1,168
Views
127
CrossRef citations to date
0
Altmetric
Review Article

Reactions and reactivity of myeloperoxidase-derived oxidants: Differential biological effects of hypochlorous and hypothiocyanous acids

, &
Pages 975-995 | Received 31 Dec 2011, Accepted 13 Feb 2012, Published online: 23 Apr 2012

References

  • Kettle AJ, Winterbourn CC. Myeloperoxidase: a key regulator of neutrophil oxidant production. Redox Rep 1997;3: 3–15.
  • van Dalen CJ, Whitehouse MW, Winterbourn CC, Kettle AJ. Thiocyanate and chloride as competing substrates for myeloperoxidase. Biochem J 1997;327:487–492.
  • Davies MJ, Hawkins CL, Pattison DI, Rees MD. Mammalian heme peroxidases: from molecular mechanisms to health implications. Antioxid Redox Signal 2008;10: 1199–1234.
  • Klebanoff SJ. Myeloperoxidase: friend and foe. J Leukoc Biol 2005;77:598–625.
  • Pattison DI, Davies MJ. Reactions of myeloperoxidase-derived oxidants with biological substrates: gaining chemical insight into human inflammatory diseases. Curr Med Chem 2006;13:3271–3290.
  • Winterbourn CC. Comparative reactivities of various biological compounds with myeloperoxidase-hydrogen peroxide-chloride, and similarity of the oxidant to hypochlorite. Biochim Biophys Acta 1985;840:204–210.
  • Winterbourn CC. Biological reactivity and biomarkers of the neutrophil oxidant, hypochlorous acid. Toxicology 2002;181–182:223–227.
  • Pullar JM, Winterbourn CC, Vissers MCM. Loss of GSH and thiol enzymes in endothelial cells exposed to sublethal concentrations of hypochlorous acid. Am J Physiol 1999;277:H1505–H1512.
  • Pullar JM, Vissers MC, Winterbourn CC. Living with a killer: the effects of hypochlorous acid on mammalian cells. IUBMB Life 2000;50:259–266.
  • Schraufstatter IU, Browne K, Harris A, Hyslop PA, Jackson JH, Quehenberger O, . Mechanisms of hypochlorite injury of target cells. J Clin Invest 1990;85:554–562.
  • Lloyd MM, Van Reyk DM, Davies MJ, Hawkins CL. HOSCN is a more potent inducer of apoptosis and protein thiol depletion in murine macrophage cells than HOCl or HOBr. Biochem J 2008;414:271–280.
  • Whiteman M, Rose P, Siau JL, Cheung NS, Tan GS, Halliwell B, . Hypochlorous acid-mediated mitochondrial dysfunction and apoptosis in human hepatoma HepG2 and fetal liver cells: role of mitochondrial permeability transition. Free Radic Biol Med 2005;38:1571–1584.
  • Lane AE, Tan JT, Hawkins CL, Heather AK, Davies MJ. The myeloperoxidase-derived oxidant hoscn inhibits protein tyrosine phosphatases and modulates cell signalling via the mitogen-activated protein kinase (MAPK) pathway in macrophages. Biochem J 2010;430:161–169.
  • Whiteman M, Chu SH, Siau JL, Rose P, Sabapathy K, Schantz JT, . The pro-inflammatory oxidant hypochlorous acid induces bax-dependent mitochondrial permeabilisation and cell death through AIF-/EndoG-dependent pathways. Cell Signal 2007;19:705–714.
  • Whiteman M, Spencer JPE, Szeto HH, Armstrong JS. Do mitochondriotropic antioxidants prevent chlorinative stress-induced mitochondrial and cellular injury? Antioxid Redox Signal 2008;10:641–650.
  • Vissers MCM, Pullar JM, Hampton MB. Hypochlorous acid causes caspase activation and apoptosis or growth arrest in human endothelial cells. Biochem J 1999;344:443–339.
  • Vissers MC, Lee WG, Hampton MB. Regulation of apoptosis by vitamin C. Specific protection of the apoptotic machinery against exposure to chlorinated oxidants. J Biol Chem 2001;276:46835–46840.
  • Wagner BA, Britigan BE, Reszka KJ, McCormick ML, Burns CP. Hydrogen peroxide-induced apoptosis of HL-60 human leukemia cells is mediated by the oxidants hypochlorous acid and chloramines. Arch Biochem Biophys 2002;401:223–234.
  • Tenovuo J, Anttila O, Lumikari M, Sievers G. Antibacterial effect of myeloperoxidase against Streptococcus mutans. Oral Microbiol Immun 1988;3:68–71.
  • Hanstrom L, Johansson A, Carlsson J. Lactoperoxidase and thiocyanate protect cultured mammalian cells against hydrogen peroxide toxicity. Med Biol 1983;61:268–274.
  • Tenovuo J, Larjava H. The protective effect of peroxidase and thiocyanate against hydrogen peroxide toxicity assessed by the uptake of [3H]-thymidine by human gingival fibroblasts cultured in vitro. Arch Oral Biol 1984;29:445–451.
  • Xu Y, Szep S, Lu Z. The antioxidant role of thiocyanate in the pathogenesis of cystic fibrosis and other inflammation-related diseases. Proc Natl Acad Sci USA 2009;106: 20515–20519.
  • Gould NS, Gauthier S, Kariya CT, Min E, Huang J, Brian DJ. Hypertonic saline increases lung epithelial lining fluid glutathione and thiocyanate: two protective CFTR-dependent thiols against oxidative injury. Respir Res 2010;11:119–128.
  • Grisham MB, Ryan EM. Cytotoxic properties of salivary oxidants. Am J Physiol 1990;258:C115–C121.
  • Arlandson M, Decker T, Roongta VA, Bonilla L, Mayo KH, MacPherson JC, . Eosinophil peroxidase oxidation of thiocyanate – characterization of major reaction products and a potential sulfhydryl-targeted cytotoxicity system. J Biol Chem 2001;276:215–224.
  • Wang JG, Mahmud SA, Nguyen J, Slungaard A. Thiocyanate-dependent induction of endothelial cell adhesion molecule expression by phagocyte peroxidases: a novel HOSCN-specific oxidant mechanism to amplify inflammation. J Immunol 2006;177:8714–8722.
  • Wang JG, Mahmud SA, Thompson JA, Geng JG, Key NS, Slungaard A. The principal eosinophil peroxidase product, HOSCN, is a uniquely potent phagocyte oxidant inducer of endothelial cell tissue factor activity: a potential mechanism for thrombosis in eosinophilic inflammatory states. Blood 2006;107:558–565.
  • Bozonet SM, Scott-Thomas AP, Nagy P, Vissers MC. Hypothiocyanous acid is a potent inhibitor of apoptosis and caspase 3 activation in endothelial cells. Free Radic Biol Med 2010;49:1054–1063.
  • Wang Z, Nicholls SJ, Rodriguez ER, Kummu O, Horkko S, Barnard J, . Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nat Med 2007;13: 1176–1184.
