142
Views
10
CrossRef citations to date
0
Altmetric
Review

Current status of NADPH oxidase research in cardiovascular pharmacology

, , , , &
Pages 401-428 | Published online: 25 Jul 2013

Abstract

The implications of reactive oxygen species in cardiovascular disease have been known for some decades. Rationally, therapeutic antioxidant strategies combating oxidative stress have been developed, but the results of clinical trials have not been as good as expected. Therefore, to move forward in the design of new therapeutic strategies for cardiovascular disease based on prevention of production of reactive oxygen species, steps must be taken on two fronts, ie, comprehension of reduction-oxidation signaling pathways and the pathophysiologic roles of reactive oxygen species, and development of new, less toxic, and more selective nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors, to clarify both the role of each NADPH oxidase isoform and their utility in clinical practice. In this review, we analyze the value of NADPH oxidase as a therapeutic target for cardiovascular disease and the old and new pharmacologic agents or strategies to prevent NADPH oxidase activity. Some inhibitors and different direct or indirect approaches are available. Regarding direct NADPH oxidase inhibition, the specificity of NADPH oxidase is the focus of current investigations, whereas the chemical structure-activity relationship studies of known inhibitors have provided pharmacophore models with which to search for new molecules. From a general point of view, small-molecule inhibitors are preferred because of their hydrosolubility and oral bioavailability. However, other possibilities are not closed, with peptide inhibitors or monoclonal antibodies against NADPH oxidase isoforms continuing to be under investigation as well as the ongoing search for naturally occurring compounds. Likewise, some different approaches include inhibition of assembly of the NADPH oxidase complex, subcellular translocation, post-transductional modifications, calcium entry/release, electron transfer, and genetic expression. High-throughput screens for any of these activities could provide new inhibitors. All this knowledge and the research presently underway will likely result in development of new drugs for inhibition of NADPH oxidase and application of therapeutic approaches based on their action, for the treatment of cardiovascular disease in the next few years.

Introduction

The implications of reactive oxygen species (ROS) in cardiovascular disease have been known for some decades. Rationally, therapeutic antioxidant strategies combating oxidative stress have been developed, but the results of clinical trials have not been as good as expected.Citation1 There is no convincing explanation for that, but we know now that ROS not only have pathologic effects but also, in the appropriate subcellular compartments and under proper antioxidant control, participate in signaling pathways that are crucial for cellular functioning. Therefore, blocking production of ROS should be selective and appropriate to have therapeutic activity. Pathways sensitive to oxidation/reduction are known by the generic name of “redox signaling” pathways. However, the participation of different ROS, the existence of different sources of ROS in the same cell, and the complex relationship between different signaling routes draw a complex picture and we do not have the necessary global view for this kind of cellular signaling. Our knowledge of the way in which ROS participate in pathophysiologic processes is also limited, in part by the absence of appropriately sensitive and selective probes for ROS and in part by having nonspecific pharmacologic tools for inhibition of production of ROS. This explains why we do not have well defined therapeutic targets for preventing the action of ROS in cardiovascular disease. In this sense, evidence has been accumulating regarding the important role of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in generation of ROS in the cardiovascular system. This membrane enzyme complex is defined mainly by its catalytic subunit, which has seven different isoforms. The characteristics and roles of this enzyme make it a suitable candidate for a good therapeutic target. However, further investigations are needed into the different roles of the NADPH oxidase isoforms and their participation in signaling pathways, in addition to the requirement for developing isoform-specific inhibitors. Therefore, to move forward in the design of new therapeutic strategies for cardiovascular disease based on prevention of ROS production, steps must be taken on both fronts, ie, comprehension of reduction-oxidation (redox) signaling pathways and the pathophysiologic roles of ROS, and the development of new, less toxic and more selective NADPH oxidase inhibitors, allowing clarification of the role of each NADPH oxidase isoform and its potential use in clinical practice. In this review, we assess the value of NADPH oxidase as a therapeutic target in cardiovascular disease as well as old and new pharmacologic agents and strategies to prevent the activity of NADPH oxidase.

Targeting NADPH oxidase

Oxidative stress: harmful or key signaling pathways

The pathologic effects of ROS in the cardiovascular system result simultaneously from their direct actions modifying vascular cell functions and from their ability to scavenge and remove several beneficial vasoprotective compounds, such as nitric oxide. ROS have direct effects on vascular cells, including endothelial cells, vascular smooth muscle cells, fibroblasts, and even resident macrophages, which are also important in the early stages of cardiovascular disease.Citation2 This includes the direct vasoregulatory effects of individual ROS, while others are regulated via toxic oxidation of proteins, lipids, and DNA.Citation3 Overproduction of ROS activates pro-oxidative genes, such as hypoxia inducible factor-1α, leading to angiogenesis and cell proliferation,Citation4 as well as a number of proinflammatory genes.

The interaction between nitric oxide and superoxide anions occurs so rapidly that it is impossible for the former molecule to have any biological effects. Therefore, many studies in cell cultures, animal models, and human vessels have shown that oxidative stress, in particular production of superoxide anions, is the single most important mechanism for endothelial dysfunction,Citation5 a situation wherein endothelial cells are unable to provide vasoprotective and functional agents for the vessel wall ().

Figure 1 Formation of reactive oxygen species (ROS) in vascular cells. The reduction of oxygen (O2) by one electron leads to the formation of superoxide anion (O2•−), which can be either dismutated to hydrogen peroxide (H2O2) spontaneously or in a reaction catalyzed by superoxide dismutase (SOD). Nitric oxide (NO•) is produced by endothelial nitric oxide synthase (eNOS) from L-arginine (L-Arg) and tetrahydrobiopterin. O2•− and NO• react spontaneously with each other to form peroxynitrite (ONOO). H2O2 can also be generated directly from oxygen by some vascular oxidases, such as xanthine oxidase (XO), glucose oxidase (GO) and NOX4-containing NADPH-oxidases (NOX4). H2O2 can be scavenged by catalase (CAT) or glutathione peroxidase (GPx) to form water and oxygen or can undergo non-enzymatic reactions to generate the hydroxyl radical (OH•) in the metal-catalyzed Haber-Weiss or Fenton reaction. OH• may be protonated to the hydroperoxyl radical. Ferrous-containing enzymes, such as myeloperoxidase (MPO) are activated by H2O2 to form a highly reactive radical that can oxidize NO• to nitrogen dioxide anion (NO2) and react with NO2 to form nitrogen dioxide radical (NO2•). NO2• can, in turn, participate in nitrating events, such as the formation of nitrotyrosines (NO2-Tyr). Alternatively, MPO can use H2O2 to form hypochlorous acid (HOCl). Singlet oxygen (1O2) is formed upon the reaction of HOCl with H2O2.

Abbreviations: 1e, one electron; 2e, two electrons; BH4, tetrahydrobiopterin; Cl, chloride anion; Fe2+, ferrous iron; H+, hydrogen cation; Tyr, tyrosine.
Figure 1 Formation of reactive oxygen species (ROS) in vascular cells. The reduction of oxygen (O2) by one electron leads to the formation of superoxide anion (O2•−), which can be either dismutated to hydrogen peroxide (H2O2) spontaneously or in a reaction catalyzed by superoxide dismutase (SOD). Nitric oxide (NO•) is produced by endothelial nitric oxide synthase (eNOS) from L-arginine (L-Arg) and tetrahydrobiopterin. O2•− and NO• react spontaneously with each other to form peroxynitrite (ONOO−). H2O2 can also be generated directly from oxygen by some vascular oxidases, such as xanthine oxidase (XO), glucose oxidase (GO) and NOX4-containing NADPH-oxidases (NOX4). H2O2 can be scavenged by catalase (CAT) or glutathione peroxidase (GPx) to form water and oxygen or can undergo non-enzymatic reactions to generate the hydroxyl radical (OH•) in the metal-catalyzed Haber-Weiss or Fenton reaction. OH• may be protonated to the hydroperoxyl radical. Ferrous-containing enzymes, such as myeloperoxidase (MPO) are activated by H2O2 to form a highly reactive radical that can oxidize NO• to nitrogen dioxide anion (NO2−) and react with NO2− to form nitrogen dioxide radical (NO2•). NO2• can, in turn, participate in nitrating events, such as the formation of nitrotyrosines (NO2-Tyr). Alternatively, MPO can use H2O2 to form hypochlorous acid (HOCl). Singlet oxygen (1O2) is formed upon the reaction of HOCl with H2O2.

On the other hand, ROS also have a role in key signaling pathways that are important for proper cell functioning. Participation of ROS has been described in a number of processes, including: regulation of gene expression by modification of the activity of transcription factorsCitation6Citation8 controlling the expression of adhesion molecules in the endotheliumCitation9,Citation10 or proteins implicated in cell proliferation and migration,Citation11Citation14 regulation of cytoskeletal activity,Citation15 and control of intracellular concentrations of second messengers like calcium.Citation16 Further, the implication of each type of ROS in modulation of responses seems to vary according to the region and type of vascular cell. Therefore, the final effects of each reactive molecule can vary depending on the region of tissue involved.Citation17 This complexity may partially explain why direct scavenging of ROS by antioxidant vitamins did not demonstrate clinical benefit in the majority of large trials.Citation18 Therefore, antioxidant strategies must be carefully designed based on a strong understanding of how ROS are involved in each pathophysiologic process.

Central role of NADPH oxidases in generation of oxidative stress

The main sources of ROS include redox enzymes such as NADPH oxidase, xanthine oxidase, lipooxygenase, and cyclooxygenase. It is important to note that these sources are continuously interacting with each other. NADPH oxidases are a family of multisubunit enzyme complexes that are unique in being the only enzymes that have been identified with the primary function of generating superoxide and/or hydrogen peroxide. ROS are produced as intermediates in redox reactions, leading from O2 to H2O. The major mechanism for generation of ROS begins with the reduction of O2, by the addition of one electron to generate a superoxide anion (O2•−), considered the primary ROS, and this is the reaction catalyzed by NADPH oxidases.

O2•− interacts with other molecules to produce secondary ROS, directly or through enzyme-catalyzed or meta-catalyzed reactions.Citation19 Reduction of O2•− leads to formation of H2O2, which is further converted to secondary metabolites, such as highly reactive hydroxyl HO. Although the favored reaction is generation of H2O2, O2•− also reacts with nitric oxide (NO) to form peroxinitrite (ONOO), with transition metals, such as iron found in iron/sulfur center-containing proteins, or it may be protonated to the hydroperoxyl radical (H2O, ). H2O is particularly important in lipid peroxidation and atherogenesis.

Seven isoforms of NADPH oxidase have been described in mammals. Each of these isoforms comprises a core catalytic subunit, ie, the so-called NADPH oxidase (NOX) and dual oxidase (DUOX) subunits, and up to five regulatory subunits. These regulatory subunits have important roles in: maturation and expression of the NOX and DUOX subunits in biological membranes, (ie, p22phox, DUOX activator 1, and DUOX activator 2), in enzyme activation (p67phox and NOX activator 1), and in spatial organization of the various components of the enzyme complex (p47phox, NOX organizer 1, and p40phox).Citation20 Some NADPH oxidase isoforms also rely on the small guanine triphosphate hydrolases (GTPases), ie, RAC1 and RAC2, for their activation. Each NADPH oxidase isoform is defined by the nature of its catalytic subunit (NOX1–NOX5, DUOX1, or DUOX2), which determines its suit of regulatory subunits ().

Figure 2 Subunit composition of the NADPH oxidase isoforms. The catalytic core subunits of the complex (NOX1–5 and DUOX1–2) are shown in grey and their stabilization partners (p22phox and DUOX activator 1 and 2) are shown in magenta. NOX4 is the only isoform that produces hydrogen peroxide instead of superoxide anion. The regulatory cytosolic subunits of each isoform are shown in each case: p40phox, p47phox, p67phox, NOX organizer 1 (NOXO1), NOX activator 1 (NOXA1), small GTPase (Rac), polymerase delta-interacting protein 2 (POLDIP2), a p47phox analog tyrosine kinase substrate with 4/5 SH3 domains (TKS4/5), EF hand motifs, (calcium-binding motifs composed of two helixes (E and F) joined by a loop), calmodulin (CaM) and heat shock protein 90 (HSP90). On DUXO1 and DUOX2 a putative additional amino-terminal transmembrane domain and extracellular peroxidase-like region (PLD) are shown.

Abbreviations: C-ter, carboxy-terminal; FAD, flavin adenine dinucleotide; GTPase, guanine triphosphate hydrolase; NADPH, nicotinamide adenine dinucleotide phosphate; N-ter, amino-terminal; O2, oxygen; H2O2, hydrogen peroxide; O2−, superoxide; SH3, Src-homology region 3.
Figure 2 Subunit composition of the NADPH oxidase isoforms. The catalytic core subunits of the complex (NOX1–5 and DUOX1–2) are shown in grey and their stabilization partners (p22phox and DUOX activator 1 and 2) are shown in magenta. NOX4 is the only isoform that produces hydrogen peroxide instead of superoxide anion. The regulatory cytosolic subunits of each isoform are shown in each case: p40phox, p47phox, p67phox, NOX organizer 1 (NOXO1), NOX activator 1 (NOXA1), small GTPase (Rac), polymerase delta-interacting protein 2 (POLDIP2), a p47phox analog tyrosine kinase substrate with 4/5 SH3 domains (TKS4/5), EF hand motifs, (calcium-binding motifs composed of two helixes (E and F) joined by a loop), calmodulin (CaM) and heat shock protein 90 (HSP90). On DUXO1 and DUOX2 a putative additional amino-terminal transmembrane domain and extracellular peroxidase-like region (PLD) are shown.

Although all NOX have the same function in generating ROS, mechanisms of activation, subunit requirements, and intracellular distributions vary between isoforms. Unlike phagocytic NADPH oxidases, which are activated only upon stimulation and generate O2•− in a burst-like manner extracellularly, nonphagocytic NADPH oxidases are constitutively active, produce O2•− intracellularly in a slow and sustained fashion, and act as intracellular signaling molecules, influencing not only transcription factors but also other molecules involved in inflammation, cell growth, and contraction, such as mitogen-activated protein kinases, tyrosine kinases, and protein phosphatases.Citation21,Citation22

ROS produced by NADPH oxidases have been shown to modify virtually all other possible sources of ROS (). Changes in the structure of several oxidases caused by the overproduction of ROS from NADPH oxidases lead to their dysfunction and to a further increase in generation of ROS, forming a vicious cycle of oxidative stress.Citation23 An example is endothelial nitric oxide synthase, which primarily produces nitric oxide, but generates ROS rather than nitric oxide when it is uncoupled under pathologic conditions. Uncoupling of nitric oxide synthase occurs by oxidation of its cofactor tetrahydrobiopterin, and this oxidation depends on peroxynitrite, which is formed by reaction of nitric oxide with superoxide anions produced by NADPH oxidases. Similarly, production of superoxide anions by NADPH oxidase induces mitochondrial oxidative damage via structural changes to the inner mitochondrial membrane and disturbs flow in the electron transport chain, which enhances ROS production.Citation24,Citation25

Validating the role of NADPH oxidase in cardiovascular health and disease

NADPH oxidases are found in virtually every tissue and are the major source of superoxide anions observed in the vasculature.Citation26 Multiple NOX oxidase isoforms are constitutively expressed in each of the predominant cell types of the vascular wall. Endothelial cells express NOX1, NOX2, NOX4, and NOX5; vascular smooth muscle cells express NOX1, NOX4, and NOX5; and adventitial fibroblasts express NOX2 and NOX4.Citation19,Citation27 In parallel with the two-way actions of ROS, NOX proteins have both beneficial and harmful effects. They are important signaling molecules that regulate vascular tone, expression, proliferation, migration, and differentiation.Citation28 On the other hand, cardiovascular risk factors and vascular diseases increase ROS and contribute to atherosclerosis, vascular dysfunction, hypertension, vascular hypertrophy, and thrombosis.Citation26,Citation28 To inhibit NADPH oxidase successfully, it is important to understand these points ().

Figure 3 NADPH oxidase and cardiovascular diseases. Risk factors for cardiovascular diseases cause vascular injury and the release of cytokines and factors that activate NADPH oxidases. ROS generation (superoxide anion (O2•−) and hydrogen peroxide (H2O2) activate redox signaling, which trigger cellular responses or cause oxidative stress, which have the potential to originate oxidative macromolecular damage, decrease nitric oxide (NO•) bioavailability and increase oxidation of low-density lipoprotein (ox-LDL). All together contribute to vascular inflammation, dysfunction and platelet or leukocyte adhesion, which lead to multiple outcomes of cardiovascular disease.

Abbreviations: ECM, extra-cellular matrix; VSMC, vascular smooth muscle cells; NADPH, nicotinamide adenine dinucleotide phosphate; ↓, decrease; ROS, reactive oxygen species; LDL, low-density lipoprotein.
Figure 3 NADPH oxidase and cardiovascular diseases. Risk factors for cardiovascular diseases cause vascular injury and the release of cytokines and factors that activate NADPH oxidases. ROS generation (superoxide anion (O2•−) and hydrogen peroxide (H2O2) activate redox signaling, which trigger cellular responses or cause oxidative stress, which have the potential to originate oxidative macromolecular damage, decrease nitric oxide (NO•) bioavailability and increase oxidation of low-density lipoprotein (ox-LDL). All together contribute to vascular inflammation, dysfunction and platelet or leukocyte adhesion, which lead to multiple outcomes of cardiovascular disease.

