Skip main navigation

Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis

Originally publishedhttps://doi.org/10.1161/CIRCRESAHA.116.309326Circulation Research. 2017;120:713–735

    Abstract

    Major reactive oxygen species (ROS)–producing systems in vascular wall include NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase, xanthine oxidase, the mitochondrial electron transport chain, and uncoupled endothelial nitric oxide (NO) synthase. ROS at moderate concentrations have important signaling roles under physiological conditions. Excessive or sustained ROS production, however, when exceeding the available antioxidant defense systems, leads to oxidative stress. Animal studies have provided compelling evidence demonstrating the roles of vascular oxidative stress and NO in atherosclerosis. All established cardiovascular risk factors such as hypercholesterolemia, hypertension, diabetes mellitus, and smoking enhance ROS generation and decrease endothelial NO production. Key molecular events in atherogenesis such as oxidative modification of lipoproteins and phospholipids, endothelial cell activation, and macrophage infiltration/activation are facilitated by vascular oxidative stress and inhibited by endothelial NO. Atherosclerosis develops preferentially in vascular regions with disturbed blood flow (arches, branches, and bifurcations). The fact that these sites are associated with enhanced oxidative stress and reduced endothelial NO production is a further indication for the roles of ROS and NO in atherosclerosis. Therefore, prevention of vascular oxidative stress and improvement of endothelial NO production represent reasonable therapeutic strategies in addition to the treatment of established risk factors (hypercholesterolemia, hypertension, and diabetes mellitus).

    Atherosclerosis remains a leading cause of death and disability. The formation of vascular plaques is a complex process involving the interaction of plasma lipids with the vascular wall and immune cells. Since the 1950s, oxidative modifications of lipids and proteins have been detected in vascular lesions and the degree of oxidation correlates with the severity of disease,1 indicating a role of oxidative stress in atherogenesis.

    Role of Reactive Oxygen Species–Producing Systems in Atherosclerosis

    A variety of important reactive oxygen species (ROS)–producing systems are present in the vascular wall, including NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase, xanthine oxidase, enzymes of the mitochondrial respiratory chain, and a dysfunctional, uncoupled endothelial NO synthase.24 Importantly, there exists cross talk between these prooxidant systems.57 NADPH oxidase can trigger endothelial NOS (eNOS) uncoupling,8 xanthine oxidase activity,9,10 and mitochondrial ROS production.1114

    NADPH Oxidases

    NADPH oxidases are expressed not only in infiltrating monocytes/macrophages but also in resident cells of the vascular wall. Consisting of 2 membrane-bound subunits (p22phox and a Nox homologue) and several cytosolic regulatory subunits, these oxidases are multisubunit enzyme complexes and produce superoxide from molecular oxygen using NADPH as the electron donor.15,16 In contrast to Nox1 and Nox2 (that require p22phox, p47phox [or NOXO1], p67phox [or NOXA1] and Rac117), Nox4 only requires p22phox and releases hydrogen peroxide instead of superoxide.18

    Three Nox isoforms are expressed in the vascular wall of mice with Nox119 and Nox420 in vascular smooth muscle cells (VSMC), Nox2,21 and Nox422,23 predominantly in endothelial cells. Recent studies have shown that Nox enzymes have differential roles in atherogenesis.24

    Genetic deletion of Nox1 in apolipoprotein E-knockout (ApoE-KO) mice reduces atherosclerosis either on an atherogenic diet25 or under the condition of streptozotocin-induced diabetes mellitus26 although some controversies exist.27 Nox1 may be especially important in diabetes mellitus–accelerated atherosclerosis as diabetic conditions lead to upregulation of this Nox isoform.26,28 Nox2 is also implicated in atherogenesis29 with some regional differences. Nox2 deficiency has no effect on atherosclerosis in the aortic sinus, but reduce atherosclerosis in the descending aorta of mice.30,31 Consistent with the atherogenic roles of Nox1 and Nox2, deficiency of p47phx, a regulatory subunit required for the activation of the both Nox isoforms, reduces atherosclerosis in mice32,33 (Table 1).

    Table 1. Role of NADPH Oxidases in Atherosclerosis

    Gene AlteredGenetic BackgroundDiet/InterventionAtherosclerosisReferences
    Nox1-/yApoE-KOAtherogenic diet25
    Nox1-/yApoE-KONormal chow/diabetes mellitus26
    Nox1-/yApoE-KOWestern diet27
    Nox2-/yApoE-KOAtherogenic diet30
    Nox2-/yApoE-KOWestern diet31
    Endothelial Nox2 TgApoE-KONormal chow±angiotensin II34
    p47phox−/−ApoE-KONormal chow32
    p47phox−/−ApoE-KONormal chow33
    p47phox−/−ApoE-KOHigh fat33
    Endothelial-Nox4 TgApoE-KOWestern diet↔ (12 w)↓ (24 w)35
    Nox4−/−ApoE-KONormal chow or high-fat diet36
    Nox4−/−LDL-R-KOHigh-fat diet37
    Nox4−/−ApoE-KONormal chow/diabetes mellitus28

    ApoE-KO indicates apolipoprotein E-knockout; LDLR-KO, low-density lipoprotein receptor-knockout; and NADPH, reduced form of nicotinamide adenine dinucleotide phosphate.

    In contrast to the proatherosclerotic role of Nox1 and Nox2, several groups have independently shown that Nox4 protects against atherosclerosis in murine models28,3537 (Table 1). The protective role of Nox4 against atherosclerosis may be explained by the following facts: (1) Nox4 releases hydrogen peroxide instead of superoxide because of spontaneous superoxide dismutation within the Nox4 enzyme.38 (2) Nox4 does not lead to peroxynitrite generation because the Nox4 product hydrogen peroxide does not interact with NO.24,38 (3) Hydrogen peroxide produced by Nox4 maintains eNOS and heme oxygenase-1 expression in the setting of vascular stress.18,39 (4) Nox4-derived hydrogen peroxide inhibits proliferation of vascular smooth muscle cells40 and prevents vascular inflammation and remodeling.28 Nevertheless, Nox4 is not universally beneficial. Detrimental roles of Nox4 have been shown in rodent models of ischemic stroke,41,42 cardiac hypertrophy,43 and diabetic cardiomyopathy.44 Like other ROS, hydrogen peroxide may have both protective and damaging functions, depending on the amounts formed, the cell type expressing the Nox enzyme and its subcellular location.

    Nox5 has been found upregulated in human atherosclerotic lesions,45 in human hypertension46 and in human diabetic nephropathy.47 The rodent genome does not contain the Nox5 gene, making it challenging to study Nox5 in experimental settings. Interestingly, transgenic mice expressing human Nox5 in a podocyte-specific manner exhibit early onset albuminuria and elevated systolic blood pressure, suggesting a role for Nox5 in disease processes.47 Nevertheless, the impact of Nox5 on atherosclerosis development remains elusive.24,48

    It is notable that Nox enzymes in both monocytes/macrophages and cells of the vascular wall are required for atherogenesis. Results from bone marrow transplantation experiments using ApoE-KO and p47phox−/− mice indicate that NADPH oxidase activity in monocytes/macrophages is essential for low-density lipoprotein (LDL) oxidation. ROS produced by NADPH oxidases in endothelial cells and smooth muscle cells, on the other hand, are involved in endothelial activation (expression of adhesion molecules and the subsequent monocyte/macrophage infiltration) and smooth muscle cell proliferation.49 Consistent with this concept, endothelial-specific Nox2 overexpression increases vascular superoxide production, endothelial vascular cell adhesion molecule-1 expression, and macrophage recruitment, but does not induce atherosclerosis in ApoE-KO mice.34

    Xanthine Oxidase

    Xanthine oxidase (XO) generates superoxide and hydrogen peroxide by using molecular oxygen as an electron acceptor.50,51 The expression and activity of endothelial XO are enhanced by proatherosclerotic stimuli such as angiotensin II treatment10 and oscillatory shear stress.9 In addition, XO can be released from the liver and the circulation XO adhere to endothelial cells by associating with endothelial glycosaminoglycans.52 The activity of both endothelial XO53 and plasma XO52 is increased in experimental atherosclerosis, as well as in human atherosclerotic plaque,54,55 suggesting a contribution of XO-derived superoxide to atherosclerosis. Inhibition of XO improves endothelium-dependent, NO-mediated vasodilation in aorta rings from hypercholesterolemic animals52 and reverses endothelial dysfunction in heavy smokers.56 XO inhibitors, such as allopurinol, tungsten,57 and febuxostat,51 reduce atherosclerosis development in ApoE-KO mice. However, definitive proof of its role in atherosclerosis remains to be established, as no data from genetically modified murine models are available.

    Mitochondria

    Normally, mitochondrial oxidative phosphorylation generates physiological levels of superoxide, which is converted to hydrogen peroxide by the manganese-dependent superoxide dismutase (MnSOD, SOD2) and subsequently by glutathione peroxidase 1 (GPx1) to water.58 Mitochondrial oxidative stress can occur under pathological conditions because of excessive ROS production or insufficient ROS detoxification. Indeed, atherosclerosis in human has been associated with mitochondrial oxidative stress.59

    The importance of mitochondrial redox balance is exemplified by a global or cardiac deletion of mitochondrial SOD2, which causes perinatal lethality because of cardiac myopathy and congestive heart failure, respectively.60,61 Heterozygous SOD2+/− knockout mice on ApoE-KO background show increased ROS levels in the mitochondria and accelerated atherogenesis at arterial branch points.62

    Uncoupled eNOS

    Under physiological conditions, eNOS produces NO, which represents a key vasoprotective factor of the endothelium.6365 Under pathological conditions associated with oxidative stress, however, eNOS may become dysfunctional.2,66,67 Oxidative stress contributes to endothelial dysfunction primarily because of rapid oxidative inactivation of NO by excess superoxide. In the second step, the persisting oxidative stress renders eNOS uncoupled (ie, uncoupling of O2 reduction from NO synthesis), such that it produces superoxide at the expense of NO. Mechanistically, deficiency of eNOS cofactor tetrahydrobiopterin (BH4), deficiency of eNOS substrate l-arginine, and eNOS S-glutathionylation are likely to be the major causes for eNOS uncoupling.66,68 Peroxynitrite and superoxide can oxidize BH4 leading to BH4 deficiency. ROS production from uncoupled eNOS has been shown in mouse models of atherosclerosis6971 and in patients with hypercholesterolemia,72 atherosclerosis,73 hypertension74 or diabetes mellitus,75 and in chronic smokers.76

    Cyclooxygenases

    Although there is evidence that cyclooxygenases may indirectly modulate vascular ROS generation,77 it is unlikely that the cyclooxygenase enzymes per se serve as sources of pathogenic ROS.78 Cyclooxygenase-1 inhibition or deletion in fact attenuates atherogenesis in mice.79,80 Experiments with cell-specific deletion of cyclooxygenase-2 in mice indicate a complex role of cyclooxygenase-2 in atherogenesis. Whereas cyclooxygenase-2 in macrophages promotes atherosclerosis development, cyclooxygenase-2 in T cells has little effect on lesion burden.81 In contrast, cyclooxygenase-2 in endothelial cells and VSMC seems to be atheroprotective.82 The beneficial effects of cyclooxygenase-2 have been attributed to prostacyclin biosynthesis,82 but prostacyclin-independent mechanisms have also been proposed.83

    Role of Antioxidant Enzymes in Atherosclerosis

    Vascular cells are equipped with a variety of antioxidant defense enzymes enabling the reduction of oxidative burden.

    Superoxide Dismutase

    There are 3 SOD isoforms expressed in mammalian tissues: (1) SOD1 (copper/zinc-SOD), which is located in the cytoplasm and in the mitochondrial intermembrane space; (2) SOD2, which is expressed in the mitochondrial matrix, and (3) SOD3 (extracellular SOD), which is found in extracellular matrix, on cell surface, and in extracellular fluids.84,85 All 3 isozymes serve key antioxidant functions by catalyzing the dismutation of superoxide into oxygen and hydrogen peroxide.4,65

    Unexpectedly, transgenic mice overexpressing SOD1 develop more pronounced atherosclerotic lesion than control mice.86 It is proposed that the effects of SOD on atherogenesis are dose dependent.87 Moderate SOD1 upregulation reduces ROS burden, whereas excessive SOD activity could enhance oxidative injury by increasing formation of distal oxidants. SOD1 overexpression generates high amount of hydrogen peroxide, which can lead to the formation proatherogenic molecules such as hydroxyl radicals or metal-associated reactive species85 (Figure 1). In supporting this concept, combined overexpression of SOD1 and catalase reduces atherosclerosis in ApoE-KO mice.88

    Figure 1.

    Figure 1. Enzymes involved in the generation and inactivation of reactive oxygen species (ROS). Superoxide anion (O2·-) can be produced in the vascular wall by NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidases (Nox1 and Nox2), xanthine oxidase, uncoupled endothelial nitric oxide synthase (eNOS), and the mitochondrial respiration chain. O2·- can be converted to hydrogen peroxide (H2O2) by the enzyme superoxide dismutase (SOD). H2O2 can undergo spontaneous conversion to hydroxyl radical (OH·) via the Fenton reaction. OH· is extremely reactive and attacks most cellular components. H2O2 can be detoxified via glutathione (GSH) peroxidase, catalase or thioredoxin (Trx) peroxidase to H2O and O2. The enzyme myeloperoxidase can use H2O2 to oxidize chloride to the strong-oxidizing agent hypochlorous acid (HOCl). HOCl can chlorinate and thereby inactivate various biomolecules including lipoproteins and the eNOS substrate l-arginine. Besides HOCl generation, myelo-peroxidase can oxidize (and thus inactivate) NO to nitrite (NO2) in the vasculature. Paraoxonase (PON) isoforms 2 and 3 can prevent mitochondrial O2·- generation. Reprinted from Li et al67 with permission of the publisher. Copyright © 2013, Elsevier.

    SOD2 is the first line of defense against superoxide as a byproduct of the mitochondrial electron transport chain. Homozygous SOD2 mutant mice die within the first 10 days of life, indicating the importance of this enzyme.60 SOD2 deficiency (SOD2+/−) leads to mitochondrial dysfunction, increased mitochondrial DNA damage, and accelerated atherosclerosis in ApoE-KO mice.62

    SOD3 is abundantly expressed in the vascular wall.89 Paradoxically, genetic deletion of SOD3 in ApoE-KO mice leads to a slight reduction in atherosclerosis after 1-month atherogenic diet, whereas no effect can be found after 3 months on the atherogenic diet or after 8 months on standard chow.90 Therefore, functional significance of SOD3 in atherogenesis is still unclear and warrants more studies in future.

    Overall, the effect of SOD enzymes on atherosclerosis is likely to be context dependent. SOD enzymes may have antiatherosclerotic effects by inhibition of oxidative damages caused by superoxide and by prevention of superoxide-mediated inactivation of NO. However, SOD may also enhance oxidative stress in case that the capacity of downstream enzymes (eg, catalase and GPx) is insufficient to detoxify SOD the product (Table 2).

    Table 2. Role of Antioxidative Enzymes in Atherosclerosis

    EnzymeGene AlteredGenetic BackgroundDiet/interventionAtherosclerosisReferences
    SOD1SOD1 TgB6Atherogenic diet86
    SOD1 TgB6Atherogenic diet+irradiation91
    SOD1 TgApoE-KONormal chow88
    SOD1 Tg+CAT TgApoE-KONormal chow88
    SOD1 TgApoE-KONormal chow+BaP92
    SOD1 Tg+CAT TgApoE-KONormal chow+BaP↓↓92
    SOD2SOD2+/−ApoE-KONormal chow62
    SOD3SOD3−/−ApoE-KONormal chow↓ (1 mo)↔ (3 mo)90
    SOD3−/−ApoE-KOAtherogenic diet↔ (8 mo)90
    CatalaseCAT TgApoE-KONormal chow88
    CAT TgApoE-KONormal chow+BaP92
    GPx1GPx1−/−B6High fat↔ (12 wk)↓ (20 wk)93
    GPx1−/−ApoE-KOWestern diet94
    GPx1−/−ApoE-KONormal chow+streptozotocin95
    GPx4GPx4 TgApoE-KONormal chow96
    PON1PON1 TgB6Atherogenic diet97
    PON1 TgApoE-KONormal chow97
    PON1−/−B6Atherogenic diet98
    PON2PON2 TgApoE-KONormal chow99
    PON2−/−B6Atherogenic diet100
    PON3PON3 TgB6Atherogenic diet101
    PON3 TgLDLR-KOWestern diet101
    Thioredoxin-2Endothelial TgApoE-KOWestern diet102

    ApoE-KO indicates apolipoprotein E-knockout; BaP, benzo(a)pyrene; GPx1, glutathione perioxidase 1; LDLR-KO, low-density lipoprotein receptor-knockout; PON, paraoxonase; and SOD, superoxide dismutase.

    Catalase

    Catalase is located exclusively in peroxisomes58 where it catalyzes the reduction of hydrogen peroxide to oxygen and water. Overexpression of catalase reduces atherosclerosis in ApoE-KO mice.88

    Although ROS indisputably participate in atherogenesis, it is likely that the relative contribution of different ROS varies in different atherosclerosis models (Table 2). This becomes apparent when SOD1 is compared with catalase in mouse atherosclerosis models. In ApoE-KO mice on high-fat diet, overexpression of catalase reduces atherosclerosis, whereas SOD1 overexpression is ineffective.88,92 It is likely that superoxide makes only a minor contribution to the atherosclerosis induced by a high-fat diet or by ApoE deficiency. It has been suggested that these atherogenic stimuli (eg, oxidized low-density lipoprotein [oxLDL]) lead to the accumulation of peroxides and not superoxide.103 In contrast, the effect of SOD1 overexpression is evident in atherosclerosis models where both hydrogen peroxide and superoxide participate in atherogenesis, such as x-ray exposure91 or treatment with the environmental pollutant benzo(a)pyrene.92

    Glutathione Peroxidases

    By reducing hydrogen peroxide to water and lipid hydroperoxides to their corresponding alcohols, glutathione peroxidases (GPx) represent the major antioxidant enzyme within many cells.104 GPx1 is the ubiquitous intracellular member of the GPx family and is expressed both in the mitochondria and in the cytoplasm. A low activity of red blood cell GPx1 is associated with an increased risk of cardiovascular events in patients with coronary artery disease (CAD).105 GPx1 deficiency increases LDL oxidation, foam cell formation, and macrophage proliferation.106 Two independent studies have shown that deficiency of GPx1 enhances atherosclerosis in ApoE-KO mice,94,95 indicating a protective role of GPx1 against atherogenesis. Consistently, seleno-organic GPx1-mimetics also reduce atherosclerotic lesions in diabetic ApoE-KO mice.107

    Similar antiatherosclerotic properties have been shown for GPx4, although this GPx enzyme differs significantly from the other GPx family members in structure, intracellular localization, and functional characteristics.108 GPx4 reduces hydrogen peroxide and a wide range of lipid hydroperoxides, including oxidized phospholipids and cholesterol hydroperoxides.96 GPx4 overexpression reduces atherosclerosis in ApoE-KO mice by preventing lipid peroxidation and oxidized lipid sensing by vascular cells.96

    Paraoxonases

    The paraoxonase family of proteins has 3 members (paraoxonase 1, paraoxonase 2, and paraoxonase 3) with overlapping and discrete esterase and lactonase activities, metabolizing/hydrolyzing arachidonic acid oxidation products (all 3 paraoxonases), organophosphates (paraoxonase 1), quorum-sensing signals of pathogenic bacteria (N-acyl-homoserine lactones by paraoxonase 2), and drugs (eg, lovastatin and spironolactone by paraoxonase 3).109 All 3 paraoxonase proteins reduce oxidative stress, decrease lipid peroxidation, and diminish atherosclerosis, but with different mechanisms.

    Paraoxonase 1 is mainly synthesized by the liver and associates with high-density lipoprotein (HDL) particles. Many of the antiatherosclerotic properties of HDL are partly attributed to the esterase, peroxidase-like, and phospholipase-like activities of paraoxonase 1.110112 HDL-associated paraoxonase 1 inhibits the formation of oxidized phospholipids and thus LDL oxidation. Overexpression of paraoxonase 197,113 reduces atherosclerosis, whereas disruption of paraoxonase 1 gene98 exacerbates atherogenesis (Table 2).

    Paraoxonase 2 is undetectable in plasma114 but abundantly expressed in the vascular wall. Paraoxonase 2 is found in intracellular structures, such as the membranes of the ER or mitochondria, where it reduces superoxide formation and ER stress signaling.115,116 Interestingly, paraoxonase 2 can translocate to the plasma membrane in response to oxidative stress where it suppresses lipid peroxidation and regulates glucosylceramide content.117 Paraoxonase 2 prevents LDL peroxidation, reduces oxidative stress in all major vascular cells,100,116 and protects against atherosclerosis in mouse models (Table 2).99,100 In humans, paraoxonase 2 expression is found decreased in plaques versus plaque-adjacent tissue, indicating that the protective effect of paraoxonase 2 could fail during atherosclerosis development.118

    Paraoxonase 3 is found both in serum and cells and prevents LDL oxidation like paraoxonase 1.119 Similar to paraoxonase 2, paraoxonase 3’s antioxidative effect results from the prevention of mitochondrial superoxide formation because of an interaction with coenzyme Q10 (ubiquinone).115,120 During the Q cycle, the unstable intermediate ubisemiquinone can donate an electron to molecular oxygen leading to superoxide production. Paraoxonase 2 and paraoxonase 3 are present in the inner mitochondrial membrane and bind to coenzyme Q10 with high affinity. The 2 paraoxonase enzymes sequester ubisemiquinone and thereby reduce mitochondrial superoxide formation.115,120,121 Paraoxonase 3 counteracts atherosclerosis in mice,101 and its expression level is reduced in vascular cells of atherosclerotic patients.122

    Thioredoxins

    The thioredoxin systems involve thioredoxin, thioredoxin reductases, and thioredoxin peroxidases.123,124 Thioredoxin can reduce target proteins via cysteine thiol-disulfide exchanges. Thioredoxin-dependent peroxidase can also directly scavenge hydrogen peroxide, and thioredoxin reductase converts oxidized thioredoxin to its reduced form to facilitate its redox activity (Figure 1). Experiments with transgenic mice have shown that both the cytosolic thioredoxin-1 and the mitochondrial thioredoxin-2 systems are essential regulators of cardiac function.123,124 For the vascular system, compelling evidence exists supporting a crucial role of the thioredoxin-2 system. Endothelial-specific deletion of thioredoxin-R2 leads to increased vascular stiffness, impaired endothelial function, and a prothrombotic, proinflammatory vascular phenotype.125 Endothelial-specific overexpression of mitochondrial thioredoxin-2 improves endothelial function and reduces atherosclerotic lesions in ApoE-KO mice.102

    Role of NO in Atherosclerosis

    Results from studies using genetically modified animals indicate that NO synthase (NOS) isoforms have different roles in atherosclerosis with eNOS and neuronal nitric oxide synthase (nNOS) being atheroprotective and inducible nitric oxide synthase (iNOS) being proatherogenic.126

    Neuronal NO Synthase

    The nNOS is expressed in not only perivascular nerve fibers but also the vascular wall127,128 and atherosclerotic plaques.129 Vascular nNOS contributes to vasodilation130 and can partly compensate the loss of eNOS in eNOS−/− mice.131133 Recent human studies suggest that nNOS plays an important role in the local regulation of vascular tone independently of its effect in the central nervous system. Inhibition of nNOS reduces the basal blood flow in human forearm and coronary circulations without affecting the eNOS-mediated vasodilatation elicited by acetylcholine, substance P, or increased shear stress,134,135 indicating distinct roles of nNOS and eNOS.

