It is the cache of ${baseHref}. It is a snapshot of the page. The current page could have changed in the meantime.
Tip: To quickly find your search term on this page, press Ctrl+F or ⌘-F (Mac) and use the find bar.

Biological Research - Nitric Oxide: Oxygen Radical Interactions in Atherosclerosis

SciELO - Scientific Electronic Library Online

 
vol.33 número2Effect of nitric oxide and plant antioxidants on microsomal content of lipid radicalsMitochondrial Function and Nitric Oxide Utilization índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Biological Research

versión impresa ISSN 0716-9760

Biol. Res. v.33 n.2 Santiago  2000

http://dx.doi.org/10.4067/S0716-97602000000200017 

Nitric Oxide - Oxygen Radicals Interactions in
Atherosclerosis

HOMERO RUBBO, CARLOS BATTHYANY AND RAFAEL RADI

Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo,
Uruguay

ABSTRACT

Atherosclerosis is one of the most common diseases and the principal cause of death in western civilization. The pathogenesis of this disease can be explained on the basis of the ‘oxidative-modification hypothesis,’ which proposes that low-density lipoprotein (LDL) oxidation represents a key early event. Nitric oxide (.NO) regulates critical lipid membrane and lipoprotein oxidation events by a) contributing to the formation of more potent secondary oxidants from superoxide (i.e.: peroxynitrite), and b) its antioxidant properties through termination reactions with lipid radicals to possibly less reactive secondary nitrogen-containing products (LONO, LOONO). Relative rates of production and steady state concentrations of superoxide and .NO and cellular sites of production will profoundly influence the expression of differential oxidant injury-enhancing and protective effects of .NO. Full understanding of the physiological roles of .NO, coupled with detailed insight into .NO regulation of oxygen radical-dependent reactions, will yield a more rational basis for intervention strategies directed toward oxidant-dependent atherogenic processes.

Key words: antioxidants, free radicals, lipid oxidation, low density lipoprotein oxidation, nitric oxide, peroxynitrite.

INTRODUCTION

Nitric oxide (.NO, nitrogen monoxide) is an endogenously-synthesized free radical first characterized as a component of endothelial-derived relaxation factor (Palmer et al., 1987). Nitric oxide is produced by a variety of mammalian cells including vascular endothelium, neurons, smooth muscle cells, macrophages, neutrophils, platelets and pulmonary epithelium (Moncada & Higgs, 1991). The physiological actions of .NO range from mediating vasodilation, neurotransmission, inhibition of platelet adherence/aggregation and the macrophage and neutrophil killing of pathogens (Moncada & Higgs, 1991). The high rate of production and broad distribution of sites of production of .NO, combined with its facile direct and indirect reactions with metalloproteins, thiols and various oxygen radical species, assures that .NO will play a central role in regulating vascular physiologic and cellular homeostasis as well as critical intravascular free radical and oxidant reactions. The multifaceted role that .NO plays in vascular disease will be emphasized during atherosclerosis, the largest single contributor to morbidity and mortality in Western countries.

A. The oxidative-modification hypothesis of atherosclerosis

Atherosclerosis is a complex disease of diverse ethiology, where the oxidation, increased deposition and altered metabolism of lipoproteins are key events associated with lesion development (Diaz et al., 1997, Ross, 1993). Functional responses of the vasculature become altered as well, resulting in impaired vasodilation and, in advanced stages of the disease, vasospasm (O’Brien & Chait, 1994). Recent evidence now compellingly reveals that reactive oxygen species are central mediators of the initiation and progression of the both structural and functional lesions characteristic of atherosclerosis. The relationship between hypercholesterolemia, elevated level of low-density lipoprotein (LDL) and premature atherosclerosis is now firmly established (Steinberg & Witztum, 1990). The cholesterol that accumulates in atherosclerotic lesions in vessel walls is derived primarily from lipoproteins, predominantly LDL (Steinberg et al., 1989). Postsecretory modifications in the structure of lipoproteins significantly affect their atherogenicity. Oxidative modification of LDL is probably the most important and is widely regarded as a critical event in the atherogenic process (Leake, 1993). Indeed, the past decade produced a series of remarkable studies that suggested that oxidative stress, particularly the oxidation of LDL, represents a risk factor and plays a key role at several steps of atherosclerosis, according to the oxidative-modification hypothesis of atherosclerosis (Westhuyzen, 1997; Witztum, 1994). This hypothesis suggests that oxidatively modified LDL (ox-LDL), but not native (unmodified) LDL, is taken by scavenger receptors on monocytes, smooth muscle cells and macrophages in the intima of blood vessel, by an unregulated process leading to the formation of lipid-laden foam cells. According to this hypothesis, LDL initially accumulates in the extracellular subendothelial space of arteries because of the augmented permeability of the endothelial cells secondary to any injury that leads to endothelial dysfunction. Through the action of resident vascular cells, LDL is mildly oxidized to a form known as minimally modified LDL (mm-LDL). This mm-LDL induces local vascular cells to produce monocyte chemotactic protein 1 (MCP-1), granulocyte-macrophage colony-stimulating factor (GM-CSF) and colony-stimulating factor 1(CSF-1), which stimulate monocyte recruitment and differentiation to macrophages in arterial walls (Parhami et al., 1993; Witztum, 1993). The accumulating activated monocytes and macrophages stimulate further peroxidation of LDL, leading to the derivatization of apolipoprotein B-100 lysine residues by products of fatty acid peroxidation (Steinbrecher et al., 1989). This completely oxidized LDL is recognized by scavenger receptors on macrophages and internalized to form foam cells (Quinn et al., 1987). In contrast to the uptake of native LDL by the LDL receptor on macrophages and other cells, the uptake of oxidized LDL by the scavenger-receptor pathway is not subject to negative-feedback regulation and thus results in massive uptake of cholesterol by macrophages (Henriksen et al., 1981).