ROS are involved in a large number of cardiovascular diseases and the causal mechanisms are complex. Validating the involvement of a protein in a biological process requires experimental pharmacologic inhibition or genetic deletion and expressional confirmation studies (for a review, see Altenhofer et al).Citation29 Vascular cells simultaneously express multiple NOX enzymes, and various cardiovascular diseases have been associated with changes in the expression of NOX1, NOX2, NOX4, and NOX5. In vascular smooth muscle cells from large arteries, NOX1 is required for migration, hypertrophy, and proliferation, NOX4 modulates differentiation, and NOX1 and NOX2 are implicated in hypertension.Citation19

Expression of the vascular NOX2 subunit is commonly increased by proinflammatory stimuli or in the presence of cardiovascular risk factors.Citation30 The endothelial injury and reduced nitric oxide signaling that occurs in the early stages of vascular disease is largely mediated by excessive levels of NOX2 oxidase-derived superoxide anion.Citation17,Citation30 Studies in transgenic and knockout animal models have shown the involvement of NOX1 and NOX2 and some of their regulatory subunits, such as p47phox, in the development of hypertension, atherosclerosis, and restenosis (for a review see Drummond et al).Citation27 Therefore, there is a strong rationale for therapeutically targeting NADPH oxidases in the arterial wall for the treatment of vascular diseases.

Vascular disease and atherosclerosis

The demonstration that expression of the NOX1 and/or NOXA1 subunit is elevated in atherosclerotic lesions of apolipoprotein E-deficient (ApoE−/−) mice and humans implies a role for the enzyme in this disease state.Citation31,Citation32 In support, there is evidence for a greater reduction in development of atherosclerotic lesions in the aortic arch of Nox1−/−/ApoE−/− double knockout mice compared with ApoE−/− single knockout mice.Citation33 NOX2 seems to participate in the redox signaling implicated in initiation and progression of atherosclerosis. Overexpression of NOX2 in ApoE−/− mice increased recruitment of macrophages, although did not alter progression of atherosclerosis.Citation34

The case of NOX4 remains controversial. NOX4 is highly expressed in endothelial cells, vascular smooth muscle cells, and adventitial fibroblasts, and owing to its lack of reliance on traditional cytosolic organizer and activator subunits, NOX4 activity was, until recently,Citation35 thought to be regulated primarily by changes in expression of its catalytic subunit. Hence, observations that levels of NOX4 in vascular cells are downregulated,Citation36,Citation37 unchanged,Citation38 or upregulated,Citation39,Citation40 depending on the pathologic stimulus under investigation, provide no clear indication of a homeostatic versus pathophysiologic function of the catalytic subunit. Consistent with this, several reports suggest a role for NOX4 in promoting endothelial cell survival, proliferation, and migration,Citation41Citation44 whereas other studies indicate that NOX4 mediates effects such as endoplasmic reticulum stress, oxidative DNA damage, and apoptotic and necrotic cell death of endothelial cells.Citation40,Citation45Citation47 Likewise, in vascular smooth muscle cells, NOX4 was initially described as a key regulator of cellular differentiation and quiescence,Citation48,Citation49 which is suggestive of a homeostatic function, but was subsequently shown to contribute to vascular smooth muscle cell proliferation, migration, and hypertrophy under certain conditions,Citation50,Citation51 all of which are important in arterial remodeling and atherogenesis.

Finally, even the fact that NOX4 generates hydrogen peroxide, which is capable of activating proinflammatory signaling cascades,Citation52 rather than superoxide,Citation53Citation55 raises questions as to whether NOX4 is likely to have a protective or detrimental role in vascular disease.

Indeed, in human microvascular endothelial cell lines and primary aortic vascular smooth cells, NOX5 activity was augmented by relevant proatherogenic stimuli, including thrombin, platelet-derived growth factor, angiotensin II, and endothelin 1; in each instance, NOX5 activity was suppressed following depletion of intracellular calcium.Citation56,Citation57 Endothelial cells and coronary arteries from patients with coronary artery disease showed greater NOX5 subunit expression and calcium-dependent ROS production compared with those from healthy subjects.Citation58 Importantly, calcium-dependent but not calcium-independent ROS production was reduced by small interfering RNA against NOX5, indicating that calcium-dependent production of ROS is due to NOX5 activity.Citation58 Although these data suggest a strong association between NOX5 activity and coronary artery disease in humans, evidence for a cause-effect relationship is not yet proven, and additional work is needed to characterize its physiologic and pathologic roles in the arterial wall.

Recent research has shown that NOX enzymes are particularly relevant in ischemia-reperfusion injury, a situation where a tissue is deprived of oxygen for a long period of time, followed by an abrupt increase in oxygen concentrations, eg, after thrombolysis in stroke and myocardial infarction or after surgical intervention. NOX are determinants of endothelial function after ischemia-reperfusion injury.Citation59

On the other hand, the situation is slightly different in the vascular system of the brain. All of the major cell types present in brain tissue, including neurons, astrocytes, microglia, and vascular endothelial cells, constitutively express NOX1, NOX2, and NOX4,Citation60 which presumably refects the physiologic roles of NOX-derived ROS in brain function. However, cerebral arteries differ from many noncerebral arteries in that they have higher levels of NADPH oxidase activity,Citation61,Citation62 and ROS more typically elicit marked vasorelaxant responses in cerebral vessels.Citation17 If NADPH oxidase-derived ROS help to maintain cerebral blood flow and neural function, then great caution will be needed in developing future therapeutic strategies involving inhibition of brain NADPH oxidases.

There is evidence from studies of global NOX2 knockout mice that elevated activity of NADPH oxidase isoforms normally contributes to worsened outcomes following stroke, at least in males,Citation63Citation67 and that the protective effects of apocynin occur via inhibition of NOX2.Citation63,Citation67,Citation68 There is evidence that NOX4-derived ROS mediate cerebral endothelial apoptosis induced by tumor necrosis factor (TNF)Citation40 and infarct damage after stroke.Citation69 However, there is conflicting evidence as to whether NOX1 activity might contribute to stroke damage.Citation70,Citation71

Heart failure

Even more than nitric oxide depletion, endothelial nitric oxide synthase uncoupling, and modulation of intracellular redox signaling pathways, ROS also affect cellular energetics in the myocardium, resulting in an energy deficit, cellular damage, and acceleration of cell death via apoptosis and necrosis.Citation72 Further, ROS modulate proliferation of fibroblasts, synthesis of collagen, and activation of matrix metalloproteinases, and their expression in the myocardium causes myocardial hypertrophy, fibrosis, and necrosis, which can lead to endothelial and myocardial dysfunction.Citation73,Citation74 At the subcellular level, ROS have been shown to be involved in the pathophysiology of pressure overload-induced left ventricular hypertrophy and heart failure.Citation75 ROS are implicated not only in the process of cellular hypertrophy and remodeling in the decompensated phase, but also in the development of compensated pressure overload left ventricular hypertrophy.Citation76

In NOX2y∕− (NOX2-deficient) mice in which pressure overload was induced by constriction of the ascending aorta, there was a large increase in messenger (m)RNA expression of p22phox and a small increase in mRNA expression of p47phox, whereas in wild-type mice there was a moderate increase in p22phox and no increase in p47phox. ROS levels in the myocardium were two times higher than in wild-type mice.Citation77 Bendall et alCitation78 and Byrne et alCitation79 reported contrasting roles for NOX isoforms in pressure overload-induced versus angiotensin II-induced myocardial hypertrophy using NOX2−∕− mice in which hypertrophy was induced by transaortic constriction or chronic angiotensin II infusion, respectively. All these findings indicate that the function of cardiac NADPH oxidases depends on the isoform and also on the specific pathologic process involved.

The NOX2 isoform and the subunits of the enzyme complex associated to it were identified as a major source of ROS in left ventricular hypertrophy induced by pressure overload, and contributed to pathophysiologic changes, such as activation of redox-sensitive kinases and progression of heart failure.Citation80

Studies of NOX4−∕− and cardiac-specific NOX4−∕− (cNOX4−∕−) mice demonstrated the important role of NOX4, although contrasting results have been observed. It was seen that attenuated left ventricular hypertrophy, interstitial fibrosis and apoptosis, and systolic function were improved in cNOX4−∕− mice compared with wild-type mice.Citation81 In NOX4−∕− mice and a cardiomyocyte-targeted NOX4 transgenic model, with the latter overexpressing NOX4 in the heart, NOX4 expression was increased in cardiomyocytes in response to pressure overload and after myocardial infarction. However, NOX4−∕− mice showed significantly more cardiac dilatation and contractile deterioration compared with wild-type mice, and NOX4 transgenic mice developed less hypertrophy and fibrosis compared with wild-type mice.Citation82 These data suggest that endogenous NOX4 has an important role in mediating cardiomyocyte hypertrophy, fibrosis, and apoptosis in response to pressure overload, although the mechanism of action remains undefined.

NOX2 is a relevant source of ROS in the pathogenesis of ischemic cardiomyopathy. It is unquestionable that NOX2 contributes to cardiac remodeling processes after myocardial infarction, because deletion of NOX2 significantly abrogates the effects.Citation83 Nevertheless, the infarct size, measured before and after remodeling, was comparable between NOX2y/ and wild-type mice,Citation83p47phox−∕− and heterozygous control mice,Citation84 and p47phox−∕− and wild-type mice.Citation85 It appears that the role of NOX2-derived ROS depends crucially on the basal level of ROS production; loss of NOX2-derived ROS under physiologic conditions where it plays a proangiogenic role is detrimental, whereas loss of NOX2 in pathophysiologic conditions, when overall production is increased, is beneficial.

Hypertension

There is abundant evidence that ROS contribute to the pathophysiology of hypertension, and many mechanisms have been implicated. As we have previously commented, increased production of superoxide anions leads to a reduction in bioavailability of the vasodilator, nitric oxide. Further, ROS have been implicated in proliferation and hypertrophy of vascular resistance.Citation11,Citation12,Citation86 There is genetic evidence from mice, rats, and humans to suggest a role of NOX in hypertension. In vascular smooth muscle cells, NOX1 is localized to the plasma membrane, caveolae, and endosomes, and its expression is induced by several substances, including vasoactive agonists, growth factors, proinflammatory cytokines, and advanced glycation end products.Citation87 Studies in animal models of cardiovascular disease, especially of angiotensin II-induced hypertension, implicate this NADPH oxidase isoform as an important contributor to the pathology of cardiovascular disease.

NOX1 expression in the vascular wall is elevated in several in vivo animal models of hypertension.Citation88Citation92 Accordingly, vascular superoxide production, endothelial dysfunction, and aortic hypertrophy were exacerbated in transgenic mice overexpressing NOX1 in vascular smooth muscle cells.Citation93,Citation94 These parameters were blunted in mice that were globally deficient in NOX1.Citation95,Citation96 However, NOX1 is associated with angiotensin II-dependent hypertension more than with noradrenaline-dependent hypertension.Citation97

Spontaneously hypertensive rats have a polymorphism in the promoter of the p22phox gene, a subunit of the NADPH oxidase complex, that leads to an overexpression of this subunit and a subsequent increase in ROS. Humans homozygous for a polymorphism in the gene encoding p22phox have reduced oxidative stress in the vascular system and probably also reduced blood pressure.Citation98,Citation99

It is likely that the type of experimental hypertension and the location of the blood vessel studied can significantly impact how NOX4 expression is regulated. In the absence of pathogenic stimuli, NOX4 knockout mice do not have an obvious phenotype and are normotensive.Citation28

NOX4 is strongly expressed in the media of small pulmonary arteries and is causally involved in development of pulmonary hypertension.Citation100 NOX4 is the major NADPH oxidase homologue expressed in human pulmonary artery smooth muscle cells,Citation51 and its expression at both the mRNA and protein levels is significantly increased in lungs from patients with idiopathic pulmonary arterial hypertension compared with healthy lungs,Citation100 suggesting a correlation between NOX4 and onset of pulmonary arterial hypertension.

Current status of pharmacologic research on NADPH oxidase

In the previous section, we highlighted the value of targeting NADPH oxidase activity for cardiovascular problems. From this starting point, intense research has been undertaken in this field to answer two important questions, ie, where to act and how to do it, meaning that we still do not know what is the best molecular target or the best inhibitor.

Points of targeting

NADPH oxidases are involved in complex mechanisms of action. Therefore, targeting these enzymes can be done at several different points of the pathways involved. It is also important to define the aim of the strategy of inhibition in terms of whether all known oxidases should be simultaneously inhibited or only specific NADPH oxidase isoforms. Equally, the subcellular location of NADPH oxidases could be critical for the expected effect of the inhibition, because local ROS production in different subcellular compartments has different pathophysiologic significance.

Moreover, some different mechanisms could be used to inhibit NADPH oxidase activity. Decreasing expression of the catalytic subunits or their regulatory subunits is one possibility. Activation of the enzyme complex can be also decreased by blocking translocation of the cytosolic subunits to the membrane or inhibiting activation of the regulatory subunits. A decrease in the signal transduction pathways upstream of NADPH oxidase activation is an indirect way to inhibit the activity of the enzyme. Finally, we consider direct inhibition of some or specific NADPH oxidase subunits. All together, they form the available “points of targeting” which should guide the final objective: NADPH oxidase inhibition. We will make some considerations about these points before reviewing the currently available inhibitory drugs or strategies.

NADPH isoforms

As we have seen, the evidence for the seven NOX isoforms is not equal. The main volume of research in all fields and particularly in the cardiovascular field includes NOX1, NOX2, and NOX4. Little is known about the involvement of NOX5 in cardiovascular disease and almost nothing is known about NOX3, DUOX1, and DUOX2. This may or may not reflect the real involvement of each isoform in the pathophysiologic process, and be an indication for future research. Therefore, the state of the art at the moment points to NOX2 in several cell types, and to a lesser extent NOX1 in vascular smooth muscle cells, as the NADPH oxidases that currently represent the most promising therapeutic targets for treating vascular diseases. However, NOX4 and NOX5 could emerge as validated drug targets from future research.

There is a strong rationale for therapeutically targeting NOX2 in the arterial wall for the treatment of vascular disease because this isoform can generate large amounts of ROS, and pharmacologic inhibition or genetic deletion reduces vascular oxidative stress in several disease models.Citation63,Citation67,Citation68,Citation101Citation105

Studies in animal models implicate NOX1 as an important contributor to the pathology of cardiovascular disease. Expression of NOX1 in the vascular wall is elevated in several in vivo animal models of hypertension.Citation88Citation92 In mice overexpressing NOX1 in vascular smooth muscle cells, angiotensin II-induced hypertension, endothelial dysfunction, and aortic hypertrophy were exacerbated.Citation93,Citation94 Finally, expression of NOX1 is elevated in atherosclerotic lesions from apolipoprotein E-deficient (ApoE−∕−) mice and humans.Citation31,Citation32 In addition, greater reduction in development of atherosclerotic lesions in the aorta of the Nox1−∕−/ApoE−∕− double knockout mice has been observed.Citation33

There is no clear indication of a homeostatic versus pathophysiologic function of NOX4 in vascular cells. In fact, NOX4 has been associated with beneficial effectsCitation41Citation44 and detrimental effects.Citation40,Citation45Citation47 Therefore, this NADPH oxidase isoform could participate in positive arterial remodeling or in atherosclerosis, depending on the physiologic conditions. NOX5 seems to function as a stand-alone protein and has a calcium-binding domain, which allows NOX5 activity to be regulated by increases in the cytosolic concentration of calcium.Citation106 NOX5 activity in vascular cells responds to proatherogenic stimuli and is suppressed by depletion of intracellular calcium.Citation37,Citation57 NOX5 expression is greater in endothelial cells and coronary arteries from patients with coronary artery disease compared with those from healthy subjects.Citation58 Although these data suggest a strong association between NOX5 activity and coronary artery disease, evidence for a cause-effect relationship is not yet proven, and further research is needed to characterize this association.

Subcellular location of NADPH oxidases

Because ROS are diffusible and short-lived, subcellular localization of NOX isoforms is crucial in their activation, in the site of ROS production, and in the coordination of redox-signaling events. NOX isoforms are now recognized to have specific subcellular localizations defined by their unique localization motifs that bind to adaptor proteins and target their cargo to the correct membrane. NADPH oxidases appear to be activated within discrete subcellular compartments, including caveolae/lipid rafts, focal adhesions, cell-cell contacts, lamellipodial leading edges, endosomes, and the nucleus.Citation107 This facilitates spatially confined ROS production with redox-sensitive targets in proximity, which may allow ROS to activate specific redox-signaling events. However, the mechanisms by which NOX isoforms traffic to the correct subcellular location in order to accomplish appropriate signaling remains poorly defined.