    Disruption of nNOS gene enhances neointimal formation and constrictive vascular remodeling in a mouse model of carotid artery ligation.128,136 Double knockout (DKO) mice deficient in nNOS and ApoE show markedly larger atherosclerotic lesion in the aortic root and descending thoracic aorta129,137 and increased mortality137 (Table 3).

    Table 3. Role of NO Synthases in Atherosclerosis

    EnzymeGene AlteredGenetic BackgroundDiet/interventionAtherosclerosisReferences
    nNOSnNOS−/−ApoE-KOWestern diet137
    iNOSiNOS−/−ApoE-KOWestern diet138
    eNOSeNOS−/−ApoE-KOWestern diet139141
    eNOS-TgApoE-KOWestern diet142
    eNOS-Tg xGCH1-TgApoE-KONormal chow↓ (vs eNOS-Tg)↔ (vs ApoE-KO)143

    ApoE-KO indicates apolipoprotein E-knockout; eNOS, endothelial nitric oxide synthase; iNOS, inducible nitric oxide synthase; and nNOS, neuronal nitric oxide synthase.

    Inducible NO Synthase

    The inducible isoform iNOS is normally absent in the vasculature under physiological conditions. Its expression is induced in blood vessels in pathological situations, such as inflammation, sepsis, or oxidative stress.144

    Genetic disruption of iNOS ameliorates atherosclerotic lesion in ApoE-KO mice without changing the lipid profile138 (Table 3). The reduced atherosclerosis in iNOS/ApoE DKO mice is associated with decreased plasma levels of lipoperoxides, indicating that peroxynitrite-mediated oxidative stress is likely to be involved in the proatherosclerotic effects of iNOS.138

    In contrast to the regulated production of NO by nNOS and eNOS, iNOS may generate large amounts of NO over long periods of time. If induced in endothelial cells, iNOS competes with eNOS for BH4 and thus reduces eNOS-mediated NO production by limiting BH4 availability for eNOS.145 Furthermore, iNOS induction in the vasculature facilitates the generation of peroxynitrite,146149 a key proatherosclerosis oxidant.150,151 Indeed, the expression of iNOS in human atherosclerosis plaque is associated with nitrotyrosine, a marker of peroxynitrite formation.149,152,153

    Endothelial NO Synthase

    The eNOS enzyme is constitutively expressed mainly in endothelial cells. Its activity is regulated by shear stress of the flowing blood and by agonists such as bradykinin and acetylcholine. Endothelial NO produced by eNOS induces vascular smooth muscle relaxation and inhibits platelet aggregation and adhesion.6365 In addition to these antihypertensive and antithrombotic properties, eNOS-derived NO also exerts multiple antiatherosclerotic effects, including inhibition of LDL oxidation, prevention of leukocyte adhesion to vascular endothelium and leukocyte migration into the vascular wall, and inhibition of vascular smooth muscle cell proliferation.6365 At low physiological levels, NO also acts as an antioxidant, abating fenton-type reactions, terminating radical chain reactions, and inhibiting peroxidases and oxidases (eg, by nitrosylation of allosteric thiols).154

    Consistent with the antiatherosclerotic role of eNOS-derived NO, genetic disruption of eNOS in ApoE-KO mice enhances atherosclerosis139,140 (Table 3). Unexpectedly, overexpression of eNOS also accelerates atherogenesis in ApoE-KO mice.142 This paradoxical phenomenon is explained by later findings that eNOS overexpression leads to eNOS uncoupling owing to a relative deficiency of eNOS cofactor BH4. Indeed, reversal of eNOS uncoupling by upregulating BH4 synthesis in ApoE-KO/eNOS-Tg mice leads to a reduction of atherosclerotic lesion.143

    The eNOS-KO mice are hypertensive. However, hypertension does not account for the accelerated atherosclerosis in the eNOS-KO animals. Treatment of ApoE/eNOS DKO mice with hydralazine lowers the blood pressure to levels seen in ApoE-KO mice, but does not change the extent of lesion formation in ApoE/eNOS-DKO mice. These results indicate that hypertension is not required for the accelerated atherosclerosis seen in apoE/eNOS DKO animals and that eNOS plays important roles in suppressing atherogenesis separate from blood pressure regulation.140

    Recent studies have shown that eNOS is also present in the perivascular adipose tissue. Under physiological conditions, NO derived from perivascular adipose tissue–eNOS contributes to the vasoprotective effects of perivascular adipose tissue. Under pathological conditions, however, perivascular adipose tissue–eNOS may become a superoxide-generating enzyme.155,156

    Differences Between Murine Models and Human Pathology

    Although the ApoE-KO and the LDL receptor-KO mice are excellent models for atherosclerosis research, there exist significant differences between these commonly used murine models and human pathology.157,158 For instance, (1) these mice have usually excessive blood cholesterol levels; they are not common in human patients. (2) The majority of lipoproteins are found in the HDL fraction in mice, whereas it is in LDL and very-low-density lipoprotein in humans. (3) Murine atherosclerosis studies are mostly performed in relatively young mice without aging. (4) There are significant differences between the murine and human immune systems. (5) Mice do not develop plaque rupture on a regular basis.157,158 Therefore, these differences must be considered when translating results from murine experiments into clinical settings.

    Because of the significance of such differences, mouse models for plaque rupture have been developed. The brachiocephalic artery in ApoE-deficient mice fed a high-fat diet, with or without angiotensin II infusion, is a practically feasible model for plaque rupture.159 It has been shown in this model that monocyte/macrophage-mediated oxidative stress and inflammation are likely to be involved in plaque destabilization and plaque rupture.160 Recently, a mouse model with a fibrillin-1 gene mutation on the ApoE-KO background has been described as a model of acute plaque rupture with human-like complications.161 Also in this model, lipid oxidation and iNOS-mediated inflammation are likely to play roles in the pathology.

    In both the ApoE-KO and LDL receptor-KO mice, atherogenesis is driven, at least in part, by non-HDL hyperlipidemia, although the underlying mechanisms are different.162 Because ApoE has antioxidative properties, the oxidation of lipoproteins is more prominent in apoE-KO mice than in LDL receptor-KO mice. Consistently, antibodies to oxidized LDL epitopes are especially high in the ApoE-KO mice.162 There are no data available directly comparing the levels of endothelial NO between the 2 mouse models.

    There exist sex differences in atherosclerosis,163 both in humans164 and mice.165 Atherosclerotic lesion formation and lipid accumulation in the aorta from ApoE-KO mice is more pronounced in men than women.165 These sex-dependent differences are associated with lower NADPH oxidase activity in the women.165 Moreover, ovariectomy enhances NADPH oxidase activity, lipid deposition, and atherosclerotic lesion formation, which can be recovered by administration of 17β-estradiol, indicating the role of female sex hormones.165,166 Furthermore, arteries from female animals also show higher expression of SOD1, SOD2, and eNOS.167 Estrogens have been shown to enhance eNOS expression and NO production in endothelial cells.168

    Gene Polymorphisms and Atherosclerosis in Humans

    Genome-wide association studies have identified 58 single-nucleotide polymorphisms that are genome-wide significantly associated with CAD.169,170 Ten of the CAD loci are associated with LDL and another 5 with blood pressure,170 which should not be surprising. On the contrary, the majority of the CAD genome-wide association study loci is not associated with known risk factors for CAD.170 The redox genes and NO synthase genes mentioned in this article are not among the 58 CAD genome-wide association study loci, either.

    In contrast to genome-wide association studies, hypothesis-driven candidate gene association studies have provided evidence for possible association of redox gene and eNOS gene polymorphisms with the risk of atherosclerosis (Table 4). Unfortunately, the results are often conflicting and inconclusive, partially because of the small sample size in each study or the differences in ethnicity.

    Table 4. Gene Polymorphisms and Atherosclerosis

    Enzymers NumberPolymorphismFunctional ConsequenceEffects on Atherosclerosis
    Nox2rs4673C242T (His72Tyr)Nox2 activity↓CAD ↓171173
    CAD ↑174,175
    SOD1rs9974610A→G (≈13.6 kb 5' from TSS)UnknownCV death risk ↓176
    rs10432782T→G (intron 2)UnknownCV death risk ↑176
    rs1041740C→T (intron 4)Unknown
    SOD2rs4880Ala16Val↓SOD2 into mitochondriaCAD ↑177179
    SOD3rs1799895Arg213Gly↓SOD3 tissue bindingCAD risk ↑180,181
    GPx1rs1050450Pro198LeuGPx1 activity↓IMT ↑182; CAD ↑183,184
    GPx1 activity ↔No effect185,186
    eNOSrs1799983Glu298AspeNOS activity↓CAD ↑187189
    rs 2070744-T786CeNOS expression↓CAD ↑189,190
    Intron 4 VNTReNOS expression↓?CAD ↑187,189

    CAD indicates coronary artery disease; CV, cardiovascular; GPx1, glutathione perioxidase 1; Nox2, NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase 2; SOD, superoxide dismutase; and TSS, transcription start site.

    NADPH Oxidase Subunits

    The p22phox subunit is required by Nox family members of NADPH oxidases. The Nox proteins and the p22phox protein are stable only as a heterodimer. A significant number of allelic variants have been identified within the promoter and exonic sequences of the p22phox gene.191 Among these, particular attention has been paid to the C242T polymorphism which results in replacement of histidine by tyrosine at amino acid position 72 (H72Y), a potential heme binding site.174 The 242T allele has been shown to be associated with reduced NADPH oxidase activity and respiratory burst in human neutrophils,192 and with decreased vascular NADPH oxidase activity in saphenous veins of patients with CAD, independently of other clinical risk factors.193 Although some studies indicate that the T allele may confer protection against atherosclerosis,171173 some other studies have shown negative or even opposite effects.174,175 Two recent meta-analyses have also provided conflicting results.194,195 Therefore, more studies are needed before a definitive conclusion can be drawn.

    Whereas the C242T variant is a rather common (T allele ≈20%194) genetic polymorphism, the chronic granulomatous disease is a rare (1 in 200 000–250 000 individuals) inherited disorder of the innate immune system and caused by genetic defects in the genes encoding 4 of the phox proteins (Nox2/gp91phox, p22phox, p47phox, and p67phox).191 The deficiency of Nox2 activity in patients with chronic granulomatous disease results in an improved flow-mediated arterial dilation196 and a protection of the vascular endothelium against ischemia/reperfusion-induced endothelial dysfunction in the brachial artery.197 Moreover, individuals with chronic granulomatous disease have lower carotid intimal-medial thickness198 and reduced carotid atherosclerosis,199 although coronary atherosclerosis is not changed by chronic granulomatous disease.199 All these observations support the concept that Nox2 activity contributes to atherogenesis in humans. Consistently, upregulation of Nox2 is evident in coronary arteries of human patients with atherosclerosis55,200

    SOD Enzymes

    Association studies for SOD1 has been scarce. A recent study has found that 3 variants in the SOD gene are associated with increased risk of death from cardiovascular causes (sudden death, fatal myocardial infarction, or stroke).176

    A functional polymorphism has been identified in the SOD2 gene resulting in the replacement of alanine 16 with a valine (Ala16Val) in the mitochondrial targeting domain. Human beings harboring this variant have an increased carotid intima-to-media thickness and are at increased risk for CAD and acute myocardial infarction.177179

    Genetic variants exist in both the coding region and the promoter region of SOD3 gene.201 Most researches have concentrated on the functional variant Arg213Gly in the heparin-binding domain. The Gly allele is associated with decreased SOD3 affinity for heparin, reduced tissue binding,202204 and reduced antioxidant effects in the vascular wall.204 This polymorphism has been linked to increased body weight, triglycerides, and higher cardiovascular risk180,181 but paradoxically decreased risk of lung disease.205

    GPx1

    The Pro198Leu polymorphism is a site located within the GPx1 C-terminal region. This amino acid substitution has been proposed to change structural conformation of the active site region and to modify the enzyme activity.206 GPx1 polymorphism has been associated with increased carotid intima-to-media thickness, peripheral arterial disease, and increased CAD risk.182184,207 However, other studies found no association of Pro198Leu polymorphism with stroke208 or coronary artery stenosis.185,186 The predicted tertiary structure of GPx1 shows that the C-terminal fragment containing the Pro198Leu polymorphic site is located on the protein surface and within a nonfunctional fragment.185

    eNOS

    The most intensively examined and functionally related common eNOS variants include Glu298Asp (G894T), -T786C, and the intron 4 variable number tandem repeat.187,209 Early studies suggested that eNOS protein containing 298Asp is subject to selective proteolytic cleavage.210,211 These observations, however, turned out to be artifacts.212,213 A later study has provided evidence that the Glu298Asp single-nucleotide polymorphism affects eNOS localization to caveolar membrane leading to diminished shear-dependent responses and impaired coordination of the eNOS regulatory cycle.214 A large number of genetic association studies exist addressing the impact of Glu298Asp polymorphisms on atherosclerosis risk with partly promising results.188,215217 However, negative findings have also been reported.184,218,219 Results from several meta-analyses indicate positive associations of Glu298Asp,187189 -786T>C,189,190 and Intron 4189 polymorphisms with CAD risk.

    Cardiovascular Risk Factors Induce Vascular Oxidative Stress and Decrease Endothelial NO Production

    All established risk factors for atherosclerosis enhance oxidative stress and induces eNOS uncoupling in the vascular wall.66,67 Uncoupling of eNOS leads to not only reduced endothelial NO production but also a potentiation of oxidative stress (Figure 2).

    Figure 2.

    Figure 2. Cardiovascular risk factors induce vascular oxidative stress and reduce endothelial nitric oxide (NO) production. Hypercholesterolemia, hypertension, smoking, and diabetes mellitus lead to activation of NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase, partly through mechanism dependent on protein kinase C (PKC). Diabetes mellitus also stimulates mitochondrial ROS production, which then triggers NADPH oxidase activation. NADPH oxidase can enhance superoxide (O2·-) production from mitochondria and xanthine oxidase. The endothelial NO synthase (eNOS) enzyme can become uncoupled through 2 major mechanisms: deficiency of the cofactor tetrahydrobiopterin (BH4) or of the substrate l-arginine. O2·- reacts with NO resulting in peroxynitrite (ONOO-). ONOO- oxidizes BH4 leading to BH4 deficiency. l-Arginine deficiency is caused by upregulation of arginase expression and activity, partly through RhoA/ROCK-dependent mechanisms. Uncoupled eNOS produces superoxide thereby potentiating oxidative stress. The uncoupling of eNOS decreases endothelial NO production, which is further exacerbated by reduced eNOS expression and activity. In addition to the risk factors at the population level, disturbed flow renders arterial bifurcations and side branches prone to atherosclerosis. Enhanced oxidative stress and reduced endothelial NO production contribute significantly to the enhanced atherosclerosis at these regions.

    Hypertension

    Hypertension is a major risk factor for atherosclerosis and for CAD and stroke. One of the underlying mechanisms for the enhanced atherogenesis in hypertension patients is oxidative stress. In fact, oxidative stress plays a crucial role in the pathogenesis of hypertension itself. This has been shown for variety animal models of hypertension types, including angiotensin II–induced hypertension, spontaneously hypertensive rats (an animal model of genetic hypertension), and deoxycorticosterone acetate-salt hypertension, which is a low renin/angiotensin hypertension model. Moreover, NADPH oxidase is likely to represent the primary ROS source. Genetic deletion or pharmacological inhibition of NADPH oxidase lowers blood pressure in hypertension models.8,220,221

    Uncoupled eNOS also contributes significantly to vascular oxidative stress in hypertension.66,67 Molecular mechanisms for eNOS uncoupling in hypertension include BH4 deficiency, l-arginine deficiency, and S-glutathionylation. The deficiency of BH4 is caused by NADPH oxidase–mediated of BH4 oxidation8 and by reduced BH4 recycling from BH2 because of a downregulation of endothelial dihydrofolate reductase.222l-Arginine deficiency in hypertension models has been attributed to upregulation of arginase expression/activity in blood vessels.223225 Uncoupling of eNOS by S-glutathionylation is evident in angiotensin II–induced hypertension.226 Reversal of eNOS uncoupling reduces blood pressure in hypertensive animals221 or contributes to blood pressure reduction by some antihypertensive drugs.226

    Hypercholesterolemia

    NADPH oxidases and XO are proposed to be the major sources of superoxide in the coronary artery of hypercholesterolemic patients with CAD.55 Uncoupling of eNOS is likely to be a subsequent event secondary to oxidative stress mediated by NADPH oxidase (and XO) because of oxidation-induced BH4 deficiency. Both native LDL and oxLDL have been shown to stimulate superoxide/peroxynitrite production and to uncouple eNOS.227,228 ROS production from uncoupled eNOS has been shown in LDL-treated endothelial cells, in hypercholesterolemic ApoE-KO mice71 and in hypercholesterolemic patients.72 In addition to the direct effects of LDL on endothelial ROS production, hypercholesterolemia may indirectly enhance oxidative stress by potentiating the effects of angiotensin II via upregulation of AT1 receptor.229

    In addition to BH4 deficiency, l-arginine deficiency also represents a cause of eNOS uncoupling in hypercholesterolemia. An upregulation of arginase expression and activity has been shown in ApoE-KO mice (Arg2)230,231 and in hyperlipidemic rabbits (Arg1 and Arg2).232 The aortic arginase activity in ApoE-KO mice is significantly reduced after the removal of the endothelium, suggesting important contribution from endothelial cells.230 The functional relevance of arginase upregulation in atherosclerosis has been shown in ApoE-KO mice. Selective endothelial overexpression of Arg2 induces endothelial dysfunction and enhances atherosclerosis in mice.233 Chronic treatment with an arginase inhibitor for 4 or 8 weeks reduces aortic plaque burden in ApoE-KO mice.230

    In addition to eNOS uncoupling, LDL and oxLDL also reduce endothelial NO production by inhibiting eNOS activity.66 Hypercholesterolemia upregulates caveolin abundance, stimulates eNOS translocation to membrane caveolae, and promotes eNOS interaction with caveolin.234 Moreover, LDL also decreases the association of eNOS with Hsp90.228 These effects additively inhibit eNOS activity because caveolin is an inhibiting and Hsp90 a stimulating interaction protein of eNOS. Finally, oxLDL decreases eNOS activity by inhibiting Akt-mediated phosphorylation of eNOS at serine 1177235 or by increased proteasomal degradation of eNOS protein.236

    Cigarette Smoking

    Cigarette smoke–containing compounds have the potential to active endothelial NADPH oxidase237 and to stimulate mitochondrial oxidative stress.238 The enhanced production of superoxide and peroxynitrite leads to vascular inflammation, DNA damage, and vascular aging.238 Moreover, compounds from cigarette smoke also induce oxidative modifications of LDL, which potentiates the pro-oxidative activity of LDL (oxLDL is more potent in activating NADPH oxidase than native LDL).239 BH4 deficiency and eNOS uncoupling have been documented in smokers and supplementation with BH4 can reverse eNOS uncoupling and improve endothelial dysfunction in smokers.75

    Diabetes Mellitus

    Hyperglycemia stimulates ROS production from different cellular sources. Among these, the mitochondrial electron-transport chain is likely to represent the initial superoxide producer.240 Mitochondria-derived superoxide overproduction leads to activation of protein kinase C and formation of advanced glycation end products.240 Protein kinase C and advanced glycation end products can activate NADPH oxidase and, at the same time, inhibit eNOS activity through post-translational modifications.241

    Protein kinase C–stimulated NADPH oxidase activation and eNOS uncoupling have been observed in streptozotocin-induced type 1 diabetes mellitus.242 This is likely to be attributable to NADPH oxidase–mediated BH4 oxidation. Indeed, BH4 oxidation and BH4 deficiency are evident in streptozotocin-treated mice243 and rats.244 In addition, diabetes mellitus also causes BH4 deficiency by reducing BH4 synthesis. Enhanced ROS production in diabetes mellitus accelerates proteasomal degradation of guanosine 5′-triphosphate cyclohydrolase I, a rate-limiting enzyme in the synthesis of BH4.245247 In addition, eNOS S-glutathionylation represents another important mechanism of eNOS uncoupling in the setting of type 1 diabetes mellitus.247

    In mouse models of type 2 diabetes mellitus, a relative BH4 deficiency is evident because of an enhanced BH4 oxidation and a low BH4:BH2 ratio.248250 The increased levels of angiotensin II in diabetic patients may additionally reduce dihydrofolate reductase expression and decrease BH4 recycling from BH2.222

    l-Arginine deficiency represents another mechanism for eNOS uncoupling under conditions of diabetes mellitus. High glucose upregulates Arg1 in (bovine and murine) endothelial cells,251,252 whereas persistent insulin stimulation upregulates the expression and activity of Arg2 in human endothelial cells.253 In addition, l-arginine deficiency and eNOS uncoupling have also been documented in rodent models of type 1251,252,254,255 and type 2 diabetes mellitus.256

    In patients with type 2 diabetes mellitus, plasma arginase activity is elevated.257 An upregulation of Arg1 in coronary arterioles of patients with (type 1 or type 2) diabetes mellitus has been shown to contribute to the reduced NO production and consequently diminished vasodilation.258 Arginase inhibition markedly improves endothelium-dependent vasodilation in the forearm of patients with type 2 diabetes mellitus and CAD, whereas it does not affect endothelial function in healthy controls.259 This observation indicates a functional role of arginase contributing to endothelial dysfunction in patients with diabetes mellitus.

    Biomechanical Factors

    Besides the risk factors at population level (hypertension, hypercholesterolemia, smoking, and diabetes mellitus), disturbed blood flow represents a key risk factor for atherosclerosis within an organism. Atherosclerosis preferentially develops at arches, arterial bifurcations, and side branches, regions that are exposed to nonuniform, disturbed patterns of blood flow.260 Several studies have demonstrated the causal relationship between disturbed flow and atherosclerosis. Disturbed flow induced by applying a constrictive cuff261,262 or by partial ligation or tandem ligations263,264 leads to rapid development of atherosclerosis in the carotid artery, which is otherwise resistant to atherogenesis.