  • Morgan PE, Pattison DI, Talib J, Summers FA, Harmer JA, Celermajer DS, . High plasma thiocyanate levels in smokers are a key determinant of thiol oxidation induced by myeloperoxidase. Free Radic Biol Med 2011;51:1815–1822.
  • Winterbourn CC, Vissers MC, Kettle AJ. Myeloperoxidase. Curr Opin Hematol 2000;7:53–58.
  • Babior BM. The respiratory burst oxidase. TIBS 1987; 12:241–243.
  • Bielski BHJ, Cabelli DE, Arudi RL, Ross AB. Reactivity of HO2/O2− radicals in aqueous solution. J Phys Chem Ref Data 1985;14:1041–1100.
  • Furtmuller PG, Zederbauer M, Jantschko W, Helm J, Bogner M, Jakopitsch C, . Active site structure and catalytic mechanisms of human peroxidases. Arch Biochem Biophys 2006;445:199–213.
  • Furtmuller PG, Burner U, Obinger C. Reaction of myeloperoxidase compound i with chloride, bromide, iodide, and thiocyanate. Biochemistry 1998;37:17923–17930.
  • Davies MJ. Myeloperoxidase-derived oxidation: mechanisms of biological damage and its prevention. J Clin Biochem Nutr 2011;48:8–19.
  • Lentner C. Geigy scientific tables: physical chemistry, composition of blood, hematology, somatometric data. Basle: Ciba-Geigy Ltd; 1984.
  • Ashby MT, Carlson AC, Scott MJ. Redox buffering of hypochlorous acid by thiocyanate in physiologic fluids. J Am Chem Soc 2004;126:15976–15977.
  • Nagy P, Beal JL, Ashby MT. Thiocyanate is an efficient endogenous scavenger of the phagocytic killing agent hypobromous acid. Chem Res Toxicol 2006;19:587–593.
  • Xulu BA, Ashby MT. Small molecular, macromolecular, and cellular chloramines react with thiocyanate to give the human defense factor hypothiocyanite. Biochemistry 2010; 49:2068–2074.
  • Winterbourn CC, Hampton MB, Livesey JH, Kettle AJ. Modeling the reactions of superoxide and myeloperoxidase in the neutrophil phagosome: implications for microbial killing. J Biol Chem 2006;281:39860–39869.
  • Chapman ALP, Hampton MB, Senthilmohan R, Winterbourn CC, Kettle AJ. Chlorination of bacterial and neutrophil cell proteins during phagocytosis and killing of Staphylococcus aureus. J Biol Chem 2002;277:9757–9762.
  • Rosen H, Crowley JR, Heinecke JW. Human neutrophils use the myeloperoxidase-hydrogen peroxide-chloride system to chlorinate but not nitrate bacterial proteins during phagocytosis. J Biol Chem 2002;34:30463–30468.
  • Segal AW. How neutrophils kill microbes. Ann Rev Immunol 2005;23:197–223.
  • Klebanoff SJ. Oxygen metabolism and the toxic properties of phagocytes. Ann Int Med 1980;93:480–489.
  • Hawkins CL. The role of hypothiocyanous acid (HOSCN) in biological systems. Free Radic Res 2009;43:1147–1158.
  • Hansson M, Olsson I, Nauseef WM. Biosynthesis, processing, and sorting of human myeloperoxidase. Arch Biochem Biophys 2006;445:214–224.
  • van der Veen BS, Winther MPJ, Herringa P. Myeloperoxidase: molecular mechanisms of action and their relevance to human health and disease. Antioxid Redox Signal 2009;11: 2899–2937.
  • Hazen SL, Heinecke JW. 3-chlorotyrosine, a specific marker of myeloperoxidase-catalysed oxidation, is markedly elevated in low density lipoprotein isolated from human atherosclerotic intima. J Clin Invest 1997;99:2075–2081.
  • Aldridge R, Chan T, van Dalen C, Senthilmohan R, Winn M, Venge P, . Eosinophil peroxidase produces hypobromous acid in the airways of stable asthmatics. Free Radic Biol Med 2002;33:847–856.
  • Buss H, Senthilmohan R, Darlow BA, Mogridge N, Kettle AJ, Winterbourn CC. 3-chlorotyrosine as a marker of protein damage by myeloperoxidase in traceal aspirates from preterm infants: association with adverse respiratory outcome. Pediatr Res 2003;53:455–462.
  • Kettle AJ, Chan T, Osberg I, Senthilmohan R, Chapman AL, Mocatta TJ, . Myeloperoxidase and protein oxidation in the airways of young children with cystic fibrosis. Am J Respir Crit Care Med 2004;170:1317–1323.
  • Hazen SL, Gaut JP, Crowley JR, Hsu FF, Heinecke JW. Elevated levels of protein-bound p-hydroxyphenylacetaldehyde, an amino-acid-derived aldehyde generated by myeloperoxidase, are present in human fatty streaks, intermediate lesions and advanced atherosclerotic lesions. Biochem J 2000;352:693–699.
  • Carr AC, Vissers MC, Domigan NM, Winterbourn CC. Modification of red cell membrane lipids by hypochlorous acid and haemolysis by preformed lipid chlorohydrins. Redox Rep 1997;3:263–271.
  • Thukkani AK, Martinson BD, Albert CJ, Vogler GA, Ford DA. Neutrophil-mediated accumulation of 2-ClHDA during myocardial infarction: 2-ClHDA-mediated myocardial injury. Am J Physiol Heart Circ Physiol 2005;288: H2955–H2964.
  • Thukkani AK, McHowat J, Hsu FF, Brennan ML, Hazen SL, Ford DA. Identification of alpha-chloro fatty aldehydes and unsaturated lysophosphatidylcholine molecular species in human atherosclerotic lesions. Circulation 2003;108: 3128–3133.
  • Asahi T, Kondo H, Masuda M, Nishino H, Aratani Y, Naito Y, . Chemical and immunochemical detection of 8-halogenated deoxyguanosines at early stage inflammation. J Biol Chem 2010;285:9282–9291.
  • Ohnishi S, Murata M, Kawanishi S. DNA damage induced by hypochlorite and hypobromite with reference to inflammation-associated carcinogenesis. Cancer Lett 2002;178:37–42.
  • Henderson JP, Byun J, Heinecke JW. Molecular chlorine generated by the myeloperoxidase-hydrogen peroxide-chloride system of phagocytes produces 5-chlorocytosine in bacterial RNA. J Biol Chem 1999;274:33440–33448.
  • Henderson JP, Byun J, Williams MV, Mueller DM, McCormick ML, Heinecke JW. Production of brominating intermediates by myeloperoxidase. A transhalogenation pathway for generating mutagenic nucleobases during inflammation. J Biol Chem 2001;276:7867–7875.
  • Kawai Y, Morinaga H, Kondo H, Miyoshi N, Nakamura Y, Uchida K, . Endogenous formation of novel halogenated 2′-deoxycytidine. Hypohalous acid-mediated DNA modification at the site of inflammation. J Biol Chem 2004;279:51241–51249.
  • Brennan ML, Anderson MM, Shih DM, Qu XD, Wang X, Mehta AC, . Increased atherosclerosis in myeloperoxidase-deficient mice. J Clin Invest 2001;107:419–430.
