444
Views
70
CrossRef citations to date
0
Altmetric
Research Article

The role of hypothiocyanous acid (HOSCN) in biological systems

Pages 1147-1158 | Received 18 Jun 2009, Published online: 12 Nov 2009

References

  • 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.
  • 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.
  • Thomas EL. Products of the lactoperoxidase-catalysed oxidation of thiocyanate and halides. KM Pruitt, JO Tenovuo. The lactoperoxidase system: chemistry and biological significance. New York: Marcel Dekker, Inc; 1985. 31–53.
  • Husgafvel-Pursiainen K, Sorsa M, Engstrom K, Einisto P. Passive smoking at work: biochemical and biological measures of exposure to environmental tobacco smoke. Int Arch Occup Environ Health 1987; 59:337–345.
  • 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 Chem2006; 3801–3811.
  • van Dalen CJ, Whitehouse MW, Winterbourn CC, Kettle AJ. Thiocyanate and chloride as competing substrates for myeloperoxidase. Biochem J 1997; 327:487–492.
  • van Dalen CJ, Kettle AJ. Substrates and products of eosinophil peroxidase. Biochem J 2001; 358:233–239.
  • Slungaard A, Mahoney JR Jr. Thiocyanate is the major substrate for eosinophil peroxidase in physiologic fluids. Implications for cytotoxicity. J Biol Chem 1991; 266:4903–4910.
  • Das D, De PK, Banerjee RK. Thiocyanate, a plausible physiological electron-donor of gastric peroxidase. Biochem J 1995; 305:59–64.
  • Contempre B, de Escobar GM, Denef JF, Dumont JE, Many MC. Thiocyanate induces cell necrosis and fibrosis in selenium- and iodine-deficient rat thyroids: a potential experimental model for myxedematous endemic cretinism in central Africa. Endocrinology 2004; 145:994–1002.
  • Tahboub YR, Galijasevic S, Diamond MP, Abu-Soud HM. Thiocyanate modulates the catalytic activity of mammalian peroxidases. J Biol Chem 2005; 280:26129–26136.
  • Modi S, Deodhar SS, Behere DV, Mitra S. Lactoperoxidase-catalyzed oxidation of thiocyanate by hydrogen peroxide: nuclear 15N magnetic resonance and optical spectral studies. Biochemistry 1991; 30:118–124.
  • Furtmuller PG, Burner U, Obinger C. Reaction of myeloperoxidase compound I with chloride, bromide, iodide, and thiocyanate. Biochemistry 1998; 37:17923–17930.
  • Furtmuller PG, Burner U, Regelsberger G, Obinger C. Spectral and kinetic studies on the formation of eosinophil peroxidase compound I and its reaction with halides and thiocyanate. Biochemistry 2000; 39:15578–15584.
  • Furtmuller PG, Jantschko W, Regelsberger G, Jakopitsch C, Arnhold J, Obinger C. Reaction of lactoperoxidase compound I with halides and thiocyanate. Biochemistry 2002; 41:11895–11900.
  • Nord G, Pedersen B, Farver O. Outer-sphere oxidation of iodide and thiocyanate by tris(2,2′-bipyridyl)- and tris(1,10-phenanthroline)osmium(III) in aqueous solutions. Inorg Chem 1978; 17:2233–2238.
  • Furtmuller PG, Arnhold J, Jantschko W, Pichler H, Obinger C. Redox properties of the couples compound I/compound II and compound II/native enzyme of human myeloperoxidase. Biochem Biophys Res Commun 2003; 301:551–557.
  • Furtmuller PG, Arnhold J, Jantschko W, Zederbauer M, Jakopitsch C, Obinger C. Standard reduction potentials of all couples of the peroxidase cycle of lactoperoxidase. J Inorg Biochem 2005; 99:1220–1229.
  • DeFelippis MR, Murthy CP, Faraggi M, Klapper MH. Pulse radiolytic measurement of redox potentials: the tyrosine and tryptophan radicals. Biochemistry 1989; 28:4847–4853.
