High performance liquid chromatography coupled mass spectrometry and tandem mass spectrometry The chromatographic system consisted of a Waters ACQUITY UPLC coupled with a photodiode array (PDA) detector and a ZQ single quadrupole or a Waters/Micromass Quattro Micro triple quadrupole mass spectrometer (Waters Corp., Milford, MA). Oxidation products or adducts were resolved on a BEH C18 column (1.7 μm, 50 mm x 2.1 mm; Waters, Milford, MA) equilibrated with 20:80 acetonitrile/water. In early experiments, the acetonitrile gradient was ramped to 100% from 0.2 – 10 min at 0.8 ml/min with ~ 1/3 split to the MS and column temperature was 35 ºC. 50 mM ammonium acetate was used as the aqueous modifier. MS data were acquired in both ES+ and ES- mode. In ES+ capillary was at 3 kV, cone voltage 30 V, source and desolvation temp were 100 & 250 ºC, respectively and desolvation gas flow 800-1000 L/h. In ES- capillary was 2.5 kV and remaining parameters the same as for ES+. OP products and especially OP-LME adducts were more efficiently ionized in ES+ mode. 59 High resolution mass measurements and MSMS data were also acquired using a quadrupole time-of-flight mass spectrometer (SYNAPT TM High Definition Mass Spectrometry TM system; Waters, Milford, MA, USA). This system was coupled to an ACQUITY ultra-performance liquid chromatography (UPLC) system similar to the chromatographic set up mentioned above. The following acetonitrile gradient was used: 0-0.2 min, 10-10%; 0.2-2.0 min, 10-15%; 2-25 min, 15-40%; 25-31 min, 40-90% at 0.3 mL/min. 20 mM ammonium acetate was used as the buffer in the aqueous phase. Inject volume was 10 µL and total run time was 35 min, including the 100% acetonitrile flush and re- equilibration. ES+ data were collected using ionization conditions similar those noted for quadrupole analyses with the instrument operating in V mode, tuned to approximately 10000 resolution and using leucine enkephalin for lock mass correction. m/z 50-1000 calibration was carried out by infusion of a sodium formate solution with root mean square deviations < 1 parts per million (ppm) for the ion clusters. Scan data were collected using 0.5 sec scans from m/z 100-600 using collision energies of 6 and 4 V, respectively in the instruments 2 collision cells. MSMS data for selected ions were collected using 1 sec scans from m/z 50-600, with the quadrupole at unit resolution and using ramps of 10-30V or 15-30 V in collision cell 1 and either 6, 12 or 16 V in collision cell 2 to yield varying degrees of fragmentation. For fluorescence detection of adducts, a Waters Alliance LC coupled with a PDA and 2475 multi wavelength Fluorescence Detector System was used (with excitation/emission set at 350/430 nm). For these measurements, oxidation products were resolved on a 2.1x150 Xterra MS C18 5 µm column with a 20 mM ammonium acetate/acetonitrile gradient as follows: 0.2-2.0, 10-15%; 2-25, 15-40%; 25- 31, 40-90%, 34.25-34.5, 90-100%, 36.25-36.5 100-10%. Total runtime was 40 minutes. Column temperature was 35ºC and flow rate was 0.3 mL/min. 2.19 Statistical analysis Unless stated otherwise, all results are expressed as means ± SD of pooled data from 3 to 5 experiments as specified in figures. Comparison of two means was done using a 2-tailed Student‟s t test. For comparing more than two means one-way ANOVA was used followed by Tukey‟s post hoc analysis to 60 measure the level of statistical significance between groups. In Figure 3.14, where the mean of control is compared to other conditions, Dunnett‟s post hoc analysis was carried out after ANOVA. The level of significance of difference is indicated in the graphs as follows: * p ≤ 0.05, ** p ≤ 0.01, and *** p ≤ 0.001. ANOVA and post hoc analyses were carried out with GraphPad Prism 4 software (La Jolla, CA). 61 3. OxLDL-mediated VEGF secretion in macrophages 3.1 Introduction/rationale As discussed in section 1.3.3, VEGF is thought to play an important role in atherosclerosis. Increased expression of VEGF has been documented in atherosclerotic plaques of humans and animals and correlates with the extent of disease progression [416-419]. Macrophages seem to be an important source of VEGF in atherosclerotic plaques [416]. OxLDL has been shown to increase VEGF expression in macrophages [418, 518, 519] however, the cellular mechanisms by which oxLDL induces VEGF secretion are not well understood. In particular, it is unclear how the type and extent of oxidation of LDL influences its induction of VEGF, or if macrophage scavenger receptors are required. Recently, LOX-1 has been implicated as a receptor for oxLDL in endothelial cells [520] and in bovine articular chondrocytes [521], and it was suggested in the latter paper that oxLDL acts through LOX-1 to induce VEGF expression in chondrocytes. However, no reports to date have addressed whether either of the two main receptors involved in oxLDL uptake in macrophages (SR-A and CD36) are required for VEGF upregulation by oxLDL in these cells. As well, only limited information has been published about the mechanism by which oxLDL stimulates VEGF secretion. Inoue et al. showed a potential role for peroxisome proliferator-activated receptor γ (PPARγ) [518], and Salomonsson et al. reported that oxLDL increased VEGF mRNA stability, partly via p38 stress activated MAPK [519]. However, many factors that increase VEGF act through receptor tyrosine kinases (RTKs) which then signal via two major downstream signal transduction pathways, PI3K and Ras-dependent MAPK pathways (section 1.3.2) but these pathways have not been examined with regard to VEGF induction by oxLDL. The objectives of the studies in this chapter were to determine how different types and degrees of LDL modification affect VEGF induction, to determine if SR-A, CD36 or LOX-1 were required, and to define the signal transduction pathways involved in enhancement of VEGF expression by oxLDL. 62 3.2 Results 3.2.1 OxLDL induces VEGF expression and secretion in macrophages To confirm the previous data, we measured the expression and secretion of VEGF in RAW264.7 with qPCR and ELISA, respectively - as described in the methods section. Figure 3.1A shows that extensively oxidized LDL, but not native LDL, increased VEGF secretion in RAW 264.7 cells in a time- dependent manner. This increase reached statistical significance as early as 12 hours. Figure 3.1B shows that the increase in VEGF secretion by oxLDL in RAW 264.7 cells was concentration-dependent. Figures 3.1C shows that the increased VEGF secretion is at least in part achieved by increasing mRNA levels. The increase in VEGF mRNA became statistically significant after 6 hours of oxLDL treatment. To verify that oxLDL can also increase VEGF expression in primary macrophages, we measured VEGF secretion in response to oxLDL in mouse peritoneal macrophages and human monocyte-derived macrophages. We found that oxLDL caused a dose-dependent increase in VEGF secretion in both hMDMs (Figure 3.2A) and in MPMs (Figure 3.2B). Compared to RAW264.7 and MPM, hMDMs showed slower kinetics with regards to oxLDL-mediated VEGF secretion. 3.2.2 Type and degree of modification influences oxLDL-mediated VEGF secretion To determine the effect of oxidative modification of LDL on VEGF secretion, cells were treated with LDL oxidized to various degrees. The mildest degree of oxidation, which resulted mainly in lipid peroxidation, was with ferrous ion (FeLDL). More extensive oxidation was obtained by exposing LDL to Cu++ for 2, 5, and 24 h to produce a range from mildly to heavily oxidized LDL (oxLDL2, oxLDL5, oxLDL24, respectively). Degree of oxidation was monitored by agarose gel electrophoresis (Figure 3.3B). FeLDL had the lowest mobility of the modified LDLs, and there was a progressive increase in migration with time of exposure to copper. For oxLDL, the electrophoretic mobility correlated with the amount of VEGF secretion (Figure 3.3A). FeLDL used in our experiments had less apoB100 modification than oxLDL2 as judged by electrophoretic mobility (Figure 3.3B), but the difference in VEGF secretion between cells incubated with FeLDL and oxLDL2 did not reach statistical significance 63 (p = 0.14). Figure 3.3A also shows that FeLDL was less potent than oxLDL5 (p = 0.004) or oxLDL24 (p < 0.001) at increasing VEGF secretion. To determine if selective modification of apoB100 by lipid peroxidation products without oxidation of LDL lipids has an effect, we tested the effect of LDL modified by fatty acid oxidation products (OP-LDL). Figure 3.4A shows that OP-LDL preparations with comparable electrophoretic mobilities to that of oxLDL24 significantly increased VEGF secretion compared to nLDL, but they appeared less potent than oxLDL24 (Figure 3.3A p = 0.003). Similar results were observed in MPMs and hMDMs using different preparations of OP-LDL (Fig. 3.4C &D). As a control we also used Ac-LDL which is a non-oxidative chemical modification of apoB100 that renders it a ligand for scavenger receptor A. Although Ac-LDL had an electrophoretic mobility similar to that of oxLDL24, suggesting that there was a similar degree of modification of lysine residues, it had no significant effect on VEGF secretion in any of the 3 cell types tested (Fig. 3.4A, C and D). This indicates that an oxidative modification, and not merely a charge change of apoB sufficient to confer scavenger receptor recognition, is required to induce VEGF. 3.2.3 OxLDL and ceramide phosphate both increase macrophage secretion through different mechanisms Ceramides are important lipid messengers which carry out important functions such as inhibition of cell proliferation and angiogenesis, and induction of apoptosis [522]. OxLDL can modulate intracellular ceramide homeostasis. For example, our lab has shown that oxLDL inhibits macrophage apoptosis at least in part through inhibiting acid sphingomyelinase activity, thereby blocking ceramide generation [314]. An anti-angiogenic effect has also been shown for ceramide [523]. Interestingly, many of the effects of oxLDL can be mimicked by Cer-P. For example, Cer-P is mitogenic for macrophages, and blocks macrophage apoptosis [522]. In addition, Cer 1-P has been involved in inflammatory responses through stimulation of the cytosolic form of PLA2 and the subsequent generation of prostaglandins [522]. We therefore decided to examine VEGF secretion in response to Cer-P. Figure 3.5 shows that Cer-P caused a dose-dependent increase of VEGF secretion in RAW267.4 cells. To inhibit 64 Cer-P activity we added sphingomyelinase (SMase) to the medium to increase ceramide levels in the cell membrane. Figure 3.6 shows that pre-treating RAW267.4 cells with SMase at 0.6 U/mL or higher led to a decrease in basal as well as in Cer-P stimulated VEGF secretion. However, it did not inhibit oxLDL- mediated VEGF secretion. In fact, a trend toward increased VEGF secretion was observed (Figure 3.6). 3.2.4 Scavenger receptors and oxLDL-mediated VEGF secretion Figure 3.7 shows that there was no difference in VEGF secretion in MPMs from wild-type and SRA -/- CD36 -/- DKO mice. It also shows that MPMs from LOX-1 knockout mice exhibited the same induction of VEGF secretion by incubation with extensively oxidized LDL as MPMs from wild-type mice. These results indicate that signal transduction mediated by SR-A, CD36, or LOX-1 does not play a significant role in VEGF secretion by macrophages. 3.2.5 VEGF secretion is independent of oxLDL uptake Our lab has already shown that in unstimulated peritoneal macrophages, LOX-1 does not significantly contribute to oxLDL uptake [524]. SR-A and CD36 however are both important for macrophage uptake of oxLDL (Section 1.2.4). To determine if uptake of oxLDL was important for VEGF secretion by oxLDL, in parallel with experiments described above, oxLDL uptake was measured in CD36/SR-A double knockout (DKO) MPMs. Figure 3.8A shows the fluorescence plot of one experiment demonstrating a significant reduction of DiI-oxLDL uptake in DKO compared with wild- type cells.. The DKO cells formed a bimodal population, with most cells showing a very low median fluorescence similar to macrophages exposed to native LDL, but also an unexplained minor population with an intermediate level of fluorescence that formed a shoulder on the low-fluorescence population. Figure 3.8B shows pooled results for mean fluorescence intensity from 4 independent experiments. DiI- LDL uptake was similar between MPMs from wild-type and DKO mice (p = 0.65) and as expected, in macrophages from DKO mice DiI-oxLDL uptake was reduced to almost 20% that of cells from wild- type mice (p < 0.001). Taken together, these uptake studies and VEGF measurements show that oxLDL uptake is not essential for its effect on VEGF secretion. 65 3.2.6 OxLDL-mediated VEGF secretion is independent of MAPKK Ras and PI3K can activate two important downstream kinases, MAPK and PKB/Akt, both of which may be involved in regulating VEGF expression (section 1.3.2). To assess the role of MAPK, RAW 264.7 cells were treated with PD 98059, a specific MAPKK inhibitor. At the incubation times and concentrations of inhibitors used, no toxicity from the inhibitor was evident by MTS assay. Figure 3.9A shows that PD 98059 reduced the basal levels of VEGF secretion but oxLDL retained the ability to increase VEGF secretion in the presence of PD 98059 at concentrations as high as 50 µM. The activity of MAPKK inhibitor on its target was verified by immunoblotting for phosphorylated Erk1/2, the downstream target of MAPKK (Figure 3.9B). 3.2.7 PI3K is important in oxLDL-induced VEGF secretion To assess the role of PI3K, RAW 264.7 cells were treated with the PI3K inhibitor LY 294002. Incubation times longer than 18 h or concentrations higher than 30 µM reduced cell viability to 80% (data not shown). Therefore a shorter time-point of 14 hours was used to test the effect of this inhibitor. Figure 3.10A shows that LY 294002 significantly inhibited VEGF secretion. Wortmannin, another inhibitor of PI3K, also significantly reduced VEGF secretion (p < 0.05 data not shown). However, the effect of LY 294002 was greater than that of wortmannin. It is possible that this difference is due to the shorter biological half-life of wortmannin. The activity of LY 294002 on their targets was verified by immunoblotting (Figure 3.10B). VEGF secretion by hMDMs was similarly inhibited by LY 294002 (Figure 3.11). 3.2.8 OxLDL-induced VEGF secretion is independent of classical and novel PKC isoforms PDK-1 acts as a central hub in phosphoinositide signaling. It phosphorylates and activates many members of AGC (cAMP-dependent, cGMP-dependent and protein kinase C) family of kinases including all isoforms of PKC [525]. PKCα and PKCδ have been implicated as key regulators of VEGF expression [391, 526, 527]. To evaluate roles of conventional and novel PKC isoforms we took two approaches. One was to test the effect of RÖ-31-7549 and RÖ-32-0432, which are both inhibitors of 66 conventional and novel, but not the atypical, PKC isoforms. Neither of these compounds inhibited oxLDL-mediated VEGF secretion (Figures 3.12A and B). Second, we exposed RAW 264.7 cells to phorbol 12-myristate 13-acetate (PMA) for 24 h to downregulate expression and activity of conventional and novel PKC isoforms [528]. PMA treatment by itself increased basal levels of VEGF secretion. OxLDL was capable of further increasing VEGF even after PKC depletion by prolonged PMA treatment (Figure 3.12C) which suggests neither conventional nor novel PKC isoenzymes are involved in VEGF upregulation by oxLDL. 3.2.9 PKCζ mediates oxLDL-induced VEGF secretion Next, to examine the role of PKCδ, we used a cell-permeable pseudosubstrate inhibitor of this atypical PKC isoform. This inhibitor slightly but significantly (p < 0.05) reduced VEGF upregulation at 1 μM, and at 5 µM it caused more than 50% inhibition of oxLDL-mediated VEGF secretion (Fig. 3.13). To verify that oxLDL in fact activates PKCδ, we carried out kinase assays on immunoprecipitated PKCδ. It is known that LPS increases PKCδ activity in macrophages [529]. OxLDL‟s ability to increase PKCδ activity was comparable to this positive control (more than 3 fold increase with LPS (1µg/mL, 10 minutes) and oxLDL24 (25µg/mL, 120 minutes). Results in Figure 3.14 show a time-dependent augmentation in PKCδ activity following oxLDL treatment with increasing to more than 500% of control at 50 minutes. This increase in PKCδ activity was reduced to baseline in the presence of a PKCδ pseudo- substrate inhibitor (not shown). 3.3 Summary/discussion In the studies presented in this chapter, we used three different types of macrophages namely RAW 264.7 cells, mouse peritoneal macrophages, and human monocyte derived macrophages. RAW 264.7 macrophages, originally generated through transformation by Abelson Leukemia virus, exhibit many properties and functions of primary macrophages without a requirement for activation (for example by PMA)[530]. Normal intima is devoid of resident macrophages and therefore the major source of intimal macrophages is blood monocytes that are recruited to the site of LDL deposition. 67 Therfore, hMDM is a frequently used model, even though hMDMs show some differences compared to macrophages isolated from atherosclerotic lesions. Mouse peritoneal macrophage is one of most commonly used primary cells in atherosclerosis research. In addition to closely resembling macrophage behavior in artery wall [531], it is an attractive model as functions of specific gene products can be assessed via their deletion in transgenic mice. Our results demonstrate a dose-dependent increase in secretion of VEGF by these macrophages in response to extensively oxidized LDL and are in agreement with previous reports in different cell types [418, 518, 519]. We also found that LDL mildly oxidized with iron, which has mostly peroxidation of lipids and little modification of apoB [472] and LDL modified by exposure to fatty acid oxidation products, which has mostly derivatization of apoB with fatty acid oxidation products but little change to LDL lipids [506] both induced VEGF secretion. This seems to suggest that oxidation of either the lipid or the protein components of LDL may lead to VEGF induction. Our findings with mildly oxidized LDL are in line with recent results that show oxidized phospholipids can induce VEGF secretion in monocyte- derived macrophages [532]. On the other hand, our findings contrast somewhat with the report of Ramos et al., who proposed that LPC in oxLDL may increase VEGF expression and secretion in macrophages [418]. In that study however, the maximal effect of LPC on VEGF was significantly less than that of oxLDL and we suspect that other components of oxLDL were probably also contributing to VEGF upregulation, as we have shown for the growth-inducing effect of LPC in MPM [418]. We found that Ac-LDL had no significant activity on VEGF secretion, indicating that an oxidative rather than a chemical lysine modification is required for VEGF induction. Other groups have suggested that ligands of PPARγ can upregulate VEGF in macrophages [533]. Of interest, 9- and 13-hydroxy-(S)-10,12-octadecadienoic acid (9- and 13-HODE, respectively) were suggested by Inoue et al to account for part of the increase in VEGF secretion mediated by oxLDL [518]. However; the concentrations of ligands used in that study were very high. The specificity of some PPARγ ligands such as ciglitazone and prostaglandin J2 (used in the above-mentioned study) is also in doubt, as many effects attributed to these ligands have been reproduced in PPARγ-null cells [534]. In addition, potent and specific PPARγ agonists were not able to induce VEGF expression in primary 68 macrophages [535]. Synthetic LXR agonists acting through LXR can induce VEGF in macrophages [535], and oxysterols (which are LXR agonists) have been shown to increase VEGF expression in rat vascular smooth muscle cells [536]. Further studies are required to define the quantitative importance of oxysterols in VEGF induction by oxLDL. In our studies, the ability of ceramide to reduce basal levels of VEGF secretion is in line with other studies showing anti-angiogenic properties for ceramide [523]. Additionally, our results show for the first time that Cer-P can increase VEGF secretion. However, it seems that oxLDL and Cer-P are using different mechanisms as ceramide antagonizes Cer-P-induced (as expected) but not oxLDL-induced VEGF secretion. OxLDL is not a simple well-defined entity. Depending on the method used to oxidize LDL, there can be wide variation in the nature and extent of protein or lipid modification. [316]. The initial stage of metal-catalyzed oxidation is accompanied by consumption of LDL antioxidants followed by a propagation phase during which lipid peroxidation of mainly polyunsaturated fatty acids (PUFAs) and cholesterol occurs. Further oxidation includes a decomposition step where oxidized PUFAs are fragmented and covalently bind to lysine residues on apoB100. This lysine derivatization leads to recognition of extensively oxidized LDL by SR-A and loss of recognition by LDL receptors [510]. Due to complexities inherent in these steps, any two preparations may not be identical. In our results with OP- LDL, oxLDL24, Ac-LDL, and FeLDL, different preparations showed very reproducible effects. However one caveat in our studies is that the experiments with oxLDL2 and oxLDL5 were done with same preparations. CD36 and SR-A are two scavenger receptors that together are responsible for more than 80% of binding and uptake of oxLDL by macrophages [269]. We found that macrophages lacking both these receptors had the same induction of VEGF secretion by oxLDL as wild-type mice, which rules out any essential role for either SR-A or CD36 signaling in VEGF induction. Secondly, it also suggests that induction of VEGF secretion is not dependent on receptor-mediated uptake of oxLDL. One could hypothesize that 20% of uptake might be still enough for increased VEGF secretion; however, considering the dose-dependent nature of response to oxLDL, one would expect to see a significant difference between wild-type and DKO cells. Our results also rule out an essential role for LOX-1 69 scavenger receptor. The observed discrepancy between our results and previous reports could be explained by the fact that previous reports used endothelial cells [520] or articular chondrocytes [521]. In addition to cell-type differences, in both studies, pre-incubation with anti-LOX-1 blocking antibody was used to demonstrate a role for this receptor whereas we used macrophages from LOX-1 knockout mice. One would expect that the knockout model would represent a more specific and complete inactivation of LOX-1 than the antibody. As discussed in section 1.3.3, VEGF has a complex role in atherosclerosis and may act as a double-edged sword in that it tends to accelerate lesion progression but can also improve revascularization in ischemic areas of myocardium. OxLDL increases VEGF secretion but can also antagonize potentially beneficial effects of VEGF. For example, oxLDL can inhibit VEGF-induced endothelial progenitor cell differentiation [537] and also can inhibit VEGF-induced NO-dependent migration of endothelial cells [538]. This suggests that if oxLDL induces VEGF in plaque, the angiogenic effect will predominate and potential beneficial effects of VEGF on endothelium would be inhibited by oxLDL. Thus, finding signaling pathways that link atherogenic stimuli (such as oxLDL) to VEGF but do not block beneficial effects of this cytokine could be therapeutically important.The regulatory mechanisms involved in increased VEGF secretion in response to oxLDL have not been clearly dissected. Previous studies indicated a role for PPARγ and P38 MAPK but in those studies each pathway was shown to have only a partial effect [518, 519]. Many stimuli act through different receptor tyrosine kinases (RTKs) followed by activation of PI3K/PKB and Ras dependent MAPK pathways and both of these pathways are activated in macrophages in response to oxLDL [310]. We found that MAPKK activity was involved in the control of baseline VEGF secretion but MAPKK inhibition did not block oxLDL-induced VEGF secretion. On the other hand, PI3K inhibitors significantly inhibited oxLDL mediated VEGF secretion. Another caveat in our studies is that pharmacological inhibitors are not 100% selective and specific for their targets. For example wortmannin may inhibit MAPK at higher doses and LY 294002 can inhibit casein kinase 2[539]. However, the fact that both these inhibitors were able to inhibit oxLDL-mediated VEGF secretion suggests that the effect is mediated by their common (and intended) target. One of the potential targets of PI3K is PKC. We found no evidence that 70 conventional and novel PKC isoforms are involved. Our finding that prolonged exposure to PMA increases VEGF expression corroborates the findings of Shih et al. In those studies PKCδ was suggested to play a key role in PMA-mediated VEGF upregulation [526]. In our hands, oxLDL was still able to further upregulate VEGF with extended PMA treatment. This also fits well with findings of Ijichi et al. who showed that brief and prolonged PMA treatment both induce VEGF expression, and that angiogenic stimuli such as hypoxia are still able to increase VEGF in the presence of PMA, consistent with a role for atypical PKC isoforms [540]. We showed that PKCδ is activated by oxLDL and also demonstrated that a cell permeable pseudosubstrate inhibitor of PKCδ significantly blocked oxLDL-mediated VEGF secretion. This suggests that activation of this atypical PKC isoform is important for VEGF induction by oxLDL. 71 Figure 3.1 OxLDL increases VEGF expression and secretion in RAW264.7. Macrophage VEGF secretion in response to native LDL (nLDL) or LDL that had been extensively oxidized with Cu ++ for 24 h (oxLDL24) was measured by ELISA as described in the methods. Panels represent mean values ± SD from at least 3 independent experiments with duplicates for each condition. A) Time course of VEGF secretion by RAW 264.7 cells. Medium from untreated cells (CTL) or cells treated with 25 µg/mL of nLDL or oxLDL24 was assayed at the indicated time points. B) Effect of oxLDL concentration in RAW 264.7 cells. VEGF was assayed in medium after 24 h incubation with increasing concentrations of nLDL or oxLDL24. Level of significance of difference between oxLDL and corresponding nLDL control is indicated as follows: * p ≤ 0.05, ** p ≤ 0.01, and *** p ≤ 0.001. No difference was observed between cells treated with nLDL or control (CTL) cells exposed to medium alone. C) VEGF mRNA was measured in RAW264.7 by qPCR at baseline, 3, and 6 h after treatment with oxLDL24 or nLDL. Results 72 were normalized to β-actin and expressed as fold increase of oxLDL24 treatment relative to nLDL. *** denotes level of significance of p ≤ 0.001 between baseline (0 h) and 6 h conditions. 73 Figure 3.2 OxLDL increases VEGF secretion in primary macrophages. Macrophage VEGF secretion in response to nLDL or oxLDL24 was measured as described in the methods. Panels represent mean values ± SD from 3 independent experiments with duplicates for each condition. A) VEGF secretion by hMDM. Medium was assayed for VEGF after 48 h of treatment with increasing concentrations of nLDL or oxLDL24. B) VEGF secretion by MPM. Medium was assayed for VEGF after 24 h with no treatment (CTL) or with 25 μg/mL of nLDL or oxLDL24. Level of significance of difference between oxLDL and corresponding nLDL control is indicated as follows: * p ≤ 0.05, ** p ≤ 0.01, and *** p ≤ 0.001. No difference was observed between cells treated with nLDL or control (CTL) cells exposed to medium alone (Not shown in panel A). 74 Figure 3.3 Effect of degree of LDL oxidation on VEGF secretion. A) VEGF secretion by RAW264.7 cells was measured after 24 h of treatment with 25 µg/mL of nLDL or the same concentration of LDL that has been modified with ferrous ion (FeLDL) or with copper ion for 2, 5, or 24 h (oxLDL2, 5, or 24, respectively). The panel represents mean values ± SD from 3 independent experiments with duplicates for each condition. Level of significance of difference between nLDL control and each condition is indicated as: * p ≤ 0.05 and *** p ≤ 0.001. B) With point of origin at the minus sign, electrophoretic mobility of nLDL and modified LDLs was assessed on agarose gel (Titan gel lipoprotein kit). Lipoprotein bands were visualized by Fat Red staining and the mobility was determined by measuring the distance between the point of origin (minus sign) and the leading edge of the lipoprotein and was expressed relative to the mobility of nLDL (relative mobilities as follows: FeLDL: 1.5, oxLDL2: 2.8, oxLDL5: 4.9, and oxLDL24: 6.2). 75 Figure 3.4 Effect of type of apoB modification on VEGF secretion. LDL was extensively modified by acetylation (Ac-LDL), by exposure to copper ion for 24 h (oxLDL24) or by exposure to oxidation products of fatty acids (OP-LDL) as described in methods. Panels represent mean values ± SD from at least 3 independent experiments with duplicates for each condition. A) VEGF secretion by RAW264.7 after 24 h treatment with 25 µg/mL of nLDL, oxLDL24 or Ac-LDL and varying concentrations of OP- LDL. B) Bars represent electrophoretic mobility of modified LDLs used in these experiments relative to nLDL C) VEGF secretion in MPMs from CD-1 mouse after 24 h incubation with 25 μg/mL of oxLDL24, nLDL, Ac-LDL or OP-LDL. D) VEGF secretion in hMDM after incubation for 48 h with 25 μg/mL of nLDL or oxLDL24, or 40 μg/mL of Ac-LDL or OP-LDL. For panels C) and D), the electrophoretic mobilities of different LDL modifications relative to nLDL were 6.8, 6.2, and 4 for Ac- LDL, oxLDL24 and OP-LDL respectively. (* p ≤ 0.05 and *** p ≤ 0.001 between nLDL control and the specified conditions). 76 Figure 3.5 Ceramide-1-phosphate increases VEGF secretion in RAW267.4 cells. Macrophage VEGF secretion was measured in medium after 24 h incubation with oxLDL, nLDL, or increasing concentrations of ceramide1-1-phosphate (Cer-P) and expressed as mean values ± SD from at least 3 independent experiments with duplicates for each condition. Level of significance of difference between each condition and nLDL control is indicated as ** p ≤ 0.01 and *** p ≤ 0.001. 77 Figure 3.6 Ceramide inhibits Cer-P-mediated, but not oxLDL -mediated VEGF secretion. Following 24 h incubation with or without indicated concentrations of sphingomyelinase (SMase), media was changed and RAW267.4 cells were treated with oxLDL (25 μg/mL), nLDL (25 μg/mL) or Cer-P (50 μM) and VEGF secretion was measured in medium after 24 h incubation. Bars represent the difference between the mean and the upper range from 2 independent experiments with triplicates for each condition. 