368
Views
11
CrossRef citations to date
0
Altmetric
Review

Potential roles of myeloperoxidase and hypochlorous acid in metabolism and toxicity of alkene hydrocarbons and drug molecules containing olefinic moieties

&
Pages 513-524 | Received 04 Oct 2016, Accepted 08 Dec 2016, Published online: 16 Dec 2016

References

  • Kalgutkar AS, Soglia JR. Minimising the potential for metabolic activation in drug discovery. Expert Opin Drug Metab Toxicol. 2005;1:91–142.
  • Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA. 1998;279:1200–1205.
  • Thompson RA, Isin EM, Ogese MO, et al. Reactive metabolites: current and emerging risk and hazard assessments. Chem Res Toxicol. 2016;29:505–533.
  • Stepan AF, Walker DP, Bauman J, et al. Structural alert/reactive metabolite concept as applied in medicinal chemistry to mitigate the risk of idiosyncratic drug toxicity: a perspective based on the critical examination of trends in the top 200 drugs marketed in the United States. Chem Res Toxicol. 2011;24:1345–1410.
  • Kalgutkar AS, Gardner I, Obach RS, et al. A comprehensive listing of bioactivation pathways of organic functional groups. Curr Drug Metab. 2005;6:161–225.
  • Obach RS, Kalgutkar AS, Ryder TF, et al. In vitro metabolism and covalent binding of enol-carboxamide derivatives and anti-inflammatory agents sudoxicam and meloxicam: insights into the hepatotoxicity of sudoxicam. Chem Res Toxicol. 2008;21:1890–1899.
  • Kalgutkar AS, Henne KR, Lame ME, et al. Metabolic activation of the nontricyclic antidepressant trazodone to electrophilic quinone-imine and epoxide intermediates in human liver microsomes and recombinant P4503A4. Chem Biol Interact. 2005;155:10–20.
  • Niwa T, Murayama N, Yamazaki H. Oxidation of endobiotics mediated by xenobiotic-metabolizing forms of human cytochrome P450. Curr Drug Metab. 2009;10:700–712.
  • Srivastava A, Lian L-Y, Maggs JL, et al. Quantifying the metabolic activation of nevirapine in patients by integrated applications of NMR and mass spectrometries. Drug Metab Dispos. 2010;38:122–132.
  • Harvey DJ, Glazener L, Johnson DB, et al. Comparative metabolism of four allylic barbiturates and hexobarbital by the rat and guinea pig. Drug Metab Dispos. 1977;5:527–546.
  • Peterson LA. Reactive metabolites in the biotransformation of molecules containing a furan ring. Chem Res Toxicol. 2013;26:6–25.
  • Gramec D, Peterlin Mašič L, Sollner Dolenc M. Bioactivation potential of thiophene-containing drugs. Chem Res Toxicol. 2014;27:1344–1358.
  • Tafazoli S, O’Brien PJ. Peroxidases: a role in the metabolism and side effects of drugs. Drug Discov Today. 2005;10:617–625.
  • IARC. Ethylene. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol. 60, Some Industrial Chemicals. Lyon, France: 1994.
  • IARC. 1,3-Butadiene. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol. 97, 1,3-Butadiene, ethylene oxide and vinyl halides (vinyl fluoride, vinyl chloride and vinyl bromide). Lyon, France: 2008.
  • IARC. Styrene. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, Vol. 82, Some Traditional Herbal Medicines, Some Mycotoxins, Naphthalene and Styrene. Lyon, France: 2002.
  • Duescher RJ, Elfarra AA. 1,3-Butadiene oxidation by human myeloperoxidase. J Biol Chem. 1992;267:19859–19865.
  • Duescher RJ, Elfarra AA. Chlorohydrins are novel metabolites of 1,3-butadiene, styrene, and cyclohexene formed by human myeloperoxidase (MPO). Toxicologist. 1992;12:63.
  • Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 2006;6:173–182.
  • Klebanoff SJ, Kettle AJ, Rosen H, et al. Myeloperoxidase: a front-line defender against phagocytosed microorganisms. J Leukoc Biol. 2013;93:185–198.
  • Malle E, Furtmüller PG, Sattler W, et al. Myeloperoxidase: a target for new drug development? Br J Pharmacol. 2007;152:838–854.