  • Wang J, Slungaard A. Role of eosinophil peroxidase in host defence and disease pathology. Arch Biochem Biophys 2006; 445:256–260.
  • Davies MJ, Hawkins CL, Pattison DI, Rees MD. Mammalian heme peroxidases: from molecular mechanisms to health implications. Antiox Redox Signal 2008; 10:1199–1234.
  • Klebanoff SJ. Myeloperoxidase: friend and foe. J Leukocyte Biol 2005; 77:598–625.
  • Daugherty A, Dunn JL, Rateri DL, Heinecke JW. Myeloperoxidase, a catalyst for lipoprotein oxidation, is expressed in human atherosclerotic lesions. J Clin Invest 1994; 94:437–444.
  • Hazell LJ, Arnold L, Flowers D, Waeg G, Malle E, Stocker R. Presence of hypochlorite-modified proteins in human atherosclerotic lesions. J Clin Invest 1996; 97:1535–1544.
  • Hazell LJ, Baernthaler G, Stocker R. Correlation between intima-to-media ratio, apolipoprotein B-100, myeloperoxidase, and hypochlorite-oxidized proteins in human atherosclerosis. Free Radic Biol Med 2001; 31:1254–1262.
  • 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.
  • 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.
  • Zhang R, Brennan ML, Fu X, Aviles RJ, Pearce GL, Penn MS, Topol EJ, Sprecher DL, Hazen SL. Association between myeloperoxidase levels and risk of coronary artery disease. J Am Med Assoc 2001; 286:2136–2142.
  • Brennan ML, Penn MS, Van Lente F, Nambi V, Shishehbor MH, Aviles RJ, Goormastic M, Pepoy ML, McErlean ES, Topol EJ, Nissen SE, Hazen SL. Prognostic value of myeloperoxidase in patients with chest pain. N Engl J Med 2003; 349:1595–1604.
  • Baldus S, Heeschen C, Meinertz T, Zeiher AM, Eiserich JP, Munzel T, Simoons ML, Hamm CW. Myeloperoxidase serum levels predict risk in patients with acute coronary syndromes. Circulation 2003; 108:1440–1445.
  • Mocatta TJ, Pilbrow AP, Cameron VA, Senthilmohan R, Frampton CM, Richards AM, Winterbourn CC. Plasma concentrations of myeloperoxidase predict mortality after myocardial infarction. J Am Coll Cardiol 2007; 49:1993–2000.
  • 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.
  • Mitra SN, Slungaard A, Hazen SL. Role of eosinophil peroxidase in the origins of protein oxidation in asthma. Redox Rep 2000; 5:215–224.
  • Heinecke JW. Eosinophil-dependent bromination in the pathogenesis of asthma. J Clin Invest 2000; 105:1331–1332.
  • 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.
  • 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.
  • Wang Z, Nicholls SJ, Rodriguez ER, Kummu O, Horkko S, Barnard J, Reynolds WF, Topol EJ, DiDonato JA, Hazen SL. Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nature Med 2007; 13:1176–1184.
  • Thomas EL, Bates KP, Jefferson MM. Peroxidase antimicrobial system of human saliva: requirements for accumulation of hypothiocyanite. J Dent Res 1981; 60:785–796.
  • Pruitt KM, Tenovuo J, Andrews RW, McKane T. Lactoperoxidase-catalyzed oxidation of thiocyanate: polarographic study of the oxidation products. Biochemistry 1982; 21:562–567.
  • 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.
  • Aune TM, Thomas EL. Accumulation of hypothiocyanite ion during peroxidase-catalyzed oxidation of thiocyanate ion. Eur J Biochem 1977; 80:209–214.
  • Wever R, Kast WM, Kasinoedin JH, Boelens R. The peroxidation of thiocyanate catalysed by myeloperoxidase and lactoperoxidase. Biochim Biophys Acta 1982; 709:212–219.
  • Pruitt KM, Tenovuo J. Kinetics of hypothiocyanite production during peroxidase-catalyzed oxidation of thiocyanate. Biochim Biophys Acta 1982; 704:204–214.