78 Figure 3.7 LOX-1, SR-A, and CD36 are not essential for oxLDL-mediated VEGF secretion. VEGF secretion in MPMs from wild-type (WT), LOX-1, and SR-A/CD36 (DKO) knockout mice was measured after 24 h incubation with 25 μg/mL of oxLDL24 or nLDL. Each bar represents pooled data ± SD from three independent experiments. 79 Figure 3.8 OxLDL uptake is significantly reduced in SR-A/CD36 double knockout MPMs. A) The histogram represents lipoprotein uptake from one experiment in wild-type (WT) and SR-A/CD36 double knockout (DKO) MPMs. Unlabeled oxLDL is used as unstained control. DiI-LDL and DiI-OxLDL represent fluorescently labeled nLDL and oxLDL24, respectively. B) DiI uptake was measured as described in the methods. Data represent the means and SD of pooled data from four independent experiments. 80 Figure 3.9 MAPKK is not essential for oxLDL-mediated VEGF secretion. A) Role of mitogen activated protein kinase was examined using the selective MAPKK inhibitor PD 98059. RAW 264.7 cells were treated with oxLDL24 or 5 µL of DMSO as vehicle control with or without pre-incubation for 45 min with the indicated concentration of PD 98059. The graph represents data from one experiment done in duplicate and was confirmed in two independent experiments. B) The activity of PD 98059 on MAPKK in RAW 264.7 cells was verified by evaluation of phosphorylation status of the downstream target MAPK1 (Erk2) by immunoblot. 81 Figure 3.10 Role of PI3K in oxLDL-mediated VEGF secretion in RAW 264.7 cells. A) RAW 264.7 cells were incubated for 14 h with oxLDL24 or nLDL, with or without 45 min pretreatment with indicated concentrations of LY 294002, a PI3K inhibitor. Bars represent mean values ± SD from three independent experiments with duplicates for each condition. (***: p ≤ 0.001 between oxLDL24 and the specified treatments). B) The activity of 15 µM LY 294002 in RAW 264.7 cells was verified by evaluation of PKB phosphorylation by immunoblot. 82 Figure 3.11 Role of PI3K in oxLDL-mediated VEGF secretion in hMDMs. Human MDMs were incubated for 48 h with oxLDL24 or nLDL, with or without 45 min pretreatment with indicated concentrations of LY 294002. Bars represent means ± SD from two independent experiments with triplicates for each condition. (**: p ≤ 0.05 between oxLDL24 and the specified conditions). 83 Figure 3.12 OxLDL-induced VEGF secretion is independent of classical and novel PKC isoforms. RAW 264.7 cells were pretreated with increasing concentrations of A) RÖ 32-0432 or B) RÖ 31-7549, inhibitors of both conventional and novel PKC isoforms. C) RAW 264.7 cells were preincubated with or without 1 μM phorbol 12-myristate 13-acetate (PMA). After 24 h, medium was replaced with fresh medium containing 25 μg/mL oxLDL, or no oxLDL. After an additional 24 h incubation, medium was harvested for VEGF immunoassay. All graphs represent data from one of two or three similar experiments all carried out in duplicate. Error bars represent mean ± range. 84 Figure 3.13 PKCζ mediates induction of VEGF secretion by oxLDL. RAW 264.7 cells were pretreated with increasing concentrations of a myristoylated PKCδ pseudo-substrate peptide (PKCδ-PS) for 1 h prior to addition of oxLDL24. After 24 h of incubation, VEGF was measured. Bars represent means ± SD from three independent experiments with duplicates for each condition. Level of significance of difference between each condition and oxLDL24 is indicated as ** p ≤ 0.01 and *** p ≤ 0.001. No significant effect of PKCδ-PS by itself was observed on basal levels of VEGF. 85 Figure 3.14 OxLDL induces PKCζ activation. RAW 264.7 cells were treated with 25 μg/mL of oxLDL24 and PKCδ kinase activity was measured as described in Methods. Results were normalized and expressed as fold increase relative to no treatment control. The graph represents pooled data from four independent experiments (*: p ≤ 0.05 between 50 minutes of oxLDL24 treatment and control). 86 4. Receptors for oxLDL and its prosurvival effect in macrophages 4.1 Introduction/rationale Macrophage survival and proliferation play an important role in pathogenesis of atherosclerosis (section 1.2.8). The signaling pathways mediating prosurvival effects of oxLDL have been partially defined (section 1.2.6). However, no studies have been reported that characterize the signaling events upstream of PI3K activation. The fact that signaling events are triggered within seconds to minutes after addition of oxLDL suggests a receptor-mediated mechanism. The studies presented in this section were carried out to determine if one of the pattern recognition receptors (PRRs) expressed by macrophages is required for anti-apoptotic signaling by oxLDL in macrophages. Important macrophage PRRs and their significance in atherogenesis and oxLDL uptake were discussed in sections1.2.2 and 1.2.4, respectively. The major scavenger receptors I studied were SR-A, CD36, and LOX-1. As well, I tested a role for signaling PRRs including TLR4 and its signaling partner CD14. FcγRIIb, a member of immunoglobulin receptor superfamily, was also studied. At least in mice, FcγRIIb has been shown to bind to oxLDL with high affinity [541]. 4.2 Results 4.2.1 LOX-1 signaling is not required for oxLDL prosurvival activity LOX-1 is a major receptor for oxLDL in endothelial cells [542], but is also expressed in monocyte-derived macrophages [542, 543] and macrophages in atherosclerotic plaques [544]. There is evidence showing that LOX-1 can trigger cell signaling in response to oxLDL [545-548]. To address the role of LOX-1 in oxLDL-mediated macrophage survival, we compared the ability of oxLDL to promote survival in cytokine-deprived BMDMs from wild-type and LOX-1 deficient mice. Figure 4.1 shows that after 24 h there was no significant difference in the viability of wild-type and LOX-1 deficient cells treated with oxLDL. Additionally, there was no significant difference between wild-type and LOX-1 BMDMs in the percentage of sub-diploid cells after incubation with oxLDL (Figure 4.2). To measure the contribution of LOX-1 to oxLDL uptake in BMDMs, uptake of fluorescently labeled oxLDL was 87 compared between LOX-1 and WT BMDMs, and as show in Figure 4.5B, there is no significant difference. 4.2.2 OxLDL-mediated macrophage survival is independent of CD36 or SR-A receptors CD36 [233-235] and SR-A [229-232] have both been shown to contribute to atherogenesis in animal models. It has also been reported that these endocytic receptors are capable of initiating cell signaling upon ligand binding [549, 550]. To test the effect of CD36 and SR-A, the ability of oxLDL to promote survival or inhibit apoptosis were compared between BMDMs from wild-type mice and from mice recombinant for deletion mutations in both these receptor genes. Figure 4.4 shows that oxLDL but not native LDL increased BMDM viability measured with the MTS assay in cells from both wild-type and DKO mice. There was no significant difference in viability after 24 h of M-CSF deprivation between wild type and DKO BMDMs at any concentration of oxLDL (Figure 4.4A). Longer time points (48 h) also did not show any difference with oxLDL in WT or DKO BMDMs (Figure 4.4B). Figure 4.5 shows the number of sub-diploid cells relative to the control condition (incubated without M-CSF or oxLDL (oxLDL 0 µg/mL)). Again, oxLDL was equally effective in reducing apoptosis in WT and DKO BMDMs. After 24 h of treatment, 50 µg/mL of oxLDL appeared slightly more effective at reducing sub- diploid population in DKO BMDMs (P = 0.03) but there was no difference after 48 h incubation and so the statistical significance at 24 h is probably spurious. 4.2.3 OxLDL-mediated macrophage survival is independent of oxLDL uptake SR-A and CD36 together are responsible for most of the uptake of oxLDL in mouse peritoneal macrophages [269]. To confirm the functional inactivation of these receptors for oxLDL, in parallel with the above-mentioned experiments, uptake of fluorescently labeled oxLDL was measured in BMDMs as described in the methods section. Figure 4.5A shows that uptake of oxLDL in DKO BMDMs was reduced by 60% compared to wild-type. 88 4.2.4 Toll-like receptor 4 (TLR4) is not essential for oxLDL-mediated survival Signaling through TLR4 is believed to play an important role in atherosclerosis [219, 222, 223]. Interestingly, minimally oxidized LDL can initiate TLR4-dependent signaling [101].At least certain oxidation specific epitopes on oxLDL have been reported to act as PAMPs to initiate TLR4-mediated signaling [551].To determine if TLR4 is responsible for the anti-apoptotic effect of oxLDL, we measured viability and apoptosis in TLR4 deficient macrophages. As shown in Figure 4.6A, there was no difference between viability of wild type and TLR4 deficient BMDMs in response to different concentrations of oxLDL. As well, oxLDL was equally effective in reducing the sub-diploid population in TLR4 -/- as in wild type BMDM (Figure 4.7). 4.2.5 CD14 is not required for oxLDL prosurvival activity In addition to the fact that CD14 acts as a signaling partner of TLR4 [552], some types of oxidatively modified LDL are reportedly recognized by (and bind to) CD14 [553]. Moreover, studies have shown that SR-A ligands can activate PI3K pathway via a CD14 dependent, SR-A independent pathway [554]. To determine whether CD14 plays a role in oxLDL-mediated survival, we compared oxLDL viability in BMDMs from wild type and CD14 knockout mice. OxLDL was able to increase viability (Figure 4.8) and inhibit apoptosis (Figure 4.9) in CD14-null BMDMs as effectively in as wild- type cells. 4.2.6 Fc gamma receptor IIb (FcγRIIb) and oxLDL-mediated survival FcγRIIb, a macrophage receptor for Fc region of IgG, also binds to oxLDL with high affinity [541]. However, oxLDL increased viability in FcγRIIb-deficient macrophages to the same extent as in wild type cells (Figure 4.10). With FcγRIIb deficiency, we observed a slight increase in the sub-diploid population in all conditions (with or without M-CSF) which reached statistical significance for oxLDL 25 µg/mL treatment (Figure 4.11). This difference represented only ~10% of oxLDL ability to inhibit apoptosis and in FcγRIIb-/- BMDMs, oxLDL was still able to significantly decrease sub-diploid 89 population compared to no M-CSF control or LDL treatment (p<0.0005 for oxLDL at 25 and 50 µg/mL, Figure 4.11). 4.3 Summary/discussion In these studies we evaluated macrophages from mice lacking a variety of receptors for oxLDL and found that none of these receptors were required for the prosurvival effect of oxLDL. LOX-1 is an endocytic scavenger receptor that has been shown to trigger cell signaling in response to oxLDL [545- 548]. Our findings however show that in BMDMs LOX-1 is not required for oxLDL-mediated pro- survival signaling events. These observations are supported by a recent publication showing that in unstimulated MPMs, LOX-1 inactivation does not reduce oxLDL internalization [524]. CD36 and SR-A together account for the majority of oxLDL uptake and are thought to play a role in foam cell formation. Kunjathoor et al. showed in their studies that ~85% of oxLDL uptake in mouse peritoneal macrophages is mediated by these two receptors [269]. In our hands, 60% of oxLDL uptake in BMDMs was dependent on the combined effect of these receptors. This difference is probably due to proportionately greater receptor-independent uptake of oxLDL in BMDM compared to peritoneal macrophages. In fact in studies presented in chapter 3, similar to [269], we found that up to 80% of oxLDL uptake in MPMs is mediated by these two receptors. Another possible explanation for this cell- type specific difference could be that the pattern of SR expression (or compensatory mechanisms) between BMDMs and MPMs is different (with CD36 and SR-A being quantitatively more important in MPMs). In any case, considering the dose-dependent nature of oxLDL-induced prosurvival effect, the fact that oxLDL sustains its ability to promote survival in spite of 60% reduction in uptake suggests that uptake of oxLDL is not required for its anti-apoptotic effect. Although endocytic scavenger receptors are not generally considered as signaling receptors, there are reports that CD36 [550, 555] and SR-A [549] can initiate signaling upon ligand/receptor binding. Our results show that neither SR-A nor CD36 is responsible for oxLDL-induced signal transduction. Toll-like receptor signaling is thought to be involved in the inflammatory processes that are important in the pathogenesis of atherosclerosis. Our results show that the TLR4 do not play a role in 90 anti-apoptotic effect of oxLDL in BMDM. This is in line with other experiments carried out in our laboratories showing that BMDMs from C3H/HeJ (LPS-unresponsive TLR4 mutant [556]) or TLR2 knockout mice also exhibited the same viability pattern in response to oxLDL [557]. In our hands, FcγRIIb-deficient BMDMs exhibited a general increase in sub-diploid population compared to wild-type which became statistically significant for only one of oxLDL treatments. One point worth noting is the genetic background of FcγRIIb knockout animals used in our experiments. FcγRIIb knockout mice come from a mixed Sv129/C57BL6 genetic background. The controls used in our studies have pure C57BL6 background and the marginal difference between the two cells could have been caused by a residual Sv129 DNA carryover. Strain-specific differences in the effect of FcγRIIb deficiency has been observed previously [558]. However this is not a likely explanation for the increased sub-diploid population as the observed trend was discordant with parallel MTS results where an increase in viability of FcγRIIb-deficient BMDMs was seen. While this marginal trend might be spurious, it is plausible in the sense that a decrease in apoptotic cell clearance in FcγRIIb-null macrophages could lead to a larger sub-diploid pool. Although FCγRs have been shown to be involved in clearance of apoptotic cells in vivo, no reports exist in literature regarding direct (immunoglobulin-independent) clearance. It is however known that that FcγRIIb deficiency leads to an autoimmune phenotype and an exaggerated IL- 12 response in antigen-presenting cells incubated with apoptotic thymocytes [559]. In any case, this difference observed in our experiments at best can account for only ~10% of oxLDL ability to inhibit apoptosis which rules out an essential role for FcγRIIb. Other receptors of interest for future studies include SR-PSOX (also known as CXCL16) receptor for advanced glycation products (RAGE), and SR-BI. SR-PSOX is a high affinity receptor for oxLDL and phosphatidylserine [560]. Aslanian et al have shown that murine SR-PSOX accounts for ~34% of oxLDL uptake. Unexpectedly however, they showed an atheroprotective role for this scavenger receptor [237]. An increased number of apoptotic cells in the plaques of SR-PSOX-null mice were observed which was attributed to ineffective clearance of apoptotic cells. This putative role in apoptotic cell clearance was suggested as a mechanism for atheroprotection by SR-PSOX. To our knowledge there are no reports on SR-PSOX signaling in response to oxLDL. It would be interesting to see if SR-PSOX 91 is required, at least partially, for oxLDL-mediated anti-apoptotic signaling in macrophages. Although not classified as a scavenger receptor, RAGE, is also another multi-ligand receptor that belongs to the immunoglobulin superfamily and is expressed in macrophages [241]. It was recently shown that RAGE deactivation attenuates atherosclerosis in LDLR -/- mice. In that study, it was shown that RAGE triggers cell signaling in response to oxLDL and in fact, part of the protective effect of RAGE deletion was attributed to a dampened inflammatory response of macrophages following oxLDL treatment. In their studies, Sun et al. show that RAGE is required for oxLDL-mediated MAPK signaling however they did not study the possible role of RAGE in other signaling pathways induced by oxLDL [242]. We have shown before that MAPK signaling by oxLDL is not essential for its prosurvival effect [310] but it would be interesting to test the effect of oxLDL-induced prosurvival pathways such as PI3K and calcium signaling in RAGE-deficient BMDMs. Interestingly, Kinslinger et al. have shown that N ε - carboxymethyllysine adducts are ligands of RAGE [561]. In addition to glycoxidation, lipid peroxidation followed by protein modification is capable of generating N ε -carboxymethyllysine adducts (section 1.3.5 and [562]) , therefore it would be interesting to study and compare the contribution of this receptor to the prosurvival effects of oxLDL and OP-LDL. As discussed in section 1.2.2, SR-BI is an atheroprotective scavenger receptor through its roles in reverse cholesterol transport [238-240]. Different groups have shown that SR-BI contributes to cellular bindings and degradation of oxLDL [563, 564]. Additionally, it has been shown that SR-BI is capable of triggering signaling events and activating kinases (such MAPK and p38) upon ligand binding [565, 566]. These properties makes SR-BI another interesting candidate for mediating the anti-apoptotic effects of oxLDL in macrophages. 92 Figure 4.1 LOX-1 is not essential for oxLDL-mediated survival. BMDMs from wild-type (WT) or LOX-1 knockout mice were seeded at 3 x 10 4 cells/well in 96 well plates. After overnight incubation in media containing M-CSF, they were washed and incubated for 24 h without M-CSF, but with indicated concentrations of oxLDL. Viability was then measured by the bioreduction of MTS as described in the methods. The results represent means ± SD of pooled data from four independent experiments. Viability is expressed as a percentage relative to that of cells treated with M-CSF. None of the differences between corresponding WT and LOX-1conditions reached statistical significance. 93 94 Figure 4.2 LOX-1 is not essential for the anti-apoptotic effect of oxLDL. BMDMs from WT or LOX- 1 mice were plated at 10 6 cells/well in 6-well plates. Following overnight incubation in M-CSF- containing media cells were washed and incubated for 24 h in medium containing M-CSF or no M-CSF and indicated concentration of oxLDL. The sub-diploid population was measured by flow cytometry after propidium iodide staining. In the upper panel, histograms of each condition from one representative experiment are shown. The sub-diploid population was measured by counting the cells with fluorescence intensity under the region depicted by the red line. Results in the lower panel represent the means ± SD of pooled data from three independent experiments which are expressed relative to control cells incubated with neither M-CSF nor oxLDL. None of the differences between corresponding WT and LOX-1conditions reached statistical significance. 95 Figure 4.3 SR-A and CD36 are not required for oxLDL-mediated survival. BMDMs from wild-type (WT) or SR-A/CD36 double knockout (DKO) mice were seeded at 3 x 10 4 cells/well in 96 well plates. After overnight incubation in media containing M-CSF, they were washed and incubated without M- CSF, but with indicated concentrations of native LDL (nLDL) or oxLDL for A) 24 h or B) 48 h. Viability was then measured by the bioreduction of MTS as described in the methods. Results represent the means ± SD of pooled data from four independent experiments. Viability is expressed as a percentage relative to that of cells treated with M-CSF. None of the differences between corresponding WT and DKO conditions reached statistical significance. 96 97 Figure 4.4 OxLDL-mediated inhibition of apoptosis is independent of SR-A and CD36. BMDMs from WT or CD36/SR-A double knockout (DKO) mice were plated at 10 6 cells/well in 6-well plates. Following overnight incubation in media containing M-CSF, cells were washed and then replenished with in medium containing M-CSF or no M-CSF together with the indicated concentration of nLDL or oxLDL. A) The sub-diploid population was measured by flow cytometry after propidium iodide staining. Histograms from one representative experiment are shown. The sub-diploid population was measured by counting the cells with fluorescence intensity under the region depicted by the red line. Results in B) and C) are means ± SD of pooled data from four independent experiments at 24 h incubation (B) or 48 h (C) expressed relative to control cells incubated with neither M-CSF nor oxLDL. Unless specified, the difference between WT and the corresponding DKO is not statistically significant. 98 Figure 4.5 OxLDL uptake in BMDMs depends on SR-A/CD36 but not LOX-1. BMDMs from wild- type (WT), CD36/SR-A double knockout (DKO) (A), or LOX-1 knockout mice (B) were seeded at 10 6 cells/well in 6-well plates. They were then washed and incubated for 24 hr with fluorescently labeled oxLDL or nLDL (DiI-oxLDL and DiI-LDL, respectively). Cell content of DiI was then measured by fluorimetry after isopropanol extraction and expressed as percentage of DiI-oxLDL uptake in corresponding WT BMDMs. (NS: not significant). 99 Figure 4.6 TLR4 does not contribute to the prosurvival effect of oxLDL. BMDMs from wild-type (WT) or Toll-like receptor 4 (TLR4) knockout mice were seeded at 3*10 4 cells/well in 96 well plates. After overnight incubation in media containing M-CSF, they were washed and incubated for 24 h without M-CSF, but with indicated concentrations of nLDL or oxLDL. Viability was then measured by the bioreduction of MTS. Results represent the means ± SD of pooled data from three independent experiments, each in triplicate. Viability is expressed as a percentage relative to that of cells treated with M-CSF. None of the differences between corresponding WT and TLR4 conditions reached statistical significance. 100 101 Figure 4.7 TLR4 is not essential for the anti-apoptotic effect of oxLDL. BMDMs from WT or CD14 mice were incubated for 24 h in medium containing M-CSF or no M-CSF and indicated concentration of nLDL or oxLDL. The sub-diploid population was measured by flow cytometry after propidium iodide staining. In the upper panel, histograms of each condition from one representative experiment are shown. The sub-diploid population was measured under the region depicted by red line. The lower panel shows means ± SD of pooled data from three independent experiments expressed relative to control cells incubated with neither M-CSF nor oxLDL. None of the differences between the WT and the corresponding TLR4 reached statistical significance. 102 Figure 4.8 CD14 is not essential for the prosurvival effect of oxLDL. BMDMs from wild-type (WT) or CD14 knockout mice were seeded at 3 x 10 4 cells/well in 96 well plates. After overnight incubation in media containing M-CSF, they were washed and incubated for 24 h without M-CSF, but with indicated concentrations of nLDL or oxLDL. Viability was then measured by the bioreduction of MTS. Results represent the means ± SD of pooled data from three independent experiments, each done in triplicate. Viability is expressed as a percentage relative to that of cells treated with M-CSF. None of the differences between the WT and corresponding TLR4 reached statistical significance. 103 104 Figure 4.9 CD14 does not contribute to the anti-apoptotic effect of oxLDL. BMDMs from WT or CD14 mice were incubated for 24 h in medium containing M-CSF or no M-CSF and indicated concentration of nLDL or oxLDL. The sub-diploid population was measured by flow cytometry after propidium iodide staining. In the upper panel, histograms of each condition from one representative experiment are shown. The sub-diploid population was measured under the region depicted by the red line. The lower panel shows the means ± SD of pooled data from three independent experiments which are expressed relative to control cells incubated with neither M-CSF nor oxLDL. Unless specified, the difference between WT and the corresponding CD14 is not statistically significant. 105 Figure 4.10 Fc gamma receptor IIb (FcγRIIb) is not required for the prosurvival effect of oxLDL. BMDMs from wild-type (WT) or FcγRIIb knockout mice were seeded at 3 x 104 cells/well in 96-well plates. After overnight incubation in media containing M-CSF, they were washed and incubated for 24 h without M-CSF, but with indicated concentrations of nLDL or oxLDL. Viability was then measured by the bioreduction of MTS. Results represent the means ± SD of pooled data from three independent experiments, each done in triplicate. Viability is expressed as a percentage relative to that of cells treated with M-CSF. With oxLDL treatment, there was no statistically significant difference between the WT and FcγRIIb knockout macrophages. 106 107 Figure 4.11 Anti-apoptotic effect of oxLDL and Fc gamma receptor IIb (FcγRIIb). BMDMs from WT or FcγRIIb -/- mice were incubated for 24 h in medium containing M-CSF or no M-CSF and indicated concentration of nLDL or oxLDL. The sub-diploid population was measured by flow cytometry after propidium iodide staining. In the upper panel, histograms of each condition from one representative experiment are shown. The sub-diploid population was measured in the area depicted by the red marker. The lower panel shows the means ± SD of pooled data from three independent experiments which are expressed relative to control cells incubated with neither M-CSF nor oxLDL. Unless specified, the difference between the WT and FcγRIIb knockout macrophages is not statistically significant. 108 5. Fluorescent adducts formed by reaction of oxidized unsaturated fatty acids with amines induce macrophage growth 5.1 Introduction/rationale Macrophages play a key role in pathogenesis of atherosclerosis from its conception to complications (section 1.2). In particular, macrophage burden correlates well with the disease progression and clinical outcome [246, 247]. As discussed in section 1.2.6, several groups including our own have shown that oxLDL promotes macrophage growth and survival. OP-LDL, or LDL modified by oxidation products of fatty acids (OPs), was used in our laboratory as a model for metal-independent oxidatively modified LDL (section 1.4.5). In OP-LDL, unlike extensive modification of LDL with copper ion, endogenous lipids are not oxidized and LPC content is not increased. Even though lysine residues of apoB become derivatized, the apoB itself is not fragmented [268, 506, 510]. Epitopes recognized by antibodies specific to this type of modification are shown to exist both in vivo and in LDL oxidized by other methods [87, 507]. Martens et al. showed that OP-induced modification of not only LDL but also proteins such as albumin or immunoglobulins confer mitogenic properties towards macrophages [307]. OPs and protein modification by them were discussed in sections 1.4.2 and 1.4.5, respectively. The studies presented in this chapter were carried out to further define OPs and adducts they generate through reacting with lysine residues with relation to macrophage growth and survival. 5.2 Results 5.2.1 Both lipid and protein moiety of oxLDL promote macrophage growth Sakai et al. had suggested that LPC is important in oxLDL-mediated survival [298, 299]. Martens et al. subsequently challenged this report with data showing that in MPMs, the effect of macrophage growth is not due to LPC [307]. In contrast, they found that lysine modification of apoB is essential, as prior methylation of apoB lysine residues abolished growth-promoting properties of oxidatively modified LDL. To determine more rigorously whether the lipid or protein moiety is contributing to the induction of growth by oxLDL, we extracted the lipid and separately reconstituted the 109 lipid and protein moieties of oxLDL. The ability of these fractions to promote macrophage growth was compared. Figure 5.1 shows that both lipid and protein fractions can promote growth of MPMs. 5.2.2 OPs react with lysine residues To confirm that the lysine residue is the target of reactive OPs, polylysine was preincubated with OPs and the mixture was then added to LDL. Figure 5.2 show that this pre-treatment with polylysine completely abolishes induction of growth. Preincubation with polylysine also significantly prevents lysine modification on apoB as judged by relative electrophoretic mobility measurements (1.2 vs. 6.6- fold that of native LDL with or without polylysine pre-treatment, respectively). 5.2.3 Amine modification by OPs is sufficient for growth induction We were somewhat surprised by the observation that lipid extracts of oxLDL can promote macrophage growth (Figure 5.1). One possibility for this finding could be the modification of amine- containing phospholipids by OPs. To explore this possibility, phosphatidylethanolamine (PE)-containing liposomes were modified by OPs from arachidonic and linoleic acid (AOP-PE and LOP-PE, respectively). Figure 5.3A shows that OP-PE liposomes are capable of promoting growth in MPMs - although not quite to the same extent of OP-LDL. On the other hand, the product of incubation of phosphatidylcholine (PC) liposomes, which lack the free amino group, with OP did not induce growth (Figure 5.3B). Additionally, Figure 5.3B shows that prior treatment (and reduction) of OPs with sodium borohydride abolished the growth-promoting effect of OP-PE liposomes. 5.2.4 Borohydride-reducible functional groups in OPs are essential for the growth-promoting effect Similar to OP-PE (Figure 5.3B), we observed that pre-treatment of OPs with borohydride will abolish the growth-promoting effect of OP-LDL (Figure 5.4). Figure 5.4 additionally shows that borohydride treatment after LDL modification does not diminish the ability to promote growth. This suggests that borohydride-reducible functional groups are important for growth-promoting adduct 110 formation while such functional groups are not required after modification. The electrophoretic mobility of LDL was reduced by 50% with borohydride pretreatment (not shown) which suggests some reactive OPs are not amenable to reduction by borohydride. 5.2.5 Alkenals do not account for the growth-inducing activity of OPs Borohydride-reducible functional groups include reactive aldehydes. Aldehydes are commonly generated during lipid peroxidation of unsaturated fatty acids (section 1.4.1) and are capable of modifying lysine residues and creating fluorescent adducts (section 1.4.6). In fact, aldehydic reactive products are reportedly the main components of oxidized fatty acids responsible for lysine modification of apoB [316, 505, 567]. To test if these reactive aldehydes could be responsible for the growth-inducing derivatization, we modified LDL with 4-hydroxynonenal (HNE) or 4-hydroxyhexenal (HHE), the major fragmented aldehydic products of arachidonic acid oxidation [316], to generate HNE-LDL and HHE- LDL, respectively. Figure 5.5 shows that neither HNE-LDL nor HHE-LDL induced macrophage growth. 