The oxidative-modification hypothesis is supported by evidence that LDL oxidation occurs in vivo. In fact, antibodies raised against oxidized LDL react with atherosclerotic lesions but not with normal arterial segments (Yla-Herttuala et al., 1989). Patients with carotid atherosclerosis have also higher levels of autoantibodies to oxidized LDL than normal subjects (Salonen et al., 1992). This link between LDL oxidation and atherogenesis provides a convenient and simple rationale for the potential beneficial effects of antioxidants on atherosclerosis disease (Gey & Puska, 1989; Riemersma et al., 1991; Rimm et al., 1993; Stampfer et al., 1993; Stephens et al., 1996).

B. Mechanisms of LDLl oxidation in vivo.

Several pathways promote LDL oxidation in vitro, but the physiologically relevant mechanisms for LDL oxidation in vivo are still to be defined, although it is clear that in vivo LDL oxidation involves free radical chain reactions (Heinecke, 1997). Next we will discuss the mechanisms that may promote LDL oxidation in the human artery wall.

B1. Metal ions: LDL oxidation by cultured arterial cells requires micromolar concentration of iron or copper, where metal chelators block LDL oxidation by most types of cells (Heinecke et al., 1984). Tissue homogenates prepared from atherosclerotic lesions contain catalitically active metal ions, suggesting that metal ions may stimulate LDL oxidation in vivo (Swain & Gutteridge, 1995). These reactions serve to modify several properties of LDL, including electrophoretic mobility, fatty acid peroxide and thiobarbituric acid-reactive material content, the extent of apoprotein amino acid oxidation, polypeptide chain scission of apolipoprotein B and ultimately, the increased uptake, degradation and accumulation of modified LDL by macrophages (Steinbrecher et al., 1984, Batthyany et al., 2000). Lipid peroxidation propagation reactions have a critical requirement for metal catalysis. Existing "seeded" lipoprotein and vascular cell lipid hydroperoxides (LOOH) play a key role in these oxidative processes and their consequences by giving rise to a variety of reactive radical species (i.e.: lipid peroxyl (LOO.) and alkoxyl (LO.) radicals) and secondary breakdown products (i.e.: reactive aldehydes) which will react with primary amines to yield fluorescent Schiff’s base products (Fruebis et al., 1992). This resultant oxidized lipoprotein product is the more anionic species, which becomes a ligand recognized by macrophage scavenger receptor(s) (Lamb et al., 1995).

B2. Lipoxygenase and myeloperoxidase: The observation that LDL may be oxidatively modified by incubation with soybean lipoxygenase and phospholipase A2 (Sparrow et al., 1988), coupled with data showing that lipoxygenase inhibitors prevent LDL oxidation by endothelial cells or macrophages (Parthasarathy et al., 1989), suggests that cellular lipoxygenases are critically involved in oxidative modification of LDL. LDL exposed to fibroblasts transfected with the gene for 15-lipoxygenase exhibited increased levels of lipid hydroperoxides and both 15-lipoxygenase mRNA and protein have been detected in human atherosclerotic lesions (Benz et al., 1995; Yla-Herttuala et al., 1990). Recent studies (Kuhn et al., 1997), have demonstrated a significant enrichment of the stereospecific isomer 13S-hydroxy-9Z,11E-octadecadienoic acid in lipids extracted from human atherosclerotic lesions, suggesting that 15-lipoxygenase may oxidize LDL in vivo.

Myeloperoxidase is a heme protein secreted in the lesion by activated neutrophiles, and catalitically active myeloperoxidase is a component of human atherosclerotic tissue, where it colocalizes with foamy macrophages in the cellular rich regions of lesions (Daugherty et al., 1994). Oxidation products of the enzyme have been detected by immunochemistry in atherosclerotic vascular lesions (Hazell et al., 1996), suggesting that myeloperoxidase promotes LDL oxidation in vivo. Recent studies demonstrate that myeloperoxidase converts tyrosine to 3-chlorotyrosine, a stable product that may therefore serve as a molecular fingerprint for the action of the enzyme (Leeuwenburgh et al., 1997). It was recently shown that the 3-chlorotyrosine level was six times greater in atherosclerotic lesion than in normal aortic tissue, and 30 fold higher than in circulating LDL (Hazen & Heinecke, 1997). These results provide evidence that myeloperoxidase constitutes a mechanism for LDL oxidation in vivo.