  • Thomas EL. Lactoperoxidase-catalyzed oxidation of thiocyanate: equilibria between oxidized forms of thiocyanate. Biochemistry 1981; 20:3273–3280.
  • Nagy P, Alguindigue SS, Ashby MT. Lactoperoxidase-catalyzed oxidation of thiocyanate by hydrogen peroxide: a reinvestigation of hypothiocyanite by nuclear magnetic resonance and optical spectroscopy. Biochemistry 2006; 45:12610–12616.
  • Pollock JR, Goff HM. Lactoperoxidase-catalyzed oxidation of thiocyanate ion: a carbon-13 nuclear magnetic resonance study of the oxidation products. Biochim Biophys Acta 1992; 1159:279–285.
  • Arlandson M, Decker T, Roongta VA, Bonilla L, Mayo KH, MacPherson JC, Hazen SL, Slungaard A. Eosinophil peroxidase oxidation of thiocyanate—characterization of major reaction products and a potential sulfhydryl-targeted cytotoxicity system. J Biol Chem 2001; 276:215–224.
  • Aune TM, Thomas EL. Oxidation of protein sulfhydryls by products of peroxidase-catalyzed oxidation of thiocyanate ion. Biochemistry 1978; 17:1005–1010.
  • 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.
  • 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.
  • 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.
  • Aune TM, Thomas EL, Morrison M. Lactoperoxidase-catalyzed incorporation of thiocyanate ion into a protein substrate. Biochemistry 1977; 16:4611–4615.
  • Ashby MT, Aneetha H, Carlson AC, Scott MJ, Beal JL. Bioorganic chemistry of hypothiocyanite. Phosphorus Sulfur Silicon Relat Elem 2005; 180:1369–1374.
  • Grisham MB, Ryan EM. Cytotoxic properties of salivary oxidants. Am J Physiol 1990; 258:C115–C121.
  • Ashby MT, Aneetha H. Reactive sulfur species: aqueous chemistry of sulfenyl thiocyanates. J Am Chem Soc 2004; 126:10216–10217.
  • 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.
  • Algunindigue Nimmo SL, Lemma K, Ashby MT. Reactions of cysteine sulfenyl thiocyanate with thiols to give unsymmetrical disulfides. Heteroatom Chem 2007; 18:467–471.
  • 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.
  • 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.
  • Janssen-Heininger YMW, Mossman BT, Heintz NH, Forman HJ, Kalyanaraman B, Finkel T, Stamler JS, Rhee SG, van der Vliet A. Redox regulation of signal transduction: principles, pitfalls and promises. Free Radic Biol Med 2008; 45:1–17.
  • 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.
  • Hawkins CL, Pattison DI, Stanley NR, Davies MJ. Tryptophan residues are targets in hypothiocyanous acid-mediated protein oxidation. Biochem J 2008; 416:441–452.
  • 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.
  • Hawkins CL, Pattison DI, Davies MJ. Hypochlorite-induced oxidation of amino acids, peptides and proteins. Amino Acids 2003; 25:259–274.
  • Stark GR. Modification of proteins with cyanate. Method Enzymol 1998; 25:579–584.
  • 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.
  • Van Antwerpen P, Zouaoui Boudjeltia K, Furtmuller PG, Dieu M, Delporte C, Raes M, Moguilevsky N, Vanhaeverbeek M, Ducobu J, Neve J, Obinger C. 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.
  • 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.
  • 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.
  • Spalteholz H, Wenske K, Arnhold J. Interaction of hypohalous acids and heme peroxidases with unsaturated phosphatidylcholines. Biofactors 2005; 24:67–76.
  • White WE Jr. Pruitt KM, Mansson-Rahemtulla B. Peroxidase-thiocyanate-peroxide antibacterial system does not damage DNA. Antimicrob Agents Chemother 1983; 23:267–272.
  • Suzuki T, Ohshima H. Modification by fluoride, bromide, iodide, thiocyanate and nitrite anions of reaction of a myeloperoxidase-H2O2-Cl- system with nucleosides. Chem Pharm Bull 2003; 51:301–304.