5.2.6 Hydroperoxides and/or endoperoxides contribute to the induction of growth by OPs Other borohydride-reducible functional groups in OP would include hydroperoxides and endoperoxides. Triphenylphosphine (TPP), a reducing agent selective for hydroperoxides and endoperoxides, was preincubated with OPs and the mixture was then used to modify LDL. Table 5.1 shows that TPP substantially, although not completely, reduced the ability of OP to modify lysine residues (based on electrophoretic mobility) and diminished the ability of modified LDL to induce growth. This suggests that at least part of OP reactivity responsible for lysine modification and growth induction is due to hydro (and/or endo) peroxides. 111 TABLE 5.1 Hydroperoxides and/or endoperoxides contribute to the induction of growth by OPs. Arachidonic acid or linoleic acid (100 μmoles) was autoxidized for 48 h, and dissolved in 1 ml CHCl3. Aliquots were then incubated with or without a 2-fold molar excess of triphenylphosphine for 12 h at 4°C. Lipid hydroperoxide content of each sample was assayed in triplicate. An aliquot equivalent to 6 μmoles of fatty acid was mixed with 1 mg LDL in PBS for 8 h, and agarose gel electrophoresis of each LDL sample was performed. Each LDL sample was also tested for effects on macrophage viability using the MTS assay. Viability results are normalized to the value with LOP-LDL. Similar results were obtained in two replicate experiments. Relative electrophoretic mobility macrophage viability LOOH assay mol/mol LOP or AOP LDL 1.0 45% <0.1 LOP-LDL 2.9 100% 2.7 ± 0.7 (LOP+TPP)-LDL 1.6 58% 0.3 ± 0.1 AOP-LDL 3.3 80% 1.8 ± 0.8 (AOP+TPP)-LDL 2.2 68% 1.1 ± 0.3 112 5.2.7 Autoxidation of arachidonic and linoleic acids produces mainly unfragmented oxygenated species The OPs of LA and AA were studied using electron ion spray-mass spectrometry (ESI-MS). Figure 5.6 shows the results of ESI-MS in positive ion mode. Unoxidized AA (MAA = 304) under our experimental conditions existed as protonated (m/z = 305) and sodated (m/z = 327) forms. Unoxidized LA (MLA = 280) mainly existed as sodated form (m/z = 303). ESI-MS of AOP shows emergence of new masses in the range m/z 325 to 420, and with LOP in the range m/z 315 to 400. Table 5.2 shows the results of MS studies of a time-course of AA and LA oxidation for 24, 48, and 72 hours. With increased degree of oxidation, more than 90% of AA or LA is consumed while the relative peak intensities for OPs increase. An aliquot of every time-point was also used to modify LDL and the degree of lysine modification of apoB was estimated by agarose electrophoresis. Table 5.2 shows that the increase in relative abundance of OPs correlates with the ability to modify LDL. The m/z values of the most abundant products are consistent with addition of up to 6 and 4 oxygens to AA and LA respectively, without fragmentation. Table 5.2 also includes predicted structural assignments based on each m/z. These studies suggest that by 24 h, most of the arachidonic acid has been oxidized by addition of two molecules of oxygen (i.e. addition of four oxygen atoms at two of the four double bonds) consistent with the formation of two hydroperoxides or epidioxides. This was further supported by olefinic proton number in NMR studies carried out by our collaborator Dr. Guenther Eigendorf. Subsequently there is an increase in the proportion of fatty acid molecules with lower degrees of oxygenation, presumably reflecting secondary rearrangement or decomposition of hydroperoxides, perhaps to epoxides, ketones, and hydroxides. The proportionate amount of the ion representing the 2-oxygen atom addition product increased in parallel with those of the 3- and 4-oxygen atom addition products, so no inferences could be drawn from the time course study as to which species was responsible for modifying LDL. We further studied autoxidized linoleic acid with MSMS. Isolation of the ion at m/z 335 and collision with helium yielded a main fragment in positive ion mode with m/z 225, consistent with 9- hydroxy-10,11-epoxy linoleic acid fragmenting at the methyl end of the epoxide (+Na). A minor ion at m/z 221 was found, consistent with fragmentation at the methyl end of the 13-hydroxy-10,11-epoxy- 113 octadecadienoic acid. No ions were detected that would have corresponded to fragmentation at either side of a 9-hydroperoxide. While 9-hydroxy-10,11-epoxy linoleic is capable of reacting with amino groups, it is also possible that the 13-hydroperoxide was responsible for reacting with amino groups but was not detected in the mass spectrometer due to decomposition. To determine if the reactive 2-oxygen addition product was a hydroperoxide or an epoxy-hydroxy compound, we subjected linoleic acid that had been oxidized to reduction with sodium borohydride. Before reduction, the 3 most abundant ions were m/z 335, m/z 357, and m/z 317 corresponding to M+32+Na + , M+32+2Na + -H + , and M+32+Na + -H2O respectively. These ions accounted for 70% of the total. After reduction with 100 mM NaBH4 about half of the total di-oxygenated product was reduced to a mono-oxygenated species. We conclude from this that the oxidation product contains an approximately equal mixture of hydroperoxy-octadecadienoic acid and epoxy-hydroxy-octadecenoic acid. Neither 9,10-epoxyoctadec-12Z-enoic acid nor 12,13-epoxyoctadec-9Z-enoic acid reacted with LDL (not shown). However, the keto, epoxy-hydroxy, or epoxy-keto forms would be more reactive and might react with amino groups under some conditions. 114 TABLE 5.2 Time course analysis of polyunsaturated fatty acid autoxidation products. 9 mg of neat arachidonic acid or linoleic acid were incubated for 24-72 h in an air atmosphere at 40°C. At each time point, a sample of oxidized fatty acid was analyzed by mass spectrometry. A parallel sample was dissolved in PBS, and incubated for 24 h with LDL. Agarose gel electrophoresis was then performed to assess the extent of LDL modification. Results are representative of three independent experiments. arachidonic acid m/z assignment unoxidized 24h 48h 72 h 305 +H 27.5 0.0 0.0 0.0 327 +Na 72.5 26.1 8.3 2.2 343 +Na+oxygen 0.0 6.9 9.6 9.0 345 +Na+H2O 0.0 1.0 4.6 10.0 359 +Na+2 oxygen 0.0 4.9 6.4 8.6 375 +Na+3 oxygen 0.0 8.4 19.7 28.3 391 +Na+4 oxygen 0.0 49.3 45.9 35.8 407 +Na+5 oxygen 0.0 1.0 3.7 4.3 423 +Na+6 oxygen 0.0 2.5 1.8 1.8 100% 100% 100% 100% LDL modification electrophoretic mobility (mm) 5 30 35 36 linoleic acid m/z assignment unoxidized 24h 48h 72h 303 +Na 18.7 29.7 7.5 2.8 319 +Na+oxygen 0.0 19.5 27.8 30.9 325 +2Na-H 81.3 0 0 0 335 +Na+2oxygen 0.0 42.4 53.5 55.2 351 +Na+3oxygen 0.0 5.9 8.6 8.8 367 +Na+4oxygen 0.0 2.5 2.7 2.2 100% 100% 100% 100% LDL modification electrophoretic mobility (mm) 5 11.5 12.0 12.5 115 5.2.8 OP modification of lysine and apoB generate fluorescent adducts As mentioned, we and others have reported that copper-oxidized LDL has characteristic fluorescence properties, with maximum excitation at 350-360 nm and maximum emission at 430 nm (section 1.4.4 and [505, 568]). To see if OP modification of LDL generates the same fluorescence pattern, we compared the fluorescence of AOP-LDL and LOP-LDL with that of oxLDL. In addition to nLDL control, Ac-LDL was used as a non-oxidative lysine modification control. Similar to oxLDL, AOP-LDL and LOP-LDL exhibited a more than 20-fold increase in fluorescent intensities (excitation/emission of 350/430 nm) compared to nLDL. Acetylation of LDL on the other hand, produced no increase in fluorescence intensity (Figure 5.7A). We next measured the fluorescence of adducts formed by modification of lysine methyl ester (LME) with OPs. Figure 5.7B represents the fluorescence intensities of serial dilutions of OP-LME reaction mixture depicting an increase in the fluorescence with OP-LME while there was no fluorescence associated with OP alone. Lysine methyl ester contains two potentially reactive amino groups. However, the alpha amine of lysine is 6 time less reactive compared to the epsilon amine [569] and it is believed that esterification of the carboxyl group further reduces reactivity of the alpha amine. Hence, although the alpha amine was not protected, at least 90% of adducts are anticipated to be on the epsilon amino group. Figure 5.7B shows that the increased fluorescent intensity of different concentrations of OP-LMEs while there was no increase in fluorescence of OP alone. Fluorescent scanning of LOP-LME revealed a fluorescence spectrum (Figure 5.7C) almost identical to what has been reported for oxLDL [505]. 5.2.9 Unfragmented linoleic acid autoxidation products can modify lysine methyl ester Modification of LME by LOP was chosen as the initial adduct to study because there are fewer potential products than with AOP modification LOP were used to modify lysine methyl ester (LME). The LOP-LME reaction mixture (equivalent to 10 μg of LOP) was injected into the ionization source of an Esquire Ion Trap instrument. Figure 5.8A shows a representative mass spectrum analysis of LOP- LME in positive ion mode. In addition to LME (m/z = 161) and masses known to exist in LOP, new products were found that only existed in LOP-LME reaction moisture. The m/z for these products (such 116 as 455, 457, 477) were all within the 450-510 range suggesting that adducts are formed by reaction of LME with unfragmented linoleic acid containing one to four atoms of oxygen . Figure 5.9B lists the predicted m/z of possible LOP-LME adducts with masses found in our experiments highlighted in red. 5.2.10 Putative structures of LOP-LME adducts Fruebis et al. had suggested that OPs of unsaturated fatty acids could modify proteins through a concerted reaction involving the amine group of lysine residues and reactive functions (such as hydroperoxy groups) on OPs ([501] and Section 1.4.5). According to this model, adducts with nitrogen- containing heterocycles (such as pyrrole, pyrroline, or pyridine) are produced (Figure 1.7). Fukuzawa et al. have shown that keto-oleic acid can modify amine containing amino acids without fragmentation leading to generation of hydroxylactam-containing adducts with the same fluorescent properties we observed in OP-LME [570]. To further characterize the adducts in LOP-LME, major ions unique to LOP-LME were analyzed by electrospray ionization tandem mass spectrometry (ESI-MSMS). Table 5.3 represents the putative parent ion composition for the major daughter ions of each mass. Figures 5.9A shows the MSMS plot of m/z = 455. The empiric formula for M = 454 is C25H46N2O5, and based on the reaction mechanisms proposed by Fruebis et al. and Fukuwaza et al., one can propose several putative structures for the parent adduct that could produce the observed daughter ions upon fragmentation (Figure 5.9B & C– schemes a) through e)). Loss of H2O is a common phenomenon in MSMS analysis of fatty acids and their derivatives and this explains the peak at m/z = 437 (Figure 5.9A). Fragmentation of the carboxylic acid-containing side chain adjacent to the heterocycle accounts for the 144, 312, and 295 daughter ions. The relative abundance of m/z = 142 compared to 144 can be explained by existence of an unsaturated bond within the aliphatic chain between the heterocyclic structure and the carboxyl (Figure 5.9C scheme e)) or by desaturation of the aliphatic chain upon fragmentation. Scheme c) depicts how such dehydrogenation might occur with transfer of the hydrogens to an oxo functional group (or the neighboring unsaturated bond of a pyrroline ring - not shown in the scheme) to produce a hydroxyl group (or to saturate the 117 double bond). Additionally, a charge-remote fragmentation, which is commonly seen with fatty acids and their derivatives [571], can explain how a daughter fragment can undergo dehydrogenation (Scheme d)). Lou et al. have reported conditions under which tertiary amines can undergo spontaneous dehydrogenation during fragmentation [572]. Scheme e) in Figure 5.9C is an application of this observation that provides an alternative explanation for the daughter ions of m/z = 455. Upon dehydrogenation, the daughter ion containing the cyclic moiety is already positively charged (N+) and does not require further protonation in positive ion mode resulting in m/z of 311 and 295 in positive ion mode. The fact that hydroxypyrrolidine has 4-fold higher fluorescence than pyrrole (see below) makes it an attractive candidate for the core structure of the adduct. An example of a structure with pyrrolidine core is given in scheme a). Table 5.3 shows the major daughter ions for other adducts including m/z = 457. The ion with m/z of 457 can be considered a hydrogenated form of 455. The detection of core fragments 312 and 296 and as well as the 144 fragment representing the carboxyl side chain (see figure 5.9A) is consistent with this assumption. Emergence of fragments with m/z of 391 and 341 however, could be a sign for the existence of other isobaric (m/z = 457) compounds within LOP-LME mixture. Another less likely explanation could be that saturation of the double bond is accompanied by intramolecular rearrangements leading to a different pattern of fragmentation. Species with higher m/z followed the same fragmentation pattern with generation of dehydrated pseudoparent ions. In addition they commonly generated fragments with m/z = 335 and 319 which is possibly the sodated form of core structures (m/z = 312 and 296, respectively) that were observed with m/z of 455 and 457. These results showed that adducts may contain 5 or 6 membered nitrogen heterocycles, most likely a pyrrole, pyrroline, or a derivative of. To determine if pyrrole derivatives might explain the fluorescence at 350/430 nm wavelengths, we compared fluorescence of pyrrole with that of its derivatives 3-pyrroline and 3-hydroxypyrrolidine. No fluorescence was seen with 3-pyrroline but that of 3-hydroxypyrrolidine was about 4-fold higher than that of pyrrole (data not shown). 118 TABLE 5.3 ESI-MSMS analysis and possible assignments of adducts of lysine methyl ester with autoxidized linoleic acid parent ion composition assignment daughter ions 455 LME+LA+OOH-H2O+ H + 437, 423, 378, 312, 296, 144, 142 457 LME+LA+O+H + 438, 391, 379, 341, 312, 296, 144 473 LME+LA+OOH+H + 455, 419, 413, 357, 335, 319, 144 477 LME+LA+OOH-H2O+Na + 459, 335, 317 489 LME+LA+2OOH-OH - 471, 453, 335, 317, 195 491 LME+LA+2OOH-2H2O+Na + 473, 335, 319, 211, 195 505 LME+LA+OOH+OOH+H + 487, 405, 351, 333, 317, 211, 195 119 5.2.11 Unfragmented arachidonic acid autoxidation products can modify lysine methyl ester As discussed in section 1.4.1, AA autoxidation yields far more products compared to LA. For example, isolevuglandins alone, which are found in AOP but not LOP, comprise 64 isomeric structures. To study adducts formed by AOP, we used HPLC coupled to a ZQ single quadrupole spectrometer. Initially, an acetonitrile gradient (20 to 100%) from 0.2 – 10 min was used. Figure 5.10 depicts the results of a representative run. The total ion current (TIC) of the eluent in positive ion mode (Figure 5.13 A) shows an increase in some peak intensities but no peak could be uniquely assigned to AOP-LME. We therefore obtained mass spectra of 30-second fractions of the AOP-LME chromatogram and compared these with spectra from AOP and AA controls. Figure 5.10B shows the MS analysis of the eluent (from 120 to 180 seconds) for AOP-LME, AOP, and AA. New masses with m/z of 475, 491, 493, and 509 were found only in AOP-LME. Each of these masses seemed to elute in a relatively large number of fractions. For better chromatographic resolution, longer runs were used and Figure 5.11 shows the results of single ion recordings (SIRs) of the above-mentioned masses with optimized running conditions (total run time of 35 minutes) as described in methods (Sections 2.18). Each plot depicts an overlay of SIR of one m/z for AOP and AOP-LME mixture which clearly demonstrates these masses exist only in the reaction mixture. Each mass produced at least a dozen chromatographic peaks. Retention times suggest that with addition of oxygen, adducts become more hydrophilic. Un-oxidized arachidonic acid eluted at 30.5 minutes (not shown) while adducts eluted between 3 and 23 minutes. The charge in positive ion mode can be due to protonation or addition of sodium or potassium. To determine if adducts found in positive ion mode are protonated we carried out SIRs of 473, 489, 491, and 507 in negative ion mode. Figure 5.125A shows the SIRs of m/z = 507 in negative ion mode and m/z of 509 in positive ion mode. The overall pattern of the negative ion SIR of m/z = 507 is almost identical to that of 509 in positive ion mode suggesting that m/z = 509 is protonated. In general, AOP-LME adducts where less efficiently ionized in negative mode. Comparing the ion intensities for example, negative ion mode underestimates the intensity of m = 507 by 34 fold compared to m/z = 509 in positive mode (Figure 5.125A). The efficiency of ionization in negative mode further decreased for adducts with lower m/z to a point that m/z = 473 was quite poorly ionized in ES-. To see if these adducts in positive 120 ion mode are singly charged, we looked at isotopic distribution of each m/z using a time-of-flight (TOF) analyzer. Inserts a, b, and c of Figure 5.12 show one-unit mass increases for different isotopic distributions which suggest that adducts were singly charged. We therefore concluded that in positive ion mode, these adducts accept a single proton. We then studied the elemental composition of adducts by a SYNAPT™ High Definition Mass Spectrometry™ (HDMS™) System operating in TOF mode for exact mass measurements. MassLynx software (4.1) was used for the analysis. The parameters used in analyses included concentration (parts per million (PPM)), estimated/calculated number of double bonds (double bond equivalent (DBE)), and i-FIT™ value. i-FIT™ measures the probability that the isotope distribution of a theoretical elemental composition will fit the experimental data (the lower the i-FIT value, the higher the probability of a match). Table 5.4 shows elemental composition analysis results of one representative peak for each m/z = 475, 491, 493, and 509. Considering all the criteria that are used in the analysis (such as i-FIT values) and also that adducts are most likely protonated (Figure 5.12), the most likely empirical formulae for adducts are as follows: C27H43N2O5 (for m/z = 475), C27H43N2O6 (for m/z = 491), C27H45N2O6 (for m/z = 493), and C27H45N2O7 (for m/z = 509). Analyses of other chromatographic peaks for each m/z produced similar compositional predictions. 121 Table 5.4 Elemental composition analysis of AOP-LME adducts. Elemental composition of m/z = 475, 491, 493, 509 was predicted based on exact mass measurements using a TOF analyzer. MassLynx software (4.1) was used with the following parameters: Tolerance = 10.0, parts per million (PPM), Double bond equivalent (DBE): min = -1.5, max = 50, Number of isotope peaks used for i-FIT = 3, Elements: C:10-40, H:0-100, N:0-5, O:0-10, 23Na:0-1. The most probable empirical formulae are highlighted. (i-FIT™: lower value increases the probability that the isotopic pattern of the elemental composition matches a cluster of peaks in the spectrum). Mass Calc. Mass mDa PPM DBE i-FIT Empiric Formula 475.3185 475.3172 1.3 2.7 7.5 0.7 C27 H43 N2 O5 475.3148 3.7 7.8 4.5 1.2 C25 H44 N2 O5 23Na 475.3188 -0.3 -0.6 8.5 2 C30 H44 O3 23Na 475.3212 -2.7 -5.7 11.5 3.5 C32 H43 O3 475.3231 -4.6 -9.7 -1.5 3.8 C20 H47 N2 O10 491.3142 491.3121 2.1 4.3 7.5 1.1 C27 H43 N2 O6 491.3097 4.5 9.2 4.5 1.1 C25 H44 N2 O6 23Na 491.3137 0.5 1 8.5 1.8 C30 H44 O4 23Na 491.3151 -0.9 -1.8 13.5 2.5 C31 H40 N4 23Na 491.3161 -1.9 -3.9 11.5 2.6 C32 H43 O4 491.3175 -3.3 -6.7 16.5 3.4 C33 H39 N4 493.3289 493.3278 1.1 2.2 6.5 0.9 C27 H45 N2 O6 493.3294 -0.5 -1 7.5 1.4 C30 H46 O4 23Na 493.3254 3.5 7.1 3.5 1.6 C25 H46 N2 O6 23Na 493.3307 -1.8 -3.6 12.5 2.5 C31 H42 N4 23Na 493.3318 -2.9 -5.9 10.5 2.7 C32 H45 O4 493.3331 -4.2 -8.5 15.5 4.2 C33 H41 N4 509.3217 509.3227 -1 -2 6.5 1.1 C27 H45 N2 O7 509.3243 -2.6 -5.1 7.5 1.5 C30 H46 O5 23Na 509.3208 0.9 1.8 19.5 2 C39 H41 509.3203 1.4 2.7 3.5 2.1 C25 H46 N2 O7 23Na 509.3256 -3.9 -7.7 12.5 2.8 C31 H42 N4 O 23Na 509.3267 -5 -9.8 10.5 3.1 C32 H45 O5 509.3184 3.3 6.5 16.5 3.3 C37 H42 23Na 509.3187 3 5.9 2.5 3.6 C22 H45 N4 O9 509.3168 4.9 9.6 15.5 4.2 C34 H41 N2 O2 122 5.2.12 AOP-LME adducts include isolevuglandin-modified LME We next carried out MSMS studies on AOP-LME adducts with a SYNAPT™ HDMS™ system operated in TOF. Figure 5.13A depicts the MSMS plot for m/z = 509 and 491. The MSMS plots represent the pooled data from all chromatographic peaks from each mass. The ion at m/z = 491 seems to be the dehydrated form of 509 as its fragmentation pattern was identical to that of 509. The fragmentation products included the pseudoparent ions (with loss of one or more molecules of water) and additional fragmentation products. An identical fragmentation pattern has been reported before by Brame et al. for hydroxylactam adducts of isolevuglandin-modified proteins [442]. Inserts (c) to (f) of Figure 5.16A, schematically represent the structure of these daughter ions. Exact m/z of 84.1 is consistent with it being generated from fragmentation followed by cyclization of the lysyl portion of molecule as has been observed before [442]. Similarly, m/z of 475 likely represents the dehydrated form of m/z = 493 (Figure 5.14). The fragmentation pattern of these adducts was identical to lactam-containing adducts formed by modification of lysine by isolevuglandins (Figure 5.14B & [442] ). HPLC-MSMS studies of AOP-LME showed that the daughter ions existed in all peaks of a single species (m/z = 493 for example). Figure 5.15 represents the HPLC-MSMS study of m/z = 493.3 after AOP-LME (equivalent to 10 μg of AA) was resolved by a ACQUITY UPLC system and the eluent was studied by SYNAPT™ as described in methods. Daughter ions 84.1, 233, and 144 are seen in all peaks reflecting that these fragments are present regardless of which isolevuglandin the adduct is derived from. Similar results were obtained with m/z = 509.3. We therefore conclude that the adducts are derived from different isolevuglandins (i.e. isoLG, iso[4]LG, iso[7]LG, and iso[10]LG). 5.2.13 Fluorescent peaks of AOP-LME correspond to isolevuglandin-derived adducts. To see if these adduct could have contributed to fluorescent signature observed in AOP-LME, AOP and AOP-LME were resolved with a Waters Alliance LC (on a 2.1x150 Xterra MS C18 5 µm column) coupled with 2475 multi wavelength fluorescence detector system. Fluorescent peaks of the eluent (excitation/emission wavelengths: 350/430) were found to exist between 10 to 25 minutes which clearly corresponds to the region were adducts are eluted. As an example, Figure 5.18 shows that many 123 peaks of the m/z = 491 correspond to fluorescent peaks of the eluent. Given the number of peaks for each mass and also the fact that there is a great overlap between the elution times of these masses, it is not possible to conclude that one mass contributes to the fluorescent peaks more than other masses. In fact, it is more likely that all adducts - containing a lactam or hydroxylactam as a core structure - may have more or less the same fluorescent properties. Therefore the fluorescent peaks in the eluent reflect the sum of contribution of different masses. 5.3 Summary/discussion Protein modification by lipid peroxidation products is a common event in many age-related and pathological conditions. Lipid peroxidation products are potentially genotoxic through modification of nucleic acids [573]. Additionally, it has been shown that fragmented aldehydic products can drastically change gene expression pattern especially genes regulating antioxidant, heat shock, ER stress, and nutrient deprivation responses [574]. The experiments described in this chapter yielded significant new observations regarding a particular form of amino group modification by unfragmented autoxidation products of linoleic or arachidonic acid. First, our NMR and MS studies indicate that this metal-free method for autoxidizing polyunsaturated fatty acids causes minimal fragmentation of the carbon backbone despite the fact that nearly all of the fatty acid molecules are oxidized. Second, these oxidized fatty acids are capable of forming covalent adducts with free amino groups on proteins or phosphatidylethanolamine. Third, fluorescence and MSMS studies strongly suggest the formation of a pyrrole-type adduct of the oxidation products with amino groups. Fourth, these adducts mimic the ability of copper-oxidized LDL to inhibit macrophage apoptosis whereas adducts with various other reactive oxidation products such as 4-HNE and 4-HHE had no biologic activity with regard to macrophage growth and survival. The biologic relevance of modified aminophospholipids is supported by reports showing that oxidized phospholipids (not covalently associated with apoB) are recognized by the scavenger receptor CD36 [478, 575] and are also biologically active [576]. Our results with OP-modified PE liposome 124 confirms that prosurvival activity requires a relatively small structure, namely a small molecular domain containing an amino group coupled to an oxidized fatty acid by a pyrrole-type adduct. Our studies do not define if there is a unique structural adduct that promotes growth upon modifying apoB. Autoxidized fatty acid preparations are expected to contain a large number of reactive products including lipid peroxides, epoxides, 2-alkenals, 4-hydroxy alkenals, and (in the case of arachidonic acid) other biologically active compounds such as isoprostanes and isolevuglandins [442, 448, 493, 577, 578]. Fragmented aldehydes such as HNE and HHE Are widely recognized for their ability to modify proteins and DNA. However in our hands, HNE or HHE -modified LDL could not promote survival in macrophages. Furthermore, Fruebis et al. showed that these hydroxy-aldehydes cannot account for the fluorescence observed with adducts of lysine and linoleic acid oxidation products [501]. Brame at al. have shown that autoxidation of arachidonic acid generates oxidation products that are several order of magnitude more reactive than HNE [442]. This is especially significant since HNE is stated by some experts to be one of the most reactive fatty acid peroxidation products [567]. Similarly, MS analysis showed no evidence that adehyde fragments were being generated. Additional evidence that the reactive products in our OP have not undergone fragmentation of the carbon backbone is derived from NMR studies carried out by our collaborator Dr. Guenter Eigendorf (Department of Chemistry, University of British Columbia) which indicate no detectable formation of aldehydes despite oxidation of more than 90% of the starting polyunsaturated fatty acid. Hence, it seems reasonable to conclude that hydroperoxides, endoperoxides, and/or unfragmented aldehydic products seem to be the main peroxidation products responsible for conferring prosurvival effect to modified LDL. While studying AOP-LME with HPLC-MS, we were able to find newly generated masses with mass to charge ratios and fragmentation patterns that very well fitted with isolevuglandin derived lactam and hydroxylactam-containing adducts. A pyrrole ring-containing adduct of lysine methyl ester derived from any of the isolevuglandins will have an m/z of 477 in positive ion mode (which was not found in our system) but further oxygenation of pyrrole ring produces lactam (+16, m/z = 493) and hydroxylactam (+32, m/z = 509) containing adducts which were found in our reaction mixture. This fits 125 very well with previous reports showing that, unless air is strictly excluded, all of lysyl-pyrrole adducts of levuglandins and isolevuglandins will be oxidized to lactam and hydroxylactam [442]. We also found two masses corresponding to dehydrated lactam and hydroxylactam adducts (m/z = 475 and 491, respectively). The MSMS studies and fragmentation patterns did corroborate the assumption that these are dehydrated forms. Similarly, Salomon et al. had reported that lysyl-lactam and lysyl-hydroxylactam adducts do exist as sodated [M+Na], protonated [M+H], and dehydrated [M-H2O+H] species [579]. The relative intensities of our major ions were similar to studies by Brame et al. A difference we observed was the existence of m/z = 144 daughter ion. Since this fragment existed in all peaks of both lactam and hydroxylactam adducts, it is most likely due to a core fragment as opposed to fatty acid side chains where different fragments are expected based on structural isomerism. This difference might be due to use of lysine methyl ester in our system (while Brame et al. used lysine). In the absence of the esterified methyl, fragmented lysine will easily cyclize to generate the m/z = 84.1 fragment. To positively determine the origins of the 144 fragment, parallel MSMS experiments with lysine, LME, and lysine ethyl ester (LEE) should be carried out. If still present in all three conditions, the fragment (m/z = 144) is more likely to have originated from an aliphatic side chain. Otherwise, it is expected to observe m/z of 144 and 168 for LME and LEE, respectively. Although we are not aware of published fluorescence spectra for isolevuglandin-modified LDL, our results suggest that lysyl-isolevuglandin adducts or related structures may be the source of fluorescent peaks (excitation/emission:350/430 nm) observed in our chromatographic studies. On the other hand, since linoleic acid autoxidation cannot generate isolevuglandins, it is clear that the isoLG- lysine structure is not essential for generation of fluorescence or (more importantly) for induction of growth in macrophages. Salomon et al. have shown that oxidized LDL contains isoLG-lysyl and LG- lysyl adducts [579]. It is also known that LG-modified LDL behaves similar to oxLDL in that it is recognized and taken up avidly but degraded poorly by macrophages [580]. It would be interesting to determine if LDL modified by iso[4]LG (E) [581] has prosurvival activity in macrophages. 