In vascular smooth muscle cells, stimulation of angiotensin II promotes angiotensin type 1 receptor (AT1) trafficking into caveolin-1-enriched membrane fractions where NOX1 is found.Citation108 This causes localized production of ROS and ROS-cSrc-dependent transactivation of the epidermal growth factor receptor (EGFR) and its egress from caveolae. Tyrosine-phosphorylated EGFR and caveolin-1 subsequently appear at focal adhesions where NOX4 and paxillin localize, thereby forming redox signaling platforms.Citation109,Citation110 These events are essential for activation of specific redox signaling pathways involved in vascular smooth muscle cell hypertrophy.Citation111 For instance, treatment with TNF-α causes activation of NOX1 and generation of ROS within endosomes with subsequent nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) activation.Citation112,Citation113 However, treatment with thrombin causes nonendosomal NOX1-dependent production of ROS, which, as in the case of stimulation with angiotensin II, leads to transactivation of EGFR.Citation112

In endothelial cells, stimulation with TNF-α, Fas ligand, and shear stress induces recruitment of NOX2, p47phox, and Rac1 into lipid rafts,Citation114 thereby promoting raft-localized activation of NADPH oxidase and production of ROS and endothelium-derived nitric oxide synthase (NOS) within raft domains. This formation of redox signaling platforms in caveolae/lipid rafts in which death receptors couple to NADPH oxidase contributes to a decrease in bioavailability of nitric oxide and production of peroxynitrite,Citation115 which uncouples NOS to produce more superoxide anions and finally contributes to endothelial dysfunction.Citation116

Integrin-mediated cell adhesion (focal adhesions and cell-matrix adhesion sites) serve as organizing centers for regulatory and structural proteins to facilitate rapid and precise control of cell functions like gene transcription for cell proliferation, survival, motility, and cytoskeletal remodeling.Citation117Citation121 A synergistic action of integrins and receptor tyrosine kinases occurs during redox signaling.Citation122 Growth factor activates NADPH oxidase, whereas activation of integrin stimulates the mitochondria and 5-lipooxygenase to generate localized production of ROS at cell-matrix adhesions. These ROS induce oxidative inactivation of protein tyrosine phosphatases,Citation123Citation126 which negatively regulate receptor tyrosine kinase and focal adhesion kinase, thereby promoting downstream redox signaling events leading to proliferation, survival, and gene expression.Citation122

The molecule primarily responsible for cell-cell contacts of endothelial cells is vascular endothelial-cadherin.Citation127 Tyrosine phosphorylation of the vascular endothelial-cadherin complex is required for disruption of the cell-cell junction,Citation128Citation130 and is mediated via cSrc, which is dependent on ROS.Citation131,Citation132 Moreover, NADPH oxidase-derived ROS are involved in vascular endothelial-cadherin/β-catenin phosphorylation and disruption of cell-cell contacts.Citation133 Stimulation of vascular endothelial growth factor promotes loss of cell-cell contacts, thereby promoting migration and proliferation of endothelial cells involved in angiogenesis as well as endothelial permeability via a ROS-dependent mechanism.Citation134,Citation135 Polymorphonuclear leukocyte adhesion to endothelial cells stimulates production of ROS, which also promote loss of cell-cell adhesions through phosphorylation of vascular endothelial-cadherin.Citation136 These responses are involved in increasing transendothelial migration of polymorphonuclear leukocytes and endothelial permeability, which contributes to inflammatory diseases, such as atherosclerosis and diabetes. Thus, NADPH oxidase-derived ROS produced near the adherent junction may activate redox signaling that disrupts cell-cell contacts, promoting angiogenesis, vascular inflammation, and permeability.Citation131,Citation137

Migration of endothelial cells is a key event for tissue repair in response to injury, angiogenesis, and wound healing. In these cells, Rac1-dependent and NOX2-dependent NADPH oxidase plays an important role in cell migration.Citation12,Citation138Citation140 NOX2 and its regulatory subunits are targeted to the focal complexes or membrane ruffles in lamellipodia.Citation141Citation143 Oxidants derived from activation of NADPH oxidase and lamellipodial dynamics are likely to be spatially and functionally coupled at the leading edge to promote directional cell migration.Citation143,Citation144 Different functional and scaffold proteins, such as WAVE1,Citation141,Citation145 IQGAP1,Citation146,Citation147 and protein tyrosine phosphatase-PEST,Citation142 have been associated with NADPH oxidase complexes in leading edges of lamellipodia and focal contacts.

The NADPH oxidase complex is also formed in intracellular compartments. NOX2,Citation148 NOX4,Citation149 or NOX1, NOX2, and NOX4Citation44 have been found in the perinuclear compartments of different types of endothelial cells. NOX4 is critical for oxidative inactivation of protein tyrosine phosphatase 1B localized mainly to the cytosolic face of the endoplasmic reticulum, thereby enhancing activation of receptor tyrosine kinase in this intracellular compartment.Citation150 Stimulation of interleukin-1 β promotes endocytosis of its receptor, which is required for NOX2-dependent ROS production in early endosomes and subsequent redox-dependent activation of the transcription factor, NF-κB.151 NOX1 and NOX2 are also involved in endosomal production of ROS after hypoxia-reoxygenation injury required for inhibitory subunit of NF-kappa B alpha (IκBα) tyrosine phosphorylation.Citation152 NOX1 can also participate in the ROS production required for NF-κB activation induced by TNF-α.Citation113 Taken together, these studies suggest that NADPH oxidase-dependent ROS production in endosomes is involved in the proinflammatory immune response.

Many transcription factors are redox-sensitive, including activator protein 1 (AP-1), NF-κB, Nrf2, p53, and the glucocorticoid receptor,Citation153Citation158 which require an oxidative signal in the cytoplasm to initiate signaling for activation, whereas oxidative stress in the nuclear compartment blocks the process.Citation159 NOX4 preferentially localizes to the nucleus in human endothelial cells,Citation160 so local NOX4-dependent ROS production in the nucleus may contribute to regulation of the redox-dependent transcription factor and gene expression involved in cell growth, differentiation, senescence, and apoptosis.

A better understanding of compartmentalization of redox signaling will provide further insights into temporally and spatially organized ROS-dependent signaling systems and the relevance of antioxidant therapy, with targeting to specific intracellular microdomains for the treatment of various oxidant stress-dependent diseases.

Expression of NADPH oxidase

NOX isoforms are differentially expressed during development of atherosclerosis. Expression of NOX4 appears to increase the stability of atherosclerotic plaques, while at earlier stages of plaque development NOX2 seems to predominate, contributing to progression of atherosclerosis.Citation161 This led to the conclusion that NOX4 might be involved in the maintenance of the differentiated vascular smooth muscle cell phenotype.Citation48 NOX5 in turn seems to be coexpressed at sites of human plaque instabilityCitation58 These aspects point to the complex nature of regulation of NADPH oxidases and their role in disease. It is also clear that we must be cautious about the pharmacologic targeting of these enzymes in humans.

The natural or pathophysiologic expression of NADPH oxidases in the vasculature can also be modified by selected drugs, which clearly introduces a new point of targeting. Docosahexaenoic acid has been demonstrated to decrease NOX4 expression and activity in human aortic endothelial cells, an effect associated with reduced production of ROS.Citation162 In the same way, resveratrol, a natural occurring antioxidant found in grapes and wine, downregulates NOX4 in a concentration-dependent way in human endothelial cells, on the contrary, it upregulates the expression of two antioxidative enzymes, ie, superoxide dismutase and glutathione peroxidase 1.Citation163

Pathophysiologic upregulation of NADPH oxidase expression could be also selectively inhibited by some drugs. Sodium hydrosulfide and the H2S-donating derivative of sildenafil (ACS6) completely inhibited NOX2 expression induced by TNF-α in porcine endothelial cells via the adenylyl cyclase-protein kinase A pathway.Citation164 Expression of p47phox could also be inhibited, but it is not clear if this effect is due to hydrosulfide or to the sildenafil structure.Citation165 CPU0213, an endothelin receptor antagonist, reversed the upregulation of p22phox and p47phox (NADPH oxidase regulatory subunits) induced by administration of isoproterenol, a beta-adrenergic sympathomimetic, in rats.Citation166 In rats with hypertension induced by deoxycorticosterone acetate (DOCA) salt, dietary sesamin prevented the enhanced increase in NADPH oxidase activity and expression.Citation91 Many other natural products have been shown to reduce expression of NADPH oxidase in vascular tissues.Citation163,Citation167Citation173 One of the main transcription factors implicated in TNF-α-induced or angiotensin II-induced upregulation of NOX isoforms and NADPH oxidase subunits is AP-1.Citation174 In contrast, enhanced expression of NOX1, NOX2, NOX4, and p22phox by nonselective, nonsteroidal anti-inflammatory drugs has been demonstrated in the aorta and heart in animal models, in isolated human endothelial cells, and in healthy volunteers.Citation175

This picture is further complicated by the fact that genetic variation of NADPH oxidase components, which is related to differential ROS production, is involved in atherosclerosis and vascular dysfunction. Genetic variation of the p22phox subunit encoding gene CYBA has been discussed in this context; however, all other subunits should be carefully examined also in the context of pharmacokinetics and immunogenetics.Citation23,Citation58,Citation161,Citation176 Although expression of p22phox is not affected, the C242T CYBA single nucleotide polymorphism is associated with altered NADPH oxidase activity in patients with cardiovascular disease.Citation98

Direct inhibition or modulation of regulatory pathways

Due to the complex relationship between NADPH oxidases and other proteins in the context of some signaling pathways and even the complex cross-talk between different signaling pathways, inhibition of NADPH oxidase activity can be done sometimes via regulation of a pathway rather than direct inhibition of the oxidase. This possibility has been considered in clinical practice because it has not been easy to obtain specific and selective inhibitors for each NADPH oxidase isoform. Therefore, at the moment, direct and indirect inhibition of NADPH oxidase activity should be considered as strategies with the same objective ().

Figure 4 Possible strategies for NADPH oxidase inhibition. Several steps that can be used to modulate NADPH oxidase activity. (1) NADPH oxidase subunits expression. (2) Signaling upstream of NADPH oxidase activation. (3) Association of cytosolic subunits and formation of the complete enzyme complex. (4) Subunit phosphorylation (P) and activation. (5) Modulation of cytosolic calcium concentration. (6) Transference of electrons through the enzyme complex.

Abbreviations: Ca2+, calcium ion; CaM, calmodulin; e, electron; FAD, flavin adenine dinucleotide; HSP90, heat shock protein 90; NADPH, nicotinamide adenine dinucleotide phosphate; O2−, superoxide anion; O2, oxygen.
Figure 4 Possible strategies for NADPH oxidase inhibition. Several steps that can be used to modulate NADPH oxidase activity. (1) NADPH oxidase subunits expression. (2) Signaling upstream of NADPH oxidase activation. (3) Association of cytosolic subunits and formation of the complete enzyme complex. (4) Subunit phosphorylation (P) and activation. (5) Modulation of cytosolic calcium concentration. (6) Transference of electrons through the enzyme complex.

Suitable inhibitors

Multiple lines of evidence for inhibition of NADPH oxidase are required to confirm the inhibitory effect. Therefore, several approaches can be used to investigate the inhibitory effect and to discriminate between compounds that act directly on the enzyme complex versus the activation process upstream of NADPH oxidase. For a summary of these approaches, see the review by Jaquet et al.Citation177 We discuss below the available NADPH oxidase inhibitors differentiated by these general mechanisms of action. Other authors have grouped the NADPH oxidase inhibitors by more precise steps of NADPH oxidase activation. In this regard, it is useful to consult the review of Streeter et al,Citation178 which included the following steps as possible targets: NOX expression, signal transduction upstream of NOX activation, trafficking of NOX to the appropriate subcellular compartment, assembly of NADPH oxidase complex, electron transfer from NADPH to oxygen, and oxidation of downstream targets.

Direct inhibitors: past and future

Several compounds have the ability to inhibit NADPH oxidases directly (). In many cases, their effects have been confirmed in vivo, but major problems with the inhibitors currently available include low specificity, associated toxicity, or their peptide character. The search for successful inhibitors continues as the prospect for use in clinical practice increases along with molecular knowledge about redox signaling (). Specific inhibitors are also essential to establish fully the role of NADPH oxidases and individual NOX isoforms in different pathologies.

Figure 5 Sites for direct NADPH oxidase inhibition. Some mechanisms of action have been reported for NADPH oxidase inhibitors. (1) Specific inhibition of the catalytic subunit (NOX). (2) Inhibition of the expression of any subunit or inhibition of the traffic for subcellular location. (3) Flavin (cofactor) inhibition or electron transfer inhibition. (4) Inhibition of correct subunits assembly. (5) Inhibition of subunit phosphorylation (P) and activation.

Abbreviations: FAD, flavin adenine dinucleotide; NADPH, reduced nicotinamide adenine dinucleotide phosphate; N-ter, amino terminal; O2−, superoxide anion.
Figure 5 Sites for direct NADPH oxidase inhibition. Some mechanisms of action have been reported for NADPH oxidase inhibitors. (1) Specific inhibition of the catalytic subunit (NOX). (2) Inhibition of the expression of any subunit or inhibition of the traffic for subcellular location. (3) Flavin (cofactor) inhibition or electron transfer inhibition. (4) Inhibition of correct subunits assembly. (5) Inhibition of subunit phosphorylation (P) and activation.

Table 1 Direct inhibitors of NADPH oxidase activity

Small-molecule NADPH oxidase inhibitors

Old and new NADPH oxidase inhibitors are presented in . The first inhibitors used were diphenyleneiodonium and apocynin, a naturally occurring inhibitor originally isolated from the roots of Picrorhiza kurroa. These have been the most widely studied and used as pharmacologic tools. However, they lack selectivity for NADPH oxidases. Diphenyleneiodonium inhibits flavin oxidases, thus affecting numerous enzymes.Citation179,Citation180 Apocynin is an orally active agent that acts as a prodrug, requiring a reaction with peroxidase to be able to block assembly of NADPH oxidase.Citation181 Its possible therapeutic value has been probed in animal models, where it blunted the development of hypertension and prevented endothelial dysfunction in rats rendered hypertensive by DOCA salt.Citation182 However, apocynin seems to have effects other than inhibition of NADPH oxidase, interfering also with metabolism of arachidonic acid,Citation183 synthesis of glutathione, and activation of AP-1 transcription factor.Citation184 Further, it has recently been shown to be a direct ROS scavenger in certain experimental conditions.Citation185

Phenylarsine oxide is a potent NOX2 inhibitor. It is ineffective once the oxidase complex is formed, suggesting that it prevents assembly of an active oxidase complex. Because phenylarsine oxide is known to form disulfide bonds with vicinal cysteine residues, it is postulated that these residues are only unmasked and accessible for phenylarsine oxide in the resting state of the NADPH oxidase complex.Citation186 However, this chemical reactivity with cysteine residues is likely to make this agent an inhibitor of a variety of other enzymes. Phenylarsine oxide has been demonstrated to decrease NADPH oxidase-derived ROS production in rat arteries,Citation187 and in vivo inhibited production of superoxide anions induced by angiotensin II and enhanced chronic heart failure in rabbits.Citation188 However, it is also shown that phenylarsine oxide may induce ROS production dependent on NADPH oxidase, which may mediate other effects like protein expression.Citation189

Gliotoxin is a mycotoxin extracted from Aspergillus species. It blocks NOX2 but is inefficient for NOX4 inhibition.Citation54 Gliotoxin interferes with phosphorylation of p47phox,Citation190 but also directly affects NOX2 activity.Citation191 It is speculated that gliotoxin acts by a mechanism similar to that of phenylarsine oxide.