  • Fiedler TJ, Davey CA, Fenna RE. X-ray crystal structure and characterization of halide-binding sites of human myeloperoxidase at 1.8 Å resolution. J Biol Chem. 2000;275:11964–11971.
  • Winterbourn CC, Kettle AJ. Redox reactions and microbial killing in the neutrophil phagosome. Antioxid Redox Signal. 2013;18:642–660.
  • Hori H, Fenna RE, Kimura S, et al. Aromatic substrate molecules bind at the distal heme pocket of myeloperoxidase. J Biol Chem. 1994;269:8388–8392.
  • Van Der Veen BS, De Winther MP, Heeringa P. Myeloperoxidase: molecular mechanisms of action and their relevance to human health and disease. Antioxid Redox Signal. 2009;11:2899–2937.
  • Wang G, Nauseef WM. Salt, chloride, bleach, and innate host defense. J Leukoc Biol. 2015;98:163–172.
  • Nussbaum C, Klinke A, Adam M, et al. Myeloperoxidase: a leukocyte-derived protagonist of inflammation and cardiovascular disease. Antioxid Redox Signal. 2013;18:692–713.
  • Gullberg U, Bengtsson N, Bülow E, et al. Processing and targeting of granule proteins in human neutrophils. J Immunol Methods. 1999;232:201–210.
  • Hansson M, Olsson I, Nauseef WM. Biosynthesis, processing, and sorting of human myeloperoxidase. Arch Biochem Biophys. 2006;445:214–224.
  • Dang Y, Lowe GM, Edwards SW, et al. The effects of GM-CSF on myeloperoxidase release in normal and myelodysplastic neutrophils. Leuk Res. 1993;17:1037–1044.
  • Holle JU, Windmöller M, Lange C, et al. Toll-like receptor TLR2 and TLR9 ligation triggers neutrophil activation in granulomatosis with polyangiitis. Rheumatology. 2013;52:1183–1189.
  • Chatham WW, Turkiewicz A, Blackburn WD Jr. Determinants of neutrophil HOCl generation: ligand-dependent responses and the role of surface adhesion. J Leukoc Biol. 1994;56:654–660.
  • Odobasic D, Kitching AR, Yang Y, et al. Neutrophil myeloperoxidase regulates T-cell-driven tissue inflammation in mice by inhibiting dendritic cell function. Blood. 2013;121:4195–4204.
  • Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–1535.
  • Brinkmann V, Zychlinsky A. Beneficial suicide: why neutrophils die to make NETs. Nat Rev Microbiol. 2007;5:577–582.
  • Parker H, Albrett AM, Kettle AJ, et al. Myeloperoxidase associated with neutrophil extracellular traps is active and mediates bacterial killing in the presence of hydrogen peroxide. J Leukoc Biol. 2012;91:369–376.
  • Aiken ML, Painter RG, Zhou Y, et al. Chloride transport in functionally active phagosomes isolated from human neutrophils. Free Radic Biol Med. 2012;53:2308–2317.
  • Painter RG, Marrero L, Lombard GA, et al. CFTR-mediated halide transport in phagosomes of human neutrophils. J Leuk Biol. 2010;87:933–942.
  • Winterbourn CC, Hampton MB, Livesey JH, et al. Modeling the reactions of superoxide and myeloperoxidase in the neutrophil phagosome: implications for microbial killing. J Biol Chem. 2006;281:39860–39869.
  • Hawkins CL, Pattison DI, Davies MJ. Hypochlorite-induced oxidation of amino acids, peptides and proteins. Amino Acids. 2003;25:259–274.
  • Panasenko OM, Gorudko IV, Sokolov AV. Hypochlorous acid as a precursor of free radicals in living systems. Biochemistry (Mosc). 2013;78:1466–1489.
  • Malle E, Marsche G, Arnhold J, et al. Modification of low-density lipoprotein by myeloperoxidase-derived oxidants and reagent hypochlorous acid. Biochim Biophys Acta. 2006;1761:392–415.
  • Stanley NR, Pattison DI, Hawkins CL. Ability of hypochlorous acid and N-chloramines to chlorinate DNA and its constituents. Chem Res Toxicol. 2010;23:1293–1302.