    In each cardiac cycle, arteries are exposed to perpendicular and longitudinal forces generated by intraluminal pressure, and axial stress (shear stress), which acts longitudinally on the surface of the arterial wall.260 Whereas laminar flow (with a high shear stress parallel to the vascular wall and a low circumferential strain) is atheroprotective, disturbed flow (with a change in the diameter and proximity to bifurcations) is proatherogenic.260,265,266 Endothelial cells exposed laminar flow show higher endothelia NO production and are resistant to inflammatory signals and have low intercellular permeability.265 In contrast, endothelial cells exposed to disturbed flow show a proinflammatory phenotype with higher ROS production, elevated cell turnover, increased cell-cell permeability, and upregulated expression of adhesion molecules and chemokines.265

    Laminar flow enhances endothelial NO production267 by stimulating eNOS phosphorylation at serine 1177268,269 and thus increasing the sensitivity of the enzyme to Ca2+ so that the enzyme can be activated at resting Ca2+ levels.270 In addition, shear stress also upregulates eNOS expression.271 In contrast, disturbed flow or oscillatory shear stress decreases eNOS expression and activity.272,273

    On the contrary, oscillatory shear stress markedly increases endothelial superoxide production,274 with NADPH oxidase being the primary superoxide source.9,275 Importantly, NADPH oxidases serve a role as a master oxidase and promote ROS production from other enzymatic sources, eg, XO.9 Xanthine oxidoreductase exists in 2 forms: xanthine dehydrogenase (XDH) and XO. XDH can be converted to XO by reversible sulfhydryl oxidation or by irreversible proteolytic modification.276 In endothelial cells, oscillatory shear stress leads to a NADPH oxidase–dependent degradation of XDH resulting in an increase in XO/XDH ratio. Because both XO and XDH use xanthine as a substrate but only XO generates superoxide, the higher XO/XDH ratio results in enhanced XO-mediated superoxide production.9

    Redox Signaling

    NO exerts its cellular effects through both cGMP-dependent and cGMP-independent mechanisms. In a cGMP-independent manner, NO modifies protein cysteine residues resulting in an S-nitrosothiol.277 A plethora of proteins (including NADPH oxidase278 and eNOS itself279) undergo S-nitrosylation, which may represent part of the mechanistical explanation of the wide range of cellular effects of NO in the cardiovascular system.277

    Protein denitrosylation can be catalyzed by 2 major enzymatic systems. Whereas thioredoxins directly denitrosylate substrate S-nitrosothiol proteins, the S-nitrosoglutathione (GSNO) reductase governs protein S-nitrosylation indirectly by acting on GSNO and thereby modulates the cellular equilibrium between S-nitrosothiol proteins and GSNO.277 Genetic deletion of GSNO reductase increases the levels of S-nitrosothiols in red blood cells and lowers systemic vascular resistance,280 demonstrating the role for GSNO in conveying the systemic activity of NO derived from eNOS.277 Interestingly, the endogenous gaseous signaling molecule hydrogen sulfide also regulates cysteine thiols,281 protects against oxidative stress,282 and has an impact on protein S-nitrosylation,283,284 including S-nitrosylation of eNOS.285

    Cysteine is a unique amino acid because of its thiol side chain, which is redox active. Numerous reactions are known to occur on cysteine thiol side chains that affect protein structure and function, including S-nitrosylation, S-sulfhydration (modification by hydrogen sulfide), S-glutathionylation, and disulfide bond formation. Moreover, stepwise oxidation of cysteine thiol by hydrogen peroxide results in the formation of sulfenic acid, sulfinic acid, and sulfonic acid.286,287 Physiological conditions (NO>ROS) favor S-nitrosylation, whereas NO/ROS disequilibrium under oxidative stress favors oxidation reactions (S-glutathionylation, intramolecular disulfide, and sulfur oxides formation).154 Thiol oxidation adversely impacts S-nitrosylation signaling.154 Indeed, disruption of protein S-nitrosylation has been documented under conditions of heart failure154,277 and high glucose.288

    Effects of ROS and NO in Key Steps of Atherosclerosis: The Molecular Links

    ROS and NO are implicated in the initiation and propagation of atherosclerosis, which provides an explantation how cardiovascular risk factors promote atherogenesis.

    LDL Accumulation in the Vascular Wall

    As mentioned above, atherosclerosis develops in the arterial system initially at predilection sites with geometries, such as arches, branches, and bifurcations.289,290 One mechanism linking the disturbed flow patterns to atherogenesis is LDL accumulation in the vascular wall.

    Theoretically, accumulation of LDL in the artery wall can be caused either by an increase in lipoprotein influx (increase in endothelial permeability and delivery of LDL into the artery wall) or a decrease in lipoprotein efflux caused by an increased binding LDL by the artery wall.

    LDL transport from the circulation to the vessel wall is promoted at sites of disturbed flow because of flow stagnation and the subsequent prolonged contact between blood and vascular endothelial cells.260,266 Moreover, structural examination of macromolecular structures reveals that a nearly confluent elastin surface layer is present throughout mouse aorta. This internal elastin layer, however, is missing at vascular branch points, which are among the sites most prone to atherosclerosis.291 LDL binding is most extensive in the arterial branch points where the elastin layer is absent,291 indicating that the absence of an elastin layer contributes to the initial LDL infiltration at these sites and the subsequent development of atherosclerosis.

    A recent study has provided compelling evidence that LDL retention represents a mechanism that could be even more important than LDL influx for LDL accumulation in flow-disturbed vascular regions.289 Infusion of normocholesterolemic mice with labeled human LDL results in LDL retention in the intimal and medial layers along the inner curvature of the aortic arch and at branch points.289 The straight segment of the common carotid artery is exposed to uniform laminar flow and is resistant to atherosclerosis. Normally, no LDL retention is found in this region.289 Implanting a mildly constrictive perivascular collar evokes disturbed laminar flow in the segment proximal to the collar characterized by low wall shear stress and cyclic circumferential stretching.292 Strikingly, this manipulated blood flow in the straight segment of the common carotid artery is sufficient to transform this otherwise atherosclerosis-resistant site into LDL-retaining regions. Importantly, the accumulation of LDL in the flow-manipulated region is a result of enhanced LDL binding by the vascular wall without any changes in endothelial permeability or LDL influx.289 Gene expression analyses have revealed that flow manipulation leads to increased expression of proteoglycan core proteins associated with LDL binding and retention.289

    The expression of proteoglycans is upregulated after vascular injury293 and in insulin resistance294 associated with increased cholesterol deposition in the vascular wall. Proatherogenic molecules such as platelet-derived growth factor stimulates proteoglycan synthesis in VSMC and enhances LDL retention in blood vessels.295,296 Interestingly, proteoglycans secreted from statin-exposed cells demonstrate a reduction in LDL-binding affinity. Thus, the atheroprotective effects of statins may be partly attributed to changes in vascular proteoglycans and lower LDL retention in the vessel wall.297

    The roles of NO and oxidative stress in LDL accumulation in the vascular wall have not been sufficiently studied to date. Previous studies have shown that endothelial NO prevents the uptake of LDL by arterial walls.298,299 Disturbed flow is known to reduce endothelial NO production300 and enhance ROS production in endothelial cells and in VSMC.9,274,301,302 However, the precise roles of NO and ROS in LDL uptake and LDL binding (eg, regulation of proteoglycan expression) are still unknown and warrants future studies.

    LDL Oxidation

    Experimental studies have demonstrated that oxLDL within the arterial wall promotes atherogenesis,303305 although direct evidence for the role of LDL oxidation in human atherosclerosis is still rare306 and many questions remain to be answered.305

    OxLDL exhibits a wide array of proatherogenic properties.304 Many of these effects are mediated by oxidized phospholipids within the LDL molecules.304 Lipid peroxidation can occur through nonenzymatic mechanisms (eg, by ROS derived from NADPH oxidase or uncoupled eNOS)307 or through enzymatic mechanisms (eg, by myeloperoxidases, lipoxygenases, cyclooxygenases, and cytochrome P450).308 The lipid peroxidation products, such as malondialdehyde, 4-hydroxynonenal, phosphocholine of oxidized phospholipid, and 2-(ω-carboxyethyl) pyrrole, are highly reactive. They modify self-molecules leading to the generation of structural neoepitopes termed oxidation-specific epitopes (OSEs).308 OSEs, including oxidized phospholipids and malondialdehyde-modified amino groups, have been documented on the surface of apoptotic cells and oxLDL molecules.303,308

    Peroxidation of phospholipids promotes a conformational change in the apoB-100 molecule leading to increased nonreceptor-mediated capture of the oxLDL particle by vascular cells.303 Moreover, OSEs are recognized by both cellular (eg, scavenger receptors and toll-like receptors [TLRs]) and soluble pattern recognition receptors (eg, proteins of the complement system, C-reactive protein, and natural IgM antibodies).308

    The recognition of OSEs by cellular and humoral immune responses has important physiological roles in maintaining tissue homeostasis by removing dying cells, cellular debris, and damaged molecules.308 In situations of increased oxidative stress, however, OSEs generation is significantly increased. Furnished with a variety of scavenger receptors and TLRs, endothelial cells, and macrophages are the major cellular sensors of OSEs in atherosclerosis. Consequently, the persistent sensing of OSEs by these cells triggers chronic inflammation through the secretion of chemokines and proinflammatory cytokines (see below).

    The role of LDL oxidation in atherogenesis has been shown in gene targeting studies.304 Genetic deletion of lipoxygenases decreases LDL oxidation and atherosclerosis lesion in mouse models of atherosclerosis.309312 Disruption of paraoxonase 1, an enzyme that indirectly inhibits LDL oxidation, increases LDL oxidation and lesion formation.313 Moreover, OSE-specific natural IgM antibodies block the binding and uptake of oxLDL by macrophages, prevent foam cell formation,314 and decrease atherosclerosis in mice.315,316 Patients with lower levels of IgM antibodies directed against malondialdehyde-LDL and oxLDL show an increased risk of cardiovascular disease.317

    Interestingly, lipid peroxidation also leads to the formation of highly reactive γ-ketoaldehydes (isoketals). Proteins oxidatively modified by isoketals are formed in hypertension and accumulate in dendritic cells. Peptides derived from isoketal adducts of proteins behave as modified self-antigens, activating dendritic cells and T cells, which has been recently identified as a novel mechanism in hypertension.318,319

    As aforementioned, NADPH oxidase–derived ROS are involved in LDL oxidation.275 Interestingly, results from bone marrow transplantation experiments in mice indicate that NADPH oxidase in infiltrating immune cells in the vascular wall may be of more importance in LDL oxidation than those in the resident cells of the vascular wall.49 In contrast, endothelial NO has been shown to inhibit LDL oxidation320,321

    Endothelial Cell Activation and Adhesion Molecule Expression

    Endothelial cells can sense OSE and take up oxLDL via the lectin-like oxidized LDL receptor-1, TLR2, and TLR4.303,308 This may lead to a variety of biological effects, such as (1) reduction of endothelial NO production, (2) upregulation of leukocyte adhesion molecules, (3) promotion of a prothrombotic surface, and (4) synthesis of smooth muscle cell mitogenic factors.303

    OSE sensing by endothelial cells is a key response in the development of atherosclerosis.308 Oxidized phospholipids, for instance, induce the expression of chemoattractants (eg, MCP-1 [monocyte chemoattractant protein-1], chemokine (C-X-C motif) ligand 8 and P selectin) and trigger monocyte binding to endothelial cells via TLR4.307 4-hydroxynonenal induces nuclear factor-κB activation in endothelial cells which is mediated by LOX1.322 2-(ω-carboxyethyl) pyrrole has been shown to activate endothelial cells by stimulating TLR2.323 Deficiency of LOX1324,325 or TLR2326 reduces vascular cell adhesion molecule-1 expression and atherosclerosis in LDL receptor-KO mice.

    The expression of adhesion molecules by endothelial cells is inhibited by laminar shear stress,327 but enhanced by disturbed flow.328,329 In addition, endothelial expression of adhesion molecules is also upregulated by proatherogenic stimuli, such as proinflammatory cytokines,330,331 angiotensin II,332 advanced glycation end products,333 and leptin.334 Interestingly, the anti-inflammatory adipokine, adiponectin, inhibits the expression of adhesion molecules in endothelial cells and prevents leukocyte–endothelium interaction.335

    NO and ROS have opposite roles in the regulation of adhesion molecule expression and endothelial–leukocyte interaction. Endothelial NO inhibits cytokine-induced nuclear factor-κB activation and upregulation of vascular cell adhesion molecule-1, E-selectin, and intercellular adhesion molecule-1.132,331,336338 Inhibition of NO production increases leukocyte adherence, indicating that endothelial NO is an endogenous modulator of leukocyte adhesion to vascular endothelium.339 On the contrary, ROS are implicated in upregulation of adhesion molecules induced by cytokines330,331 or by leptin.334 ROS not only potentiate OSE sensing by upregulation of endothelial TLR2 expression,326 but are also involved in the intercellular signaling cascades leading to adhesion molecules expression induced by stretch or oscillatory shear stress.340,341

    In agreement with this concept, targeted delivery of SOD to endothelial cells in mice inhibits nuclear factor-κB signaling and vascular cell adhesion molecule-1 expression.342 In contrast, genetic disruption of peroxiredoxin1343 or GPx1344 potentiates chemoattractant expression and leukocyte–endothelial cell adhesion.

    Macrophage Activation and Foam Cell Formation

    A hallmark of atherosclerosis is the formation of foam cells due to enhanced uptake of oxLDL by macrophages mediated by OSE binding to scavenger receptors (eg, CD36, TLRs, SRA1 [scavenger receptor A1] and lectin-like oxidized LDL receptor-1).308,345 OxLDL is taken up much more rapidly than native LDL by monocytes/macrophages.304 The scavenger receptors are not downregulated by intracellular LDL, allowing progressive accumulation of cholesterol to the point of foam cell generation.304

    The uptake of oxLDL by macrophages has an important physiological role, as this facilitates oxLDL removal.308 However, under pathological situations, the enhanced uptake of cholesterol-rich LDL and subsequent inefficient removal of intracellular cholesterol can trigger signaling pathways that induce the secretion of inflammatory chemokines and cytokines.308 SRA and CD36 account for ≈90% of macrophage uptake of oxLDL. Genetic disruption of either CD36 or SRA1 reduces atherosclerosis in mice.304,345,346 Supersaturation of cholesterol in macrophages results in the formation of cholesterol crystals leading to the damage of lysosomes, inflammasome activation, and interleukin-1β secretion.347 Excessive foam cell formation induces macrophage apoptosis as observed in advanced atherosclerotic lesions.308,348

    Oxidative stress plays a crucial role in macrophage activation and foam cell formation. ROS derived from NADPH oxidase and uncoupled eNOS are involved in the generation of OSE.307 Moreover, XO also plays an important role in cholesterol crystal-induced ROS formation and inflammatory cytokine release by macrophages. XO inhibition reduces arterial ROS levels, improves endothelial dysfunction, and suppresses plaque formation in ApoE-KO mice.51

    Therapeutic Strategies

    Despite the role of oxidative stress in the pathogenesis of atherosclerosis and other diseases, dietary antioxidant supplements to human subjects do show preventative or therapeutic effect. In a large meta-analysis, antioxidant supplements show no benefit (vitamin C and selenium) or even increase mortality (beta carotene, vitamin A, and vitamin E).349,350

    However, the ineffectiveness of the antioxidant therapy does not disprove the role of oxidative stress in atherogenesis. Rather, the used antioxidant compounds are likely to be unspecific, not correctly dosed, cannot reach the intracellular compartment of ROS generation, and some may even have pro-oxidative properties.4,351,352 Therefore, better antioxidant strategies need to be developed.

    Among the ROS-producing enzymes, NADPH oxidases are proposed to be the master oxidases.9 NADPH oxidase activation promotes ROS production from other sources. Therefore, NADPH oxidases represent attractive therapeutic targets. However, because of the protective role of Nox4 shown in recent studies, a promising Nox inhibitor should be specific for Nox1 or Nox2. Nox inhibitors with improved specificity for NADPH oxidases and moderate Nox isoform selectivity have been developed.353 The first results in mouse atherosclerosis models are encouraging.

    In addition to inhibition of XO activity51 and prevention of mitochondrial oxidative stress,354 pharmacological reversal of eNOS uncoupling represents further fascinating strategies. Among the drugs currently in clinical use, inhibitors of the renin–angiotensin–aldosterone system, statins, nebivolol, and pentaerythritol tetranitrate have been shown to prevent or reverse eNOS uncoupling under experimental conditions. Other compounds, such as resveratrol, sepiapterin, folic acid, and AVE3085, may also recouple eNOS and improve endothelial function although the long-term benefit of these compounds is still unknown (see our recent reviews66,67,355).

    Conclusion

    Endothelial NO protects against, whereas vascular oxidative stress promotes, atherosclerosis. Cardiovascular risk factors decrease endothelial NO production and stimulate ROS production from various ROS sources including NADPH oxidases, XO, mitochondria, and uncoupled eNOS. ROS and NO have opposite roles in the process of atherogenesis, such as LDL oxidation, endothelial cell activation, and macrophage infiltration/activation. Prevention of vascular oxidative stress and improvement of endothelia NO production may represent feasible therapeutic strategies in addition to the treatment of established cardiovascular risk factors.

    Nonstandard Abbreviations and Acronyms

    ApoE-KO

    apolipoprotein E-knockout

    BH4

    tetrahydrobiopterin

    CAD

    coronary artery disease

    CCA

    common carotid artery

    CEP

    2-(ω-carboxyethyl) pyrrole

    COX

    cyclooxygenase

    DKO

    double knockout

    eNOS

    endothelial nitric oxide synthase

    GPx1

    glutathione perioxidase 1

    GSNO

    S-nitrosoglutathione

    HDL

    high-density lipoprotein

    iNOS

    inducible nitric oxide synthase

    LDL

    low-density lipoprotein

    nNOS

    neuronal nitric oxide synthase

    OSE

    oxidation-specific epitopes

    oxLDL

    oxidized low-density lipoprotein

    ROS

    reactive oxygen species

    SOD

    superoxide dismutase

    TLR

    toll-like receptor

    XDH

    xanthine dehydrogenase

    XO

    xanthine oxidase

    Acknowledgments

    Original work from the authors’ laboratory contributing to this review was supported by the German Research Foundation (DFG; LI-1042/1–1 and LI-1042/3-1), the German Federal Ministry of Education and Research (BMBF; 01EO1003), and intramural fund (Stufe I) of the Johannes Gutenberg University Medical Center Mainz.

    Footnotes

    Correspondence to Ulrich Förstermann, MD, PhD, Department of Pharmacology, Johannes Gutenberg University Medical Center, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany. E-mail