  • McMillen TS, Heinecke JW, LeBoeuf RC. Expression of human myeloperoxidase by macrophages promotes atherosclerosis in mice. Circulation 2005;111:2798–2804.
  • Stark GR. Reactions of cyanate with functional groups of proteins. III. Reactions with amino and carboxyl groups. Biochemistry 1965;4:1030–1036.
  • Stark GR. Modification of proteins with cyanate. Meth Enzymol 1998;25:579–584.
  • Koelsch M, Mallak R, Graham GG, Kajer T, Milligan MK, Nguyen LQ, . Acetaminophen (paracetamol) inhibits myeloperoxidase-catalyzed oxidant production and biological damage at therapeutically achievable concentrations. Biochem Pharmacol 2010;79:1156–1164.
  • Burner U, Obinger C, Paumann M, Furtmuller PG, Kettle AJ. Transient and steady-state kinetics of the oxidation of substituted benzoic acid hydrazides by myeloperoxidase. J Biol Chem 1999;274:9494–9502.
  • Kettle AJ, Candaeis LP. Oxidation of tryptophan by redox intermediates of myeloperoxidase and inhibition of hypochlorous acid production. Redox Rep 2000;5: 179–184.
  • Kettle AJ, Gedye CA, Hampton MB, Winterbourn CC. Inhibition of myeloperoxidase by benzoic acid hydrazides. Biochem J 1995;308:559–563.
  • Kettle AJ, Geyde CA, Winterbourn CC. Mechanism of inactivation of myeloperoxidase by 4-aminobenzoic hydrazide. Biochem J 1997;321:503–508.
  • Kettle AJ, Robertson IGC, Palmer BD, Anderson RF, Patel KB, Winterbourn CC. Oxidative metabolism of amsacrine by the neutrophil enzyme myeloperoxidase. Biochem Pharmacol 1992;44:1731–1738.
  • Kettle AJ, Winterbourn CC. Oxidation of hydroquinone by myeloperoxidase – mechanism of stimulation by benzoquinone. J Biol Chem 1992;267:8319–8324.
  • Ximenes VF, Silva SO, Rodrigues MR, Catalani LH, Maghzal GJ, Kettle AJ, . Superoxide-dependent oxidation of melatonin by myeloperoxidase. J Biol Chem 2005; 280:38160–38169.
  • Tiden AK, Sjogren T, Svensson M, Bernlind A, Senthilmohan R, Auchere F, . 2-thioxanthines are mechanism-based inactivators of myeloperoxidase that block oxidative stress during inflammation. J Biol Chem 2011;286:37578–37589.
  • Peto RL. Future worldwide health effects of current smoking patterns. In: Koop CE, Pearson CE, Schwartz MR (eds). Critical issues in global health. San Francisco: Jossey-Bass; 2001.
  • O'Donnell CJ, Kannel WB. Epidemiology of atherosclerotic vascular disease. In: Lanzer PT, Topel EJ (eds). Panvascular medicine. Berlin: Springer-Verlag; 2003.
  • Yamagishi K, Iso H, Kitamura A, Sankai T, Tanigawa T, Naito Y, . Smoking raises the risk of total and ischemic strokes in hypertensive men. Hypertens Res 2003;26: 209–217.
  • Chelland Campbell S, Moffatt RJ, Stamford BA. Smoking and smoking cessation - the relationship between cardiovascular disease and lipoprotein metabolism: a review. Atherosclerosis 2008;201:225–235.
  • Wilson J. Cyanide in human disease: a review of clinical and laboratory evidence. Fundam. Appl Toxicol 1983;3: 397–399.
  • Cipollone R, Ascenzi P, Tomao P, Imperi F, Visca P. Enzymatic detoxification of cyanide: clues from Pseudomonas aeruginosa rhodanese. J Mol Microbiol Biotechnol 2008; 15:199–211.
  • Spagnolo A, Torsello S, Morisi G, Petrozzi E, Antonini R, Ricci G, . Serum thiocyanate levels as an objective measure of smoking habits in epidemiological studies. Eur J Epidemiol 1988;4:206–211.
  • Jaimes EA, DeMaster EG, Tian RX, Raij L. Stable compounds of cigarette smoke induce endothelial superoxide anion production via NADPH oxidase activation. Arterioscler. Thromb Vasc Biol 2004;24:1031–1036.
  • Phillips TP. Overview of cassava consumption and production. Cassava, toxicity and thyroid: research and public health issues. Ottawa: IDRC; 1983. pp. 83–88.
  • Carlsson L, Mlingi N, Juma A, Ronquist G, Rosling H. Metabolic fates in humans of linamarin in cassava flour ingested as stiff porridge. Food Chem Toxicol 1999;37:307–312.
  • Okafor PN, Okorowkwo CO, Maduagwu EN. Occupational and dietary exposures of humans to cyanide poisoning from large-scale cassava processing and ingestion of cassava foods. Food Chem Toxicol 2002;40:1001–1005.
  • Vetter J. Plant cyanogenic glycosides. Toxicon 2000;38: 11–36.
  • Shibamoto T, Bjeldanes LF. Introduction to food toxicology. San Diego, California: Academic Press; 1993.
  • Vesey CJ, Cole PV. Blood cyanide and thiocyanate concentrations produced by long-term therapy with sodium nitroprusside. Br J Anaesth 1985;57:148–155.
  • Abdel-Naim AB, Mohamadin AM. Myeloperoxidase-catalyzed oxidation of chloroacetonitrile to cyanide. Toxicol Lett 2004;146:249–257.
  • Agency USEP Toxicological review of acetonitrile (Agency USEP, ed.), U.S. Environmental Protection Agency, Washington, DC; 1999.
  • Arnhold J, Monzani E, Furtmuller PG, Zederbauer M, Casella L, Obinger C. Kinetics and thermodynamics of halide and nitrite oxidation by mammalian heme peroxidases. Eur J Inorg Chem 2006;3801–3811.
  • Armesto XL, Canle ML, Garcia MV, Santaballa JA. Aqueous chemistry of N-halo-compounds. Chem Soc Rev 1998;27:453–460.
  • Deborde M, von Gunten U. Reactions of chlorine with inorganic and organic compounds during water treatment-kinetics and mechanisms: a critical review. Water Res 2008; 42:13–51.
  • Skaff O, Pattison DI, Davies MJ. Hypothiocyanous acid reactivity with low-molecular-mass and protein thiols: absolute rate constants and assessment of biological relevance. Biochem J 2009;422:111–117.
  • Skaff O, Pattison DI, Morgan PE, Bachana R, Jain VK, Priyadarsin KI, . Selenium-containing amino acids are targets for myeloperoxidase-derived hypothiocyanous acid: determination of absolute rate constants and implications for biological damage. Biochem J 2012;441:305–316.
  • Nagy P, Jameson GN, Winterbourn CC. Kinetics and mechanisms of the reaction of hypothiocyanous acid with 5-thio-2-nitrobenzoic acid and reduced glutathione. Chem Res Toxicol 2009;22:1833–1840.