B3. Reactive nitrogen species: Nitric oxide reacts with superoxide (O2.) to form peroxynitrite (ONOO- ), a reactive nitrogen species that promotes LDL oxidation. The radical-radical reaction between O2.- and .NO is extremely fast and almost diffusionally limited in rate (~1010 M-1.s-1) (Kissner et al., 1997), giving rise to significant quantities of a molecule with strong oxidizing properties. Peroxynitrite is also a nitrating agent: this is the addition of a nitro group (-NO2) to a biomolecule. It reacts with tyrosine in vitro to yield the stable product 3-nitrotyrosine, which can then be used as a fingerprint for ONOO- reaction in tissues. Monoclonal and polyclonal antibodies to nitrotyrosine formation show immunoreactivity in fatty streaks of coronary arteries of young autopsy subjects (Beckmann et al., 1994). In older patients, nitrotyrosine immunoreactivity is found in close association with foam cells, vascular endothelium and in the neointima of advanced atherosclerotic lesions. We should note that myeloperoxidase may also play a role in nitrotyrosine formation, because it can convert nitrite, the autoxidation product of .NO, to a reactive intermediate that nitrates the aromatic ring of tyrosine and proteins in vitro (Eiserich et al., 1998). In fact, the enzyme catalyzes aromatic nitration in media containing nitrite concentrations approximating those found in biologic fluids, confirming that this major .NO metabolite can serve as a physiological substrate for myeloperoxidase in the presence of plasma levels of chloride (Eiserich et al., 1998). It has been demonstrated that reactive nitrogen species generated by the myeloperoxidase/hydrogen peroxide/nitrite system of monocytes convert LDL to a form (NO2-LDL) that is avidly taken up and degraded by macrophages, leading to massive cholesterol deposition and foam cell formation (Podrez et al., 1999).

C) Antioxidant properties of nitric oxide in atherosclerosis

Nitric oxide has many physiological actions that can be interpreted to be potentially antiatherosclerotic (Boger et al., 1998; Bult, 1996). It inhibits 1) platelet aggregation and adherence to endothelial cells, 2) monocyte adherence to endothelial cells, 3) the expression of the monocyte chemoattractant protein, 4) vascular smooth muscle cell migration and proliferation and 5) the in vivo intimal proliferative response to ballon injury. Nitric oxide reduces oxidant stress in the vascular wall, which in turn may lower the rate of LDL oxidation and the expression of redox-sensitive genes that contribute to atherogenesis. In fact, vascular .NO either suppresses the expression of adhesion molecules by endothelial cells or the generation of products that are chemotactic for monocytes such as oxidized LDL. There is accumulating evidence that the salutary effects of .NO are diminished in atherosclerotic vessels due to its reactions with reactive oxygen species. In particular, the reaction of .NO with O2•-, as well as its reaction with LO. and LOO. to inhibit lipid oxidation, suggests that .NO can both enhance and inhibit lipoprotein oxidation in the vessel wall. The removal of .NO from the vascular compartment by its rapid reactions with these free radical species will concomitantly lower its steady state concentration, thus increasing platelet and inflammatory cell adhesion to the vessel wall and impairing endothelial-dependent mechanisms of relaxation. The following sections develop these concepts in more detail.

C1. Nitric oxide reaction with lipid peroxyl radicals. Nitric oxide has been observed to play a critical role in regulating lipid oxidation induced by reactive oxygen and nitrogen species (Rubbo et al., 1994; Rubbo et al., 2000). Lipid reactions of .NO are an important area of focus for multiple reasons. First, this reactive species significantly concentrates in lipophilic cell compartments, with an n-octanol:water partition coefficient of 6-8:1. This property will further enhance the ability of .NO to regulate oxidant-induced membrane lipid oxidation. Second, .NO reacts with LO. and LOO. at near diffusion-limited rates (Padmaja & Huie, 1993), inferring that both lipid peroxidation processes and reactions of lipophilic antioxidants will be influenced by local .NO concentrations.