126 It is also known that oxLDL competes for the same receptor(s) with isoLG (or LG)-modified LDL while Ac-LDL is not a competitive inhibitor [580]. This suggests that isoLG-LDL is recognized by a receptor other than SR-A. It would be interesting to study the contribution of other scavenger receptors such as CD36, LOX-1, or RAGE to isoLG-LDL uptake. One caveat to consider in interpreting such experiments however, is the possibility that isoLG is cooperating with other reactive OPs for modification of lysine and induction of macrophage growth. It has been reported that, even when in excess molar ratio, only 16 molecules of LG bind to a molecule of BSA [445]. Even though it appears that in oxLDL the isoLG-lysyl epitopes are four times more abundant than LG-lysyl epitopes [579], if similar limitations exist for apoB modification with isoLG, then it would difficult to measure/interpret partial contributions of isoLG. The finding that the prosurvival effect of OPs was almost completely abolished by pretreatment with NaBH4 suggests that this biologic activity involves formation of adducts with ketones, hydroperoxides, or aldehydes. The partial inhibition of formation of bioactive adducts by triphenylphosphine supports the conclusion that at least part of the prosurvival effect was mediated by adducts with hydroperoxides (and/or endoperoxides). The mass spectrum for LOP-LME showed only very low-intensity new ions in the region between m/z 250 and 343 where one might expect to find adducts of lysine methyl ester with fragmented aldehydes derived from linoleic acid hydroperoxide. However, there were several prominent new ions in the range m/z 434-505 that could represent lysine adducts with linoleate-derived oxidation products having an intact 18-carbon backbone. It has been proposed that linoleic acid-derived hydroperoxides [501] and keto oleic acid [570] can modify lysine residues to produce 5 membered pyrrole, lactam, or hydroxylactam (and possibly 6 membered pyridine) heterocycles with characteristic fluorescent spectra very close to what we found. The fragmentation pattern of m/z = 455 is consistent with products suggested by both mechanisms. We postulated that the existence of 142/144 doublet, and 312 daughter ions is consistent with fragmentation of hydrocarbon chain adjacent to the heterocycle (Figure 5.9B). However, a fragmentation of LME, just below N of the heterocycle is also consistent with these daughter ions. This type of fragmentation is similar to what is believed to happen in isoLG-LME adducts. To 127 positively trace the origin of these daughter ions, it is possible to compare the MSMS profile of adducts of LME with lysine and [U- 13 C] lysine. If m/z = 144 still present in MSMS analysis of LOP-lysine and LOP-[U- 13 C] lysine, it would be an indication that m/z = 144 originated from the fatty acid and contains the carboxy terminus. Although it is quite possible for autoxidation products of arachidonic acid to proceed with the same chemical reactions proposed above (and by Fruebis et al. and Fukuzawa et al.) for linoleic acid, the only major masses identified in our HPLC-MS studies of AOP-LME were shown to be isoLG-LME adducts. Considering the high reactivity of isoLGs, it is not surprising that, when present, they would be modifying amine groups long before any other reactive components. It is still likely however that other adducts are present in the mix but have not been discovered due to lower intensities. On the other hand, it is also known that higher number of double bonds (triene systems and beyond) increases the likelihood of producing bicyclic endoperoxides that are prostaglandin-like precursors [582] and can generate γ- ketoaldehydes (such as isoLGs) by an spontaneous cleavage of the endoperoxide moiety. Pryor et al. have proposed a mechanism by which hydroperoxides of a diene system (e.g. linoleic acid) may produce such bicyclic endoperoxides [582]. Zhang et al. ([583]) have also shown the production of hydroperoxide and dihydroperoxide unconjugated dienes which are capable of using mechanisms proposed by Pryor et al. to generate such bicyclic endoperoxides. This suggests that LOP may contain γ-ketoaldehydes with similar reactivity and behavior to isoLGs. However to our knowledge, there are no reports of unfragmented γ-ketoaldehyes being produced from linoleic acid autoxidation. Additionally, adducts produced from γ-linolenic acid (18:3) but not linoleic acid are recognized by anti-iso[4]LG-lysine antibody [579] which suggests that prostaglandin-like precursors do not exist in LOP. The structures we proposed here for LOP-LME adducts may contain pyrrole, lactam, or hydroxylactam core, a feature that is shared by isoLG-lysine. However, the hydrocarbon side chains in the LOP-LME adducts are on positions 2 and 5 of the heterocycle while in the isoLG-adducts, they are on positions 3 and 4. Salomon et al. have shown that the length of residues on positions 3 and 4 is the major determinant for recognition by anti-LG-lysine and anti-iso[4]LG-lysine antibodies while differences on positions 2 and 5 are not recognized [579]. 128 In BMDMs, oxLDL inhibits apoptosis by activation of PI3K/PKB axis [310], blocking ceramide generation [314], and inducing calcium oscillations and activation of calcium-dependent kinases such as eF2K [315]. Similarly, in peritoneal macrophages, oxLDL induces growth by activation of PI3K/PKB axis [308]. It would be interesting if oxLDL, AOP-LDL, LOP-LDL use the same intracellular signaling pathways for inducing macrophage survival. Our results in chapter 4 suggest that the survival signaling by oxLDL may not require internalization of the modified LDL, and it would be of interest to identify what proteins oxLDL or OP-LDL interact with on the cell surface. Since linoleic and arachidonic acids are present in significant amounts in vivo, our findings have potential importance not only for atherosclerosis but also for any other condition with increased oxidative stress. For example, advanced lipid peroxidation modified proteins have been reported to correlate significantly with periodontal disease (inflammatory disease of the gingiva and adjacent soft tissue) [584] and lipid-induced glomerular disease [585]. Numerous articles in the literature implicate fragmented aldehydes as the key injurious lipid peroxidation products in atherosclerosis [316, 586], Alzheimer‟s disease [587], as well as liver damage induced by ethanol [588], iron overload [589], prooxidants [590]. In addition, 4-HNE is reported to increase AP-1 binding to DNA and thereby to induce expression of the pro-fibrotic cytokine TGF-1beta [591]. In contrast, our results with adducts of unfragmented oxidation products show that they can activate prosurvival pathways in macrophages. Reagents that can detect these unfragmented adducts in vivo might lead to novel screening tools for diseases related to lipid peroxidation. Furthermore, strategies to modulate their generation might be of therapeutic value. 129 Figure 5.1 Lipid and protein fractions of oxLDL promote macrophage growth. Lipid and protein of native or oxLDL were extracted by the Bligh-Dyer method. The fractions were subsequently reconstituted as described in the methods. MPMs from CD-1 mice were isolated and seeded in 96 well plates at 10*10 4 cells/well density and incubated for 24 hours with indicated concentrations of oxLDL or lipoprotein fraction. Viability was then assessed with the XTT method. The graph represents the means ± SD of pooled data from three independent experiments, each done in quadruplicate. The difference between nLDL and all other conditions (at corresponding concentration) was statistically significant (P < 0.05). 130 Figure 5.2 OPs react with lysine residues. Arachidonic acid was thermally autoxidized (AOP) as described in the methods. A 300 µL aliquot was preincubated for 2 hr with 400 µL PBS containing 1 mg polylysine and then used to modify LDL (AOP/Lys-LDL). The control (AOP-LDL) was not treated with polylysine. Mouse peritoneal macrophages (MPMs) from CD-1 mice were isolated and plated in 96 well plates at 10*10 4 cells/well density and incubated with indicated concentrations of AOP-LDL or AOP/Lys-LDL for 24 hours. Viability was then assessed by the XTT method. 131 Figure 5.3 Amine modification by OPs is sufficient for growth induction. A) LDL and phosphatidylethanolamine (PE) liposomes were modified by thermally oxidized arachidonic acid and linoleic acid (AOP-LDL, AOP-PE, and LOP-PE, respectively). MPMs were isolated and seeded in 96- well plates at 10*10 4 cells/well density and incubated with indicated concentrations of LDL or the above-mentioned modifications and viability was assessed after 24 h by the XTT method. B) Phosphatidylcholine (PC) or phosphatidylethanolamine (PE) containing liposomes were modified by AOP or AOP that had been pretreated with 75mM sodium borohydride for 2 h ((AOP+ BH4)-PE). Viability was assessed as in A). Similar results were observed in two other experiments. 132 Figure 5.4 Borohydride-reducible functional groups in OPs are essential for the growth-promoting effect. Sodium borohydride (100mM) was used to reduce arachidonic acid oxidation products (AOP) before or after LDL modification ((AOP+BH4) LDL and (AOP-LDL)+BH4, respectively). Viability of MPMs was assessed by XTT method after 24 h incubation with indicated concentrations of these modifications. The graph represents the means ± SD of pooled data from three independent experiments, each done in triplicate. P < 0.05 between (AOP+BH4)-LDL and AOP-LDL or (AOP-LDL)+BH4. 133 Figure 5.5 Alkenals do not account for the growth-inducing activity of OPs. LDL was modified with 4-hydroxyhexenal or 4-hydroxynonenal (HHE-LDL and HNE-LDL, respectively) as described in methods. MPMs were isolated and plated in 96 well plates at 10*10 4 cells/well density and incubated with indicated concentrations of modified lipoproteins for 24 hours. Viability was then assessed by XTT method. Similar results were observed in 2 independent experiments. 134 Figure 5.6 ESI-MS analysis of neat and thermally oxidized linoleic and arachidonic acids. 10μg arachidonic (AA) or linoleic acid (LA) or their thermally oxidized (72 h) products (AOP and LOP, respectively) were dissolved in methanol and injected into the electrospray ionization source of an Esquire Ion Trap instrument. In positive ion mode, AA (MAA = 304) existed as protonated [MAA+H] + or sodated [MAA+Na] + ions. In this set of experiments, major masses found in AOP, in descending order of abundance, are as follows: 375 [MAA+Na+3O] + , 359 [MAA+Na+2O] + , 391 [MAA+Na+4O] + , and 343 [MAA+Na+1O] + . LA (MLA = 280) mainly existed as sodated forms (303: [MLA+Na] + or 325: [MLA+2Na- H] + ). The most abundant masses in LOP correspond to addition of up to three oxygens in sodated state (335: [MLA+Na+2O] + , 319: [MLA+Na+1O] + , and 351: [MLA+Na+3O] + . 135 Figure 5.7 OP modification of lysine and apoB generate fluorescent adducts. Fluorescent intensities of 25 μg/mL of nLDL and LDL that had been modified by copper sulfate, AOP, LOP, or acetylation (oxLDL, AOP-LDL, LOP-LDL, and Ac-LDL, respectively) (A) or increasing concentrations of OP- LME and OP (B) was measured with excitation/emission wavelengths set at 350/430. The results in A) and B) represent pooled data from at least 3 preparations and expressed as fold increase relative to nLDL. C) Fluorescent spectra of LOP and LOP-LME. 136 Figure 5.8 ESI-MS analysis of adducts formed by modification of lysine methyl ester with oxidized linoleic acid. A) Mass spectrum (100-600 range) of LOP-LME acquired on an Esquire Ion Trap instrument. m/z of 161 = LME and 300 < m/z < 370 corresponds to OPs. New masses (420 < m/z < 510) were found exclusively in LOP-LME reaction mixture. B) Predicted masses of given assignments assuming no fragmentation of acyl chain. Masses highlighted in red represent the most abundant masses found in our analyses. 137 138 Figure 5.9 MSMS analysis and putative structures of m/z = 455 in LOP-LME. A) LOP-LME was analyzed by ESI-MSMS in ES+ scan mode which reveals two pseudoparent ions ([M-H2O] + and [M- 2H2O] + ) plus major daughter ions. B) Shows the empirical formula and molecular weight of m/z = 455. Scheme a) depicts several putative structures based on the concerted reaction suggested by [501] while scheme b) represents a pathway proposed by Fukuzawa et al. ([570]) where a hydroxylactam (m/z = 469) can be produced by modification of lysine with keto-oleic acid. A derivative of such compound with a m/z = 455 with daughter ions is also depicted. C) Explains how a daughter ion with an unsaturated c-c bond can account for the 142 fragment. The upper section of scheme d) exhibits a charge-remote fragmentation commonly seen in MSMS analysis of fatty acids and their derivatives leading to creation of unsaturated bonds with a loss of hydrogen [571]. Lower section of scheme d) exhibits predicted fragmentation products and their m/z based on this model. Scheme e) exhibits the possibility of dehydrogenation and generation of C=N double bond. 1 3 9 Figure 5.10 HPLC-MS study of AOP-LME adducts. A) 10μg (or equivalent of) arachidonic acid (AA), AA oxidation products (AOP), and AOP-LME reaction mixtures were resolved by HPLC on a BEH C18 column (1.7 μm, 50 mm x 2.1 mm) and eluted with a 20-100% acetonitrile gradient. The effluent was analyzed with a ZQ single quadrupole mass spectrometer. Total ion current in positive ionization scan mode is illustrated in (A). B) The mass spectrum (200-700 range) of each sample for the 2.00 to 2.50 minutes (red marker) of chromatogram is shown. New masses with m/z of 475, 491, 493, and 509 were found only in AOP-LME reaction mixture. 1 4 0 141 Figure 5.11 m/z of 475, 491, 493, and 509 are unique to AOP-LME reaction mixture. AOP and AOP-LME (equivalent to 10μg AA) were resolved on a BEH C18 column (1.7 μm, 50 mm x 2.1 mm). An acetonitrile gradient (10- 90%) was used with a total run time of 35min, including the 100% acetonitrile flush and re-equilibration. The eluents were directed to a ZQ single quadrupole mass spectrometer. Single ion recordings in ES+ mode for m/z of 475, 491, 493, and 509 were compared between AOP and AOP-LME. 142 Figure 5.12 AOP-LME adducts are singly charged and protonated species. AOP-LME (equivalent of 10μg of AA) was resolved on a BEH column and directed to a ZQ single quadrupole mass spectrometer. Panels represent the single ion recordings of A) 507 (ES-) and 509 (ES+), B) 491 (ES-) and 493 (ES+), and C) 489 (ES-) and 491 (ES+). High resolution isotopic distribution for m/z of 509, 493, and 491 are shown in a), b), and c), respectively. 1 4 3 Figure 5.13 m/z = 509 is an isolevuglandin-derived hydroxylactam-containing adduct. A) MSMS analysis of AOP-LME (equivalent of 10μg of AA) acquired on a SYNAPT system operating in TOF mode for m/z = 509 and 491. Plots represent the pooled data from all the chromatographic peaks of each mass. Intensities are normalized to the highest peak (226 and 1.42e3 for m/z = 491 and 509, respectively). B) Molecular weight, empirical formula and structures of parent ions (491 and 509) and their fragmentation products. Iso[4]LG derived adduct is used as an example. 1 4 4 1 4 5 Figure 5.14 m/z = 493 is an isolevuglandin-derived lactam-containing adduct. A) MSMS analysis of AOP-LME (equivalent of 10μg of AA) acquired on a SYNAPT system operating in TOF mode for m/z = 493 and 475. Plots represent the pooled data from all the chromatographic peaks of each mass. Intensities are normalized to the highest peak (884 and 119 for m/z = 493 and 475, respectively). B) Molecular weight, empirical formula and structures of parent ions (495 and 493) and their fragmentation products. Iso[4]LG derived adduct is used as an example. 1 4 6 147 Figure 5.15 HPLC-MSMS analysis of m/z = 493. AOP-LME (equivalent of 10μg of AA) was resolved on a BEH column with acetonitrile gradient (10-90%) on an ACQUITY UPLC system coupled to SYNAPT™ High Definition Mass Spectrometry™ (HDMS™) System operating in TOF mode. Panels represent the total ion current for MSMS study of m/z = 493.3 (lowest panel) and chromatograms for m/z of 84.1, 332, and 144 daughter ions. In the lowest panel, the numbers below retention times (in red) represent the most abundant fragment within that peak. 148 Figure 5.16 Fluorescent peaks of AOP-LME correspond to m/z of 491. AOP or AOP-LME (equivalent of 10μg of AA) was resolved by a Waters Alliance LC with an Xterra 5µm C18 column coupled with a PDA and 2475 multi wavelength Fluorescence Detector System. Acetonitrile gradient (10-100%) was used as described in methods. The fluorescence of the eluent was measured with excitation/emission set at 350/430 nm (upper panel). In parallel, similar chromatographic set up was coupled to a PDA and the ZQ single quadrupole mass spectrometer. Single ion recordings for adducts was carried out with the lower panel representing that of m/z = 491in ES+. PDA readings at 205 nm (for AA) and 234 nm (for conjugated dienes) were used to align the chromatograms. In both panels, the numbers associated with the peaks represent retention times. 149 6. Summary This thesis focuses on oxidized LDL and its effects on macrophages. The first project dealt with the mechanisms by which oxLDL induces production and secretion of VEGF. This project focused on oxLDL-mediated VEGF secretion at three different levels: 1) the ligand: what moiety of oxLDL triggers this event? 2) the receptor: are scavenger receptors involved? and 3) the intracellular events: what signaling pathways are required? Our results with nLDL, acetylated LDL, and LDL that had been modified by oxidation with copper or ferrous ions or by exposure to autoxidation products of arachidonic acid showed that oxidative modification of either its lipid or protein component can induce VEGF expression. Higher degrees of oxidation of LDL conferred higher potency to induce VEGF. Studies from knockout animals revealed that LOX-1, SR-A, and CD36, three of major scavenger receptors, are not required for this effect. Additionally, we found that uptake of oxLDL may not be essential for VEGF induction as scavenger receptor-deficient macrophages still secreted VEGF efficiently in spite of an 80% decrease in oxLDL uptake. Signaling studies revealed that mitogen-activated protein kinase (MAPK) pathway is not important for oxLDL-mediated VEGF secretion in macrophages even though it is important for baseline levels of VEGF secretion. PI3K pathway, on the other hand, was shown to be essential for VEGF induction by oxLDL. We also show that an atypical protein kinase C isoform (PKCδ) is required for VEGF secretion. Although we did not directly demonstrate this, we believe that PKCδ activation is downstream of PI3K, possibly through phosphoinositide-dependent kinase-1 (PDK-1) activation. We also, for the first time, show that oxLDL increases the activity of PKCδ in macrophages. The importance of finding a pathway essential for oxLDL-mediated VEGF secretion lies in the Janus-like effects of this growth factor in atherogenesis. VEGF on one hand is potentially atherogenic because it can increase vascular permeability, recruit inflammatory cells, and promote angiogenesis within the plaque, which could drive plaque growth. On the other hand, VEGF has therapeutic potential by promoting the establishment of collateral circulation to compensate for an occluded segment in an artery. Interestingly, not only does oxLDL increase the intra-plaque VEGF, but it also antagonizes some 150 of potential effects of this growth factor, tipping the balance towards an atherogenic profile. Therefore, targeting molecular mechanisms that link oxLDL to VEGF expression in lesions (as opposed to a non- selective suppression of VEGF) can restore the more favorable balance. Our laboratory has been studying the prosurvival and mitogenic properties of oxLDL towards macrophages, as this may be a mechanism by which macrophage populations in the intima are expanded. We have shown that the PI3K-PKB pathway as well as EF2K activation are required, but to date we have not defined upstream signaling events or if a cell surface receptor is involved. I took advantage of the availability of transgenic mice lacking one or more scavenger receptors to see which ones, if any, are required for the antiapoptotic effects of oxLDL in bone marrow derived macrophages. My results indicated that none of these receptors (SR-A and CD36, LOX-1, TLR4, CD14, or FcγRIIb) are essential for activating the oxLDL prosurvival pathway. Furthermore, the anti-apoptotic effect was not affected by the rate of oxLDL uptake. Three major receptors that remain to be explored in this regard are SR-PSOX, SR-BI, and RAGE. LDL modified by oxidation products of unsaturated fatty acids was developed in our laboratory to facilitate the study of oxidative protein modification of LDL. The final project I undertook was centered on characterization of this type of modification especially with respect to its prosurvival potential. These studies showed that both protein and lipid moieties of oxLDL are capable of promoting survival and growth in macrophages. It appears that oxidation products of unsaturated fatty acids can derivatize amine groups (of whether the lysine residues in apoB or phosphatidylethanolamine) resulting in promotion of growth and survival. Studies with borohydride and triphenylphosphine show that the reactive functions required for these modifications include aldehydes, ketons, hydroperoxides, or endoperoxides. LDL modified with alkenals (eg. 4-HNE) derived from fragmentation of fatty acid hydroperoxides showed no prosurvival activity. MS analysis showed that the OPs we generated to modify LDL were largely unfragmented even though more than 80% of the fatty acid molecules were peroxidized. To study the structure of the adducts formed when these OP react with amines, we mixed oxidation products with lysine methyl ester 151 (LME). Mass spectrometry showed that LOP-LME and AOP-LME both contain masses that correspond to formation of a covalent adduct with an unfragmented oxygenated fatty acid. In the case of AOP-LME, some fragments generated by MSMS were consistent with isolevuglandin adducts. Another aspect of this project studied the fluorescent properties of these adducts. Similar to oxLDL (and modification of any protein by lipid peroxidation product) a specific fluorescent pattern with maximum emission and excitation of 350/430 nm was observed with LDL and LME modification by AOP and LOP. It also appears that isolevuglandin-lysyl adducts could have contributed to this flluorescent pattern within AOP-LME mix. This project has many potential applications in the field of atherosclerosis and beyond. Firstly, it characterizes a metal-free method for oxidative modification of LDL. Secondly, since arachidonic acid and linoleic acid are the two most abundant unsaturated fatty acids in vivo, any condition with increased oxidative stress could lead to the generation of similar adducts. These adducts have therefore the potential to become markers of oxidative damage. Unlike many current markers of oxidative damage, which are relatively unstable and have a short half-life, these adducts are quite stable. Analogous to diabetes and glycated hemoglobin (HbA1C), a long-term (months) marker of glucose control in diabetic patients, these adducts may be used to give information on the cumulative effect of oxidative damage over a longer period of time. This by itself can have many other applications such as defining a biological age (as opposed to a chronological age) or monitoring cases with chronic oxidative injury, such as smoking, and create more incentive for behavior modification. Thirdly, our results highlight the fact that such adducts may not merely be markers of oxidative damage, but they may in fact contribute to disease pathogenesis by virtue of their biological activities. Therefore developing strategies that isolate or neutralize these adducts or block their recognition by PRRs that mediate detrimental effects have therapeutic applications. 152 7. References [1] Tegos TJ, Kalodiki E, Sabetai MM, Nicolaides AN. The genesis of atherosclerosis and risk factors: a review. Angiology. 2001 Feb;52(2):89-98. [2] The global burden of disease : 2004 update Geneva: World Health Organization.; 2008. [3] The Growing Burden of Heart Disease and Stroke in Canada Ottawa, Canada: Heart and Stroke Foundation of Canada; 2003. [4] Lloyd-Jones D, Adams R, Carnethon M, De Simone G, Ferguson TB, Flegal K, et al. Heart disease and stroke statistics -- 2009 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation. 2009 Jan 27;119(3):480-6. [5] Stary HC, Chandler AB, Dinsmore RE, Fuster V, Glagov S, Insull W, Jr., et al. A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation. 1995 Sep 1;92(5):1355-74. [6] Stary HC, Chandler AB, Glagov S, Guyton JR, Insull W, Jr., Rosenfeld ME, et al. A definition of initial, fatty streak, and intermediate lesions of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation. 1994 May;89(5):2462-78. [7] Stary HC, Blankenhorn DH, Chandler AB, Glagov S, Insull W, Jr., Richardson M, et al. A definition of the intima of human arteries and of its atherosclerosis-prone regions. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation. 1992 Jan;85(1):391-405. [8] Munro JM, van der Walt JD, Munro CS, Chalmers JA, Cox EL. An immunohistochemical analysis of human aortic fatty streaks. Human pathology. 1987 Apr;18(4):375-80. [9] Glass CK, Witztum JL. Atherosclerosis. the road ahead. Cell. 2001 Feb 23;104(4):503-16. [10] Steinberg D. Thematic review series: the pathogenesis of atherosclerosis. An interpretive history of the cholesterol controversy: part I. Journal of lipid research. 2004 Sep;45(9):1583-93. [11] Gofman JW, Lindgren FT, Elliott H. Ultracentrifugal studies of lipoproteins of human serum. The Journal of biological chemistry. 1949 Jun;179(2):973-9. [12] Havel RJ, Eder HA, Bragdon JH. The distribution and chemical composition of ultracentrifugally separated lipoproteins in human serum. The Journal of clinical investigation. 1955 Sep;34(9):1345-53. [13] Lees RS, Hatch FT. Sharper separation of lipoprotein species by paper electrophoresis in albumin-containing buffer. The Journal of laboratory and clinical medicine. 1963 Mar;61:518- 28. [14] Vance DE, Vance JE. Biochemistry of Lipids, Lipoproteins and membranes, 4th ed. New Comprehensive Biochemistry. 2002: Elsevier B.V. 607. 2002. [15] Lewis GF, Rader DJ. New insights into the regulation of HDL metabolism and reverse cholesterol transport. Circulation research. 2005 Jun 24;96(12):1221-32. [16] Ebara T, Endo Y, Yoshiike S, Tsuji M, Taguchi S, Murase T, et al. A 60-y-old chylomicronemia patient homozygous for missense mutation (G188E) in the lipoprotein lipase gene showed no accelerated atherosclerosis. Clinica chimica acta; international journal of clinical chemistry. 2007 Nov-Dec;386(1-2):100-4. [17] Ebara T, Okubo M, Horinishi A, Adachi M, Murase T, Hirano T. No evidence of accelerated atherosclerosis in a 66-yr-old chylomicronemia patient homozygous for the nonsense mutation (Tyr61-->stop) in the lipoprotein lipase gene. Atherosclerosis. 2001 Dec;159(2):375-9. [18] Ebara T, Ramakrishnan R, Steiner G, Shachter NS. Chylomicronemia due to apolipoprotein CIII overexpression in apolipoprotein E-null mice. Apolipoprotein CIII-induced hypertriglyceridemia is not mediated by effects on apolipoprotein E. The Journal of clinical investigation. 1997 Jun 1;99(11):2672-81. [19] Nordestgaard BG, Zilversmit DB. Large lipoproteins are excluded from the arterial wall in diabetic cholesterol-fed rabbits. Journal of lipid research. 1988 Nov;29(11):1491-500. 153 [20] Mahley RW, Innerarity TL, Rall SC, Jr., Weisgraber KH. Plasma lipoproteins: apolipoprotein structure and function. Journal of lipid research. 1984 Dec 1;25(12):1277-94. [21] Goldstein JL, Brown MS. Lipoprotein receptors and the control of plasma LDL cholesterol levels. European heart journal. 1992 Jul;13 Suppl B:34-6. [22] Keys A, Aravanis C, Blackburn HW, Van Buchem FS, Buzina R, Djordjevic BD, et al. Epidemiological studies related to coronary heart disease: characteristics of men aged 40-59 in seven countries. Acta medica Scandinavica. 1966;460:1-392. [23] Keys A, Menotti A, Aravanis C, Blackburn H, Djordevic BS, Buzina R, et al. The seven countries study: 2,289 deaths in 15 years. Preventive medicine. 1984 Mar;13(2):141-54. [24] Kannel WB, Dawber TR, Kagan A, Revotskie N, Stokes J, 3rd. Factors of risk in the development of coronary heart disease--six year follow-up experience. The Framingham Study. Annals of internal medicine. 1961 Jul;55:33-50. [25] Wilson PW, Garrison RJ, Castelli WP, Feinleib M, McNamara PM, Kannel WB. Prevalence of coronary heart disease in the Framingham Offspring Study: role of lipoprotein cholesterols. The American journal of cardiology. 1980 Oct;46(4):649-54. [26] Steinberg D. Thematic review series: the pathogenesis of atherosclerosis: an interpretive history of the cholesterol controversy, part III: mechanistically defining the role of hyperlipidemia. Journal of lipid research. 2005 Oct;46(10):2037-51. [27] The Lipid Research Clinics Coronary Primary Prevention Trial results. II. The relationship of reduction in incidence of coronary heart disease to cholesterol lowering. Jama. 1984 Jan 20;251(3):365-74. [28] The Lipid Research Clinics Coronary Primary Prevention Trial results. I. Reduction in incidence of coronary heart disease. Jama. 1984 Jan 20;251(3):351-64. [29] Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). Lancet. 1994 Nov 19;344(8934):1383-9. [30] Baigent C, Keech A, Kearney PM, Blackwell L, Buck G, Pollicino C, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet. 2005 Oct 8;366(9493):1267-78. [31] Hollander W, Colombo MA, Kirkpatrick B, Paddock J. Soluble proteins in the human atherosclerotic plaque. With spectral reference to immunoglobulins, C3-complement component, alpha 1-antitrypsin and alpha 2-macroglobulin. Atherosclerosis. 1979 Dec;34(4):391-405. [32] Gutstein WH, Schneck DJ. In vitro boundary layer studies of blood flow in branched tubes. Journal of atherosclerosis research. 1967 May-Jun;7(3):295-9. [33] Scharfstein H, Gutstein WH, Lewis L. Changes of Boundary Layer Flow in Model Systems: Implications for Initiation of Endothelial Injury. Circulation research. 1963 Dec;13:580-4. [34] Ross R, Glomset JA. The pathogenesis of atherosclerosis (second of two parts). The New England journal of medicine. 1976 Aug 19;295(8):420-5. [35] Ross R, Glomset JA. The pathogenesis of atherosclerosis (first of two parts). The New England journal of medicine. 1976 Aug 12;295(7):369-77. [36] Asakura T, Karino T. Flow patterns and spatial distribution of atherosclerotic lesions in human coronary arteries. Circulation research. 1990 Apr;66(4):1045-66. [37] Huo Y, Wischgoll T, Kassab GS. Flow patterns in three-dimensional porcine epicardial coronary arterial tree. American journal of physiology. 2007 Nov;293(5):H2959-70. [38] Hahn C, Schwartz MA. Mechanotransduction in vascular physiology and atherogenesis. Nature reviews. 2009 Jan;10(1):53-62. [39] Brooks AR, Lelkes PI, Rubanyi GM. Gene expression profiling of vascular endothelial cells exposed to fluid mechanical forces: relevance for focal susceptibility to atherosclerosis. Endothelium. 