  • Pattison DI, Hawkins CL, Davies MJ. What are the plasma targets of the oxidant hypochlorous acid? A kinetic modeling approach. Chem Res Toxicol 2009;22:807–817.
  • Nagy P, Winterbourn CC. Redox chemistry of biological thiols. In: Fishbein JC (ed) Advances in molecular toxicology. Elsevier; 2010. pp. 183–222.
  • Armesto XL, Canle ML, Santaballa JA. α-Amino acids chlorination in aqueous media. Tetrahedron 1993;49:275–284.
  • Folkes LK, Candeias LP, Wardman P. Kinetics and mechanisms of hypochlorous acid reactions. Arch Biochem Biophys 1995;323:120–126.
  • Pattison DI, Davies MJ. Absolute rate constants for the reaction of hypochlorous acid with protein side-chains and peptide bonds. Chem Res Toxicol 2001;14:1453–1464.
  • Antelo JM, Arce F, Parajo M. Kinetic study of the formation of N-chloramines. Int J Chem Kinet 1995;27:637–647.
  • Armesto XL, Canle ML, Fernandez MI, Garcia MV, Santaballa J. First steps in the oxidation of sulfur-containing amino acids by hypohalogenation: very fast generation of intermediate sulfenyl halides and halosulfonium cations. Tetrahedron 2000;56:1103–1109.
  • Peskin AV, Winterbourn CC. Kinetics of the reactions of hypochlorous acid and amino acid chloramines with thiols, methionine, and ascorbate. Free Radic Biol Med 2001;30: 572–579.
  • Prutz WA. Hypochlorous acid interactions with thiols, nucleotides, DNA, and other biological substrates. Arch Biochem Biophys 1996;332:110–120.
  • Peskin AV, Winterbourn CC. Histamine chloramine reactivity with thiol compounds, ascorbate and methionine and with intracellular glutathione. Free Radic Biol Med 2003; 35:1252–1260.
  • Lu J, Holmgren A. Selenoproteins. J Biol Chem 2009; 284:723–727.
  • Morris JC. Kinetics of reactions between aqueous chlorine and nitrogen compounds. In: Faust ED, Hunter JV (eds) Principles and applications of water chemistry. New York: John Wiley and Sons; 1967. pp. 23–53.
  • Pattison DI, Davies MJ. Evidence for rapid inter- and intra-molecular chlorine transfer reactions of histamine and carnosine chloramines: implications for the prevention of hypochlorous acid mediated damage. Biochemistry 2006; 45:8152–8162.
  • Prutz WA. Consecutive halogen transfer between various functional groups induced by reaction of hypohalous acids: NADH oxidation by halogenated amide groups. Arch Biochem Biophys 1999;371:107–114.
  • Curtis MP, Hicks AJ, Neidigh JW. Kinetics of 3-chlorotyrosine formation and loss due to hypochlorous acid and chloramines. Chem Res Toxicol 2011;24:418–428.
  • Winterbourn CC, Kettle AJ. Biomarkers of myeloperoxidase-derived hypochlorous acid. Free Radic Biol Med 2000; 29:403–409.
  • Fu S, Wang H, Davies MJ, Dean RT. Reaction of hypochlorous acid with tyrosine and peptidyl-tyrosyl residues gives dichlorinated and aldehydic products in addition to 3-chlorotyrosine. J Biol Chem 2000;275:10851–10857.
  • Kalmar J, Woldegiorgis KL, Biri B, Ashby MT. Mechanism of decomposition of the human defense factor hypothiocyanite near physiological pH. J Am Chem Soc 2011;133: 19911–19921.
  • Ashby MT, Aneetha H. Reactive sulfur species: aqueous chemistry of sulfenyl thiocyanates. J Am Chem Soc 2004; 126:10216–10217.
  • Ashby MT, Aneetha H, Carlson AC, Scott MJ, Beal JL. Bioorganic chemistry of hypothiocyanite. Phosphorus Sulfur Silicon Relat Elem 2005;180:1369–1374.
  • Lemma K, Ashby MT. Reactive sulfur species: kinetics and mechanism of the equilibrium between cysteine sulfenyl thiocyanate and cysteine thiosulfinate ester in acidic aqueous solution. J Org Chem 2008;73:3017–3023.
  • Nagy P, Lemma K, Ashby MT. Kinetics and mechanism of the comproportionation of hypothiocyanous acid and thiocyanate to give thiocyanogen in acidic aqueous solution. Inorg Chem 2007;46:285–292.
  • Nagy P, Lemma K, Ashby MT. Reactive sulfur species: kinetics and mechanisms of the reaction of cysteine thiosulfinate ester with cysteine to give cysteine sulfenic acid. J Org Chem 2007;72:8838–8846.
  • Nagy P, Wang X, Lemma K, Ashby MT. Reactive sulfur species: hydrolysis of hypothiocyanite to give thiocarbamate-S-oxide. J Am Chem Soc 2007;129:15756–15757.
  • Wang XG, Ashby MT. Reactive sulfur species: kinetics and mechanism of the reaction of thiocarbamate-S-oxide with cysteine. Chem Res Toxicol 2008;21:2120–2126.
  • Tenovuo J, Pruitt KM, Mansson-Rahemtulla B, Harrington P, Baldone DC. Products of thiocyanate peroxidation: properties and reaction mechanisms. Biochim Biophys Acta 1986;870:377–384.
  • Barrett TJ, Hawkins CL. Hypothiocyanous acid: benign or deadly? Chem Res Toxicol 2012;25:263–273.
  • Carr AC, Winterbourn CC. Oxidation of neutrophil glutathione and protein thiols by myeloperoxidase-derived hypochlorous acid. Biochem J 1997;327:275–281.
  • Davies MJ, Hawkins CL. Hypochlorite-induced oxidation of thiols: formation of thiyl radicals and the role of sulfenyl chlorides as intermediates. Free Radic Res 2000;33: 719–729.
  • Hu ML, Louie S, Cross CE, Motchnik P, Halliwell B. Antioxidant protection against hypochlorous acid in human plasma. J Lab Clin Med 1993;121:257–262.
  • Aune TM, Thomas EL. Accumulation of hypothiocyanite ion during peroxidase-catalyzed oxidation of thiocyanate ion. Eur J Biochem 1977;80:209–214.
  • Aune TM, Thomas EL, Morrison M. Lactoperoxidase-catalyzed incorporation of thiocyanate ion into a protein substrate. Biochemistry 1977;16:4611–4615.
  • Harwood DT, Kettle AJ, Winterbourn CC. Production of glutathione sulfonamide and dehydroglutathione from GSH by myeloperoxidase-derived oxidants and detection using a novel LC-MS/MS method. Biochem J 2006;399:161–168.
  • Drozdz R, Naskalski JW, Sznajd J. Oxidation of amino acids and peptides in reaction with myeloperoxidase, chloride and hydrogen peroxide. Biochim Biophys Acta 1988;957:47–52.
  • Pereira WE, Hoyano Y, Summons RE, Bacon VA, Duffield AM. Chlorination studies. II. The reaction of aqueous hypochlorous acid with α−αmino acids and dipeptides. Biochim Biophys Acta 1973;313:170–180.
