<<

Physiol Rev 99: 311–379, 2019 Published October 31, 2018; doi:10.1152/physrev.00036.2017 SOURCES OF VASCULAR NITRIC OXIDE AND REACTIVE OXYGEN SPECIES AND THEIR REGULATION

X Jesús Tejero, Sruti Shiva, and Mark T. Gladwin

Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania

Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 99: 311–379, 2019. Published October 31, 2018; doi:10.1152/physrev.00036.2017.—Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of suffi- cient NO levels to regulate the resistance of the blood vessels and hence the mainte- Lnance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concen- tration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreduc- tases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the thera- peutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.

I. INTRODUCTION 311 These include oxygen radicals and peroxides, such as super- ·Ϫ II. NITRIC OXIDE GENERATION AND... 312 oxide (O2 ) and hydrogen peroxide (H2O2), nitrogen rad- · III. SUPEROXIDE AND HYDROGEN... 327 ical species, such as NO and nitrogen dioxide (NO2 ), and Ϫ IV. OTHER ROS 344 other species, such as peroxynitrite (ONOO ) and hypo- Ϫ V. CROSS-TALK BETWEEN ROS AND... 346 chlorite (ClO ). The species containing nitrogen are often VI. CONCLUDING REMARKS:... 347 treated separately as reactive nitrogen species (RNS). It is worth indicating that despite being long considered toxic I. INTRODUCTION species, most of these molecules have been shown to exert important signaling functions (249, 778, 937, 960). There- Nitric oxide (NO) is a small free radical molecule with fore, the role of many of these molecules in health and critical signaling roles. The discovery of the function of NO disease is related to their production rates, steady-state con- in the vascular endothelium as endothelium-derived relax- centrations, and the ability of the cellular antioxidant sys- ing factor led to the awarding of the 1998 Nobel Prize to tems to modulate their activity. Drs. Furchgott, Ignarro and Murad (36, 324, 449, 491, 716). The functions of NO in mammalian systems extend In general, dysregulated production of ROS/RNS, as is the beyond vascular signaling and are relevant in all organ sys- case for NO, leads to oxidative stress and deleterious con- tems, including but not limited to neuronal signaling, and sequences for living systems. However, as pointed out host defense (448, 659, 738). above, these molecules often have important signaling roles at low concentrations. For instance, the differences in re- A number of oxygen-related species of high chemical reac- sponse to NO at varying concentrations have attracted con- tivity are referred to as reactive oxygen species (ROS). siderable attention. It has been shown that low levels (pM/

0031-9333/19 Copyright © 2019 the American Physiological Society 311 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

nM) are physiological and related to the activation of high donors or supplementing with NOS substrates to reverse affinity primary binding targets such as soluble guanylyl endothelial dysfunction have had limited success. The use of cyclase (sGC) and cytochrome c oxidase (433, 863). An general antioxidants for the treatment of oxidative stress emerging paradigm proposes that intermediate levels (50– has also failed in most cases. Recent advances in the field 300 nM) can activate a range of positive and negative re- have provided many clues on why these approaches have sponses from wound healing to oncogenic pathways (938). been unsuccessful. We will discuss these and other relevant Higher concentrations of NO (Ͼ1 ␮M) can lead not only to physiological and pathophysiological issues and indicate oxidative stress but also nitrative and nitrosative stress via how advances in basic biochemistry of the generation of the generation of peroxynitrite and nitrosating species (411, NO/ROS have evolved our understanding and set new di- 412, 938, 939), and in combination with oxygen, can trig- rections in the field. ger posttranslational modification of proteins, lipids, and DNA (277, 433, 938). In this review, we will present the biology of NO and ROS in the cardiovascular system with special emphasis on their The production of adequate levels of NO in the vascular routes of formation, chemistry, mode of action, and dys- endothelium is critical for the regulation of blood flow and regulation in vascular disease. The formation pathways of vasodilation, as will be discussed at length in this review NO and the mechanisms of NO signaling will be discussed (299, 565, 573, 600, 786). In this context, it has become in sect. II. The proteins and biological systems generating increasingly appreciated that oxygen levels can impact the hydrogen peroxide and superoxide are treated in sect. III. oxidation/reduction properties of different proteins and Other ROS of particular relevance in the vascular system regulate NO levels (FIGURE 1) (367, 578, 595, 931). For are discussed in sect. IV. Section V will study the cross-talk example, nitric oxide synthases (NOSs) produce NO using between NO- and ROS-generating systems. Finally, in sect.

L-arginine and molecular oxygen (O2) as substrates. Thus, VI, we will discuss the current challenges and opportunities under hypoxic or anoxic conditions, the generation of NO for the treatment of cardiovascular disease through the reg- via NOS is compromised. However, a number of proteins ulation of NO and ROS levels in pathological conditions. that are involved in oxidative processes at basal oxygen levels can become de facto reductases as oxygen is depleted. The biological role of this transition is particularly promi- II. NITRIC OXIDE GENERATION AND nent in the case of heme- and molybdopterin-containing VASCULAR FUNCTION proteins such as hemoglobin (Hb), myoglobin (Mb), and xanthine oxidase (XO) (185, 575, 578, 862, 880, 945, The generation of sufficient NO levels to regulate the resis- 990). Clinical intervention through these pathways contin- tance of the blood vessels and hence the maintenance of an ues attracting intense research. adequate blood flow is critical to the healthy performance of the vasculature (277, 299, 573, 600, 786). A number of The concept of oxygen-regulated oxidation and reduction mechanisms are involved in both the generation of NO and processes in the metabolism of NO is not only relevant to the response to NO signaling in the vasculature. In this NO generation but also to the scavenging of NO in the section, we will overview the mammalian proteins involved vasculature (FIGURE 1). In this regard, the role of globins in the generation and sensing of NO in the cardiovascular like ␣-Hb and cytoglobin (Cygb) as catalytic NO dioxyge- system. The production of NO in basal conditions is largely nases that scavenge NO is a topic of current research (25, regulated by the activity of endothelial NOS (eNOS) in the 593, 594, 898). vascular endothelium (324, 449, 716). Nevertheless, the contribution of other agents cannot be ignored. For in- The translation of our knowledge about the biology of NO stance, nitrite reduction by heme proteins can mediate hy- and ROS has encountered significant challenges. For in- poxic vasodilation and other physiological responses (367, stance, initial attempts to enhance NO levels using NO 591, 968); neuronal NOS (nNOS) and inducible NOS (iNOS) can provide compensatory NO generation or exac- erbated RNS synthesis (386, 444, 547, 704, 727). In this - NO NO3 study, we will review NO synthases and other NO-gener- ating biological systems. Fe2+-O 2 - NO2 NO A. Oxygen-Dependent Nitric Oxide Synthesis O 2 Fe2+ The canonical pathways of NO formation rely on the spe- cialized NOS enzymes. NOS are dimeric, multidomain en- FIGURE 1. Oxygen and oxidoreductase enzymes regulate nitric oxide (NO) homeostasis. The gradient in the concentration of oxygen zymes that synthesize NO from molecular oxygen and L-ar- shifts the function of globins from oxidizing, NO-scavenging proteins ginine and use iron protoporphyrin-IX (heme), tetrahydro- to nitrite-reducing, NO-generating proteins. biopterin (BH4), FAD, flavin mononucleotide (FMN), and

312 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

NADPH as cofactors (204, 904). The architecture of these 1. eNOS proteins is complex, with a oxygenase/heme domain that binds BH4 and heme, in which the oxidation of arginine to eNOS is the constitutive NOS form in endothelial cells NO using molecular oxygen takes place, and a reductase (ECs), and as such, it is the main contributor to vascular domain evolutionarily related to cytochrome P450 reduc- NO levels in physiological conditions (446). For exam- tase (CYPOR) that binds the cofactors FAD and FMN (204, ple, the infusion of a competitive inhibitor of eNOS into 377). The reductase domain uses NADPH as electron the human systemic or coronary circulation decreases source to reduce the FAD and FMN and finally transfer the basal blood flow by ~25% (146, 722). Basal levels of NO electrons to the oxygenase/heme domain (FIGURE 2). Be- produced by eNOS not only regulate blood flow but ton- tween both domains, there is a calmodulin (CaM) binding ically inhibit platelet activation and the expression of domain that regulates the electron shuttling between the inflammatory adhesion molecules on endothelium. The reductase and oxygenase domains. To add to this complex- role of eNOS in the maintenance of optimal cardiovas- ity, the electron transfer between domains occurs between cular function is also highlighted by the experiments with the reductase domain of one monomer and the oxygenase eNOS-deficient mice. eNOS deletion has some limited domain of the other monomer; thus, only the dimer is able effects on mouse blood pressure and vasodilation, as to generate NO catalytically. adaptive processes increase the production of nNOS and prostaglandins (444, 913). However, eNOSϪ/Ϫ mice The role of NOS enzymes in vascular function and patho- show impairments in angiogenesis and wound healing biology has been extensively studied (277, 299, 569, 790). (563). In the apolipoprotein E (ApoE) knockout (KO) In this study, we will summarize the most relevant concepts atherosclerosis model, addition of the eNOS deletion ac- about the function and regulation of the NOS isoforms in celerates the atherosclerotic process (533). Notably, health and disease states. overexpression of eNOS is also detrimental (708), high- lighting the delicate balance of eNOS function. NOS enzymes comprise three main isoforms: nNOS (NOS I), iNOS (NOS II), and eNOS (NOS III). Among these iso- Although eNOS is predominantly expressed in the endo- forms, eNOS is the enzyme more abundant in vascular en- thelium (TABLE 1), and the ECs are proposed to regulate dothelial cells and has the most significant impact on vas- NO signaling in the vasculature, a number of recent stud- cular function, with a variety of pathologies related with ies using chimeric cross bone marrow transplant mouse eNOS dysfunction. However, the specific roles of iNOS and models have shown that the red blood cell also expresses nNOS in vascular disease are not to be ignored and will be a functional eNOS that contributes in part to systemic also discussed below. blood pressure responses (181, 536, 1025). This erythro-

ACHeme Oxygenase H B 4 domain

CaM binding FIGURE 2. Architecture of nitric oxide synthases FMN (NOS). A: the arrangement of the domains in the NOS Reductase monomer. The oxygenase/heme domain (red) is con- domain NADPH nected to the reductase domain by a flexible linker, FAD containing a calmodulin (CaM) binding sequence. The reductase domain includes a flavin mononucleotide (FMN)-binding domain (orange) that shuttles electrons B Heme NADPH FMN from NADPH/FAD to the heme group and a FAD-con- H4B FAD e– taining domain (yellow) that uses NADPH as an electron NADPHADP HemeHem source. B: the binding of CaM (blue) to NOS promotes FMNMN H B FAD 4 electron transfer from the FMN domain of one mono- mer to the heme domain of the other monomer. C: three-dimensional structure model of NOS. The figure CaM CaM is assembled from the separated structures of the human endothelial NOS oxygenase domain (PDB: NO e– e– L-Citrulline + 4D1O) (574), the CaM binding peptide bound to CaM

L-Arginine + O2 (PDB:2N8J) (740), and the structure of the neuronal NADPHADP Heme NOS reductase (PDB:1TLL) (338). FMNMN CaM FAD H4B

HemeHemem NADPH CaM FMN H4B FAD

L-Arginine + O2 L-Citrulline + NO e– e–

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 313 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

Table 1. Tissue distribution of NOS isoforms in the cardiovascular system

Tissue Cell Types Cellular Location References

nNOS (NOS1) Heart ECs Plasma membrane 114, 126, 599, 841, 977, 1037 Lung VSMCs Caveolae Blood vessels Adventitial fibroblasts Sarcoplasmic Cardiomyocytes reticulum iNOS (NOS2) Heart ECs Plasma membrane 126–128, 521, 525, 604, 688, 698, 867, 903, Lung VSMCs Phagosomes 984, 1064 Blood vessels Adventitial fibroblasts Golgi Cardiomyocytes Mitochondria Macrophages and other leukocytes

eNOS (NOS3) Heart ECs Plasma membrane 126, 181, 283, 358, 518, 819, 878, 1025 Lung VSMCs Lipid rafts Blood vessels Cardiomyocytes Caveolae Erythrocytes Platelets

EC, endothelial cell; eNOS, endothelial NOS; iNOS, inducible NOS; NOS, nitric oxide synthase; nNOS, neuronal NOS; VSMC, vascular smooth muscle cell.

cyte eNOS can contribute to endocrine NO signaling to Because of their impact in eNOS activity, the more relevant modulate both blood pressure and myocardial injury and phosphorylation sites are Thr495, Ser1177 (Ser1179 in bo- appears to be tonically regulated by arginase through the vine eNOS) and Tyr657. Other phosphorylation sites in- availability of the eNOS substrate L-arginine (1044). clude Tyr81, Ser114, Ser615, and Ser633 (TABLE 2).

eNOS is constitutively expressed; a number of posttransla- The activation of eNOS requires Ca2ϩ and CaM; however, tional modifications impact the ability of eNOS to produce changes in resting Ca2ϩ concentrations are not strictly nec- NO, from changes in the NO production rates to, in some essary to regulate NO synthesis by eNOS. In turn, CaM cases, the complete blockade of NO synthesis (296). The rel- affinity to eNOS is regulated via phosphorylation of ative presence of these modifications is critical to enzyme ac- Thr495. Thr495 is located in the CaM-binding site of tivity. General measurements of eNOS protein such as the eNOS, and the phosphorylation of Thr495 impairs CaM determination of monomer/dimer ratio by Western blot or a binding, thus blocking Ca2ϩ/CaM-dependent activation of single phosphorylation site status show a necessarily simplistic the enzyme (741). In resting endothelial cells, Thr495 is view of the activity of eNOS in cells. A deeper analysis of the generally phosphorylated (297). The phosphorylation has main regulatory factors of eNOS activity, including individual been attributed to protein kinase C (PKC) (297, 646), and phosphorylation sites, other posttranslational modifications, the residue is dephosphorylated by protein phosphatase 1 and the levels of the cofactors and substrates, with special (PP1) (646). The equilibrium between phosphorylated

interest on tetrahydropterin/dihydropterin (BH4/BH2) ra- and dephosphorylated Thr495 is mainly modulated by 2ϩ tios and L-arginine/asymmetric dimethylarginine (ADMA) the changes in intracellular Ca levels. Thus, bradykinin concentrations are necessary for a better assessment of the and Ca2ϩ ionophores promote Thr495 dephosphoryla- eNOS function in vivo and the assessment of endothelial tion and eNOS activation. Ser1177 phosphorylation ac- dysfunction conditions. In this section, we will describe the tivates electron flow through the reductase domain and, regulation of eNOS activity by posttranslational modifica- hence, increases NO synthesis in functional eNOS.

tions; the role of the cofactors BH4 and L-arginine and their Ser1177 is located in the C-terminal portion of eNOS in counterparts BH2 and ADMA, and the role of arginases and a helical C-terminal tail element that blocks flavin reduc- L-arginine transport metabolons will be treated in sect. IIIB. tion and regulates the conformational equilibrium of the A detailed study on other regulatory mechanisms, including reductase domain (404, 631). The mechanism is similar protein/protein interactions, shear stress, and other mecha- to that described for nNOS Ser1412 (7, 947). In resting nisms, has been provided by other reviews (59, 296, 299, cells, Ser1177 is usually not phosphorylated. Phosphor- 870). ylation is induced by a variety of signals, and the kinases involved are dependent on the inducing factor (TABLE 2). 2ϩ A) PHOSPHORYLATION. A number of amino acids have been Some agonists like bradykinin and Ca ionophores can shown to be phosphorylated in human eNOS (296, 669). induce Thr495 dephosphorylation via PP1 and Ser1177

314 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

Table 2. Posttranslational modification sites in human eNOS

Site Kinase Effect of Modification References

Phosphorylation Thr495 PKC Impairs CaM binding 297, 407, 646 AMP kinase Decreases NO synthesis rates Constitutive Can cause uncoupling through the reductase domain Ser1177 Akt Activates electron transfer through the reductase domain 74, 236, 321 PKA Increases NO synthesis rates AMP kinase CaMKII Tyr81 Src Slight increase in activity 320, 322 May be involved in Ca2ϩ/CaM sensitivity Ser114 Constitutive 74, 327 Ser615 PKA No change in NO synthesis 74, 647 Akt May modulate protein/protein binding interactions Ser633 PKA No change in NO synthesis 74, 647 PKG Tyr657 PYK2 Decrease in NO synthesis 292 Glutathionylation Cys382 Not determined 160 Cys689 Decrease flow through the reductase domain 162 Decreased NO synthesis Can cause uncoupling through the reductase domain Cys908 Decrease flow through the reductase domain 162 Decreased NO synthesis Can cause uncoupling through the reductase domain

CaM, calmodulin; NO, nitric oxide phosphorylation via activation of CaMKII at the same and the formation of superoxide with diminished NO syn- time. Shear stress activates Ser1177 via PKA-dependent thesis activity. Glutathionylation appears to be a detrimen- phosphorylation. VEGF, insulin, and estrogens promote tal modification caused by oxidative stress conditions (162, Ser1177 phosphorylation via Akt kinase. 522) and is very sensitive to the ratio of oxidized and re- duced glutathione (160). Tyr657 is located in the FMN domain of eNOS in close proximity to the FMN cofactor. The mutation of Tyr657 C) S-NITROSATION. S-nitrosation of eNOS has also been re- (292) causes complete loss of NO synthesis and L-citrulline ported (777). This reaction is also dependent on eNOS my- formation, suggesting a blockade of the intramolecular ristoylation and Ser1177 phosphorylation and involves the electron transfer. Thus, Tyr657 phosphorylation effectively nitrosation of the Zn-binding cysteines (Cys96 and Cys101 inactivates eNOS. The phosphorylation of Tyr657 is cata- in the bovine eNOS) (271, 272). As a consequence of S- lyzed by proline-rich tyrosine kinase 2 (PYK2). This kinase nitrosation, the formation of the eNOS dimer is blocked, is activated in endothelial cells by several stimuli including resulting in the loss of NO synthesis. Denitrosation of angiotensin II, oxidative stress, and insulin (296). The det- Cys96-NO and Cys101-NO restores eNOS activity. Addi- rimental role of this phosphorylation on NO synthesis has tional studies using mutant eNOS Cys96Ser and spurred interest in the pharmacological inhibition of PYK2 Cys101Ser, which cannot be S-nitrosated, indicate that the for the treatment of cardiovascular disease (94, 628, 869, enzymatic activity is not altered by treatment with NO do- 983). nors, confirming a role for the modification of these thiols in the regulation of eNOS activity (271). B) GLUTATHIONYLATION. Recent reports indicate that eNOS can also be modified posttranslationally by S-glutathionylation D) OTHER POSTTRANSLATIONAL MODIFICATIONS. eNOS can be acy- (162). At least three residues have been shown to be suscep- lated at different residues. The myristoylation of the N-ter- tible: Cys689, Cys908 (162), and Cys382 (160) (TABLE 2). minal glycine targets eNOS to the membrane (284), The process can be reversed by glutaredoxin-1 and thiore- whereas the palmitoylation of Cys15 and Cys26 targets doxin (160, 907). The reaction increases eNOS uncoupling eNOS specifically to caveolae (82, 336). These modifica-

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 315 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

tions are not expected to change the intrinsic NOS activity As pointed out, iNOS expression is prevalent in macro- but are important to modulate eNOS localization and phages. In these cells, iNOS induction is necessary for the function. generation of high levels of NO in the phagosome and bac- terial lysis and, therefore, is a basic tool for the innate im- Hyperglycemia can result in N-acetyl glycosylation of mune response (519, 688). However, in pathological situa- Ser1177. This modification results in a nonphosphorylable tions, increased NO synthesis can be deleterious (e.g., in Ser1177 and limits eNOS response to agonists and vasore- macrophages recruited to the atherosclerotic plaque). The laxation (257, 280, 1035). high rate of NO production by iNOS, together with super- oxide formation from iNOS or other sources, can lead to In summary, we want to remark that the generation of NO the production of peroxynitrite (see sect. IVA) (222, 624, by eNOS in optimal conditions, at least as studied in vitro 981, 1034), a very toxic oxidant and nitrating agent (78, with saturating concentrations of coenzymes and substrates 709). Studies in which iNOS deletion was added to the Ϫ/Ϫ and absence of deleterious modifications, is a well-balanced ApoE atherosclerosis model indicate improved cardio- vascular function in the ApoEϪ/ϪiNOSϪ/Ϫ mice compared process with a very limited production of superoxide as a Ϫ/Ϫ side reaction. However, a number of circumstances can lead with ApoE controls, indicating a detrimental role of to changes in the NO generation cycle and trigger the pro- iNOS in atherosclerosis progression (532). duction of superoxide instead of NO. These processes are Finally, it should be noted that iNOS-derived NO can also generally termed “eNOS uncoupling” as the consumption have positive effects in certain conditions. iNOS activation of reducing equivalents by eNOS is no longer coupled to the has been identified as a mechanism of protection against formation of NO and is instead driven to the generation of ischemia/reperfusion damage. This observation appears to superoxide from molecular oxygen. This process is largely be related to a preconditioning effect caused by increased deleterious and has been linked to endothelial dysfunction levels of NO (109, 483, 579, 1009). and other vascular pathologies. eNOS uncoupling will be discussed in sect. IIIB along with other mechanisms of su- 3. nNOS peroxide generation. Similar to eNOS, nNOS is a constitutively expressed form 2. iNOS of NOS activated by CaM binding. NO synthesis rates for nNOS are generally higher than eNOS, but also tightly 2ϩ Unlike eNOS and nNOS, which are constitutively ex- regulated by Ca /CaM (unlike iNOS), and nNOS is also pressed enzymes regulated via CaM binding and posttrans- very susceptible to feedback inhibition by NO (4, 902). This lational modifications, iNOS has a much higher CaM affin- enables nNOS to produce NO in a pulsatile manner instead ity, so it binds CaM at very low Ca2ϩ concentrations, and of generating sustained low levels. These features appear to thus, its activity is not regulated by Ca2ϩ/CaM but mainly be more related to its role in synaptic transmission (811, 812). Although nNOS is commonly found in neurons (120), at the level of gene transcription (169). The kinetic param- it is also found in other tissues including vascular smooth eters of iNOS lead to a catalytic activity that generates muscle cells (VSMCs), adventitial fibroblasts, ECs, and car- higher NO levels than eNOS and nNOS, is less sensitive to diomyocytes (114, 126, 599, 841, 1037) (TABLE 1). Expres- NO-dependent autoinhibition, and generates higher levels sion levels of nNOS in the vasculature are lower than for of other nitrogen species, such as nitrate (816, 904). eNOS; however, there is increasing evidence of important functions for nNOS-derived NO in vascular physiology The expression of iNOS is usually limited to airway epithe- (188). lium and neuronal cells (TABLE 1), where it is highly ex- pressed and active under basal conditions (604, 984, 1064), Early experiments in rodents indicated an important role of and activated macrophages and hepatocytes, where it is nNOS in the regulation of cerebral blood flow (731). Selec- expressed in the setting of inflammatory stimuli (688). tive inhibition of nNOS causes increases in blood pressure and decreased response to acetylcholine in normotensive Although healthy cells in the vasculature do not present rats (139). In eNOSϪ/Ϫ mice, nNOS can be activated by significant levels of iNOS, several pathologic processes shear stress, partially compensating the loss of eNOS in the show increased iNOS activity in blood vessels (386, 704, vasculature (444, 547). Experiments with nNOS KO mice 727). As iNOS can generate higher NO levels than eNOS, indicate increased neointima formation in carotid artery this iNOS activation leads to excess NO and severe impair- ligation and balloon injury models (666), indicating that ment of vascular function. This effect is mediated by several nNOS appears to limit vascular injury independently of pathways including continuous activation of sGC and com- eNOS. To investigate the role of nNOS in atherosclerosis, Ϫ/Ϫ petition for BH4 with eNOS (386). Overall, the excess NO the nNOS deletion was incorporated in mice carrying limits the response of blood vessels to vasodilators and de- the ApoEϪ/Ϫ deletion. The double deletion indicates a pro- creases NO sensitivity (263). tective role for nNOS in the development of atherosclerosis,

316 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

G as the mice carrying both deletions showed accelerated pro- nine (L-NNA) and its precursor N -nitro-L-arginine methyl gression of the disease (534). ester (L-NAME) were characterized as general NOS inhibi- tors and continue to be broadly used in research, especially The effects of nNOS in the human vascular system have L-NAME (FIGURE 3). been demonstrated by studies with nNOS-specific inhibi- tors. These works indicate that NO synthesis from nNOS B) 7-NITROINDAZOLE. 7-Nitroindazole (7-NI) (FIGURE 3) is one has definite roles in the vasodilatory response, including, of the earliest inhibitors showing isoform specificity. Al- but not limited to, microvascular tone and coronary flow though it was shown that all three isoforms can bind 7-NI (498, 843, 844). with very similar affinity (17), in vivo studies showed inhi- bition of nNOS without significant effects in blood pres- Another relevant pathway for nNOS-dependent NO signal- sure, a surrogate of eNOS inhibition (664). It appears that ing not mediated by sGC is the formation of S-nitrosothiols 7-NI may have differential properties in cell permeability, (462). This function has special relevance in brain function with limited uptake in endothelial cells (402). Thus, al- in health and neurodegenerative diseases (686). A more though 7-NI cannot be accurately described as a specific specific description of S-nitrosation and its signaling poten- nNOS inhibitor, it can behave as such in vivo. tial is presented in sect. IID. C) 1400W. N-[3-(aminomethyl)benzyl] acetamidine (FIGURE 4. Pharmacological regulation of NOS enzymes 3) is a specific inhibitor of iNOS (344). It remains one of the most widely used NOS inhibitors in research because of its There have been significant efforts to develop specific NOS cell and tissue permeability. Its selectivity seems related to inhibitors for research and pharmacological purposes (17, the irreversible effect on the faster reacting iNOS, whereas 752). the inhibition on nNOS and eNOS is reversible. A similar effect is observed in N(5)-(1-iminoethyl)-L-ornithine A) NONISOZYME-SPECIFIC NOS INHIBITORS. Early research indicated (L-NIO) (FIGURE 3) (279). that analogs of the substrate L-arginine had inhibitory G properties on the three NOS isoforms. Among these, N - D) GW273629 AND GW274150. These sulfur-substituted acetami- G monomethyl-L-arginine (L-NMMA) and N -nitro-L-argi- dine amino acids (FIGURE 3) are specific inhibitors for iNOS

FIGURE 3. Chemical structure of the nitric oxide synthase (NOS) substrate L-arginine, tetrahydrobiopterin, and selected NOS inhibitors.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 317 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

(16). With safer toxicity profiles than 1400W, GW273629 Nitrate and nitrite can enter the circulation through the and GW274150 have been used in clinical trials for mi- dietary intake of nitrate-rich foods, particularly leafy green graine (437, 965) (NCT00242866; NCT00319137). Al- vegetables (1002). In addition, NO generated from NOS though both compounds were ineffective, it is not clear if enzymes is eventually oxidized to nitrate and nitrite as well. this is due to pharmacokinetic issues (965). Mammals do not possess efficient systems for nitrate reduc- tion, but oral commensal bacteria have enzymatic nitrate E) VAS203. The pterin 4-amino-tetrahydrobiopterin (VAS- reduction pathways able to generate significant amounts of

203) (FIGURE 3) isaBH4 analog that can inhibit all NOS nitrite from nitrate reduction in the saliva (467, 602). Ni- proteins by replacing the BH4 cofactor (1008). VAS203 has trate is not only acquired from dietary sources but is also shown efficacy in the treatment of traumatic brain injury concentrated from the blood into the saliva via the salivary and has been used in phase II clinical trials (897) glands, pumped through the sialin transporter (761). After (NCT02012582). consumption of nitrate-rich foods (beet root, spinach, kale, etc.), the concentration of nitrate and nitrite in saliva can The structural similarities between isoforms (and particu- reach levels as high as 10 mM and 2 mM, respectively (601). larly between eNOS and nNOS) have made the develop- A portion of this nitrite is eventually absorbed into the ment of specific eNOS and nNOS inhibitors particularly circulation. Studies using antiseptic killing of the mouth challenging. Structure-based inhibitors have allowed the microbiome, or systemic NOS inhibition, suggest that development of new inhibitors with improved isoform spec- about half of basal plasma nitrite comes from the salivary ificity (337, 752). Cell permeability and other pharmacoki- reduction of dietary nitrate and the other half from the netic considerations have precluded the clinical use of novel oxidation of NO produced mainly by eNOS (485, 557, inhibitors; however, newer compounds have been devel- 763, 865). NO oxidation to nitrite is proposed to occur via oped; for instance, novel-specific nNOS inhibitors have the oxidase activity of plasma ceruloplasmin (865). been tested in animal studies (29, 254, 255, 1051). The further development of new clinically available specific in- Once nitrite accumulates in the plasma, the reduction of hibitors for NOS, in particular for the iNOS and nNOS nitrite to NO in the vasculature is mediated by several pro- isoforms, is to be expected. teins, including deoxygenated Hb (deoxyHb) (185). In- creased physiological levels of nitrite in plasma from a ni- trate-rich diet can lower blood pressure (554). The effect is B. Nitrite-Dependent Nitric Oxide Synthesis dependent on the generation of nitrite from oral bacteria, as antiseptic mouthwash can eliminate both the increase in circulating nitrite and the blood pressure decrease (485). Despite early indications to the contrary (124, 329), nitrate Subsequent studies have further validated the link between and nitrite have been often overlooked as inert NO oxida- nitrite levels and improved cardiovascular function (486, tion metabolites circulating in plasma and in cells. How- 502). ever, during the last two decades, a novel paradigm of ni- trite as a source of NO, especially in hypoxia, has emerged Numerous metal-containing proteins have been shown to (184, 185, 216, 364, 366, 369, 600, 603, 861, 968). Indeed, catalyze the reduction of nitrite to NO. These proteins gen- it has become increasingly accepted that the routes for the erally contain a heme or molybdopterin cofactor. The pres- production of NO in vivo are not only oxidative (as in ence of many of these proteins in diverse components of the NOS) but also reductive (particularly by nitrite-reducing vasculature, including blood cells, blood vessels, and heart proteins such as globins and molybdopterin enzymes). tissue, makes them relevant for the generation of NO in These pathways can work synergistically to maintain NO vascular biology. In the subsequent sections, we will discuss levels in response to changes in oxygen tension. We foresee the known mechanisms for nitrite reduction that are rele- this theme of synergistic oxidative and reductive pathways vant for cardiovascular function. We also note that, to date, to be potentially relevant to the generation of other reactive existing evidence indicates a role for Hb, Mb, and XO on species in health and disease. biological NO generation, whereas the role of the other possible mechanisms in vivo are still a matter of discussion. A few years after the discovery of the role of NO as the endothelial-derived relaxing factor, increasing evidence 1. Inorganic nitrite reduction supported the presence of nonenzymatic NO generation (89, 605, 1075). These studies and observations of arterial- Along with the role of different proteins in the reduction of to-venous gradients of nitrite in the human circulation in- nitrite, a number of nonenzymatic processes can reduce dicated a potential role for nitrite as a NO source in vivo nitrite to NO in vivo. In general, these systems require con- (369). Subsequent studies have extended this notion to a certed electron and proton donation, which is optimal at more global paradigm that encompasses the role of nitrate lower pH and hypoxic conditions, thus making them par- and nitrite as part of a cycle of NO generation that includes ticularly relevant in ischemic events (1074, 1075). The sim- both enzymatic and nonenzymatic processes. plest mechanism involves the disproportionation of nitrite,

318 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS a process accelerated at acidic pH as it requires the proto- A) Hb. The ability of deoxyHb to reduce nitrite has been nation of nitrite to nitrous acid (HNO2). Two molecules of extensively documented (124, 248, 366, 447). Initial re- nitrous acid, then disproportionate, to form N2O3 and wa- ports by Brooks demonstrated that deoxyHb reacts with ter (Eqs. 1–4) nitrite, generating nitrosyl Hb and methemoglobin, consis- 2NOϪϩ 2Hϩ → 2HNO (1) tent with electron transfer from a ferrous Hb to nitrite 2 2 (124). The studies by Huang et al. (445) confirmed the ϩ → ϩ HNO2 HNO2 N2O3 H2O(2) overall stoichiometry of two molecules of deoxyHb gener- → · ϩ · ating one molecule of nitrosyl Hb and one molecule of N2O3 NO NO2 (3) methemoglobin (Eq. 7), consistent with electron transfer from a ferrous Hb to nitrite and indicated the critical influ- Ϫ ϩ ϩ → · ϩ · ϩ 2NO2 2H NO NO2 H2O(4) ence of pH in the process. By studying the same reaction using alkyl nitrites, which do not show a pH dependence in This process is prevalent in very low pH conditions, as in their reaction rates with deoxyHb, it was established that the stomach (89, 605). In fact, this mechanism has been the reaction of nitrite requires a proton and thus formally shown to be responsible for nitrite-dependent increases in requires nitrous acid (247, 248). This property also makes gastric mucosal blood flow (98) and may relate to the role of the reaction faster in low pH conditions. The overall pro- nitrite in host defense as acidified nitrite is a potent antimi- cess can be written as (Eqs. 5–7) crobial agent (99, 261). 2ϩ ϩ ϩ ϩ Ϫ → 3ϩ ϩ DeoxyHb ͑Fe ͒ H NO2 MetHb͑Fe ͒ NO Several molecules present in cells have been shown to re- ϩ OHϪ (5) duce nitrite to NO. Ascorbic acid (vitamin C) is a widely 2ϩ present cellular antioxidant that can catalyze the reduction DeoxyHb͑Fe ͒ ϩ NO → Hb Ϫ NO (6) of nitrite to NO in vitro (205). This effect is also observed with in vivo infusions of ascorbic acid and nitrite (217). 2DeoxyHb͑Fe2ϩ͒ ϩ Hϩ ϩ NO Ϫ → MetHb͑Fe3ϩ͒ Polyphenols can be assimilated through the diet and, like 2 ϩ ϩ Ϫ vitamin C, can reduce nitrite to NO (325, 733, 834). HbNO OH (7)

Dietary interventions have indicated improved vascular This scheme has been found to apply to virtually all the function linked to the consumption of polyphenols from heme protein–mediated nitrite reduction reactions studied different sources, including cocoa and dealcoholized red to date, including Hb (248, 447), Mb (247, 862), neuroglo- wine (60, 168, 225, 417, 834). Combined use of polyphe- bin (Ngb) (932, 945), Cygb (182, 571, 780), cystathionine nols and nitrite appears to have additive effects in vascular ␤-synthase (CBS) (149, 350), globin X (184), plant and protection (791). The effects of these dietary components cyanobacterial Hb (phytohemoglobins) (905, 946), flavo- can be, in part, due to the interaction with different enzy- hemoglobin (340), heme-albumin (43), cytochrome c (41), matic systems (30, 558, 824), but the link between these protoglobin (40), and microperoxidase-11 (42). compounds and NO formation deserves further investiga- tion (600, 789). The reaction of deoxyHb with nitrite shows an increase in the instantaneous reaction rate as the reaction progresses 2. Heme proteins (447). Studies with T-state and R-state Hb–stabilizing com- pounds have unambiguously identified the reason for this Heme proteins have emerged as critical parts in the gener- change is the shift from T-state to R-state during the course ation of NO under hypoxia. The specific role of globins in of the reaction, as the NO formed in the reaction binds to nitrite reduction and NO homeostasis has been studied in the unreacted deoxyHb to stabilize the R-state. This process recent reviews (39, 770, 931). It should be noted that com- is referred to as R-state autocatalysis. The rate constants for Ϫ Ϫ peting reactions, namely the scavenging of NO by the fer- the reaction of Hb with nitrite change from 0.12 M 1s 1 Ϫ Ϫ rous heme [rate constants in the order of 1.7 ϫ 107 to for T-state Hb to 6.0 M 1s 1 for R-state Hb (TABLE 3) 1.5 ϫ 108 MϪ1sϪ1 (180, 967), Eq. 6] and the rate of NO (447). As the distribution of R-state and T-state Hb in vivo dioxygenation [rate constants in the order of 3.4 ϫ 107 to are regulated by oxygen concentration, this phenomenon 8.9 ϫ 107 MϪ1sϪ1 for mammalian Hb/Mb (264, 341), Eq. has important relevance for the reaction in vivo. In fact, the 8] greatly decrease the amount of bioavailable NO gener- reaction of Hb with nitrite has been modeled mathemati- ated by the reactions of nitrite with heme proteins (931). cally at different oxygen tensions. The combination of a However, nitrite-dependent NO generation in vivo cata- faster rate at higher oxygen, but increased availability of lyzed by Hb and Mb is well documented (183, 185, 365, deoxyHb at low oxygen, leads to bell-shaped dependence of 862, 975); it is very likely that the measurement of this NO the observed rates versus the oxygen concentration with the generation is due to the fact that the high concentrations of maximal rates of NO generation around 50% Hb oxygen Hb and Mb (in the mM range) can overcome the low effi- saturation, consistent with experimental data (367, 368, ciency of the nitrite reduction reaction. 794).

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 319 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

lation, questioning the role of SNO-Hb in physiologic hy- Table 3. Rate constants for the reaction of nitrite with poxic vasodilation (454). Importantly, more recent studies selected deoxygenated heme proteins have shown that the ␤-93 cysteine KO mouse shows more Protein K, M؊1s؊1 Reference cardiac injury in ischemic conditions, suggesting that SNO-Hb may play a more prominent role in cardiac rather Hemoglobin than vascular function (1060). Human (T-state)a 0.12 447 Human (R-state)a 6 447 With regards to the nitrite reduction hypothesis, human Myoglobin studies show a significant gradient of nitrite from the arte- Horse hearta 2.9 945 rial to venous circulation, concomitant with the production Sperm whalea 5.6 945 of NO (detected as iron-nitrosyl Hb) in the venous circula- tion (185, 369). In vitro experiments in isolated aortic rings Neuroglobin demonstrate that nitrite mediates vasodilation in conditions Human (S-S)a 0.12 945 of Hb deoxygenation (185, 196). Consistent with these ex- Human (SH)a 0.062 945 periments, infusions of physiological nitrite concentrations Cytoglobin mediate vasodilation in humans, which is enhanced during b Human 0.14 571 exercise and not affected by NOS inhibition. This effect is c Human 1.14 182 accompanied by iron-nitrosyl Hb formation, consistent Cystathionine-␤-synthase with nitrite reductase chemistry (185). Humand 0.6 350

a100 mM sodium phosphate, pH 7.4, 25°C; b100 mM sodium A central controversy relevant to all theories of erythrocytic phosphate, pH 7.0, 25°C; c100 mM phosphate, pH 7.4, 37°C; NO transport is the question of how NO generated can d 100 mM HEPES, pH 7.4, 37°C. escape the red cell without reacting with Hb. Although theoretical calculations suggest that this type of NO escape should be limited, accumulating studies by many research B) PHYSIOLOGICAL IMPLICATIONS OF Hb-DEPENDENT NITRITE REDUC- groups now demonstrate the production of bioavailable TION. The functional implications of the Hb-dependent ni- NO generated from the incubation of erythrocytes with trite reductase reaction remains a subject of intense re- nitrite (185, 196, 883, 975). The production of NO gas has search, particularly in the context of viable physiological been measured by chemiluminescence in vitro from the re- models of erythrocytic NO transport (366, 424, 591). action of deoxygenated Hb, Mb, or erythrocytes with ni- There is consensus in the field that NO-erythrocytic inter- actions regulate vascular function, particularly that oxygen trite. More physiological biosensor experiments demon- desaturation of red blood cell Hb stimulates increased vas- strate that incubation of nitrite with erythrocytes can in- cular NO bioavailability, leading to NO-dependent hy- duce cGMP production and inhibit activation in platelets poxic vasodilation (266). However, the molecular mecha- coincubated with this reaction (591, 883, 975). The mech- anism for NO release remains unclear but has been postu- nism of this regulation is more contentious. Three models ϩ by which this erythrocytic regulation of NO production lated to involve the formation of NO (684) or N2O3 as occurs have been proposed: 1) hypoxic release of ATP from intermediates (69). Additionally, NO escape may relate to the red blood cell (882) that stimulates endothelial NO the localization of its formation, particularly at the surface production by binding to endothelial purinergic receptors, of the erythrocyte (813) or via concerted reactions of NO 2) the S-nitrosation of cysteine 93 of the ␤-chain of Hb and autoxidation of oxygen to form NO and NO2 (N2O3) (SNO-Hb) (866), and 3) the reduction of nitrite by de- at partial oxygen saturations (i.e., the reactions of nitrite oxyHb (as described above) (425). with both oxygenated Hb and deoxyHb at around 50% Hb oxygen saturation) (384). More work is needed to under- Understanding the role of each of these potential mecha- stand the biophysics of NO signaling from the erythrocyte nisms is an ongoing area of interest. The SNO-Hb hypoth- during nitrite reactions with deoxygenated red blood cells esis proposes that SNO-Hb is formed when Hb is oxygen- and Hb. ated (R-state) in the lungs and remains stable. Once Hb becomes deoxygenated (T-state), the SNO-Hb reacts with C) Mb. The physiological relevance of the reaction of Hb with existing thiols to release a vasodilatory signal (471, 728, nitrite suggested that similar processes could involve the 886). In the last 25 years since this proposal, numerous closely related globin, Mb. The ability of Mb to catalyze groups have debated key elements of the hypothesis includ- nitrite reduction to NO was first confirmed in vitro. Con- ing the mechanism of SNO-Hb formation and release and sistent with the monomeric structure of Mb and its lack of physiological levels of SNO-Hb. Additionally, a genetic allosteric regulation, the reaction rate is constant through murine model in which the cysteine 93 of ␤-Hb was re- the reaction, with a bimolecular rate constant similar to that placed with alanine (and in which SNO-Hb formation was of R-state Hb (TABLE 3) (447). In vitro studies of isolated abolished) demonstrated no impairment in hypoxic vasodi- mitochondria in the presence of Mb and nitrite have shown

320 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS that Mb-dependent NO is bioavailable and can inhibit mi- species is more abundant. Thus, both processes can syner- tochondrial respiration in a similar manner to a conven- gize to regulate the concentration of NO (593, 594). The tional NO donor (862). In the cardiovascular system, the presence of an efficient reduction system for Cygb in heart is the most relevant organ for the function of Mb as a smooth muscle cells via cytochrome b5 and cytochrome b5 nitrite reductase. In a murine myocardial infarction (MI) reductase further supports the possible relevance of these model, nitrite has been shown to decrease infarct size, and processes in vivo (25, 593). this effect was lost in Mb KO mice, which cannot reduce nitrite to NO (774). Through this mechanism, cardiac Mb is F) CBS. CBS is a pivotal enzyme for the metabolism of homo- thought to be critical for the reduction of circulating nitrite cysteine (649). Mutations in the CBS gene are associated accumulated through diet or through the therapeutic pro- with hereditary homocystinuria (531). CBS binds heme and cess of remote ischemic preconditioning, which mediates 5=-pyridoxal phosphate. The 5=-pyridoxal phosphate group cardioprotection (267, 775). is critical for the reaction of homocysteine with serine or cys- teine, whereas the role of the heme domain in the catalysis of D) Ngb. Ngb is a recently discovered six-coordinate globin CBS in unknown, but this heme appears to be functional and that is evolutionarily related to Hb and Mb (134). The redox active (868). The CBS-mediated reduction of nitrite to physiological function of Ngb is unknown, although be- NO by the CBS heme has been recently reported (149, 350). cause of its generally low concentration in cells (with the The reaction proceeds with rate constants similar to these of exception of the retina in the eye), it is probably not related T-state Hb, Ngb, or Cygb (TABLE 3) (149, 350). The presence to oxygen transport and storage as Hb and Mb are (38, 39, of CBS in endothelial cells (809) suggests that CBS could play 133). It was recently demonstrated that, as observed with a role in vascular nitrite reduction in vivo. other globins, deoxygenated Ngb can also reduce nitrite to NO (932, 945). Furthermore, this process is redox regu- G) NOS. As NOS proteins contain a heme group, it is conceiv- lated by the redox state of two surface thiols. Thus, oxida- able that they can catalyze the reduction of nitrite as ob- tion of two cysteine residues to form an intramolecular served in other heme enzymes. Indeed, in anoxic conditions, disulfide bond doubles the rate of nitrite reduction (TABLE eNOS has been shown to catalyze nitrite reduction to NO 3). This redox transition occurs in a physiologically relevant (970). The contribution of eNOS to nitrite reduction could range and can be controlled by the cellular GSSG/GSH ra- be significant in specific tissues, including red blood cells tios (945). It appears that Ngb is not present in vascular and kidney (654, 998). cells, except for sympathetic nerves (898). The reaction of Ngb with nitrite may have implications for microcircula- 3. Molybdoproteins tion in the brain, where several studies have found a vaso- protective effect of Ngb (140, 503, 917). The relevance of molybdopterins in the generation of NO in the vasculature has been increasingly appreciated since this E) Cygb. Like Ngb, Cygb is a six-coordinate globin discovered in reaction was first described for XO (650, 1061). As the the early 2000s (132, 493, 951). Cygb is ubiquitous in human molybdopterin group does not react directly with oxygen or tissues, mainly found in fibroblasts and cells of related lineage NO, these reactions that decrease NO release from heme such as osteoblasts and chondroblasts, but also in other cell proteins do not limit Mo-dependent nitrite reduction reac- types, including neurons (403, 687). Interestingly, it has been tions. Thus, the reduction rates by molybdoproteins can be identified in VSMCs (398, 571, 594). Cygb can reduce nitrite relevant in vivo at lower rate constant values as compared to NO at rate constants similar or slightly higher than Ngb for heme protein reduction rates. The reduction of nitrite by (182, 571) (TABLE 3). In addition, the oxygenated form of all four mammalian molybdenum-containing enzymes has Cygb reacts with NO to form nitrate in a NO dioxygenation been characterized. Recent reviews on the nitrite metabo- reaction that scavenges NO (339) (Eq. 8) lism by molybdenum enzymes are available (614, 616). 2ϩ Ϫ ϩ → 3ϩ ϩ Ϫ Fe O2 NO Fe NO3 (8) A) XO. Mammalian molybdopterin-containing enzymes cata- lyze different oxidation-reduction reactions including, but This reaction is common to most heme proteins (264, 341, not limited to, xanthine oxidation, sulfite oxidation, and 342, 931). In practice, this indicates that Cygb expression drug metabolism processes (839, 840). To our knowledge, can regulate the diffusion of NO and possibly prevent ex- the ability of mammalian Mo-containing enzymes to reduce cessive vasodilation at high levels of NO, not unlike the nitrite to NO was first described for XO (370, 578, 650, proposed role of ␣-Hb in vascular endothelial cells (898). 1061). Earlier work had shown the ability of XO to also This role has been proposed for Cygb in the vascular endo- reduce nitrate to nitrite (50, 308). Subsequently, several thelium (398, 593, 594). It should be noted that the scav- reports have shown that all mammalian Mo-containing en- enging of NO is more efficient at high O2 tensions, which zymes [XO, aldehyde oxidase (AO), sulfite oxidase, and the favor the formation of the ferrous/oxygenated heme spe- mitochondrial amidoxime-reducing component (mARC)] cies, whereas nitrite reduction would be prevalent when the are able to reduce nitrite at different intrinsic rates (TABLE 4) oxygen concentration is low, and the ferrous/deoxygenated (575, 880, 990).

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 321 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

B) AO. AO is a molybdenum-containing protein with high Table 4. Kinetic parameters for the reaction of nitrite with sequence and structural similarity to XO. Its function is yet selected molybdopterin proteins unknown, although it has been related to the metabolism of

-؊1 ؊1 ؊1 retinoic acid, neurotransmitters, and a variety of xenobiot Protein kcat,s KM,mM K,M s Reference ics (513, 756, 920). AO has been shown to reduce nitrite to Xanthine oxidase NO, albeit a slower rate than XO (TABLE 4) (568, 575, 617, Human, pH 7.4a 0.41 2.2 186 616 1001). As observed for XO, the pH dependence of nitrite Human, pH 6.3b 1.2 0.67 1800 616 reduction shows a bell-shaped pattern with a maximal rate Rat, pH 7.4a 0.55 1.9 289 616 around pH 6.5 (617). The relevance of AO-dependent ni- Rat, pH 6.3a 0.58 0.25 2300 616 trite reduction has not been studied in so much detail as for XO, but depending on their tissue abundance and the avail- Aldehyde oxidase ability of their reducing substrates, some studies suggest Human, pH 7.4a 0.47 4.1 115 616 that the magnitude of AO activity in the vasculature can be Rat, pH 7.4a 0.67 3.6 186 616 similar to XO (568, 745, 1073). Rat, pH 6.3a 0.66 0.43 1500 616 Sulfite oxidase C) SULFITE OXIDASE. The molydopterin-containg sulfite oxidase c Human, pH 7.4 0.002 1.6 1.3 990 catalyzes the oxidation of sulfite to sulfate, preventing the c Human, pH 6.5 0.004 1.7 2.4 990 accumulation of toxic levels of sulfite. Impaired sulfite oxi- mARC-1 dase function leads to severe neurological damage and Human, pH 7.4d 0.1 9.5 11 880 death (510). Like XO and AO, the molybdopterin cofactor aReaction contains 50 ␮M of reductant (aldehyde); breaction con- of sulfite oxidase is able to catalyze nitrite reduction to tains 750 ␮M of reductant (aldehyde); creaction contains 5 ␮Mof produce NO (TABLE 4) (990). Unlike other molydopterin reductant (sulfite); dreaction contains 1 mM NADH, 0.2 ␮M cyto- proteins, in sulfite reductase only the Mo4ϩ center, and not chrome b reductase and 2 ␮M cytochrome b . ϩ 5 5 the Mo5 species, is able to catalyze the reaction (990). The

reaction has a marked O2 dependence with very low NO formation in the presence of O2. This is most probably Although the architecture of these enzymes is variable, ex- related to the fast oxidation of the Mo4ϩ species to Mo6ϩ isting evidence indicates that the reduction of nitrite takes by molecular oxygen as observed in other molybdopterin place in the molybdopterin site, where Mo reduces nitrite to proteins (880, 990). NO in a single electron transfer reaction similar to that of the heme proteins (615, 616) (Eqs. 9 and 10) D) mARC 1 AND 2. Two isoforms of the mARC are expressed in 4ϩ ϩ ϩ ϩ Ϫ → 5ϩ ϩ ϩ Ϫ mammals (705). Sequence homology suggests that mARC Mo H NO2 Mo NO OH (9) proteins belong to the sulfite oxidase family. The structure 5ϩ ϩ ϩ ϩ Ϫ → 6ϩ ϩ ϩ Ϫ Mo H NO2 Mo NO OH (10) of the mARC proteins has not been elucidated, but electron paramagnetic resonance data seem to support these similar- Depending on the redox potential of the enzyme, nitrite ities (1043). mARC proteins have a not yet understood reduction may occur by either of the two reactions or only physiological function. Existing evidence suggests that they ϩ by Mo4 (Eq. 9) pathways or one specific pathway (614, may be involved in the detoxification of N-hydroxylated 616). The pH dependence of the reaction is not linear as in substrates; mARC enzymes are required for the detoxifica- the case of the heme-dependent reactions, probably because tion of some hydroxylamines (706). mARC can also cata- of the protonation of important catalytic residues at low pH lyze the reduction of the NOS reaction-intermediate NG- that offsets the effect of the increased proton concentration. hydroxy-L-arginine to L-arginine in a reaction that may be These circumstances lead to a maximal rate for XO-depen- involved in NO biosynthesis (528). dent nitrite reduction around pH 6.3 (TABLE 4) (617). As noted above, unlike nitrite reduction by heme, the Mo co- Both mARC enzymes can reduce nitrite to NO in a process factor does not bind NO. Thus, this reaction potentially has very sensitive to oxygen-mediated inhibition (TABLE 4)

a higher yield of free NO. (880). As mARC enzymes can use the cytochrome b5 reduc- tase/cytochrome b5 system as a source of electrons, the three In the vasculature, XO has been identified in the surface of proteins can form a mitochondrial metabolon for the reduc- epithelial cells, endothelial cells (5, 798, 979, 1010), and tion of nitrite to NO under hypoxic conditions (880). erythrocytes (357, 998). To date, XO is the most relevant nonheme nitrite reductase in vivo. Many studies have 4. Other proteins shown the relevance of XO in mammalian physiology (357, 568, 577, 578, 998). The use of specific XO inhibitors such A) CARBONIC ANHYDRASE. The reduction of nitrite by carbonic as allopurinol and oxypurinol has allowed the determina- anhydrase has been described (1). Unlike heme- or molyb- tion of specific XO effects on nitrite reduction and NO denum-containing proteins, the Zn2ϩ in the active site of metabolism (357, 568, 575, 998). the carbonic anhydrase is not redox active, thus invoking a

322 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS nitrite anhydrase mechanism as described in Eqs. 1–4. Such C. The NO Receptor sGC and the reaction would be of notable interest in vascular physiology Modulation of Vascular Tone because of the ubiquitous presence of carbonic anhydrase in erythrocytes; however, this activity remains highly contro- 1. Structure and function of sGC versial (615). The effects of NO in the vasculature at concentrations in the B) CYTOCHROME C. The electron carrier cytochrome c has also nM/pM range (397) are mainly mediated through the acti- been noted as a situational nitrite reductase. As cytochrome vation of the canonical NO receptor sGC (223, 662, 711, c is a six-coordinate heme protein with no distal site avail- 751). sGC is a cytosolic, heterodimeric protein comprising able for nitrite ligation, the reaction is only observable in two homologous subunits, ␣ and ␤. Although at least two conditions where the protein is partly unfolded, a situation isoforms for either ␣ and ␤ subunit exist (termed ␣1, ␣2 and that can be elicited by cardiolipin or other anionic phospho- ␤1, ␤2) with different tissue distributions (129), the ␣1␤1 lipids (477). In the presence of cardiolipin-containing lipo- heterodimer is the most common form. Each monomer con- somes, cytochrome c has been shown to catalyze nitrite tains four domains: an N-terminal heme-binding domain, a reduction to NO in a reaction that may be prevalent in Per-Arnt-Sim (PAS) domain, a helical/coiled coil motif, and apoptotic processes (68). a C-terminal, guanylyl cyclase domain that catalyzes the formation of cGMP from GTP (FIGURE 4). It should be C) CYP AND CYPOR. The heme P450 cytochromes (CYPs) are noted that despite the homology between the N-terminal involved in a wide range of detoxification processes. CYP domains, only the N-terminal domain of the ␤ subunit 2B4 and microsomal rat liver CYP fractions have been char- binds heme (489). The heme domain in the ␤ subunit is the acterized as nitrite reductases (576). Notably, the associ- NO-sensing center of sGC. Remarkably, unlike most other ated CYP-reducing protein, CYPOR, can reduce nitrate to hemoproteins that bind oxygen, CO and NO, this heme nitrite, forming a metabolon that can tentatively catalyze group shows a high selectivity toward NO and can also the complete process from nitrate to NO (576). bind CO weakly but shows no affinity toward molecular

H-NOX PAS Helix GTP Cyclase -subunit

-subunit HemeHeme

GTP Cyclase Domain FIGURE 4. Architecture of soluble guany- lyl cyclase (sGC). Top: arrangement of sGC domains in the sequence of ␣ and ␤ sub- units. Bottom: model for the interaction of the N-termini domains of the ␣ and ␤ sub- units of human sGC. Domains are colored according to the top panel. Catalytic GTP cyclase domains are not shown. The model for the human sGC protein is based on the model for Manduca sexta sGC derived from chemical cross-linking, small-angle X-ray scattering, and homology modeling (312, 313, 662).

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 323 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

oxygen (625). When NO binds to the heme, changes in the In addition to the presence of heme-free protein, it is in- heme coordination cause a conformational change in the creasingly appreciated that a pool of ferric sGC can also N-terminal heme domain that relieves an inhibitory inter- exist in smooth muscle cells (641, 769, 891). As noted, the action between the heme domain and the catalytic C-termi- oxidation of the heme iron is probably the intermediate step nal domain, activating cGMP synthesis (1017). The exact in the generation of free-heme sGC (312, 363, 888). The details of the conformational changes that mediate enzyme generation of ferric sGC is one of the mechanisms of sGC activation are not yet known and are the focus of ongoing desensitization, and this form of the protein, whereas non- structural studies (145, 313, 958). responsive to stimulator drugs, can be rescued by sGC ac- tivators (302, 891). Recent work indicates that CYB5R3 The activation of sGC by NO leads to an increase in cGMP (methemoglobin reductase) may mediate the reduction of synthesis of at least two orders of magnitude (895). cGMP ferric sGC to NO-responsive ferrous sGC in cells (769). exerts a number of downstream signaling effects on phos- These findings open the possibility of novel pathways to phodiesterases such as PDE5 and other targets including regulate sGC responses in blood flow regulation and for cGMP-dependent kinases and cGMP-gated ion channels, targeted therapy in diseases associated with oxidative stress eliciting a vasodilatory response (673, 674, 993). and endothelial dysfunction.

Apart from changes in NO levels or downstream signaling, Posttranslational modifications of sGC have been also de- scribed. Prolonged exposure to NO has been long known to the activity of sGC itself is compromised in a number of cause a decrease in sGC activity, usually characterized as pathological conditions (355, 363, 600, 853, 888, 1022). sGC desensitization (85, 671, 831). This process is related Situations directly related to sGC that can account for a to some of the tolerance profiles observed for NO donor decrease in sGC activity include changes in the expression treatments (459). sGC desensitization is probably due to of sGC, defective incorporation or loss of sGC heme, a different concurrent mechanisms, including S-nitrosation deficient reduction of the sGC heme iron, leading to an NO and heme iron–nitrosylation. Recent reports indicate that unresponsive ferric enzyme, posttranslational modifica- several cysteine residues in sGC can undergo S-nitrosation, tions, ubiquitination and proteosomal degradation, and de- leading to decreased activity (93, 822, 823). The formation fects in cGMP synthesis. of a heme iron–nitrosylated form of sGC, which has de- creased guanylyl cyclase activity, has been reported (953). Changes in sGC related to the mRNA transcript have been Ubiquitination of sGC targets the protein for proteosomal ␣ described. For example, expression of a variant 2 subunit degradation, a process that can be inhibited by sGC activa- ␣ ( 2i) containing a 31 amino acid insertion has been shown tors (643). Further research on these sGC modifications, in different tissues. This subunit can form a dimer with ␤1 and interventions to prevent their occurrence, is ongoing but produces an inactive sGC form (80). Alternatively, sev- (92). eral splice variants of the ␣1 and ␤1 monomers have been identified in vivo (626, 787, 852). These splice forms can be 2. Pharmacological regulation of sGC related to decreased sGC activity and appear to be more abundant in disease conditions such as aortic aneurysm Because of the pivotal role of sGC in the regulation of the (626). NO response, substantial efforts have been made to develop pharmacological regulators of sGC function. In theory, di- The incorporation of the heme group in the sGC ␤1 mono- rect targeting of sGC offers notable advantages, bypassing mer is regulated by the chaperone heat shock protein 90 the complex mechanisms that may limit NO generation and (Hsp90) (95, 354, 356, 818). This process is also regulated availability under pathological conditions (FIGURE 5). Two by NO, with low NO doses improving Hsp90/sGC interac- main groups of compounds, activators and stimulators, tion and heme insertion, whereas high NO blocks heme have been developed with different mechanisms of action. insertion (354, 985). The relevance of heme-free sGC in the In both cases, the target of the drugs appears to be the context of cardiovascular disease is not well characterized. NO-sensing, heme domain of the ␤ subunit (662). A large Several lines of evidence suggest the presence of pools of number of sGC agonists have been developed over the last heme-free sGC in vivo (354, 356, 799, 943), and it is rea- three decades; we discuss below a few of the more widely sonable to expect that the exacerbated ROS conditions that studied compounds. For a more complete view of the devel- exist in the origin and development of many cardiovascular opment of these and other sGC-related drugs, the reader is pathologies can promote heme oxidation and subsequent referred to more specific reviews (274, 302, 888). heme loss (312, 363, 888). Thus, a plausible therapeutic strategy can target the inactive, heme-depleted sGC by use A) STIMULATORS. Stimulators are compounds that enhance the of agonists (activators) that can bind to the empty heme site activity of functional sGC and require the presence of fer- activating the enzyme. Some examples of these drugs are rous (Fe2ϩ) heme-bound sGC. These compounds can coop- discussed in the next section. erate with endogenous NO-mediated activation.

324 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

Normal Endothelial dysfunction

Blood flow

Endothelial cells L-arginine L-citrulline L-arginine L-citrulline

eNOS OxidativeOxidative sstresstress eNOS

BH4 BH eeNOSNOS uuncouplingncoupling 2 eNOS eNOS

•- NO O2

ONOO •- sGC Fe (II) heme sGC Fe (III) heme (Reduced) Oxidation and heme loss (Oxidized)

GTP cGMP Vasodilation

Smooth muscle cells

FIGURE 5. Soluble guanylyl cyclase (sGC) function in healthy and endothelial dysfunction states. Oxidative

stress conditions cause oxidation of BH4 to BH2, superoxide production in the endothelial cells, and promote oxidation and heme loss in smooth muscle cell sGC.

The benzyl indazole YC-1 (FIGURE 6) was the first devel- the treatment of pulmonary arterial hypertension and oped sGC stimulator (524). The effects of YC-1 on sGC chronic thromboembolic pulmonary hypertension (351, activation were found to be synergistic with NO donors 352). and required heme-bound sGC (310, 436, 896). The compound presented some limitations and a lack of spec- Another second-generation stimulator compound is BAY ificity with cGMP-independent effects and inhibition of 1021189 (vericiguat) (301) (FIGURE 6), which has completed PDE5 (309, 326, 991). Based on YC-1 structure, a num- Phase IIB trials (287, 349, 744) and is currently in Phase III ber of optimized compounds have been developed such as clinical trials for the treatment of heart failure with reduced BAY 41–2272 and BAY 41–8543 (889). These com- ejection fraction (301) (NCT02861534). pounds are more specific than YC-1 and despite some poor pharmacokinetic properties, have been extensively B) ACTIVATORS. sGC activators are compounds that can trigger used in research (105, 353, 670, 760, 764, 852, 889). the catalytic activity of sGC independently of the redox state of the sGC heme or even in the absence of bound heme. Further studies led to the development of another YC-1– In general, these compounds work as heme analogs, replac- related compound, BAY 63–2521 (riociguat) (83, 655) (FIG- ing the heme group and triggering a similar conformational URE 6). Riociguat (commercialized as Adempas) was ap- change to that of NO-bound heme in the native enzyme, proved in 2013 by the Food and Drug Administration for thus activating cGMP formation (302).