2004 Jan-Feb;11(1):45-57. [40] Passerini AG, Polacek DC, Shi C, Francesco NM, Manduchi E, Grant GR, et al. Coexisting proinflammatory and antioxidative endothelial transcription profiles in a disturbed flow region of the adult porcine aorta. Proceedings of the National Academy of Sciences of the United States of America. 2004 Feb 24;101(8):2482-7. 154 [41] Cheng C, van Haperen R, de Waard M, van Damme LC, Tempel D, Hanemaaijer L, et al. Shear stress affects the intracellular distribution of eNOS: direct demonstration by a novel in vivo technique. Blood. 2005 Dec 1;106(12):3691-8. [42] Ziegler T, Bouzourene K, Harrison VJ, Brunner HR, Hayoz D. Influence of oscillatory and unidirectional flow environments on the expression of endothelin and nitric oxide synthase in cultured endothelial cells. Arteriosclerosis, thrombosis, and vascular biology. 1998 May;18(5):686-92. [43] Shyy YJ, Hsieh HJ, Usami S, Chien S. Fluid shear stress induces a biphasic response of human monocyte chemotactic protein 1 gene expression in vascular endothelium. Proceedings of the National Academy of Sciences of the United States of America. 1994 May 24;91(11):4678-82. [44] Tzima E, Del Pozo MA, Kiosses WB, Mohamed SA, Li S, Chien S, et al. Activation of Rac1 by shear stress in endothelial cells mediates both cytoskeletal reorganization and effects on gene expression. The EMBO journal. 2002 Dec 16;21(24):6791-800. [45] Hajra L, Evans AI, Chen M, Hyduk SJ, Collins T, Cybulsky MI. The NF-kappa B signal transduction pathway in aortic endothelial cells is primed for activation in regions predisposed to atherosclerotic lesion formation. Proceedings of the National Academy of Sciences of the United States of America. 2000 Aug 1;97(16):9052-7. [46] Hwang J, Ing MH, Salazar A, Lassegue B, Griendling K, Navab M, et al. Pulsatile versus oscillatory shear stress regulates NADPH oxidase subunit expression: implication for native LDL oxidation. Circulation research. 2003 Dec 12;93(12):1225-32. [47] McNally JS, Davis ME, Giddens DP, Saha A, Hwang J, Dikalov S, et al. Role of xanthine oxidoreductase and NAD(P)H oxidase in endothelial superoxide production in response to oscillatory shear stress. American journal of physiology. 2003 Dec;285(6):H2290-7. [48] Liu Y, Chen BP, Lu M, Zhu Y, Stemerman MB, Chien S, et al. Shear stress activation of SREBP1 in endothelial cells is mediated by integrins. Arteriosclerosis, thrombosis, and vascular biology. 2002 Jan;22(1):76-81. [49] Buchanan JR, Jr., Kleinstreuer C, Truskey GA, Lei M. Relation between non-uniform hemodynamics and sites of altered permeability and lesion growth at the rabbit aorto-celiac junction. Atherosclerosis. 1999 Mar;143(1):27-40. [50] Himburg HA, Grzybowski DM, Hazel AL, LaMack JA, Li XM, Friedman MH. Spatial comparison between wall shear stress measures and porcine arterial endothelial permeability. American journal of physiology. 2004 May;286(5):H1916-22. [51] Berceli SA, Warty VS, Sheppeck RA, Mandarino WA, Tanksale SK, Borovetz HS. Hemodynamics and low density lipoprotein metabolism. Rates of low density lipoprotein incorporation and degradation along medial and lateral walls of the rabbit aorto-iliac bifurcation. Arteriosclerosis (Dallas, Tex. 1990 Sep-Oct;10(5):686-94. [52] Williams KJ, Tabas I. The response-to-retention hypothesis of atherogenesis reinforced. Current opinion in lipidology. 1998 Oct;9(5):471-4. [53] Williams KJ, Tabas I. The response-to-retention hypothesis of early atherogenesis. Arteriosclerosis, thrombosis, and vascular biology. 1995 May;15(5):551-61. [54] Galis ZS, Alavi MZ, Moore S. Co-localization of aortic apolipoprotein B and chondroitin sulfate in an injury model of atherosclerosis. The American journal of pathology. 1993 May;142(5):1432-8. [55] Cardoso LE, Mourao PA. Glycosaminoglycan fractions from human arteries presenting diverse susceptibilities to atherosclerosis have different binding affinities to plasma LDL. Arterioscler Thromb. 1994 Jan;14(1):115-24. [56] Williams KJ. Arterial wall chondroitin sulfate proteoglycans: diverse molecules with distinct roles in lipoprotein retention and atherogenesis. Current opinion in lipidology. 2001 Oct;12(5):477-87. [57] Nakashima Y, Fujii H, Sumiyoshi S, Wight TN, Sueishi K. Early human atherosclerosis: accumulation of lipid and proteoglycans in intimal thickenings followed by macrophage infiltration. Arteriosclerosis, thrombosis, and vascular biology. 2007 May;27(5):1159-65. 155 [58] Boren J, Olin K, Lee I, Chait A, Wight TN, Innerarity TL. Identification of the principal proteoglycan-binding site in LDL. A single-point mutation in apo-B100 severely affects proteoglycan interaction without affecting LDL receptor binding. The Journal of clinical investigation. 1998 Jun 15;101(12):2658-64. [59] Flood C, Gustafsson M, Richardson PE, Harvey SC, Segrest JP, Boren J. Identification of the proteoglycan binding site in apolipoprotein B48. The Journal of biological chemistry. 2002 Aug 30;277(35):32228-33. [60] Skalen K, Gustafsson M, Rydberg EK, Hulten LM, Wiklund O, Innerarity TL, et al. Subendothelial retention of atherogenic lipoproteins in early atherosclerosis. Nature. 2002 Jun 13;417(6890):750-4. [61] Tabas I. Secretory sphingomyelinase. Chemistry and physics of lipids. 1999 Nov;102(1-2):123- 30. [62] Oorni K, Pentikainen MO, Ala-Korpela M, Kovanen PT. Aggregation, fusion, and vesicle formation of modified low density lipoprotein particles: molecular mechanisms and effects on matrix interactions. Journal of lipid research. 2000 Nov;41(11):1703-14. [63] Brown MS, Goldstein JL. Cholesterol feedback: from Schoenheimer's bottle to Scap's MELADL. Journal of lipid research. 2008 Oct 29. [64] Brown MS, Goldstein JL. Receptor-mediated control of cholesterol metabolism. Science (New York, NY. 1976 Jan 16;191(4223):150-4. [65] Kar S, Day AJ. Composition and metabolism of lipid in macrophages from normally fed and cholesterol-fed rabbits. Experimental and molecular pathology. 1978 Feb;28(1):65-75. [66] Wurster NB, Zilversmit DB. The role of phagocytosis in the development of atherosclerotic lesions in the rabbit. Atherosclerosis. 1971 Nov-Dec;14(3):309-22. [67] Goldstein JL, Ho YK, Basu SK, Brown MS. Binding site on macrophages that mediates uptake and degradation of acetylated low density lipoprotein, producing massive cholesterol deposition. Proceedings of the National Academy of Sciences of the United States of America. 1979 Jan;76(1):333-7. [68] Brown MS, Goldstein JL, Krieger M, Ho YK, Anderson RG. Reversible accumulation of cholesteryl esters in macrophages incubated with acetylated lipoproteins. The Journal of cell biology. 1979 Sep;82(3):597-613. [69] Henriksen T, Evensen SA, Carlander B. Injury to human endothelial cells in culture induced by low density lipoproteins. Scandinavian journal of clinical and laboratory investigation. 1979 Jun;39(4):361-8. [70] Hessler JR, Robertson AL, Jr., Chisolm GM, 3rd. LDL-induced cytotoxicity and its inhibition by HDL in human vascular smooth muscle and endothelial cells in culture. Atherosclerosis. 1979 Mar;32(3):213-29. [71] Henriksen T, Mahoney EM, Steinberg D. Enhanced macrophage degradation of low density lipoprotein previously incubated with cultured endothelial cells: recognition by receptors for acetylated low density lipoproteins. Proceedings of the National Academy of Sciences of the United States of America. 1981 Oct;78(10):6499-503. [72] Steinbrecher UP, Parthasarathy S, Leake DS, Witztum JL, Steinberg D. Modification of low density lipoprotein by endothelial cells involves lipid peroxidation and degradation of low density lipoprotein phospholipids. Proceedings of the National Academy of Sciences of the United States of America. 1984 Jun;81(12):3883-7. [73] Khoo JC, Miller E, McLoughlin P, Steinberg D. Enhanced macrophage uptake of low density lipoprotein after self-aggregation. Arteriosclerosis (Dallas, Tex. 1988 Jul-Aug;8(4):348-58. [74] Xu XX, Tabas I. Sphingomyelinase enhances low density lipoprotein uptake and ability to induce cholesteryl ester accumulation in macrophages. The Journal of biological chemistry. 1991 Dec 25;266(36):24849-58. [75] Brown BE, Dean RT, Davies MJ. Glycation of low-density lipoproteins by methylglyoxal and glycolaldehyde gives rise to the in vitro formation of lipid-laden cells. Diabetologia. 2005 Feb;48(2):361-9. 156 [76] Bhakdi S, Dorweiler B, Kirchmann R, Torzewski J, Weise E, Tranum-Jensen J, et al. On the pathogenesis of atherosclerosis: enzymatic transformation of human low density lipoprotein to an atherogenic moiety. The Journal of experimental medicine. 1995 Dec 1;182(6):1959-71. [77] Kapinsky M, Torzewski M, Buchler C, Duong CQ, Rothe G, Schmitz G. Enzymatically degraded LDL preferentially binds to CD14(high) CD16(+) monocytes and induces foam cell formation mediated only in part by the class B scavenger-receptor CD36. Arteriosclerosis, thrombosis, and vascular biology. 2001 Jun;21(6):1004-10. [78] Khoo JC, Miller E, Pio F, Steinberg D, Witztum JL. Monoclonal antibodies against LDL further enhance macrophage uptake of LDL aggregates. Arterioscler Thromb. 1992 Nov;12(11):1258- 66. [79] Steinberg D, Parthasarathy S, Carew TE, Khoo JC, Witztum JL. Beyond cholesterol. Modifications of low-density lipoprotein that increase its atherogenicity. The New England journal of medicine. 1989 Apr 6;320(14):915-24. [80] Navab M, Imes SS, Hama SY, Hough GP, Ross LA, Bork RW, et al. Monocyte transmigration induced by modification of low density lipoprotein in cocultures of human aortic wall cells is due to induction of monocyte chemotactic protein 1 synthesis and is abolished by high density lipoprotein. The Journal of clinical investigation. 1991 Dec;88(6):2039-46. [81] Navab M, Liao F, Hough GP, Ross LA, Van Lenten BJ, Rajavashisth TB, et al. Interaction of monocytes with cocultures of human aortic wall cells involves interleukins 1 and 6 with marked increases in connexin43 message. The Journal of clinical investigation. 1991 May;87(5):1763- 72. [82] Yla-Herttuala S, Palinski W, Rosenfeld ME, Parthasarathy S, Carew TE, Butler S, et al. Evidence for the presence of oxidatively modified low density lipoprotein in atherosclerotic lesions of rabbit and man. The Journal of clinical investigation. 1989 Oct;84(4):1086-95. [83] Palinski W, Rosenfeld ME, Yla-Herttuala S, Gurtner GC, Socher SS, Butler SW, et al. Low density lipoprotein undergoes oxidative modification in vivo. Proceedings of the National Academy of Sciences of the United States of America. 1989 Feb;86(4):1372-6. [84] Palinski W, Ord VA, Plump AS, Breslow JL, Steinberg D, Witztum JL. ApoE-deficient mice are a model of lipoprotein oxidation in atherogenesis. Demonstration of oxidation-specific epitopes in lesions and high titers of autoantibodies to malondialdehyde-lysine in serum. Arterioscler Thromb. 1994 Apr;14(4):605-16. [85] Salonen JT, Yla-Herttuala S, Yamamoto R, Butler S, Korpela H, Salonen R, et al. Autoantibody against oxidised LDL and progression of carotid atherosclerosis. Lancet. 1992 Apr 11;339(8798):883-7. [86] Palinski W, Tangirala RK, Miller E, Young SG, Witztum JL. Increased autoantibody titers against epitopes of oxidized LDL in LDL receptor-deficient mice with increased atherosclerosis. Arteriosclerosis, thrombosis, and vascular biology. 1995 Oct;15(10):1569-76. [87] Palinski W, Horkko S, Miller E, Steinbrecher UP, Powell HC, Curtiss LK, et al. Cloning of monoclonal autoantibodies to epitopes of oxidized lipoproteins from apolipoprotein E-deficient mice. Demonstration of epitopes of oxidized low density lipoprotein in human plasma. The Journal of clinical investigation. 1996 Aug 1;98(3):800-14. [88] Napoli C, D'Armiento FP, Mancini FP, Postiglione A, Witztum JL, Palumbo G, et al. Fatty streak formation occurs in human fetal aortas and is greatly enhanced by maternal hypercholesterolemia. Intimal accumulation of low density lipoprotein and its oxidation precede monocyte recruitment into early atherosclerotic lesions. The Journal of clinical investigation. 1997 Dec 1;100(11):2680-90. [89] Rajavashisth TB, Andalibi A, Territo MC, Berliner JA, Navab M, Fogelman AM, et al. Induction of endothelial cell expression of granulocyte and macrophage colony-stimulating factors by modified low-density lipoproteins. Nature. 1990 Mar 15;344(6263):254-7. [90] Cushing SD, Berliner JA, Valente AJ, Territo MC, Navab M, Parhami F, et al. Minimally modified low density lipoprotein induces monocyte chemotactic protein 1 in human endothelial cells and smooth muscle cells. Proceedings of the National Academy of Sciences of the United States of America. 1990 Jul;87(13):5134-8. 157 [91] Smith JD, Trogan E, Ginsberg M, Grigaux C, Tian J, Miyata M. Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proceedings of the National Academy of Sciences of the United States of America. 1995 Aug 29;92(18):8264-8. [92] Boring L, Gosling J, Cleary M, Charo IF. Decreased lesion formation in CCR2-/- mice reveals a role for chemokines in the initiation of atherosclerosis. Nature. 1998 Aug 27;394(6696):894-7. [93] Gosling J, Slaymaker S, Gu L, Tseng S, Zlot CH, Young SG, et al. MCP-1 deficiency reduces susceptibility to atherosclerosis in mice that overexpress human apolipoprotein B. The Journal of clinical investigation. 1999 Mar;103(6):773-8. [94] Quinn MT, Parthasarathy S, Fong LG, Steinberg D. Oxidatively modified low density lipoproteins: a potential role in recruitment and retention of monocyte/macrophages during atherogenesis. Proceedings of the National Academy of Sciences of the United States of America. 1987 May;84(9):2995-8. [95] McMurray HF, Parthasarathy S, Steinberg D. Oxidatively modified low density lipoprotein is a chemoattractant for human T lymphocytes. The Journal of clinical investigation. 1993 Aug;92(2):1004-8. [96] Khan BV, Parthasarathy SS, Alexander RW, Medford RM. Modified low density lipoprotein and its constituents augment cytokine-activated vascular cell adhesion molecule-1 gene expression in human vascular endothelial cells. The Journal of clinical investigation. 1995 Mar;95(3):1262-70. [97] Amberger A, Maczek C, Jurgens G, Michaelis D, Schett G, Trieb K, et al. Co-expression of ICAM-1, VCAM-1, ELAM-1 and Hsp60 in human arterial and venous endothelial cells in response to cytokines and oxidized low-density lipoproteins. Cell stress & chaperones. 1997 Jun;2(2):94-103. [98] Cybulsky MI, Iiyama K, Li H, Zhu S, Chen M, Iiyama M, et al. A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. The Journal of clinical investigation. 2001 May;107(10):1255-62. [99] Dong ZM, Chapman SM, Brown AA, Frenette PS, Hynes RO, Wagner DD. The combined role of P- and E-selectins in atherosclerosis. The Journal of clinical investigation. 1998 Jul 1;102(1):145-52. [100] Keiper T, Al-Fakhri N, Chavakis E, Athanasopoulos AN, Isermann B, Herzog S, et al. The role of junctional adhesion molecule-C (JAM-C) in oxidized LDL-mediated leukocyte recruitment. Faseb J. 2005 Dec;19(14):2078-80. [101] Miller YI, Viriyakosol S, Worrall DS, Boullier A, Butler S, Witztum JL. Toll-like receptor 4- dependent and -independent cytokine secretion induced by minimally oxidized low-density lipoprotein in macrophages. Arteriosclerosis, thrombosis, and vascular biology. 2005 Jun;25(6):1213-9. [102] Terkeltaub R, Banka CL, Solan J, Santoro D, Brand K, Curtiss LK. Oxidized LDL induces monocytic cell expression of interleukin-8, a chemokine with T-lymphocyte chemotactic activity. Arterioscler Thromb. 1994 Jan;14(1):47-53. [103] Bae YS, Lee JH, Choi SH, Kim S, Almazan F, Witztum JL, et al. Macrophages generate reactive oxygen species in response to minimally oxidized low-density lipoprotein: toll-like receptor 4- and spleen tyrosine kinase-dependent activation of NADPH oxidase 2. Circulation research. 2009 Jan 30;104(2):210-8, 21p following 8. [104] Kugiyama K, Kerns SA, Morrisett JD, Roberts R, Henry PD. Impairment of endothelium- dependent arterial relaxation by lysolecithin in modified low-density lipoproteins. Nature. 1990 Mar 8;344(6262):160-2. [105] Rajavashisth TB, Liao JK, Galis ZS, Tripathi S, Laufs U, Tripathi J, et al. Inflammatory cytokines and oxidized low density lipoproteins increase endothelial cell expression of membrane type 1-matrix metalloproteinase. The Journal of biological chemistry. 1999 Apr 23;274(17):11924-9. [106] Marathe GK, Davies SS, Harrison KA, Silva AR, Murphy RC, Castro-Faria-Neto H, et al. Inflammatory platelet-activating factor-like phospholipids in oxidized low density lipoproteins 158 are fragmented alkyl phosphatidylcholines. The Journal of biological chemistry. 1999 Oct 1;274(40):28395-404. [107] Cyrus T, Witztum JL, Rader DJ, Tangirala R, Fazio S, Linton MF, et al. Disruption of the 12/15- lipoxygenase gene diminishes atherosclerosis in apo E-deficient mice. The Journal of clinical investigation. 1999 Jun;103(11):1597-604. [108] Cyrus T, Pratico D, Zhao L, Witztum JL, Rader DJ, Rokach J, et al. Absence of 12/15- lipoxygenase expression decreases lipid peroxidation and atherogenesis in apolipoprotein e- deficient mice. Circulation. 2001 May 8;103(18):2277-82. [109] George J, Afek A, Shaish A, Levkovitz H, Bloom N, Cyrus T, et al. 12/15-Lipoxygenase gene disruption attenuates atherogenesis in LDL receptor-deficient mice. Circulation. 2001 Oct 2;104(14):1646-50. [110] Zhao L, Cuff CA, Moss E, Wille U, Cyrus T, Klein EA, et al. Selective interleukin-12 synthesis defect in 12/15-lipoxygenase-deficient macrophages associated with reduced atherosclerosis in a mouse model of familial hypercholesterolemia. The Journal of biological chemistry. 2002 Sep 20;277(38):35350-6. [111] Huo Y, Zhao L, Hyman MC, Shashkin P, Harry BL, Burcin T, et al. Critical role of macrophage 12/15-lipoxygenase for atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2004 Oct 5;110(14):2024-31. [112] Podrez EA, Febbraio M, Sheibani N, Schmitt D, Silverstein RL, Hajjar DP, et al. Macrophage scavenger receptor CD36 is the major receptor for LDL modified by monocyte-generated reactive nitrogen species. The Journal of clinical investigation. 2000 Apr;105(8):1095-108. [113] Zhang R, Brennan ML, Shen Z, MacPherson JC, Schmitt D, Molenda CE, et al. Myeloperoxidase functions as a major enzymatic catalyst for initiation of lipid peroxidation at sites of inflammation. The Journal of biological chemistry. 2002 Nov 29;277(48):46116-22. [114] Brennan ML, Penn MS, Van Lente F, Nambi V, Shishehbor MH, Aviles RJ, et al. Prognostic value of myeloperoxidase in patients with chest pain. The New England journal of medicine. 2003 Oct 23;349(17):1595-604. [115] McMillen TS, Heinecke JW, LeBoeuf RC. Expression of human myeloperoxidase by macrophages promotes atherosclerosis in mice. Circulation. 2005 May 31;111(21):2798-804. [116] Nicholls SJ, Hazen SL. Myeloperoxidase and cardiovascular disease. Arteriosclerosis, thrombosis, and vascular biology. 2005 Jun;25(6):1102-11. [117] Nicholls SJ, Zheng L, Hazen SL. Formation of dysfunctional high-density lipoprotein by myeloperoxidase. Trends in cardiovascular medicine. 2005 Aug;15(6):212-9. [118] Wang Z, Nicholls SJ, Rodriguez ER, Kummu O, Horkko S, Barnard J, et al. Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nature medicine. 2007 Oct;13(10):1176-84. [119] Nicholls SJ, Hazen SL. Myeloperoxidase, modified lipoproteins and atherogenesis. Journal of lipid research. 2008 Dec 16. [120] Warnholtz A, Nickenig G, Schulz E, Macharzina R, Brasen JH, Skatchkov M, et al. Increased NADH-oxidase-mediated superoxide production in the early stages of atherosclerosis: evidence for involvement of the renin-angiotensin system. Circulation. 1999 Apr 20;99(15):2027-33. [121] Guzik TJ, West NE, Black E, McDonald D, Ratnatunga C, Pillai R, et al. Functional effect of the C242T polymorphism in the NAD(P)H oxidase p22phox gene on vascular superoxide production in atherosclerosis. Circulation. 2000 Oct 10;102(15):1744-7. [122] Barry-Lane PA, Patterson C, van der Merwe M, Hu Z, Holland SM, Yeh ET, et al. p47phox is required for atherosclerotic lesion progression in ApoE(-/-) mice. The Journal of clinical investigation. 2001 Nov;108(10):1513-22. [123] Sorescu D, Weiss D, Lassegue B, Clempus RE, Szocs K, Sorescu GP, et al. Superoxide production and expression of nox family proteins in human atherosclerosis. Circulation. 2002 Mar 26;105(12):1429-35. [124] Hwang J, Saha A, Boo YC, Sorescu GP, McNally JS, Holland SM, et al. Oscillatory shear stress stimulates endothelial production of O2- from p47phox-dependent NAD(P)H oxidases, leading to monocyte adhesion. The Journal of biological chemistry. 2003 Nov 21;278(47):47291-8. 159 [125] Cardillo C, Kilcoyne CM, Cannon RO, 3rd, Quyyumi AA, Panza JA. Xanthine oxidase inhibition with oxypurinol improves endothelial vasodilator function in hypercholesterolemic but not in hypertensive patients. Hypertension. 1997 Jul;30(1 Pt 1):57-63. [126] Spiekermann S, Landmesser U, Dikalov S, Bredt M, Gamez G, Tatge H, et al. Electron spin resonance characterization of vascular xanthine and NAD(P)H oxidase activity in patients with coronary artery disease: relation to endothelium-dependent vasodilation. Circulation. 2003 Mar 18;107(10):1383-9. [127] Ballinger SW, Patterson C, Knight-Lozano CA, Burow DL, Conklin CA, Hu Z, et al. Mitochondrial integrity and function in atherogenesis. Circulation. 2002 Jul 30;106(5):544-9. [128] Ohashi M, Runge MS, Faraci FM, Heistad DD. MnSOD deficiency increases endothelial dysfunction in ApoE-deficient mice. Arteriosclerosis, thrombosis, and vascular biology. 2006 Oct;26(10):2331-6. [129] Stroes E, Kastelein J, Cosentino F, Erkelens W, Wever R, Koomans H, et al. Tetrahydrobiopterin restores endothelial function in hypercholesterolemia. The Journal of clinical investigation. 1997 Jan 1;99(1):41-6. [130] Martasek P, Miller RT, Liu Q, Roman LJ, Salerno JC, Migita CT, et al. The C331A mutant of neuronal nitric-oxide synthase is defective in arginine binding. The Journal of biological chemistry. 1998 Dec 25;273(52):34799-805. [131] Guzik TJ, Black E, West NE, McDonald D, Ratnatunga C, Pillai R, et al. Relationship between the G894T polymorphism (Glu298Asp variant) in endothelial nitric oxide synthase and nitric oxide-mediated endothelial function in human atherosclerosis. American journal of medical genetics. 2001 Apr 22;100(2):130-7. [132] Hsiai TK, Hwang J, Barr ML, Correa A, Hamilton R, Alavi M, et al. Hemodynamics influences vascular peroxynitrite formation: Implication for low-density lipoprotein apo-B-100 nitration. Free radical biology & medicine. 2007 Feb 15;42(4):519-29. [133] Kuhlencordt PJ, Chen J, Han F, Astern J, Huang PL. Genetic deficiency of inducible nitric oxide synthase reduces atherosclerosis and lowers plasma lipid peroxides in apolipoprotein E-knockout mice. Circulation. 2001 Jun 26;103(25):3099-104. [134] Ishikawa K, Navab M, Leitinger N, Fogelman AM, Lusis AJ. Induction of heme oxygenase-1 inhibits the monocyte transmigration induced by mildly oxidized LDL. The Journal of clinical investigation. 1997 Sep 1;100(5):1209-16. [135] Ishikawa K, Sugawara D, Goto J, Watanabe Y, Kawamura K, Shiomi M, et al. Heme oxygenase- 1 inhibits atherogenesis in Watanabe heritable hyperlipidemic rabbits. Circulation. 2001 Oct 9;104(15):1831-6. [136] Ishikawa K, Sugawara D, Wang X, Suzuki K, Itabe H, Maruyama Y, et al. Heme oxygenase-1 inhibits atherosclerotic lesion formation in ldl-receptor knockout mice. Circulation research. 2001 Mar 16;88(5):506-12. [137] Juan SH, Lee TS, Tseng KW, Liou JY, Shyue SK, Wu KK, et al. Adenovirus-mediated heme oxygenase-1 gene transfer inhibits the development of atherosclerosis in apolipoprotein E- deficient mice. Circulation. 2001 Sep 25;104(13):1519-25. [138] Yet SF, Layne MD, Liu X, Chen YH, Ith B, Sibinga NE, et al. Absence of heme oxygenase-1 exacerbates atherosclerotic lesion formation and vascular remodeling. Faseb J. 2003 Sep;17(12):1759-61. [139] Exner M, Minar E, Wagner O, Schillinger M. The role of heme oxygenase-1 promoter polymorphisms in human disease. Free radical biology & medicine. 2004 Oct 15;37(8):1097- 104. [140] Bhattacharyya T, Nicholls SJ, Topol EJ, Zhang R, Yang X, Schmitt D, et al. Relationship of paraoxonase 1 (PON1) gene polymorphisms and functional activity with systemic oxidative stress and cardiovascular risk. Jama. 2008 Mar 19;299(11):1265-76. [141] Shih DM, Gu L, Xia YR, Navab M, Li WF, Hama S, et al. Mice lacking serum paraoxonase are susceptible to organophosphate toxicity and atherosclerosis. Nature. 1998 Jul 16;394(6690):284- 7. 160 [142] Ng CJ, Shih DM, Hama SY, Villa N, Navab M, Reddy ST. The paraoxonase gene family and atherosclerosis. Free radical biology & medicine. 2005 Jan 15;38(2):153-63. [143] Ng CJ, Bourquard N, Grijalva V, Hama S, Shih DM, Navab M, et al. Paraoxonase-2 deficiency aggravates atherosclerosis in mice despite lower apolipoprotein-B-containing lipoproteins: anti- atherogenic role for paraoxonase-2. The Journal of biological chemistry. 2006 Oct 6;281(40):29491-500. [144] Ng CJ, Hama SY, Bourquard N, Navab M, Reddy ST. Adenovirus mediated expression of human paraoxonase 2 protects against the development of atherosclerosis in apolipoprotein E- deficient mice. Molecular genetics and metabolism. 2006 Dec;89(4):368-73. [145] Ng CJ, Bourquard N, Hama SY, Shih D, Grijalva VR, Navab M, et al. Adenovirus-mediated expression of human paraoxonase 3 protects against the progression of atherosclerosis in apolipoprotein E-deficient mice. Arteriosclerosis, thrombosis, and vascular biology. 2007 Jun;27(6):1368-74. [146] Parthasarathy S, Young SG, Witztum JL, Pittman RC, Steinberg D. Probucol inhibits oxidative modification of low density lipoprotein. The Journal of clinical investigation. 1986 Feb;77(2):641-4. [147] Carew TE, Schwenke DC, Steinberg D. Antiatherogenic effect of probucol unrelated to its hypocholesterolemic effect: evidence that antioxidants in vivo can selectively inhibit low density lipoprotein degradation in macrophage-rich fatty streaks and slow the progression of atherosclerosis in the Watanabe heritable hyperlipidemic rabbit. Proceedings of the National Academy of Sciences of the United States of America. 1987 Nov;84(21):7725-9. [148] Kita T, Nagano Y, Yokode M, Ishii K, Kume N, Ooshima A, et al. Probucol prevents the progression of atherosclerosis in Watanabe heritable hyperlipidemic rabbit, an animal model for familial hypercholesterolemia. Proceedings of the National Academy of Sciences of the United States of America. 1987 Aug;84(16):5928-31. [149] Bjorkhem I, Henriksson-Freyschuss A, Breuer O, Diczfalusy U, Berglund L, Henriksson P. The antioxidant butylated hydroxytoluene protects against atherosclerosis. Arterioscler Thromb. 1991 Jan-Feb;11(1):15-22. [150] Sparrow CP, Doebber TW, Olszewski J, Wu MS, Ventre J, Stevens KA, et al. Low density lipoprotein is protected from oxidation and the progression of atherosclerosis is slowed in cholesterol-fed rabbits by the antioxidant N,N'-diphenyl-phenylenediamine. The Journal of clinical investigation. 1992 Jun;89(6):1885-91. [151] Tangirala RK, Casanada F, Miller E, Witztum JL, Steinberg D, Palinski W. Effect of the antioxidant N,N'-diphenyl 1,4-phenylenediamine (DPPD) on atherosclerosis in apoE-deficient mice. Arteriosclerosis, thrombosis, and vascular biology. 1995 Oct;15(10):1625-30. [152] Gey KF, Moser UK, Jordan P, Stahelin HB, Eichholzer M, Ludin E. Increased risk of cardiovascular disease at suboptimal plasma concentrations of essential antioxidants: an epidemiological update with special attention to carotene and vitamin C. The American journal of clinical nutrition. 1993 May;57(5 Suppl):787S-97S. [153] Gey KF. Cardiovascular disease and vitamins. Concurrent correction of 'suboptimal' plasma antioxidant levels may, as important part of 'optimal' nutrition, help to prevent early stages of cardiovascular disease and cancer, respectively. Bibliotheca nutritio et dieta. 1995(52):75-91. [154] Heinecke JW. Lipoprotein oxidation in cardiovascular disease: chief culprit or innocent bystander? The Journal of experimental medicine. 2006 Apr 17;203(4):813-6. [155] Steinberg D, Witztum JL. Is the oxidative modification hypothesis relevant to human atherosclerosis? Do the antioxidant trials conducted to date refute the hypothesis? Circulation. 2002 Apr 30;105(17):2107-11. [156] Witztum JL, Steinberg D. The oxidative modification hypothesis of atherosclerosis: does it hold for humans? Trends in cardiovascular medicine. 2001 Apr-May;11(3-4):93-102. [157] Steinberg D. The LDL modification hypothesis of atherogenesis: an update. Journal of lipid research. 2008 Nov 15. 161 [158] Natural history of aortic and coronary atherosclerotic lesions in youth. Findings from the PDAY Study. Pathobiological Determinants of Atherosclerosis in Youth (PDAY) Research Group. Arterioscler Thromb. 1993 Sep;13(9):1291-8. [159] Zieske AW, Malcom GT, Strong JP. Natural history and risk factors of atherosclerosis in children and youth: the PDAY study. Pediatric pathology & molecular medicine. 2002 Mar- Apr;21(2):213-37. [160] Davies DF. Hypothesis. An immunological view of atherogenesis. Journal of atherosclerosis research. 1969 Sep-Oct;10(2):253-9. [161] Ross R. Atherosclerosis--an inflammatory disease. The New England journal of medicine. 1999 Jan 14;340(2):115-26. [162] Libby P. The forgotten majority: unfinished business in cardiovascular risk reduction. Journal of the American College of Cardiology. 2005 Oct 4;46(7):1225-8. [163] Steinberg D. Atherogenesis in perspective: hypercholesterolemia and inflammation as partners in crime. Nature medicine. 2002 Nov;8(11):1211-7. [164] Hansson GK, Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol. 2006 Jul;6(7):508-19. [165] Horkko S, Binder CJ, Shaw PX, Chang MK, Silverman G, Palinski W, et al. Immunological responses to oxidized LDL. Free radical biology & medicine. 2000 Jun 15;28(12):1771-9. [166] Bochkov VN. Inflammatory profile of oxidized phospholipids. Thrombosis and haemostasis. 2007 Mar;97(3):348-54. [167] Buono C, Come CE, Witztum JL, Maguire GF, Connelly PW, Carroll M, et al. Influence of C3 deficiency on atherosclerosis. Circulation. 2002 Jun 25;105(25):3025-31. [168] Persson L, Boren J, Robertson AK, Wallenius V, Hansson GK, Pekna M. Lack of complement factor C3, but not factor B, increases hyperlipidemia and atherosclerosis in apolipoprotein E-/- low-density lipoprotein receptor-/- mice. Arteriosclerosis, thrombosis, and vascular biology. 2004 Jun;24(6):1062-7. [169] Schmiedt W, Kinscherf R, Deigner HP, Kamencic H, Nauen O, Kilo J, et al. Complement C6 deficiency protects against diet-induced atherosclerosis in rabbits. Arteriosclerosis, thrombosis, and vascular biology. 1998 Nov;18(11):1790-5. [170] Vanderlaan PA, Reardon CA. Thematic review series: the immune system and atherogenesis. The unusual suspects:an overview of the minor leukocyte populations in atherosclerosis. Journal of lipid research. 2005 May;46(5):829-38. [171] Berland R, Wortis HH. Origins and functions of B-1 cells with notes on the role of CD5. Annual review of immunology. 