    References

    • 1. Stocker R, Keaney JFRole of oxidative modifications in atherosclerosis.Physiol Rev. 2004; 84:1381–1478. doi: 10.1152/physrev.00047.2003.CrossrefMedlineGoogle Scholar
    • 2. Förstermann U.Oxidative stress in vascular disease: causes, defense mechanisms and potential therapies.Nat Clin Pract Cardiovasc Med. 2008; 5:338–349. doi: 10.1038/ncpcardio1211.CrossrefMedlineGoogle Scholar
    • 3. Förstermann U.Nitric oxide and oxidative stress in vascular disease.Pflugers Arch. 2010; 459:923–939. doi: 10.1007/s00424-010-0808-2.CrossrefMedlineGoogle Scholar
    • 4. Li H, Horke S, Förstermann U.Vascular oxidative stress, nitric oxide and atherosclerosis.Atherosclerosis. 2014; 237:208–219. doi: 10.1016/j.atherosclerosis.2014.09.001.CrossrefMedlineGoogle Scholar
    • 5. Daiber A, Di Lisa F, Oelze M, Kroller-Schon S, Steven S, Schulz E, Munzel T.Crosstalk of mitochondria with nadph oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function [published online ahead of print December 9, 2015].Br J Pharmacol. doi: 10.1111/bph.13403. http://onlinelibrary.wiley.com/doi/10.1111/bph.13403/abstract;jsessionid=150E9A30F843D95F53F162452858D588.f04t04.Google Scholar
    • 6. Daiber A.Redox signaling (cross-talk) from and to mitochondria involves mitochondrial pores and reactive oxygen species.Biochim Biophys Acta. 2010; 1797:897–906. doi: 10.1016/j.bbabio.2010.01.032.CrossrefMedlineGoogle Scholar
    • 7. Itani HA, Dikalova AE, McMaster WG, Nazarewicz RR, Bikineyeva AT, Harrison DG, Dikalov SI.Mitochondrial cyclophilin D in vascular oxidative stress and hypertension.Hypertension. 2016; 67:1218–1227. doi: 10.1161/HYPERTENSIONAHA.115.07085.LinkGoogle Scholar
    • 8. Landmesser U, Dikalov S, Price SR, McCann L, Fukai T, Holland SM, Mitch WE, Harrison DG.Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension.J Clin Invest. 2003; 111:1201–1209. doi: 10.1172/JCI14172.CrossrefMedlineGoogle Scholar
    • 9. McNally JS, Davis ME, Giddens DP, Saha A, Hwang J, Dikalov S, Jo H, Harrison DG.Role of xanthine oxidoreductase and NAD(P)H oxidase in endothelial superoxide production in response to oscillatory shear stress.Am J Physiol Heart Circ Physiol. 2003; 285:H2290–H2297. doi: 10.1152/ajpheart.00515.2003.CrossrefMedlineGoogle Scholar
    • 10. Landmesser U, Spiekermann S, Preuss C, Sorrentino S, Fischer D, Manes C, Mueller M, Drexler H.Angiotensin II induces endothelial xanthine oxidase activation: role for endothelial dysfunction in patients with coronary disease.Arterioscler Thromb Vasc Biol. 2007; 27:943–948. doi: 10.1161/01.ATV.0000258415.32883.bf.LinkGoogle Scholar
    • 11. Dikalova AE, Bikineyeva AT, Budzyn K, Nazarewicz RR, McCann L, Lewis W, Harrison DG, Dikalov SI.Therapeutic targeting of mitochondrial superoxide in hypertension.Circ Res. 2010; 107:106–116. doi: 10.1161/CIRCRESAHA.109.214601.LinkGoogle Scholar
    • 12. Kröller-Schön S, Steven S, Kossmann S, et al. Molecular mechanisms of the crosstalk between mitochondria and NADPH oxidase through reactive oxygen species-studies in white blood cells and in animal models.Antioxid Redox Signal. 2014; 20:247–266. doi: 10.1089/ars.2012.4953.CrossrefMedlineGoogle Scholar
    • 13. Wenzel P, Mollnau H, Oelze M, Schulz E, Wickramanayake JM, Müller J, Schuhmacher S, Hortmann M, Baldus S, Gori T, Brandes RP, Münzel T, Daiber A.First evidence for a crosstalk between mitochondrial and NADPH oxidase-derived reactive oxygen species in nitroglycerin-triggered vascular dysfunction.Antioxid Redox Signal. 2008; 10:1435–1447. doi: 10.1089/ars.2007.1969.CrossrefMedlineGoogle Scholar
    • 14. Doughan AK, Harrison DG, Dikalov SI.Molecular mechanisms of angiotensin II-mediated mitochondrial dysfunction: linking mitochondrial oxidative damage and vascular endothelial dysfunction.Circ Res. 2008; 102:488–496. doi: 10.1161/CIRCRESAHA.107.162800.LinkGoogle Scholar
    • 15. Drummond GR, Selemidis S, Griendling KK, Sobey CG.Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets.Nat Rev Drug Discov. 2011; 10:453–471. doi: 10.1038/nrd3403.CrossrefMedlineGoogle Scholar
    • 16. Bedard K, Krause KH.The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology.Physiol Rev. 2007; 87:245–313. doi: 10.1152/physrev.00044.2005.CrossrefMedlineGoogle Scholar
    • 17. Brandes RP, Kreuzer J.Vascular NADPH oxidases: molecular mechanisms of activation.Cardiovasc Res. 2005; 65:16–27. doi: 10.1016/j.cardiores.2004.08.007.CrossrefMedlineGoogle Scholar
    • 18. Schröder K, Zhang M, Benkhoff S, Mieth A, Pliquett R, Kosowski J, Kruse C, Luedike P, Michaelis UR, Weissmann N, Dimmeler S, Shah AM, Brandes RP.Nox4 is a protective reactive oxygen species generating vascular NADPH oxidase.Circ Res. 2012; 110:1217–1225. doi: 10.1161/CIRCRESAHA.112.267054.LinkGoogle Scholar
    • 19. Lassègue B, Sorescu D, Szöcs K, Yin Q, Akers M, Zhang Y, Grant SL, Lambeth JD, Griendling KK.Novel gp91(phox) homologues in vascular smooth muscle cells: nox1 mediates angiotensin II-induced superoxide formation and redox-sensitive signaling pathways.Circ Res. 2001; 88:888–894.LinkGoogle Scholar
    • 20. Ellmark SH, Dusting GJ, Fui MN, Guzzo-Pernell N, Drummond GR.The contribution of Nox4 to NADPH oxidase activity in mouse vascular smooth muscle.Cardiovasc Res. 2005; 65:495–504. doi: 10.1016/j.cardiores.2004.10.026.CrossrefMedlineGoogle Scholar
    • 21. Görlach A, Brandes RP, Nguyen K, Amidi M, Dehghani F, Busse R.A gp91phox containing NADPH oxidase selectively expressed in endothelial cells is a major source of oxygen radical generation in the arterial wall.Circ Res. 2000; 87:26–32.LinkGoogle Scholar
    • 22. Ago T, Kitazono T, Ooboshi H, Iyama T, Han YH, Takada J, Wakisaka M, Ibayashi S, Utsumi H, Iida M.Nox4 as the major catalytic component of an endothelial NAD(P)H oxidase.Circulation. 2004; 109:227–233. doi: 10.1161/01.CIR.0000105680.92873.70.LinkGoogle Scholar
    • 23. Xu H, Goettsch C, Xia N, Horke S, Morawietz H, Förstermann U, Li H.Differential roles of PKCalpha and PKCepsilon in controlling the gene expression of Nox4 in human endothelial cells.Free Radic Biol Med. 2008; 44:1656–1667. doi: 10.1016/j.freeradbiomed.2008.01.023.CrossrefMedlineGoogle Scholar
    • 24. Fulton DJ, Barman SA.Clarity on the isoform-specific roles of NADPH oxidases and NADPH oxidase-4 in atherosclerosis.Arterioscler Thromb Vasc Biol. 2016; 36:579–581. doi: 10.1161/ATVBAHA.116.307096.LinkGoogle Scholar
    • 25. Sheehan AL, Carrell S, Johnson B, Stanic B, Banfi B, Miller FJRole for Nox1 NADPH oxidase in atherosclerosis.Atherosclerosis. 2011; 216:321–326. doi: 10.1016/j.atherosclerosis.2011.02.028.CrossrefMedlineGoogle Scholar
    • 26. Gray SP, Di Marco E, Okabe J, et al. NADPH oxidase 1 plays a key role in diabetes mellitus-accelerated atherosclerosis.Circulation. 2013; 127:1888–1902. doi: 10.1161/CIRCULATIONAHA.112.132159.LinkGoogle Scholar
    • 27. Sobey CG, Judkins CP, Rivera J, Lewis CV, Diep H, Lee HW, Kemp-Harper BK, Broughton BR, Selemidis S, Gaspari TA, Samuel CS, Drummond GR.NOX1 deficiency in apolipoprotein E-knockout mice is associated with elevated plasma lipids and enhanced atherosclerosis.Free Radic Res. 2015; 49:186–198. doi: 10.3109/10715762.2014.992893.CrossrefMedlineGoogle Scholar
    • 28. Gray SP, Di Marco E, Kennedy K, Chew P, Okabe J, El-Osta A, Calkin AC, Biessen EA, Touyz RM, Cooper ME, Schmidt HH, Jandeleit-Dahm KA.Reactive oxygen species can provide atheroprotection via NOX4-dependent inhibition of inflammation and vascular remodeling.Arterioscler Thromb Vasc Biol. 2016; 36:295–307. doi: 10.1161/ATVBAHA.115.307012.LinkGoogle Scholar
    • 29. Quesada IM, Lucero A, Amaya C, Meijles DN, Cifuentes ME, Pagano PJ, Castro C.Selective inactivation of NADPH oxidase 2 causes regression of vascularization and the size and stability of atherosclerotic plaques.Atherosclerosis. 2015; 242:469–475. doi: 10.1016/j.atherosclerosis.2015.08.011.CrossrefMedlineGoogle Scholar
    • 30. Kirk EA, Dinauer MC, Rosen H, Chait A, Heinecke JW, LeBoeuf RC.Impaired superoxide production due to a deficiency in phagocyte NADPH oxidase fails to inhibit atherosclerosis in mice.Arterioscler Thromb Vasc Biol. 2000; 20:1529–1535.LinkGoogle Scholar
    • 31. Judkins CP, Diep H, Broughton BR, Mast AE, Hooker EU, Miller AA, Selemidis S, Dusting GJ, Sobey CG, Drummond GR.Direct evidence of a role for Nox2 in superoxide production, reduced nitric oxide bioavailability, and early atherosclerotic plaque formation in ApoE-/- mice.Am J Physiol Heart Circ Physiol. 2010; 298:H24–H32. doi: 10.1152/ajpheart.00799.2009.CrossrefMedlineGoogle Scholar
    • 32. Hsich E, Segal BH, Pagano PJ, Rey FE, Paigen B, Deleonardis J, Hoyt RF, Holland SM, Finkel T.Vascular effects following homozygous disruption of p47(phox): An essential component of NADPH oxidase.Circulation. 2000; 101:1234–1236.LinkGoogle Scholar
    • 33. Barry-Lane PA, Patterson C, van der Merwe M, Hu Z, Holland SM, Yeh ET, Runge MS.p47phox is required for atherosclerotic lesion progression in ApoE(-/-) mice.J Clin Invest. 2001; 108:1513–1522. doi: 10.1172/JCI11927.CrossrefMedlineGoogle Scholar
    • 34. Douglas G, Bendall JK, Crabtree MJ, Tatham AL, Carter EE, Hale AB, Channon KM.Endothelial-specific Nox2 overexpression increases vascular superoxide and macrophage recruitment in ApoE-/- mice.Cardiovasc Res. 2012; 94:20–29. doi: 10.1093/cvr/cvs026.CrossrefMedlineGoogle Scholar
    • 35. Craige SM, Kant S, Reif M, Chen K, Pei Y, Angoff R, Sugamura K, Fitzgibbons T, Keaney JFEndothelial NADPH oxidase 4 protects ApoE-/- mice from atherosclerotic lesions.Free Radic Biol Med. 2015; 89:1–7. doi: 10.1016/j.freeradbiomed.2015.07.004.CrossrefMedlineGoogle Scholar
    • 36. Schürmann C, Rezende F, Kruse C, Yasar Y, Löwe O, Fork C, van de Sluis B, Bremer R, Weissmann N, Shah AM, Jo H, Brandes RP, Schröder K.The NADPH oxidase Nox4 has anti-atherosclerotic functions.Eur Heart J. 2015; 36:3447–3456. doi: 10.1093/eurheartj/ehv460.CrossrefMedlineGoogle Scholar
    • 37. Langbein H, Brunssen C, Hofmann A, Cimalla P, Brux M, Bornstein SR, Deussen A, Koch E, Morawietz H.NADPH oxidase 4 protects against development of endothelial dysfunction and atherosclerosis in LDL receptor deficient mice.Eur Heart J. 2016; 37:1753–1761. doi: 10.1093/eurheartj/ehv564.CrossrefMedlineGoogle Scholar
    • 38. Takac I, Schröder K, Zhang L, Lardy B, Anilkumar N, Lambeth JD, Shah AM, Morel F, Brandes RP.The E-loop is involved in hydrogen peroxide formation by the NADPH oxidase Nox4.J Biol Chem. 2011; 286:13304–13313. doi: 10.1074/jbc.M110.192138.CrossrefMedlineGoogle Scholar
    • 39. Craige SM, Chen K, Pei Y, Li C, Huang X, Chen C, Shibata R, Sato K, Walsh K, Keaney JFNADPH oxidase 4 promotes endothelial angiogenesis through endothelial nitric oxide synthase activation.Circulation. 2011; 124:731–740. doi: 10.1161/CIRCULATIONAHA.111.030775.LinkGoogle Scholar
    • 40. Di Marco E, Gray SP, Kennedy K, Szyndralewiez C, Lyle AN, Lassègue B, Griendling KK, Cooper ME, Schmidt HH, Jandeleit-Dahm KA.NOX4-derived reactive oxygen species limit fibrosis and inhibit proliferation of vascular smooth muscle cells in diabetic atherosclerosis.Free Radic Biol Med. 2016; 97:556–567. doi: 10.1016/j.freeradbiomed.2016.07.013.CrossrefMedlineGoogle Scholar
    • 41. Kleinschnitz C, Grund H, Wingler K, Armitage ME, Jones E, Mittal M, Barit D, Schwarz T, Geis C, Kraft P, Barthel K, Schuhmann MK, Herrmann AM, Meuth SG, Stoll G, Meurer S, Schrewe A, Becker L, Gailus-Durner V, Fuchs H, Klopstock T, de Angelis MH, Jandeleit-Dahm K, Shah AM, Weissmann N, Schmidt HH.Post-stroke inhibition of induced nadph oxidase type 4 prevents oxidative stress and neurodegeneration.PLoS Biol. 2010; 8CrossrefMedlineGoogle Scholar
    • 42. Nishimura A, Ago T, Kuroda J, Arimura K, Tachibana M, Nakamura K, Wakisaka Y, Sadoshima J, Iihara K, Kitazono T.Detrimental role of pericyte Nox4 in the acute phase of brain ischemia.J Cereb Blood Flow Metab. 2016; 36:1143–1154. doi: 10.1177/0271678X15606456.CrossrefMedlineGoogle Scholar
    • 43. Kuroda J, Ago T, Matsushima S, Zhai P, Schneider MD, Sadoshima J.NADPH oxidase 4 (Nox4) is a major source of oxidative stress in the failing heart.Proc Natl Acad Sci U S A. 2010; 107:15565–15570. doi: 10.1073/pnas.1002178107.CrossrefMedlineGoogle Scholar
    • 44. Maalouf RM, Eid AA, Gorin YC, Block K, Escobar GP, Bailey S, Abboud HE.Nox4-derived reactive oxygen species mediate cardiomyocyte injury in early type 1 diabetes.Am J Physiol Cell Physiol. 2012; 302:C597–C604. doi: 10.1152/ajpcell.00331.2011.CrossrefMedlineGoogle Scholar
    • 45. Guzik TJ, Chen W, Gongora MC, Guzik B, Lob HE, Mangalat D, Hoch N, Dikalov S, Rudzinski P, Kapelak B, Sadowski J, Harrison DG.Calcium-dependent NOX5 nicotinamide adenine dinucleotide phosphate oxidase contributes to vascular oxidative stress in human coronary artery disease.J Am Coll Cardiol. 2008; 52:1803–1809. doi: 10.1016/j.jacc.2008.07.063.CrossrefMedlineGoogle Scholar
    • 46. Yu P, Han W, Villar VA, Yang Y, Lu Q, Lee H, Li F, Quinn MT, Gildea JJ, Felder RA, Jose PA.Unique role of NADPH oxidase 5 in oxidative stress in human renal proximal tubule cells.Redox Biol. 2014; 2:570–579. doi: 10.1016/j.redox.2014.01.020.CrossrefMedlineGoogle Scholar
    • 47. Holterman CE, Thibodeau JF, Towaij C, Gutsol A, Montezano AC, Parks RJ, Cooper ME, Touyz RM, Kennedy CR.Nephropathy and elevated BP in mice with podocyte-specific NADPH oxidase 5 expression.J Am Soc Nephrol. 2014; 25:784–797. doi: 10.1681/ASN.2013040371.CrossrefMedlineGoogle Scholar
    • 48. Montezano AC, Tsiropoulou S, Dulak-Lis M, Harvey A, Camargo Lde L, Touyz RM.Redox signaling, Nox5 and vascular remodeling in hypertension.Curr Opin Nephrol Hypertens. 2015; 24:425–433. doi: 10.1097/MNH.0000000000000153.CrossrefMedlineGoogle Scholar
    • 49. Vendrov AE, Hakim ZS, Madamanchi NR, Rojas M, Madamanchi C, Runge MS.Atherosclerosis is attenuated by limiting superoxide generation in both macrophages and vessel wall cells.Arterioscler Thromb Vasc Biol. 2007; 27:2714–2721. doi: 10.1161/ATVBAHA.107.152629.LinkGoogle Scholar
    • 50. Nishino T, Okamoto K, Eger BT, Pai EF, Nishino T.Mammalian xanthine oxidoreductase - mechanism of transition from xanthine dehydrogenase to xanthine oxidase.FEBS J. 2008; 275:3278–3289. doi: 10.1111/j.1742-4658.2008.06489.x.CrossrefMedlineGoogle Scholar
    • 51. Nomura J, Busso N, Ives A, Matsui C, Tsujimoto S, Shirakura T, Tamura M, Kobayashi T, So A, Yamanaka Y.Xanthine oxidase inhibition by febuxostat attenuates experimental atherosclerosis in mice.Sci Rep. 2014; 4:4554. doi: 10.1038/srep04554.CrossrefMedlineGoogle Scholar
    • 52. White CR, Darley-Usmar V, Berrington WR, McAdams M, Gore JZ, Thompson JA, Parks DA, Tarpey MM, Freeman BA.Circulating plasma xanthine oxidase contributes to vascular dysfunction in hypercholesterolemic rabbits.Proc Natl Acad Sci U S A. 1996; 93:8745–8749.CrossrefMedlineGoogle Scholar
    • 53. Ohara Y, Peterson TE, Harrison DG.Hypercholesterolemia increases endothelial superoxide anion production.J Clin Invest. 1993; 91:2546–2551. doi: 10.1172/JCI116491.CrossrefMedlineGoogle Scholar
    • 54. Patetsios P, Song M, Shutze WP, Pappas C, Rodino W, Ramirez JA, Panetta TF.Identification of uric acid and xanthine oxidase in atherosclerotic plaque.Am J Cardiol. 2001; 88:188–91, A6.CrossrefMedlineGoogle Scholar
    • 55. Guzik TJ, Sadowski J, Guzik B, Jopek A, Kapelak B, Przybylowski P, Wierzbicki K, Korbut R, Harrison DG, Channon KM.Coronary artery superoxide production and nox isoform expression in human coronary artery disease.Arterioscler Thromb Vasc Biol. 2006; 26:333–339. doi: 10.1161/01.ATV.0000196651.64776.51.LinkGoogle Scholar
    • 56. Guthikonda S, Sinkey C, Barenz T, Haynes WG.Xanthine oxidase inhibition reverses endothelial dysfunction in heavy smokers.Circulation. 2003; 107:416–421.LinkGoogle Scholar
    • 57. Schröder K, Vecchione C, Jung O, Schreiber JG, Shiri-Sverdlov R, van Gorp PJ, Busse R, Brandes RP.Xanthine oxidase inhibitor tungsten prevents the development of atherosclerosis in ApoE knockout mice fed a Western-type diet.Free Radic Biol Med. 2006; 41:1353–1360. doi: 10.1016/j.freeradbiomed.2006.03.026.CrossrefMedlineGoogle Scholar
    • 58. Wang Y, Wang GZ, Rabinovitch PS, Tabas I.Macrophage mitochondrial oxidative stress promotes atherosclerosis and nuclear factor-κB-mediated inflammation in macrophages.Circ Res. 2014; 114:421–433. doi: 10.1161/CIRCRESAHA.114.302153.LinkGoogle Scholar
    • 59. Corral-Debrinski M, Shoffner JM, Lott MT, Wallace DC.Association of mitochondrial DNA damage with aging and coronary atherosclerotic heart disease.Mutat Res. 1992; 275:169–180.CrossrefMedlineGoogle Scholar
    • 60. Li Y, Huang TT, Carlson EJ, Melov S, Ursell PC, Olson JL, Noble LJ, Yoshimura MP, Berger C, Chan PH, Wallace DC, Epstein CJ.Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase.Nat Genet. 1995; 11:376–381. doi: 10.1038/ng1295-376.CrossrefMedlineGoogle Scholar
    • 61. Nojiri H, Shimizu T, Funakoshi M, Yamaguchi O, Zhou H, Kawakami S, Ohta Y, Sami M, Tachibana T, Ishikawa H, Kurosawa H, Kahn RC, Otsu K, Shirasawa T.Oxidative stress causes heart failure with impaired mitochondrial respiration.J Biol Chem. 2006; 281:33789–33801. doi: 10.1074/jbc.M602118200.CrossrefMedlineGoogle Scholar
    • 62. Ballinger SW, Patterson C, Knight-Lozano CA, Burow DL, Conklin CA, Hu Z, Reuf J, Horaist C, Lebovitz R, Hunter GC, McIntyre K, Runge MS.Mitochondrial integrity and function in atherogenesis.Circulation. 2002; 106:544–549.LinkGoogle Scholar
    • 63. Li H, Förstermann U.Nitric oxide in the pathogenesis of vascular disease.J Pathol. 2000; 190:244–254. doi: 10.1002/(SICI)1096-9896(200002)190:3<244::AID-PATH575>3.0.CO;2-8.CrossrefMedlineGoogle Scholar
    • 64. Li H, Förstermann U.Prevention of atherosclerosis by interference with the vascular nitric oxide system.Curr Pharm Des. 2009; 15:3133–3145.CrossrefMedlineGoogle Scholar
    • 65. Förstermann U, Sessa WC.Nitric oxide synthases: regulation and function.Eur Heart J. 2012; 33:829–37, 837a. doi: 10.1093/eurheartj/ehr304.CrossrefMedlineGoogle Scholar
    • 66. Li H, Förstermann U.Uncoupling of endothelial NO synthase in atherosclerosis and vascular disease.Curr Opin Pharmacol. 2013; 13:161–167. doi: 10.1016/j.coph.2013.01.006.CrossrefMedlineGoogle Scholar
    • 67. Li H, Horke S, Förstermann U.Oxidative stress in vascular disease and its pharmacological prevention.Trends Pharmacol Sci. 2013; 34:313–319. doi: 10.1016/j.tips.2013.03.007.CrossrefMedlineGoogle Scholar
    • 68. Förstermann U, Münzel T.Endothelial nitric oxide synthase in vascular disease: from marvel to menace.Circulation. 2006; 113:1708–1714. doi: 10.1161/CIRCULATIONAHA.105.602532.LinkGoogle Scholar
    • 69. Alp NJ, McAteer MA, Khoo J, Choudhury RP, Channon KM.Increased endothelial tetrahydrobiopterin synthesis by targeted transgenic GTP-cyclohydrolase I overexpression reduces endothelial dysfunction and atherosclerosis in ApoE-knockout mice.Arterioscler Thromb Vasc Biol. 2004; 24:445–450. doi: 10.1161/01.ATV.0000115637.48689.77.LinkGoogle Scholar
    • 70. Wohlfart P, Xu H, Endlich A, Habermeier A, Closs EI, Hübschle T, Mang C, Strobel H, Suzuki T, Kleinert H, Förstermann U, Ruetten H, Li H.Antiatherosclerotic effects of small-molecular-weight compounds enhancing endothelial nitric-oxide synthase (eNOS) expression and preventing eNOS uncoupling.J Pharmacol Exp Ther. 2008; 325:370–379. doi: 10.1124/jpet.107.128009.CrossrefMedlineGoogle Scholar