Another chemical inhibitor of NADPH oxidase is aminoethyl benzenesulfonyl fluoride (AEBSF), but its action is at high concentrations (half maximal inhibitory concentration [IC50] > 1 mM) and it has off-target effects as a serine protease inhibitor. AEBSF interferes with the binding of p47phox and/or p67phox to the membrane of the NADPH oxidase complex, probably by a direct effect on NADPH oxidase.Citation192 It also interferes with the most commonly used assays for ROS.Citation193

VAS2870 is a cell-permeable thiotriazolopyrimidine compound that acts as a rapid and reversible inhibitor of NADPH oxidase activity. It does not seem to have direct ROS scavenging properties and had no effects on xanthine oxidase-mediated superoxide production or on endothelial nitric oxide synthase activity.Citation193,Citation194 VAS2870 inhibited elevated ROS production and restored endothelium-dependent relaxation in aortas from spontaneously hypertensive ratsCitation38 and limited phenylephrine-induced contraction in rat tail arteries.Citation195 Intrathecal administration of VAS2870 to mice following experimental stroke was shown to reduce brain infarct volume and improve the neurologic outcome.Citation69 Interestingly, in this study, Nox4−/− mice were similarly protected against brain infarction and neurologic deficits following stroke as those administered VAS2870. A more recently introduced triazolopyrimidine, VAS3947, has been shown to be relatively specific for NADPH oxidases versus nitric oxide synthase and xanthine oxidase, but it cannot be excluded that VAS3947 interferes with an alternative source of ROS, such as the mitochondrial electron chain.Citation193

GK-136901 is a pyrazolopyridine derivative, structurally related to VAS2870 and VAS3947, and recently identified as a potent selective inhibitor of NOX1 and NOX4 isoform-dependent ROS generation from disrupted cell membrane preparations; it had no detectable inhibitory effects against ROS generated in vitro by xanthine oxidase. GK-136901 has high oral bioavailability, lacks off-target effects against a panel of 135 enzymes, and has a good safety profile in vivo.Citation196 All together, this makes it a good drug candidate as a NADPH oxidase inhibitor, and it has been shown to attenuate hypoxia-induced pulmonary vascular cell proliferation in mice, suggesting its value in pulmonary hypertension.Citation197

S17834 blocks NADPH oxidase activity in intact endothelial cells and in isolated endothelial cell membranes without inhibiting ROS generated by xanthine oxidase in a cell-free preparation system, indicating that it does not have superoxide scavenger properties.Citation198 Further, when administered to apolipoprotein E-deficient and low-density lipoprotein receptor-deficient mice, S17834 suppressed TNF-induced leukocyte adhesion, reduced expression of plasminogen activator inhibitor 1, augmented nitric oxide production in endothelial monolayers, and showed atheroprotective properties.Citation198Citation200 Its major mechanism of action involves prevention of binding of cytosolic subunits (mainly p47phox) to the membrane complex of the enzyme. Although its exact mechanism of action remains unclear, plumbagin, a plant-derived naphthoquinone, has been used to inhibit NADPH oxidase-dependent superoxide production in cell lines that express NOX4Citation201 and as an antiatherosclerotic drug in animals.Citation202

Several phenothiazines have been identified as NOX1 inhibitors by high-throughput screening using an HT29 cell-based assay.Citation203 The inhibitor 2-acetylphenothiazine (ML171) had an IC50 for NOX1 in the nanomolar range, whereas the IC50 values for NOX2, NOX3, and NOX4 were in the micromolar range, similar to the IC50 value for xanthine oxidase. NOX5 was not included in the assays. ML171 appears to target the NOX1 catalytic subunit but not its cytosolic regulators. Due to the presence of the phenothiazine structure, a possible side effect of ML171 is a potential antipsychotic action. However, binding assays for a large battery of human receptors and channels in the central nervous system led to the conclusion that ML171 does not likely exert unwanted antipsychotic effects.Citation203

Fulvene-5 is a recently described NADPH oxidase inhibitor from the family of fulvenes, which are highly water-soluble aromatic ring structures. Fulvene-5 showed inhibitory activity against NOX2 and NOX4. However, there are no data on the IC50 values or its specificity and activity toward other NOX isoforms.Citation204 Although little public information is available, some other small NADPH oxidase inhibitors have been investigated by pharmaceutical companies. An overview of some of these products can be found in the review published by Jaquet et al.Citation177

In some cases, comparative studies between old (normally diphenyleneiodonium and/or apocynin) and new small NADPH oxidase inhibitors have been reported to clarify their potential value as NADPH oxidase inhibitors, selectivity versus other sources of ROS, and inherent ROS scavenger properties.Citation193

Peptide inhibitors

In an attempt to eliminate off-target effects and gain selectivity for some NOX isoforms, the focus has been shifted on some occasions to the development of peptide inhibitors, which truly inhibit activation of NADPH oxidase. A more detailed review of these peptides has been published recently by El-Benna et al.Citation205 Nox2ds-tat, a cell-permeative chimeric form targeting the assembly of NOX2, is a peptide inhibitor that binds to the p47phox subunit and prevents its interaction with other NADPH oxidase subunits.Citation206 Therefore, it inhibits the NOX1 and NOX2 isoforms, but not NOX4, because this subunit is p47phox-independent. It decreased superoxide production and inhibited angiotensin II-induced blood pressure increases in animal models.Citation103 Nox2ds, a nonchimeric peptide that mimics a sequence in the cytosolic B-loop of NOX2, was shown to inhibit superoxide anion production by the NOX2 isoform but not ROS production by NOX1 oxidase or NOX4 oxidase.Citation207 Other peptide inhibitors were derived from p22phox, p47phox, or Rac1 subunits to prevent their phosphorylation, activation, or assembly to form the final enzyme complex.Citation205 The proline-argininerich peptide, PR39 protein, is an inhibitor not derived from NADPH oxidase components that binds to the Src-homology region 3 (SH3) domain of p47phox subunits, limiting its regulatory activity.Citation208 However, in terms of its mechanism of action, PR39 is not completely selective for NADPH oxidase and can affect any other protein possessing the SH3 domain. All these peptide inhibitors have shown low stability and lack of oral availability, making them clinically unattractive.

Naturally occurring antioxidants

The search for novel and specific NADPH oxidase inhibitors continues, and naturally occurring antioxidants represent another possible approach. In addition to apocynin, the best studied molecule of this origin, other natural components have NADPH oxidase inhibition properties. It has been found that polyphenols, apart from their well known superoxide radical scavenging ability, also decrease the activity of NADPH oxidase.Citation209 In fact, the beneficial effects on cardiovascular disease may partly explain the protective role of polyphenol-containing foods and beverages such as fruit, vegetables, green tea, and red wine.Citation210Citation214 It should be kept in mind that designing antioxidant strategies with this type of polyphenol or other natural compounds requires additional randomized studies with dietary interventions.

According to the criteria suggested by Jaquet et alCitation177 for defining new NADPH oxidase inhibitors, procyanidin-rich fractions from distilled grape pomace should contain such type of drugs. Procyanidins from this origin, which are polyphenol compounds of the flavonoid group, inhibit NADPH oxidase activity in endothelial cells and cell disrupted systems. They also exerted inhibition at lower concentrations than those needed to scavenge superoxide anions in a cell-free in vitro system.Citation215 Therefore, natural procyanidins from grapes seem to be a new source of NADPH oxidase inhibitors.

Other natural products have been shown to inhibit NADPH oxidase activity or prevent overexpression of some of its subunits in in vitro cell models under certain conditions. Flavonoids from Inula Britannica L reduced p47phox expression and phosphorylation in vascular smooth muscle cells, with a subsequent reduction in production of superoxide anions.Citation168 Ginkgolide B, from the ginkgo biloba tree, attenuated the enhancement of NOX4 expression and ROS generation in human endothelial cells treated with oxidized low-density lipoprotein.Citation170 Rutaecarpine reversed the NADPH oxidase activity induced by hypoxia-reoxygenation and upregulation of NOX2, NOX4, and p47phox in myocardial cells in vitro.Citation171 Reinioside C, a major compound of Polygala fallax H, attenuated the upregulation of NADPH oxidase (NOX2 and p22phox) induced by oxidized low-density lipoprotein, generation of ROS, and activation of NF-κB in endothelial cells.Citation172 Resveratrol, a stilbene found in grapes and wine, and without superoxide scavenger properties, has been shown to inhibit NADPH oxidase activity in vascular tissues.Citation216 It also downregulated NOX4 and upregulated superoxide dismutase-1 and glutathione peroxidase 1 (antioxidative enzymes) in a model of human endothelial cells.Citation163 Therefore, resveratrol modifies the expression profile of pro-oxidative and antioxidative enzymes which could contribute to its vascular protective effect. Quercetin and its methylated metabolite, isorhamnetin, reduced endothelin 1-induced upregulation of the p47phox subunit and endothelial nitric oxide synthase uncoupling in the endothelium, but it is not clear if this is a direct or a protein kinase C-mediated mechanism.Citation173

Studies in animal models under pathophysiologic conditions have also shown the inhibitory activity of some of these compounds. Epicatechin reduced the increased NADPH oxidase activity and overexpression of p47phox and p22phox in DOCA salt animalsCitation167 and rats after chronic inhibition of nitric oxide synthesis.Citation217 Tanshinone IIA, a major constituent of Salvia miltiorrhiza B, inhibited increased NADPH oxidase activity and expression as well as superoxide anion production in a hypertensive rat model.Citation169 Folic acid supplementation in the diet attenuated NADPH oxidase and xanthine oxidase activity and restored superoxide dismutase activity in the kidney of the hyperhomocysteinemic rat.Citation218

Magnolol and honokiol are active components of Magnolia offcinalis that inhibit NADPH oxidase activity,Citation219 which explains some of their antioxidative and anti-inflammatory effects.Citation220 Honokiol has been shown to reduce myocardial infarct size and cardiac arrhythmias in rats.Citation221,Citation222 This compound also suppresses apoptosis in high glucose-induced human endothelial cells via NADPH oxidase inhibition.Citation223 Interestingly, this compound is able to inhibit NADPH oxidase after the enzyme is activated, but not before, which could be useful in pathologic conditions. In some cases, the natural product does not directly inhibit NADPH oxidase activity, but affects its regulation. Xuezhikang, an extract of red yeast rice, reduces NADPH oxidase activity by decreasing membrane translocation of its regulatory subunit p47phox by inhibition of extracellular signal-regulated kinase 1/2 activation.Citation224

Puerarin, an isofavone-C-glucoside, interferes with the NADPH oxidase-related ROS pathways and blocks NF-κB activation by inhibiting phosphorylation of p47phox and Rac1.Citation225Tripterygium wilfordii Hook F is a traditional Chinese medicinal plant that contains some compounds with inhibitory effects on NADPH oxidase activity. One of these compounds is tripterine, which prevents Janus kinase-dependent activation of inducible nitric oxide synthase and NOX1, an effect that consequently protects endothelial barrier function.Citation226 Celastrol, another compound of the same plant, was demonstrated to be a potent nonspecific inhibitor of NOX1, NOX2, NOX4, and NOX5, with increased potency against NOX1 and NOX2.Citation227 The suggested mechanism for this action is inhibition of the functional association between cytosolic subunits and the membrane enzyme complex.

Pharmacophore models

In cell systems overexpressing NOX4, several NOX4 oxidase inhibitors have been identified from compounds containing one of the following five core structures: oxalyl hydrazides, flavonoids, oxindoles, benzoquinolines, and benzothiophenes.Citation228 From these data, the authors mapped the common pharmacophore points and constructed a pharmacophore model of a NOX4 oxidase inhibitor that comprised two hydrogen bond donors and acceptors and two aromatic-hydrophobic rings.Citation228 Similarly, structure-activity relationship investigations around the pyrazolopyridine dione core and high-throughput screening led to the discovery of new inhibitors selective for NOX4 and NOX1 isoforms and with little affinity for the NOX2 isoform.Citation196 Additionally, they showed good oral bioavailability. Independent of NOX isoform specificity, many NADPH oxidase inhibitors show some common structural features, ie, they are generally flat and lipophilic aromatic heterocyclic compounds. Further work is needed to determine the isoform selectivity of compound bases in pharmacophore models as well as their specificity for inhibition of ROS production via NADPH oxidases versus other sources.

Novel perspectives

It has been discovered recently that zinc signals have an essential role in the redox signaling network, and this implies that regulation of zinc homeostasis by zinc administration could be a novel strategic pharmacologic intervention for oxidative stress. In fact, intracellular zinc has a protective role in myocardial recovery from oxidative stress enhanced by ischemia-reperfusion injury.Citation229 Pyrithione, a zinc ionophore, reversed expression of NOX2 induced by hypoxia-reoxygenation and p47phox phosphorylation in rat cardiac myocytes, indicating that this compound protects cardiomyocytes from reperfusion injury by attenuation of NADPH oxidase activity.Citation230

A novel approach in the search for specific NADPH oxidase inhibitors has been targeted to the binding of the proline-rich domain of p22phox to p47phox, a prerequisite for activation of some NADPH oxidase isoforms, ie, NOX1 and NOX2. A high-throughput screen monitoring this interaction identified ebselen, a glutathione peroxidase mimetic and hydrogen peroxide scavenger, and several of its analogs as potent inhibitors of this binding.Citation231 This finding led to the proposal of ebselen as a new anti-inflammatory drug in view of its antioxidant properties.Citation232 Naturally occurring products like celastrol have also been shown to disrupt binding of the proline-rich region of p22phox to the tandem SH3 domain of p47phox and NOX organizer 1 (NOXO1), the p47phox homologous. Naloxone, an opioid antagonist, has recently been shown to bind to NOX2 and inhibit both its action, and translocation of the p47phox subunit to the membrane.Citation233 This suggests a new mechanism of nonopioid action of naloxone, which has been suggested to be useful in the prevention of inflammation-mediated neurodegeneration.

Modulation of signal transduction upstream of NADPH oxidase activation

In addition to direct inhibition of NADPH oxidase, some drugs have the ability to regulate signaling pathways that participate in activation of NADPH oxidase activity. The main mechanisms involved in this effect are inhibition of the regulating NADPH oxidase subunits, by reduction of their phosphorylation or by blocking their translocation to the membrane complex of the enzyme, or reduction of the expression of one or more NADPH oxidase subunits (). Interestingly, some drugs with this ability are commonly used in cardiovascular disease. Therefore, it is reasonable to think that, in addition to the primary mechanisms of action of these drugs, regulation of NADPH oxidase activity at the vascular level could also contribute to their beneficial effects.

Figure 6 Indirect inhibition of NADPH oxidase. Points for NADPH oxidase activity inhibition or reduction of the enzyme’s expression. The stimulating effects of the renin-angiotensin-aldosterone system (RAAS) on NADPH oxidase activity can be stopped by the inhibition of renin, angiotensin converting enzyme (ACE) or blockage of angiotensin II receptor 1 (AT1) or aldosterone receptor. Protein kinase C inhibition reduces NADPH oxidase phosphorylation (P) and expression. Glucagon-like peptide-1 and tyrosine kinase inhibition reduce the activity and the expression of NADPH oxidases. Sirtuins reduce NOX1 expression, whereas calcium channel blockers reduce NOX5 activity and statins decrease cytosolic subunit translocation to the membrane-fixed enzyme complex.

Abbreviations: Ca2+, calcium ion; CaM, calmodulin; FAD, flavin adenine dinucleotide; HSP90, heat shock protein 90; NADPH, reduced nicotinamide adenine dinucleotide phosphate; Tyr-K, tyrosine kinase.
Figure 6 Indirect inhibition of NADPH oxidase. Points for NADPH oxidase activity inhibition or reduction of the enzyme’s expression. The stimulating effects of the renin-angiotensin-aldosterone system (RAAS) on NADPH oxidase activity can be stopped by the inhibition of renin, angiotensin converting enzyme (ACE) or blockage of angiotensin II receptor 1 (AT1) or aldosterone receptor. Protein kinase C inhibition reduces NADPH oxidase phosphorylation (P) and expression. Glucagon-like peptide-1 and tyrosine kinase inhibition reduce the activity and the expression of NADPH oxidases. Sirtuins reduce NOX1 expression, whereas calcium channel blockers reduce NOX5 activity and statins decrease cytosolic subunit translocation to the membrane-fixed enzyme complex.

Blockade of the renin-angiotensin-aldosterone system

The renin-angiotensin-aldosterone system is well characterized as being critically involved in cardiovascular disease. Its participation in the regulation of NADPH oxidase function has been demonstrated.Citation234,Citation235 Therefore, activation of NADPH oxidase is interrupted by inhibition of the renin-angiotensin-aldosterone system at any level, ie, renin inhibition, angiotensin-converting enzyme inhibition, angiotensin II blockade, or mineralocorticoid receptor blockade. Angiotensin-converting enzyme inhibitors affect several mechanisms of NADPH oxidase activity, including expression of the subunits, enzyme activation, and even AT1 receptor levels in animal models of cardiovascular disease or nephropathy.Citation236Citation238 The effects of angiotensin II receptor antagonists on NADPH oxidase are superior to those exerted by angiotensin-converting enzyme inhibitors.Citation239 Blockade of the AT1 receptor causes inhibition of NADPH oxidase and a decrease in superoxide anion production with noticeable improvement of endothelial function.Citation234,Citation240 Hence, AT1 receptor blockers can be considered as one of the most potent drugs clinically available for inhibiting activation of NADPH oxidase in the cardiovascular system.

Aliskiren, a new direct renin inhibitor, reduced vascular NADPH oxidase activity and atherosclerotic plaque in apolipoprotein E-deficient mice.Citation241 However, neither aliskiren alone nor the combination of aliskiren and valsartan resulted in a greater reduction in myocardial fibrosis or hypertrophy or markers of oxidative stress in a transgenic model of tissue renin overexpression.Citation242

Mineralocorticoid receptor activation by aldosterone includes mechanisms by which cardiomyocytes trigger NOX-dependent and reactive oxygen species-mediated coronary endothelial dysfunction.Citation243 Expression of NOX isoforms like NOX1 can be also regulated by aldosterone in vascular tissue.Citation244 Concordantly, use of eplerenone to antagonize mineralocorticoid receptors improved endothelial dysfunction and inhibited NADPH oxidase subunits in the post-myocardial infarction phase.Citation245 In summary, inhibition of the renin-angiotensin-aldosterone system is a promising mechanism for regulation of NADPH oxidase in cardiovascular disease, and should be investigated further in clinical trials.