  • Storkey C, Davies MJ, Pattison DI. Reevaluation of the rate constants for the reaction of hypochlorous acid (HOCl) with cysteine, methionine, and peptide derivatives using a new competition kinetic approach. Free Radic Biol Med. 2014;73:60–66.
  • Hawkins CL, Davies MJ. Reaction of HOCl with amino acids and peptides: EPR evidence for rapid rearrangement and fragmentation reactions of nitrogen-centred radicals. J Chem Soc Perkin Trans. 1998;2:1937–1945.
  • 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.
  • Jeitner TM, Kalogiannis M, Krasnikov BF, et al. Linking inflammation and Parkinson disease: hypochlorous acid generates parkinsonian poisons. Toxicol Sci. 2016;151:388–402.
  • Elfarra AA, Duescher RJ, Pasch CM. Mechanisms of 1,3-butadiene oxidations to butadiene monoxide and crotonaldehyde by mouse liver microsomes and chloroperoxidase. Arch Biochem Biophys. 1991;286:244–251.
  • Elfarra AA, Krause RJ, Selzer RR. Biochemistry of 1,3-butadiene metabolism and its relevance to 1,3-butadiene-induced carcinogenicity. Toxicology. 1996;113:23–30.
  • Hrycay EG, Bandiera SM. Monooxygenase, peroxidase and peroxygenase properties and reaction mechanisms of cytochrome P450 enzymes. Adv Exp Med Biol. 2015;851:1–61.
  • Davies MJ, Hawkins CL, Pattison DI, et al. Mammalian heme peroxidases: from molecular mechanisms to health implications. Antioxid Redox Signal. 2008;10:1199–1234.
  • Forbes LV, Sjögren T, Auchère F, et al. Potent reversible inhibition of myeloperoxidase by aromatic hydroxamates. J Biol Chem. 2013;288:36636–36647.
  • Arnhold J, Osipov AN, Spalteholz H, et al. Effects of hypochlorous acid on unsaturated phosphatidylcholines. Free Radic Biol Med. 2001;31:1111–1119.
  • Carr AC, van den Berg JJ, Winterbourn CC. Differential reactivities of hypochlorous and hypobromous acids with purified Escherichia coli phospholipid: formation of haloamines and halohydrins. Biochim Biophys Acta. 1998;1392:254–264.
  • Carr AC, van den Berg JJ, Winterbourn CC. Chlorination of cholesterol in cell membranes by hypochlorous acid. Arch Biochem Biophys. 1996;332:63–69.
  • Winterbourn CC, van den Berg JJ, Roitman E, et al. Chlorohydrin formation from unsaturated fatty acids reacted with hypochlorous acid. Arch Biochem Biophys. 1992;296:547–555.
  • O’Brien PJ. Peroxidases. Chem Biol Interact. 2000;129:113–139.
  • Brown AM, Skamarauskas J, Lister T, et al. Differential susceptibility of astrocytic and neuronal function to 3-chloropropanediol in the rat inferior colliculus. J Neurochem. 2011;116:996–1004.
  • 3-Chloro-1,2-Propandiol. WHO Food Add. Ser. 48. Geneva: WHO Available from: www.inchem.org/documents/jecfa/jecmono/v48je18.htm [cited 2016 Oct 4].
  • Zeiger E, Anderson B, Haworth S, et al. Salmonella mutagenicity tests: IV. Results from the testing of 300 chemicals. Environ Mol Mutagen. 1988;11(Suppl. 12):1–157.
  • Piasecki A, Ruge A, Marquardt H. Malignant transformation of mouse M2-fibroblasts by glycerol chlorohydrines contained in protein hydrolysates and commercial food. Arzneimittelforschung. 1990;40:1054–1055.
  • IARC. 1,3-Dichloro-2-propanol, 3-monochloro-1,2-propanediol. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol. 101, Some Chemicals Present in Industrial and Consumer Products, Food and Drinking-Water. Lyon, France: 2013.
  • Greenberg HL, Ott MG, Shore RE. Men assigned to ethylene oxide production or other ethylene oxide related chemical manufacturing: a mortality study. Br J Ind Med. 1990;47:221–230.
  • Liu X-J, Zeng F-M, An J, et al. Cytotoxicity, genotoxicity, and mutagenicity of 1-chloro-2-hydroxy-3-butene and 1-chloro-3-buten-2-one, two alternative metabolites of 1,3-butadiene. Toxicol Appl Pharmacol. 2013;271:13–19.