  • 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.
  • Mikola H, Waris M, Tenovuo J. Inhibition of herpes simplex virus type 1, respiratory syncytial virus and echovirus type 11 by peroxidase-generated hypothiocyanite. Antiviral Res 1995; 26:161–171.
  • Thomas EL, Aune TM. Lactoperoxidase, peroxide, thiocyanate antimicrobial system: correlation of sulfhydryl oxidation with antimicrobial action. Infect Immun 1978; 20:456–463.
  • 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.
  • Lenander-Lumikari M, Tenovuo J, Emilson CG, Vilja P. Viability of Streptococcus mutans and Streptococcus sobrinus in whole saliva with varying concentrations of indigenous antimicrobial agents. Caries Res 1992; 26:371–378.
  • Klebanoff SJ, Clem WH, Luebke RG. Peroxidase-thiocyanate-hydrogen peroxide antimicrobial system. Biochim Biophys Acta 1966; 117:63–72.
  • Oram JD, Reiter B. The inhibition of Streptococci by lacteroperoxidase, thiocyanate and hydrogen peroxide—the oxidation of thiocyanate and the nature of the inhibitory compound. Biochem J 1966; 100:382–388.
  • Thomas EL, Pera KA, Smith KW, Chwang AK. Inhibition of Streptococcus mutans by the lactoperoxidase antimicrobial system. Infect Immun 1983; 39:767–778.
  • Carlsson J, Edlund MB, Hanstrom L. Bactericidal and cytotoxic effects of hypothiocyanite-hydrogen peroxide mixtures. Infect Immun 1984; 44:581–586.
  • Lumikari M, Soukka T, Nurmio S, Tenovuo J. Inhibition of the growth of Streptococcus mutans, Streptococcus sobrinus and Lactobacillus casei by oral peroxidase systems in human saliva. Arch Oral Biol 1991; 36:155–160.
  • Wolfson LM, Sumner SS. Antibacterial activity of the lactoperoxidase system—a review. J Food Protect 1993; 56:887–892.
  • Hogg DM, Jago GR. The antibacterial action of lactoperoxidase: the nature of the bacterial inhibitor. Biochem J 1970; 117:779–790.
  • 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.
  • 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.
  • Pruitt KM, Reiter B. Biochemistry of peroxidase system: antimicrobial effects. KM Pruitt, J Tenovuo. The lactoperoxidase system: Chemistry and biological significance. New York: Marcel Dekker; 1985; 143–178.
  • 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.
  • Thomas EL, Milligan TW, Joyner RE, Jefferson MM. Antibacterial activity of hydrogen peroxide and the lactoperoxidase-hydrogen peroxide-thiocyanate system against oral streptococci. Infect Immun 1994; 62:529–535.
  • Adamson M, Pruitt K. Lactoperoxidase-catalysed inactivation of hexokinase. Biochim Biophys Acta 1981; 658:238–247.
  • 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.
  • Mahmud SA, Wang JG, Slungaard A. Thiocyanate-dependent inhibition of spontaneous and agonist-induced eosinophil apoptosis by eosinophil peroxidase (EPO): a potential physiologic role for endogenously generated HOSCN in maintaining eosinophil viability. Blood 2006; 108:466A.
  • Yang L, Froio RM, Sciuto TE, Dvorak AM, Alon R, Luscinskas FW. ICAM-1 regulates neutrophil adhesion and transcellular migration of TNF-alpha activated endothelium under flow. Blood 2005; 106:584–592.
  • Lenander-Lumikari M. Inhibition of Candida-Albicans by the peroxidase/SCN−/H2O2 system. Oral Microbiol Immunol 1992; 7:315–320.
  • 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.
  • Thomas EL, Bates KP, Jefferson MM. Hypothiocyanite ion: detection of the antimicrobial agent in human saliva. J Dent Res 1980; 59:1466–1472.
  • 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.
  • 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.
  • 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.
