AHA Scientific Statement Measurement of Reactive Oxygen Species, Reactive Nitrogen Species, and Redox-Dependent Signaling in the Cardiovascular System A Scientific Statement From the American Heart Association Kathy K. Griendling, PhD, FAHA, Co-Chair*; Rhian M. Touyz, MD, PhD, FAHA, Co-Chair*; Jay L. Zweier, MD, FAHA; Sergey Dikalov, PhD; William Chilian, PhD, FAHA; Yeong-Renn Chen, PhD; David G. Harrison, MD, FAHA; Aruni Bhatnagar, PhD, FAHA; on behalf of the American Heart Association Council on Basic Cardiovascular Sciences Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Abstract—Reactive oxygen species and reactive nitrogen species are biological molecules that play important roles in cardiovascular physiology and contribute to disease initiation, progression, and severity. Because of their ephemeral nature and rapid reactivity, these species are difficult to measure directly with high accuracy and precision. In this statement, we review current methods for measuring these species and the secondary products they generate and suggest approaches for measuring redox status, oxidative stress, and the production of individual reactive oxygen and nitrogen species. We discuss the strengths and limitations of different methods and the relative specificity and suitability of these methods for measuring the concentrations of reactive oxygen and reactive nitrogen species in cells, tissues, and biological fluids. We provide specific guidelines, through expert opinion, for choosing reliable and reproducible assays for different experimental and clinical situations. These guidelines are intended to help investigators and clinical researchers avoid experimental error and ensure high-quality measurements of these important biological species. (Circ Res. 2016;119:e39-e75. DOI: 10.1161/RES.0000000000000110.) Key Words: AHA Scientific Statements ◼ free radicals ◼ oxidants ◼ peroxides ◼ reactive nitrogen species ◼ reactive oxygen species R eactive oxygen species (ROS) and reactive nitrogen species (RNS) are produced in virtually all eukaryotic cells. These molecules regulate several physiological processes including proliferation, migration, hypertrophy, differentiation, cytoskeletal dynamics, and metabolism, but when available in excess, they react with lipids, proteins, and nucleic acids, thereby altering structural and functional properties of target molecules and leading to extensive tissue dysfunction and injury. ROS and RNS have been implicated in the development of many cardiovascular diseases, including hypertension, heart failure, atherosclerosis, and cardiovascular and renal complications of diabetes mellitus. Cardiovascular injury caused by ischemia-reperfusion or exposure to environmental pollutants, tobacco smoke, and ionizing radiation has also been linked to excessive ROS or RNS accumulation. Despite extensive data implicating oxidative stress in animal models of cardiovascular disease and dysfunction, there is still no conclusive evidence that ROS/RNS are fundamentally involved in the pathogenesis of cardiovascular disease in humans, and there is a paucity of convincing evidence that *Drs Griendling and Touyz contributed equally to this work. The American Heart Association makes every effort to avoid any actual or potential conflicts of interest that may arise as a result of an outside relationship or a personal, professional, or business interest of a member of the writing panel. Specifically, all members of the writing group are required to complete and submit a Disclosure Questionnaire showing all such relationships that might be perceived as real or potential conflicts of interest. This statement was approved by the American Heart Association Science Advisory and Coordinating Committee on February 15, 2016, and the American Heart Association Executive Committee on April 25, 2016. A copy of the document is available at http://professional.heart.org/statements by using either “Search for Guidelines & Statements” or the “Browse by Topic” area. To purchase additional reprints, call 843-216-2533 or e-mail kelle.ramsay@ wolterskluwer.com. The American Heart Association requests that this document be cited as follows: Griendling KK, Touyz RM, Zweier JL, Dikalov S, Chilian W, Chen Y-R, Harrison DG, Bhatnagar A; on behalf of the American Heart Association Council on Basic Cardiovascular Sciences. Measurement of reactive oxygen species, reactive nitrogen species, and redox-dependent signaling in the cardiovascular system: a scientific statement from the American Heart Association. Circ Res. 2016;119:e39–e75. doi: 10.1161/RES.0000000000000110. Expert peer review of AHA Scientific Statements is conducted by the AHA Office of Science Operations. For more on AHA statements and guidelines development, visit http://professional.heart.org/statements. Select the “Guidelines & Statements” drop-down menu, then click “Publication Development.” Permissions: Multiple copies, modification, alteration, enhancement, and/or distribution of this document are not permitted without the express permission of the American Heart Association. Instructions for obtaining permission are located at http://www.heart.org/HEARTORG/General/CopyrightPermission-Guidelines_UCM_300404_Article.jsp. A link to the “Copyright Permissions Request Form” appears on the right side of the page. © 2016 American Heart Association, Inc. Circulation Research is available at http://circ.ahajournals.org DOI: 10.1161/RES.0000000000000110 e39 e40 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 antioxidants are effective therapies in cardiovascular medicine. Yet the belief remains that oxidative stress contributes to many cardiovascular pathologies. Putative reasons for this paradox relate to the relatively weak nature of the antioxidants used in clinical trials, an incomplete understanding of the complex molecular mechanisms whereby ROS cause pathological changes, the difficulty in extrapolating findings from experimental models to clinical scenarios, and the methodological challenges relating to accurate measurement of ROS in the cardiovascular system. Although myriad techniques, protocols, assays, and commercial kits have been developed to measure ROS, and numerous biomarkers have been used to study human oxidative stress, many of these assays are used inappropriately or without due experimental diligence, resulting in potential inaccuracies and artifacts, as well as much confusion in the literature. Moreover, the cardiovascular researcher who is a novice in the field of free radical biology may be overwhelmed in choosing from the many tools and methods currently available to measure ROS, RNS, and oxidative stress. Objectives The purpose of this scientific statement is to review current methods for measuring ROS, RNS, and their secondary products; to discuss the strengths and limitations of specific methodological approaches; and to provide guidelines, through expert opinion, for reliable and reproducible techniques for quantifying these species and for elucidating their role in redox-dependent signaling and cell injury. Because of the diversity of experimental systems in which the roles of these species are interrogated, we offer here only general methodological guidelines and assistance with decisions as to which assay to choose. This review does not focus on specific protocols, and the reader is referred to appropriate methodology-based articles for specific protocols. Measurements of ROS and RNS in clinical studies are discussed separately to recommend a distinct set of measurements that should be made to assess systemic oxidative stress and to document oxidative injury in clinical scenarios. Background Chemically reactive molecules containing ROS or RNS are generated endogenously in most cells during the course of normal metabolism, during disease development, and in response to tissue injury. These species are also produced on exposure to exogenous insults such as environmental pollutants, tobacco smoke, and ionizing radiation. Most ROS and RNS are relatively unstable oxygen- or nitrogen-centered free radicals that contain unpaired electrons (biological half-life is microseconds to milliseconds); however, these species also include more stable species such as peroxides and nitrosothiols (biological half-life is seconds). Major ROS and RNS generated in biological systems are listed in Table 1. Although nitrogen itself is a stable diatomic gas, molecular oxygen in the ground state is a free radical with 2 unpaired electrons in its outmost shell. Because both these electrons have parallel spin, diatomic oxygen is relatively unreactive. However, during its utilization in the mitochondria, oxygen accepts 1 electron at a time, which leads to the generation of .− partially reduced species such as superoxide (O2 ). Most of the oxygen is reduced to water, but incomplete reduction of .− O2 leads to the generation of O2 , hydrogen peroxide (H2O2), and hydroxyl radical (•OH). Hence, normal utilization of oxygen during mitochondrial respiration results in continuous ROS formation. Superoxide is unstable, with a lifetime of milliseconds at neutral pH, and in aqueous solution it spontaneously reacts or dismutates to yield H2O2 and O2. Hydrogen peroxide by itself is fairly unreactive, but it oxidizes Fe(II) to Fe(III) to generate hydroxyl radicals in a .− process known as the Fenton reaction.1,2 In vitro, O2 can reduce the oxidized metal generated by the Fenton reaction to regenerate Fe(II) (Haber-Weiss reaction); however, the in vivo identity of the reductant is less clear, and Fe(III) can be reduced with variable efficiency by a variety of cell reductants. In addition to iron, other metals such as Cu, Cr, Co, V, and to a lesser extent Ni can catalyze Fenton chemistry,3 but in most biological systems the role of iron is critical. In most vertebrates, iron is bound to ferritin, which oxidizes Fe(II) at the ferroxidase center, thereby decreasing Fe(II) available for the Fenton reaction.4 Notably, dysregulation of iron homeostasis because of injury or hereditary ferritinopathies results in increased oxidative damage. In the mitochondria, destruction of the [4Fe-4S] cluster (eg, aconitase) can also result in the release of Fenton-active Fe(II), leading to increased ROS production.5 This could be a mechanism whereby non-Fenton metals (such as Ag and Hg) exert toxicity by increasing ROS production.6 The hydroxyl radical generated by the Fenton reaction is extremely reactive and short-lived. It reacts indiscriminately with most cell constituents and leads to the generation of secondary free radical species that deplete antioxidants and oxidize thiols and unsaturated lipids. In contrast to the indiscriminate reactivity of the hydroxyl radicals, ROS generated by enzymatic reactions such as those catalyzed by nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, cytochrome P450 enzymes, and glucose oxidase are generally considered to serve signaling functions, although localized generation of hydroxyl-like species (eg, in the endoplasmic reticulum) could also affect the expression of specific genes or their coactivators.7 NO is a diatomic molecule containing 1 atom of oxygen (O, 8 electrons) and 1 atom of nitrogen (N, 7 electrons) and therefore contains 1 unpaired electron, which makes it a free radical.8 NO is a π-radical where the unpaired electron is delocalized between nitrogen and oxygen atoms, rendering it relatively stable. NO can be isolated as a gas and is stable in the absence of oxygen. In mammalian cells, NO is enzymatically formed by nitric oxide synthases (NOS), which oxidize l-arginine. NO acts as a second messenger to modulate numerous biological processes, including endothelial function, vascular smooth muscle cell contraction/dilation, inflamma.− tion, neuroplasticity, and cytotoxicity. In the presence of O2 , NO is converted into the strong oxidant peroxynitrite. NO can also react with oxygen to form other RNS and with thiols to generate nitrosothiols (RSNO). Both ROS and RNS are highly reactive, and as a result, they have short half-lives in biological environments. Therefore, these species are difficult to measure directly, and even indirect estimates of their abundance and reactivity are challenging. Although many approaches have been adopted Griendling et al Measurement of Reactive Oxygen Species e41 Table 1. Major Biological Reactive Oxygen and Reactive Nitrogen Species Formula Name Comment Reactive oxygen species O2 1 Singlet oxygen H2O2 Hydrogen peroxide HOO∙ Hydroperoxyl radical HO∙ Hydroxy radical O2 Superoxide anion .- O3 Ozone Electronically excited state of molecular oxygen Simplest peroxide with oxygen-oxygen single bond Free radical; protonated form of superoxide Free radical; neutral form of hydroxide ion (HO−) Free radical; dioxide (1-); product of 1 e− reduction of dioxygen Trioxygen; an allotrope of oxygen OCl− Hypochloride anion Salt of hypochlorous acid ROOH Hydroperoxide Organic peroxide, protonated peroxyl radical ROO∙ Peroxyl radical Free radical; lipid peroxyl radical when R is a lipid carbon RO∙ Alkoxyl radical Free radical; lipid alkoxyl radical when R is a lipid carbon Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Reactive nitrogen species ∙NO Nitric oxide N2O Nitrous oxide “Laughing gas,” reacts with oxygen to generate nitric oxide ONOO− . NO2 Peroxynitrite Product of the reaction between superoxide and nitric oxide Nitrogen dioxide N2O3 Dinitrogen trioxide ONOOH Peroxynitrous acid Nitrogen-centered free radical Free radical, derived from peroxynitrite Product of reaction between nitric oxide and nitrogen dioxide Protonated form of peroxynitrite NO− Nitroxyl anion Conjugate base of nitroxyl NO+ Nitrosyl cation Also, nitrosonium cation HNO2 Nitrous acid NO2Cl Nitrosyl chloride NO2− Nitrite NO2+ Nitronium ion Also nitryl ion, generated from the removal of e- from nitrogen dioxide or protonation of nitric acid RSNOs Nitrosothiols Formed by covalent addition of nitric oxide to cysteine and protein or nonprotein sulfhydryl residues to circumvent these limitations, and a wide range of methods have been devised to quantify ROS, RNS, and their secondary products, there are many considerations in the choice of assay and its application to a particular system. Following are specific methodological guidelines to assist researchers in choosing appropriate assays and designing specific controls to obtain reliable measures. Measurement of Superoxide, Hydrogen Peroxide, and Redox Status Introduction Many methods have been devised to measure oxygen free radicals and their derived oxidants with various levels of sen.− sitivity and accuracy. These include chemical assays for O2 , H2O2, or •OH generation, direct chemiluminescent assays or measurement of signal in the presence of chemiluminigenic probes, fluorescence detection in the presence of redox-sensitive probes, and either direct or spin trapping–based electron paramagnetic resonance (EPR) spectroscopy. Only EPR Weak monobasic acid Derived from nitrite and hypochlorous acid Anion, generated from nitric oxide enables the direct detection of free radicals, but other assays can be informative if used with proper controls. . Measurement of Superoxide Anion Radical (O2− ) EPR Assay: EPR Spin-Trapping EPR spectroscopy is the “gold standard” for unambiguous assessment of oxygen-centered free radical generation in biological systems. EPR, also known as electron spin resonance, is a method to detect molecules with unpaired electrons. When combined with an appropriate spin trap, it is a powerful and .− reliable technique to unequivocally measure O2 , •OH, and NO in biological samples (for NO, see Measurement of RNS). A variety of spin traps have been used to detect and quantify free radicals in cardiovascular tissues. Spin trapping with DMPO (5, 5-dimethyl-1-pyrroline-N-oxide) is the .− strategy most widely used to detect O2 or •OH generated in endothelial cells9,10 or myocytes (see Table 2 for comparison of spin traps).11–14 DMPO has also been used to assay for .− mitochondria-derived O2 generation by EPR.13,15 Because of the short half-life of DMPO/•OOH (t1/2 ≈45 seconds) and e42 Circulation Research August 19, 2016 Table 2. Summary of EPR Spin Probes Used to Measure Oxygen Free Radical(s) and Redox Status In Vitro, Ex Vivo, and In Vivo Spin Probes Nature of Probes System Applied DMPO Nitrone spin trap In vitro and ex vivo O2 , OH, and carbon-centered radicals in vitro and ex vivo DEPMPO Nitrone spin trap In vitro O2 and •OH generation mediated by the enzymes of cellular systems of endothelium and myocytes PBN or POBN Nitrone spin trap In vitro, ex vivo, and in vivo In vivo spin trapping of O2 -derived hydroxyl and alkyl radicals TAM radical (OXO63 and CT02-H) Trityl free radical In vitro and ex vivo Simultaneous determination of O2 and oxygen consumption in vitro and ex vivo TEMPOL Stable nitroxide In vitro and ex vivo Redox status in vitro and ex vivo with X-band EPR PCA Stable nitroxide In vitro and in vivo In vivo redoximetry with L-band EPR TEMPONE Stable nitroxide In vivo Biradical spin probe In vitro and ex vivo RSSR .− Measurements • .− .− .− In vivo redoximetry with PEDRI Detection of reduced thiols with X-band EPR .− Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 CM-H Hydroxylamine spin probe In vitro, ex vivo, and In vivo Detection of total cellular and tissue O2 by X-band EPR CP-H Hydroxylamine spin probe In vitro, ex vivo, and in vivo Measurements of O2 in membrane fractions, cells, tissue PP-H Hydroxylamine spin probe In vitro, ex vivo, and in vivo Measurements of extracellular O2 using X-band EPR CAT1-H Hydroxylamine spin probe In vitro and ex vivo Measurements of extracellular O2 TM-H Hydroxylamine spin probe In vitro and ex vivo Detection of cellular and mitochondrial O2 mitoTEMPO-H Hydroxylamine spin probe In vitro, ex vivo, and in vivo .− .− .− .− .− Mitochondria-targeted detection of O2 EPR indicates electron paramagnetic resonance spectroscopy; and PEDRI, proton-electron double-resonance imaging. .− the possibility that the O2 adduct can decompose to give an • OH adduct, a superoxide dismutase (SOD)–sensitive 4-line spectrum of DMPO/•OH is often detected. Therefore, a suitable SOD, polyethylene glycol–conjugated SOD (PEG-SOD), or SOD mimetic must be used to confirm that the detected .− DMPO/•OH is dependent on the formation of O2 .16–18 To dis.− tinguish O2 from •OH in biological systems or for in vivo • OH detection, DMSO (dimethyl sulfoxide), a specific hydroxyl radical scavenger, is used.19 An alternative to DMPO is DEPMPO [5-(diethoxyphosphoryl)-5-methyl-1-pyrroline-Noxide], a diethoxylphosphoryl derivative of DMPO, which .− has the advantage that it forms a more stable O2 adduct (t1/2 ≈15 minutes).20 PBN (α-phenyl-N-t-butylnitrone) or POBN [α-(4-pyridyl-1-oxide)-N-tert-butylnitrone] have been used effectively for in vivo spin trapping of ROS production.21,22 .− Although these traps are useful for trapping O2 -derived hy.− droxyl and alkyl radicals, stable O2 adducts are not typically formed with PBN or POBN. In vivo or in situ direct EPR or EPR spin trapping with DMPO has been demonstrated in the Langendorff-perfused .− heart, which provides direct evidence of O2 generation during myocardial ischemia and reperfusion injury.11,14,23,24 These measurements also detect the carbon-centered radical formed by ischemia and reperfusion injury–mediated lipid oxidation or lipid peroxidation (DMPO/•R).14,24 Ex vivo EPR spin trapping with DMPO has been used recently to quantify mi.− tochondria-derived O2 in the rat heart.15 DEPMPO has been used extensively to investigate molecular mechanisms of ROS generation mediated by enzymes such as endothelial NOS (eNOS),25 NADPH oxidases, xanthine oxidase,26 and the mitochondrial electron transport chain in vitro.27–30 In summary, EPR spin trapping with either DMPO or DEPMPO is a useful approach for measuring oxygen-, carbon-, nitrogen-, and sulfur-centered radicals. These spin traps .− .− can measure O2 generation in cells, as well as O2 or •OH generation mediated by cardiovascular enzymes and mitochondria in vitro and in the isolated heart ex vivo. However, when used with tissues or cells, they have the limitation that spin adducts can be converted to EPR silent products, so that failure to detect a signal does not rule out low-level radical generation.31 In vivo spin trapping of ROS with nitrones can be performed but might not provide the adduct stability and resultant sensitivity to detect low levels of radical generation. Recent advances in the development of stabilized/protected nitrone spin traps bound to cyclodextran or calixpyrrole groups offer promise in addressing this limitation.32,33 Alternative approaches for the detection of spin trap adducts or their metabolites include high-performance liquid chromatography (HPLC) and mass spectroscopy,34,35 high-field nuclear magnetic resonance,36 and antibody-immune based detection.37,38 EPR Assay: Spin Probes With Cyclic Hydroxylamine or Other Hydroxylamines Several cyclic hydroxylamines, including CM-H (1-hydroxy3-methoxycarbonyl-2,2,5,5-tetramethylpyrrolidine), CP-H, PP-H, CAT1-H, and mitochondria-targeted mitoTEMPO-H, .− have been used widely as spin probes to measure O2 production in the cardiovascular system, an approach that requires confirmation with SOD, PEG-SOD, or SOD mimetics.31,39,40 .− Cyclic hydroxylamines react rapidly with O2 , producing .− stable nitroxides, and allow site-specific O2 detection in the intracellular, extracellular, and mitochondrial compartments. However, 1-electron oxidation of cyclic hydroxylamine to nitroxide is also mediated by other cellular oxidants, including H2O2, peroxynitrite, hypochlorite, and compound I- and compound II-like species. This makes it necessary to use specific scavengers to identify the oxidant generating the signal. Because of their favorable cell permeability, cyclic Griendling et al Measurement of Reactive Oxygen Species e43 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 hydroxylamines can be used as spin probes to determine the redox status of the cardiovascular system in situ by EPR redoximetry as described in Measurement of Redox Status. .− CM-H has been used to detect intracellular O2 in cultured cells and tissue samples. To minimize hydroxylamine autoxidation, the spin probe solution must be prepared in argonbubbled ice-cold NaCl in the presence of the chelating agent diethylenetriamine-penta-acetic acid or a combination of deferoxamine and diethyldithiocarbamate.31 In complex in vivo .− systems, O2 -independent oxidation of the hydroxylamine to the nitroxide is possible, and the nitroxide product could be reduced enzymatically. Hence, these possibilities should be considered and appropriate controls included, such as SOD, PEG-SOD, or SOD mimetics.31,39,40 EPR with the spin probe CM-H has been used to detect and quantify angiotensin II (Ang II)–induced and Nox1.− dependent O2 production in the aorta,39 as well as NADPH.− and xanthine oxidase–dependent O2 production in models of atrial fibrillation.40 EPR with the cyclic hydroxylamine spin .− probe is an effective approach for studying intracellular O2 in vessels and other tissues.31 EPR Assay: Triarylmethyl (Trityl) Free Radical Probes Triarylmethyl (trityl)-based radicals (also called TAM radicals or probes) can be used for simultaneous measurements .− of O2 and oxygen.41 These probes provide >10-fold higher EPR sensitivity compared with all other types of spin probes and are well suited for in vivo EPR spectroscopy and imaging. CT02-H is an analogue of the fully substituted TAM radical .− CT-03, with a higher rate of reaction with O2 and an EPR doublet signal because of its aromatic hydrogen.42 On reac.− tion with O2 , CT02-H loses its EPR signal, accompanied by a change in color from green to purple, which provides an .− additional indication of O2 flux. In the cardiovascular system, stimulated decay of the EPR signal of TAMs (CT-03, CT-02, .− or OXO63) can be initiated by O2 , •OH, and alkyl peroxyl radical (ROO•).41 In vitro studies suggest that the reactivity of .− O2 with these TAM probes is much higher than that of •OH and ROO•. However, these probes are stable in the presence of biological reductants or oxidants, including ascorbate, glutathione, NADPH, H2O2, and NO. In combination with suitable SOD, PEG-SOD, or SOD mimetic controls, the use of TAM or CT02-H is advantageous over cyclic hydroxylamines .− for the detection of O2 because the probe does not react with H2O2, and very low concentrations of the probe are needed for the measurement. TAMs can also be used for EPR oximetry to determine the oxygen concentration and consumption rate in cardiovascular systems of cultured cells or tissue homogenates.41 Unlike cyclic hydroxylamine and nitrone spin traps, conventional TAM radical probes (OXO63, CT03, and CT02-H) have 3 or 2 carboxylate groups, and their negative .− charge limits their use to extracellular O2 detection. This limitation can be overcome by esterified TAM probes, such as AMT-02, that are taken up in cells and can provide intracellular .− O2 detection. EPR with TAM has been applied to monitor NADPH .− oxidase–mediated O2 release from stimulated neutrophils, endothelial cells, and other cellular systems.41 TAM spin .− probes enable simultaneous measurements of O2 and oxygen concentration and consumption in biological systems associated with respiratory burst and anoxia-reoxygenation.42 The probe CT02-H is more sensitive than nitrone spin traps in mea.− suring low O2 flux. It is also uniquely well suited for in vivo EPR imaging applications (see Measurement of Redox Status). Cytochrome c Reduction Assay Cytochrome c (or acetylated cytochrome c) reduction is an.− other assay used to measure O2 production in vascular and myocardial cells and tissue.40,43,44 Ferricytochrome c accepts .− an electron from O2 to form ferrocytochrome c, which results in an increase in absorbance at 550 nm. As a control, the assay must also be performed in the presence of SOD or PEG-SOD. Acetylation of cytochrome c reduces the charge effect of the lysine residues of cytochrome c; thus, use of acetylated cytochrome c improves the electron transfer .− specificity for O2 .45 Potential drawbacks of the use of cytochrome c relate to its reactivity in biological systems. Both cytochrome c and acetylated cytochrome c show peroxidase activity in the presence of ROS because of oxidation of the sixth heme ligand methionine.46 Furthermore, cytochrome c can be reduced by a variety of biological reducing agents, including the NOS and P450 reductases. In the mitochondrial system, higher electron transfer efficiency mediated by com.− plex III than from O2 to cytochrome c limits its usefulness. A suitable mitochondrial inhibitor and SOD should be used to distinguish whether the reduction of ferrocytochrome c is caused by electron leakage or electron transfer. Thus, depending on the system, specificity could be a concern. Controls with SOD, PEG-SOD, or an SOD mimetic or other appropriate scavengers are essential. The cytochrome c reduction assay has been used to mea.− sure O2 production in biological systems in vitro and oc.− casionally ex vivo.44 It is best suited for measuring O2 in controlled systems that are free of enzymes that can directly .− reduce it or for measuring extracellular O2 in cellular systems. A major limitation of the cytochrome c assay is its rela.− tive insensitivity except for high levels of O2 production. For this reason, it has been used predominantly for measurement .− of O2 production by phagocytic cells and is not often used to .− detect the low levels of O2 produced by nonphagocytic cells. Nitroblue Tetrazolium Assay The nitroblue tetrazolium assay is based on the conversion of the water-soluble yellow-colored dye to its blue formazan .− product by O2 . This assay is well suited for use in in situ .− microscopic assays detecting O2 release from individual cells. The method can be modified for quantitative purposes by dissolving nitroblue tetrazolium in DMSO under alkaline conditions and measuring its absorbance at 620 nm 47 with . − a microplate reader, and it has been used to measure O2 generation by neutrophils or phagocytes during the inflammatory process.47,48 Unfortunately, this assay is not .− specific for O2 because of the enzyme-mediated (NADPH oxidase and NOS reductase) formation of blue formazan. Because nonspecific oxidation of nitroblue tetrazolium can occur, appropriate controls with SOD, PEG-SOD, or SOD mimetics are essential. Therefore, it is recommended that this assay be used mostly for high-throughput, in vitro ROS measurements. e44 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Fluorescence Analysis of Dihydroethidium or Hydroethidium .− Dihydroethidium (DHE) is widely used to detect O2 generation in situ, in vitro, and ex vivo in cardiovascular systems because the DHE probe is user-friendly and has high sensi.− tivity. Oxidation of DHE by O2 yields 2-hydoxyethidium + 49 (2-OH-E ). However, other oxidants such as ONOO−, •OH, H2O2, compound I, and compound II can produce a 2-electron oxidation product, E+, with similar fluorescence charac.− teristics. Detection of O2 by these fluorescent probes is not specific because of overlapping fluorescence of the superoxide-specific 2OH-E+ and the nonspecific oxidation ethidium (E+) product.49 DHE can also be oxidized spontaneously because of atmospheric oxidation or light-induced oxidation, resulting in the formation of ethidium. Therefore, to avoid artificial signals, it is essential to pretreat DHE with Dowex cation exchange beads immediately before use to absorb any ethidium contamination. New protocols in which fluorescent .− settings are optimized to measure the O2 -specific product and to minimize interference by nonspecific DHE oxidation products have been developed by changing the excitation from 480 to 405 nm, which significantly reduces the emission intensity of E+ compared with 2OH-E+.50 The most unequivocal and .