2002;20:253-300. [172] Ochsenbein AF, Fehr T, Lutz C, Suter M, Brombacher F, Hengartner H, et al. Control of early viral and bacterial distribution and disease by natural antibodies. Science (New York, NY. 1999 Dec 10;286(5447):2156-9. [173] Chou MY, Hartvigsen K, Hansen LF, Fogelstrand L, Shaw PX, Boullier A, et al. Oxidation- specific epitopes are important targets of innate immunity. Journal of internal medicine. 2008 May;263(5):479-88. [174] Tupin E, Nicoletti A, Elhage R, Rudling M, Ljunggren HG, Hansson GK, et al. CD1d-dependent activation of NKT cells aggravates atherosclerosis. The Journal of experimental medicine. 2004 Feb 2;199(3):417-22. [175] Dansky HM, Charlton SA, Harper MM, Smith JD. T and B lymphocytes play a minor role in atherosclerotic plaque formation in the apolipoprotein E-deficient mouse. Proceedings of the National Academy of Sciences of the United States of America. 1997 Apr 29;94(9):4642-6. [176] Reardon CA, Blachowicz L, White T, Cabana V, Wang Y, Lukens J, et al. Effect of immune deficiency on lipoproteins and atherosclerosis in male apolipoprotein E-deficient mice. Arteriosclerosis, thrombosis, and vascular biology. 2001 Jun;21(6):1011-6. [177] Song L, Leung C, Schindler C. Lymphocytes are important in early atherosclerosis. The Journal of clinical investigation. 2001 Jul;108(2):251-9. 162 [178] Zhou X, Nicoletti A, Elhage R, Hansson GK. Transfer of CD4(+) T cells aggravates atherosclerosis in immunodeficient apolipoprotein E knockout mice. Circulation. 2000 Dec 12;102(24):2919-22. [179] Buono C, Pang H, Uchida Y, Libby P, Sharpe AH, Lichtman AH. B7-1/B7-2 costimulation regulates plaque antigen-specific T-cell responses and atherogenesis in low-density lipoprotein receptor-deficient mice. Circulation. 2004 Apr 27;109(16):2009-15. [180] Mach F, Schonbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature. 1998 Jul 9;394(6689):200-3. [181] Davenport P, Tipping PG. The role of interleukin-4 and interleukin-12 in the progression of atherosclerosis in apolipoprotein E-deficient mice. The American journal of pathology. 2003 Sep;163(3):1117-25. [182] Elhage R, Jawien J, Rudling M, Ljunggren HG, Takeda K, Akira S, et al. Reduced atherosclerosis in interleukin-18 deficient apolipoprotein E-knockout mice. Cardiovascular research. 2003 Jul 1;59(1):234-40. [183] Miller AM, Xu D, Asquith DL, Denby L, Li Y, Sattar N, et al. IL-33 reduces the development of atherosclerosis. The Journal of experimental medicine. 2008 Feb 18;205(2):339-46. [184] Mallat Z, Gojova A, Brun V, Esposito B, Fournier N, Cottrez F, et al. Induction of a regulatory T cell type 1 response reduces the development of atherosclerosis in apolipoprotein E-knockout mice. Circulation. 2003 Sep 9;108(10):1232-7. [185] Ait-Oufella H, Salomon BL, Potteaux S, Robertson AK, Gourdy P, Zoll J, et al. Natural regulatory T cells control the development of atherosclerosis in mice. Nature medicine. 2006 Feb;12(2):178-80. [186] Binder CJ, Hartvigsen K, Chang MK, Miller M, Broide D, Palinski W, et al. IL-5 links adaptive and natural immunity specific for epitopes of oxidized LDL and protects from atherosclerosis. The Journal of clinical investigation. 2004 Aug;114(3):427-37. [187] Caligiuri G, Rudling M, Ollivier V, Jacob MP, Michel JB, Hansson GK, et al. Interleukin-10 deficiency increases atherosclerosis, thrombosis, and low-density lipoproteins in apolipoprotein E knockout mice. Molecular medicine (Cambridge, Mass. 2003 Jan-Feb;9(1-2):10-7. [188] Robertson AK, Rudling M, Zhou X, Gorelik L, Flavell RA, Hansson GK. Disruption of TGF- beta signaling in T cells accelerates atherosclerosis. The Journal of clinical investigation. 2003 Nov;112(9):1342-50. [189] Helgadottir A, Manolescu A, Thorleifsson G, Gretarsdottir S, Jonsdottir H, Thorsteinsdottir U, et al. The gene encoding 5-lipoxygenase activating protein confers risk of myocardial infarction and stroke. Nature genetics. 2004 Mar;36(3):233-9. [190] Swanberg M, Lidman O, Padyukov L, Eriksson P, Akesson E, Jagodic M, et al. MHC2TA is associated with differential MHC molecule expression and susceptibility to rheumatoid arthritis, multiple sclerosis and myocardial infarction. Nature genetics. 2005 May;37(5):486-94. [191] Wang X, Ria M, Kelmenson PM, Eriksson P, Higgins DC, Samnegard A, et al. Positional identification of TNFSF4, encoding OX40 ligand, as a gene that influences atherosclerosis susceptibility. Nature genetics. 2005 Apr;37(4):365-72. [192] Ameli S, Hultgardh-Nilsson A, Regnstrom J, Calara F, Yano J, Cercek B, et al. Effect of immunization with homologous LDL and oxidized LDL on early atherosclerosis in hypercholesterolemic rabbits. Arteriosclerosis, thrombosis, and vascular biology. 1996 Aug;16(8):1074-9. [193] Fredrikson GN, Soderberg I, Lindholm M, Dimayuga P, Chyu KY, Shah PK, et al. Inhibition of atherosclerosis in apoE-null mice by immunization with apoB-100 peptide sequences. Arteriosclerosis, thrombosis, and vascular biology. 2003 May 1;23(5):879-84. [194] Freigang S, Horkko S, Miller E, Witztum JL, Palinski W. Immunization of LDL receptor- deficient mice with homologous malondialdehyde-modified and native LDL reduces progression of atherosclerosis by mechanisms other than induction of high titers of antibodies to oxidative neoepitopes. Arteriosclerosis, thrombosis, and vascular biology. 1998 Dec;18(12):1972-82. 163 [195] George J, Afek A, Gilburd B, Levkovitz H, Shaish A, Goldberg I, et al. Hyperimmunization of apo-E-deficient mice with homologous malondialdehyde low-density lipoprotein suppresses early atherogenesis. Atherosclerosis. 1998 May;138(1):147-52. [196] Palinski W, Miller E, Witztum JL. Immunization of low density lipoprotein (LDL) receptor- deficient rabbits with homologous malondialdehyde-modified LDL reduces atherogenesis. Proceedings of the National Academy of Sciences of the United States of America. 1995 Jan 31;92(3):821-5. [197] Zhou X, Caligiuri G, Hamsten A, Lefvert AK, Hansson GK. LDL immunization induces T-cell- dependent antibody formation and protection against atherosclerosis. Arteriosclerosis, thrombosis, and vascular biology. 2001 Jan;21(1):108-14. [198] Yamashita T, Freigang S, Eberle C, Pattison J, Gupta S, Napoli C, et al. Maternal immunization programs postnatal immune responses and reduces atherosclerosis in offspring. Circulation research. 2006 Sep 29;99(7):e51-64. [199] Nicoletti A, Paulsson G, Caligiuri G, Zhou X, Hansson GK. Induction of neonatal tolerance to oxidized lipoprotein reduces atherosclerosis in ApoE knockout mice. Molecular medicine (Cambridge, Mass. 2000 Apr;6(4):283-90. [200] Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nature immunology. 2005 Dec;6(12):1191-7. [201] Cecchini MG, Dominguez MG, Mocci S, Wetterwald A, Felix R, Fleisch H, et al. Role of colony stimulating factor-1 in the establishment and regulation of tissue macrophages during postnatal development of the mouse. Development (Cambridge, England). 1994 Jun;120(6):1357-72. [202] Dai XM, Ryan GR, Hapel AJ, Dominguez MG, Russell RG, Kapp S, et al. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood. 2002 Jan 1;99(1):111-20. [203] Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003 Jul;19(1):71-82. [204] Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, et al. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science (New York, NY. 2007 Aug 3;317(5838):666-70. [205] Gordon S. Macrophage heterogeneity and tissue lipids. The Journal of clinical investigation. 2007 Jan;117(1):89-93. [206] Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453-61. [207] Landsman L, Varol C, Jung S. Distinct differentiation potential of blood monocyte subsets in the lung. J Immunol. 2007 Feb 15;178(4):2000-7. [208] Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, et al. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. The Journal of experimental medicine. 2007 Nov 26;204(12):3037-47. [209] Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, et al. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. The Journal of experimental medicine. 2007 May 14;204(5):1057-69. [210] Janeway CA, Jr., Medzhitov R. Innate immune recognition. Annual review of immunology. 2002;20:197-216. [211] Medzhitov R, Preston-Hurlburt P, Janeway CA, Jr. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997 Jul 24;388(6640):394-7. [212] Uematsu S, Akira S. Toll-Like receptors (TLRs) and their ligands. Handbook of experimental pharmacology. 2008(183):1-20. [213] Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006 Feb 24;124(4):783-801. [214] Seimon TA, Obstfeld A, Moore KJ, Golenbock DT, Tabas I. Combinatorial pattern recognition receptor signaling alters the balance of life and death in macrophages. Proceedings of the National Academy of Sciences of the United States of America. 2006 Dec 26;103(52):19794-9. 164 [215] Trinchieri G, Sher A. Cooperation of Toll-like receptor signals in innate immune defence. Nat Rev Immunol. 2007 Mar;7(3):179-90. [216] Hoebe K, Georgel P, Rutschmann S, Du X, Mudd S, Crozat K, et al. CD36 is a sensor of diacylglycerides. Nature. 2005 Feb 3;433(7025):523-7. [217] Kawai T, Akira S. TLR signaling. Seminars in immunology. 2007 Feb;19(1):24-32. [218] Mullick AE, Tobias PS, Curtiss LK. Modulation of atherosclerosis in mice by Toll-like receptor 2. The Journal of clinical investigation. 2005 Nov;115(11):3149-56. [219] Lehr HA, Sagban TA, Ihling C, Zahringer U, Hungerer KD, Blumrich M, et al. Immunopathogenesis of atherosclerosis: endotoxin accelerates atherosclerosis in rabbits on hypercholesterolemic diet. Circulation. 2001 Aug 21;104(8):914-20. [220] Liu X, Ukai T, Yumoto H, Davey M, Goswami S, Gibson FC, 3rd, et al. Toll-like receptor 2 plays a critical role in the progression of atherosclerosis that is independent of dietary lipids. Atherosclerosis. 2008 Jan;196(1):146-54. [221] Madan M, Amar S. Toll-like receptor-2 mediates diet and/or pathogen associated atherosclerosis: proteomic findings. PLoS ONE. 2008;3(9):e3204. [222] Bjorkbacka H, Kunjathoor VV, Moore KJ, Koehn S, Ordija CM, Lee MA, et al. Reduced atherosclerosis in MyD88-null mice links elevated serum cholesterol levels to activation of innate immunity signaling pathways. Nature medicine. 2004 Apr;10(4):416-21. [223] Michelsen KS, Wong MH, Shah PK, Zhang W, Yano J, Doherty TM, et al. Lack of Toll-like receptor 4 or myeloid differentiation factor 88 reduces atherosclerosis and alters plaque phenotype in mice deficient in apolipoprotein E. Proceedings of the National Academy of Sciences of the United States of America. 2004 Jul 20;101(29):10679-84. [224] Kodama T, Reddy P, Kishimoto C, Krieger M. Purification and characterization of a bovine acetyl low density lipoprotein receptor. Proceedings of the National Academy of Sciences of the United States of America. 1988 Dec;85(23):9238-42. [225] Rohrer L, Freeman M, Kodama T, Penman M, Krieger M. Coiled-coil fibrous domains mediate ligand binding by macrophage scavenger receptor type II. Nature. 1990 Feb 8;343(6258):570-2. [226] Kodama T, Freeman M, Rohrer L, Zabrecky J, Matsudaira P, Krieger M. Type I macrophage scavenger receptor contains alpha-helical and collagen-like coiled coils. Nature. 1990 Feb 8;343(6258):531-5. [227] Pluddemann A, Neyen C, Gordon S. Macrophage scavenger receptors and host-derived ligands. Methods (San Diego, Calif. 2007 Nov;43(3):207-17. [228] Pluddemann A, Mukhopadhyay S, Gordon S. The interaction of macrophage receptors with bacterial ligands. Expert reviews in molecular medicine. 2006;8(28):1-25. [229] Suzuki H, Kurihara Y, Takeya M, Kamada N, Kataoka M, Jishage K, et al. A role for macrophage scavenger receptors in atherosclerosis and susceptibility to infection. Nature. 1997 Mar 20;386(6622):292-6. [230] Sakaguchi H, Takeya M, Suzuki H, Hakamata H, Kodama T, Horiuchi S, et al. Role of macrophage scavenger receptors in diet-induced atherosclerosis in mice. Laboratory investigation; a journal of technical methods and pathology. 1998 Apr;78(4):423-34. [231] Kamada N, Kodama T, Suzuki H. Macrophage scavenger receptor (SR-A I/II) deficiency reduced diet-induced atherosclerosis in C57BL/6J mice. Journal of atherosclerosis and thrombosis. 2001;8(1):1-6. [232] Babaev VR, Gleaves LA, Carter KJ, Suzuki H, Kodama T, Fazio S, et al. Reduced atherosclerotic lesions in mice deficient for total or macrophage-specific expression of scavenger receptor-A. Arteriosclerosis, thrombosis, and vascular biology. 2000 Dec;20(12):2593-9. [233] Febbraio M, Podrez EA, Smith JD, Hajjar DP, Hazen SL, Hoff HF, et al. Targeted disruption of the class B scavenger receptor CD36 protects against atherosclerotic lesion development in mice. The Journal of clinical investigation. 2000 Apr;105(8):1049-56. [234] Febbraio M, Guy E, Silverstein RL. Stem cell transplantation reveals that absence of macrophage CD36 is protective against atherosclerosis. Arteriosclerosis, thrombosis, and vascular biology. 2004 Dec;24(12):2333-8. 165 [235] Marleau S, Harb D, Bujold K, Avallone R, Iken K, Wang Y, et al. EP 80317, a ligand of the CD36 scavenger receptor, protects apolipoprotein E-deficient mice from developing atherosclerotic lesions. Faseb J. 2005 Nov;19(13):1869-71. [236] Mehta JL, Sanada N, Hu CP, Chen J, Dandapat A, Sugawara F, et al. Deletion of LOX-1 reduces atherogenesis in LDLR knockout mice fed high cholesterol diet. Circulation research. 2007 Jun 8;100(11):1634-42. [237] Aslanian AM, Charo IF. Targeted disruption of the scavenger receptor and chemokine CXCL16 accelerates atherosclerosis. Circulation. 2006 Aug 8;114(6):583-90. [238] Arai T, Wang N, Bezouevski M, Welch C, Tall AR. Decreased atherosclerosis in heterozygous low density lipoprotein receptor-deficient mice expressing the scavenger receptor BI transgene. The Journal of biological chemistry. 1999 Jan 22;274(4):2366-71. [239] Braun A, Trigatti BL, Post MJ, Sato K, Simons M, Edelberg JM, et al. Loss of SR-BI expression leads to the early onset of occlusive atherosclerotic coronary artery disease, spontaneous myocardial infarctions, severe cardiac dysfunction, and premature death in apolipoprotein E- deficient mice. Circulation research. 2002 Feb 22;90(3):270-6. [240] Kozarsky KF, Donahee MH, Glick JM, Krieger M, Rader DJ. Gene transfer and hepatic overexpression of the HDL receptor SR-BI reduces atherosclerosis in the cholesterol-fed LDL receptor-deficient mouse. Arteriosclerosis, thrombosis, and vascular biology. 2000 Mar;20(3):721-7. [241] Brett J, Schmidt AM, Yan SD, Zou YS, Weidman E, Pinsky D, et al. Survey of the distribution of a newly characterized receptor for advanced glycation end products in tissues. The American journal of pathology. 1993 Dec;143(6):1699-712. [242] Sun L, Ishida T, Yasuda T, Kojima Y, Honjo T, Yamamoto Y, et al. RAGE mediates oxidized LDL-induced pro-inflammatory effects and atherosclerosis in non-diabetic LDL receptor- deficient mice. Cardiovascular research. 2009 May 1;82(2):371-81. [243] Mariathasan S, Monack DM. Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol. 2007 Jan;7(1):31-40. [244] Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006 May 4;441(7089):101-5. [245] Yan ZQ, Hansson GK. Innate immunity, macrophage activation, and atherosclerosis. Immunological reviews. 2007 Oct;219:187-203. [246] Jonasson L, Holm J, Skalli O, Bondjers G, Hansson GK. Regional accumulations of T cells, macrophages, and smooth muscle cells in the human atherosclerotic plaque. Arteriosclerosis (Dallas, Tex. 1986 Mar-Apr;6(2):131-8. [247] Moreno PR, Falk E, Palacios IF, Newell JB, Fuster V, Fallon JT. Macrophage infiltration in acute coronary syndromes. Implications for plaque rupture. Circulation. 1994 Aug;90(2):775-8. [248] Qiao JH, Tripathi J, Mishra NK, Cai Y, Tripathi S, Wang XP, et al. Role of macrophage colony- stimulating factor in atherosclerosis: studies of osteopetrotic mice. The American journal of pathology. 1997 May;150(5):1687-99. [249] Rajavashisth T, Qiao JH, Tripathi S, Tripathi J, Mishra N, Hua M, et al. Heterozygous osteopetrotic (op) mutation reduces atherosclerosis in LDL receptor- deficient mice. The Journal of clinical investigation. 1998 Jun 15;101(12):2702-10. [250] Bursill CA, Choudhury RP, Ali Z, Greaves DR, Channon KM. Broad-spectrum CC-chemokine blockade by gene transfer inhibits macrophage recruitment and atherosclerotic plaque formation in apolipoprotein E-knockout mice. Circulation. 2004 Oct 19;110(16):2460-6. [251] Dawson TC, Kuziel WA, Osahar TA, Maeda N. Absence of CC chemokine receptor-2 reduces atherosclerosis in apolipoprotein E-deficient mice. Atherosclerosis. 1999 Mar;143(1):205-11. [252] Gu L, Okada Y, Clinton SK, Gerard C, Sukhova GK, Libby P, et al. Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice. Molecular cell. 1998 Aug;2(2):275-81. 166 [253] Combadiere C, Potteaux S, Gao JL, Esposito B, Casanova S, Lee EJ, et al. Decreased atherosclerotic lesion formation in CX3CR1/apolipoprotein E double knockout mice. Circulation. 2003 Feb 25;107(7):1009-16. [254] Lesnik P, Haskell CA, Charo IF. Decreased atherosclerosis in CX3CR1-/- mice reveals a role for fractalkine in atherogenesis. The Journal of clinical investigation. 2003 Feb;111(3):333-40. [255] Teupser D, Pavlides S, Tan M, Gutierrez-Ramos JC, Kolbeck R, Breslow JL. Major reduction of atherosclerosis in fractalkine (CX3CL1)-deficient mice is at the brachiocephalic artery, not the aortic root. Proceedings of the National Academy of Sciences of the United States of America. 2004 Dec 21;101(51):17795-800. [256] Saederup N, Chan L, Lira SA, Charo IF. Fractalkine deficiency markedly reduces macrophage accumulation and atherosclerotic lesion formation in CCR2-/- mice: evidence for independent chemokine functions in atherogenesis. Circulation. 2008 Apr 1;117(13):1642-8. [257] Swirski FK, Libby P, Aikawa E, Alcaide P, Luscinskas FW, Weissleder R, et al. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. The Journal of clinical investigation. 2007 Jan;117(1):195-205. [258] Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. The Journal of clinical investigation. 2007 Jan;117(1):185-94. [259] Combadiere C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, et al. Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 2008 Apr 1;117(13):1649-57. [260] An G, Wang H, Tang R, Yago T, McDaniel JM, McGee S, et al. P-selectin glycoprotein ligand-1 is highly expressed on Ly-6Chi monocytes and a major determinant for Ly-6Chi monocyte recruitment to sites of atherosclerosis in mice. Circulation. 2008 Jun 24;117(25):3227-37. [261] Galis ZS, Sukhova GK, Kranzhofer R, Clark S, Libby P. Macrophage foam cells from experimental atheroma constitutively produce matrix-degrading proteinases. Proceedings of the National Academy of Sciences of the United States of America. 1995 Jan 17;92(2):402-6. [262] Galis ZS, Sukhova GK, Libby P. Microscopic localization of active proteases by in situ zymography: detection of matrix metalloproteinase activity in vascular tissue. Faseb J. 1995 Jul;9(10):974-80. [263] Sukhova GK, Schonbeck U, Rabkin E, Schoen FJ, Poole AR, Billinghurst RC, et al. Evidence for increased collagenolysis by interstitial collagenases-1 and -3 in vulnerable human atheromatous plaques. Circulation. 1999 May 18;99(19):2503-9. [264] Wilcox JN, Smith KM, Schwartz SM, Gordon D. Localization of tissue factor in the normal vessel wall and in the atherosclerotic plaque. Proceedings of the National Academy of Sciences of the United States of America. 1989 Apr;86(8):2839-43. [265] Lendon CL, Davies MJ, Born GV, Richardson PD. Atherosclerotic plaque caps are locally weakened when macrophages density is increased. Atherosclerosis. 1991 Mar;87(1):87-90. [266] Shah PK, Falk E, Badimon JJ, Fernandez-Ortiz A, Mailhac A, Villareal-Levy G, et al. Human monocyte-derived macrophages induce collagen breakdown in fibrous caps of atherosclerotic plaques. Potential role of matrix-degrading metalloproteinases and implications for plaque rupture. Circulation. 1995 Sep 15;92(6):1565-9. [267] Feng J, Han J, Pearce SF, Silverstein RL, Gotto AM, Jr., Hajjar DP, et al. Induction of CD36 expression by oxidized LDL and IL-4 by a common signaling pathway dependent on protein kinase C and PPAR-gamma. Journal of lipid research. 2000 May;41(5):688-96. [268] Lougheed M, Lum CM, Ling W, Suzuki H, Kodama T, Steinbrecher U. High affinity saturable uptake of oxidized low density lipoprotein by macrophages from mice lacking the scavenger receptor class A type I/II. The Journal of biological chemistry. 1997 May 16;272(20):12938-44. [269] Kunjathoor VV, Febbraio M, Podrez EA, Moore KJ, Andersson L, Koehn S, et al. Scavenger receptors class A-I/II and CD36 are the principal receptors responsible for the uptake of modified low density lipoprotein leading to lipid loading in macrophages. The Journal of biological chemistry. 2002 Dec 20;277(51):49982-8. 167 [270] Jovinge S, Ares MP, Kallin B, Nilsson J. Human monocytes/macrophages release TNF-alpha in response to Ox-LDL. Arteriosclerosis, thrombosis, and vascular biology. 1996 Dec;16(12):1573- 9. [271] Shen LH, Zhou L, Wang BY, Pu J, Hu LH, Chai DJ, et al. Oxidized low-density lipoprotein induces differentiation of RAW264.7 murine macrophage cell line into dendritic-like cells. Atherosclerosis. 2008 Aug;199(2):257-64. [272] Burger-Kentischer A, Goebel H, Seiler R, Fraedrich G, Schaefer HE, Dimmeler S, et al. Expression of macrophage migration inhibitory factor in different stages of human atherosclerosis. Circulation. 2002 Apr 2;105(13):1561-6. [273] Schlittenhardt D, Schmiedt W, Bonaterra GA, Metz J, Kinscherf R. Colocalization of oxidized low-density lipoprotein, caspase-3, cyclooxygenase-2, and macrophage migration inhibitory factor in arteriosclerotic human carotid arteries. Cell and tissue research. 2005 Dec;322(3):425- 35. [274] Lougheed M, Zhang HF, Steinbrecher UP. Oxidized low density lipoprotein is resistant to cathepsins and accumulates within macrophages. The Journal of biological chemistry. 1991 Aug 5;266(22):14519-25. [275] Kritharides L, Jessup W, Mander EL, Dean RT. Apolipoprotein A-I-mediated efflux of sterols from oxidized LDL-loaded macrophages. Arteriosclerosis, thrombosis, and vascular biology. 1995 Feb;15(2):276-89. [276] Dhaliwal BS, Steinbrecher UP. Cholesterol delivered to macrophages by oxidized low density lipoprotein is sequestered in lysosomes and fails to efflux normally. Journal of lipid research. 2000 Oct;41(10):1658-65. [277] Hessler JR, Morel DW, Lewis LJ, Chisolm GM. Lipoprotein oxidation and lipoprotein-induced cytotoxicity. Arteriosclerosis (Dallas, Tex. 1983 May-Jun;3(3):215-22. [278] Morel DW, Hessler JR, Chisolm GM. Low density lipoprotein cytotoxicity induced by free radical peroxidation of lipid. Journal of lipid research. 1983 Aug;24(8):1070-6. [279] Vicca S, Hennequin C, Nguyen-Khoa T, Massy ZA, Descamps-Latscha B, Drueke TB, et al. Caspase-dependent apoptosis in THP-1 cells exposed to oxidized low-density lipoproteins. Biochemical and biophysical research communications. 2000 Jul 14;273(3):948-54. [280] Vicca S, Massy ZA, Hennequin C, Rihane D, Nguyen-Khoa T, Drueke TB, et al. New insights into the effects of the protein moiety of oxidized LDL (oxLDL). Kidney international. 2003 May(84):S125-7. [281] Colles SM, Maxson JM, Carlson SG, Chisolm GM. Oxidized LDL-induced injury and apoptosis in atherosclerosis. Potential roles for oxysterols. Trends in cardiovascular medicine. 2001 Apr- May;11(3-4):131-8. [282] Harada K, Ishibashi S, Miyashita T, Osuga J, Yagyu H, Ohashi K, et al. Bcl-2 protein inhibits oxysterol-induced apoptosis through suppressing CPP32-mediated pathway. FEBS letters. 1997 Jul 7;411(1):63-6. [283] Yang L, Sinensky MS. 25-Hydroxycholesterol activates a cytochrome c release-mediated caspase cascade. Biochemical and biophysical research communications. 2000 Nov 30;278(3):557-63. [284] Panini SR, Yang L, Rusinol AE, Sinensky MS, Bonventre JV, Leslie CC. Arachidonate metabolism and the signaling pathway of induction of apoptosis by oxidized LDL/oxysterol. Journal of lipid research. 2001 Oct;42(10):1678-86. [285] Rusinol AE, Thewke D, Liu J, Freeman N, Panini SR, Sinensky MS. AKT/protein kinase B regulation of BCL family members during oxysterol-induced apoptosis. The Journal of biological chemistry. 2004 Jan 9;279(2):1392-9. [286] Berthier A, Lemaire-Ewing S, Prunet C, Monier S, Athias A, Bessede G, et al. Involvement of a calcium-dependent dephosphorylation of BAD associated with the localization of Trpc-1 within lipid rafts in 7-ketocholesterol-induced THP-1 cell apoptosis. Cell death and differentiation. 2004 Aug;11(8):897-905. 168 [287] Li W, Yuan XM. Increased expression and translocation of lysosomal cathepsins contribute to macrophage apoptosis in atherogenesis. Annals of the New York Academy of Sciences. 2004 Dec;1030:427-33. [288] Li W, Dalen H, Eaton JW, Yuan XM. Apoptotic death of inflammatory cells in human atheroma. Arteriosclerosis, thrombosis, and vascular biology. 2001 Jul;21(7):1124-30. [289] Guicciardi ME, Deussing J, Miyoshi H, Bronk SF, Svingen PA, Peters C, et al. Cathepsin B contributes to TNF-alpha-mediated hepatocyte apoptosis by promoting mitochondrial release of cytochrome c. The Journal of clinical investigation. 2000 Nov;106(9):1127-37. [290] Stoka V, Turk B, Schendel SL, Kim TH, Cirman T, Snipas SJ, et al. Lysosomal protease pathways to apoptosis. Cleavage of bid, not pro-caspases, is the most likely route. The Journal of biological chemistry. 2001 Feb 2;276(5):3149-57. [291] Gordon D, Reidy MA, Benditt EP, Schwartz SM. Cell proliferation in human coronary arteries. Proceedings of the National Academy of Sciences of the United States of America. 1990 Jun;87(12):4600-4. [292] Villaschi S, Spagnoli LG. Autoradiographic and ultrastructural studies on the human fibro- atheromatous plaque. Atherosclerosis. 1983 Jul;48(1):95-100. [293] Katsuda S, Coltrera MD, Ross R, Gown AM. Human atherosclerosis. IV. Immunocytochemical analysis of cell activation and proliferation in lesions of young adults. The American journal of pathology. 1993 Jun;142(6):1787-93. [294] Rekhter MD, Gordon D. Active proliferation of different cell types, including lymphocytes, in human atherosclerotic plaques. The American journal of pathology. 1995 Sep;147(3):668-77. [295] Yui S, Sasaki T, Miyazaki A, Horiuchi S, Yamazaki M. Induction of murine macrophage growth by modified LDLs. Arterioscler Thromb. 1993 Mar;13(3):331-7. [296] Sakai M, Matsumura T, Biwa T, Hakamata H, Yi D, Shichiri M, et al. Role of the macrophage scavenger receptor for internalization of lysophosphatidylcholine in oxidized low density lipoprotein-induced macrophage growth. Annals of the New York Academy of Sciences. 1997 Apr 15;811:378-84. [297] Sakai M, Miyazaki A, Hakamata H, Kodama T, Suzuki H, Kobori S, et al. The scavenger receptor serves as a route for internalization of lysophosphatidylcholine in oxidized low density lipoprotein-induced macrophage proliferation. The Journal of biological chemistry. 1996 Nov 1;271(44):27346-52. [298] Sakai M, Miyazaki A, Hakamata H, Sasaki T, Yui S, Yamazaki M, et al. Lysophosphatidylcholine plays an essential role in the mitogenic effect of oxidized low density lipoprotein on murine macrophages. The Journal of biological chemistry. 1994 Dec 16;269(50):31430-5. [299] Sakai M, Miyazaki A, Hakamata H, Sato Y, Matsumura T, Kobori S, et al. Lysophosphatidylcholine potentiates the mitogenic activity of modified LDL for human monocyte-derived macrophages. Arteriosclerosis, thrombosis, and vascular biology. 1996 Apr;16(4):600-5. [300] Senokuchi T, Matsumura T, Sakai M, Matsuo T, Yano M, Kiritoshi S, et al. Extracellular signal- regulated kinase and p38 mitogen-activated protein kinase mediate macrophage proliferation induced by oxidized low-density lipoprotein. Atherosclerosis. 2004 Oct;176(2):233-45. [301] Matsumura T, Sakai M, Kobori S, Biwa T, Takemura T, Matsuda H, et al. Two intracellular signaling pathways for activation of protein kinase C are involved in oxidized low-density lipoprotein-induced macrophage growth. Arteriosclerosis, thrombosis, and vascular biology. 1997 Nov;17(11):3013-20. [302] Biwa T, Hakamata H, Sakai M, Miyazaki A, Suzuki H, Kodama T, et al. Induction of murine macrophage growth by oxidized low density lipoprotein is mediated by granulocyte macrophage colony-stimulating factor. The Journal of biological chemistry. 1998 Oct 23;273(43):28305-13. [303] Biwa T, Sakai M, Matsumura T, Kobori S, Kaneko K, Miyazaki A, et al. Sites of action of protein kinase C and phosphatidylinositol 3-kinase are distinct in oxidized low density lipoprotein-induced macrophage proliferation. The Journal of biological chemistry. 2000 Feb 25;275(8):5810-6. 169 [304] Sakai M, Biwa T, Matsumura T, Takemura T, Matsuda H, Anami Y, et al. Glucocorticoid inhibits oxidized LDL-induced macrophage growth by suppressing the expression of granulocyte/macrophage colony-stimulating factor. Arteriosclerosis, thrombosis, and vascular biology. 1999 Jul;19(7):1726-33. [305] Sakai M, Kobori S, Matsumura T, Biwa T, Sato Y, Takemura T, et al. HMG-CoA reductase inhibitors suppress macrophage growth induced by oxidized low density lipoprotein. Atherosclerosis. 1997 Aug;133(1):51-9. [306] Senokuchi T, Matsumura T, Sakai M, Yano M, Taguchi T, Matsuo T, et al. Statins suppress oxidized low density lipoprotein-induced macrophage proliferation by inactivation of the small G protein-p38 MAPK pathway. The Journal of biological chemistry. 2005 Feb 25;280(8):6627- 33. [307] Martens JS, Lougheed M, Gomez-Munoz A, Steinbrecher UP. A modification of apolipoprotein B accounts for most of the induction of macrophage growth by oxidized low density lipoprotein. The Journal of biological chemistry. 1999 Apr 16;274(16):10903-10. [308] Martens JS, Reiner NE, Herrera-Velit P, Steinbrecher UP. Phosphatidylinositol 3-kinase is involved in the induction of macrophage growth by oxidized low density lipoprotein. The Journal of biological chemistry. 1998 Feb 27;273(9):4915-20. [309] Hamilton JA, Myers D, Jessup W, Cochrane F, Byrne R, Whitty G, et al. Oxidized LDL can induce macrophage survival, DNA synthesis, and enhanced proliferative response to CSF-1 and GM-CSF. Arteriosclerosis, thrombosis, and vascular biology. 1999 Jan;19(1):98-105. [310] Hundal RS, Salh BS, Schrader JW, Gomez-Munoz A, Duronio V, Steinbrecher UP. Oxidized low density lipoprotein inhibits macrophage apoptosis through activation of the PI 3-kinase/PKB pathway. Journal of lipid research. 2001 Sep;42(9):1483-91. [311] Tushinski RJ, Stanley ER. The regulation of mononuclear phagocyte entry into S phase by the colony stimulating factor CSF-1. Journal of cellular physiology. 1985 Feb;122(2):221-8. [312] Hamilton JA, Byrne R, Jessup W, Kanagasundaram V, Whitty G. Comparison of macrophage responses to oxidized low-density lipoprotein and macrophage colony-stimulating factor (M- CSF or CSF-1). The Biochemical journal. 