Statins

Statins, also known as 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, aid in the prevention of endothelial dysfunction by lowering low-density lipoprotein cholesterol levels and enhancing nitric oxide-dependent vasodilatation.Citation246 Increasing evidence suggests that, at the same time, the pleiotropic effects of statins involve direct reduction of oxidative stress in blood vessels. Statins reduce both activity and expression of NOX1Citation247 and NOX2Citation248 and the activity of NOX4 in endothelial cells.Citation39 Therefore, several NOX isoforms may be affected. The effects of statins on activation of NADPH oxidase have been associated with inhibition of the geranylgeranylation of Rac1-GTPase (a regulatory subunit of NADPH oxidase complex), which is a required condition for Rac1 activation.Citation247,Citation249 However, this mechanism could explain the inhibition of Rac1 translocation but not the changes in expression of NOX isoforms and some NADPH oxidase subunits observed with statins. In fact, the mechanisms of action seem to differ from one vascular cell type to another, and there are conflicting data about the NADPH oxidase subunit implicated in each case.Citation250 Atorvastatin, simvastatin, rosuvastatin, and fluvastatin have been shown to inhibit NADPH oxidase activity and expression of at least the p22phox and NOX1 isoforms.Citation248 Moreover, the inhibitory effects of pitavastatin on NADPH oxidase activity have been shown to be independent of endothelial nitric oxide synthase, which was observed in mice deficient in this enzyme.Citation251 Together, these data suggest that inhibition of NADPH oxidases may contribute to the beneficial cardiovascular effects of the statins, although relatively few human trials have been carried out to date and more specific biomarkers for the antioxidant effects of statins are needed to separate this action from their lipid-lowering effects.Citation252

Calcium channel blockers

The effects of calcium channel blockers have been studied in the context of NOX5, which is a calcium-dependent NADPH oxidase. Such phenomena link calcium to NOX5 signaling, which is potentially important in the regulation of endothelial function by angiotensin II and endothelin-1.Citation253 However, because NOX5 is expressed predominantly in endothelial cells, concerns have been raised that the endothelium lacks the channels where calcium antagonists work. Amlodipine and manidipine reduced superoxide generation by inhibiting overexpression of NADPH oxidase in angiotensin II-stimulated rat endothelial cells.Citation254 These effects may also lead to decreased expression of adhesion and chemoattractant molecules, thereby explaining the antiatherosclerotic effects of calcium channel blockers.Citation255Citation257

Protein kinase C inhibitors

Several studies have demonstrated that protein kinase C is a strong NADPH oxidase activator,Citation258 because this kinase phosphorylates NOX2Citation259 and even NOX5, elevating the calcium sensitivity of this isoform.Citation260 However, protein kinase C participates in the signaling pathways of important stimuli for endothelial cells, such as TNF-α,Citation258,Citation261 angiotensin II,Citation262 and low-density lipoprotein cholesterol.Citation263 Clinical studies have related the inhibition of protein kinase C to a decrease in superoxide anion generation and decreased hyperglycemia-induced endothelial dysfunction.Citation264 However, the difficulty in translating this knowledge to clinical practice lies in the divergent and different roles of protein kinase C in the regulation of several cellular functions and their differences between distinct cell types.

Tyrosine kinase inhibitors

Tyrosine kinase inhibitors (tyrphostins) participate in regulation of NADPH oxidase activity and expression. Tyrphostin AG490 reduced the increased NADPH oxidase activity and expression of NOX1, NOX2, and NOX4 isoforms in apolipoprotein E-deficient mice.Citation265 This effect ran parallel to a marked reduction in atherosclerotic lesions of the aorta. In vascular tissue, the main target for the NADPH oxidase inhibitory effect of tyrphostins could be the EGFR kinase. This enzyme has been related to activation of NADPH oxidase in vascular tissue, and tyrosine kinase inhibition has been associated with restoration of impaired endothelium-dependent relaxation,Citation102 inhibition of angiotensin II-induced and pyrogallol-induced potentiation of contraction,Citation266 and even reduction of NOX2 and NOX4 overexpression.Citation267 Together, these results implicate EGFR kinase and NADPH oxidase activation downstream in endothelial dysfunction.

Sirtuins

A relationship has been shown recently between sirtuin 1, a nicotinamide adenine dinucleotide-dependent deacetylase, and NADPH oxidases, more specifically with the NOX1 isoform.Citation268 Sirtuin 1 exerts protective effects in atherosclerosis by promoting endothelium-dependent vascular relaxation. In vascular smooth muscle cells, overexpression of sirtuin 1 significantly inhibited angiotensin II-induced hypertrophy, while knockdown of sirtuin-1 resulted in increased growth of vascular smooth muscle cells. Expression of NOX1 induced by angiotensin II was inhibited by sirtuin 1 in these cells, whereas sirtuin 1 inhibition increased NOX1 expression.Citation269

In isolated aorta from Wistar rats, inhibition of sirtuin 1 significantly increased vascular superoxide production and enhanced NADPH oxidase activity and expression of p22phox and NOX4 subunits, together contributing to endothelial dysfunction, as evidenced by reduced relaxation in response to endothelium-dependent vasodilators.Citation270

Glucagon-like peptide-1

Glucagon-like peptide-1, one of the incretin hormones, has been reported to have beneficial cardiovascular effects. At least part of these effects has been related to antioxidant properties via NADPH oxidases. Glucagon-like peptide-1 decreased high-glucose-induced ROS production, as well as levels of NADPH oxidase subunits, such as p47phox and NOX2, in microvascular cardiac endothelial cells from rats.Citation271 In human umbilical vein endothelial cells, liraglutide, a glucagon-like peptide-1 analog, significantly decreased TNF-α-induced ROS production and overexpression of NOX2 and p22phox.Citation272 Together, these results suggest that the glucagon-like peptide-1 signaling pathway is another interesting point to target for reduction of NADPH oxidase activity.

Perspectives

The NOX1 and NOX2 isoforms currently appear to be the major contributors to oxidative stress in cardiovascular disease, so are the most logical therapeutic targets. However, none of the NADPH oxidase inhibitors found or developed thus far show selectivity for these isoforms. Taking into account that the other NADPH oxidase isoforms have important roles in some processes, inhibitors displaying a dual selectivity for NOX1 and NOX2 or for NOX2 and NOX4 over other NADPH oxidases will probably offer great benefits in terms of therapeutic efficacy versus side effects. In this way, some authors have postulated rational approaches to identify new target sites for novel dual inhibitors of vascular NOX1 and NOX2 isoforms.Citation27 The potential implications of inhibiting the important extravascular effects of NOX2 must be considered, given that its crucial role in the innate immune response will severely compromise immunologic function. This consideration will mandate dose titration of an NADPH oxidase inhibitor to partially inhibit the target NADPH oxidase in the vascular wall without affecting immune function.

Assembly and activation of the NADPH oxidase complex

Inhibiting formation of the NADPH oxidase complex is a known way of preventing activation of NADPH oxidase, particularly with the NOX1 and NOX2 isoforms. This approach has been most successful in inhibiting NOX2 because activation of this isoform depends on its association with the p47phox, p67phox, and p40phox cytosolic subunits. P47phox contains an autoinhibitory domain that prevents translocation of this subunit to the membrane NADPH oxidase subcomplex. Phosphorylation of p47phox relieves this autoinhibition and allows translocation to form the final NADPH oxidase complex.Citation273 NOX1 does not necessarily rely on complex assembly for activation because it can associate with NOXO1, the p47phox homologous, which does not have an autoinhibitory domain.Citation274 NOX4 and NOX5 do not use cytosolic subunits, so do not rely on complex assembly for activation.

Several drugs interfere with assembly of the NADPH oxidase complex as discussed above. AEBSF, apocynin, phenylarsine oxide, gliotoxin, and Nox2ds-tat prevent binding of p47phox to the membrane complex. Ebselen and celastrol block the union between p22phox and p47phox. A particular mechanism to inhibit complex formation is that used by the statins, which inhibit geranylgeranylation of Rac1, thereby preventing translocation and activation of the NADPH oxidase complex.

Specific inhibitors of phosphorylation

Phosphorylation is an important mechanism in regulating assembly and activation of the NADPH oxidase complex.Citation275 As mentioned above, p47phox must be phosphorylated in order to translocate and bind to the NADPH oxidase membrane complex. Phosphorylation of NOX2Citation259 or NOX5Citation260 by protein kinase C enhances its assembly or sensitivity to activation, respectively. Therefore, approaches to prevent specific subunit phosphorylation would be another useful strategy to inhibit some specific NADPH oxidase isoforms.

Calcium dependence of NOX5

The calcium dependence of the NOX5 isoform offers a unique point of action with therapeutic utility. Inhibition of calcium release is expected to interfere with NOX5 signaling, which could be important with regard to responses of endothelial cells to relevant stimuli at the endothelium, such as angiotensin II or endothelin 1.Citation253 This observation could explain the observation that calcium channel blockers potentiate the protective effects of angiotensin II receptor blockers on vascular endothelium in the rat.Citation276

Optimization of electron transfer inhibition by NADPH oxidase

NADPH oxidase inhibitors like diphenyleneiodonium block generation of ROS by preventing electron flow through the enzyme, so oxygen does not receive the electrons and ROS production is stopped. However, the activity of diphenyleneiodonium is nonspecific for other electron-transporting enzymes. Therefore, it will be interesting to develop an electron extractor capable of binding specifically to a particular NOX isoform. Another approach to interfering with electron transfer is anion channel inhibition because transfer of electrons across the membrane where NADPH oxidase is fixed is electrogenic and requires charge compensation through anion channels, such as chloride-proton antiporter ClC-3.Citation277,Citation278 Therefore, specific inhibitors of ClC-3 or similarly relevant antiporters may be an effective strategy to reduce NOX-mediated cardiovascular disease.

Reaching the target

Due to the specific pattern of expression of each NOX isoform in tissues and in each type of cell and because of the specific subcellular location of each NOX isoform, therapeutic targeting to the correct NADPH oxidase in its appropriate localization is not a banal question. In this sense, localization motifs in protein sequences provide an attractive therapeutic target for new NADPH oxidase inhibitors. However, additional studies identifying and characterizing NOX trafficking motifs and the adaptor proteins interacting with them are needed. A related question, which supposes an important limitation of antioxidant therapy, is the potential disruption of physiologic ROS-dependent cell signaling or the intracellular redox state. Therefore, creating antioxidants that target cellular microdomains containing NOX may be a solution to this limitation.

Controversies

One important question arises from the recent findings about the hypothetical beneficial role of NOX4. Mice deficient in this isoform show reduced expression of endothelial nitric oxide synthase and heme oxygenase-1 (the vascular source of nitric oxide and carbon monoxide, respectively), both of which are known for their protective vascular roles.Citation279 On the contrary, overexpression of NOX4 enhances vasodilatation induced by acetylcholine or histamine, which explains the lower systemic blood pressure found in these animals.Citation280 Moreover, NOX4 seems to produce hydrogen peroxide indirectlyCitation54 or directly,Citation55 although the exact mechanism for formation of hydrogen peroxide is not clear. Together, these results confirm the need for further investigations of the role of the specific contribution of each NADPH oxidase isoform in physiologic and pathophysiologic cardiovascular signaling.

Conclusion

The role of NADPH oxidases in cardiovascular disease has been confirmed. The combination of each NOX isoform and its particular cellular and subcellular locations may determine the specific signaling pathway where NADPH oxidases participate. However, further investigation is needed to clarify this issue in order to define the ideal target for NADPH oxidase inhibition. Regarding the method of inhibiting NADPH oxidase, NOX specificity is the focus of current investigations. Some inhibitors as well as direct and indirect approaches are available, as discussed above. Studies of the chemical structure-activity relationship for known inhibitors have provided pharmacophore models to assist in the search for new molecules. Likewise, some of the old and newer approaches include inhibition of assembly of the NADPH oxidase complex, subcellular translocation, post-transductional modifications, calcium entry/ release, and electron transfer. High-throughput screens for any of these activities could provide new inhibitors. From a general point of view, small-molecule inhibitors are preferred because of their hydrosolubility and oral bioavailability. However, other possibilities are not closed, with peptide inhibitors and monoclonal antibodies against NOX isoforms continuing to be under investigation, as well as a search for naturally occurring compounds. We are confident that, in the next few years, all this knowledge and research in progress will likely result in the development of new drugs for inhibition of NADPH oxidase and novel therapeutic approaches to the treatment of cardiovascular disease.

Acknowledgments

This study was supported in part by the Plan Nacional Español de I+D, 2008–2011, and the Instituto de Salud Carlos III, Subdirección General de Evaluación y Fomento de la Investigación (PI10/01403) cofinanced by European Regional Development Fund. The Isidro Parga Pondal program of the Xunta de Galicia supported the work of EÁ. The work of BP-D is supported by the Instituto de Salud Carlos III (FI11/00325).

Disclosure

The authors report no conflicts of interest in this work.