Nitric oxide has been reported to have contrasting effects on LDL oxidation, with the pro-oxidant versus antioxidant outcome of .NO extremely dependent on relative concentrations of individual reactive species. For both macrophage and endothelial cell model systems, increased rates of cell .NO production via cytokine-mediated stimulation of inducible macrophage nitric oxide synthase gene expression and activity or exogenous addition of .N have been shown to inhibit cell and O2.--mediated lipoprotein oxidation (Hogg et al., 1993; Jessup et al., 1992; Rubbo et al., 1995). In contrast to these examples, the simultaneous production of .NO and O2.- by 1,3-morpholino-sydnonimine-HCl (SIN-1) or the direct addition of ONOO- has been shown to oxidize lipoproteins to potentially atherogenic forms (Darley-Usmar et al., 1992). It has also been shown that either ONOO- or the myeloperoxidase/hydrogen peroxide/nitrite system depletes LDL of native antioxidants and converts the LDL to a form readily recognized by macrophage scavenger receptors (Graham et al., 1993; Podrez et al., 1999, Panasenko et al., 2000). We have learned that LDL oxidation is inhibited by .NO via the termination of lipid radical-mediated chain propagation reactions (Rubbo et al., 1995). This reaction is now shown to occur both in vitro and in lipid extracts of atherosclerotic vascular lesions, in the major oxidizable lipid in LDL, cholesteryl linoleate, yielding nitrogen-containing oxidized lipid derivatives (unpublished data). Some nitrogen-containing lipid intermediates appear to be highly unstable and may decompose to reinitiate radical processes. In particular, the product of the LOO·/.NO combination reaction (LOONO) may be cleaved by homolysis to LO. and .NO2 with rearrangement of LO. to an epoxyallylic acid radical L(O). followed by recombination of L(O). with .NO2 (Odonnell et al, 1999). These observations taken together with the fact that .NO can diffuse into the hydrophobic core of the LDL particle (unpublished data) are in agreement with our hypothesis that .NO can represent the major lipophilic antioxidant in LDL.

C2. Nitric oxide and a-tocopherol inhibition of lipid oxidation. a-Tocopherol (a-TH), a lipophilic chain-breaking antioxidant in biological membranes and lipoproteins, acts by donating hydrogen atoms to chain-propagating LOO·to form the corresponding LOOH (Liebler, 1993). Since the reaction of LOO.with a-TH occurs at a rate three orders of magnitude less than for the reaction of LOO.with .NO, .NO could act more readily than or in concert with a-TH, lycopene, retinyl derivatives and ß-carotene as an antioxidant defense against oxygen radical-derived oxidized lipid species. In fact, based on a comparison of relative rate constants, it is predicted that the termination of LOO by NO (k=2.0 x 109 M-1. s-1) will be significantly more facile than the reaction of LOO.with a-tocopherol (k=2.5 x 106 M-1. s-1), thus protecting a-TH from oxidation (Rubbo et al., 2000). Because of a high reactivity with other radical species, a relatively lower reactivity of lipid radical-.NO termination products and an ability of .NO to readily traverse membranes and lipoproteins, .NO can effectively terminate radical species throughout all aspects of membrane and lipoprotein microenvironments. This can also help spare other tissue antioxidant defenses as well during periods of oxidant stress.


Figure 1. Pro- and antioxidant fates of nitric oxide on low density lipoprotein oxidation

C3. Nitric oxide and mechanisms underlying impaired vasomotor responses in atherosclerosis. Endothelium plays an important role in maintaining vascular integrity by the synthesis and release of vasoactive substances such as .NO. The changes that occur during atherosclerosis include the loss of the control of vascular tone, a .NO-dependent event. The mechanisms accounting for endothelial dysfunction in hypercholesterolemia have not been completely elucidated, but may be explained by decreased bioavailability of .NO due to either decreased expression of the eNOS, decreased substrate availability, presence of an endogenous eNOS inhibitor or increased .NO degradation by reactive oxygen-and nitrogen species (Busse & Fleming, 1996).

Dietary L-arginine and other strategies for the enhancement of vessel wall .NO synthesis have been shown to be antiatherogenic in this vascular disease associated with excess production and reactions of reactive oxygen species. In fact, both animal models and clinical studies show that chronic administration of L-arginine restores endogenous .NO production, improves endothelial dependent relaxation, decreases inflammatory cell accumulation at the vessel wall and reduces intimal hyperplasia, all hallmarks of atherosclerotic disease (Boger et al., 1998; Clarkson et al., 1996; Cooke, 1998; Davies et al., 1995; Hayward & Lefer, 1998; Wang et al., 1994). Furthermore, balloon angioplasty is often used to treat atherosclerotic vaso occlusive problems. Both the administration of .NO donors and the transfection of constitutive nitric oxide synthase to balloon-injured vessels reduce intimal cell hyperplasia, often the cause for repeat angioplasty, aortocoronary bypass graft surgery or myocardial infarction (Hoshida et al., 1996).