2001 Feb 15;354(Pt 1):179-87. [313] Boullier A, Li Y, Quehenberger O, Palinski W, Tabas I, Witztum JL, et al. Minimally oxidized LDL offsets the apoptotic effects of extensively oxidized LDL and free cholesterol in macrophages. Arteriosclerosis, thrombosis, and vascular biology. 2006 May;26(5):1169-76. [314] Hundal RS, Gomez-Munoz A, Kong JY, Salh BS, Marotta A, Duronio V, et al. Oxidized low density lipoprotein inhibits macrophage apoptosis by blocking ceramide generation, thereby maintaining protein kinase B activation and Bcl-XL levels. The Journal of biological chemistry. 2003 Jul 4;278(27):24399-408. [315] Chen JH, Riazy M, Smith EM, Proud CG, Steinbrecher UP, Duronio V. Oxidized LDL-mediated macrophage survival involves elongation factor-2 kinase. Arteriosclerosis, thrombosis, and vascular biology. 2009 Jan;29(1):92-8. [316] Esterbauer H, Gebicki J, Puhl H, Jurgens G. The role of lipid peroxidation and antioxidants in oxidative modification of LDL. Free radical biology & medicine. 1992 Oct;13(4):341-90. [317] Carpenter KL, Challis IR, Arends MJ. Mildly oxidised LDL induces more macrophage death than moderately oxidised LDL: roles of peroxidation, lipoprotein-associated phospholipase A2 and PPARgamma. FEBS letters. 2003 Oct 9;553(1-2):145-50. [318] Han CY, Pak YK. Oxidation-dependent effects of oxidized LDL: proliferation or cell death. Experimental & molecular medicine. 1999 Dec 31;31(4):165-73. [319] Shatrov VA, Brune B. Induced expression of manganese superoxide dismutase by non-toxic concentrations of oxidized low-density lipoprotein (oxLDL) protects against oxLDL-mediated cytotoxicity. The Biochemical journal. 2003 Sep 1;374(Pt 2):505-11. [320] Steinbrecher UP, Gomez-Munoz A, Duronio V. Acid sphingomyelinase in macrophage apoptosis. Current opinion in lipidology. 2004 Oct;15(5):531-7. [321] Chisolm GM, Ma G, Irwin KC, Martin LL, Gunderson KG, Linberg LF, et al. 7 beta- hydroperoxycholest-5-en-3 beta-ol, a component of human atherosclerotic lesions, is the primary 170 cytotoxin of oxidized human low density lipoprotein. Proceedings of the National Academy of Sciences of the United States of America. 1994 Nov 22;91(24):11452-6. [322] Kosugi K, Morel DW, DiCorleto PE, Chisolm GM. Toxicity of oxidized low-density lipoprotein to cultured fibroblasts is selective for S phase of the cell cycle. Journal of cellular physiology. 1987 Mar;130(3):311-20. [323] Marchant CE, Law NS, van der Veen C, Hardwick SJ, Carpenter KL, Mitchinson MJ. Oxidized low-density lipoprotein is cytotoxic to human monocyte-macrophages: protection with lipophilic antioxidants. FEBS letters. 1995 Jan 23;358(2):175-8. [324] Reid VC, Mitchinson MJ, Skepper JN. Cytotoxicity of oxidized low-density lipoprotein to mouse peritoneal macrophages: an ultrastructural study. The Journal of pathology. 1993 Dec;171(4):321-8. [325] Bjorkerud B, Bjorkerud S. Contrary effects of lightly and strongly oxidized LDL with potent promotion of growth versus apoptosis on arterial smooth muscle cells, macrophages, and fibroblasts. Arteriosclerosis, thrombosis, and vascular biology. 1996 Mar;16(3):416-24. [326] Hardwick SJ, Hegyi L, Clare K, Law NS, Carpenter KL, Mitchinson MJ, et al. Apoptosis in human monocyte-macrophages exposed to oxidized low density lipoprotein. The Journal of pathology. 1996 Jul;179(3):294-302. [327] Chatterjee S. Role of oxidized human plasma low density lipoproteins in atherosclerosis: effects on smooth muscle cell proliferation. Molecular and cellular biochemistry. 1992 Apr;111(1- 2):143-7. [328] Balla G, Jacob HS, Eaton JW, Belcher JD, Vercellotti GM. Hemin: a possible physiological mediator of low density lipoprotein oxidation and endothelial injury. Arterioscler Thromb. 1991 Nov-Dec;11(6):1700-11. [329] Cathcart MK, McNally AK, Chisolm GM. Lipoxygenase-mediated transformation of human low density lipoprotein to an oxidized and cytotoxic complex. Journal of lipid research. 1991 Jan;32(1):63-70. [330] Steffen Y, Schewe T, Sies H. Epicatechin protects endothelial cells against oxidized LDL and maintains NO synthase. Biochemical and biophysical research communications. 2005 Jun 17;331(4):1277-83. [331] Negre-Salvayre A, Lopez M, Levade T, Pieraggi MT, Dousset N, Douste-Blazy L, et al. Ultraviolet-treated lipoproteins as a model system for the study of the biological effects of lipid peroxides on cultured cells. II. Uptake and cytotoxicity of ultraviolet-treated LDL on lymphoid cell lines. Biochimica et biophysica acta. 1990 Aug 6;1045(3):224-32. [332] Yuan XM, Li W, Olsson AG, Brunk UT. The toxicity to macrophages of oxidized low-density lipoprotein is mediated through lysosomal damage. Atherosclerosis. 1997 Sep;133(2):153-61. [333] Auge N, Escargueil-Blanc I, Lajoie-Mazenc I, Suc I, Andrieu-Abadie N, Pieraggi MT, et al. Potential role for ceramide in mitogen-activated protein kinase activation and proliferation of vascular smooth muscle cells induced by oxidized low density lipoprotein. The Journal of biological chemistry. 1998 May 22;273(21):12893-900. [334] Auge N, Garcia V, Maupas-Schwalm F, Levade T, Salvayre R, Negre-Salvayre A. Oxidized LDL-induced smooth muscle cell proliferation involves the EGF receptor/PI-3 kinase/Akt and the sphingolipid signaling pathways. Arteriosclerosis, thrombosis, and vascular biology. 2002 Dec 1;22(12):1990-5. [335] Dersch K, Ichijo H, Bhakdi S, Husmann M. Fatty acids liberated from low-density lipoprotein trigger endothelial apoptosis via mitogen-activated protein kinases. Cell death and differentiation. 2005 Aug;12(8):1107-14. [336] Klouche M, Gottschling S, Gerl V, Hell W, Husmann M, Dorweiler B, et al. Atherogenic properties of enzymatically degraded LDL: selective induction of MCP-1 and cytotoxic effects on human macrophages. Arteriosclerosis, thrombosis, and vascular biology. 1998 Sep;18(9):1376-85. [337] Morel DW, DiCorleto PE, Chisolm GM. Endothelial and smooth muscle cells alter low density lipoprotein in vitro by free radical oxidation. Arteriosclerosis (Dallas, Tex. 1984 Jul- Aug;4(4):357-64. 171 [338] Cathcart MK, Morel DW, Chisolm GM, 3rd. Monocytes and neutrophils oxidize low density lipoprotein making it cytotoxic. Journal of leukocyte biology. 1985 Aug;38(2):341-50. [339] Sata M, Walsh K. Oxidized LDL activates fas-mediated endothelial cell apoptosis. The Journal of clinical investigation. 1998 Nov 1;102(9):1682-9. [340] Napoli C, Quehenberger O, De Nigris F, Abete P, Glass CK, Palinski W. Mildly oxidized low density lipoprotein activates multiple apoptotic signaling pathways in human coronary cells. Faseb J. 2000 Oct;14(13):1996-2007. [341] Thomas JP, Geiger PG, Girotti AW. Lethal damage to endothelial cells by oxidized low density lipoprotein: role of selenoperoxidases in cytoprotection against lipid hydroperoxide- and iron- mediated reactions. Journal of lipid research. 1993 Mar;34(3):479-90. [342] Hammad SM, Taha TA, Nareika A, Johnson KR, Lopes-Virella MF, Obeid LM. Oxidized LDL immune complexes induce release of sphingosine kinase in human U937 monocytic cells. Prostaglandins & other lipid mediators. 2006 Mar;79(1-2):126-40. [343] Morel DW, Chisolm GM. Antioxidant treatment of diabetic rats inhibits lipoprotein oxidation and cytotoxicity. Journal of lipid research. 1989 Dec;30(12):1827-34. [344] Hodis HN, Kramsch DM, Avogaro P, Bittolo-Bon G, Cazzolato G, Hwang J, et al. Biochemical and cytotoxic characteristics of an in vivo circulating oxidized low density lipoprotein (LDL-). Journal of lipid research. 1994 Apr;35(4):669-77. [345] van Vlijmen BJ, Gerritsen G, Franken AL, Boesten LS, Kockx MM, Gijbels MJ, et al. Macrophage p53 deficiency leads to enhanced atherosclerosis in APOE*3-Leiden transgenic mice. Circulation research. 2001 Apr 27;88(8):780-6. [346] Liu J, Thewke DP, Su YR, Linton MF, Fazio S, Sinensky MS. Reduced macrophage apoptosis is associated with accelerated atherosclerosis in low-density lipoprotein receptor-null mice. Arteriosclerosis, thrombosis, and vascular biology. 2005 Jan;25(1):174-9. [347] Arai S, Shelton JM, Chen M, Bradley MN, Castrillo A, Bookout AL, et al. A role for the apoptosis inhibitory factor AIM/Spalpha/Api6 in atherosclerosis development. Cell metabolism. 2005 Mar;1(3):201-13. [348] Boesten LS, Zadelaar AS, van Nieuwkoop A, Hu L, Jonkers J, van de Water B, et al. Macrophage retinoblastoma deficiency leads to enhanced atherosclerosis development in ApoE- deficient mice. Faseb J. 2006 May;20(7):953-5. [349] Merched AJ, Williams E, Chan L. Macrophage-specific p53 expression plays a crucial role in atherosclerosis development and plaque remodeling. Arteriosclerosis, thrombosis, and vascular biology. 2003 Sep 1;23(9):1608-14. [350] De Meyer GR, Kockx MM, Knaapen MW, Martinet W, De Cleen DM, Bult H, et al. Nitric oxide donor molsidomine favors features of atherosclerotic plaque stability during cholesterol lowering in rabbits. Journal of cardiovascular pharmacology. 2003 Jun;41(6):970-8. [351] Secchiero P, Candido R, Corallini F, Zacchigna S, Toffoli B, Rimondi E, et al. Systemic tumor necrosis factor-related apoptosis-inducing ligand delivery shows antiatherosclerotic activity in apolipoprotein E-null diabetic mice. Circulation. 2006 Oct 3;114(14):1522-30. [352] Croons V, Martinet W, Herman AG, Timmermans JP, De Meyer GR. Selective clearance of macrophages in atherosclerotic plaques by the protein synthesis inhibitor cycloheximide. The Journal of pharmacology and experimental therapeutics. 2007 Mar;320(3):986-93. [353] Verheye S, Martinet W, Kockx MM, Knaapen MW, Salu K, Timmermans JP, et al. Selective clearance of macrophages in atherosclerotic plaques by autophagy. Journal of the American College of Cardiology. 2007 Feb 13;49(6):706-15. [354] Ball RY, Stowers EC, Burton JH, Cary NR, Skepper JN, Mitchinson MJ. Evidence that the death of macrophage foam cells contributes to the lipid core of atheroma. Atherosclerosis. 1995 Apr 7;114(1):45-54. [355] Kolodgie FD, Narula J, Burke AP, Haider N, Farb A, Hui-Liang Y, et al. Localization of apoptotic macrophages at the site of plaque rupture in sudden coronary death. The American journal of pathology. 2000 Oct;157(4):1259-68. 172 [356] Tabas I. Consequences and therapeutic implications of macrophage apoptosis in atherosclerosis: the importance of lesion stage and phagocytic efficiency. Arteriosclerosis, thrombosis, and vascular biology. 2005 Nov;25(11):2255-64. [357] Stoneman V, Braganza D, Figg N, Mercer J, Lang R, Goddard M, et al. Monocyte/macrophage suppression in CD11b diphtheria toxin receptor transgenic mice differentially affects atherogenesis and established plaques. Circulation research. 2007 Mar 30;100(6):884-93. [358] Schrijvers DM, De Meyer GR, Kockx MM, Herman AG, Martinet W. Phagocytosis of apoptotic cells by macrophages is impaired in atherosclerosis. Arteriosclerosis, thrombosis, and vascular biology. 2005 Jun;25(6):1256-61. [359] Ait-Oufella H, Kinugawa K, Zoll J, Simon T, Boddaert J, Heeneman S, et al. Lactadherin deficiency leads to apoptotic cell accumulation and accelerated atherosclerosis in mice. Circulation. 2007 Apr 24;115(16):2168-77. [360] Thorp E, Cui D, Schrijvers DM, Kuriakose G, Tabas I. Mertk receptor mutation reduces efferocytosis efficiency and promotes apoptotic cell accumulation and plaque necrosis in atherosclerotic lesions of apoe-/- mice. Arteriosclerosis, thrombosis, and vascular biology. 2008 Aug;28(8):1421-8. [361] Ait-Oufella H, Pouresmail V, Simon T, Blanc-Brude O, Kinugawa K, Merval R, et al. Defective mer receptor tyrosine kinase signaling in bone marrow cells promotes apoptotic cell accumulation and accelerates atherosclerosis. Arteriosclerosis, thrombosis, and vascular biology. 2008 Aug;28(8):1429-31. [362] Gautier EL, Huby T, Witztum JL, Ouzilleau B, Miller ER, Saint-Charles F, et al. Macrophage apoptosis exerts divergent effects on atherogenesis as a function of lesion stage. Circulation. 2009 Apr 7;119(13):1795-804. [363] Ho QT, Kuo CJ. Vascular endothelial growth factor: biology and therapeutic applications. The international journal of biochemistry & cell biology. 2007;39(7-8):1349-57. [364] Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science (New York, NY. 1983 Feb 25;219(4587):983-5. [365] Ferrara N, Henzel WJ. Pituitary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cells. Biochemical and biophysical research communications. 1989 Jun 15;161(2):851-8. [366] Keck PJ, Hauser SD, Krivi G, Sanzo K, Warren T, Feder J, et al. Vascular permeability factor, an endothelial cell mitogen related to PDGF. Science (New York, NY. 1989 Dec 8;246(4935):1309-12. [367] Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science (New York, NY. 1989 Dec 8;246(4935):1306-9. [368] Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, et al. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature. 1996 Apr 4;380(6573):435-9. [369] Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O'Shea KS, et al. Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature. 1996 Apr 4;380(6573):439- 42. [370] Miquerol L, Langille BL, Nagy A. Embryonic development is disrupted by modest increases in vascular endothelial growth factor gene expression. Development (Cambridge, England). 2000 Sep;127(18):3941-6. [371] Gerber HP, Hillan KJ, Ryan AM, Kowalski J, Keller GA, Rangell L, et al. VEGF is required for growth and survival in neonatal mice. Development (Cambridge, England). 1999 Mar;126(6):1149-59. [372] Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocrine reviews. 2004 Aug;25(4):581-611. [373] Yang R, Thomas GR, Bunting S, Ko A, Ferrara N, Keyt B, et al. Effects of vascular endothelial growth factor on hemodynamics and cardiac performance. Journal of cardiovascular pharmacology. 1996 Jun;27(6):838-44. 173 [374] Fukumura D, Gohongi T, Kadambi A, Izumi Y, Ang J, Yun CO, et al. Predominant role of endothelial nitric oxide synthase in vascular endothelial growth factor-induced angiogenesis and vascular permeability. Proceedings of the National Academy of Sciences of the United States of America. 2001 Feb 27;98(5):2604-9. [375] Morbidelli L, Chang CH, Douglas JG, Granger HJ, Ledda F, Ziche M. Nitric oxide mediates mitogenic effect of VEGF on coronary venular endothelium. The American journal of physiology. 1996 Jan;270(1 Pt 2):H411-5. [376] Parenti A, Morbidelli L, Cui XL, Douglas JG, Hood JD, Granger HJ, et al. Nitric oxide is an upstream signal of vascular endothelial growth factor-induced extracellular signal-regulated kinase1/2 activation in postcapillary endothelium. The Journal of biological chemistry. 1998 Feb 13;273(7):4220-6. [377] Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nature medicine. 2003 Jun;9(6):669-76. [378] Alon T, Hemo I, Itin A, Pe'er J, Stone J, Keshet E. Vascular endothelial growth factor acts as a survival factor for newly formed retinal vessels and has implications for retinopathy of prematurity. Nature medicine. 1995 Oct;1(10):1024-8. [379] Gerber HP, McMurtrey A, Kowalski J, Yan M, Keyt BA, Dixit V, et al. Vascular endothelial growth factor regulates endothelial cell survival through the phosphatidylinositol 3'-kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR activation. The Journal of biological chemistry. 1998 Nov 13;273(46):30336-43. [380] Byzova TV, Goldman CK, Pampori N, Thomas KA, Bett A, Shattil SJ, et al. A mechanism for modulation of cellular responses to VEGF: activation of the integrins. Molecular cell. 2000 Oct;6(4):851-60. [381] Morales-Ruiz M, Fulton D, Sowa G, Languino LR, Fujio Y, Walsh K, et al. Vascular endothelial growth factor-stimulated actin reorganization and migration of endothelial cells is regulated via the serine/threonine kinase Akt. Circulation research. 2000 Apr 28;86(8):892-6. [382] Clauss M, Gerlach M, Gerlach H, Brett J, Wang F, Familletti PC, et al. Vascular permeability factor: a tumor-derived polypeptide that induces endothelial cell and monocyte procoagulant activity, and promotes monocyte migration. The Journal of experimental medicine. 1990 Dec 1;172(6):1535-45. [383] Barleon B, Sozzani S, Zhou D, Weich HA, Mantovani A, Marme D. Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood. 1996 Apr 15;87(8):3336-43. [384] Yoo SA, Bae DG, Ryoo JW, Kim HR, Park GS, Cho CS, et al. Arginine-rich anti-vascular endothelial growth factor (anti-VEGF) hexapeptide inhibits collagen-induced arthritis and VEGF-stimulated productions of TNF-alpha and IL-6 by human monocytes. J Immunol. 2005 May 1;174(9):5846-55. [385] Kim I, Moon SO, Kim SH, Kim HJ, Koh YS, Koh GY. Vascular endothelial growth factor expression of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and E-selectin through nuclear factor-kappa B activation in endothelial cells. The Journal of biological chemistry. 2001 Mar 9;276(10):7614-20. [386] Yoo SA, Kwok SK, Kim WU. Proinflammatory role of vascular endothelial growth factor in the pathogenesis of rheumatoid arthritis: prospects for therapeutic intervention. Mediators of inflammation. 2008;2008:129873. [387] Danese S. VEGF in inflammatory bowel disease: a master regulator of mucosal immune-driven angiogenesis. Dig Liver Dis. 2008 Aug;40(8):680-3. [388] Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF system. Nature medicine. 2003 Jun;9(6):677-84. [389] Pages G, Pouyssegur J. Transcriptional regulation of the Vascular Endothelial Growth Factor gene--a concert of activating factors. Cardiovascular research. 2005 Feb 15;65(3):564-73. [390] Gardner AM, Olah ME. Distinct protein kinase C isoforms mediate regulation of vascular endothelial growth factor expression by A2A adenosine receptor activation and phorbol esters in 174 pheochromocytoma PC12 cells. The Journal of biological chemistry. 2003 Apr 25;278(17):15421-8. [391] Pal S, Datta K, Khosravi-Far R, Mukhopadhyay D. Role of protein kinase Czeta in Ras-mediated transcriptional activation of vascular permeability factor/vascular endothelial growth factor expression. The Journal of biological chemistry. 2001 Jan 26;276(4):2395-403. [392] Birk DM, Barbato J, Mureebe L, Chaer RA. Current insights on the biology and clinical aspects of VEGF regulation. Vascular and endovascular surgery. 2008 Dec-2009 Jan;42(6):517-30. [393] Niu G, Wright KL, Huang M, Song L, Haura E, Turkson J, et al. Constitutive Stat3 activity up- regulates VEGF expression and tumor angiogenesis. Oncogene. 2002 Mar 27;21(13):2000-8. [394] Wei D, Le X, Zheng L, Wang L, Frey JA, Gao AC, et al. Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene. 2003 Jan 23;22(3):319-29. [395] Wei LH, Kuo ML, Chen CA, Chou CH, Lai KB, Lee CN, et al. Interleukin-6 promotes cervical tumor growth by VEGF-dependent angiogenesis via a STAT3 pathway. Oncogene. 2003 Mar 13;22(10):1517-27. [396] Page EL, Robitaille GA, Pouyssegur J, Richard DE. Induction of hypoxia-inducible factor- 1alpha by transcriptional and translational mechanisms. The Journal of biological chemistry. 2002 Dec 13;277(50):48403-9. [397] Blouin CC, Page EL, Soucy GM, Richard DE. Hypoxic gene activation by lipopolysaccharide in macrophages: implication of hypoxia-inducible factor 1alpha. Blood. 2004 Feb 1;103(3):1124- 30. [398] Dery MA, Michaud MD, Richard DE. Hypoxia-inducible factor 1: regulation by hypoxic and non-hypoxic activators. The international journal of biochemistry & cell biology. 2005 Mar;37(3):535-40. [399] Richard DE, Berra E, Gothie E, Roux D, Pouyssegur J. p42/p44 mitogen-activated protein kinases phosphorylate hypoxia-inducible factor 1alpha (HIF-1alpha) and enhance the transcriptional activity of HIF-1. The Journal of biological chemistry. 1999 Nov 12;274(46):32631-7. [400] Chua CC, Hamdy RC, Chua BH. Upregulation of vascular endothelial growth factor by H2O2 in rat heart endothelial cells. Free radical biology & medicine. 1998 Nov 15;25(8):891-7. [401] Castilla MA, Caramelo C, Gazapo RM, Martin O, Gonzalez-Pacheco FR, Tejedor A, et al. Role of vascular endothelial growth factor (VEGF) in endothelial cell protection against cytotoxic agents. Life sciences. 2000;67(9):1003-13. [402] Schafer G, Cramer T, Suske G, Kemmner W, Wiedenmann B, Hocker M. Oxidative stress regulates vascular endothelial growth factor-A gene transcription through Sp1- and Sp3- dependent activation of two proximal GC-rich promoter elements. The Journal of biological chemistry. 2003 Mar 7;278(10):8190-8. [403] Huez I, Creancier L, Audigier S, Gensac MC, Prats AC, Prats H. Two independent internal ribosome entry sites are involved in translation initiation of vascular endothelial growth factor mRNA. Molecular and cellular biology. 1998 Nov;18(11):6178-90. [404] Stein I, Itin A, Einat P, Skaliter R, Grossman Z, Keshet E. Translation of vascular endothelial growth factor mRNA by internal ribosome entry: implications for translation under hypoxia. Molecular and cellular biology. 1998 Jun;18(6):3112-9. [405] Crew JP, Fuggle S, Bicknell R, Cranston DW, de Benedetti A, Harris AL. Eukaryotic initiation factor-4E in superficial and muscle invasive bladder cancer and its correlation with vascular endothelial growth factor expression and tumour progression. British journal of cancer. 2000 Jan;82(1):161-6. [406] Graff JR, Zimmer SG. Translational control and metastatic progression: enhanced activity of the mRNA cap-binding protein eIF-4E selectively enhances translation of metastasis-related mRNAs. Clinical & experimental metastasis. 2003;20(3):265-73. [407] Levy NS, Chung S, Furneaux H, Levy AP. Hypoxic stabilization of vascular endothelial growth factor mRNA by the RNA-binding protein HuR. The Journal of biological chemistry. 1998 Mar 13;273(11):6417-23. 175 [408] Goldberg-Cohen I, Furneauxb H, Levy AP. A 40-bp RNA element that mediates stabilization of vascular endothelial growth factor mRNA by HuR. The Journal of biological chemistry. 2002 Apr 19;277(16):13635-40. [409] Pages G, Berra E, Milanini J, Levy AP, Pouyssegur J. Stress-activated protein kinases (JNK and p38/HOG) are essential for vascular endothelial growth factor mRNA stability. The Journal of biological chemistry. 2000 Aug 25;275(34):26484-91. [410] Langheinrich AC, Michniewicz A, Sedding DG, Walker G, Beighley PE, Rau WS, et al. Correlation of vasa vasorum neovascularization and plaque progression in aortas of apolipoprotein E(-/-)/low-density lipoprotein(-/-) double knockout mice. Arteriosclerosis, thrombosis, and vascular biology. 2006 Feb;26(2):347-52. [411] Moulton KS, Heller E, Konerding MA, Flynn E, Palinski W, Folkman J. Angiogenesis inhibitors endostatin or TNP-470 reduce intimal neovascularization and plaque growth in apolipoprotein E- deficient mice. Circulation. 1999 Apr 6;99(13):1726-32. [412] Virmani R, Kolodgie FD, Burke AP, Finn AV, Gold HK, Tulenko TN, et al. Atherosclerotic plaque progression and vulnerability to rupture: angiogenesis as a source of intraplaque hemorrhage. Arteriosclerosis, thrombosis, and vascular biology. 2005 Oct;25(10):2054-61. [413] Koga J, Matoba T, Egashira K, Kubo M, Miyagawa M, Iwata E, et al. Soluble Flt-1 gene transfer ameliorates neointima formation after wire injury in flt-1 tyrosine kinase-deficient mice. Arteriosclerosis, thrombosis, and vascular biology. 2009 Apr;29(4):458-64. [414] Lucerna M, Zernecke A, de Nooijer R, de Jager SC, Bot I, van der Lans C, et al. Vascular endothelial growth factor-A induces plaque expansion in ApoE knock-out mice by promoting de novo leukocyte recruitment. Blood. 2007 Jan 1;109(1):122-9. [415] Zhao Q, Egashira K, Inoue S, Usui M, Kitamoto S, Ni W, et al. Vascular endothelial growth factor is necessary in the development of arteriosclerosis by recruiting/activating monocytes in a rat model of long-term inhibition of nitric oxide synthesis. Circulation. 2002 Mar 5;105(9):1110- 5. [416] Inoue M, Itoh H, Ueda M, Naruko T, Kojima A, Komatsu R, et al. Vascular endothelial growth factor (VEGF) expression in human coronary atherosclerotic lesions: possible pathophysiological significance of VEGF in progression of atherosclerosis. Circulation. 1998 Nov 17;98(20):2108-16. [417] Panutsopulos D, Papalambros E, Sigala F, Zafiropoulos A, Arvanitis DL, Spandidos DA. Protein and mRNA expression levels of VEGF-A and TGF-beta1 in different types of human coronary atherosclerotic lesions. International journal of molecular medicine. 2005 Apr;15(4):603-10. [418] Ramos MA, Kuzuya M, Esaki T, Miura S, Satake S, Asai T, et al. Induction of macrophage VEGF in response to oxidized LDL and VEGF accumulation in human atherosclerotic lesions. Arteriosclerosis, thrombosis, and vascular biology. 1998 Jul;18(7):1188-96. [419] Chen YX, Nakashima Y, Tanaka K, Shiraishi S, Nakagawa K, Sueishi K. Immunohistochemical expression of vascular endothelial growth factor/vascular permeability factor in atherosclerotic intimas of human coronary arteries. Arteriosclerosis, thrombosis, and vascular biology. 1999 Jan;19(1):131-9. [420] Celletti FL, Hilfiker PR, Ghafouri P, Dake MD. Effect of human recombinant vascular endothelial growth factor165 on progression of atherosclerotic plaque. Journal of the American College of Cardiology. 2001 Jun 15;37(8):2126-30. [421] Celletti FL, Waugh JM, Amabile PG, Brendolan A, Hilfiker PR, Dake MD. Vascular endothelial growth factor enhances atherosclerotic plaque progression. Nature medicine. 2001 Apr;7(4):425- 9. [422] Celletti FL, Waugh JM, Amabile PG, Kao EY, Boroumand S, Dake MD. Inhibition of vascular endothelial growth factor-mediated neointima progression with angiostatin or paclitaxel. J Vasc Interv Radiol. 2002 Jul;13(7):703-7. [423] Moulton KS, Vakili K, Zurakowski D, Soliman M, Butterfield C, Sylvin E, et al. Inhibition of plaque neovascularization reduces macrophage accumulation and progression of advanced atherosclerosis. Proceedings of the National Academy of Sciences of the United States of America. 2003 Apr 15;100(8):4736-41. 176 [424] Petrovan RJ, Kaplan CD, Reisfeld RA, Curtiss LK. DNA vaccination against VEGF receptor 2 reduces atherosclerosis in LDL receptor-deficient mice. Arteriosclerosis, thrombosis, and vascular biology. 2007 May;27(5):1095-100. [425] Tsutsumi Y, Losordo DW. Double face of VEGF. Circulation. 2005 Aug 30;112(9):1248-50. [426] Henry TD, Annex BH, McKendall GR, Azrin MA, Lopez JJ, Giordano FJ, et al. The VIVA trial: Vascular endothelial growth factor in Ischemia for Vascular Angiogenesis. Circulation. 2003 Mar 18;107(10):1359-65. [427] Fam NP, Verma S, Kutryk M, Stewart DJ. Clinician guide to angiogenesis. Circulation. 2003 Nov 25;108(21):2613-8. [428] Howell WM, Ali S, Rose-Zerilli MJ, Ye S. VEGF polymorphisms and severity of atherosclerosis. Journal of medical genetics. 2005 Jun;42(6):485-90. [429] Asahara T, Chen D, Tsurumi Y, Kearney M, Rossow S, Passeri J, et al. Accelerated restitution of endothelial integrity and endothelium-dependent function after phVEGF165 gene transfer. Circulation. 1996 Dec 15;94(12):3291-302. [430] Shiojima I, Walsh K. The role of vascular endothelial growth factor in restenosis: the controversy continues. Circulation. 2004 Oct 19;110(16):2283-6. [431] Bhardwaj S, Roy H, Heikura T, Yla-Herttuala S. VEGF-A, VEGF-D and VEGF- D(DeltaNDeltaC) induced intimal hyperplasia in carotid arteries. European journal of clinical investigation. 2005 Nov;35(11):669-76. [432] Hedman M, Hartikainen J, Syvanne M, Stjernvall J, Hedman A, Kivela A, et al. Safety and feasibility of catheter-based local intracoronary vascular endothelial growth factor gene transfer in the prevention of postangioplasty and in-stent restenosis and in the treatment of chronic myocardial ischemia: phase II results of the Kuopio Angiogenesis Trial (KAT). Circulation. 2003 Jun 3;107(21):2677-83. [433] Ford-Hutchinson AW, Gresser M, Young RN. 5-Lipoxygenase. Annual review of biochemistry. 1994;63:383-417. [434] Stankova J, Rola-Pleszczynski M, Dubois CM. Granulocyte-macrophage colony-stimulating factor increases 5-lipoxygenase gene transcription and protein expression in human neutrophils. Blood. 1995 Jun 15;85(12):3719-26. [435] Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural, cellular, and molecular biology. Annual review of biochemistry. 2000;69:145-82. [436] Brash AR. Autoxidation of methyl linoleate: identification of the bis-allylic 11-hydroperoxide. Lipids. 2000 Sep;35(9):947-52. [437] Schneider C, Tallman KA, Porter NA, Brash AR. Two distinct pathways of formation of 4- hydroxynonenal. Mechanisms of nonenzymatic transformation of the 9- and 13-hydroperoxides of linoleic acid to 4-hydroxyalkenals. The Journal of biological chemistry. 2001 Jun 15;276(24):20831-8. [438] Neff WE, Frankel EN, Weisleder D. High-pressure liquid chromatography of autoxidized lipids. II. Hydroperoxy-cyclic peroxides and other secondary products from methyl linolenate Lipids. 1981;16 (6):439-48. [439] Frankel EN. Lipid Oxidation. Chapter 2. 2nd ed: THE OILY PRESS 2005. [440] Porter NA, Funk MO. Letter: Peroxy radical cyclization as a model for prostaglandin biosynthesis. The Journal of organic chemistry. 1975 Nov 28;40(24):3614-5. [441] Morrow JD, Hill KE, Burk RF, Nammour TM, Badr KF, Roberts LJ, 2nd. A series of prostaglandin F2-like compounds are produced in vivo in humans by a non-cyclooxygenase, free radical-catalyzed mechanism. Proceedings of the National Academy of Sciences of the United States of America. 1990 Dec;87(23):9383-7. [442] Brame CJ, Salomon RG, Morrow JD, Roberts LJ, 2nd. Identification of extremely reactive gamma-ketoaldehydes (isolevuglandins) as products of the isoprostane pathway and characterization of their lysyl protein adducts. The Journal of biological chemistry. 1999 May 7;274(19):13139-46. [443] Roberts LJ, Morrow JD. Measurement of F(2)-isoprostanes as an index of oxidative stress in vivo. Free radical biology & medicine. 2000 Feb 15;28(4):505-13. 177 [444] Salomon RG, Miller DB, Zagorski MG, Coughlin DJ. Prostaglandin endoperoxides. 14. Solvent- induced fragmentation of prostaglandin endoperoxides. New aldehyde products from PGH2 and a novel intramolecular 1,2-hydride shift during endoperoxide fragmentation in aqueous solution. Journal of the American Chemical Society. 1984;106(20):6049–60. [445] Salomon RG, Jirousek MR, Ghosh S, Sharma RB. Prostaglandin endoperoxides 21. Covalent binding of levuglandin E2 with proteins. Prostaglandins. 1987 Nov;34(5):643-56. [446] Fessel JP, Porter NA, Moore KP, Sheller JR, Roberts LJ, 2nd. Discovery of lipid peroxidation products formed in vivo with a substituted tetrahydrofuran ring (isofurans) that are favored by increased oxygen tension. Proceedings of the National Academy of Sciences of the United States of America. 2002 Dec 24;99(26):16713-8. [447] Yin H, Morrow JD, Porter NA. Identification of a novel class of endoperoxides from arachidonate autoxidation. The Journal of biological chemistry. 2004 Jan 30;279(5):3766-76. [448] Frankel EN. Secondary products of lipid oxidation. Chemistry and physics of lipids. 1987 Jul- Sep;44(2-4):73-85. [449] Frankel EN. Volatile lipid oxidation products. Progress in lipid research. 