− quantitative detection of intracellular O2 by 2-OH-E+ in tissue specimens can be obtained with an HPLC equipped with a fluorescence or electrochemical detector, or by liquid chromatography (LC)/mass spectrometry (MS).31,51 In combination with the technique of EPR redoximetry, .− DHE or hydroethidium has been used to monitor O2 production during myocardial ischemia and reperfusion in vivo52 .− and ex vivo.53 In situ DHE imaging of O2 generation in the myocardium has been applied in experimental models of cardiac hypertrophy induced by pressure overload.54 DHE has also been used extensively in the vasculature.55–57 Combined with HPLC instrumentation, 2-OH-E+ formation from frozen myocardial tissue extracts preincubated with DHE has been .− reported as the indicator of O2 production associated with pathological cardiac hypertrophy.58 In all of the above applications, it is critical that appropriate controls with SOD, PEG-SOD, or SOD mimetics are used.59 When combined with HPLC and LC/MS instrumentation, the DHE probe and the fingerprint of 2-OH-E+ are a .− useful approach to measure and quantify intracellular O2 production in cells and tissues. The approach is highly sensitive and valuable in a range of applications including translational .− research in obtaining O2 measurements from patient-derived cells and tissues. When used with microscopy, it is imperative to understand that DHE is detected as blue fluorescence within the cytoplasm and only becomes a bright fluorescent red when it intercalates within the DNA. Hence, in the absence of DNA, for example, in platelets, DHE should not be used for ROS measurement. Fluorescence Analysis of Hydrocyans Hydrocyanines or deuterocyanines are reduced dyes capable of detecting 1 or more types of ROS.60,61 These dyes exhibit little or negligible fluorescence in the reduced state; however, on reaction with ROS, they are oxidized, which results in a large increase in fluorescence intensity. The stability of reduced dyes is enhanced by replacing hydrogen with deuterium. Deuterocyanines are thus more protected from auto-oxidation and exhibit lower levels of background fluorescence in cell culture. The cyanine dyes have emission wavelengths from 500 to 1100 nm, and the corresponding reduced dyes have longer half-lives than DHE and thus have potential for in vivo application. In many cases, hydrocyanines or deuterocyanines are membrane permeable and can accumulate in cells or tissues. Once inside cells, hydrocyanines or deuterocyanines are reoxidized by ROS, become membrane impermeable, and are trapped within the cells. For example, the reduced dye hydroIR-676 exhibits negligible fluorescence; however, on reaction .− with O2 , IR-676 exhibits a 100-fold increase in fluorescence (λex=675 nm and λem=693 nm). In vitro studies indicate that hydro-Cy7 and hydro-Cy3 exhibit much higher selectivity for .− O2 and •OH than other ROS. Hydro-Cy3 and deuteron-Cy3 have been used to detect Ang II–mediated intracellular ROS production from aortic muscle cells and ROS production in en face preparations of live and explanted aortas.60 In the mouse model of lipopolysaccharide-induced acute inflammation, hydro-Cy7 has been used to image in vivo ROS generation by activated macrophages and neutrophils.60 As with other fluorescent dyes, SOD controls are important to ensure specificity. Mitochondria-Specific Fluorescent Probes Mitochondria-targeted hydroethidium and Mito-SOX (3,8-phenanthridinediamine, 5-[6′-triphenylphosphoniumhexyl]5,6 dihydro-6-phenyl), which show a chemistry similar to DHE or hydroethidium,49 are widely used to measure mitochondrial .− O2 in situ. Their use and limitations are similar to those described above for DHE.49 Measurement of mitochondrial .− O2 by Mito-SOX tracks well with electron spin resonance measurements and the signal produced by the more sensitive MitoTracker Red CM-H2XRos.62 Both Mito-SOX and MitoTracker Red CM-H2XRos are nonfluorescent in their reduced form and fluoresce red when oxidized. The advantage of these probes is that they provide information about .− mitochondria-localized O2 production; the disadvantage is that like other fluorescent probes, they are not fully quantitative and must be used with appropriate controls, such as mitochondria-targeted antioxidants. Mito-SOX has been used with flow cytometry to better quantify ROS, as described in H9c2 myocytes and coronary artery endothelial cells.63 Chemiluminescence Assays Several chemiluminescent probes have been used to measure .− O2 , including lucigenin, coelenterazine, and the luminol analogue L-012. Lucigenin (N,N′-dimethyl-9,9′-biacridinium dinitrate) undergoes 1 electron reduction to emit an amplified chemiluminescence signal that correlates with the amount of .− O2 present in the sample. This assay has been widely used .− for O2 detection in the cardiovascular system.40 In general, it is more applicable to cell-free assays than to intracellular .− O2 because of its limited permeability. Coelenterazine (2-(4-hydroxybenzyl)-6-(4-hydroxyphenyl)-8-benzyl-3,7-dihydroimidazo [1,2-a]pyrazin-3-1) is the luciferin chromophore in aquaporin. It has been used in membrane preparations to .− detect O2 and purportedly produces 15 times more light than .− lucigenin.64 Coelenterazine has been used to detect O2 in vas65,66 L-012 cular smooth muscle cells and adventitial fibroblasts. Griendling et al Measurement of Reactive Oxygen Species e45 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 (8-amino-5-chloro-7-phenylpyridol[3,4-d]pyridazine-1,4(2H,3H)dione sodium salt) produces luminescence at long wavelengths (530 nm) and is more sensitive than lucigenin.67 .− It has been used to detect extracellular O2 in whole blood and in membrane fractions from cells and tissue.67 However, L-012 has recently been shown to react with H2O2 and peroxidase to .− produce O2 , which makes the signal artificially inhibitable by .− SOD and thus unsuitable for O2 detection.68 The validity of the lucigenin assay has been questioned .− because O2 is generated by lucigenin itself.69 Lucigenin can .− uncouple flavoenzymes, thus triggering O2 in a concentration-dependent manner. The concentration of lucigenin used is therefore a critical parameter affecting the validity of this assay.70 It has been shown that the concentration of lucigenin should be ≤5 μmol/L, and extensive controls are required to rule out redox cycling.71,72 It is also important to document that the concentration used does not significantly increase oxygen consumption. Higher concentrations of lucigenin can attenuate endothelium-dependent relaxation by acetylcholine and have been shown to uncouple eNOS.70 An additional consideration is the concentration of NADPH used in cell-free assays. NADPH at a concentration of 100 μmol/L is most commonly used, because higher concentrations can lead to aberrant results. Thus, lucigenin is only useful if used at low concentrations (≤5 μmol/L) in fractionated cells or tissues with the appropriate concentration of NADPH, as well as SOD controls. . Recommendations for Measurement of O2− Figure 1 provides a comparison of methods for measuring .− O2 and H2O2. The cytochrome c reduction assay is suitable .− for measurements of O2 in controlled systems free of reducing equivalents or enzymes that couple to it. Therefore, cytochrome c is not recommended for use in cellular and tissue applications. In contrast, the nitroblue tetrazolium assay is ef.− .− fective in measuring O2 generation from cells with high O2 production and can be used as a high- throughput ROS assay system in vitro. Suitable SOD controls are important to establish specificity. Lucigenin is not recommended for use in .− measuring intracellular O2 because of its ability to uncouple .− flavoenzymes, resulting in O2 generation, but it has been used extensively to measure ROS generation in membrane fractions using NADPH as a substrate. For such an application, the concentration of lucigenin (≤5 μmol/L) is critical, because high concentrations of lucigenin can produce anomalous results.70 If lucigenin is used, it should be complemented with a second, independent assay. Similar considerations should also be applied to coelenterazine. Combined with appropriate SOD controls, fluorescence analysis with DHE is a useful approach to measure and image .− intracellular O2 production from cells and tissues because of its high sensitivity. Ideally, it should be combined with HPLC to ensure specificity. DHE should only be used in cells containing DNA. Mitochondria-specific fluorescent probes are useful for qualitative assessment, for example, ROS localization, but not for quantitative studies. Localization of ROS production can be confirmed by supplementation with cell-impermeable Cu,Zn-SOD, cell-permeable PEG-SOD, or mitochondria-targeted antioxidants such as mitoTEMPO.57 Hydrocyanines and deuterocyanines are useful as fluorescence probes with high persistence and high sensitivity, but further work is needed to characterize their specificity. These fluorescence-based methods are useful in a wide range of applications including translational research from patient-derived cells and tissues. The EPR spin-trapping technique provides the most direct and specific identification of oxygen free radicals, including .− O2 and •OH, in cellular and ex vivo systems; however, EPR with nitrone spin traps is not feasible with intact tissue samples. Failure to detect a signal does not rule out free radical or other ROS generation. Hydoxylamine probes with appro.− priate SOD controls are useful for the measurement of O2 . Hydroxylamine probes and nitroxides also enable assessment of redox state and stress and are useful in ex vivo and in vivo systems. Newly developed trityl probes or trityl-nitroxide bi.− radicals can provide more sensitive O2 measurements and are well suited for in vivo EPR and EPR imaging. Figure 1. Choice of assays to measure .− O2 and H2O2.. When choosing an assay to measure O2− or H2O2, one must first consider whether reactive oxygen species (ROS) production is intracellular or extracellular. Assays listed as primary (1°) are the most well accepted, secondary (2°) are more widely used, and tertiary (3°) are either less sensitive (cytochrome c) or should be verified with a second method (chemiluminescence). Cyt c indicates cytochrome c; DCFH-DA, dichlorodihydrofluorescein diacetate; DHE, dihydroethidium; EC, electrochemical; EPR, electron paramagnetic resonance spectroscopy; HPLC, high-performance liquid chromatography; and NBT, nitroblue tetrazolium. e46 Circulation Research August 19, 2016 Measurement of H2O2 Electrochemical Detection With H2O2 Electrode Electrochemical detection of H2O2 using an H2O2-specific electrode is a direct and reliable method for measuring H2O2 in vitro and has been used to assess H2O2 produced in endothelial cells and neutrophils.73 In combination with spin-trapping agents, electrochemical detection is effective for distinguish.− ing the production of H2O2 and O2 that results from NOS uncoupling.74 Electrochemical detection is useful in assessing H2O2 generated by isolated enzymes and for measuring H2O2 metabolism in the isolated mitochondria and cellular systems. However, this method is not widely used for in vivo measurements because of its poor sensitivity and difficulties with in vivo use of polarographic electrodes. Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Amplex Red In the presence of H2O2, Amplex Red is oxidized efficiently by horseradish peroxidase to a fluorescent product, resorufin, which can be measured using a fluorescent plate reader. The assay is a feasible and reliable method to measure extracellular H2O2 in vessels75 and the heart.76,77 The assay is also highly effective for assessing extramitochondrial H2O2 generated from isolated myocardial mitochondria76,77 and has been used to measure H2O2 released from mouse aorta.31,75 It has also been used to quantify external H2O2 released from mitochondria of the ischemic heart.76 The Amplex Red assay is highly recommended for measuring H2O2 released from mitochondria, cells, vessels, and cell-free systems. However, in tissues, the product (resorufin) induces nonspecific oxidation via other oxidant and radical-mediated reactions. Therefore, controls to correct for the fluorescent background must be performed with addition of SOD and catalase. Dichlorodihydrofluorescein Diacetate Dichlorodihydrofluorescein diacetate (DCFH-DA) is a cellpermeable and widely used probe to detect intracellular production of H2O2. It is commonly used for measuring H2O2 or oxidant stress in cells or tissues in combination with confocal microscopy or flow cytometry. DCFH has been used to image H2O2 in vessels and to evaluate mitochondrial H2O2 as the source of flow-induced dilation in coronary resistance arteries,78 as well as H2O2 levels of carotid artery and aortic smooth muscle cells during the progression of atherosclerotic lesion development.79 DCFH-DA has also been used to explore iron-mediated redox-signaling mechanisms in the cardiovascular system80 and to investigate doxorubicin-induced cardiotoxicity.81 Because DCF does not directly react with H2O2 to form a fluorescent product, DCF fluorescence must be used with caution as an indicator of H2O2. Compounds I and II are reactive 1-electron oxidizing intermediates that are formed from peroxidase or heme proteins on reaction with H2O2.82,83 They can oxidize DCFH to DCF, leading to artificial fluorescence in cells and tissue. Fe2+, a redox-active metal, can promote DCFH oxidation in the presence of O2 or H2O2, which can pose a serious problem when the assay is applied to myocardial reperfusion or vascular inflammation. The 1-electron oxidation intermediate, DCF− (DCF semiquinone anion radical), .− is a source of O2 ,83,84 which triggers redox cycling and leads to artificial amplification of DCF fluorescence. In view of these limitations, assays using DCFH or DCFH-DA must be conducted along with SOD and catalase controls to establish specificity; however, because of the potential for artifacts, this probe is not recommended for measuring intracellular ROS.59 HyPer Probe A redox-active biosensor, HyPer, is another approach for monitoring intracellular H2O2 levels. The design of HyPer is based on OxyR, a natural bacterial H2O2 sensor. HyPer is the first genetically encoded fluorescent sensor to detect intracellular H2O2. HyPer is highly sensitive to H2O2 (~ submicromo.− lar) but is insensitive to other oxidants, such as O2 , GSSG (oxidized glutathione), NO, and peroxynitrite. It does not cause artificial ROS generation and is capable of detecting fast changes in H2O2 concentration. HyPer has been used for in vivo imaging of H2O2 in cardiac myocytes from eNOS−/− mice and mice deficient in neuronal NOS (nNOS−/−)85 and for detecting in situ H2O2 synthesis stimulated by Ang II in cardiac myocytes. HyPer can be expressed in cardiac myocytes of adult mice through tail vein injection of HyPer lentivirus.85,86 HyPer is useful in monitoring intracellular H2O2 in situ. A major advantage is that it is genetically encoded and so can be targeted to specific compartments, such as the endoplasmic reticulum and mitochondria.87 Care must be taken, however, to avoid quenching of H2O2 in these compartments by the presence of the probe. Moreover, despite its high specificity for H2O2, the fluorescence signal of cytosolic HyPer can respond to pH changes, decreasing with acidosis and increasing with alkalosis. Thus, the HyPer assay should be conducted with side-by-side pH monitoring.88 Furthermore, the HyPer fluorescent response can adapt to repeated oxidation and can be insensitive to reducing stimuli. HyPer responses thus must be interpreted carefully.88 Recent modifications (eg, HyPer-3) have overcome some of these difficulties and allow expanded dynamic range and ratiometric imaging.89 Mitochondrial Probes MitoB ([3-hydroxybenzyl]triphenylphosphonium bromide) accumulates in the mitochondria by virtue of a triphenylphosphonium cation component. The arylboronic moiety of MitoB reacts with H2O2 to form a phenol product, MitoP, in the mitochondrial matrix. H2O2 concentration is determined by assessing the ratio of MitoP to MitoB as measured by MS with d15-MitoP and d15-MitoB used as standards.90,91 As such, this assay can be used in cultured cells and tissue. A limitation of this method is the need for sophisticated equipment not readily available in many laboratories. Dickinson and Chang92 recently described a mitochondriatargeted boronate-based peroxy yellow 1 probe containing a triphenylphosphonium group that allows selective detection of H2O2 within the mitochondria (MitoPY1). This new fluorescent probe permits imaging H2O2 within the mitochondria of living cells using confocal microscopy and flow cytometry. Recommendations for Measurement of H2O2 Figure 1 provides decision support for choosing a method to measure H2O2. Electrochemical detection is useful in assessing H2O2 generation by isolated enzymatic, mitochondrial and cellular systems; however, it is not suited for in vivo systems Griendling et al Measurement of Reactive Oxygen Species e47 because of its limited sensitivity and difficulties associated with in vivo use of polarigraphic electrodes. The Amplex Red assay is suitable for measuring H2O2 released from mitochondria, cells, vessels, and cell-free systems; however, in tissues, there is the potential for nonspecific oxidation so that controls to correct for the fluorescent background must be performed with addition of SOD and catalase. HyPer can be used to monitor intracellular H2O2 in situ. Because it is genetically encoded, the protein can be targeted to specific cellular compartments. The fluorescence ratio of HyPer changes with pH; therefore, it should be used with parallel pH monitoring. Mitochondriaspecific production of H2O2 can be measured with MitoB. Measurement of Redox Status Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Redox status refers to the tendency or ability of the tissue or cell to accept (more oxidized) or donate (more reduced) electrons. The biological redox status is often reflected by the balance of GSH (reduced glutathione)/GSSG, NAD+/ NADH, and NADP+/NADPH in a cell or organ, as well as by the balance of several sets of metabolites (eg, lactate and pyruvate, β-hydroxybutyrate, and acetoacetate) whose interconversion is dependent on these ratios, free radical reactions, and reversible/irreversible modifications of redox proteins. Although abnormal redox status has been linked to several conditions such as hypoxia, ischemia and reperfusion, hypertension, atherosclerosis, shock, and sepsis, accurate assessment of redox status is difficult. EPR-based techniques and measurement of redox couples such as GSH/ GSSG and cysteine/cystine (Cys/CySS) represent the most common approaches. The NADPH/NADP+ ratio is also an important determinant of the cellular redox status, but it is difficult to measure because no methods are available to measure the concentration of free NADPH or NADP+ in the cell. The NADH:NAD+ ratio, however, can be estimated indirectly by measuring the pyruvate to lactate ratio, which is regulated by the reaction catalyzed by lactate dehydrogenase, although in such measurements it is important to ensure that the reaction is at or near equilibrium.93 Levels of free NAD+/NADH can also be measured with newly developed biosensors or nuclear magnetic resonance using the magnetic enhancement technique,94,95 but these methods will not be discussed further here. EPR Redoximetry With Spin Probe Hydroxylamines and Nitroxides In oxidized conditions such as those in the postischemic myocardium, cyclic hydroxylamines can serve as a redox substrate to determine the redox activity of tissue specimens using EPR. The assay can be combined with a nitroxide probe such as TEMPOL to assess the level of reductive stress in the tissue specimen. This approach is applicable to in vivo measurements of whole animals and translational research using human samples; however, it cannot determine the redox status of thiols that are involved in oxidative posttranslational modifications (Oxidative Modification of Proteins). To obtain a complete picture of redox status, additional assays should be conducted, including analysis of GSH and GSSG (by an enzymatic recycling assay), measurement of free thiols by the biradical spin probe RSSR [bis-(2,2,5,5-tetramethyl3-imidazoline-1-oxyl-4-yl) disulfide], redox immunoblotting to assess protein oxidation, and HPLC analysis of oxidatively modified amino acids. In Vivo Redoximetry With Low-Frequency EPR Spectroscopy and Imaging In vivo redoximetry provides noninvasive in situ measurement of redox status. With the use of nitroxide probes, this approach has proved useful in monitoring in situ redox alterations of the myocardium during ischemia and reperfusion.96–98 Nitroxides are paramagnetic and are reduced or reoxidized depending on the redox environment. By measuring the shift of redox status from more reduced to more oxidized, one can determine how redox alterations correlate with disease progression.52,96 Trityl-nitroxide is a biradical that can also be used for in vivo redoximetry. With this biradical, the sharp trityl signal increases with nitroxide reduction as opposed to the loss of the nitroxide EPR signal. It can be used to simultaneously monitor oxygenation and redox alterations.99 With the addition of magnetic field gradients, EPR images of the free radical probe and its reductive clearance can be performed.100–102 This oxy-redoximetric assay is highly promising, and ex vivo cardiac applications have been performed with nitroxide probes.103 For in vivo redoximetry using nitroxides, it is important to confirm that the signal decay is caused by reduction of the nitroxides to the diamagnetic hydroxylamine. The hydroxylamine can be reoxidized to paramagnetic nitroxide by ferricyanide. Ferricyanide is thus used to confirm reduction to the hydroxylamine. In vivo EPR redoximetry is considered to be the “gold standard” for measuring redox physiology in the in vivo setting under physiological and pathological conditions. In Vivo Redox Imaging Using Proton-Electron DoubleResonance Imaging This approach utilizes the Overhauser effect to enable in vivo imaging of free radicals in the heart. The redox state of the tissue is measured by combining a nitroxide spin probe or biradical with proton-electron double-resonance imaging (PEDRI) instrumentation, enabling nuclear magnetic resonance/magnetic resonance imaging–based detection and imaging of the redox state based on saturation of the EPR resonance. As described above, nitroxide spin probes enable measurement of the tissue redox state based on their reduction to diamagnetic hydroxylamines.93,98,101 PEDRI instrumentation enables imaging of the in vivo distribution and metabolism of nitroxide radicals in living mice, which enables the redox status of the vascular system to be visualized.98 PEDRI has been applied to image nitroxide radicals in the isolated beating rat heart, with visualization of radical uptake, distribution, and clearance.101 When PEDRI is applied to acquire in vivo redox images of the beating heart using nitroxides as the redox-sensitive probe, a nitroxide radical with longer relaxation time and sharp EPR line width can offer better sensitivity. In vivo, PEDRI provides an enhanced form of in vivo redoximetry along with spatial mapping. It is a powerful tool for measuring and visualizing the redox distribution and clearance in the isolated heart and in animal models of disease, but it requires very specialized equipment and expertise that are not commonly available in most laboratories. e48 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 GSH/GSSG and Cys/CySS A simpler method for estimating redox potential is assessment of the ratio of the concentration of reduced to oxidized glutathione or reduced to oxidized Cys. As the most abundant redox couple in the cell, and because it is maintained by the combined activity of several redox couples, GSH/GSSG is a useful indicator of redox status and correlates with many cell functions, such as proliferation, differentiation, and apoptosis.104 Virtually all cardiovascular cell types express transporters for GSH, GSSG, Cys, and CySS, so that redox potentials measured in plasma reflect tissue redox state, but only indirectly. GSH can be measured using enzymatic recycling of GSSG reductase, HPLC with UV detection, and electrochemical detection, but these methods are not sensitive enough to detect low levels of GSSG. A better assay uses iodoacetate to alkylate free thiols, derivatization with dansyl chloride to fluorescently tag amino groups, and HPLC and fluorescence to separate, detect, and quantify GSH, GSSG, Cys, and CySS.105 Subsequent calculation of the nonequilibrium redox potential (EhGSH or EhCys) is a reflection of the redox state of the tissue of interest. During the assay, care must be taken to prevent oxidation and degradation. These measurements can be performed in plasma, tissue biopsy specimens, and cultured cells.105 Ashfaq et al106 showed that EhGSH/GSSG in plasma correlates with changes in intima-media thickness, a subclinical indicator of atherosclerosis. RoGFP Another fluorescence-based method to measure redox status involves the use of reduction-oxidation sensitive green fluorescent protein (RoGFP) probes.107 These probes consist of a green fluorescent protein in which 2 cysteines were introduced, the oxidation state of which serves as a readout of redox status in the particular subcellular compartment in which the RoGFP is expressed. When a disulfide bond is formed within the molecule, its fluorescent properties are altered such that there is a reciprocal change in emission (525 nm) after excitation at 2 different wavelengths (400 and 484 nm).108 This ratiometric property makes the signal independent of expression levels of the probe. Another advantage is the ability to add a targeting sequence so that the probe is expressed in specific subcellular compartments.109,110 Early versions of this probe (RoGFP2) are sufficient for reducing environments such as the cytosol and mitochondria. Subsequent modifications, in which the thermodynamic stability of the disulfide is lowered by inserting a single amino acid adjacent to one of the cysteines (eg, roGFP1-iL),111 have rendered the redox potential closer to that of the endoplasmic reticulum. Thus, one must know the redox potential of the compartment to be studied and choose the version of RoGFP that most closely aligns with it. RoGFP has been used successfully to measure redox status in the cytosol, mitochondria, and mitochondrial inner membrane space in vascular smooth muscle cells exposed to hypoxia as well as glutathione-induced reductive stress in the heart.110,112 Recommendations for Measurement of Redox Status If EPR is not available, measurement of GSH/GSSG or Cys/ CySS couples can be used instead. Care should be taken during sample preparation to avoid oxidation and degradation, as well as contributions from inadvertent lysis of cells. RoGFP probes are a useful addition to our armamentarium and allow measurement of redox status in specific subcellular compartments. Measurement of RNS Introduction The short half-life of NO in biological systems presents unique challenges for its detection and quantification.113 NO is lipophilic and diffuses rapidly through cells. The main sources of NO include NOS and reductive metabolism of nitrite (NO2−) and RSNO.114 In biological systems, NO is rapidly oxidized by several nonenzymatic and enzymatic pathways. In the vasculature, NO is oxidized by oxyhemoglobin to nitrate (NO3−). NO bioavailability can be reduced by its reaction with free .− .