References

  • MyungSKJuWChoBKorean Meta-Analysis Study GroupEfficacy of vitamin and antioxidant supplements in prevention of cardiovascular disease: systematic review and meta-analysis of randomised controlled trialsBMJ2013346f1023335472
  • GriendlingKKSorescuDUshio-FukaiMNAD(P)H oxidase: role in cardiovascular biology and diseaseCirc Res200086549450110720409
  • CaiHHarrisonDGEndothelial dysfunction in cardiovascular diseases: the role of oxidant stressCirc Res2000871084084411073878
  • GaoLMannGEVascular NAD(P)H oxidase activation in diabetes: a double-edged sword in redox signallingCardiovasc Res200982192019179352
  • MontezanoACTouyzRMReactive oxygen species and endothelial function – role of nitric oxide synthase uncoupling and Nox family nicotinamide adenine dinucleotide phosphate oxidasesBasic Clin Pharmacol Toxicol20121101879421883939
  • OkamotoAIwamotoYMaruYOxidative stress-responsive transcription factor ATF3 potentially mediates diabetic angiopathyMol Cell Biol20062631087109716428460
  • LinSJShyueSKHungY YSuperoxide dismutase inhibits the expression of vascular cell adhesion molecule-1 and intracellular cell adhesion molecule-1 induced by tumor necrosis factor-alpha in human endothelial cells through the JNK/p38 pathwaysArterioscler Thromb Vasc Biol200525233434015576639
  • WarabiETakabeWMinamiTShear stress stabilizes NF-E2-related factor 2 and induces antioxidant genes in endothelial cells: role of reactive oxygen/nitrogen speciesFree Radic Biol Med200742226026917189831
  • ChenXLZhangQZhaoRDingXTummalaPEMedfordRMRac1 and superoxide are required for the expression of cell adhesion molecules induced by tumor necrosis factor-alpha in endothelial cellsJ Pharmacol Exp Ther2003305257358012606638
  • TrueALRahmanAMalikABActivation of NF-kappaB induced by H(2) O(2) and TNF-alpha and its effects on ICAM-1 expression in endothelial cellsAm J Physiol Lung Cell Mol Physiol20002792L302L31110926553
  • Wolfram KuhlmannCRWiebke LuddersDSchaeferCALysophosphatidylcholine-induced modulation of Ca(2+)-activated K(+)channels contributes to ROS-dependent proliferation of cultured human endothelial cellsJ Mol Cell Cardiol200436567568215135662
  • AbidMRKachraZSpokesKCAirdWCNADPH oxidase activity is required for endothelial cell proliferation and migrationFEBS Lett2000486325225611119713
  • MoldovanLMythreyeKGoldschmidt-ClermontPJSatterwhiteLLReactive oxygen species in vascular endothelial cell motility. Roles of NAD(P)H oxidase and Rac1Cardiovasc Res200671223624616782079
  • EyriesMCollinsTKhachigianLMModulation of growth factor gene expression in vascular cells by oxidative stressEndothelium200411213313915370072
  • HuTRamachandraraoS PSivaSReactive oxygen species production via NADPH oxidase mediates TGF-beta-induced cytoskeletal alterations in endothelial cellsAm J Physiol Renal Physiol20052894F816F82516159901
  • HuQZhengGZweierJLDeshpandeSIraniKZiegelsteinRCNADPH oxidase activation increases the sensitivity of intracellular Ca2+ stores to inositol 1,4,5-trisphosphate in human endothelial cellsJ Biol Chem200027521157491575710747906
  • MillerAADrummondGRSobeyCGNovel isoforms of NADPH-oxidase in cerebral vascular controlPharmacol Ther2006111392894816616784
  • WidderJDHarrisonDGCan vitamin E prevent cardiovascular events and cancer? Nat Clin Pract Cardiovasc Med200521051051116186847
  • BriegerKSchiavoneSMillerFJJrKrauseKHReactive oxygen species: from health to diseaseSwiss Med Wkly2012142w13659
  • LetoTLMorandSHurtDUeyamaTTargeting and regulation of reactive oxygen species generation by Nox family NADPH oxidasesAntioxid Redox Signal200911102607261919438290
  • CosentinoFFranciaPCamiciGGPelicciPGLüscherTFVolpeMFinal common molecular pathways of aging and cardiovascular disease: role of the p66Shc proteinArterioscler Thromb Vasc Biol200828462262818162611
  • CruzadoMCRislerNRMiatelloRMYa oGSchiffrinELTouyzRMVascular smooth muscle cell NAD(P)H oxidase activity during the development of hypertension: effect of angiotensin II and role of insulinlike growth factor-1 receptor transactivationAm J Hypertens2005181818715691621
  • SchrammAMatusikPOsmendaGGuzikTJTargeting NADPH oxidases in vascular pharmacologyVascul Pharmacol2012565–621623122405985
  • DoughanAKHarrisonDGDikalovSIMolecular mechanisms of angiotensin II-mediated mitochondrial dysfunction: linking mitochondrial oxidative damage and vascular endothelial dysfunctionCirc Res2008102448849618096818
  • RayRShahAMNADPH oxidase and endothelial cell functionClin Sci (Lond)2005109321722616104842
  • GuzikTJWestNEBlackEVascular superoxide production by NAD(P)H oxidase: association with endothelial dysfunction and clinical risk factorsCirc Res2000869E85E9010807876
  • DrummondGRSelemidisSGriendlingKKSobeyCGCombating oxidative stress in vascular disease: NADPH oxidases as therapeutic targetsNat Rev Drug Discov201110645347121629295
  • TakacISchroderKBrandesR PThe Nox family of NADPH oxidases: friend or foe of the vascular system? Curr Hypertens Rep2012141707822071588
  • AltenhoferSKleikersP WRadermacherKAThe NOX toolbox: validating the role of NADPH oxidases in physiology and diseaseCell Mol Life Sci201269142327234322648375
  • BrandesR PSchroderKComposition and functions of vascular nicotinamide adenine dinucleotide phosphate oxidasesTrends Cardiovasc Med2008181151918206804
  • BengtssonSHGulluyanLMDustingGJDrummondGRNovel isoforms of NADPH oxidase in vascular physiology and pathophysiologyClin Exp Pharmacol Physiol2003301184985414678249
  • NiuXLMadamanchiNRVendrovAENox activator 1: a potential target for modulation of vascular reactive oxygen species in atherosclerotic arteriesCirculation2010121454955920083677
  • SheehanALCarrellSJohnsonBStanicBBanfiBMillerFJJrRole for Nox1 NADPH oxidase in atherosclerosisAtherosclerosis2011216232132621411092
  • DouglasGBendallJKCrabtreeMJEndothelial-specific Nox2 overexpression increases vascular superoxide and macrophage recruitment in ApoE−/− miceCardiovasc Res2012941202922287576
  • LyleANDeshpandeNNTaniyamaYPoldip2, a novel regulator of Nox4 and cytoskeletal integrity in vascular smooth muscle cellsCirc Res2009105324925919574552
  • JudkinsC PDiepHBroughtonBRDirect evidence of a role for Nox2 in superoxide production, reduced nitric oxide bioavailability, and early atherosclerotic plaque formation in ApoE−/− miceAm J Physiol Heart Circ Physiol20102981H24H3219837950
  • EllmarkSHDustingGJFuiMNGuzzo-PernellNDrummondGRThe contribution of Nox4 to NADPH oxidase activity in mouse vascular smooth muscleCardiovasc Res200565249550415639489
  • WindSBeuerleinKArmitageMEOxidative stress and endothelial dysfunction in aortas of aged spontaneously hypertensive rats by NOX1/2 is reversed by NADPH oxidase inhibitionHypertension201056349049720606112
  • AlvarezERodino-JaneiroBKUcieda-SomozaRGonzález-JuanateyJRPravastatin counteracts angiotensin II-induced upregulation and activation of NADPH oxidase at plasma membrane of human endothelial cellsJ Cardiovasc Pharmacol201055220321220010434
  • BasuroySBhattacharyaSLefferC WParfenovaHNox4 NADPH oxidase mediates oxidative stress and apoptosis caused by TNF-alpha in cerebral vascular endothelial cellsAm J Physiol Cell Physiol20092963C422C43219118162
  • BasuroySTcheranovaDBhattacharyaSLefferC WParfenovaHNox4 NADPH oxidase-derived reactive oxygen species, via endogenous carbon monoxide, promote survival of brain endothelial cells during TNF-alpha-induced apoptosisAm J Physiol Cell Physiol20113002C256C26521123734
  • DatlaSRPeshavariyaHDustingGJMahadevKGoldsteinBJJiangFImportant role of Nox4 type NADPH oxidase in angiogenic responses in human microvascular endothelial cells in vitroArterioscler Thromb Vasc Biol200727112319232417717289
  • PeshavariyaHDustingGJJiangFNADPH oxidase isoform selective regulation of endothelial cell proliferation and survivalNaunyn Schmiedebergs Arch Pharmacol2009380219320419337723
  • PetryADjordjevicTWeitnauerMKietzmannTHessJGörlachANOX2 and NOX4 mediate proliferative response in endothelial cellsAntioxid Redox Signal200689–101473148416987004
  • WuRFMaZLiuZTeradaLSNox4-derived H2O2 mediates endoplasmic reticulum signaling through local Ras activationMol Cell Biol201030143553356820457808
  • GordilloGFangHParkHRoySNox-4-dependent nuclear H2O2 drives DNA oxidation resulting in 8-OHdG as urinary biomarker and hemangioendothelioma formationAntioxid Redox Signal201012893394319817625
  • PedruzziEGuichardCOllivierVNAD(P)H oxidase Nox-4 mediates 7-ketocholesterol-induced endoplasmic reticulum stress and apoptosis in human aortic smooth muscle cellsMol Cell Biol20042424107031071715572675
  • ClempusRESorescuDDikalovaAENox4 is required for maintenance of the differentiated vascular smooth muscle cell phenotypeArterioscler Thromb Vasc Biol2007271424817082491
  • SzocsKLassegueBSorescuDUpregulation of Nox-based NAD(P)H oxidases in restenosis after carotid injuryArterioscler Thromb Vasc Biol2002221212711788456
  • IsmailSSturrockAWuPNOX4 mediates hypoxia-induced proliferation of human pulmonary artery smooth muscle cells: the role of autocrine production of transforming growth factor-{beta}1 and insulin-like growth factor binding protein-3Am J Physiol Lung Cell Mol Physiol20092963L489L49919036873
  • SturrockAHuecksteadtT PNormanKNox4 mediates TGF-beta1-induced retinoblastoma protein phosphorylation, proliferation, and hypertrophy in human airway smooth muscle cellsAm J Physiol Lung Cell Mol Physiol20072926L1543L155517369289
  • ThomasSRWittingPKDrummondGRRedox control of endothelial function and dysfunction: molecular mechanisms and therapeutic opportunitiesAntioxid Redox Signal200810101713176518707220
  • DikalovSIDikalovaAEBikineyevaATSchmidtHHHarrisonDGGriendlingKKDistinct roles of Nox1 and Nox4 in basal and angiotensin II-stimulated superoxide and hydrogen peroxide productionFree Radic Biol Med20084591340135118760347
  • SerranderLCartierLBedardKNOX4 activity is determined by mRNA levels and reveals a unique pattern of ROS generationBiochem J2007406110511417501721
  • TakacISchroderKZhangLThe E-loop is involved in hydrogen peroxide formation by the NADPH oxidase Nox4J Biol Chem201128615133041331321343298
  • BelAibaRSDjordjevicTPetryANOX5 variants are functionally active in endothelial cellsFree Radic Biol Med200742444645917275676
  • JayDBPapaharalambusCASeidel-RogolBNox5 mediates PDGF-induced proliferation in human aortic smooth muscle cellsFree Radic Biol Med200845332933518466778
  • GuzikTJChenWGongoraMCCalcium-dependent NOX5 nicotinamide adenine dinucleotide phosphate oxidase contributes to vascular oxidative stress in human coronary artery diseaseJ Am Coll Cardiol200852221803180919022160
  • LoukogeorgakisS Pvan den BergMJSofatRRole of NADPH oxidase in endothelial ischemia/reperfusion injury in humansCirculation2010121212310231620479156
  • BedardKKrauseKHThe NOX family of ROS-generating NADPH oxidases: physiology and pathophysiologyPhysiol Rev200787124531317237347
  • GirouardHWangGGalloEFNMDA receptor activation increases free radical production through nitric oxide and NOX2J Neurosci20092982545255219244529
  • MillerAADrummondGRSchmidtHHSobeyCGNADPH oxidase activity and function are profoundly greater in cerebral versus systemic arteriesCirc Res200597101055106216210546
  • JackmanKAMillerAADe SilvaTMCrackPJDrummondGRSobeyCGReduction of cerebral infarct volume by apocynin requires pretreatment and is absent in Nox2-deficient miceBr J Pharmacol2009156468068819175604
  • KunzAAnratherJZhouPOrioMIadecolaCCyclooxygenase-2 does not contribute to postischemic production of reactive oxygen speciesJ Cereb Blood Flow Metab200727354555116820798
  • WalderCEGreenS PDarbonneWCIschemic stroke injury is reduced in mice lacking a functional NADPH oxidaseStroke19972811225222589368573
  • BraitVHJackmanKAWalduckAKMechanisms contributing to cerebral infarct size after stroke: gender, reperfusion, T lymphocytes, and Nox2-derived superoxideJ Cereb Blood Flow Metab20103071306131720145655
  • KahlesTLuedikePEndresMNADPH oxidase plays a central role in blood-brain barrier damage in experimental strokeStroke200738113000300617916764
  • ChenHSongYSChanPHInhibition of NADPH oxidase is neuroprotective after ischemia-reperfusionJ Cereb Blood Flow Metab20092971262127219417757
  • KleinschnitzCGrundHWinglerKPost-stroke inhibition of induced NADPH oxidase type 4 prevents oxidative stress and neurodegenerationPLoS Biol201089pii e1000479
  • JackmanKAMillerAADrummondGRSobeyCGImportance of NOX1 for angiotensin II-induced cerebrovascular superoxide production and cortical infarct volume following ischemic strokeBrain Res2009128621522019559686
  • KahlesTKohnenAHeumuellerSNADPH oxidase Nox1 contributes to ischemic injury in experimental stroke in miceNeurobiol Dis201040118519220580928
  • GiordanoFJOxygen, oxidative stress, hypoxia, and heart failureJ Clin Invest2005115350050815765131
  • GroteKFlachILuchtefeldMMechanical stretch enhances mRNA expression and proenzyme release of matrix metalloproteinase-2 (MMP-2) via NAD(P)H oxidase-derived reactive oxygen speciesCirc Res20039211e80e8612750313
  • SiwikDAPaganoPJColucciWSOxidative stress regulates collagen synthesis and matrix metalloproteinase activity in cardiac fibroblastsAm J Physiol Cell Physiol20012801C53C6011121376
  • DhallaNSTemsahRMNetticadanTRole of oxidative stress in cardiovascular diseasesJ Hypertens200018665567310872549
  • DhallaAKHillMFSingalPKRole of oxidative stress in transition of hypertrophy to heart failureJ Am Coll Cardiol19962825065148800132
  • MaytinMSiwikDAItoMPressure overload-induced myocardial hypertrophy in mice does not require gp91phoxCirculation200410991168117114981002
  • BendallJKCaveACHeymesCGallNShahAMPivotal role of a gp91(phox)-containing NADPH oxidase in angiotensin II-induced cardiac hypertrophy in miceCirculation2002105329329611804982
  • ByrneJAGrieveDJBendallJKContrasting roles of NADPH oxidase isoforms in pressure-overload versus angiotensin II-induced cardiac hypertrophyCirc Res200393980280514551238
  • LiJMGallN PGrieveDJChenMShahAMActivation of NADPH oxidase during progression of cardiac hypertrophy to failureHypertension200240447748412364350
  • KurodaJAgoTMatsushimaSZhaiPSchneiderMDSadoshimaJNADPH oxidase 4 (Nox4) is a major source of oxidative stress in the failing heartProc Natl Acad Sci U S A201010735155651557020713697
  • ZhangMBrewerACSchroderKNADPH oxidase-4 mediates protection against chronic load-induced stress in mouse hearts by enhancing angiogenesisProc Natl Acad Sci U S A201010742181211812620921387
  • LooiYHGrieveDJSivaAInvolvement of Nox2 NADPH oxidase in adverse cardiac remodeling after myocardial infarctionHypertension200851231932518180403
  • HoffmeyerMRJonesS PRossCRMyocardial ischemia/reperfusion injury in NADPH oxidase-deficient miceCirc Res200087981281711055986
  • DoerriesCGroteKHilfiker-KleinerDCritical role of the NAD(P)H oxidase subunit p47phox for left ventricular remodeling/ dysfunction and survival after myocardial infarctionCirc Res2007100689490317332431
  • WangJNShiNChenSYManganese superoxide dismutase inhibits neointima formation through attenuation of migration and proliferation of vascular smooth muscle cellsFree Radic Biol Med201252117318122062629
  • LassegueBGriendlingKKNADPH oxidases: functions and pathologies in the vasculatureArterioscler Thromb Vasc Biol201030465366119910640
  • AkasakiTOhyaYKurodaJIncreased expression of gp91phox homologues of NAD(P)H oxidase in the aortic media during chronic hypertension: involvement of the renin-angiotensin systemHypertens Res2006291081382017283869
  • OelzeMWarnholtzAFaulhaberJNADPH oxidase accounts for enhanced superoxide production and impaired endothelium-dependent smooth muscle relaxation in BKbeta1−/− miceArterioscler Thromb Vasc Biol20062681753175916763163
  • WangPTangFLiRContribution of different Nox homologues to cardiac remodeling in two-kidney two-clip renovascular hypertensive rats: effect of valsartanPharmacol Res200755540841717324585
  • NakanoDKurumazukaDNagaiYNishiyamaAKisoYMatsumuraYDietary sesamin suppresses aortic NADPH oxidase in DOCA salt hypertensive ratsClin Exp Pharmacol Physiol200835332432617941888
  • NakamuraTYamamotoEKataokaKBeneficial effects of pioglitazone on hypertensive cardiovascular injury are enhanced by combination with candesartanHypertension200851229630118158350
  • DikalovaAClempusRLassegueBNox1 overexpression potentiates angiotensin II-induced hypertension and vascular smooth muscle hypertrophy in transgenic miceCirculation2005112172668267616230485
  • DikalovaAEGongoraMCHarrisonDGLambethJDDikalovSGriendlingKKUpregulation of Nox1 in vascular smooth muscle leads to impaired endothelium-dependent relaxation via eNOS uncouplingAm J Physiol Heart Circ Physiol20102993H673H67920639222