    • 71. Xia N, Daiber A, Habermeier A, Closs EI, Thum T, Spanier G, Lu Q, Oelze M, Torzewski M, Lackner KJ, Münzel T, Förstermann U, Li H.Resveratrol reverses endothelial nitric-oxide synthase uncoupling in apolipoprotein E knockout mice.J Pharmacol Exp Ther. 2010; 335:149–154. doi: 10.1124/jpet.110.168724.CrossrefMedlineGoogle Scholar
    • 72. Stroes E, Kastelein J, Cosentino F, Erkelens W, Wever R, Koomans H, Lüscher T, Rabelink T.Tetrahydrobiopterin restores endothelial function in hypercholesterolemia.J Clin Invest. 1997; 99:41–46. doi: 10.1172/JCI119131.CrossrefMedlineGoogle Scholar
    • 73. Antoniades C, Shirodaria C, Crabtree M, Rinze R, Alp N, Cunnington C, Diesch J, Tousoulis D, Stefanadis C, Leeson P, Ratnatunga C, Pillai R, Channon KM.Altered plasma versus vascular biopterins in human atherosclerosis reveal relationships between endothelial nitric oxide synthase coupling, endothelial function, and inflammation.Circulation. 2007; 116:2851–2859. doi: 10.1161/CIRCULATIONAHA.107.704155.LinkGoogle Scholar
    • 74. Porkert M, Sher S, Reddy U, Cheema F, Niessner C, Kolm P, Jones DP, Hooper C, Taylor WR, Harrison D, Quyyumi AA.Tetrahydrobiopterin: a novel antihypertensive therapy.J Hum Hypertens. 2008; 22:401–407. doi: 10.1038/sj.jhh.1002329.CrossrefMedlineGoogle Scholar
    • 75. Heitzer T, Krohn K, Albers S, Meinertz T.Tetrahydrobiopterin improves endothelium-dependent vasodilation by increasing nitric oxide activity in patients with Type II diabetes mellitus.Diabetologia. 2000; 43:1435–1438. doi: 10.1007/s001250051551.CrossrefMedlineGoogle Scholar
    • 76. Ueda S, Matsuoka H, Miyazaki H, Usui M, Okuda S, Imaizumi T.Tetrahydrobiopterin restores endothelial function in long-term smokers.J Am Coll Cardiol. 2000; 35:71–75.CrossrefMedlineGoogle Scholar
    • 77. Li H, Hortmann M, Daiber A, Oelze M, Ostad MA, Schwarz PM, Xu H, Xia N, Kleschyov AL, Mang C, Warnholtz A, Münzel T, Förstermann U.Cyclooxygenase 2-selective and nonselective nonsteroidal anti-inflammatory drugs induce oxidative stress by up-regulating vascular NADPH oxidases.J Pharmacol Exp Ther. 2008; 326:745–753. doi: 10.1124/jpet.108.139030.CrossrefMedlineGoogle Scholar
    • 78. Goncharov NV, Avdonin PV, Nadeev AD, Zharkikh IL, Jenkins RO.Reactive oxygen species in pathogenesis of atherosclerosis.Curr Pharm Des. 2015; 21:1134–1146.CrossrefMedlineGoogle Scholar
    • 79. McClelland S, Gawaz M, Kennerknecht E, Konrad CS, Sauer S, Schuerzinger K, Massberg S, Fitzgerald DJ, Belton O.Contribution of cyclooxygenase-1 to thromboxane formation, platelet-vessel wall interactions and atherosclerosis in the ApoE null mouse.Atherosclerosis. 2009; 202:84–91. doi: 10.1016/j.atherosclerosis.2008.04.016.CrossrefMedlineGoogle Scholar
    • 80. Praticò D, Tillmann C, Zhang ZB, Li H, FitzGerald GA.Acceleration of atherogenesis by COX-1-dependent prostanoid formation in low density lipoprotein receptor knockout mice.Proc Natl Acad Sci U S A. 2001; 98:3358–3363. doi: 10.1073/pnas.061607398.CrossrefMedlineGoogle Scholar
    • 81. Hui Y, Ricciotti E, Crichton I, Yu Z, Wang D, Stubbe J, Wang M, Puré E, FitzGerald GA.Targeted deletions of cyclooxygenase-2 and atherogenesis in mice.Circulation. 2010; 121:2654–2660. doi: 10.1161/CIRCULATIONAHA.109.910687.LinkGoogle Scholar
    • 82. Tang SY, Monslow J, Todd L, Lawson J, Puré E, FitzGerald GA.Cyclooxygenase-2 in endothelial and vascular smooth muscle cells restrains atherogenesis in hyperlipidemic mice.Circulation. 2014; 129:1761–1769. doi: 10.1161/CIRCULATIONAHA.113.007913.LinkGoogle Scholar
    • 83. Kirkby NS, Lundberg MH, Wright WR, Warner TD, Paul-Clark MJ, Mitchell JA.COX-2 protects against atherosclerosis independently of local vascular prostacyclin: identification of COX-2 associated pathways implicate Rgl1 and lymphocyte networks.PLoS One. 2014; 9:e98165. doi: 10.1371/journal.pone.0098165.CrossrefMedlineGoogle Scholar
    • 84. Faraci FM, Didion SP.Vascular protection: superoxide dismutase isoforms in the vessel wall.Arterioscler Thromb Vasc Biol. 2004; 24:1367–1373. doi: 10.1161/01.ATV.0000133604.20182.cf.LinkGoogle Scholar
    • 85. Fukai T, Ushio-Fukai M.Superoxide dismutases: role in redox signaling, vascular function, and diseases.Antioxid Redox Signal. 2011; 15:1583–1606. doi: 10.1089/ars.2011.3999.CrossrefMedlineGoogle Scholar
    • 86. Tribble DL, Gong EL, Leeuwenburgh C, Heinecke JW, Carlson EL, Verstuyft JG, Epstein CJ.Fatty streak formation in fat-fed mice expressing human copper-zinc superoxide dismutase.Arterioscler Thromb Vasc Biol. 1997; 17:1734–1740.LinkGoogle Scholar
    • 87. Nelson SK, Bose SK, McCord JM.The toxicity of high-dose superoxide dismutase suggests that superoxide can both initiate and terminate lipid peroxidation in the reperfused heart.Free Radic Biol Med. 1994; 16:195–200.CrossrefMedlineGoogle Scholar
    • 88. Yang H, Roberts LJ, Shi MJ, Zhou LC, Ballard BR, Richardson A, Guo ZM.Retardation of atherosclerosis by overexpression of catalase or both Cu/Zn-superoxide dismutase and catalase in mice lacking apolipoprotein E.Circ Res. 2004; 95:1075–1081. doi: 10.1161/01.RES.0000149564.49410.0d.LinkGoogle Scholar
    • 89. Strålin P, Karlsson K, Johansson BO, Marklund SL.The interstitium of the human arterial wall contains very large amounts of extracellular superoxide dismutase.Arterioscler Thromb Vasc Biol. 1995; 15:2032–2036.LinkGoogle Scholar
    • 90. Sentman ML, Brännström T, Westerlund S, Laukkanen MO, Ylä-Herttuala S, Basu S, Marklund SL.Extracellular superoxide dismutase deficiency and atherosclerosis in mice.Arterioscler Thromb Vasc Biol. 2001; 21:1477–1482.LinkGoogle Scholar
    • 91. Tribble DL, Barcellos-Hoff MH, Chu BM, Gong EL.Ionizing radiation accelerates aortic lesion formation in fat-fed mice via SOD-inhibitable processes.Arterioscler Thromb Vasc Biol. 1999; 19:1387–1392.LinkGoogle Scholar
    • 92. Yang H, Zhou L, Wang Z, Roberts LJ, Lin X, Zhao Y, Guo Z.Overexpression of antioxidant enzymes in ApoE-deficient mice suppresses benzo(a)pyrene-accelerated atherosclerosis.Atherosclerosis. 2009; 207:51–58. doi: 10.1016/j.atherosclerosis.2009.03.052.CrossrefMedlineGoogle Scholar
    • 93. de Haan JB, Witting PK, Stefanovic N, Pete J, Daskalakis M, Kola I, Stocker R, Smolich JJ.Lack of the antioxidant glutathione peroxidase-1 does not increase atherosclerosis in C57BL/J6 mice fed a high-fat diet.J Lipid Res. 2006; 47:1157–1167. doi: 10.1194/jlr.M500377-JLR200.CrossrefMedlineGoogle Scholar
    • 94. Torzewski M, Ochsenhirt V, Kleschyov AL, Oelze M, Daiber A, Li H, Rossmann H, Tsimikas S, Reifenberg K, Cheng F, Lehr HA, Blankenberg S, Förstermann U, Münzel T, Lackner KJ.Deficiency of glutathione peroxidase-1 accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice.Arterioscler Thromb Vasc Biol. 2007; 27:850–857. doi: 10.1161/01.ATV.0000258809.47285.07.LinkGoogle Scholar
    • 95. Lewis P, Stefanovic N, Pete J, Calkin AC, Giunti S, Thallas-Bonke V, Jandeleit-Dahm KA, Allen TJ, Kola I, Cooper ME, de Haan JB.Lack of the antioxidant enzyme glutathione peroxidase-1 accelerates atherosclerosis in diabetic apolipoprotein E-deficient mice.Circulation. 2007; 115:2178–2187. doi: 10.1161/CIRCULATIONAHA.106.664250.LinkGoogle Scholar
    • 96. Guo Z, Ran Q, Roberts LJ, Zhou L, Richardson A, Sharan C, Wu D, Yang H.Suppression of atherogenesis by overexpression of glutathione peroxidase-4 in apolipoprotein E-deficient mice.Free Radic Biol Med. 2008; 44:343–352. doi: 10.1016/j.freeradbiomed.2007.09.009.CrossrefMedlineGoogle Scholar
    • 97. Tward A, Xia YR, Wang XP, Shi YS, Park C, Castellani LW, Lusis AJ, Shih DM.Decreased atherosclerotic lesion formation in human serum paraoxonase transgenic mice.Circulation. 2002; 106:484–490.LinkGoogle Scholar
    • 98. Shih DM, Gu L, Xia YR, Navab M, Li WF, Hama S, Castellani LW, Furlong CE, Costa LG, Fogelman AM, Lusis AJ.Mice lacking serum paraoxonase are susceptible to organophosphate toxicity and atherosclerosis.Nature. 1998; 394:284–287. doi: 10.1038/28406.CrossrefMedlineGoogle Scholar
    • 99. 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.Mol Genet Metab. 2006; 89:368–373. doi: 10.1016/j.ymgme.2006.07.004.CrossrefMedlineGoogle Scholar
    • 100. Ng CJ, Bourquard N, Grijalva V, Hama S, Shih DM, Navab M, Fogelman AM, Lusis AJ, Young S, Reddy ST.Paraoxonase-2 deficiency aggravates atherosclerosis in mice despite lower apolipoprotein-B-containing lipoproteins: anti-atherogenic role for paraoxonase-2.J Biol Chem. 2006; 281:29491–29500. doi: 10.1074/jbc.M605379200.CrossrefMedlineGoogle Scholar
    • 101. Shih DM, Xia YR, Wang XP, Wang SS, Bourquard N, Fogelman AM, Lusis AJ, Reddy ST.Decreased obesity and atherosclerosis in human paraoxonase 3 transgenic mice.Circ Res. 2007; 100:1200–1207. doi: 10.1161/01.RES.0000264499.48737.69.LinkGoogle Scholar
    • 102. Zhang H, Luo Y, Zhang W, He Y, Dai S, Zhang R, Huang Y, Bernatchez P, Giordano FJ, Shadel G, Sessa WC, Min W.Endothelial-specific expression of mitochondrial thioredoxin improves endothelial cell function and reduces atherosclerotic lesions.Am J Pathol. 2007; 170:1108–1120. doi: 10.2353/ajpath.2007.060960.CrossrefMedlineGoogle Scholar
    • 103. Asmis R, Begley JG.Oxidized LDL promotes peroxide-mediated mitochondrial dysfunction and cell death in human macrophages: a caspase-3-independent pathway.Circ Res. 2003; 92:e20–e29.LinkGoogle Scholar
    • 104. Lubos E, Loscalzo J, Handy DE.Glutathione peroxidase-1 in health and disease: from molecular mechanisms to therapeutic opportunities.Antioxid Redox Signal. 2011; 15:1957–1997. doi: 10.1089/ars.2010.3586.CrossrefMedlineGoogle Scholar
    • 105. Blankenberg S, Rupprecht HJ, Bickel C, Torzewski M, Hafner G, Tiret L, Smieja M, Cambien F, Meyer J, Lackner KJ; AtheroGene Investigators. Glutathione peroxidase 1 activity and cardiovascular events in patients with coronary artery disease.N Engl J Med. 2003; 349:1605–1613. doi: 10.1056/NEJMoa030535.CrossrefMedlineGoogle Scholar
    • 106. Cheng F, Torzewski M, Degreif A, Rossmann H, Canisius A, Lackner KJ.Impact of glutathione peroxidase-1 deficiency on macrophage foam cell formation and proliferation: implications for atherogenesis.PLoS One. 2013; 8:e72063. doi: 10.1371/journal.pone.0072063.CrossrefMedlineGoogle Scholar
    • 107. Chew P, Yuen DY, Koh P, Stefanovic N, Febbraio MA, Kola I, Cooper ME, de Haan JB.Site-specific antiatherogenic effect of the antioxidant ebselen in the diabetic apolipoprotein E-deficient mouse.Arterioscler Thromb Vasc Biol. 2009; 29:823–830. doi: 10.1161/ATVBAHA.109.186619.LinkGoogle Scholar
    • 108. Imai H, Nakagawa Y.Biological significance of phospholipid hydroperoxide glutathione peroxidase (PHGPx, GPx4) in mammalian cells.Free Radic Biol Med. 2003; 34:145–169.CrossrefMedlineGoogle Scholar
    • 109. Draganov DI, Teiber JF, Speelman A, Osawa Y, Sunahara R, La Du BN.Human paraoxonases (PON1, PON2, and PON3) are lactonases with overlapping and distinct substrate specificities.J Lipid Res. 2005; 46:1239–1247. doi: 10.1194/jlr.M400511-JLR200.CrossrefMedlineGoogle Scholar
    • 110. Mackness M, Mackness B.Targeting paraoxonase-1 in atherosclerosis.Expert Opin Ther Targets. 2013; 17:829–837. doi: 10.1517/14728222.2013.790367.CrossrefMedlineGoogle Scholar
    • 111. Aviram M, Vaya J.Paraoxonase 1 activities, regulation, and interactions with atherosclerotic lesion.Curr Opin Lipidol. 2013; 24:339–344. doi: 10.1097/MOL.0b013e32835ffcfd.CrossrefMedlineGoogle Scholar
    • 112. Costa LG, Cole TB, Furlong CE.Paraoxonase (PON1): from toxicology to cardiovascular medicine.Acta Biomed. 2005; 76 Suppl 2:50–57.MedlineGoogle Scholar
    • 113. Mackness MI, Arrol S, Abbott C, Durrington PN.Protection of low-density lipoprotein against oxidative modification by high-density lipoprotein associated paraoxonase.Atherosclerosis. 1993; 104:129–135.CrossrefMedlineGoogle Scholar
    • 114. Ng CJ, Wadleigh DJ, Gangopadhyay A, Hama S, Grijalva VR, Navab M, Fogelman AM, Reddy ST.Paraoxonase-2 is a ubiquitously expressed protein with antioxidant properties and is capable of preventing cell-mediated oxidative modification of low density lipoprotein.J Biol Chem. 2001; 276:44444–44449. doi: 10.1074/jbc.M105660200.CrossrefMedlineGoogle Scholar
    • 115. Witte I, Foerstermann U, Devarajan A, Reddy ST, Horke S.Protectors or traitors: the roles of PON2 and PON3 in atherosclerosis and cancer.J Lipids. 2012; 2012:342806. doi: 10.1155/2012/342806.CrossrefMedlineGoogle Scholar
    • 116. Horke S, Witte I, Wilgenbus P, Krüger M, Strand D, Förstermann U.Paraoxonase-2 reduces oxidative stress in vascular cells and decreases endoplasmic reticulum stress-induced caspase activation.Circulation. 2007; 115:2055–2064. doi: 10.1161/CIRCULATIONAHA.106.681700.LinkGoogle Scholar
    • 117. Hagmann H, Kuczkowski A, Ruehl M, Lamkemeyer T, Brodesser S, Horke S, Dryer S, Schermer B, Benzing T, Brinkkoetter PT.Breaking the chain at the membrane: paraoxonase 2 counteracts lipid peroxidation at the plasma membrane.FASEB J. 2014; 28:1769–1779. doi: 10.1096/fj.13-240309.CrossrefMedlineGoogle Scholar
    • 118. Fortunato G, Di Taranto MD, Bracale UM, Del Guercio L, Carbone F, Mazzaccara C, Morgante A, D’Armiento FP, D’Armiento M, Porcellini M, Sacchetti L, Bracale G, Salvatore F.Decreased paraoxonase-2 expression in human carotids during the progression of atherosclerosis.Arterioscler Thromb Vasc Biol. 2008; 28:594–600. doi: 10.1161/ATVBAHA.107.154658.LinkGoogle Scholar
    • 119. Draganov DI, Stetson PL, Watson CE, Billecke SS, La Du BN.Rabbit serum paraoxonase 3 (PON3) is a high density lipoprotein-associated lactonase and protects low density lipoprotein against oxidation.J Biol Chem. 2000; 275:33435–33442. doi: 10.1074/jbc.M004543200.CrossrefMedlineGoogle Scholar
    • 120. Schweikert EM, Devarajan A, Witte I, Wilgenbus P, Amort J, Förstermann U, Shabazian A, Grijalva V, Shih DM, Farias-Eisner R, Teiber JF, Reddy ST, Horke S.PON3 is upregulated in cancer tissues and protects against mitochondrial superoxide-mediated cell death.Cell Death Differ. 2012; 19:1549–1560. doi: 10.1038/cdd.2012.35.CrossrefMedlineGoogle Scholar
    • 121. Altenhöfer S, Witte I, Teiber JF, Wilgenbus P, Pautz A, Li H, Daiber A, Witan H, Clement AM, Förstermann U, Horke S.One enzyme, two functions: PON2 prevents mitochondrial superoxide formation and apoptosis independent from its lactonase activity.J Biol Chem. 2010; 285:24398–24403. doi: 10.1074/jbc.M110.118604.CrossrefMedlineGoogle Scholar
    • 122. Marsillach J, Camps J, Beltran-Debón R, Rull A, Aragones G, Maestre-Martínez C, Sabench F, Hernández M, Castillo DD, Joven J, Mackness M, Mackness B.Immunohistochemical analysis of paraoxonases-1 and 3 in human atheromatous plaques.Eur J Clin Invest. 2011; 41:308–314. doi: 10.1111/j.1365-2362.2010.02411.x.CrossrefMedlineGoogle Scholar
    • 123. Hanschmann EM, Godoy JR, Berndt C, Hudemann C, Lillig CH.Thioredoxins, glutaredoxins, and peroxiredoxins–molecular mechanisms and health significance: from cofactors to antioxidants to redox signaling.Antioxid Redox Signal. 2013; 19:1539–1605. doi: 10.1089/ars.2012.4599.CrossrefMedlineGoogle Scholar
    • 124. Huang Q, Zhou HJ, Zhang H, Huang Y, Hinojosa-Kirschenbaum F, Fan P, Yao L, Belardinelli L, Tellides G, Giordano FJ, Budas GR, Min W.Thioredoxin-2 inhibits mitochondrial reactive oxygen species generation and apoptosis stress kinase-1 activity to maintain cardiac function.Circulation. 2015; 131:1082–1097. doi: 10.1161/CIRCULATIONAHA.114.012725.LinkGoogle Scholar
    • 125. Kirsch J, Schneider H, Pagel JI, et al. Endothelial Dysfunction, and A Prothrombotic, Proinflammatory Phenotype Is Caused by Loss of Mitochondrial Thioredoxin Reductase in Endothelium.Arterioscler Thromb Vasc Biol. 2016; 36:1891–1899. doi: 10.1161/ATVBAHA.116.307843.LinkGoogle Scholar
    • 126. Liu VW, Huang PL.Cardiovascular roles of nitric oxide: a review of insights from nitric oxide synthase gene disrupted mice.Cardiovasc Res. 2008; 77:19–29. doi: 10.1016/j.cardiores.2007.06.024.MedlineGoogle Scholar
    • 127. Schwarz PM, Kleinert H, Förstermann U.Potential functional significance of brain-type and muscle-type nitric oxide synthase I expressed in adventitia and media of rat aorta.Arterioscler Thromb Vasc Biol. 1999; 19:2584–2590.LinkGoogle Scholar
    • 128. Tsutsui M.Neuronal nitric oxide synthase as a novel anti-atherogenic factor.J Atheroscler Thromb. 2004; 11:41–48.CrossrefMedlineGoogle Scholar
    • 129. Schödel J, Padmapriya P, Marx A, Huang PL, Ertl G, Kuhlencordt PJ.Expression of neuronal nitric oxide synthase splice variants in atherosclerotic plaques of apoE knockout mice.Atherosclerosis. 2009; 206:383–389. doi: 10.1016/j.atherosclerosis.2009.02.033.CrossrefMedlineGoogle Scholar
    • 130. Capettini LS, Cortes SF, Silva JF, Alvarez-Leite JI, Lemos VS.Decreased production of neuronal NOS-derived hydrogen peroxide contributes to endothelial dysfunction in atherosclerosis.Br J Pharmacol. 2011; 164:1738–1748. doi: 10.1111/j.1476-5381.2011.01500.x.CrossrefMedlineGoogle Scholar
    • 131. Benkhoff S, Loot AE, Pierson I, Sturza A, Kohlstedt K, Fleming I, Shimokawa H, Grisk O, Brandes RP, Schröder K.Leptin potentiates endothelium-dependent relaxation by inducing endothelial expression of neuronal NO synthase.Arterioscler Thromb Vasc Biol. 2012; 32:1605–1612. doi: 10.1161/ATVBAHA.112.251140.LinkGoogle Scholar
    • 132. Sanz MJ, Hickey MJ, Johnston B, McCafferty DM, Raharjo E, Huang PL, Kubes P.Neuronal nitric oxide synthase (NOS) regulates leukocyte-endothelial cell interactions in endothelial NOS deficient mice.Br J Pharmacol. 2001; 134:305–312. doi: 10.1038/sj.bjp.0704234.CrossrefMedlineGoogle Scholar
    • 133. Talukder MA, Fujiki T, Morikawa K, Motoishi M, Kubota H, Morishita T, Tsutsui M, Takeshita A, Shimokawa H.Up-regulated neuronal nitric oxide synthase compensates coronary flow response to bradykinin in endothelial nitric oxide synthase-deficient mice.J Cardiovasc Pharmacol. 2004; 44:437–445.CrossrefMedlineGoogle Scholar
    • 134. Seddon MD, Chowienczyk PJ, Brett SE, Casadei B, Shah AM.Neuronal nitric oxide synthase regulates basal microvascular tone in humans in vivo.Circulation. 2008; 117:1991–1996. doi: 10.1161/CIRCULATIONAHA.107.744540.LinkGoogle Scholar
    • 135. Seddon M, Melikian N, Dworakowski R, Shabeeh H, Jiang B, Byrne J, Casadei B, Chowienczyk P, Shah AM.Effects of neuronal nitric oxide synthase on human coronary artery diameter and blood flow in vivo.Circulation. 2009; 119:2656–2662. doi: 10.1161/CIRCULATIONAHA.108.822205.LinkGoogle Scholar
    • 136. Morishita T, Tsutsui M, Shimokawa H, Horiuchi M, Tanimoto A, Suda O, Tasaki H, Huang PL, Sasaguri Y, Yanagihara N, Nakashima Y.Vasculoprotective roles of neuronal nitric oxide synthase.FASEB J. 2002; 16:1994–1996. doi: 10.1096/fj.02-0155fje.CrossrefMedlineGoogle Scholar
    • 137. Kuhlencordt PJ, Hötten S, Schödel J, Rützel S, Hu K, Widder J, Marx A, Huang PL, Ertl G.Atheroprotective effects of neuronal nitric oxide synthase in apolipoprotein e knockout mice.Arterioscler Thromb Vasc Biol. 2006; 26:1539–1544. doi: 10.1161/01.ATV.0000223143.88128.19.LinkGoogle Scholar
    • 138. 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; 103:3099–3104.LinkGoogle Scholar
    • 139. Kuhlencordt PJ, Gyurko R, Han F, Scherrer-Crosbie M, Aretz TH, Hajjar R, Picard MH, Huang PL.Accelerated atherosclerosis, aortic aneurysm formation, and ischemic heart disease in apolipoprotein E/endothelial nitric oxide synthase double-knockout mice.Circulation. 2001; 104:448–454.LinkGoogle Scholar
    • 140. Chen J, Kuhlencordt PJ, Astern J, Gyurko R, Huang PL.Hypertension does not account for the accelerated atherosclerosis and development of aneurysms in male apolipoprotein e/endothelial nitric oxide synthase double knockout mice.Circulation. 2001; 104:2391–2394.LinkGoogle Scholar