1983;22(1):1-33. [450] Schroepfer GJ, Jr. Oxysterols: modulators of cholesterol metabolism and other processes. Physiological reviews. 2000 Jan;80(1):361-554. [451] Bjorkhem I. Mechanism of degradation of the steroid side chain in the formation of bile acids. Journal of lipid research. 1992 Apr;33(4):455-71. [452] van Reyk DM, Brown AJ, Hult'en LM, Dean RT, Jessup W. Oxysterols in biological systems: sources, metabolism and pathophysiological relevance. Redox Rep. 2006;11(6):255-62. [453] Huff MW, Telford DE. Lord of the rings--the mechanism for oxidosqualene:lanosterol cyclase becomes crystal clear. Trends in pharmacological sciences. 2005 Jul;26(7):335-40. [454] Rowe AH, Argmann CA, Edwards JY, Sawyez CG, Morand OH, Hegele RA, et al. Enhanced synthesis of the oxysterol 24(S),25-epoxycholesterol in macrophages by inhibitors of 2,3- oxidosqualene:lanosterol cyclase: a novel mechanism for the attenuation of foam cell formation. Circulation research. 2003 Oct 17;93(8):717-25. [455] Wong J, Quinn CM, Brown AJ. Statins inhibit synthesis of an oxysterol ligand for the liver x receptor in human macrophages with consequences for cholesterol flux. Arteriosclerosis, thrombosis, and vascular biology. 2004 Dec;24(12):2365-71. [456] Saito Y, Yoshida Y, Niki E. Cholesterol is more susceptible to oxidation than linoleates in cultured cells under oxidative stress induced by selenium deficiency and free radicals. FEBS letters. 2007 Sep 4;581(22):4349-54. [457] Gardner HW. Oxygen radical chemistry of polyunsaturated fatty acids. Free radical biology & medicine. 1989;7(1):65-86. [458] Murphy RC, Johnson KM. Cholesterol, reactive oxygen species, and the formation of biologically active mediators. The Journal of biological chemistry. 2008 Jun 6;283(23):15521-5. [459] Niki E, Yoshida Y, Saito Y, Noguchi N. Lipid peroxidation: mechanisms, inhibition, and biological effects. Biochemical and biophysical research communications. 2005 Dec 9;338(1):668-76. [460] Chang YH, Abdalla DS, Sevanian A. Characterization of cholesterol oxidation products formed by oxidative modification of low density lipoprotein. Free radical biology & medicine. 1997;23(2):202-14. [461] Watabe T, Tsubaki A, Isobe M, Ozawa N, Hiratsuka A. A mechanism for epoxidation of cholesterol by hepatic microsomal lipid hydroperoxides. Biochimica et biophysica acta. 1984 Aug 15;795(1):60-6. [462] Spiteller G. Peroxyl radicals: inductors of neurodegenerative and other inflammatory diseases. Their origin and how they transform cholesterol, phospholipids, plasmalogens, polyunsaturated fatty acids, sugars, and proteins into deleterious products. Free radical biology & medicine. 2006 Aug 1;41(3):362-87. [463] Hazen SL, Hsu FF, Duffin K, Heinecke JW. Molecular chlorine generated by the myeloperoxidase-hydrogen peroxide-chloride system of phagocytes converts low density 178 lipoprotein cholesterol into a family of chlorinated sterols. The Journal of biological chemistry. 1996 Sep 20;271(38):23080-8. [464] van den Berg JJ, Winterbourn CC, Kuypers FA. Hypochlorous acid-mediated modification of cholesterol and phospholipid: analysis of reaction products by gas chromatography-mass spectrometry. Journal of lipid research. 1993 Nov;34(11):2005-12. [465] Dzeletovic S, Breuer O, Lund E, Diczfalusy U. Determination of cholesterol oxidation products in human plasma by isotope dilution-mass spectrometry. Analytical biochemistry. 1995 Feb 10;225(1):73-80. [466] Brown AJ, Jessup W. Oxysterols and atherosclerosis. Atherosclerosis. 1999 Jan;142(1):1-28. [467] Dzeletovic S, Babiker A, Lund E, Diczfalusy U. Time course of oxysterol formation during in vitro oxidation of low density lipoprotein. Chemistry and physics of lipids. 1995 Nov 17;78(2):119-28. [468] Brown AJ, Leong SL, Dean RT, Jessup W. 7-Hydroperoxycholesterol and its products in oxidized low density lipoprotein and human atherosclerotic plaque. Journal of lipid research. 1997 Sep;38(9):1730-45. [469] Garcia-Cruset S, Carpenter KL, Guardiola F, Stein BK, Mitchinson MJ. Oxysterol profiles of normal human arteries, fatty streaks and advanced lesions. Free radical research. 2001 Jul;35(1):31-41. [470] Ashraf MZ, Kar NS, Podrez EA. Oxidized phospholipids: biomarker for cardiovascular diseases. The international journal of biochemistry & cell biology. 2009 Jun;41(6):1241-4. [471] Leitinger N, Watson AD, Faull KF, Fogelman AM, Berliner JA. Monocyte binding to endothelial cells induced by oxidized phospholipids present in minimally oxidized low density lipoprotein is inhibited by a platelet activating factor receptor antagonist. Advances in experimental medicine and biology. 1997;433:379-82. [472] Watson AD, Leitinger N, Navab M, Faull KF, Horkko S, Witztum JL, et al. Structural identification by mass spectrometry of oxidized phospholipids in minimally oxidized low density lipoprotein that induce monocyte/endothelial interactions and evidence for their presence in vivo. The Journal of biological chemistry. 1997 May 23;272(21):13597-607. [473] Kadl A, Huber J, Gruber F, Bochkov VN, Binder BR, Leitinger N. Analysis of inflammatory gene induction by oxidized phospholipids in vivo by quantitative real-time RT-PCR in comparison with effects of LPS. Vascular pharmacology. 2002 Apr;38(4):219-27. [474] Reddy ST, Grijalva V, Ng C, Hassan K, Hama S, Mottahedeh R, et al. Identification of genes induced by oxidized phospholipids in human aortic endothelial cells. Vascular pharmacology. 2002 Apr;38(4):211-8. [475] Subbanagounder G, Wong JW, Lee H, Faull KF, Miller E, Witztum JL, et al. Epoxyisoprostane and epoxycyclopentenone phospholipids regulate monocyte chemotactic protein-1 and interleukin-8 synthesis. Formation of these oxidized phospholipids in response to interleukin- 1beta. The Journal of biological chemistry. 2002 Mar 1;277(9):7271-81. [476] Bochkov VN, Mechtcheriakova D, Lucerna M, Huber J, Malli R, Graier WF, et al. Oxidized phospholipids stimulate tissue factor expression in human endothelial cells via activation of ERK/EGR-1 and Ca(++)/NFAT. Blood. 2002 Jan 1;99(1):199-206. [477] Marathe GK, Prescott SM, Zimmerman GA, McIntyre TM. Oxidized LDL contains inflammatory PAF-like phospholipids. Trends in cardiovascular medicine. 2001 Apr-May;11(3- 4):139-42. [478] Boullier A, Gillotte KL, Horkko S, Green SR, Friedman P, Dennis EA, et al. The binding of oxidized low density lipoprotein to mouse CD36 is mediated in part by oxidized phospholipids that are associated with both the lipid and protein moieties of the lipoprotein. The Journal of biological chemistry. 2000 Mar 31;275(13):9163-9. [479] Fruhwirth GO, Loidl A, Hermetter A. Oxidized phospholipids: from molecular properties to disease. Biochimica et biophysica acta. 2007 Jul;1772(7):718-36. [480] Sinnhuber RO, Yu TC, Yu TC. Characterization of the red pigment formed in the 2-thio- barbituric acid determination of oxidative rancidity Food Res. 1958:626-33 179 [481] Chio KS, Tappel AL. Synthesis and characterization of the fluorescent products derived from malonaldehyde and amino acids. Biochemistry. 1969 Jul;8(7):2821-6. [482] Reiss U, Tappel AL. Fluorescent product formation and changes in structure of DNA reacted with peroxidizing arachidonic acid. Lipids. 1973 Apr;8(4):199-202. [483] Christopoulou CN, Perkins EG. Chromatographic studies on fatty acid dinners: Gas-liquid chromatography, high performance liquid chromatography and thin-layer chromatography J Am Oil Chem Soc 1989;66(9):1353-9. [484] Snyder JM, Frankel EN, Selke E, Warner K. Comparison of gas chromatographic methods for volatile lipid oxidation compounds in soybean oil. J Am Oil Chem Soc 1988;65(10):1617-20. [485] Selke E, Frankel EN. Dynamic headspace capillary gas chromatographic analysis of soybean oil volatiles J Am Oil Chem Soc 1987;64(5):749-53. [486] Chan HW, Levett G. Autoxidation of methyl linoleate. Separation and analysis of isomeric mixtures of methyl linoleate hydroperoxides and methyl hydroxylinoleates. Lipids. 1977 Jan;12(1):99-104. [487] Frankel EN, Evans CD, Mcconnell DG, Jones EP. Analyses of lipids and oxidation products by partition chromatography. Fatty acid hydroperoxides J Am Oil Chem Soc 1961;38(3):134-7. [488] Marquez-Ruiz G, Jorge N, Martin-Polvillo M, Dobarganes C. Rapid, quantitative determination of polar compounds in fats and oils by solid-phase extraction and size-exclusion chromatography using monostearin as internal standard Journal of chromatography. 1996;749(1-2):55-60. [489] Whitehouse CM, Dreyer RN, Yamashita M, Fenn JB. Electrospray interface for liquid chromatographs and mass spectrometers. Analytical chemistry. 1985 Mar;57(3):675-9. [490] Karas M, Hillenkamp F. Laser desorption ionization of proteins with molecular masses exceeding 10,000 daltons. Analytical chemistry. 1988 Oct 15;60(20):2299-301. [491] Singh G, Gutierrez A, Xu K, Blair IA. Liquid chromatography/electron capture atmospheric pressure chemical ionization/mass spectrometry: analysis of pentafluorobenzyl derivatives of biomolecules and drugs in the attomole range. Analytical chemistry. 2000 Jul 15;72(14):3007- 13. [492] Rentel C, Gfrorer P, Bayer E. Coupling of capillary electrochromatography to coordination ion spray mass spectrometry, a novel detection method. Electrophoresis. 1999 Sep;20(12):2329-36. [493] Harvilla CM, Hachey DL, Porter NA. Coordination (Ag+) Ion Spray−Mass Spectrometry of Peroxidation Products of Cholesterol Linoleate and Cholesterol Arachidonate: High- Performance Liquid Chromatography−Mass Spectrometry Analysis of Peroxide Products from Polyunsaturated Lipid Autoxidation. JACS. 2000;122(33):8042–55. [494] Sharma RA, Farmer PB. Biological relevance of adduct detection to the chemoprevention of cancer. Clin Cancer Res. 2004 Aug 1;10(15):4901-12. [495] Chio KS, Reiss U, Fletcher B, Tappel AL. Peroxidation of subcellular organelles: formation of lipofuscinlike fluorescent pigments. Science (New York, NY. 1969 Dec 19;166(3912):1535-6. [496] Ball RY, Brodley H, Brooks PN, Mitchinson MJ. The production of ceroid by mouse peritoneal macrophages in vitro. British journal of experimental pathology. 1984 Dec;65(6):719-24. [497] Mitchinson MJ, Hothersall DC, Brooks PN, De Burbure CY. The distribution of ceroid in human atherosclerosis. The Journal of pathology. 1985 Feb;145(2):177-83. [498] Oe T, Arora JS, Lee SH, Blair IA. A novel lipid hydroperoxide-derived cyclic covalent modification to histone H4. The Journal of biological chemistry. 2003 Oct 24;278(43):42098- 105. [499] Zhang WH, Liu J, Xu G, Yuan Q, Sayre LM. Model studies on protein side chain modification by 4-oxo-2-nonenal. Chemical research in toxicology. 2003 Apr;16(4):512-23. [500] Boutaud O, Brame CJ, Salomon RG, Roberts LJ, 2nd, Oates JA. Characterization of the lysyl adducts formed from prostaglandin H2 via the levuglandin pathway. Biochemistry. 1999 Jul 20;38(29):9389-96. [501] Fruebis J, Parthasarathy S, Steinberg D. Evidence for a concerted reaction between lipid hydroperoxides and polypeptides. Proceedings of the National Academy of Sciences of the United States of America. 1992 Nov 15;89(22):10588-92. 180 [502] Kim JG, Sabbagh F, Santanam N, Wilcox JN, Medford RM, Parthasarathy S. Generation of a polyclonal antibody against lipid peroxide-modified proteins. Free radical biology & medicine. 1997;23(2):251-9. [503] Haberland ME, Fogelman AM, Edwards PA. Specificity of receptor-mediated recognition of malondialdehyde-modified low density lipoproteins. Proceedings of the National Academy of Sciences of the United States of America. 1982 Mar;79(6):1712-6. [504] Haberland ME, Olch CL, Folgelman AM. Role of lysines in mediating interaction of modified low density lipoproteins with the scavenger receptor of human monocyte macrophages. The Journal of biological chemistry. 1984 Sep 25;259(18):11305-11. [505] Steinbrecher UP. Oxidation of human low density lipoprotein results in derivatization of lysine residues of apolipoprotein B by lipid peroxide decomposition products. The Journal of biological chemistry. 1987 Mar 15;262(8):3603-8. [506] Steinbrecher UP, Lougheed M, Kwan WC, Dirks M. Recognition of oxidized low density lipoprotein by the scavenger receptor of macrophages results from derivatization of apolipoprotein B by products of fatty acid peroxidation. The Journal of biological chemistry. 1989 Sep 15;264(26):15216-23. [507] Magil AB, Frohlich JJ, Innis SM, Steinbrecher UP. Oxidized low-density lipoprotein in experimental focal glomerulosclerosis. Kidney Int. 1993 Jun;43(6):1243-50. [508] Wieland E, Parthasarathy S, Steinberg D. Peroxidase-dependent metal-independent oxidation of low density lipoprotein in vitro: a model for in vivo oxidation? Proceedings of the National Academy of Sciences of the United States of America. 1993 Jul 1;90(13):5929-33. [509] Zhang W, Salomon RG. Oxidized phospholipids, isolevuglandins, and atherosclerosis. Molecular nutrition & food research. 2005 Nov;49(11):1050-62. [510] Zhang H, Yang Y, Steinbrecher UP. Structural requirements for the binding of modified proteins to the scavenger receptor of macrophages. The Journal of biological chemistry. 1993 Mar 15;268(8):5535-42. [511] Liao F, Berliner JA, Mehrabian M, Navab M, Demer LL, Lusis AJ, et al. Minimally modified low density lipoprotein is biologically active in vivo in mice. The Journal of clinical investigation. 1991 Jun;87(6):2253-7. [512] Basu SK, Goldstein JL, Anderson GW, Brown MS. Degradation of cationized low density lipoprotein and regulation of cholesterol metabolism in homozygous familial hypercholesterolemia fibroblasts. Proceedings of the National Academy of Sciences of the United States of America. 1976 Sep;73(9):3178-82. [513] Hazen SL, Hsu FF, Heinecke JW. p-Hydroxyphenylacetaldehyde is the major product of L- tyrosine oxidation by activated human phagocytes. A chloride-dependent mechanism for the conversion of free amino acids into reactive aldehydes by myeloperoxidase. The Journal of biological chemistry. 1996 Jan 26;271(4):1861-7. [514] Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. The Journal of biological chemistry. 1951 Nov;193(1):265-75. [515] Bligh EG, Dyer WJ. A rapid method of total lipid extraction and purification. Canadian journal of biochemistry and physiology. 1959 Aug;37(8):911-7. [516] Hajra AK. On extraction of acyl and alkyl dihydroxyacetone phosphate from incubation mixtures. Lipids. 1974 Aug;9(8):502-5. [517] Allen JW, Johnson RS, Bhatia SN. Hypoxic inhibition of 3-methylcholanthrene-induced CYP1A1 expression is independent of HIF-1alpha. Toxicology letters. 2005 Jan 15;155(1):151- 9. [518] Inoue M, Itoh H, Tanaka T, Chun TH, Doi K, Fukunaga Y, et al. Oxidized LDL regulates vascular endothelial growth factor expression in human macrophages and endothelial cells through activation of peroxisome proliferator-activated receptor-gamma. Arteriosclerosis, thrombosis, and vascular biology. 2001 Apr;21(4):560-6. [519] Salomonsson L, Pettersson S, Englund MC, Wiklund O, Ohlsson BG. Post-transcriptional regulation of VEGF expression by oxidised LDL in human macrophages. European journal of clinical investigation. 2002 Oct;32(10):767-74. 181 [520] Dandapat A, Hu C, Sun L, Mehta JL. Small concentrations of oxLDL induce capillary tube formation from endothelial cells via LOX-1-dependent redox-sensitive pathway. Arteriosclerosis, thrombosis, and vascular biology. 2007 Nov;27(11):2435-42. [521] Kanata S, Akagi M, Nishimura S, Hayakawa S, Yoshida K, Sawamura T, et al. Oxidized LDL binding to LOX-1 upregulates VEGF expression in cultured bovine chondrocytes through activation of PPAR-gamma. Biochemical and biophysical research communications. 2006 Sep 29;348(3):1003-10. [522] Gomez-Munoz A. Ceramide 1-phosphate/ceramide, a switch between life and death. Biochimica et biophysica acta. 2006 Dec;1758(12):2049-56. [523] Gupta N, Nodzenski E, Khodarev NN, Yu J, Khorasani L, Beckett MA, et al. Angiostatin effects on endothelial cells mediated by ceramide and RhoA. EMBO reports. 2001 Jun;2(6):536-40. [524] Schaeffer DF, Riazy M, Parhar KS, Chen JH, Duronio V, Sawamura T, et al. LOX-1 augments oxLDL uptake by lysophosphatidylcholine-stimulated murine macrophages but is not required for oxLDL clearance from plasma. Journal of lipid research. 2009 Apr 9. [525] Mora A, Komander D, van Aalten DM, Alessi DR. PDK1, the master regulator of AGC kinase signal transduction. Seminars in cell & developmental biology. 2004 Apr;15(2):161-70. [526] Shih SC, Mullen A, Abrams K, Mukhopadhyay D, Claffey KP. Role of protein kinase C isoforms in phorbol ester-induced vascular endothelial growth factor expression in human glioblastoma cells. The Journal of biological chemistry. 1999 May 28;274(22):15407-14. [527] Suzuma I, Suzuma K, Ueki K, Hata Y, Feener EP, King GL, et al. Stretch-induced retinal vascular endothelial growth factor expression is mediated by phosphatidylinositol 3-kinase and protein kinase C (PKC)-zeta but not by stretch-induced ERK1/2, Akt, Ras, or classical/novel PKC pathways. The Journal of biological chemistry. 2002 Jan 11;277(2):1047-57. [528] Paul A, Doherty K, Plevin R. Differential regulation by protein kinase C isoforms of nitric oxide synthase induction in RAW 264.7 macrophages and rat aortic smooth muscle cells. British journal of pharmacology. 1997 Mar;120(5):940-6. [529] Monick MM, Carter AB, Flaherty DM, Peterson MW, Hunninghake GW. Protein kinase C zeta plays a central role in activation of the p42/44 mitogen-activated protein kinase by endotoxin in alveolar macrophages. J Immunol. 2000 Oct 15;165(8):4632-9. [530] Raschke WC, Baird S, Ralph P, Nakoinz I. Functional macrophage cell lines transformed by Abelson leukemia virus. Cell. 1978 Sep;15(1):261-7. [531] Witztum JL. You are right too! The Journal of clinical investigation. 2005 Aug;115(8):2072-5. [532] Bochkov VN, Philippova M, Oskolkova O, Kadl A, Furnkranz A, Karabeg E, et al. Oxidized phospholipids stimulate angiogenesis via autocrine mechanisms, implicating a novel role for lipid oxidation in the evolution of atherosclerotic lesions. Circulation research. 2006 Oct 13;99(8):900-8. [533] Jozkowicz A, Dulak J, Piatkowska E, Placha W, Dembinska-Kiec A. Ligands of peroxisome proliferator-activated receptor-gamma increase the generation of vascular endothelial growth factor in vascular smooth muscle cells and in macrophages. Acta biochimica Polonica. 2000;47(4):1147-57. [534] Chawla A, Barak Y, Nagy L, Liao D, Tontonoz P, Evans RM. PPAR-gamma dependent and independent effects on macrophage-gene expression in lipid metabolism and inflammation. Nature medicine. 2001 Jan;7(1):48-52. [535] Walczak R, Joseph SB, Laffitte BA, Castrillo A, Pei L, Tontonoz P. Transcription of the vascular endothelial growth factor gene in macrophages is regulated by liver X receptors. The Journal of biological chemistry. 2004 Mar 12;279(11):9905-11. [536] Dulak J, Jozkowicz A, Dichtl W, Alber H, Schwarzacher SP, Pachinger O, et al. Vascular endothelial growth factor synthesis in vascular smooth muscle cells is enhanced by 7- ketocholesterol and lysophosphatidylcholine independently of their effect on nitric oxide generation. Atherosclerosis. 2001 Dec;159(2):325-32. [537] Imanishi T, Hano T, Matsuo Y, Nishio I. Oxidized low-density lipoprotein inhibits vascular endothelial growth factor-induced endothelial progenitor cell differentiation. Clinical and experimental pharmacology & physiology. 2003 Sep;30(9):665-70. 182 [538] Chavakis E, Dernbach E, Hermann C, Mondorf UF, Zeiher AM, Dimmeler S. Oxidized LDL inhibits vascular endothelial growth factor-induced endothelial cell migration by an inhibitory effect on the Akt/endothelial nitric oxide synthase pathway. Circulation. 2001 Apr 24;103(16):2102-7. [539] Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. The Biochemical journal. 2000 Oct 1;351(Pt 1):95- 105. [540] Ijichi A, Sakuma S, Tofilon PJ. Hypoxia-induced vascular endothelial growth factor expression in normal rat astrocyte cultures. Glia. 1995 Jun;14(2):87-93. [541] Stanton LW, White RT, Bryant CM, Protter AA, Endemann G. A macrophage Fc receptor for IgG is also a receptor for oxidized low density lipoprotein. The Journal of biological chemistry. 1992 Nov 5;267(31):22446-51. [542] Sawamura T, Kume N, Aoyama T, Moriwaki H, Hoshikawa H, Aiba Y, et al. An endothelial receptor for oxidized low-density lipoprotein. Nature. 1997 Mar 6;386(6620):73-7. [543] Yoshida H, Kondratenko N, Green S, Steinberg D, Quehenberger O. Identification of the lectin- like receptor for oxidized low-density lipoprotein in human macrophages and its potential role as a scavenger receptor. The Biochemical journal. 1998 Aug 15;334 ( Pt 1):9-13. [544] Kataoka H, Kume N, Miyamoto S, Minami M, Moriwaki H, Murase T, et al. Expression of lectinlike oxidized low-density lipoprotein receptor-1 in human atherosclerotic lesions. Circulation. 1999 Jun 22;99(24):3110-7. [545] Li D, Mehta JL. Upregulation of endothelial receptor for oxidized LDL (LOX-1) by oxidized LDL and implications in apoptosis of human coronary artery endothelial cells: evidence from use of antisense LOX-1 mRNA and chemical inhibitors. Arteriosclerosis, thrombosis, and vascular biology. 2000 Apr;20(4):1116-22. [546] Li D, Mehta JL. Antisense to LOX-1 inhibits oxidized LDL-mediated upregulation of monocyte chemoattractant protein-1 and monocyte adhesion to human coronary artery endothelial cells. Circulation. 2000 Jun 27;101(25):2889-95. [547] Li D, Liu L, Chen H, Sawamura T, Mehta JL. LOX-1, an oxidized LDL endothelial receptor, induces CD40/CD40L signaling in human coronary artery endothelial cells. Arteriosclerosis, thrombosis, and vascular biology. 2003 May 1;23(5):816-21. [548] Li D, Liu L, Chen H, Sawamura T, Ranganathan S, Mehta JL. LOX-1 mediates oxidized low- density lipoprotein-induced expression of matrix metalloproteinases in human coronary artery endothelial cells. Circulation. 2003 Feb 4;107(4):612-7. [549] Coller SP, Paulnock DM. Signaling pathways initiated in macrophages after engagement of type A scavenger receptors. Journal of leukocyte biology. 2001 Jul;70(1):142-8. [550] Rahaman SO, Lennon DJ, Febbraio M, Podrez EA, Hazen SL, Silverstein RL. A CD36- dependent signaling cascade is necessary for macrophage foam cell formation. Cell metabolism. 2006 Sep;4(3):211-21. [551] Walton KA, Hsieh X, Gharavi N, Wang S, Wang G, Yeh M, et al. Receptors involved in the oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine-mediated synthesis of interleukin-8. A role for Toll-like receptor 4 and a glycosylphosphatidylinositol-anchored protein. The Journal of biological chemistry. 2003 Aug 8;278(32):29661-6. [552] Ingalls RR, Heine H, Lien E, Yoshimura A, Golenbock D. Lipopolysaccharide recognition, CD14, and lipopolysaccharide receptors. Infectious disease clinics of North America. 1999 Jun;13(2):341-53, vii. [553] Miller YI, Viriyakosol S, Binder CJ, Feramisco JR, Kirkland TN, Witztum JL. Minimally modified LDL binds to CD14, induces macrophage spreading via TLR4/MD-2, and inhibits phagocytosis of apoptotic cells. The Journal of biological chemistry. 2003 Jan 17;278(3):1561-8. [554] Kim WS, Ordija CM, Freeman MW. Activation of signaling pathways by putative scavenger receptor class A (SR-A) ligands requires CD14 but not SR-A. Biochemical and biophysical research communications. 2003 Oct 17;310(2):542-9. 183 [555] Moore KJ, El Khoury J, Medeiros LA, Terada K, Geula C, Luster AD, et al. A CD36-initiated signaling cascade mediates inflammatory effects of beta-amyloid. The Journal of biological chemistry. 2002 Dec 6;277(49):47373-9. [556] Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science (New York, NY. 1998 Dec 11;282(5396):2085-8. [557] Chen JH. Oxidized low density lipoprotein-mediated macrophage survival: a role for calcium and regulation of cellular energy, Ph.D. Thesis, University of British Columbia 2009. [558] Bolland S, Ravetch JV. Spontaneous autoimmune disease in Fc(gamma)RIIB-deficient mice results from strain-specific epistasis. Immunity. 2000 Aug;13(2):277-85. [559] McGaha TL, Karlsson MC, Ravetch JV. FcgammaRIIB deficiency leads to autoimmunity and a defective response to apoptosis in Mrl-MpJ mice. J Immunol. 2008 Apr 15;180(8):5670-9. [560] Shimaoka T, Kume N, Minami M, Hayashida K, Kataoka H, Kita T, et al. Molecular cloning of a novel scavenger receptor for oxidized low density lipoprotein, SR-PSOX, on macrophages. The Journal of biological chemistry. 2000 Dec 29;275(52):40663-6. [561] Kislinger T, Fu C, Huber B, Qu W, Taguchi A, Du Yan S, et al. N(epsilon)- (carboxymethyl)lysine adducts of proteins are ligands for receptor for advanced glycation end products that activate cell signaling pathways and modulate gene expression. The Journal of biological chemistry. 1999 Oct 29;274(44):31740-9. [562] Fu MX, Requena JR, Jenkins AJ, Lyons TJ, Baynes JW, Thorpe SR. The advanced glycation end product, Nepsilon-(carboxymethyl)lysine, is a product of both lipid peroxidation and glycoxidation reactions. The Journal of biological chemistry. 1996 Apr 26;271(17):9982-6. [563] Acton SL, Scherer PE, Lodish HF, Krieger M. Expression cloning of SR-BI, a CD36-related class B scavenger receptor. The Journal of biological chemistry. 1994 Aug 19;269(33):21003-9. [564] Sun B, Boyanovsky BB, Connelly MA, Shridas P, van der Westhuyzen DR, Webb NR. Distinct mechanisms for OxLDL uptake and cellular trafficking by class B scavenger receptors CD36 and SR-BI. Journal of lipid research. 2007 Dec;48(12):2560-70. [565] Bulat N, Waeber G, Widmann C. LDLs stimulate p38 MAPKs and wound healing through SR- BI independently of Ras and PI3 kinase. Journal of lipid research. 2009 Jan;50(1):81-9. [566] Osada Y, Sunatani T, Kim IS, Nakanishi Y, Shiratsuchi A. Signalling pathway involving GULP, MAPK and Rac1 for SR-BI-induced phagocytosis of apoptotic cells. Journal of biochemistry. 2009 Mar;145(3):387-94. [567] Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free radical biology & medicine. 1991;11(1):81-128. [568] Quehenberger O, Koller E, Jurgens G, Esterbauer H. Investigation of lipid peroxidation in human low density lipoprotein. Free radical research communications. 1987;3(1-5):233-42. [569] DeCaprio AP, Olajos EJ, Weber P. Covalent binding of a neurotoxic n-hexane metabolite: conversion of primary amines to substituted pyrrole adducts by 2,5-hexanedione. Toxicology and applied pharmacology. 1982 Sep 30;65(3):440-50. [570] Fukuzawa K, Kishikawa K, Tokumura A, Tsukatani H, Shibuya M. Fluorescent pigments by covalent binding of lipid peroxidation by-products to protein and amino acids. Lipids. 1985 Dec;20(12):854-61. [571] Cheng C, Gross ML. Applications and mechanisms of charge-remote fragmentation. Mass spectrometry reviews. 2000 Nov-Dec;19(6):398-420. [572] Lou X, Spiering AJ, de Waal BF, van Dongen JL, Vekemans JA, Meijer EW. Dehydrogenation of tertiary amines in matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. J Mass Spectrom. 2008 Aug;43(8):1110-22. [573] Blair IA. DNA adducts with lipid peroxidation products. The Journal of biological chemistry. 2008 Jun 6;283(23):15545-9. [574] West JD, Marnett LJ. Alterations in gene expression induced by the lipid peroxidation product, 4-hydroxy-2-nonenal. Chemical research in toxicology. 2005 Nov;18(11):1642-53. 184 [575] Podrez EA, Hoppe G, O'Neil J, Hoff HF. Phospholipids in oxidized LDL not adducted to apoB are recognized by the CD36 scavenger receptor. Free radical biology & medicine. 2003 Feb 1;34(3):356-64. [576] Watson AD, Subbanagounder G, Welsbie DS, Faull KF, Navab M, Jung ME, et al. Structural identification of a novel pro-inflammatory epoxyisoprostane phospholipid in mildly oxidized low density lipoprotein. The Journal of biological chemistry. 1999 Aug 27;274(35):24787-98. [577] Nakamura T. Red pigment-forming substances from autoxidized linolenate: identification of prostaglandin-like substances. Lipids. 1985 Mar;20(3):180-6. [578] Spiteller G. Lipid peroxidation in aging and age-dependent diseases. Experimental gerontology. 2001 Sep;36(9):1425-57. [579] Salomon RG, Sha W, Brame C, Kaur K, Subbanagounder G, O'Neil J, et al. Protein adducts of iso[4]levuglandin E2, a product of the isoprostane pathway, in oxidized low density lipoprotein. The Journal of biological chemistry. 1999 Jul 16;274(29):20271-80. [580] Hoppe G, Subbanagounder G, O'Neil J, Salomon RG, Hoff HF. Macrophage recognition of LDL modified by levuglandin E2, an oxidation product of arachidonic acid. Biochimica et biophysica acta. 1997 Jan 7;1344(1):1-5. [581] Subbanagounder G, Salomon RG, Murthi KK, Brame C, Roberts LJ. Total synthesis of Iso[4]- levuglandin E-2. Journal of Organic Chemistry. 1997 Oct 31;62(22):7658-66. [582] Pryor WA, Stanley JP, Blair E. Autoxidation of polyunsaturated fatty acids: II. A suggested mechanism for the formation of TBA-reactive materials from prostaglandin-like endoperoxides. Lipids. 1976 May;11(5):370-9. [583] Zhang W, Sun M, Salomon RG. Preparative singlet oxygenation of linoleate provides doubly allylic dihydroperoxides: putative intermediates in the generation of biologically active aldehydes in vivo. The Journal of organic chemistry. 2006 Jul 21;71(15):5607-15. [584] Su H, Gornitsky M, Velly AM, Yu H, Benarroch M, Schipper HM. Salivary DNA, Lipid and Protein Oxidation in Non-smokers with Periodontal Disease. Free radical biology & medicine. 2009 Jan 20. [585] Iacobini C, Menini S, Ricci C, Scipioni A, Sansoni V, Mazzitelli G, et al. Advanced lipoxidation end-products mediate lipid-induced glomerular injury: role of receptor-mediated mechanisms. The Journal of pathology. 2009 Jul;218(3):360-9. [586] Leonarduzzi G, Chiarpotto E, Biasi F, Poli G. 4-Hydroxynonenal and cholesterol oxidation products in atherosclerosis. Molecular nutrition & food research. 2005 Nov;49(11):1044-9. [587] Liu Q, Raina AK, Smith MA, Sayre LM, Perry G. Hydroxynonenal, toxic carbonyls, and Alzheimer disease. Molecular aspects of medicine. 2003 Aug-Oct;24(4-5):305-13. [588] Sampey BP, Korourian S, Ronis MJ, Badger TM, Petersen DR. Immunohistochemical characterization of hepatic malondialdehyde and 4-hydroxynonenal modified proteins during early stages of ethanol-induced liver injury. Alcoholism, clinical and experimental research. 2003 Jun;27(6):1015-22. [589] Houglum K, Filip M, Witztum JL, Chojkier M. Malondialdehyde and 4-hydroxynonenal protein adducts in plasma and liver of rats with iron overload. The Journal of clinical investigation. 1990 Dec;86(6):1991-8. [590] Hartley DP, Kroll DJ, Petersen DR. Prooxidant-initiated lipid peroxidation in isolated rat hepatocytes: detection of 4-hydroxynonenal- and malondialdehyde-protein adducts. Chemical research in toxicology. 1997 Aug;10(8):895-905. [591] Chiarpotto E, Castello L, Leonarduzzi G, Biasi And F, Poli G. Role of 4-hydroxy-2,3-nonenal in the pathogenesis of fibrosis. BioFactors (Oxford, England). 2005;24(1-4):229-36.