− radicals such as O2 . One such reaction between NO and O2 occurs at a diffusion-limited rate and yields OONO−, a strong oxidant implicated in numerous pathological processes.115 In addition, O2 reacts with NO to produce N2O3. Other RNS include N-nitrosation products and nitrated unsaturated fatty acids (Oxidative Modification of Proteins).116 Detection of NO Detection of NO in biological systems is complicated because of its short half-life, contaminants in laboratory reagents, and the multiple reactions that it undergoes, leading to a variety of end products. Because of their short half-lives, most RNS cannot be detected in frozen or fixed tissue samples, but they can be detected in live tissue or intact cells. Colorimetric (Griess Reaction) and Related Methods In the Griess reaction, nitrite-containing media is first treated with sulfanilamide in acidic media to form a transient diazonium salt, which then reacts with a coupling reagent, Nnaphthyl-ethylenediamine, to form a stable azocompound. These products can be measured by spectrophotometry, fluorescence,117 or HPLC.118 A major problem with the unmodified Griess reaction is its relative insensitivity (≈500 nmol/L), which makes measurements of endothelium-derived nitrite difficult. With this method, it is feasible to measure the production of nitrite generated by the inducible or neuronal NOS isoforms, which have higher Vmax values. The sensitivity of the method can be enhanced 10-fold by fluorometric detection, which makes it suitable for clinical applications.117 Another drawback of the Griess reaction is that a substantial portion of NO production by NOS enzymes is oxidized to nitrate, which is not detected by this technique unless NO is reduced to nitrite by a nitrate reductase reaction. Nitrite and nitrate contaminants in laboratory reagents often yield strong signals that can overwhelm true biological responses. In pathological states in which NO is oxidized rapidly, its bioavailability is reduced, but nitrate and nitrite levels are not affected. Diet is a major source of nitrite and nitrate and must be controlled for, particularly when measurements are made in the plasma. The main advantage of measuring nitrite and nitrate is that these products are relatively stable. Nitrate and nitrite are incapable of eliciting vasodilatation or other signals themselves; however, they can be biologically relevant because they can generate NO via nitrate reductases or on reaction with deoxyhemoglobin.119 Griendling et al Measurement of Reactive Oxygen Species e49 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Fluorescent Techniques There are several fluorescent probes for detecting NO based on diaminofluorescein (DAF), including DAF-2 and DAF-FM. DAF-FM is significantly more photostable than DAF-2 and is thus more sensitive for NO detection.120 The NO detection limit for DAF-FM is ≈3 nmol/L120 versus ≈5 nmol/L for DAF2.121 In this assay, cells are loaded with DAF-FM, washed, and analyzed for DAF-FM fluorescence by either a fluorescent plate reader, fluorescence microscopy, HPLC, or occasionally by flow cytometry.122 NO donors are generally used as a positive control. Some samples should also be preincubated with NOS inhibitors such as L-NAME (Nω-nitro-l-arginine methyl ester) or L-NIO [N5-(1-iminoethyl)-l-ornithine hydrochloride] to ensure specificity of the signal. A potential problem with DAF-2 and DAF-FM is that ascorbic acid and dihydroascorbic acid react with these probes and reduce their ability to detect NO.123,124 This reaction is concentration dependent, and as long as the concentrations of ascorbic acid and dihydroascorbic acid are stable, this issue does not affect the semiquantitative value of these probes. Chemiluminescence Ozone-based chemiluminescence provides sensitive NO detection.125 Liquid samples from either cells or vessels are injected into a reflux chamber that contains inert gas (nitrogen or argon), which carries NO to the NO analyzer where it reacts with ozone to produce the excited state NO2*, which emits light on decay.126 Typically, the reaction chamber that contains either sodium iodide (NaI) in acetic acid or vanadium in hydrochloric acid is maintained at 100°C. When NaI is used, nitrite is converted to NO, whereas vanadium can reduce both nitrite and nitrate. Other 1-electron oxidation state nitroso species, such as nitrosothiols, can also be detected with this approach. To differentiate between RNS without having to change reaction solutions, samples can be pretreated with RNS-specific reagents before analysis.127 Recently, an improved method for detecting NO with chemiluminescence has been reported that uses a reaction with soluble guanylyl cyclase-luciferin-luciferase.128 This approach is based on NO-mediated activation of soluble guanylyl cyclase, which catalyzes the conversion of GTP to cGMP and inorganic pyrophosphate. The assay measures the pyrophosphate, which is converted into ATP with the help of ATP sulfurylase. The resulting ATP activates the luciferin-luciferase reaction, which yields light. This approach allows detection of NO in the low nanomolar range. Electrochemical Approaches NO can be detected with microelectrodes, most often coated to be selectively permeable to NO and to eliminate other oxidizable substances.126 For example, polymerization of o-phenylenediamine on the electrode can increase NO specificity.126 In electrochemical detection, the voltage of an electrode is controlled to catalyze NO oxidation on the electrode surface. The current generated is a linear function of NO concentration at the electrode surface.126 This approach requires shielding from extraneous electrical signals, usually by a Faraday cage. In addition, it is essential that the electrode be placed at a constant distance from the source of the NO, for example, a set distance from the cell surface, using a micromanipulator. An advantage of electrochemical detection is that the sample size can be quite small; however, the commercially available devices purported to measure NO are insensitive to the low levels released by endothelial cells. Measurements of NO by this approach are less sensitive than those obtained with lipophilic probes, such as Fe[DETC]2 (DETC=N, N-diethyldithiocarbamate) (see Electron Paramagnetic Resonance below). This is likely because of the lipophilic nature of NO and the integrative effect of the trap. NO can also be measured with hemin-functionalized graphene transistors with subnanomolar sensitivity.129 The graphene is noncovalently functionalized with hemin chloride to form an NO sensor, because hemin has a high binding constant and high selectivity toward NO. Electrochemical detection with NO electrodes has been used in studies of cultured cells and tissue.126,130 Common challenges in such measurements include the specificity of the electrode to NO, the reproducibility of results with different electrodes, and the dependence of data on the distance of the electrode from the cell surface. Electron Paramagnetic Resonance EPR is an extremely specific and sensitive approach to detect NO in biological systems. Because of its short half-life and very short relaxation time, NO cannot be directly detected by EPR; however, compounds such as the nitronyl nitroxides PTIO (2-Phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide) and CPTIO [2-(4-Carboxyphenyl)-4,4,5,5-tetramethylimidazoline1-oxyl-3-oxide]131,132 and the iron-dithiocarbamate complexes Fe[DETC]2 and Fe[MGD]2 (MGD=N-methyl-D-glucaminedithiocarbamate) that form stable adducts with the NO radical can be used to detect NO.133 Detection of NO by nitronyl nitroxides is limited by bioreduction of nitroxides32 and potential cross-reaction with HNO and NO2.134 Fe[DETC]2 and Fe[MGD]2, in contrast, specifically react with NO radical to produce relatively stable complexes with specific EPR spectra, which provides unambiguous NO detection. A natural spin trap for NO is hemoglobin. On reaction with NO, ferrous hemoglobin forms a stable hemoglobin-NO complex that yields specific high-resolution spectra in rat and mouse blood.133,135 Typically, blood is snap-frozen in a tuberculin syringe and placed in a liquid nitrogen quartz Dewar flask designed for low-temperature EPR.135 The average lifetime of hemoglobin-NO is 4 hours, and it can accumulate in the blood in substantial concentrations. Blood hemoglobin-NO levels have been used to estimate vascular NO production; however, other sources of NO, such as inducible NOS or neuronal NOS, also likely contribute to the signal.133 Indeed, endotoxin-induced inflammation has been shown to increase the electron spin resonance hemoglobin-NO signal.136 The contribution of inducible NOS and eNOS in the circulating hemoglobin-NO can be determined by treating experimental animals with specific NOS inhibitors such as 1400W and L-NIO or by using mice deficient in specific NOS enzymes.133 It should also be kept in mind that dietary nitrite can be reduced to NO by deoxygenated hemoglobin and yield an identical hemoglobin-NO signal; however, this can be minimized by use of a standard or low-nitrate/nitrite diet.133 The hemoglobin-NO signal in humans is very difficult to detect, likely because of lower levels of hemoglobin-NO and a shift to the low-resolution EPR spectra of 6-coordinate hemoglobin-NO. e50 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Fe[DETC]2 is used in a second method to detect NO with EPR and can be produced by reaction of ferrous iron with DETC under anaerobic conditions. The resulting colloid is lipophilic and appears to detect NO that accumulates in cell membranes. The maximum signals from vessels or cultured endothelial cells are obtained when they are stimulated with the calcium ionophore A23187. Fe[DETC]2 can be used for in vitro, ex vivo, and in vivo NO measurements.126,133,137 The stability of the NO-Fe[DETC]2 complex allows measurement of the accumulation of bioactive NO released from the tissue or cells over time. Limitations of this technique include the special handling required for Fe[DETC]2 colloid to prevent oxidation and the requirement for an EPR spectrometer and expertise with such measurements.138 The third EPR method to detect NO uses Fe[MGD]2, which is water soluble and much more hydrophilic than Fe[DETC]2 and does not penetrate cell membranes. It remains in the extracellular compartment, which allows detection of NO released from cells.138,139 A limitation of Fe[MGD]2 and also to some extent Fe[DETC]2 is that the ferrous state can reduce nitrite to NO.140 This problem is largely prevented when these traps are present in the ferric state, but the trapping efficiency drops by a factor of 2.67 Detection of Peroxynitrite .− Peroxynitrite, formed by the rapid reaction of NO with O2 , reacts with thiols, lipids, proteins, and DNA, which makes it very short-lived.141 Excessive production of OONO− contributes to cellular and end-organ dysfunction in inflammation and cardiovascular disease.141 Peroxynitrite rapidly oxidizes tetrahydrobiopterin and thus induces eNOS uncoupling and vascular dysfunction.142 Its reaction with lipids leads to formation of isoprostanes and highly reactive ketoaldehydes, which can be used as indirect measures of OONO−. Peroxynitrite is also frequently measured by accumulation of stable nitration products such as 8-nitroguanine143 and nitrotyrosine.144 Other methods include EPR detection with spin probes and spin traps such as CM-H and EMPO [5-(ethoxycarbonyl)-5-methyl-1-pyrroline N-oxide]. The recently developed boronate probes have proved useful in direct detection of peroxynitrite and related species.66 Uric acid has been used as a relatively specific scavenger of peroxynitrite. Thus, inhibition of boronate, CM-H or luminol signals with BH4 or uric acid can provide confirmation of peroxynitrite detection. Dihydrorhodamine and luminol chemiluminescence have often been used to detect peroxynitrite; however, these probes undergo nonspecific oxidation in the presence of other oxidants and are therefore not specific for peroxynitrite. Their use is not recommended unless accompanied by controls to validate specificity.59 Citrulline Assay NOS convert l-arginine to l-citrulline; therefore, accumulation of l-citrulline was used as a marker of NOS activity and NO production in the past.145,146 The main source of citrulline, however, is the urea cycle, and alteration of urea production affects citrulline levels.147 Furthermore, citrulline is a marker of a wide range of pathologies such as bowel diseases148 and rheumatoid arthritis.149 Hence, l-citrulline is not a specific biomarker of NO bioavailability and as such should not be used as an index of NO status. Recommendations for RNS Measurement The selection of an assay depends on the experimental conditions and which RNS the investigator wishes to detect. Given the caveats presented above, several assays are available to measure either NO, oxidation products of NO, or other RNS (Figure 2). Either EPR with the NO spin trap Fe[DETC]2 or the fluorescent dye DAF-2 can be used to detect NO with suitable accuracy. Ozone-based chemiluminescence is also quite accurate and can be used to detect not only NO but also nitrite, nitrate, and nitrosothiols. Proper controls, such as inhibition of the NOS enzymes, control for contaminants in the media used, and in some cases, dietary controls for nitrate-rich foods, are essential to yield valid experimental results. For detection of peroxynitrite, no probe or detection method is entirely specific, but when combined with known scavengers of peroxynitrite such as uric acid or BH4, oxidation of CM-H or boronate probes can be used. Although peroxynitrite can react with protein tyrosines to form nitrotyrosines, immunostaining for nitrotyrosine is not recommended as a specific signal for peroxynitrite, because nitrotyrosine can also be formed via other pathways. Variations of the Griess assay can be used to detect nitrite, but one needs to be Figure 2. Decision tree for measuring reactive nitrogen species (RNS). When choosing an assay, one must first consider the identity of the RNS to be measured, the type of sample (cells, tissue, or in vivo), and the product to be analyzed. Assays are listed along with the appropriate probe. Ab indicates antibody; DAF-FM, diaminofluorescein-FM; EPR, electron paramagnetic resonance spectroscopy; Fe[DETC]2, Fe-diethyldithiocarbamate; Hgb, hemoglobin; and Mass Spec, mass spectrometry. Griendling et al Measurement of Reactive Oxygen Species e51 aware of nitrite and nitrate in laboratory reagents, plasticware, and the diet as potential contaminants. Oxidative Modification of Proteins Introduction Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Amino acid residues of proteins can act as electron donors, leading to covalent modifications of the amino acid chain. Oxidation of proteins can be induced by many stimuli, including ROS/RNS, metal cations, γ-irradiation, ultraviolet light and ozone, leakage of the electron transport chain in mitochondria, oxidoreductase enzymes, and products of lipid peroxidation.150,151 Increased bioavailability of ROS/RNS can lead to protein oxidation in cardiovascular cells. Oxidative modification of proteins has functional consequences such as gain or loss of protein function or enzymatic activity, altered protein-protein interactions, conformational changes, changes in protein localization, susceptibility to proteolysis, increased immunogenicity, and modified gene transcription. Such biochemical modifications alter cell signaling and impair or promote cell functions.150,152–154 Common Protein Oxidative Modifications in Cardiovascular Cells Proteins are highly sensitive to posttranslational oxidative modifications, which can be reversible (by biological antioxidant systems) or irreversible.155 Whereas irreversible modification usually leads to loss of protein function and protein degradation, reversible oxidative modification influences cell signaling and regulatory and homeostatic functions. Most amino acids can be irreversibly modified either directly via oxidation by ROS/RNS or indirectly by secondary by-products of oxidative stress. Some amino acids, such as Cys and methionine (Met), are more susceptible to oxidation than other amino acids because of the nucleophilic nature of sulfur (S) atoms present on their side chains.156,157 N-nitrosation of lysine, arginine, and histidine also contributes to protein modification. The most common type of irreversible modification is the formation of carbonyl groups (C=O). Amino acids prone to carbonylation include proline, arginine, threonine, lysine, histidine, and Cys. Assessment of the extent of such generalized oxidation serves as a marker of oxidative stress.158 Sulfur-containing Cys and Met, the amino acids most susceptible to oxidation, undergo reversible modifications by cellular enzymes and accordingly have a regulatory role in redox signaling.153 Oxidation of Cys leads to production of sulfenic acid (SOH), which could further react with another Cys residue to form an intermolecular or intramolecular disulfide bond or which could be further oxidized in a stepwise manner to sulfinic acid and sulfonic acid (Figure 3).161 Sulfenic acid can be redox recycled back to thiol by the glutaredoxin or thioredoxin systems.161,162 Cys sulfinic acids, previously thought to be irreversibly oxidized, can be reduced to the catalytically active thiol by sulfiredoxin.162 Because Cys is both an e- donor and acceptor, it is considered a redox-active amino acid that plays an important role in the regulation of protein function by ROS in cardiovascular cells. Met is readily oxidized to form methionine sulfoxide (MetO; Figure 4), which can be further oxidized to produce methionine sulfone or radicals. These latter molecules contribute to oxidative damage.164 Protein methionine oxidation can be reversed enzymatically by methionine sulfoxide reductases.164 Methionine sulfoxide reductases are repair enzymes that reduce methionine sulfoxide residues in oxidatively injured proteins to methionine residues. Accordingly, these enzymes protect cells from oxidative stress and have been considered as antioxidant and protein repair enzymes. Proteins with oxidation-sensitive methionine residues that are important in the cardiovascular system include calcium/calmodulin-dependent protein kinase II, α2-macroglobulin, 15-lipoxygenases, apolipoprotein A-1, thrombomodulin, and von Willebrand factor. Approaches for Detection of Oxidative Modifications Protein oxidation can be assessed in cultured cells, circulating cells, and tissue by direct and indirect approaches. Direct approaches detect changes by exploiting the properties of oxidized forms of proteins or structural changes induced by oxidation. Such approaches detect both reversible and irreversible modifications with specific antibodies165,166 or chemoselective probes167 and can be applied at a global or targeted level. Cell-permeable probes have the advantage of labeling within the system, thus preventing any artifactual labeling caused by oxidation on cell lysis.168 Indirect approaches are most commonly used in cardiovascular research because they exploit conserved biochemical properties of the reversible oxidative modifications, as well as the catalytic activity of certain proteins. Most of these assays are based on the ability of unmodified, active sites (eg, free thiols, S−) to be irreversibly alkylated and the resistance of oxidized sites (eg, SOH, SNO[S-nitrosothiol]) to alkylation. Indirect approaches monitor reversible modifications by using labeled substrates169 or probes170 and can be classified as negative and positive methods. In negative methods, the unmodified sites (eg, S−) are labeled with the probe, and oxidation is measured as loss of signal. On the other hand, in positive methods, the unmodified sites are alkylated initially, and the reversibly oxidized sites are then reduced and labeled with the probe; therefore, an increase in signal intensity indicates elevated levels of oxidation. Indirect approaches are advantageous because they can provide a relative quantification of the active versus the oxidized state. The major limitation of indirect approaches is that the fraction of reversibly oxidized proteins is overestimated if a significant proportion of oxidized proteins within the sample are in the irreversible state.171 The sensitivity of these methods can be improved by preparing subcellular enrichments before the assay. Methods to Assess Protein Oxidation Irreversible Oxidative Modification: Measurement of Protein Carbonylation (C=O) The most commonly used direct method to assess carbonylation levels is based on the derivatization of the stable carbonyl groups by 2,4-dinitrophenylhydrazine, which results in the formation of a stable product, dinitrophenyl hydrazone.172 Dinitrophenyl hydrazone is generally detected either by spectrophotometric assay (absorbance at 370 nm)173 or by immunoassays. Development of dinitrophenyl hydrazone–specific e52 Circulation Research August 19, 2016 Figure 3. Oxidative posttranslational modifications on protein thiols (cysteine).159,160 The side chain of cysteine residues possess a terminal thiol (-SH) functional group. The sulfur at the core of the thiol is electron rich, allowing multiple oxidation states as shown in the Figure. Protein thiols can form various oxidative modifications, including reversible and irreversible forms. SNO indicates S-nitrosylation; and SOH, S-sulfenylation. Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 antibodies has permitted quantification of carbonyl content by ELISA,174 immunohistochemistry,175 or Western blotting.176 Immunodetection by Western blotting offers the advantage of increased specificity compared with all other assays, because it allows for the assessment of carbonylation of individual proteins, which can further be extracted as gel bands and identified by tandem MS (MS/MS). Alternatively, carbonyl groups can be quantified after reduction by sodium borotritide to the corresponding 3H-radiolabeled hydroxy compound, which is detected by standard methods.177 An advantage of the methods used for assessment of protein carbonylation is that only standard laboratory equipment is needed. Reversible Oxidation: Measurement of Cys Thiol Oxidation Most approaches used to monitor reversible oxidative modifications of Cys thiols rely generally on the efficient trapping of the native redox state of the thiol proteome. For this purpose, all free thiols are selectively alkylated (or trapped) by alkylating agents, such as iodoacetamide and N-ethylmaleimide.178 Modified Cys residues can be labeled by direct or indirect approaches. The labeled probes used in these strategies can be fluorescently tagged, biotinylated, or isotopic derivatives of thiol alkylating agents. Labeled proteins are typically either separated by 2-dimensional gel electrophoresis and subjected to MS or LC/MS identification in proteomics approaches; pulled down and detected by immunoblotting; or analyzed by Western blotting with specific antibodies, as in the case of a modified cysteinyl-labeling assay.179 The most commonly used approaches involve alkylation of free thiols with subsequent reduction of the reversibly oxidized thiols and labeling with a probe. Increase in the signal is indicative of elevated oxidation.170 This approach can assess either global or specific types of reversible thiol modifications, depending on the selectivity of the reductant being used. Dithiothreitol and Tris(2-carboxyethyl)phosphine are considered general reductants and are therefore used for untargeted screening of protein thiol modifications. Detection of sulfenylation, which occurs when free thiols react with H2O2 to form sulfenic acid (R-SOH), is typically based on the specific reduction of sulfenic acid with arsenite.162 Snitrosylation (RSNO) of free thiols on reaction with NOderived species such as N2O3 (NO+) or by interaction with Figure 4. Oxidative posttranslational modifications on methionine residues in proteins. Addition of O2 to the sulfur atom of methionine (Met) leads to production of methionine sulfoxide (MetO).163,164 MetO is reduced back to Met by methioinine sulfoxide reductases. Methionine sulfoxide residues can be further targeted by H2O2 to methionine sulfone, which is an irreversible form of methionine oxidation. Griendling et al Measurement of Reactive Oxygen Species e53 metalloprotein-linked enzymes is another Cys thiol modification that is discussed in more detail below in Modification of Proteins by NO. In addition, formation of mixed disulfides, referred to as S-glutathionylation as a result of other thiol redox reactions can be detected via reduction by glutaredoxin.162 Although this methodology is widely used, especially during proteomic screenings of the thiol proteome, it does not allow for identification of the specific site of the thiol modification. A more direct approach in assessing Cys thiol oxidation under nonreducing conditions is the antibody-based strategy. Specific antibodies have been raised to detect S-glutathiolated proteins180; however, their use has limitations because of their low sensitivity. Antibodies have also been generated to react with the hyperoxidized (SO3H) form of proteins, such as peroxiredoxin.181 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Measurement of Methionine Oxidation Methionine oxidation is a promising marker of oxidative stress. Conventional methods for assessing MetO generation and reduction depend on HPLC and MS. More recently, novel antibodies and Met-rich proteins have been used to measure methionine/selenomethionine oxidation and methionine sulfoxide reductase.182 Measurement of Sulfenylation Sulfenylation is typically measured with a specific alkylating agent, 5,5-dimethyl-1,3-cyclohexanedione (dimedone). This approach is based on the principle that sulfenylated proteins react irreversibly with dimedone and can then be isolated by affinity-tagged purification and identified by MS/MS, Western blotting, or fluorescence measurements. Several dimedone-based probes and antibodies have been developed to provide direct methods for assessment of protein sulfenylation. The probes typically consist of the dimedone reactive group, a linker, and a reporter/affinity tag or an azide group.183,184 The tag is usually a fluorophore or biotin, which enables detection of the oxidized proteins with fluorescence. The introduction of an azide group has improved the reactivity of probes (eg, DAz1, DCP-N3), because it allows connection of the bulky reporter/affinity tag to the probe by “click chemistry” after the probe is already bound to the Cys-SOH.184 Moreover, the recent development of cell-permeable probes (eg, DCP-Rho1, DYn-2) has permitted capture of sulfenylation in living cells, which can be analyzed by confocal or fluorescence imaging.168,183 A limitation of using dimedone-based probes is their reduced reactivity with certain protein families, such as protein tyrosine phosphatases, important in signaling in vascular and cardiac cells185; however, this issue has been addressed by generation of dimedone-based probes with a protein tyrosine phosphatase–binding module, which binds irreversibly to the oxidized catalytic site of protein tyrosine phosphatases (eg, BiPhaz-1, NAPhaz-1).186 In the case of dimedone-specific antibodies, cells need to be treated with dimedone before they are lysed. A limitation of this method is that dimedone reacts irreversibly with SOH groups; therefore, treatment with dimedone for long periods of time will alter the cellular redox state and could generate false-positive results.187 Another more targeted approach monitors sulfenylation of specific proteins of interest using conformation-specific antibodies, which recognize the oxidized structure of the protein.188 The limitation of this method is that the crystallographic structure of the unmodified and modified form of the protein has to be known in order for the specific conformation-sensing antibody to be developed. Most studies examining oxidative modifications of proteins have detected protein S-glutathionylation S-sulfenylation, and S-nitrosylation (see Modification of Proteins by NO). These modifications are increasingly recognized as important signaling events in cardiovascular physiology and disease. The significance of protein S-glutathionylation and S-sulfenylation in cardiovascular disease has been suggested in various experimental models. In rat aortic rings, S-glutathiolated eNOS is associated with impaired vasodilatation.189 In atherosclerosis, S-glutathionylation induced activation of sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) is substituted by irreversible sulfonylation.190 The cardiac sarcolemmal Na+-K+ pump is inhibited by S-glutathionylation on the β1-subunit in ventricular myocytes.191 In models of ischemia-reperfusion, there is a loss of function of GAPDH,159 aldose reductase,192 and actin because of S-glutathionylation193 Studies that used selective reduction of sulfenic acid and specific probes to assess S-sulfenylation levels have shown that multiple structural and mitochondrial proteins of cardiac myocytes, including cytoskeletal components, undergo sulfenylation after exposure to H2O2.194,195 However, it is still unclear whether sulfenic acids can act as signaling molecules or whether they are intermediates in the process of irreversible oxidation that results in the formation of sulfinic and sulfonic acids. Modification of Proteins by NO Nitrosothiols are important mediators of NO signaling pathways196 and are likely formed by multiple mechanisms, including nitrosation by N2O3, radical recombination of thiyl radicals with NO, and transition metal-catalyzed reactions.197 Cellular levels of nitrosothiols can be reduced by denitrosylating enzymes such as S-nitrosoglutathione reductase.198 The major cellular S-nitrosothiol is S-nitrosoglutathione reductase.199 An important reaction is transnitrosylation, in which NO is transferred from one thiol to another.200 Several methods have been developed for detection of protein and low-molecular-weight nitrosothiols; however, these techniques are challenging and prone to artifacts.201 These measurements can be affected by light, reducing agents, and pH because of an effect on nitrosothiol stability. Chung et al202 offer a comprehensive review on the utility of Cys tandem mass tag (cysTMT) to detect and determine the reactivity of an individual Cys’s reactivity for S-nitrosylation. This approach has facilitated the identification and quantification of Cys oxidative modifications in cardiovascular cells and tissue.202 Other major methods of nitrosothiol detection include chemiluminescence-based methods and switch-based methods,201 and recently, nitrosothiol-specific antibodies have been developed.203,204 The biotin-switch method of detecting nitrosothiols is similar to that described in Reversible Oxidation: Measurement of Cys Thiol Oxidation. For measurement of nitrosothiols, this method consists of 4 major steps: (1) thiol blocking, (2) selective reduction of RSNO, (3) labeling of resultant free thiols, e54 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 and (4) detection of biotin-labeled thiols.201 Significant concerns have arisen regarding the potential artifacts associated with high levels of ascorbate that can potentially reduce some protein disulfides.205 Phosphine-mediated selective reduction appears to be preferable because it is specific for nitrosothiols and does not reduce protein disulfides.206 Both polyclonal and monoclonal antibodies have been developed that can detect RSNO moieties.203,204,207 An example is the anti-S-nitrosocysteine antibody, which detects S-nitrosocysteines but not reduced cysteines.203 It is generally recommended that one use positive and negative controls with chemical nitrosation and cleaving of S-NO bonds in situ.208 If cleaving the S-NO bonds does not reduce the signal, then immunostaining is likely nonspecific. A third method of detecting NO-mediated protein modification is anaerobic reductive chemiluminescence, which uses excess Cys to drive the transnitrosation equilibrium in favor of CySS-NO and reduce Cu2+ to Cu+. Reductive RSNO assays are performed under helium or argon in a purge vessel that contains Cys and CuCl.201,208 The free NO that is released is purged in the inert gas stream to a chemiluminescence-based NO analyzer. Photolysis-chemiluminescence is based on photosensitive homolytic cleavage of RSNO after exposure to light; however, ultraviolet photolysis has been criticized for overestimating RSNO values because of the presence of endogenous nitrite and nitrate that can also be converted to NO by ultraviolet irradiation.209 The Cu-Cys assay is not useful for solutions with high protein content (plasma, hemoglobin) and has been replaced by triodiode (KI3)-based reductive chemiluminescence, which provides a more specific assay of RSNO levels.210,211 Triodiode-based assays have become the most accepted and widely used method for RSNO measurement, providing sensitive and specific measurements in complex systems. A cardioprotective role of S-nitrosylation has been demonstrated for cyclooxygenase 2212 and mitochondria complex I,213 translated as attenuated infarct size during ischemia-reperfusion. In rat hypoxic myocardium, decreased S-nitrosylation of Na+-K+ ATPase was followed by increased S-glutathionylation and loss of activity.214 S-nitrosylation of p47phox by NO has been shown to suppress Nox-induced ROS production in human endothelial cells.215 On the other hand, impaired relaxation in mouse aorta has been related to protein kinase C S-nitrosylation,216 as well as to augmented global S-nitrosylation in Ang II–induced hypertension.217 Studies in bovine aortic endothelial cells demonstrated increased levels of S-nitrosylated endothelial NOS, which was reduced on stimulation with vascular endothelial growth factor.218 Measurement of Protein-Derived Radicals Immuno-spin trapping with anti-DMPO antibodies is a useful approach for detecting protein-derived radical intermediates both in vitro and in vivo. Antibodies produced in rabbits immunized against the hapten 5,5-dimethyl-2-octanoic acid-1-pyrroline N-oxide (OA-DMPO, a structural analog of DMPO with an additional carbon chain terminating in a carboxylgroup for amide linking) bind specifically to the protein adduct of DMPO.219–221 This approach expands the utility of spin trapping without EPR and can be highly sensitive in detecting protein radicals formed under physiological or pathological conditions. It can be used in conjunction with immunoblotting analysis, immunofluorescent staining, flow cytometry, and even magnetic resonance imaging. In combination with MS, immuno-spin trapping has been used to define the role of protein thiol radicals in mediating S-glutathionylation of mitochondrial complex I at the level of isolated enzymes, myocytes, and hearts of eNOS−/− mice.13,30,222 In endothelial cells, the same approach has been used to examine a similar mechanism in regulating protein S-glutathionylation coupling, and function of eNOS.223 Anti-DMPO antibodies can also be covalently bound to an albumin-gadolinium-diethylenetriaminepentaacetic acid-biotin magnetic resonance imaging contrast agent and used for molecular magnetic resonance imaging.224,225 This approach has been used to detect in vivo levels of membrane-bound protein/lipid radicals in kidney, liver, and lung of diabetic mice.225 Immuno-spin trapping is a unique and powerful approach to detect protein radical intermediates in time and space; however, the approach is limited to the specific protein radical adducts of DMPO, mainly Cys- and tyrosine-based protein radicals. Additional antibodies against different type of spin traps (eg, nitroso spin traps) could provide broader utility in determining other types of protein radicals. Tyrosine or Tryptophan Modifications Tyrosine and tryptophan nitration are common posttranslational modifications that affect protein structure and function. 