    • 141. Ponnuswamy P, Schröttle A, Ostermeier E, Grüner S, Huang PL, Ertl G, Hoffmann U, Nieswandt B, Kuhlencordt PJ.eNOS protects from atherosclerosis despite relevant superoxide production by the enzyme in apoE mice.PLoS One. 2012; 7:e30193. doi: 10.1371/journal.pone.0030193.CrossrefMedlineGoogle Scholar
    • 142. Ozaki M, Kawashima S, Yamashita T, Hirase T, Namiki M, Inoue N, Hirata K, Yasui H, Sakurai H, Yoshida Y, Masada M, Yokoyama M.Overexpression of endothelial nitric oxide synthase accelerates atherosclerotic lesion formation in apoE-deficient mice.J Clin Invest. 2002; 110:331–340. doi: 10.1172/JCI15215.CrossrefMedlineGoogle Scholar
    • 143. Takaya T, Hirata K, Yamashita T, Shinohara M, Sasaki N, Inoue N, Yada T, Goto M, Fukatsu A, Hayashi T, Alp NJ, Channon KM, Yokoyama M, Kawashima S.A specific role for eNOS-derived reactive oxygen species in atherosclerosis progression.Arterioscler Thromb Vasc Biol. 2007; 27:1632–1637. doi: 10.1161/ATVBAHA.107.142182.LinkGoogle Scholar
    • 144. Pautz A, Art J, Hahn S, Nowag S, Voss C, Kleinert H.Regulation of the expression of inducible nitric oxide synthase.Nitric Oxide. 2010; 23:75–93. doi: 10.1016/j.niox.2010.04.007.CrossrefMedlineGoogle Scholar
    • 145. Gunnett CA, Lund DD, McDowell AK, Faraci FM, Heistad DD.Mechanisms of inducible nitric oxide synthase-mediated vascular dysfunction.Arterioscler Thromb Vasc Biol. 2005; 25:1617–1622. doi: 10.1161/01.ATV.0000172626.00296.ba.LinkGoogle Scholar
    • 146. Xia Y, Zweier JL.Superoxide and peroxynitrite generation from inducible nitric oxide synthase in macrophages.Proc Natl Acad Sci U S A. 1997; 94:6954–6958.CrossrefMedlineGoogle Scholar
    • 147. Marfella R, Di Filippo C, Esposito K, Nappo F, Piegari E, Cuzzocrea S, Berrino L, Rossi F, Giugliano D, D’Amico M.Absence of inducible nitric oxide synthase reduces myocardial damage during ischemia reperfusion in streptozotocin-induced hyperglycemic mice.Diabetes. 2004; 53:454–462.CrossrefMedlineGoogle Scholar
    • 148. Virdis A, Colucci R, Fornai M, Blandizzi C, Duranti E, Pinto S, Bernardini N, Segnani C, Antonioli L, Taddei S, Salvetti A, Del Tacca M.Cyclooxygenase-2 inhibition improves vascular endothelial dysfunction in a rat model of endotoxic shock: role of inducible nitric-oxide synthase and oxidative stress.J Pharmacol Exp Ther. 2005; 312:945–953. doi: 10.1124/jpet.104.077644.CrossrefMedlineGoogle Scholar
    • 149. Depre C, Havaux X, Renkin J, Vanoverschelde JL, Wijns W.Expression of inducible nitric oxide synthase in human coronary atherosclerotic plaque.Cardiovasc Res. 1999; 41:465–472.CrossrefMedlineGoogle Scholar
    • 150. Pacher P, Beckman JS, Liaudet L.Nitric oxide and peroxynitrite in health and disease.Physiol Rev. 2007; 87:315–424. doi: 10.1152/physrev.00029.2006.CrossrefMedlineGoogle Scholar
    • 151. Liaudet L, Rosenblatt-Velin N, Pacher P.Role of peroxynitrite in the cardiovascular dysfunction of septic shock.Curr Vasc Pharmacol. 2013; 11:196–207.MedlineGoogle Scholar
    • 152. Wilcox JN, Subramanian RR, Sundell CL, Tracey WR, Pollock JS, Harrison DG, Marsden PA.Expression of multiple isoforms of nitric oxide synthase in normal and atherosclerotic vessels.Arterioscler Thromb Vasc Biol. 1997; 17:2479–2488.LinkGoogle Scholar
    • 153. Baker CS, Hall RJ, Evans TJ, Pomerance A, Maclouf J, Creminon C, Yacoub MH, Polak JM.Cyclooxygenase-2 is widely expressed in atherosclerotic lesions affecting native and transplanted human coronary arteries and colocalizes with inducible nitric oxide synthase and nitrotyrosine particularly in macrophages.Arterioscler Thromb Vasc Biol. 1999; 19:646–655.LinkGoogle Scholar
    • 154. Hare JM, Stamler JS.NO/redox disequilibrium in the failing heart and cardiovascular system.J Clin Invest. 2005; 115:509–517. doi: 10.1172/JCI24459.CrossrefMedlineGoogle Scholar
    • 155. Xia N, Li H.The role of perivascular adipose tissue in obesity-induced vascular dysfunction [published online ahead of print October 20, 2016].Br J Pharmacol. doi: 10.1111/bph.13650. http://onlinelibrary.wiley.com/doi/10.1111/bph.13650/abstract.Google Scholar
    • 156. Xia N, Horke S, Habermeier A, Closs EI, Reifenberg G, Gericke A, Mikhed Y, Münzel T, Daiber A, Förstermann U, Li H.Uncoupling of endothelial nitric oxide synthase in perivascular adipose tissue of diet-induced obese mice.Arterioscler Thromb Vasc Biol. 2016; 36:78–85. doi: 10.1161/ATVBAHA.115.306263.LinkGoogle Scholar
    • 157. Bentzon JF, Falk E.Atherosclerotic lesions in mouse and man: is it the same disease?Curr Opin Lipidol. 2010; 21:434–440. doi: 10.1097/MOL.0b013e32833ded6a.CrossrefMedlineGoogle Scholar
    • 158. Gleissner CA.Translational atherosclerosis research: From experimental models to coronary artery disease in humans.Atherosclerosis. 2016; 248:110–116. doi: 10.1016/j.atherosclerosis.2016.03.013.CrossrefMedlineGoogle Scholar
    • 159. Matoba T, Sato K, Egashira K.Mouse models of plaque rupture.Curr Opin Lipidol. 2013; 24:419–425. doi: 10.1097/MOL.0b013e3283646e4d.CrossrefMedlineGoogle Scholar
    • 160. Sato K, Nakano K, Katsuki S, Matoba T, Osada K, Sawamura T, Sunagawa K, Egashira K.Dietary cholesterol oxidation products accelerate plaque destabilization and rupture associated with monocyte infiltration/activation via the MCP-1-CCR2 pathway in mouse brachiocephalic arteries: therapeutic effects of ezetimibe.J Atheroscler Thromb. 2012; 19:986–998.CrossrefMedlineGoogle Scholar
    • 161. Van der Donckt C, Van Herck JL, Schrijvers DM, et al. Elastin fragmentation in atherosclerotic mice leads to intraplaque neovascularization, plaque rupture, myocardial infarction, stroke, and sudden death.Eur Heart J. 2015; 36:1049–1058. doi: 10.1093/eurheartj/ehu041.CrossrefMedlineGoogle Scholar
    • 162. Getz GS, Reardon CA.Do the Apoe-/- and Ldlr-/- Mice Yield the Same Insight on Atherogenesis?Arterioscler Thromb Vasc Biol. 2016; 36:1734–1741. doi: 10.1161/ATVBAHA.116.306874.LinkGoogle Scholar
    • 163. Miller AA, De Silva TM, Jackman KA, Sobey CG.Effect of gender and sex hormones on vascular oxidative stress.Clin Exp Pharmacol Physiol. 2007; 34:1037–1043. doi: 10.1111/j.1440-1681.2007.04732.x.CrossrefMedlineGoogle Scholar
    • 164. Joakimsen O, Bonaa KH, Stensland-Bugge E, Jacobsen BK.Age and sex differences in the distribution and ultrasound morphology of carotid atherosclerosis: the Tromsø Study.Arterioscler Thromb Vasc Biol. 1999; 19:3007–3013.LinkGoogle Scholar
    • 165. Tsuda M, Iwai M, Li JM, Li HS, Min LJ, Ide A, Okumura M, Suzuki J, Mogi M, Suzuki H, Horiuchi M.Inhibitory effects of AT1 receptor blocker, olmesartan, and estrogen on atherosclerosis via anti-oxidative stress.Hypertension. 2005; 45:545–551. doi: 10.1161/01.HYP.0000157409.88971.fc.LinkGoogle Scholar
    • 166. Miller AA, Drummond GR, Mast AE, Schmidt HH, Sobey CG.Effect of gender on NADPH-oxidase activity, expression, and function in the cerebral circulation: role of estrogen.Stroke. 2007; 38:2142–2149. doi: 10.1161/STROKEAHA.106.477406.LinkGoogle Scholar
    • 167. Laughlin MH, Welshons WV, Sturek M, Rush JW, Turk JR, Taylor JA, Judy BM, Henderson KK, Ganjam VK.Gender, exercise training, and eNOS expression in porcine skeletal muscle arteries.J Appl Physiol (1985). 2003; 95:250–264. doi: 10.1152/japplphysiol.00061.2003.CrossrefMedlineGoogle Scholar
    • 168. Kleinert H, Wallerath T, Euchenhofer C, Ihrig-Biedert I, Li H, Förstermann U.Estrogens increase transcription of the human endothelial NO synthase gene: analysis of the transcription factors involved.Hypertension. 1998; 31:582–588.LinkGoogle Scholar
    • 169. Nikpay M, Goel A, Won HH, et al; CARDIoGRAMplusC4D Consortium. A comprehensive 1,000 genomes-based genome-wide association meta-analysis of coronary artery disease.Nat Genet. 2015; 47:1121–1130. doi: 10.1038/ng.3396.CrossrefMedlineGoogle Scholar
    • 170. Nurnberg ST, Zhang H, Hand NJ, Bauer RC, Saleheen D, Reilly MP, Rader DJ.From loci to biology: functional genomics of genome-wide association for coronary disease.Circ Res. 2016; 118:586–606. doi: 10.1161/CIRCRESAHA.115.306464.LinkGoogle Scholar
    • 171. Inoue N, Kawashima S, Kanazawa K, Yamada S, Akita H, Yokoyama M.Polymorphism of the NADH/NADPH oxidase p22 phox gene in patients with coronary artery disease.Circulation. 1998; 97:135–137.LinkGoogle Scholar
    • 172. Shimokata K, Yamada Y, Kondo T, Ichihara S, Izawa H, Nagata K, Murohara T, Ohno M, Yokota M.Association of gene polymorphisms with coronary artery disease in individuals with or without nonfamilial hypercholesterolemia.Atherosclerosis. 2004; 172:167–173.CrossrefMedlineGoogle Scholar
    • 173. Arca M, Conti B, Montali A, Pignatelli P, Campagna F, Barillà F, Tanzilli G, Verna R, Vestri A, Gaudio C, Violi F.C242T polymorphism of NADPH oxidase p22phox and recurrence of cardiovascular events in coronary artery disease.Arterioscler Thromb Vasc Biol. 2008; 28:752–757. doi: 10.1161/ATVBAHA.107.154823.LinkGoogle Scholar
    • 174. Cahilly C, Ballantyne CM, Lim DS, Gotto A, Marian AJ.A variant of p22(phox), involved in generation of reactive oxygen species in the vessel wall, is associated with progression of coronary atherosclerosis.Circ Res. 2000; 86:391–395.LinkGoogle Scholar
    • 175. Andreassi MG, Adlerstein D, Carpeggiani C, Shehi E, Fantinato S, Ghezzi E, Botto N, Coceani M, L’abbate A.Individual and summed effects of high-risk genetic polymorphisms on recurrent cardiovascular events following ischemic heart disease.Atherosclerosis. 2012; 223:409–415. doi: 10.1016/j.atherosclerosis.2012.05.029.CrossrefMedlineGoogle Scholar
    • 176. Neves AL, Mohammedi K, Emery N, Roussel R, Fumeron F, Marre M, Velho G.Allelic variations in superoxide dismutase-1 (SOD1) gene and renal and cardiovascular morbidity and mortality in type 2 diabetic subjects.Mol Genet Metab. 2012; 106:359–365. doi: 10.1016/j.ymgme.2012.04.023.CrossrefMedlineGoogle Scholar
    • 177. Kakko S, Päivänsalo M, Koistinen P, Kesäniemi YA, Kinnula VL, Savolainen MJ.The signal sequence polymorphism of the MnSOD gene is associated with the degree of carotid atherosclerosis.Atherosclerosis. 2003; 168:147–152.CrossrefMedlineGoogle Scholar
    • 178. Fujimoto H, Taguchi J, Imai Y, Ayabe S, Hashimoto H, Kobayashi H, Ogasawara K, Aizawa T, Yamakado M, Nagai R, Ohno M.Manganese superoxide dismutase polymorphism affects the oxidized low-density lipoprotein-induced apoptosis of macrophages and coronary artery disease.Eur Heart J. 2008; 29:1267–1274. doi: 10.1093/eurheartj/ehm500.CrossrefMedlineGoogle Scholar
    • 179. Möllsten A, Jorsal A, Lajer M, Vionnet N, Tarnow L.The V16A polymorphism in SOD2 is associated with increased risk of diabetic nephropathy and cardiovascular disease in type 1 diabetes.Diabetologia. 2009; 52:2590–2593. doi: 10.1007/s00125-009-1550-1.CrossrefMedlineGoogle Scholar
    • 180. Marklund SL, Nilsson P, Israelsson K, Schampi I, Peltonen M, Asplund K.Two variants of extracellular-superoxide dismutase: relationship to cardiovascular risk factors in an unselected middle-aged population.J Intern Med. 1997; 242:5–14.CrossrefMedlineGoogle Scholar
    • 181. Juul K, Tybjaerg-Hansen A, Marklund S, Heegaard NH, Steffensen R, Sillesen H, Jensen G, Nordestgaard BG.Genetically reduced antioxidative protection and increased ischemic heart disease risk: The Copenhagen City Heart Study.Circulation. 2004; 109:59–65. doi: 10.1161/01.CIR.0000105720.28086.6C.LinkGoogle Scholar
    • 182. Hamanishi T, Furuta H, Kato H, Doi A, Tamai M, Shimomura H, Sakagashira S, Nishi M, Sasaki H, Sanke T, Nanjo K.Functional variants in the glutathione peroxidase-1 (GPx-1) gene are associated with increased intima-media thickness of carotid arteries and risk of macrovascular diseases in japanese type 2 diabetic patients.Diabetes. 2004; 53:2455–2460.CrossrefMedlineGoogle Scholar
    • 183. Heslop CL, Tebbutt SJ, Podder M, Ruan J, Hill JS.Combined polymorphisms in oxidative stress genes predict coronary artery disease and oxidative stress in coronary angiography patients.Ann Hum Genet. 2012; 76:435–447. doi: 10.1111/j.1469-1809.2012.00731.x.CrossrefMedlineGoogle Scholar
    • 184. Nemoto M, Nishimura R, Sasaki T, Hiki Y, Miyashita Y, Nishioka M, Fujimoto K, Sakuma T, Ohashi T, Fukuda K, Eto Y, Tajima N.Genetic association of glutathione peroxidase-1 with coronary artery calcification in type 2 diabetes: a case control study with multi-slice computed tomography.Cardiovasc Diabetol. 2007; 6:23. doi: 10.1186/1475-2840-6-23.CrossrefMedlineGoogle Scholar
    • 185. Najafi M, Ghasemi H, Roustazadeh A, Farajollahi M.Lack of association between glutathione peroxidase1 (GPx1) activity, Pro198Leu polymorphism and stenosis of coronary arteries: A population-based prediction.Meta Gene. 2014; 2:722–729. doi: 10.1016/j.mgene.2014.09.007.CrossrefMedlineGoogle Scholar
    • 186. Souiden Y, Mallouli H, Meskhi S, Chaabouni Y, Rebai A, Chéour F, Mahdouani K.MnSOD and GPx1 polymorphism relationship with coronary heart disease risk and severity.Biol Res. 2016; 49:22. doi: 10.1186/s40659-016-0083-6.CrossrefMedlineGoogle Scholar
    • 187. Casas JP, Cavalleri GL, Bautista LE, Smeeth L, Humphries SE, Hingorani AD.Endothelial nitric oxide synthase gene polymorphisms and cardiovascular disease: a HuGE review.Am J Epidemiol. 2006; 164:921–935. doi: 10.1093/aje/kwj302.CrossrefMedlineGoogle Scholar
    • 188. Casas JP, Bautista LE, Humphries SE, Hingorani AD.Endothelial nitric oxide synthase genotype and ischemic heart disease: meta-analysis of 26 studies involving 23028 subjects.Circulation. 2004; 109:1359–1365. doi: 10.1161/01.CIR.0000121357.76910.A3.LinkGoogle Scholar
    • 189. Rai H, Parveen F, Kumar S, Kapoor A, Sinha N.Association of endothelial nitric oxide synthase gene polymorphisms with coronary artery disease: an updated meta-analysis and systematic review.PLoS One. 2014; 9:e113363. doi: 10.1371/journal.pone.0113363.CrossrefMedlineGoogle Scholar
    • 190. Liu D, Jiang Z, Dai L, Zhang X, Yan C, Han Y.Association between the - 786T>C 1polymorphism in the promoter region of endothelial nitric oxide synthase (eNOS) and risk of coronary artery disease: a systematic review and meta-analysis.Gene. 2014; 545:175–183. doi: 10.1016/j.gene.2013.09.099.CrossrefMedlineGoogle Scholar
    • 191. San José G, Fortuño A, Beloqui O, Díez J, Zalba G.NADPH oxidase CYBA polymorphisms, oxidative stress and cardiovascular diseases.Clin Sci (Lond). 2008; 114:173–182. doi: 10.1042/CS20070130.CrossrefMedlineGoogle Scholar
    • 192. Wyche KE, Wang SS, Griendling KK, Dikalov SI, Austin H, Rao S, Fink B, Harrison DG, Zafari AM.C242T CYBA polymorphism of the NADPH oxidase is associated with reduced respiratory burst in human neutrophils.Hypertension. 2004; 43:1246–1251. doi: 10.1161/01.HYP.0000126579.50711.62.LinkGoogle Scholar
    • 193. Guzik TJ, West NE, Black E, McDonald D, Ratnatunga C, Pillai R, Channon KM.Functional effect of the C242T polymorphism in the NAD(P)H oxidase p22phox gene on vascular superoxide production in atherosclerosis.Circulation. 2000; 102:1744–1747.LinkGoogle Scholar
    • 194. Wu Z, Lou Y, Jin W, Liu Y, Lu L, Chen Q, Xie Y, Lu G.Relationship of the p22phox (CYBA) gene polymorphism C242T with risk of coronary artery disease: a meta-analysis.PLoS One. 2013; 8:e70885. doi: 10.1371/journal.pone.0070885.CrossrefMedlineGoogle Scholar
    • 195. Xu Q, Yuan F, Shen X, Wen H, Li W, Cheng B, Wu J.Polymorphisms of C242T and A640G in CYBA gene and the risk of coronary artery disease: a meta-analysis.PLoS One. 2014; 9:e84251. doi: 10.1371/journal.pone.0084251.CrossrefMedlineGoogle Scholar
    • 196. Violi F, Sanguigni V, Carnevale R, et al. Hereditary deficiency of gp91(phox) is associated with enhanced arterial dilatation: results of a multicenter study.Circulation. 2009; 120:1616–1622. doi: 10.1161/CIRCULATIONAHA.109.877191.LinkGoogle Scholar
    • 197. Loukogeorgakis SP, van den Berg MJ, Sofat R, Nitsch D, Charakida M, Haiyee B, de Groot E, MacAllister RJ, Kuijpers TW, Deanfield JE.Role of NADPH oxidase in endothelial ischemia/reperfusion injury in humans.Circulation. 2010; 121:2310–2316. doi: 10.1161/CIRCULATIONAHA.108.814731.LinkGoogle Scholar
    • 198. Violi F, Pignatelli P, Pignata C, Plebani A, Rossi P, Sanguigni V, Carnevale R, Soresina A, Finocchi A, Cirillo E, Catasca E, Angelico F, Loffredo L.Reduced atherosclerotic burden in subjects with genetically determined low oxidative stress.Arterioscler Thromb Vasc Biol. 2013; 33:406–412. doi: 10.1161/ATVBAHA.112.300438.LinkGoogle Scholar
    • 199. Sibley CT, Estwick T, Zavodni A, et al. Assessment of atherosclerosis in chronic granulomatous disease.Circulation. 2014; 130:2031–2039. doi: 10.1161/CIRCULATIONAHA.113.006824.LinkGoogle Scholar
    • 200. Sorescu D, Weiss D, Lassègue B, Clempus RE, Szöcs K, Sorescu GP, Valppu L, Quinn MT, Lambeth JD, Vega JD, Taylor WR, Griendling KK.Superoxide production and expression of nox family proteins in human atherosclerosis.Circulation. 2002; 105:1429–1435.LinkGoogle Scholar
    • 201. Mohammedi K, Bellili-Muñoz N, Marklund SL, Driss F, Le Nagard H, Patente TA, Fumeron F, Roussel R, Hadjadj S, Marre M, Velho G.Plasma extracellular superoxide dismutase concentration, allelic variations in the SOD3 gene and risk of myocardial infarction and all-cause mortality in people with type 1 and type 2 diabetes.Cardiovasc Diabetol. 2015; 14:845. doi: 10.1186/s12933-014-0163-2.CrossrefMedlineGoogle Scholar
    • 202. Sandström J, Nilsson P, Karlsson K, Marklund SL.10-fold increase in human plasma extracellular superoxide dismutase content caused by a mutation in heparin-binding domain.J Biol Chem. 1994; 269:19163–19166.CrossrefMedlineGoogle Scholar
    • 203. Adachi T, Yamada H, Yamada Y, Morihara N, Yamazaki N, Murakami T, Futenma A, Kato K, Hirano K.Substitution of glycine for arginine-213 in extracellular-superoxide dismutase impairs affinity for heparin and endothelial cell surface.Biochem J. 1996; 313 (Pt 1):235–239.CrossrefMedlineGoogle Scholar
    • 204. Chu Y, Alwahdani A, Iida S, Lund DD, Faraci FM, Heistad DD.Vascular effects of the human extracellular superoxide dismutase R213G variant.Circulation. 2005; 112:1047–1053. doi: 10.1161/CIRCULATIONAHA.104.531251.LinkGoogle Scholar
    • 205. Hartney JM, Stidham T, Goldstrohm DA, et al. A common polymorphism in extracellular superoxide dismutase affects cardiopulmonary disease risk by altering protein distribution.Circ Cardiovasc Genet. 2014; 7:659–666. doi: 10.1161/CIRCGENETICS.113.000504.LinkGoogle Scholar
    • 206. Ratnasinghe D, Tangrea JA, Andersen MR, Barrett MJ, Virtamo J, Taylor PR, Albanes D.Glutathione peroxidase codon 198 polymorphism variant increases lung cancer risk.Cancer Res. 2000; 60:6381–6383.MedlineGoogle Scholar
    • 207. Zhang JX, Wang ZM, Zhang JJ, Zhu LL, Gao XF, Chen SL.Association of glutathione peroxidase-1 (GPx-1) rs1050450 Pro198Leu and Pro197Leu polymorphisms with cardiovascular risk: a meta-analysis of observational studies.J Geriatr Cardiol. 2014; 11:141–150. doi: 10.3969/j.issn.1671-5411.2014.02.003.MedlineGoogle Scholar
    • 208. Forsberg L, de Faire U, Marklund SL, Andersson PM, Stegmayr B, Morgenstern R.Phenotype determination of a common Pro-Leu polymorphism in human glutathione peroxidase 1.Blood Cells Mol Dis. 2000; 26:423–426. doi: 10.1006/bcmd.2000.0325.CrossrefMedlineGoogle Scholar
    • 209. Vecoli C.Endothelial nitric oxide synthase gene polymorphisms in cardiovascular disease.Vitam Horm. 2014; 96:387–406. doi: 10.1016/B978-0-12-800254-4.00015-5.CrossrefMedlineGoogle Scholar