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 325 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

ample, as described above in sect. IIB2, S-nitrosation of Hb is an active area of research and SNO-Hb has been consid- ered a reservoir of NO activity. In this regard, decreased levels of SNO-Hb have been reported in conditions such as pulmonary hypertension and hypothesized to contribute to disease pathogenesis (586, 729).

Accumulating studies demonstrate that beyond simply rep- resenting a reservoir of NO activity, S-nitrosation is a mech- anism of enzymatic regulation. S-nitrosation of specific cys- teine residues can modulate enzymatic activity and func- tion. One prototypical example of this paradigm is the type 2 ryanodine receptor (RyR2). The RyR2 is a Ca2ϩ release channel that releases Ca2ϩ from the sarcoplasmic reticulum to mediate cardiac excitation/contraction coupling. The magni- tude and duration of Ca2ϩ release from through the RyR2 determines contractility of the myocyte. Although the RyR2 contains many surface-exposed thiols, physiological S-nitrosa- tion of specific cysteine residues activate the protein and con- FIGURE 6. Chemical structure of selected soluble guanylyl cyclase tribute to maintainance of healthy excitation contraction cou- (sGC) stimulators and activators. pling (1038). A lack of this S-nitrosation has been associated with cardiac arrhythmias as well as heart failure (131, 372, 373). Similar to the RyR2, a multitude of enzymes have now High-throughput screening assays led to the discovery of a been shown to be activated or inhibited by S-nitrosation. Al- novel series of sGC regulators that do not require the sGC though review of all these proteins is beyond the scope of this heme. The first developed drug in this category was the manuscript, the S-nitrosation of mitochondrial complex I and compound BAY 582667 (cinaciguat) (890) (FIGURE 6). its cardioprotective signaling is discussed in sect. IIIC3, and the Apart from the recovery of sGC synthesis, it was noted that S-nitrosation of eNOS as a regulator of its activity is outlined BAY 582667 could limit the degradation of heme-oxidized in sect. IIA1. The role of S-nitrosation in regulating cardiovas- and heme-free sGC (643). Cinaciguat showed promising cular function has been reviewed elsewhere (586, 677, 914). results in animal studies and acute decompensated heart failure (553), but the trials were stopped because of the In the following section, we review the chemical mecha- development of low blood pressure (269). nisms by which S-nitrosothiols are formed. It is important to note that physiological levels of S-nitrosothiols are deter- Another sGC stimulator that has reached clinical use is mined not only by the rate of S-nitrosation but also by HMR 1766 (ataciguat) (827) (FIGURE 6). Phase II clinical protein denitrosation. Although S-nitrosothiols degrade trials are currently evaluating the effect of ataciguat on through transnitrosation reactions (outlined below) and in- aortic valve calcification (NCT02481258). teraction with other low molecular–weight thiols, the exis- tence of denitrosating enzymes has been reported. The best D. S-Nitrosothiols and the Modulation of characterized enzyme in this class is S-nitrosoglutathione Vascular Function reductase (GSNOR), a member of the aldehyde dehydroge- nase enzyme group, which catalyzes the denitrosation of 1. S-nitrosation and vascular function glutathione and thus regulates the equilibrium of S-nitrosa- tion (592). Although activation of sGC is considered the predominant mechanism of NO-dependent signaling, it is now recog- 2. Formation of S-nitrosothiols nized that posttranslational modification of cysteine resi- dues by S-nitrosation comprises a significant alternative The formation of S-nitrosothiols is, on occasion, described mechanism of NO signaling. The first protein shown to be as the reaction of NO with a thiol (R-SH) group to generate S-nitrosated was albumin (885), and its existence was first a protein-bound nitrosothiol (R-SNO). However, it should proposed to represent a reservoir of NO bioactivity. How- be noted that NO is not a nitrosating species and does not ever, S-nitrosated albumin is now considered to be a prod- react directly with thiols. Formation of S-nitrosothiols in- uct of more detrimental NO “scavenging” by the protein volves the reaction of the deprotonated thiol (R-SϪ) and a (332, 956). It is now well recognized that a plethora of formal nitrosonium ion (NOϩ)(Eq. 11) proteins of various function and localization are S-nitro- sated physiologically or in pathological conditions. For ex- R-SϪ ϩ NOϩ → R-SNO (11)

326 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

In physiological conditions, the thiolate is generally pro- Finally, the reaction of an existing S-nitrosothiol with a vided by glutathione or cysteine, yielding GSNO or Cys- thiolate group can result in transnitrosation (Eq. 17) NO, respectively. The nature of the nitrosating species can R-SNO ϩ R’-SϪ ↔ R-S- ϩ R’-SNO (17) vary, generating different possible mechanisms for the reac- tion. Most of these routes require the formation of the ni- · Transnitrosation is a fundamental reaction of S-nitrosothi- trogen dioxide radical intermediate (NO2 )(Eqs. 12a, 12b) ols and allows for the formation of GSNO or S-nitrosated · ϩ ↔ · NO O2 O2NO (12a) proteins to mediate the formation of other S-nitrosothiols O NO· ϩ NO· → 2NO · (12b) and probably serve as a reservoir for S-nitrosothiols in the 2 2 cellular environment. Altogether, we want to note that the formation of the S-nitrosothiols is a complex process, and The reaction of NO · with a thiolate in the presence of NO 2 existing evidence indicates the requirement of significant can generate the S-nitrosothiol via a thiyl (RS·) radical (Eqs. concentrations of NO and usually the presence of superox- 13a, 13b) ide and/or peroxynitrite. · ϩ Ϫ → · ϩ Ϫ NO2 R-S R-S NO2 (13a) The formation of S-nitrosothiols and their functional roles R-S· ϩ NO· → R-SNO (13b) in vivo is a growing field, with protein S-nitrosation identi-

· · fied in most physiological pathways. It should be considered The reaction of NO2 with another molecule of NO2 or NO that the stability and precision of this modification is under generates dinitrogen tetroxide (N2O4) or dinitrogen triox- intense study to determine its parallels with other protein ide (N2O3), respectively, which are strong nitrosating spe- modifications such as phosphorylation (1023, 1024). Re- cies and can react directly with the thiolate (691, 692, 1018) cent studies of the proteome of eNOSϪ/Ϫ mice compared (Eqs. 14a–d) with wild type mice identify a more limited set of S-nitro- · ϩ ↔ NO2 NO N2O3 (14a) sated proteins (125, 378). Altogether, it appears that the detection of S-nitrosated proteins in the presence of exoge- NO · ϩ NO · ↔ N O (14b) 2 2 2 4 nous NO or R-SNO species can lead to nonphysiological ϩ Ϫ → ϩ Ϫ N2O3 R-S R-SNO NO2 (14c) S-nitrosation and more sensitive methods, monitoring pro- N O ϩ R-SϪ → R-SNO ϩ NO Ϫ (14d) tein S-nitrosation levels in vivo are necessary for accurate 2 4 3 detection of these modifications. Peroxynitrite (sect. IVA), formed by the reaction of NO and · superoxide, is another source of NO2 , either through ho- III. SUPEROXIDE AND HYDROGEN · molytic decomposition, via peroxynitrous acid, into NO2 PEROXIDE GENERATION AND · and OH radicals (Eqs. 15a–b) or via reactions with CO2 VASCULAR FUNCTION (Eq. 15c) or metal centers (Eq. 15d) ONOOϪ ϩ Hϩ → ONOOH (15a) The presence of ROS in the vasculature has historically been considered detrimental and a consequence of abnormal → · ϩ · ONOOH NO2 OH (15b) generation and/or an inability of the endogenous reductant Ϫ ϩ → · ϩ Ϫ· and antioxidant systems to scavenge these reactive species. ONOO CO2 NO2 CO3 (15c) The excess of superoxide, or other ROS, in the cell causes Ϫ ϩ nϩ ϪϾ · ϩ (nϩ1) ϭ ONOO M NO2 M O(15d) the modification of different parts of the cellular machinery and is a known player in the genesis and progression of · These reactions leading to the formation of NO2 are well cardiovascular disease (227, 408, 409, 570, 672, 700, 893). characterized (123, 548, 585, 1018). Global kinetic analy- Conversely, the signaling properties of many of these mol- sis of the reactions involved suggests that the main source of ecules are increasingly recognized. Indeed, the generation of · NO2 in vivo may be the decomposition of peroxynitrite superoxide and hydrogen peroxide, among others, has been (585). This result is consistent with the unfavorable rates shown to mediate critical processes such as angiogenesis, · for the direct formation of NO2 from oxygen and NO in hypoxic adaptation, and energy homeostasis (96, 249, 291, vivo, (1018) although the exact mechanisms of formation 720, 778, 825, 960). In this section, we will discuss the main remain a topic of current research (123, 872). sources of superoxide and hydrogen peroxide generation in the vasculature and their endogenous and pharmacological An additional pathway involves the formation of a ferric regulation. Among these, NADPH oxidases (NOXs) ap- nitrosyl heme which can be in transient equilibrium with a pear to be the most important sources quantitatively. In ferrous-nitrosonium species (Eqs. 16a–c) healthy situations, the generation of ROS by eNOS, mito- Fe3ϩ ϩ NO ↔ Fe3ϩ Ϫ NO (16a) chondria, or XO is limited, and in the case of mitochondria, it is probably more related to signaling pathways (289, Fe3ϩ Ϫ NO ↔ Fe2ϩ Ϫ NOϩ (16b) 290). However, there is significant evidence that not only Fe2ϩ Ϫ NOϩ ϩ R-SϪ → Fe2ϩ ϩ R-SNO (16c) NOXs but also the generation of ROS by uncoupled eNOS,

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 327 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

mitochondria, or XO has particular relevance in the origin and development of cardiovascular disease. Loop C

A. NOXs

NOXs are multiprotein enzyme complexes that catalyze the electron transfer to molecular oxygen to form ROS, more commonly superoxide, but also hydrogen peroxide (79, 252, 383, 546, 912). To date, seven isoforms have been described [NOX 1–5 and dual oxidases (DUOX) 1 and 2]. Each isoform consists of a core, catalytic subunit with a Cytosol transmembrane domain and a variable number of regula- tory subunits. The core subunit defines the name of the NOX complex. Thus, NOXs complexes show differences in Loop B their molecular architecture, activator proteins, localiza- tion, and function. NADPH is the preferred substrate for all NOX isoforms, with the NOX2 isoform being particularly NADPH specific (48, 63, 177, 1047).

The putative mechanism of all NOX proteins can be de- scribed as an electron transfer chain in which electrons are transferred from NADPH to the FAD cofactor in the flavo- protein/dehydrogenase domain and from FAD to the heme moieties (Eqs. 18, 19), where molecular oxygen is reduced FIGURE 7. Structure of the NADPH oxidase (NOX) core protein to superoxide. The specific mechanism of superoxide gen- membrane and cytosolic domains. The sequence comprises six eration is still unknown. According to one hypothesis, the transmembrane helices and a cytosolic flavoprotein/dehydroge- 2ϩ nase domain. The transmembrane helices are indicated by colors: I, reduced (Fe ) heme moiety binds oxygen to form a fer- red; II, orange; III, yellow; IV, green; V, cyan; and VI, dark blue. The rous/oxy species that decays to ferric heme and superoxide location of loops B and C is indicated. The dehydrogenase domain is (Eq. 20) (245, 980). Routes that do not involve the forma- shown in purple. The cofactors heme and FAD are shown as red and tion of a stable heme/oxy complex are also possible (Eq. 21) yellow sticks, respectively. Figure was drawn with PyMOL based on (456). Notably, the reaction of NOX1–3 and 5 generates the structures for the transmembrane (PDB: 5O0T) and dehydro- genase (PDB: 5O0X) domains of NOX5 (613). superoxide, whereas NOX4 and DUOX1/2 produce hydro- gen peroxide (26, 936). Existing evidence suggests that the primary species generated is superoxide in all isoforms, and NOX core subunits 1–4 are structurally similar with a N- the formation of hydrogen peroxide is mediated by the per- terminal, membrane domain and a C-terminal flavoprotein, oxidase-like domain in DUOX or by the dismutation of dehydrogenase domain with high sequence homology to the superoxide molecules in NOX4 (Eq. 22). The mechanism of ferredoxin/NADP reductase family (490). NOX5 has a sim- NOX4 is still controversial, but elegant work by Takac et ilar architecture but has an additional N-terminal domain al. indicates that the E-loop (sequence between helices V before the transmembrane domain with four EF hand mo- and VI) of NOX4 shows notable differences with the other ϩ tifs that are responsive to Ca2 . The reductase domain of NOX isoforms (925). This loop in NOX4 seems to provide NOX5 also includes a CaM-binding motif not found in the steric hindrance to slow down superoxide release, forcing ϩ NOX1–4 proteins. When Ca2 is bound, the EF hands fold two superoxide molecules to react with each other in a over the reductase CaM element to relieve autoinhibition reaction that appears to be catalyzed, in part, by a histidine (63, 944). DUOX1 and 2 have a similar architecture to residue (925). NOX5 and include an additional transmembrane domain ϩ ϩ ϩ → ϩ ϩ NADPH FAD H NADP FADH2 (18) connected to a putative extracellular peroxidase domain ϩ 3ϩ → ϩ 2ϩ (262). The ability of this domain to act as a peroxidase is FADH2 2Fe FAD 2Fe (19) still under debate (638, 639). 2ϩ ϩ → 2ϩ Ϫ Fe O2 Fe O2 (20a) 2ϩ Ϫ → 3ϩ ϩ Ϫ· Fe O2 Fe O2 (20b) Recently, the crystal structure of a NOX core subunit (NOX5) has been determined (613). This structure is con- Fe2ϩ ϩ O → Fe3ϩ ϩ O Ϫ· (21) 2 2 sistent with previous evidence suggesting the presence of six Ϫ· ϩ ϩ → ϩ 2O2 2H O2 H2O2 (22) transmembrane ␣ helices with five connecting loops (FIG- URE 7) (613). The transmembrane domain includes four The architecture of the NOX/DUOX core subunits has been conserved histidines that serve as binding sites for two heme described in detail elsewhere (13, 79, 383, 546) (FIGURE 7). moieties bound between helices III and V (288, 613), and

328 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS the flavoprotein domain binds the cofactor FAD and in- domain (694). However, a number of NOX4-interacting cludes a binding site for NADPH (49, 480, 613, 980). proteins have been recently identified. The protein polymer- ase delta–interacting protein 2 (poldip2) seems to exert reg- NOX isoforms are regulated by a large number of protein/ ulatory effects on NOX4 (212, 607), although poldip2 is protein interactions, and most isoforms are not functional involved on many other pathways (316). The membrane without the assembly of the NOX protein with several ad- protein Toll-like receptor 4 (TLR4) has been shown to in- ditional proteins. NOX1–4 are associated to another mem- crease NOX4 activity (86, 724), and Tks4/5, a homolog of brane component, p22phox (24). Although there is no struc- NOXO1, also regulates NOX4 activity (231). Additional tural data available for p22phox, sequence analysis and com- interactions with protein disulfide isomerase have also been putational models have been used to elaborate a consensus reported (465). In addition, gene splicing also appears to prediction of a three-helix transmembrane domain and a modulate NOX4 activation and play important roles in cytoplasmic C-terminal domain (637). Other models are cardiovascular disease (971). consistent with the presence of two transmembrane helices instead of three (135, 451, 930). The regulation of NOX expression is modulated by a vari- ety of agonist species and transcription factors. We will only In the human vasculature, the most relevant isoforms ap- highlight some of the main agonists for each isoform; a pear to be NOX1, NOX2, NOX4, and NOX5 (252, 253, more detailed view is covered in recent reviews (79, 555, 555). The relative architecture of these three proteins and 737, 912). their activator proteins are shown in FIGURE 8. NOX1 ac- tivation requires p22phox, NOX-organizer 1 (NOXO1), 1. NOX2 NOX-activator 1 (NOXA1), and the GTPase Rac1 (24, 108, 348). The activator protein p47phox can replace NOX2 is the main NOX isoform in neutrophils and mac- NOXO1 in a noncanonical mode (61). Recent studies indi- rophages where it mediates the oxidative burst (198, 845). cate a possible role of EBP50 in NOX1 assembly (14). Genetic defects in the main constituents of NOX2 can trig- NOX2 activation requires p22phox, p47phox, p67phox, ger an inability of the immune system cells to kill invading p40phox, and Rac1 (276). As described for NOX1, a non- bacteria and fungi. This condition, characterized by an in- canonical mode of activation can use NOXO1 instead of creased susceptibility to infections, is known as chronic p47phox (927). The activation mechanisms of NOX4 are granulomatous disease. Most mutations causing the disease not completely understood. NOX4 appears to be constitu- (around 90%) are found in the membrane component tively active in the presence of p22phox without the partici- NOX2 (also known historically as gp91phox) and the inter- pation of cytosolic proteins (24, 627). This, in part, can be acting protein p47phox. Mutations in p22phox, p67phox, and explained by the constitutive activity of its dehydrogenase Rac2 have been also identified (428, 438).

-• O -• O2 2 NOX1 NOX2 p22NOX1NOX1 p22 NOX2NOX2

NOXO1/ p47 p40

Rac1 Rac1/ p47/ NOXA1/ p67/ Rac2 NOXO1 FIGURE 8. Putative assembly architecture of NADPH p67p6 NOXA1 oxidase (NOX)1, NOX2, NOX4, and NOX5 and their regulatory proteins.

H O 2 2 -• O2 p22 NOX4NOX4 NOX5N OX5

Ca2+ Ca2+ Ca2+ Ca2+

Poldip2

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 329 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

Blood vessels NOX2

L-citrulline Endothelial NOX1NOX1 NOX4NOX4 cells NOX2NOX2 NOX5NOX5 FIGURE 9. Cellular distribution of NADPH oxidase (NOX) species in the blood vessels. Predominant forms NNOX1OX1 Smooth in each cell type are shown in bold. muscle NOX2NOX2 cells NOX4NOX4 NOX5NOX5

Adventitial NOX2NOX2 fibroblasts NOX4NOX4 NOX2

NOX2 expression has been also reported in other cell types and increased NOX2 activity is linked to vascular disease including endothelial cells and adventitial fibroblasts (376, (142, 252, 478). 382, 657, 714, 715, 736, 876) (FIGURE 9, TABLE 5). Thus, the effects of NOX2 activity depend on different cell types In the vasculature, different effectors can increase NOX2 and have important links to the immune response (392, expression and activity (555, 737). A large number of 488). The current knowledge about NOX enzymes in the cytokines and hormones have been shown to induce cardiovascular system indicates that NOX2 is usually the NOX2 expression, including angiotensin II (658, 714, main player in the development of pathological conditions 802), thrombin (725), and TNF-␣ (275, 964). Circulat-

Table 5. Tissue distribution of NOX isoforms in the cardiovascular system

Tissue Cell Types Cellular Location References

NOX1 Heart ECs Plasma membrane 9, 423, 431, 652, Blood vessels VSMCs Endosomes 876, 879, 909 Adventitial fibroblasts Caveolae Cardiomyocytes NOX2 Heart ECs Plasma membrane 9, 166, 376, 382, Lung VSMCs Phagosomes 427, 468, 473, 580, 657, 714, Blood vessels Adventitial fibroblasts Endoplasmic reticulum 715, 736, 742, Cardiomyocytes 876, 950, 964 Hematopoietic stem cells Macrophages neutrophils, monocytes, dendritic cells NOX4 Heart ECs Plasma membrane 9, 10, 100, 118, Lung VSMCs Focal adhesions 166, 179, 201, 228, 265, 347, Blood vessels Adventitial fibroblasts Endoplasmic reticulum 423, 431, 540, Cardiomyocytes Nucleus 541, 556, 736, Cardiac fibroblasts Mitochondria 742, 876, 906, Hematopoietic stem cells 964 NOX5 Heart ECs Plasma membrane 81, 166, 201, Blood vessels VSMCs Endoplasmic reticulum 396, 469, 494, 848 Cardiomyocytes Cardiac fibroblasts

EC, endothelial cell; NOX, NADPH oxidase; VSMC, vascular smooth muscle cell.

330 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS ing lipids also increase NOX2 production in ECs in vitro 2. NOX1 (159, 208, 856, 1063). The role of endothelial NOX2 in the development of cardiovascular disease is controver- The isoform NOX1 is expressed in VSMCs and in lower sial. Some studies have described that endothelial over- levels is also found in ECs (556, 909) (FIGURE 9, TABLE 5). expression of NOX2 increases endothelial dysfunction NOX1 is found in caveolae and signalosomes of vascular (88, 676). However, in other cases, an increased expres- cells (423, 431, 652). NOX1 expression is activated by sion of endothelial NOX2 has not been linked to patho- several factors including angiotensin II, PDGF, and throm- logical conditions even though an increased production bin (382, 492, 630, 695, 999). The nature of the agonist can of superoxide and macrophage recruitment was observed vary in different cellular compartments (651, 652, 1055). (243, 443). Increased NOX1 activity is associated with vascular dis- ease, and studies have shown links between high NOX1 Global KO studies in mice indicate a negative role of NOX2 levels and hypertension (1015), diabetes (163, 380, 1003), in disease conditions. The deletion of the organizer subunit ischemia/reperfusion injury (479), and coronary artery dis- phox p47 ameliorates the hypertensive response to angioten- ease (393). NOX1-dependent signaling has been linked to sin II, decreases endothelial superoxide formation and angiogenesis. For instance, knockdown of NOX1 can de- eNOS uncoupling, and improves survival after MI (238, crease angiogenesis (343), whereas increased NOX1 pro- Ϫ Ϫ 549, 550). The mice combining the mutation of p47phox / motes tumor angiogenesis and growth (582). These findings with ApoEϪ/Ϫ appear to be protected from atherosclerotic indicate a critical role of NOX1-derived superoxide in an- lesions (253) compared with ApoEϪ/Ϫ mice (64, 976), al- giogenesis signaling via PPAR␣-, AKT-, and ERK1/2-medi- though some reports do not find significant differences in ated pathways. atherosclerosis progression or lesion size (443). It should be noted that the use of p47phoxϪ/Ϫ as a surrogate for the In agreement with these observations, the overexpression of NOX2 deletion is partly limited by the involvement of NOX1 in VSMCs increases blood pressure and hypertro- p47phox in other protein complexes, including not only the phy in mouse hypertension models (235). siRNA against noncanonical activation of NOX1, as already mentioned NOX1 or NOXO1 decreased superoxide production and above but also its interaction with protein disulfide isomer- eNOS uncoupling in diabetic mice (1049). NOXA1 has ase (214, 559). been also associated with atherosclerotic injury, with over- expression of NOXA1 increasing ROS production and in- creased NOXA1 levels detected in atherosclerotic lesions Other studies have directly targeted the NOX2 protein. (695). Mouse models of NOX2 deletion have generally shown phox similar benefits as those using p47 -deficient mice. The Studies of NOX1 function have also taken advantage of the Ϫ/Ϫ NOX2 mice are also protected against angiotensin II– generation of several KO lines. Mice with deletions of dependent hypertension (151, 988). The combination of NOX1 (345, 630), NOXO1 (511), and NOXA1 (293) have Ϫ Ϫ Ϫ Ϫ NOX2 / with ApoE / can decrease the atherosclerotic been developed. NOX1 deletion limits the blood pressure lesions (475), but this is not always the case (508). Other increase elicited by angiotensin II, although it is unclear if works have found a positive effect of the NOX2 deletion in basal blood pressure levels are reduced (345, 346, 630). mice treated with high fat diet. In these studies, mice carry- ing the deletion gain weight but show no increase in ROS The effect of NOX1 in atherosclerosis has been investigated Ϫ Ϫ Ϫ Ϫ production or endothelial dysfunction as compared with similarly to NOX2, using a Nox1 / ApoE / mouse the control animals (256, 609). model. The results of different studies are contradictory, with examples of improved outcomes for NOX1 deficient mice subjected to high fat diet (854) and diabetic mouse Finally, it is worth noting that the role of NOX2 in the models (380), whereas in other cases, NOX1 deletion did immune system and in vascular endothelial cells will not seem to have an effect (875). have, as noted, different impacts in cardiovascular dis- ease (573). The relative contribution of these sources for The effects of the p47phox deletions, often used as a surro- the initiation and development of atherosclerotic lesions gate for the NOX2 deletion, can be partly due to changes in and other pathologies remains an issue of active research. NOX1 activity. This notion is supported by a diminished The current paradigm indicates that macrophage NOX2 effect of angiotensin II in the p47phox KO (549, 550) as is more detrimental in the development of cardiovascular compared with NOX1 (345, 630) or NOX2 deficient mod- disease (395, 876), with endothelial NOX2 probably in- els (988). volved in the initiation, but not so much in the develop- ment of the disease, and a subsequent progress of the Altogether, the role of NOX1 in cardiovascular disease atherosclerotic lesion dependent on immune cells (243, seems secondary to NOX2, but still significant, as shown in 573, 976). several models (345, 380, 630, 854). It is important to

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 331 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

consider that therapies focused on NOX2 can overlook can modulate the role of VEGF-induced dimerization of compensatory effects of NOX1 in part by the sharing of a CD146 necessary for cell adhesion (1065), and NOX4-de- number of regulatory elements that can activate the produc- rived ROS can induce the reversible glutathionylation of tion of superoxide in both proteins. SERCA and increase Ca2ϩ levels, thus regulating VEGF- signaling cell migration (273). 3. NOX4 In a final note, it should be also considered that although NOX4 is the most abundant isoform in ECs, and it is also NOX4 activity has been shown to reduce fibrosis in athero- present in VSMCs and adventitial fibroblasts (10, 265, 736, sclerosis (230), it can also promote fibrosis, particularly in 832, 876, 1036). In the heart, NOX4 has been identified in the lung, but also in kidney and other tissues (110, 201, ECs, cardiomyocytes, and fibroblasts (682, 1058). The in- 416, 596, 820). The basis of these differences in NOX4 tracellular location of NOX4 suggests a role in signaling roles are not completely understood, but could be based on and may be involved in endoplasmic reticulum stress (817) the specific roles of NOX4 in different cell types and/or (FIGURE 9, TABLE 5). The expression of NOX4 is upregu- detrimental effect of eNOS activation in conditions in lated by TGF-␤ (201, 906). Other regulators include TNF-␣ which eNOS is uncoupled (323). (70, 681, 884, 1048), hypoxia (455), and PDGF (487). 4. NOX5 The role of NOX4 in vascular biology appears to be gener- ally protective. Endothelial-targeted overexpression of NOX5 is the last discovered NOX isoform (62). In human NOX4 has beneficial effects and decreases blood pressure blood vessels, NOX5 is expressed in ECs and VSMCs (81, (779). Existing evidence supports a role of NOX4 in main- 469) (FIGURE 9, TABLE 5). taining blood pressure levels (552, 723, 779, 832). NOX5 expression can be activated by most of the factors Endothelial-specific overexpression of NOX4 in mice pro- that activate other vascular NOX enzymes such as angio- motes angiogenesis and eNOS function (193) and decreases tensin II, TNF-␣, PDGF, thrombin, and endothelin-1 (81, blood pressure (779). Comparable results were achieved 660, 681). As indicated above, the particular architecture of with NOX4 knockdown studies in cells (642). A beneficial NOX5 indicates a regulation of its activity through Ca2ϩ effect of NOX4 in ischemia/reperfusion has also been ob- levels (62, 469). Phosphorylation of Ser475, Ser490, served (989). Thr494, and Ser498 within the flavoprotein domain in- creases Ca2ϩ sensitivity (463, 717, 718); this phosphoryla- Ϫ Ϫ Studies with NOX4 / mice indicate protection from en- tion is mediated by different kinases, including PKC iso- dothelial dysfunction in wild-type mice compared with the forms (164). NOX4 deletion (832). In general, NOX4 shows protective Ϫ Ϫ effects in ApoE / mice (194, 229, 230, 379, 552, 838). Unlike the rest of the NOX proteins, NOX5 does not have One study has related NOX4 to hyperglycemia and insulin an homolog in rodents, which has hampered the research of resistance in mice (1012). However, NOX4 deletion in the this isoform (871). New approaches, such as mice strains Ϫ Ϫ ApoE / mouse does not ameliorate diabetes-associated incorporating the human NOX5 gene, may provide new vasculopathy (380). Other studies have found no change in information about NOX5 function (439, 517). NOX4 levels in diabetic rat models (1003). Heart-specific NOX4 overexpression has protective effects in the response Despite these difficulties, increasing data suggest a role of to chronic load stress in part due to increased HIF1␣ re- NOX5 in cardiovascular disease. Higher NOX5 expression sponse and increased angiogenesis (1058). It should be levels have been observed in patients with coronary artery noted that some aspects of NOX4 function in the cardio- disease (391), but causation has not been proven. NOX5 vascular system remain controversial. For instance, splicing expression also increases in the human heart after MI (396). of NOX4 appears to influence cardiovascular disease, with Expression of NOX5 in VSMCs and in macrophages and a shorter protein (NOX4D) exerting beneficial signaling monocytes can contribute to the development of atheroscle- functions in the nucleus, whereas full-length NOX4 is over- rotic lesions (622, 623). NOX5 also appears to modulate expressed in ischemia with detrimental effects (971). proliferation and cell migration processes (371, 719). Alto- gether, increased NOX5 activity seems to be involved in a NOX4 protective role may be due to its generation of hy- pathological response (661). Further work is required to drogen peroxide, and not superoxide (253). As hydrogen delineate the role of NOX5 in human cardiovascular dis- peroxide does not scavenge NO, changes in NOX4 activity ease. do not directly impact NO levels. The signaling mechanisms of hydrogen peroxide in ECs (941) lead to the increased 5. Pharmacological regulation of NOX enzymes activity of eNOS and other enzymes (121, 141, 193, 251, 815, 940). Consistent with this role of NOX4-derived ROS A plethora of NOX inhibitors, including small molecules in angiogenesis (642, 989), it has been shown that NOX4 and peptides, have been developed (22). Most of the inhib-

332 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS itors have shown poor specificity between isoforms, ham- the inhibitory effects of apocynin; notably, apocynin is an pering the progress in the field (174). In this section, we will antioxidant and scavenger of ROS like HOCl and hydrogen present some of the most relevant inhibitors because of their peroxide (426, 735). Thus, although apocynin remains wide use or high isoform specificity. A more comprehensive widely used in research and has led to important insights in analysis of the available inhibitors for NOX enzymes can be the function of NOX enzymes (237, 413, 581), the results found in recent reviews (22, 174, 175, 252). of experiments with apocynin need be considered with cau- tion. In particular, effects in isoforms other than NOX1 and A) DIPHENYLENE IODONIUM. Diphenylene iodonium (DPI) (FIG- NOX2 should be validated with other approaches. URE 10) is a nonspecific inhibitor of all NOX isoforms (18, 197, 246). Its mechanism of action is based on the reaction C) NOX2 DOCKING SEQUENCE-TAT. NOX2 docking sequence-tat with the FAD moiety of the NOX dehydrogenase domain (NOX2ds-tat), the first isoform-specific NOX inhibitor, is a (699). Unfortunately, this reaction is general for any flavin- peptide based on the sequence of the B-loop of NOX2 (199, containing protein, which means that DPI is also an inhib- 218, 782). NOX2ds blocks specifically the interaction be- itor of many other ROS-producing enzymes, including, but tween NOX2 and p47phox (199). The addition of the tat not limited to, NOS, mitochondrial complex I and XO (18). sequence (nine amino acids from HIV-tat) confers mem- Some reports indicate that the use of low levels of DPI may brane penetration ability to the peptide (782). The inhibitor selectively target NOX in the presence of other flavopro- shows high specificity toward NOX2 (199). The chemical teins (241); however, the lack of specificity makes unlikely nature of the compound makes oral delivery unsuitable and the general use of DPI as a NOX inhibitor in vivo. has limited its pharmacological application, however, be- cause of its isoform specificity and lack of off-target effects B) APOCYNIN. Apocynin (FIGURE 10) is a methoxy-substituted remains widely used in research (244, 461, 476, 762, 782, catechol generally considered to be a specific NOX2 iso- 910). Notably, a similar approach with NOX4 did not re- form inhibitor. Apocynin inhibits NOX2 by reacting with phox sult in significant inhibition of NOX4 (200), which suggests cysteine residues in p47 that appear to be necessary for a strong interaction between dehydrogenase and transmem- enzyme assembly (894). It should be noted that this mech- brane domains in NOX4 and points out a critical function anism of action converts apocynin into a NOX1 inhibitor of the B-loop in this interaction (460), very probably linked when NOX1 uses p47phox for activation in the noncanoni- to the constitutive activity of this NOX isoform. cal mode. Moreover, there are several factors that question

D) NOXA1 DOCKING SEQUENCE. NOXA1 docking sequence (NOXA1ds) is a peptide developed to block the interac- tion between NOX1 and NOXA1. The sequence con- tains 11 amino acids from the putative binding region of NOXA1 to NOX1. The selected sequence shows low similarity with p67phox to prevent cross reactivity with NOX2. The peptide prevents NOX1 assembly and does not inhibit NOX2, NOX4, or NOX5 as tested in in vitro assays (772, 792).

E) ML171. ML171 (2-acetyl phenothiazine) (FIGURE 10) is one of the few small-molecule, NOX isoform–specific in- hibitors reported to date. ML171 inhibits NOX1 at nano-

molar concentrations, with IC50 values at least 20-fold higher for NOX2, NOX3, NOX4, and XO (359). The mechanism of action of ML171 seems to involve only inter- action with the NOX1 protein and not with its activating subunits. ML171 showed off-target effects with the inhibi- tion of serotonin and adrenergic receptors in the micromo- lar range (359). The study of differently substituted phe- nothiazines indicates some isoform selectivity that can rep- resent a promising pathway to novel small-molecule, selective NOX inhibitors (847).

F) VAS2870 AND VAS3947. VAS2870 and VAS3947 (FIGURE 10) are recently described NOX inhibitors based on a triazol- FIGURE 10. Chemical structures of selected NADPH oxidase opyrimidine scaffold (892, 934, 1016). VAS2870 appears (NOX) inhibitors. to inhibit all isoforms, with proven inhibitory effects at least

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 333 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

for NOX1, NOX2, NOX4, and NOX5 isoforms (21, 520). tant to recognize and understand the causes of eNOS un- The mode of action of these compounds is unknown, al- coupling to develop adequate interventions. In addition, a though for NOX2, they appear to block complex assembly number of situations impinge NOS function and NO gen- (21). These inhibitors do not have a noticeable effect on eration, but not necessarily cause increased superoxide pro- XO- or eNOS-dependent ROS production, so they can be duction, despite being usually characterized as uncoupling. considered specific pan-NOX inhibitors. VAS3947, a more Factors that can trigger eNOS uncoupling are discussed soluble derivative of VAS2870, shows similar inhibitory below. properties (1016). Off-target effects for VAS2870, namely nonspecific alkylation of thiols, have been reported (915). Initial reports indicated that nNOS had the ability to pro- duce superoxide in a reaction dependent on NADPH and 2ϩ G) GKT-136901 AND GKT-137831. GKT-136901 and GKT- Ca /CaM (750). The effect of NOS inhibitors on this pro- 137831 are pyrazolopyridines structurally related to cess was diverse, with some inhibitors efficiently suppress- VAS2870 and VAS3947. GKT-136901 and GKT-137831 ing superoxide production (e.g., L-NAME), whereas others G (FIGURE 10) are the first inhibitors suitable for oral admin- had little effect (e.g., N -monomethyl-L-arginine) (749). istration, and currently GKT-137831 is in phase 2 clinical The reaction is observed in the three NOS isoforms (750, trials (472). GKT-136901 was first characterized as a selec- 973, 1034). The effect of cofactors and NOS inhibitors was tive NOX1 and NOX4 inhibitor, although it can also in- thoroughly investigated (150, 250, 590, 749, 973, 1033). hibit NOX 2 at higher concentrations (545). Inhibitory ef- Superoxide can be generated by the oxygenase/heme do- fects against NOX5 have also been documented (680). No main, in a Ca2ϩ/CaM-dependent way, or independently by off-target effects were detected against a panel of 135 en- the reductase domain from Ca2ϩ/CaM (653, 1032). In con- zymes, including XO and eNOS (545). GKT-137831 shows ditions that promote uncoupling, the heme group is usually similar inhibitory properties with potent inhibition of the main generator of superoxide species through the for- NOX1 and NOX4 and inhibitory effects on NOX2 and mation of a ferrous/oxy complex that decays to ferric heme NOX5 at higher concentrations (35). The basis of the inhi- and superoxide (Eq. 20) (FIGURE 11) (250, 901, 973, 1033). bition by these compounds is not known, although the ac- tivity of GKT-136901 as peroxynitrite scavengers has been The loss of the dimer structure renders eNOS unable to recently reported (826). produce NO, as the heme group of one monomer must receive electrons from the FMN domain of the other mono- H) GSK2795039. The discovery of a novel, specific NOX2 in- mer. Thus, conditions that increase the monomer/dimer ra- hibitor was recently reported (435). GSK2795039 (FIGURE 10) appears to be specific toward NOX2 and does not show - • - • off-target effects toward eNOS or XO. Existing evidence O2 O2 O2 O2 suggests that GSK2795039 inhibits NOX2 through compe- tition with NADPH for the binding site of the NOX2 de- NOS hydrogenase domain (435). monomer HEME FMN FAD

O O - • B. Uncoupling of Endothelial NOS 2 2 - • - • O2 O2 O2 O2 NO production is critical for vascular function as described NOS dimer in previous sections (299, 573, 600, 786). In conditions in -CaM which the substrates and cofactors are present in saturating HEME FMN FAD concentrations, NOS enzymes catalyze the synthesis of NO L O O - • from molecular oxygen and -arginine with high efficiency. FAD FMN HEME 2 2 The electrons transferred from NADPH are almost quanti- tatively used to catalyze L-arginine oxidation, and the ratio NOS dimer - • of NADPH to L-citrulline is close to the optimal value of 1.5 O2 O2 +CaM molecules of NADPH per L-citrulline (and NO) generated - • (3, 590). However, a number of factors can alter the elec- CaM O2 O2 tron transfer in NOS so the amount of NADPH required to FAD FMN HEME

generate NO becomes much larger. This is largely due to the CaM reduction of other substrates, usually molecular oxygen, to HEME FMN FAD generate superoxide. This situation when the consumption - • of NADPH is not correlated with a stoichiometric produc- O2 O2 tion of NO is generally termed eNOS uncoupling (488, 570, FIGURE 11. Superoxide generation by nitric oxide synthase (NOS) 870). Although the superoxide generation is the common monomer and dimer in the presence or absence of calmodulin manifestation of this NOS dysfunctional state, it is impor- (CaM).

334 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

tio decrease NO output and can increase superoxide pro- The conversion of BH4 to BH2 in the setting of endothelial duction through the reductase domain but also decrease dysfunction has been widely reported (192, 258, 550). The superoxide formation in the oxygenase domain (FIGURE accumulation of BH2 becomes deleterious as the binding 11). Therefore, it should be noted that measurement of site of BH4 in NOS can also bind this pterin. This is partic- dimer/monomer ratios as an indication of eNOS uncou- ularly problematic in the case of eNOS (190). iNOS and pling is not a reliable and reproducible method, and more nNOS show higher affinity toward BH4 than for BH2 specific methods monitoring posttranslational modifica- (around 10-fold in the absence of L-arginine) (6, 514, 753); tions and substrate levels are preferable. however, eNOS shows almost identical binding affinities for BH4 and BH2, and it is thus very sensitive to imbalances The concomitant formation of NO and superoxide can be in the concentrations of BH4 and BH2 (190). Thus, rather especially deleterious as both radicals can combine to form than the relative amount of BH2, the BH4/BH2 ratio gives a peroxynitrite at diffusion-limited rates. Peroxynitrite is a more precise estimation of the endothelial function (190, strong oxidant and nitrosating species (78). Therefore, the 192, 974). superoxide generated by the uncoupled eNOS not only in- Mechanistically, when NOS binds BH2 instead of BH4, the duces toxic effects on its own but 1) acts as a NO scavenger, G decreasing NO availability and 2) generates peroxynitrite, a ferrous/oxy heme cannot hydroxylate L-arginine to N -hy- droxy-L-arginine, NO synthesis is blocked, and the oxygen more harmful species (78). Peroxynitrite can in turn oxidize is eventually oxidized to superoxide. Thus, not only is the BH to BH , further promoting eNOS uncoupling (543). 4 2 NO synthesis completely abolished but the protein also be- Notably, the deleterious effect of superoxide on the endo- comes an oxidase, generating superoxide instead of NO thelium relaxing effect of NO was discovered even before (191, 972, 1007) (TABLE 6). the chemical nature of NO as endothelium-derived relaxing factor was elucidated (385). In the following sections, we 2. L-Arginine/ADMA dissect the different causes of NOS uncoupling and over- view the current status of pharmacological approaches to In saturating conditions of the NOS cofactors, the synthesis prevent or reverse this dysfunctional state. Special emphasis of NO only requires L-Arginine and O2 as substrates (904). is dedicated to the two most relevant aspects in pathophys- L-Arginine is also important for the dimerization of NOS iology, namely the dysregulation of BH4 and L-arginine lev- (51, 515) and increases BH4 binding affinity (514, 753). els. The absence of L-arginine leads to the production of super- oxide in an otherwise active form of NOS; thus, L-arginine

1. BH4/BH2 availability can have an important effect on eNOS uncou- pling (TABLE 6). Given the concentration of L-arginine in The binding of the cofactor tetrahydrobiopterin (BH )isa ECs [0.1–0.8 mM (1030)] and the high affinity of NOS 4 ϭ ␮ enzymes toward L-arginine [eNOS KM 2.9 M (746)], it requisite for NO synthesis by NOS enzymes. BH4 binding synergistically enhances the affinity toward the substrate was long thought that L-arginine availability would not be a functional issue for NOS in vivo. However, in recent years, L-arginine. In addition, BH4 plays a critical role in the cat- alytic cycle of NOS (87, 204, 377, 904, 933, 1000, 1007). it has become clear that extracellular L-arginine and ADMA concentrations, compartmentalization, and membrane The ability of BH4 to support electron delivery to the heme group at critical steps, generating a transient radical species, is specific to BH4. Other pterins are, in general, unable to support NO synthesis in mammalian NOS enzymes, or can Table 6. Relative effect of different factors on eNOS only support a NO synthesis rate much slower than BH4 superoxide and NO production (753). BH4 can be oxidized in vivo by molecular oxygen or Superoxide NO ROS to generate dihydrobiopterin (BH2), a pterin very Production Pterin Substrate Other Synthesis structurally similar to BH4 but unable to support NO syn- thesis (544, 753, 933). ϩϩϩ None L-arginine None ϩϩϩ None ADMA None Biological de novo synthesis of BH4 is mediated by three ϩϩϩ None None None ϩϩ enzymes that catalyze the stepwise process from GTP to BH2 L-arginine None ϩϩ BH4, namely GTP cyclohydrolase I (GCH1), pyruvoyl tet- BH2 None None ϩ ϩ rahydropterin synthase (PTPS), and sepiapterin reductase. BH4 L-arginine S1177p ϭ ϭ In this route, the reaction of GTP with GCH1 is the limiting BH4 L-arginine ϭ step. An additional salvage pathway can produce BH4 from BH4 L-arginine T495p – sepiapterin and/or BH . In this pathway, sepiapterin reduc- 2 Qualitative assessment based on data from Refs. 150, 161, 250, tase reduces sepiapterin to BH2, whereas dihydrofolate re- 334, 753, 973, 974, 1033. ADMA, asymmetric dimethylarginine; ductase can reduce BH2 to BH4 (87, 1007). eNOS, endothelial nitric oxide synthase; NO, nitric oxide.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 335 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

transporters are important for NOS function, as will be Coupled Uncoupled discussed below in sect. IIIB4 (167, 668, 1030).

ADMA (FIGURE 3) is mainly produced from the breakdown of proteins containing methylated arginine residues. The NO synthesis same processes also yield the methyl arginines NG-methyl Superoxide output monoarginine (FIGURE 3) and symmetric dimethylarginine G NO output (FIGURE 3) (668). Notably, ADMA and N -monomethyl-L- arginine (but not symmetric dimethylarginine) are NOS in- hibitors and can be important regulators of NOS activity in Peroxynitrite output vivo (566, 963). The metabolism of ADMA and its effects on cardiovascular disease have been covered by recent re- BH BH 2 views (668, 790, 962). Soon after its discovery in cells and 4 tissue, ADMA was recognized as an important marker for monomer ↑ monomer ↑

cardiovascular disease progression (656, 829, 961, 1011, (Zn-S breakdown) (BH4 deficiency) 1053, 1068). More recent observations indicate that the FIGURE 12. Progression of nitric oxide (NO) synthesis, superox- ADMA/L-arginine ratios in plasma may be a better marker ide, and peroxynitrite during the development of endothelial nitric than either metabolite assessed independently (75, 589, oxide synthase (eNOS) uncoupling. 606, 790, 966).

3. Other causes of eNOS uncoupling reversed by glutaredoxin-1 (160). The glutathionylation of Cys689 and Cys908 causes eNOS uncoupling. These resi- A) DISRUPTION OF THE Zn/S CLUSTER. The dimeric structure of NOS dues, located in the reductase domain, appear to disturb the is held together by interactions between the oxygenase do- electron transfer between FAD and FMN cofactors, and mains, and among these interactions, the formation of a indirectly limit the rate of reduction of the oxygenase do- Zn/4S cluster with the participation of two cysteines from main. Consistent with this hypothesis, the addition of the each NOS monomer (195). The oxidation of the cysteine inhibitor L-NAME decreases superoxide production only in residues involved in the formation of the Zn/S cluster dis- part, indicating that most of the superoxide production oc- rupts the dimer structure. Work by Zou et al. indicated that curs in the reductase domain via electron transfer from the this cluster is particularly sensitive to peroxynitrite (1070). reduced FAD and FMN to molecular oxygen (162) (TABLE This observation is not unexpected, as metal-thiol bonds 6, FIGS. 11, 12). are some of the main targets of peroxynitrite (78, 526). S-glutathionylation has been involved in the development The breakdown of the Zn/S cluster is, in most cases, sec- of angiotensin II–mediated endothelial dysfunction (328) ondary to eNOS uncoupling. As discussed above, the mo- and nitroglycerin resistance (522). Both processes appear to nomeric eNOS cannot catalyze the NO synthesis reactions, be initiated by mitochondrial ROS. Consistent with this and the monomerization renders the heme group inactive. view, the increase in ROS levels, BH depletion, and gluta- The superoxide formation from the monomer is low as 4 compared with the uncoupled dimer, and thus, this mecha- thione oxidation, with subsequent eNOS S-glutathionyla- nism does not increase the rate of superoxide formation. tion, appears to be intimately related (189, 215). However, the breakdown of the Zn/S cluster is a marker that indicates the generation of peroxynitrite in the cellular Along with glutaredoxin-1, thioredoxin has also been pro- environment. Although this is not a 100% specific marker posed to deglutathionylate eNOS, and this process is inde- of eNOS uncoupling (the superoxide source may be differ- pendent of glutathione oxidation (907). This offers thera- ent from NOS, for example, a NOX enzyme), the correla- peutical opportunities in oxidative stress situations in tion of the Zn/S cluster breakdown and eNOS uncoupling is which glutathione levels are low (432). probably elevated (TABLE 6, FIGURE 12). C) PHOSPHORYLATION. Phosphorylation of Thr495 is a negative Another modification that disrupts the Zn/S cluster is S- regulator of CaM binding to eNOS (297, 407, 646). The nitrosation of the cysteine residues Cys96 and Cys101, as Thr495-phosphorylated NOS shows decreased electron discussed in sect. IIA1. transfer from the reductase domain to the heme domain and decreased NO synthesis (588). The accumulation of elec- B) GLUTATHIONYLATION OF CYSTEINE RESIDUES. Recent reports indi- trons in the reductase domain causes an increased uncou- cate that eNOS can be modified posttranslationally by S- pling from the reductase domain, which, as discussed pre- glutathionylation (162). At least three residues have been viously, is less pronounced than when this occurs in the shown to be susceptible of modification: Cys 689, Cys908 heme but can cause significant production of superoxide (162), and Cys382 (160) (TABLE 2). The process can be (TABLE 6).

336 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

4. Pharmacological interventions in eNOS uncoupling In most examples, BH4 is able to improve endothelial func- tion, but the study by Cunnington et al. (202) clearly indi- The sensitivity of the eNOS system to oxidative stress con- cated that in some conditions, BH4 supplementation is not a verts eNOS into a main target in conditions of ROS imbal- practical therapeutic approach. In severe oxidative stress ance. According to the kindling hypothesis of endothelial conditions, the BH4 will be rapidly oxidized to BH2, further dysfunction, the initial formation of ROS by other sources increasing the BH2/BH4 ratio (FIGURE 12). As the treatment can promote uncoupling of NOS, amplifying the initial ex- with antioxidants has been shown to improve endothelial cess of ROS (488). The uncoupling causes eNOS activity to function in certain conditions (56, 419, 420, 422, 572), it is change from healthy (producing NO) to toxic (generating possible that the coadministration of BH4 with ascorbic superoxide) (303). As such, the uncoupling of eNOS has acid or other antioxidants can circumvent these issues. Of been noted as a target for therapeutic potential (488, 569, note, BH4 is also an antioxidant, and it is not clear if some 570, 600, 784). Different interventions have been devel- of the observed effects at the supraphysiological concentra- oped to address this misdirected reactivity. As the status of tions used are just due to short-term antioxidant effects; eNOS uncoupling changes with the severity of the cause, related pterins, such as folate, can also produce similar ef- different treatments may prove more efficacious at different fects (32, 33, 860). Techniques that can assess the oxidative stages (FIGURE 12). Current strategies are discussed below. stress conditions in tissues can certainly improve the use of BH4 therapy with a more personalized approach. Pleiotro- pic effects of some drugs can improve BH availability; this A) BH4. Many studies have shown that BH4 levels correlate 4 with eNOS activity (796, 1006). Conversely, low levels of has been observed for statins such as atorvastatin (31).

BH4 are found in endothelial dysfunction and cardiovascu- lar disease (421, 429, 440, 550, 560, 858, 899). Spear- As an alternative to BH4, supplementation using other re- headed by these observations, interventions to reverse lated compounds such as sepiapterin has been also used. eNOS uncoupling via BH4 supplementation have been nu- Sepiapterin can be converted into BH4 via the salvage path- merous, and in general, the supplementation of BH4 or way by sepiapterin reductase and dihydrofolate reductase positive modifications in the BH4 synthesis pathways have as described in the previous sections. Sepiapterin supple- been positive. A large number of studies on mice and other mentation improves endothelial function in several mouse Ϫ Ϫ animals have been described; for the sake of conciseness, we models, including the atherosclerosis model ApoE / mice will only discuss some of the recent human studies. For a (560), diabetes models (721), or obesity models (527). more complete overview of the BH4 supplementation ex- Mouse GCH1 KO mice show abolished endothelial NO periments including studies in other organisms, the reader is synthesis and increased superoxide production that can be referred to recent reviews (87, 828). reversed by sepiapterin supplementation (173). It should be noted that this pathway has BH2 as an intermediate, which

Acute supplementation of BH4, either orally or injected, could lead to detrimental effects. In conditions in which generally improves vasodilatation. This effect has been ob- dihydrofolate reductase is inactive or absent, the recovery served in a number of human studies, including healthy of eNOS coupling by sepiapterin supplementation is inef- individuals (450, 743, 987) and patients with hypertension fective (452). (429), hypercholesterolemia (318, 899, 1031), chronic heart failure (849), coronary artery disease (618, 850), Folic acid has wide effects on the metabolism, impacting heart failure with reduced ejection fraction (178), or rheu- cysteine/homocysteine metabolism, but also pathways in- matoid arthritis (619) or smokers (418, 929, 957). Studies, volving tetrahydrofolate and dihydrofolate and hence BH4 including the assessment of coronary artery function in pa- biosynthesis (598). The effects on eNOS uncoupling seem tients with atherosclerosis, did not find an improvement related to the antioxidant properties of the folic acid metab- after BH4 treatment (1028). olites (900). Supplementation of folic acid (5-methyltetra- hydrofolate) shows positive effects on eNOS function (900, Several studies monitoring endothelial function after 978) and has been shown to prevent nitroglycerin tolerance chronic BH4 administration have been conducted. In hyper- in short-term human studies (375). Folic acid supplementa- cholesterolemia patients, oral BH4 for 4 wk (400 mg twice tion studies in human blood vessel samples have shown daily) improved endothelial function (186). A treatment increases in BH4 levels and BH4/BH2 ratios (33). with oral BH4 for 2–6 wk in patients with coronary artery disease did not show improvement; and actually increased To improve endogenous BH4 synthesis, efforts have focused BH2 levels (202). In patients with hypertension, a 4–8-wk, in GCH1, the rate limiting enzyme during de novo BH4 400 mg/day treatment was able to decrease blood pressure synthesis (143). Overexpression of GCH1 in cells and (747). Studies on pulmonary hypertension also show im- mouse models improves eNOS function (19, 143). Human provement in the 6-min walking distance (788). A recent variants of GCH1 show measurable effects in blood pres- study using 400 mg daily for 1 wk improved endothelial sure, indicating the direct relationship between GCH1 and function in rheumatoid arthritis patients (619). eNOS function (1057).

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 337 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

B) L-ARGININE. Several lines of evidence indicate that the avail- of L-arginine levels as a biomarker a poor indicator of out- ability of L-arginine may be jeopardized under endothelial comes (668, 857, 1029). Studies of a panel of L-arginine dysfunction, and the concentrations of inhibitory L-arginine metabolites indicate some correlations, particularly for metabolites, such as ADMA, can also increase in patholog- ADMA, but also the need to consider other factors such as ical conditions (107, 1053). In this regard, different studies age, sex, and body mass index (75, 771). The interaction of have investigated alternatives to improve L-arginine supply L-arginine levels with ADMA and other metabolites is the either via direct L-arginine supplementation, the use of L-ar- focus of current studies. As pointed out above, the ratio of ginine precursors, or through the inactivation of L-arginine L-arginine to ADMA appears to be a better indicator of consuming arginases. cardiovascular risk (28, 103, 104, 606, 1050). It has been noted that when ADMA levels are low, the effects of exog- The direct supplementation of L-arginine was shown to in- enous L-arginine are limited; however, the therapy seems to crease NO production in cells. This finding was rather sur- be more beneficial in patients with higher ADMA levels, in prising, as most cells have intracellular L-arginine concen- which the treatment increases the L-arginine/ADMA ratios and improves endothelial function (103). trations manyfold higher than the eNOS KM value, and thus, this observation has been termed the “arginine para- dox” (12, 797). Further studies showed that L-arginine im- Other L-arginine–related species may allow for interventions port via the CAT1 transporter is critical for eNOS activity of L-arginine levels. For example, L-citrulline has also been (167, 634). A membrane metabolon including arginine suc- used to increase L-arginine levels (842, 1029). This supplemen- cinate lyase, argininosuccinate synthetase, hsp90, and tation can avoid the formation of detrimental high ADMA eNOS associates in the caveolae and is necessary for regular levels (632). The studies of L-citrulline in humans show mixed eNOS function (270, 294, 634). Another factor contribut- results. In some cases, L-citrulline increased NO synthesis, but ing to the arginine paradox, as will be discussed below, is no changes in blood pressure were observed (506). Other stud- the concentration of endogenous eNOS inhibitors, mainly ies have observed positive effects of L-citrulline including re- duced blood pressure (20, 285, 286). ADMA. At high ADMA concentrations, L-arginine supple- mentation may not overcome eNOS inhibition, indicating Finally, it must be noted that arginases play an important the need to determine L-arginine/ADMA ratios to better role in the regulation of L-arginine levels (144, 259, 734, assess L-arginine availability (102, 106, 954). 804). Exacerbated arginase activity can decrease L-arginine levels and cause eNOS uncoupling (91, 144, 667, 804, In animal models of MI, L-arginine supplementation shows 1039). Thus, the use of arginase inhibitors is gaining in- mixed results. Some studies show improvement in functional creasing attention. Several human studies have used the recovery (712), whereas studies in L-arginine–treated dogs G arginase inhibitor nor-N -hydroxy-L-arginine (FIGURE 3). show detrimental outcomes (665). Further studies have ex- In these cases, arginase inhibition showed protection plored L-arginine supplementation in human patients. Again, against ischemia-reperfusion injury (530) and improved en- the results have been heterogeneous. The causes of this vari- dothelial function in patients with coronary artery disease ability are still unclear. Many studies have reported a lack of and diabetes (855) and in patients with hypercholesterol- effect on L-arginine in endothelial function markers, and on emia (529). Further studies in this field and the search for occasion, the supplementation of L-arginine has been associ- novel natural and synthetic arginase inhibitors are war- ated with higher mortality effects, such as tests of L-arginine in ranted (757). patients after MI (835). Other studies show impaired recovery in peripheral artery disease patients (1014). In contrast, most studies detect decreases in blood pressure with supplementa- C. Mitochondria tion. A recent meta-analysis indicates that doses between 4 and 24 g/day decrease blood pressure (systolic, 5 mmHg; dia- The mitochondrial electron transport chain (ETC) repre- stolic, 3 mmHg) (240). Other recent studies show improved sents a physiologically significant generator of ROS within endothelial function and insulin sensitivity in patients with vascular cells. Mitochondria generate ATP through the impaired glucose uptake and metabolic syndrome (6.6 g/day transfer of electrons from ETC complexes I and II to com- L-arginine for 2 wk) (663) and improved endothelial function plex IV, where oxygen binds and is reduced to water. and decreased homocysteine levels in patients with elevated Through electron transfer steps, protons are pumped from blood pressure and hyperhomocysteinuria (2.4 g/day for the matrix to the inner membrane space, establishing a pro- 4 wk) (781). ton-motive force that couples electron transfer to the phos- phorylation of ADP to ATP at complex V (FIGURE 13). Altogether, it is not clear if the levels of L-arginine are the Although the majority of electrons entering the ETC ulti- best target for intervention. In some cases high, L-arginine mately reduce oxygen to water at complex IV, a small pro- levels have even been correlated with higher incidence of portion (~2–11%) of electrons escape from the chain at ischemic heart disease (1045). It seems that the involvement complex I or III to generate superoxide (34, 115, 678). of L-arginine in different metabolic pathways makes the use Although this superoxide, being a charged molecule, likely

338 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

Complex I Complex II Complex III Complex IV Complex V NADH: Ubiquinone Succinate Quinol: Cytochrome c Cytochrome c ATP synthase reductase dehydrogenase reductase oxidase

+ -• + -• + + H O2 H O2 H H Intermembrane space

e– e– Cyt c e– Q cycle – – e e

Mitochondrial matrix + NADH NAD O2 2H2O ADP ATP

Succinate Fumarate

FIGURE 13. Mitochondrial electron transfer chain. Main sites of superoxide production are indicated.

does not escape the mitochondrion, manganese superoxide (FIGURE 13). In this process, ubiquinol (QH2) binds to the dismutase (MnSOD) located within the mitochondria cata- Q0 site of complex III and undergoes a one-electron oxida- lyzes the dismutation of superoxide to hydrogen peroxide, tion that produces an unstable semiquinone radical. Al- which can freely diffuse out of the organelle to mediate though in most cases this semiquinone is rapidly oxidized, cellular signaling or contribute to oxidative stress. its reaction with oxygen leads to superoxide production at this site (567, 678). Importantly, specific complex III inhib- 1. Mechanisms of ROS production itors, such as Antimycin A, can stabilize this semiquinone radical, significantly enhancing superoxide production by Mitochondrial ROS production is regulated by a number of complex III (678). Physiologically, it is unclear which fac- factors including oxygen levels, substrate availability, and mi- tors stabilize this species to regulate superoxide production. tochondrial morphology/dynamics, all of which ultimately However, a number of studies demonstrate that complex modulate the redox state of the ETC. Complex I, a 1,000-kDa III–dependent ROS production is important in physiologi- protein comprised of ~45 subunits, is the most significant site cal signaling pathways, including hypoxic sensing (997). of superoxide generation within the ETC and produces super- oxide by two distinct mechanisms depending on the reduction Although complexes I and III are considered the predomi- status of the ETC and strength of the proton-motive force nant sources of ROS within the mitochondrion, it is impor- across the inner mitochondrial membrane (FIGURE 13).A tant to note that mitochondrial enzymes beyond the ETC FMN center within the complex, which accepts electrons from have been reported to generate ROS. Pyruvate dehydroge- NADH, serves as the entry point to the ETC. In conditions in nase (887), ␣-ketoglutarate dehydrogenase (130, 887, 952), which electron transport through the ETC is slow, this FMN and monoamine oxygenase A and B (290) have all been can become reduced and react with oxygen, catalyzing super- demonstrated to serve as ROS sources. These sources, al- oxide production. This occurs in conditions of high NADH/ though lower in their ROS-generating capacity, potentially NADϩ ratios, such as when cellular ATP demand is low or the play a role in specific mitochondrial-dependent redox sig- ETC has sustained damage. In contrast, when electron supply naling pathways. Notably, other reactive species–produc- is high through the ETC, coenzyme Q downstream of complex ing enzymes have also been demonstrated to translocate I becomes reduced. A high proton-motive force present in this into the mitochondrion in specific conditions. For example, condition can then push electrons in the reverse direction from eNOS has been shown to enter the mitochondrion of pul- coenzyme Q to the complex I FMN, generating superoxide by monary smooth muscle cells upon stimulation with asym- reverse electron transport. This type of ROS generation occurs metric dimethyl arginine or endothelin-1 (768, 916). Addi- when succinate, the substrate for complex II, is present in high tionally, although controversial, NOX4 has been reported concentration and is responsible in vivo for the ROS generated to be localized within the mitochondrion either constitu- after ischemia upon reperfusion (171, 172). tively or in specific pathological conditions (152, 306, 540).