  • Aune TM, Thomas EL. Oxidation of protein sulfhydryls by products of peroxidase-catalyzed oxidation of thiocyanate ion. Biochemistry 1978;17:1005–1010.
  • Hawkins CL, Pattison DI, Davies MJ. Hypochlorite-induced oxidation of amino acids, peptides and proteins. Amino Acids 2003;25:259–274.
  • Nagy P, Ashby MT. Reactive sulfur species: kinetics and mechanisms of the oxidation of cysteine by hypohalous acid to give cysteine sulfenic acid. J Am Chem Soc 2007; 129:14082–14091.
  • Barrett TJ, Pattison DI, Leonard SE, Carroll KS, Davies MJ, Hawkins CL. Inactivation of thiol-dependent enzymes by hypothiocyanous acid: role of sulfenyl thiocyanate and sulfenic acid intermediates. Free Radic Biol Med 2012;52: 1075–1085.
  • Algunindigue Nimmo SL, Lemma K, Ashby MT. Reactions of cysteine sulfenyl thiocyanate with thiols to give unsymmetrical disulfides. Heteroatom Chem 2007;18:467–471.
  • Pullar JM, Vissers MCM, Winterbourn CC. Glutathione oxidation by hypochlorous acid in endothelial cells produces glutathione sulfonamide as a major product but not glutathione disulfide. J Biol Chem 2001;276:22120–22125.
  • Harwood DT, Nimmo SL, Kettle AJ, Winterbourn CC, Ashby MT. Molecular structure and dynamic properties of a sulfonamide derivative of glutathione that is produced under conditions of oxidative stress by hypochlorous acid. Chem Res Toxicol 2008;21:1011–1016.
  • Harwood DT, Darlow BA, Cheah FC, McNeill N, Graham P, Winterbourn CC. Biomarkers of neutrophil-mediated glutathione and protein oxidation in tracheal aspirates from preterm infants: association with bacterial infection. Pediatr Res 2011;69:28–33.
  • Harwood DT, Kettle AJ, Brennan S, Winterbourn CC. Simultaneous determination of reduced glutathione, glutathione disulphide and glutathione sulphonamide in cells and physiological fluids by isotope dilution liquid chromatography-tandem mass spectrometry. J Chromatogr B 2009; 877:3393–3399.
  • Yuan W, Wang Y, Heinecke JW, Fu XY. Hypochlorous acid converts the gamma-glutamyl group of glutathione disulfide to 5-hydroxybutyrolactam, a potential marker for neutrophil activation. J Biol Chem 2009;284:26908–26917.
  • Hawkins CL, Pattison DI, Stanley NR, Davies MJ. Tryptophan residues are targets in hypothiocyanous acid-mediated protein oxidation. Biochem J 2008;416:441–452.
  • Thomas EL. Products of the lactoperoxidase-catalysed oxidation of thiocyanate and halides. In: Pruitt KM Tenovuo JO (eds) The lactoperoxidase system: chemistry and biological significance. New York: Marcel Dekker, Inc.; 1985. pp. 31–53.
  • Raftery MJ, Yang Z, Valenzuela SM, Geczy CL. Novel intra- and inter-molecular sulfinamide bonds in S100A8 produced by hypochlorite oxidation. J Biol Chem 2001;276:33 393–33401.
  • Dickinson BC, Chang CJ. Chemistry and biology of reactive oxygen species in signaling or stress responses. Nat Chem Biol 2011;7:504–511.
  • Kettenhofen NJ, Wood MJ. Formation, reactivity, and detection of protein sulfenic acids. Chem Res Toxicol 2010; 23:1633–1646.
  • Klomsiri C, Karplus PA, Poole LB. Cysteine-based redox switches in enzymes. Antioxid Redox Signal 2011;14: 1065–1077.
  • Roos G, Messens J. Protein sulfenic acid formation: from cellular damage to redox regulation. Free Radic Biol Med 2011;51:314–326.
  • Chang TS, Jeong W, Woo HA, Lee SM, Park S, Rhee SG. Characterization of mammalian sulfiredoxin and its reactivation of hyperoxidized peroxiredoxin through reduction of cysteine sulfinic acid in the active site to cysteine. J Biol Chem 2004;279:50994–51001.
  • Flohe L, Toppo S, Cozza G, Ursini F. A comparison of thiol peroxidase mechanisms. Antioxid Redox Signal 2011;15: 763–780.
  • Cho CS, Lee S, Lee GT, Woo HA, Choi EJ, Rhee SG. Irreversible inactivation of glutathione peroxidase 1 and reversible inactivation of peroxiredoxin II by H2O2 in red blood cells. Antioxid Redox Signal 2010;12:1235–1246.
  • Lee BC, Dikiy A, Kim HY, Gladyshev VN. Functions and evolution of selenoprotein methionine sulfoxide reductases. Biochim Biophys Acta 2009;1790:1471–1477.
  • Beal JL, Foster SB, Ashby MT. Hypochlorous acid reacts with the N-terminal methionines of proteins to give dehydromethionine, a potential biomarker for neutrophil-induced oxidative stress. Biochemistry 2009;48:11142–11148.
  • Peskin AV, Turner R, Maghzal GJ, Winterbourn CC, Kettle AJ. Oxidation of methionine to dehydromethionine by reactive halogen species generated by neutrophils. Biochemistry 2009;48:10175–10182.
  • Assmann A, Briviba K, Sies H. Reduction of methionine selenoxide to selenomethionine by glutathione. Arch Biochem Biophys 1998;349:201–203.
  • Suryo Rahmanto A, Davies MJ. Catalytic activity of selenomethionine in removing amino acid, peptide, and protein hydroperoxides Free Radic Biol Med 2011;51:2288–2299.
  • Thomas EL, Grisham MB, Jefferson MM. Preparation and characterization of chloramines. Meth Enzymol 1986;132: 569–585.
  • Thomas EL, Bozeman PM, Jefferson MM, King CC. Oxidation of bromide by the human leukocyte enzymes myeloperoxidase and eosinophil peroxidase. Formation of bromamines. J Biol Chem 1995;270:2906–2913.
  • Hawkins CL, Davies MJ. Reaction of hocl with amino acids and peptides: EPR evidence for rapid rearrangement and fragmentation reactions of nitrogen-centered radicals. J Chem Soc Perkin Trans 2 1998;1937–1945.
  • Hawkins CL, Davies MJ. Hypochlorite-induced damage to proteins: formation of nitrogen-centred radicals from lysine residues and their role in protein fragmentation. Biochem J 1998;332:617–625.
  • Peskin AV, Midwinter RG, Harwood DT, Winterbourn CC. Chlorine transfer between glycine, taurine and histamine: reaction rates and impact on cellular reactivity. Free Radic Biol Med 2004;37:1622–1630.
  • Pattison DI, Hawkins CL, Davies MJ. Hypochlorous acid-mediated protein oxidation: how important are chloramine transfer reactions and protein tertiary structure? Biochemistry 2007;46:9853–9864.