    • 210. Tesauro M, Thompson WC, Rogliani P, Qi L, Chaudhary PP, Moss J.Intracellular processing of endothelial nitric oxide synthase isoforms associated with differences in severity of cardiopulmonary diseases: cleavage of proteins with aspartate vs. glutamate at position 298.Proc Natl Acad Sci U S A. 2000; 97:2832–2835.CrossrefMedlineGoogle Scholar
    • 211. Persu A, Stoenoiu MS, Messiaen T, Davila S, Robino C, El-Khattabi O, Mourad M, Horie S, Feron O, Balligand JL, Wattiez R, Pirson Y, Chauveau D, Lens XM, Devuyst O.Modifier effect of ENOS in autosomal dominant polycystic kidney disease.Hum Mol Genet. 2002; 11:229–241.CrossrefMedlineGoogle Scholar
    • 212. Fairchild TA, Fulton D, Fontana JT, Gratton JP, McCabe TJ, Sessa WC.Acidic hydrolysis as a mechanism for the cleavage of the Glu(298)–>Asp variant of human endothelial nitric-oxide synthase.J Biol Chem. 2001; 276:26674–26679. doi: 10.1074/jbc.M103647200.CrossrefMedlineGoogle Scholar
    • 213. McDonald DM, Alp NJ, Channon KM.Functional comparison of the endothelial nitric oxide synthase Glu298Asp polymorphic variants in human endothelial cells.Pharmacogenetics. 2004; 14:831–839.CrossrefMedlineGoogle Scholar
    • 214. Joshi MS, Mineo C, Shaul PW, Bauer JA.Biochemical consequences of the NOS3 Glu298Asp variation in human endothelium: altered caveolar localization and impaired response to shear.FASEB J. 2007; 21:2655–2663. doi: 10.1096/fj.06-7088com.CrossrefMedlineGoogle Scholar
    • 215. Hingorani AD, Liang CF, Fatibene J, Lyon A, Monteith S, Parsons A, Haydock S, Hopper RV, Stephens NG, O’Shaughnessy KM, Brown MJ.A common variant of the endothelial nitric oxide synthase (Glu298–>Asp) is a major risk factor for coronary artery disease in the UK.Circulation. 1999; 100:1515–1520.LinkGoogle Scholar
    • 216. Wang XL, Sim AS, Badenhop RF, McCredie RM, Wilcken DE.A smoking-dependent risk of coronary artery disease associated with a polymorphism of the endothelial nitric oxide synthase gene.Nat Med. 1996; 2:41–45.CrossrefMedlineGoogle Scholar
    • 217. Colombo MG, Paradossi U, Andreassi MG, Botto N, Manfredi S, Masetti S, Biagini A, Clerico A.Endothelial nitric oxide synthase gene polymorphisms and risk of coronary artery disease.Clin Chem. 2003; 49:389–395.CrossrefMedlineGoogle Scholar
    • 218. Karvonen J, Kauma H, Kervinen K, Rantala M, Ikäheimo M, Päivänsalo M, Savolainen MJ, Kesäniemi YA.Endothelial nitric oxide synthase gene Glu298Asp polymorphism and blood pressure, left ventricular mass and carotid artery atherosclerosis in a population-based cohort.J Intern Med. 2002; 251:102–110.CrossrefMedlineGoogle Scholar
    • 219. Granath B, Taylor RR, van Bockxmeer FM, Mamotte CD.Lack of evidence for association between endothelial nitric oxide synthase gene polymorphisms and coronary artery disease in the Australian Caucasian population.J Cardiovasc Risk. 2001; 8:235–241.CrossrefMedlineGoogle Scholar
    • 220. Matsuno K, Yamada H, Iwata K, Jin D, Katsuyama M, Matsuki M, Takai S, Yamanishi K, Miyazaki M, Matsubara H, Yabe-Nishimura C.Nox1 is involved in angiotensin II-mediated hypertension: a study in Nox1-deficient mice.Circulation. 2005; 112:2677–2685. doi: 10.1161/CIRCULATIONAHA.105.573709.LinkGoogle Scholar
    • 221. Li H, Witte K, August M, Brausch I, Gödtel-Armbrust U, Habermeier A, Closs EI, Oelze M, Münzel T, Förstermann U.Reversal of endothelial nitric oxide synthase uncoupling and up-regulation of endothelial nitric oxide synthase expression lowers blood pressure in hypertensive rats.J Am Coll Cardiol. 2006; 47:2536–2544. doi: 10.1016/j.jacc.2006.01.071.CrossrefMedlineGoogle Scholar
    • 222. Chalupsky K, Cai H.Endothelial dihydrofolate reductase: critical for nitric oxide bioavailability and role in angiotensin II uncoupling of endothelial nitric oxide synthase.Proc Natl Acad Sci U S A. 2005; 102:9056–9061. doi: 10.1073/pnas.0409594102.CrossrefMedlineGoogle Scholar
    • 223. Demougeot C, Prigent-Tessier A, Bagnost T, André C, Guillaume Y, Bouhaddi M, Marie C, Berthelot A.Time course of vascular arginase expression and activity in spontaneously hypertensive rats.Life Sci. 2007; 80:1128–1134. doi: 10.1016/j.lfs.2006.12.003.CrossrefMedlineGoogle Scholar
    • 224. Rodriguez S, Richert L, Berthelot A.Increased arginase activity in aorta of mineralocorticoid-salt hypertensive rats.Clin Exp Hypertens. 2000; 22:75–85.CrossrefMedlineGoogle Scholar
    • 225. Johnson FK, Johnson RA, Peyton KJ, Durante W.Arginase inhibition restores arteriolar endothelial function in Dahl rats with salt-induced hypertension.Am J Physiol Regul Integr Comp Physiol. 2005; 288:R1057–R1062. doi: 10.1152/ajpregu.00758.2004.CrossrefMedlineGoogle Scholar
    • 226. Galougahi KK, Liu CC, Gentile C, Kok C, Nunez A, Garcia A, Fry NA, Davies MJ, Hawkins CL, Rasmussen HH, Figtree GA.Glutathionylation mediates angiotensin II-induced eNOS uncoupling, amplifying NADPH oxidase-dependent endothelial dysfunction.J Am Heart Assoc. 2014; 3:e000731. doi: 10.1161/JAHA.113.000731.LinkGoogle Scholar
    • 227. Pritchard KA, Groszek L, Smalley DM, Sessa WC, Wu M, Villalon P, Wolin MS, Stemerman MB.Native low-density lipoprotein increases endothelial cell nitric oxide synthase generation of superoxide anion.Circ Res. 1995; 77:510–518.LinkGoogle Scholar
    • 228. Stepp DW, Ou J, Ackerman AW, Welak S, Klick D, Pritchard KANative LDL and minimally oxidized LDL differentially regulate superoxide anion in vascular endothelium in situ.Am J Physiol Heart Circ Physiol. 2002; 283:H750–H759. doi: 10.1152/ajpheart.00029.2002.CrossrefMedlineGoogle Scholar
    • 229. Nickenig G, Bäumer AT, Temur Y, Kebben D, Jockenhövel F, Böhm M.Statin-sensitive dysregulated AT1 receptor function and density in hypercholesterolemic men.Circulation. 1999; 100:2131–2134.LinkGoogle Scholar
    • 230. Ryoo S, Gupta G, Benjo A, et al. Endothelial arginase II: a novel target for the treatment of atherosclerosis.Circ Res. 2008; 102:923–932. doi: 10.1161/CIRCRESAHA.107.169573.LinkGoogle Scholar
    • 231. Erdely A, Kepka-Lenhart D, Salmen-Muniz R, Chapman R, Hulderman T, Kashon M, Simeonova PP, Morris SMArginase activities and global arginine bioavailability in wild-type and ApoE-deficient mice: responses to high fat and high cholesterol diets.PLoS One. 2010; 5:e15253. doi: 10.1371/journal.pone.0015253.CrossrefMedlineGoogle Scholar
    • 232. Hayashi T, Esaki T, Sumi D, Mukherjee T, Iguchi A, Chaudhuri G.Modulating role of estradiol on arginase II expression in hyperlipidemic rabbits as an atheroprotective mechanism.Proc Natl Acad Sci U S A. 2006; 103:10485–10490. doi: 10.1073/pnas.0603918103.CrossrefMedlineGoogle Scholar
    • 233. Vaisman BL, Andrews KL, Khong SM, Wood KC, Moore XL, Fu Y, Kepka-Lenhart DM, Morris SM, Remaley AT, Chin-Dusting JP.Selective endothelial overexpression of arginase II induces endothelial dysfunction and hypertension and enhances atherosclerosis in mice.PLoS One. 2012; 7:e39487. doi: 10.1371/journal.pone.0039487.CrossrefMedlineGoogle Scholar
    • 234. Feron O, Dessy C, Moniotte S, Desager JP, Balligand JL.Hypercholesterolemia decreases nitric oxide production by promoting the interaction of caveolin and endothelial nitric oxide synthase.J Clin Invest. 1999; 103:897–905. doi: 10.1172/JCI4829.CrossrefMedlineGoogle Scholar
    • 235. 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; 103:2102–2107.LinkGoogle Scholar
    • 236. Steffen Y, Jung T, Klotz LO, Schewe T, Grune T, Sies H.Protein modification elicited by oxidized low-density lipoprotein (LDL) in endothelial cells: protection by (-)-epicatechin.Free Radic Biol Med. 2007; 42:955–970. doi: 10.1016/j.freeradbiomed.2006.12.024.CrossrefMedlineGoogle Scholar
    • 237. Jaimes EA, DeMaster EG, Tian RX, Raij L.Stable compounds of cigarette smoke induce endothelial superoxide anion production via NADPH oxidase activation.Arterioscler Thromb Vasc Biol. 2004; 24:1031–1036. doi: 10.1161/01.ATV.0000127083.88549.58.LinkGoogle Scholar
    • 238. Csiszar A, Labinskyy N, Pinto JT, Ballabh P, Zhang H, Losonczy G, Pearson K, de Cabo R, Pacher P, Zhang C, Ungvari Z.Resveratrol induces mitochondrial biogenesis in endothelial cells.Am J Physiol Heart Circ Physiol. 2009; 297:H13–H20. doi: 10.1152/ajpheart.00368.2009.CrossrefMedlineGoogle Scholar
    • 239. Steffen Y, Vuillaume G, Stolle K, Roewer K, Lietz M, Schueller J, Lebrun S, Wallerath T.Cigarette smoke and LDL cooperate in reducing nitric oxide bioavailability in endothelial cells via effects on both eNOS and NADPH oxidase.Nitric Oxide. 2012; 27:176–184. doi: 10.1016/j.niox.2012.06.006.CrossrefMedlineGoogle Scholar
    • 240. Giacco F, Brownlee M.Oxidative stress and diabetic complications.Circ Res. 2010; 107:1058–1070. doi: 10.1161/CIRCRESAHA.110.223545.LinkGoogle Scholar
    • 241. Funk DM.Coagulation assays and anticoagulant monitoring.Hematology Am Soc Hematol Educ Program. 2012; 2012:460–465. doi: 10.1182/asheducation-2012.1.460.CrossrefMedlineGoogle Scholar
    • 242. Hink U, Li H, Mollnau H, Oelze M, Matheis E, Hartmann M, Skatchkov M, Thaiss F, Stahl RA, Warnholtz A, Meinertz T, Griendling K, Harrison DG, Forstermann U, Munzel T.Mechanisms underlying endothelial dysfunction in diabetes mellitus.Circ Res. 2001; 88:E14–E22.LinkGoogle Scholar
    • 243. Alp NJ, Mussa S, Khoo J, Cai S, Guzik T, Jefferson A, Goh N, Rockett KA, Channon KM.Tetrahydrobiopterin-dependent preservation of nitric oxide-mediated endothelial function in diabetes by targeted transgenic GTP-cyclohydrolase I overexpression.J Clin Invest. 2003; 112:725–735. doi: 10.1172/JCI17786.CrossrefMedlineGoogle Scholar
    • 244. Faria AM, Papadimitriou A, Silva KC, Lopes de Faria JM, Lopes de Faria JB.Uncoupling endothelial nitric oxide synthase is ameliorated by green tea in experimental diabetes by re-establishing tetrahydrobiopterin levels.Diabetes. 2012; 61:1838–1847. doi: 10.2337/db11-1241.CrossrefMedlineGoogle Scholar
    • 245. Wenzel P, Daiber A, Oelze M, Brandt M, Closs E, Xu J, Thum T, Bauersachs J, Ertl G, Zou MH, Förstermann U, Münzel T.Mechanisms underlying recoupling of eNOS by HMG-CoA reductase inhibition in a rat model of streptozotocin-induced diabetes mellitus.Atherosclerosis. 2008; 198:65–76. doi: 10.1016/j.atherosclerosis.2007.10.003.CrossrefMedlineGoogle Scholar
    • 246. Xu J, Wu Y, Song P, Zhang M, Wang S, Zou MH.Proteasome-dependent degradation of guanosine 5’-triphosphate cyclohydrolase I causes tetrahydrobiopterin deficiency in diabetes mellitus.Circulation. 2007; 116:944–953. doi: 10.1161/CIRCULATIONAHA.106.684795.LinkGoogle Scholar
    • 247. Schuhmacher S, Oelze M, Bollmann F, et al. Vascular dysfunction in experimental diabetes is improved by pentaerithrityl tetranitrate but not isosorbide-5-mononitrate therapy.Diabetes. 2011; 60:2608–2616. doi: 10.2337/db10-1395.CrossrefMedlineGoogle Scholar
    • 248. Pannirselvam M, Simon V, Verma S, Anderson T, Triggle CR.Chronic oral supplementation with sepiapterin prevents endothelial dysfunction and oxidative stress in small mesenteric arteries from diabetic (db/db) mice.Br J Pharmacol. 2003; 140:701–706. doi: 10.1038/sj.bjp.0705476.CrossrefMedlineGoogle Scholar
    • 249. Pannirselvam M, Verma S, Anderson TJ, Triggle CR.Cellular basis of endothelial dysfunction in small mesenteric arteries from spontaneously diabetic (db/db -/-) mice: role of decreased tetrahydrobiopterin bioavailability.Br J Pharmacol. 2002; 136:255–263. doi: 10.1038/sj.bjp.0704683.CrossrefMedlineGoogle Scholar
    • 250. Shinozaki K, Nishio Y, Okamura T, Yoshida Y, Maegawa H, Kojima H, Masada M, Toda N, Kikkawa R, Kashiwagi A.Oral administration of tetrahydrobiopterin prevents endothelial dysfunction and vascular oxidative stress in the aortas of insulin-resistant rats.Circ Res. 2000; 87:566–573.LinkGoogle Scholar
    • 251. Romero MJ, Platt DH, Tawfik HE, Labazi M, El-Remessy AB, Bartoli M, Caldwell RB, Caldwell RW.Diabetes-induced coronary vascular dysfunction involves increased arginase activity.Circ Res. 2008; 102:95–102. doi: 10.1161/CIRCRESAHA.107.155028.LinkGoogle Scholar
    • 252. Yao L, Chandra S, Toque HA, Bhatta A, Rojas M, Caldwell RB, Caldwell RW.Prevention of diabetes-induced arginase activation and vascular dysfunction by Rho kinase (ROCK) knockout.Cardiovasc Res. 2013; 97:509–519. doi: 10.1093/cvr/cvs371.CrossrefMedlineGoogle Scholar
    • 253. Giri H, Muthuramu I, Dhar M, Rathnakumar K, Ram U, Dixit M.Protein tyrosine phosphatase SHP2 mediates chronic insulin-induced endothelial inflammation.Arterioscler Thromb Vasc Biol. 2012; 32:1943–1950. doi: 10.1161/ATVBAHA.111.239251.LinkGoogle Scholar
    • 254. Romero MJ, Iddings JA, Platt DH, Ali MI, Cederbaum SD, Stepp DW, Caldwell RB, Caldwell RW.Diabetes-induced vascular dysfunction involves arginase I.Am J Physiol Heart Circ Physiol. 2012; 302:H159–H166. doi: 10.1152/ajpheart.00774.2011.CrossrefMedlineGoogle Scholar
    • 255. Toque HA, Tostes RC, Yao L, Xu Z, Webb RC, Caldwell RB, Caldwell RW.Arginase II deletion increases corpora cavernosa relaxation in diabetic mice.J Sex Med. 2011; 8:722–733. doi: 10.1111/j.1743-6109.2010.02098.x.CrossrefMedlineGoogle Scholar
    • 256. Grönros J, Jung C, Lundberg JO, Cerrato R, Ostenson CG, Pernow J.Arginase inhibition restores in vivo coronary microvascular function in type 2 diabetic rats.Am J Physiol Heart Circ Physiol. 2011; 300:H1174–H1181. doi: 10.1152/ajpheart.00560.2010.CrossrefMedlineGoogle Scholar
    • 257. Kashyap SR, Lara A, Zhang R, Park YM, DeFronzo RA.Insulin reduces plasma arginase activity in type 2 diabetic patients.Diabetes Care. 2008; 31:134–139. doi: 10.2337/dc07-1198.CrossrefMedlineGoogle Scholar
    • 258. Beleznai T, Feher A, Spielvogel D, Lansman SL, Bagi Z.Arginase 1 contributes to diminished coronary arteriolar dilation in patients with diabetes.Am J Physiol Heart Circ Physiol. 2011; 300:H777–H783. doi: 10.1152/ajpheart.00831.2010.CrossrefMedlineGoogle Scholar
    • 259. Shemyakin A, Kövamees O, Rafnsson A, Böhm F, Svenarud P, Settergren M, Jung C, Pernow J.Arginase inhibition improves endothelial function in patients with coronary artery disease and type 2 diabetes mellitus.Circulation. 2012; 126:2943–2950. doi: 10.1161/CIRCULATIONAHA.112.140335.LinkGoogle Scholar
    • 260. Kwak BR, Bäck M, Bochaton-Piallat ML, et al. Biomechanical factors in atherosclerosis: mechanisms and clinical implications.Eur Heart J. 2014; 35:3013–20, 3020a. doi: 10.1093/eurheartj/ehu353.CrossrefMedlineGoogle Scholar
    • 261. Cheng C, Tempel D, van Haperen R, van der Baan A, Grosveld F, Daemen MJ, Krams R, de Crom R.Atherosclerotic lesion size and vulnerability are determined by patterns of fluid shear stress.Circulation. 2006; 113:2744–2753. doi: 10.1161/CIRCULATIONAHA.105.590018.LinkGoogle Scholar
    • 262. Cheng C, van Haperen R, de Waard M, van Damme LC, Tempel D, Hanemaaijer L, van Cappellen GW, Bos J, Slager CJ, Duncker DJ, van der Steen AF, de Crom R, Krams R.Shear stress affects the intracellular distribution of eNOS: direct demonstration by a novel in vivo technique.Blood. 2005; 106:3691–3698. doi: 10.1182/blood-2005-06-2326.CrossrefMedlineGoogle Scholar
    • 263. Nam D, Ni CW, Rezvan A, Suo J, Budzyn K, Llanos A, Harrison D, Giddens D, Jo H.Partial carotid ligation is a model of acutely induced disturbed flow, leading to rapid endothelial dysfunction and atherosclerosis.Am J Physiol Heart Circ Physiol. 2009; 297:H1535–H1543. doi: 10.1152/ajpheart.00510.2009.CrossrefMedlineGoogle Scholar
    • 264. Chen YC, Bui AV, Diesch J, Manasseh R, Hausding C, Rivera J, Haviv I, Agrotis A, Htun NM, Jowett J, Hagemeyer CE, Hannan RD, Bobik A, Peter K.A novel mouse model of atherosclerotic plaque instability for drug testing and mechanistic/therapeutic discoveries using gene and microRNA expression profiling.Circ Res. 2013; 113:252–265. doi: 10.1161/CIRCRESAHA.113.301562.LinkGoogle Scholar
    • 265. Hopkins PN.Molecular biology of atherosclerosis.Physiol Rev. 2013; 93:1317–1542. doi: 10.1152/physrev.00004.2012.CrossrefMedlineGoogle Scholar
    • 266. Chatzizisis YS, Coskun AU, Jonas M, Edelman ER, Feldman CL, Stone PH.Role of endothelial shear stress in the natural history of coronary atherosclerosis and vascular remodeling: molecular, cellular, and vascular behavior.J Am Coll Cardiol. 2007; 49:2379–2393. doi: 10.1016/j.jacc.2007.02.059.CrossrefMedlineGoogle Scholar
    • 267. Noris M, Morigi M, Donadelli R, Aiello S, Foppolo M, Todeschini M, Orisio S, Remuzzi G, Remuzzi A.Nitric oxide synthesis by cultured endothelial cells is modulated by flow conditions.Circ Res. 1995; 76:536–543.LinkGoogle Scholar
    • 268. Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM.Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation.Nature. 1999; 399:601–605. doi: 10.1038/21224.CrossrefMedlineGoogle Scholar
    • 269. Gallis B, Corthals GL, Goodlett DR, Ueba H, Kim F, Presnell SR, Figeys D, Harrison DG, Berk BC, Aebersold R, Corson MA.Identification of flow-dependent endothelial nitric-oxide synthase phosphorylation sites by mass spectrometry and regulation of phosphorylation and nitric oxide production by the phosphatidylinositol 3-kinase inhibitor LY294002.J Biol Chem. 1999; 274:30101–30108.CrossrefMedlineGoogle Scholar
    • 270. Fleming I.Molecular mechanisms underlying the activation of eNOS.Pflugers Arch. 2010; 459:793–806. doi: 10.1007/s00424-009-0767-7.CrossrefMedlineGoogle Scholar
    • 271. Lam CF, Peterson TE, Richardson DM, Croatt AJ, d’Uscio LV, Nath KA, Katusic ZS.Increased blood flow causes coordinated upregulation of arterial eNOS and biosynthesis of tetrahydrobiopterin.Am J Physiol Heart Circ Physiol. 2006; 290:H786–H793. doi: 10.1152/ajpheart.00759.2005.CrossrefMedlineGoogle Scholar
    • 272. Gambillara V, Chambaz C, Montorzi G, Roy S, Stergiopulos N, Silacci P.Plaque-prone hemodynamics impair endothelial function in pig carotid arteries.Am J Physiol Heart Circ Physiol. 2006; 290:H2320–H2328. doi: 10.1152/ajpheart.00486.2005.CrossrefMedlineGoogle Scholar
    • 273. Ziegler T, Bouzourène 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.Arterioscler Thromb Vasc Biol. 1998; 18:686–692.LinkGoogle Scholar
    • 274. De Keulenaer GW, Chappell DC, Ishizaka N, Nerem RM, Alexander RW, Griendling KK.Oscillatory and steady laminar shear stress differentially affect human endothelial redox state: role of a superoxide-producing NADH oxidase.Circ Res. 1998; 82:1094–1101.LinkGoogle Scholar
    • 275. Hwang J, Ing MH, Salazar A, Lassègue B, Griendling K, Navab M, Sevanian A, Hsiai TK.Pulsatile versus oscillatory shear stress regulates NADPH oxidase subunit expression: implication for native LDL oxidation.Circ Res. 2003; 93:1225–1232. doi: 10.1161/01.RES.0000104087.29395.66.LinkGoogle Scholar
    • 276. Harris CM, Sanders SA, Massey V.Role of the flavin midpoint potential and NAD binding in determining NAD versus oxygen reactivity of xanthine oxidoreductase.J Biol Chem. 1999; 274:4561–4569.CrossrefMedlineGoogle Scholar
    • 277. Lima B, Forrester MT, Hess DT, Stamler JS.S-nitrosylation in cardiovascular signaling.Circ Res. 2010; 106:633–646. doi: 10.1161/CIRCRESAHA.109.207381.LinkGoogle Scholar
    • 278. Selemidis S, Dusting GJ, Peshavariya H, Kemp-Harper BK, Drummond GR.Nitric oxide suppresses NADPH oxidase-dependent superoxide production by S-nitrosylation in human endothelial cells.Cardiovasc Res. 2007; 75:349–358. doi: 10.1016/j.cardiores.2007.03.030.CrossrefMedlineGoogle Scholar
    • 279. Ravi K, Brennan LA, Levic S, Ross PA, Black SM.S-nitrosylation of endothelial nitric oxide synthase is associated with monomerization and decreased enzyme activity.Proc Natl Acad Sci U S A. 2004; 101:2619–2624.CrossrefMedlineGoogle Scholar