3-Nitrotyrosine and nitrotryptophan are formed on reaction of OONO− with proteins containing tyrosine or tryptophan residues. Alternatively, these modifications can occur by peroxidase-mediated reactions between protein tyrosines or tryptophans and nitrite. Such changes have been detected by MS and immunostaining in various disease processes, including aging.226,227 They have also been used to serve as a footprint of peroxynitrite formation in vivo228; however, peroxidase-based reactions of nitrite with tyrosines can also yield nitrotyrosine and in fact might be its major source in vivo. HPLC, biotin tagging, MS/MS, and immunological methods have been used for nitrotyrosine and nitrotryptophan detection.144,229 MS/MSbased peptide sequencing provides unambiguous confirmation of nitrotyrosine formation. Antibodies purported to detect nitrotyrosine also detect nitrotryptophan,230 and so must be used with caution. Formation of dityrosine residues as a result of oxidative damage has also been used as a marker of oxidant stress. In this method, dityrosines are first released by acid hydrolysis in antioxidant-containing buffers and then derivatized with heptafluorobutyric anhydride/ethyl acetate before analysis by MS. This method can be used on whole tissue, but usually dityrosine formation on individual proteins is below the detection limit of the assay. Recommendations for Measurement Considering the importance of oxidative stress and redox signaling in cardiovascular pathophysiology, unraveling exactly how protein function is changed by oxidation is critical to elucidate molecular mechanisms of disease. Accordingly assessment of oxidative modifications of proteins, a posttranslational Griendling et al Measurement of Reactive Oxygen Species e55 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 phenomenon, should be a priority in the tool kit of redox methodologies in cardiovascular research. Although MS- and electrophoresis-based proteomics are commonly used, these approaches separate proteins as individual polypeptides rather than as functional complexes. Moreover, data generated are rarely quantitative and do not indicate modification levels per functional unit, and full resolution of basic or hydrophobic proteins, including membrane-associated proteins, cannot be attained with current technology. Hence, these techniques provide information on global oxidative changes. Another system that is gaining popularity in cardiovascular research is the OxyBlot (Merck Millipore) protein oxidation assay, which is ideal for rapid detection of proteins modified by ROS. This assay, which can be performed in crude or purified protein preparation, measures carbonyl groups introduced into protein side chains and also gives an assessment of global rather than specific protein oxidation. In-depth characterization and quantification of oxidative modification of individual proteins requires complex oxidative proteomics. Such studies should be performed in collaboration with specialized proteomics laboratories. However, it is becoming relatively simple to detect individual oxidized proteins within a mixture of proteins using immune-based techniques, such as Western blotting, ELISA, and immunohistochemistry/immunocytochemistry. Numerous commercial antibodies to assess nitrosylation, glutathionylation, carbonylation, and sulfenation of proteins are now available, which makes assessment of protein oxidation in cardiovascular cells and tissues relatively simple. Immunostaining can also be used to measure 3-nitrotyrosine, a marker of RNS. Other immune-based methods rely on 5-iodoacetamido fluorescein to label oxidized sulfhydryl groups, which can then be pulled down with fluorescein antibody.154 Alternatively, proteins such as thioredoxin can be derivatized at the active-site Cys thiols using a large maleimide derivative, and then nonreducing gel electrophoresis can be used to separate reduced (labeled) and oxidized (unlabeled) proteins based on their size.231 Choice of method depends on the availability of species-specific antibodies, the type of modification to be studied, and the particular protein of interest. As with other redox methods, appropriate controls must be included to demonstrate specificity of the protein modification under study. Measurement of Lipid Peroxidation Introduction Because of their highly reactive state, ROS are unable to diffuse away from their site of formation, which often leads to only site-specific alterations. Thus, it has been suggested that lipid peroxidation products amplify and propagate the cellular effects of ROS.232,233 Measurements of lipid peroxidation products are therefore not only sensitive assessments of ROS generation but also provide mechanistic insights into the pathogenesis of ROS-induced injury. Common Lipid Oxidative Modifications Oxidation of lipids (peroxidation when biological material is involved) is a complex process, with several free radicals, metastable intermediates, and end products.234,235 Several products of lipid peroxidation can be measured to document ROS-mediated lipid peroxidation in biological tissue. Because of their extremely short half-life, C-centered radicals or peroxyl radicals are rarely measured, except in detailed mechanistic studies. Measurement of Lipid Peroxidation Lipid Hydroperoxides Lipid hydroperoxides (LOOH) are usually measured by HPLC, because they are largely degraded during MS. Older methods for LOOH separation/detection used either chemiluminescence236 or mercury cathode electrochemical detection.237 These methods have been improved considerably by the inclusion of appropriate stable isotope internal standards238,239 and are suitable for the measurement of LOOH in plasma, lowdensity lipoprotein, and tissue samples. Phospholipid LOOH in tissues and cells can also be identified by HPLC followed by fluorescence detection.240 The method has high sensitivity (1–2 pmol of LOOH), although its specificity remains unclear. The formation of specific hydroperoxides can be used to study the reactivity of specific ROS; for instance, singlet oxygen, ozone, or ionizing radiation in biological tissues can be specifically documented by measuring specific cholesterol hydroperoxides.233 Unsaturated Aldehydes Aldehydes such as 4-hydroxy-trans-2-nonenal (HNE), malondialdehyde (MDA), isoprostanes, and acrolein are major end products of lipid peroxidation.232 Among the free aldehydes generated from lipid peroxidation, the C9 unsaturated aldehyde HNE is the most abundant product of arachidonic acid241 and is one of the most studied products of lipid peroxidation. Tissue and plasma concentrations of free lipid peroxidation-derived aldehydes such as HNE can be measured by HPLC or gas chromatography (GC)/MS. HPLC methods usually use aldehyde-reactive probes such as 2,4-dinitrophenylhydrazine and 1,3-cyclohexandione or other similar chemical probes.242 Derivatization by reagents such as pentafluorobenzyl oxime followed by silylation is also required for detection with negative chemical ionization by GC/MS.243,244 With the use of an appropriate deuterated internal standard, GC/MS can be a sensitive and reliable method for the measurement of free HNE245,246; however, current approaches that use LC/MS247,248 are preferable because they do not require derivatization to enhance optical activity or to generate a volatile product. In addition, measurement of HNE-modified proteins has been widely used as a measure of oxidative stress and ROS generation in a variety of disease conditions, and coupled with MS identification of the modified proteins,249–251 it can provide mechanistic understanding of oxidative stress–induced injury and dysfunction. Proteins modified by lipid peroxidation–derived aldehydes such as HNE, MDA, and acrolein are commonly detected by Western analysis or immunohistochemistry with antibodies against aldehyde-modified proteins. Several anti-protein MDA and anti-protein HNE antibodies are commercially available and suitable for immunohistochemistry, ELISA, and Western blotting. Polyclonal or monoclonal e56 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 antibodies raised against HNE-treated keyhole limpet haemocyanin252,253 or bovine serum albumin254 have been used to identify HNE-histidine adducts, with little cross-reactivity with HNE-lys or HNE-cys. Increased formation of HNE and related aldehydes such as acrolein in animal models and humans can also be measured by quantification of their urinary metabolites. The major urinary metabolites are mercapturic acid derivatives. In mouse and rat urine, the most abundant HNE metabolite is 1,4-dihydroxynonane-mercapturic acid (DHN-MA),255,256 which is derived from the reduction of the glutathione conjugate of HNE (GS-HNE) to GS-DHN by the enzyme aldose reductase.257 Similarly, the reduced form of the mercapturate derivative (3-hydroxypropanal mercapturic acid; 3-HPMA) is also the most abundant urinary metabolite of acrolein in rats.258 Both DHN-MA255,259 and 3-HPMA260,261 have also been detected in human urine and have been linked to oxidative stress; however, because in addition to lipid peroxidation acrolein can also be derived from exposure to combustion products and cigarette smoke, DHN-MA might be a more selective marker of lipid peroxidation than 3-HPMA in humans. Oxidized Phospholipids In addition to free aldehydes, peroxidation of phospholipids generates carbonyls that remain esterified to the phospholipid backbone. The most commonly studied oxidized phospholipids are 1-palmitoyl-2-oxo-valeroyl-sn-glycero-phosphocholine (POVPC), 1-palmitoyl-2-glutaroyl-sn-glycero-phosphocholine (PGPC), and 1-palmitoyl-2-(5,6-epoxyisopro-stane E2)-snglycero-phosphocholine (PEIPC).262,263 These phospholipids are derived from the oxidation of 1-palmitoyl-2-arachidonylglycerol-3-phosphocholine (PAPC), one of the most abundant phospholipids in low-density lipoprotein. The levels of these oxidized phospholipids can be measured by electrospray ionization MS in tissues after chloroform-methanol extraction and solid-phase extraction chromatography using dimyristoylphosphatidylcholine as an internal standard.262 Such measurements have shown that aortas obtained from rabbits fed an atherogenic diet contain higher levels of POVPC, PGPC, and PEIPC than those from rabbits fed a control diet.262 Moreover, oxidation products of cardiolipin, which is abundant in the mitochondria, can be measured as specific biomarkers of mitochondrial oxidative stress with LC/MS.264 Because oxidation creates new epitopes, oxidized phospholipids are recognized by antibodies of innate immunity. Several natural monoclonal IgM antibodies have been cloned, including E06,265–267 which was cloned from apolipoprotein E–null mice.268 These IgM autoantibodies have been shown to identify oxidized phospholipids in arterial plaques268 and in lipoproteins, although the specific epitopes recognized by these antibodies have not been well-defined because they also recognize MDA-modified low-density lipoprotein or high-density lipoprotein, which suggests that they recognize the adduct between oxidized lipids and apoproteins.268 Of these antibodies, E06 appears to specifically recognize the perturbed phosphorylcholine headgroup of oxidized phospholipids and can be used to recognize these phospholipids in cells, tissues, and lipoproteins. Therefore, positive reactivity to the antibody can be a useful indicator of the oxidation of phosphocholine phospholipids in cells and tissues. However, in most cells, oxidized phospholipids are readily removed, either by cytosolic enzymes with phospholipase A2 activity, which removes the oxidized sn2 side chain,269 or by the reduction of sn-2 aldehydes by aldose reductase.270 Hence, the absence of oxidized phospholipids does not necessarily indicate a lack of lipid peroxidation, and the measured levels are reflective of a steady state achieved between the generation and the metabolism of these phospholipids. Oxidized phospholipids can also be measured in the plasma, and the levels of oxidized phospholipids bound to lipoproteins correlate with the extent of angiographically documented atherosclerotic disease,266 which suggests that oxidized phospholipid might be involved in the formation of atherosclerotic lesions.271 Isoprostanes Nonenzymatic peroxidation of arachidonic acid generates prostaglandin-like compounds that have been measured as reliable biomarkers of oxidative stress in general and lipid peroxidation in particular. Several chemically distinct species of isoprostanes are generated from the oxidation of arachidonic acid.272,273 From 4 F2-isoprostane (IsoP) regioisomers, each composed of 8 racemic diastereomers, 64 different species are generated. Although oxidation products of arachidonic acid have been classically measured as markers of lipid peroxidation, other isoprostanes derived from eicosapentaenoic acid, docosahexaenoic acid, adrenic acid, and α-linolenic acid provide a more comprehensive picture of fatty acid–specific lipid peroxidation.274 In principle, any or all of these isoprostanes could be measured; however, practically, only a few of these species are quantified, of which the 5-series F2-IsoPs and the 15-series F2-IsoPs derived from arachidonic acid are most frequently measured. Because of the high structural diversity of isoprostanes, GC/MS or LC/MS is the method of choice for measuring these species, although ELISA-based immunoassays are also used frequently. The immunoassays, although simpler, cheaper, and faster, have high variability. There is poor correlation between different immunoassays, and the values obtained from ELISA often do not correspond to those measured by GC or LC/MS.275 Moreover, the antibodies used in these assays exhibit cross-reactivity with molecules of related structure, such as the products of cyclooxygenase.276 Immunoassays are also highly sensitive to interference by biological impurities present in complex biological systems, such as cell extracts, urine, or blood. Hence, the results obtained from ELISA and other immunoassays should be interpreted with caution. The most sensitive and accurate measurements of isoprostanes involve quantification by GC/MS with appropriate deuterium-labeled internal standards.277,278 These methods can measure isoprostanes to a lower limit of quantification of 2 to 100 pg/ml and can be optimized to measure multiple species of isoprostanes and isofurans.277 However, these techniques require elaborate sample preparation steps, such as thin-layer chromatography, Griendling et al Measurement of Reactive Oxygen Species e57 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 solid-phase extraction, liquid-liquid extraction, or affinity chromatography. Moreover, the samples must undergo chemical derivatization to pentafluorobenzyl ester, trimethylsilyl ether analogs before GC/MS. As a result, only a few specialized laboratories have the necessary experience and infrastructure to reliably measure isoprostanes by GC/MS. In contrast with the GC/MS methods, LC/MS measurements of isoprostanes require shorter procedures for sample preparation, with no derivatization step. Although earlier methods suffered from poor resolution, long run times, and low sensitivity, newer methods using ultraperformance LC with tandem MS detection using multiple reaction monitoring rival or exceed the sensitivity and specificity of GC/MS techniques.279,280 These methods generate high resolution of analytes to minimize matrix effects281 and can measure isoprostanes in the low picomolar range. With such high-sensitivity methods, changes in the levels of 15-F2-IsoP can be measured in as few as 10 000 cells.280 These methods can also be adapted to simultaneously measure both enzymatically and nonenzymatically generated eicosanoids.282 Although these techniques require sophisticated mass spectrometers and ultraperformance LC, they enable rapid, accurate, and sensitive detection of a wider range of isoprostanes and related species than has been possible thus far. Isoprostanes are more stable than other products of lipid peroxidation and are therefore measured as reliable indices of oxidative stress. Levels of F2-IsoP in blood and urine have been measured to assess systemic oxidative stress in both animals283 and humans284 under conditions of validated oxidative stress and in animal models of atherosclerosis, transplant arteriosclerosis, flow-induced vascular remodeling,285 and age-accelerated atherosclerosis.286 Additionally, urinary levels of isoprostanes have been used to document an increase in oxidative stress in humans with high cardiovascular disease risk,287 diabetes mellitus,288 or pulmonary hypertension289 and to document increased free radical generation after reperfusion in patients undergoing acute coronary angioplasty.290,291 In the plasma, F2-IsoPs circulate in their free unesterified form, although they can also be bound to the phospholipid backbone, from which they can be released by platelet-activating factor acetylhydrolase. Because the levels of free and bound isoprostanes are highly correlated, usually only levels of free isoprostanes are measured. The metabolism of F2-IsoPs in the liver generates multiple metabolites, including those generated by their conjugation with taurine or glucuronide. In human studies, these metabolites are measured along with the unmetabolized parent compound.273 Recommendations for Measurement In the design of experiments, the choice of the lipid peroxidation product to be measured will depend on the overall objective of the study (Figure 5); however, in general, measurement of lipid peroxidation products can provide a useful and informative index of oxidative stress. Direct measurements of lipid peroxidation products such as HNE, acrolein, or lipid hydroperoxides, however, are difficult because they are rapidly metabolized or react readily with other cell constituents. Therefore, the formation of these products is usually assessed indirectly by measuring their covalent protein adducts. Measurements of protein-HNE or protein-MDA adducts provide unambiguous evidence for lipid peroxidation, Figure 5. Analytical methods for measuring products of lipid peroxidation in experimental and clinical studies. The choice of the assay depends on the experimental system. Assays suitable for different experimental systems are shown in horizontal bars. Measurements of conjugated dienes are suitable only for isolated membranes and cells, whereas thiobarbituric acid reactive substances (TBARS), free 4-hydroxy-trans-2-nonenal (HNE) and malonaldehyde (MDA) levels, hydroperoxides, isoprostanes, oxidized lipids, and protein-aldehyde adducts can be measured in most experimental systems. Autoantibodies against protein conjugates of aldehydes can be measured in the animal and human plasma, whereas urinary isoprostanes and metabolites of aldehydes (mercapurates) can be measured in the urine. Assays listed as primary (1°) are the most well accepted, secondary (2°) are more widely used, and tertiary (3°) are either less sensitive or less specific (TBARS) or should be verified with a second method. e58 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 and an increase in abundance of these adducts is likely to be reflective of increased oxidative stress. However, these assays, although qualitative, are not very quantitative. When generated, HNE and MDA react with multiple proteins that show variable levels of modification, making quantification of the total extent of lipid peroxidation somewhat challenging. Moreover, some proteins are adducted to HNE or MDA at baseline, and therefore their presence interferes with quantification of the total adduct formation. In addition, proteins covalently modified by HNE are readily degraded by proteolysis,292 and thus, measurements of the levels of these adducts at a single time point could underestimate their formation. Hence, it is preferable to measure more stable products of lipid peroxidation, such as isoprostanes, in urine, blood, or tissue if the intent is to document total local or systemic oxidative stress. F2-IsoPs have been well validated as markers of oxidative stress both in animal and clinical studies.273 Nevertheless, measurements of specific products of lipid peroxidation such as lipid hydroperoxides, proteinHNE adducts, or oxidized phospholipids are useful in addressing specific mechanistic questions. Oxidative Modification of DNA Introduction One of the more severe consequences of excessive ROS production is oxidative modification of DNA.293 Characteristic DNA modification has been detected under several pathological conditions associated with increased oxidative stress; however, whether these modifications contribute to disease progression or are a consequence of pathologically increased ROS levels remains to be determined.294 Techniques to measure DNA oxidation can be applied to both nuclear and mitochondrial DNA, and many are applicable to RNA, which also undergoes oxidative modification.295 Oxidation of guanine has been considered to be a surrogate measure of oxidative DNA modification, but there are several other chemical modifications of DNA that are induced by ROS and RNS, including (1) purine and pyrimidine base lesions generated by oxidation, deamination, depurination, and depyrimidination; (2) modification of the sugar moiety; (3) formation of protein-DNA cross-links; (4) formation of DNA-DNA cross links; and (5) single- and double-strand breaks.296 Typically, strand breaks occur when a base is oxidized, for instance, conversion of guanosine to 8-hydroxy-deoxyguanosine, on which endogenous enzymes excise the modified base, creating an abasic site. Sometimes, the newly added base is a mismatch, which can lead to errors in replication. Modification of the sugar moiety by ROS such the alkoxyl radical generates reactive aldehydes such as base propenals and MDA.297 These aldehydes, generated from radical oxidation of 2-deoxyribose, like those generated from lipid peroxidation, react with nucleophilic centers in the DNA and modify DNA bases. In aggregate, between 50 and 75 base modifications have been reported, although to date, some of these have been found only under in vitro conditions in which oxidative modification was specifically induced.293,294,298 Under normal conditions, there appears to be a background level of ROS-induced damage in both nuclear and mitochondrial DNA that is normally repaired by enzymes such as the DNA glycosylases. These repair mechanisms can be broadly summarized as base excision repair, nucleotide excision repair, and mismatch repair. Although estimates of DNA damage range from 100 000 to 1 000 000 lesions per genome per cell division,299 the overall rate in humans appears to be about 1 mutation per 30 million base pairs per cell generation.300 Mechanisms of Oxidative Damage to Nucleotides Although several ROS can modify proteins, their reactivity with DNA is more limited. Some species such as the •OH radicals and crypto-OH or ferryl-OH react avidly with DNA, .− whereas other species such as O2 or H2O2 are not known to directly modify DNA.298,301 Additionally, RNS such as OONO− can react directly with DNA.302 In vitro, the •OH radical reacts with DNA by addition to double bonds of DNA bases and by abstraction of an H atom (also known as hydrogen atom transfer) from the methyl group of thymine and the C-H bonds of 2′-deoxyribose. The •OH radical also reacts with carbons at the C5-C6 double bond of pyrimidines, which results in the production of C5-OH and C6OH adduct radicals. Hydrogen atom transfer from thymine results in the allyl radical. Adduct radicals differ in terms of their redox properties, with some being reducing (C5-OH) whereas others are oxidizing (C6-OH).298 These radicals can form many different products (see references 294, 296, and 298 for detailed lists of the many products) depending on their redox properties and environment (eg, presence or absence of oxygen or reaction partners), but for the purposes of this statement, we will focus on the measurement of oxidation modification in broad categories: chemical modification, fragmentation, and abasic sites. Measurement of DNA Damage Oxidative DNA base damage is measured by either direct or indirect approaches.303 Direct approaches use sophisticated analytical procedures, for example, MS, to evaluate chemical modification of extracted and purified DNA. Indirect approaches involve the use of a specific DNA repair enzyme and measurement of DNA strand breaks. For indirect analysis, a DNA N-glycosylase repair enzyme is used to excise the modified bases, which induces a strand break that can be measured by reliable methods such as the comet assay and the alkaline elution technique. More recent techniques use polymerase chain reaction (PCR) to evaluate strand breaks in mitochondrial DNA or antibodies directed against oxidized bases to quantify base modification. Among DNA bases, guanine has the lowest ionization potential, which makes it more nucleophilic and therefore more susceptible to oxidation than other bases.296 For this reason, some assays have focused specifically on the oxidation of guanine, or 8-oxoguanine; however, a caveat for these measurements is that sometimes the nature of the procedure affects the measurements. For instance, LC or LC/MS techniques can potentially overestimate 8-oxoguanine levels in the sample because of spontaneous oxidation of the guanine during sample preparation.304 Hence, it is imperative that all methods of Griendling et al Measurement of Reactive Oxygen Species e59 procedure are performed with extreme care to avoid spurious oxidation during sample preparation. Comet Assay The comet assay is a simple method based on gel electrophoresis that enables measurement of DNA strand breaks in cells.305 Isolated cells are mixed with an agarose solution, which is then allowed to gel. The imbedded cells are then lysed and electrophoresed at an alkaline pH, which produces comet-like structures when the DNA is observed by fluorescence microscopy. The “intensity” of the fluorescent signal of the comet tail relative to the head reflects the number of DNA breaks.306 The comet assay has been used to evaluate DNA strand breaks in cardiac myocytes subjected to oxidative injury.307 Although this assay is simple and easy to use, it provides only general information about strand breaks and does not provide insight into the nature of the oxidative modification. Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Mass Spectrometry Typically, MS analysis of ROS-induced DNA damage involves GC-MS, HPLC-MS, or electrospray ionization MS analysis.308,309 MS can detect a range of specific oxidations of DNA bases, including 8-oxoguanine, thymidine hydroperoxides, and oxidizing or reducing radicals. Although MS is highly sensitive (femtomolar range), the approach is not without limitations. For example, conditions of sample preparation (derivatization of the nucleotides) for GC-MS are conducive to oxidation and can lead to overestimates of the amount of oxidized DNA. Therefore, for GC-MS analysis of ROS-induced DNA modification, it is important to incorporate procedures to prevent excessive sample oxidation, such as the addition of antioxidants. Other MS methods, HPLC-MS and electrospray ionization MS, are less prone to induce sample oxidation, and these approaches also allow precise identification of the nature of the chemical modifications. MS has been used to study oxidative and DNA damage in experimental models of cardiovascular disease. For example, in Ang II–induced hy.− pertension in mice, O2 formation, double-strand DNA breaks, and mutagenic DNA base modification 7,8-dihydro-8-oxoguanine were increased in the heart and kidney, effects that were attenuated by TEMPOL treatment.310 Similarly, Daehn et al311 described 8-oxoguanosine accumulation and evidence for DNA fragmentation linked to endothelial dysfunction in glomerulosclerosis. High-Pressure Liquid Chromatography HPLC coupled to electrochemical detection is another useful technique for measuring oxidatively modified DNA.309 It is a fairly sensitive technique and is also relatively inexpensive to use; however, it is not as sensitive as the LC/MS methods and does not enable identification of the molecular modification of the base. Immunoassays The use of antibodies that recognize 8-oxoguanine is another useful technique for analyzing oxidatively modified guanine.312,313 This approach has been used to “visualize” the base lesions with immunostaining or to provide a more comprehensive view of the modifications via Western blotting. For example, Martinet et al314 showed elevated 8-oxo-guanine in rabbit atherosclerotic plaques, and Mayr et al315 used anti8-oxoguanine antibodies to detect DNA damage in vascular smooth muscle cells exposed to mechanical stress. An ELISA can also be performed to quantify these modifications with a high-throughput plate reader approach. The caveats to this approach are similar to any measurements that use antibodybased analytical approaches; namely, that it is imperative to establish the specificity of the antibody because methods reported in the literature often use different concentrations of primary and secondary antibodies and different blocking agents. Also, the use of a secondary antibody in the absence of the primary antibody serves as an important control in immunostaining approaches to determine background or nonspecific staining. The principal advantage of this technique is that the use of antibody-based analyses is routine in many laboratories, and hence, these techniques can be readily extended to oxidatively modified DNA. The limitations are that only 1 base modification is analyzed, and as mentioned above, sometimes the sample preparation induces oxidative modification of guanine; thus, these measurements could overestimate the amount of injury that is occurring in a cell or tissue. Another limitation is that some antibodies raised against 8-oxoguanine cross-react with guanine, which would overestimate the number of oxidized bases.316,317 Hence, this approach is better suited for qualitative rather than quantitative analysis of DNA damage. Southern Blotting Oxidative damage to DNA can also be assessed by Southern blotting,318–320 and this can be applied to nuclear or mitochondrial DNA. For this procedure, known amounts of isolated DNA are incubated in sodium hydroxide to produce single-strand breaks at abasic sites present in the DNA because of repair of oxidatively modified bases. This mixture is electrophoresed and transferred to a nylon membrane, where it is hybridized with a 32P-labeled probe for DNA sequencing. The break frequency can be derived from the band intensity of the oxidatively modified DNA as a function of the intensity of the control DNA bands. An advantage of this technique is that there is no requirement for expensive, high-end equipment; only routine electrophoresis equipment, commonly available in most laboratories, is needed. The disadvantages are that the exact nature of the base lesion or lesions is not revealed and that the site where the lesion occurs cannot be identified. Polymerase Chain Reaction Several approaches have been adopted to evaluate oxidative DNA modification with PCR.321–323 One such approach involves the treatment of DNA with formamidopyrimidine glycosylase to produce strand cleavage at sites of oxidized purines, thus creating single-strand breaks that block PCR amplification. The difference in PCR amplification between formamidopyrimidine glycosylase–treated and untreated DNA is then used as an estimate of oxidative base damage. Data can be expressed as the change in lesion frequency normalized to a 1-kb sequence, calculated from the quotient of band intensities in the formamidopyrimidine glycosylase–treated and untreated samples. Another PCR strategy involves determining the concentration of e60 Circulation Research August 19, 2016 different sized PCR products (0.2, 2.9, and 16 kb) amplified from mitochondrial DNA.324 If there are no lesions, the products should all have the same concentration, but if there are lesions, there will be less concentration of the larger products than the smaller ones. Using this approach, DNA lesion frequencies (break points in DNA) are calculated as the amplification of damaged (treated) samples relative to the amplification of nondamaged controls (from genomic DNA), which results in a relative amplification ratio (treated/control). The principal advantage of the PCR approach is the relative ease of the assay; a disadvantage is that the specific nature of the base lesion is not known, nor is the identity of the nucleotide revealed by this approach. Wosniak et al325 used this method to assess mitochondrial DNA damage in rabbit vascular smooth muscle cells exposed to Ang II. Recommendations for Measurement Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 A variety of techniques can be used to assess oxidative DNA base damage, but the choice of the method depends on the nature of the research question (Figure 6). If the question pertains to the formation of a particular sugar or base modification, then an approach using MS is more informative. If instead the question pertains to general base modifications, then a less precise technique might Figure 6. Analytical methods of determining oxidative modification of DNA. If the desired measurement is chemical modification of bases or sugars, the best method is mass spectroscopic analysis. If this technique is not available, then high-performance liquid chromatography (HPLC) can be used, but with less capability of determining the specific modification. Antibody-based methods are only applicable for deoxyguanosine. If the goal is to determine reactive oxygen species–induced fragmentation or base lesions that can disrupt transcription, quantitative polymerase chain reaction (qPCR) offers a reasonably simple and quantitative approach. Southern analysis can also provide quantitative information but requires the use of radioactivity. The comet assay can show fragmentation but is difficult to quantify. Assays listed as primary (1°) are the most well accepted, secondary (2°) are more widely used, and tertiary (3°) are either less sensitive or less specific or should be verified with a second method. be applicable. The most important aspect of application of these techniques is careful preparation of the sample, as oxidation of bases during the analysis procedures overestimates oxidative modification of DNA and RNA. We refer the reader to Collins et al (Table I),326 in which oxidation of guanine was evaluated by a number of methods, with a range of nearly 4 orders of magnitude. This variation is in part attributable to the method used and to the sample preparation. Thus, no matter which technique is used to determine the oxidative modification of DNA or RNA by ROS, the measurements are only as good as the care and preparation of the samples. This is likely the most critical and overlooked aspect of these measurements. As a general measure of DNA damage, the comet assay or immunoassays for 8-oxoguanine can be used; alternatively, HPLC-based approaches represent a sensitive and economical alternative. However, for more detailed analysis and for identification and quantification of DNA oxidation, MS-based approaches (GC-MS, HPLC-MS, or electrospray ionization MS) are recommended. Measuring ROS and RNS and Oxidative Stress in Clinical/Translational Studies On the basis of preclinical studies showing that ROS/RNSinduced oxidative stress plays a key role in cardiovascular physiology and pathology, measurements of these species and oxidative stress should be useful in understanding cardiovascular function and disease in humans. However, because free radicals and other ROS/RNS are very reactive, they are difficult to measure. As a result, accurate .− measurements of ROS/RNS such as O2 , H2O2, or NO in human tissues and biological fluids is challenging. Hence, indirect methods that involve the measurement of stable end products or biomarkers of oxidative stress have been used to assess oxidative status in clinical and translational studies. Although several circulating biomarkers are currently used to assess oxidative stress in humans, an ideal biomarker, as defined by Giustarini et al327 (Table 3), has not yet been identified. Therefore, in human studies, measurements of multiple independent indices of oxidative stress are recommended. Several such biomarkers are currently being measured, primarily in research laboratories, but are not yet routine assays in clinical laboratories because their diagnostic value remains uncertain. Lipid Peroxidation Because of their relatively high stability, products of lipid peroxidation in blood and urine are frequently measured to assess oxidative stress in clinical and translational studies, including lipid hydroperoxides, MDA, F2-IsoPs, and HNE. Lipid Hydroperoxides Several clinical studies have used LOOH as an index of peroxidative injury of membrane lipids and oxidative stress.328,329 Such studies have shown that plasma levels of lipid hydroperoxides are higher in patients with ischemic heart disease, peripheral artery disease,330 heart failure,331 and type 1 diabetes mellitus.332 Increased levels of lipid hydroperoxides have also been detected in the venous effluent Griendling et al Measurement of Reactive Oxygen Species e61 Table 3. Criteria for an Ideal Clinical Biomarker of ROS/RNS 1. Chemically stable molecule 2. Unsusceptible to artificial generation or loss during storage or processing 3. Directly implicated in the onset or progression of disease 4. Specific for the ROS/RNS in question 5. Detectable in the target tissue 6. Present in sufficient and quantifiable concentrations 7. Low intravariability and intervariability in humans 8. Determined by an assay that is accurate, precise, specific, sensitive, robust, and interference-free 9. Measurable within a detection range of a reliable and robust analytical method Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 10.Free of confounding from dietary input or environmental factors 11.Consensus and establishment of reference values and intervals 12.Consensus and establishment of animal models RNS indicates reactive nitrogen species; and ROS, reactive oxygen species. of reperfused myocardium333 and in the plasma after thrombolytic therapy,334 which indicates that measurements of lipid hydroperoxide can be a reliable index of acute oxidative stress. Changes in plasma levels of lipid peroxides have also been measured to document changes in oxidative stress after drug intervention.335 In PREVENT (Prospective Randomized Evaluation of the Vascular Effects of Norvasc Trial), baseline serum LOOH levels, measured by the ferrous oxidation of xylenol orange (FOX) assay, were found to correlate with thiobarbituric reactive substances (TBARS) and with higher risk of vascular events.336 Overall, measurements of plasma LOOH levels are a useful index of oxidative stress, although it is unclear whether this is reflective of systemic oxidative stress or local oxidation of circulating lipoproteins. Malondialdehyde MDA is a stable end product of lipid peroxidation and can be measured as an index of oxidative stress; however, few studies measure MDA directly but instead rely on measurement of TBARS in the plasma. It has been reported that plasma TBARS levels are increased in patients with coronary artery disease, hypertension, atherosclerosis, diabetes mellitus, heart failure, stroke, and aging.337–341 Cigarette smoking is also associated with elevated levels of TBARS, which suggests that proatherogenic and vascular injury effects of smoking might be related to oxidative damage induced by lipid peroxidation.342 Some studies have shown that plasma TBARS can predict cardiovascular events. For example, it has been reported that elevated plasma TBARS predict carotid atherosclerotic plaque progression over a 3-year period, as validated by carotid wall thickness measured with ultrasound.343 In the PREVENT trial, patients with high TBARS levels at baseline were at increased risk of vascular events.336 The vascular risk reflected by TBARS was found to be independent of C-reactive protein, interleukin 6, and traditional risk factors.336 Of the oxidative stress biomarkers used clinically, plasma TBARS is among the most common; however, because of the nonspecificity of the test, it is not clear which specific species are being measured, and there is a high probability of increased artifactual generation of TBARS during sample processing. Isoprostanes Increased circulating and urine levels of F2-IsoPs have been demonstrated in various cardiovascular diseases associated with vascular injury, including hypertension, atherosclerosis, ischemia-reperfusion injury, and heart failure.344,345 In addition, in healthy people with cardiovascular disease risk factors, such as obesity, diabetes mellitus, and hyperlipidemia, plasma concentrations of F2-IsoPs are elevated,346 which suggests that indices of lipid peroxidation could be clinically relevant biomarkers of cardiovascular risk. In support of this concept, a recent prospective study that included 1002 patients with atrial fibrillation who had undergone anticoagulation therapy who were studied for 25 months demonstrated that the levels of 8-iso-PGF2α and sNOX2-dp (a marker of Nox2 levels) correlated with cardiovascular events.347 Using various modeling paradigms, it was shown that 8-iso-PGF2α predicted cardiovascular events and death. As such, it was suggested that F2IsoP might complement conventional risk factors in prediction of cardiovascular events. 4-Hydroxy-Trans-2-Nonenal Increasing experimental evidence indicates that HNE might have a dual role as both a marker of systemic oxidative stress and a contributor to the pathogenesis of cardiovascular disease.348,349 Despite experimental evidence linking HNE and cardiovascular disease, there is a paucity of information in humans. A few studies in dialyzed patients demonstrated that the plasma levels of MDA and HNE correlate with severity of cardiovascular disease.350,351 Nonenzymatic Total Antioxidant Capacity Total antioxidant capacity (TAC) is a measure of the combined antioxidant effect of the nonenzymatic defenses in biological fluids and does not take into account the enzymatic antioxidant systems such as superoxide dismutase, catalase, and peroxidase. The assay measures low-molecular-weight antioxidants, both water soluble and lipid soluble, and includes urate, bilirubin, vitamin C, thiols, flavonoids, carotenoids, and vitamin E.352 Experimental and clinical studies have shown, for the most part, low levels of TAC in various cardiovascular diseases.353–355TAC assays have also been used extensively in human studies evaluating the effects of dietary antioxidants. A recent comprehensive study reviewed prospective cohort studies and clinical trials relating to associations between plasma/dietary antioxidants (TAC) and cardiovascular events.356,357 In long-term, large-scale, population-based cohort studies, higher levels of TAC were associated with a lower risk of cardiovascular disease, which supports a protective effect of dietary antioxidant vitamins, carotenoids, and polyphenols. However, results from large randomized, controlled trials fail to support long-term use of single-antioxidant supplements for cardiovascular prevention because of their lack of benefit or even adverse effects on major cardiovascular events or e62 Circulation Research August 19, 2016 cancer. Although the use of antioxidant supplements has been reported to have no benefit on cardiovascular events in several large randomized, controlled trials, cohort studies still support the protective effects of dietary antioxidants on preventing cardiovascular disease. In particular, antioxidant vitamins and polyphenols exhibit high antioxidant capacity in vitro and cardioprotective effects in vivo. Although TAC assays could shed light on dietary antioxidant effects in specific patient cohorts, there are technical concerns regarding specificity and sensitivity of these assays, and as such, TAC of foods or populations should not yet be used to make decisions affecting population health.358 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 NO and Peroxynitrite as Biomarkers of Endothelial Dysfunction Biomarkers of impaired endothelial function reflect altered NO bioavailability, increased oxidative stress, coagulation, and endothelial inflammation. NO, produced by endothelial cells, is a major determinant of endothelium-dependent vasodilation and is an inhibitor of coagulation, inflammation, and oxidative stress359,360 and consequently has been considered as an important marker reflecting endothelial status. Because NO has a short half-life, plasma levels of oxidative degradation products of NO, including nitrite, nitrate, and nitrosothiols, have been used as surrogate indices of NO generation.361 The most commonly used approach to assess NO breakdown products is the Griess reaction assay, which can be used in various clinical samples such as urine, plasma, serum, and cerebrospinal fluid. The Griess assay, which is now available in commercial kits, can detect nitrite levels in clinical samples as low as 0.5 mmol/L.362,363 NO products can also be measured with chemiluminescence and fluorescence methodology,364,365 but the clinical utility of these approaches is still unclear. In addition to assessing levels of NO and its metabolites, measurement of asymmetric dimethylarginine, a potent endogenous inhibitor of NOS-derived NO production, can reflect NO bioavailability.366 Plasma asymmetric dimethylarginine levels are measured by LC/MS367 and have been shown to correlate with endothelial NOS activity and to be associated with endothelial dysfunction.368,369 Vascular injury is also associated with increased production of endothe.− lial O2 , which readily reacts with NO to form peroxynitrite (ONOO−), an injurious free radical that further contributes to endothelial dysfunction.370 Similarly, lipid peroxyl radicals react with NO and could also be a source of NO inactivation.371 Plasma measures of endothelial oxidative stress include indices of lipid peroxidation (F2-IsoPs and TBARS) and nitrotyrosine levels, reflecting peroxynitrite generation, although these measurements are not specific for endothelial ROS production.372 Lipid peroxidation is increased in diseases associated with endothelial dysfunction, and there is a strong negative correlation between oxidative stress and endothelial function as assessed by forearm blood flow responses to acetylcholine in the presence of the antioxidant vitamin C.373,374 Recommendations for Measurement Considering the important role of ROS and oxidative stress in the pathophysiology of cardiovascular disease, there is an increasing need to accurately measure oxidative status in the clinical setting, as well as in translational studies. Current approaches rely on the measurement of circulating biomarkers of oxidative stress (Figure 7). However, there are limitations to using such an approach, because (1) circulating levels of ROS biomarkers reflect generalized oxidative status without any information about the tissue or enzymatic source from which the ROS are generated; (2) biomarkers are indirect indices of ROS; and (3) current methodologies lack sensitivity and specificity. Development of new analytical methodologies should focus on direct measurement of free radicals, using assays that are user-friendly, practical, and of clinical utility. Because the ideal biomarker of oxidative stress has not yet been identified, it is prudent to measure multiple independent biomarkers when evaluating ROS status in humans. A practical guide to suggested biomarkers to evaluate ROS/RNS status in the clinic is summarized in Figure 8. Measurements of ROS/RNS and oxidative stress are often made in the context of antioxidant supplementation; Figure 7. Approaches to assess redox status in clinical and translational studies. Schematic showing simplified practical approaches to measuring oxidative status in circulating cells, blood, urine, and cerebral spinal fluid (CSF). These approaches are used primarily in clinical and translational research and are not used as routine assays in clinical laboratories. EPR indicates electron paramagnetic resonance spectroscopy; FL, fluorescence; GSH, reduced glutathione; GSSG, oxidized glutathione; HPLC, high-performance liquid chromatography; LC/MS, liquid chromatography/mass spectrometry; RBC, red blood cells; TAC, total antioxidant capacity; and WBC, white blood cells. Griendling et al Measurement of Reactive Oxygen Species e63 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 however, antioxidant supplementation does not automatically have an antioxidant effect, an issue incompletely considered in many previous clinical trials that failed to show benefit. Failure to detect changes in oxidative stress after antioxidant supplementation could have several possible causes: (1) Antioxidant pharmacokinetics did not allow sufficient accumulation of antioxidant in the target tissue. (2) Antioxidants did not have sufficient antioxidant potency; for example, intracellular superoxide dismutase .− reacts 10 000 times faster with O2 than the frequently used antioxidant supplement ascorbic acid. (3) Antioxidant supplements were not targeted to specific subcellular sites of ROS/RNS overproduction, such as mitochondria. It is conceivable that the use of antioxidants targeted to compartments where ROS are generated, such as the mitochondria, might be more effective.375 Indeed, recent studies with transgenic mice support the therapeutic potential of SOD overexpression in mitochondria and cytoplasm. (4) Finally, and relevant to this statement, many of these studies lack validation of an antioxidant effect by measurement of relevant biomarkers discussed herein. Hence, in assessing changes in the oxidative stress, it is important not only to measure changes in the ROS/RNS themselves but in their secondary products as well. Summary and Overall Recommendations Reliable and reproducible measures of ROS/RNS are difficult to achieve given the complexity of the biological systems with which they interact, their short half-lives and high reactivity, the balance between production and antioxidant capacity, and compartmentalization of reactivity and short diffusion distances. However, these molecules have both physiological and pathological roles, and precise measurements are not only desirable but also imperative for consistency across studies. The principles and analyses in this statement provide a basis for deciding which assays to use in specific experimental conditions, as well as recommendations for common use in various settings. As general guidance, we recommend that any method or assay used for measuring ROS or RNS should be specific, Figure 8. Decision tree for measuring oxidant status in clinical tissue. Clinical samples (blood, tissue, urine) can be assayed for pro-oxidant and antioxidant biomarkers with a variety of bioassays. For measuring pro-oxidants, levels of lipid peroxidation or DNA damage can be measured in biological fluids. Metabolites of lipid peroxidation products (aldehydes, isoprostanes, and hydroperoxides) by gas chromatography (GC)/liquid chromatography (LC) mass spectroscopy (MS) provide the most sensitive and specific assessment of oxidative stress and are shown as 1° assays. Proteins modified by 4-hydroxy-trans-2-nonenal and malondialdehyde can also be measured by Western analysis or other immunoassays, and isoprostanes can be measured by ELISA, but these measurements are generally less sensitive and less specific (2° assays). Lipid hydroperoxides (LOOH) can also be measured by high-performance liquid chromatography (HPLC). The use of nonspecific and nonselective measurements of lipid peroxidation products (thiobarbituric reactive substances [TBARS]) is not recommended (3° assay). DNA damage can be measured by quantifying modified bases either by GC/LC/MS (1° assay), HPLC, or reverse transcription polymerase chain reaction (RT-PCR; 2° assays) or by less specific immunoassays (3° assay). Assessment of antioxidant status could involve measurements of the total antioxidant capacity (TAC) or measurement of reduced glutathione (GSH)/oxidized glutathione (GSSG) or reduced cysteine (Cys)/oxidized cysteine (CySS) ratio either by HPLC (2° assays) or by colorimetric/fluorometric techniques, although these measurements are relatively less sensitive and prone to artifacts (3° assays) than other techniques. ORAC indicates oxygen radical absorbance capacity. e64 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Figure 9. Recommended approaches for the detection of reactive oxygen species (ROS)/reactive nitrogen species (RNS) and their effects: ROS and RNS can be quantified either by direct detection or by measuring secondary products or protein and DNA modification induced by ROS/RNS-initiated reactions. For rigorous documentation of significant ROS/RNS production, it is recommended that the ROS/RNS species be measured directly, along with the measurement of one of the consequences of their production (protein/DNA modification, thiol depletion, or lipid oxidation). Assays that provide the most sensitive and selective measurements of the species/product of interest are listed. Although the choice of the assay will depend on the specific research question of interest, at a minimum, measurement of >1 outcome of ROS/RNS production is recommended. DAF-2 indicates diaminofluorescein 2; EPR, electron paramagnetic resonance spectroscopy; Fe[DETC]2, Fe-diethyldithiocarbamate; GSH/GSSG, reduced glutathione/oxidized glutathione; HNE, 4-hydroxy-trans-2-nonenal; LC, liquid chromatography; MDA, malondialdehyde; MS, mass spectroscopy; SNO, S-nitrosylation; SOH, S-sulfenylation; and SSG, S-glutathionylation. sensitive, and reproducible. Given the highly reactive nature of these species, care should be taken to ensure the stability of the sample during isolation, preparation, storage, and analysis, which is particularly challenging when measuring oxidative modifications that can be influenced by exposure to ambient oxygen. Because of the ephemeral nature of many of these modifications, best practice is to confirm findings with >1 assay and to use appropriate controls. In addition, we recommend that investigators choose an array of assays that directly and specifically measure the ROS of interest, as well as its particular endpoints, rather than relying on global measures of oxidative stress (Figure 9). We also suggest that the values of antioxidants (such as glutathione) or products of oxidation (such as lipid peroxidation products) should be reported in absolute rather than relative values. Values of these metabolites expressed in molar concentrations are particularly useful for intertissue comparisons and for comparison of results obtained in different laboratories. Acknowledgment The authors thank Sofia Tsiropoulou for her help with the section on protein oxidation. Griendling et al Measurement of Reactive Oxygen Species e65 Disclosures Writing Group Disclosures Writing Group Member Other Research Support Speakers’ Bureau/ Honoraria Expert Witness Ownership Interest Consultant/ Advisory Board Other Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Employment Research Grant Kathy K. Griendling Emory University NIH (HL038206, HL095070)† None None None Ownership interest in hydrocyan dyes (intellectual property)* None None Rhian M. Touyz University of Glasgow Institute of Cardiovascular & Medical Sciences British Heart Foundation (RG/13/7/30099, RE/13/5/30177 and CH/12/4/29762)†; Canadian Institutes of Health Research† None None None None None None Aruni Bhatnagar University of Louisville NIH (HL55477, GM103492)† None None None None None None Yeong-Renn Chen Northeast Ohio Medical University None None None None None None None William Chilian Northeast Ohio Medical University NIH (RO1HL115114-04)† None None None None None None Sergey Dikalov Vanderbilt University Medical Center AHA*; NIH (R01 HL124116-01A1)† None None None None None None David G. Harrison Vanderbilt University Medical Center NIH† None None Covington and Burling† None None None Davis Heart and Lung Research Institute, The Ohio State University None None None None None None None Jay L. Zweier This table represents the relationships of writing group members that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure Questionnaire, which all members of the writing group are required to complete and submit. A relationship is considered to be “significant” if (1) the person receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (2) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition. *Modest. †Significant. Reviewer Disclosures Other Research Support Speakers’ Bureau/ Honoraria Expert Witness Ownership Interest Consultant/ Advisory Board Other Reviewer Employment Research Grant Ruhul Abid Brown University AHA (I have a Grant-in-Aid award that involves roles of ROS in coronary endothelium)*; NIH (I am a PI of COBRE Project grant that involves ROS measurement in vascular endothelium in vivo and in vitro)* None None None None None None CNR, Institute of Clinical Physiology Genetics Unit (Italy) None None None None None None None Northwestern University None None None None None None None University Hospital Jena (Germany) None None None None None None None Maria Grazia Andreassi Hossein Ardehali P. Christian Schulze This table represents the relationships of reviewers that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure Questionnaire, which all reviewers are required to complete and submit. A relationship is considered to be “significant” if (1) the person receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (2) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition. *Significant. e66 Circulation Research August 19, 2016 References Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 1.Imlay JA, Chin SM, Linn S. Toxic DNA damage by hydrogen peroxide through the Fenton reaction in vivo and in vitro. Science. 