  • Prutz WA, Kissner R, Koppenol WH. Oxidation of nadh by chloramines and chloramides and its activation by iodide and by tertiary amines. Arch Biochem Biophys 2001;393:297–307.
  • Thomas EL. Lactoperoxidase-catalyzed oxidation of thiocyanate: equilibria between oxidized forms of thiocyanate. Biochemistry 1981;20:3273–3280.
  • Fu X, Wang Y, Kao J, Irwin A, d'Avignon A, Mecham RP, . Specific sequence motifs direct the oxygenation and chlorination of tryptophan by myeloperoxidase. Biochemistry 2006;45:3961–3971.
  • Fu X, Kao JLF, Bergt C, Kassim SY, Huq NP, d'Avignon A, . Oxidative cross-linking of tryptophan to glycine restrains matrix metalloproteinase activity: specific structural motifs control protein oxidation. J Biol Chem 2004;279:6209–6212.
  • Hawkins CL, Davies MJ. Inactivation of protease inhibitors and lysozyme by hypochlorous acid: role of side-chain oxidation and protein unfolding in loss of biological function. Chem Res Toxicol 2005;18:1600–1610.
  • Hawkins CL, Davies MJ. The role of aromatic amino acid oxidation, protein unfolding, and aggregation in the hypobromous acid-induced inactivation of trypsin inhibitor and lysozyme. Chem Res Toxicol 2005;18:1669–1677.
  • Peng DQ, Brubaker G, Wu Z, Zheng L, Willard B, Kinter M, . Apolipoprotein A-I tryptophan substitution leads to resistance to myeloperoxidase-mediated loss of function. Arterioscler Thromb Vasc Biol 2008;28:2063–2070.
  • Domigan NM, Charlton TS, Duncan MW, Winterbourn CC, Kettle AJ. Chlorination of tyrosyl residues in peptides by myeloperoxidase and human neutrophils. J Biol Chem 1995;270:16542–16548.
  • Bergt C, Fu X, Huq NP, Kao J, Heinecke JW. Lysine residues direct the chlorination of tyrosines in YXXK motifs of apolipoprotein A-I when hypochlorous acid oxidizes HDL. J Biol Chem 2004;279:7856–7866.
  • Zheng L, Settle M, Brubaker G, Schmitt D, Hazen SL, Smith JD, . Localization of nitration and chlorination sites on apolipoprotein A-I catalyzed by myeloperoxidase in human atheroma and associated oxidative impairment in ABCA1-dependent cholesterol efflux from macrophages. J Biol Chem 2005;280:38–47.
  • Shao B, Bergt C, Fu X, Green P, Voss JC, Oda MN, . Tyrosine 192 in apolipoprotein A-I is the major site of nitration and chlorination by myeloperoxidase, but only chlorination markedly impairs ABCA1-dependent cholesterol transport. J Biol Chem 2005;280:5983–5993.
  • Hampton MB, Kettle AJ, Winterbourn CC. Inside the neutrophil phagosome: oxidants, myeloperoxidase, and bacterial killing. Blood 1998;92:3007–3017.
  • Seifu E, Buys EM, Donkin EF. Significance of the lactoperoxidase system in the dairy industry and its potential applications: a review. Trends Food Sci Technol 2005;16: 137–154.
  • Wolfson LM, Sumner SS. Antibacterial activity of the lactoperoxidase system – a review. J Food Protect 1993;56: 887–892.
  • de Wit JN, van Hooydonk ACM. Structure, functions and applications of lactoperoxidase in natural antimicrobial systems. Neth Milk Dairy J 1996;50:227–244.
  • Reiter B, Harnulv G. Lactoperoxidase antibacterial system – natural occurrence, biological functions and practical applications. J Food Protect 1984;47:724–732.
  • Bjorck L, Claesson O, Schulthess W. The lactoperoxidase/thiocyanate/hydrogen peroxide system as a temporary preservative for raw milk in developing countries. Milchwissenschaft 1979;34:726–729.
  • Harnulv BG, Kandasamy C. Increasing the keeping quality of raw milk by activation of the lactoperoxidase system. Results from Sri Lanka. Milchwissenschaft 1982;37:454–457.
  • Thomas EL, Aune TM. Lactoperoxidase, peroxide, thiocyanate antimicrobial system: correlation of sulfhydryl oxidation with antimicrobial action. Infect Immun 1978;20:456–463.
  • Oram JD, Reiter B. The inhibition of Streptococci by lactoperoxidase, thiocyanate and hydrogen peroxide – the oxidation of thiocyanate and the nature of the inhibitory compound. Biochem J 1966;100:382–388.
  • Mickelson MN. Antibacterial action of lactoperoxidase-thiocyanate-hydrogen peroxide on Streptococcus agalactiae. Appl Environ Microbiol 1979;38:821–826.
  • Hoogendoorn H, Piessens JP, Scholtes W, Stoddard LA. Hypothiocyanite ion; the inhibitor formed by the system lactoperoxidase-thiocyanate-hydrogen peroxide. I. Identification of the inhibiting compound. Caries Res 1977;11:77–84.
  • Carlsson J, Iwami Y, Yamada T. Hydrogen peroxide excretion by oral Streptococci and effect of lactoperoxidase-thiocyanate-hydrogen peroxide. Infect Immun 1983;40:70–80.
  • Pruitt KM, Reiter B. Biochemistry of peroxidase system: antimicrobial effects. In: Pruitt KM, Tenovuo J (eds) The lactoperoxidase system: chemistry and biological significance. New York: Marcel Dekker; 1985. pp. 143–178.
  • Shin K, Hayasawa H, Lonnerdal B. Inhibition of Escherichia coli respiratory enzymes by the lactoperoxidase-hydrogen peroxide-thiocyanate antimicrobial system. J Appl Microbiol 2001;90:489–493.
  • Shin K, Yamauchi K, Teraguchi S, Hayasawa H, Imoto I. Susceptibility of Helicobacter pylori and its urease activity to the peroxidase-hydrogen peroxide-thiocyanate antimicrobial system. J Med Microbiol 2002;51:231–237.
  • Das D, De PK, Banerjee RK. Thiocyanate, a plausible physiological electron-donor of gastric peroxidase. Biochem J 1995;305:59–64.
  • Ashby MT. Inorganic chemistry of defensive peroxidases in the human oral cavity. J Dent Res 2008;87:900–914.
  • Pruitt KM. The salivary peroxidase system: thermodynamic, kinetic and antibacterial properties. J Oral Pathol 1987;16: 417–420.
  • Cao CF. Crevicular fluid myeloperoxidase at healthy, gingivitis and periodontitis sites. J Clin Periodontol 1989;16: 17–20.
  • Schultz CP, Ahmed AH, Dawes C, Mantsch HH. Thiocyanate levels in human saliva – quantification by fourier transform infrared spectroscopy. Anal Biochem 1996;240:7–12.
  • Fragoso MA, Fernandez V, Forteza R, Randell RH, Salathe M, Conner GE. Transcellular thiocyanate transport by human airway epithelia. J Physiol 2004;561:183–194.