  • Waidyanatha S, Gaudette NF, Hong Y, et al. Formation of epichlorohydrin, a known rodent carcinogen, following oral administration of 1,3-dichloro-2-propanol in rats. Chem Res Toxicol. 2014;27:1787–1795.
  • Elfarra AA, Zhang X-Y. Alcohol dehydrogenase- and rat liver cytosol-dependent bioactivation of 1-chloro-2-hydroxy-3-butene to 1-chloro-3-buten-2-one, a bifunctional alkylating agent. Chem Res Toxicol. 2012;25:2600–2607.
  • Irving RM, Elfarra AA. Role of reactive metabolites in the circulation in extrahepatic toxicity. Expert Opin Drug Metab Toxicol. 2012;8:1157–1172.
  • Duescher RJ, Elfarra AA. Human liver microsomes are efficient catalysts of 1,3-butadiene oxidation: evidence for major roles by cytochromes P450 2A6 and 2E1. Arch Biochem Biophys. 1994;311:342–349.
  • Bernauer U, Vieth B, Ellrich R, et al. CYP2E1-dependent benzene toxicity: the role of extrahepatic benzene metabolism. Arch Toxicol. 1999;73:189–196.
  • Bernauer U, Vieth B, Ellrich R, et al. CYP2E1 expression in bone marrow and its intra- and interspecies variability: approaches for a more reliable extrapolation from one species to another in the risk assessment of chemicals. Arch Toxicol. 2000;73:618–624.
  • Hodges VM, Molloy GY, Wickramasinghe SN. Demonstration of mRNA for five species of cytochrome P450 in human bone marrow, bone marrow-derived macrophages and human haemopoietic cell lines. Br J Haematol. 2000;108:151–156.
  • Koppikar AM, Wu C, Leavens T, et al. Health assessment of 1,3-Butadiene, EPA/600/P-98/001F. Washington, DC: U.S. Environmental Protection Agency, Office of Research and Development, National Center for Environmental Assessment-Washington Office, U.S. Government Printing Office; 2002. Available from: http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=54499 [cited 2016 Oct 4].
  • Sathiakumar N, Delzell E. A follow-up study of mortality among women in the North American synthetic rubber industry. J Occup Environ Med. 2009;51:1314–1325.
  • Sathiakumar N, Brill I, Delzell E. 1,3-Butadiene, styrene and lung cancer among synthetic rubber industry workers. J Occup Environ Med. 2009;51:1326–1332.
  • Eschner MS, Selmani I, Groger TM, et al. Online comprehensive two-dimensional characterization of puff-by-puff resolved cigarette smoke by hyphenation of fast gas chromatography to single-photon ionization time-of-flight mass spectrometry: quantification of hazardous volatile organic compounds. Anal Chem. 2011;83:6619–6627.
  • Sathiakumar N, Brill I, Leader M, et al. 1,3-Butadiene, styrene and lymphohematopoietic cancer among male synthetic rubber industry workers - preliminary exposure-response analyses. Chem Biol Interact. 2015;241:40–49.
  • Heck JE, Park AS, Qiu J, et al. Risk of leukemia in relation to exposure to ambient air toxics in pregnancy and early childhood. Int J Hyg Environ Health. 2014;217:662–668.
  • 1,3-Butadiene, CASRN 106-99-0. Integrated risk information system. Washington, DC: U.S. Environmental Protection Agency; 2002. Available from: cfpub.epa.gov/ncea/iris/iris_documents/documents/subst/0139_summary.pdf [cited 2016 Oct 4].
  • Benzene, CASRN 71-43-2. Integrated risk information system. Washington, DC: U.S. Environmental Protection Agency; 2003. Available from: cfpub.epa.gov/ncea/iris/iris_documents/documents/subst/0276_summary.pdf [cited 2016 Oct 4].
  • McCarthy MC, O’Brien TE, Charrier JG, et al. Characterization of the chronic risk and hazard of hazardous air pollutants in the United States using ambient monitoring data. Environ Health Perspect. 2009;117:790–796.