  • Lenander-Lumikari M, Tenovuo J, Mikola H. Effects of a lactoperoxidase system-containing toothpaste on levels of hypothiocyanite and bacteria in saliva. Caries Res 1993; 27:285–291.
  • 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.
  • Courtois PH, Pourtois M. Purification of NADH: hypothiocyanite oxidoreductase in Streptococcus sanguis. Biochem Mol Med 1996; 57:134–138.
  • Pourtois M, Binet C, Van Tieghem N, Courtois P, Vandenabbeele A, Thiry L. Inhibition of HIV infectivity by lactoperoxidase-produced hypothiocyanite. J Biol Buccale 1990; 18:251–253.
  • Courtois P, van Beers D, de Foor M, Mandelbaum IM, Pourtois M. Abolition of herpes simplex cytopathic effect after treatment with peroxidase generated hypothiocyanite. J Biol Buccale 1990; 18:71–74.
  • Valimaa H, Waris M, Hukkanen V, Blankenvoorde MFJ, Amerongen AVN, Tenovuo J. Salivary defense factors in herpes simplex virus infection. J Dent Res 2002; 81:416–421.
  • Fitzsimmons N, Berry DR. Inhibition of Candida-Albicans by Lactobacillus-Acidophilus—evidence for the involvement of a peroxidase system. Microbios 1994; 80:125–133.
  • 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.
  • Kamau DN, Doores S, Pruitt KM. Activation of the lactoperoxidase system prior to pasteurisation for shelf-life extension of milk. Milchwissenschaft 1991; 46:213–214.
  • Kamau DN, Doores S, Pruitt KM. Enhanced thermal destruction of Listeria monocytogenes and Staphylococcus aureus by the lactoperoxidase system. Appl Environ Microbiol 1990; 56:2711–2716.
  • Christensen TG. The distribution and function of peroxidases in the respiratory tract. Surv Synth Pathol Res 1984; 3:201–218.
  • Ratner AJ, Prince A. Lactoperoxidase—new recognition of an ‘old’ enzyme in airway defenses. Am J Respir Cell Mol Biol 2000; 22:642–644.
  • Gerson C, Sabater J, Scuri M, Torbati A, Coffey R, Abraham JW, Lauredo I, Forteza R, Wanner A, Salathe M, Abraham WM, Conner GE. The lactoperoxidase system functions in bacterial clearance of airways. Am J Respir Cell Mol Biol 2000; 22:665–671.
  • Conner GE, Salathe M, Forteza R. Lactoperoxidase and hydrogen peroxide metabolism in the airway. Am J Respir Crit Care Med 2002; 166:S57–S61.
  • Wijkstrom-Frei C, El-Chemaly S, Ali-Rachedi R, Gerson C, Cobas MA, Forteza R, Salathe M, Conner GE. Lactoperoxidase and human airway host defense. Am J Respir Cell Mol Biol 2003; 29:206–212.
  • 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.
  • Pedemonte N, Caci E, Sondo E, Caputo A, Rhoden K, Pfeffer U, Di Candia M, Bandettini R, Ravazzolo R, Zegarra-Moran O, Galietta LJV. Thiocyanate transport in resting and IL-4-stimulated human bronchial epithelial cells: Role of pendrin and anion channels. J Immunol 2007; 178:5144–5153.
  • Forteza R, Salathe M, Miot F, Forteza R, Conner GE. Regulated hydrogen peroxide production by Duox in human airway epithelial cells. Am J Respir Cell Mol Biol 2005; 32:462–469.
  • 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.
  • Moskwa P, Lorentzen D, Excoffon K, Zabner J, McCray PB, Nauseef WM, Dupuy C, Banfi B. A novel host defense system of airways is defective in cystic fibrosis. Am J Respir Crit Care Med 2007; 175:174–183.
  • Scanlon CE, Berger B, Malcom G, Wissler RW. Evidence for more extensive deposits of epitopes of oxidized low density lipoprotein 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. PDAY Research Group. Pathobiological Determinants of Atherosclerosis in Youth. Atherosclerosis 1996; 124:191–202.

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.