Antioxidants can improve endothelium-dependent vasodilation in humans with atherosclerosis documented by coronary angiography. In fact, acute administration of superoxide dismutase or a combination of lovastatin with probucol for one year reverts the abnormal vasoconstrictor response of the coronary arteries to acetylcholine in atherosclerotic patients (Anderson et al., 1995). In essential hypertensive patients, impaired endothelial vasodilation can also be improved by intrabrachial administration of ascorbic acid (vitamin C), the main water-soluble antioxidant in human plasma, an effect that can be reversed by nitric oxide synthase inhibitors, supporting the hypothesis that .NO inactivation by oxidant species contributes to endothelial dysfunction (Taddei et al., 1998). In addition, the administration of ascorbic acid improves endothelium-dependent vasodilatation in patients with hypercholesterolaemia in the absence of clinical evidence of atherosclerosis (Ting et al., 1997). Intracellular ascorbic acid is able to enhance .NO synthesis in endothelial cells, and this may explain, in part, its beneficial vascular effects (Heller et al., 1999; Huang et al., 2000).

CONCLUSIONS

The recent observations of a) the potent inhibitory effects of .NO towards platelet function and neutrophil margination on the vessel wall, b) the extremely fast and direct reactivity of.NO with oxidizing lipids and O2.-, c) the tenuous balance between O2.-, oxidized lipoproteins and .NO in regulating endothelial-dependent relaxation, and d) the diversity of pro-atherogenic oxidizing events that occur in the vascular compartment all strongly support a central role for .NO in regulating vascular atherogenic processes. The reaction between .NO and O2.- in the vasculature has the combined effect of eliminating a putative antioxidant (.NO) while at the same time generating a potent oxidant (ONOO-). Since the rate constant for .NO reaction with LOO. is greater than that for a-TH reaction with LOO. and the ability of .NO to concentrate in membranes, .NO can act more readily than or in concert with lipophilic antioxidants as an adjunct antioxidant defense against oxygen radical-derived oxidized lipid species. The anti-atherogenic and endothelial-dependent relaxation restoring effects observed following L-arginine dietary supplementation provides strong support for these concepts. Future directions may include the development of novel pharmacological strategies against atherosclerosis, investigating the antioxidant capacity of various compounds. Special attention will be paid to the direct reactions and indirect protective effects that antioxidants exert toward both lipids and lipoproteins through different mechanisms, such as .NO donors, ONOO- scavengers, and superoxide dismutase mimetics.

ACKNOWLEDGMENTS

This work was supported from CONICYT, Parke-Davies Laboratory; Fundación Manuel Pérez, Facultad de Medicina and PEDECIBA

Corresponding Author:Dr. Homero Rubbo. Departamento de Bioquímica, Facultad de Medicina. General Flores 2125, 11800 Montevideo, Uruguay. Phone: 5982-9249561 Fax: 5982-9249563 E-mail: hrubbo@fmed.edu.uy

Received: October 1, 1999. Accepted: January 10, 2000

REFERENCES

ANDERSON TJ, MEREDITH IT, YEUNG AC, FREI B, SELWYN AP, GANZ P (1995) The effect of cholesterol-lowering and antioxidant therapy on endothelium-dependent coronary vasomotion. N Engl J Med 332: 488-493         [ Links ]

BATTHYANY C., SANTOS C., BOTTI M., CERVEÑANSKY E., RADI R., AUGUSTO H., RUBBO M. (2000)( Direct evidence for APO b-100 mediated copper reduction: studeis. Arch Biochem Biophys (in press)         [ Links ]

BECKMANN JS, YE YZ, ANDERSON PG, CHEN J, ACCAVITTI MA, TARPEY MM, WHITE CR (1994) Extensive nitration of protein tyrosines in human atherosclerosis detected by immunohistochemistry. Biol Chem Hoppe Seyler 375: 81-88         [ Links ]

BENZ DJ, MOL M, EZAKI M, MORI-ITO N, ZELAN I, MIYANOHARA A, FRIEDMANN T, PARTHASARATHY S, STEINBERG D, WITZTUM JL (1995) Enhanced levels of lipoperoxides in low density lipoprotein incubated with murine fibroblast expressing high levels of human 15-lipoxygenase. J Biol Chem 270: 5191-5197         [ Links ]

BOGER RH, BODE-BOGER SM, KIENKE S, STAN AC, NAFE R., FROLICH JC (1998) Dietary L-arginine decreases myointimal cell proliferation and vascular monocyte accumulation in cholesterol-fed rabbits. Atherosclerosis 136: 67-77         [ Links ]

BULT H (1996) Nitric oxide and atherosclerosis: possible implications for therapy. Mol Med Today 2: 510-518         [ Links ]

BUSSE R, FLEMING I (1996) Endothelial dysfunction in atherosclerosis. J Vasc Res 33: 181-194         [ Links ]

CLARKSON P, ADAMS MR, POWE AJ, DONALD AE, MCCREDIE R, ROBINSON J, MCCARTHY SN, KEECH A, CELERMAJER DS, DEANFIELD JE (1996) Oral L-arginine improves endothelium-dependent dilation in hypercholesterolemic young adults. J Clin Invest 97: 1989-1994         [ Links ]

COOKE JP (1998) Is atherosclerosis an arginine deficiency disease? J Invest Med 46: 377-380         [ Links ]