Complex III also represents a site of superoxide production 2. Physiological role of mitochondrial ROS within the ETC, although the rate of generation at this site is significantly lower than by reverse electron transport Although it has been recognized since the 1960s that mito- through complex I. Complex III catalyzes the Q cycle in chondria produce ROS, the physiological role of this phe- which electrons are accepted from ubiquinone to be used to nomenon remained unclear until decades later. Accumulat- reduce cytochrome c for subsequent transfer to complex IV ing data now demonstrate that mitochondrial ROS both

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 339 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

mediate essential vascular signaling pathways and can also 997). A similar pathway exists in the pulmonary vascula- contribute to the pathogenesis of cardiovascular disease. ture in which mitochondria-generated oxidants signal the This section will provide an overview of what is known conserved phenomenon of hypoxic vasoconstriction (995). about the role of mitochondrial ROS in cardiovascular On a cellular level, mitochondrial ROS have also been health and disease. shown to be required for the differentiation of some hema- topoietic stem cells as well as human mesenchymal stem Early studies viewed mitochondrial ROS production as cells (707, 948). Additionally, mitochondrial ROS have pathological and focused on the deleterious effects of mito- been implicated in pathways leading to cell proliferation chondrial superoxide production. Mice completely lacking and senescence (233, 1046) as well as in the activation of MnSOD, leading to greater mitochondrial superoxide con- other major cell signaling molecules such as the MAP kinase centrations, died within weeks of birth (584), confirming JNK and NF-␬B (330, 689). the essentiality of mitochondrial superoxide scavenging. Further, mice partially or conditionally lacking MnSOD Notably, mitochondrial ROS modulate mitochondrial showed dilated cardiomyopathy characterized by left ven- function, number, and cellular metabolic balance through a tricle dilation, decreased wall thickness, and hypertrophy number of mechanisms. Mitochondrial hydrogen peroxide (584, 696). Notably, tissue from the left ventricle of heart can oxidize the catalytic subunit of AMP kinase, leading to failure patients showed an increase in superoxide produc- its autophosphorylation and subsequent activation to mod- tion and a decrease in MnSOD protein levels and activity, ulate metabolism within the cell (481, 1067). Although ac- consistent with a role for increased mitochondrial superox- tivation of AMP kinase can lead to mitochondrial biogene- ide in the pathogenesis of heart failure (814). It is now sis and increased mitochondrial number, this pathway can evident that mitochondrial ROS production also plays a also directly be stimulated by mitochondrial hydrogen per- major role in the pathogenesis of MI. During myocardial oxide-dependent activation of the MAP kinase Akt (911, ischemia, the ETC as well as the NADH and quinone pools 919). As a counterbalance, mitochondrial ROS have also become maximally reduced. Additionally, there is a signif- been shown to induce organellar degradation (e.g., mi- icant buildup of succinate, the substrate for complex II, tophagy) under specific conditions (810). Through these which further supports the reduction of the quinone pool. mechanisms, mitochondrial ROS potentially play a role as a Together, these conditions promote reverse electron trans- mediator in elaborate feedback pathways to regulate mito- fer upon reperfusion, which results in significant superoxide chondrial energetics and redox signaling. production from complex I (171). Pharmacological inhibi- tors of complex I, including classical inhibitors as well as The role of mitochondrial oxidants in the pathogenesis of NO donors and nitrite, decrease this reverse electron trans- some disease processes remains controversial. For example, fer–mediated ROS production, leading to attenuated in- two competing hypotheses have been proposed for the role farct size and protection of myocardial function after isch- of mitochondrial ROS in the pathogenesis of pulmonary emia/reperfusion in a number of animal models (discussed arterial hypertension. Studies by Schumacker and col- in the next section) (136, 165, 170, 864). leagues suggest that increases in pulmonary mitochondrial superoxide production leads to the release of intracellular 2ϩ Beyond the heart, changes in mitochondrial redox status Ca stores and subsequent vasoconstriction (996). To the can also have detrimental effects in the vascular wall and contrary, Archer and colleagues demonstrate in animal circulating cells. Decreased MnSOD activity or increased models of pulmonary arterial hypertension that mitochon- mitochondrial superoxide generation causes endothelial drial ROS production in the pulmonary artery smooth mus- dysfunction and impairs acetylcholine-dependent vasodila- cle cells is decreased, leading to inhibition of the KV1.5 channel, which ultimately results in membrane depolariza- tion, particularly with age (1005). This dysfunction occurs 2ϩ not only through the direct reaction of superoxide with NO tion, intracellular Ca increases, and vasoconstriction but is also associated with the oxidative damage of mito- (645). Although the role of mitochondrial ROS in patho- chondrial DNA and proteins. Notably, specific polymor- genesis remains controversial and a hot area of research, phisms in the gene for MnSOD have been established as an there is no doubt that changes in mitochondrial redox status independent risk factor for coronary artery disease (317). play a role in disease progression.

Importantly, although mitochondrial ROS can propagate 3. Pharmacological interventions at the level of cellular damage, low physiological concentrations of mito- mitochondrial function chondrial oxidants have been shown to mediate a number of essential cellular signaling pathways (290). For example, With the recognition that mitochondrial ROS play a role in mitochondria have been proposed to serve as a cellular disease pathogenesis, the development of antioxidants tar- oxygen sensor, such that in hypoxic conditions, mitochon- geted to the mitochondrion is an active area of research. drial ROS production by complex III leads to the stabiliza- Several compounds have been developed with encouraging tion of HIF1␣ and downstream hypoxic adaptation (84, results in vascular disease and other pathologies.

340 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

A) MITOQ. The most extensively tested mitochondrial antiox- cumulates within the mitochondrion and binds to cardiolipin idant is probably MitoQ (FIGURE 14). This small molecule in the inner mitochondrial membrane where it scavenges ROS consists of the antioxidant ubiquinol linked to the lipophilic and prevents lipid peroxidation (921) (FIGURE 14). These pep- cation triphenylphosphonium (500). The positive charge of tides have been shown to mediate protection in animal models the triphenylphosphonium cation is delocalized over its of ischemia/reperfusion (922) and insulin resistance (27). large hydrophobic area, which allows it to freely be taken Phase I clinical trials established that the molecule was safe and into the mitochondria based on the mitochondrial mem- well tolerated (360). Multiple phase II trials are underway, brane potential (873). MitoQ has been used extensively in examining the effects of the drug in conditions such as skeletal animal models of disease and been shown to be protective muscle disorders (NCT02245620), mitochondrial disorders against ischemia/reperfusion injury, cardiac hypertrophy, (NCT02976038, NCT02805790, NCT02367014), and heart and hypertension (210, 211) among other pathologies. failure (NCT02814097, NCT02788747, NCT02914665).

MitoQ has also been tested in phase II clinical trials for C) MITOSNO AND SODIUM NITRITE. In addition to pure mitochondri- both Parkinson’s disease and hepatitis C (NCT00329056; al-targeted oxidant scavengers, potential therapeutics that NCT00433108). Although Parkinson’s patients taking Mi- modulate ETC activity to decrease mitochondrial ROS pro- toQ showed no difference in motor function compared with duction are being investigated. For example, MitoSNO those on placebo treatment, this trial did establish that ad- (FIGURE 14), a mitochondrially targeted S-nitrosothiol, was ministration of MitoQ to humans for 1 yr is safe (874). In shown in rodent models to significantly attenuate infarct contrast, chronic hepatitis C patients treated with MitoQ size after MI (170). Mechanistically, this protection is de- for 28 days showed significant lowering of liver injury pendent on MitoSNO transnitrosating a critical cysteine in markers such as circulating alanine transaminase (333). the active site of complex I. This S-nitrosation of Cys39 in These positive results have led to further phase IIB trials in the ND3 subunit of complex I inhibits its catalytic activity hepatitis as well as the initiation of clinical trials to test and, hence, attenuates reverse electron transport and ROS MitoQ in other vascular pathologies. production (170). Notably, other nitrogen-centered mole- cules, such as nitrite, have been shown to promote S-nitro- B) SZETO–SCHILLER-31. Szeto–Schiller peptides are another class sation of complex I and attenuate ischemia/reperfusion in- of molecules that target antioxidant activity to the mito- jury (864). Nitrite administered before or during cardiac chondrion. Szeto–Schiller-31 (SS-31) (Bendavia/elamipretide; ischemia has been shown to protect cardiac function and Stealth Peptides) is a small water-soluble tetrapeptide that ac- attenuate infarct size in a number of small and large animal models (226, 374, 864). These preclinical studies have led to ongoing clinical testing of nitrite as a therapeutic for MI as well as cardiac arrest.

D. XO

Xanthine oxidoreductase (XOR) is expressed as a xanthine dehydrogenase (XDH) but can become XO by posttransla- tional modifications (FIGURE 15). XOR is a dimeric protein containing one molybdopterin cofactor, two iron/sulfur clusters (2Fe/2S), and one molecule of FAD per 145-kDa subunit. The N-terminal domain contains the two Fe/S clus- ters, a central domain contains the FAD cofactor, and a NADϩ/H binding site and the C-terminal domain contains the molybdopterin cofactor and a purine binding site (268). The enzyme has an important role in the catabolism of purines, catalyzing the stepwise oxidations of hypoxan- thine to xanthine and xanthine to uric acid. Purine oxida- tion takes place in the molybdopterin site where the reac- tion leads to the reduction of the Mo6ϩ to Mo4ϩ. The elec- trons are then transported within the enzyme, first to the

Fe/S clusters and then to the FAD. The fully reduced FADH2 can reduce NADϩ to form NADH.

1. ROS generation by XOR

FIGURE 14. Structures of selected mitochondria-targeted During normal operation, XDH shows a limited produc- compounds. tion of superoxide/hydrogen peroxide via electron leak

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 341 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

A C Xanthine Hypoxanthine

MPT e–

Fe2S2 e–

Fe2S2 e– FIGURE 15. Architecture of xanthine dehydrogenase FAD (XDH)/zanthine oxidase. A and B indicate the arrange- ment of the domains in the XDH/xanthine oxidoreduc- tase (XOR) monomer. The middle, FAD-containing do- NAD+ NADH main (yellow), is connected to the N-terminal domain

containing two iron/sulfur clusters (Fe2S2) (green) and the molybdopterin domain (pink) by two flexible linkers. B Posttranslational changes modify the protein activity from a XDH (A), NADH-producing enzyme, to the xan- Xanthine Hypoxanthine thine oxidase (XO) (B), hydrogen peroxide/superoxide- generating enzyme. C: three-dimensional structure of the XDH monomer (PDB:1FO4) (268). MPT e–

Fe2S2 e–

Fe2S2 e– FAD

• – O2 H2O2 / O2

through the reduced FAD cofactor to O2. This reaction only 2. Pharmacological interventions targeting XO produces significant levels of ROS if NADϩ levels are low; however, these conditions can happen in the setting of vas- Extensive evidence supports a role of XO in the develop- cular hypoxia (406, 562). ment of cardiovascular disease (137, 148, 157, 282, 430, 466, 561, 633). As XO has become an interesting pharma- Oxidation of cysteine residues 535 and 992 in rat or bovine cological target in the treatment of heart and cardiovascular XDH or limited proteolysis turns XDH into XO (23, 72, diseases, numerous studies have used different strategies for 693, 773, 803, 994). XO shows a decreased affinity for the inhibition of XO. ϩ NAD and increased O2 affinity. The comparison of the A) TUNGSTEN. Tungsten can be used as a global inhibitor of crystal structures for XDH and XO indicates the movement ϩ molybdenum-containing enzymes as it replaces the Mo of a loop that partially blocks the NAD binding site and atom in the molybdopterin, rendering the enzymes that use alters the electrostatics of the FAD environment (268, 542). this cofactor inactive. Because of this mechanism of action, Altogether, these changes in substrate affinity lead to an it should be noted that effects independent of XO will also increased electron flow from the molybdopterin domain to ϩ be observed because of inhibition of AO, sulfite oxidase, oxygen instead of NAD . Thus, XO catalyzes the oxidation and mARC. Tungsten is a powerful tool for the study of of hypoxanthine to xanthine and subsequently xanthine to molybdenum enzymes in cell and animal studies and has uric acid, generating hydrogen peroxide and superoxide in a been found to alleviate atherosclerosis in ApoEϪ/Ϫ mice reaction amplified under ischemia and hypoxia (147, 307, (833). However, because of its broad effects, the clinical use 496, 497, 499). of tungsten is not feasible.

XO is expressed in ECs (393), and its activity is increased B) ALLOPURINOL. Allopurinol is structurally similar to the XO by angiotensin II (551), TNF-␣, LPS (117, 311), IL-6, and substrate hypoxanthine and is metabolized to the active hypoxia (410, 748). There are low-circulating levels of compound oxypurinol (FIGURE 16). Both allopurinol and plasma XO in baseline conditions that can increase in oxypurinol have been used for the treatment of gout for disease conditions by the release from damaged cells in decades (305, 710). Beyond its use for hyperuricemia and liver or other tissues (1010). The binding of plasma XO gout, more recent studies have used allopurinol and oxy- to ECs can lead to vascular dysfunction (45, 71, 562, purinol as XO inhibitors for the treatment of cardiovascu- 612, 1010). lar disease in human studies. In small trials, the pharmaco-

342 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

covalent modification in the enzyme that inactivates XO after complex dissociation (629, 703, 821). Topiroxostat is currently under study for the treatment of hyperuricemia in clinical trials (NCT02327754; NCT02837198) (441, 442).

E) NITRITE. Nitrite has also been used to inhibit XO-catalyzed ROS generation and to take advantage of its nitrite reduc- tase activity to generate NO (reviewed in sect. IIB3) while decreasing superoxide production (1071). Recent work in- dicates that this treatment can be effective in the reduction of blood pressure in hypertensive patients (357).

FIGURE 16. Structures of xanthine oxidase substrates and inhibitors. E. Other Sources of ROS

Other proteins have been related to the direct or indirect logical inhibition of XO with allopurinol/oxypurinol im- production of ROS in the cardiovascular system, including, proved cardiovascular conditions in several pathologies, but not limited to, peroxidases, cyclooxygenases (COX), including heart failure (58, 278), dilated cardiomyopathy lipoxygenases, monoamine oxidase, heme oxygenase (HO), (148), and diabetes (137, 224). Amelioration of endothelial function in smokers was also observed (387). Despite these glucose oxidase, cytochrome b5 reductase, and CYPOR initial results, subsequent trials with larger number of pa- (209, 505, 790, 908). In this section, we will briefly describe tients have failed to report substantial positive effects as some of these ROS sources. compared with the placebo groups (361, 405), although the meta-analysis of available studies indicates some improve- 1. Peroxidases and COX ment of cardiovascular function (430). Altogether, the ef- fect of allopurinol in cardiovascular disease is still contro- Mammalian peroxidases are heme proteins that catalyze versial. It has also been pointed out that allopurinol and the oxidation of a wide range of substrates. These proteins oxypurinol have limited capacity to inhibit XO bound to are activated by their reaction with hydrogen peroxide to glycosaminoglycans (620). It is very possible that the use of generate an oxidizing heme species that can hydroxylate or XO inhibitors needs to be complemented with other thera- oxygenate a substrate. Mammalian peroxidases include pies targeting other sources of endothelial dysfunction thyroid peroxidase, myeloperoxidase (MPO), lactoperoxi- (928). dase, eosinophil peroxidase, and COX. MPO, lactoperoxi- dase, and eosinophil peroxidase are integral components of C) FEBUXOSTAT. Febuxostat (2-(3-cyano-4-isobutoxyphenyl)- the immune defense (516). 4-methyl-5-thiazole carboxylic acid) (TEI-6720, TMX-67) (FIGURE 16) is a XO inhibitor developed by Teijin, with A) MPO. MPO is found predominantly in neutrophils and higher selectivity and potency than allopurinol and oxy- monocytes and catalyzes the reaction of hydrogen peroxide purinol (702). The selectivity is partly due to a lack of to produce several reactive species, mainly hypochlorous structural resemblance with purines; this also reduces side acid (HOCl) (discussed in sect. IVB) (516, 1021). Macro- effects of purine and pyrimidine metabolism (1041). The higher affinity and specificity of febuxostat can have advan- phage localization in the atherosclerotic plaque leads to tages over allopurinol and oxypurinol. Notably, the activity detectable MPO activity and products of HOCl oxidation of febuxostat is not impaired by XO immobilization on the (213, 414). Several studies have correlated low circulating surface of ECs (620). Febuxostat was approved in 2008 in MPO levels with better cardiovascular prognosis (239, 260, Europe and by the Food and Drug Administration in 2009 644, 690, 1059). for the treatment of gout (commercial name Uloric in United States, Adenuric in Europe) (76, 77, 837). Studies in B) COX. COX are proteins from the peroxidase family, evo- rodents have shown improvement in atherosclerosis and lutionarily related to the peroxidase enzymes of the immune hypertension after treatment with febuxostat (697, 859). system (1052). They utilize lipid substrates, generally ara- The safety of febuxostat for the treatment of cardiovascular chidonic acid, and produce a variety of prostaglandins, lipid disease in humans is still under investigation (331, 610). oxidation products with important physiological effects (156). Two isoforms are expressed in mammals: COX-1 D) TOPIROXOSTAT. Topiroxostat (FYX-051, 4-[5-pyridin-4-yl- and COX-2, and both of them have been related to endo- 1H-[1, 2, 4] triazol-3-yl]pyridine-2-carbonitrile) (FIGURE thelial dysfunction (981, 982). Interestingly, some interplay 16) is a novel XO inhibitor developed by Fuji Yahuhin Co. between COX-2 and other ROS/RNS-generating proteins Ltd. Topiroxostat is a potent inhibitor of XO and induces a such as iNOS (52) and XO (701) have been described.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 343 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

2. Lipoxygenases sistent with an important role in the processing of heme from red blood cell turnover (713, 805). The interplay be- Lipoxygenases catalyze the insertion of oxygen in polyun- tween HO-1, CO production, and NOX inactivation is a saturated fatty acids (119, 535, 1040) and are also biolog- focus of current interest in cardiovascular research (685, ical sources of superoxide (537). Several lipoxygenases have 793, 805, 924). been related to inflammation and cardiovascular disease, in particular, 5-lipoxygenase and 12/15-lipoxygenase (203, IV. OTHER ROS 597, 636).

3. AO A. Peroxynitrite

Ϫ The Mo-containing protein AO has also been described as a Peroxynitrite (ONOO ) is a strong oxidizing and nitrating potential source of superoxide in vivo (538, 539). Although agent formed by the reaction of NO with superoxide (Eq. the function of AO in mammals is yet to be established 23). This reaction occurs at a diffusion-limited rate and is (434, 935), AO appears to play an important role in drug governed by the flux of NO and superoxide production metabolism and other metabolic pathways (335, 512, 513, (78). Another physiological route by which peroxynitrite 756, 920). Studies with rat AO have shown a significant can be formed is through the reaction of hydrogen peroxide production of superoxide when the enzyme uses NADH as with nitrite in acidic conditions (Eq. 24) (808). a substrate (539). Recent work on the human enzyme shows Ϫ Ϫ NO· ϩ O · → ONOO (23) that the production of superoxide by the human AOX1 2 ϩ → ϩ isoform is lower than that of the rat enzymes, and the en- H2O2 HNO2 ONOOH H2O(24a) zyme is not reactive toward NADH (304). However, a Ϫ Ϫ ONOOH ϩ OH → ONOO ϩ H O(24b) known single nucleotide polymorphism in the AOX1 gene 2 was shown to produce a mutant enzyme with increased Despite its rapid rate of formation, peroxynitrite reacts rel- superoxide formation rates in vitro (304). The possible im- atively slowly in comparison with other RNS, with many pact of these single nucleotide polymorphisms in vivo re- biomolecules, including metal centers, proteins, DNA, and mains to be determined. lipids (78, 526, 709). Additionally, studies show peroxyni- trite can traverse cell membranes by both diffusion and 4. CYPOR and CYP through ion channels, suggesting that peroxynitrite can me- diate oxidation/nitration distant from its site of formation CYPOR is a diflavin protein, structurally and evolutionarily (221, 611). related to the reductase domain of the NOS enzymes (377) that functions as a general reductase of the many CYP pro- The reaction of peroxynitrite with biomolecules is generally teins. The P450 system is conspicuous for its role in detox- considered as a detrimental event. Peroxynitrite can directly ification and hormone synthesis (675, 1013). CYPOR, ei- react with a variety of metal centers, resulting in their oxi- ther directly or in combination with the CYP proteins, has dation. In the cardiovascular system, peroxynitrite can ox- been described as a source of superoxide (758, 851). The idize heme proteins, such as Hb and Mb, resulting in the presence of these proteins in endothelial cells makes them a oxidation of their heme oxygen–carrying site from ferrous significant source of superoxide in the vasculature (295, iron to ferric heme (101, 942). In the mitochondrion, a 298). Because of the functional similarities with the dehy- similar reaction oxidizes the electron transport protein cy- drogenase domain of NOX, some NADPH-dependent su- tochrome c (942). Reaction of peroxynitrite with Zn/S clus- peroxide measurements can be unable to differentiate ters such as that present in eNOS, causes dysfunction of the CYPOR from NOX as the source of superoxide (783). enzyme as outlined above (sect. IIIB3) (78, 526). Addition- ally, peroxynitrite reacts rapidly with Fe/S clusters leading 5. HO to the inactivation of critical enzymes such as mitochondrial aconitase (155, 401). HO mediates the degradation of heme in mammals. This enzyme catalyzes the degradation of the protoporphyrin IX Peroxynitrite is perhaps most well recognized for its reac- (heme b) to biliverdin (805, 806). This process generates tion with cysteine and tyrosine residues in proteins. Per- ferrous iron (Fe2ϩ) and CO, which are well-known toxic oxynitrite-dependent cysteine oxidation results in the for- species (122, 739, 795, 959), but CO can also be, in low mation of a sulfenic acid intermediate that can further be concentrations, a signaling molecule (713, 739, 805, 807). oxidized to a disulfide. This type of oxidation modulates the Three HO isoforms exist; the inducible isoform HO-1 is, to activity of a number of enzymes. For example, peroxyni- date, the most relevant to vascular physiology. HO-1 ex- trite-mediated oxidation of ETC components such as com- pression in basal conditions is low or undetectable in most plex I and complex V results in inhibition of mitochondrial tissues. High levels of HO-1 are present in the spleen, con- function (767). Oxidation of tyrosine phosphatases results

344 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS in the inhibition of their enzymatic activity and the aberrant ease (113), protection after ischemia/reperfusion (683, propagation of phosphorylative signaling (926). In con- 801), decreases in atherosclerosis (800), and reversal of trast, oxidation of cysteine residues on metalloproteinases obesity-induced pulmonary hypertension (495). results in the activation of these enzymes, which may also be detrimental to tissue integrity and repair (992). In addition B. HOCl to proteins, peroxynitrite can directly oxidize reduced glu- tathione, depleting the antioxidant capacity of the cell (37). Hypochlorous acid (HOCl) is generated by the enzyme MPO, localized within leukocytes by the following reaction Beyond cysteine oxidation, peroxynitrite can mediate the (300) (Eq. 25) Ϫ covalent addition of a nitro group ( NO2) to the aromatic H O ϩ ClϪ → HOCl ϩ OHϪ (25) ring of tyrosine, resulting in tyrosine nitration. This modi- 2 2 fication is measured as the hallmark of peroxynitrite pres- This highly reactive oxidizing and chlorinating agent plays ence and is traditionally viewed as a sign of nitrative or a major role in the microbicidal actions of neutrophils (15, oxidative stress (78, 765). Importantly, protein nitration 1021). Although HOCl reacts with a variety of biomolecule can also occur through the reaction of nitrite with heme targets (755), in proteins, it predominantly oxidizes cys- proteins in the presence of hydrogen peroxide (153, 154). teine and methionine residues to disulfide and methionine Thus, methodology beyond nitrotyrosine measurement is sulfoxide, respectively (726). With regards to metal centers, required to confirm the presence of peroxynitrite in biolog- HOCl can react with heme and Fe/S centers to mediate ical systems. oxidative bleaching. Additionally, HOCl reacts more slowly with tyrosine residues to mediate the formation of Although a growing number of proteins have now been 3-chlorotyrosine (1019). Notably, 3-chlorotyrosine is uti- shown to be nitrated, multiple studies have shown that lized as a marker of the presence of both MPO (as this is the nitration is a selective process (47, 484). Tyrosines for only enzyme known to generate HOCl) and HOCl. Oxida- which the aromatic ring is closer to the surface of a protein, tion of amino acids and metal centers of enzymes results in in which there is a neighboring negative charge, and those in the HOCl-dependent impairment of a number of critical hydrophobic regions are more likely to be nitrated (66, enzymes. 877). Several studies provide comprehensive lists of pro- teins identified to be nitrated in physiology and disease (44, The role of HOCl in the modification of proteins and lipids 73, 381, 709). These proteins include, but are not limited is particularly well studied and pertinent to the pathogene- to, enzymes involved in antioxidant defenses such as Mn- sis of vascular disease. In the context of atherosclerosis, SOD (219, 220), detoxification enzymes such as CYP (587), HOCl has been shown to oxidize both high and low density nucleic acid regulatory proteins such as histone deacetylases lipoproteins (90, 470, 881, 1042). Furthermore, chlori- (457) and histones (453, 504), and cell structural proteins nated proteins and lipids are present in atherosclerotic such as tubulin (730, 732) and myosin heavy chain (648). plaques (90, 415, 621). Hypochlorous acid decreases eNOS Importantly, the nitration of these proteins decreases en- activity and NO bioavailability by a number of direct and zyme activity, disrupts metabolism and cellular detoxifica- indirect mechanisms, disrupting protective NO signaling tion, perturbs cytoskeletal organization, and ultimately (2, 464, 1054). The oxidation and chlorination of proteins, contributes to the cytotoxic effects of peroxynitrite. such as matrix metalloproteinases, leads to detrimental vas- cular remodeling (314). Although peroxynitrite is traditionally regarded as a harm- ful species, it should be noted that it may have some bene- C. Hydroxyl Radical ficial signaling properties. For example, lipids are also a significant biological target for peroxynitrite, and although The hydroxyl radical (OH·) is one of the most reactive spe- the species can abstract a hydrogen atom to initiate lipid cies formed in biological systems. In this context, OH· is peroxidation, a detrimental process that can lead to cyto- generally formed as a product of Fenton reactions involving toxicity in many cases (65, 766), peroxynitrite can also free iron or copper ions (Fe2ϩ;Cuϩ) and hydrogen peroxide mediate lipid nitration (54, 1027). Nitrated fatty acids are a (Eq. 26) (281, 986) or from the reaction of hydrogen per- class of electrophilic signaling molecules which have been oxide with superoxide (Haber–Weiss reaction) (Eq. 27) shown to react with a number of cellular targets to mediate (394). protective inflammatory and metabolic signaling (830). Pri- 2ϩ ϩ → 3ϩ ϩ · ϩ Ϫ Fe H2O2 Fe OH OH (26a) mary mechanisms of nitrated fatty acid signaling include ϩ ϩ → 2ϩ ϩ · ϩ Ϫ activation of PPAR␥ (55, 583) and Michael addition to Cu H2O2 Cu OH OH (26b) Ϫ· ϩ → · ϩ ϩ Ϫ thiol-containing proteins in a number of signaling pathways O2 H2O2 OH O2 OH (27) (53, 955). Through these signaling mechanisms, nitrated lipids mediate a number of physiological effects in animal The process is generally more complex than indicated in Eq. models including attenuation of inflammatory bowel dis- 26 and includes the formation of a transient metal/peroxide

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 345 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

· ϩ · → complex that can react in a different fashion depending on NO2 NO N2O3 (29) the metal ligands and possible substrates. Therefore, in some cases, the reaction may not produce free hydroxyl As discussed previously in section IIB2, the formation of radical but other metal-based oxidizing species (759, 1020). N2O3 in the red blood cell via an anhydrase reaction involv- ing Hb, NO, and nitrite (69) constitutes a possible pathway · Although the endogenous production of OH in healthy to preserve NO bioactivity (367). conditions is apparently negligible (474), the excess of hy- drogen peroxide and superoxide in oxidative stress condi- 3. Carbonate radical tions can yield an increased amount of this toxic species (1072). The carbonate radical can be formed in vivo by the reaction of the hydroxyl radical with carbonate or bicarbonate (138) In physiology, perhaps the most common source of Fenton or via the reaction of peroxynitrite with carbon dioxide chemistry is the presence of increased concentrations of free (Eq. 30) (112, 608, 640). iron and/or copper (122, 458, 959). In the cardiovascular · ϩ 2Ϫ → Ϫ ϩ ·Ϫ system, this can be related to aging (122) or hemolytic dis- OH CO3 OH CO3 (30a) ease (1026). The source of this iron is most likely plasma- · ϩ Ϫ → Ϫ ϩ ·Ϫ OH HCO3 H2O CO3 (30b) released Hb; whereas Hb or heme are not catalysts for the Fenton reaction, the reaction of plasma Hb with peroxides Several examples of biological oxidations mediated by the can cause the release of free iron (389, 759). bicarbonate anion, in particular through interactions with peroxynitrite, have been reported in the literature [e.g., (46, The hydroxyl radical reacts with multiple organic sub- 635, 918, 1056)]. In relation to cardiovascular biology, it is strates including, but not limited to, DNA, lipids, and pro- important to note that XO has also been characterized as a teins at rates near the diffusion limits. This lack of specific- target and source of carbonate radical (111). ity makes the use of antioxidants to prevent hydroxyl rad- ical toxicity impractical. Ideally, the best strategies will be based on the chelation of the active metal ions in an envi- V. CROSS-TALK BETWEEN ROS AND RNS ronment that blocks their redox reactivity. Proteins like SOURCES ceruloplasmin, transferrin, or lactoferrin limit the reaction (388, 1020). The binding of NO to heme iron can also limit Our description of sources of NO and ROS may give the heme reactivity and prevent iron release from heme (482). impression of a rigorous separation of roles for each protein or enzymatic system. This is certainly not the situation in physiological systems. We have discussed how some en- D. Other Species zymes, in particular eNOS, can produce different species in response to posttranslational modifications or lack of co- The reactions of NO can generate other reactive species that factors or substrates (sects. IIA1 and IIIB). Current research have a poorly characterized role in vivo. Among these spe- has greatly advanced our knowledge on how different sys- · cies, we will mention the nitrogen dioxide radical (NO2 ), tems can influence the NO/ROS generation from other ·Ϫ dinitrogen trioxide (N2O3), and carbonate radical (CO3 ) sources in which it has been termed ROS-induced ROS (46, 692). release (1066, 1069).

1. Nitrogen dioxide Mitochondria and NOX are the main effectors of down- stream signaling to other sources and to each other. Mito- · Nitrogen dioxide (NO2 ) is formed in the reaction of NO chondrial uncoupling and increased ROS generation with oxygen (Eq. 28) (509, 1018). A more reactive species activates the mitochondrial permeability transition pore than NO, it is often used as a surrogate for the detection of (mPTP), allowing the release of ROS to the cytosol (1069). NO. Other formation routes include the reactions of nitrite In response to the increased levels of ROS, the PKC is acti- with oxygenated Hb (501) and peroxynitrite decomposi- vated, which in turn, can activate NOXs (319, 776). Other tion (46). routes of mitochondrial triggered activation of NOX in- NO· ϩ O ↔ O NO· (28a) volve the activation of NOX1 via activation of PI3K and 2 2 Rac1. The blockade of mitochondrial ROS by rotenone was · ϩ · → · O2NO NO 2NO2 (28b) shown to prevent NOX1 activation (FIGURE 17) (564).

2. Dinitrogen trioxide Indeed, activated NOX can in turn, increase ROS produc- tion by the mitochondria (11, 116). This process appears to

Dinitrogen trioxide (N2O3) is a strong nitrosating species require the function of mitochondrial ATP-dependent po- also formed in the oxidation of NO (Eq. 29) (691, 692, tassium channels (KATP), as shown by the ability of KATP 1018). inhibitors to block the increase of mitochondrial ROS pro-

346 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

Mitochondria

mPTP PI3K K ATP PKC FIGURE 17. Cross-talk between mi- tochondria, NADPH oxidase (NOX), and other reactive oxygen species NOS XO (ROS) sources. Mitochondrial ROS are released to the cytosol via the mito- chondrial permeability transition pore (mPTP). These can cause activation of ROSROS ROSROS PKC and phosphatidylinositol 3-kinase (PI3K) kinases and Rac1 GTPase with subsequent activation of NOX enzymes and further ROS production. Alterna- tively, NOX can release ROS that acti-  PKC vate protein kinase C, isoform ␧ (PKC␧) PI3K and in turn activate the mitochondrial Rac1 ATP-dependent potassium channels

(KATP), increasing mitochondrial ROS. Cytosolic ROS can increase ROS-gen- erating activity of endothelial nitric ox- ide synthase (eNOS) and xanthine oxi- dase (XO).

NOX

duction after angiotensin II–induced activation of NOX the direct inhibition of a single source of oxidative stress. (242, 507). Of note, the activation of NOX via angiotensin Recent work indicates that this can be a feasible approach II and the angiotensin II type 1 receptor is mediated by PKC; (206, 207, 234, 836). The development of novel, specific ␧ however, the isoform PKC is also an activator of the KATP mitochondrial inhibitors (discussed in sect. IIIC3)isa channel, which, as indicated, also stimulates mitochondrial promising step in this direction (8, 679, 923, 1062). ROS production (FIGURE 17) (187, 242).

Another case of significant cross-talk between NOX and VI. CONCLUDING REMARKS: ADVANCES, mitochondria has been provided by studies of nitroglycerin CHALLENGES, AND THERAPEUTIC resistance. In this case, the development of tolerance was OPPORTUNITIES mainly due to mitochondrial ROS release and was indepen- dent of NOX, whereas the activation of NOX, triggered by Over the last decade, our knowledge about NO and ROS mitochondrial ROS, was the effector of the endothelial dys- generation has increased substantially. From the initial function associated to nitroglycerin tolerance (1004). ideas considering that oxidative species need to be sup- pressed and that oxidative stress could be combated with Secondary to the main role of mitochondria and NOX, but adequate antioxidant treatment, we now can appreciate the not to be ignored, XO and eNOS can also amplify the critical signaling properties of NO, hydrogen peroxide, or formation of ROS; for instance, XO is indirectly activated by angiotensin II, increasing cellular ROS production (551). superoxide. A direct role of mitochondrial ROS on XO activation has also been described (362). As discussed in previous section, The initial approach to the treatment of oxidative stress was increased ROS eventually causes eNOS uncoupling via BH based on the supplementation with different antioxidants, 4 ␤ depletion, Zn/S cluster breakdown, and other routes, fur- in particular vitamins C and E and -carotene. The use of ther perpetuating the generation of superoxide and other antioxidants has been markedly ineffective when not dam- ROS (FIGURE 17) (488, 570). aging (97, 390, 400, 785). In fact, the situation has been termed the “antioxidant paradox” (399, 400). The failure The existence of many different routes interconnecting of these studies, along with their limitations, has been dis- ROS/RNS sources may suggest that the normalization of cussed at length in this review, and a number of explana- dysfunctional conditions is a herculean task; however, it tions for this phenomenon have been proposed (67, 158, also presents the promise of normalizing a disease state by 252, 400, 672, 949).

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 347 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

At the same time, we are increasingly appreciative of the sGC. Advances on NOX research have led to the develop- cross-talk between different radical species, generation ment of novel inhibitors on their way to widespread use for pathways, and cellular compartments. Different examples the treatment of cardiovascular disease and other patholo- of species interaction have been discussed throughout this gies. New highly specific inhibitors of other ROS sources

review; for instance, the oxidation of BH4 by superoxide like the XO-targeted febuxostat and mitochondrial-tar- will trigger eNOS uncoupling with decreased NO produc- geted drugs provide additional pathways to modulate cel- tion and increased superoxide production. In another ex- lular ROS production. We foresee that the availability of ample, NO, through its high affinity for heme groups or via these specific drugs and a better biomarker-based diagnosis S-nitrosation, can act as an important modulator of NOX of the dysregulated systems will allow for more personal- (176, 315, 846) or can also modulate the insertion of heme ized treatments. in the receptor sGC. The generation of NO through NOS enzymes and oxidative pathways in normoxic conditions is Finally, antioxidants targeted to specific cells or organelles supplemented by the actions of reductive, nitrite-dependent can still become relevant in the management of oxidative pathways in hypoxia (603, 968). We also note how some stress pathologies, in particular, but not limited to, mito- proteins are able to switch, in an oxygen-dependent man- chondria-targeted antioxidants (873). The development of ner, from NO scavengers into nitrite-reducing, NO-produc- these treatments directed toward specific organs, tissue, and ing systems. The ability to interact with different oxidation cell types can be one of the next big advances in the field. states of sGC and use pharmacological tools to recover ACKNOWLEDGMENTS sGC-dependent signaling provides a new tool for the treat- ment of conditions in which NO signaling is compromised. We thank Patrick Pagano (University of Pittsburgh), Maria Further research aimed to the understanding of these dy- Eugenia Cifuentes-Pagano (University of Pittsburgh), Eric namics will be critical for the treatment of endothelial dys- E. Kelley (West Virginia University), Mauro Siragusa function and other redox imbalance-mediated pathologies. (Goethe University, Frankfurt am Main, Germany), and members of the Gladwin laboratory for critical reading of The interactions between mitochondria and ROS-produc- our manuscript. Coordinates for the sGC model were ing proteins, especially NOXs, are of particular current in- kindly provided by William Montfort (University of Ari- terest (116, 206, 234, 836). Mitochondria are a physiolog- zona). Because of the immense volume of literature in the ically significant source of ROS with important implica- field, we are sure that many important works are not in- tions in physiology, not only in cardiovascular disease but cluded in the reference list. We sincerely apologize to the in many other conditions like aging (57, 84, 136, 165, 170, authors for these unintentional omissions. 290, 864, 997). GRANTS It is also significant that many of our advances have evolved not only from the clinical field but also from basic science. This work was supported by National Institutes of Health Chemical knowledge of NO and ROS reactivity has been Grants R01 HL098032, R01 HL125886, P01 HL103455, available well before its biological relevance was estab- T32 HL110849, and T32 HL007563 (to M.T. Gladwin), lished. This knowledge is critical for the understanding of R21 ES027390 (to J. Tejero), and funding from the Institute the biology of ROS and can provide new tools and strate- for Transfusion Medicine and the Hemophilia Center of gies for the study of ROS biology (232). The studies on Western Pennsylvania (to M.T. Gladwin and S. Shiva). purified protein systems, although necessarily simplistic, have provided insights on the reactivity of most proteins DISCLOSURES involved in NO and ROS biology that have been widely validated in vivo. Conversely, the study of genetic patholo- M.T. Gladwin is a coinventor on a National Institutes of gies such as chronic granulomatous disease has helped to Health government patent application for the use of nitrite establish the relevance of NOX proteins even before these salts in the treatment of cardiovascular diseases. J. Tejero were thoroughly characterized. and S. Shiva do not declare any conflicts of interest, finan- cial or otherwise. In summary, we have progressed from a simpler approach leveraging antioxidant vitamins and ROS scavengers, REFERENCES which failed, to a more fundamental discovery of the enzy- matic sources and reaction pathways of ROS. The future 1. Aamand R, Dalsgaard T, Jensen FB, Simonsen U, Roepstorff A, Fago A. Generation of treatments need to address these issues. It is clearer now nitric oxide from nitrite by carbonic anhydrase: a possible link between metabolic activity and vasodilation. Am J Physiol Heart Circ Physiol 297: H2068–H2074, 2009. that specific inhibitors directed to the sources of ROS can doi:10.1152/ajpheart.00525.2009. yield more effective therapies. A better understanding of 2. Abdo AI, Rayner BS, van Reyk DM, Hawkins CL. Low-density lipoprotein modified NOS function has allowed the development of new strate- by myeloperoxidase oxidants induces endothelial dysfunction. Redox Biol 13: 623– gies to couple eNOS and target downstream effectors like 632, 2017. doi:10.1016/j.redox.2017.08.004.

348 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

3. Abu-Soud HM, Presta A, Mayer B, Stuehr DJ. Analysis of neuronal NO synthase 20. Alsop P, Hauton D. Oral nitrate and citrulline decrease blood pressure and increase under single-turnover conditions: conversion of Nomega-hydroxyarginine to nitric vascular conductance in young adults: a potential therapy for heart failure. Eur J Appl oxide and citrulline. Biochemistry 36: 10811–10816, 1997. doi:10.1021/bi971414g. Physiol 116: 1651–1661, 2016. doi:10.1007/s00421-016-3418-7.

4. Abu-Soud HM, Wang J, Rousseau DL, Fukuto JM, Ignarro LJ, Stuehr DJ. Neuronal 21. Altenhöfer S, Kleikers PWM, Radermacher KA, Scheurer P, Rob Hermans JJ, nitric oxide synthase self-inactivates by forming a ferrous-nitrosyl complex during Schiffers P, Ho H, Wingler K, HHHW. The NOX toolbox: validating the role aerobic catalysis. J Biol Chem 270: 22997–23006, 1995. doi:10.1074/jbc.270.39. of NADPH oxidases in physiology and disease. Cell Mol Life Sci 69: 2327–2343, 2012. 22997. doi:10.1007/s00018-012-1010-9.

5. Adachi T, Fukushima T, Usami Y, Hirano K. Binding of human xanthine oxidase to 22. Altenhöfer S, Radermacher KA, Kleikers PW, Wingler K, Schmidt HH. Evolution of sulphated glycosaminoglycans on the endothelial-cell surface. Biochem J 289: 523– NADPH oxidase inhibitors: selectivity and mechanisms for target engagement. An- 527, 1993. doi:10.1042/bj2890523. tioxid Redox Signal 23: 406–427, 2015. doi:10.1089/ars.2013.5814.

6. Adak S, Aulak KS, Stuehr DJ. Direct evidence for nitric oxide production by a nitric- 23. Amaya Y, Yamazaki K, Sato M, Noda K, Nishino T, Nishino T. Proteolytic conversion oxide synthase-like protein from Bacillus subtilis. J Biol Chem 277: 16167–16171, of xanthine dehydrogenase from the NAD-dependent type to the O2-dependent 2002. doi:10.1074/jbc.M201136200. type. Amino acid sequence of rat liver xanthine dehydrogenase and identification of the cleavage sites of the enzyme protein during irreversible conversion by trypsin. J 7. Adak S, Santolini J, Tikunova S, Wang Q, Johnson JD, Stuehr DJ. Neuronal nitric- Biol Chem 265: 14170–14175, 1990. oxide synthase mutant (Ser-1412 --Ͼ Asp) demonstrates surprising connections between heme reduction, NO complex formation, and catalysis. J Biol Chem 276: 24. Ambasta RK, Kumar P, Griendling KK, Schmidt HH, Busse R, Brandes RP. Direct 1244–1252, 2001. doi:10.1074/jbc.M006857200. interaction of the novel Nox proteins with p22phox is required for the formation of a functionally active NADPH oxidase. J Biol Chem 279: 45935–45941, 2004. doi:10. 8. Adlam VJ, Harrison JC, Porteous CM, James AM, RAJ, Murphy MP, Sammut IA. 1074/jbc.M406486200. Targeting an antioxidant to mitochondria decreases cardiac ischemia-reperfusion injury. FASEB J 19: 1088–1095, 2005. doi:10.1096/fj.05-3718com. 25. Amdahl MB, Sparacino-Watkins CE, Corti P, Gladwin MT, Tejero J. Efficient reduc- 9. Ago T, Kitazono T, Kuroda J, Kumai Y, Kamouchi M, Ooboshi H, Wakisaka M, tion of vertebrate cytoglobins by the cytochrome b5/cytochrome b5 reductase/ Kawahara T, Rokutan K, Ibayashi S, Iida M. NAD(P)H oxidases in rat basilar arterial NADH system. Biochemistry 56: 3993–4004, 2017. doi:10.1021/acs.biochem. endothelial cells. Stroke 36: 1040–1046, 2005. doi:10.1161/01.STR.0000163111. 7b00224. 05825.0b. 26. Ameziane-El-Hassani R, Morand S, Boucher JL, Frapart YM, Apostolou D, Agnandji 10. Ago T, Kitazono T, Ooboshi H, Iyama T, Han YH, Takada J, Wakisaka M, Ibayashi S, D, Gnidehou S, Ohayon R, Noël-Hudson MS, Francon J, Lalaoui K, Virion A, Dupuy ϩ Utsumi H, Iida M. Nox4 as the major catalytic component of an endothelial NA- C. Dual oxidase-2 has an intrinsic Ca2 -dependent H2O2-generating activity. J Biol D(P)H oxidase. Circulation 109: 227–233, 2004. doi:10.1161/01.CIR.0000105680. Chem 280: 30046–30054, 2005. doi:10.1074/jbc.M500516200. 92873.70. 27. Anderson EJ, Lustig ME, Boyle KE, Woodlief TL, Kane DA, Lin CT, Price JW III, Kang 11. Ago T, Kuroda J, Pain J, Fu C, Li H, Sadoshima J. Upregulation of Nox4 by hypertro- L, Rabinovitch PS, Szeto HH, Houmard JA, Cortright RN, Wasserman DH, Neufer phic stimuli promotes apoptosis and mitochondrial dysfunction in cardiac myocytes. PD. Mitochondrial H2O2 emission and cellular redox state link excess fat intake to Circ Res 106: 1253–1264, 2010. doi:10.1161/CIRCRESAHA.109.213116. insulin resistance in both rodents and humans. J Clin Invest 119: 573–581, 2009. doi:10.1172/JCI37048. 12. Aisaka K, Gross SS, Griffith OW, Levi R. L-arginine availability determines the dura- tion of acetylcholine-induced systemic vasodilation in vivo. Biochem Biophys Res Com- 28. Anderssohn M, Rosenberg M, Schwedhelm E, Zugck C, Lutz M, Lüneburg N, Frey mun 163: 710–717, 1989. doi:10.1016/0006-291X(89)92281-X. N, Böger RH. The L-Arginine-asymmetric dimethylarginine ratio is an independent predictor of mortality in dilated cardiomyopathy. J Card Fail 18: 904–911, 2012. 13. Al Ghouleh I, Khoo NKH, Knaus UG, Griendling KK, Touyz RM, Thannickal VJ, doi:10.1016/j.cardfail.2012.10.011. Barchowsky A, Nauseef WM, Kelley EE, Bauer PM, Darley-Usmar V, Shiva S, Cifu- entes-Pagano E, Freeman BA, Gladwin MT, Pagano PJ. Oxidases and peroxidases in 29. Annedi SC, Maddaford SP, Ramnauth J, Renton P, Rybak T, Silverman S, Rakhit S, cardiovascular and lung disease: new concepts in reactive oxygen species signaling. Mladenova G, Dove P, Andrews JS, Zhang D, Porreca F. Discovery of a potent, orally Free Radic Biol Med 51: 1271–1288, 2011. doi:10.1016/j.freeradbiomed.2011.06. bioavailable and highly selective human neuronal nitric oxide synthase (nNOS) inhib- 011. itor, N-(1-(piperidin-4-yl)indolin-5-yl)thiophene-2-carboximidamide as a pre-clini- cal development candidate for the treatment of migraine. Eur J Med Chem 55: 94– 14. Al Ghouleh I, Meijles DN, Mutchler S, Zhang Q, Sahoo S, Gorelova A, Henrich 107, 2012. doi:10.1016/j.ejmech.2012.07.006. Amaral J, Rodríguez AI, Mamonova T, Song GJ, Bisello A, Friedman PA, Cifuentes- Pagano ME, Pagano PJ. Binding of EBP50 to Nox organizing subunit p47phox is 30. Anter E, Thomas SR, Schulz E, Shapira OM, Vita JA, Keaney JF Jr. Activation of pivotal to cellular reactive species generation and altered vascular phenotype. Proc endothelial nitric-oxide synthase by the p38 MAPK in response to black tea poly- Natl Acad Sci USA 113: E5308–E5317, 2016. doi:10.1073/pnas.1514161113. phenols. J Biol Chem 279: 46637–46643, 2004. doi:10.1074/jbc.M405547200.

15. Albrich JM, McCarthy CA, Hurst JK. Biological reactivity of hypochlorous acid: im- 31. Antoniades C, Bakogiannis C, Leeson P, Guzik TJ, Zhang M-H, Tousoulis D, Anto- plications for microbicidal mechanisms of leukocyte myeloperoxidase. Proc Natl nopoulos AS, Demosthenous M, Marinou K, Hale A, Paschalis A, Psarros C, Trian- Acad Sci USA 78: 210–214, 1981. doi:10.1073/pnas.78.1.210. tafyllou C, Bendall J, Casadei B, Stefanadis C, Channon KM. Rapid, direct effects of statin treatment on arterial redox state and nitric oxide bioavailability in human 16. Alderton WK, Angell AD, Craig C, Dawson J, Garvey E, Moncada S, Monkhouse J, atherosclerosis via tetrahydrobiopterin-mediated endothelial nitric oxide synthase Rees D, Russell LJ, Russell RJ, Schwartz S, Waslidge N, Knowles RG. GW274150 and coupling. Circulation 124: 335–345, 2011. doi:10.1161/CIRCULATIONAHA.110. GW273629 are potent and highly selective inhibitors of inducible nitric oxide syn- 985150. thase in vitro and in vivo. Br J Pharmacol 145: 301–312, 2005. doi:10.1038/sj.bjp. 0706168. 32. Antoniades C, Shirodaria C, Crabtree M, Rinze R, Alp N, Cunnington C, Diesch J, Tousoulis D, Stefanadis C, Leeson P, Ratnatunga C, Pillai R, Channon KM. Altered 17. Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function plasma versus vascular biopterins in human atherosclerosis reveal relationships be- and inhibition. Biochem J 357: 593–615, 2001. doi:10.1042/bj3570593. tween endothelial nitric oxide synthase coupling, endothelial function, and inflam- 18. Aldieri E, Riganti C, Polimeni M, Gazzano E, Lussiana C, Campia I, Ghigo D. Classical mation. Circulation 116: 2851–2859, 2007. doi:10.1161/CIRCULATIONAHA.107. inhibitors of NOX NAD(P)H oxidases are not specific. Curr Drug Metab 9: 686–696, 704155. 2008. doi:10.2174/138920008786049285. 33. Antoniades C, Shirodaria C, Warrick N, Cai S, de Bono J, Lee J, Leeson P, Neubauer 19. Alp NJ, McAteer MA, Khoo J, Choudhury RP, Channon KM. Increased endothelial S, Ratnatunga C, Pillai R, Refsum H, Channon KM. 5-methyltetrahydrofolate rapidly tetrahydrobiopterin synthesis by targeted transgenic GTP-cyclohydrolase I overex- improves endothelial function and decreases superoxide production in human pression reduces endothelial dysfunction and atherosclerosis in ApoE-knockout vessels: effects on vascular tetrahydrobiopterin availability and endothelial nitric mice. Arterioscler Thromb Vasc Biol 24: 445–450, 2004. doi:10.1161/01.ATV. oxide synthase coupling. Circulation 114: 1193–1201, 2006. doi:10.1161/ 0000115637.48689.77. CIRCULATIONAHA.106.612325.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 349 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

34. Aon MA, Stanley BA, Sivakumaran V, Kembro JM, O’Rourke B, Paolocci N, Cortassa 53. Baker LM, Baker PR, Golin-Bisello F, Schopfer FJ, Fink M, Woodcock SR, Branchaud S. Glutathione/thioredoxin systems modulate mitochondrial H2O2 emission: an ex- BP, Radi R, Freeman BA. Nitro-fatty acid reaction with glutathione and cysteine. perimental-computational study. J Gen Physiol 139: 479–491, 2012. doi:10.1085/jgp. Kinetic analysis of thiol alkylation by a Michael addition reaction. J Biol Chem 282: 201210772. 31085–31093, 2007. doi:10.1074/jbc.M704085200.

35. Aoyama T, Paik Y-H, Watanabe S, Laleu B, Gaggini F, Fioraso-Cartier L, Molango S, 54. Baker PR, Schopfer FJ, Sweeney S, Freeman BA. Red cell membrane and plasma Heitz F, Merlot C, Szyndralewiez C, Page P, Brenner DA. Nicotinamide adenine linoleic acid nitration products: synthesis, clinical identification, and quantitation. Proc dinucleotide phosphate oxidase in experimental liver fibrosis: GKT137831 as a novel Natl Acad Sci USA 101: 11577–11582, 2004. doi:10.1073/pnas.0402587101. potential therapeutic agent. Hepatology 56: 2316–2327, 2012. doi:10.1002/hep. 25938. 55. Baker PRS, Lin Y, Schopfer FJ, Woodcock SR, Groeger AL, Batthyany C, Sweeney S, Long MH, Iles KE, Baker LMS, Branchaud BP, Chen YE, Freeman BA. Fatty acid 36. Arnold WP, Mittal CK, Katsuki S, Murad F. Nitric oxide activates guanylate cyclase transduction of nitric oxide signaling: multiple nitrated unsaturated fatty acid deriv- and increases guanosine 3=:5=-cyclic monophosphate levels in various tissue prepa- atives exist in human blood and urine and serve as endogenous peroxisome prolif- rations. Proc Natl Acad Sci USA 74: 3203–3207, 1977. doi:10.1073/pnas.74.8.3203. erator-activated receptor ligands. J Biol Chem 280: 42464–42475, 2005. doi:10. 1074/jbc.M504212200. 37. Arteel GE, Briviba K, Sies H. Protection against peroxynitrite. FEBS Lett 445: 226– 230, 1999. doi:10.1016/S0014-5793(99)00073-3. 56. Baker TA, Milstien S, Katusic ZS. Effect of vitamin C on the availability of tetrahy- drobiopterin in human endothelial cells. J Cardiovasc Pharmacol 37: 333–338, 2001. 38. Ascenzi P, di Masi A, Leboffe L, Fiocchetti M, Nuzzo MT, Brunori M, Marino M. doi:10.1097/00005344-200103000-00012. Neuroglobin: From structure to function in health and disease. Mol Aspects Med 52: 1–48, 2016. doi:10.1016/j.mam.2016.10.004. 57. Balaban RS, Nemoto S, Finkel T. Mitochondria, oxidants, and aging. Cell 120: 483– 495, 2005. doi:10.1016/j.cell.2005.02.001. 39. Ascenzi P, Gustincich S, Marino M. Mammalian nerve globins in search of functions. IUBMB Life 66: 268–276, 2014. doi:10.1002/iub.1267. 58. Baldus S, Müllerleile K, Chumley P, Steven D, Rudolph V, Lund GK, Staude HJ, Stork A, Köster R, Kähler J, Weiss C, Münzel T, Meinertz T, Freeman BA, Heitzer T. 40. Ascenzi P, Leboffe L, Pesce A, Ciaccio C, Sbardella D, Bolognesi M, Coletta M. Inhibition of xanthine oxidase improves myocardial contractility in patients with Nitrite-reductase and peroxynitrite isomerization activities of Methanosarcina ace- ischemic cardiomyopathy. Free Radic Biol Med 41: 1282–1288, 2006. doi:10.1016/j. tivorans protoglobin. PLoS One 9: e95391, 2014. doi:10.1371/journal.pone.0095391. freeradbiomed.2006.07.010. 41. Ascenzi P, Marino M, Polticelli F, Santucci R, Coletta M. Cardiolipin modulates 59. Balligand JL, Feron O, Dessy C. eNOS activation by physical forces: from short-term allosterically the nitrite reductase activity of horse heart cytochrome c. J Biol Inorg regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol Rev Chem 19: 1195–1201, 2014. doi:10.1007/s00775-014-1175-9. 89: 481–534, 2009. doi:10.1152/physrev.00042.2007. 42. Ascenzi P, Sbardella D, Fiocchetti M, Santucci R, Coletta M. NO2(-)-mediated ni- 60. Balzer J, Rassaf T, Heiss C, Kleinbongard P, Lauer T, Merx M, Heussen N, Gross HB, trosylation of ferrous microperoxidase-11. J Inorg Biochem 153: 121–127, 2015. Keen CL, Schroeter H, Kelm M. Sustained benefits in vascular function through doi:10.1016/j.jinorgbio.2015.06.022. flavanol-containing cocoa in medicated diabetic patients a double-masked, random- 43. Ascenzi P, Tundo GR, Fanali G, Coletta M, Fasano M. Warfarin modulates the nitrite ized, controlled trial. J Am Coll Cardiol 51: 2141–2149, 2008. doi:10.1016/j.jacc.2008. reductase activity of ferrous human serum heme-albumin. J Biol Inorg Chem 18: 01.059. 939–946, 2013. doi:10.1007/s00775-013-1040-2. 61. Bánfi B, Clark RA, Steger K, Krause KH. Two novel proteins activate superoxide 44. Aslan M, Dogan S. Proteomic detection of nitroproteins as potential biomarkers for generation by the NADPH oxidase NOX1. J Biol Chem 278: 3510–3513, 2003. cardiovascular disease. J Proteomics 74: 2274–2288, 2011. doi:10.1016/j.jprot.2011. doi:10.1074/jbc.C200613200. 05.029. 62. Bánfi B, Molnár G, Maturana A, Steger K, Hegedûs B, Demaurex N, Krause KHA. A 45. Aslan M, Ryan TM, Adler B, Townes TM, Parks DA, Thompson JA, Tousson A, Ca(2ϩ)-activated NADPH oxidase in testis, spleen, and lymph nodes. J Biol Chem Gladwin MT, Patel RP, Tarpey MM, Batinic-Haberle I, White CR, Freeman BA. 276: 37594–37601, 2001. doi:10.1074/jbc.M103034200. Oxygen radical inhibition of nitric oxide-dependent vascular function in sickle cell 63. Bánfi B, Tirone F, Durussel I, Knisz J, Moskwa P, Molnár GZ, Krause KH, Cox JA. disease. Proc Natl Acad Sci USA 98: 15215–15220, 2001. doi:10.1073/pnas. Mechanism of Ca2ϩ activation of the NADPH oxidase 5 (NOX5). J Biol Chem 279: 221292098. 18583–18591, 2004. doi:10.1074/jbc.M310268200. 46. Augusto O, Bonini MG, Amanso AM, Linares E, Santos CC, De Menezes SL. Nitro- 64. Barry-Lane PA, Patterson C, van der Merwe M, Hu Z, Holland SM, Yeh ETH, Runge gen dioxide and carbonate radical anion: two emerging radicals in biology. Free Radic MS. p47phox is required for atherosclerotic lesion progression in ApoE(Ϫ/Ϫ) mice. Biol Med 32: 841–859, 2002. doi:10.1016/S0891-5849(02)00786-4. J Clin Invest 108: 1513–1522, 2001. doi:10.1172/JCI200111927. 47. Aulak KS, Miyagi M, Yan L, West KA, Massillon D, Crabb JW, Stuehr DJ. Proteomic 65. Bartesaghi S, D, Martinez DM, Petruk A, Demicheli V, Trujillo M, Martí MA, method identifies proteins nitrated in vivo during inflammatory challenge. Proc Natl Estrín DA, Radi R. Tyrosine oxidation and nitration in transmembrane peptides is Acad Sci USA 98: 12056–12061, 2001. doi:10.1073/pnas.221269198. connected to lipid peroxidation. Arch Biochem Biophys 622: 9–25, 2017. doi:10.1016/ 48. Babior BM, Curnutte JT, Kipnes BS. Pyridine nucleotide-dependent superoxide pro- j.abb.2017.04.006. duction by a cell-free system from human granulocytes. J Clin Invest 56: 1035–1042, 1975. doi:10.1172/JCI108150. 66. Bartesaghi S, Valez V, Trujillo M, Peluffo G, Romero N, Zhang H, Kalyanaraman B, Radi R. Mechanistic studies of peroxynitrite-mediated tyrosine nitration in mem- 49. Babior BM, Kipnes RS. Superoxide-forming enzyme from human neutrophils: evi- branes using the hydrophobic probe N-t-BOC-L-tyrosine tert-butyl ester. Biochem- dence for a flavin requirement. Blood 50: 517–524, 1977. istry 45: 6813–6825, 2006. doi:10.1021/bi060363x.

50. Bach A. Zur Kenntnis der Reduktionsfermente. I. Mitteilung über das Schardinger- 67. Bast A, Haenen GRMM. Ten misconceptions about antioxidants. Trends Pharmacol Enzym (Perhydridase). Biochem Z 31: 443–449, 1911. Sci 34: 430–436, 2013. doi:10.1016/j.tips.2013.05.010.

51. Baek KJ, Thiel BA, Lucas S, Stuehr DJ. Macrophage nitric oxide synthase subunits. 68. Basu S, Azarova NA, Font MD, King SB, Hogg N, Gladwin MT, Shiva S, Kim-Shapiro Purification, characterization, and role of prosthetic groups and substrate in regulat- DB. Nitrite reductase activity of cytochrome c. J Biol Chem 283: 32590–32597, ing their association into a dimeric enzyme. J Biol Chem 268: 21120–21129, 1993. 2008. doi:10.1074/jbc.M806934200.

52. Baker CSR, Hall RJC, Evans TJ, Pomerance A, Maclouf J, Creminon C, Yacoub MH, 69. Basu S, Grubina R, Huang J, Conradie J, Huang Z, Jeffers A, Jiang A, He X, Azarov I, Polak JM. Cyclooxygenase-2 is widely expressed in atherosclerotic lesions affecting Seibert R, Mehta A, Patel R, King SB, Hogg N, Ghosh A, Gladwin MT, Kim-Shapiro

native and transplanted human coronary arteries and colocalizes with inducible nitric DB. Catalytic generation of N2O3 by the concerted nitrite reductase and anhydrase oxide synthase and nitrotyrosine particularly in macrophages. Arterioscler Thromb activity of hemoglobin. Nat Chem Biol 3: 785–794, 2007. doi:10.1038/nchembio. Vasc Biol 19: 646–655, 1999. doi:10.1161/01.ATV.19.3.646. 2007.46.