1988;240:640–642. 2. Stohs SJ, Bagchi D. Oxidative mechanisms in the toxicity of metal ions. Free Radic Biol Med. 1995;18:321–336. 3. Valko M, Morris H, Cronin MT. Metals, toxicity and oxidative stress. Curr Med Chem. 2005;12:1161–1208. 4. Arosio P, Ingrassia R, Cavadini P. Ferritins: a family of molecules for iron storage, antioxidation and more. Biochim Biophys Acta. 2009;1790:589– 599. doi: 10.1016/j.bbagen.2008.09.004. 5. Vasquez-Vivar J, Kalyanaraman B, Kennedy MC. Mitochondrial aconitase is a source of hydroxyl radical: an electron spin resonance investigation. J Biol Chem. 2000;275:14064–14069. 6. Xu H, Qu F, Xu H, Lai W, Andrew Wang Y, Aguilar ZP, Wei H. Role of reactive oxygen species in the antibacterial mechanism of silver nanoparticles on Escherichia coli O157:H7. Biometals. 2012;25:45–53. doi: 10.1007/s10534-011-9482-x. 7. Liu Q, Berchner-Pfannschmidt U, Möller U, Brecht M, Wotzlaw C, Acker H, Jungermann K, Kietzmann T. A Fenton reaction at the endoplasmic reticulum is involved in the redox control of hypoxia-inducible gene expression. Proc Natl Acad Sci U S A. 2004;101:4302–4307. doi: 10.1073/ pnas.0400265101. 8.Hughes MN. Chemistry of nitric oxide and related species. Methods Enzymol. 2008;436:3–19. doi: 10.1016/S0076-6879(08)36001-7. 9. Rosen GM, Freeman BA. Detection of superoxide generated by endothelial cells. Proc Natl Acad Sci U S A. 1984;81:7269–7273. 10. Zweier JL, Kuppusamy P, Lutty GA. Measurement of endothelial cell free radical generation: evidence for a central mechanism of free radical injury in postischemic tissues. Proc Natl Acad Sci U S A. 1988;85:4046–4050. 11.Wang P, Chen H, Qin H, Sankarapandi S, Becher MW, Wong PC, Zweier JL. Overexpression of human copper, zinc-superoxide dismutase (SOD1) prevents postischemic injury. Proc Natl Acad Sci U S A. 1998;95:4556–4560. 12. Zorov DB, Filburn CR, Klotz LO, Zweier JL, Sollott SJ. Reactive oxygen species (ROS)-induced ROS release: a new phenomenon accompanying induction of the mitochondrial permeability transition in cardiac myocytes. J Exp Med. 2000;192:1001–1014. 13. Kang PT, Zhang L, Chen CL, Chen J, Green KB, Chen YR. Protein thiyl radical mediates S-glutathionylation of complex I. Free Radic Biol Med. 2012;53:962–973. doi: 10.1016/j.freeradbiomed.2012.05.025. 14. Zweier JL. Measurement of superoxide-derived free radicals in the reperfused heart: evidence for a free radical mechanism of reperfusion injury. J Biol Chem. 1988;263:1353–1357. 15. Kang PT, Chen CL, Zen P, Guarini G, Chen YR. BCNU-induced gR2 defect mediates S-glutathionylation of complex I and respiratory uncoupling in myocardium. Biochem Pharmacol. 2014;89:490–502. doi: 10.1016/j. bcp.2014.03.012. 16. Tamura Y, Chi LG, Driscoll EM Jr, Hoff PT, Freeman BA, Gallagher KP, Lucchesi BR. Superoxide dismutase conjugated to polyethylene glycol provides sustained protection against myocardial ischemia/reperfusion injury in canine heart. Circ Res. 1988;63:944–959. 17. Galiñanes M, Qiu Y, Ezrin A, Hearse DJ. PEG-SOD and myocardial protection: studies in the blood- and crystalloid-perfused rabbit and rat hearts. Circulation. 1992;86:672–682. 18.Cai H, Li Z, Dikalov S, Holland SM, Hwang J, Jo H, Dudley SC Jr, Harrison DG. NAD(P)H oxidase-derived hydrogen peroxide mediates endothelial nitric oxide production in response to angiotensin II. J Biol Chem. 2002;277:48311–48317. doi: 10.1074/jbc.M208884200. 19. Burkitt MJ, Mason RP. Direct evidence for in vivo hydroxyl-radical generation in experimental iron overload: an ESR spin-trapping investigation. Proc Natl Acad Sci U S A. 1991;88:8440–8444. 20.Frejaville C, Karoui H, Tuccio B, Le Moigne F, Culcasi M, Pietri S, Lauricella R, Tordo P. 5-(Diethoxyphosphoryl)-5-methyl-1-pyrroline N-oxide: a new efficient phosphorylated nitrone for the in vitro and in vivo spin trapping of oxygen-centered radicals. J Med Chem. 1995;38:258–265. 21. Kadiiska MB, Burkitt MJ, Xiang QH, Mason RP. Iron supplementation generates hydroxyl radical in vivo: an ESR spin-trapping investigation. J Clin Invest. 1995;96:1653–1657. doi: 10.1172/JCI118205. 22. Sato K, Kadiiska MB, Ghio AJ, Corbett J, Fann YC, Holland SM, Thurman RG, Mason RP. In vivo lipid-derived free radical formation by NADPH oxidase in acute lung injury induced by lipopolysaccharide: a model for ARDS. FASEB J. 2002;16:1713–1720. doi: 10.1096/fj.02-0331com. 23. Zweier JL, Flaherty JT, Weisfeldt ML. Direct measurement of free radical generation following reperfusion of ischemic myocardium. Proc Natl Acad Sci U S A. 1987;84:1404–7. 24. Zweier JL, Kuppusamy P, Williams R, Rayburn BK, Smith D, Weisfeldt ML, Flaherty JT. Measurement and characterization of postischemic free radical generation in the isolated perfused heart. J Biol Chem. 1989;264:18890–18895. 25.Vásquez-Vivar J, Kalyanaraman B, Martásek P, Hogg N, Masters BS, Karoui H, Tordo P, Pritchard KA Jr. Superoxide generation by endothelial nitric oxide synthase: the influence of cofactors. Proc Natl Acad Sci U S A. 1998;95:9220–5. 26. Lee CI, Liu X, Zweier JL. Regulation of xanthine oxidase by nitric oxide and peroxynitrite. J Biol Chem. 2000;275:9369–9376. 27. Chen J, Chen CL, Alevriadou BR, Zweier JL, Chen YR. Excess NO predisposes mitochondrial succinate-cytochrome c reductase to produce hydroxyl radical. Biochim Biophys Acta. 2011;1807:491–502. doi: 10.1016/j. bbabio.2011.03.001. 28. Chen J, Chen CL, Rawale S, Chen CA, Zweier JL, Kaumaya PT, Chen YR. Peptide-based antibodies against glutathione-binding domains suppress superoxide production mediated by mitochondrial complex I. J Biol Chem. 2010;285:3168–3180. doi: 10.1074/jbc.M109.056846. 29. Chen YR, Chen CL, Yeh A, Liu X, Zweier JL. Direct and indirect roles of cytochrome b in the mediation of superoxide generation and NO catabolism by mitochondrial succinate-cytochrome c reductase. J Biol Chem. 2006;281:13159–13168. 30. Chen YR, Chen CL, Zhang L, Green-Church KB, Zweier JL. Superoxide generation from mitochondrial NADH dehydrogenase induces selfinactivation with specific protein radical formation. J Biol Chem. 2005;280:37339–37348. doi: 10.1074/jbc.M503936200. 31. Dikalov S, Griendling KK, Harrison DG. Measurement of reactive oxygen species in cardiovascular studies. Hypertension. 2007;49:717–727. doi: 10.1161/01.HYP.0000258594.87211.6b. 32.Han Y, Liu Y, Rockenbauer A, Zweier JL, Durand G, Villamena FA. Lipophilic beta-cyclodextrin cyclic-nitrone conjugate: synthesis and spin trapping studies. J Org Chem. 2009;74:5369–5380. doi: 10.1021/ jo900856x. 33.Kim SU, Liu Y, Nash KM, Zweier JL, Rockenbauer A, Villamena FA. Fast reactivity of a cyclic nitrone-calix[4]pyrrole conjugate with superoxide radical anion: theoretical and experimental studies. J Am Chem Soc. 2010;132:17157–17173. doi: 10.1021/ja105198c. 34.Lardinois OM, Detweiler CD, Tomer KB, Mason RP, Deterding LJ. Identifying the site of spin trapping in proteins by a combination of liquid chromatography, ELISA, and off-line tandem mass spectrometry. Free Radic Biol Med. 2008;44:893–906. doi: 10.1016/j. freeradbiomed.2007.11.015. 35.Michail K, Siraki AG. Post-trapping derivatization of radical-derived EPR-silent adducts: application to free radical detection by HPLC/UV in chemical, biochemical, and biological systems and comparison with EPR spectroscopy. Anal Chem. 2012;84:6739–6746. doi: 10.1021/ac301142c. 36. Khramtsov V, Berliner LJ, Clanton TL. NMR spin trapping: detection of free radical reactions using a phosphorus-containing nitrone spin trap. Magn Reson Med. 1999;42:228–234. 37. Gomez-Mejiba SE, Zhai Z, Akram H, Deterding LJ, Hensley K, Smith N, Towner RA, Tomer KB, Mason RP, Ramirez DC. Immuno-spin trapping of protein and DNA radicals: “tagging” free radicals to locate and understand the redox process. Free Radic Biol Med. 2009;46:853–865. doi: 10.1016/j.freeradbiomed.2008.12.020. 38. Ramirez DC, Mejiba SE, Mason RP. Immuno-spin trapping of DNA radicals. Nat Methods. 2006;3:123–127. doi: 10.1038/nmeth852. 39. Dikalova A, Clempus R, Lassègue B, Cheng G, McCoy J, Dikalov S, San Martin A, Lyle A, Weber DS, Weiss D, Taylor WR, Schmidt HH, Owens GK, Lambeth JD, Griendling KK. Nox1 overexpression potentiates angiotensin II-induced hypertension and vascular smooth muscle hypertrophy in transgenic mice. Circulation. 2005;112:2668–2676. doi: 10.1161/ CIRCULATIONAHA.105.538934. 40. Dudley SC Jr, Hoch NE, McCann LA, Honeycutt C, Diamandopoulos L, Fukai T, Harrison DG, Dikalov SI, Langberg J. Atrial fibrillation increases production of superoxide by the left atrium and left atrial appendage: role of the NADPH and xanthine oxidases. Circulation. 2005;112:1266–1273. doi: 10.1161/CIRCULATIONAHA.105.538108. 41. Rizzi C, Samouilov A, Kutala VK, Parinandi NL, Zweier JL, Kuppusamy P. Application of a trityl-based radical probe for measuring superoxide. Free Radic Biol Med. 2003;35:1608–1618. 42.Liu Y, Song Y, De Pascali F, Liu X, Villamena FA, Zweier JL. Tetrathiatriarylmethyl radical with a single aromatic hydrogen as a Griendling et al Measurement of Reactive Oxygen Species e67 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 highly sensitive and specific superoxide probe. Free Radic Biol Med. 2012;53:2081–2091. doi: 10.1016/j.freeradbiomed.2012.09.011. 43. Landmesser U, Dikalov S, Price SR, McCann L, Fukai T, Holland SM, Mitch WE, Harrison DG. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J Clin Invest. 2003;111:1201–1209. doi: 10.1172/JCI14172. 44.Guzik TJ, Mussa S, Gastaldi D, Sadowski J, Ratnatunga C, Pillai R, Channon KM. Mechanisms of increased vascular superoxide production in human diabetes mellitus: role of NAD(P)H oxidase and endothelial nitric oxide synthase. Circulation. 2002;105:1656–1662. 45. Azzi A, Montecucco C, Richter C. The use of acetylated ferricytochrome c for the detection of superoxide radicals produced in biological membranes. Biochem Biophys Res Commun. 1975;65:597–603. 46.Chen YR, Deterding LJ, Sturgeon BE, Tomer KB, Mason RP. Protein oxidation of cytochrome C by reactive halogen species enhances its peroxidase activity. J Biol Chem. 2002;277:29781–29791. doi: 10.1074/jbc. M200709200. 47.Choi HS, Kim JW, Cha YN, Kim C. A quantitative nitroblue tetrazolium assay for determining intracellular superoxide anion production in phagocytic cells. J Immunoassay Immunochem. 2006;27:31–44. doi: 10.1080/15321810500403722. 48.Tan AS, Berridge MV. Superoxide produced by activated neutrophils efficiently reduces the tetrazolium salt, WST-1 to produce a soluble formazan: a simple colorimetric assay for measuring respiratory burst activation and for screening anti-inflammatory agents. J Immunol Methods. 2000;238:59–68. 49. Zielonka J, Kalyanaraman B. Hydroethidine- and MitoSOX-derived red fluorescence is not a reliable indicator of intracellular superoxide formation: another inconvenient truth. Free Radic Biol Med. 2010;48:983–1001. doi: 10.1016/j.freeradbiomed.2010.01.028. 50. Nazarewicz RR, Bikineyeva A, Dikalov SI. Rapid and specific measurements of superoxide using fluorescence spectroscopy. J Biomol Screen. 2013;18:498–503. doi: 10.1177/1087057112468765. 51. Zhao H, Joseph J, Fales HM, Sokoloski EA, Levine RL, Vasquez-Vivar J, Kalyanaraman B. Detection and characterization of the product of hydroethidine and intracellular superoxide by HPLC and limitations of fluorescence [published correction appears in Proc Natl Acad Sci U S A. 2005;102:9086]. Proc Natl Acad Sci U S A. 2005;102:5727–5732. doi: 10.1073/pnas.0501719102. 52. Zhu X, Zuo L, Cardounel AJ, Zweier JL, He G. Characterization of in vivo tissue redox status, oxygenation, and formation of reactive oxygen species in postischemic myocardium. Antioxid Redox Signal. 2007;9:447–455. doi: 10.1089/ars.2006.1389. 53. Zuo L, Chen YR, Reyes LA, Lee HL, Chen CL, Villamena FA, Zweier JL. The radical trap 5,5-dimethyl-1-pyrroline N-oxide exerts dose-dependent protection against myocardial ischemia-reperfusion injury through preservation of mitochondrial electron transport. J Pharmacol Exp Ther. 2009;329:515–523. doi: 10.1124/jpet.108.143479. 54.Takimoto E, Champion HC, Li M, Ren S, Rodriguez ER, Tavazzi B, Lazzarino G, Paolocci N, Gabrielson KL, Wang Y, Kass DA. Oxidant stress from nitric oxide synthase-3 uncoupling stimulates cardiac pathologic remodeling from chronic pressure load. J Clin Invest. 2005;115:1221–1231. doi: 10.1172/JCI21968. 55.Laurindo FR, Fernandes DC, Santos CX. Assessment of superoxide production and NADPH oxidase activity by HPLC analysis of dihydroethidium oxidation products. Methods Enzymol. 2008;441:237–260. doi: 10.1016/S0076-6879(08)01213-5. 56. Xu S, Chamseddine AH, Carrell S, Miller FJ Jr. Nox4 NADPH oxidase contributes to smooth muscle cell phenotypes associated with unstable atherosclerotic plaques. Redox Biol. 2014;2:642–650. doi: 10.1016/j. redox.2014.04.004. 57.Dikalov SI, Nazarewicz RR, Bikineyeva A, Hilenski L, Lassègue B, Griendling KK, Harrison DG, Dikalova AE. Nox2-induced production of mitochondrial superoxide in angiotensin II-mediated endothelial oxidative stress and hypertension. Antioxid Redox Signal. 2014;20:281–294. doi: 10.1089/ars.2012.4918. 58.Zhang M, Takimoto E, Lee DI, Santos CX, Nakamura T, Hsu S, Jiang A, Nagayama T, Bedja D, Yuan Y, Eaton P, Shah AM, Kass DA. Pathological cardiac hypertrophy alters intracellular targeting of phosphodiesterase type 5 from nitric oxide synthase-3 to natriuretic peptide signaling. Circulation. 2012;126:942–951. doi: 10.1161/ CIRCULATIONAHA.112.090977. 59. Kalyanaraman B, Darley-Usmar V, Davies KJ, Dennery PA, Forman HJ, Grisham MB, Mann GE, Moore K, Roberts LJ 2nd, Ischiropoulos H. Measuring reactive oxygen and nitrogen species with fluorescent probes: challenges and limitations. Free Radic Biol Med. 2012;52:1–6. doi: 10.1016/j.freeradbiomed.2011.09.030. 60.Murphy N, Taylor W, Kundu K, Knight S, Lee S, inventors; Emory University, Georgia Tech Research Corporation, assignees. Reduced dye probes for the detection of radical oxygen species. US patent US8,628,753 B2. January 14, 2014. 61.Kundu K, Knight SF, Willett N, Lee S, Taylor WR and Murthy N. Hydrocyanines: a class of fluorescent sensors that can image reactive oxygen species in cell culture, tissue, and in vivo. Angew Chem Int Ed Engl. 2009;48:299–303. doi: 10.1002/anie.200804851. 62.Kuznetsov AV, Kehrer I, Kozlov AV, Haller M, Redl H, Hermann M, Grimm M, Troppmair J. Mitochondrial ROS production under cellular stress: comparison of different detection methods. Anal Bioanal Chem. 2011;400:2383–2390. doi: 10.1007/s00216-011-4764-2. 63.Mukhopadhyay P, Rajesh M, Yoshihiro K, Haskó G, Pacher P. Simple quantitative detection of mitochondrial superoxide production in live cells. Biochem Biophys Res Commun. 2007;358:203–208. doi: 10.1016/j. bbrc.2007.04.106. 64. Kervinen M, Pätsi J, Finel M, Hassinen IE. Lucigenin and coelenterazine as superoxide probes in mitochondrial and bacterial membranes. Anal Biochem. 2004;324:45–51. 65.Kirabo A, Fontana V, de Faria AP, Loperena R, Galindo CL, Wu J, Bikineyeva AT, Dikalov S, Xiao L, Chen W, Saleh MA, Trott DW, Itani HA, Vinh A, Amarnath V, Amarnath K, Guzik TJ, Bernstein KE, Shen XZ, Shyr Y, Chen SC, Mernaugh RL, Laffer CL, Elijovich F, Davies SS, Moreno H, Madhur MS, Roberts J 2nd, Harrison DG. DC isoketalmodified proteins activate T cells and promote hypertension. J Clin Invest. 2014;124:4642–4656. doi: 10.1172/JCI74084. 66. Zielonka J, Sikora A, Adamus J, Kalyanaraman B. Detection and differentiation between peroxynitrite and hydroperoxides using mitochondriatargeted arylboronic acid. Methods Mol Biol. 2015;1264:171–181. doi: 10.1007/978-1-4939-2257-4_16. 67. Vanin AF, Liu X, Samouilov A, Stukan RA, Zweier JL. Redox properties of iron-dithiocarbamates and their nitrosyl derivatives: implications for their use as traps of nitric oxide in biological systems. Biochim Biophys Acta. 2000;1474:365–377. 68. Zielonka J, Lambeth JD, Kalyanaraman B. On the use of L-012, a luminol-based chemiluminescent probe, for detecting superoxide and identifying inhibitors of NADPH oxidase: a reevaluation. Free Radic Biol Med. 2013;65:1310–1314. doi: 10.1016/j.freeradbiomed.2013.09.017. 69. Vásquez-Vivar J, Hogg N, Pritchard KA Jr, Martasek P, Kalyanaraman B. Superoxide anion formation from lucigenin: an electron spin resonance spin-trapping study. FEBS Lett. 1997;403:127–130. 70. Skatchkov MP, Sperling D, Hink U, Mülsch A, Harrison DG, Sindermann I, Meinertz T, Münzel T. Validation of lucigenin as a chemiluminescent probe to monitor vascular superoxide as well as basal vascular nitric oxide production. Biochem Biophys Res Commun. 1999;254:319–324. doi: 10.1006/bbrc.1998.9942. 71.Tarpey MM, White CR, Suarez E, Richardson G, Radi R, Freeman BA. Chemiluminescent detection of oxidants in vascular tissue: lucigenin but not coelenterazine enhances superoxide formation. Circ Res. 1999;84:1203–1211. 72.Guzik TJ, Channon KM. Measurement of vascular reactive oxy gen species production by chemiluminescence. Methods Mol Med. 2005;108:73–89. 73.Liu X, Zweier JL. A real-time electrochemical technique for measurement of cellular hydrogen peroxide generation and consumption: evaluation in human polymorphonuclear leukocytes. Free Radic Biol Med. 2001;31:894–901. 74.Druhan L, Liu X, Zweier JL. Tetrahydrobiopterin enhances hydrogen peroxide production by nitric oxide synthase while suppressing superoxide generation: implications for cardiovascular disease. Circulation. 2010;122:A21538. Abstract. 75.Weber DS, Rocic P, Mellis AM, Laude K, Lyle AN, Harrison DG, Griendling KK. Angiotensin II-induced hypertrophy is potentiated in mice overexpressing p22phox in vascular smooth muscle. Am J Physiol Heart Circ Physiol. 2005;288:H37–H42. doi: 10.1152/ajpheart.00638.2004. 76.Chen Q, Moghaddas S, Hoppel CL, Lesnefsky EJ. Ischemic defects in the electron transport chain increase the production of reactive oxygen species from isolated rat heart mitochondria. Am J Physiol Cell Physiol. 2008;294:C460–C466. doi: 10.1152/ajpcell.00211.2007. 77. Korge P, Ping P, Weiss JN. Reactive oxygen species production in energized cardiac mitochondria during hypoxia/reoxygenation: modulation by nitric oxide. Circ Res. 2008;103:873–880. doi: 10.1161/CIRCRESAHA. 108.180869. e68 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 78.Liu Y, Zhao H, Li H, Kalyanaraman B, Nicolosi AC, Gutterman DD. Mitochondrial sources of H2O2 generation play a key role in flow-mediated dilation in human coronary resistance arteries. Circ Res. 2003;93:573– 580. doi: 10.1161/01.RES.0000091261.19387.AE. 79.Khatri JJ, Johnson C, Magid R, Lessner SM, Laude KM, Dikalov SI, Harrison DG, Sung HJ, Rong Y, Galis ZS. Vascular oxidant stress enhances progression and angiogenesis of experimental atheroma. Circulation. 2004;109:520–525. doi: 10.1161/01.CIR.0000109698.70638.2B. 80. Tampo Y, Kotamraju S, Chitambar CR, Kalivendi SV, Keszler A, Joseph J, Kalyanaraman B. Oxidative stress-induced iron signaling is responsible for peroxide-dependent oxidation of dichlorodihydrofluorescein in endothelial cells: role of transferrin receptor-dependent iron uptake in apoptosis. Circ Res. 2003;92:56–63. 81.Kotamraju S, Kalivendi SV, Konorev E, Chitambar CR, Joseph J, Kalyanaraman B. Oxidant-induced iron signaling in doxorubicin-mediated apoptosis. Methods Enzymol. 2004;378:362–382. doi: 10.1016/ S0076-6879(04)78026-X. 82. Rota C, Fann YC, Mason RP. Phenoxyl free radical formation during the oxidation of the fluorescent dye 2′,7′-dichlorofluorescein by horseradish peroxidase: possible consequences for oxidative stress measurements. J Biol Chem. 1999;274:28161–28168. 83.Rota C, Chignell CF, Mason RP. Evidence for free radical formation during the oxidation of 2′-7′-dichlorofluorescin to the fluorescent dye 2′-7′-dichlorofluorescein by horseradish peroxidase: possible implications for oxidative stress measurements. Free Radic Biol Med. 1999;27:873–881. 84. Marchesi E, Rota C, Fann YC, Chignell CF, Mason RP. Photoreduction of the fluorescent dye 2′-7′-dichlorofluorescein: a spin trapping and direct electron spin resonance study with implications for oxidative stress measurements. Free Radic Biol Med. 1999;26:148–161. 85.Sartoretto JL, Kalwa H, Pluth MD, Lippard SJ, Michel T. Hydrogen peroxide differentially modulates cardiac myocyte nitric oxide synthesis. Proc Natl Acad Sci U S A. 2011;108:15792–15797. doi: 10.1073/ pnas.1111331108. 86. Sartoretto JL, Kalwa H, Romero N, Michel T. In vivo imaging of nitric oxide and hydrogen peroxide in cardiac myocytes. Methods Enzymol. 2013;528:61–78. doi: 10.1016/B978-0-12-405881-1.00004-5. 87.Malinouski M, Zhou Y, Belousov VV, Hatfield DL, Gladyshev VN. Hydrogen peroxide probes directed to different cellular compartments. PLoS One. 2011;6:e14564. doi: 10.1371/journal.pone.0014564. 88. Weller J, Kizina KM, Can K, Bao G, Müller M. Response properties of the genetically encoded optical H2O2 sensor HyPer. Free Radic Biol Med. 2014;76:227–241. doi: 10.1016/j.freeradbiomed.2014.07.045. 89. Bilan DS, Pase L, Joosen L, Gorokhovatsky AY, Ermakova YG, Gadella TW, Grabher C, Schultz C, Lukyanov S, Belousov VV. HyPer-3: a genetically encoded H(2)O(2) probe with improved performance for ratiometric and fluorescence lifetime imaging. ACS Chem Biol. 2013;8:535–542. doi: 10.1021/cb300625g. 90.Cairns AG, McQuaker SJ, Murphy MP, Hartley RC. Targeting mitochondria with small molecules: the preparation of MitoB and MitoP as exomarkers of mitochondrial hydrogen peroxide. Methods Mol Biol. 2015;1265:25–50. doi: 10.1007/978-1-4939-2288-8_3. 91.Cochemé HM, Logan A, Prime TA, Abakumova I, Quin C, McQuaker SJ, Patel JV, Fearnley IM, James AM, Porteous CM, Smith RA, Hartley RC, Partridge L, Murphy MP. Using the mitochondria-targeted ratiometric mass spectrometry probe MitoB to measure H2O2 in living Drosophila. Nat Protoc. 2012;7:946–958. doi: 10.1038/nprot.2012.035. 92.Dickinson BC, Chang CJ. A targetable fluorescent probe for imaging hydrogen peroxide in the mitochondria of living cells [published correction appears in J Am Chem Soc. 2008;130:11561]. J Am Chem Soc. 2008;130:9638–9639. doi: 10.1021/ja802355u. 93. Sun Z, Li H, Petryakov S, Samouilov A, Zweier JL. In vivo proton electron double resonance imaging of mice with fast spin echo pulse sequence. J Magn Reson Imaging. 2012;35:471–475. doi: 10.1002/jmri.22874. 94. Christensen CE, Karlsson M, Winther JR, Jensen PR, Lerche MH. Noninvasive in-cell determination of free cytosolic [NAD+]/[NADH] ratios using hyperpolarized glucose show large variations in metabolic phenotypes. J Biol Chem. 2014;289:2344–2352. doi: 10.1074/jbc.M113.498626. 95. Zhao Y, Jin J, Hu Q, Zhou HM, Yi J, Yu Z, Xu L, Wang X, Yang Y, Loscalzo J. Genetically encoded fluorescent sensors for intracellular NADH detection. Cell Metab. 2011;14:555–566. doi: 10.1016/j.cmet.2011.09.004. 96. Zhu X, Liu B, Zhou S, Chen YR, Deng Y, Zweier JL, He G. Ischemic preconditioning prevents in vivo hyperoxygenation in postischemic myocardium with preservation of mitochondrial oxygen consumption. Am J Physiol Heart Circ Physiol. 2007;293:H1442–H1450. 97.He G. Electron paramagnetic resonance oximetry and redoximetry. Methods Mol Biol. 2010;594:85–105. doi: 10.1007/978-1-60761-411-1_6. 98. Li H, He G, Deng Y, Kuppusamy P, Zweier JL. In vivo proton electron double resonance imaging of the distribution and clearance of nitroxide radicals in mice. Magn Reson Med. 2006;55:669–675. doi: 10.1002/ mrm.20804. 99. Liu Y, Villamena FA, Rockenbauer A, Zweier JL. Trityl-nitroxide biradicals as unique molecular probes for the simultaneous measurement of redox status and oxygenation. Chem Commun (Camb). 2010;46:628–630. doi: 10.1039/b919279d. 100. Kuppusamy P, Chzhan M, Vij K, Shteynbuk M, Lefer DJ, Giannella E, Zweier JL. Three-dimensional spectral-spatial EPR imaging of free radicals in the heart: a technique for imaging tissue metabolism and oxygenation. Proc Natl Acad Sci U S A. 1994;91:3388–92. 101. Liebgott T, Li H, Deng Y, Zweier JL. Proton electron double resonance imaging (PEDRI) of the isolated beating rat heart. Magn Reson Med. 2003;50:391–399. doi: 10.1002/mrm.10534. 102. Velayutham M, Li H, Kuppusamy P, Zweier JL. Mapping ischemic risk region and necrosis in the isolated heart using EPR imaging. Magn Reson Med. 2003;49:1181–1187. doi: 10.1002/mrm.10473. 103. Caia GL, Efimova OV, Velayutham M, El-Mahdy MA, Abdelghany TM, Kesselring E, Petryakov S, Sun Z, Samouilov A, Zweier JL. Organ specific mapping of in vivo redox state in control and cigarette smoke-exposed mice using EPR/NMR co-imaging. J Magn Reson. 2012;216:21–27. doi: 10.1016/j.jmr.2011.10.017. 104.Schafer FQ, Buettner GR. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med. 2001;30:1191–1212. 105.Jones DP, Liang Y. Measuring the poise of thiol/disulfide couples in vivo. Free Radic Biol Med. 2009;47:1329–1338. doi: 10.1016/j. freeradbiomed.2009.08.021. 106. Ashfaq S, Abramson JL, Jones DP, Rhodes SD, Weintraub WS, Hooper WC, Vaccarino V, Harrison DG, Quyyumi AA. The relationship between plasma levels of oxidized and reduced thiols and early atherosclerosis in healthy adults. J Am Coll Cardiol. 2006;47:1005–1011. doi: 10.1016/j. jacc.2005.09.063. 107. Meyer AJ, Dick TP. Fluorescent protein-based redox probes. Antioxid Redox Signal. 2010;13:621–650. doi: 10.1089/ars.2009.2948. 108. Dooley CT, Dore TM, Hanson GT, Jackson WC, Remington SJ, Tsien RY. Imaging dynamic redox changes in mammalian cells with green fluorescent protein indicators. J Biol Chem. 2004;279:22284–22293. doi: 10.1074/jbc.M312847200. 109. van Lith M, Tiwari S, Pediani J, Milligan G, Bulleid NJ. Real-time monitoring of redox changes in the mammalian endoplasmic reticulum. J Cell Sci. 2011;124(pt 14):2349–2356. doi: 10.1242/jcs.085530. 110.Waypa GB, Marks JD, Guzy R, Mungai PT, Schriewer J, Dokic D, Schumacker PT. Hypoxia triggers subcellular compartmental redox signaling in vascular smooth muscle cells. Circ Res. 2010;106:526–535. doi: 10.1161/CIRCRESAHA.109.206334. 111. Lohman JR, Remington SJ. Development of a family of redox-sensitive green fluorescent protein indicators for use in relatively oxidizing subcellular environments. Biochemistry. 2008;47:8678–8688. doi: 10.1021/ bi800498g. 112.Zhang H, Limphong P, Pieper J, Liu Q, Rodesch CK, Christians E, Benjamin IJ. Glutathione-dependent reductive stress triggers mitochondrial oxidation and cytotoxicity. FASEB J. 2012;26:1442–1451. doi: 10.1096/fj.11-199869. 113. Bryan NS, Grisham MB. Methods to detect nitric oxide and its metabolites in biological samples. Free Radic Biol Med. 2007;43:645–657. doi: 10.1016/j.freeradbiomed.2007.04.026. 114. Kalyanaraman B. Teaching the basics of redox biology to medical and graduate students: oxidants, antioxidants and disease mechanisms. Redox Biol. 2013;1:244–257. doi: 10.1016/j.redox.2013.01.014. 115.Radi R. Peroxynitrite, a stealthy biological oxidant. J Biol Chem. 2013;288:26464–26472. doi: 10.1074/jbc.R113.472936. 116. Koenitzer JR, Freeman BA. Redox signaling in inflammation: interactions of endogenous electrophiles and mitochondria in cardiovascular disease. Ann N Y Acad Sci. 2010;1203:45–52. doi: 10.1111/j.17496632.2010.05559.x. 117. Sun J, Zhang X, Broderick M, Fein H. Measurement of nitric oxide production in biological systems by using Griess reaction assay. Sensors. 2003;3:276–284. doi: 10.3390/s30800276. 118. Kleinbongard P, Dejam A, Lauer T, Jax T, Kerber S, Gharini P, Balzer J, Zotz RB, Scharf RE, Willers R, Schechter AN, Feelisch M, Kelm M. Plasma nitrite concentrations reflect the degree of endothelial Griendling et al Measurement of Reactive Oxygen Species e69 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 dysfunction in humans. Free Radic Biol Med. 2006;40:295–302. doi: 10.1016/j.freeradbiomed.2005.08.025. 119. Gladwin MT. Haldane, hot dogs, halitosis, and hypoxic vasodilation: the emerging biology of the nitrite anion. J Clin Invest. 2004;113:19–21. doi: 10.1172/JCI20664. 120.Kojima H, Urano Y, Kikuchi K, Higuchi T, Hirata Y, Nagano T. Fluorescent indicators for imaging nitric oxide production. Angew Chem Int Ed Engl. 1999;38:3209–3212. 121. Kojima H, Nakatsubo N, Kikuchi K, Kawahara S, Kirino Y, Nagoshi H, Hirata Y, Nagano T. Detection and imaging of nitric oxide with novel fluorescent indicators: diaminofluoresceins. Anal Chem. 1998;70:2446–2453. 122.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, Kelm M. Human red blood cells at work: identification and visualization of erythrocytic eNOS activity in health and disease. Blood. 2012;120:4229–4237. doi: 10.1182/ blood-2012-07-442277. 