DARLEY-USMAR VM, HOGG N, OLEARY V, WILSON MT, MONCADA S (1992) The simultaneous generation of superoxide and nitric oxide can initiate lipid peroxidation in human low density lipoprotein. Free Radic Res Commun 17: 9-20         [ Links ]

DAUGHERTY A, DUNN JL, RATERI DL, HEINECKE JW (1994) Myeloperoxidase, a catalyst for lipoprotein oxidation, is expressed in human atherosclerotic lesions. J Clin Invest 94: 437-444         [ Links ]

DAVIES MG, DALEN H, KIM JH, BARBER L, SVENDSEN E, HAGEN PO (1995) Control of accelerated vein graft atheroma with the nitric oxide precursor: L-arginine. J Surg Res 59: 35-42         [ Links ]

DIAZ MN, FREI B, VITA JA, KEANEY JF JR (1997) Antioxidants and atherosclerotic heart disease. N Engl J Med 337: 408-416         [ Links ]

EISERICH JP, HRISTOVA M, CROSS CE, JONES AD, FREEMAN BA, HALLIWELL B, VAN DER VLIET A (1998) Formation of nitric oxide-derived inflammatory oxidants by myeloperoxidase in neutrophils. Nature 391: 393-397         [ Links ]

FRUEBIS J, PARTHASARATHY S, STEINBERG D (1992) Evidence for a concerted reaction between lipid hydroperoxides and polypeptides. Proc Natl Acad Sci USA 89: 10588-10592         [ Links ]

GEY KF, PUSKA P (1989) Plasma vitamins E and A inversely correlated to mortality from ischemic heart disease in cross-cultural epidemiology. Ann NY Acad Sci 570: 268-282         [ Links ]

GRAHAM A, HOGG N, KALYANARAMAN B, OLEARY V, DARLEY-USMAR V, MONCADA S (1993) Peroxynitrite modification of low-density lipoprotein leads to recognition by the macrophage scavenger receptor. FEBS Lett 330: 181-185         [ Links ]

HAYWARD R, LEFER AM (1998) L-arginine attenuates endothelial dysfunction and prolongs survival in rats subjected to traumatic shock. Endothelium 6: 71-79         [ Links ]

HAZELL LJ, ARNOLD L, FLOWERS D, WAEG G, MALLE E, STOCKER R (1996) Presence of hypochlorite-modified proteins in human atherosclerotic lesions. J Clin Invest 97: 1535-1544         [ Links ]

HAZEN SL, HEINECKE JW (1997) 3-Chlorotyrosine, a specific marker of myeloperoxidase-catalyzed oxidation, is markedly elevated in low density lipoprotein isolated from human atherosclerotic intima. J Clin Invest 99: 2075-2081         [ Links ]

HEINECKE JW (1997) Mechanisms of oxidative damage of low density lipoprotein in human atherosclerosis. Curr Opin Lipidol 8: 268-74         [ Links ]

HEINECKE JW, ROSEN H, CHAIT A (1984) Iron and copper promote modification of low density lipoprotein by human arterial smooth muscle cells in culture. J Clin Invest 74: 1890-1894         [ Links ]

HELLER R, MUNSCHER-PAULIG F, GRABNER R, TILL U (1999) L-Ascorbic acid potentiates nitric oxide synthesis in endothelial cells. J Biol Chem 274: 8254-8260         [ Links ]

HENRIKSEN T, MAHONEY EM, STEINBERG D (1981) Enhanced macrophage degradation of low density lipoprotein previously incubated with cultured endothelial cells: recognition by receptors for acetylated low density lipoproteins. Proc Natl Acad Sci USA 78: 6499-6503         [ Links ]

HOGG N, KALYANARAMAN B, JOSEPH J, STRUCK A, PARTHASARATHY S (1993) Inhibition of low-density lipoprotein oxidation by nitric oxide. Potential role in atherogenesis. FEBS Lett 334: 170-174         [ Links ]

HOSHIDA S, NISHIDA M, YAMASHITA N, IGARASHI J, HORI M, KAMADA T, KUZUYA T, TADA M (1996) Amelioration of severity of myocardial injury by a nitric oxide donor in rabbits fed a cholesterol-rich diet. J Am Coll Cardiol 27: 902-909         [ Links ]

HUANG A, VITA JA, VENEMA RC, KEANEY JF JR (2000) Ascorbic acid enhances endothelial nitric-oxide synthase activity by increasing intracellular tetrahydrobiopterin. J Biol Chem 275: 17399-17406         [ Links ]

JESSUP W, MOHR D, GIESEG SP, DEAN RT, STOCKER R (1992) The participation of nitric oxide in cell free- and its restriction of macrophage-mediated oxidation of low-density lipoprotein. Biochim Biophys Acta 1180: 73-82         [ Links ]

KISSNER R, NAUSER T, BUGNON P, LYE PG, KOPPENOL WH (1997) Formation and properties of peroxynitrite as studied by laser flash photolysis, high-pressure stopped-flow technique, and pulse radiolysis [published erratum appears in Chem Res Toxicol 1998 May;11(5):557]. Chem Res Toxicol 10: 1285-1292         [ Links ]