350 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

70. Basuroy S, Bhattacharya S, Leffler CW, Parfenova H. Nox4 NADPH oxidase medi- 87. Bendall JK, Douglas G, McNeill E, Channon KM, Crabtree MJ. Tetrahydrobiopterin ates oxidative stress and apoptosis caused by TNF-alpha in cerebral vascular endo- in cardiovascular health and disease. Antioxid Redox Signal 20: 3040–3077, 2014. thelial cells. Am J Physiol Cell Physiol 296: C422–C432, 2009. doi:10.1152/ajpcell. doi:10.1089/ars.2013.5566. 00381.2008. 88. Bendall JK, Rinze R, Adlam D, Tatham AL, de Bono J, Wilson N, Volpi E, Channon 71. Battelli MG, Bolognesi A, Polito L. Pathophysiology of circulating xanthine oxi- KM. Endothelial Nox2 overexpression potentiates vascular oxidative stress and he- doreductase: new emerging roles for a multi-tasking enzyme. Biochim Biophys Acta modynamic response to angiotensin II: studies in endothelial-targeted Nox2 trans- 1842: 1502–1517, 2014. doi:10.1016/j.bbadis.2014.05.022. genic mice. Circ Res 100: 1016–1025, 2007. doi:10.1161/01.RES.0000263381. 83835.7b. 72. Battelli MG, Polito L, Bolognesi A. Xanthine oxidoreductase in atherosclerosis pathogenesis: not only oxidative stress. Atherosclerosis 237: 562–567, 2014. doi:10. 89. Benjamin N, O’Driscoll F, Dougall H, Duncan C, Smith L, Golden M, McKenzie H. 1016/j.atherosclerosis.2014.10.006. Stomach NO synthesis. Nature 368: 502, 1994. doi:10.1038/368502a0.

73. Batthyány C, Bartesaghi S, Mastrogiovanni M, Lima A, Demicheli V, Radi R. Tyrosine- 90. Bergt C, Pennathur S, Fu X, Byun J, O’Brien K, McDonald TO, Singh P, Ananthara- Nitrated Proteins: Proteomic and Bioanalytical Aspects. Antioxid Redox Signal 26: maiah GM, Chait A, Brunzell J, Geary RL, Oram JF, Heinecke JW. The myeloperox- 313–328, 2017. doi:10.1089/ars.2016.6787. idase product hypochlorous acid oxidizes HDL in the human artery wall and impairs ABCA1-dependent cholesterol transport. Proc Natl Acad Sci USA 101: 13032–13037, 74. Bauer PM, Fulton D, Boo YC, Sorescu GP, Kemp BE, Jo H, Sessa WC. Compensa- 2004. doi:10.1073/pnas.0405292101. tory phosphorylation and protein-protein interactions revealed by loss of function and gain of function mutants of multiple serine phosphorylation sites in endothelial 91. Berkowitz DE, White R, Li D, Minhas KM, Cernetich A, Kim S, Burke S, Shoukas AA, Nyhan D, Champion HC, Hare JM. Arginase reciprocally regulates nitric oxide syn- nitric-oxide synthase. J Biol Chem 278: 14841–14849, 2003. doi:10.1074/jbc. thase activity and contributes to endothelial dysfunction in aging blood vessels. Cir- M211926200. culation 108: 2000–2006, 2003. doi:10.1161/01.CIR.0000092948.04444.C7. 75. Baum C, Johannsen SS, Zeller T, Atzler D, Ojeda FM, Wild PS, Sinning CR, Lackner 92. Beuve A. Thiol-Based Redox Modulation of Soluble Guanylyl Cyclase, the Nitric KJ, Gori T, Schwedhelm E, Böger RH, Blankenberg S, Münzel T, Schnabel RB; Oxide Receptor. Antioxid Redox Signal 26: 137–149, 2017. doi:10.1089/ars.2015. Gutenberg Health Study investigators. ADMA and arginine derivatives in relation to 6591. non-invasive vascular function in the general population. Atherosclerosis 244: 149– 156, 2016. doi:10.1016/j.atherosclerosis.2015.10.101. 93. Beuve A, Wu C, Cui C, Liu T, Jain MR, Huang C, Yan L, Kholodovych V, Li H. Identification of novel S-nitrosation sites in soluble guanylyl cyclase, the nitric oxide 76. Becker MA, Schumacher HR, MacDonald PA, Lloyd E, Lademacher C. Clinical effi- receptor. J Proteomics 138: 40–47, 2016. doi:10.1016/j.jprot.2016.02.009. cacy and safety of successful longterm urate lowering with febuxostat or allopurinol in subjects with gout. J Rheumatol 36: 1273–1282, 2009. doi:10.3899/jrheum. 94. Bibli SI, Zhou Z, Zukunft S, Fisslthaler B, Andreadou I, Szabo C, Brouckaert P, 080814. Fleming I, Papapetropoulos A. Tyrosine phosphorylation of eNOS regulates myo- cardial survival after an ischaemic insult: role of PYK2. Cardiovasc Res 113: 926–937, 77. Becker MA, Schumacher HR Jr, Wortmann RL, MacDonald PA, Eustace D, Palo WA, 2017. doi:10.1093/cvr/cvx058. Streit J, Joseph-Ridge N. Febuxostat compared with allopurinol in patients with hyperuricemia and gout. N Engl J Med 353: 2450–2461, 2005. doi:10.1056/ 95. Billecke SS, Bender AT, Kanelakis KC, Murphy PJM, Lowe ER, Kamada Y, Pratt WB, NEJMoa050373. Osawa Y. hsp90 is required for heme binding and activation of apo-neuronal nitric- oxide synthase: geldanamycin-mediated oxidant generation is unrelated to any ac- 78. Beckman JS, Koppenol WH. Nitric oxide, superoxide, and peroxynitrite: the good, tion of hsp90. J Biol Chem 277: 20504–20509, 2002. doi:10.1074/jbc.M201940200. the bad, and ugly. Am J Physiol 271: C1424–C1437, 1996. doi:10.1152/ajpcell.1996. 271.5.C1424. 96. Bir SC, Shen X, Kavanagh TJ, Kevil CG, Pattillo CB. Control of angiogenesis dictated by picomolar superoxide levels. Free Radic Biol Med 63: 135–142, 2013. doi:10.1016/ 79. Bedard K, Krause K-H. The NOX family of ROS-generating NADPH oxidases: j.freeradbiomed.2013.05.015. physiology and pathophysiology. Physiol Rev 87: 245–313, 2007. doi:10.1152/ physrev.00044.2005. 97. Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C. Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic 80. Behrends S, Harteneck C, Schultz G, Koesling D. A variant of the ␣ 2 subunit of review and meta-analysis. JAMA 297: 842–857, 2007. doi:10.1001/jama.297.8.842. soluble guanylyl cyclase contains an insert homologous to a region within adenylyl cyclases and functions as a dominant negative protein. J Biol Chem 270: 21109– 98. Björne H H, Petersson J, Phillipson M, Weitzberg E, Holm L, Lundberg JO. Nitrite in 21113, 1995. doi:10.1074/jbc.270.36.21109. saliva increases gastric mucosal blood flow and mucus thickness. J Clin Invest 113: 106–114, 2004. doi:10.1172/JCI19019. 81. BelAiba RS, Djordjevic T, Petry A, Diemer K, Bonello S, Banfi B, Hess J, Pogrebniak A, Bickel C, Görlach A. NOX5 variants are functionally active in endothelial cells. 99. Björne H, Weitzberg E, Lundberg JO. Intragastric generation of antimicrobial nitro- Free Radic Biol Med 42: 446–459, 2007. doi:10.1016/j.freeradbiomed.2006.10.054. gen oxides from saliva–physiological and therapeutic considerations. Free Radic Biol Med 41: 1404–1412, 2006. doi:10.1016/j.freeradbiomed.2006.07.020. 82. Belhassen L, Feron O, Kaye DM, Michel T, Kelly RA. Regulation by cAMP of post- 100. Block K, Gorin Y, Abboud HE. Subcellular localization of Nox4 and regulation in translational processing and subcellular targeting of endothelial nitric-oxide synthase diabetes. Proc Natl Acad Sci USA 106: 14385–14390, 2009. doi:10.1073/pnas. (type 3) in cardiac myocytes. J Biol Chem 272: 11198–11204, 1997. doi:10.1074/jbc. 0906805106. 272.17.11198. 101. Boccini F, Herold S. Mechanistic studies of the oxidation of oxyhemoglobin by 83. Belik J. Riociguat, an oral soluble guanylate cyclase stimulator for the treatment of peroxynitrite. Biochemistry 43: 16393–16404, 2004. doi:10.1021/bi0482250. pulmonary hypertension. Curr Opin Investig Drugs 10: 971–979, 2009. 102. Bode-Böger SM, Scalera F, Ignarro LJ. The L-arginine paradox: Importance of the 84. Bell EL, Klimova TA, Eisenbart J, Moraes CT, Murphy MP, Budinger GRS, Chandel L-arginine/asymmetrical dimethylarginine ratio. Pharmacol Ther 114: 295–306, 2007. NS. The Qo site of the mitochondrial complex III is required for the transduction of doi:10.1016/j.pharmthera.2007.03.002. hypoxic signaling via reactive oxygen species production. J Cell Biol 177: 1029–1036, 2007. doi:10.1083/jcb.200609074. 103. Böger RH. The pharmacodynamics of L-arginine. J Nutr 137, Suppl 2: 1650S–1655S, 2007. doi:10.1093/jn/137.6.1650S. 85. Bellamy TC, Wood J, Goodwin DA, Garthwaite J. Rapid desensitization of the nitric oxide receptor, soluble guanylyl cyclase, underlies diversity of cellular cGMP re- 104. Böger RH, Sullivan LM, Schwedhelm E, Wang TJ, Maas R, Benjamin EJ, Schulze F, sponses. Proc Natl Acad Sci USA 97: 2928–2933, 2000. doi:10.1073/pnas.97.6.2928. Xanthakis V, Benndorf RA, Vasan RS. Plasma asymmetric dimethylarginine and inci- dence of cardiovascular disease and death in the community. Circulation 119: 1592– 86. Ben Mkaddem S, Pedruzzi E, Werts C, Coant N, Bens M, Cluzeaud F, Goujon JM, 1600, 2009. doi:10.1161/CIRCULATIONAHA.108.838268. Ogier-Denis E, Vandewalle A. Heat shock protein gp96 and NAD(P)H oxidase 4 play key roles in Toll-like receptor 4-activated apoptosis during renal ischemia/reperfu- 105. Boerrigter G, Costello-Boerrigter LC, Cataliotti A, Tsuruda T, Harty GJ, Lapp H, sion injury. Cell Death Differ 17: 1474–1485, 2010. doi:10.1038/cdd.2010.26. Stasch JP, Burnett JC Jr. Cardiorenal and humoral properties of a novel direct soluble

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 351 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

guanylate cyclase stimulator BAY 41-2272 in experimental congestive heart failure. 125. Bruegger JJ, Smith BC, Wynia-Smith SL, Marletta MA. Comparative and integrative Circulation 107: 686–689, 2003. doi:10.1161/01.CIR.0000055737.15443.F8. metabolomics reveal that S-nitrosation inhibits physiologically relevant metabolic enzymes. J Biol Chem 293: 6282–6296, 2018. doi:10.1074/jbc.M117.817700. 106. Böger RH. Asymmetric dimethylarginine, an endogenous inhibitor of nitric oxide synthase, explains the “L-arginine paradox” and acts as a novel cardiovascular risk 126. Buchwalow IB, Podzuweit T, Bocker W, Samoilova VE, Thomas S, Wellner M, Baba factor. J Nutr 134, Suppl: 2842S–2847S, 2004. doi:10.1093/jn/134.10.2842S. HA, Robenek H, Schnekenburger J, Lerch MM. Vascular smooth muscle and nitric oxide synthase. FASEB J 16: 500–508, 2002. doi:10.1096/fj.01-0842com. 107. Böger RH, Bode-Böger SM, Szuba A, Tsao PS, Chan JR, Tangphao O, Blaschke TF, Cooke JP. Asymmetric dimethylarginine (ADMA): a novel risk factor for endothelial 127. Buchwalow IB, Schulze W, Karczewski P, Kostic MM, Wallukat G, Morwinski R, dysfunction: its role in hypercholesterolemia. Circulation 98: 1842–1847, 1998. doi: Krause E-G, Müller J, Paul M, Slezak J, Luft FC, Haller H. Inducible nitric oxide 10.1161/01.CIR.98.18.1842. synthase in the myocard. Mol Cell Biochem 217: 73–82, 2001. doi:10.1023/ A:1007286602865. 108. Bokoch GM, Diebold BA. Current molecular models for NADPH oxidase regulation by Rac GTPase. Blood 100: 2692–2696, 2002. doi:10.1182/blood-2002-04-1149. 128. Buchwalow IB, Schulze W, Kostic MM, Wallukat G, Morwinski R. Intracellular local- ization of inducible nitric oxide synthase in neonatal rat cardiomyocytes in culture. 109. Bolli R. Cardioprotective function of inducible nitric oxide synthase and role of nitric Acta Histochem 99: 231–240, 1997. doi:10.1016/S0065-1281(97)80046-3. oxide in myocardial ischemia and preconditioning: an overview of a decade of re- 129. Budworth J, Meillerais S, Charles I, Powell K. Tissue distribution of the human search. J Mol Cell Cardiol 33: 1897–1918, 2001. doi:10.1006/jmcc.2001.1462. soluble guanylate cyclases. Biochem Biophys Res Commun 263: 696–701, 1999. doi: 110. Bondi CD, Manickam N, Lee DY, Block K, Gorin Y, Abboud HE, Barnes JL. NA- 10.1006/bbrc.1999.1444. D(P)H oxidase mediates TGF-beta1-induced activation of kidney myofibroblasts. J 130. Bunik VI, Sievers C. Inactivation of the 2-oxo acid dehydrogenase complexes upon Am Soc Nephrol 21: 93–102, 2010. doi:10.1681/ASN.2009020146. generation of intrinsic radical species. Eur J Biochem 269: 5004–5015, 2002. doi:10. 111. Bonini MG, Miyamoto S, Di Mascio P, Augusto O. Production of the carbonate 1046/j.1432-1033.2002.03204.x. radical anion during xanthine oxidase turnover in the presence of bicarbonate. J Biol 131. Burger DE, Lu X, Lei M, Xiang FL, Hammoud L, Jiang M, Wang H, Jones DL, Sims SM, Chem 279: 51836–51843, 2004. doi:10.1074/jbc.M406929200. Feng Q. Neuronal nitric oxide synthase protects against myocardial infarction-in- duced ventricular arrhythmia and mortality in mice. Circulation 120: 1345–1354, 112. Bonini MG, Radi R, -Sueta G, AM, Augusto O. Direct EPR detection 2009. doi:10.1161/CIRCULATIONAHA.108.846402. of the carbonate radical anion produced from peroxynitrite and carbon dioxide. J Biol Chem 274: 10802–10806, 1999. doi:10.1074/jbc.274.16.10802. 132. Burmester T, Ebner B, Weich B, Hankeln T. Cytoglobin: a novel globin type ubiqui- tously expressed in vertebrate tissues. Mol Biol Evol 19: 416–421, 2002. doi:10. 113. Borniquel S, Jansson EA, Cole MP, Freeman BA, Lundberg JO. Nitrated oleic acid 1093/oxfordjournals.molbev.a004096. up-regulates PPARgamma and attenuates experimental inflammatory bowel disease. Free Radic Biol Med 48: 499–505, 2010. doi:10.1016/j.freeradbiomed.2009.11.014. 133. Burmester T, Hankeln T. Function and evolution of vertebrate globins. Acta Physiol (Oxf) 211: 501–514, 2014. doi:10.1111/apha.12312. 114. Boulanger CM, Heymes C, Benessiano J, Geske RS, Lévy BI, Vanhoutte PM. Neuro- nal nitric oxide synthase is expressed in rat vascular smooth muscle cells: activation 134. Burmester T, Weich B, Reinhardt S, Hankeln T. A vertebrate globin expressed in the by angiotensin II in hypertension. Circ Res 83: 1271–1278, 1998. doi:10.1161/01.RES. brain. Nature 407: 520–523, 2000. doi:10.1038/35035093. 83.12.1271. 135. Burritt JB, Busse SC, Gizachew D, Siemsen DW, Quinn MT, Bond CW, Dratz EA, 115. Boveris A, Chance B. The mitochondrial generation of hydrogen peroxide. General Jesaitis AJ. Antibody imprint of a membrane protein surface. Phagocyte flavocyto- properties and effect of hyperbaric oxygen. Biochem J 134: 707–716, 1973. doi:10. chrome b. J Biol Chem 273: 24847–24852, 1998. doi:10.1074/jbc.273.38.24847. 1042/bj1340707. 136. Burwell LS, Nadtochiy SM, Brookes PS. Cardioprotection by metabolic shut-down 116. Brandes RP. Triggering mitochondrial radical release: a new function for NADPH and gradual wake-up. J Mol Cell Cardiol 46: 804–810, 2009. doi:10.1016/j.yjmcc. oxidases. Hypertension 45: 847–848, 2005. doi:10.1161/01.HYP.0000165019. 2009.02.026. 32059.b2. 137. Butler R, Morris AD, Belch JJF, Hill A, Struthers AD. Allopurinol normalizes endo- 117. Brandes RP, Koddenberg G, Gwinner W, Kim D-y, Kruse H-J, Busse R, Mügge A. thelial dysfunction in type 2 diabetics with mild hypertension. Hypertension 35: 746– Role of increased production of superoxide anions by NAD(P)H oxidase and xan- 751, 2000. doi:10.1161/01.HYP.35.3.746. thine oxidase in prolonged endotoxemia. Hypertension 33: 1243–1249, 1999. doi: 138. Buxton GV, Elliot AJ. Rate-constant for reaction of hydroxyl radicals with bicarbon- 10.1161/01.HYP.33.5.1243. ate ions. Radiat Phys Chem 27: 241–243, 1986. doi:10.1016/1359-0197(86)90059-7. 118. Brar SS, Kennedy TP, Sturrock AB, Huecksteadt TP, Quinn MT, Whorton AR, 139. Cacanyiova S, Kristek F, Gerova M, Krenek P, Klimas J. Effect of chronic nNOS Hoidal JR. An NAD(P)H oxidase regulates growth and transcription in melanoma inhibition on blood pressure, vasoactivity, and arterial wall structure in Wistar rats. cells. Am J Physiol Cell Physiol 282: C1212–C1224, 2002. doi:10.1152/ajpcell.00496. Nitric Oxide 20: 304–310, 2009. doi:10.1016/j.niox.2009.03.002. 2001. 140. Cai B, Lin Y, Xue XH, Fang L, Wang N, Wu ZY. TAT-mediated delivery of neuro- 119. Brash AR. Lipoxygenases: occurrence, functions, catalysis, and acquisition of sub- globin protects against focal cerebral ischemia in mice. Exp Neurol 227: 224–231, strate. J Biol Chem 274: 23679–23682, 1999. doi:10.1074/jbc.274.34.23679. 2011. doi:10.1016/j.expneurol.2010.11.009.

120. Bredt DS, Hwang PM, Snyder SH. Localization of nitric oxide synthase indicating a 141. Cai H, Davis ME, Drummond GR, Harrison DG. Induction of endothelial NO syn- neural role for nitric oxide. Nature 347: 768–770, 1990. doi:10.1038/347768a0. thase by hydrogen peroxide via a Ca(2ϩ)/calmodulin-dependent protein kinase II/janus kinase 2-dependent pathway. Arterioscler Thromb Vasc Biol 21: 1571–1576, 121. Bretón-Romero R, González de Orduña C, Romero N, Sánchez-Gómez FJ, de 2001. doi:10.1161/hq1001.097028. Álvaro C, Porras A, Rodríguez-Pascual F, Laranjinha J, Radi R, Lamas S. Critical role of hydrogen peroxide signaling in the sequential activation of p38 MAPK and eNOS 142. Cai H, Griendling KK, Harrison DG. The vascular NAD(P)H oxidases as therapeutic in laminar shear stress. Free Radic Biol Med 52: 1093–1100, 2012. doi:10.1016/j. targets in cardiovascular diseases. Trends Pharmacol Sci 24: 471–478, 2003. doi:10. freeradbiomed.2011.12.026. 1016/S0165-6147(03)00233-5.

122. Brewer GJ. Iron and copper toxicity in diseases of aging, particularly atherosclerosis 143. Cai S, Alp NJ, McDonald D, Smith I, Kay J, Canevari L, Heales S, Channon KM. GTP and Alzheimer’s disease. Exp Biol Med (Maywood) 232: 323–335, 2007. cyclohydrolase I gene transfer augments intracellular tetrahydrobiopterin in human endothelial cells: effects on nitric oxide synthase activity, protein levels and dimeri- 123. Broniowska KA, Hogg N. The chemical biology of S-nitrosothiols. Antioxid Redox sation. Cardiovasc Res 55: 838–849, 2002. doi:10.1016/S0008-6363(02)00460-1. Signal 17: 969–980, 2012. doi:10.1089/ars.2012.4590. 144. Caldwell RW, Rodriguez PC, Toque HA, Narayanan SP, Caldwell RB. Arginase: A 124. Brooks J. The action of nitrite on haemoglobin in the absence of oxygen. Proc R Soc Multifaceted Enzyme Important in Health and Disease. Physiol Rev 98: 641–665, Med 123: 368–382, 1937. doi:10.1098/rspb.1937.0057. 2018. doi:10.1152/physrev.00037.2016.

352 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

145. Campbell MG, Underbakke ES, Potter CS, Carragher B, Marletta MA. Single-particle 163. Chen F, Qian L-H, Deng B, Liu Z-M, Zhao Y, Le YY. Resveratrol protects vascular EM reveals the higher-order domain architecture of soluble guanylate cyclase. Proc endothelial cells from high glucose-induced apoptosis through inhibition of NADPH Natl Acad Sci USA 111: 2960–2965, 2014. doi:10.1073/pnas.1400711111. oxidase activation-driven oxidative stress. CNS Neurosci Ther 19: 675–681, 2013. doi:10.1111/cns.12131. 146. Cannon RO III, Schechter AN, Panza JA, Ognibene FP, Pease-Fye ME, Waclawiw MA, Shelhamer JH, Gladwin MT. Effects of inhaled nitric oxide on regional blood 164. Chen F, Yu Y, Haigh S, Johnson J, Lucas R, Stepp DW, Fulton DJR. Regulation of flow are consistent with intravascular nitric oxide delivery. J Clin Invest 108: 279– NADPH oxidase 5 by protein kinase C isoforms. PLoS One 9: e88405, 2014. doi:10. 287, 2001. doi:10.1172/JCI200112761. 1371/journal.pone.0088405.

147. Cantu-Medellin N, Kelley EE. Xanthine oxidoreductase-catalyzed reactive species 165. Chen Q, Moghaddas S, Hoppel CL, Lesnefsky EJ. Reversible blockade of electron generation: A process in critical need of reevaluation. Redox Biol 1: 353–358, 2013. transport during ischemia protects mitochondria and decreases myocardial injury doi:10.1016/j.redox.2013.05.002. following reperfusion. J Pharmacol Exp Ther 319: 1405–1412, 2006. doi:10.1124/jpet. 106.110262. 148. Cappola TP, Kass DA, Nelson GS, Berger RD, Rosas GO, Kobeissi ZA, Marbán E, 166. Cheng G, Cao Z, Xu X, van Meir EG, Lambeth JD. Homologs of gp91phox: cloning Hare JM. Allopurinol improves myocardial efficiency in patients with idiopathic di- and tissue expression of Nox3, Nox4, and Nox5. Gene 269: 131–140, 2001. doi:10. lated cardiomyopathy. Circulation 104: 2407–2411, 2001. doi:10.1161/hc4501. 1016/S0378-1119(01)00449-8. 098928. 167. Chin-Dusting JPF, Willems L, Kaye DM. L-arginine transporters in cardiovascular 149. Carballal S, Cuevasanta E, Yadav PK, Gherasim C, Ballou DP, Alvarez B, Banerjee R. disease: a novel therapeutic target. Pharmacol Ther 116: 428–436, 2007. doi:10. Kinetics of Nitrite Reduction and Peroxynitrite Formation by Ferrous Heme in 1016/j.pharmthera.2007.08.001. Human Cystathionine ␤-Synthase. J Biol Chem 291: 8004–8013, 2016. doi:10.1074/ jbc.M116.718734. 168. Chiva-Blanch G, Urpi-Sarda M, Ros E, Arranz S, Valderas-Martínez P, Casas R, Sacanella E, Llorach R, Lamuela-Raventos RM, Andres-Lacueva C, Estruch R. Deal- 150. Cardounel AJ, Cui H, Samouilov A, Johnson W, Kearns P, Tsai A-L, Berka V, Zweier coholized red wine decreases systolic and diastolic blood pressure and increases JL. Evidence for the pathophysiological role of endogenous methylarginines in regu- plasma nitric oxide: short communication. Circ Res 111: 1065–1068, 2012. doi:10. lation of endothelial NO production and vascular function. J Biol Chem 282: 879– 1161/CIRCRESAHA.112.275636. 887, 2007. doi:10.1074/jbc.M603606200. 169. Cho HJ, Xie QW, Calaycay J, Mumford RA, Swiderek KM, Lee TD, Nathan C. 151. Carlström M, Lai EY, Ma Z, Patzak A, Brown RD, Persson AEG. Role of NOX2 in the Calmodulin is a subunit of nitric oxide synthase from macrophages. J Exp Med 176: regulation of afferent arteriole responsiveness. Am J Physiol Regul Integr Comp Physiol 599–604, 1992. doi:10.1084/jem.176.2.599. 296: R72–R79, 2009. doi:10.1152/ajpregu.90718.2008. 170. Chouchani ET, Methner C, Nadtochiy SM, Logan A, Pell VR, Ding S, James AM, 152. Case AJ, Li S, Basu U, Tian J, Zimmerman MC. Mitochondrial-localized NADPH Cochemé HM, Reinhold J, Lilley KS, Partridge L, Fearnley IM, Robinson AJ, Hartley oxidase 4 is a source of superoxide in angiotensin II-stimulated neurons. Am J Physiol RC, Smith RAJ, Krieg T, Brookes PS, Murphy MP. Cardioprotection by S-nitrosation Heart Circ Physiol 305: H19–H28, 2013. doi:10.1152/ajpheart.00974.2012. of a cysteine switch on mitochondrial complex I. Nat Med 19: 753–759, 2013. doi:10.1038/nm.3212. 153. Cassina AM, Hodara R, Souza JM, Thomson L, Castro L, Ischiropoulos H, Freeman BA, Radi R. Cytochrome c nitration by peroxynitrite. J Biol Chem 275: 21409–21415, 171. Chouchani ET, Pell VR, Gaude E, Aksentijevic´ D, Sundier SY, Robb EL, Logan A, 2000. doi:10.1074/jbc.M909978199. Nadtochiy SM, Ord ENJ, Smith AC, Eyassu F, Shirley R, Hu CH, Dare AJ, James AM, Rogatti S, Hartley RC, Eaton S, Costa ASH, Brookes PS, Davidson SM, Duchen MR, 154. Castro L, Eiserich JP, Sweeney S, Radi R, Freeman BA. Cytochrome c: a catalyst and Saeb-Parsy K, Shattock MJ, Robinson AJ, Work LM, Frezza C, Krieg T, Murphy MP. target of nitrite-hydrogen peroxide-dependent protein nitration. Arch Biochem Bio- Ischaemic accumulation of succinate controls reperfusion injury through mitochon- phys 421: 99–107, 2004. doi:10.1016/j.abb.2003.08.033. drial ROS. Nature 515: 431–435, 2014. doi:10.1038/nature13909.

155. Castro L, Rodriguez M, Radi R. Aconitase is readily inactivated by peroxynitrite, but 172. Chouchani ET, Pell VR, James AM, Work LM, Saeb-Parsy K, Frezza C, Krieg T, not by its precursor, nitric oxide. J Biol Chem 269: 29409–29415, 1994. Murphy MP. A Unifying Mechanism for Mitochondrial Superoxide Production during Ischemia-Reperfusion Injury. Cell Metab 23: 254–263, 2016. doi:10.1016/j.cmet. 156. Caughey GE, Cleland LG, Penglis PS, Gamble JR, James MJ. Roles of cyclooxygenase 2015.12.009. (COX)-1 and COX-2 in prostanoid production by human endothelial cells: selective up-regulation of prostacyclin synthesis by COX-2. J Immunol 167: 2831–2838, 2001. 173. Chuaiphichai S, McNeill E, Douglas G, Crabtree MJ, Bendall JK, Hale AB, Alp NJ, doi:10.4049/jimmunol.167.5.2831. Channon KM. Cell-autonomous role of endothelial GTP cyclohydrolase 1 and tet- rahydrobiopterin in blood pressure regulation. Hypertension 64: 530–540, 2014. 157. Chambers DE, Parks DA, Patterson G, Roy R, McCord JM, Yoshida S, Parmley doi:10.1161/HYPERTENSIONAHA.114.03089. LF, Downey JM. Xanthine oxidase as a source of free radical damage in myocar- dial ischemia. J Mol Cell Cardiol 17: 145–152, 1985. doi:10.1016/S0022- 174. Cifuentes-Pagano E, Meijles DN, Pagano PJ. The quest for selective nox inhibitors and therapeutics: challenges, triumphs and pitfalls. Antioxid Redox Signal 20: 2741– 2828(85)80017-1. 2754, 2014. doi:10.1089/ars.2013.5620. 158. Chandel NS, Tuveson DA. The promise and perils of antioxidants for cancer pa- 175. Cifuentes-Pagano ME, Meijles DN, Pagano PJ. Nox Inhibitors & Therapies: Rational tients. N Engl J Med 371: 177–178, 2014. doi:10.1056/NEJMcibr1405701. Design of Peptidic and Small Molecule Inhibitors. Curr Pharm Des 21: 6023–6035, 159. Chen CY, Yi L, Jin X, Zhang T, Fu YJ, Zhu JD, Mi MT, Zhang QY, Ling WH, Yu B. 2015. doi:10.2174/1381612821666151029112013. Inhibitory effect of delphinidin on monocyte-endothelial cell adhesion induced by 176. Clancy RM, Leszczynska-Piziak J, Abramson SB. Nitric oxide, an endothelial cell oxidized low-density lipoprotein via ROS/p38MAPK/NF-␬B pathway. Cell Biochem relaxation factor, inhibits neutrophil superoxide anion production via a direct action Biophys 61: 337–348, 2011. doi:10.1007/s12013-011-9216-2. on the NADPH oxidase. J Clin Invest 90: 1116–1121, 1992. doi:10.1172/JCI115929.

160. Chen CA, De Pascali F, Basye A, Hemann C, Zweier JL. Redox modulation of 177. Clark RA, Leidal KG, Pearson DW, Nauseef WM. NADPH oxidase of human neu- endothelial nitric oxide synthase by glutaredoxin-1 through reversible oxidative trophils. Subcellular localization and characterization of an arachidonate-activatable post-translational modification. Biochemistry 52: 6712–6723, 2013. doi:10.1021/ superoxide-generating system. J Biol Chem 262: 4065–4074, 1987. bi400404s. 178. Clifton HL, Garten RS, Lee JF, Rossman MJ, Clifford JR, Hydren JR, Stehlik J, Rich- 161. Chen CA, Druhan LJ, Varadharaj S, Chen YR, Zweier JL. Phosphorylation of endo- ardson RS, Wray DW. The impact of acute oral tetrahydrobiopterin on vascular thelial nitric-oxide synthase regulates superoxide generation from the enzyme. J Biol function in heart failure patients with reduced ejection fraction. FASEB J 30: 735.5, Chem 283: 27038–27047, 2008. doi:10.1074/jbc.M802269200. 2016.

162. Chen CA, Wang TY, Varadharaj S, Reyes LA, Hemann C, Talukder MA, Chen YR, 179. Colston JT, de la Rosa SD, Strader JR, Anderson MA, Freeman GL. H2O2 activates Druhan LJ, Zweier JL. S-glutathionylation uncouples eNOS and regulates its cellular Nox4 through PLA2-dependent arachidonic acid production in adult cardiac fibro- and vascular function. Nature 468: 1115–1118, 2010. doi:10.1038/nature09599. blasts. FEBS Lett 579: 2533–2540, 2005. doi:10.1016/j.febslet.2005.03.057.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 353 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

180. Cooper CE. Nitric oxide and iron proteins. Biochim Biophys Acta 1411: 290–309, 195. Crane BR, Rosenfeld RJ, Arvai AS, Ghosh DK, Ghosh S, Tainer JA, Stuehr DJ, Getzoff 1999. doi:10.1016/S0005-2728(99)00021-3. ED. N-terminal domain swapping and metal ion binding in nitric oxide synthase dimerization. EMBO J 18: 6271–6281, 1999. doi:10.1093/emboj/18.22.6271. 181. Cortese-Krott MM, Rodriguez-Mateos A, Sansone R, Kuhnle GG, Thasian-Sivarajah S, Krenz T, Horn P, Krisp C, Wolters D, Heiß C, Kröncke KD, Hogg N, Feelisch M, 196. Crawford JH, Isbell TS, Huang Z, Shiva S, Chacko BK, Schechter AN, Darley-Usmar Kelm M. Human red blood cells at work: identification and visualization of erythro- VM, Kerby JD, Lang JD Jr, Kraus D, Ho C, Gladwin MT, Patel RP. Hypoxia, red blood cytic eNOS activity in health and disease. Blood 120: 4229–4237, 2012. doi:10.1182/ cells, and nitrite regulate NO-dependent hypoxic vasodilation. Blood 107: 566–574, blood-2012-07-442277. 2006. doi:10.1182/blood-2005-07-2668.

182. Corti P, Ieraci M, Tejero J. Characterization of zebrafish neuroglobin and cytoglobins 197. Cross AR, Jones OTG. The effect of the inhibitor diphenylene iodonium on the 1 and 2: Zebrafish cytoglobins provide insights into the transition from six-coordi- superoxide-generating system of neutrophils. Specific labelling of a component poly- nate to five-coordinate globins. Nitric Oxide 53: 22–34, 2016. doi:10.1016/j.niox. peptide of the oxidase. Biochem J 237: 111–116, 1986. doi:10.1042/bj2370111. 2015.12.004. 198. Cross AR, Segal AW. The NADPH oxidase of professional phagocytes–prototype of the NOX electron transport chain systems. Biochim Biophys Acta 1657: 1–22, 2004. 183. Corti P, Tejero J, Gladwin MT. Evidence mounts that red cells and deoxyhemoglobin doi:10.1016/j.bbabio.2004.03.008. can reduce nitrite to bioactive NO to mediate intravascular endocrine NO signaling: commentary on “Anti-platelet effects of dietary nitrate in healthy volunteers: in- 199. Csányi G, Cifuentes-Pagano E, Al Ghouleh I, Ranayhossaini DJ, Egaña L, Lopes LR, volvement of cGMP and influence of sex”. Free Radic Biol Med 65: 1518–1520, 2013. Jackson HM, Kelley EE, Pagano PJ. Nox2 B-loop peptide, Nox2ds, specifically inhibits doi:10.1016/j.freeradbiomed.2013.09.020. the NADPH oxidase Nox2. Free Radic Biol Med 51: 1116–1125, 2011. doi:10.1016/ j.freeradbiomed.2011.04.025. 184. Corti P, Xue J, Tejero J, Wajih N, Sun M, Stolz DB, Tsang M, Kim-Shapiro DB, Gladwin MT. Globin X is a six-coordinate globin that reduces nitrite to nitric oxide in 200. Csányi G, Pagano PJ. Strategies aimed at Nox4 oxidase inhibition employing peptides fish red blood cells. Proc Natl Acad Sci USA 113: 8538–8543, 2016. doi:10.1073/pnas. from Nox4 B-loop and C-terminus and p22 (phox) N-terminus: an elusive target. Int 1522670113. J Hypertens 2013: 842827, 2013. doi:10.1155/2013/842827.

185. Cosby K, Partovi KS, Crawford JH, Patel RP, Reiter CD, Martyr S, Yang BK, Wac- 201. Cucoranu I, Clempus R, Dikalova A, Phelan PJ, Ariyan S, Dikalov S, Sorescu D. lawiw MA, Zalos G, Xu X, Huang KT, Shields H, Kim-Shapiro DB, Schechter AN, NAD(P)H oxidase 4 mediates transforming growth factor-beta1-induced differenti- Cannon RO III, Gladwin MT. Nitrite reduction to nitric oxide by deoxyhemoglobin ation of cardiac fibroblasts into myofibroblasts. Circ Res 97: 900–907, 2005. doi:10. vasodilates the human circulation. Nat Med 9: 1498–1505, 2003. doi:10.1038/ 1161/01.RES.0000187457.24338.3D. nm954. 202. Cunnington C, Van Assche T, Shirodaria C, Kylintireas I, Lindsay AC, Lee JM, An- 186. Cosentino F, Hürlimann D, Delli Gatti C, Chenevard R, Blau N, Alp NJ, Channon toniades C, Margaritis M, Lee R, Cerrato R, Crabtree MJ, Francis JM, Sayeed R, KM, Eto M, Lerch P, Enseleit F, Ruschitzka F, Volpe M, Lüscher TF, Noll G. Chronic Ratnatunga C, Pillai R, Choudhury RP, Neubauer S, Channon KM. Systemic and treatment with tetrahydrobiopterin reverses endothelial dysfunction and oxidative vascular oxidation limits the efficacy of oral tetrahydrobiopterin treatment in pa- stress in hypercholesterolaemia. Heart 94: 487–492, 2008. doi:10.1136/hrt.2007. tients with coronary artery disease. Circulation 125: 1356–1366, 2012. doi:10.1161/ 122184. CIRCULATIONAHA.111.038919.

187. Costa AD, Garlid KD. Intramitochondrial signaling: interactions among mitoKATP, 203. Cyrus T, Witztum JL, Rader DJ, Tangirala R, Fazio S, Linton MF, Funk CD. Disruption of the 12/15-lipoxygenase gene diminishes atherosclerosis in apo E-deficient mice. J PKCepsilon, ROS, and MPT. Am J Physiol Heart Circ Physiol 295: H874–H882, 2008. Clin Invest 103: 1597–1604, 1999. doi:10.1172/JCI5897. doi:10.1152/ajpheart.01189.2007. 204. Daff S. NO synthase: structures and mechanisms. Nitric Oxide 23: 1–11, 2010. 188. Costa ED, Rezende BA, Cortes SF, Lemos VS. Neuronal Nitric Oxide Synthase in doi:10.1016/j.niox.2010.03.001. Vascular Physiology and Diseases. Front Physiol 7: 206, 2016. doi:10.3389/fphys. 2016.00206. 205. Dahn H, Loewe L, Bunton C. Über die Oxydation von Ascorbinsäure durch sal- petrige Säure Teil VI: Übersicht und Diskussion der Ergebnisse. 18. Mitteilung über 189. Crabtree MJ, Brixey R, Batchelor H, Hale AB, Channon KM. Integrated redox sensor Reduktone und 1, 2, 3᎑Tricarbonylverbindungen. Helv Chim Acta 43: 320–333, 1960. and effector functions for tetrahydrobiopterin- and glutathionylation-dependent en- doi:10.1002/hlca.19600430143. dothelial nitric-oxide synthase uncoupling. J Biol Chem 288: 561–569, 2013. doi:10. 1074/jbc.M112.415992. 206. Daiber A. Redox signaling (cross-talk) from and to mitochondria involves mitochon- drial pores and reactive oxygen species. Biochim Biophys Acta 1797: 897–906, 2010. 190. Crabtree MJ, Smith CL, Lam G, Goligorsky MS, Gross SS. Ratio of 5,6,7,8-tetrahy- doi:10.1016/j.bbabio.2010.01.032. drobiopterin to 7,8-dihydrobiopterin in endothelial cells determines glucose-elicited changes in NO vs. superoxide production by eNOS. Am J Physiol Heart Circ Physiol 207. Daiber A, Steven S, Weber A, Shuvaev VV, Muzykantov VR, Laher I, Li H, Lamas S, 294: H1530–H1540, 2008. doi:10.1152/ajpheart.00823.2007. Münzel T. Targeting vascular (endothelial) dysfunction. Br J Pharmacol 174: 1591– 1619, 2017. doi:10.1111/bph.13517. 191. Crabtree MJ, Tatham AL, Al-Wakeel Y, Warrick N, Hale AB, Cai S, Channon KM, Alp NJ. Quantitative regulation of intracellular endothelial nitric-oxide synthase (eNOS) 208. Dandapat A, Hu C, Sun L, Mehta JL. Small concentrations of oxLDL induce capillary coupling by both tetrahydrobiopterin-eNOS stoichiometry and biopterin redox sta- tube formation from endothelial cells via LOX-1-dependent redox-sensitive path- tus: insights from cells with tet-regulated GTP cyclohydrolase I expression. J Biol way. Arterioscler Thromb Vasc Biol 27: 2435–2442, 2007. doi:10.1161/ATVBAHA. Chem 284: 1136–1144, 2009. doi:10.1074/jbc.M805403200. 107.152272.

192. Crabtree MJ, Tatham AL, Hale AB, Alp NJ, Channon KM. Critical role for tetrahy- 209. Dao VT, Casas AI, Maghzal GJ, Seredenina T, Kaludercic N, Robledinos-Anton N, Di drobiopterin recycling by dihydrofolate reductase in regulation of endothelial nitric- Lisa F, Stocker R, Ghezzi P, Jaquet V, Cuadrado A, Schmidt HH. Pharmacology and Clinical Drug Candidates in Redox Medicine. Antioxid Redox Signal 23: 1113–1129, oxide synthase coupling: relative importance of the de novo biopterin synthesis 2015. doi:10.1089/ars.2015.6430. versus salvage pathways. J Biol Chem 284: 28128–28136, 2009. doi:10.1074/jbc. M109.041483. 210. Dare AJ, Bolton EA, Pettigrew GJ, Bradley JA, Saeb-Parsy K, Murphy MP. Protection against renal ischemia-reperfusion injury in vivo by the mitochondria targeted anti- 193. Craige SM, Chen K, Pei Y, Li C, Huang X, Chen C, Shibata R, Sato K, Walsh K, Keaney oxidant MitoQ. Redox Biol 5: 163–168, 2015. doi:10.1016/j.redox.2015.04.008. JF Jr. NADPH oxidase 4 promotes endothelial angiogenesis through endothelial nitric oxide synthase activation. Circulation 124: 731–740, 2011. doi:10.1161/ 211. Dare AJ, Logan A, Prime TA, Rogatti S, Goddard M, Bolton EM, Bradley JA, Petti- CIRCULATIONAHA.111.030775. grew GJ, Murphy MP, Saeb-Parsy K. The mitochondria-targeted anti-oxidant MitoQ decreases ischemia-reperfusion injury in a murine syngeneic heart transplant model. 194. Craige SM, Kant S, Reif M, Chen K, Pei Y, Angoff R, Sugamura K, Fitzgibbons T, J Heart Lung Transplant 34: 1471–1480, 2015. doi:10.1016/j.healun.2015.05.007. Keaney JF Jr. Endothelial NADPH oxidase 4 protects ApoEϪ/Ϫ mice from athero- sclerotic lesions. Free Radic Biol Med 89: 1–7, 2015. doi:10.1016/j.freeradbiomed. 212. Datla SR, McGrail DJ, Vukelic S, Huff LP, Lyle AN, Pounkova L, Lee M, Seidel-Rogol 2015.07.004. B, Khalil MK, Hilenski LL, Terada LS, Dawson MR, Lassègue B, Griendling KK.

354 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

Poldip2 controls vascular smooth muscle cell migration by regulating focal adhesion 230. Di Marco E, Gray SP, Kennedy K, Szyndralewiez C, Lyle AN, Lassègue B, Griendling turnover and force polarization. Am J Physiol Heart Circ Physiol 307: H945–H957, KK, Cooper ME, Schmidt HHHW, Jandeleit-Dahm KAM. NOX4-derived reactive 2014. doi:10.1152/ajpheart.00918.2013. oxygen species limit fibrosis and inhibit proliferation of vascular smooth muscle cells in diabetic atherosclerosis. Free Radic Biol Med 97: 556–567, 2016. doi:10.1016/j. 213. Daugherty A, Dunn JL, Rateri DL, Heinecke JW. Myeloperoxidase, a catalyst for freeradbiomed.2016.07.013. lipoprotein oxidation, is expressed in human atherosclerotic lesions. J Clin Invest 94: 437–444, 1994. doi:10.1172/JCI117342. 231. Diaz B, Shani G, Pass I, Anderson D, Quintavalle M, Courtneidge SA. Tks5-depen- dent, nox-mediated generation of reactive oxygen species is necessary for invado- 214. de A Paes AM, Veríssimo-Filho S, Guimarães LL, Silva AC, Takiuti JT, Santos CX, podia formation. Sci Signal 2: ra53, 2009. doi:10.1126/scisignal.2000368. Janiszewski M, Laurindo FR, Lopes LR. Protein disulfide isomerase redox-dependent association with p47(phox): evidence for an organizer role in leukocyte NADPH 232. Dickinson BC, Chang CJ. Chemistry and biology of reactive oxygen species in sig- oxidase activation. J Leukoc Biol 90: 799–810, 2011. doi:10.1189/jlb.0610324. naling or stress responses. Nat Chem Biol 7: 504–511, 2011. doi:10.1038/nchembio. 607. 215. De Pascali F, Hemann C, Samons K, Chen CA, Zweier JL. Hypoxia and reoxygen- ation induce endothelial nitric oxide synthase uncoupling in endothelial cells through 233. Diebold L, Chandel NS. Mitochondrial ROS regulation of proliferating cells. Free tetrahydrobiopterin depletion and S-glutathionylation. Biochemistry 53: 3679–3688, Radic Biol Med 100: 86–93, 2016. doi:10.1016/j.freeradbiomed.2016.04.198. 2014. doi:10.1021/bi500076r. 234. Dikalov S. Cross talk between mitochondria and NADPH oxidases. Free Radic Biol 216. Dejam A, Hunter CJ, Schechter AN, Gladwin MT. Emerging role of nitrite in human Med 51: 1289–1301, 2011. doi:10.1016/j.freeradbiomed.2011.06.033. biology. Blood Cells Mol Dis 32: 423–429, 2004. doi:10.1016/j.bcmd.2004.02.002. 235. Dikalova A, Clempus R, Lassègue B, Cheng G, McCoy J, Dikalov S, San Martin A, Lyle 217. Dejam A, Hunter CJ, Tremonti C, Pluta RM, Hon YY, Grimes G, Partovi K, A, Weber DS, Weiss D, Taylor WR, Schmidt HH, Owens GK, Lambeth JD, Grien- Pelletier MM, Oldfield EH, Cannon RO III, Schechter AN, Gladwin MT. Nitrite dling KK. Nox1 overexpression potentiates angiotensin II-induced hypertension and infusion in humans and nonhuman primates: endocrine effects, pharmacokinetics, vascular smooth muscle hypertrophy in transgenic mice. Circulation 112: 2668– and tolerance formation. Circulation 116: 1821–1831, 2007. doi:10.1161/ 2676, 2005. doi:10.1161/CIRCULATIONAHA.105.538934. CIRCULATIONAHA.107.712133. 236. Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM. Activation of 218. DeLeo FR, Yu L, Burritt JB, Loetterle LR, Bond CW, Jesaitis AJ, Quinn MT. Mapping nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature sites of interaction of p47-phox and flavocytochrome b with random-sequence 399: 601–605, 1999. doi:10.1038/21224. peptide phage display libraries. Proc Natl Acad Sci USA 92: 7110–7114, 1995. doi:10. 1073/pnas.92.15.7110. 237. Dodd-O JM, Pearse DB. Effect of the NADPH oxidase inhibitor apocynin on isch- emia-reperfusion lung injury. Am J Physiol Heart Circ Physiol 279: H303–H312, 2000. 219. Demicheli V, Moreno DM, Jara GE, Lima A, Carballal S, Ríos N, Batthyany C, Ferrer- doi:10.1152/ajpheart.2000.279.1.H303. Sueta G, Quijano C, Estrín DA, Martí MA, Radi R. Mechanism of the Reaction of Human Manganese Superoxide Dismutase with Peroxynitrite: Nitration of Critical 238. Doerries C, Grote K, Hilfiker-Kleiner D, Luchtefeld M, Schaefer A, Holland SM, Tyrosine 34. Biochemistry 55: 3403–3417, 2016. doi:10.1021/acs.biochem.6b00045. Sorrentino S, Manes C, Schieffer B, Drexler H, Landmesser U. Critical role of the NAD(P)H oxidase subunit p47phox for left ventricular remodeling/dysfunction and 220. Demicheli V, Quijano C, Alvarez B, Radi R. Inactivation and nitration of human survival after myocardial infarction. Circ Res 100: 894–903, 2007. doi:10.1161/01. superoxide dismutase (SOD) by fluxes of nitric oxide and superoxide. Free Radic Biol RES.0000261657.76299.ff. Med 42: 1359–1368, 2007. doi:10.1016/j.freeradbiomed.2007.01.034. 239. Dominguez-Rodriguez A, Samimi-Fard S, Abreu-Gonzalez P, Garcia-Gonzalez MJ, 221. Denicola A, Souza JM, Radi R. Diffusion of peroxynitrite across erythrocyte mem- Kaski JC. Prognostic value of admission myeloperoxidase levels in patients with branes. Proc Natl Acad Sci USA 95: 3566–3571, 1998. doi:10.1073/pnas.95.7.3566. ST-segment elevation myocardial infarction and cardiogenic shock. Am J Cardiol 101: 222. Depre C, Havaux X, Renkin J, Vanoverschelde JLJ, Wijns W. Expression of inducible 1537–1540, 2008. doi:10.1016/j.amjcard.2008.02.032. nitric oxide synthase in human coronary atherosclerotic plaque. Cardiovasc Res 41: 240. Dong J-Y, Qin L-Q, Zhang Z, Zhao Y, Wang J, Arigoni F, Zhang W. Effect of oral 465–472, 1999. doi:10.1016/S0008-6363(98)00304-6. L-arginine supplementation on blood pressure: a meta-analysis of randomized, dou- 223. Derbyshire ER, Marletta MA. Structure and regulation of soluble guanylate cyclase. ble-blind, placebo-controlled trials. Am Heart J 162: 959–965, 2011. doi:10.1016/j. Annu Rev Biochem 81: 533–559, 2012. doi:10.1146/annurev-biochem-050410- ahj.2011.09.012. 100030. 241. Doroshow JH, Gaur S, Markel S, Lu J, van Balgooy J, Synold TW, Xi B, Wu X, Juhasz 224. Desco MC, Asensi M, Márquez R, Martínez-Valls J, Vento M, Pallardó FV, Sastre J, A. Effects of iodonium-class flavin dehydrogenase inhibitors on growth, reactive Viña J. Xanthine oxidase is involved in free radical production in type 1 diabetes: oxygen production, cell cycle progression, NADPH oxidase 1 levels, and gene ex- protection by allopurinol. Diabetes 51: 1118–1124, 2002. doi:10.2337/diabetes.51. pression in human colon cancer cells and xenografts. Free Radic Biol Med 57: 162– 4.1118. 175, 2013. doi:10.1016/j.freeradbiomed.2013.01.002.

225. Desideri G, Kwik-Uribe C, Grassi D, Necozione S, Ghiadoni L, Mastroiacovo D, 242. Doughan AK, Harrison DG, Dikalov SI. Molecular mechanisms of angiotensin II- Raffaele A, Ferri L, Bocale R, Lechiara MC, Marini C, Ferri C. Benefits in cognitive mediated mitochondrial dysfunction: linking mitochondrial oxidative damage and function, blood pressure, and insulin resistance through cocoa flavanol consump- vascular endothelial dysfunction. Circ Res 102: 488–496, 2008. doi:10.1161/ tion in elderly subjects with mild cognitive impairment: the Cocoa, Cognition, CIRCRESAHA.107.162800. and Aging (CoCoA) study. Hypertension 60: 794–801, 2012. doi:10.1161/ 243. Douglas G, Bendall JK, Crabtree MJ, Tatham AL, Carter EE, Hale AB, Channon KM. HYPERTENSIONAHA.112.193060. Endothelial-specific Nox2 overexpression increases vascular superoxide and mac- 226. Dezfulian C, Raat N, Shiva S, Gladwin MT. Role of the anion nitrite in ischemia- rophage recruitment in ApoEϪ/Ϫ mice. Cardiovasc Res 94: 20–29, 2012. doi:10. reperfusion cytoprotection and therapeutics. Cardiovasc Res 75: 327–338, 2007. 1093/cvr/cvs026. doi:10.1016/j.cardiores.2007.05.001. 244. Dourron HM, Jacobson GM, Park JL, Liu J, Reddy DJ, Scheel ML, Pagano PJ. Perivas- 227. Dhalla NS, Temsah RM, Netticadan T. Role of oxidative stress in cardiovascular cular gene transfer of NADPH oxidase inhibitor suppresses angioplasty-induced diseases. J Hypertens 18: 655–673, 2000. doi:10.1097/00004872-200018060- neointimal proliferation of rat carotid artery. Am J Physiol Heart Circ Physiol 288: 00002. H946–H953, 2005. doi:10.1152/ajpheart.00413.2004.

228. Dhaunsi GS, Paintlia MK, Kaur J, Turner RB. NADPH oxidase in human lung fibro- 245. Doussière J, Gaillard J, Vignais PV. Electron transfer across the O2- generating blasts. J Biomed Sci 11: 617–622, 2004. doi:10.1007/BF02256127. flavocytochrome b of neutrophils. Evidence for a transition from a low-spin state to a high-spin state of the heme iron component. Biochemistry 35: 13400–13410, 1996. 229. Di Marco E, Gray SP, Chew P, Kennedy K, Cooper ME, Schmidt HH, Jandeleit- doi:10.1021/bi960916b. Dahm KA. Differential effects of NOX4 and NOX1 on immune cell-mediated in- flammation in the aortic sinus of diabetic ApoEϪ/Ϫ mice. Clin Sci (Lond) 130: 1363– 246. Doussière J, Vignais PV. Diphenylene iodonium as an inhibitor of the NADPH oxi- 1374, 2016. doi:10.1042/CS20160249. dase complex of bovine neutrophils. Factors controlling the inhibitory potency of

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 355 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

diphenylene iodonium in a cell-free system of oxidase activation. Eur J Biochem 208: 265. Ellmark SHM, Dusting GJ, Fui MNT, Guzzo-Pernell N, Drummond GR. The contri- 61–71, 1992. doi:10.1111/j.1432-1033.1992.tb17159.x. bution of Nox4 to NADPH oxidase activity in mouse vascular smooth muscle. Cardiovasc Res 65: 495–504, 2005. doi:10.1016/j.cardiores.2004.10.026. 247. Doyle MP, LePoire DM, Pickering RA. Oxidation of hemoglobin and myoglobin by alkyl nitrites inhibition by oxygen. J Biol Chem 256: 12399–12404, 1981. 266. Ellsworth ML. The red blood cell as an oxygen sensor: what is the evidence? Acta Physiol Scand 168: 551–559, 2000. doi:10.1046/j.1365-201x.2000.00708.x. 248. Doyle MP, Pickering RA, DeWeert TM, Hoekstra JW, Pater D. Kinetics and mech- anism of the oxidation of human deoxyhemoglobin by nitrites. J Biol Chem 256: 267. Elrod JW, Calvert JW, Gundewar S, Bryan NS, Lefer DJ. Nitric oxide promotes 12393–12398, 1981. distant organ protection: evidence for an endocrine role of nitric oxide. Proc Natl Acad Sci USA 105: 11430–11435, 2008. doi:10.1073/pnas.0800700105. 249. Dröge W. Free radicals in the physiological control of cell function. Physiol Rev 82: 47–95, 2002. doi:10.1152/physrev.00018.2001. 268. Enroth C, Eger BT, Okamoto K, Nishino T, Nishino T, Pai EF. Crystal structures of bovine milk xanthine dehydrogenase and xanthine oxidase: structure-based mech- 250. Druhan LJ, Forbes SP, Pope AJ, Chen CA, Zweier JL, Cardounel AJ. Regulation of anism of conversion. Proc Natl Acad Sci USA 97: 10723–10728, 2000. doi:10.1073/ eNOS-derived superoxide by endogenous methylarginines. Biochemistry 47: 7256– pnas.97.20.10723. 7263, 2008. doi:10.1021/bi702377a. 269. Erdmann E, Semigran MJ, Nieminen MS, Gheorghiade M, Agrawal R, Mitrovic V, 251. Drummond GR, Cai H, Davis ME, Ramasamy S, Harrison DG. Transcriptional and posttranscriptional regulation of endothelial nitric oxide synthase expression by hy- Mebazaa A. Cinaciguat, a soluble guanylate cyclase activator, unloads the heart but drogen peroxide. Circ Res 86: 347–354, 2000. doi:10.1161/01.RES.86.3.347. also causes hypotension in acute decompensated heart failure. Eur Heart J 34: 57–67, 2013. doi:10.1093/eurheartj/ehs196. 252. Drummond GR, Selemidis S, Griendling KK, Sobey CG. Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nat Rev Drug Discov 10: 270. Erez A, Nagamani SC, Shchelochkov OA, Premkumar MH, Campeau PM, Chen Y, 453–471, 2011. doi:10.1038/nrd3403. Garg HK, Li L, Mian A, Bertin TK, Black JO, Zeng H, Tang Y, Reddy AK, Summar M, O’Brien WE, Harrison DG, Mitch WE, Marini JC, Aschner JL, Bryan NS, Lee B. 253. Drummond GR, Sobey CG. Endothelial NADPH oxidases: which NOX to target in Requirement of argininosuccinate lyase for systemic nitric oxide production. Nat vascular disease? Trends Endocrinol Metab 25: 452–463, 2014. doi:10.1016/j.tem. Med 17: 1619–1626, 2011. doi:10.1038/nm.2544. 2014.06.012. 271. Erwin PA, Lin AJ, Golan DE, Michel T. Receptor-regulated dynamic S-nitrosylation of 254. Drury PP, Davidson JO, Mathai S, van den Heuij LG, Ji H, Bennet L, Tan S, Silverman endothelial nitric-oxide synthase in vascular endothelial cells. J Biol Chem 280: RB, Gunn AJ. nNOS inhibition during profound asphyxia reduces seizure burden and 19888–19894, 2005. doi:10.1074/jbc.M413058200. improves survival of striatal phenotypic neurons in preterm fetal sheep. Neurophar- macology 83: 62–70, 2014. doi:10.1016/j.neuropharm.2014.03.017. 272. Erwin PA, Mitchell DA, Sartoretto J, Marletta MA, Michel T. Subcellular targeting and differential S-nitrosylation of endothelial nitric-oxide synthase. J Biol Chem 281: 151– 255. Drury PP, Davidson JO, van den Heuij LG, Tan S, Silverman RB, Ji H, Blood AB, 157, 2006. doi:10.1074/jbc.M510421200. Fraser M, Bennet L, Gunn AJ. Partial neuroprotection by nNOS inhibition during profound asphyxia in preterm fetal sheep. Exp Neurol 250: 282–292, 2013. doi:10. 273. Evangelista AM, Thompson MD, Bolotina VM, Tong X, Cohen RA. Nox4- and 1016/j.expneurol.2013.10.003. Nox2-dependent oxidant production is required for VEGF-induced SERCA cys- teine-674 S-glutathiolation and endothelial cell migration. Free Radic Biol Med 53: 256. Du J, Fan LM, Mai A, Li J-M. Crucial roles of Nox2-derived oxidative stress in 2327–2334, 2012. doi:10.1016/j.freeradbiomed.2012.10.546. deteriorating the function of insulin receptors and endothelium in dietary obesity of middle-aged mice. Br J Pharmacol 170: 1064–1077, 2013. doi:10.1111/bph.12336. 274. Evgenov OV, Pacher P, Schmidt PM, Haskó G, Schmidt HHHW, Stasch J-P. NO- independent stimulators and activators of soluble guanylate cyclase: discovery and 257. Du XL, Edelstein D, Dimmeler S, Ju Q, Sui C, Brownlee M. Hyperglycemia inhibits therapeutic potential. Nat Rev Drug Discov 5: 755–768, 2006. doi:10.1038/nrd2038. endothelial nitric oxide synthase activity by posttranslational modification at the Akt site. J Clin Invest 108: 1341–1348, 2001. doi:10.1172/JCI11235. 275. Fan J, Frey RS, Rahman A, Malik AB. Role of neutrophil NADPH oxidase in the mechanism of tumor necrosis factor-alpha -induced NF-kappa B activation and in- 258. Dumitrescu C, Biondi R, Xia Y, Cardounel AJ, Druhan LJ, Ambrosio G, Zweier JL. tercellular adhesion molecule-1 expression in endothelial cells. J Biol Chem 277: Myocardial ischemia results in tetrahydrobiopterin (BH4) oxidation with impaired 3404–3411, 2002. doi:10.1074/jbc.M110054200. endothelial function ameliorated by BH4. Proc Natl Acad Sci USA 104: 15081–15086, 2007. doi:10.1073/pnas.0702986104. 276. Fan LM, Teng L, Li JM. Knockout of p47 phox uncovers a critical role of p40 phox in 259. Durante W, Johnson FK, Johnson RA. Arginase: a critical regulator of nitric oxide reactive oxygen species production in microvascular endothelial cells. Arterioscler synthesis and vascular function. Clin Exp Pharmacol Physiol 34: 906–911, 2007. doi: Thromb Vasc Biol 29: 1651–1656, 2009. doi:10.1161/ATVBAHA.109.191502. 10.1111/j.1440-1681.2007.04638.x. 277. Farah C, Michel LYM, Balligand JL. Nitric oxide signalling in cardiovascular health and 260. Düzgünçinar O, Yavuz B, Hazirolan T, Deniz A, Tokgözog˘lu SL, Akata D, Demir- disease. Nat Rev Cardiol 15: 292–316, 2018. doi:10.1038/nrcardio.2017.224. pençe E. Plasma myeloperoxidase is related to the severity of coronary artery dis- 278. Farquharson CA, Butler R, Hill A, Belch JJF, Struthers AD. Allopurinol improves ease. Acta Cardiol 63: 147–152, 2008. doi:10.2143/AC.63.2.2029520. endothelial dysfunction in chronic heart failure. Circulation 106: 221–226, 2002. 261. Dykhuizen RS, Frazer R, Duncan C, Smith CC, Golden M, Benjamin N, Leifert C. doi:10.1161/01.CIR.0000022140.61460.1D. Antimicrobial effect of acidified nitrite on gut pathogens: importance of dietary 279. Fast W, Nikolic D, Van Breemen RB, Silverman RB. Mechanistic studies of the nitrate in host defense. Antimicrob Agents Chemother 40: 1422–1425, 1996. inactivation of inducible nitric oxide synthase by N-5-(1-iminoethyl)-L-ornithine (L- 262. Edens WA, Sharling L, Cheng G, Shapira R, Kinkade JM, Lee T, Edens HA, Tang X, NIO). J Am Chem Soc 121: 903–916, 1999. doi:10.1021/ja982318l. Sullards C, Flaherty DB, Benian GM, Lambeth JD. Tyrosine cross-linking of extra- cellular matrix is catalyzed by Duox, a multidomain oxidase/peroxidase with homol- 280. Federici M, Menghini R, Mauriello A, Hribal ML, Ferrelli F, Lauro D, Sbraccia P, ogy to the phagocyte oxidase subunit gp91phox. J Cell Biol 154: 879–891, 2001. Spagnoli LG, Sesti G, Lauro R. Insulin-dependent activation of endothelial nitric oxide doi:10.1083/jcb.200103132. synthase is impaired by O-linked glycosylation modification of signaling proteins in human coronary endothelial cells. Circulation 106: 466–472, 2002. doi:10.1161/01. 263. Eguchi D, D’Uscio LV, Wambi C, Weiler D, Kovesdi I, O’Brien T, Katusic ZS. CIR.0000023043.02648.51. Inhibitory effect of recombinant iNOS gene expression on vasomotor function of canine basilar artery. Am J Physiol Heart Circ Physiol 283: H2560–H2566, 2002. 281. Fenton HJH. LXXIII. Oxidation of tartaric acid in presence of iron. J Chem Soc Trans doi:10.1152/ajpheart.00415.2002. 65: 899–910, 1894. doi:10.1039/CT8946500899.