  • GavazziGBanfiBDeffertCDecreased blood pressure in NOX1-deficient miceFEBS Lett2006580249750416386251
  • MatsunoKYamadaHIwataKNox1 is involved in angiotensin II-mediated hypertension: a study in Nox1-deficient miceCirculation2005112172677268516246966
  • GavazziGDeffertCTrocmeCSchäppiMHerrmannFRKrauseKHNOX1 deficiency protects from aortic dissection in response to angiotensin IIHypertension200750118919617502491
  • MehranpourPWangSSBlancoRRThe C242T CYBA polymorphism as a major determinant of NADPH oxidase activity in patients with cardiovascular diseaseCardiovasc Hematol Agents Med Chem20097325125919689263
  • MorenoMUSan JoseGFortunoABeloquiODíezJZalbaGThe C242T CYBA polymorphism of NADPH oxidase is associated with essential hypertensionJ Hypertens20062471299130616794479
  • MittalMRothMKonigPHypoxia-dependent regulation of nonphagocytic NADPH oxidase subunit NOX4 in the pulmonary vasculatureCirc Res2007101325826717585072
  • SelemidisSSobeyCGWinglerKSchmidtHHDrummondGRNADPH oxidases in the vasculature: molecular features, roles in disease and pharmacological inhibitionPharmacol Ther2008120325429118804121
  • JungOSchreiberJGGeigerHPedrazziniTBusseRBrandesR Pgp91phox-containing NADPH oxidase mediates endothelial dysfunction in renovascular hypertensionCirculation2004109141795180115037533
  • ReyFECifuentesMEKiarashAQuinnMTPaganoPJNovel competitive inhibitor of NAD(P)H oxidase assembly attenuates vascular O(2)(−) and systolic blood pressure in miceCirc Res200189540841411532901
  • BrennanAMSuhS WWonSJNADPH oxidase is the primary source of superoxide induced by NMDA receptor activationNat Neurosci200912785786319503084
  • ParkLZhouPPitstickRNox2-derived radicals contribute to neurovascular and behavioral dysfunction in mice overexpressing the amyloid precursor proteinProc Natl Acad Sci U S A200810541347135218202172
  • BanfiBMolnarGMaturanaAA Ca(2+)-activated NADPH oxidase in testis, spleen, and lymph nodesJ Biol Chem200127640375943760111483596
  • Ushio-FukaiMCompartmentalization of redox signaling through NADPH oxidase-derived ROSAntioxid Redox Signal20091161289129918999986
  • HilenskiLLClempusREQuinnMTLambethJDGriendlingKKDistinct subcellular localizations of Nox1 and Nox4 in vascular smooth muscle cellsArterioscler Thromb Vasc Biol200424467768314670934
  • Ushio-FukaiMZafariAMFukuiTIshizakaNGriendlingKKp22phox is a critical component of the superoxide-generating NADH/NADPH oxidase system and regulates angiotensin II-induced hypertrophy in vascular smooth muscle cellsJ Biol Chem19962713823317233218798532
  • ZafariAMUshio-FukaiMAkersMRole of NADH/NADPH oxidase-derived H2O2 in angiotensin II-induced vascular hypertrophyHypertension19983234884959740615
  • CalleraGEMontezanoACYogiATostesRCTouyzRMVascular signaling through cholesterol-rich domains: implications in hypertensionCurr Opin Nephrol Hypertens20071629010417293683
  • MillerFJJrChuXStanicBA differential role for endocytosis in receptor-mediated activation of Nox1Antioxid Redox Signal201012558359319737091
  • MillerFJJrFilaliMHussGJCytokine activation of nuclear factor kappa B in vascular smooth muscle cells requires signaling endosomes containing Nox1 and ClC-3Circ Res2007101766367117673675
  • ZhangAYYiFZhangGGulbinsELiPLLipid raft clustering and redox signaling platform formation in coronary arterial endothelial cellsHypertension2006471748016344372
  • YangBRizzoVTNF-alpha potentiates protein-tyrosine nitration through activation of NADPH oxidase and eNOS localized in membrane rafts and caveolae of bovine aortic endothelial cellsAm J Physiol Heart Circ Physiol20072922H954H96217028163
  • LiPLGulbinsELipid rafts and redox signalingAntioxid Redox Signal2007991411141517627466
  • ChenKDLiYSKimMMechanotransduction in response to shear stress. Roles of receptor tyrosine kinases, integrins, and ShcJ Biol Chem199927426183931840010373445
  • IshidaTPetersonTEKovachNLBerkBCMAP kinase activation by flow in endothelial cells. Role of beta 1 integrins and tyrosine kinasesCirc Res19967923103168756009
  • JalaliSLiYSSotoudehMShear stress activates p60 src-Ras-MAPK signaling pathways in vascular endothelial cellsArterioscler Thromb Vasc Biol19981822272349484987
  • SiegDJHauckCRIlicDFAK integrates growth-factor and integrin signals to promote cell migrationNat Cell Biol20002524925610806474
  • TzimaEdel PozoMAShattilSJChienSSchwartzMAActivation of integrins in endothelial cells by fluid shear stress mediates Rho-dependent cytoskeletal alignmentEMBO J200120174639464711532928
  • ChiarugiPFrom anchorage dependent proliferation to survival: lessons from redox signallingIUBMB Life200860530130718421772
  • ChiarugiPCirriPRedox regulation of protein tyrosine phosphatases during receptor tyrosine kinase signal transductionTrends Biochem Sci200328950951413678963
  • FinkelTSignal transduction by reactive oxygen speciesJ Cell Biol2011194171521746850
  • LeeSRKwonKSKimSRRheeSGReversible inactivation of protein-tyrosine phosphatase 1B in A431 cells stimulated with epidermal growth factorJ Biol Chem19982732515366153729624118
  • MengTCFukadaTTonksNKReversible oxidation and inactivation of protein tyrosine phosphatases in vivoMol Cell20029238739911864611
  • DejanaECoradaMLampugnaniMGEndothelial cell-to-cell junctionsFASEB J19959109109187615160
  • EsserSWolburgKWolburgHBreierGKurzchaliaTRisauWVascular endothelial growth factor induces endothelial fenestrations in vitroJ Cell Biol199814049479599472045
  • NawrothRPoellGRanftAVE-PTP and VE-cadherin ectodomains interact to facilitate regulation of phosphorylation and cell contactsEMBO J200221184885489512234928
  • WrightTJLeachLShawPEJonesPDynamics of vascular endothelial-cadherin and beta-catenin localization by vascular endothelial growth factor-induced angiogenesis in human umbilical vein cellsExp Cell Res2002280215916812413882
  • LinMTYe nMLLinC YKuoMLInhibition of vascular endothelial growth factor-induced angiogenesis by resveratrol through interruption of Src-dependent vascular endothelial cadherin tyrosine phosphorylationMol Pharmacol20036451029103614573751
  • van WeteringSvan BuulJDQuikSReactive oxygen species mediate Rac-induced loss of cell-cell adhesion in primary human endothelial cellsJ Cell Sci2002115Pt 91837184611956315
  • NwariakuFELiuZZhuXNADPH oxidase mediates vascular endothelial cadherin phosphorylation and endothelial dysfunctionBlood2004104103214322015271797
  • Yamaoka-TojoMTojoTKimHWIQGAP1 mediates VE-cadherin-based cell-cell contacts and VEGF signaling at adherence junctions linked to angiogenesisArterioscler Thromb Vasc Biol20062691991199716763158
  • Yamaoka-TojoMUshio-FukaiMHilenskiLIQGAP1, a novel vascular endothelial growth factor receptor binding protein, is involved in reactive oxygen species-dependent endothelial migration and proliferationCirc Res200495327628315217908
  • AllinghamMJvan BuulJDBurridgeKICAM-1-mediated, Src- and Pyk2-dependent vascular endothelial cadherin tyrosine phosphorylation is required for leukocyte transendothelial migrationJ Immunol200717964053406417785844
  • TangFYNguyenNMeydaniMGreen tea catechins inhibit VEGF-induced angiogenesis in vitro through suppression of VE-cadherin phosphorylation and inactivation of Akt moleculeInt J Cancer2003106687187812918064
  • ColavittiRPaniGBedogniBReactive oxygen species as downstream mediators of angiogenic signaling by vascular endothelial growth factor receptor-2/KDRJ Biol Chem200227753101310811719508
  • IkedaSUshio-FukaiMZuoLNovel role of ARF6 in vascular endothelial growth factor-induced signaling and angiogenesisCirc Res200596446747515692085
  • Ushio-FukaiMTangYFukaiTNovel role of gp91(phox)-containing NAD(P)H oxidase in vascular endothelial growth factor-induced signaling and angiogenesisCirc Res200291121160116712480817
  • WuRFGuYXuYCNwariakuFETeradaLSVascular endothelial growth factor causes translocation of p47phox to membrane ruffes through WAVE1J Biol Chem200327838368303684012855698
  • WuRFXuYCMaZNwariakuFESarosiGAJrTeradaLSSubcellular targeting of oxidants during endothelial cell migrationJ Cell Biol2005171589390416330715
  • IkedaSYamaoka-TojoMHilenskiLIQGAP1 regulates reactive oxygen species-dependent endothelial cell migration through interacting with Nox2Arterioscler Thromb Vasc Biol200525112295230016179592
  • MoldovanLMoldovanNISohnRHParikhSAGoldschmidt-ClermontPJRedox changes of cultured endothelial cells and actin dynamicsCirc Res200086554955710720417
  • WientjesFBReevesE PSoskicVFurthmayrHSegalAWThe NADPH oxidase components p47(phox) and p40(phox) bind to moesin through their PX domainBiochem Biophys Res Commun2001289238238811716484
  • BriggsM WSacksDBIQGAP proteins are integral components of cytoskeletal regulationEMBO Rep20034657157412776176
  • MateerSCWangNBloomGSIQGAPs: integrators of the cytoskeleton, cell adhesion machinery, and signaling networksCell Motil Cytoskeleton200355314715512789660
  • LiJMShahAMIntracellular localization and preassembly of the NADPH oxidase complex in cultured endothelial cellsJ Biol Chem200227722199521996011893732
  • Van BuulJDFernandez-BorjaMAnthonyECHordijkPLExpression and localization of NOX2 and NOX4 in primary human endothelial cellsAntioxid Redox Signal200573–430831715706079
  • ChenKKirberMTXiaoHYangYKeaneyJFJrRegulation of ROS signal transduction by NADPH oxidase 4 localizationJ Cell Biol200818171129113918573911
  • LiQHarrazMMZhouWNox2 and Rac1 regulate H2O2-dependent recruitment of TRAF6 to endosomal interleukin-1 receptor complexesMol Cell Biol200626114015416354686
  • LiQZhangYMardenJJBanfiBEngelhardtJFEndosomal NADPH oxidase regulates c-Src activation following hypoxia/reoxygenation injuryBiochem J2008411353154118397177
  • AbateCPatelLRauscherFJIIICurranTRedox regulation of fos and jun DNA-binding activity in vitroScience19902494973115711612118682
  • AllenRGTresiniMOxidative stress and gene regulationFree Radic Biol Med200028346349910699758
  • BloomDDhakshinamoorthySJaiswalAKSite-directed mutagenesis of cysteine to serine in the DNA binding region of Nrf2 decreases its capacity to upregulate antioxidant response element-mediated expression and antioxidant induction of NAD(P)H:quinone oxidoreductase1 geneOncogene200221142191220011948402
  • BodwellJEHolbrookNJMunckASulfhydryl-modifying reagents reversibly inhibit binding of glucocorticoid-receptor complexes to DNA-celluloseBiochemistry1984237139213986722099
  • GalterDMihmSDrogeWDistinct effects of glutathione disulphide on the nuclear transcription factor kappa B and the activator protein-1Eur J Biochem199422126396488174544
  • HainautPMilnerJRedox modulation of p53 conformation and sequence-specific DNA binding in vitroCancer Res19935319446944738402615
  • HansenJMGoYMJonesD PNuclear and mitochondrial compartmentation of oxidative stress and redox signalingAnnu Rev Pharmacol Toxicol20064621523416402904
  • KurodaJNakagawaKYamasakiTThe superoxide-producing NAD(P)H oxidase Nox4 in the nucleus of human vascular endothelial cellsGenes Cells200510121139115116324151
  • SorescuDWeissDLassegueBSuperoxide production and expression of nox family proteins in human atherosclerosisCirculation2002105121429143511914250
  • RichardDWolfCBarbeUKefiKBauseroPVisioliFDocosahexaenoic acid down-regulates endothelial Nox 4 through a sPLA2 signalling pathwayBiochem Biophys Res Commun2009389351652219737538
  • SpanierGXuHXiaNResveratrol reduces endothelial oxidative stress by modulating the gene expression of superoxide dismutase 1 (SOD1), glutathione peroxidase 1 (GPx1) and NADPH oxidase subunit (Nox4)J Physiol Pharmacol200960Suppl41114116
  • MuzaffarSJeremyJ YSparatoreAResveratrol reduces endothelial oxidative stress by modulating the gene expression of superoxide dismutase 1Br J Pharmacol2008155798499418846041
  • ShuklaNRossoniGHotstonMEffect of hydrogen sulphide-donating sildenafil (ACS6) on erectile function and oxidative stress in rabbit isolated corpus cavernosum and in hypertensive ratsBJU Int2009103111522152919245441
  • XuJLiNDaiDZYuFDaiYThe endothelin receptor antagonist CPU0213 is more effective than aminoguanidine to attenuate isoproterenol-induced vascular abnormality by suppressing overexpression of NADPH oxidase [correction of oxidas], ETA, ETB, and MMP9 in the vasculatureJ Cardiovasc Pharmacol2008521424818594475
  • Gomez-GuzmanMJimenezRSanchezMEpicatechin lowers blood pressure, restores endothelial function, and decreases oxidative stress and endothelin-1 and NADPH oxidase activity in DOCA-salt hypertensionFree Radic Biol Med2012521707922001745
  • ZhangHBWenJKZhangJFlavonoids from Inula britannica reduces oxidative stress through inhibiting expression and phosphorylation of p47(phox) in VSMCsPharm Biol201149881582021500971
  • WangPWuXBaoYTanshinone IIA prevents cardiac remodeling through attenuating NAD (P)H oxidase-derived reactive oxygen species production in hypertensive ratsPharmazie201166751752421812327
  • ZhangSChenBWuWBaoLQiRGinkgolide B reduces inflammatory protein expression in oxidized low-density lipoprotein-stimulated human vascular endothelial cellsJ Cardiovasc Pharmacol201157672172721436722
  • BaoMHDaiWLiYJHuC PRutaecarpine prevents hypoxia-reoxygenation-induced myocardial cell apoptosis via inhibition of NADPH oxidasesCan J Physiol Pharmacol201189317718621423291
  • BaiY PHuC PChenMFInhibitory effect of reinioside C on monocyte-endothelial cell adhesion induced by oxidized low-density lipoprotein via inhibiting NADPH oxidase/ROS/NF-kappaB pathwayNaunyn Schmiedebergs Arch Pharmacol2009380539940619730822
  • RomeroMJimenezRSanchezMQuercetin inhibits vascular superoxide production induced by endothelin-1: role of NADPH oxidase, uncoupled eNOS and PKCAtherosclerosis20092021586718436224
  • ManeaAManeaSAGafencuAVRaicuMSimionescuMAP-1-dependent transcriptional regulation of NADPH oxidase in human aortic smooth muscle cells: role of p22phox subunitArterioscler Thromb Vasc Biol200828587888518309110
  • LiHHortmannMDaiberACyclooxygenase 2-selective and nonselective nonsteroidal anti-inflammatory drugs induce oxidative stress by up-regulating vascular NADPH oxidasesJ Pharmacol Exp Ther2008326374575318550689
  • GuzikTJWestNEBlackEFunctional effect of the C242T polymorphism in the NAD(P)H oxidase p22phox gene on vascular superoxide production in atherosclerosisCirculation2000102151744174711023926
  • JaquetVScapozzaLClarkRAKrauseKHLambethJDSmall-molecule NOX inhibitors: ROS-generating NADPH oxidases as therapeutic targetsAntioxid Redox Signal200911102535255219309261
  • StreeterJThielWBriegerKMillerFJJrOpportunity Nox: the future of NADPH oxidases as therapeutic targets in cardiovascular diseaseCardiovasc Ther201331312513722280098
  • O’DonnellB VTewDGJonesOTEnglandPJStudies on the inhibitory mechanism of iodonium compounds with special reference to neutrophil NADPH oxidaseBiochem J1993290Pt 141498439298
  • O’DonnellVBSmithGCJonesOTInvolvement of phenyl radicals in iodonium inhibition of flavoenzymesMol Pharmacol19944647787857969060
  • StolkJHiltermannTJDijkmanJHVerhoevenAJCharacteristics of the inhibition of NADPH oxidase activation in neutrophils by apocynin, a methoxy-substituted catecholAm J Respir Cell Mol Biol1994111951028018341
  • GhoshMWangHDMcNeillJRRole of oxidative stress and nitric oxide in regulation of spontaneous tone in aorta of DOCA-salt hypertensive ratsBr J Pharmacol2004141456257314744820
  • EngelsFRenirieBFHartBALabadieR PNijkampF PEffects of apocynin, a drug isolated from the roots of Picrorhiza kurroa, on arachidonic acid metabolismFEBS Lett199230532542561299626
  • LapperreTSJimenezLAAntonicelliFApocynin increases glutathione synthesis and activates AP-1 in alveolar epithelial cellsFEBS Lett199944322352399989612
  • HeumullerSWindSBarbosa-SicardEApocynin is not an inhibitor of vascular NADPH oxidases but an antioxidantHypertension200851221121718086956
  • KutsumiHKawaiKJohnstonRBJrRokutanKEvidence for participation of vicinal dithiols in the activation sequence of the respiratory burst of human neutrophilsBlood1995859255925697727784
  • HamiltonCABrosnanMJAl-BennaSBergGDominiczakAFNAD(P)H oxidase inhibition improves endothelial function in rat and human blood vesselsHypertension200240575576212411473
  • LiYLGaoLZuckerIHSchultzHDNADPH oxidase-derived superoxide anion mediates angiotensin II-enhanced carotid body chemoreceptor sensitivity in heart failure rabbitsCardiovasc Res200775354655417499230
  • FanelusIDesrosiersRRReactive oxygen species generated by thiol-modifying phenylarsine oxide stimulate the expression of protein L-isoaspartyl methyltransferaseBiochem Biophys Res Commun2008371220320818407833
  • TsunawakiSYoshidaLSNishidaSKobayashiTShimoyamaTFungal metabolite gliotoxin inhibits assembly of the human respiratory burst NADPH oxidaseInfect Immun20047263373338215155643