    • 280. Liu L, Yan Y, Zeng M, Zhang J, Hanes MA, Ahearn G, McMahon TJ, Dickfeld T, Marshall HE, Que LG, Stamler JS.Essential roles of S-nitrosothiols in vascular homeostasis and endotoxic shock.Cell. 2004; 116:617–628.CrossrefMedlineGoogle Scholar
    • 281. Song P, Zou MH.Redox regulation of endothelial cell fate.Cell Mol Life Sci. 2014; 71:3219–3239. doi: 10.1007/s00018-014-1598-z.CrossrefMedlineGoogle Scholar
    • 282. King AL, Polhemus DJ, Bhushan S, et al. Hydrogen sulfide cytoprotective signaling is endothelial nitric oxide synthase-nitric oxide dependent.Proc Natl Acad Sci U S A. 2014; 111:3182–3187. doi: 10.1073/pnas.1321871111.CrossrefMedlineGoogle Scholar
    • 283. Wang R, Szabo C, Ichinose F, Ahmed A, Whiteman M, Papapetropoulos A.The role of H2S bioavailability in endothelial dysfunction.Trends Pharmacol Sci. 2015; 36:568–578. doi: 10.1016/j.tips.2015.05.007.CrossrefMedlineGoogle Scholar
    • 284. Yuan S, Patel RP, Kevil CG.Working with nitric oxide and hydrogen sulfide in biological systems.Am J Physiol Lung Cell Mol Physiol. 2015; 308:L403–L415. doi: 10.1152/ajplung.00327.2014.CrossrefMedlineGoogle Scholar
    • 285. Altaany Z, Ju Y, Yang G, Wang R.The coordination of S-sulfhydration, S-nitrosylation, and phosphorylation of endothelial nitric oxide synthase by hydrogen sulfide.Sci Signal. 2014; 7:ra87. doi: 10.1126/scisignal.2005478.CrossrefMedlineGoogle Scholar
    • 286. Chung HS, Wang SB, Venkatraman V, Murray CI, Van Eyk JE.Cysteine oxidative posttranslational modifications: emerging regulation in the cardiovascular system.Circ Res. 2013; 112:382–392. doi: 10.1161/CIRCRESAHA.112.268680.LinkGoogle Scholar
    • 287. Brown DI, Griendling KK.Regulation of signal transduction by reactive oxygen species in the cardiovascular system.Circ Res. 2015; 116:531–549. doi: 10.1161/CIRCRESAHA.116.303584.LinkGoogle Scholar
    • 288. Wadham C, Parker A, Wang L, Xia P.High glucose attenuates protein S-nitrosylation in endothelial cells: role of oxidative stress.Diabetes. 2007; 56:2715–2721. doi: 10.2337/db06-1294.CrossrefMedlineGoogle Scholar
    • 289. Steffensen LB, Mortensen MB, Kjolby M, Hagensen MK, Oxvig C, Bentzon JF.Disturbed Laminar Blood Flow Vastly Augments Lipoprotein Retention in the Artery Wall: A Key Mechanism Distinguishing Susceptible From Resistant Sites.Arterioscler Thromb Vasc Biol. 2015; 35:1928–1935. doi: 10.1161/ATVBAHA.115.305874.LinkGoogle Scholar
    • 290. Peiffer V, Sherwin SJ, Weinberg PD.Does low and oscillatory wall shear stress correlate spatially with early atherosclerosis? A systematic review.Cardiovasc Res. 2013; 99:242–250. doi: 10.1093/cvr/cvt044.CrossrefMedlineGoogle Scholar
    • 291. Kwon GP, Schroeder JL, Amar MJ, Remaley AT, Balaban RS.Contribution of macromolecular structure to the retention of low-density lipoprotein at arterial branch points.Circulation. 2008; 117:2919–2927. doi: 10.1161/CIRCULATIONAHA.107.754614.LinkGoogle Scholar
    • 292. von der Thüsen JH, van Berkel TJ, Biessen EA.Induction of rapid atherogenesis by perivascular carotid collar placement in apolipoprotein E-deficient and low-density lipoprotein receptor-deficient mice.Circulation. 2001; 103:1164–1170.LinkGoogle Scholar
    • 293. Wight TN, Merrilees MJ.Proteoglycans in atherosclerosis and restenosis: key roles for versican.Circ Res. 2004; 94:1158–1167. doi: 10.1161/01.RES.0000126921.29919.51.LinkGoogle Scholar
    • 294. Mangat R, Warnakula S, Borthwick F, Hassanali Z, Uwiera RR, Russell JC, Cheeseman CI, Vine DF, Proctor SD.Arterial retention of remnant lipoproteins ex vivo is increased in insulin resistance because of increased arterial biglycan and production of cholesterol-rich atherogenic particles that can be improved by ezetimibe in the JCR:LA-cp rat.J Am Heart Assoc. 2012; 1:e003434. doi: 10.1161/JAHA.112.003434.LinkGoogle Scholar
    • 295. Ballinger ML, Osman N, Hashimura K, de Haan JB, Jandeleit-Dahm K, Allen T, Tannock LR, Rutledge JC, Little PJ.Imatinib inhibits vascular smooth muscle proteoglycan synthesis and reduces LDL binding in vitro and aortic lipid deposition in vivo.J Cell Mol Med. 2010; 14:1408–1418. doi: 10.1111/j.1582-4934.2009.00902.x.CrossrefMedlineGoogle Scholar
    • 296. Osman N, Getachew R, Thach L, Wang H, Su X, Zheng W, Little PJ.Platelet-derived growth factor-stimulated versican synthesis but not glycosaminoglycan elongation in vascular smooth muscle is mediated via Akt phosphorylation.Cell Signal. 2014; 26:912–916. doi: 10.1016/j.cellsig.2014.01.019.CrossrefMedlineGoogle Scholar
    • 297. Meyers CD, Tannock LR, Wight TN, Chait A.Statin-exposed vascular smooth muscle cells secrete proteoglycans with decreased binding affinity for LDL.J Lipid Res. 2003; 44:2152–2160. doi: 10.1194/jlr.M300252-JLR200.CrossrefMedlineGoogle Scholar
    • 298. Cardona-Sanclemente LE, Born GV.Effect of inhibition of nitric oxide synthesis on the uptake of LDL and fibrinogen by arterial walls and other organs of the rat.Br J Pharmacol. 1995; 114:1490–1494.CrossrefMedlineGoogle Scholar
    • 299. Weinberg PD.Rate-limiting steps in the development of atherosclerosis: the response-to-influx theory.J Vasc Res. 2004; 41:1–17. doi: 10.1159/000076124.CrossrefMedlineGoogle Scholar
    • 300. Won D, Zhu SN, Chen M, Teichert AM, Fish JE, Matouk CC, Bonert M, Ojha M, Marsden PA, Cybulsky MI.Relative reduction of endothelial nitric-oxide synthase expression and transcription in atherosclerosis-prone regions of the mouse aorta and in an in vitro model of disturbed flow.Am J Pathol. 2007; 171:1691–1704. doi: 10.2353/ajpath.2007.060860.CrossrefMedlineGoogle Scholar
    • 301. Hahn C, Schwartz MA.Mechanotransduction in vascular physiology and atherogenesis.Nat Rev Mol Cell Biol. 2009; 10:53–62. doi: 10.1038/nrm2596.CrossrefMedlineGoogle Scholar
    • 302. Pan S.Molecular mechanisms responsible for the atheroprotective effects of laminar shear stress.Antioxid Redox Signal. 2009; 11:1669–1682. doi: 10.1089/ars.2009.2487.CrossrefMedlineGoogle Scholar
    • 303. Leopold JA, Loscalzo J.Oxidative risk for atherothrombotic cardiovascular disease.Free Radic Biol Med. 2009; 47:1673–1706. doi: 10.1016/j.freeradbiomed.2009.09.009.CrossrefMedlineGoogle Scholar
    • 304. Steinberg D.The LDL modification hypothesis of atherogenesis: an update.J Lipid Res. 2009; 50 Suppl:S376–S381. doi: 10.1194/jlr.R800087-JLR200.CrossrefMedlineGoogle Scholar
    • 305. Steinberg D, Witztum JL.Oxidized low-density lipoprotein and atherosclerosis.Arterioscler Thromb Vasc Biol. 2010; 30:2311–2316. doi: 10.1161/ATVBAHA.108.179697.LinkGoogle Scholar
    • 306. Libby P, Ridker PM, Hansson GK.Progress and challenges in translating the biology of atherosclerosis.Nature. 2011; 473:317–325. doi: 10.1038/nature10146.CrossrefMedlineGoogle Scholar
    • 307. Bochkov VN, Oskolkova OV, Birukov KG, Levonen AL, Binder CJ, Stöckl J.Generation and biological activities of oxidized phospholipids.Antioxid Redox Signal. 2010; 12:1009–1059. doi: 10.1089/ars.2009.2597.CrossrefMedlineGoogle Scholar
    • 308. Binder CJ, Papac-Milicevic N, Witztum JL.Innate sensing of oxidation-specific epitopes in health and disease.Nat Rev Immunol. 2016; 16:485–497. doi: 10.1038/nri.2016.63.CrossrefMedlineGoogle Scholar
    • 309. Cyrus T, Witztum JL, Rader DJ, Tangirala R, Fazio S, Linton MF, Funk CD.Disruption of the 12/15-lipoxygenase gene diminishes atherosclerosis in apo E-deficient mice.J Clin Invest. 1999; 103:1597–1604. doi: 10.1172/JCI5897.CrossrefMedlineGoogle Scholar
    • 310. Cyrus T, Praticò D, Zhao L, Witztum JL, Rader DJ, Rokach J, FitzGerald GA, Funk CD.Absence of 12/15-lipoxygenase expression decreases lipid peroxidation and atherogenesis in apolipoprotein e-deficient mice.Circulation. 2001; 103:2277–2282.LinkGoogle Scholar
    • 311. George J, Afek A, Shaish A, Levkovitz H, Bloom N, Cyrus T, Zhao L, Funk CD, Sigal E, Harats D.12/15-Lipoxygenase gene disruption attenuates atherogenesis in LDL receptor-deficient mice.Circulation. 2001; 104:1646–1650.LinkGoogle Scholar
    • 312. Zhao L, Cuff CA, Moss E, Wille U, Cyrus T, Klein EA, Praticò D, Rader DJ, Hunter CA, Puré E, Funk CD.Selective interleukin-12 synthesis defect in 12/15-lipoxygenase-deficient macrophages associated with reduced atherosclerosis in a mouse model of familial hypercholesterolemia.J Biol Chem. 2002; 277:35350–35356. doi: 10.1074/jbc.M205738200.CrossrefMedlineGoogle Scholar
    • 313. Shih DM, Xia YR, Wang XP, Miller E, Castellani LW, Subbanagounder G, Cheroutre H, Faull KF, Berliner JA, Witztum JL, Lusis AJ.Combined serum paraoxonase knockout/apolipoprotein E knockout mice exhibit increased lipoprotein oxidation and atherosclerosis.J Biol Chem. 2000; 275:17527–17535. doi: 10.1074/jbc.M910376199.CrossrefMedlineGoogle Scholar
    • 314. Hörkkö S, Bird DA, Miller E, Itabe H, Leitinger N, Subbanagounder G, Berliner JA, Friedman P, Dennis EA, Curtiss LK, Palinski W, Witztum JL.Monoclonal autoantibodies specific for oxidized phospholipids or oxidized phospholipid-protein adducts inhibit macrophage uptake of oxidized low-density lipoproteins.J Clin Invest. 1999; 103:117–128. doi: 10.1172/JCI4533.CrossrefMedlineGoogle Scholar
    • 315. Binder CJ, Hörkkö S, Dewan A, Chang MK, Kieu EP, Goodyear CS, Shaw PX, Palinski W, Witztum JL, Silverman GJ.Pneumococcal vaccination decreases atherosclerotic lesion formation: molecular mimicry between Streptococcus pneumoniae and oxidized LDL.Nat Med. 2003; 9:736–743. doi: 10.1038/nm876.CrossrefMedlineGoogle Scholar
    • 316. Faria-Neto JR, Chyu KY, Li X, Dimayuga PC, Ferreira C, Yano J, Cercek B, Shah PK.Passive immunization with monoclonal IgM antibodies against phosphorylcholine reduces accelerated vein graft atherosclerosis in apolipoprotein E-null mice.Atherosclerosis. 2006; 189:83–90. doi: 10.1016/j.atherosclerosis.2005.11.033.CrossrefMedlineGoogle Scholar
    • 317. Tsiantoulas D, Diehl CJ, Witztum JL, Binder CJ.B cells and humoral immunity in atherosclerosis.Circ Res. 2014; 114:1743–1756. doi: 10.1161/CIRCRESAHA.113.301145.LinkGoogle Scholar
    • 318. Kirabo A, Fontana V, de Faria AP, et al. DC isoketal-modified proteins activate T cells and promote hypertension.J Clin Invest. 2014; 124:4642–4656. doi: 10.1172/JCI74084.CrossrefMedlineGoogle Scholar
    • 319. Pober JS.Is hypertension an autoimmune disease?J Clin Invest. 2014; 124:4234–4236. doi: 10.1172/JCI77766.CrossrefMedlineGoogle Scholar
    • 320. Goss SP, Hogg N, Kalyanaraman B.The effect of nitric oxide release rates on the oxidation of human low density lipoprotein.J Biol Chem. 1997; 272:21647–21653.CrossrefMedlineGoogle Scholar
    • 321. Hogg N, Kalyanaraman B, Joseph J, Struck A, Parthasarathy S.Inhibition of low-density lipoprotein oxidation by nitric oxide. Potential role in atherogenesis.FEBS Lett. 1993; 334:170–174.CrossrefMedlineGoogle Scholar
    • 322. Kumano-Kuramochi M, Shimozu Y, Wakita C, Ohnishi-Kameyama M, Shibata T, Matsunaga S, Takano-Ishikawa Y, Watanabe J, Goto M, Xie Q, Komba S, Uchida K, Machida S.Identification of 4-hydroxy-2-nonenal-histidine adducts that serve as ligands for human lectin-like oxidized LDL receptor-1.Biochem J. 2012; 442:171–180. doi: 10.1042/BJ20111029.CrossrefMedlineGoogle Scholar
    • 323. West XZ, Malinin NL, Merkulova AA, Tischenko M, Kerr BA, Borden EC, Podrez EA, Salomon RG, Byzova TV.Oxidative stress induces angiogenesis by activating TLR2 with novel endogenous ligands.Nature. 2010; 467:972–976. doi: 10.1038/nature09421.CrossrefMedlineGoogle Scholar
    • 324. Mehta JL, Sanada N, Hu CP, et al. Deletion of LOX-1 reduces atherogenesis in LDLR knockout mice fed high cholesterol diet.Circ Res. 2007; 100:1634–1642. doi: 10.1161/CIRCRESAHA.107.149724.LinkGoogle Scholar
    • 325. Yokoyama C, Aoyama T, Ido T, Kakino A, Shiraki T, Tanaka T, Nishigaki K, Hasegawa A, Fujita Y, Sawamura T, Minatoguchi S.Deletion of LOX-1 Protects against Heart Failure Induced by Doxorubicin.PLoS One. 2016; 11:e0154994. doi: 10.1371/journal.pone.0154994.CrossrefMedlineGoogle Scholar
    • 326. Mullick AE, Soldau K, Kiosses WB, Bell TA, Tobias PS, Curtiss LK.Increased endothelial expression of Toll-like receptor 2 at sites of disturbed blood flow exacerbates early atherogenic events.J Exp Med. 2008; 205:373–383. doi: 10.1084/jem.20071096.CrossrefMedlineGoogle Scholar
    • 327. Korenaga R, Ando J, Kosaki K, Isshiki M, Takada Y, Kamiya A.Negative transcriptional regulation of the VCAM-1 gene by fluid shear stress in murine endothelial cells.Am J Physiol. 1997; 273:C1506–C1515.CrossrefMedlineGoogle Scholar
    • 328. Chappell DC, Varner SE, Nerem RM, Medford RM, Alexander RW.Oscillatory shear stress stimulates adhesion molecule expression in cultured human endothelium.Circ Res. 1998; 82:532–539.LinkGoogle Scholar
    • 329. Nakashima Y, Raines EW, Plump AS, Breslow JL, Ross R.Upregulation of VCAM-1 and ICAM-1 at atherosclerosis-prone sites on the endothelium in the ApoE-deficient mouse.Arterioscler Thromb Vasc Biol. 1998; 18:842–851.LinkGoogle Scholar
    • 330. Marui N, Offermann MK, Swerlick R, Kunsch C, Rosen CA, Ahmad M, Alexander RW, Medford RM.Vascular cell adhesion molecule-1 (VCAM-1) gene transcription and expression are regulated through an antioxidant-sensitive mechanism in human vascular endothelial cells.J Clin Invest. 1993; 92:1866–1874. doi: 10.1172/JCI116778.CrossrefMedlineGoogle Scholar
    • 331. Khan BV, Harrison DG, Olbrych MT, Alexander RW, Medford RM.Nitric oxide regulates vascular cell adhesion molecule 1 gene expression and redox-sensitive transcriptional events in human vascular endothelial cells.Proc Natl Acad Sci U S A. 1996; 93:9114–9119.CrossrefMedlineGoogle Scholar
    • 332. Pueyo ME, Gonzalez W, Nicoletti A, Savoie F, Arnal JF, Michel JB.Angiotensin II stimulates endothelial vascular cell adhesion molecule-1 via nuclear factor-kappaB activation induced by intracellular oxidative stress.Arterioscler Thromb Vasc Biol. 2000; 20:645–651.LinkGoogle Scholar
    • 333. Schmidt AM, Hori O, Chen JX, Li JF, Crandall J, Zhang J, Cao R, Yan SD, Brett J, Stern D.Advanced glycation endproducts interacting with their endothelial receptor induce expression of vascular cell adhesion molecule-1 (VCAM-1) in cultured human endothelial cells and in mice. A potential mechanism for the accelerated vasculopathy of diabetes.J Clin Invest. 1995; 96:1395–1403. doi: 10.1172/JCI118175.CrossrefMedlineGoogle Scholar
    • 334. Bouloumie A, Marumo T, Lafontan M, Busse R.Leptin induces oxidative stress in human endothelial cells.FASEB J. 1999; 13:1231–1238.CrossrefMedlineGoogle Scholar
    • 335. Ouedraogo R, Gong Y, Berzins B, Wu X, Mahadev K, Hough K, Chan L, Goldstein BJ, Scalia R.Adiponectin deficiency increases leukocyte-endothelium interactions via upregulation of endothelial cell adhesion molecules in vivo.J Clin Invest. 2007; 117:1718–1726. doi: 10.1172/JCI29623.CrossrefMedlineGoogle Scholar
    • 336. De Caterina R, Libby P, Peng HB, Thannickal VJ, Rajavashisth TB, Gimbrone MA, Shin WS, Liao JK.Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines.J Clin Invest. 1995; 96:60–68. doi: 10.1172/JCI118074.CrossrefMedlineGoogle Scholar
    • 337. Spiecker M, Peng HB, Liao JK.Inhibition of endothelial vascular cell adhesion molecule-1 expression by nitric oxide involves the induction and nuclear translocation of IkappaBalpha.J Biol Chem. 1997; 272:30969–30974.CrossrefMedlineGoogle Scholar
    • 338. Niu XF, Smith CW, Kubes P.Intracellular oxidative stress induced by nitric oxide synthesis inhibition increases endothelial cell adhesion to neutrophils.Circ Res. 1994; 74:1133–1140.LinkGoogle Scholar
    • 339. Kubes P, Suzuki M, Granger DN.Nitric oxide: an endogenous modulator of leukocyte adhesion.Proc Natl Acad Sci U S A. 1991; 88:4651–4655.CrossrefMedlineGoogle Scholar
    • 340. Lehoux S.Redox signalling in vascular responses to shear and stretch.Cardiovasc Res. 2006; 71:269–279. doi: 10.1016/j.cardiores.2006.05.008.CrossrefMedlineGoogle Scholar
    • 341. Chiu JJ, Wung BS, Shyy JY, Hsieh HJ, Wang DL.Reactive oxygen species are involved in shear stress-induced intercellular adhesion molecule-1 expression in endothelial cells.Arterioscler Thromb Vasc Biol. 1997; 17:3570–3577.LinkGoogle Scholar
    • 342. Shuvaev VV, Han J, Tliba S, Arguiri E, Christofidou-Solomidou M, Ramirez SH, Dykstra H, Persidsky Y, Atochin DN, Huang PL, Muzykantov VR.Anti-inflammatory effect of targeted delivery of SOD to endothelium: mechanism, synergism with NO donors and protective effects in vitro and in vivo.PLoS One. 2013; 8:e77002. doi: 10.1371/journal.pone.0077002.CrossrefMedlineGoogle Scholar
    • 343. Kisucka J, Chauhan AK, Patten IS, Yesilaltay A, Neumann C, Van Etten RA, Krieger M, Wagner DD.Peroxiredoxin1 prevents excessive endothelial activation and early atherosclerosis.Circ Res. 2008; 103:598–605. doi: 10.1161/CIRCRESAHA.108.174870.LinkGoogle Scholar
    • 344. Sharma A, Yuen D, Huet O, Pickering R, Stefanovic N, Bernatchez P, de Haan JB.Lack of glutathione peroxidase-1 facilitates a pro-inflammatory and activated vascular endothelium.Vascul Pharmacol. 2016; 79:32–42. doi: 10.1016/j.vph.2015.11.001.CrossrefMedlineGoogle Scholar
    • 345. Moore KJ, Sheedy FJ, Fisher EA.Macrophages in atherosclerosis: a dynamic balance.Nat Rev Immunol. 2013; 13:709–721. doi: 10.1038/nri3520.CrossrefMedlineGoogle Scholar
    • 346. Kunjathoor VV, Febbraio M, Podrez EA, Moore KJ, Andersson L, Koehn S, Rhee JS, Silverstein R, Hoff HF, Freeman MW.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.J Biol Chem. 2002; 277:49982–49988. doi: 10.1074/jbc.M209649200.CrossrefMedlineGoogle Scholar
    • 347. Duewell P, Kono H, Rayner KJ, et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals.Nature. 2010; 464:1357–1361. doi: 10.1038/nature08938.CrossrefMedlineGoogle Scholar
    • 348. Tabas I.Macrophage death and defective inflammation resolution in atherosclerosis.Nat Rev Immunol. 2010; 10:36–46. doi: 10.1038/nri2675.CrossrefMedlineGoogle Scholar
    • 349. Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C.Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis.JAMA. 2007; 297:842–857. doi: 10.1001/jama.297.8.842.CrossrefMedlineGoogle Scholar
    • 350. Schmidt HH, Stocker R, Vollbracht C, Paulsen G, Riley D, Daiber A, Cuadrado A.Antioxidants in Translational Medicine.Antioxid Redox Signal. 2015; 23:1130–1143. doi: 10.1089/ars.2015.6393.CrossrefMedlineGoogle Scholar
    • 351. Bast A, Haenen GR.Ten misconceptions about antioxidants.Trends Pharmacol Sci. 2013; 34:430–436. doi: 10.1016/j.tips.2013.05.010.CrossrefMedlineGoogle Scholar
    • 352. Münzel T, Gori T, Bruno RM, Taddei S.Is oxidative stress a therapeutic target in cardiovascular disease?Eur Heart J. 2010; 31:2741–2748. doi: 10.1093/eurheartj/ehq396.CrossrefMedlineGoogle Scholar
    • 353. Altenhöfer S, Radermacher KA, Kleikers PW, Wingler K, Schmidt HH.Evolution of NADPH Oxidase Inhibitors: Selectivity and Mechanisms for Target Engagement.Antioxid Redox Signal. 2015; 23:406–427. doi: 10.1089/ars.2013.5814.CrossrefMedlineGoogle Scholar
    • 354. Apostolova N, Victor VM.Molecular strategies for targeting antioxidants to mitochondria: therapeutic implications.Antioxid Redox Signal. 2015; 22:686–729. doi: 10.1089/ars.2014.5952.CrossrefMedlineGoogle Scholar
    • 355. Li H, Forstermann U.Pharmacological prevention of eNOS uncoupling.Curr Pharm Des. 2014; 20:3595–3606.CrossrefMedlineGoogle Scholar

    eLetters(0)

    eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

    Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.