264. Eich RF, Li T, Lemon DD, Doherty DH, Curry SR, Aitken JF, Mathews AJ, Johnson 282. Feoli AM, Macagnan FE, Piovesan CH, Bodanese LC, Siqueira IR. Xanthine oxidase KA, Smith RD, Phillips GN Jr, Olson JS. Mechanism of NO-induced oxidation of activity is associated with risk factors for cardiovascular disease and inflammatory myoglobin and hemoglobin. Biochemistry 35: 6976–6983, 1996. doi:10.1021/ and oxidative status markers in metabolic syndrome: effects of a single exercise bi960442g. session. Oxid Med Cell Longev 2014: 587083, 2014. doi:10.1155/2014/587083.

356 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

283. Feron O, Belhassen L, Kobzik L, Smith TW, Kelly RA, Michel T. Endothelial nitric Stimulator Vericiguat (BAY 1021189) for the Treatment of Chronic Heart Failure. J oxide synthase targeting to caveolae. Specific interactions with caveolin isoforms in Med Chem 60: 5146–5161, 2017. doi:10.1021/acs.jmedchem.7b00449. cardiac myocytes and endothelial cells. J Biol Chem 271: 22810–22814, 1996. doi: 10.1074/jbc.271.37.22810. 302. Follmann M, Griebenow N, Hahn MG, Hartung I, Mais F-J, Mittendorf J, Schäfer M, Schirok H, Stasch J-P, Stoll F, Straub A. The chemistry and biology of soluble guan- 284. Feron O, Michel JB, Sase K, Michel T. Dynamic regulation of endothelial nitric oxide ylate cyclase stimulators and activators. Angew Chem Int Ed Engl 52: 9442–9462, synthase: complementary roles of dual acylation and caveolin interactions. Biochem- 2013. doi:10.1002/anie.201302588. istry 37: 193–200, 1998. doi:10.1021/bi972307p. 303. Förstermann U, Münzel T. Endothelial nitric oxide synthase in vascular disease: 285. Figueroa A, Alvarez-Alvarado S, Jaime SJ, Kalfon R. l-Citrulline supplementation from marvel to menace. Circulation 113: 1708–1714, 2006. doi:10.1161/ attenuates blood pressure, wave reflection and arterial stiffness responses to CIRCULATIONAHA.105.602532. metaboreflex and cold stress in overweight men. Br J Nutr 116: 279–285, 2016. doi:10.1017/S0007114516001811. 304. Foti A, Dorendorf F, Leimkühler S. A single nucleotide polymorphism causes en- hanced radical oxygen species production by human aldehyde oxidase. PLoS One 12: 286. Figueroa A, Trivino JA, Sanchez-Gonzalez MA, Vicil F. Oral L-citrulline supplemen- e0182061, 2017. doi:10.1371/journal.pone.0182061. tation attenuates blood pressure response to cold pressor test in young men. Am J Hypertens 23: 12–16, 2010. doi:10.1038/ajh.2009.195. 305. Fravel MA, Ernst ME. Management of gout in the older adult. Am J Geriatr Pharma- cother 9: 271–285, 2011. doi:10.1016/j.amjopharm.2011.07.004. 287. Filippatos G, Maggioni AP, Lam CSP, Pieske-Kraigher E, Butler J, Spertus J, Pon- ikowski P, Shah SJ, Solomon SD, Scalise AV, Mueller K, Roessig L, Bamber L, Gheo- 306. Frazziano G, Al Ghouleh I, Baust J, Shiva S, Champion HC, Pagano PJ. Nox-derived rghiade M, Pieske B. Patient-reported outcomes in the SOluble guanylate Cyclase ROS are acutely activated in pressure overload pulmonary hypertension: indications stimulatoR in heArT failurE patientS with PRESERVED ejection fraction (SO- for a seminal role for mitochondrial Nox4. Am J Physiol Heart Circ Physiol 306: CRATES-PRESERVED) study. Eur J Heart Fail 19: 782–791, 2017. doi:10.1002/ejhf. H197–H205, 2014. doi:10.1152/ajpheart.00977.2012. 800. 307. Fridovich I. Quantitative aspects of the production of superoxide anion radical by 288. Finegold AA, Shatwell KP, Segal AW, Klausner RD, Dancis A. Intramembrane bis- milk xanthine oxidase. J Biol Chem 245: 4053–4057, 1970. heme motif for transmembrane electron transport conserved in a yeast iron reduc- 308. Fridovich I, Handler P. Xanthine oxidase. V. Differential inhibition of the reduction of tase and the human NADPH oxidase. J Biol Chem 271: 31021–31024, 1996. doi:10. various electron acceptors. J Biol Chem 237: 916–921, 1962. 1074/jbc.271.49.31021. 309. Friebe A, Müllershausen F, Smolenski A, Walter U, Schultz G, Koesling D. YC-1 289. Finkel T. Oxygen radicals and signaling. Curr Opin Cell Biol 10: 248–253, 1998. potentiates nitric oxide- and carbon monoxide-induced cyclic GMP effects in human doi:10.1016/S0955-0674(98)80147-6. platelets. Mol Pharmacol 54: 962–967, 1998. doi:10.1124/mol.54.6.962. 290. Finkel T. Signal transduction by mitochondrial oxidants. J Biol Chem 287: 4434–4440, 310. Friebe A, Schultz G, Koesling D. Sensitizing soluble guanylyl cyclase to become a 2012. doi:10.1074/jbc.R111.271999. highly CO-sensitive enzyme. EMBO J 15: 6863–6868, 1996. 291. Finkel T. Signal transduction by reactive oxygen species. J Cell Biol 194: 7–15, 2011. 311. Friedl HP, Till GO, Ryan US, Ward PA. Mediator-induced activation of xanthine doi:10.1083/jcb.201102095. oxidase in endothelial cells. FASEB J 3: 2512–2518, 1989. doi:10.1096/fasebj.3.13. 292. Fisslthaler B, Loot AE, Mohamed A, Busse R, Fleming I. Inhibition of endothelial nitric 2806779. oxide synthase activity by proline-rich tyrosine kinase 2 in response to fluid shear 312. Fritz BG, Hu X, Brailey JL, Berry RE, Walker FA, Montfort WR. Oxidation and loss of stress and insulin. Circ Res 102: 1520–1528, 2008. doi:10.1161/CIRCRESAHA.108. heme in soluble guanylyl cyclase from Manduca sexta. Biochemistry 50: 5813–5815, 172072. 2011. doi:10.1021/bi200794c. 293. Flaherty JP, Spruce CA, Fairfield HE, Bergstrom DE. Generation of a conditional null 313. Fritz BG, Roberts SA, Ahmed A, Breci L, Li W, Weichsel A, Brailey JL, Wysocki VH, allele of NADPH oxidase activator 1 (NOXA1). Genesis 48: 568–575, 2010. doi:10. Tama F, Montfort WR. Molecular model of a soluble guanylyl cyclase fragment 1002/dvg.20655. determined by small-angle X-ray scattering and chemical cross-linking. Biochemistry 294. Flam BR, Hartmann PJ, Harrell-Booth M, Solomonson LP, Eichler DC. Caveolar 52: 1568–1582, 2013. doi:10.1021/bi301570m. localization of arginine regeneration enzymes, argininosuccinate synthase, and lyase, 314. Fu X, Kassim SY, Parks WC, Heinecke JW. Hypochlorous acid oxygenates the with endothelial nitric oxide synthase. Nitric Oxide 5: 187–197, 2001. doi:10.1006/ cysteine switch domain of pro-matrilysin (MMP-7). A mechanism for matrix metal- niox.2001.0340. loproteinase activation and atherosclerotic plaque rupture by myeloperoxidase. J 295. Fleming I. Cytochrome p450 and vascular homeostasis. Circ Res 89: 753–762, 2001. Biol Chem 276: 41279–41287, 2001. doi:10.1074/jbc.M106958200. doi:10.1161/hh2101.099268. 315. Fujii H, Ichimori K, Hoshiai K, Nakazawa H. Nitric oxide inactivates NADPH oxidase 296. Fleming I. Molecular mechanisms underlying the activation of eNOS. Pflugers Arch in pig neutrophils by inhibiting its assembling process. J Biol Chem 272: 32773–32778, 459: 793–806, 2010. doi:10.1007/s00424-009-0767-7. 1997. doi:10.1074/jbc.272.52.32773.

297. Fleming I, Fisslthaler B, Dimmeler S, Kemp BE, Busse R. Phosphorylation of Thr(495) 316. Fujii M, Amanso A, Abrahão TB, Lassègue B, Griendling KK. Polymerase delta- regulates Ca(2ϩ)/calmodulin-dependent endothelial nitric oxide synthase activity. interacting protein 2 regulates collagen accumulation via activation of the Akt/mTOR Circ Res 88: E68–E75, 2001. doi:10.1161/hh1101.092677. pathway in vascular smooth muscle cells. J Mol Cell Cardiol 92: 21–29, 2016. doi:10. 1016/j.yjmcc.2016.01.016. 298. Fleming I, Michaelis UR, Bredenkötter D, Fisslthaler B, Dehghani F, Brandes RP, Busse R. Endothelium-derived hyperpolarizing factor synthase (Cytochrome P450 317. Fujimoto H, Taguchi J, Imai Y, Ayabe S, Hashimoto H, Kobayashi H, Ogasawara K, 2C9) is a functionally significant source of reactive oxygen species in coronary arter- Aizawa T, Yamakado M, Nagai R, Ohno M. Manganese superoxide dismutase poly- ies. Circ Res 88: 44–51, 2001. doi:10.1161/01.RES.88.1.44. morphism affects the oxidized low-density lipoprotein-induced apoptosis of macro- phages and coronary artery disease. Eur Heart J 29: 1267–1274, 2008. doi:10.1093/ 299. Förstermann U, Sessa WC. Nitric oxide synthases: regulation and function. Eur Heart eurheartj/ehm500. J 33: 829–837, 2012. doi:10.1093/eurheartj/ehr304. 318. Fukuda Y, Teragawa H, Matsuda K, Yamagata T, Matsuura H, Chayama K. Tetrahy- 300. Folkes LK, Candeias LP, Wardman P. Kinetics and mechanisms of hypochlorous acid drobiopterin restores endothelial function of coronary arteries in patients with hy- reactions. Arch Biochem Biophys 323: 120–126, 1995. doi:10.1006/abbi.1995.0017. percholesterolaemia. Heart 87: 264–269, 2002. doi:10.1136/heart.87.3.264.

301. Follmann M, Ackerstaff J, Redlich G, Wunder F, Lang D, Kern A, Fey P, Griebenow 319. Fukui T, Ishizaka N, Rajagopalan S, Laursen JB, Capers Q IV, Taylor WR, Harrison N, Kroh W, Becker-Pelster EM, Kretschmer A, Geiss V, Li V, Straub A, Mittendorf J, DG, de Leon H, Wilcox JN, Griendling KK. p22phox mRNA expression and NADPH Jautelat R, Schirok H, Schlemmer KH, Lustig K, Gerisch M, Knorr A, Tinel H, Mon- oxidase activity are increased in aortas from hypertensive rats. Circ Res 80: 45–51, dritzki T, Trübel H, Sandner P, Stasch JP. Discovery of the Soluble Guanylate Cyclase 1997. doi:10.1161/01.RES.80.1.45.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 357 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

320. Fulton D, Church JE, Ruan L, Li C, Sood SG, Kemp BE, Jennings IG, Venema RC. Src Mete A, Cheshire DR, Connolly S, Stuehr DJ, Aberg A, Wallace AV, Tainer JA, kinase activates endothelial nitric-oxide synthase by phosphorylating Tyr-83. J Biol Getzoff ED. Anchored plasticity opens doors for selective inhibitor design in nitric Chem 280: 35943–35952, 2005. doi:10.1074/jbc.M504606200. oxide synthase. Nat Chem Biol 4: 700–707, 2008. doi:10.1038/nchembio.115.

321. Fulton D, Gratton JP, McCabe TJ, Fontana J, Fujio Y, Walsh K, Franke TF, Papapetro- 338. Garcin ED, Bruns CM, Lloyd SJ, Hosfield DJ, Tiso M, Gachhui R, Stuehr DJ, Tainer JA, poulos A, Sessa WC. Regulation of endothelium-derived nitric oxide production by Getzoff ED. Structural basis for isozyme-specific regulation of electron transfer in the protein kinase Akt. Nature 399: 597–601, 1999. doi:10.1038/21218. Erratum at: nitric-oxide synthase. J Biol Chem 279: 37918–37927, 2004. doi:10.1074/jbc. Nature 399: 597–601, 1999. doi:10.1038/23523. M406204200.

322. Fulton D, Ruan L, Sood SG, Li C, Zhang Q, Venema RC. Agonist-stimulated endo- 339. Gardner AM, Cook MR, Gardner PR. Nitric-oxide dioxygenase function of human thelial nitric oxide synthase activation and vascular relaxation. Role of eNOS phos- cytoglobin with cellular reductants and in rat hepatocytes. J Biol Chem 285: 23850– phorylation at Tyr83. Circ Res 102: 497–504, 2008. doi:10.1161/CIRCRESAHA.107. 23857, 2010. doi:10.1074/jbc.M110.132340. 162933. 340. Gardner AM, Gardner PR. Flavohemoglobin detoxifies nitric oxide in aerobic, but 323. Fulton DJ, Barman SA. Clarity on the Isoform-Specific Roles of NADPH Oxidases not anaerobic, Escherichia coli. Evidence for a novel inducible anaerobic nitric oxide- and NADPH Oxidase-4 in Atherosclerosis. Arterioscler Thromb Vasc Biol 36: 579– scavenging activity. J Biol Chem 277: 8166–8171, 2002. doi:10.1074/jbc. 581, 2016. doi:10.1161/ATVBAHA.116.307096. M110470200.

324. Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the relaxation 341. Gardner PR. Nitric oxide dioxygenase function and mechanism of flavohemoglobin, of arterial smooth muscle by acetylcholine. Nature 288: 373–376, 1980. doi:10.1038/ hemoglobin, myoglobin and their associated reductases. J Inorg Biochem 99: 247– 288373a0. 266, 2005. doi:10.1016/j.jinorgbio.2004.10.003.

325. Gago B, Lundberg JO, Barbosa RM, Laranjinha J. Red wine-dependent reduction of 342. Gardner PR, Gardner AM, Martin LA, Salzman AL. Nitric oxide dioxygenase: an nitrite to nitric oxide in the stomach. Free Radic Biol Med 43: 1233–1242, 2007. enzymic function for flavohemoglobin. Proc Natl Acad Sci USA 95: 10378–10383, doi:10.1016/j.freeradbiomed.2007.06.007. 1998. doi:10.1073/pnas.95.18.10378.

326. Galle J, Zabel U, Hübner U, Hatzelmann A, Wagner B, Wanner C, Schmidt HH. 343. Garrido-Urbani S, Jemelin S, Deffert C, Carnesecchi S, Basset O, Szyndralewiez C, Effects of the soluble guanylyl cyclase activator, YC-1, on vascular tone, cyclic GMP Heitz F, Page P, Montet X, Michalik L, Arbiser J, Rüegg C, Krause KH, Imhof BA. levels and phosphodiesterase activity. Br J Pharmacol 127: 195–203, 1999. doi:10. Targeting vascular NADPH oxidase 1 blocks tumor angiogenesis through a PPAR␣ 1038/sj.bjp.0702495. mediated mechanism. PLoS One 6: e14665, 2011. doi:10.1371/journal.pone. 0014665. Erratum at: http://dx.doi.org/10.1371/annotation/a392bbef-b0ec-4c70- 327. Gallis B, Corthals GL, Goodlett DR, Ueba H, Kim F, Presnell SR, Figeys D, Harrison b403-74a7bad85178. DG, Berk BC, Aebersold R, Corson MA. Identification of flow-dependent endothe- lial nitric-oxide synthase phosphorylation sites by mass spectrometry and regulation 344. Garvey EP, Oplinger JA, Furfine ES, Kiff RJ, Laszlo F, Whittle BJ, Knowles RG. 1400W of phosphorylation and nitric oxide production by the phosphatidylinositol 3-kinase is a slow, tight binding, and highly selective inhibitor of inducible nitric-oxide synthase inhibitor LY294002. J Biol Chem 274: 30101–30108, 1999. doi:10.1074/jbc.274.42. in vitro and in vivo. J Biol Chem 272: 4959–4963, 1997. doi:10.1074/jbc.272.8.4959. 30101. 345. Gavazzi G, Banfi B, Deffert C, Fiette L, Schappi M, Herrmann F, Krause KH. De- 328. Galougahi KK, Liu CC, Gentile C, Kok C, Nunez A, Garcia A, Fry NA, Davies MJ, creased blood pressure in NOX1-deficient mice. FEBS Lett 580: 497–504, 2006. Hawkins CL, Rasmussen HH, Figtree GA. Glutathionylation mediates angiotensin doi:10.1016/j.febslet.2005.12.049. II-induced eNOS uncoupling, amplifying NADPH oxidase-dependent endothelial dysfunction. J Am Heart Assoc 3: e000731, 2014. doi:10.1161/JAHA.113.000731. 346. Gavazzi G, Deffert C, Trocme C, Schäppi M, Herrmann FR, Krause K-H. NOX1 deficiency protects from aortic dissection in response to angiotensin II. Hypertension 329. Gamgee A. Researches on the blood. On the action of nitrites on blood. Philos Trans 50: 189–196, 2007. doi:10.1161/HYPERTENSIONAHA.107.089706. R Soc Lond 158: 589–625, 1868. doi:10.1098/rstl.1868.0025. 347. Geiszt M, Kopp JB, Várnai P, Leto TL. Identification of renox, an NAD(P)H oxidase 330. Gan P, Gao Z, Zhao X, Qi G. Surfactin inducing mitochondria-dependent ROS to in kidney. Proc Natl Acad Sci USA 97: 8010–8014, 2000. doi:10.1073/pnas. activate MAPKs, NF-␬B and inflammasomes in macrophages for adjuvant activity. Sci 130135897. Rep 6: 39303, 2016. doi:10.1038/srep39303. 348. Geiszt M, Lekstrom K, Witta J, Leto TL. Proteins homologous to p47phox and 331. Gandhi PK, Gentry WM, Bottorff MB. Cardiovascular thromboembolic events asso- p67phox support superoxide production by NAD(P)H oxidase 1 in colon epithelial ciated with febuxostat: investigation of cases from the FDA adverse event reporting cells. J Biol Chem 278: 20006–20012, 2003. doi:10.1074/jbc.M301289200. system database. Semin Arthritis Rheum 42: 562–566, 2013. doi:10.1016/j. semarthrit.2012.11.002. 349. Gheorghiade M, Greene SJ, Butler J, Filippatos G, Lam CS, Maggioni AP, Ponikowski P, Shah SJ, Solomon SD, Kraigher-Krainer E, Samano ET, Müller K, Roessig L, Pieske 332. Gandley RE, Tyurin VA, Huang W, Arroyo A, Daftary A, Harger G, Jiang J, Pitt B, B; SOCRATES-REDUCED Investigators and Coordinators. Effect of Vericiguat, a Taylor RN, Hubel CA, Kagan VE. S-nitrosoalbumin-mediated relaxation is enhanced Soluble Guanylate Cyclase Stimulator, on Natriuretic Peptide Levels in Patients With by ascorbate and copper: effects in pregnancy and preeclampsia plasma. Hyperten- Worsening Chronic Heart Failure and Reduced Ejection Fraction: The SOCRATES- sion 45: 21–27, 2005. doi:10.1161/01.HYP.0000150158.42620.3e. REDUCED Randomized Trial. JAMA 314: 2251–2262, 2015. doi:10.1001/jama.2015. 15734. 333. Gane EJ, Weilert F, Orr DW, Keogh GF, Gibson M, Lockhart MM, Frampton CM, Taylor KM, Smith RA, Murphy MP. The mitochondria-targeted anti-oxidant mito- 350. Gherasim C, Yadav PK, Kabil O, Niu WN, Banerjee R. Nitrite reductase activity and quinone decreases liver damage in a phase II study of hepatitis C patients. Liver Int 30: inhibition of H2S biogenesis by human cystathionine ß-synthase. PLoS One 9: e85544, 1019–1026, 2010. doi:10.1111/j.1478-3231.2010.02250.x. 2014. doi:10.1371/journal.pone.0085544.

334. Gao YT, Roman LJ, Martásek P, Panda SP, Ishimura Y, Masters BS. Oxygen metab- 351. Ghofrani H-A, D’Armini AM, Grimminger F, Hoeper MM, Jansa P, Kim NH, Mayer E, olism by endothelial nitric-oxide synthase. J Biol Chem 282: 28557–28565, 2007. Simonneau G, Wilkins MR, Fritsch A, Neuser D, Weimann G, Wang C; CHEST-1 doi:10.1074/jbc.M704890200. Study Group. Riociguat for the treatment of chronic thromboembolic pulmonary hypertension. N Engl J Med 369: 319–329, 2013. doi:10.1056/NEJMoa1209657. 335. Garattini E, Terao M. The role of aldehyde oxidase in drug metabolism. Expert Opin Drug Metab Toxicol 8: 487–503, 2012. doi:10.1517/17425255.2012.663352. 352. Ghofrani H-A, Galiè N, Grimminger F, Grünig E, Humbert M, Jing Z-C, Keogh AM, Langleben D, Kilama MO, Fritsch A, Neuser D, Rubin LJ; PATENT-1 Study Group. 336. García-Cardeña G, Oh P, Liu J, Schnitzer JE, Sessa WC. Targeting of nitric oxide Riociguat for the treatment of pulmonary arterial hypertension. N Engl J Med 369: synthase to endothelial cell caveolae via palmitoylation: implications for nitric oxide 330–340, 2013. doi:10.1056/NEJMoa1209655. signaling. Proc Natl Acad Sci USA 93: 6448–6453, 1996. doi:10.1073/pnas.93.13. 6448. 353. Ghosh A, Koziol-White CJ, Asosingh K, Cheng G, Ruple L, Groneberg D, Friebe A, Comhair SA, Stasch JP, Panettieri RA Jr, Aronica MA, Erzurum SC, Stuehr DJ. Soluble 337. Garcin ED, Arvai AS, Rosenfeld RJ, Kroeger MD, Crane BR, Andersson G, Andrews guanylate cyclase as an alternative target for bronchodilator therapy in asthma. Proc G, Hamley PJ, Mallinder PR, Nicholls DJ, St-Gallay SA, Tinker AC, Gensmantel NP, Natl Acad Sci USA 113: E2355–E2362, 2016. doi:10.1073/pnas.1524398113.

358 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

354. Ghosh A, Stasch JP, Papapetropoulos A, Stuehr DJ. Nitric oxide and heat shock 370. Godber BL, Doel JJ, Sapkota GP, Blake DR, Stevens CR, Eisenthal R, Harrison R. protein 90 activate soluble guanylate cyclase by driving rapid change in its subunit Reduction of nitrite to nitric oxide catalyzed by xanthine oxidoreductase. J Biol Chem interactions and heme content. J Biol Chem 289: 15259–15271, 2014. doi:10.1074/ 275: 7757–7763, 2000. doi:10.1074/jbc.275.11.7757. jbc.M114.559393. 371. Gole HK, Tharp DL, Bowles DK. Upregulation of intermediate-conductance Ca2ϩ- 355. Ghosh A, Stuehr DJ. Regulation of sGC via hsp90, Cellular Heme, sGC Agonists, and activated Kϩ channels (KCNN4) in porcine coronary smooth muscle requires NA- NO: New Pathways and Clinical Perspectives. Antioxid Redox Signal 26: 182–190, DPH oxidase 5 (NOX5). PLoS One 9: e105337, 2014. doi:10.1371/journal.pone. 2017. doi:10.1089/ars.2016.6690. 0105337.

356. Ghosh A, Stuehr DJ. Soluble guanylyl cyclase requires heat shock protein 90 for 372. Gonzalez DR, Beigi F, Treuer AV, Hare JM. Deficient ryanodine receptor S-nitrosy- heme insertion during maturation of the NO-active enzyme. Proc Natl Acad Sci USA lation increases sarcoplasmic reticulum calcium leak and arrhythmogenesis in cardi- 109: 12998–13003, 2012. doi:10.1073/pnas.1205854109. omyocytes. Proc Natl Acad Sci USA 104: 20612–20617, 2007. doi:10.1073/pnas. 0706796104. 357. Ghosh SM, Kapil V, Fuentes-Calvo I, Bubb KJ, Pearl V, Milsom AB, Khambata R, Maleki-Toyserkani S, Yousuf M, Benjamin N, Webb AJ, Caulfield MJ, Hobbs AJ, 373. Gonzalez DR, Treuer AV, Castellanos J, Dulce RA, Hare JM. Impaired S-nitrosylation Ahluwalia A. Enhanced vasodilator activity of nitrite in hypertension: critical role for of the ryanodine receptor caused by xanthine oxidase activity contributes to calcium erythrocytic xanthine oxidoreductase and translational potential. Hypertension 61: leak in heart failure. J Biol Chem 285: 28938–28945, 2010. doi:10.1074/jbc.M110. 154948. 1091–1102, 2013. doi:10.1161/HYPERTENSIONAHA.111.00933. 374. Gonzalez FM, Shiva S, Vincent PS, Ringwood LA, Hsu LY, Hon YY, Aletras AH, 358. Giaid A, Saleh D. Reduced expression of endothelial nitric oxide synthase in the lungs Cannon RO III, Gladwin MT, Arai AE. Nitrite anion provides potent cytoprotective of patients with pulmonary hypertension. N Engl J Med 333: 214–221, 1995. doi:10. and antiapoptotic effects as adjunctive therapy to reperfusion for acute myocardial 1056/NEJM199507273330403. infarction. Circulation 117: 2986–2994, 2008. doi:10.1161/CIRCULATIONAHA. 359. Gianni D, Taulet N, Zhang H, DerMardirossian C, Kister J, Martinez L, Roush WR, 107.748814. Brown SJ, Bokoch GM, Rosen H. A novel and specific NADPH oxidase-1 (Nox1) 375. Gori T, Burstein JM, Ahmed S, Miner SES, Al-Hesayen A, Kelly S, Parker JD. Folic acid small-molecule inhibitor blocks the formation of functional invadopodia in human prevents nitroglycerin-induced nitric oxide synthase dysfunction and nitrate toler- colon cancer cells. ACS Chem Biol 5: 981–993, 2010. doi:10.1021/cb100219n. ance: a human in vivo study. Circulation 104: 1119–1123, 2001. doi:10.1161/hc3501. 095358. 360. Gibson CM, Giugliano RP, Kloner RA, Bode C, Tendera M, Jánosi A, Merkely B, Godlewski J, Halaby R, Korjian S, Daaboul Y, Chakrabarti AK, Spielman K, Neal BJ, 376. Görlach A, Brandes RP, Nguyen K, Amidi M, Dehghani F, Busse R. A gp91phox Weaver WD. EMBRACE STEMI study: a Phase 2a trial to evaluate the safety, toler- containing NADPH oxidase selectively expressed in endothelial cells is a major ability, and efficacy of intravenous MTP-131 on reperfusion injury in patients under- source of oxygen radical generation in the arterial wall. Circ Res 87: 26–32, 2000. going primary percutaneous coronary intervention. Eur Heart J 37: 1296–1303, doi:10.1161/01.RES.87.1.26. 2016. doi:10.1093/eurheartj/ehv597. 377. Gorren AC, Mayer B. Nitric-oxide synthase: a cytochrome P450 family foster child. 361. Givertz MM, Anstrom KJ, Redfield MM, Deswal A, H, Butler J, Tang WH, Biochim Biophys Acta 1770: 432–445, 2007. doi:10.1016/j.bbagen.2006.08.019. Dunlap ME, LeWinter MM, Mann DL, Felker GM, O’Connor CM, Goldsmith SR, Ofili EO, Saltzberg MT, Margulies KB, Cappola TP, Konstam MA, Semigran MJ, 378. Gould NS, Evans P, Martínez-Acedo P, Marino SM, Gladyshev VN, Carroll KS, McNulty SE, Lee KL, Shah MR, Hernandez AF; NHLBI Heart Failure Clinical Re- Ischiropoulos H. Site-Specific Proteomic Mapping Identifies Selectively Modified search Network. Effects of Xanthine Oxidase Inhibition in Hyperuricemic Heart Regulatory Cysteine Residues in Functionally Distinct Protein Networks. Chem Biol Failure Patients: The Xanthine Oxidase Inhibition for Hyperuricemic Heart Failure 22: 965–975, 2015. doi:10.1016/j.chembiol.2015.06.010. Patients (EXACT-HF) Study. Circulation 131: 1763–1771, 2015. doi:10.1161/ 379. Gray SP, Di Marco E, Kennedy K, Chew P, Okabe J, El-Osta A, Calkin AC, Biessen CIRCULATIONAHA.114.014536. EA, Touyz RM, Cooper ME, Schmidt HH, Jandeleit-Dahm KA. Reactive Oxygen 362. Gladden JD, Zelickson BR, Wei CC, Ulasova E, Zheng J, Ahmed MI, Chen Y, Bam- Species Can Provide Atheroprotection via NOX4-Dependent Inhibition of Inflam- man M, Ballinger S, Darley-Usmar V, Dell’Italia LJ. Novel insights into interactions mation and Vascular Remodeling. Arterioscler Thromb Vasc Biol 36: 295–307, 2016. between mitochondria and xanthine oxidase in acute cardiac volume overload. Free doi:10.1161/ATVBAHA.115.307012. Radic Biol Med 51: 1975–1984, 2011. doi:10.1016/j.freeradbiomed.2011.08.022. 380. Gray SP, Di Marco E, Okabe J, Szyndralewiez C, Heitz F, Montezano AC, de Haan JB, Koulis C, El-Osta A, Andrews KL, Chin-Dusting JPF, Touyz RM, Wingler K, Cooper 363. Gladwin MT. Deconstructing endothelial dysfunction: soluble guanylyl cyclase oxi- ME, Schmidt HHHW, Jandeleit-Dahm KA. NADPH oxidase 1 plays a key role in dation and the NO resistance syndrome. J Clin Invest 116: 2330–2332, 2006. doi:10. diabetes mellitus-accelerated atherosclerosis. Circulation 127: 1888–1902, 2013. 1172/JCI29807. doi:10.1161/CIRCULATIONAHA.112.132159. 364. Gladwin MT. Haldane, hot dogs, halitosis, and hypoxic vasodilation: the emerging 381. Greenacre SA, Ischiropoulos H. Tyrosine nitration: localisation, quantification, con- biology of the nitrite anion. J Clin Invest 113: 19–21, 2004. doi:10.1172/JCI20664. sequences for protein function and signal transduction. Free Radic Res 34: 541–581, 365. Gladwin MT. Hemoglobin as a nitrite reductase regulating red cell-dependent hy- 2001. doi:10.1080/10715760100300471. poxic vasodilation. Am J Respir Cell Mol Biol 32: 363–366, 2005. doi:10.1165/rcmb. 382. Griendling KK, Minieri CA, Ollerenshaw JD, Alexander RW. Angiotensin II stimulates F294. NADH and NADPH oxidase activity in cultured vascular smooth muscle cells. Circ Res 74: 1141–1148, 1994. doi:10.1161/01.RES.74.6.1141. 366. Gladwin MT, Grubina R, Doyle MP. The new chemical biology of nitrite reactions with hemoglobin: R-state catalysis, oxidative denitrosylation, and nitrite reductase/ 383. Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H oxidase: role in cardiovascular anhydrase. Acc Chem Res 42: 157–167, 2009. doi:10.1021/ar800089j. biology and disease. Circ Res 86: 494–501, 2000. doi:10.1161/01.RES.86.5.494.

367. Gladwin MT, Kim-Shapiro DB. The functional nitrite reductase activity of the heme- 384. Grubina R, Huang Z, Shiva S, Joshi MS, Azarov I, Basu S, Ringwood LA, Jiang A, Hogg globins. Blood 112: 2636–2647, 2008. doi:10.1182/blood-2008-01-115261. N, Kim-Shapiro DB, Gladwin MT. Concerted nitric oxide formation and release from the simultaneous reactions of nitrite with deoxy- and oxyhemoglobin. J Biol 368. Gladwin MT, Raat NJ, Shiva S, Dezfulian C, Hogg N, Kim-Shapiro DB, Patel RP. Chem 282: 12916–12927, 2007. doi:10.1074/jbc.M700546200. Nitrite as a vascular endocrine nitric oxide reservoir that contributes to hypoxic signaling, cytoprotection, and vasodilation. Am J Physiol Heart Circ Physiol 291: 385. Gryglewski RJ, Palmer RMJ, Moncada S. Superoxide anion is involved in the break- H2026–H2035, 2006. doi:10.1152/ajpheart.00407.2006. down of endothelium-derived vascular relaxing factor. Nature 320: 454–456, 1986. doi:10.1038/320454a0. 369. Gladwin MT, Shelhamer JH, Schechter AN, Pease-Fye ME, Waclawiw MA, Panza JA, Ognibene FP, Cannon RO III. Role of circulating nitrite and S-nitrosohemoglobin in 386. Gunnett CA, Lund DD, McDowell AK, Faraci FM, Heistad DD. Mechanisms of the regulation of regional blood flow in humans. Proc Natl Acad Sci USA 97: 11482– inducible nitric oxide synthase-mediated vascular dysfunction. Arterioscler Thromb 11487, 2000. doi:10.1073/pnas.97.21.11482. Vasc Biol 25: 1617–1622, 2005. doi:10.1161/01.ATV.0000172626.00296.ba.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 359 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

387. Guthikonda S, Sinkey C, Barenz T, Haynes WG. Xanthine oxidase inhibition reverses 406. Harris CM, Massey V. The reaction of reduced xanthine dehydrogenase with mo- endothelial dysfunction in heavy smokers. Circulation 107: 416–421, 2003. doi:10. lecular oxygen. Reaction kinetics and measurement of superoxide radical. J Biol Chem 1161/01.CIR.0000046448.26751.58. 272: 8370–8379, 1997. doi:10.1074/jbc.272.13.8370.

388. Gutteridge JM. Inhibition of the Fenton reaction by the protein caeruloplasmin and 407. Harris MB, Ju H, Venema VJ, Liang H, Zou R, Michell BJ, Chen ZP, Kemp BE, Venema other copper complexes. Assessment of ferroxidase and radical scavenging activi- RC. Reciprocal phosphorylation and regulation of endothelial nitric-oxide synthase in ties. Chem Biol Interact 56: 113–120, 1985. doi:10.1016/0009-2797(85)90043-2. response to bradykinin stimulation. J Biol Chem 276: 16587–16591, 2001. doi:10. 1074/jbc.M100229200. 389. Gutteridge JM. Iron promoters of the Fenton reaction and lipid peroxidation can be released from haemoglobin by peroxides. FEBS Lett 201: 291–295, 1986. doi:10. 408. Harrison D, Griendling KK, Landmesser U, Hornig B, Drexler H. Role of oxidative 1016/0014-5793(86)80626-3. stress in atherosclerosis. Am J Cardiol 91: 7A–11A, 2003. doi:10.1016/S0002- 9149(02)03144-2. 390. Gutteridge JMC, Halliwell B. Antioxidants: Molecules, medicines, and myths. Biochem Biophys Res Commun 393: 561–564, 2010. doi:10.1016/j.bbrc.2010.02.071. 409. Harrison DG, Ohara Y. Physiologic consequences of increased vascular oxidant stresses in hypercholesterolemia and atherosclerosis: implications for impaired va- 391. Guzik TJ, Chen W, Gongora MC, Guzik B, Lob HE, Mangalat D, Hoch N, Dikalov S, somotion. Am J Cardiol 75: 75B–81B, 1995. doi:10.1016/0002-9149(95)80018-N. Rudzinski P, Kapelak B, Sadowski J, Harrison DG. Calcium-dependent NOX5 nico- tinamide adenine dinucleotide phosphate oxidase contributes to vascular oxidative 410. Hassoun PM, Yu F-S, Cote CG, Zulueta JJ, Sawhney R, Skinner KA, Skinner HB, Parks stress in human coronary artery disease. J Am Coll Cardiol 52: 1803–1809, 2008. DA, Lanzillo JJ. Upregulation of xanthine oxidase by lipopolysaccharide, interleu- doi:10.1016/j.jacc.2008.07.063. kin-1, and hypoxia. Role in acute lung injury. Am J Respir Crit Care Med 158: 299–305, 1998. doi:10.1164/ajrccm.158.1.9709116. 392. Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis of angiotensin II induced 411. Hausladen A, AJ, Stamler JS. Nitrosative stress: metabolic pathway involving hypertension and vascular dysfunction. J Exp Med 204: 2449–2460, 2007. doi:10. the flavohemoglobin. Proc Natl Acad Sci USA 95: 14100–14105, 1998. doi:10.1073/ pnas.95.24.14100. 1084/jem.20070657. 412. Hausladen A, Privalle CT, Keng T, DeAngelo J, Stamler JS. Nitrosative stress: acti- 393. Guzik TJ, Sadowski J, Guzik B, Jopek A, Kapelak B, Przybylowski P, Wierzbicki K, vation of the transcription factor OxyR. Cell 86: 719–729, 1996. doi:10.1016/S0092- Korbut R, Harrison DG, Channon KM. Coronary artery superoxide production and 8674(00)80147-6. nox isoform expression in human coronary artery disease. Arterioscler Thromb Vasc Biol 26: 333–339, 2006. doi:10.1161/01.ATV.0000196651.64776.51. 413. Hayashi T, Juliet PAR, Kano-Hayashi H, Tsunekawa T, Dingqunfang D, Sumi D, Matsui-Hirai H, Fukatsu A, Iguchi A. NADPH oxidase inhibitor, apocynin, restores 394. Haber F, Weiss J. The catalytic decomposition of hydrogen peroxide by iron salts. the impaired endothelial-dependent and -independent responses and scavenges su- Proc R Soc Lond A Math Phys Sci 147: 332–351, 1934. doi:10.1098/rspa.1934.0221. peroxide anion in rats with type 2 diabetes complicated by NO dysfunction. Diabetes 395. Haendeler J, Eckers A, Lukosz M, Unfried K, Altschmied J. Endothelial NADPH Obes Metab 7: 334–343, 2005. doi:10.1111/j.1463-1326.2004.00393.x. oxidase 2: when does it matter in atherosclerosis? Cardiovasc Res 94: 1–2, 2012. 414. Hazell LJ, Arnold L, Flowers D, Waeg G, Malle E, Stocker R. Presence of hypochlo- doi:10.1093/cvr/cvs106. rite-modified proteins in human atherosclerotic lesions. J Clin Invest 97: 1535–1544, 396. Hahn NE, Meischl C, Kawahara T, Musters RJ, Verhoef VM, van der Velden J, 1996. doi:10.1172/JCI118576. AB, Paulus WJ, van Rossum AC, Niessen HW, Krijnen PA. NOX5 expression is 415. Hazen SL, Heinecke JW. 3-Chlorotyrosine, a specific marker of myeloperoxidase- increased in intramyocardial blood vessels and cardiomyocytes after acute myocar- catalyzed oxidation, is markedly elevated in low density lipoprotein isolated from dial infarction in humans. Am J Pathol 180: 2222–2229, 2012. doi:10.1016/j.ajpath. human atherosclerotic intima. J Clin Invest 99: 2075–2081, 1997. doi:10.1172/ 2012.02.018. JCI119379.

397. Hall CN, Garthwaite J. What is the real physiological NO concentration in vivo? Nitric 416. Hecker L, Vittal R, Jones T, Jagirdar R, Luckhardt TR, Horowitz JC, Pennathur S, Oxide 21: 92–103, 2009. doi:10.1016/j.niox.2009.07.002. Martinez FJ, Thannickal VJ. NADPH oxidase-4 mediates myofibroblast activation and fibrogenic responses to lung injury. Nat Med 15: 1077–1081, 2009. doi:10.1038/ 398. Halligan KE, Jourd’heuil FL, Jourd’heuil D. Cytoglobin is expressed in the vasculature nm.2005. and regulates cell respiration and proliferation via nitric oxide dioxygenation. J Biol Chem 284: 8539–8547, 2009. doi:10.1074/jbc.M808231200. 417. Heiss C, Keen CL, Kelm M. Flavanols and cardiovascular disease prevention. Eur Heart J 31: 2583–2592, 2010. doi:10.1093/eurheartj/ehq332. 399. Halliwell B. The antioxidant paradox. Lancet 355: 1179–1180, 2000. doi:10.1016/ S0140-6736(00)02075-4. 418. Heitzer T, Brockhoff C, Mayer B, Warnholtz A, Mollnau H, Henne S, Meinertz T, Münzel T. Tetrahydrobiopterin improves endothelium-dependent vasodilation in 400. Halliwell B. The antioxidant paradox: less paradoxical now? Br J Clin Pharmacol 75: chronic smokers: evidence for a dysfunctional nitric oxide synthase. Circ Res 86: 637–644, 2013. doi:10.1111/j.1365-2125.2012.04272.x. E36–E41, 2000. doi:10.1161/01.RES.86.2.e36.

401. Han D, Canali R, Garcia J, Aguilera R, Gallaher TK, Cadenas E. Sites and mechanisms 419. Heitzer T, Finckh B, Albers S, Krohn K, Kohlschütter A, Meinertz T. Beneficial of aconitase inactivation by peroxynitrite: modulation by citrate and glutathione. effects of alpha-lipoic acid and ascorbic acid on endothelium-dependent, nitric ox- Biochemistry 44: 11986–11996, 2005. doi:10.1021/bi0509393. ide-mediated vasodilation in diabetic patients: relation to parameters of oxidative stress. Free Radic Biol Med 31: 53–61, 2001. doi:10.1016/S0891-5849(01)00551-2. 402. Handy RL, Moore PK. A comparison of the effects of L-NAME, 7-NI and L-NIL on carrageenan-induced hindpaw oedema and NOS activity. Br J Pharmacol 123: 1119– 420. Heitzer T, Just H, Münzel T. Antioxidant vitamin C improves endothelial dysfunction 1126, 1998. doi:10.1038/sj.bjp.0701735. in chronic smokers. Circulation 94: 6–9, 1996. doi:10.1161/01.CIR.94.1.6.

403. Hankeln T, Wystub S, Laufs T, Schmidt M, Gerlach F, Saaler-Reinhardt S, Reuss S, 421. Heitzer T, Krohn K, Albers S, Meinertz T. Tetrahydrobiopterin improves endo- Burmester T. The cellular and subcellular localization of neuroglobin and cytoglo- thelium-dependent vasodilation by increasing nitric oxide activity in patients with bin–a clue to their function? IUBMB Life 56: 671–679, 2004. doi:10.1080/ Type II diabetes mellitus. Diabetologia 43: 1435–1438, 2000. doi:10.1007/ 15216540500037794. s001250051551.

404. Haque MM, Ray SS, Stuehr DJ. Phosphorylation Controls Endothelial Nitric-oxide 422. Heller R, Unbehaun A, Schellenberg B, Mayer B, Werner-Felmayer G, Werner ER. Synthase by Regulating Its Conformational Dynamics. J Biol Chem 291: 23047–23057, L-ascorbic acid potentiates endothelial nitric oxide synthesis via a chemical stabiliza- 2016. doi:10.1074/jbc.M116.737361. tion of tetrahydrobiopterin. J Biol Chem 276: 40–47, 2001. doi:10.1074/jbc. M004392200. 405. Hare JM, Mangal B, Brown J, Fisher C Jr, Freudenberger R, Colucci WS, Mann DL, Liu P, Givertz MM, Schwarz RP; OPT-CHF Investigators. Impact of oxypurinol in 423. Helmcke I, Heumüller S, Tikkanen R, Schröder K, Brandes RP. Identification of patients with symptomatic heart failure. Results of the OPT-CHF study. J Am Coll structural elements in Nox1 and Nox4 controlling localization and activity. Antioxid Cardiol 51: 2301–2309, 2008. doi:10.1016/j.jacc.2008.01.068. Redox Signal 11: 1279–1287, 2009. doi:10.1089/ars.2008.2383.

360 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

424. Helms C, Kim-Shapiro DB. Hemoglobin-mediated nitric oxide signaling. Free Radic 442. Hosoya T, Sasaki T, Ohashi T. Clinical efficacy and safety of topiroxostat in Japanese Biol Med 61: 464–472, 2013. doi:10.1016/j.freeradbiomed.2013.04.028. hyperuricemic patients with or without gout: a randomized, double-blinded, con- trolled phase 2b study. Clin Rheumatol 36: 649–656, 2017. doi:10.1007/s10067-016- 425. Helms CC, Gladwin MT, Kim-Shapiro DB. Erythrocytes and vascular function: ox- 3474-8. ygen and nitric oxide. Front Physiol 9: 125, 2018. doi:10.3389/fphys.2018.00125. 443. Hsich E, Segal BH, Pagano PJ, Rey FE, Paigen B, Deleonardis J, Hoyt RF, Holland SM, 426. Heumüller S, Wind S, Barbosa-Sicard E, Schmidt HHHW, Busse R, Schröder K, Finkel T. Vascular effects following homozygous disruption of p47(phox): An essen- Brandes RP. Apocynin is not an inhibitor of vascular NADPH oxidases but an anti- tial component of NADPH oxidase. Circulation 101: 1234–1236, 2000. doi:10.1161/ oxidant. Hypertension 51: 211–217, 2008. doi:10.1161/HYPERTENSIONAHA.107. 01.CIR.101.11.1234. 100214. 444. Huang A, Sun D, Shesely EG, Levee EM, Koller A, Kaley G. Neuronal NOS-depen- 427. Heymes C, Bendall JK, Ratajczak P, Cave AC, Samuel JL, Hasenfuss G, Shah AM. dent dilation to flow in coronary arteries of male eNOS-KO mice. Am J Physiol Heart Increased myocardial NADPH oxidase activity in human heart failure. J Am Coll Circ Physiol 282: H429–H436, 2002. doi:10.1152/ajpheart.00501.2001. Cardiol 41: 2164–2171, 2003. doi:10.1016/S0735-1097(03)00471-6. 445. Huang KT, Keszler A, Patel N, Patel RP, Gladwin MT, Kim-Shapiro DB, Hogg N. The 428. Heyworth PG, Cross AR, Curnutte JT. Chronic granulomatous disease. Curr Opin reaction between nitrite and deoxyhemoglobin. Reassessment of reaction kinetics Immunol 15: 578–584, 2003. doi:10.1016/S0952-7915(03)00109-2. and stoichiometry. J Biol Chem 280: 31126–31131, 2005. doi:10.1074/jbc. M501496200. 429. Higashi Y, Sasaki S, Nakagawa K, Fukuda Y, Matsuura H, Oshima T, Chayama K. Tetrahydrobiopterin enhances forearm vascular response to acetylcholine in both 446. Huang PL, Huang Z, Mashimo H, Bloch KD, Moskowitz MA, Bevan JA, Fishman MC. normotensive and hypertensive individuals. Am J Hypertens 15: 326–332, 2002. Hypertension in mice lacking the gene for endothelial nitric oxide synthase. Nature doi:10.1016/S0895-7061(01)02317-2. 377: 239–242, 1995. doi:10.1038/377239a0.

430. Higgins P, Dawson J, Lees KR, McArthur K, Quinn TJ, Walters MR. Xanthine oxidase 447. Huang Z, Shiva S, Kim-Shapiro DB, Patel RP, Ringwood LA, Irby CE, Huang KT, Ho inhibition for the treatment of cardiovascular disease: a systematic review and meta- C, Hogg N, Schechter AN, Gladwin MT. Enzymatic function of hemoglobin as a nitrite reductase that produces NO under allosteric control. J Clin Invest 115: 2099– analysis. Cardiovasc Ther 30: 217–226, 2012. doi:10.1111/j.1755-5922.2011. 2107, 2005. doi:10.1172/JCI24650. 00277.x. 448. Ignarro LJ. Nitric Oxide: Biology and Pathobiology. Cambridge, MA: Academic, 2009. 431. Hilenski LL, Clempus RE, Quinn MT, Lambeth JD, Griendling KK. Distinct subcellu- lar localizations of Nox1 and Nox4 in vascular smooth muscle cells. Arterioscler 449. Ignarro LJ, Buga GM, Wood KS, Byrns RE, Chaudhuri G. Endothelium-derived re- Thromb Vasc Biol 24: 677–683, 2004. doi:10.1161/01.ATV.0000112024.13727.2c. laxing factor produced and released from artery and vein is nitric oxide. Proc Natl Acad Sci USA 84: 9265–9269, 1987. doi:10.1073/pnas.84.24.9265. 432. Hilgers RH, Kundumani-Sridharan V, Subramani J, Chen LC, Cuello LG, Rusch NJ, Das KC. Thioredoxin reverses age-related hypertension by chronically improving 450. Ihlemann N, Rask-Madsen C, Perner A, Dominguez H, Hermann T, Køber L, Torp- vascular redox and restoring eNOS function. Sci Transl Med 9: eaaf6094, 2017. Pedersen C. Tetrahydrobiopterin restores endothelial dysfunction induced by an doi:10.1126/scitranslmed.aaf6094. oral glucose challenge in healthy subjects. Am J Physiol Heart Circ Physiol 285: H875– H882, 2003. doi:10.1152/ajpheart.00008.2003. 433. Hill BG, Dranka BP, Bailey SM, Lancaster JR Jr, Darley-Usmar VM. What part of NO don’t you understand? Some answers to the cardinal questions in nitric oxide biol- 451. Imajoh-Ohmi S, Tokita K, Ochiai H, Nakamura M, Kanegasaki S. Topology of cyto- ogy. J Biol Chem 285: 19699–19704, 2010. doi:10.1074/jbc.R110.101618. chrome b558 in neutrophil membrane analyzed by anti-peptide antibodies and pro- teolysis. J Biol Chem 267: 180–184, 1992. 434. Hille R, Hall J, Basu P. The mononuclear molybdenum enzymes. Chem Rev 114: 3963–4038, 2014. doi:10.1021/cr400443z. 452. Ionova IA, Vásquez-Vivar J, Whitsett J, Herrnreiter A, Medhora M, Cooley BC, Pieper GM. Deficient BH4 production via de novo and salvage pathways regulates 435. Hirano K, Chen WS, Chueng AL, Dunne AA, Seredenina T, Filippova A, Ramachan- NO responses to cytokines in adult cardiac myocytes. Am J Physiol Heart Circ Physiol dran S, Bridges A, Chaudry L, Pettman G, Allan C, Duncan S, Lee KC, Lim J, Ma MT, 295: H2178–H2187, 2008. doi:10.1152/ajpheart.00748.2008. Ong AB, Ye NY, Nasir S, Mulyanidewi S, Aw CC, Oon PP, Liao S, Li D, Johns DG, Miller ND, Davies CH, Browne ER, Matsuoka Y, Chen DW, Jaquet V, Rutter AR. 453. Irie Y, Saeki M, Kamisaki Y, Martin E, Murad F. Histone H1.2 is a substrate for Discovery of GSK2795039, a Novel Small Molecule NADPH Oxidase 2 Inhibitor. denitrase, an activity that reduces nitrotyrosine immunoreactivity in proteins. Proc Antioxid Redox Signal 23: 358–374, 2015. doi:10.1089/ars.2014.6202. Natl Acad Sci USA 100: 5634–5639, 2003. doi:10.1073/pnas.1131756100.

436. Hoenicka M, Becker EM, Apeler H, Sirichoke T, Schröder H, Gerzer R, Stasch JP. 454. Isbell TS, Sun CW, Wu LC, Teng X, Vitturi DA, Branch BG, Kevil CG, Peng N, Wyss Purified soluble guanylyl cyclase expressed in a baculovirus/Sf9 system: stimulation JM, Ambalavanan N, Schwiebert L, Ren J, Pawlik KM, Renfrow MB, Patel RP, Townes TM. SNO-hemoglobin is not essential for red blood cell-dependent hypoxic vasodi- by YC-1, nitric oxide, and carbon monoxide. J Mol Med (Berl) 77: 14–23, 1999. lation. Nat Med 14: 773–777, 2008. doi:10.1038/nm1771. doi:10.1007/s001090050292. 455. Ismail S, Sturrock A, Wu P, Cahill B, Norman K, Huecksteadt T, Sanders K, Kennedy 437. Høivik HO, Laurijssens BE, Harnisch LO, Twomey CK, Dixon RM, Kirkham AJ, T, Hoidal J. NOX4 mediates hypoxia-induced proliferation of human pulmonary Williams PM, Wentz AL, Lunnon MW. Lack of efficacy of the selective iNOS inhibitor artery smooth muscle cells: the role of autocrine production of transforming growth GW274150 in prophylaxis of migraine headache. Cephalalgia 30: 1458–1467, 2010. factor-beta1 and insulin-like growth factor binding protein-3. Am J Physiol Lung Cell doi:10.1177/0333102410370875. Mol Physiol 296: L489–L499, 2009. doi:10.1152/ajplung.90488.2008. 438. Holland SM. Chronic granulomatous disease. Clin Rev Allergy Immunol 38: 3–10, 456. Isogai Y, Iizuka T, Shiro Y. The mechanism of electron donation to molecular oxygen 2010. doi:10.1007/s12016-009-8136-z. by phagocytic cytochrome b558. J Biol Chem 270: 7853–7857, 1995. doi:10.1074/ jbc.270.14.7853. 439. Holterman CE, Thibodeau JF, Towaij C, Gutsol A, Montezano AC, Parks RJ, Cooper ME, Touyz RM, Kennedy CR. Nephropathy and elevated BP in mice with podocyte- 457. Ito K, Hanazawa T, Tomita K, Barnes PJ, Adcock IM. Oxidative stress reduces specific NADPH oxidase 5 expression. J Am Soc Nephrol 25: 784–797, 2014. doi:10. histone deacetylase 2 activity and enhances IL-8 gene expression: role of tyrosine 1681/ASN.2013040371. nitration. Biochem Biophys Res Commun 315: 240–245, 2004. doi:10.1016/j.bbrc. 2004.01.046. 440. Hong HJ, Hsiao G, Cheng TH, Yen MH. Supplemention with tetrahydrobiopterin suppresses the development of hypertension in spontaneously hypertensive rats. 458. Iyamu EW, Perdew H, Woods GM. Cysteine-iron promotes arginase activity by Hypertension 38: 1044–1048, 2001. doi:10.1161/hy1101.095331. driving the Fenton reaction. Biochem Biophys Res Commun 376: 116–120, 2008. doi:10.1016/j.bbrc.2008.08.102. 441. Hosoya T, Ogawa Y, Hashimoto H, Ohashi T, Sakamoto R. Comparison of topir- oxostat and allopurinol in Japanese hyperuricemic patients with or without gout: a 459. Jabs A, Oelze M, Mikhed Y, Stamm P, Kröller-Schön S, Welschof P, Jansen T, Haus- phase 3, multicentre, randomized, double-blind, double-dummy, active-controlled, ding M, Kopp M, Steven S, Schulz E, Stasch JP, Münzel T, Daiber A. Effect of soluble parallel-group study. J Clin Pharm Ther 41: 290–297, 2016. doi:10.1111/jcpt.12391. guanylyl cyclase activator and stimulator therapy on nitroglycerin-induced nitrate

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 361 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

tolerance in rats. Vascul Pharmacol 71: 181–191, 2015. doi:10.1016/j.vph.2015.03. 477. Kagan VE, Tyurin VA, Jiang J, Tyurina YY, Ritov VB, Amoscato AA, Osipov AN, 007. Belikova NA, Kapralov AA, Kini V, Vlasova II, Zhao Q, Zou M, Di P, Svistunenko DA, Kurnikov IV, Borisenko GG. Cytochrome c acts as a cardiolipin oxygenase required 460. Jackson HM, Kawahara T, Nisimoto Y, Smith SME, Lambeth JD. Nox4 B-loop cre- for release of proapoptotic factors. Nat Chem Biol 1: 223–232, 2005. doi:10.1038/ ates an interface between the transmembrane and dehydrogenase domains. J Biol nchembio727. Chem 285: 10281–10290, 2010. doi:10.1074/jbc.M109.084939. 478. Kahles T, Brandes RP. Which NADPH oxidase isoform is relevant for ischemic 461. Jacobson GM, Dourron HM, Liu J, Carretero OA, Reddy DJ, Andrzejewski T, Pagano stroke? The case for nox 2. Antioxid Redox Signal 18: 1400–1417, 2013. doi:10.1089/ PJ. Novel NAD(P)H oxidase inhibitor suppresses angioplasty-induced superoxide ars.2012.4721. and neointimal hyperplasia of rat carotid artery. Circ Res 92: 637–643, 2003. doi:10. 1161/01.RES.0000063423.94645.8A. 479. Kahles T, Kohnen A, Heumueller S, Rappert A, Bechmann I, Liebner S, Wittko IM, Neumann-Haefelin T, Steinmetz H, Schroeder K, Brandes RP. NADPH oxidase 462. Jaffrey SR, Erdjument-Bromage H, Ferris CD, Tempst P, Snyder SH. Protein S- Nox1 contributes to ischemic injury in experimental stroke in mice. Neurobiol Dis 40: nitrosylation: a physiological signal for neuronal nitric oxide. Nat Cell Biol 3: 193–197, 185–192, 2010. doi:10.1016/j.nbd.2010.05.023. 2001. doi:10.1038/35055104. 480. Kakinuma K, Kaneda M, Chiba T, Ohnishi T. Electron spin resonance studies on a 463. Jagnandan D, Church JE, Banfi B, Stuehr DJ, Marrero MB, Fulton DJ. Novel mecha- flavoprotein in neutrophil plasma membranes. Redox potentials of the flavin and its nism of activation of NADPH oxidase 5. calcium sensitization via phosphorylation. J participation in NADPH oxidase. J Biol Chem 261: 9426–9432, 1986. Biol Chem 282: 6494–6507, 2007. doi:10.1074/jbc.M608966200. 481. Kamga Pride C, Mo L, Quesnelle K, Dagda RK, Murillo D, Geary L, Corey C, 464. Jaimes EA, Sweeney C, Raij L. Effects of the reactive oxygen species hydrogen Portella R, Zharikov S, St Croix C, Maniar S, Chu CT, Khoo NK, Shiva S. Nitrite peroxide and hypochlorite on endothelial nitric oxide production. Hypertension 38: activates protein kinase A in normoxia to mediate mitochondrial fusion and 877–883, 2001. tolerance to ischaemia/reperfusion. Cardiovasc Res 101: 57–68, 2014. doi:10. 465. Janiszewski M, Lopes LR, Carmo AO, Pedro MA, Brandes RP, Santos CXC, Laurindo 1093/cvr/cvt224. FRM. Regulation of NAD(P)H oxidase by associated protein disulfide isomerase in 482. Kanner J, Harel S, Granit R. Nitric oxide as an antioxidant. Arch Biochem Biophys 289: vascular smooth muscle cells. J Biol Chem 280: 40813–40819, 2005. doi:10.1074/jbc. 130–136, 1991. doi:10.1016/0003-9861(91)90452-O. M509255200. 483. Kanno S, Lee PC, Zhang Y, Ho C, Griffith BP, Shears LL II, Billiar TR. Attenuation of 466. Jankov RP, Kantores C, Pan J, Belik J. Contribution of xanthine oxidase-derived myocardial ischemia/reperfusion injury by superinduction of inducible nitric oxide superoxide to chronic hypoxic pulmonary hypertension in neonatal rats. Am J Physiol synthase. Circulation 101: 2742–2748, 2000. doi:10.1161/01.CIR.101.23.2742. Lung Cell Mol Physiol 294: L233–L245, 2008. doi:10.1152/ajplung.00166.2007. 484. Kanski J, Behring A, Pelling J, Schöneich C. Proteomic identification of 3-nitroty- 467. Jansson EA, Huang L, Malkey R, Govoni M, Nihlén C, Olsson A, Stensdotter M, rosine-containing rat cardiac proteins: effects of biological aging. Am J Physiol Heart Petersson J, Holm L, Weitzberg E, Lundberg JO. A mammalian functional nitrate Circ Physiol 288: H371–H381, 2005. doi:10.1152/ajpheart.01030.2003. reductase that regulates nitrite and nitric oxide homeostasis. Nat Chem Biol 4: 411– 417, 2008. doi:10.1038/nchembio.92. 485. Kapil V, Haydar SMA, Pearl V, Lundberg JO, Weitzberg E, Ahluwalia A. Physiological role for nitrate-reducing oral bacteria in blood pressure control. Free Radic Biol Med 468. Javeshghani D, Hussain SN, Scheidel J, Quinn MT, Magder SA. Superoxide produc- 55: 93–100, 2013. doi:10.1016/j.freeradbiomed.2012.11.013. tion in the vasculature of lipopolysaccharide-treated rats and pigs. Shock 19: 486– 493, 2003. doi:10.1097/01.shk.0000054374.88889.37. 486. Kapil V, Khambata RS, Robertson A, Caulfield MJ, Ahluwalia A. Dietary nitrate pro- 469. Jay DB, Papaharalambus CA, Seidel-Rogol B, Dikalova AE, Lassègue B, Griendling vides sustained blood pressure lowering in hypertensive patients: a randomized, KK. Nox5 mediates PDGF-induced proliferation in human aortic smooth muscle phase 2, double-blind, placebo-controlled study. Hypertension 65: 320–327, 2015. cells. Free Radic Biol Med 45: 329–335, 2008. doi:10.1016/j.freeradbiomed.2008.04. doi:10.1161/HYPERTENSIONAHA.114.04675. 024. 487. Kaplan-Albuquerque N, Bogaert YE, Van Putten V, Weiser-Evans MC, Nemenoff 470. Jerlich A, Horakova L, Fabjan JS, Giessauf A, Jürgens G, Schaur RJ. Correlation of RA. Patterns of gene expression differentially regulated by platelet-derived growth low-density lipoprotein modification by myeloperoxidase with hypochlorous acid factor and hypertrophic stimuli in vascular smooth muscle cells: markers for pheno- formation. Int J Clin Lab Res 29: 155–161, 1999. doi:10.1007/s005990050083. typic modulation and response to injury. J Biol Chem 280: 19966–19976, 2005. doi:10.1074/jbc.M500917200. 471. Jia L, Bonaventura C, Bonaventura J, Stamler JS. S-nitrosohaemoglobin: a dynamic activity of blood involved in vascular control. Nature 380: 221–226, 1996. doi:10. 488. Karbach S, Wenzel P, Waisman A, Munzel T, Daiber A. eNOS uncoupling in cardio- 1038/380221a0. vascular diseases–the role of oxidative stress and inflammation. Curr Pharm Des 20: 3579–3594, 2014. doi:10.2174/13816128113196660748. 472. Jiang JX, Chen X, Serizawa N, Szyndralewiez C, Page P, Schröder K, Brandes RP, Devaraj S, Török NJ. Liver fibrosis and hepatocyte apoptosis are attenuated by 489. Karow DS, Pan D, Davis JH, Behrends S, Mathies RA, Marletta MA. Characterization GKT137831, a novel NOX4/NOX1 inhibitor in vivo. Free Radic Biol Med 53: 289– of functional heme domains from soluble guanylate cyclase. Biochemistry 44: 16266– 296, 2012. doi:10.1016/j.freeradbiomed.2012.05.007. 16274, 2005. doi:10.1021/bi051601b.