123. Nakatsubo N, Kojima H, Kikuchi K, Nagoshi H, Hirata Y, Maeda D, Imai Y, Irimura T, Nagano T. Direct evidence of nitric oxide production from bovine aortic endothelial cells using new fluorescence indicators: diaminofluoresceins. FEBS Lett. 1998;427:263–266. 124.Zhang X, Kim WS, Hatcher N, Potgieter K, Moroz LL, Gillette R, Sweedler JV. Interfering with nitric oxide measurements. 4,5-diaminofluorescein reacts with dehydroascorbic acid and ascorbic acid. J Biol Chem. 2002;277:48472–48478. doi: 10.1074/jbc.M209130200. 125. Pinder AG, Rogers SC, Khalatbari A, Ingram TE, James PE. The measurement of nitric oxide and its metabolites in biological samples by ozone-based chemiluminescence. Methods Mol Biol. 2008;476:11–28. 126.Hart CM, Kleinhenz DJ, Dikalov SI, Boulden BM, Dudley SC Jr. The measurement of nitric oxide production by cultured endothelial cells. Methods Enzymol. 2005;396:502–514. doi: 10.1016/ S0076-6879(05)96042-4. 127. Hendgen-Cotta U, Grau M, Rassaf T, Gharini P, Kelm M, Kleinbongard P. Reductive gas-phase chemiluminescence and flow injection analysis for measurement of the nitric oxide pool in biological matrices. Methods Enzymol. 2008;441:295–315. doi: 10.1016/S0076-6879(08)01216-0. 128.Woldman YY, Sun J, Zweier JL, Khramtsov VV. Direct chemiluminescence detection of nitric oxide in aqueous solutions using the natural nitric oxide target soluble guanylyl cyclase. Free Radic Biol Med. 2009;47:1339–1345. doi: 10.1016/j.freeradbiomed.2009.09.007. 129. Jiang S, Cheng R, Wang X, Xue T, Liu Y, Nel A, Huang Y, Duan X. Real-time electrical detection of nitric oxide in biological systems with sub-nanomolar sensitivity. Nat Commun. 2013;4:2225. doi: 10.1038/ ncomms3225. 130. Griveau S, Bedioui F. Overview of significant examples of electrochemical sensor arrays designed for detection of nitric oxide and relevant species in a biological environment. Anal Bioanal Chem. 2013;405:3475–3488. doi: 10.1007/s00216-012-6671-6. 131. Woldman YYu, Khramtsov VV, Grigor’ev IA, Kiriljuk IA, Utepbergenov DI. Spin trapping of nitric oxide by nitronylnitroxides: measurement of the activity of no synthase from rat cerebellum. Biochem Biophys Res Commun. 1994;202:195–203. 132.Bobko AA, Ivanov A, Khramtsov VV. Discriminative EPR detection of NO and HNO by encapsulated nitronyl nitroxides. Free Radic Res. 2013;47:74–81. doi: 10.3109/10715762.2012.746460. 133. Dikalov S, Fink B. ESR techniques for the detection of nitric oxide in vivo and in tissues. Methods Enzymol. 2005;396:597–610. doi: 10.1016/ S0076-6879(05)96052-7. 134.Samuni U, Samuni Y, Goldstein S. On the distinction between ni troxyl and nitric oxide using nitronyl nitroxides. J Am Chem Soc. 2010;132:8428–8432. doi: 10.1021/ja101945j. 135. Jiang J, Corbett J, Hogg N, Mason RP. An electron paramagnetic resonance investigation of the oxygen dependence of the arterial-venous gradient of nitrosyl hemoglobin in blood circulation. Free Radic Biol Med. 2007;43:1208–1215. doi: 10.1016/j.freeradbiomed.2007.06.024. 136.Kozlov AV, Szalay L, Umar F, Fink B, Kropik K, Nohl H, Redl H, Bahrami S. EPR analysis reveals three tissues responding to endotoxin by increased formation of reactive oxygen and nitrogen species. Free Radic Biol Med. 2003;34:1555–1562. 137. van Faassen EE, Koeners MP, Joles JA, Vanin AF. Detection of basal NO production in rat tissues using iron-dithiocarbamate complexes. Nitric Oxide. 2008;18:279–286. doi: 10.1016/j.niox.2008.02.003. 138. Cai H, Dikalov S, Griendling KK, Harrison DG. Detection of reactive oxygen species and nitric oxide in vascular cells and tissues: comparison of sensitivity and specificity. Methods Mol Med. 2007;139:293–311. 139. Zweier JL, Fertmann J, Wei G. Nitric oxide and peroxynitrite in postischemic myocardium. Antioxid Redox Signal. 2001;3:11–22. doi: 10.1089/152308601750100443. 140. Tsuchiya K, Kirima K, Yoshizumi M, Houchi H, Tamaki T, Mason RP. The role of thiol and nitrosothiol compounds in the nitric oxide-forming reactions of the iron-N-methyl-d-glucamine dithiocarbamate complex. Biochem J. 2002;367(pt 3):771–779. doi: 10.1042/BJ20020310. 141. Szabó C, Ischiropoulos H, Radi R. Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nat Rev Drug Discov. 2007;6:662–680. doi: 10.1038/nrd2222. 142. Laursen JB, Somers M, Kurz S, McCann L, Warnholtz A, Freeman BA, Tarpey M, Fukai T, Harrison DG. Endothelial regulation of vasomotion in apoE-deficient mice: implications for interactions between peroxynitrite and tetrahydrobiopterin. Circulation. 2001;103:1282–1288. 143. Niles JC, Wishnok JS, Tannenbaum SR. Peroxynitrite-induced oxidation and nitration products of guanine and 8-oxoguanine: structures and mechanisms of product formation. Nitric Oxide. 2006;14:109–121. doi: 10.1016/j.niox.2005.11.001. 144. Kooy NW, Royall JA, Ye YZ, Kelly DR, Beckman JS. Evidence for in vivo peroxynitrite production in human acute lung injury. Am J Respir Crit Care Med. 1995;151:1250–1254. doi: 10.1164/ajrccm.151.4.7697261. 145. Siervo M, Stephan BC, Feelisch M, Bluck LJ. Measurement of in vivo nitric oxide synthesis in humans using stable isotopic methods: a systematic review. Free Radic Biol Med. 2011;51:795–804. doi: 10.1016/j. freeradbiomed.2011.05.032. 146. Wu G, Meininger CJ. Analysis of citrulline, arginine, and methylarginines using high-performance liquid chromatography. Methods Enzymol. 2008;440:177–189. doi: 10.1016/S0076-6879(07)00810-5. 147.Curis E, Nicolis I, Moinard C, Osowska S, Zerrouk N, Bénazeth S, Cynober L. Almost all about citrulline in mammals. Amino Acids. 2005;29:177–205. doi: 10.1007/s00726-005-0235-4. 148. Bennike T, Birkelund S, Stensballe A, Andersen V. Biomarkers in inflammatory bowel diseases: current status and proteomics identification strategies. World J Gastroenterol. 2014;20:3231–3244. doi: 10.3748/wjg. v20.i12.3231. 149.Gertel S, Amital H. Putative approaches to bypass the citrulline-specific autoimmune response in rheumatoid arthritis. Isr Med Assoc J. 2014;16:587–590. 150. Dean RT, Fu S, Stocker R, Davies MJ. Biochemistry and pathology of radical-mediated protein oxidation. Biochem J. 1997;324(pt 1):1–18. 151. Shacter E. Quantification and significance of protein oxidation in biological samples. Drug Metab Rev. 2000;32:307–326. doi: 10.1081/ DMR-100102336. 152. Maron BA, Tang SS, Loscalzo J. S-nitrosothiols and the S-nitrosoproteome of the cardiovascular system. Antioxid Redox Signal. 2013;18:270–287. doi: 10.1089/ars.2012.4744. 153.Pimentel D, Haeussler DJ, Matsui R, Burgoyne JR, Cohen RA, Bachschmid MM. Regulation of cell physiology and pathology by protein S-glutathionylation: lessons learned from the cardiovascular system. Antioxid Redox Signal. 2012;16:524–542. doi: 10.1089/ ars.2011.4336. 154. Maheswaranathan M, Gole HK, Fernandez I, Lassègue B, Griendling KK, San Martín A. Platelet-derived growth factor (PDGF) regulates Slingshot phosphatase activity via Nox1-dependent auto-dephosphorylation of serine 834 in vascular smooth muscle cells. J Biol Chem. 2011;286:35430–35437. doi: 10.1074/jbc.M111.268284. 155. Cai Z, Yan LJ. Protein oxidative modifications: beneficial roles in disease and health. J Biochem Pharmacol Res. 2013;1:15–26. 156.Nagahara N, Matsumura T, Okamoto R, Kajihara Y. Protein cysteine modifications: (1) medical chemistry for proteomics. Curr Med Chem. 2009;16:4419–4444. 157. Vogt W. Oxidation of methionyl residues in proteins: tools, targets, and reversal. Free Radic Biol Med. 1995;18:93–105. 158. Dalle-Donne I, Giustarini D, Colombo R, Rossi R, Milzani A. Protein carbonylation in human diseases. Trends Mol Med. 2003;9:169–176. 159. Eaton P, Wright N, Hearse DJ, Shattock MJ. Glyceraldehyde phosphate dehydrogenase oxidation during cardiac ischemia and reperfusion. J Mol Cell Cardiol. 2002;34:1549–1560. 160. Rudyk O, Eaton P. Biochemical methods for monitoring protein thiol redox states in biological systems. Redox Biol. 2014;2:803–813. doi: 10.1016/j.redox.2014.06.005. 161.Rhee SG, Jeong W, Chang TS, Woo HA. Sulfiredoxin, the cysteine sulfinic acid reductase specific to 2-Cys peroxiredoxin: its discovery, mechanism of action, and biological significance. Kidney Int Suppl. 2007:S3–8. e70 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 162.Kumar V, Calamaras TD, Haeussler D, Colucci WS, Cohen RA, McComb ME, Pimentel D, Bachschmid MM. Cardiovascular redox and ox stress proteomics. Antioxid Redox Signal. 2012;17:1528–1559. doi: 10.1089/ars.2012.4706. 163. Burgoyne JR, Oka S, Ale-Agha N, Eaton P. Hydrogen peroxide sensing and signaling by protein kinases in the cardiovascular system. Antioxid Redox Signal. 2013;18:1042–1052. doi: 10.1089/ars.2012.4817. 164. Kim G, Weiss SJ, Levine RL. Methionine oxidation and reduction in proteins. Biochim Biophys Acta. 2014;1840:901–905. doi: 10.1016/j. bbagen.2013.04.038. 165. Haque A, Andersen JN, Salmeen A, Barford D, Tonks NK. Conformationsensing antibodies stabilize the oxidized form of PTP1B and inhibit its phosphatase activity. Cell. 2011;147:185–198. doi: 10.1016/j. cell.2011.08.036. 166. Tong X, Hou X, Jourd’heuil D, Weisbrod RM, Cohen RA. Upregulation of Nox4 by TGFβ1 oxidizes SERCA and inhibits NO in arterial smooth muscle of the prediabetic Zucker rat [published correction appears in Circ Res. 2012;110:e88–e89]. Circ Res. 2010;107:975–983. doi: 10.1161/CIRCRESAHA.110.221242. 167. Nelson KJ, Klomsiri C, Codreanu SG, Soito L, Liebler DC, Rogers LC, Daniel LW, Poole LB. Use of dimedone-based chemical probes for sulfenic acid detection methods to visualize and identify labeled proteins. Methods Enzymol. 2010;473:95–115. doi: 10.1016/S0076-6879(10)73004-4. 168. Paulsen CE, Truong TH, Garcia FJ, Homann A, Gupta V, Leonard SE, Carroll KS. Peroxide-dependent sulfenylation of the EGFR catalytic site enhances kinase activity. Nat Chem Biol. 2012;8:57–64. doi: 10.1038/ nchembio.736. 169. Meng TC, Hsu SF, Tonks NK. Development of a modified in-gel assay to identify protein tyrosine phosphatases that are oxidized and inactivated in vivo. Methods. 2005;35:28–36. doi: 10.1016/j.ymeth.2004.07.005. 170.Chouchani ET, James AM, Fearnley IM, Lilley KS, Murphy MP. Proteomic approaches to the characterization of protein thiol modification. Curr Opin Chem Biol. 2011;15:120–128. doi: 10.1016/j. cbpa.2010.11.003. 171. Held JM, Danielson SR, Behring JB, Atsriku C, Britton DJ, Puckett RL, Schilling B, Campisi J, Benz CC, Gibson BW. Targeted quantitation of site-specific cysteine oxidation in endogenous proteins using a differential alkylation and multiple reaction monitoring mass spectrometry approach. Mol Cell Proteomics. 2010;9:1400–1410. doi: 10.1074/mcp. M900643-MCP200. 172. Levine RL, Garland D, Oliver CN, Amici A, Climent I, Lenz AG, Ahn BW, Shaltiel S, Stadtman ER. Determination of carbonyl content in oxidatively modified proteins. Methods Enzymol. 1990;186:464–478. 173. Mohanty JG, Bhamidipaty S, Evans MK, Rifkind JM. A fluorimetric semi-microplate format assay of protein carbonyls in blood plasma. Anal Biochem. 2010;400:289–294. doi: 10.1016/j.ab.2010.01.032. 174. Yeh CC, Barr RG, Powell CA, Mesia-Vela S, Wang Y, Hamade NK, Austin JH, Santella RM. No effect of cigarette smoking dose on oxidized plasma proteins. Environ Res. 2008;106:219–225. doi: 10.1016/j. envres.2007.09.008. 175. Kostal V, Levar K, Swift M, Skillrud E, Chapman M, Thompson LV, Arriaga EA. Semi-automated image analysis: detecting carbonylation in subcellular regions of skeletal muscle. Anal Bioanal Chem. 2011;400:213–222. doi: 10.1007/s00216-011-4725-9. 176. Gao S, Qin T, Liu Z, Caceres MA, Ronchi CF, Chen CY, Yeum KJ, Taylor A, Blumberg JB, Liu Y, Shang F. Lutein and zeaxanthin supplementation reduces H2O2-induced oxidative damage in human lens epithelial cells. Mol Vis. 2011;17:3180–3190. 177. Winter ML, Liehr JG. Free radical-induced carbonyl content in protein of estrogen-treated hamsters assayed by sodium boro[3H]hydride reduction. J Biol Chem. 1991;266:14446–14450. 178. Hansen RE, Winther JR. An introduction to methods for analyzing thiols and disulfides: reactions, reagents, and practical considerations. Anal Biochem. 2009;394:147–158. doi: 10.1016/j.ab.2009.07.051. 179.Boivin B, Zhang S, Arbiser JL, Zhang ZY, Tonks NK. A modified cysteinyl-labeling assay reveals reversible oxidation of protein tyrosine phosphatases in angiomyolipoma cells. Proc Natl Acad Sci U S A. 2008;105:9959–9964. doi: 10.1073/pnas.0804336105. 180. De Pascali F, Hemann C, Samons K, Chen CA, Zweier JL. Hypoxia and reoxygenation induce endothelial nitric oxide synthase uncoupling in endothelial cells through tetrahydrobiopterin depletion and S-glutathionylation. Biochemistry. 2014;53:3679–3688. doi: 10.1021/ bi500076r. 181. Fu Y, Yang G, Zhu F, Peng C, Li W, Li H, Kim HG, Bode AM, Dong Z, Dong Z. Antioxidants decrease the apoptotic effect of 5-Fu in colon cancer by regulating Src-dependent caspase-7 phosphorylation. Cell Death Dis. 2014;5:e983. doi: 10.1038/cddis.2013.509. 182.Liang X, Kaya A, Zhang Y, Le DT, Hua D, Gladyshev VN. Characterization of methionine oxidation and methionine sulfoxide reduction using methionine-rich cysteine-free proteins. BMC Biochem. 2012;13:21. doi: 10.1186/1471-2091-13-21. 183. Klomsiri C, Rogers LC, Soito L, McCauley AK, King SB, Nelson KJ, Poole LB, Daniel LW. Endosomal H2O2 production leads to localized cysteine sulfenic acid formation on proteins during lysophosphatidic acid-mediated cell signaling. Free Radic Biol Med. 2014;71:49–60. doi: 10.1016/j.freeradbiomed.2014.03.017. 184. Poole LB, Klomsiri C, Knaggs SA, Furdui CM, Nelson KJ, Thomas MJ, Fetrow JS, Daniel LW, King SB. Fluorescent and affinity-based tools to detect cysteine sulfenic acid formation in proteins. Bioconjug Chem. 2007;18:2004–2017. doi: 10.1021/bc700257a. 185.Leonard SE, Reddie KG, Carroll KS. Mining the thiol proteome for sulfenic acid modifications reveals new targets for oxidation in cells. ACS Chem Biol. 2009;4:783–799. doi: 10.1021/cb900105q. 186.Leonard SE, Garcia FJ, Goodsell DS, Carroll KS. Redox-based probes for protein tyrosine phosphatases. Angew Chem Int Ed Engl. 2011;50:4423–7. 187.Seo YH, Carroll KS. Profiling protein thiol oxidation in tumor cells using sulfenic acid-specific antibodies. Proc Natl Acad Sci U S A. 2009;106:16163–16168. doi: 10.1073/pnas.0903015106. 188. Haque A, Tonks NK. The use of phage display to generate conformation-sensor recombinant antibodies. Nat Protoc. 2012;7:2127–2143. doi: 10.1038/nprot.2012.132. 189. Chen CA, Wang TY, Varadharaj S, Reyes LA, Hemann C, Talukder MA, Chen YR, Druhan LJ, Zweier JL. S-glutathionylation uncouples eNOS and regulates its cellular and vascular function. Nature. 2010;468:1115– 1118. doi: 10.1038/nature09599. 190. Adachi T, Weisbrod RM, Pimentel DR, Ying J, Sharov VS, Schöneich C, Cohen RA. S-Glutathiolation by peroxynitrite activates SERCA during arterial relaxation by nitric oxide. Nat Med. 2004;10:1200–1207. doi: 10.1038/nm1119. 191. Figtree GA, Liu CC, Bibert S, Hamilton EJ, Garcia A, White CN, Chia KK, Cornelius F, Geering K, Rasmussen HH. Reversible oxidative modification: a key mechanism of Na+-K+ pump regulation. Circ Res. 2009;105:185–193. doi: 10.1161/CIRCRESAHA.109.199547. 192.Wetzelberger K, Baba SP, Thirunavukkarasu M, Ho YS, Maulik N, Barski OA, Conklin DJ, Bhatnagar A. Postischemic deactivation of cardiac aldose reductase: role of glutathione S-transferase P and glutaredoxin in regeneration of reduced thiols from sulfenic acids. J Biol Chem. 2010;285:26135–26148. doi: 10.1074/jbc.M110.146423. 193. Chen FC, Ogut O. Decline of contractility during ischemia-reperfusion injury: actin glutathionylation and its effect on allosteric interaction with tropomyosin. Am J Physiol Cell Physiol. 2006;290:C719–C727. doi: 10.1152/ajpcell.00419.2005. 194. Charles RL, Schröder E, May G, Free P, Gaffney PR, Wait R, Begum S, Heads RJ, Eaton P. Protein sulfenation as a redox sensor: proteomics studies using a novel biotinylated dimedone analogue. Mol Cell Proteomics. 2007;6:1473–1484. doi: 10.1074/mcp.M700065-MCP200. 195. Saurin AT, Neubert H, Brennan JP, Eaton P. Widespread sulfenic acid formation in tissues in response to hydrogen peroxide. Proc Natl Acad Sci U S A. 2004;101:17982–17987. doi: 10.1073/pnas.0404762101. 196.Broniowska KA, Hogg N. The chemical biology of S-nitrosothiols. Antioxid Redox Signal. 2012;17:969–980. doi: 10.1089/ars.2012.4590. 197. Smith BC, Marletta MA. Mechanisms of S-nitrosothiol formation and selectivity in nitric oxide signaling. Curr Opin Chem Biol. 2012;16:498– 506. doi: 10.1016/j.cbpa.2012.10.016. 198. Foster MW, Hess DT, Stamler JS. Protein S-nitrosylation in health and disease: a current perspective. Trends Mol Med. 2009;15:391–404. doi: 10.1016/j.molmed.2009.06.007. 199. Corti A, Franzini M, Scataglini I, Pompella A. Mechanisms and targets of the modulatory action of S-nitrosoglutathione (GSNO) on inflammatory cytokines expression. Arch Biochem Biophys. 2014;562:80–91. doi: 10.1016/j.abb.2014.08.002. 200. Marino SM, Gladyshev VN. Structural analysis of cysteine S-nitrosylation: a modified acid-based motif and the emerging role of trans-nitrosylation. J Mol Biol. 2010;395:844–859. doi: 10.1016/j. jmb.2009.10.042. 201.Diers AR, Keszler A, Hogg N. Detection of S-nitrosothiols. Biochim Biophys Acta. 2014;1840:892–900. doi: 10.1016/j.bbagen.2013.07.026. 202. Chung HS, Wang SB, Venkatraman V, Murray CI, Van Eyk JE. Cysteine oxidative posttranslational modifications: emerging regulation in the Griendling et al Measurement of Reactive Oxygen Species e71 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 cardiovascular system. Circ Res. 2013;112:382–392. doi: 10.1161/ CIRCRESAHA.112.268680. 203. Gow AJ, Chen Q, Hess DT, Day BJ, Ischiropoulos H, Stamler JS. Basal and stimulated protein S-nitrosylation in multiple cell types and tissues. J Biol Chem. 2002;277:9637–9640. doi: 10.1074/jbc.C100746200. 204. Munson DA, Grubb PH, Kerecman JD, McCurnin DC, Yoder BA, Hazen SL, Shaul PW, Ischiropoulos H. Pulmonary and systemic nitric oxide metabolites in a baboon model of neonatal chronic lung disease. Am J Respir Cell Mol Biol. 2005;33:582–588. doi: 10.1165/rcmb.2005-0182OC. 205. Huang B, Chen C. An ascorbate-dependent artifact that interferes with the interpretation of the biotin switch assay. Free Radic Biol Med. 2006;41:562–567. doi: 10.1016/j.freeradbiomed.2006.03.006. 206. Li S, Wang H, Xian M, Whorton AR. Identification of protein nitrosothiols using phosphine-mediated selective reduction. Nitric Oxide. 2012;26:20–26. doi: 10.1016/j.niox.2011.11.001. 207.Chung KK, Dawson VL, Dawson TM. S-nitrosylation in Parkinson’s disease and related neurodegenerative disorders. Methods Enzymol. 2005;396:139–150. doi: 10.1016/S0076-6879(05)96014-X. 208. Gow A, Doctor A, Mannick J, Gaston B. S-Nitrosothiol measurements in biological systems. J Chromatogr B Analyt Technol Biomed Life Sci. 2007;851:140–151. doi: 10.1016/j.jchromb.2007.01.052. 209. Riccio DA, Nutz ST, Schoenfisch MH. Visible photolysis and amperometric detection of S-nitrosothiols. Anal Chem. 2012;84:851–856. doi: 10.1021/ac2031805. 210. Gladwin MT, Shelhamer JH, Schechter AN, Pease-Fye ME, Waclawiw MA, Panza JA, Ognibene FP, Cannon RO 3rd. Role of circulating nitrite and S-nitrosohemoglobin in the regulation of regional blood flow in humans. Proc Natl Acad Sci U S A. 2000;97:11482–11487. doi: 10.1073/ pnas.97.21.11482. 211.Samouilov A, Zweier JL. Development of chemiluminescence-based methods for specific quantitation of nitrosylated thiols. Anal Biochem. 1998;258:322–330. doi: 10.1006/abio.1998.2609. 212.Atar S, Ye Y, Lin Y, Freeberg SY, Nishi SP, Rosanio S, Huang MH, Uretsky BF, Perez-Polo JR, Birnbaum Y. Atorvastatin-induced cardioprotection is mediated by increasing inducible nitric oxide synthase and consequent S-nitrosylation of cyclooxygenase-2. Am J Physiol Heart Circ Physiol. 2006;290:H1960–H1968. doi: 10.1152/ajpheart.01137.2005. 213. Chouchani ET, Methner C, Nadtochiy SM, Logan A, Pell VR, Ding S, James AM, Cochemé HM, Reinhold J, Lilley KS, Partridge L, Fearnley IM, Robinson AJ, Hartley RC, Smith RA, Krieg T, Brookes PS, Murphy MP. Cardioprotection by S-nitrosation of a cysteine switch on mitochondrial complex I. Nat Med. 2013;19:753–759. doi: 10.1038/nm.3212. 214. Yakushev S. Cross talk between S-nitrosylation and S-glutathionylation in control of the Na,K-ATPase regulation in hypoxia. Am J Physiol Heart Circ Physiol. 2012;303:H1332–H1343. doi: 10.1152/ ajpheart.00145.2012. 215. Selemidis S, Dusting GJ, Peshavariya H, Kemp-Harper BK, Drummond GR. Nitric oxide suppresses NADPH oxidase-dependent superoxide production by S-nitrosylation in human endothelial cells. Cardiovasc Res. 2007;75:349–358. doi: 10.1016/j.cardiores.2007.03.030. 216. Choi H, Tostes RC, Webb RC. S-nitrosylation Inhibits protein kinase C-mediated contraction in mouse aorta. J Cardiovasc Pharmacol. 2011;57:65–71. doi: 10.1097/FJC.0b013e3181fef9cb. 217. Choi H, Allahdadi KJ, Tostes RC, Webb RC. Augmented S-nitrosylation contributes to impaired relaxation in angiotensin II hypertensive mouse aorta: role of thioredoxin reductase. J Hypertens. 2011;29:2359–2368. doi: 10.1097/HJH.0b013e32834d2554. 218. Erwin PA, Lin AJ, Golan DE, Michel T. Receptor-regulated dynamic S-nitrosylation of endothelial nitric-oxide synthase in vascular endothelial cells. J Biol Chem. 2005;280:19888–19894. doi: 10.1074/jbc. M413058200. 219.Detweiler CD, Deterding LJ, Tomer KB, Chignell CF, Germolec D, Mason RP. Immunological identification of the heart myoglobin radical formed by hydrogen peroxide. Free Radic Biol Med. 2002;33:364–369. 220. Gomez-Mejiba SE, Zhai Z, Della-Vedova MC, Muñoz MD, Chatterjee S, Towner RA, Hensley K, Floyd RA, Mason RP, Ramirez DC. Immunospin trapping from biochemistry to medicine: advances, challenges, and pitfalls: focus on protein-centered radicals. Biochim Biophys Acta. 2014;1840:722–729. doi: 10.1016/j.bbagen.2013.04.039. 221.Mason RP. Using anti-5,5-dimethyl-1-pyrroline N-oxide (anti-DM PO) to detect protein radicals in time and space with immuno-spin trapping. Free Radic Biol Med. 2004;36:1214–1223. doi: 10.1016/j. freeradbiomed.2004.02.077. 222. Kang PT, Chen CL, Chen YR. Increased mitochondrial prooxidant activity mediates up-regulation of complex I S-glutathionylation via protein thiyl radical in the murine heart of eNOS(-/-). Free Radic Biol Med. 2015;79:56–68. doi: 10.1016/j.freeradbiomed.2014.11.016. 223. Chen CA, Lin CH, Druhan LJ, Wang TY, Chen YR, Zweier JL. Superoxide induces endothelial nitric-oxide synthase protein thiyl radical formation, a novel mechanism regulating eNOS function and coupling. J Biol Chem. 2011;286:29098–29107. doi: 10.1074/jbc.M111.240127. 224. Towner RA, Garteiser P, Bozza F, Smith N, Saunders D, d’ Avila JC, Magno F, Oliveira MF, Ehrenshaft M, Lupu F, Silasi-Mansat R, Ramirez DC, Gomez-Mejiba SE, Mason RP, Castro Faria-Neto HC. In vivo detection of free radicals in mouse septic encephalopathy using molecular MRI and immuno-spin trapping. Free Radic Biol Med. 2013;65:828– 837. doi: 10.1016/j.freeradbiomed.2013.08.172. 225. Towner RA, Smith N, Saunders D, Henderson M, Downum K, Lupu F, Silasi-Mansat R, Ramirez DC, Gomez-Mejiba SE, Bonini MG, Ehrenshaft M, Mason RP. In vivo imaging of immuno-spin trapped radicals with molecular magnetic resonance imaging in a diabetic mouse model. Diabetes. 2012;61:2405–2413. doi: 10.2337/db11-1540. 226. Feeney MB, Schöneich C. Proteomic approaches to analyze protein tyrosine nitration. Antioxid Redox Signal. 2013;19:1247–1256. doi: 10.1089/ ars.2012.5058. 227. Nuriel T, Hansler A, Gross SS. Protein nitrotryptophan: formation, significance and identification. J Proteomics. 2011;74:2300–2312. doi: 10.1016/j.jprot.2011.05.032. 228. Carballal S, Bartesaghi S, Radi R. Kinetic and mechanistic considerations to assess the biological fate of peroxynitrite. Biochim Biophys Acta. 2014;1840:768–780. doi: 10.1016/j.bbagen.2013.07.005. 229.Nuriel T, Deeb RS, Hajjar DP, Gross SS. Protein 3-nitrotyrosine in complex biological samples: quantification by high-pressure liquid chromatography/electrochemical detection and emergence of proteomic approaches for unbiased identification of modification sites. Methods Enzymol. 2008;441:1–17. doi: 10.1016/S0076-6879(08)01201-9. 230. Ikeda K, Yukihiro Hiraoka B, Iwai H, Matsumoto T, Mineki R, Taka H, Takamori K, Ogawa H, Yamakura F. Detection of 6-nitrotryptophan in proteins by Western blot analysis and its application for peroxynitrite-treated PC12 cells. Nitric Oxide. 2007;16:18–28. doi: 10.1016/j. niox.2006.04.263. 231. Go YM, Jones DP. Thioredoxin redox Western analysis. In: Curr Protoc Toxicol. 2009;chapter 17:unit17.12. doi: 10.1002/0471140856.tx1712s41. 232.Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med. 1991;11:81–128. 233. Girotti AW. Lipid hydroperoxide generation, turnover, and effector action in biological systems. J Lipid Res. 1998;39:1529–1542. 234. Yin H, Xu L, Porter NA. Free radical lipid peroxidation: mechanisms and analysis. Chem Rev. 2011;111:5944–5972. doi: 10.1021/cr200084z. 235. Guéraud F, Atalay M, Bresgen N, Cipak A, Eckl PM, Huc L, Jouanin I, Siems W, Uchida K. Chemistry and biochemistry of lipid peroxidation products. Free Radic Res. 2010;44:1098–1124. doi: 10.3109/10715762.2010.498477. 236. Yamamoto Y, Frei B, Ames BN. Assay of lipid hydroperoxides using high-performance liquid chromatography with isoluminal chemiluminescence detection. Methods Enzymol. 1990;186:371–380. 237. Korytowski W, Geiger PG, Girotti AW. High-performance liquid chromatography with mercury cathode electrochemical detection: application to lipid hydroperoxide analysis. J Chromatogr B Biomed Appl. 1995;670:189–197. 238.Hui SP, Chiba H, Sakurai T, Asakawa C, Nagasaka H, Murai T, Ide H, Kurosawa T. An improved HPLC assay for phosphatidylcholine hydroperoxides (PCOOH) in human plasma with synthetic PCOOH as internal standard. J Chromatogr B Analyt Technol Biomed Life Sci. 2007;857:158–163. doi: 10.1016/j.jchromb.2007.06.019. 239.Hui SP, Murai T, Yoshimura T, Chiba H, Nagasaka H, Kurosawa T. Improved HPLC assay for lipid peroxides in human plasma using the internal standard of hydroperoxide. Lipids. 2005;40:515–522. 240.Tyurin VA, Tyurina YY, Ritov VB, Lysytsya A, Amoscato AA, Kochanek PM, Hamilton R, Dekosky ST, Greenberger JS, Bayir H, Kagan VE. Oxidative lipidomics of apoptosis: quantitative assessment of phospholipid hydroperoxides in cells and tissues. Methods Mol Biol. 2010;610:353–374. doi: 10.1007/978-1-60327-029-8_21. 241. Benedetti A, Comporti M, Esterbauer H. Identification of 4-hydroxynonenal as a cytotoxic product originating from the peroxidation of liver microsomal lipids. Biochim Biophys Acta. 1980;620:281–296. 242.Yan LJ, Forster MJ. Chemical probes for analysis of carbonylated proteins: a review. J Chromatogr B Analyt Technol Biomed Life Sci. 2011;879:1308–1315. doi: 10.1016/j.jchromb.2010.08.004. e72 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 243.van Kuijk FJ, Siakotos AN, Fong LG, Stephens RJ, Thomas DW. Quantitative measurement of 4-hydroxyalkenals in oxidized low-density lipoprotein by gas chromatography-mass spectrometry. Anal Biochem. 1995;224:420–424. 244.Selley ML. Determination of the lipid peroxidation product (E)-4 hydroxy-2-nonenal in clinical samples by gas chromatography–negativeion chemical ionisation mass spectrometry of the O-pentafluorobenzyl oxime. J Chromatogr B Biomed Sci Appl. 1997;691:263–268. 245. Shinmura K, Bolli R, Liu SQ, Tang XL, Kodani E, Xuan YT, Srivastava S, Bhatnagar A. Aldose reductase is an obligatory mediator of the late phase of ischemic preconditioning. Circ Res. 2002;91:240–246. 246.Srivastava S, Vladykovskaya E, Barski OA, Spite M, Kaiserova K, Petrash JM, Chung SS, Hunt G, Dawn B, Bhatnagar A. Aldose reductase protects against early atherosclerotic lesion formation in apolipoprotein E-null mice. Circ Res. 2009;105:793–802. doi: 10.1161/ CIRCRESAHA.109.200568. 247.Gioacchini AM, Calonghi N, Boga C, Cappadone C, Masotti L, Roda A, Traldi P. Determination of 4-hydroxy-2-nonenal at cellular levels by means of electrospray mass spectrometry. Rapid Commun Mass Spectrom. 1999;13:1573–1579. doi: 10.1002/(SICI)1097-0231 (19990815)13:15<1573::AID-RCM675>3.0.CO;2-Z. 248. Zanardi E, Jagersma CG, Ghidini S, Chizzolini R. Solid phase extraction and liquid chromatography-tandem mass spectrometry for the evaluation of 4-hydroxy-2-nonenal in pork products. J Agric Food Chem. 2002;50:5268–5272. 249. Méndez D, Hernáez ML, Kamali AN, Diez A, Puyet A, Bautista JM. Differential carbonylation of cytoskeletal proteins in blood group O erythrocytes: potential role in protection against severe malaria. Infect Genet Evol. 2012;12:1780–1787. doi: 10.1016/j.meegid.2012.06.013. 250.Vladykovskaya E, Sithu SD, Haberzettl P, Wickramasinghe NS, Merchant ML, Hill BG, McCracken J, Agarwal A, Dougherty S, Gordon SA, Schuschke DA, Barski OA, O’Toole T, D’Souza SE, Bhatnagar A, Srivastava S. Lipid peroxidation product 4-hydroxy-trans-2-nonenal causes endothelial activation by inducing endoplasmic reticulum stress. J Biol Chem. 2012;287:11398–11409. doi: 10.1074/jbc.M111.320416. 251. Chavez JD, Wu J, Bisson W, Maier CS. Site-specific proteomic analysis of lipoxidation adducts in cardiac mitochondria reveals chemical diversity of 2-alkenal adduction. J Proteomics. 2011;74:2417–2429. doi: 10.1016/j.jprot.2011.03.031. 252. Uchida K, Szweda LI, Chae HZ, Stadtman ER. Immunochemical detection of 4-hydroxynonenal protein adducts in oxidized hepatocytes. Proc Natl Acad Sci U S A. 1993;90:8742–8746. 253. Toyokuni S, Miyake N, Hiai H, Hagiwara M, Kawakishi S, Osawa T, Uchida K. The monoclonal antibody specific for the 4-hydroxy-2-nonenal histidine adduct. FEBS Lett. 1995;359:189–191. 254. Waeg G, Dimsity G, Esterbauer H. Monoclonal antibodies for detection of 4-hydroxynonenal modified proteins. Free Radic Res. 1996;25:149–159. 255.Alary J, Debrauwer L, Fernandez Y, Cravedi JP, Rao D, Bories G. 1,4-Dihydroxynonene mercapturic acid, the major end metabolite of exogenous 4-hydroxy-2-nonenal, is a physiological component of rat and human urine. Chem Res Toxicol. 1998;11:130–135. doi: 10.1021/ tx970139w. 256. Kuiper HC, Miranda CL, Sowell JD, Stevens JF. Mercapturic acid conjugates of 4-hydroxy-2-nonenal and 4-oxo-2-nonenal metabolites are in vivo markers of oxidative stress. J Biol Chem. 2008;283:17131–17138. doi: 10.1074/jbc.M802797200. 