KUHN H, HEYDECK D, HUGOU I, GNIWOTTA C (1997) In vivo action of 15-lipoxygenase in early stages of human atherogenesis. J Clin Invest 99: 888-893         [ Links ]

LAMB DJ, MITCHINSON MJ, LEAKE DS (1995) Transition metal ions within human atherosclerotic lesions can catalyse the oxidation of low density lipoprotein by macrophages. FEBS Lett 374: 12-16         [ Links ]

LEAKE DS (1993) Oxidised low density lipoproteins and atherogenesis. Br Heart J 69: 476-478         [ Links ]

LEEUWENBURGH C, RASMUSSEN JE, HSU FF, MUELLER DM, PENNATHUR S, HEINECKE JW (1997) Mass spectrometric quantification of markers for protein oxidation by tyrosyl radical, copper, and hydroxyl radical in low density lipoprotein isolated from human atherosclerotic plaques. J Biol Chem 272: 3520-3526         [ Links ]

LIEBLER DC (1993) The role of metabolism in the antioxidant function of vitamin E. Crit Rev Toxicol 23: 147-169         [ Links ]

MONCADA S, HIGGS EA (1991) Endogenous nitric oxide: physiology, pathology and clinical relevance. Eur J Clin Invest 21: 361-374         [ Links ]

OBRIEN K, CHAIT A (1994) The biology of the artery wall in atherogenesis. Med Clin North Am 78: 41-67         [ Links ]

ODONNELL V, EISERICH J, CHUMLEY P, JABLONSKY M, RAMA N, KIRK M, BARNES S, DARLEY-USMAR V, FREEMAN BA (1999) Nitration of unsaturated fatty acids by nitric oxide-derived reactive nitrogen species peroxynitrite, nitrous acid, nitrogen dioxide and nitronium ion. Chem. Res. Toxicol. 12: 83-92.         [ Links ]

PADMAJA S, HUIE RE (1993) The reaction of nitric oxide with organic peroxyl radicals. Biochem Biophys Res Commun 195: 539-544         [ Links ]

PALMER RM, FERRIGE AG, MONCADA S (1987) Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature 327: 524-526         [ Links ]

PANASENKO OM, SHARON VS, BRIVIBA K, SIES H (2000). Interaction of peroxynitrite with carotenoids in human low density lipoproteins. Arch Biochem Biophys 373: 302-305         [ Links ]

PARHAMI F, FANG ZT, FOGELMAN AM, ANDALIBI A, TERRITO MC, BERLINER JA (1993) Minimally modified low density lipoprotein-induced inflammatory responses in endothelial cells are mediated by cyclic adenosine monophosphate. J Clin Invest 92: 471-478         [ Links ]

PARTHASARATHY S, WIELAND E, STEINBERG D (1989) A role for endothelial cell lipoxygenase in the oxidative modification of low density lipoprotein. Proc Natl Acad Sci USA 86: 1046-1050         [ Links ]

PODREZ EA, SCHMITT D, HOFF HF, HAZEN SL (1999) Myeloperoxidase-generated reactive nitrogen species convert LDL into an atherogenic form in vitro. J Clin Invest 103: 1547-1560         [ Links ]

QUINN MT, PARTHASARATHY S, FONG LG, STEINBERG D (1987) Oxidatively modified low density lipoproteins: a potential role in recruitment and retention of monocyte/macrophages during atherogenesis. Proc Natl Acad Sci USA 84: 2995-2998         [ Links ]

RIEMERSMA RA, WOOD DA, MACINTYRE CC, ELTON RA, GEY KF, OLIVER MF (1991) Risk of angina pectoris and plasma concentrations of vitamins A, C, and E and carotene. Lancet 337: 1-5         [ Links ]

RIMM EB, STAMPFER MJ, ASCHERIO A, GIOVANNUCCI E, COLDITZ GA, WILLETT WC (1993) Vitamin E consumption and the risk of coronary heart disease in men. N Engl J Med 328: 1450-1456         [ Links ]

ROSS R (1993) The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature 362: 801-809         [ Links ]

RUBBO H, RADI R, TRUJILLO M, TELLERI R, KALYANARAMAN B, BARNES S, KIRK M, FREEMAN BA (1994) Nitric oxide regulation of superoxide and peroxynitrite-dependent lipid peroxidation. Formation of novel nitrogen-containing oxidized lipid derivatives. J Biol Chem 269: 26066-26075         [ Links ]

RUBBO H, PARTHASARATHY S, BARNES S, KIRK M, KALYANARAMAN B, FREEMAN BA (1995) Nitric oxide inhibition of lipoxygenase-dependent liposome and low- density lipoprotein oxidation: termination of radical chain propagation reactions and formation of nitrogen-containing oxidized lipid derivatives. Arch Biochem Biophys 324: 15-25         [ Links ]