473. Jones SA, O’Donnell VB, Wood JD, Broughton JP, Hughes EJ, Jones OTG. Expression 490. Karplus PA, Daniels MJ, Herriott JR. Atomic structure of ferredoxin-NADPϩ reduc- of phagocyte NADPH oxidase components in human endothelial cells. Am J Physiol tase: prototype for a structurally novel flavoenzyme family. Science 251: 60–66, 271: H1626–H1634, 1996. 1991. doi:10.1126/science.1986412.

474. Josephson RA, Silverman HS, Lakatta EG, Stern MD, Zweier JL. Study of the mech- 491. Katsuki S, Arnold W, Mittal C, Murad F. Stimulation of guanylate cyclase by sodium anisms of hydrogen peroxide and hydroxyl free radical-induced cellular injury and nitroprusside, nitroglycerin and nitric oxide in various tissue preparations and com- calcium overload in cardiac myocytes. J Biol Chem 266: 2354–2361, 1991. parison to the effects of sodium azide and hydroxylamine. J Cyclic Nucleotide Res 3: 23–35, 1977. 475. Judkins CP, Diep H, Broughton BRS, Mast AE, Hooker EU, Miller AA, Selemidis S, Dusting GJ, Sobey CG, Drummond GR. Direct evidence of a role for Nox2 in 492. Katsuyama M, Fan C, Yabe-Nishimura C. NADPH oxidase is involved in prostaglan- superoxide production, reduced nitric oxide bioavailability, and early atherosclerotic din F2alpha-induced hypertrophy of vascular smooth muscle cells: induction of plaque formation in ApoEϪ/Ϫ mice. Am J Physiol Heart Circ Physiol 298: H24–H32, NOX1 by PGF2alpha. J Biol Chem 277: 13438–13442, 2002. doi:10.1074/jbc. 2010. doi:10.1152/ajpheart.00799.2009. M111634200.

476. Jung O, Schreiber JG, Geiger H, Pedrazzini T, Busse R, Brandes RP. gp91phox- 493. Kawada N, Kristensen DB, Asahina K, Nakatani K, Minamiyama Y, Seki S, Yoshizato containing NADPH oxidase mediates endothelial dysfunction in renovascular K. Characterization of a stellate cell activation-associated protein (STAP) with per- hypertension. Circulation 109: 1795–1801, 2004. doi:10.1161/01.CIR. oxidase activity found in rat hepatic stellate cells. J Biol Chem 276: 25318–25323, 0000124223.00113.A4. 2001. doi:10.1074/jbc.M102630200.

362 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

494. Kawahara T, Lambeth JD. Phosphatidylinositol (4,5)-bisphosphate modulates Nox5 512. Kitamura S, Sugihara K. Current status of prediction of drug disposition and toxicity localization via an N-terminal polybasic region. Mol Biol Cell 19: 4020–4031, 2008. in humans using chimeric mice with humanized liver. Xenobiotica 44: 123–134, 2014. doi:10.1091/mbc.e07-12-1223. doi:10.3109/00498254.2013.868062.

495. Kelley EE, Baust J, Bonacci G, Golin-Bisello F, Devlin JE, St Croix CM, Watkins SC, 513. Kitamura S, Sugihara K, Ohta S. Drug-metabolizing ability of molybdenum hydroxy- Gor S, Cantu-Medellin N, Weidert ER, Frisbee JC, Gladwin MT, Champion HC, lases. Drug Metab Pharmacokinet 21: 83–98, 2006. doi:10.2133/dmpk.21.83. Freeman BA, Khoo NK. Fatty acid nitroalkenes ameliorate glucose intolerance and pulmonary hypertension in high-fat diet-induced obesity. Cardiovasc Res 101: 352– 514. Klatt P, M, Leopold E, Schmidt K, Werner ER, Mayer B. The pteridine binding 363, 2014. doi:10.1093/cvr/cvt341. site of brain nitric oxide synthase. Tetrahydrobiopterin binding kinetics, specificity, and allosteric interaction with the substrate domain. J Biol Chem 269: 13861–13866, 496. Kelley EE, Hock T, Khoo NK, Richardson GR, Johnson KK, Powell PC, Giles GI, 1994. Agarwal A, Lancaster JR Jr, Tarpey MM. Moderate hypoxia induces xanthine oxi- doreductase activity in arterial endothelial cells. Free Radic Biol Med 40: 952–959, 515. Klatt P, Schmidt K, Lehner D, Glatter O, Bächinger HP, Mayer B. Structural analysis 2006. doi:10.1016/j.freeradbiomed.2005.11.008. of porcine brain nitric oxide synthase reveals a role for tetrahydrobiopterin and L-arginine in the formation of an SDS-resistant dimer. EMBO J 14: 3687–3695, 1995. 497. Kelley EE, Khoo NK, Hundley NJ, Malik UZ, Freeman BA, Tarpey MM. Hydrogen peroxide is the major oxidant product of xanthine oxidase. Free Radic Biol Med 48: 516. Klebanoff SJ. Myeloperoxidase: friend and foe. J Leukoc Biol 77: 598–625, 2005. 493–498, 2010. doi:10.1016/j.freeradbiomed.2009.11.012. doi:10.1189/jlb.1204697.

498. Kellogg DL Jr, Zhao JL, Wu Y. Neuronal nitric oxide synthase control mechanisms in 517. Kleikers PW, Dao VT, Göb E, Hooijmans C, Debets J, van Essen H, Kleinschnitz C, the cutaneous vasculature of humans in vivo. J Physiol 586: 847–857, 2008. doi:10. Schmidt HH. SFRR-E Young Investigator AwardeeNOXing out stroke: Identification 1113/jphysiol.2007.144642. of NOX4 and 5as targets in blood-brain-barrier stabilisation and neuroprotection. Free Radic Biol Med 75, Suppl 1: S16, 2014. doi:10.1016/j.freeradbiomed.2014.10. 499. Kellogg EW III, Fridovich I. Superoxide, hydrogen peroxide, and singlet oxygen in 593. lipid peroxidation by a xanthine oxidase system. J Biol Chem 250: 8812–8817, 1975. 518. Kleinbongard P, Schulz R, Rassaf T, Lauer T, Dejam A, Jax T, Kumara I, Gharini P, 500. Kelso GF, Porteous CM, Coulter CV, Hughes G, Porteous WK, Ledgerwood EC, Kabanova S, Ozüyaman B, Schnürch HG, Gödecke A, Weber AA, Robenek M, Smith RA, Murphy MP. Selective targeting of a redox-active ubiquinone to mitochon- Robenek H, Bloch W, Rösen P, Kelm M. Red blood cells express a functional endo- dria within cells: antioxidant and antiapoptotic properties. J Biol Chem 276: 4588– thelial nitric oxide synthase. Blood 107: 2943–2951, 2006. doi:10.1182/blood-2005- 4596, 2001. doi:10.1074/jbc.M009093200. 10-3992.

501. Keszler A, Piknova B, Schechter AN, Hogg N. The reaction between nitrite and 519. Kleinert H, Schwarz PM, Förstermann U. Regulation of the expression of inducible oxyhemoglobin: a mechanistic study. J Biol Chem 283: 9615–9622, 2008. doi:10. nitric oxide synthase. Biol Chem 384: 1343–1364, 2003. doi:10.1515/BC.2003.152. 1074/jbc.M705630200. 520. Kleinschnitz C, Grund H, Wingler K, Armitage ME, Jones E, Mittal M, Barit D, 502. Khambata RS, Ghosh SM, Rathod KS, Thevathasan T, Filomena F, Xiao Q, Ahluwalia Schwarz T, Geis C, Kraft P, Barthel K, Schuhmann MK, Herrmann AM, Meuth SG, A. Antiinflammatory actions of inorganic nitrate stabilize the atherosclerotic plaque. Stoll G, Meurer S, Schrewe A, Becker L, Gailus-Durner V, Fuchs H, Klopstock T, de Proc Natl Acad Sci USA 114: E550–E559, 2017. doi:10.1073/pnas.1613063114. Angelis MH, Jandeleit-Dahm K, Shah AM, Weissmann N, Schmidt HHHW. Post- stroke inhibition of induced NADPH oxidase type 4 prevents oxidative stress and 503. Khan AA, Wang Y, Sun Y, Mao XO, Xie L, Miles E, Graboski J, Chen S, Ellerby LM, neurodegeneration. PLoS Biol 8: e1000479, 2010. doi:10.1371/journal.pbio. Jin K, Greenberg DA. Neuroglobin-overexpressing transgenic mice are resistant to 1000479. cerebral and myocardial ischemia. Proc Natl Acad Sci USA 103: 17944–17948, 2006. doi:10.1073/pnas.0607497103. 521. Kleschyov AL, Muller B, Schott C, Stoclet JC. Role of adventitial nitric oxide in vascular hyporeactivity induced by lipopolysaccharide in rat aorta. Br J Pharmacol 504. Khan MA, Alam K, Zafaryab M, Rizvi MMA. Peroxynitrite-modified histone as a 124: 623–626, 1998. doi:10.1038/sj.bjp.0701916. pathophysiological biomarker in autoimmune diseases. Biochimie 140: 1–9, 2017. doi:10.1016/j.biochi.2017.06.006. 522. Knorr M, Hausding M, Kröller-Schuhmacher S, Steven S, Oelze M, Heeren T, Scholz A, Gori T, Wenzel P, Schulz E, Daiber A, Münzel T. Nitroglycerin-induced endothe- 505. Kietzmann T, Petry A, Shvetsova A, Gerhold JM, Görlach A. The epigenetic land- lial dysfunction and tolerance involve adverse phosphorylation and S-Glutathionyla- scape related to reactive oxygen species formation in the cardiovascular system. Br tion of endothelial nitric oxide synthase: beneficial effects of therapy with the AT1 J Pharmacol 174: 1533–1554, 2017. doi:10.1111/bph.13792. receptor blocker telmisartan. Arterioscler Thromb Vasc Biol 31: 2223–2231, 2011. doi:10.1161/ATVBAHA.111.232058. 506. Kim IY, Schutzler SE, Schrader A, Spencer HJ, Azhar G, Deutz NE, Wolfe RR. Acute ingestion of citrulline stimulates nitric oxide synthesis but does not increase blood 524. Ko FN, Wu CC, Kuo SC, Lee FY, Teng CM. YC-1, a novel activator of platelet flow in healthy young and older adults with heart failure. Am J Physiol Endocrinol guanylate cyclase. Blood 84: 4226–4233, 1994. Metab 309: E915–E924, 2015. doi:10.1152/ajpendo.00339.2015. 525. Kobzik L, Bredt DS, Lowenstein CJ, Drazen J, Gaston B, Sugarbaker D, Stamler JS. 507. Kimura S, Zhang GX, Nishiyama A, Shokoji T, Yao L, Fan YY, Rahman M, Suzuki T, Nitric oxide synthase in human and rat lung: immunocytochemical and histochemical Maeta H, Abe Y. Role of NAD(P)H oxidase- and mitochondria-derived reactive localization. Am J Respir Cell Mol Biol 9: 371–377, 1993. doi:10.1165/ajrcmb/9.4.371. oxygen species in cardioprotection of ischemic reperfusion injury by angiotensin II. Hypertension 45: 860–866, 2005. doi:10.1161/01.HYP.0000163462.98381.7f. 526. Koppenol WH. Peroxynitrite: the basics. In: Peroxynitrite Detection in Biological Me- dia: Challenges and Advances, edited by Peteu SFS, Bayachou SM. London: Royal 508. Kirk EA, Dinauer MC, Rosen H, Chait A, Heinecke JW, LeBoeuf RC. Impaired Society of Chemistry, 2015, p. 1–11. doi:10.1039/9781782622352-00001. superoxide production due to a deficiency in phagocyte NADPH oxidase fails to inhibit atherosclerosis in mice. Arterioscler Thromb Vasc Biol 20: 1529–1535, 2000. 527. Korda M, Kubant R, Patton S, Malinski T. Leptin-induced endothelial dysfunction in doi:10.1161/01.ATV.20.6.1529. obesity. Am J Physiol Heart Circ Physiol 295: H1514–H1521, 2008. doi:10.1152/ ajpheart.00479.2008. 509. Kirsch M, Korth HG, Sustmann R, de Groot H. The pathobiochemistry of nitrogen dioxide. Biol Chem 383: 389–399, 2002. doi:10.1515/BC.2002.043. 528. Kotthaus J, Wahl B, Havemeyer A, Kotthaus J, Schade D, Garbe-Schönberg D, Mendel R, Bittner F, Clement B. Reduction of N(␻)-hydroxy-L-arginine by the mi- 510. Kisker C, Schindelin H, Pacheco A, Wehbi WA, Garrett RM, Rajagopalan KV, En- tochondrial amidoxime reducing component (mARC). Biochem J 433: 383–391, emark JH, Rees DC. Molecular basis of sulfite oxidase deficiency from the structure 2011. doi:10.1042/BJ20100960. of sulfite oxidase. Cell 91: 973–983, 1997. doi:10.1016/S0092-8674(00)80488-2. 529. Kovamees O, Shemyakin A, Eriksson M, Angelin B, Pernow J. Arginase inhibition 511. Kiss PJ, Knisz J, Zhang Y, Baltrusaitis J, Sigmund CD, Thalmann R, Smith RJH, Verpy improves endothelial function in patients with familial hypercholesterolaemia irre- E, Bánfi B. Inactivation of NADPH oxidase organizer 1 results in severe imbalance. spective of their cholesterol levels. J Intern Med 279: 477–484, 2016. doi:10.1111/ Curr Biol 16: 208–213, 2006. doi:10.1016/j.cub.2005.12.025. joim.12461.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 363 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

530. Kövamees O, Shemyakin A, Pernow J. Effect of arginase inhibition on ischemia- Physiol Heart Circ Physiol 279: H1906–H1912, 2000. doi:10.1152/ajpheart.2000.279. reperfusion injury in patients with coronary artery disease with and without diabetes 4.H1906. mellitus. PLoS One 9: e103260, 2014. doi:10.1371/journal.pone.0103260. 548. Lancaster JR Jr. Nitroxidative, nitrosative, and nitrative stress: kinetic predictions of 531. Kraus JP, Janosík M, Kozich V, Mandell R, Shih V, Sperandeo MP, Sebastio G, de reactive nitrogen species chemistry under biological conditions. Chem Res Toxicol 19: Franchis R, Andria G, Kluijtmans LA, Blom H, Boers GH, Gordon RB, Kamoun P, Tsai 1160–1174, 2006. doi:10.1021/tx060061w. MY, Kruger WD, Koch HG, Ohura T, Gaustadnes M. Cystathionine beta-synthase mutations in homocystinuria. Hum Mutat 13: 362–375, 1999. doi:10.1002/ 549. Landmesser U, Cai H, Dikalov S, McCann L, Hwang J, Jo H, Holland SM, Harrison (SICI)1098-1004(1999)13:5Ͻ362::AID-HUMU4Ͼ3.0.CO;2-K. DG. Role of p47(phox) in vascular oxidative stress and hypertension caused by angiotensin II. Hypertension 40: 511–515, 2002. doi:10.1161/01.HYP.0000032100. 532. Kuhlencordt PJ, Chen J, Han F, Astern J, Huang PL. Genetic deficiency of inducible 23772.98. nitric oxide synthase reduces atherosclerosis and lowers plasma lipid peroxides in apolipoprotein E-knockout mice. Circulation 103: 3099–3104, 2001. doi:10.1161/ 550. Landmesser U, Dikalov S, Price SR, McCann L, Fukai T, Holland SM, Mitch WE, 01.CIR.103.25.3099. Harrison DG. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J Clin Invest 111: 1201–1209, 2003. doi: 533. Kuhlencordt PJ, Gyurko R, Han F, Scherrer-Crosbie M, Aretz TH, Hajjar R, Picard 10.1172/JCI200314172. MH, Huang PL. Accelerated atherosclerosis, aortic aneurysm formation, and isch- emic heart disease in apolipoprotein E/endothelial nitric oxide synthase double- 551. Landmesser U, Spiekermann S, Preuss C, Sorrentino S, Fischer D, Manes C, Mueller knockout mice. Circulation 104: 448–454, 2001. doi:10.1161/hc2901.091399. M, Drexler H. Angiotensin II induces endothelial xanthine oxidase activation: role for endothelial dysfunction in patients with coronary disease. Arterioscler Thromb Vasc 534. Kuhlencordt PJ, Hötten S, Schödel J, Rützel S, Hu K, Widder J, Marx A, Huang PL, Ertl Biol 27: 943–948, 2007. doi:10.1161/01.ATV.0000258415.32883.bf. G. Atheroprotective effects of neuronal nitric oxide synthase in apolipoprotein e knockout mice. Arterioscler Thromb Vasc Biol 26: 1539–1544, 2006. doi:10.1161/01. 552. Langbein H, Brunssen C, Hofmann A, Cimalla P, Brux M, Bornstein SR, Deussen A, ATV.0000223143.88128.19. Koch E, Morawietz H. NADPH oxidase 4 protects against development of endothe- lial dysfunction and atherosclerosis in LDL receptor deficient mice. Eur Heart J 37: 535. Kuhn H, Banthiya S, van Leyen K. Mammalian lipoxygenases and their biological 1753–1761, 2016. doi:10.1093/eurheartj/ehv564. relevance. Biochim Biophys Acta 1851: 308–330, 2015. doi:10.1016/j.bbalip.2014.10. 002. 553. Lapp H, Mitrovic V, Franz N, Heuer H, Buerke M, Wolfertz J, Mueck W, Unger S, Wensing G, Frey R. Cinaciguat (BAY 58-2667) improves cardiopulmonary hemody- 536. Kuhn V, Diederich L, Keller TCS IV, Kramer CM, Lückstädt W, Panknin C, Suvorava namics in patients with acute decompensated heart failure. Circulation 119: 2781– T, Isakson BE, Kelm M, Cortese-Krott MM. Red Blood Cell Function and Dysfunc- 2788, 2009. doi:10.1161/CIRCULATIONAHA.108.800292. tion: Redox Regulation, Nitric Oxide Metabolism, Anemia. Antioxid Redox Signal 26: 718–742, 2017. doi:10.1089/ars.2016.6954. 554. Larsen FJ, Ekblom B, Sahlin K, Lundberg JO, Weitzberg E. Effects of dietary nitrate on blood pressure in healthy volunteers. N Engl J Med 355: 2792–2793, 2006. doi:10. 537. Kukreja RC, Kontos HA, Hess ML, Ellis EF. PGH synthase and lipoxygenase generate 1056/NEJMc062800. superoxide in the presence of NADH or NADPH. Circ Res 59: 612–619, 1986. doi:10.1161/01.RES.59.6.612. 555. Lassègue B, San Martín A, Griendling KK. Biochemistry, physiology, and pathophys- iology of NADPH oxidases in the cardiovascular system. Circ Res 110: 1364–1390, 538. Kundu TK, Hille R, Velayutham M, Zweier JL. Characterization of superoxide pro- 2012. doi:10.1161/CIRCRESAHA.111.243972. duction from aldehyde oxidase: an important source of oxidants in biological tissues. Arch Biochem Biophys 460: 113–121, 2007. doi:10.1016/j.abb.2006.12.032. 556. Lassègue B, Sorescu D, Szöcs K, Yin Q, Akers M, Zhang Y, Grant SL, Lambeth JD, Griendling KK. Novel gp91(phox) homologues in vascular smooth muscle cells: nox1 539. Kundu TK, Velayutham M, Zweier JL. Aldehyde oxidase functions as a superoxide mediates angiotensin II-induced superoxide formation and redox-sensitive signaling generating NADH oxidase: an important redox regulated pathway of cellular oxygen pathways. Circ Res 88: 888–894, 2001. doi:10.1161/hh0901.090299. radical formation. Biochemistry 51: 2930–2939, 2012. doi:10.1021/bi3000879. 557. Lauer T, Preik M, Rassaf T, Strauer BE, Deussen A, Feelisch M, Kelm M. Plasma 540. Kuroda J, Ago T, Matsushima S, Zhai P, Schneider MD, Sadoshima J. NADPH oxidase nitrite rather than nitrate reflects regional endothelial nitric oxide synthase activity 4 (Nox4) is a major source of oxidative stress in the failing heart. Proc Natl Acad Sci but lacks intrinsic vasodilator action. Proc Natl Acad Sci USA 98: 12814–12819, 2001. USA 107: 15565–15570, 2010. doi:10.1073/pnas.1002178107. doi:10.1073/pnas.221381098. 541. Kuroda J, Nakagawa K, Yamasaki T, Nakamura K, Takeya R, Kuribayashi F, Imajoh- 558. Laurent C, Chabi B, Fouret G, Py G, Sairafi B, Elong C, Gaillet S, Cristol JP, Coudray Ohmi S, Igarashi K, Shibata Y, Sueishi K, Sumimoto H. The superoxide-producing C, Feillet-Coudray C. Polyphenols decreased liver NADPH oxidase activity, in- NAD(P)H oxidase Nox4 in the nucleus of human vascular endothelial cells. Genes creased muscle mitochondrial biogenesis and decreased gastrocnemius age-depen- Cells 10: 1139–1151, 2005. doi:10.1111/j.1365-2443.2005.00907.x. dent autophagy in aged rats. Free Radic Res 46: 1140–1149, 2012. doi:10.3109/ 542. Kuwabara Y, Nishino T, Okamoto K, Matsumura T, Eger BT, Pai EF, Nishino T. 10715762.2012.694428. Unique amino acids cluster for switching from the dehydrogenase to oxidase form of 559. Laurindo FRM, Fernandes DC, Amanso AM, Lopes LR, Santos CXC. Novel role of xanthine oxidoreductase. Proc Natl Acad Sci USA 100: 8170–8175, 2003. doi:10. protein disulfide isomerase in the regulation of NADPH oxidase activity: pathophys- 1073/pnas.1431485100. iological implications in vascular diseases. Antioxid Redox Signal 10: 1101–1113, 2008. 543. Kuzkaya N, Weissmann N, Harrison DG, Dikalov S. Interactions of peroxynitrite, doi:10.1089/ars.2007.2011. tetrahydrobiopterin, ascorbic acid, and thiols: implications for uncoupling endothe- 560. Laursen JB, Somers M, Kurz S, McCann L, Warnholtz A, Freeman BA, Tarpey M, lial nitric-oxide synthase. J Biol Chem 278: 22546–22554, 2003. doi:10.1074/jbc. M302227200. Fukai T, Harrison DG. Endothelial regulation of vasomotion in apoE-deficient mice: implications for interactions between peroxynitrite and tetrahydrobiopterin. Circu- 544. Kwon NS, Nathan CF, Stuehr DJ. Reduced biopterin as a cofactor in the generation lation 103: 1282–1288, 2001. doi:10.1161/01.CIR.103.9.1282. of nitrogen oxides by murine macrophages. J Biol Chem 264: 20496–20501, 1989. 561. Lee BE, Toledo AH, Anaya-Prado R, Roach RR, Toledo-Pereyra LH. Allopurinol, 545. Laleu B, Gaggini F, Orchard M, Fioraso-Cartier L, Cagnon L, Houngninou-Molango xanthine oxidase, and cardiac ischemia. J Investig Med 57: 902–909, 2009. doi:10. S, Gradia A, Duboux G, Merlot C, Heitz F, Szyndralewiez C, Page P. First in class, 2310/JIM.0b013e3181bca50c. potent, and orally bioavailable NADPH oxidase isoform 4 (Nox4) inhibitors for the treatment of idiopathic pulmonary fibrosis. J Med Chem 53: 7715–7730, 2010. doi: 562. Lee MC, Velayutham M, Komatsu T, Hille R, Zweier JL. Measurement and charac- 10.1021/jm100773e. terization of superoxide generation from xanthine dehydrogenase: a redox-regu- lated pathway of radical generation in ischemic tissues. Biochemistry 53: 6615–6623, 546. Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol 4: 2014. doi:10.1021/bi500582r. 181–189, 2004. doi:10.1038/nri1312. 563. Lee PC, Salyapongse AN, Bragdon GA, Shears LL II, Watkins SC, Edington HDJ, 547. Lamping KG, Nuno DW, Shesely EG, Maeda N, Faraci FM. Vasodilator mechanisms Billiar TR. Impaired wound healing and angiogenesis in eNOS-deficient mice. Am J in the coronary circulation of endothelial nitric oxide synthase-deficient mice. Am J Physiol 277: H1600–H1608, 1999.

364 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

564. Lee SB, Bae IH, Bae YS, Um HD. Link between mitochondria and NADPH oxidase and ERK1/2 signaling pathways in prostate cancer. Biochim Biophys Acta 1833: 3375– 1 isozyme for the sustained production of reactive oxygen species and cell death. J 3385, 2013. doi:10.1016/j.bbamcr.2013.09.018. Biol Chem 281: 36228–36235, 2006. doi:10.1074/jbc.M606702200. 583. Li Y, Zhang J, Schopfer FJ, Martynowski D, Garcia-Barrio MT, Kovach A, Suino- 565. Lei J, Vodovotz Y, Tzeng E, Billiar TR. Nitric oxide, a protective molecule in the Powell K, Baker PRS, Freeman BA, Chen YE, Xu HE. Molecular recognition of cardiovascular system. Nitric Oxide 35: 175–185, 2013. doi:10.1016/j.niox.2013.09. nitrated fatty acids by PPAR gamma. Nat Struct Mol Biol 15: 865–867, 2008. doi:10. 004. 1038/nsmb.1447.

566. Leiper J, Vallance P. Biological significance of endogenous methylarginines that inhibit 584. Li Y, Huang TT, Carlson EJ, Melov S, Ursell PC, Olson JL, Noble LJ, Yoshimura MP, nitric oxide synthases. Cardiovasc Res 43: 542–548, 1999. doi:10.1016/S0008- Berger C, Chan PH, Wallace DC, Epstein CJ. Dilated cardiomyopathy and neonatal 6363(99)00162-5. lethality in mutant mice lacking manganese superoxide dismutase. Nat Genet 11: 376–381, 1995. doi:10.1038/ng1295-376. 567. Lenaz G. The mitochondrial production of reactive oxygen species: mechanisms and implications in human pathology. IUBMB Life 52: 159–164, 2001. doi:10.1080/ 585. Lim CH, Dedon PC, Deen WM. Kinetic analysis of intracellular concentrations of 15216540152845957. reactive nitrogen species. Chem Res Toxicol 21: 2134–2147, 2008. doi:10.1021/ tx800213b. 568. Li H, Cui H, Kundu TK, Alzawahra W, Zweier JL. Nitric oxide production from nitrite occurs primarily in tissues not in the blood: critical role of xanthine oxidase 586. Lima B, Forrester MT, Hess DT, Stamler JS. S-nitrosylation in cardiovascular signal- and aldehyde oxidase. J Biol Chem 283: 17855–17863, 2008. doi:10.1074/jbc. ing. Circ Res 106: 633–646, 2010. doi:10.1161/CIRCRESAHA.109.207381. M801785200. 587. Lin HL, Kenaan C, Zhang H, Hollenberg PF. Reaction of human cytochrome P450 569. Li H, Forstermann U. Pharmacological prevention of eNOS uncoupling. Curr Pharm 3A4 with peroxynitrite: nitrotyrosine formation on the proximal side impairs its Des 20: 3595–3606, 2014. doi:10.2174/13816128113196660749. interaction with NADPH-cytochrome P450 reductase. Chem Res Toxicol 25: 2642– 570. Li H, Förstermann U. Uncoupling of endothelial NO synthase in atherosclerosis and 2653, 2012. doi:10.1021/tx3002753. vascular disease. Curr Opin Pharmacol 13: 161–167, 2013. doi:10.1016/j.coph.2013. 588. Lin MI, Fulton D, Babbitt R, Fleming I, Busse R, Pritchard KA Jr, Sessa WC. Phos- 01.006. phorylation of threonine 497 in endothelial nitric-oxide synthase coordinates the 571. Li H, Hemann C, Abdelghany TM, El-Mahdy MA, Zweier JL. Characterization of the coupling of L-arginine metabolism to efficient nitric oxide production. J Biol Chem mechanism and magnitude of cytoglobin-mediated nitrite reduction and nitric oxide 278: 44719–44726, 2003. doi:10.1074/jbc.M302836200. generation under anaerobic conditions. J Biol Chem 287: 36623–36633, 2012. doi: 589. Lind L, Larsson A, Teerlink T. L-Arginine is related to endothelium-dependent va- 10.1074/jbc.M112.342378. sodilation in resistance and conduit arteries in divergent ways-The Prospective In- 572. Li H, Horke S, Förstermann U. Oxidative stress in vascular disease and its pharma- vestigation of the Vasculature in Uppsala Seniors (PIVUS) study. Atherosclerosis 203: cological prevention. Trends Pharmacol Sci 34: 313–319, 2013. doi:10.1016/j.tips. 544–549, 2009. doi:10.1016/j.atherosclerosis.2008.07.016. 2013.03.007. 590. List BM, Klösch B, Völker C, Gorren AC, Sessa WC, Werner ER, Kukovetz WR, 573. Li H, Horke S, Förstermann U. Vascular oxidative stress, nitric oxide and athero- Schmidt K, Mayer B. Characterization of bovine endothelial nitric oxide synthase as sclerosis. Atherosclerosis 237: 208–219, 2014. doi:10.1016/j.atherosclerosis.2014. a homodimer with down-regulated uncoupled NADPH oxidase activity: tetrahydro- 09.001. biopterin binding kinetics and role of haem in dimerization. Biochem J 323: 159–165, 1997. doi:10.1042/bj3230159. 574. Li H, Jamal J, Plaza C, Pineda SH, Chreifi G, Jing Q, Cinelli MA, Silverman RB, Poulos TL. Structures of human constitutive nitric oxide synthases. Acta Crystallogr D Biol 591. Liu C, Wajih N, Liu X, Basu S, Janes J, Marvel M, Keggi C, Helms CC, Lee AN, Crystallogr 70: 2667–2674, 2014. doi:10.1107/S1399004714017064. Belanger AM, Diz DI, Laurienti PJ, Caudell DL, Wang J, Gladwin MT, Kim-Shapiro DB. Mechanisms of human erythrocytic bioactivation of nitrite. J Biol Chem 290: 575. Li H, Kundu TK, Zweier JL. Characterization of the magnitude and mechanism of 1281–1294, 2015. doi:10.1074/jbc.M114.609222. aldehyde oxidase-mediated nitric oxide production from nitrite. J Biol Chem 284: 33850–33858, 2009. doi:10.1074/jbc.M109.019125. 592. Liu L, Hausladen A, Zeng M, Que L, Heitman J, Stamler JS. A metabolic enzyme for S-nitrosothiol conserved from bacteria to humans. Nature 410: 490–494, 2001. 576. Li H, Liu X, Cui H, Chen YR, Cardounel AJ, Zweier JL. Characterization of the doi:10.1038/35068596. mechanism of cytochrome P450 reductase-cytochrome P450-mediated nitric oxide and nitrosothiol generation from organic nitrates. J Biol Chem 281: 12546–12554, 593. Liu X, El-Mahdy MA, Boslett J, Varadharaj S, Hemann C, Abdelghany TM, Ismail RS, 2006. doi:10.1074/jbc.M511803200. Little SC, Zhou D, Thuy LT, Kawada N, Zweier JL. Cytoglobin regulates blood pressure and vascular tone through nitric oxide metabolism in the vascular wall. Nat 577. Li H, Samouilov A, Liu X, Zweier JL. Characterization of the effects of oxygen on Commun 8: 14807, 2017. doi:10.1038/ncomms14807. xanthine oxidase-mediated nitric oxide formation. J Biol Chem 279: 16939–16946, 2004. doi:10.1074/jbc.M314336200. 594. Liu X, Follmer D, Zweier JR, Huang X, Hemann C, Liu K, Druhan LJ, Zweier JL. Characterization of the function of cytoglobin as an oxygen-dependent regulator of 578. Li H, Samouilov A, Liu X, Zweier JL. Characterization of the magnitude and kinetics nitric oxide concentration. Biochemistry 51: 5072–5082, 2012. doi:10.1021/ of xanthine oxidase-catalyzed nitrite reduction. Evaluation of its role in nitric oxide bi300291h. generation in anoxic tissues. J Biol Chem 276: 24482–24489, 2001. doi:10.1074/jbc. M011648200. 595. Liu X, Tong J, Zweier JR, Follmer D, Hemann C, Ismail RS, Zweier JL. Differences in 579. Li J, Billiar TR. Nitric Oxide. IV. Determinants of nitric oxide protection and toxicity oxygen-dependent nitric oxide metabolism by cytoglobin and myoglobin account for in liver. Am J Physiol 276: G1069–G1073, 1999. their differing functional roles. FEBS J 280: 3621–3631, 2013. doi:10.1111/febs. 12352. 580. Li JM, Shah AM. Intracellular localization and preassembly of the NADPH oxidase complex in cultured endothelial cells. J Biol Chem 277: 19952–19960, 2002. doi:10. 596. Liu XH, Zhang QY, Pan LL, Liu SY, Xu P, Luo XL, Zou SL, Xin H, Qu LF, Zhu YZ. 1074/jbc.M110073200. NADPH oxidase 4 contributes to connective tissue growth factor expression through Smad3-dependent signaling pathway. Free Radic Biol Med 94: 174–184, 581. Li L, Fink GD, Watts SW, Northcott CA, Galligan JJ, Pagano PJ, Chen AF. Endothe- 2016. doi:10.1016/j.freeradbiomed.2016.02.031. lin-1 increases vascular superoxide via endothelin(A)-NADPH oxidase pathway in low-renin hypertension. Circulation 107: 1053–1058, 2003. doi:10.1161/01.CIR. 597. Lötzer K, Funk CD, Habenicht AJR. The 5-lipoxygenase pathway in arterial wall 0000051459.74466.46. biology and atherosclerosis. Biochim Biophys Acta 1736: 30–37, 2005. doi:10.1016/ j.bbalip.2005.07.001. 582. Li Q, Fu GB, Zheng JT, He J, Niu XB, Chen QD, Yin Y, Qian X, Xu Q, Wang M, Sun AF, Shu Y, Rui H, Liu LZ, Jiang BH. NADPH oxidase subunit p22(phox)-mediated 598. Lucock M. Folic acid: nutritional biochemistry, molecular biology, and role in disease reactive oxygen species contribute to angiogenesis and tumor growth through AKT processes. Mol Genet Metab 71: 121–138, 2000. doi:10.1006/mgme.2000.3027.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 365 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

599. Lührs H, Papadopoulos T, Schmidt HH, Menzel T. Type I nitric oxide synthase in the 616. Maia LB, Moura JJG. Nitrite reduction by molybdoenzymes: a new class of nitric human lung is predominantly expressed in capillary endothelial cells. Respir Physiol oxide-forming nitrite reductases. J Biol Inorg Chem 20: 403–433, 2015. doi:10.1007/ 129: 367–374, 2002. doi:10.1016/S0034-5687(01)00323-1. s00775-014-1234-2.

600. Lundberg JO, Gladwin MT, Weitzberg E. Strategies to increase nitric oxide signalling 617. Maia LB, Pereira V, Mira L, Moura JJ. Nitrite reductase activity of rat and human in cardiovascular disease. Nat Rev Drug Discov 14: 623–641, 2015. doi:10.1038/ xanthine oxidase, xanthine dehydrogenase, and aldehyde oxidase: evaluation of their nrd4623. contribution to NO formation in vivo. Biochemistry 54: 685–710, 2015. doi:10.1021/ bi500987w. 601. Lundberg JO, Govoni M. Inorganic nitrate is a possible source for systemic genera- tion of nitric oxide. Free Radic Biol Med 37: 395–400, 2004. doi:10.1016/j. 618. Maier W, Cosentino F, Lütolf RB, Fleisch M, Seiler C, Hess OM, Meier B, Lüscher TF. freeradbiomed.2004.04.027. Tetrahydrobiopterin improves endothelial function in patients with coronary artery disease. J Cardiovasc Pharmacol 35: 173–178, 2000. doi:10.1097/00005344- 602. Lundberg JO, Weitzberg E, Cole JA, Benjamin N. Nitrate, bacteria and human health. 200002000-00001. Nat Rev Microbiol 2: 593–602, 2004. doi:10.1038/nrmicro929. Erratum at: doi:10. 619. Mäki-Petäjä KM, Day L, Cheriyan J, Hall FC, Östör AJ, Shenker N, Wilkinson IB. 1038/nrmicro982. Tetrahydrobiopterin supplementation improves endothelial function but does not 603. Lundberg JO, Weitzberg E, Gladwin MT. The nitrate-nitrite-nitric oxide pathway in alter aortic stiffness in patients with rheumatoid arthritis. J Am Heart Assoc 5: physiology and therapeutics. Nat Rev Drug Discov 7: 156–167, 2008. doi:10.1038/ e002762, 2016. doi:10.1161/JAHA.115.002762. nrd2466. 620. Malik UZ, Hundley NJ, Romero G, Radi R, Freeman BA, Tarpey MM, Kelley EE. Febuxostat inhibition of endothelial-bound XO: implications for targeting vascular 604. Lundberg JO, Farkas-Szallasi T, Weitzberg E, Rinder J, Lidholm J, Anggåard A, Hök- ROS production. Free Radic Biol Med 51: 179–184, 2011. doi:10.1016/j. felt T, Lundberg JM, Alving K. High nitric oxide production in human paranasal freeradbiomed.2011.04.004. sinuses. Nat Med 1: 370–373, 1995. doi:10.1038/nm0495-370. 621. Malle E, Waeg G, Schreiber R, Gröne EF, Sattler W, Gröne HJ. Immunohistochem- 605. Lundberg JO, Weitzberg E, Lundberg JM, Alving K. Intragastric nitric oxide produc- ical evidence for the myeloperoxidase/H2O2/halide system in human atheroscle- tion in humans: measurements in expelled air. Gut 35: 1543–1546, 1994. doi:10. rotic lesions: colocalization of myeloperoxidase and hypochlorite-modified proteins. 1136/gut.35.11.1543. Eur J Biochem 267: 4495–4503, 2000. doi:10.1046/j.1432-1327.2000.01498.x.

606. Lüneburg N, Xanthakis V, Schwedhelm E, Sullivan LM, Maas R, Anderssohn M, 622. Manea A, Manea SA, Florea IC, Luca CM, Raicu M. Positive regulation of NADPH Riederer U, Glazer NL, Vasan RS, Böger RH. Reference intervals for plasma L-argi- oxidase 5 by proinflammatory-related mechanisms in human aortic smooth muscle nine and the L-arginine:asymmetric dimethylarginine ratio in the Framingham Off- cells. Free Radic Biol Med 52: 1497–1507, 2012. doi:10.1016/j.freeradbiomed.2012. spring Cohort. J Nutr 141: 2186–2190, 2011. doi:10.3945/jn.111.148197. 02.018.

607. Lyle AN, Deshpande NN, Taniyama Y, Seidel-Rogol B, Pounkova L, Du P, Papaha- 623. Manea A, Manea SA, Gan AM, Constantin A, Fenyo IM, Raicu M, Muresian H, ralambus C, Lassègue B, Griendling KK. Poldip2, a novel regulator of Nox4 and Simionescu M. Human monocytes and macrophages express NADPH oxidase 5; a cytoskeletal integrity in vascular smooth muscle cells. Circ Res 105: 249–259, 2009. potential source of reactive oxygen species in atherosclerosis. Biochem Biophys Res doi:10.1161/CIRCRESAHA.109.193722. Commun 461: 172–179, 2015. doi:10.1016/j.bbrc.2015.04.021.

608. Lymar SV, Hurst JK. Rapid reaction between peroxonitrite ion and carbon-dioxide - 624. Marfella R, Di Filippo C, Esposito K, Nappo F, Piegari E, Cuzzocrea S, Berrino L, implications for biological-activity. J Am Chem Soc 117: 8867–8868, 1995. doi:10. Rossi F, Giugliano D, D’Amico M. Absence of inducible nitric oxide synthase reduces 1021/ja00139a027. myocardial damage during ischemia reperfusion in streptozotocin-induced hyper- glycemic mice. Diabetes 53: 454–462, 2004. doi:10.2337/diabetes.53.2.454. 609. Lynch CM, Kinzenbaw DA, Chen X, Zhan S, Mezzetti E, Filosa J, Ergul A, Faulkner JL, Faraci FM, Didion SP. Nox2-derived superoxide contributes to cerebral vascular 625. Martin E, Berka V, Bogatenkova E, Murad F, Tsai A-L. Ligand selectivity of soluble dysfunction in diet-induced obesity. Stroke 44: 3195–3201, 2013. doi:10.1161/ guanylyl cyclase: effect of the hydrogen-bonding tyrosine in the distal heme pocket on binding of oxygen, nitric oxide, and carbon monoxide. J Biol Chem 281: 27836– STROKEAHA.113.001366. 27845, 2006. doi:10.1074/jbc.M601078200. 610. MacDonald TM, Ford I, Nuki G, Mackenzie IS, De Caterina R, Findlay E, Hallas J, 626. Martin E, Golunski E, Laing ST, Estrera AL, Sharina IG. Alternative splicing impairs Hawkey CJ, Ralston S, Walters M, Webster J, McMurray J, Perez Ruiz F, Jennings CG, soluble guanylyl cyclase function in aortic aneurysm. Am J Physiol Heart Circ Physiol MacDonald T, Ford I, Nuki G, Mackenzie I, Hallas J, Webster J, Walters M, Ralston S, 307: H1565–H1575, 2014. doi:10.1152/ajpheart.00222.2014. Hawkey C, De Caterina R, Perez-Ruiz F, Findlay E, McMurray J, Maseri A, Murray G, Bird H, McMurray J, Petrie M, MacDonald M, Jhund P, Saywood W, Flynn R, Ford I, 627. Martyn KD, Frederick LM, von Loehneysen K, Dinauer MC, Knaus UG. Functional Kean S; Members of the FAST Study Group. Protocol of the Febuxostat versus analysis of Nox4 reveals unique characteristics compared to other NADPH oxi- Allopurinol Streamlined Trial (FAST): a large prospective, randomised, open, blinded dases. Cell Signal 18: 69–82, 2006. doi:10.1016/j.cellsig.2005.03.023. endpoint study comparing the cardiovascular safety of allopurinol and febuxostat in the management of symptomatic hyperuricaemia. BMJ Open 4: e005354, 2014. 628. Matsui A, Okigaki M, Amano K, Adachi Y, Jin D, Takai S, Yamashita T, Kawashima S, doi:10.1136/bmjopen-2014-005354. Kurihara T, Miyazaki M, Tateishi K, Matsunaga S, Katsume A, Honshou S, Takahashi T, Matoba S, Kusaba T, Tatsumi T, Matsubara H. Central role of calcium-dependent 611. Macfadyen AJ, Reiter C, Zhuang Y, Beckman JS. A novel superoxide dismutase-based tyrosine kinase PYK2 in endothelial nitric oxide synthase-mediated angiogenic re- trap for peroxynitrite used to detect entry of peroxynitrite into erythrocyte ghosts. sponse and vascular function. Circulation 116: 1041–1051, 2007. doi:10.1161/ Chem Res Toxicol 12: 223–229, 1999. doi:10.1021/tx980253u. CIRCULATIONAHA.106.645416. 629. Matsumoto K, Okamoto K, Ashizawa N, Nishino T. FYX-051: a novel and potent 612. Madamanchi NR, Vendrov A, Runge MS. Oxidative stress and vascular disease. hybrid-type inhibitor of xanthine oxidoreductase. J Pharmacol Exp Ther 336: 95–103, Arterioscler Thromb Vasc Biol 25: 29–38, 2005. doi:10.1161/01.ATV.0000150649. 2011. doi:10.1124/jpet.110.174540. 39934.13. 630. Matsuno K, Yamada H, Iwata K, Jin D, Katsuyama M, Matsuki M, Takai S, Yamanishi 613. Magnani F, Nenci S, Millana Fananas E, Ceccon M, Romero E, Fraaije MW, Mattevi A. K, Miyazaki M, Matsubara H, Yabe-Nishimura C. Nox1 is involved in angiotensin Crystal structures and atomic model of NADPH oxidase. Proc Natl Acad SciUSA II-mediated hypertension: a study in Nox1-deficient mice. Circulation 112: 2677– 114: 6764–6769, 2017. doi:10.1073/pnas.1702293114. 2685, 2005. doi:10.1161/CIRCULATIONAHA.105.573709.

614. Maia LB, Moura JJ. Nitrite reduction by xanthine oxidase family enzymes: a new class 631. McCabe TJ, Fulton D, Roman LJ, Sessa WC. Enhanced electron flux and reduced of nitrite reductases. J Biol Inorg Chem 16: 443–460, 2011. doi:10.1007/s00775-010- calmodulin dissociation may explain “calcium-independent” eNOS activation by 0741-z. phosphorylation. J Biol Chem 275: 6123–6128, 2000. doi:10.1074/jbc.275.9.6123.

615. Maia LB, Moura JJG. How biology handles nitrite. Chem Rev 114: 5273–5357, 2014. 632. McCarty MF. Asymmetric dimethylarginine is a well established mediating risk factor doi:10.1021/cr400518y. for cardiovascular morbidity and mortality-should patients with elevated levels be

366 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

supplemented with citrulline? Healthcare (Basel) 4: E40, 2016. doi:10.3390/ 650. Millar TM, Stevens CR, Benjamin N, Eisenthal R, Harrison R, Blake DR. Xanthine healthcare4030040. oxidoreductase catalyses the reduction of nitrates and nitrite to nitric oxide under hypoxic conditions. FEBS Lett 427: 225–228, 1998. doi:10.1016/S0014- 633. McCord JM. Oxygen-derived free radicals in postischemic tissue injury. N Engl J Med 5793(98)00430-X. 312: 159–163, 1985. doi:10.1056/NEJM198501173120305. 651. Miller FJ Jr, Chu X, Stanic B, Tian X, Sharma RV, Davisson RL, Lamb FS. A differential 634. McDonald KK, Zharikov S, Block ER, Kilberg MS. A caveolar complex between the role for endocytosis in receptor-mediated activation of Nox1. Antioxid Redox Signal cationic amino acid transporter 1 and endothelial nitric-oxide synthase may explain 12: 583–593, 2010. doi:10.1089/ars.2009.2857. the “arginine paradox”. J Biol Chem 272: 31213–31216, 1997. doi:10.1074/jbc.272. 50.31213. 652. Miller FJ Jr, Filali M, Huss GJ, Stanic B, Chamseddine A, Barna TJ, Lamb FS. Cytokine activation of nuclear factor kappa B in vascular smooth muscle cells requires signaling 635. Medinas DB, Cerchiaro G, Trindade DF, Augusto O. The carbonate radical and endosomes containing Nox1 and ClC-3. Circ Res 101: 663–671, 2007. doi:10.1161/ related oxidants derived from bicarbonate buffer. IUBMB Life 59: 255–262, 2007. CIRCRESAHA.107.151076. doi:10.1080/15216540701230511. 653. Miller RT, Martásek P, Roman LJ, Nishimura JS, Masters BSS. Involvement of the 636. Mehrabian M, Allayee H, Wong J, Shi W, Wang XP, Shaposhnik Z, Funk CD, Lusis AJ. reductase domain of neuronal nitric oxide synthase in superoxide anion production. Identification of 5-lipoxygenase as a major gene contributing to atherosclerosis sus- Biochemistry 36: 15277–15284, 1997. doi:10.1021/bi972022c. ceptibility in mice. Circ Res 91: 120–126, 2002. doi:10.1161/01.RES.0000028008. 654. Milsom AB, Patel NSA, Mazzon E, Tripatara P, Storey A, Mota-Filipe H, Sepodes B, 99774.7F. Webb AJ, Cuzzocrea S, Hobbs AJ, Thiemermann C, Ahluwalia A. Role for endothelial 637. Meijles DN, Howlin BJ, Li JM. Consensus in silico computational modelling of the nitric oxide synthase in nitrite-induced protection against renal ischemia-reperfusion p22phox subunit of the NADPH oxidase. Comput Biol Chem 39: 6–13, 2012. doi:10. injury in mice. Nitric Oxide 22: 141–148, 2010. doi:10.1016/j.niox.2009.10.010. 1016/j.compbiolchem.2012.05.001. 655. Mittendorf J, Weigand S, Alonso-Alija C, Bischoff E, Feurer A, Gerisch M, Kern A, Knorr A, Lang D, Muenter K, Radtke M, Schirok H, Schlemmer K-H, Stahl E, Straub 638. Meitzler JL, Ortiz de Montellano PR. Caenorhabditis elegans and human dual oxidase A, Wunder F, Stasch J-P. Discovery of riociguat (BAY 63-2521): a potent, oral 1 (DUOX1) “peroxidase” domains: insights into heme binding and catalytic activity. stimulator of soluble guanylate cyclase for the treatment of pulmonary hypertension. J Biol Chem 284: 18634–18643, 2009. doi:10.1074/jbc.M109.013581. ChemMedChem 4: 853–865, 2009. doi:10.1002/cmdc.200900014. 639. Meitzler JL, Ortiz de Montellano PR. Structural stability and heme binding potential 656. Miyazaki H, Matsuoka H, Cooke JP, Usui M, Ueda S, Okuda S, Imaizumi T. Endog- of the truncated human dual oxidase 2 (DUOX2) peroxidase domain. Arch Biochem enous nitric oxide synthase inhibitor: a novel marker of atherosclerosis. Circulation Biophys 512: 197–203, 2011. doi:10.1016/j.abb.2011.05.021. 99: 1141–1146, 1999. doi:10.1161/01.CIR.99.9.1141.

640. Meli R, Nauser T, Koppenol WH. Direct observation of intermediates in the reaction 657. Mohazzab KM, Kaminski PM, Wolin MS. NADH oxidoreductase is a major source of of peroxynitrite with carbon dioxide. Helv Chim Acta 82: 722–725, 1999. superoxide anion in bovine coronary artery endothelium. Am J Physiol Heart Circ Physiol 266: H2568–H2572, 1994. 641. Mendes-Silverio CB, Leiria LOS, Morganti RP, Anhê GF, Marcondes S, Mónica FZ, De Nucci G, Antunes E. Activation of haem-oxidized soluble guanylyl cyclase with 658. Mollnau H, Wendt M, Szöcs K, Lassègue B, Schulz E, Oelze M, Li H, Bodenschatz M, BAY 60-2770 in human platelets lead to overstimulation of the cyclic GMP signaling August M, Kleschyov AL, Tsilimingas N, Walter U, Förstermann U, Meinertz T, pathway. PLoS One 7: e47223, 2012. doi:10.1371/journal.pone.0047223. Griendling K, Münzel T. Effects of angiotensin II infusion on the expression and function of NAD(P)H oxidase and components of nitric oxide/cGMP signaling. Circ 642. Meng D, Mei A, Liu J, Kang X, Shi X, Qian R, Chen S. NADPH oxidase 4 mediates Res 90: E58–E65, 2002. doi:10.1161/01.RES.0000012569.55432.02. insulin-stimulated HIF-1␣ and VEGF expression, and angiogenesis in vitro. PLoS One 7: e48393, 2012. doi:10.1371/journal.pone.0048393. 659. Moncada S, Palmer RMJ, Higgs EA. Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev 43: 109–142, 1991. 643. Meurer S, Pioch S, Pabst T, Opitz N, Schmidt PM, Beckhaus T, Wagner K, Matt S, Gegenbauer K, Geschka S, Karas M, Stasch J-P, Schmidt HHHW, Müller-Esterl W. 660. Montezano AC, Burger D, Paravicini TM, Chignalia AZ, Yusuf H, Almasri M, He Y, Nitric oxide-independent vasodilator rescues heme-oxidized soluble guanylate cy- Callera GE, He G, Krause K-H, Lambeth D, Quinn MT, Touyz RM. Nicotinamide clase from proteasomal degradation. Circ Res 105: 33–41, 2009. doi:10.1161/ adenine dinucleotide phosphate reduced oxidase 5 (Nox5) regulation by angiotensin CIRCRESAHA.109.198234. II and endothelin-1 is mediated via calcium/calmodulin-dependent, rac-1-indepen- dent pathways in human endothelial cells. Circ Res 106: 1363–1373, 2010. doi:10. 644. Meuwese MC, Stroes ESG, Hazen SL, van Miert JN, Kuivenhoven JA, Schaub RG, 1161/CIRCRESAHA.109.216036. Wareham NJ, Luben R, Kastelein JJP, Khaw K-T, Boekholdt SM. Serum myeloper- oxidase levels are associated with the future risk of coronary artery disease in 661. Montezano AC, Tsiropoulou S, Dulak-Lis M, Harvey A, Camargo LL, Touyz RM. apparently healthy individuals: the EPIC-Norfolk Prospective Population Study. JAm Redox signaling, Nox5 and vascular remodeling in hypertension. Curr Opin Nephrol Coll Cardiol 50: 159–165, 2007. doi:10.1016/j.jacc.2007.03.033. Hypertens 24: 425–433, 2015. doi:10.1097/MNH.0000000000000153. 662. Montfort WR, Wales JA, Weichsel A. Structure and Activation of Soluble Guanylyl 645. Michelakis ED, Hampl V, Nsair A, Wu X, Harry G, Haromy A, Gurtu R, Archer SL. Cyclase, the Nitric Oxide Sensor. Antioxid Redox Signal 26: 107–121, 2017. doi:10. Diversity in mitochondrial function explains differences in vascular oxygen sensing. 1089/ars.2016.6693. Circ Res 90: 1307–1315, 2002. doi:10.1161/01.RES.0000024689.07590.C2. 663. Monti LD, Casiraghi MC, Setola E, Galluccio E, Pagani MA, Quaglia L, Bosi E, Piatti P. 646. Michell BJ, Chen Zp, Tiganis T, Stapleton D, Katsis F, Power DA, Sim AT, Kemp BE. L-arginine enriched biscuits improve endothelial function and glucose metabolism: a Coordinated control of endothelial nitric-oxide synthase phosphorylation by protein pilot study in healthy subjects and a cross-over study in subjects with impaired kinase C and the cAMP-dependent protein kinase. J Biol Chem 276: 17625–17628, glucose tolerance and metabolic syndrome. Metabolism 62: 255–264, 2013. doi:10. 2001. doi:10.1074/jbc.C100122200. 1016/j.metabol.2012.08.004.

647. Michell BJ, Harris MB, Chen ZP, Ju H, Venema VJ, Blackstone MA, Huang W, Ven- 664. Moore PK, Babbedge RC, Wallace P, Gaffen ZA, Hart SL. 7-Nitro indazole, an ema RC, Kemp BE. Identification of regulatory sites of phosphorylation of the bovine inhibitor of nitric oxide synthase, exhibits anti-nociceptive activity in the mouse endothelial nitric-oxide synthase at serine 617 and serine 635. J Biol Chem 277: without increasing blood pressure. Br J Pharmacol 108: 296–297, 1993. doi:10.1111/ 42344–42351, 2002. doi:10.1074/jbc.M205144200. j.1476-5381.1993.tb12798.x.

648. Mihm MJ, Yu F, Carnes CA, Reiser PJ, McCarthy PM, Van Wagoner DR, Bauer JA. 665. Mori E, Haramaki N, Ikeda H, Imaizumi T. Intra-coronary administration of L-argi- Impaired myofibrillar energetics and oxidative injury during human atrial fibrillation. nine aggravates myocardial stunning through production of peroxynitrite in dogs. Circulation 104: 174–180, 2001. doi:10.1161/01.CIR.104.2.174. Cardiovasc Res 40: 113–123, 1998. doi:10.1016/S0008-6363(98)00146-1.

649. Miles EW, Kraus JP. Cystathionine beta-synthase: structure, function, regulation, and 666. Morishita T, Tsutsui M, Shimokawa H, Horiuchi M, Tanimoto A, Suda O, Tasaki H, location of homocystinuria-causing mutations. J Biol Chem 279: 29871–29874, 2004. Huang PL, Sasaguri Y, Yanagihara N, Nakashima Y. Vasculoprotective roles of neu- doi:10.1074/jbc.R400005200. ronal nitric oxide synthase. FASEB J 16: 1994–1996, 2002. doi:10.1096/fj.02-0155fje.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 367 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

667. Morris CR, Kato GJ, Poljakovic M, Wang X, Blackwelder WC, Sachdev V, Hazen SL, 686. Nakamura T, Lipton SA. Protein S-Nitrosylation as a Therapeutic Target for Neu- Vichinsky EP, Morris SM Jr, Gladwin MT. Dysregulated arginine metabolism, hemo- rodegenerative Diseases. Trends Pharmacol Sci 37: 73–84, 2016. doi:10.1016/j.tips. lysis-associated pulmonary hypertension, and mortality in sickle cell disease. JAMA 2015.10.002. 294: 81–90, 2005. doi:10.1001/jama.294.1.81. 687. Nakatani K, Okuyama H, Shimahara Y, Saeki S, Kim DH, Nakajima Y, Seki S, Kawada 668. Morris SM Jr. Arginine Metabolism Revisited. J Nutr 146: 2579S–2586S, 2016. doi: N, Yoshizato K. Cytoglobin/STAP, its unique localization in splanchnic fibroblast-like 10.3945/jn.115.226621. cells and function in organ fibrogenesis. Lab Invest 84: 91–101, 2004. doi:10.1038/ labinvest.3700013. 669. Mount PF, Kemp BE, Power DA. Regulation of endothelial and myocardial NO synthesis by multi-site eNOS phosphorylation. J Mol Cell Cardiol 42: 271–279, 2007. 688. Nathan C. Inducible nitric oxide synthase: what difference does it make? J Clin Invest doi:10.1016/j.yjmcc.2006.05.023. 100: 2417–2423, 1997. doi:10.1172/JCI119782.

670. Mullershausen F, Russwurm M, Friebe A, Koesling D. Inhibition of phosphodiester- 689. NaveenKumar SK, Hemshekhar M, Sundaram MS, Kemparaju K, Girish KS. Cell-free ase type 5 by the activator of nitric oxide-sensitive guanylyl cyclase BAY 41-2272. methemoglobin drives platelets to apoptosis via mitochondrial ROS-mediated acti- vation of JNK and p38 MAP kinase. Biochem Biophys Res Commun 491: 183–191, Circulation 109: 1711–1713, 2004. doi:10.1161/01.CIR.0000126286.47618.BD. 2017. doi:10.1016/j.bbrc.2017.07.073. 671. Mülsch A, Busse R, Bassenge E. Desensitization of guanylate cyclase in nitrate toler- 690. Ndrepepa G, Braun S, Mehilli J, von Beckerath N, Schömig A, Kastrati A. Myeloper- ance does not impair endothelium-dependent responses. Eur J Pharmacol 158: 191– oxidase level in patients with stable coronary artery disease and acute coronary 198, 1988. doi:10.1016/0014-2999(88)90066-0. syndromes. Eur J Clin Invest 38: 90–96, 2008. doi:10.1111/j.1365-2362.2007. 672. Münzel T, Camici GG, Maack C, Bonetti NR, Fuster V, Kovacic JC. Impact of Oxi- 01908.x. dative Stress on the Heart and Vasculature: Part 2 of a 3-Part Series. J Am Coll Cardiol 691. Nedospasov A, Rafikov R, Beda N, Nudler E. An autocatalytic mechanism of protein 70: 212–229, 2017. doi:10.1016/j.jacc.2017.05.035. nitrosylation. Proc Natl Acad Sci USA 97: 13543–13548, 2000. doi:10.1073/pnas. 250398197. 673. Münzel T, Feil R, Mülsch A, Lohmann SM, Hofmann F, Walter U. Physiology and pathophysiology of vascular signaling controlled by guanosine 3=,5=-cyclic mono- 692. Nedospasov AA. Is N2O3 the main nitrosating intermediate in aerated nitric oxide phosphate-dependent protein kinase [corrected]. Circulation 108: 2172–2183, (NO) solutions in vivo? If so, where, when, and which one? J Biochem Mol Toxicol 16: 2003. doi:10.1161/01.CIR.0000094403.78467.C3. 109–120, 2002. doi:10.1002/jbt.10029.