257.Dixit BL, Balendiran GK, Watowich SJ, Srivastava S, Ramana KV, Petrash JM, Bhatnagar A, Srivastava SK. Kinetic and structural characterization of the glutathione-binding site of aldose reductase. J Biol Chem. 2000;275:21587–21595. doi: 10.1074/jbc.M909235199. 258.Linhart I, Frantík E, Vodicková L, Vosmanská M, Smejkal J, Mitera J. Biotransformation of acrolein in rat: excretion of mercapturic acids after inhalation and intraperitoneal injection. Toxicol Appl Pharmacol. 1996;136:155–160. 259. Kuiper HC, Bruno RS, Traber MG, Stevens JF. Vitamin C supplementation lowers urinary levels of 4-hydroperoxy-2-nonenal metabolites in humans. Free Radic Biol Med. 2011;50:848–853. doi: 10.1016/j. freeradbiomed.2011.01.004. 260. Carmella SG, Chen M, Zhang Y, Zhang S, Hatsukami DK, Hecht SS. Quantitation of acrolein-derived (3-hydroxypropyl)mercapturic acid in human urine by liquid chromatography-atmospheric pressure chemical ionization tandem mass spectrometry: effects of cigarette smoking. Chem Res Toxicol. 2007;20:986–990. doi: 10.1021/tx700075y. 261. DeJarnett N, Conklin DJ, Riggs DW, Myers JA, O’Toole TE, Hamzeh I, Wagner S, Chugh A, Ramos KS, Srivastava S, Higdon D, Tollerud DJ, DeFilippis A, Becher C, Wyatt B, McCracken J, Abplanalp W, Rai SN, Ciszewski T, Xie Z, Yeager R, Prabhu SD, Bhatnagar A. Acrolein exposure is associated with increased cardiovascular disease risk. J Am Heart Assoc. 2014;3:e000934. doi: 10.1161/ JAHA.114.000934. 262. Watson AD, Leitinger N, Navab M, Faull KF, Hörkkö S, Witztum JL, Palinski W, Schwenke D, Salomon RG, Sha W, Subbanagounder G, Fogelman AM, Berliner JA. Structural identification by mass spectrometry of oxidized phospholipids in minimally oxidized low density lipoprotein that induce monocyte/endothelial interactions and evidence for their presence in vivo. J Biol Chem. 1997;272:13597–13607. 263. Watson AD, Subbanagounder G, Welsbie DS, Faull KF, Navab M, Jung ME, Fogelman AM, Berliner JA. Structural identification of a novel proinflammatory epoxyisoprostane phospholipid in mildly oxidized low density lipoprotein. J Biol Chem. 1999;274:24787–24798. 264.Tyurina YY, Domingues RM, Tyurin VA, Maciel E, Domingues P, Amoscato AA, Bayir H, Kagan VE. Characterization of cardiolipins and their oxidation products by LC-MS analysis. Chem Phys Lipids. 2014;179:3–10. doi: 10.1016/j.chemphyslip.2013.12.003. 265. Tsimikas S, Bergmark C, Beyer RW, Patel R, Pattison J, Miller E, Juliano J, Witztum JL. Temporal increases in plasma markers of oxidized lowdensity lipoprotein strongly reflect the presence of acute coronary syndromes. J Am Coll Cardiol. 2003;41:360–370. 266. Tsimikas S, Brilakis ES, Miller ER, McConnell JP, Lennon RJ, Kornman KS, Witztum JL, Berger PB. Oxidized phospholipids, Lp(a) lipoprotein, and coronary artery disease. N Engl J Med. 2005;353:46–57. doi: 10.1056/NEJMoa043175. 267.Leibundgut G, Arai K, Orsoni A, Yin H, Scipione C, Miller ER, Koschinsky ML, Chapman MJ, Witztum JL, Tsimikas S. Oxidized phospholipids are present on plasminogen, affect fibrinolysis, and increase following acute myocardial infarction. J Am Coll Cardiol. 2012;59:1426–1437. doi: 10.1016/j.jacc.2011.12.033. 268. Palinski W, Hörkkö S, Miller E, Steinbrecher UP, Powell HC, Curtiss LK, Witztum JL. Cloning of monoclonal autoantibodies to epitopes of oxidized lipoproteins from apolipoprotein E-deficient mice: demonstration of epitopes of oxidized low density lipoprotein in human plasma. J Clin Invest. 1996;98:800–814. doi: 10.1172/JCI118853. 269. Vladykovskaya E, Ozhegov E, Hoetker JD, Xie Z, Ahmed Y, Suttles J, Srivastava S, Bhatnagar A, Barski OA. Reductive metabolism increases the proinflammatory activity of aldehyde phospholipids. J Lipid Res. 2011;52:2209–2225. doi: 10.1194/jlr.M013854. 270.Srivastava S, Spite M, Trent JO, West MB, Ahmed Y, Bhatnagar A. Aldose reductase-catalyzed reduction of aldehyde phospholipids. J Biol Chem. 2004;279:53395–53406. doi: 10.1074/jbc.M403416200. 271. Berliner JA, Watson AD. A role for oxidized phospholipids in atherosclerosis [published correction appears in N Engl J Med. 2005;353:1869]. N Engl J Med. 2005;353:9–11. doi: 10.1056/NEJMp058118. 272.Morrow JD, Roberts LJ 2nd. Mass spectrometry of prostanoids: F2isoprostanes produced by non-cyclooxygenase free radical-catalyzed mechanism. Methods Enzymol. 1994;233:163–174. 273.Milne GL, Dai Q, Roberts LJ, 2nd. The isoprostanes: 25 years later. Biochim Biophys Acta. 2014;1851:433–445. 274. Vigor C, Bertrand-Michel J, Pinot E, Oger C, Vercauteren J, Le Faouder P, Galano JM, Lee JC, Durand T. Non-enzymatic lipid oxidation products in biological systems: assessment of the metabolites from polyunsaturated fatty acids. J Chromatogr B Analyt Technol Biomed Life Sci. 2014;964:65–78. doi: 10.1016/j.jchromb.2014.04.042. 275.Klawitter J, Haschke M, Shokati T, Klawitter J, Christians U. Quantification of 15-F2t-isoprostane in human plasma and urine: results from enzyme-linked immunoassay and liquid chromatography/tandem mass spectrometry cannot be compared. Rapid Commun Mass Spectrom. 2011;25:463–468. doi: 10.1002/rcm.4871. 276. Il’yasova D, Morrow JD, Ivanova A, Wagenknecht LE. Epidemiological marker for oxidant status: comparison of the ELISA and the gas chromatography/mass spectrometry assay for urine 2,3-dinor-5,6-dihydro15-F2t-isoprostane. Ann Epidemiol. 2004;14:793–797. doi: 10.1016/j. annepidem.2004.03.003. 277. Milne GL, Gao B, Terry ES, Zackert WE, Sanchez SC. Measurement of F2- isoprostanes and isofurans using gas chromatography-mass spectrometry. Free Radic Biol Med. 2013;59:36–44. doi: 10.1016/j. freeradbiomed.2012.09.030. 278. Briskey DR, Wilson GR, Fassett RG, Coombes JS. Optimized method for quantification of total F(2)-isoprostanes using gas chromatographytandem mass spectrometry. J Pharm Biomed Anal. 2014;90:161–166. doi: 10.1016/j.jpba.2013.11.028. Griendling et al Measurement of Reactive Oxygen Species e73 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 279.Brose SA, Baker AG, Golovko MY. A fast one-step extraction and UPLC-MS/MS analysis for E2/D 2 series prostaglandins and isoprostanes. Lipids. 2013;48:411–419. doi: 10.1007/s11745-013-3767-5. 280. Labuschagne CF, van den Broek NJ, Postma P, Berger R, Brenkman AB. A protocol for quantifying lipid peroxidation in cellular systems by F2isoprostane analysis. PLoS One. 2013;8:e80935. doi: 10.1371/journal. pone.0080935. 281. Petrosino T, Serafini M. Matrix effect in F2-isoprostanes quantification by HPLC-MS/MS: a validated method for analysis of iPF2α-III and iPF2α-VI in human urine. J Chromatogr B Analyt Technol Biomed Life Sci. 2014;965:100–106. doi: 10.1016/j.jchromb.2014.06.021. 282. Sterz K, Scherer G, Ecker J. A simple and robust UPLC-SRM/MS method to quantify urinary eicosanoids. J Lipid Res. 2012;53:1026–1036. doi: 10.1194/jlr.D023739. 283. Kadiiska MB, Gladen BC, Baird DD, Germolec D, Graham LB, Parker CE, Nyska A, Wachsman JT, Ames BN, Basu S, Brot N, Fitzgerald GA, Floyd RA, George M, Heinecke JW, Hatch GE, Hensley K, Lawson JA, Marnett LJ, Morrow JD, Murray DM, Plastaras J, Roberts LJ 2nd, Rokach J, Shigenaga MK, Sohal RS, Sun J, Tice RR, Van Thiel DH, Wellner D, Walter PB, Tomer KB, Mason RP, Barrett JC. Biomarkers of oxidative stress study II: are oxidation products of lipids, proteins, and DNA markers of CCl4 poisoning? Free Radic Biol Med. 2005;38:698– 710. doi: 10.1016/j.freeradbiomed.2004.09.017. 284. Il’yasova D, Spasojevic I, Wang F, Tolun AA, Base K, Young SP, Marcom PK, Marks J, Mixon G, DiGiulio R, Millington DS. Urinary biomarkers of oxidative status in a clinical model of oxidative assault. Cancer Epidemiol Biomarkers Prev. 2010;19:1506–1510. doi: 10.1158/10559965.EPI-10-0211. 285. Rudic RD, Brinster D, Cheng Y, Fries S, Song WL, Austin S, Coffman TM, FitzGerald GA. COX-2-derived prostacyclin modulates vascular remodeling. Circ Res. 2005;96:1240–1247. doi: 10.1161/01. RES.0000170888.11669.28. 286. Collins AR, Lyon CJ, Xia X, Liu JZ, Tangirala RK, Yin F, Boyadjian R, Bikineyeva A, Praticò D, Harrison DG, Hsueh WA. Age-accelerated atherosclerosis correlates with failure to upregulate antioxidant genes. Circ Res. 2009;104:e42–e54. doi: 10.1161/CIRCRESAHA.108.188771. 287. Schwedhelm E, Bartling A, Lenzen H, Tsikas D, Maas R, Brümmer J, Gutzki FM, Berger J, Frölich JC, Böger RH. Urinary 8-iso-prostaglandin F2α as a risk marker in patients with coronary heart disease: a matched case-control study. Circulation. 2004;109:843–848. doi: 10.1161/01. CIR.0000116761.93647.30. 288. Davì G, Ciabattoni G, Consoli A, Mezzetti A, Falco A, Santarone S, Pennese E, Vitacolonna E, Bucciarelli T, Costantini F, Capani F, Patrono C. In vivo formation of 8-iso-prostaglandin F2α and platelet activation in diabetes mellitus: effects of improved metabolic control and vitamin E supplementation. Circulation. 1999;99:224–229. 289. Zhang R, Sun ML, Fan YF, Jiang X, Zhao QH, He J, Wang L, Shailendra PK, Safdar Z, Jing ZC. Plasma 15-F2t-isoprostane in idiopathic pulmonary arterial hypertension. Int J Cardiol. 2014;175:268–273. doi: 10.1016/j.ijcard.2014.05.014. 290. Reilly MP, Delanty N, Roy L, Rokach J, Callaghan PO, Crean P, Lawson JA, FitzGerald GA. Increased formation of the isoprostanes IPF2α-I and 8-epi-prostaglandin F2α in acute coronary angioplasty: evidence for oxidant stress during coronary reperfusion in humans. Circulation. 1997;96:3314–3320. 291. Delanty N, Reilly MP, Pratico D, Lawson JA, McCarthy JF, Wood AE, Ohnishi ST, Fitzgerald DJ, FitzGerald GA. 8-epi PGF2α generation during coronary reperfusion: a potential quantitative marker of oxidant stress in vivo. Circulation. 1997;95:2492–2499. 292. Hill BG, Haberzettl P, Ahmed Y, Srivastava S, Bhatnagar A. Unsaturated lipid peroxidation-derived aldehydes activate autophagy in vascular smooth-muscle cells. Biochem J. 2008;410:525–534. doi: 10.1042/ BJ20071063. 293. Cadet J, Berger M, Douki T, Ravanat JL. Oxidative damage to DNA: formation, measurement, and biological significance. Rev Physiol Biochem Pharmacol. 1997;131:1–87. 294. Cooke MS, Evans MD, Dizdaroglu M, Lunec J. Oxidative DNA damage: mechanisms, mutation, and disease. FASEB J. 2003;17:1195–1214. doi: 10.1096/fj.02-0752rev. 295. Nunomura A, Honda K, Takeda A, Hirai K, Zhu X, Smith MA, Perry G. Oxidative damage to RNA in neurodegenerative diseases. J Biomed Biotechnol. 2006;2006:82323. doi: 10.1155/JBB/2006/82323. 296.Cadet J, Douki T, Gasparutto D, Ravanat JL. Oxidative damage to DNA: formation, measurement and biochemical features. Mutat Res. 2003;531:5–23. 297. Shuck SC, Rose KL, Marnett LJ. Mass spectrometric methods for the analysis of nucleoside-protein cross-links: application to oxopropenyldeoxyadenosine. Chem Res Toxicol. 2014;27:136–146. doi: 10.1021/ tx400384e. 298.Cadet J, Delatour T, Douki T, Gasparutto D, Pouget JP, Ravanat JL, Sauvaigo S. Hydroxyl radicals and DNA base damage. Mutat Res. 1999;424:9–21. 299.Preston BD, Albertson TM, Herr AJ. DNA replication fidel ity and cancer. Semin Cancer Biol. 2010;20:281–293. doi: 10.1016/j. semcancer.2010.10.009. 300. Krebs JE. Lewin’s Cells. Burlington, MA: Jones & Bartlett Publishers; 2013. 301. Mouret JF, Polverelli M, Sarrazini F, Cadet J. Ionic and radical oxidations of DNA by hydrogen peroxide. Chem Biol Interact. 1991;77:187–201. 302.Tripathi P, Moinuddin, Dixit K, Mir AR, Habib S, Alam K, Ali A. Peroxynitrite modified DNA presents better epitopes for anti-DNA autoantibodies in diabetes type 1 patients. Cell Immunol. 2014;290:30–38. doi: 10.1016/j.cellimm.2014.04.012. 303. Ravanat JL, Douki T, Cadet J. Direct and indirect effects of UV radiation on DNA and its components. J Photochem Photobiol B. 2001;63:88–102. 304. Collins AR, Cadet J, Möller L, Poulsen HE, Viña J. Are we sure we know how to measure 8-oxo-7,8-dihydroguanine in DNA from human cells? Arch Biochem Biophys. 2004;423:57–65. 305. Collins AR. The comet assay for DNA damage and repair: principles, applications, and limitations. Mol Biotechnol. 2004;26:249–261. doi: 10.1385/MB:26:3:249. 306. Nandhakumar S, Parasuraman S, Shanmugam MM, Rao KR, Chand P, Bhat BV. Evaluation of DNA damage using single-cell gel electrophoresis (comet assay). J Pharmacol Pharmacother. 2011;2:107–111. doi: 10.4103/0976-500X.81903. 307. Bhatnagar A. Contribution of ATP to oxidative stress-induced changes in action potential of isolated cardiac myocytes. Am J Physiol. 1997;272(pt 2):H1598–H1608. 308. Cadet J, Poulsen H. Measurement of oxidatively generated base damage in cellular DNA and urine. Free Radic Biol Med. 2010;48:1457–1459. doi: 10.1016/j.freeradbiomed.2010.03.004. 309. Cadet J, Douki T, Ravanat JL. Measurement of oxidatively generated base damage in cellular DNA. Mutat Res. 2011;711:3–12. doi: 10.1016/j. mrfmmm.2011.02.004. 310.Brand S, Amann K, Schupp N. Angiotensin II-induced hypertension dose-dependently leads to oxidative stress and DNA damage in mouse kidneys and hearts. J Hypertens. 2013;31:333–344. doi: 10.1097/ HJH.0b013e32835ba77e. 311.Daehn I, Casalena G, Zhang T, Shi S, Fenninger F, Barasch N, Yu L, D’Agati V, Schlondorff D, Kriz W, Haraldsson B, Bottinger EP. Endothelial mitochondrial oxidative stress determines podocyte depletion in segmental glomerulosclerosis. J Clin Invest. 2014;124:1608– 1621. doi: 10.1172/JCI71195. 312.Pohjoismäki JL, Boettger T, Liu Z, Goffart S, Szibor M, Braun T. Oxidative stress during mitochondrial biogenesis compromises mtDNA integrity in growing hearts and induces a global DNA repair response. Nucleic Acids Res. 2012;40:6595–6607. doi: 10.1093/nar/gks301. 313. Curtis CD, Thorngren DL, Nardulli AM. Immunohistochemical analysis of oxidative stress and DNA repair proteins in normal mammary and breast cancer tissues. BMC Cancer. 2010;10:9. doi: 10.1186/1471-2407-10-9. 314.Martinet W,Knaapen MW,De Meyer GR,Herman AG,Kockx MM. Oxidative DNA damage and repair in experimental atherosclerosis are reversed by dietary lipid lowering. Circ Res. 2001;88:733–739. 315. Mayr M, Hy Y, Hainaut H, Xu Q. Mechanical stress-induced DNA damage and rac-p38MAPK signal pathways mediate p53-dependent apoptosis in vascular smooth muscle cells. FASEB J. 2002;16:1423–1425. 316. Yin B, Whyatt RM, Perera FP, Randall MC, Cooper TB, Santella RM. Determination of 8-hydroxydeoxyguanosine by an immunoaffinity chromatography-monoclonal antibody-based ELISA. Free Radic Biol Med. 1995;18:1023–1032. 317.Cadet J, Douki T, Frelon S, Sauvaigo S, Pouget JP, Ravanat JL. Assessment of oxidative base damage to isolated and cellular DNA by HPLC-MS/MS measurement. Free Radic Biol Med. 2002;33:441–449. 318. Druzhyna NM, Musiyenko SI, Wilson GL, LeDoux SP. Cytokines induce nitric oxide-mediated mtDNA damage and apoptosis in oligodendrocytes: protective role of targeting 8-oxoguanine glycosylase to mitochondria. J Biol Chem. 2005;280:21673–21679. doi: 10.1074/jbc. M411531200. 319. Rachek LI, Grishko VI, Alexeyev MF, Pastukh VV, LeDoux SP, Wilson GL. Endonuclease III and endonuclease VIII conditionally targeted into e74 Circulation Research August 19, 2016 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 mitochondria enhance mitochondrial DNA repair and cell survival following oxidative stress. Nucleic Acids Res. 2004;32:3240–3247. doi: 10.1093/nar/gkh648. 320.Ruchko M, Gorodnya O, LeDoux SP, Alexeyev MF, Al-Mehdi AB, Gillespie MN. Mitochondrial DNA damage triggers mitochondrial dysfunction and apoptosis in oxidant-challenged lung endothelial cells. Am J Physiol Lung Cell Mol Physiol. 2005;288:L530–L535. doi: 10.1152/ ajplung.00255.2004. 321. Lu T, Pan Y, Kao SY, Li C, Kohane I, Chan J, Yankner BA. Gene regulation and DNA damage in the ageing human brain. Nature. 2004;429:883– 891. doi: 10.1038/nature02661. 322. Bohr VA, Smith CA, Okumoto DS, Hanawalt PC. DNA repair in an active gene: removal of pyrimidine dimers from the DHFR gene of CHO cells is much more efficient than in the genome overall. Cell. 1985;40:359–369. 323.Pastukh V, Ruchko M, Gorodnya O, Wilson GL, Gillespie MN. Sequence-specific oxidative base modifications in hypoxia-inducible genes. Free Radic Biol Med. 2007;43:1616–1626. doi: 10.1016/j. freeradbiomed.2007.08.027. 324. Rothfuss O, Gasser T, Patenge N. Analysis of differential DNA damage in the mitochondrial genome employing a semi-long run real-time PCR approach. Nucleic Acids Res. 2010;38:e24. doi: 10.1093/nar/gkp1082. 325. Wosniak J Jr, Santos CX, Kowaltowski AJ, Laurindo FR. Cross-talk between mitochondria and NADPH oxidase: effects of mild mitochondrial dysfunction on angiotensin II-mediated increase in Nox isoform expression and activity in vascular smooth muscle cells. Antioxid Redox Signal. 2009;11:1265–1278. doi: 10.1089/ars.2009.2392. 326.Collins A, Cadet J, Epe B, Gedik C. Problems in the measurement of 8-oxoguanine in human DNA: report of a workshop, DNA oxidation, held in Aberdeen, UK, 19-21 January, 1997. Carcinogenesis. 1997;18:1833–1836. 327.Giustarini D, Dalle-Donne I, Tsikas D, Rossi R. Oxidative stress and human diseases: origin, link, measurement, mechanisms, and biomarkers. Crit Rev Clin Lab Sci. 2009;46:241–281. doi: 10.3109/ 10408360903142326. 328. Khan BV, Navalkar S, Khan QA, Rahman ST, Parthasarathy S. Irbesartan, an angiotensin type 1 receptor inhibitor, regulates the vascular oxidative state in patients with coronary artery disease. J Am Coll Cardiol. 2001;38:1662–1667. 329.Azarsiz E, Kayikcioglu M, Payzin S, Yildirim Sözmen E. PON1 activities and oxidative markers of LDL in patients with angiographically proven coronary artery disease. Int J Cardiol. 2003;91:43–51. 330. Stringer MD, Görög PG, Freeman A, Kakkar VV. Lipid peroxides and atherosclerosis. BMJ. 1989;298:281–284. 331. Keith M, Geranmayegan A, Sole MJ, Kurian R, Robinson A, Omran AS, Jeejeebhoy KN. Increased oxidative stress in patients with congestive heart failure. J Am Coll Cardiol. 1998;31:1352–1356. 332.Marra G, Cotroneo P, Pitocco D, Manto A, Di Leo MA, Ruotolo V, Caputo S, Giardina B, Ghirlanda G, Santini SA. Early increase of oxidative stress and reduced antioxidant defenses in patients with uncomplicated type 1 diabetes: a case for gender difference. Diabetes Care. 2002;25:370–375. 333. Buffon A, Santini SA, Ramazzotti V, Rigattieri S, Liuzzo G, Biasucci LM, Crea F, Giardina B, Maseri A. Large, sustained cardiac lipid peroxidation and reduced antioxidant capacity in the coronary circulation after brief episodes of myocardial ischemia. J Am Coll Cardiol. 2000;35:633–639. 334.Ozmen D, Boydak B, Mutaf I, Zoghi M, Kumanlioğlu K, Güner I, Bayindir O. The state of lipid peroxidation and antioxidants following thrombolytic therapy with rt-PA and streptokinase in acute myocardial infarction. Jpn Heart J. 1999;40:267–273. 335.Singh RB, Wander GS, Rastogi A, Shukla PK, Mittal A, Sharma JP, Mehrotra SK, Kapoor R, Chopra RK. Randomized, double-blind placebo-controlled trial of coenzyme Q10 in patients with acute myocardial infarction. Cardiovasc Drugs Ther. 1998;12:347–353. 336. Walter MF, Jacob RF, Bjork RE, Jeffers B, Buch J, Mizuno Y, Mason RP; PREVENT Investigators. Circulating lipid hydroperoxides predict cardiovascular events in patients with stable coronary artery disease: the PREVENT study. J Am Coll Cardiol. 2008;51:1196–1202. doi: 10.1016/j.jacc.2007.11.051. 337. White M, Ducharme A, Ibrahim R, Whittom L, Lavoie J, Guertin MC, Racine N, He Y, Yao G, Rouleau JL, Schiffrin EL, Touyz RM. Increased systemic inflammation and oxidative stress in patients with worsening congestive heart failure: improvement after short-term inotropic support. Clin Sci (Lond). 2006;110:483–489. doi: 10.1042/CS20050317. 338. White M, Cantin B, Haddad H, Kobashigawa JA, Ross H, Carrier M, Pflugfelder PW, Isaac D, Cecere R, Whittom L, Ali IS, Wang SH, He Y, Groulx A, Touyz RM. Cardiac signaling molecules and plasma biomarkers after cardiac transplantation: impact of tacrolimus versus cyclosporine. J Heart Lung Transplant. 2013;32:1222–1232. doi: 10.1016/j. healun.2013.09.010. 339. Kurlak LO, Green A, Loughna P, Broughton Pipkin F. Oxidative stress markers in hypertensive states of pregnancy: preterm and term disease. Front Physiol. 2014;5:310. doi: 10.3389/fphys.2014.00310. 340. da Cruz AC, Petronilho F, Heluany CC, Vuolo F, Miguel SP, Quevedo J, Romano-Silva MA, Dal-Pizzol F. Oxidative stress and aging: correlation with clinical parameters. Aging Clin Exp Res. 2014;26:7–12. doi: 10.1007/s40520-013-0176-9. 341. Lee WC, Wong HY, Chai YY, Shi CW, Amino N, Kikuchi S, Huang SH. Lipid peroxidation dysregulation in ischemic stroke: plasma 4-HNE as a potential biomarker? Biochem Biophys Res Commun. 2012;425:842– 847. doi: 10.1016/j.bbrc.2012.08.002. 342. Tanaka S, Miki T, Sha S, Hirata K, Ishikawa Y, Yokoyama M. Serum levels of thiobarbituric acid-reactive substances are associated with risk of coronary heart disease. J Atheroscler Thromb. 2011;18:584–591. 343. Salonen JT, Nyyssönen K, Salonen R, Porkkala-Sarataho E, Tuomainen TP, Diczfalusy U, Björkhem I. Lipoprotein oxidation and progression of carotid atherosclerosis. Circulation. 1997;95:840–845. 344. Davies SS, Roberts LJ 2nd. F2-isoprostanes as an indicator and risk factor for coronary heart disease. Free Radic Biol Med. 2011;50:559–566. doi: 10.1016/j.freeradbiomed.2010.11.023. 345.Basu S. Bioactive eicosanoids: role of prostaglandin F(2α) and F2isoprostanes in inflammation and oxidative stress related pathology. Mol Cells. 2010;30:383–391. doi: 10.1007/s10059-010-0157-1. 346. Morrow JD. Quantification of isoprostanes as indices of oxidant stress and the risk of atherosclerosis in humans. Arterioscler Thromb Vasc Biol. 2005;25:279–286. doi: 10.1161/01.ATV.0000152605.64964.c0. 347. Pignatelli P, Pastori D, Carnevale R, Farcomeni A, Cangemi R, Nocella C, Bartimoccia S, Vicario T, Saliola M, Lip GY, Violi F. Serum NOX2 and urinary isoprostanes predict vascular events in patients with atrial fibrillation. Thromb Haemost. 2015;113:617–624. doi: 10.1160/TH14-07-0571. 348. Mali VR, Ning R, Chen J, Yang XP, Xu J, Palaniyandi SS. Impairment of aldehyde dehydrogenase-2 by 4-hydroxy-2-nonenal adduct formation and cardiomyocyte hypertrophy in mice fed a high-fat diet and injected with low-dose streptozotocin. Exp Biol Med (Maywood). 2014;239:610– 618. doi: 10.1177/1535370213520109. 349. Zhang Y, Sano M, Shinmura K, Tamaki K, Katsumata Y, Matsuhashi T, Morizane S, Ito H, Hishiki T, Endo J, Zhou H, Yuasa S, Kaneda R, Suematsu M, Fukuda K. 4-Hydroxy-2-nonenal protects against cardiac ischemia-reperfusion injury via the Nrf2-dependent pathway. J Mol Cell Cardiol. 2010;49:576–586. doi: 10.1016/j.yjmcc.2010.05.011. 350. Usberti M, Gerardi GM, Gazzotti RM, Benedini S, Archetti S, Sugherini L, Valentini M, Tira P, Bufano G, Albertini A, Di Lorenzo D. Oxidative stress and cardiovascular disease in dialyzed patients. Nephron. 2002;91:25–33. doi: 57601. 351. Gerardi G, Usberti M, Martini G, Albertini A, Sugherini L, Pompella A, Di LD. Plasma total antioxidant capacity in hemodialyzed patients and its relationships to other biomarkers of oxidative stress and lipid peroxidation. Clin Chem Lab Med. 2002;40:104–110. doi: 10.1515/CCLM.2002.019. 352. Fraga CG, Oteiza PI, Galleano M. In vitro measurements and interpretation of total antioxidant capacity. Biochim Biophys Acta. 2014;1840:931– 934. doi: 10.1016/j.bbagen.2013.06.030. 353. Pinchuk I, Shoval H, Dotan Y, Lichtenberg D. Evaluation of antioxidants: scope, limitations and relevance of assays. Chem Phys Lipids. 2012;165:638–647. doi: 10.1016/j.chemphyslip.2012.05.003. 354. Lotito SB, Frei B. Consumption of flavonoid-rich foods and increased plasma antioxidant capacity in humans: cause, consequence, or epiphenomenon? Free Radic Biol Med. 2006;41:1727–1746. doi: 10.1016/j. freeradbiomed.2006.04.033. 355. Hollman PC, Cassidy A, Comte B, Heinonen M, Richelle M, Richling E, Serafini M, Scalbert A, Sies H, Vidry S. The biological relevance of direct antioxidant effects of polyphenols for cardiovascular health in humans is not established. J Nutr. 2011;141:989S–1009S. doi: 10.3945/jn.110.131490. 356. Wang Y, Chun OK, Song WO. Plasma and dietary antioxidant status as cardiovascular disease risk factors: a review of human studies. Nutrients. 2013;5:2969–3004. doi: 10.3390/nu5082969. 357. Gedikli O, Ozturk S, Yilmaz H, Baykan M, Kiris A, Durmus I, Karaman K, Karahan C, Celik S. Low total antioxidative capacity levels are associated with augmentation index but not pulse-wave velocity. Heart Vessels. 2009;24:366–370. doi: 10.1007/s00380-008-1129-7. Griendling et al Measurement of Reactive Oxygen Species e75 Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 358.Bartosz G. Non-enzymatic antioxidant capacity assays: limita tions of use in biomedicine. Free Radic Res. 2010;44:711–720. doi: 10.3109/10715761003758114. 359.Kietadisorn R, Juni RP, Moens AL. Tackling endothelial dysfunction by modulating NOS uncoupling: new insights into its pathogenesis and therapeutic possibilities [published correction appears in Am J Physiol Endocrinol Metab. 2012;303:E1505]. Am J Physiol Endocrinol Metab. 2012;302:E481–E495. doi: 10.1152/ajpendo.00540.2011. 360.Zhang YH, Casadei B. Sub-cellular targeting of constitutive NOS in health and disease. J Mol Cell Cardiol. 2012;52:341–350. doi: 10.1016/j. yjmcc.2011.09.006. 361. Nagababu E, Rifkind JM. Measurement of plasma nitrite by chemiluminescence. Methods Mol Biol. 2010;610:41–49. doi: 10.1007/9781-60327-029-8_3. 362.Beda N, Nedospasov A. A spectrophotometric assay for nitrate in an excess of nitrite. Nitric Oxide. 2005;13:93–97. doi: 10.1016/j. niox.2005.05.002. 363. Berkels R, Purol-Schnabel S, Roesen R. Measurement of nitric oxide by reconversion of nitrate/nitrite to NO. Methods Mol Biol. 2004;279:1–8. doi: 10.1385/1-59259-807-2:001. 364. Ghasemi A, Zahediasl S, Azizi F. Reference values for serum nitric oxide metabolites in an adult population. Clin Biochem. 2010;43:89–94. doi: 10.1016/j.clinbiochem.2009.09.011. 365.Giustarini D, Rossi R, Milzani A, Dalle-Donne I. Nitrite and nitrate measurement by Griess reagent in human plasma: evaluation of interferences and standardization. Methods Enzymol. 2008;440:361–380. doi: 10.1016/S0076-6879(07)00823-3. 366. Bouras G, Deftereos S, Tousoulis D, Giannopoulos G, Chatzis G, Tsounis D, Cleman MW, Stefanadis C. Asymmetric dimethylarginine (ADMA): a promising biomarker for cardiovascular disease? Curr Top Med Chem. 2013;13:180–200. 367. Schwedhelm E, Xanthakis V, Maas R, Sullivan LM, Schulze F, Riederer U, Benndorf RA, Böger RH, Vasan RS. Asymmetric dimethylarginine reference intervals determined with liquid chromatography-tandem mass spectrometry: results from the Framingham offspring cohort. Clin Chem. 2009;55:1539–1545. doi: 10.1373/clinchem.2009.124263. 368. Juonala M, Viikari JS, Alfthan G, Marniemi J, Kähönen M, Taittonen L, Laitinen T, Raitakari OT. Brachial artery flow-mediated dilation and asymmetrical dimethylarginine in the Cardiovascular Risk in Young Finns study. Circulation. 2007;116:1367–1373. doi: 10.1161/ CIRCULATIONAHA.107.690016. 369.Päivä H, Kähönen M, Lehtimäki T, Alfthan G, Viikari J, Laaksonen R, Hutri-Kähönen N, Laitinen T, Taittonen L, Raitakari OT, Juonala M. Levels of asymmetrical dimethylarginine are predictive of brachial artery flow-mediated dilation 6 years later: the Cardiovascular Risk in Young Finns Study. Atherosclerosis. 2010;212:512–515. doi: 10.1016/j. atherosclerosis.2010.06.041. 370.Wolin MS, Gupte SA, Neo BH, Gao Q, Ahmad M. Oxidant-redox regulation of pulmonary vascular responses to hypoxia and nitric oxide-cGMP signaling. Cardiol Rev. 2010;18:89–93. doi: 10.1097/ CRD.0b013e3181c9f088. 371. O’Donnell VB, Chumley PH, Hogg N, Bloodsworth A, Darley-Usmar VM, Freeman BA. Nitric oxide inhibition of lipid peroxidation: kinetics of reaction with lipid peroxyl radicals and comparison with alpha-tocopherol. Biochemistry. 1997;36:15216–15223. doi: 10.1021/bi971891z. 372. Roberts LJ, Morrow JD. Measurement of F(2)-isoprostanes as an index of oxidative stress in vivo. Free Radic Biol Med. 2000;28:505–513. 373.Heitzer T, Schlinzig T, Krohn K, Meinertz T, Münzel T. Endothelial dysfunction, oxidative stress, and risk of cardiovascular events in patients with coronary artery disease [published correction appears in Circulation. 2003;108:500]. Circulation. 2001;104:2673–2678. 374. Annuk M, Zilmer M, Fellstrom B. Endothelium-dependent vasodilation and oxidative stress in chronic renal failure: impact on cardiovascular disease. Kidney Int Suppl. 2003:S50–3. 375. Dikalova AE, Bikineyeva AT, Budzyn K, Nazarewicz RR, McCann L, Lewis W, Harrison DG, Dikalov SI. Therapeutic targeting of mitochondrial superoxide in hypertension. Circ Res. 2010;107:106–116. doi: 10.1161/CIRCRESAHA.109.214601. Downloaded from http://circres.ahajournals.org/ by guest on June 16, 2017 Measurement of Reactive Oxygen Species, Reactive Nitrogen Species, and Redox-Dependent Signaling in the Cardiovascular System: A Scientific Statement From the American Heart Association Kathy K. Griendling, Rhian M. Touyz, Jay L. Zweier, Sergey Dikalov, William Chilian, Yeong-Renn Chen, David G. Harrison and Aruni Bhatnagar on behalf of the American Heart Association Council on Basic Cardiovascular Sciences Circ Res. 2016;119:e39-e75; originally published online July 14, 2016; doi: 10.1161/RES.0000000000000110 Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231 Copyright © 2016 American Heart Association, Inc. All rights reserved. Print ISSN: 0009-7330. Online ISSN: 1524-4571 The online version of this article, along with updated information and services, is located on the World Wide Web at: http://circres.ahajournals.org/content/119/5/e39 Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for which permission is being requested is located, click Request Permissions in the middle column of the Web page under Services. Further information about this process is available in the Permissions and Rights Question and Answer document. Reprints: Information about reprints can be found online at: http://www.lww.com/reprints Subscriptions: Information about subscribing to Circulation Research is online at: http://circres.ahajournals.org//subscriptions/
© Copyright 2025 Paperzz