RUBBO H, RADI R, ANSELMI D, KIRK M, BARNES S, BUTLER J, EISERICH JP, FREEMAN BA (2000) Nitric oxide reaction with lipid peroxyl radicals spares alpha-tocopherol during lipid peroxidation. Greater oxidant protection from the pair nitric oxide/alpha-tocopherol than alpha-tocopherol/ascorbate. J Biol Chem 275: 10812-10818         [ Links ]

SALONEN JT, YLA-HERTTUALA S, YAMAMOTO R, BUTLER S, KORPELA H, SALONEN R, NYYSSONEN K, PALINSKI W, WITZTUM JL (1992) Autoantibody against oxidised LDL and progression of carotid atherosclerosis. Lancet 339: 883-887         [ Links ]

SPARROW CP, PARTHASARATHY S, STEINBERG D (1988) Enzymatic modification of low density lipoprotein by purified lipoxygenase plus phospholipase A2 mimics cell-mediated oxidative modification. J Lipid Res 29: 745-753         [ Links ]

STAMPFER MJ, HENNEKENS CH, MANSON JE, COLDITZ GA, ROSNER B, WILLETT WC (1993) Vitamin E consumption and the risk of coronary disease in women. N Engl J Med 328: 1444-1449         [ Links ]

STEINBERG D, PARTHASARATHY S, CAREW TE, KHOO JC, WITZTUM JL (1989) Beyond cholesterol. Modifications of low-density lipoprotein that increase its atherogenicity. N Engl J Med 320: 915-924         [ Links ]

STEINBERG D, WITZTUM JL (1990) Lipoproteins and atherogenesis. Current concepts. JAMA 264: 3047-3052         [ Links ]

STEINBRECHER UP, PARTHASARATHY S, LEAKE DS, WITZTUM JL, STEINBERG D (1984) Modification of low density lipoprotein by endothelial cells involves lipid peroxidation and degradation of low density lipoprotein phospholipids. Proc Natl Acad Sci USA 81: 3883-3887         [ Links ]

STEINBRECHER UP, LOUGHEED M, KWAN WC, DIRKS M (1989) Recognition of oxidized low density lipoprotein by the scavenger receptor of macrophages results from derivatization of apolipoprotein B by products of fatty acid peroxidation. J Biol Chem 264: 15216-15223         [ Links ]

STEPHENS NG, PARSONS A, SCHOFIELD PM, KELLY F, CHEESEMAN K, MITCHINSON MJ (1996) Randomised controlled trial of vitamin E in patients with coronary disease: Cambridge Heart Antioxidant Study (CHAOS). Lancet 347: 781-786         [ Links ]

SWAIN J, GUTTERIDGE JM (1995) Prooxidant iron and copper, with ferroxidase and xanthine oxidase activities in human atherosclerotic material. FEBS Lett 368: 513-515         [ Links ]

TADDEI S, VIRDIS A, GHIADONI L, MAGAGNA A, SALVETTI A (1998) Vitamin C improves endothelium-dependent vasodilation by restoring nitric oxide activity in essential hypertension. Circulation 97: 2222-2229         [ Links ]

TING HH, TIMIMI FK, HALEY EA, RODDY MA, GANZ P, CREAGER MA (1997) Vitamin C improves endothelium-dependent vasodilation in forearm resistance vessels of humans with hypercholesterolemia. Circulation 95: 2617-2622         [ Links ]

WANG BY, SINGER AH, TSAO PS, DREXLER H, KOSEK J, COOKE JP (1994) Dietary arginine prevents atherogenesis in the coronary artery of the hypercholesterolemic rabbit. J Am Coll Cardiol 23: 452-458         [ Links ]

WESTHUYZEN J (1997) The oxidation hypothesis of atherosclerosis: an update. Ann Clin Lab Sci 27: 1-10         [ Links ]

WITZTUM JL (1993) Role of oxidised low density lipoprotein in atherogenesis. Br Heart J 69: S12-18         [ Links ]

WITZTUM JL (1994) The oxidation hypothesis of atherosclerosis. Lancet 344: 793-795         [ Links ]

YLA-HERTTUALA S, PALINSKI W, ROSENFELD ME, PARTHASARATHY S, CAREW T E, BUTLER S, WITZTUM JL, STEINBERG D (1989) Evidence for the presence of oxidatively modified low density lipoprotein in atherosclerotic lesions of rabbit and man. J Clin Invest 84: 1086-1095         [ Links ]

YLA-HERTTUALA S, ROSENFELD ME, PARTHASARATHY S, GLASS CK, SIGAL E, WITZTUM JL, STEINBERG D (1990) Colocalization of 15-lipoxygenase mRNA and protein with epitopes of oxidized low density lipoprotein in macrophage-rich areas of atherosclerotic lesions. Proc Natl Acad Sci USA 87: 6959-6963         [ Links ]