674. Murad F. Shattuck Lecture. Nitric oxide and cyclic GMP in cell signaling and drug 693. Nishino T, Nishino T. The conversion from the dehydrogenase type to the oxidase development. N Engl J Med 355: 2003–2011, 2006. doi:10.1056/NEJMsa063904. type of rat liver xanthine dehydrogenase by modification of cysteine residues with fluorodinitrobenzene. J Biol Chem 272: 29859–29864, 1997. doi:10.1074/jbc.272. 675. Murataliev MB, Feyereisen R, Walker FA. Electron transfer by diflavin reductases. 47.29859. Biochim Biophys Acta 1698: 1–26, 2004. doi:10.1016/j.bbapap.2003.10.003. 694. Nisimoto Y, Jackson HM, Ogawa H, Kawahara T, Lambeth JD. Constitutive NA- 676. Murdoch CE, Alom-Ruiz SP, Wang M, Zhang M, Walker S, Yu B, Brewer A, Shah AM. DPH-dependent electron transferase activity of the Nox4 dehydrogenase domain. Role of endothelial Nox2 NADPH oxidase in angiotensin II-induced hypertension Biochemistry 49: 2433–2442, 2010. doi:10.1021/bi9022285. and vasomotor dysfunction. Basic Res Cardiol 106: 527–538, 2011. doi:10.1007/ s00395-011-0179-7. Erratum at: doi:10.1007/s00395-014-0410-4. 695. Niu X-L, Madamanchi NR, Vendrov AE, Tchivilev I, Rojas M, Madamanchi C, Brandes RP, Krause K-H, Humphries J, Smith A, Burnand KG, Runge MS. Nox activator 1: a 677. Murphy E, Kohr M, Menazza S, Nguyen T, Evangelista A, Sun J, Steenbergen C. potential target for modulation of vascular reactive oxygen species in atherosclerotic Signaling by S-nitrosylation in the heart. J Mol Cell Cardiol 73: 18–25, 2014. doi:10. arteries. Circulation 121: 549–559, 2010. doi:10.1161/CIRCULATIONAHA.109. 1016/j.yjmcc.2014.01.003. 908319.

678. Murphy MP. How mitochondria produce reactive oxygen species. Biochem J 417: 696. Nojiri H, Shimizu T, Funakoshi M, Yamaguchi O, Zhou H, Kawakami S, Ohta Y, Sami 1–13, 2009. doi:10.1042/BJ20081386. M, Tachibana T, Ishikawa H, Kurosawa H, Kahn RC, Otsu K, Shirasawa T. Oxidative stress causes heart failure with impaired mitochondrial respiration. J Biol Chem 281: 679. Murphy MP, Smith RAJ. Targeting antioxidants to mitochondria by conjugation to 33789–33801, 2006. doi:10.1074/jbc.M602118200. lipophilic cations. Annu Rev Pharmacol Toxicol 47: 629–656, 2007. doi:10.1146/ annurev.pharmtox.47.120505.105110. 697. Nomura J, Busso N, Ives A, Matsui C, Tsujimoto S, Shirakura T, Tamura M, Ko- bayashi T, So A, Yamanaka Y. Xanthine oxidase inhibition by febuxostat attenuates 680. Musset B, Clark RA, DeCoursey TE, Petheo GL, Geiszt M, Chen Y, Cornell JE, Eddy experimental atherosclerosis in mice. Sci Rep 4: 4554, 2014. doi:10.1038/srep04554. CA, Brzyski RG, El Jamali A. NOX5 in human spermatozoa: expression, function, and regulation. J Biol Chem 287: 9376–9388, 2012. doi:10.1074/jbc.M111.314955. 698. Nunokawa Y, Ishida N, Tanaka S. Cloning of inducible nitric oxide synthase in rat vascular smooth muscle cells. Biochem Biophys Res Commun 191: 89–94, 1993. doi: 681. Muzaffar S, Jeremy JY, Angelini GD, Shukla N. NADPH oxidase 4 mediates upregu- 10.1006/bbrc.1993.1188. lation of type 4 phosphodiesterases in human endothelial cells. J Cell Physiol 227: 699. O’Donnell BV, Tew DG, Jones OTG, England PJ. Studies on the inhibitory mecha- 1941–1950, 2012. doi:10.1002/jcp.22922. nism of iodonium compounds with special reference to neutrophil NADPH oxidase. 682. Nabeebaccus A, Zhang M, Shah AM. NADPH oxidases and cardiac remodelling. Biochem J 290: 41–49, 1993. doi:10.1042/bj2900041. Heart Fail Rev 16: 5–12, 2011. doi:10.1007/s10741-010-9186-2. 700. Ohara Y, Peterson TE, Harrison DG. Hypercholesterolemia increases endothelial 683. Nadtochiy SM, Zhu QM, Urciuoli W, Rafikov R, Black SM, Brookes PS. Nitroalkenes superoxide anion production. J Clin Invest 91: 2546–2551, 1993. doi:10.1172/ confer acute cardioprotection via adenine nucleotide translocase 1. J Biol Chem 287: JCI116491. 3573–3580, 2012. doi:10.1074/jbc.M111.298406. Erratum at: doi:10.1074/jbc. 701. Ohtsubo T, Rovira II, Starost MF, Liu C, Finkel T. Xanthine oxidoreductase is an A111.298406. endogenous regulator of cyclooxygenase-2. Circ Res 95: 1118–1124, 2004. doi:10. 1161/01.RES.0000149571.96304.36. 684. Nagababu E, Ramasamy S, Rifkind JM. Intermediates detected by visible spectros- copy during the reaction of nitrite with deoxyhemoglobin: the effect of nitrite con- 702. Okamoto K, Eger BT, Nishino T, Kondo S, Pai EF, Nishino T. An extremely potent centration and diphosphoglycerate. Biochemistry 46: 11650–11659, 2007. doi:10. inhibitor of xanthine oxidoreductase. Crystal structure of the enzyme-inhibitor com- 1021/bi700364e. plex and mechanism of inhibition. J Biol Chem 278: 1848–1855, 2003. doi:10.1074/ jbc.M208307200. 685. Nakahira K, Kim HP, Geng XH, Nakao A, Wang X, Murase N, Drain PF, Wang X, Sasidhar M, Nabel EG, Takahashi T, Lukacs NW, Ryter SW, Morita K, Choi AMK. 703. Okamoto K, Matsumoto K, Hille R, Eger BT, Pai EF, Nishino T. The crystal structure Carbon monoxide differentially inhibits TLR signaling pathways by regulating ROS- of xanthine oxidoreductase during catalysis: implications for reaction mechanism and induced trafficking of TLRs to lipid rafts. J Exp Med 203: 2377–2389, 2006. doi:10. enzyme inhibition. Proc Natl Acad Sci USA 101: 7931–7936, 2004. doi:10.1073/pnas. 1084/jem.20060845. 0400973101.

368 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

704. Oller J, Méndez-Barbero N, Ruiz EJ, Villahoz S, Renard M, Canelas LI, Briones AM, 722. Panza JA, García CE, Kilcoyne CM, Quyyumi AA, Cannon RO III. Impaired endothe- Alberca R, Lozano-Vidal N, Hurlé MA, Milewicz D, Evangelista A, Salaices M, Nistal lium-dependent vasodilation in patients with essential hypertension. Evidence that JF, Jiménez-Borreguero LJ, De Backer J, Campanero MR, Redondo JM. Nitric oxide nitric oxide abnormality is not localized to a single signal transduction pathway. mediates aortic disease in mice deficient in the metalloprotease Adamts1 and in a Circulation 91: 1732–1738, 1995. doi:10.1161/01.CIR.91.6.1732. mouse model of Marfan syndrome. Nat Med 23: 200–212, 2017. doi:10.1038/nm. 4266. 723. Paravicini TM, Chrissobolis S, Drummond GR, Sobey CG. Increased NADPH-oxi- dase activity and Nox4 expression during chronic hypertension is associated with 705. Ott G, Havemeyer A, Clement B. The mammalian molybdenum enzymes of mARC. enhanced cerebral vasodilatation to NADPH in vivo. Stroke 35: 584–589, 2004. J Biol Inorg Chem 20: 265–275, 2015. doi:10.1007/s00775-014-1216-4. doi:10.1161/01.STR.0000112974.37028.58.

706. Ott G, Plitzko B, Krischkowski C, Reichmann D, Bittner F, Mendel RR, Kunze T, 724. Park HS, Jung HY, Park EY, Kim J, Lee WJ, Bae YS. Cutting edge: direct interaction of Clement B, Havemeyer A. Reduction of sulfamethoxazole hydroxylamine (SMX- TLR4 with NAD(P)H oxidase 4 isozyme is essential for lipopolysaccharide-induced HA) by the mitochondrial amidoxime reducing component (mARC). Chem Res Toxi- production of reactive oxygen species and activation of NF-kappa B. J Immunol 173: col 27: 1687–1695, 2014. doi:10.1021/tx500174u. 3589–3593, 2004. doi:10.4049/jimmunol.173.6.3589.

707. Owusu-Ansah E, Banerjee U. Reactive oxygen species prime Drosophila haemato- 725. Patterson C, Ruef J, Madamanchi NR, Barry-Lane P, Hu Z, Horaist C, Ballinger CA, poietic progenitors for differentiation. Nature 461: 537–541, 2009. doi:10.1038/ Brasier AR, Bode C, Runge MS. Stimulation of a vascular smooth muscle cell NA- nature08313. D(P)H oxidase by thrombin. Evidence that p47(phox) may participate in forming this oxidase in vitro and in vivo. J Biol Chem 274: 19814–19822, 1999. doi:10.1074/jbc. 708. Ozaki M, Kawashima S, Yamashita T, Hirase T, Namiki M, Inoue N, Hirata K, Yasui 274.28.19814. H, Sakurai H, Yoshida Y, Masada M, Yokoyama M. Overexpression of endothelial nitric oxide synthase accelerates atherosclerotic lesion formation in apoE-deficient 726. Pattison DI, Davies MJ. Absolute rate constants for the reaction of hypochlorous acid mice. J Clin Invest 110: 331–340, 2002. doi:10.1172/JCI0215215. with protein side chains and peptide bonds. Chem Res Toxicol 14: 1453–1464, 2001. doi:10.1021/tx0155451. 709. Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev 87: 315–424, 2007. doi:10.1152/physrev.00029.2006. 727. Pautz A, Art J, Hahn S, Nowag S, Voss C, Kleinert H. Regulation of the expression of inducible nitric oxide synthase. Nitric Oxide 23: 75–93, 2010. doi:10.1016/j.niox. 710. Pacher P, Nivorozhkin A, Szabó C. Therapeutic effects of xanthine oxidase inhibi- 2010.04.007. tors: renaissance half a century after the discovery of allopurinol. Pharmacol Rev 58: 87–114, 2006. doi:10.1124/pr.58.1.6. 728. Pawloski JR, Hess DT, Stamler JS. Export by red blood cells of nitric oxide bioactivity. Nature 409: 622–626, 2001. doi:10.1038/35054560. 711. Padayatti PS, Pattanaik P, Ma X, van den Akker F. Structural insights into the regu- lation and the activation mechanism of mammalian guanylyl cyclases. Pharmacol Ther 729. Pawloski JR, Hess DT, Stamler JS. Impaired vasodilation by red blood cells in sickle 104: 83–99, 2004. doi:10.1016/j.pharmthera.2004.08.003. cell disease. Proc Natl Acad Sci USA 102: 2531–2536, 2005. doi:10.1073/pnas. 0409876102. 712. Padilla F, Garcia-Dorado D, Agulló L, Inserte J, Paniagua A, Mirabet S, Barrabés JA, Ruiz-Meana M, Soler-Soler J. L-Arginine administration prevents reperfusion-in- 730. Peinado MA, Hernández R, Peragón J, Ovelleiro D, Pedrosa JA, Blanco S. Proteomic duced cardiomyocyte hypercontracture and reduces infarct size in the pig. Cardio- characterization of nitrated cell targets after hypobaric hypoxia and reoxygenation in rat brain. J Proteomics 109: 309–321, 2014. doi:10.1016/j.jprot.2014.07.015. vasc Res 46: 412–420, 2000. doi:10.1016/S0008-6363(00)00048-1. 731. Pelligrino DA, Koenig HM, Albrecht RF. Nitric oxide synthesis and regional cerebral 713. Pae H-O, Lee YC, Chung H-T. Heme oxygenase-1 and carbon monoxide: emerging blood flow responses to hypercapnia and hypoxia in the rat. J Cereb Blood Flow Metab therapeutic targets in inflammation and allergy. Recent Pat Inflamm Allergy Drug 13: 80–87, 1993. doi:10.1038/jcbfm.1993.10. Discov 2: 159–165, 2008. doi:10.2174/187221308786241929. 732. Peluffo H, Shacka JJ, Ricart K, Bisig CG, Martìnez-Palma L, Pritsch O, Kamaid A, 714. Pagano PJ, Clark JK, Cifuentes-Pagano ME, Clark SM, Callis GM, Quinn MT. Local- Eiserich JP, Crow JP, Barbeito L, Estèvez AG. Induction of motor neuron apoptosis ization of a constitutively active, phagocyte-like NADPH oxidase in rabbit aortic by free 3-nitro-L-tyrosine. J Neurochem 89: 602–612, 2004. doi:10.1046/j.1471- adventitia: enhancement by angiotensin II. Proc Natl Acad Sci USA 94: 14483–14488, 4159.2004.02363.x. 1997. doi:10.1073/pnas.94.26.14483. 733. Peri L, Pietraforte D, Scorza G, Napolitano A, Fogliano V, Minetti M. Apples increase 715. Pagano PJ, Ito Y, Tornheim K, Gallop PM, Tauber AI, Cohen RA. An NADPH oxidase nitric oxide production by human saliva at the acidic pH of the stomach: a new superoxide-generating system in the rabbit aorta. Am J Physiol 268: H2274–H2280, biological function for polyphenols with a catechol group? Free Radic Biol Med 39: 1995. 668–681, 2005. doi:10.1016/j.freeradbiomed.2005.04.021. 716. Palmer RMJ, Ferrige AG, Moncada S. Nitric oxide release accounts for the biological 734. Pernow J, Jung C. Arginase as a potential target in the treatment of cardiovascular activity of endothelium-derived relaxing factor. Nature 327: 524–526, 1987. doi:10. disease: reversal of arginine steal? Cardiovasc Res 98: 334–343, 2013. doi:10.1093/ 1038/327524a0. cvr/cvt036. 717. Pandey D, Fulton DJ. Molecular regulation of NADPH oxidase 5 via the MAPK 735. Petrônio MS, Zeraik ML, Fonseca LM, Ximenes VF. Apocynin: chemical and bio- pathway. Am J Physiol Heart Circ Physiol 300: H1336–H1344, 2011. doi:10.1152/ physical properties of a NADPH oxidase inhibitor. Molecules 18: 2821–2839, 2013. ajpheart.01163.2010. doi:10.3390/molecules18032821. 718. Pandey D, Gratton JP, Rafikov R, Black SM, Fulton DJ. Calcium/calmodulin-depen- 736. Petry A, Djordjevic T, Weitnauer M, Kietzmann T, Hess J, Görlach A. NOX2 and dent kinase II mediates the phosphorylation and activation of NADPH oxidase 5. Mol NOX4 mediate proliferative response in endothelial cells. Antioxid Redox Signal 8: Pharmacol 80: 407–415, 2011. doi:10.1124/mol.110.070193. 1473–1484, 2006. doi:10.1089/ars.2006.8.1473.

719. Pandey D, Patel A, Patel V, Chen F, Qian J, Wang Y, Barman SA, Venema RC, Stepp 737. Petry A, Weitnauer M, Görlach A. Receptor activation of NADPH oxidases. Antioxid DW, Rudic RD, Fulton DJ. Expression and functional significance of NADPH oxidase Redox Signal 13: 467–487, 2010. doi:10.1089/ars.2009.3026. 5 (Nox5) and its splice variants in human blood vessels. Am J Physiol Heart Circ Physiol 302: H1919–H1928, 2012. doi:10.1152/ajpheart.00910.2011. 738. Pfeiffer S, Mayer B, Hemmens B. Nitric oxide: Chemical puzzles posed by a biolog- ical messenger. Angew Chem Int Ed Engl 38: 1714–1731, 1999. 720. Panieri E, Santoro MM. ROS signaling and redox biology in endothelial cells. Cell Mol Life Sci 72: 3281–3303, 2015. doi:10.1007/s00018-015-1928-9. 739. Piantadosi CA. Carbon monoxide, reactive oxygen signaling, and oxidative stress. Free Radic Biol Med 45: 562–569, 2008. doi:10.1016/j.freeradbiomed.2008.05.013. 721. Pannirselvam M, Simon V, Verma S, Anderson T, Triggle CR. Chronic oral supple- mentation with sepiapterin prevents endothelial dysfunction and oxidative stress in 740. Piazza M, Dieckmann T, Guillemette JG. Structural Studies of a Complex Between small mesenteric arteries from diabetic (db/db) mice. Br J Pharmacol 140: 701–706, Endothelial Nitric Oxide Synthase and Calmodulin at Physiological Calcium Concen- 2003. doi:10.1038/sj.bjp.0705476. tration. Biochemistry 55: 5962–5971, 2016. doi:10.1021/acs.biochem.6b00821.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 369 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

741. Piazza M, Taiakina V, Guillemette SR, Guillemette JG, Dieckmann T. Solution struc- 760. Purohit R, Fritz BG, The J, Issaian A, Weichsel A, David CL, Campbell E, Hausrath ture of calmodulin bound to the target peptide of endothelial nitric oxide synthase AC, Rassouli-Taylor L, Garcin ED, Gage MJ, Montfort WR. YC-1 binding to the ␤ phosphorylated at Thr495. Biochemistry 53: 1241–1249, 2014. doi:10.1021/ subunit of soluble guanylyl cyclase overcomes allosteric inhibition by the ␣ subunit. bi401466s. Biochemistry 53: 101–114, 2014. doi:10.1021/bi4015133.

742. Piccoli C, Ria R, Scrima R, Cela O, D’Aprile A, Boffoli D, Falzetti F, Tabilio A, 761. Qin L, Liu X, Sun Q, Fan Z, Xia D, Ding G, Ong HL, Adams D, Gahl WA, Zheng C, Capitanio N. Characterization of mitochondrial and extra-mitochondrial oxygen Qi S, Jin L, Zhang C, Gu L, He J, Deng D, Ambudkar IS, Wang S. Sialin (SLC17A5) consuming reactions in human hematopoietic stem cells. Novel evidence of the functions as a nitrate transporter in the plasma membrane. Proc Natl Acad Sci USA occurrence of NAD(P)H oxidase activity. J Biol Chem 280: 26467–26476, 2005. 109: 13434–13439, 2012. doi:10.1073/pnas.1116633109. doi:10.1074/jbc.M500047200. 762. Quesada IM, Lucero A, Amaya C, Meijles DN, Cifuentes ME, Pagano PJ, Castro C. 743. Pierce GL, Jablonski KL, Walker AE, Seibert SM, DeVan AE, Black SM, Sharma S, Selective inactivation of NADPH oxidase 2 causes regression of vascularization and Seals DR. Tetrahydrobiopterin supplementation enhances carotid artery compliance the size and stability of atherosclerotic plaques. Atherosclerosis 242: 469–475, 2015. doi:10.1016/j.atherosclerosis.2015.08.011. in healthy older men: a pilot study. Am J Hypertens 25: 1050–1054, 2012. doi:10. 1038/ajh.2012.70. 763. Raat NJ, Noguchi AC, Liu VB, Raghavachari N, Liu D, Xu X, Shiva S, Munson PJ, Gladwin MT. Dietary nitrate and nitrite modulate blood and organ nitrite and the 744. Pieske B, Maggioni AP, Lam CSP, Pieske-Kraigher E, Filippatos G, Butler J, Pon- cellular ischemic stress response. Free Radic Biol Med 47: 510–517, 2009. doi:10. ikowski P, Shah SJ, Solomon SD, Scalise AV, Mueller K, Roessig L, Gheorghiade M. 1016/j.freeradbiomed.2009.05.015. Vericiguat in patients with worsening chronic heart failure and preserved ejection fraction: results of the SOluble guanylate Cyclase stimulatoR in heArT failurE pa- 764. Raat NJ, Tabima DM, Specht PA, Tejero J, Champion HC, Kim-Shapiro DB, Baust J, tientS with PRESERVED EF (SOCRATES-PRESERVED) study. Eur Heart J 38: 1119– Mik EG, Hildesheim M, Stasch JP, Becker EM, Truebel H, Gladwin MT. Direct sGC 1127, 2017. doi:10.1093/eurheartj/ehw593. activation bypasses NO scavenging reactions of intravascular free oxy-hemoglobin and limits vasoconstriction. Antioxid Redox Signal 19: 2232–2243, 2013. doi:10.1089/ 745. Pinder AG, Pittaway E, Morris K, James PE. Nitrite directly vasodilates hypoxic ars.2013.5181. vasculature via nitric oxide-dependent and -independent pathways. Br J Pharmacol 157: 1523–1530, 2009. doi:10.1111/j.1476-5381.2009.00340.x. 765. Radi R. Peroxynitrite, a stealthy biological oxidant. J Biol Chem 288: 26464–26472, 2013. doi:10.1074/jbc.R113.472936. 746. Pollock JS, Förstermann U, Mitchell JA, Warner TD, Schmidt HH, Nakane M, Murad F. Purification and characterization of particulate endothelium-derived relaxing fac- 766. Radi R, Beckman JS, Bush KM, Freeman BA. Peroxynitrite-induced membrane lipid tor synthase from cultured and native bovine aortic endothelial cells. Proc Natl Acad peroxidation: the cytotoxic potential of superoxide and nitric oxide. Arch Biochem Sci USA 88: 10480–10484, 1991. doi:10.1073/pnas.88.23.10480. Biophys 288: 481–487, 1991. doi:10.1016/0003-9861(91)90224-7.

747. Porkert M, Sher S, Reddy U, Cheema F, Niessner C, Kolm P, Jones DP, Hooper C, 767. Radi R, Rodriguez M, Castro L, Telleri R. Inhibition of mitochondrial electron trans- Taylor WR, Harrison D, Quyyumi AA. Tetrahydrobiopterin: a novel antihyperten- port by peroxynitrite. Arch Biochem Biophys 308: 89–95, 1994. doi:10.1006/abbi. sive therapy. J Hum Hypertens 22: 401–407, 2008. doi:10.1038/sj.jhh.1002329. 1994.1013.

748. Poss WB, Huecksteadt TP, Panus PC, Freeman BA, Hoidal JR. Regulation of xanthine 768. Rafikov R, Rafikova O, Aggarwal S, Gross C, Sun X, Desai J, Fulton D, Black SM. dehydrogenase and xanthine oxidase activity by hypoxia. Am J Physiol 270: L941– Asymmetric dimethylarginine induces endothelial nitric-oxide synthase mitochon- drial redistribution through the nitration-mediated activation of Akt1. J Biol Chem L946, 1996. 288: 6212–6226, 2013. doi:10.1074/jbc.M112.423269. 749. Pou S, Keaton L, Surichamorn W, Rosen GM. Mechanism of superoxide generation 769. Rahaman MM, Nguyen AT, Miller MP, Hahn SA, Sparacino-Watkins C, Jobbagy S, by neuronal nitric-oxide synthase. J Biol Chem 274: 9573–9580, 1999. doi:10.1074/ Carew NT, Cantu-Medellin N, Wood KC, Baty CJ, Schopfer FJ, Kelley EE, Gladwin jbc.274.14.9573. MT, Martin E, and Straub AC. Cytochrome b5 Reductase 3 Modulates Soluble 750. Pou S, Pou WS, Bredt DS, Snyder SH, Rosen GM. Generation of superoxide by Guanylate Cyclase Redox State and cGMP Signaling. Circulation Research 121: 137– purified brain nitric oxide synthase. J Biol Chem 267: 24173–24176, 1992. 148, 2017. doi:10.1161/CIRCRESAHA.117.310705. 770. Rahaman MM, Straub AC. The emerging roles of somatic globins in cardiovascular 751. Poulos TL. Soluble guanylate cyclase. Curr Opin Struct Biol 16: 736–743, 2006. doi: redox biology and beyond. Redox Biol 1: 405–410, 2013. doi:10.1016/j.redox.2013. 10.1016/j.sbi.2006.09.006. 08.001. 752. Poulos TL, Li H. Nitric oxide synthase and structure-based inhibitor design. Nitric 771. Ramuschkat M, Appelbaum S, Atzler D, Zeller T, Bauer C, Ojeda FM, Sinning CR, Oxide 63: 68–77, 2017. doi:10.1016/j.niox.2016.11.004. Hoffmann B, Lackner KJ, Böger RH, Wild PS, Münzel T, Blankenberg S, Schwedhelm 753. Presta A, Siddhanta U, Wu C, Sennequier N, Huang L, Abu-Soud HM, Erzurum S, E, Schnabel RB; Gutenberg Health Study Investigators. ADMA, subclinical changes Stuehr DJ. Comparative functioning of dihydro- and tetrahydropterins in supporting and atrial fibrillation in the general population. Int J Cardiol 203: 640–646, 2016. doi:10.1016/j.ijcard.2015.05.102. electron transfer, catalysis, and subunit dimerization in inducible nitric oxide syn- thase. Biochemistry 37: 298–310, 1998. doi:10.1021/bi971944c. 772. Ranayhossaini DJ, Rodriguez AI, Sahoo S, Chen BB, Mallampalli RK, Kelley EE, Csanyi G, Gladwin MT, Romero G, Pagano PJ. Selective recapitulation of conserved and 755. Prütz WA. Hypochlorous acid interactions with thiols, nucleotides, DNA, and other nonconserved regions of putative NOXA1 protein activation domain confers iso- biological substrates. Arch Biochem Biophys 332: 110–120, 1996. doi:10.1006/abbi. form-specific inhibition of Nox1 oxidase and attenuation of endothelial cell migra- 1996.0322. tion. J Biol Chem 288: 36437–36450, 2013. doi:10.1074/jbc.M113.521344. 756. Pryde DC, Dalvie D, Hu Q, Jones P, Obach RS, Tran TD. Aldehyde oxidase: an 773. Rasmussen JT, Rasmussen MS, Petersen TE. Cysteines involved in the interconver- enzyme of emerging importance in drug discovery. J Med Chem 53: 8441–8460, sion between dehydrogenase and oxidase forms of bovine xanthine oxidoreductase. 2010. doi:10.1021/jm100888d. J Dairy Sci 83: 499–506, 2000. doi:10.3168/jds.S0022-0302(00)74909-5.

757. Pudlo M, Demougeot C, Girard-Thernier C. Arginase Inhibitors: A Rational Ap- 774. Rassaf T, Flögel U, Drexhage C, Hendgen-Cotta U, Kelm M, Schrader J. Nitrite proach Over One Century. Med Res Rev 37: 475–513, 2017. doi:10.1002/med. reductase function of deoxymyoglobin: oxygen sensor and regulator of cardiac en- 21419. ergetics and function. Circ Res 100: 1749–1754, 2007. doi:10.1161/CIRCRESAHA. 107.152488. 758. Puntarulo S, Cederbaum AI. Production of reactive oxygen species by microsomes enriched in specific human cytochrome P450 enzymes. Free Radic Biol Med 24: 775. Rassaf T, Totzeck M, Hendgen-Cotta UB, Shiva S, Heusch G, Kelm M. Circulating 1324–1330, 1998. doi:10.1016/S0891-5849(97)00463-2. nitrite contributes to cardioprotection by remote ischemic preconditioning. Circ Res 114: 1601–1610, 2014. doi:10.1161/CIRCRESAHA.114.303822. 759. Puppo A, Halliwell B. Formation of hydroxyl radicals from hydrogen peroxide in the presence of iron. Is haemoglobin a biological Fenton reagent? Biochem J 249: 185– 776. Rathore R, Zheng Y-M, Niu C-F, Liu Q-H, Korde A, Ho Y-S, Wang Y-X. Hypoxia 190, 1988. doi:10.1042/bj2490185. activates NADPH oxidase to increase [ROS]i and [Ca2ϩ]i through the mitochondrial

370 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

ROS-PKCepsilon signaling axis in pulmonary artery smooth muscle cells. Free Radic 793. Rodriguez AI, Gangopadhyay A, Kelley EE, Pagano PJ, Zuckerbraun BS, Bauer PM. Biol Med 45: 1223–1231, 2008. doi:10.1016/j.freeradbiomed.2008.06.012. HO-1 and CO decrease platelet-derived growth factor-induced vascular smooth muscle cell migration via inhibition of Nox1. Arterioscler Thromb Vasc Biol 30: 98– 777. Ravi K, Brennan LA, Levic S, Ross PA, Black SM. S-nitrosylation of endothelial nitric 104, 2010. doi:10.1161/ATVBAHA.109.197822. oxide synthase is associated with monomerization and decreased enzyme activity. Proc Natl Acad Sci USA 101: 2619–2624, 2004. doi:10.1073/pnas.0300464101. 794. Rong Z, Wilson MT, Cooper CE. A model for the nitric oxide producing nitrite reductase activity of hemoglobin as a function of oxygen saturation. Nitric Oxide 33: 778. Ray PD, Huang BW, Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox 74–80, 2013. doi:10.1016/j.niox.2013.06.008. regulation in cellular signaling. Cell Signal 24: 981–990, 2012. doi:10.1016/j.cellsig. 2012.01.008. 795. Rose JJ, Wang L, Xu Q, McTiernan CF, Shiva S, Tejero J, Gladwin MT. Carbon Monoxide Poisoning: Pathogenesis, Management, and Future Directions of Therapy. 779. Ray R, Murdoch CE, Wang M, Santos CX, Zhang M, Alom-Ruiz S, Anilkumar N, Am J Respir Crit Care Med 195: 596–606, 2017. doi:10.1164/rccm.201606-1275CI. Ouattara A, Cave AC, Walker SJ, Grieve DJ, Charles RL, Eaton P, Brewer AC, Shah AM. Endothelial Nox4 NADPH oxidase enhances vasodilatation and reduces blood 796. Rosenkranz-Weiss P, Sessa WC, Milstien S, Kaufman S, Watson CA, Pober JS. Reg- pressure in vivo. Arterioscler Thromb Vasc Biol 31: 1368–1376, 2011. doi:10.1161/ ulation of nitric oxide synthesis by proinflammatory cytokines in human umbilical ATVBAHA.110.219238. vein endothelial cells. Elevations in tetrahydrobiopterin levels enhance endothelial nitric oxide synthase specific activity. J Clin Invest 93: 2236–2243, 1994. doi:10.1172/ 780. Reeder BJ, Ukeri J. Strong modulation of nitrite reductase activity of cytoglobin by JCI117221. disulfide bond oxidation: Implications for nitric oxide homeostasis. Nitric Oxide 72: 16–23, 2018. doi:10.1016/j.niox.2017.11.004. 797. Rossitch E Jr, Alexander E III, Black PM, Cooke JP. L-arginine normalizes endothelial function in cerebral vessels from hypercholesterolemic rabbits. J Clin Invest 87: 781. Reule CA, Goyvaerts B, Schoen C. Effects of an L-arginine-based multi ingredient 1295–1299, 1991. doi:10.1172/JCI115132. product on endothelial function in subjects with mild to moderate hypertension and hyperhomocysteinemia - a randomized, double-blind, placebo-controlled, cross- 798. Rouquette M, Page S, Bryant R, Benboubetra M, Stevens CR, Blake DR, Whish WD, over trial. BMC Complement Altern Med 17: 92, 2017. doi:10.1186/s12906-017- Harrison R, Tosh D. Xanthine oxidoreductase is asymmetrically localised on the 1603-9. outer surface of human endothelial and epithelial cells in culture. FEBS Lett 426: 397–401, 1998. doi:10.1016/S0014-5793(98)00385-8. 782. Rey FE, Cifuentes ME, Kiarash A, Quinn MT, Pagano PJ. Novel competitive inhibitor of NAD(P)H oxidase assembly attenuates vascular O(2)(-) and systolic blood pres- 799. Roy B, Mo E, Vernon J, Garthwaite J. Probing the presence of the ligand-binding sure in mice. Circ Res 89: 408–414, 2001. doi:10.1161/hh1701.096037. haem in cellular nitric oxide receptors. Br J Pharmacol 153: 1495–1504, 2008. doi: 10.1038/sj.bjp.0707687. 783. Rezende F, Prior KK, Löwe O, Wittig I, Strecker V, Moll F, Helfinger V, Schnütgen F, Kurrle N, Wempe F, Walter M, Zukunft S, Luck B, Fleming I, Weissmann N, Brandes 800. Rudolph TK, Rudolph V, Edreira MM, Cole MP, Bonacci G, Schopfer FJ, Woodcock RP, Schröder K. Cytochrome P450 enzymes but not NADPH oxidases are the SR, Franek A, Pekarova M, Khoo NKH, Hasty AH, Baldus S, Freeman BA. Nitro-fatty source of the NADPH-dependent lucigenin chemiluminescence in membrane as- acids reduce atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb says. Free Radic Biol Med 102: 57–66, 2017. doi:10.1016/j.freeradbiomed.2016.11. Vasc Biol 30: 938–945, 2010. doi:10.1161/ATVBAHA.109.201582. 019. 801. Rudolph V, Rudolph TK, Schopfer FJ, Bonacci G, Woodcock SR, Cole MP, Baker 784. Risbano MG, Gladwin MT. Therapeutics targeting of dysregulated redox equilibrium PRS, Ramani R, Freeman BA. Endogenous generation and protective effects of nitro- and endothelial dysfunction. Handb Exp Pharmacol 218: 315–349, 2013. doi:10.1007/ fatty acids in a murine model of focal cardiac ischaemia and reperfusion. Cardiovasc 978-3-662-45805-1_13. Res 85: 155–166, 2010. doi:10.1093/cvr/cvp275.

785. Ristow M, Zarse K, Oberbach A, Klöting N, Birringer M, Kiehntopf M, Stumvoll M, 802. Rueckschloss U, Quinn MT, Holtz J, Morawietz H. Dose-dependent regulation of Kahn CR, Blüher M. Antioxidants prevent health-promoting effects of physical ex- NAD(P)H oxidase expression by angiotensin II in human endothelial cells: protective ercise in humans. Proc Natl Acad Sci USA 106: 8665–8670, 2009. doi:10.1073/pnas. effect of angiotensin II type 1 receptor blockade in patients with coronary artery 0903485106. disease. Arterioscler Thromb Vasc Biol 22: 1845–1851, 2002. doi:10.1161/01.ATV. 0000035392.38687.65. 786. Ritchie RH, Drummond GR, Sobey CG, De Silva TM, Kemp-Harper BK. The oppos- ing roles of NO and oxidative stress in cardiovascular disease. Pharmacol Res 116: 803. Ryan MG, Balendran A, Harrison R, Wolstenholme A, Bulkley GB. Xanthine oxi- 57–69, 2017. doi:10.1016/j.phrs.2016.12.017. doreductase: dehydrogenase to oxidase conversion. Biochem Soc Trans 25: 530S, 1997. doi:10.1042/bst025530s. 787. Ritter D, Taylor JF, Hoffmann JW, Carnaghi L, Giddings SJ, Zakeri H, Kwok PY. Alternative splicing for the alpha1 subunit of soluble guanylate cyclase. Biochem J 346: 804. Ryoo S, Gupta G, Benjo A, Lim HK, Camara A, Sikka G, Lim HK, Sohi J, Santhanam 811–816, 2000. doi:10.1042/bj3460811. L, Soucy K, Tuday E, Baraban E, Ilies M, Gerstenblith G, Nyhan D, Shoukas A, Christianson DW, Alp NJ, Champion HC, Huso D, Berkowitz DE. Endothelial argi- 788. Robbins IM, Hemnes AR, Gibbs JS, Christman BW, Howard L, Meehan S, Cabrita I, nase II: a novel target for the treatment of atherosclerosis. Circ Res 102: 923–932, Gonzalez R, Oyler T, Zhao L, Du RH, Mendes LA, Wilkins MR. Safety of sapropterin 2008. doi:10.1161/CIRCRESAHA.107.169573. dihydrochloride (6r-bh4) in patients with pulmonary hypertension. Exp Lung Res 37: 26–34, 2011. doi:10.3109/01902148.2010.512972. 805. Ryter SW, Alam J, Choi AMK. Heme oxygenase-1/carbon monoxide: from basic science to therapeutic applications. Physiol Rev 86: 583–650, 2006. doi:10.1152/ 789. Rocha BS, Nunes C, Pereira C, Barbosa RM, Laranjinha J. A shortcut to wide-ranging physrev.00011.2005. biological actions of dietary polyphenols: modulation of the nitrate-nitrite-nitric ox- ide pathway in the gut. Food Funct 5: 1646–1652, 2014. doi:10.1039/C4FO00124A. 806. Ryter SW, Morse D, Choi AMK. Carbon monoxide and bilirubin: potential therapies for pulmonary/vascular injury and disease. Am J Respir Cell Mol Biol 36: 175–182, 790. Rochette L, Lorin J, Zeller M, Guilland JC, Lorgis L, Cottin Y, Vergely C. Nitric oxide 2007. doi:10.1165/rcmb.2006-0333TR. synthase inhibition and oxidative stress in cardiovascular diseases: possible therapeu- tic targets? Pharmacol Ther 140: 239–257, 2013. doi:10.1016/j.pharmthera.2013.07. 807. Ryter SW, Otterbein LE, Morse D, Choi AM. Heme oxygenase/carbon monoxide 004. signaling pathways: regulation and functional significance. Mol Cell Biochem 234-235: 249–263, 2002. doi:10.1023/A:1015957026924. 791. Rodriguez-Mateos A, Hezel M, Aydin H, Kelm M, Lundberg JO, Weitzberg E, Spen- cer JP, Heiss C. Interactions between cocoa flavanols and inorganic nitrate: additive 808. Saha A, Goldstein S, Cabelli D, Czapski G. Determination of optimal conditions for effects on endothelial function at achievable dietary amounts. Free Radic Biol Med 80: synthesis of peroxynitrite by mixing acidified hydrogen peroxide with nitrite. Free 121–128, 2015. doi:10.1016/j.freeradbiomed.2014.12.009. Radic Biol Med 24: 653–659, 1998. doi:10.1016/S0891-5849(97)00365-1.

792. Rodríguez AI, Csányi G, Ranayhossaini DJ, Feck DM, Blose KJ, Assatourian L, Vorp 809. Saha S, Chakraborty PK, Xiong X, Dwivedi SK, Mustafi SB, Leigh NR, Ramchandran DA, Pagano PJ. MEF2B-Nox1 signaling is critical for stretch-induced phenotypic R, Mukherjee P, Bhattacharya R. Cystathionine ␤-synthase regulates endothelial modulation of vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 35: 430– function via protein S-sulfhydration. FASEB J 30: 441–456, 2016. doi:10.1096/fj.15- 438, 2015. doi:10.1161/ATVBAHA.114.304936. 278648.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 371 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

810. Sakellariou GK, Pearson T, Lightfoot AP, Nye GA, Wells N, Giakoumaki II, Vasilaki A, 827. Schindler U, Strobel H, Schönafinger K, Linz W, Löhn M, Martorana PA, Rütten H, Griffiths RD, Jackson MJ, McArdle A. Mitochondrial ROS regulate oxidative damage Schindler PW, Busch AE, Sohn M, Töpfer A, Pistorius A, Jannek C, Mülsch A. Bio- and mitophagy but not age-related muscle fiber atrophy. Sci Rep 6: 33944, 2016. chemistry and pharmacology of novel anthranilic acid derivatives activating heme- doi:10.1038/srep33944. oxidized soluble guanylyl cyclase. Mol Pharmacol 69: 1260–1268, 2006. doi:10.1124/ mol.105.018747. 811. Salerno JC. Neuronal nitric oxide synthase: prototype for pulsed enzymology. FEBS Lett 582: 1395–1399, 2008. doi:10.1016/j.febslet.2008.03.051. 828. Schmidt TS, Alp NJ. Mechanisms for the role of tetrahydrobiopterin in endothelial function and vascular disease. Clin Sci (Lond) 113: 47–63, 2007. doi:10.1042/ 812. Salerno JC, Ghosh DK. Space, time and nitric oxide–neuronal nitric oxide synthase CS20070108. generates signal pulses. FEBS J 276: 6677–6688, 2009. doi:10.1111/j.1742-4658. 2009.07382.x. 829. Schnabel R, Blankenberg S, Lubos E, Lackner KJ, Rupprecht HJ, Espinola-Klein C, Jachmann N, Post F, Peetz D, Bickel C, Cambien F, Tiret L, Münzel T. Asymmetric 813. Salgado MT, Cao Z, Nagababu E, Mohanty JG, Rifkind JM. Red blood cell membrane- dimethylarginine and the risk of cardiovascular events and death in patients with facilitated release of nitrite-derived nitric oxide bioactivity. Biochemistry 54: 6712– coronary artery disease: results from the AtheroGene Study. Circ Res 97: e53–e59, 6723, 2015. doi:10.1021/acs.biochem.5b00643. 2005. doi:10.1161/01.RES.0000181286.44222.61.

814. Sam F, Kerstetter DL, Pimental DR, Mulukutla S, Tabaee A, Bristow MR, Colucci 830. Schopfer FJ, Cipollina C, Freeman BA. Formation and signaling actions of electro- WS, Sawyer DB. Increased reactive oxygen species production and functional alter- philic lipids. Chem Rev 111: 5997–6021, 2011. doi:10.1021/cr200131e. ations in antioxidant enzymes in human failing myocardium. J Card Fail 11: 473–480, 2005. doi:10.1016/j.cardfail.2005.01.007. 831. Schröder H, Leitman DC, Bennett BM, Waldman SA, Murad F. Glyceryl trinitrate- induced desensitization of guanylate cyclase in cultured rat lung fibroblasts. J Phar- 815. Sánchez-Gómez FJ, Calvo E, Bretón-Romero R, Fierro-Fernández M, Anilkumar N, macol Exp Ther 245: 413–418, 1988. Shah AM, Schröder K, Brandes RP, Vázquez J, Lamas S. NOX4-dependent Hydro- gen peroxide promotes shear stress-induced SHP2 sulfenylation and eNOS activa- 832. Schröder K, Zhang M, Benkhoff S, Mieth A, Pliquett R, Kosowski J, Kruse C, Luedike tion. Free Radic Biol Med 89: 419–430, 2015. doi:10.1016/j.freeradbiomed.2015.08. P, Michaelis UR, Weissmann N, Dimmeler S, Shah AM, Brandes RP. Nox4 is a 014. protective reactive oxygen species generating vascular NADPH oxidase. Circ Res 110: 1217–1225, 2012. doi:10.1161/CIRCRESAHA.112.267054. 816. Santolini J, Meade AL, Stuehr DJ. Differences in three kinetic parameters underpin the unique catalytic profiles of nitric-oxide synthases I, II, and III. J Biol Chem 276: 833. Schröder K, Vecchione C, Jung O, Schreiber JG, Shiri-Sverdlov R, van Gorp PJ, Busse 48887–48898, 2001. doi:10.1074/jbc.M108666200. R, Brandes RP. Xanthine oxidase inhibitor tungsten prevents the development of atherosclerosis in ApoE knockout mice fed a Western-type diet. Free Radic Biol Med 817. Santos CXC, Tanaka LY, Wosniak J Jr, Laurindo FRM. Mechanisms and implications 41: 1353–1360, 2006. doi:10.1016/j.freeradbiomed.2006.03.026. of reactive oxygen species generation during the unfolded protein response: roles of 834. Schroeter H, Heiss C, Balzer J, Kleinbongard P, Keen CL, Hollenberg NK, Sies H, endoplasmic reticulum oxidoreductases, mitochondrial electron transport, and NA- Kwik-Uribe C, HH, Kelm M. (-)-Epicatechin mediates beneficial effects of DPH oxidase. Antioxid Redox Signal 11: 2409–2427, 2009. doi:10.1089/ars.2009. flavanol-rich cocoa on vascular function in humans. Proc Natl Acad Sci USA 103: 2625. 1024–1029, 2006. doi:10.1073/pnas.0510168103. 818. Sarkar A, Dai Y, Haque MM, Seeger F, Ghosh A, Garcin ED, Montfort WR, Hazen SL, 835. Schulman SP, Becker LC, Kass DA, Champion HC, Terrin ML, Forman S, Ernst KV, Misra S, Stuehr DJ. Heat Shock Protein 90 Associates with the Per-Arnt-Sim Domain Kelemen MD, Townsend SN, Capriotti A, Hare JM, Gerstenblith G. L-arginine ther- of Heme-free Soluble Guanylate Cyclase: Implications for Enzyme Maturation. J Biol apy in acute myocardial infarction: the Vascular Interaction With Age in Myocardial Chem 290: 21615–21628, 2015. doi:10.1074/jbc.M115.645515. Infarction (VINTAGE MI) randomized clinical trial. JAMA 295: 58–64, 2006. doi:10. 819. Sase K, Michel T. Expression of constitutive endothelial nitric oxide synthase in 1001/jama.295.1.58. human blood platelets. Life Sci 57: 2049–2055, 1995. doi:10.1016/0024- 836. Schulz E, Wenzel P, Münzel T, Daiber A. Mitochondrial redox signaling: Interaction 3205(95)02191-K. of mitochondrial reactive oxygen species with other sources of oxidative stress. 820. Sato N, Takasaka N, Yoshida M, Tsubouchi K, Minagawa S, Araya J, Saito N, Fujita Y, Antioxid Redox Signal 20: 308–324, 2014. doi:10.1089/ars.2012.4609. Kurita Y, Kobayashi K, Ito S, Hara H, Kadota T, Yanagisawa H, Hashimoto M, Utsumi 837. Schumacher HR Jr, Becker MA, Wortmann RL, Macdonald PA, Hunt B, Streit J, H, Wakui H, Kojima J, Numata T, Kaneko Y, Odaka M, Morikawa T, Nakayama K, Lademacher C, Joseph-Ridge N. Effects of febuxostat versus allopurinol and placebo Kohrogi H, Kuwano K. Metformin attenuates lung fibrosis development via NOX4 in reducing serum urate in subjects with hyperuricemia and gout: a 28-week, phase suppression. Respir Res 17: 107, 2016. doi:10.1186/s12931-016-0420-x. III, randomized, double-blind, parallel-group trial. Arthritis Rheum 59: 1540–1548, 2008. doi:10.1002/art.24209. 821. Sato T, Ashizawa N, Matsumoto K, Iwanaga T, Nakamura H, Inoue T, Nagata O. Discovery of 3-(2-cyano-4-pyridyl)-5-(4-pyridyl)-1,2,4-triazole, FYX-051 - a xan- 838. Schürmann C, Rezende F, Kruse C, Yasar Y, Löwe O, Fork C, van de Sluis B, Bremer thine oxidoreductase inhibitor for the treatment of hyperuricemia. Corrigendum at: R, Weissmann N, Shah AM, Jo H, Brandes RP, Schröder K. The NADPH oxidase doi:10.1016/j.bmcl.2010.02.026. Bioorg Med Chem Lett 19: 6225–6229, 2009. doi: Nox4 has anti-atherosclerotic functions. Eur Heart J 36: 3447–3456, 2015. doi:10. 10.1016/j.bmcl.2009.08.091. 1093/eurheartj/ehv460.

822. Sayed N, Baskaran P, Ma X, van den Akker F, Beuve A. Desensitization of soluble 839. Schwarz G. Molybdenum cofactor and human disease. Curr Opin Chem Biol 31: guanylyl cyclase, the NO receptor, by S-nitrosylation. Proc Natl Acad Sci USA 104: 179–187, 2016. doi:10.1016/j.cbpa.2016.03.016. 12312–12317, 2007. doi:10.1073/pnas.0703944104. 840. Schwarz G, Mendel RR, Ribbe MW. Molybdenum cofactors, enzymes and pathways. 823. Sayed N, Kim DD, Fioramonti X, Iwahashi T, Durán WN, Beuve A. Nitroglycerin- Nature 460: 839–847, 2009. doi:10.1038/nature08302. induced S-nitrosylation and desensitization of soluble guanylyl cyclase contribute to nitrate tolerance. Circ Res 103: 606–614, 2008. doi:10.1161/CIRCRESAHA.108. 841. Schwarz PM, Kleinert H, Förstermann U. Potential functional significance of brain- 175133. type and muscle-type nitric oxide synthase I expressed in adventitia and media of rat aorta. Arterioscler Thromb Vasc Biol 19: 2584–2590, 1999. doi:10.1161/01.ATV.19. 824. Schewe T, Steffen Y, Sies H. How do dietary flavanols improve vascular function? A 11.2584. position paper. Arch Biochem Biophys 476: 102–106, 2008. doi:10.1016/j.abb.2008. 03.004. 842. Schwedhelm E, Maas R, Freese R, Jung D, Lukacs Z, Jambrecina A, Spickler W, Schulze F, Böger RH. Pharmacokinetic and pharmacodynamic properties of oral 825. Schieber M, Chandel NS. ROS function in redox signaling and oxidative stress. Curr L-citrulline and L-arginine: impact on nitric oxide metabolism. Br J Clin Pharmacol 65: Biol 24: R453–R462, 2014. doi:10.1016/j.cub.2014.03.034. 51–59, 2008. doi:10.1111/j.1365-2125.2007.02990.x.

826. Schildknecht S, Weber A, Gerding HR, Pape R, Robotta M, Drescher M, Marquardt 843. Seddon M, Melikian N, Dworakowski R, Shabeeh H, Jiang B, Byrne J, Casadei B, A, Daiber A, Ferger B, Leist M. The NOX1/4 inhibitor GKT136901 as selective and Chowienczyk P, Shah AM. Effects of neuronal nitric oxide synthase on human cor- direct scavenger of peroxynitrite. Curr Med Chem 21: 365–376, 2014. doi:10.2174/ onary artery diameter and blood flow in vivo. Circulation 119: 2656–2662, 2009. 09298673113209990179. doi:10.1161/CIRCULATIONAHA.108.822205.

372 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

844. Seddon MD, Chowienczyk PJ, Brett SE, Casadei B, Shah AM. Neuronal nitric oxide 861. Shiva S, Gladwin MT. Nitrite mediates cytoprotection after ischemia/reperfusion by synthase regulates basal microvascular tone in humans in vivo. Circulation 117: 1991– modulating mitochondrial function. Basic Res Cardiol 104: 113–119, 2009. doi:10. 1996, 2008. doi:10.1161/CIRCULATIONAHA.107.744540. 1007/s00395-009-0009-3.

845. Segal AW, Jones OTG. Novel cytochrome b system in phagocytic vacuoles of human 862. Shiva S, Huang Z, Grubina R, Sun J, Ringwood LA, MacArthur PH, Xu X, Murphy E, granulocytes. Nature 276: 515–517, 1978. doi:10.1038/276515a0. Darley-Usmar VM, Gladwin MT. Deoxymyoglobin is a nitrite reductase that gener- ates nitric oxide and regulates mitochondrial respiration. Circ Res 100: 654–661, 846. Selemidis S, Dusting GJ, Peshavariya H, Kemp-Harper BK, Drummond GR. Nitric 2007. doi:10.1161/01.RES.0000260171.52224.6b. oxide suppresses NADPH oxidase-dependent superoxide production by S-nitrosy- lation in human endothelial cells. Cardiovasc Res 75: 349–358, 2007. doi:10.1016/j. 863. Shiva S, Oh JY, Landar AL, Ulasova E, Venkatraman A, Bailey SM, Darley-Usmar VM. cardiores.2007.03.030. Nitroxia: the pathological consequence of dysfunction in the nitric oxide-cyto- chrome c oxidase signaling pathway. Free Radic Biol Med 38: 297–306, 2005. doi:10. 847. Seredenina T, Chiriano G, Filippova A, Nayernia Z, Mahiout Z, Fioraso-Cartier L, 1016/j.freeradbiomed.2004.10.037. Plastre O, Scapozza L, Krause KH, Jaquet V. A subset of N-substituted phenothia- zines inhibits NADPH oxidases. Free Radic Biol Med 86: 239–249, 2015. doi:10.1016/ 864. Shiva S, Sack MN, Greer JJ, Duranski M, Ringwood LA, Burwell L, Wang X, j.freeradbiomed.2015.05.023. MacArthur PH, Shoja A, Raghavachari N, Calvert JW, Brookes PS, Lefer DJ, Gladwin MT. Nitrite augments tolerance to ischemia/reperfusion injury via the 848. Serrander L, Jaquet V, Bedard K, Plastre O, Hartley O, Arnaudeau S, Demaurex N, modulation of mitochondrial electron transfer. J Exp Med 204: 2089–2102, 2007. Schlegel W, Krause K-H. NOX5 is expressed at the plasma membrane and generates doi:10.1084/jem.20070198. superoxide in response to protein kinase C activation. Biochimie 89: 1159–1167, 2007. doi:10.1016/j.biochi.2007.05.004. 865. Shiva S, Wang X, Ringwood LA, Xu X, Yuditskaya S, Annavajjhala V, Miyajima H, Hogg N, Harris ZL, Gladwin MT. Ceruloplasmin is a NO oxidase and nitrite synthase 849. Setoguchi S, Hirooka Y, Eshima K, Shimokawa H, Takeshita A. Tetrahydrobiopterin that determines endocrine NO homeostasis. Nat Chem Biol 2: 486–493, 2006. improves impaired endothelium-dependent forearm vasodilation in patients with doi:10.1038/nchembio813. heart failure. J Cardiovasc Pharmacol 39: 363–368, 2002. doi:10.1097/00005344- 200203000-00007. 866. Singel DJ, Stamler JS. Chemical physiology of blood flow regulation by red blood cells: the role of nitric oxide and S-nitrosohemoglobin. Annu Rev Physiol 67: 99–145, 850. Setoguchi S, Mohri M, Shimokawa H, Takeshita A. Tetrahydrobiopterin improves 2005. doi:10.1146/annurev.physiol.67.060603.090918. endothelial dysfunction in coronary microcirculation in patients without epicardial coronary artery disease. J Am Coll Cardiol 38: 493–498, 2001. doi:10.1016/S0735- 867. Singh K, Balligand JL, Fischer TA, Smith TW, Kelly RA. Glucocorticoids increase 1097(01)01382-1. osteopontin expression in cardiac myocytes and microvascular endothelial cells. 851. Sevanian A, Nordenbrand K, Kim E, Ernster L, Hochstein P. Microsomal lipid per- Role in regulation of inducible nitric oxide synthase. J Biol Chem 270: 28471–28478, oxidation: the role of NADPH–cytochrome P450 reductase and cytochrome P450. 1995. doi:10.1074/jbc.270.47.28471. Free Radic Biol Med 8: 145–152, 1990. doi:10.1016/0891-5849(90)90087-Y. 868. Singh S, Madzelan P, Banerjee R. Properties of an unusual heme cofactor in PLP- 852. Sharina IG, Jelen F, Bogatenkova EP, Thomas A, Martin E, Murad F. Alpha1 soluble dependent cystathionine beta-synthase. Nat Prod Rep 24: 631–639, 2007. doi:10. guanylyl cyclase (sGC) splice forms as potential regulators of human sGC activity. J 1039/B604182P. Biol Chem 283: 15104–15113, 2008. doi:10.1074/jbc.M710269200. 869. Siragusa M, Fisslthaler B. Insulin Keeps PYK-ing on eNOS: Enhanced Insulin Receptor 853. Sharina IG, Martin E. The Role of Reactive Oxygen and Nitrogen Species in the Signaling Induces Endothelial Dysfunction. Circ Res 120: 748–750, 2017. doi:10. Expression and Splicing of Nitric Oxide Receptor. Antioxid Redox Signal 26: 122–136, 1161/CIRCRESAHA.117.310576. 2017. doi:10.1089/ars.2016.6687. 870. Siragusa M, Fleming I. The eNOS signalosome and its link to endothelial dysfunction. 854. Sheehan AL, Carrell S, Johnson B, Stanic B, Banfi B, Miller FJ Jr. Role for Nox1 Pflugers Arch 468: 1125–1137, 2016. doi:10.1007/s00424-016-1839-0. NADPH oxidase in atherosclerosis. Atherosclerosis 216: 321–326, 2011. doi:10. 871. Sirokmány G, Donkó Á, Geiszt M. Nox/Duox Family of NADPH Oxidases: Lessons 1016/j.atherosclerosis.2011.02.028. from Knockout Mouse Models. Trends Pharmacol Sci 37: 318–327, 2016. doi:10. 855. Shemyakin A, Kövamees O, Rafnsson A, Böhm F, Svenarud P, Settergren M, Jung C, 1016/j.tips.2016.01.006. Pernow J. Arginase inhibition improves endothelial function in patients with coronary artery disease and type 2 diabetes mellitus. Circulation 126: 2943–2950, 2012. doi: 872. Smith BC, Marletta MA. Mechanisms of S-nitrosothiol formation and selectivity in 10.1161/CIRCULATIONAHA.112.140335. nitric oxide signaling. Curr Opin Chem Biol 16: 498–506, 2012. doi:10.1016/j.cbpa. 2012.10.016. 856. Shin HK, Kim YK, Kim KY, Lee JH, Hong KW. Remnant lipoprotein particles induce apoptosis in endothelial cells by NAD(P)H oxidase-mediated production of super- 873. Smith RA, Hartley RC, Murphy MP. Mitochondria-targeted small molecule thera- oxide and cytokines via lectin-like oxidized low-density lipoprotein receptor-1 acti- peutics and probes. Antioxid Redox Signal 15: 3021–3038, 2011. doi:10.1089/ars. vation: prevention by cilostazol. Circulation 109: 1022–1028, 2004. doi:10.1161/01. 2011.3969. CIR.0000117403.64398.53. 874. Snow BJ, Rolfe FL, Lockhart MM, Frampton CM, O’Sullivan JD, Fung V, Smith RA, 857. Shin S, Thapa SK, Fung HL. Cellular interactions between L-arginine and asymmetric Murphy MP, Taylor KM; Protect Study Group. A double-blind, placebo-controlled dimethylarginine: Transport and metabolism. PLoS One 12: e0178710, 2017. doi:10. study to assess the mitochondria-targeted antioxidant MitoQ as a disease-modifying 1371/journal.pone.0178710. therapy in Parkinson’s disease. Mov Disord 25: 1670–1674, 2010. doi:10.1002/mds. 23148. 858. Shinozaki K, Kashiwagi A, Nishio Y, Okamura T, Yoshida Y, Masada M, Toda N, Kikkawa R. Abnormal biopterin metabolism is a major cause of impaired endotheli- 875. Sobey CG, Judkins CP, Rivera J, Lewis CV, Diep H, Lee HW, Kemp-Harper BK, um-dependent relaxation through nitric oxide/O2- imbalance in insulin-resistant rat Broughton BR, Selemidis S, Gaspari TA, Samuel CS, Drummond GR. NOX1 defi- aorta. Diabetes 48: 2437–2445, 1999. doi:10.2337/diabetes.48.12.2437. ciency in apolipoprotein E-knockout mice is associated with elevated plasma lipids and enhanced atherosclerosis. Free Radic Res 49: 186–198, 2015. doi:10.3109/ 859. Shirakura T, Nomura J, Matsui C, Kobayashi T, Tamura M, Masuzaki H. Febuxostat, 10715762.2014.992893. a novel xanthine oxidoreductase inhibitor, improves hypertension and endothelial dysfunction in spontaneously hypertensive rats. Naunyn Schmiedebergs Arch Pharma- 876. Sorescu D, Weiss D, Lassègue B, Clempus RE, Szöcs K, Sorescu GP, Valppu L, Quinn col 389: 831–838, 2016. doi:10.1007/s00210-016-1239-1. MT, Lambeth JD, Vega JD, Taylor WR, Griendling KK. Superoxide production and expression of nox family proteins in human atherosclerosis. Circulation 105: 1429– 860. Shirodaria C, Antoniades C, Lee J, Jackson CE, Robson MD, Francis JM, Moat SJ, 1435, 2002. doi:10.1161/01.CIR.0000012917.74432.66. Ratnatunga C, Pillai R, Refsum H, Neubauer S, Channon KM. Global improvement of vascular function and redox state with low-dose folic acid: implications for folate 877. Souza JM, Daikhin E, Yudkoff M, Raman CS, Ischiropoulos H. Factors determining therapy in patients with coronary artery disease. Circulation 115: 2262–2270, 2007. the selectivity of protein tyrosine nitration. Arch Biochem Biophys 371: 169–178, doi:10.1161/CIRCULATIONAHA.106.679084. 1999. doi:10.1006/abbi.1999.1480.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 373 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

878. Sowa G, Pypaert M, Sessa WC. Distinction between signaling mechanisms in lipid 895. Stone JR, Marletta MA. Soluble guanylate cyclase from bovine lung: activation with rafts vs. caveolae. Proc Natl Acad Sci USA 98: 14072–14077, 2001. doi:10.1073/pnas. nitric oxide and carbon monoxide and spectral characterization of the ferrous and 241409998. ferric states. Biochemistry 33: 5636–5640, 1994. doi:10.1021/bi00184a036.