  • Zhou J, You Y, Bai Z, et al. Health risk assessment of personal inhalation exposure to volatile organic compounds in Tianjin, China. Sci Total Environ. 2011;409:452–459.
  • Mo Z, Shao M, Lu S, et al. Process-specific emission characteristics of volatile organic compounds (VOCs) from petrochemical facilities in the yangtze river delta, China. Sci Total Environ. 2015;533:422–431.
  • Du Z, Mo J, Zhang Y. Risk assessment of population inhalation exposure to volatile organic compounds and carbonyls in urban China. Environ Int. 2014;73:33–45.
  • Loh MM, Levy JI, Spengler JD, et al. Ranking cancer risks of organic hazardous air pollutants in the United States. Environ Health Perspect. 2007;115:1160–1168.
  • Yacovitch TI, Herndon SC, Roscioli JR, et al. Air pollutant mapping with a mobile laboratory during the BEE-TEX field study. Environ Health Insights. 2015;9(S4):7–13.
  • Technology Transfer Network - Air Toxics Web Site. Original list of hazardous air pollutants. Washington, DC: U.S. Environmental Protection Agency Available from: www.epa.gov/ttn/atw/188polls.html [cited 2016 Oct 4].
  • Fowles J, Dybing E. Application of toxicological risk assessment principles to the chemical constituents of cigarette smoke. Tob Control. 2003;12:424–430.
  • Sleiman M, Logue JM, Luo W, et al. Inhalable constituents of thirdhand tobacco smoke: chemical characterization and health impact considerations. Environ Sci Technol. 2014;48:13093–13101.
  • Kirman CR, Albertini RJ, Sweeney LM, et al. 1,3-Butadiene: I. Review of metabolism and the implications to human health risk assessment. Crit Rev Toxicol. 2010;40(Suppl. 1):1–11.
  • Albertini RJ, Carson ML, Kirman CR, et al. 1,3-Butadiene: II. Genotoxicity profile. Crit Rev Toxicol. 2010;40(Suppl. 1):12–73.
  • Kohn MC, Melnick RL. Species differences in the production and clearance of 1,3-butadiene metabolites: a mechanistic model indicates predominantly physiological, not biochemical, control. Carcinogenesis. 1993;14:619–628.
  • Sweeney LM, Schlosser PM, Medinsky MA, et al. Physiologically based pharmacokinetic modeling of 1,3-butadiene, 1,2-epoxy-3-butene, and 1, 2:3,4-diepoxybutanetoxicokinetics in mice and rats. Carcinogenesis. 1997;18:611–625.
  • Sun L, Pelah A, D-P Z, et al. Formation of fused-ring 2ʹ-deoxycytidine adducts from 1-chloro-3-buten-2-one, an in vitro 1,3-butadiene metabolite, under in vitro physiological conditions. Chem Res Toxicol. 2013;26:1545–1553.
  • Zeng F-M, Liu L-Y, Zheng J, et al. Identification of a fused-ring 2ʹ-deoxyadenosine adduct formed in human cells incubated with 1-chloro-3-buten-2-one, a potential reactive metabolite of 1,3-butadiene. Chem Res Toxicol. 2016;29:1041–1050.
  • Zheng J, Li Y, Yu Y-X, et al. Novel adducts from the reaction of 1-chloro-3-buten-2-one with 2ʹ-deoxyguanosine. Structural characterization and potential as tools to investigate 1,3-butadiene carcinogenicity. Chem Biol Interact. 2015;226:40–48.
  • Liu L-Y, Zheng J, Kong C, et al. Characterization of the major purine and pyrimidine adducts formed after incubations of 1-chloro-3-buten-2-one with single-/double-stranded DNA and human cells. Chem Res Toxicol. 2016 cited 6 December 2016. DOI:10.1021/acs.chemrestox.6b00282
  • Maniglier-Poulet C, Cheng X, Ruth JA, et al. Metabolism of 1,3-butadiene to butadiene monoxide in mouse and human bone marrow cells. Chem Biol Interact. 1995;97:119–129.
  • Mayadas TN, Cullere X, Lowell CA. The multifaceted functions of neutrophils. Annu Rev Pathol. 2014;9:181–218.
  • Kotas ME, Medzhitov R. Homeostasis, inflammation, and disease susceptibility. Cell. 2015;160:816–827.
  • Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899.

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.