  • Tenovuo J, Pruitt KM, Thomas EL. Peroxidase antimicrobial system of human saliva: hypothiocyanite levels in resting and stimulated saliva. J Dent Res 1982;61:982–985.
  • Clem W, Klebanof S. Inhibitory effect of saliva on glutamic acid accumulation by Lactobacillus acidophilus and role of lactoperoxidase-thiocyanate system. J Bacteriol 1966;91: 1848–1853.
  • Germaine GR, Tellefson LM. Streptococcus mutans, Streptococcus mitis, and Actinomyces viscosus in the presence of human saliva. Infect Immun 1982;38:1060–1067.
  • Tenovuo J, Mansson-Rahemtulla B, Pruitt KM, Arnold R. Inhibition of dental plaque acid production by the salivary lactoperoxidase antimicrobial system. Infect Immun 1981; 34:208–214.
  • Tenovuo J, Jentsch H, Soukka T, Karhuvaara L. Antimicrobial factors of saliva in relation to dental caries and salivary levels of mutans Streptococci. J Biol Buccale 1992;20:85–90.
  • Carlsson J, Edlund MB, Hanstrom L. Bactericidal and cytotoxic effects of hypothiocyanite-hydrogen peroxide mixtures. Infect Immun 1984;44:581–586.
  • Fadel M, Courtois P. Effect of peroxidase-generated hypothiocyanite on the survival rate of Porphyromonas gingivalisNCTC11834. Med Sci Res 1999;27:667–669.
  • Carlsson J. Bactericidal effect of hydrogen peroxide is prevented by the lactoperoxidase-thiocyanate system under anaerobic conditions. Infect Immun 1980;29:1190–1192.
  • Adamson M, Carlsson J. Lactoperoxidase and thiocyanate protect bacteria from hydrogen-peroxide. Infect Immun 1982;35:20–24.
  • Courtois PH, Pourtois M. Purification of nadh: hypothiocyanite oxidoreductase in Streptococcus sanguis. Biochem Mol Med 1996;57:134–138.
  • Oram JD, Reiter B. The inhibition of Streptococci by lactoperoxidase, thiocyanate and hydrogen peroxide – the effect of the inhibitory system on susceptible and resistant strains of group N Streptococci. Biochem J 1966;100:373–381.
  • Takahama U, Hirotab S, Nishioka T, Onikia T. Human salivary peroxidase-catalyzed oxidation of nitrite and nitration of salivary components 4-hydroxyphenylacetic acid and proteins. Arch Oral Biol 2003;48:679–690.
  • Christensen TG. The distribution and function of peroxidases in the respiratory tract. Surv Synth Pathol Res 1984;3:201–218.
  • Wijkstrom-Frei C, El-Chemaly S, Ali-Rachedi R, Gerson C, Cobas MA, Forteza R, . Lactoperoxidase and human airway host defense. Am J Respir Cell Mol Biol 2003;29:206–212.
  • Gerson C, Sabater J, Scuri M, Torbati A, Coffey R, Abraham JW, . The lactoperoxidase system functions in bacterial clearance of airways. Am J Respir Cell Mol Biol 2000;22:665–671.
  • Lorentzen D, Durairaj L, Pezzulo AA, Nakano Y, Launspach J, Stoltz DA, . Concentration of the antibacterial precursor thiocyanate in cystic fibrosis airway secretions. Free Radic Biol Med 2011;50:1144–1150.
  • McCarty N. Permeation through the CFTR chloride channel. J Exp Biol 2000;203:1947–1962.
  • Moskwa P, Lorentzen D, Excoffon K, Zabner J, McCray PB, Nauseef WM, . A novel host defense system of airways is defective in cystic fibrosis. Am J Respir Crit Care Med 2007;175:174–183.
  • Conner GE, Wijkstrom-Frei C, Randell SH, Fernandez VE, Salathe M. The lactoperoxidase system links anion transport to host defense in cystic fibrosis. FEBS Lett 2007;581:271–278.
  • Illek B, Tam AW, Fischer H, Machen TE. Anion selectivity of apical membrane conductance of CALU 3 human airway epithelium. Eur J Physiol 1999;437:812–822.
  • Van Sande J, Massart C, Beauwens R, Schoutens A, Costagliola S, Dumont J, . Anion selectivity by the sodium iodide symporter. Endocrinology 2003;144:247–252.
  • Minarowski L, Sands D, Minarowska A, Karwowska A, Sulewska A, Gacko M, . Thiocyanate concentrations in saliva of cystic fibrosis patients. Folia Histochem Cytobiol 2008;46:245–246.
  • Dawson D, Smith S, Mansoura M. Cftr: mechanism of anion conduction. Physiol Rev 1999;79:S47–S75.
  • Elizur A, Cannon C, Ferkol T. Airway inflammation in cystic fibrosis. Chest 2008;133:489–495.
  • van der Vliet A, Nguyen MN, Shigenaga MK, Eiserich JP, Marelich GP, Cross CE. Myeloperoxidase and protein oxidation in cystic fibrosis. Am J Physiol Lung Cell Mol Physiol 2000;279:L537–L546.
  • Koller DY, Nilsson M, Enander I, Venge P, Eichler I. Serum eosinophil cationic protein, eosinophil protein X and eosinophil peroxidase in relation to pulmonary function in cystic fibrosis. Clin Exp Allergy 1998;28:241–248.
  • Koller DY, Gotz M, Eichler I, Urbanek R. Eosinophilic activation in cystic-fibrosis. Thorax 1994;49:496–499.
  • McElhinney B, Poynter ME, Shrivastava P, Hazen SL, Janssen-Heininger YMW. Eosinophil peroxidase catalyzes JNK-mediated membrane blebbing in a Rho kinase-dependent manner. J Leukocyte Biol 2003;74:897–907.
  • Saude EJ, Lacy P, Musat-Marcu S, Mayes DC, Bagu J, Man SFP, . NMR analysis of neutrophil samples from patients with activation in sputum cystic fibrosis. Magn Reson Med 2004;52:807–814.
  • Hoy A, Leininger-Muller B, Kutter D, Siest G, Visvikis S. Growing significance of myeloperoxidase in non-infectious diseases. Clin Chem Lab Med 2002;40:2–8.
  • Scanlon CEO, Berger B, Malcom G, Wissler RW. Evidence for more extensive deposits of epitopes of oxidized low density lipoproteins in aortas of young people with elevated serum thiocyanate levels. Atherosclerosis 1996;121:23–33.
  • Botti TP, Amin H, Hiltscher L, Wissler RW. A comparison of the quantitation of macrophage foam cell populations and the extent of apolipoprotein E deposition in developing atherosclerotic lesions in young people: high and low serum thiocyanate groups as an indication of smoking. Atherosclerosis 1996;124:191–202.
  • Zhang R, Shen Z, Nauseef WM, Hazen SL. Defects in leukocyte-mediated initiation of lipid peroxidation in plasma as studied in myeloperoxidase-deficient subjects: systematic identification of multiple endogenous diffusible substrates for myeloperoxidase in plasma. Blood 2002;99:1802–1810.