879. Spallarossa P, Altieri P, Garibaldi S, Ghigliotti G, Barisione C, Manca V, Fabbi P, 896. Stone JR, Marletta MA. Synergistic activation of soluble guanylate cyclase by YC-1 Ballestrero A, Brunelli C, Barsotti A. Matrix metalloproteinase-2 and -9 are induced and carbon monoxide: implications for the role of cleavage of the iron-histidine bond differently by doxorubicin in H9c2 cells: The role of MAP kinases and NAD(P)H during activation by nitric oxide. Chem Biol 5: 255–261, 1998. doi:10.1016/S1074- oxidase. Cardiovasc Res 69: 736–745, 2006. doi:10.1016/j.cardiores.2005.08.009. 5521(98)90618-4.

880. Sparacino-Watkins CE, Tejero J, Sun B, Gauthier MC, Thomas J, Ragireddy V, Mer- 897. Stover JF, Belli A, Boret H, Bulters D, Sahuquillo J, Schmutzhard E, Zavala E, Unger- chant BA, Wang J, Azarov I, Basu P, Gladwin MT. Nitrite reductase and nitric-oxide stedt U, Schinzel R, Tegtmeier F; NOSTRA Investigators. Nitric oxide synthase synthase activity of the mitochondrial molybdopterin enzymes mARC1 and mARC2. inhibition with the antipterin VAS203 improves outcome in moderate and severe J Biol Chem 289: 10345–10358, 2014. doi:10.1074/jbc.M114.555177. traumatic brain injury: a placebo-controlled randomized Phase IIa trial (NOSTRA). J Neurotrauma 31: 1599–1606, 2014. doi:10.1089/neu.2014.3344. 881. Spickett CM, Jerlich A, Panasenko OM, Arnhold J, Pitt AR, Stelmaszyn´ska T, Schaur RJ. The reactions of hypochlorous acid, the reactive oxygen species produced by 898. Straub AC, Lohman AW, Billaud M, Johnstone SR, Dwyer ST, Lee MY, Bortz PS, Best myeloperoxidase, with lipids. Acta Biochim Pol 47: 889–899, 2000. AK, Columbus L, Gaston B, Isakson BE. Endothelial cell expression of haemoglobin ␣ regulates nitric oxide signalling. Nature 491: 473–477, 2012. doi:10.1038/ 882. Sprague RS, Stephenson AH, Ellsworth ML. Red not dead: signaling in and from nature11626. erythrocytes. Trends Endocrinol Metab 18: 350–355, 2007. doi:10.1016/j.tem.2007. 08.008. 899. Stroes E, Kastelein J, Cosentino F, Erkelens W, Wever R, Koomans H, Lüscher T, Rabelink T. Tetrahydrobiopterin restores endothelial function in hypercholesterol- 883. Srihirun S, Sriwantana T, Unchern S, Kittikool D, Noulsri E, Pattanapanyasat K, emia. J Clin Invest 99: 41–46, 1997. doi:10.1172/JCI119131. Fucharoen S, Piknova B, Schechter AN, Sibmooh N. Platelet inhibition by nitrite is dependent on erythrocytes and deoxygenation. PLoS One 7: e30380, 2012. doi:10. 900. Stroes ES, van Faassen EE, Yo M, Martasek P, Boer P, Govers R, Rabelink TJ. Folic 1371/journal.pone.0030380. acid reverts dysfunction of endothelial nitric oxide synthase. Circ Res 86: 1129–1134, 2000. doi:10.1161/01.RES.86.11.1129. 884. St Hilaire C, Koupenova M, Carroll SH, Smith BD, Ravid K. TNF-alpha upregulates the A2B adenosine receptor gene: The role of NAD(P)H oxidase 4. Biochem Biophys 901. Stuehr D, Pou S, Rosen GM. Oxygen reduction by nitric-oxide synthases. J Biol Chem Res Commun 375: 292–296, 2008. doi:10.1016/j.bbrc.2008.07.059. 276: 14533–14536, 2001. doi:10.1074/jbc.R100011200.

885. Stamler JS, Jaraki O, Osborne J, Simon DI, Keaney J, Vita J, Singel D, Valeri CR, 902. Stuehr DJ, Abu-Soud HM, Rousseau DL, Feldman PL, Wang J. Control of electron Loscalzo J. Nitric oxide circulates in mammalian plasma primarily as an S-nitroso transfer in neuronal nitric oxide synthase by calmodulin, substrate, substrate analogs, adduct of serum albumin. Proc Natl Acad Sci USA 89: 7674–7677, 1992. doi:10.1073/ and nitric oxide. Adv Pharmacol 34: 207–213, 1995. doi:10.1016/S1054- pnas.89.16.7674. 3589(08)61087-X.

886. Stamler JS, Jia L, Eu JP, McMahon TJ, Demchenko IT, Bonaventura J, Gernert K, 903. Stuehr DJ, Cho HJ, Kwon NS, Weise MF, Nathan CF. Purification and characteriza- Piantadosi CA. Blood flow regulation by S-nitrosohemoglobin in the physiological tion of the cytokine-induced macrophage nitric oxide synthase: an FAD- and FMN- oxygen gradient. Science 276: 2034–2037, 1997. doi:10.1126/science.276.5321. containing flavoprotein. Proc Natl Acad Sci USA 88: 7773–7777, 1991. doi:10.1073/ 2034. pnas.88.17.7773.

887. Starkov AA, Fiskum G, Chinopoulos C, Lorenzo BJ, Browne SE, Patel MS, Beal MF. 904. Stuehr DJ, Santolini J, Wang ZQ, Wei CC, Adak S. Update on mechanism and Mitochondrial alpha-ketoglutarate dehydrogenase complex generates reactive oxy- catalytic regulation in the NO synthases. J Biol Chem 279: 36167–36170, 2004. gen species. J Neurosci 24: 7779–7788, 2004. doi:10.1523/JNEUROSCI.1899-04. doi:10.1074/jbc.R400017200. 2004. 905. Sturms R, DiSpirito AA, Hargrove MS. Plant and cyanobacterial hemoglobins reduce 888. Stasch J-P, Pacher P, Evgenov OV. Soluble guanylate cyclase as an emerging thera- nitrite to nitric oxide under anoxic conditions. Biochemistry 50: 3873–3878, 2011. peutic target in cardiopulmonary disease. Circulation 123: 2263–2273, 2011. doi:10. doi:10.1021/bi2004312. 1161/CIRCULATIONAHA.110.981738. 906. Sturrock A, Cahill B, Norman K, Huecksteadt TP, Hill K, Sanders K, Karwande SV, 889. Stasch JP, Dembowsky K, Perzborn E, Stahl E, Schramm M. Cardiovascular actions Stringham JC, Bull DA, Gleich M, Kennedy TP, Hoidal JR. Transforming growth of a novel NO-independent guanylyl cyclase stimulator, BAY 41-8543: in vivo stud- factor-beta1 induces Nox4 NAD(P)H oxidase and reactive oxygen species-depen- ies. Br J Pharmacol 135: 344–355, 2002. doi:10.1038/sj.bjp.0704483. dent proliferation in human pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 290: L661–L673, 2006. doi:10.1152/ajplung.00269.2005. 890. Stasch JP, Schmidt P, Alonso-Alija C, Apeler H, Dembowsky K, Haerter M, Heil M, Minuth T, Perzborn E, Pleiss U, Schramm M, Schroeder W, Schröder H, Stahl E, 907. Subramani J, Kundumani-Sridharan V, Hilgers RH, Owens C, Das KC. Thioredoxin Steinke W, Wunder F. NO- and haem-independent activation of soluble guanylyl Uses a GSH-independent Route to Deglutathionylate Endothelial Nitric-oxide Syn- cyclase: molecular basis and cardiovascular implications of a new pharmacological thase and Protect against Myocardial Infarction. J Biol Chem 291: 23374–23389, principle. Br J Pharmacol 136: 773–783, 2002. doi:10.1038/sj.bjp.0704778. 2016. doi:10.1074/jbc.M116.745034.

891. Stasch JP, Schmidt PM, Nedvetsky PI, Nedvetskaya TY, H S AK, Meurer S, Deile M, 908. Sugamura K, Keaney JF Jr. Reactive oxygen species in cardiovascular disease. Free Taye A, Knorr A, Lapp H, Müller H, Turgay Y, Rothkegel C, Tersteegen A, Kemp- Radic Biol Med 51: 978–992, 2011. doi:10.1016/j.freeradbiomed.2011.05.004. Harper B, Müller-Esterl W, Schmidt HH. Targeting the heme-oxidized nitric oxide receptor for selective vasodilatation of diseased blood vessels. J Clin Invest 116: 909. Suh YA, Arnold RS, Lassegue B, Shi J, Xu X, Sorescu D, Chung AB, Griendling KK, 2552–2561, 2006. doi:10.1172/JCI28371. Lambeth JD. Cell transformation by the superoxide-generating oxidase Mox1. Na- ture 401: 79–82, 1999. doi:10.1038/43459. 892. Stielow C, Catar RA, Muller G, Wingler K, Scheurer P, Schmidt HH, Morawietz H. Novel Nox inhibitor of oxLDL-induced reactive oxygen species formation in human 910. Sukumar P, Viswambharan H, Imrie H, Cubbon RM, Yuldasheva N, Gage M, Gallo- endothelial cells. Biochem Biophys Res Commun 344: 200–205, 2006. doi:10.1016/j. way S, Skromna A, Kandavelu P, Santos CX, Gatenby VK, Smith J, Beech DJ, Wheat- bbrc.2006.03.114. croft SB, Channon KM, Shah AM, Kearney MT. Nox2 NADPH oxidase has a critical role in insulin resistance-related endothelial cell dysfunction. Diabetes 62: 2130– 893. Stocker R, Keaney JF Jr. Role of oxidative modifications in atherosclerosis. Physiol Rev 2134, 2013. doi:10.2337/db12-1294. 84: 1381–1478, 2004. doi:10.1152/physrev.00047.2003. 911. Suliman HB, Carraway MS, Welty-Wolf KE, Whorton AR, Piantadosi CA. Lipopoly- 894. Stolk J, Hiltermann TJ, Dijkman JH, Verhoeven AJ. Characteristics of the inhibition of saccharide stimulates mitochondrial biogenesis via activation of nuclear respira- NADPH oxidase activation in neutrophils by apocynin, a methoxy-substituted cate- tory factor-1. J Biol Chem 278: 41510–41518, 2003. doi:10.1074/jbc. chol. Am J Respir Cell Mol Biol 11: 95–102, 1994. doi:10.1165/ajrcmb.11.1.8018341. M304719200.

374 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

912. Sumimoto H. Structure, regulation and evolution of Nox-family NADPH oxidases flavocytochrome b: characterization of six monoclonal antibodies to the p22phox that produce reactive oxygen species. FEBS J 275: 3249–3277, 2008. doi:10.1111/j. subunit. J Immunol 173: 7349–7357, 2004. doi:10.4049/jimmunol.173.12.7349. 1742-4658.2008.06488.x. 931. Tejero J, Gladwin MT. The globin superfamily: functions in nitric oxide formation and 913. Sun D, Huang A, Smith CJ, Stackpole CJ, Connetta JA, Shesely EG, Koller A, Kaley G. decay. Biol Chem 395: 631–639, 2014. doi:10.1515/hsz-2013-0289. Enhanced release of prostaglandins contributes to flow-induced arteriolar dilation in eNOS knockout mice. Circ Res 85: 288–293, 1999. doi:10.1161/01.RES.85.3.288. 932. Tejero J, Sparacino-Watkins CE, Ragireddy V, Frizzell S, Gladwin MT. Exploring the mechanisms of the reductase activity of neuroglobin by site-directed mutagenesis of 914. Sun J, Murphy E. Protein S-nitrosylation and cardioprotection. Circ Res 106: 285– the heme distal pocket. Erratum at: doi:10.1021/asc.biochem.5b00579. Biochemistry 296, 2010. doi:10.1161/CIRCRESAHA.109.209452. 54: 722–733, 2015. doi:10.1021/bi501196k.

915. Sun Q-A, Hess DT, Wang B, Miyagi M, Stamler JS. Off-target thiol alkylation by the 933. Tejero J, Stuehr D. Tetrahydrobiopterin in nitric oxide synthase. IUBMB Life 65: NADPH oxidase inhibitor 3-benzyl-7-(2-benzoxazolyl)thio-1,2,3-triazolo[4,5-d]py- 358–365, 2013. doi:10.1002/iub.1136. rimidine (VAS2870). Free Radic Biol Med 52: 1897–1902, 2012. doi:10.1016/j. freeradbiomed.2012.02.046. 934. ten Freyhaus H, Huntgeburth M, Wingler K, Schnitker J, Bäumer AT, Vantler M, Bekhite MM, Wartenberg M, Sauer H, Rosenkranz S. Novel Nox inhibitor VAS2870 916. Sun X, Kumar S, Sharma S, Aggarwal S, Lu Q, Gross C, Rafikova O, Lee SG, Dasara- attenuates PDGF-dependent smooth muscle cell chemotaxis, but not proliferation. thy S, Hou Y, Meadows ML, Han W, Su Y, Fineman JR, Black SM. Endothelin-1 Cardiovasc Res 71: 331–341, 2006. doi:10.1016/j.cardiores.2006.01.022. induces a glycolytic switch in pulmonary arterial endothelial cells via the mitochon- drial translocation of endothelial nitric oxide synthase. Am J Respir Cell Mol Biol 50: 935. Terao M, Romão MJ, Leimkühler S, Bolis M, Fratelli M, Coelho C, Santos-Silva T, 1084–1095, 2014. doi:10.1165/rcmb.2013-0187OC. Garattini E. Structure and function of mammalian aldehyde oxidases. Arch Toxicol 90: 753–780, 2016. doi:10.1007/s00204-016-1683-1. 917. Sun Y, Jin K, Peel A, Mao XO, Xie L, Greenberg DA. Neuroglobin protects the brain from experimental stroke in vivo. Proc Natl Acad Sci USA 100: 3497–3500, 2003. 936. Thannickal VJ, Fanburg BL. Activation of an H2O2-generating NADH oxidase in doi:10.1073/pnas.0637726100. human lung fibroblasts by transforming growth factor beta 1. J Biol Chem 270: 30334–30338, 1995. doi:10.1074/jbc.270.51.30334. 918. Surmeli NB, Litterman NK, Miller AF, Groves JT. Peroxynitrite mediates active site tyrosine nitration in manganese superoxide dismutase. Evidence of a role for the 937. Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. Am J Physiol Lung carbonate radical anion. J Am Chem Soc 132: 17174–17185, 2010. doi:10.1021/ Cell Mol Physiol 279: L1005–L1028, 2000. doi:10.1152/ajplung.2000.279.6.L1005. ja105684w. 938. Thomas DD, Heinecke JL, Ridnour LA, Cheng RY, Kesarwala AH, Switzer CH, 919. Suzuki J, Kaziro Y, Koide H. Synergistic action of R-Ras and IGF-1 on Bcl-xL expres- McVicar DW, Roberts DD, Glynn S, Fukuto JM, Wink DA, Miranda KM. Signaling and sion and caspase-3 inhibition in BaF3 cells: R-Ras and IGF-1 control distinct anti- stress: The redox landscape in NOS2 biology. Free Radic Biol Med 87: 204–225, apoptotic kinase pathways. FEBS Lett 437: 112–116, 1998. doi:10.1016/S0014- 2015. doi:10.1016/j.freeradbiomed.2015.06.002. 5793(98)01213-7.

920. Swenson TL, Casida JE. Aldehyde oxidase importance in vivo in xenobiotic metab- 939. Thomas DD, Ridnour LA, Isenberg JS, Flores-Santana W, Switzer CH, Donzelli S, olism: imidacloprid nitroreduction in mice. Toxicol Sci 133: 22–28, 2013. doi:10. Hussain P, Vecoli C, Paolocci N, Ambs S, Colton CA, Harris CC, Roberts DD, Wink 1093/toxsci/kft066. DA. The chemical biology of nitric oxide: implications in cellular signaling. Free Radic Biol Med 45: 18–31, 2008. doi:10.1016/j.freeradbiomed.2008.03.020. 921. Szeto HH. Cell-permeable, mitochondrial-targeted, peptide antioxidants. AAPS J 8: E277–E283, 2006. doi:10.1007/BF02854898. 940. Thomas SR, Chen K, Keaney JF Jr. Hydrogen peroxide activates endothelial nitric- oxide synthase through coordinated phosphorylation and dephosphorylation via a 922. Szeto HH. Mitochondria-targeted cytoprotective peptides for ischemia-reperfusion phosphoinositide 3-kinase-dependent signaling pathway. J Biol Chem 277: 6017– injury. Antioxid Redox Signal 10: 601–619, 2008. doi:10.1089/ars.2007.1892. 6024, 2002. doi:10.1074/jbc.M109107200.

923. Szeto HH. Mitochondria-targeted peptide antioxidants: novel neuroprotective 941. Thomas SR, Witting PK, Drummond GR. Redox control of endothelial function and agents. AAPS J 8: E521–E531, 2006. doi:10.1208/aapsj080362. dysfunction: molecular mechanisms and therapeutic opportunities. Antioxid Redox Signal 10: 1713–1765, 2008. doi:10.1089/ars.2008.2027. 924. Taillé C, El-Benna J, Lanone S, Boczkowski J, Motterlini R. Mitochondrial respiratory chain and NAD(P)H oxidase are targets for the antiproliferative effect of carbon 942. Thomson L, Trujillo M, Telleri R, Radi R. Kinetics of cytochrome c2ϩ oxidation by monoxide in human airway smooth muscle. J Biol Chem 280: 25350–25360, 2005. peroxynitrite: implications for superoxide measurements in nitric oxide-producing doi:10.1074/jbc.M503512200. biological systems. Arch Biochem Biophys 319: 491–497, 1995. doi:10.1006/abbi. 1995.1321. 925. Takac I, Schröder K, Zhang L, Lardy B, Anilkumar N, Lambeth JD, Shah AM, Morel F, Brandes RP. The E-loop is involved in hydrogen peroxide formation by the NA- 943. Thoonen R, Cauwels A, Decaluwe K, Geschka S, Tainsh RE, Delanghe J, Hochepied DPH oxidase Nox4. J Biol Chem 286: 13304–13313, 2011. doi:10.1074/jbc.M110. T, De Cauwer L, Rogge E, Voet S, Sips P, Karas RH, Bloch KD, Vuylsteke M, Stasch 192138. JP, Van de Voorde J, Buys ES, Brouckaert P. Cardiovascular and pharmacological 926. Takakura K, Beckman JS, MacMillan-Crow LA, Crow JP. Rapid and irreversible implications of haem-deficient NO-unresponsive soluble guanylate cyclase knock-in inactivation of protein tyrosine phosphatases PTP1B, CD45, and LAR by peroxyni- mice. Nat Commun 6: 8482, 2015. doi:10.1038/ncomms9482. trite. Arch Biochem Biophys 369: 197–207, 1999. doi:10.1006/abbi.1999.1374. 944. Tirone F, Radu L, Craescu CT, Cox JA. Identification of the binding site for the 927. Takeya R, Ueno N, Kami K, Taura M, Kohjima M, Izaki T, Nunoi H, Sumimoto H. regulatory calcium-binding domain in the catalytic domain of NOX5. Biochemistry Novel human homologues of p47phox and p67phox participate in activation of 49: 761–771, 2010. doi:10.1021/bi901846y. superoxide-producing NADPH oxidases. J Biol Chem 278: 25234–25246, 2003. doi: 945. Tiso M, Tejero J, Basu S, Azarov I, Wang X, Simplaceanu V, Frizzell S, Jayaraman T, 10.1074/jbc.M212856200. Geary L, Shapiro C, Ho C, Shiva S, Kim-Shapiro DB, Gladwin MT. Human neuro- 928. Tamariz L, Hare JM. Xanthine oxidase inhibitors in heart failure: where do we go globin functions as a redox-regulated nitrite reductase. J Biol Chem 286: 18277– from here? Circulation 131: 1741–1744, 2015. doi:10.1161/CIRCULATIONAHA. 18289, 2011. doi:10.1074/jbc.M110.159541. 115.016379. 946. Tiso M, Tejero J, Kenney C, Frizzell S, Gladwin MT. Nitrite reductase activity of 929. Taylor BA, Zaleski AL, Dornelas EA, Thompson PD. The impact of tetrahydrobiop- nonsymbiotic hemoglobins from Arabidopsis thaliana. Biochemistry 51: 5285–5292, terin administration on endothelial function before and after smoking cessation in 2012. doi:10.1021/bi300570v. chronic smokers. Hypertens Res 39: 144–150, 2016. doi:10.1038/hr.2015.130. 947. Tiso M, Tejero J, Panda K, Aulak KS, Stuehr DJ. Versatile regulation of neuronal nitric 930. Taylor RM, Burritt JB, Baniulis D, Foubert TR, Lord CI, Dinauer MC, Parkos CA, oxide synthase by specific regions of its C-terminal tail. Biochemistry 46: 14418– Jesaitis AJ. Site-specific inhibitors of NADPH oxidase activity and structural probes of 14428, 2007. doi:10.1021/bi701646k.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 375 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

948. Tormos KV, Anso E, Hamanaka RB, Eisenbart J, Joseph J, Kalyanaraman B, Chandel 967. Van Doorslaer S, Dewilde S, Kiger L, Nistor SV, Goovaerts E, Marden MC, Moens L. NS. Mitochondrial complex III ROS regulate adipocyte differentiation. Cell Metab 14: Nitric oxide binding properties of neuroglobin. A characterization by EPR and flash 537–544, 2011. doi:10.1016/j.cmet.2011.08.007. photolysis. J Biol Chem 278: 4919–4925, 2003. doi:10.1074/jbc.M210617200.

949. Touyz RM. Reactive oxygen species, vascular oxidative stress, and redox signaling in 968. van Faassen EE, Bahrami S, Feelisch M, Hogg N, Kelm M, Kim-Shapiro DB, Kozlov hypertension: what is the clinical significance? Hypertension 44: 248–252, 2004. AV, Li H, Lundberg JO, Mason R, Nohl H, Rassaf T, Samouilov A, Slama-Schwok A, doi:10.1161/01.HYP.0000138070.47616.9d. Shiva S, Vanin AF, Weitzberg E, Zweier J, Gladwin MT. Nitrite as regulator of hypoxic signaling in mammalian physiology. Med Res Rev 29: 683–741, 2009. doi:10. 950. Touyz RM, Chen X, Tabet F, Yao G, He G, Quinn MT, Pagano PJ, Schiffrin EL. 1002/med.20151. Expression of a functionally active gp91phox-containing neutrophil-type NAD(P)H oxidase in smooth muscle cells from human resistance arteries: regulation by angio- 970. Vanin AF, Bevers LM, Slama-Schwok A, van Faassen EE. Nitric oxide synthase re- tensin II. Circ Res 90: 1205–1213, 2002. doi:10.1161/01.RES.0000020404.01971.2F. duces nitrite to NO under anoxia. Cell Mol Life Sci 64: 96–103, 2007. doi:10.1007/ s00018-006-6374-2. 951. Trent JT III, Hargrove MS. A ubiquitously expressed human hexacoordinate hemo- globin. J Biol Chem 277: 19538–19545, 2002. doi:10.1074/jbc.M201934200. 971. Varga ZV, Pipicz M, Baán JA, Baranyai T, Koncsos G, Leszek P, Kus´mierczyk M, Sánchez-Cabo F, García-Pavía P, Brenner GJ, Giricz Z, Csont T, Mendler L, Lara- 952. Tretter L, Adam-Vizi V. Generation of reactive oxygen species in the reaction cata- Pezzi E, Pacher P, Ferdinandy P. Alternative splicing of NOX4 in the failing human lyzed by alpha-ketoglutarate dehydrogenase. J Neurosci 24: 7771–7778, 2004. doi: heart. Front Physiol 8: 935, 2017. doi:10.3389/fphys.2017.00935. 10.1523/JNEUROSCI.1842-04.2004. 972. Vásquez-Vivar J. Tetrahydrobiopterin, superoxide, and vascular dysfunction. Free 953. Tsai A-L, Berka V, Sharina I, Martin E. Dynamic ligand exchange in soluble guanylyl Radic Biol Med 47: 1108–1119, 2009. doi:10.1016/j.freeradbiomed.2009.07.024. cyclase (sGC): implications for sGC regulation and desensitization. J Biol Chem 286: 43182–43192, 2011. doi:10.1074/jbc.M111.290304. 973. Vásquez-Vivar J, Kalyanaraman B, Martásek P, Hogg N, Masters BSS, Karoui H, Tordo P, Pritchard KA Jr. Superoxide generation by endothelial nitric oxide synthase: 954. Tsikas D, Böger RH, Sandmann J, Bode-Böger SM, Frölich JC. Endogenous nitric the influence of cofactors. Proc Natl Acad Sci USA 95: 9220–9225, 1998. doi:10.1073/ oxide synthase inhibitors are responsible for the L-arginine paradox. FEBS Lett 478: pnas.95.16.9220. 1–3, 2000. doi:10.1016/S0014-5793(00)01686-0. 974. Vásquez-Vivar J, Martásek P, Whitsett J, Joseph J, Kalyanaraman B. The ratio be- 955. Turell L, Vitturi DA, Coitiño EL, Lebrato L, Möller MN, Sagasti C, Salvatore SR, tween tetrahydrobiopterin and oxidized tetrahydrobiopterin analogues controls su- Woodcock SR, Alvarez B, Schopfer FJ. The Chemical Basis of Thiol Addition to peroxide release from endothelial nitric oxide synthase: an EPR spin trapping study. Nitro-conjugated Linoleic Acid, a Protective Cell-signaling Lipid. J Biol Chem 292: Biochem J 362: 733–739, 2002. doi:10.1042/bj3620733. 1145–1159, 2017. doi:10.1074/jbc.M116.756288. 975. Velmurugan S, Kapil V, Ghosh SM, Davies S, McKnight A, Aboud Z, Khambata RS, 956. Tyurin VA, Liu SX, Tyurina YY, Sussman NB, Hubel CA, Roberts JM, Taylor RN, Webb AJ, Poole A, Ahluwalia A. Antiplatelet effects of dietary nitrate in healthy Kagan VE. Elevated levels of S-nitrosoalbumin in preeclampsia plasma. Circ Res 88: volunteers: involvement of cGMP and influence of sex. Free Radic Biol Med 65: 1210–1215, 2001. doi:10.1161/hh1101.092179. 1521–1532, 2013. doi:10.1016/j.freeradbiomed.2013.06.031. Erratum at: doi:10. 1016/j.freeradbiomed.2015.03.015. 957. Ueda S, Matsuoka H, Miyazaki H, Usui M, Okuda S, Imaizumi T. Tetrahydrobiop- terin restores endothelial function in long-term smokers. J Am Coll Cardiol 35: 71–75, 976. Vendrov AE, Hakim ZS, Madamanchi NR, Rojas M, Madamanchi C, Runge MS. 2000. doi:10.1016/S0735-1097(99)00523-9. Atherosclerosis is attenuated by limiting superoxide generation in both macrophages and vessel wall cells. Arterioscler Thromb Vasc Biol 27: 2714–2721, 2007. doi:10. 958. Underbakke ES, Iavarone AT, Chalmers MJ, Pascal BD, Novick S, Griffin PR, Mar- 1161/ATVBAHA.107.152629. letta MA. Nitric oxide-induced conformational changes in soluble guanylate cyclase. Structure 22: 602–611, 2014. doi:10.1016/j.str.2014.01.008. 977. Venema VJ, Ju H, Zou R, Venema RC. Interaction of neuronal nitric-oxide synthase with caveolin-3 in skeletal muscle. Identification of a novel caveolin scaffolding/ 959. Valko M, Jomova K, Rhodes CJ, Kucˇa K, Musílek K. Redox- and non-redox-metal- inhibitory domain. J Biol Chem 272: 28187–28190, 1997. doi:10.1074/jbc.272.45. induced formation of free radicals and their role in human disease. Arch Toxicol 90: 28187. 1–37, 2016. doi:10.1007/s00204-015-1579-5. 978. Verhaar MC, Wever RM, Kastelein JJ, van Dam T, Koomans HA, Rabelink TJ. 960. Valko M, Leibfritz D, Moncol J, Cronin MTD, Mazur M, Telser J. Free radicals and 5-methyltetrahydrofolate, the active form of folic acid, restores endothelial function antioxidants in normal physiological functions and human disease. Int J Biochem Cell in familial hypercholesterolemia. Circulation 97: 237–241, 1998. doi:10.1161/01.CIR. Biol 39: 44–84, 2007. doi:10.1016/j.biocel.2006.07.001. 97.3.237.

961. Valkonen VP, Päivä H, Salonen JT, Lakka TA, Lehtimäki T, Laakso J, Laaksonen R. 979. Vickers S, Schiller HJ, Hildreth JE, Bulkley GB. Immunoaffinity localization of the Risk of acute coronary events and serum concentration of asymmetrical dimethy- enzyme xanthine oxidase on the outside surface of the endothelial cell plasma mem- larginine. Lancet 358: 2127–2128, 2001. doi:10.1016/S0140-6736(01)07184-7. brane. Surgery 124: 551–560, 1998. doi:10.1016/S0039-6060(98)70102-3.

962. Vallance P, Leiper J. Cardiovascular biology of the asymmetric dimethylarginine: 980. Vignais PV. The superoxide-generating NADPH oxidase: structural aspects and ac- dimethylarginine dimethylaminohydrolase pathway. Arterioscler Thromb Vasc Biol 24: tivation mechanism. Cell Mol Life Sci 59: 1428–1459, 2002. doi:10.1007/s00018-002- 1023–1030, 2004. doi:10.1161/01.ATV.0000128897.54893.26. 8520-9.

963. Vallance P, Leone A, Calver A, Collier J, Moncada S. Accumulation of an endogenous 981. Virdis A, Colucci R, Fornai M, Blandizzi C, Duranti E, Pinto S, Bernardini N, Segnani inhibitor of nitric oxide synthesis in chronic renal failure. Lancet 339: 572–575, 1992. C, Antonioli L, Taddei S, Salvetti A, Del Tacca M. Cyclooxygenase-2 inhibition im- doi:10.1016/0140-6736(92)90865-Z. proves vascular endothelial dysfunction in a rat model of endotoxic shock: role of inducible nitric-oxide synthase and oxidative stress. J Pharmacol Exp Ther 312: 945– 964. Van Buul JD, Fernandez-Borja M, Anthony EC, Hordijk PL. Expression and localiza- 953, 2005. doi:10.1124/jpet.104.077644. tion of NOX2 and NOX4 in primary human endothelial cells. Antioxid Redox Signal 7: 308–317, 2005. doi:10.1089/ars.2005.7.308. 982. Virdis A, Colucci R, Fornai M, Duranti E, Giannarelli C, Bernardini N, Segnani C, Ippolito C, Antonioli L, Blandizzi C, Taddei S, Salvetti A, Del Tacca M. Cyclooxygen- 965. Van der Schueren BJ, Lunnon MW, Laurijssens BE, Guillard F, Palmer J, Van ase-1 is involved in endothelial dysfunction of mesenteric small arteries from angio- Hecken A, Depré M, Vanmolkot FH, de Hoon JN. Does the unfavorable phar- tensin II-infused mice. Hypertension 49: 679–686, 2007. doi:10.1161/01.HYP. macokinetic and pharmacodynamic profile of the iNOS inhibitor GW273629 lead 0000253085.56217.11. to inefficacy in acute migraine? J Clin Pharmacol 49: 281–290, 2009. doi:10.1177/ 0091270008329548. 983. Viswambharan H, Yuldasheva NY, Sengupta A, Imrie H, Gage MC, Haywood N, Walker AM, Skromna A, Makova N, Galloway S, Shah P, Sukumar P, Porter KE, 966. van der Zwan LP, Scheffer PG, Dekker JM, Stehouwer CD, Heine RJ, Teerlink T. Grant PJ, Shah AM, Santos CX, Li J, Beech DJ, Wheatcroft SB, Cubbon RM, Kearney Systemic inflammation is linked to low arginine and high ADMA plasma levels result- MT. Selective Enhancement of Insulin Sensitivity in the Endothelium In Vivo Reveals ing in an unfavourable NOS substrate-to-inhibitor ratio: the Hoorn Study. Clin Sci a Novel Proatherosclerotic Signaling Loop. Circ Res 120: 784–798, 2017. doi:10. (Lond) 121: 71–78, 2011. doi:10.1042/CS20100595. 1161/CIRCRESAHA.116.309678.

376 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

984. Voraphani N, Gladwin MT, Contreras AU, Kaminski N, Tedrow JR, Milosevic J, 1001. Weidert ER, Schoenborn SO, Cantu-Medellin N, Choughule KV, Jones JP, Kelley EE. Bleecker ER, Meyers DA, Ray A, Ray P, Erzurum SC, Busse WW, Zhao J, Trudeau JB, Inhibition of xanthine oxidase by the aldehyde oxidase inhibitor raloxifene: implica- Wenzel SE. An airway epithelial iNOS-DUOX2-thyroid peroxidase metabolome tions for identifying molybdopterin nitrite reductases. Nitric Oxide 37: 41–45, 2014. drives Th1/Th2 nitrative stress in human severe asthma. Mucosal Immunol 7: 1175– doi:10.1016/j.niox.2013.12.010. 1185, 2014. doi:10.1038/mi.2014.6. 1002. Weitzberg E, Lundberg JO. Novel aspects of dietary nitrate and human health. Annu 985. Waheed SM, Ghosh A, Chakravarti R, Biswas A, Haque MM, Panda K, Stuehr DJ. Rev Nutr 33: 129–159, 2013. doi:10.1146/annurev-nutr-071812-161159. Nitric oxide blocks cellular heme insertion into a broad range of heme proteins. Free Radic Biol Med 48: 1548–1558, 2010. doi:10.1016/j.freeradbiomed.2010.02.038. 1003. Wendt MC, Daiber A, Kleschyov AL, Mülsch A, Sydow K, Schulz E, Chen K, Keaney JF Jr, Lassègue B, Walter U, Griendling KK, Münzel T. Differential effects of diabetes 986. Walling C. Fenton’s reagent revisited. Acc Chem Res 8: 125–131, 1975. doi:10.1021/ on the expression of the gp91phox homologues nox1 and nox4. Free Radic Biol Med ar50088a003. 39: 381–391, 2005. doi:10.1016/j.freeradbiomed.2005.03.020.

987. Walter R, Kaufmann PA, Buck A, Berthold T, Wyss C, von Schulthess GK, Schaffner 1004. Wenzel P, Mollnau H, Oelze M, Schulz E, Wickramanayake JM, Müller J, Schuhm- A, Schoedon G. Tetrahydrobiopterin increases myocardial blood flow in healthy acher S, Hortmann M, Baldus S, Gori T, Brandes RP, Münzel T, Daiber A. First volunteers: a double-blind, placebo-controlled study. Swiss Med Wkly 131: 91–94, evidence for a crosstalk between mitochondrial and NADPH oxidase-derived reac- 2001. tive oxygen species in nitroglycerin-triggered vascular dysfunction. Antioxid Redox Signal 10: 1435–1447, 2008. doi:10.1089/ars.2007.1969. 988. Wang HD, Xu S, Johns DG, Du Y, Quinn MT, Cayatte AJ, Cohen RA. Role of NADPH oxidase in the vascular hypertrophic and oxidative stress response to angiotensin II in 1005. Wenzel P, Schuhmacher S, Kienhöfer J, Müller J, Hortmann M, Oelze M, Schulz E, mice. Circ Res 88: 947–953, 2001. doi:10.1161/hh0901.089987. Treiber N, Kawamoto T, Scharffetter-Kochanek K, Münzel T, Bürkle A, Bachschmid MM, Daiber A. Manganese superoxide dismutase and aldehyde dehydrogenase de- 989. Wang J, Hong Z, Zeng C, Yu Q, Wang H. NADPH oxidase 4 promotes cardiac ficiency increase mitochondrial oxidative stress and aggravate age-dependent vascu- microvascular angiogenesis after hypoxia/reoxygenation in vitro. Free Radic Biol Med lar dysfunction. Cardiovasc Res 80: 280–289, 2008. doi:10.1093/cvr/cvn182. 69: 278–288, 2014. doi:10.1016/j.freeradbiomed.2014.01.027. 1006. Werner-Felmayer G, Werner ER, Fuchs D, Hausen A, Reibnegger G, Schmidt K, 990. Wang J, Krizowski S, Fischer-Schrader K, Niks D, Tejero J, Sparacino-Watkins C, Weiss G, Wachter H. Pteridine biosynthesis in human endothelial cells. Impact on Wang L, Ragireddy V, Frizzell S, Kelley EE, Zhang Y, Basu P, Hille R, Schwarz G, nitric oxide-mediated formation of cyclic GMP. J Biol Chem 268: 1842–1846, 1993. Gladwin MT. Sulfite Oxidase Catalyzes Single-Electron Transfer at Molybdenum Domain to Reduce Nitrite to Nitric Oxide. Antioxid Redox Signal 23: 283–294, 2015. 1007. Werner ER, Blau N, Thöny B. Tetrahydrobiopterin: biochemistry and pathophysiol- doi:10.1089/ars.2013.5397. ogy. Biochem J 438: 397–414, 2011. doi:10.1042/BJ20110293. ϩ 991. Wang JP, Chang LC, Huang LJ, Kuo SC. Inhibition of extracellular Ca(2 ) entry by 1008. Werner ER, Pitters E, Schmidt K, Wachter H, Werner-Felmayer G, Mayer B. Iden- YC-1, an activator of soluble guanylyl cyclase, through a cyclic GMP-independent tification of the 4-amino analogue of tetrahydrobiopterin as a dihydropteridine re- pathway in rat neutrophils. Biochem Pharmacol 62: 679–684, 2001. doi:10.1016/ ductase inhibitor and a potent pteridine antagonist of rat neuronal nitric oxide syn- S0006-2952(01)00725-0. thase. Biochem J 320: 193–196, 1996. doi:10.1042/bj3200193.

992. Wang W, Sawicki G, Schulz R. Peroxynitrite-induced myocardial injury is mediated 1009. West MB, Rokosh G, Obal D, Velayutham M, Xuan Y-T, Hill BG, Keith RJ, Schrader through matrix metalloproteinase-2. Cardiovasc Res 53: 165–174, 2002. doi:10. J, Guo Y, Conklin DJ, Prabhu SD, Zweier JL, Bolli R, Bhatnagar A. Cardiac myocyte- 1016/S0008-6363(01)00445-X. specific expression of inducible nitric oxide synthase protects against ischemia/rep- 993. Warner TD, Mitchell JA, Sheng H, Murad F. Effects of cyclic GMP on smooth erfusion injury by preventing mitochondrial permeability transition. Circulation 118: muscle relaxation. Adv Pharmacol 26: 171–194, 1994. doi:10.1016/S1054- 1970–1978, 2008. doi:10.1161/CIRCULATIONAHA.108.791533. 3589(08)60054-X. 1010. White CR, Darley-Usmar V, Berrington WR, McAdams M, Gore JZ, Thompson JA, 994. Waud WR, Rajagopalan KV. The mechanism of conversion of rat liver xanthine Parks DA, Tarpey MM, Freeman BA. Circulating plasma xanthine oxidase contrib- dehydrogenase from an NADϩ-dependent form (type D) to an O2-dependent form utes to vascular dysfunction in hypercholesterolemic rabbits. Proc Natl Acad Sci USA (type O). Arch Biochem Biophys 172: 365–379, 1976. doi:10.1016/0003- 93: 8745–8749, 1996. doi:10.1073/pnas.93.16.8745. 9861(76)90088-6. 1011. Willeit P, Freitag DF, Laukkanen JA, Chowdhury S, Gobin R, Mayr M, Di Angelanto- 995. Waypa GB, Chandel NS, Schumacker PT. Model for hypoxic pulmonary vasocon- nio E, Chowdhury R. Asymmetric dimethylarginine and cardiovascular risk: system- striction involving mitochondrial oxygen sensing. Circ Res 88: 1259–1266, 2001. atic review and meta-analysis of 22 prospective studies. J Am Heart Assoc 4: e001833, doi:10.1161/hh1201.091960. 2015. doi:10.1161/JAHA.115.001833. ␬ 996. Waypa GB, Marks JD, Guzy RD, Mungai PT, Schriewer JM, Dokic D, Ball MK, 1012. Williams CR, Lu X, Sutliff RL, Hart CM. Rosiglitazone attenuates NF- B-mediated Schumacker PT. Superoxide generated at mitochondrial complex III triggers acute Nox4 upregulation in hyperglycemia-activated endothelial cells. Am J Physiol Cell responses to hypoxia in the pulmonary circulation. Am J Respir Crit Care Med 187: Physiol 303: C213–C223, 2012. doi:10.1152/ajpcell.00227.2011. 424–432, 2013. doi:10.1164/rccm.201207-1294OC. 1013. Williams PA, Cosme J, Sridhar V, Johnson EF, McRee DE. Mammalian microsomal 997. Waypa GB, Smith KA, Schumacker PT. O2 sensing, mitochondria and ROS signaling: cytochrome P450 monooxygenase: structural adaptations for membrane binding The fog is lifting. Mol Aspects Med 47-48: 76–89, 2016. doi:10.1016/j.mam.2016.01. and functional diversity. Mol Cell 5: 121–131, 2000. doi:10.1016/S1097- 002. 2765(00)80408-6.

998. Webb AJ, Milsom AB, Rathod KS, Chu WL, Qureshi S, Lovell MJ, Lecomte FMJ, 1014. Wilson AM, Harada R, Nair N, Balasubramanian N, Cooke JP. L-arginine supplemen- Perrett D, Raimondo C, Khoshbin E, Ahmed Z, Uppal R, Benjamin N, Hobbs AJ, tation in peripheral arterial disease: no benefit and possible harm. Circulation 116: Ahluwalia A. Mechanisms underlying erythrocyte and endothelial nitrite reduction to 188–195, 2007. doi:10.1161/CIRCULATIONAHA.106.683656. nitric oxide in hypoxia: role for xanthine oxidoreductase and endothelial nitric oxide synthase. Circ Res 103: 957–964, 2008. doi:10.1161/CIRCRESAHA.108.175810. 1015. Wind S, Beuerlein K, Armitage ME, Taye A, Kumar AHS, Janowitz D, Neff C, Shah AM, Wingler K, Schmidt HHHW. Oxidative stress and endothelial dysfunction in 999. Weber DS, Taniyama Y, Rocic P, Seshiah PN, Dechert MA, Gerthoffer WT, Grien- aortas of aged spontaneously hypertensive rats by NOX1/2 is reversed by NA- dling KK. Phosphoinositide-dependent kinase 1 and p21-activated protein kinase DPH oxidase inhibition. Hypertension 56: 490–497, 2010. doi:10.1161/ mediate reactive oxygen species-dependent regulation of platelet-derived growth HYPERTENSIONAHA.109.149187. factor-induced smooth muscle cell migration. Circ Res 94: 1219–1226, 2004. doi:10. 1161/01.RES.0000126848.54740.4A. 1016. Wind S, Beuerlein K, Eucker T, Müller H, Scheurer P, Armitage ME, Ho H, Schmidt HHHW, Wingler K. Comparative pharmacology of chemically distinct NADPH ox- 1000. Wei CC, Crane BR, Stuehr DJ. Tetrahydrobiopterin radical enzymology. Chem Rev idase inhibitors. Br J Pharmacol 161: 885–898, 2010. doi:10.1111/j.1476-5381.2010. 103: 2365–2383, 2003. doi:10.1021/cr0204350. 00920.x.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 377 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. TEJERO ET AL.

1017. Winger JA, Marletta MA. Expression and characterization of the catalytic domains of human endothelial cells. Free Radic Biol Med 44: 1656–1667, 2008. doi:10.1016/j. soluble guanylate cyclase: interaction with the heme domain. Biochemistry 44: 4083– freeradbiomed.2008.01.023. 4090, 2005. doi:10.1021/bi047601d. 1037. Xu KY, Huso DL, Dawson TM, Bredt DS, Becker LC. Nitric oxide synthase in cardiac 1018. Wink DA, Darbyshire JF, Nims RW, Saavedra JE, Ford PC. Reactions of the bioregu- sarcoplasmic reticulum. Proc Natl Acad Sci USA 96: 657–662, 1999. doi:10.1073/ latory agent nitric oxide in oxygenated aqueous media: determination of the kinetics pnas.96.2.657. for oxidation and nitrosation by intermediates generated in the NO/O2 reaction. Chem Res Toxicol 6: 23–27, 1993. doi:10.1021/tx00031a003. 1038. Xu L, Eu JP, Meissner G, Stamler JS. Activation of the cardiac calcium release channel (ryanodine receptor) by poly-S-nitrosylation. Science 279: 234–237, 1998. doi:10. 1019. Winterbourn CC. Biological reactivity and biomarkers of the neutrophil oxidant, 1126/science.279.5348.234. hypochlorous acid. Toxicology 181-182: 223–227, 2002. doi:10.1016/S0300- 483X(02)00286-X. 1039. Xu W, Kaneko FT, Zheng S, Comhair SAA, Janocha AJ, Goggans T, Thunnissen FB, Farver C, Hazen SL, Jennings C, Dweik RA, Arroliga AC, Erzurum SC. Increased 1020. Winterbourn CC. Toxicity of iron and hydrogen peroxide: the Fenton reaction. arginase II and decreased NO synthesis in endothelial cells of patients with pulmo- Toxicol Lett 82-83: 969–974, 1995. doi:10.1016/0378-4274(95)03532-X. nary arterial hypertension. FASEB J 18: 1746–1748, 2004. doi:10.1096/fj.04-2317fje. 1021. Winterbourn CC, Kettle AJ. Redox reactions and microbial killing in the neutrophil 1040. Yamamoto S. Mammalian lipoxygenases: molecular structures and functions. phagosome. Antioxid Redox Signal 18: 642–660, 2013. doi:10.1089/ars.2012.4827. Biochim Biophys Acta 1128: 117–131, 1992. doi:10.1016/0005-2760(92)90297-9. 1022. Wobst J, Kessler T, Dang TA, Erdmann J, Schunkert H. Role of sGC-dependent NO 1041. Yamamoto T, Moriwaki Y, Fujimura Y, Takahashi S, Tsutsumi Z, Tsutsui T, Hi- signalling and myocardial infarction risk. J Mol Med (Berl) 93: 383–394, 2015. doi:10. gashino K, Hada T. Effect of TEI-6720, a xanthine oxidase inhibitor, on the nucleo- 1007/s00109-015-1265-3. side transport in the lung cancer cell line A549. Pharmacology 60: 34–40, 2000. 1023. Wolhuter K, Eaton P. How widespread is stable protein S-nitrosylation as an end- doi:10.1159/000028344. effector of protein regulation? Free Radic Biol Med 109: 156–166, 2017. doi:10.1016/ j.freeradbiomed.2017.02.013. 1042. Yang CY, Gu ZW, Yang M, Lin SN, Garcia-Prats AJ, Rogers LK, Welty SE, Smith CV. Selective modification of apoB-100 in the oxidation of low density lipoproteins by 1024. Wolhuter K, Whitwell HJ, Switzer CH, Burgoyne JR, Timms JF, Eaton P. Evidence myeloperoxidase in vitro. J Lipid Res 40: 686–698, 1999. against stable protein S-nitrosylation as a widespread mechanism of post-transla- tional regulation. Mol Cell 69: 438–450.e5, 2018. doi:10.1016/j.molcel.2017.12.019. 1043. Yang J, Giles LJ, Ruppelt C, Mendel RR, Bittner F, Kirk ML. Oxyl and hydroxyl radical transfer in mitochondrial amidoxime reducing component-catalyzed nitrite reduc- 1025. Wood KC, Cortese-Krott MM, Kovacic JC, Noguchi A, Liu VB, Wang X, Ragha- tion. J Am Chem Soc 137: 5276–5279, 2015. doi:10.1021/jacs.5b01112. vachari N, Boehm M, Kato GJ, Kelm M, Gladwin MT. Circulating blood endothelial nitric oxide synthase contributes to the regulation of systemic blood pressure and 1044. Yang J, Gonon AT, Sjöquist P-O, Lundberg JO, Pernow J. Arginase regulates red nitrite homeostasis. Arterioscler Thromb Vasc Biol 33: 1861–1871, 2013. doi:10.1161/ blood cell nitric oxide synthase and export of cardioprotective nitric oxide bioactiv- ATVBAHA.112.301068. ity. Proc Natl Acad Sci USA 110: 15049–15054, 2013. doi:10.1073/pnas.1307058110.

1026. Wood KC, Hsu LL, Gladwin MT. Sickle cell disease vasculopathy: a state of nitric 1045. Au Yeung SL, Lin SL, Lam HSHS, Schooling CM. Effect of l-arginine, asymmetric oxide resistance. Free Radic Biol Med 44: 1506–1528, 2008. doi:10.1016/j. dimethylarginine, and symmetric dimethylarginine on ischemic heart disease risk: A freeradbiomed.2008.01.008. Mendelian randomization study. Am Heart J 182: 54–61, 2016. doi:10.1016/j.ahj. 2016.07.021. 1027. Woodcock SR, Bonacci G, Gelhaus SL, Schopfer FJ. Nitrated fatty acids: synthesis and measurement. Free Radic Biol Med 59: 14–26, 2013. doi:10.1016/j. 1046. Yoboue ED, Mougeolle A, Kaiser L, Averet N, Rigoulet M, Devin A. The role of freeradbiomed.2012.11.015. mitochondrial biogenesis and ROS in the control of energy supply in proliferating cells. Biochim Biophys Acta 1837: 1093–1098, 2014. doi:10.1016/j.bbabio.2014.02. 1028. Worthley MI, Kanani RS, Sun YH, Sun Y, Goodhart DM, Curtis MJ, Anderson TJ. 023. Effects of tetrahydrobiopterin on coronary vascular reactivity in atherosclerotic human coronary arteries. Cardiovasc Res 76: 539–546, 2007. doi:10.1016/j. 1047. Yoshida LS, Nishida S, Shimoyama T, Kawahara T, Kondo-Teshima S, Rokutan K, cardiores.2007.07.009. Kobayashi T, Tsunawaki S. Superoxide generation by Nox1 in guinea pig gastric 1029. Wu G, Bazer FW, Davis TA, Kim SW, Li P, Marc Rhoads J, Carey Satterfield M, Smith mucosal cells involves a component with p67(phox)-ability. Biol Pharm Bull 27: 147– SB, Spencer TE, Yin Y. Arginine metabolism and nutrition in growth, health and 155, 2004. doi:10.1248/bpb.27.147. disease. Amino Acids 37: 153–168, 2009. doi:10.1007/s00726-008-0210-y. 1048. Yoshida LS, Tsunawaki S. Expression of NADPH oxidases and enhanced H(2)O(2)- 1030. Wu G, Morris SM Jr. Arginine metabolism: nitric oxide and beyond. Biochem J 336: generating activity in human coronary artery endothelial cells upon induction with 1–17, 1998. doi:10.1042/bj3360001. tumor necrosis factor-alpha. Int Immunopharmacol 8: 1377–1385, 2008. doi:10. 1016/j.intimp.2008.05.004. 1031. Wyss CA, Koepfli P, Namdar M, Siegrist PT, Luscher TF, Camici PG, Kaufmann PA. Tetrahydrobiopterin restores impaired coronary microvascular dysfunction in hy- 1049. Youn JY, Gao L, Cai H. The p47phox- and NADPH oxidase organiser 1 (NOXO1)- percholesterolaemia. Eur J Nucl Med Mol Imaging 32: 84–91, 2005. doi:10.1007/ dependent activation of NADPH oxidase 1 (NOX1) mediates endothelial nitric s00259-004-1621-y. oxide synthase (eNOS) uncoupling and endothelial dysfunction in a streptozotocin- induced murine model of diabetes. Diabetologia 55: 2069–2079, 2012. doi:10.1007/ 1032. Xia Y, Roman LJ, Masters BSS, Zweier JL. Inducible nitric-oxide synthase generates s00125-012-2557-6. superoxide from the reductase domain. J Biol Chem 273: 22635–22639, 1998. doi: 10.1074/jbc.273.35.22635. 1050. Yu E, Ruiz-Canela M, Hu FB, Clish CB, Corella D, Salas-Salvadó J, Hruby A, Fitó M, Liang L, Toledo E, Ros E, Estruch R, Gómez-Gracia E, Lapetra J, Arós F, Romaguera 1033. Xia Y, Tsai AL, Berka V, Zweier JL. Superoxide generation from endothelial nitric- D, Serra-Majem L, Guasch-Ferré M, Wang DD, Martínez-González MA. Plasma oxide synthase. A Ca2ϩ/calmodulin-dependent and tetrahydrobiopterin regulatory Arginine/Asymmetric Dimethylarginine Ratio and Incidence of Cardiovascular process. J Biol Chem 273: 25804–25808, 1998. doi:10.1074/jbc.273.40.25804. Events: A Case-Cohort Study. J Clin Endocrinol Metab 102: 1879–1888, 2017. doi: 1034. Xia Y, Zweier JL. Superoxide and peroxynitrite generation from inducible nitric 10.1210/jc.2016-3569. oxide synthase in macrophages. Proc Natl Acad Sci USA 94: 6954–6958, 1997. doi: 1051. Yu L, Derrick M, Ji H, Silverman RB, Whitsett J, Vásquez-Vivar J, Tan S. Neuronal 10.1073/pnas.94.13.6954. nitric oxide synthase inhibition prevents cerebral palsy following hypoxia-ischemia in 1035. Xu B, Chibber R, Ruggiero D, Kohner E, Ritter J, Ferro A. Impairment of vascular fetal rabbits: comparison between JI-8 and 7-nitroindazole. Dev Neurosci 33: 312– endothelial nitric oxide synthase activity by advanced glycation end products. FASEB 319, 2011. doi:10.1159/000327244. J 17: 1289–1291, 2003. doi:10.1096/fj.02-0490fje. 1052. Zamocky M, Jakopitsch C, Furtmüller PG, Dunand C, Obinger C. The peroxidase- 1036. Xu H, Goettsch C, Xia N, Horke S, Morawietz H, Förstermann U, Li H. Differential cyclooxygenase superfamily: Reconstructed evolution of critical enzymes of the in- roles of PKCalpha and PKCepsilon in controlling the gene expression of Nox4 in nate immune system. Proteins 72: 589–605, 2008. doi:10.1002/prot.21950.

378 Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved. SYNTHESIS AND REGULATION OF VASCULAR NO AND ROS

1053. Zeller M, Korandji C, Guilland J-C, Sicard P, Vergely C, Lorgis L, Beer J-C, Duvillard 1064. Zhao XJ, Wang L, Shiva S, Tejero J, Myerburg MM, Wang J, Frizzell S, Gladwin MT. L, Lagrost A-C, Moreau D, Gambert P, Cottin Y, Rochette L. Impact of asymmetric Mechanisms for cellular NO oxidation and nitrite formation in lung epithelial cells. dimethylarginine on mortality after acute myocardial infarction. Arterioscler Thromb Free Radic Biol Med 61: 428–437, 2013. doi:10.1016/j.freeradbiomed.2013.04.031. Vasc Biol 28: 954–960, 2008. doi:10.1161/ATVBAHA.108.162768. 1065. Zhuang J, Jiang T, Lu D, Luo Y, Zheng C, Feng J, Yang D, Chen C, Yan X. NADPH 1054. Zhang C, Reiter C, Eiserich JP, Boersma B, Parks DA, Beckman JS, Barnes S, Kirk M, oxidase 4 mediates reactive oxygen species induction of CD146 dimerization in Baldus S, Darley-Usmar VM, White CR. L-arginine chlorination products inhibit VEGF signal transduction. Free Radic Biol Med 49: 227–236, 2010. doi:10.1016/j. endothelial nitric oxide production. J Biol Chem 276: 27159–27165, 2001. doi:10. freeradbiomed.2010.04.007. 1074/jbc.M100191200. 1066. Zinkevich NS, Gutterman DD. ROS-induced ROS release in vascular biology: redox- 1055. Zhang G, Zhang F, Muh R, Yi F, Chalupsky K, Cai H, Li PL. Autocrine/paracrine redox signaling. Am J Physiol Heart Circ Physiol 301: H647–H653, 2011. doi:10.1152/ pattern of superoxide production through NAD(P)H oxidase in coronary arterial ajpheart.01271.2010. myocytes. Am J Physiol Heart Circ Physiol 292: H483–H495, 2007. doi:10.1152/ ajpheart.00632.2006. 1067. Zmijewski JW, Banerjee S, Bae H, Friggeri A, Lazarowski ER, Abraham E. Exposure to hydrogen peroxide induces oxidation and activation of AMP-activated protein 1056. Zhang H, Andrekopoulos C, Joseph J, Crow J, Kalyanaraman B. The carbonate kinase. J Biol Chem 285: 33154–33164, 2010. doi:10.1074/jbc.M110.143685. radical anion-induced covalent aggregation of human copper, zinc superoxide dis- mutase, and alpha-synuclein: intermediacy of tryptophan- and tyrosine-derived ox- 1068. Zoccali C, Bode-Böger S, Mallamaci F, Benedetto F, Tripepi G, Malatino L, idation products. Free Radic Biol Med 36: 1355–1365, 2004. doi:10.1016/j. Cataliotti A, Bellanuova I, Fermo I, Frölich J, Böger R. Plasma concentration of freeradbiomed.2004.02.038. asymmetrical dimethylarginine and mortality in patients with end-stage renal disease: a prospective study. Lancet 358: 2113–2117, 2001. doi:10.1016/S0140- 1057. Zhang L, Rao F, Zhang K, Khandrika S, Das M, Vaingankar SM, Bao X, Rana BK, Smith 6736(01)07217-8. DW, Wessel J, Salem RM, Rodriguez-Flores JL, Mahata SK, Schork NJ, Ziegler MG, O’Connor DT. Discovery of common human genetic variants of GTP cyclohydrolase 1069. Zorov DB, Juhaszova M, Sollott SJ. Mitochondrial reactive oxygen species (ROS) and 1 (GCH1) governing nitric oxide, autonomic activity, and cardiovascular risk. J Clin ROS-induced ROS release. Physiol Rev 94: 909–950, 2014. doi:10.1152/physrev. Invest 117: 2658–2671, 2007. doi:10.1172/JCI31093. 00026.2013. 1058. Zhang M, Brewer AC, Schröder K, Santos CXC, Grieve DJ, Wang M, Anilkumar N, 1070. Zou MH, Shi C, Cohen RA. Oxidation of the zinc-thiolate complex and uncoupling of Yu B, Dong X, Walker SJ, Brandes RP, Shah AM. NADPH oxidase-4 mediates endothelial nitric oxide synthase by peroxynitrite. J Clin Invest 109: 817–826, 2002. protection against chronic load-induced stress in mouse hearts by enhancing angio- doi:10.1172/JCI0214442. genesis. Proc Natl Acad Sci USA 107: 18121–18126, 2010. doi:10.1073/pnas. 1009700107. 1071. Zuckerbraun BS, Shiva S, Ifedigbo E, Mathier MA, Mollen KP, Rao J, Bauer PM, Choi JJ, Curtis E, Choi AM, Gladwin MT. Nitrite potently inhibits hypoxic and inflamma- 1059. Zhang R, Brennan M-L, Fu X, Aviles RJ, Pearce GL, Penn MS, Topol EJ, Sprecher DL, tory pulmonary arterial hypertension and smooth muscle proliferation via xanthine Hazen SL. Association between myeloperoxidase levels and risk of coronary artery oxidoreductase-dependent nitric oxide generation. Circulation 121: 98–109, 2010. disease. JAMA 286: 2136–2142, 2001. doi:10.1001/jama.286.17.2136. doi:10.1161/CIRCULATIONAHA.109.891077. 1060. Zhang R, Hess DT, Reynolds JD, Stamler JS. Hemoglobin S-nitrosylation plays an essential role in cardioprotection. J Clin Invest 126: 4654–4658, 2016. doi:10.1172/ 1072. Zweier JL, Duke SS, Kuppusamy P, Sylvester JT, Gabrielson EW. Electron paramag- JCI90425. netic resonance evidence that cellular oxygen toxicity is caused by the generation of superoxide and hydroxyl free radicals. FEBS Lett 252: 12–16, 1989. doi:10.1016/ 1061. Zhang Z, Naughton DP, Blake DR, Benjamin N, Stevens CR, Winyard PG, Symons 0014-5793(89)80881-6. MC, Harrison R. Human xanthine oxidase converts nitrite ions into nitric oxide (NO). Biochem Soc Trans 25: 524S, 1997. doi:10.1042/bst025524s. 1073. Zweier JL, Li H, Samouilov A, Liu X. Mechanisms of nitrite reduction to nitric oxide in the heart and vessel wall. Nitric Oxide 22: 83–90, 2010. doi:10.1016/j.niox.2009. 1062. Zhao K, Luo G, Giannelli S, Szeto HH. Mitochondria-targeted peptide prevents 12.004. mitochondrial depolarization and apoptosis induced by tert-butyl hydroperoxide in neuronal cell lines. Biochem Pharmacol 70: 1796–1806, 2005. doi:10.1016/j.bcp. 1074. Zweier JL, Samouilov A, Kuppusamy P. Non-enzymatic nitric oxide synthesis in 2005.08.022. biological systems. Biochim Biophys Acta 1411: 250–262, 1999. doi:10.1016/S0005- 2728(99)00018-3. 1063. Zhao R, Ma X, Xie X, Shen GX. Involvement of NADPH oxidase in oxidized LDL- induced upregulation of heat shock factor-1 and plasminogen activator inhibitor-1 in 1075. Zweier JL, Wang P, Samouilov A, Kuppusamy P. Enzyme-independent formation vascular endothelial cells. Am J Physiol Endocrinol Metab 297: E104–E111, 2009. of nitric oxide in biological tissues. Nat Med 1: 804–809, 1995. doi:10.1152/ajpendo.91023.2008. doi:10.1038/nm0895-804.

Physiol Rev • VOL 99 • JANUARY 2019 • www.prv.org 379 Downloaded from www.physiology.org/journal/physrev by ${individualUser.givenNames} ${individualUser.surname} (130.070.008.131) on November 29, 2018. Copyright © 2019 the American Physiological Society. All rights reserved.