  • NishidaSYoshidaLSShimoyamaTNunoiHKobayashiTTsunawakiSFungal metabolite gliotoxin targets flavocytochrome b558 in the activation of the human neutrophil NADPH oxidaseInfect Immun200573123524415618159
  • DiatchukVLotanOKoshkinVWikstroemPPickEInhibition of NADPH oxidase activation by 4-(2-aminoethyl)-benzenesulfonyl fluoride and related compoundsJ Biol Chem19972722013292133019148950
  • WindSBeuerleinKEuckerTComparative pharmacology of chemically distinct NADPH oxidase inhibitorsBr J Pharmacol2010161488589820860666
  • StielowCCatarRAMullerGNovel Nox inhibitor of oxLDL-induced reactive oxygen species formation in human endothelial cellsBiochem Biophys Res Commun2006344120020516603125
  • TsaiMHJiangMJReactive oxygen species are involved in regulating alpha1-adrenoceptor-activated vascular smooth muscle contractionJ Biomed Sci2010176720727219
  • LaleuBGagginiFOrchardMFirst in class, potent, and orally bioavailable NADPH oxidase isoform 4 (Nox4) inhibitors for the treatment of idiopathic pulmonary fibrosisJ Med Chem201053217715773020942471
  • GreenDEMurphyTCKangB YThe Nox4 inhibitor GKT137831 attenuates hypoxia-induced pulmonary vascular cell proliferationAm J Respir Cell Mol Biol201247571872622904198
  • CayatteAJRupinAOliver-KrasinskiJS17834, a new inhibitor of cell adhesion and atherosclerosis that targets NADPH oxidaseArterioscler Thromb Vasc Biol200121101577158411597929
  • JaulmesASansilvestri-MorelPRolland-ValognesGNox4 mediates the expression of plasminogen activator inhibitor-1 via p38 MAPK pathway in cultured human endothelial cellsThromb Res2009124443944619540572
  • ZangMXuSMaitland-ToolanKAPolyphenols stimulate AMP-activated protein kinase, lower lipids, and inhibit accelerated atherosclerosis in diabetic LDL receptor-deficient miceDiabetes20065582180219116873680
  • DingYChenZJLiuSCheDVetterMChangCHInhibition of Nox-4 activity by plumbagin, a plant-derived bioactive naphthoquinoneJ Pharm Pharmacol200557111111615638999
  • SharmaIGusainDDixitV PHypolipidaemic and antiatherosclerotic effects of plumbagin in rabbitsIndian J Physiol Pharmacol199135110141917004
  • GianniDTauletNZhangHA novel and specific NADPH oxidase-1 (Nox1) small-molecule inhibitor blocks the formation of functional invadopodia in human colon cancer cellsACS Chem Biol201051098199320715845
  • BhandarkarSSJaconiMFriedLEFulvene-5 potently inhibits NADPH oxidase 4 and blocks the growth of endothelial tumors in miceJ Clin Invest200911982359236519620773
  • El-BennaJDangPMPerianinATowards specific NADPH oxidase inhibition by small synthetic peptidesCell Mol Life Sci201269142307231422562604
  • DeLeoFRNauseefWMJesaitisAJBurrittJBClarkRAQuinnMTA domain of p47phox that interacts with human neutrophil flavocytochrome b558J Biol Chem19952704426246262517592831
  • CsanyiGCifuentes-PaganoEAl GhoulehINox2 B-loop peptide, Nox2ds, specifically inhibits the NADPH oxidase Nox2Free Radic Biol Med20115161116112521586323
  • ShiJRossCRLetoTLBlechaFPR-39, a proline-rich antibacterial peptide that inhibits phagocyte NADPH oxidase activity by binding to Src homology 3 domains of p47 phoxProc Natl Acad Sci U S A19969312601460188650211
  • Al-AwwadiNAAraizCBornetAExtracts enriched in different polyphenolic families normalize increased cardiac NADPH oxidase expression while having differential effects on insulin resistance, hypertension, and cardiac hypertrophy in high-fructose-fed ratsJ Agric Food Chem200553115115715631522
  • JimenezRLopez-SepulvedaRKadmiriMPolyphenols restore endothelial function in DOCA-salt hypertension: role of endothelin-1 and NADPH oxidaseFree Radic Biol Med200743346247317602962
  • BadimonLVilahurGPadroTNutraceuticals and atherosclerosis: human trialsCardiovasc Ther201028420221520633023
  • AgouniALagrue-Lak-HalAHMostefaiHARed wine polyphenols prevent metabolic and cardiovascular alterations associated with obesity in Zucker fatty rats (Fa/Fa)PLoS One200945e555719440378
  • Lopez-SepulvedaRGomez-GuzmanMZarzueloMJRed wine polyphenols prevent endothelial dysfunction induced by endothelin-1 in rat aorta: role of NADPH oxidaseClin Sci (Lond)2011120832133320977430
  • Dal-RosSZollJLangALChronic intake of red wine polyphenols by young rats prevents aging-induced endothelial dysfunction and decline in physical performance: role of NADPH oxidaseBiochem Biophys Res Commun2011404274374921167817
  • AlvarezERodino-JaneiroBKJerezMUcieda-SomozaRNúñezMJGonzález-JuanateyJRProcyanidins from grape pomace are suitable inhibitors of human endothelial NADPH oxidaseJ Cell Biochem201211341386139622134950
  • OralloFAlvarezECaminaMLeiroJMGómezEFernándezPThe possible implication of trans-resveratrol in the cardioprotective effects of long-term moderate wine consumptionMol Pharmacol200261229430211809853
  • Gomez-GuzmanMJimenezRSanchezMChronic (−)-epicatechin improves vascular oxidative and inflammatory status but not hypertension in chronic nitric oxide-deficient ratsBr J Nutr201110691337134821910946
  • HwangS YSiowYLAu-YeungKKHouseJFolic acid supplementation inhibits NADPH oxidase-mediated superoxide anion production in the kidneyAm J Physiol Renal Physiol20113001F189F19820980407
  • WangJ PHsuMFRaungSLInhibition by magnolol of formylmethionyl-leucyl-phenyl alanine-induced respiratory burst in rat neutrophilsJ Pharm Pharmacol199951328529410344629
  • ChuangDYChanMHZongYMagnolia polyphenols attenuate oxidative and inflammatory responses in neurons and microglial cellsJ Neuroinflammation2013101523356518
  • TsaiSKHuangCHHuangSSHungLMHongCYAntiarrhythmic effect of magnolol and honokiol during acute phase of coronary occlusion in anesthetized rats: influence of L-NAME and aspirinPharmacolog y1999595227233
  • TsaiSKHuangSSHongC YMyocardial protective effect of honokiol: an active component in Magnolia officinalisPlanta Med19966265035069000881
  • SheuMLChiangCKTsaiKSInhibition of NADPH oxidase-related oxidative stress-triggered signaling by honokiol suppresses high glucose-induced human endothelial cell apoptosisFree Radic Biol Med200844122043205018423412
  • LiPYangYLiuMXuezhikang, extract of red yeast rice, inhibited tissue factor and hypercoagulable state through suppressing nicotinamide adenine dinucleotide phosphate oxidase and extracellular signal-regulated kinase activationJ Cardiovasc Pharmacol201158330731821697731
  • KimJKimKMKimCSPuerarin inhibits the retinal pericyte apoptosis induced by advanced glycation end products in vitro and in vivo by inhibiting NADPH oxidase-related oxidative stressFree Radic Biol Med201253235736522609359
  • WuFHanMWilsonJXTripterine prevents endothelial barrier dysfunction by inhibiting endogenous peroxynitrite formationBr J Pharmacol200915761014102319508391
  • JaquetVMarcouxJForestENADPH oxidase (NOX) isoforms are inhibited by celastrol with a dual mode of actionBr J Pharmacol20111642b50752021501142
  • BorbelyGSzabadkaiIHorvathZSmall-molecule inhibitors of NADPH oxidase 4J Med Chem201053186758676220731357
  • KaragulovaGYueYMoreyraABoutjdirMKorichnevaIProtective role of intracellular zinc in myocardial ischemia/ reperfusion is associated with preservation of protein kinase C isoformsJ Pharmacol Exp Ther2007321251752517322024
  • KasiVBodigaSKommuguriUNSankuruSBodigaVLZinc pyrithione salvages reperfusion injury by inhibiting NADPH oxidase activation in cardiomyocytesBiochem Biophys Res Commun2011410227027521651898
  • SmithSMMinJGaneshTEbselen and congeners inhibit NADPH oxidase 2-dependent superoxide generation by interrupting the binding of regulatory subunitsChem Biol201219675276322726689
  • [No authors listed]Ebselen could be used as anti-inflammatory Nox inhibitorFuture Med Chem20124121533153522917242
  • WangQZhouHGaoHNaloxone inhibits immune cell function by suppressing superoxide production through a direct interaction with gp91phox subunit of NADPH oxidaseJ Neuroinflammation201293222340895
  • ZhangHSchmeisserAGarlichsCDAngiotensin II-induced superoxide anion generation in human vascular endothelial cells: role of membrane-bound NADH-/NADPH-oxidasesCardiovasc Res199944121522210615405
  • GriendlingKKMinieriCAOllerenshawJDAlexanderRWAngiotensin II stimulates NADH and NADPH oxidase activity in cultured vascular smooth muscle cellsCirc Res1994746114111488187280
  • Miguel-CarrascoJLZambranoSBlancaAJMateAVázquezCMCaptopril reduces cardiac inflammatory markers in spontaneously hypertensive rats by inactivation of NF-kBJ Inflamm (Lond)201072120462420
  • LiuX PPangYJZhuW WBenazepril, an angiotensin-converting enzyme inhibitor, alleviates renal injury in spontaneously hypertensive rats by inhibiting advanced glycation end-product-mediated pathwaysClin Exp Pharmacol Physiol200936328729619018797
  • OttoAFontaineJBerkenboomGRamipril treatment protects against nitrate-induced oxidative stress in eNOS−/− mice: an implication of the NADPH oxidase pathwayJ Cardiovasc Pharmacol200648184284916891913
  • RueckschlossUQuinnMTHoltzJMorawietzHDose-dependent regulation of NAD(P)H oxidase expression by angiotensin II in human endothelial cells: protective effect of angiotensin II type 1 receptor blockade in patients with coronary artery diseaseArterioscler Thromb Vasc Biol200222111845185112426214
  • BerryCHamiltonCABrosnanMJInvestigation into the sources of superoxide in human blood vessels: angiotensin II increases superoxide production in human internal mammary arteriesCirculation2000101182206221210801763
  • PossJWernerCLorenzDGenschCBöhmMLaufsUThe renin inhibitor aliskiren upregulates pro-angiogenic cells and reduces atherogenesis in miceBasic Res Cardiol2010105672573520857126
  • Whaley-ConnellAHabibiJRehmerNRenin inhibition and AT(1)R blockade improve metabolic signaling, oxidant stress and myocardial tissue remodelingMetabolism201362686187223352204
  • FavreJGaoJZhangADCoronary endothelial dysfunction after cardiomyocyte-specific mineralocorticoid receptor overexpressionAm J Physiol Heart Circ Physiol20113006H2035H204321441311
  • ShiGFuYJiangWActivation of Src-ATF1 pathway is involved in upregulation of Nox1, a catalytic subunit of NADPH oxidase, by aldosteroneEndocr J201158649149921505267
  • SartorioCLFraccarolloDGaluppoPMineralocorticoid receptor blockade improves vasomotor dysfunction and vascular oxidative stress early after myocardial infarctionHypertension200750591992517846350
  • AndersonTJMeredithITYeungACFreiBSelwynA PGanzPThe effect of cholesterol-lowering and antioxidant therapy on endothelium-dependent coronary vasomotionN Engl J Med199533284884937830729
  • WassmannSLaufsUMullerKCellular antioxidant effects of atorvastatin in vitro and in vivoArterioscler Thromb Vasc Biol200222230030511834532
  • RueckschlossUGalleJHoltzJZerkowskiHRMorawietzHInduction of NAD(P)H oxidase by oxidized low-density lipoprotein in human endothelial cells: antioxidative potential of hydroxymethylglutaryl coenzyme A reductase inhibitor therapyCirculation2001104151767177211591612
  • VecchioneCGentileMTAretiniAA novel mechanism of action for statins against diabetes-induced oxidative stressDiabetologia200750487488017279352
  • Martinez-GonzalezJBadimonLInfluence of statin use on endothelial function: from bench to clinicsCurr Pharm Des200713171771178617584107
  • YagiSAkaikeMAiharaKEndothelial nitric oxide synthase-independent protective action of statin against angiotensin II-induced atrial remodeling via reduced oxidant injuryHypertension201055491892320194307
  • KwokJMMaCCMaSRecent development in the effects of statins on cardiovascular disease through Rac1 and NADPH oxidaseVascul Pharmacol2013581–2213023085091
  • MontezanoACBurgerDParaviciniTMNicotinamide adenine dinucleotide phosphate reduced oxidase 5 (Nox5) regulation by angiotensin II and endothelin-1 is mediated via calcium/calmodulin-dependent, rac-1-independent pathways in human endothelial cellsCirc Res201010681363137320339118
  • TobaHShimizuTMikiSCalcium [corrected] channel blockers reduce angiotensin II-induced superoxide generation and inhibit lectin-like oxidized low-density lipoprotein receptor-1 expression in endothelial cellsHypertens Res200629210511616755144
  • YoshiiTIwaiMLiZRegression of atherosclerosis by amlodipine via anti-inflammatory and anti-oxidative stress actionsHypertens Res200629645746616940709
  • YamamotoEKataokaKDongYFBenidipine, a dihydropyridine L-type/T-type calcium channel blocker, affords additive benefits for prevention of cardiorenal injury in hypertensive ratsJ Hypertens20102861321132920224431
  • YaoRChengXLiaoYHMolecular mechanisms of felodipine suppressing atherosclerosis in high-cholesterol-diet apolipoprotein E-knockout miceJ Cardiovasc Pharmacol200851218819518287887
  • FreyRSRahmanAKeferJCMinshallRDMalikABPKCzeta regulates TNF-alpha-induced activation of NADPH oxidase in endothelial cellsCirc Res20029091012101912016268
  • RaadHPacletMHBoussettaTRegulation of the phagocyte NADPH oxidase activity: phosphorylation of gp91phox/NOX2 by protein kinase C enhances its diaphorase activity and binding to Rac2, p67phox, and p47phoxFASEB J20092341011102219028840
  • JagnandanDChurchJEBanfiBStuehrDJMarreroMBFultonDJNovel mechanism of activation of NADPH oxidase 5. Calcium sensitization via phosphorylationJ Biol Chem200728296494650717164239
  • LiJMFanLMChristieMRShahAMAcute tumor necrosis factor alpha signaling via NADPH oxidase in microvascular endothelial cells: role of p47phox phosphorylation and binding to TRAF4Mol Cell Biol20052562320233015743827
  • MollnauHWendtMSzocsKEffects of angiotensin II infusion on the expression and function of NAD(P)H oxidase and components of nitric oxide/cGMP signalingCirc Res2002904E58E6511884382
  • O’DonnellRWJohnsonDKZieglerLMEndothelial NADPH oxidase: mechanism of activation by low-density lipoproteinEndothelium200310629129714741844
  • GuttermanDDVascular dysfunction in hyperglycemia: is protein kinase C the culprit? Circ Res20029015711786509
  • FenyoIMFloreaICRaicuMManeaATyrphostin AG490 reduces NAPDH oxidase activity and expression in the aorta of hypercholesterolemic apolipoprotein E-deficient miceVascul Pharmacol2011543–610010621457788
  • LuCSuLYLeeRMGaoYJSuperoxide anion mediates angiotensin II-induced potentiation of contractile response to sympathetic stimulationEur J Pharmacol20085891–318819318538762
  • GalanMKassanMChoiSKA novel role for epidermal growth factor receptor tyrosine kinase and its downstream endo-plasmic reticulum stress in cardiac damage and microvascular dysfunction in type 1 diabetes mellitusHypertension2012601718022665120
  • PucaRNardinocchiLStaraceGNox1 is involved in p53 deacetylation and suppression of its transcriptional activity and apoptosisFree Radic Biol Med201048101338134620171273
  • LiLGaoPZhangHSIRT1 inhibits angiotensin II-induced vascular smooth muscle cell hypertrophyActa Biochim Biophys Sin (Shanghai)201143210310921118843
  • ZarzueloMJLopez-SepulvedaRSanchezMSIRT1 inhibits NADPH oxidase activation and protects endothelial function in the rat aorta: implications for vascular agingBiochem Pharmacol2011432103109
  • WangDLuoPWangYGlucagon-like peptide-1 protects against cardiac microvascular injury in diabetes via a cAMP/PKA/ Rho-dependent mechanismDiabetes20136251697170823364453
  • ShirakiAOyamaJKomodaHThe glucagon-like peptide 1 analog liraglutide reduces TNF-alpha-induced oxidative stress and inflammation in endothelial cellsAtherosclerosis2012221237538222284365
  • El-BennaJDangPMGougerot-PocidaloMAPriming of the neutrophil NADPH oxidase activation: role of p47phox phosphorylation and NOX2 mobilization to the plasma membraneSemin Immunopathol200830327928918536919
  • BanfiBClarkRAStegerKKrauseKHTwo novel proteins activate superoxide generation by the NADPH oxidase NOX1J Biol Chem200327863510351312473664
  • BokochGMDieboldBKimJSGianniDEmerging evidence for the importance of phosphorylation in the regulation of NADPH oxidasesAntioxid Redox Signal200911102429244119358632
  • YamamotoEKataokaKDongYFCalcium channel blockers, more than diuretics, enhance vascular protective effects of angiotensin receptor blockers in salt-loaded hypertensive ratsPLoS One201276e3916222720058
  • MorelandJGDavisA PBaileyGNauseefWMLambFSAnion channels, including ClC-3, are required for normal neutrophil oxidative function, phagocytosis, and transendothelial migrationJ Biol Chem200628118122771228816522634
  • LambFSMorelandJGMillerFJJrElectrophysiology of reactive oxygen production in signaling endosomesAntioxid Redox Signal20091161335134719207039
  • SchroderKZhangMBenkhoffSNox4 is a protective reactive oxygen species generating vascular NADPH oxidaseCirc Res201211091217122522456182
  • RayRMurdochCEWangMEndothelial Nox4 NADPH oxidase enhances vasodilatation and reduces blood pressure in vivoArterioscler Thromb Vasc Biol20113161368137621415386