  • Exner M, Hermann M, Hofbauer R, Hartmann B, Kapiotis S, Gmeiner B. Thiocyanate catalyzes myeloperoxidase-initiated lipid oxidation in LDL. Free Radic Biol Med 2004; 37:146–155.
  • Lovaas E. Free radical generation and coupled thiol oxidation by lactoperoxidase/SCN−/H2O2. Free Radic Biol Med 1992;13:187–195.
  • Ashfaq S, Abramson JL, Jones DP, Rhodes SD, Weintraub WS, Hooper WC, . The relationship between plasma levels of oxidized and reduced thiols and early atherosclerosis in healthy adults. J Am Coll Cardiol 2006;47:1005–1011.
  • Stark GR, Stein WH, Moore S. Reactions of the cyanate present in aqueous urea with amino acids and proteins. J Biol Chem 1960;235:3177–3181.
  • Holzer M, Gauster M, Pfeifer T, Wadsack C, Fauler G, Stiegler P, . Protein carbamylation renders high-density lipoprotein dysfunctional. Antioxid Redox Signal 2011;14: 2337–2346.
  • Horkko S, Savolainen MJ, Kervinen K, Kesaniemi YA. Carbamylation-induced alterations in low-density-lipoprotein metabolism. Kidney Int 1992;41:1175–1181.
  • Roxborough HE, Young IS. Carbamylation of proteins and atherogenesis in renal failure. Med Hypoth 1995;45: 125–128.
  • Ok E, Basnakian AG, Apostolov EO, Barri YM, Shah PK. Carbamylated low density lipoprotein induces death of endothelial cells: a link to atherosclerosis in patients with kidney disease. Kidney Int 2005;68:173–178.
  • Sirpal S. Myeloperoxidase-mediated lipoprotein carbamylation as a mechanistic pathway for atherosclerotic vascular disease. Clin Sci 2009;116:681–695.
  • Holzer M, Birner-Gruenberger R, Stojakovic T, El-Gamal D, Binder V, Wadsack C, . Uremia alters HDL composition and function. J Am Soc Nephrol 2011;22:1631–1641.
  • Libby P. Inflammation in atherosclerosis. Nature 2002;420: 868–874.
  • Rehani T, Jonas A, Slungaard A. The major phagocyte peroxidase-derived oxidant hoscn stimulates the gene specific p38 MAPK-dependent NF-κB mediated activation of tissue factor, VCAM-1 and ICAM-1 expression in endothelium. Blood 2010;116:633.
  • Yamamoto Y, Gaynor RB. Role of the NF-κB pathway in the pathogenesis of human disease states. Curr Mol Med 2001;1:287–296.
  • Gareus R, Kotsaki E, Xanthoulea S, van der Made I, Gijbels MJ, Kardakaris R, . Endothelial cell-specific NF-κB inhibition protects mice from atherosclerosis. Cell Metab 2008;8:372–383.
  • Kockx MM. Apoptosis in the atherosclerotic plaque: quantitative and qualitative aspects. Arterioscler Thromb Vasc Biol 1998;18:1519–1522.
  • Tabas I. Macrophage apoptosis in atherosclerosis: consequences on plaque progression and the role of endoplasmic reticulum stress. Antioxid Redox Signal 2009;11: 2333–2339.
  • Virmani R, Burke AP, Kolodgie FD, Farb A. Pathology of the thin-cap fibroatheroma: a type of vulnerable plaque. J Interv Cardiol 2003;16:267–272.
  • Libby P. Changing concepts of atherogenesis. J Intern Med 2000;247:349–358.
  • Wagner BA, Reszka KJ, McCormick ML, Britigan BE, Evig CB, Burns CP. Role of thiocyanate, bromide and hypobromous acid in hydrogen peroxide-induced apoptosis. Free Radic Res 2004;38:167–175.
  • Wu WJ, Chen YH, d'Avignon A, Hazen SL. 3-bromotyrosine and 3,5-dibromotyrosine are major products of protein oxidation by eosinophil peroxidase: potential markers for eosinophil-dependent tissue injury in vivo. Biochemistry 1999;38:3538–3548.
  • Wang J, Slungaard A. Role of eosinophil peroxidase in host defence and disease pathology. Arch Biochem Biophys 2006;445:256–260.
  • Forman HJ, Fukuto JM, Torres M. Redox signaling: thiol chemistry defines which reactive oxygen and nitrogen species can act as second messengers. Am J Physiol Cell Physiol 2004;287:C246–C256.
  • Janssen-Heininger YMW, Mossman BT, Heintz NH, Forman HJ, Kalyanaraman B, Finkel T, . Redox regulation of signal transduction: principles, pitfalls and promises. Free Radic Biol Med 2008;45:1–17.
  • Van Antwerpen P, Zouaoui Boudjeltia K, Furtmuller PG, Dieu M, Delporte C, Raes M, Oxidation of cyanide to cyanate by myeloperoxidase: a new route for protein carbamylation. In: Abstracts from 6th International Human Peroxidase Meeting, Chapel Hill, North Carolina, USA; 2009.
  • Furtmuller PG, Van Antwerpen P, Zouaoui Boudjeltia K, Obinger C. Role of cyanide as low spin ligand and electron donor in catalysis of mammalian peroxidases. In: Abstracts from 6th International Human Peroxidase Meeting, Chapel Hill, North Carolina, USA; 2009.
  • Gerritsen CM, Margerum DW. Non-metal redox kinetics: hypochlorite and hypochlorous acid reactions with cyanide. Inorg Chem 1990;29:2757–2762.
  • Radi R, Beckman JS, Bush KM, Freeman BA. Peroxynitrite oxidation of sulfhydryls – the cytotoxic potential of superoxide and nitric-oxide. J Biol Chem 1991;266:4244–4250.
  • Koppenol WH, Moreno JJ, Pryor WA, Ischiropoulos H, Beckman JS. Peroxynitrite, a cloaked oxidant formed by nitric oxide and superoxide. Chem Res Toxicol 1992;5: 834–842.
  • Trujillo M, Radi R. Peroxynitrite reaction with the reduced and the oxidized forms of lipoic acid: new insights into the reaction of peroxynitrite with thiols. Arch Biochem Biophys 2002;397:91–98.
  • Perrin D, Koppenol WH. The quantitative oxidation of methionine to methionine sulfoxide by peroxynitrite. Arch Biochem Biophys 2000;377:266–272.
  • Pryor WA, Jin X, Squadrito GL. One-electron and 2-electron oxidations of methionine by peroxynitrite. Proc Natl Acad Sci USA 1994;91:11173–11177.
  • Padmaja S, Squadrito GL, Lemercier JN, Cueto R, Pryor WA. Rapid oxidation of DL-selenomethionine by peroxynitrite. Free Radic Biol Med 1996;21:317–322.
  • Winterbourn CC, Metodiewa D. Reactivity of biologically important thiol compounds with superoxide and hydrogen peroxide. Free Radic Biol Med 1999;27:322–328.
  • Nguyen TH, Burnier J, Meng W. The kinetics of relaxin oxidation by hydrogen-peroxide. Pharmaceut Res 1993;10: 1563–1571.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.