922
Views
0
CrossRef citations to date
0
Altmetric
Review Articles

Acrylonitrile’s genotoxicity profile: mutagenicity in search of an underlying molecular mechanism

ORCID Icon, &
Pages 69-116 | Received 27 Sep 2022, Accepted 08 Feb 2023, Published online: 06 Jun 2023

References

  • Abdel-Naim AB, Mohamadin AM. 2004. Myeloperoxidase-catalyzed oxidation of chloroacetonitrile to cyanide. Toxicol Lett. 146(3):249–257.
  • Abdel-Naim AB, Hamada F, Abdel Aziz AH, Ahmed AE. 1994. Acrylonitrile (VCN)-induced testicular toxicity in the rat. Toxicologist. 14(1):268.
  • Abdel-Rahman SZ, Nouraldeen AM, Ahmed AE. 1994. Molecular interaction of [2,3-14C]acrylonitrile with DNA in gastric tissue of rat. J Biochem Toxicol. 9(4):191–198.
  • Abrahamson S, Würgler FE, DeJongh C, Meyer HU. 1980. How many loci on the X-chromosome of Drosophila melanogaster can mutate to recessive lethals? Environ Mutagen. 2(4):447–453.
  • Adewoye AB, Lindsay SJ, Dubrova YE, Hurles ME. 2015. The genome-wide effects of ionizing radiation on mutation induction in the mammalian germline. Nat Commun. 6:6684.
  • Adler ID, Pacchierotti F, Russo A. 2012. The measurement of induced genetic change in mammalian germ cells. Methods Mol Biol. 817:335–375.
  • Ahmed AE, Farooqui YH, Upreti RK, El-Shabrawy O. 1982. Distribution and covalent interactions of [1-14C]acrylonitrile in the rat. Toxicology. 23(2–3):159–175.
  • Ahmed AE, Abdel-Aziz AH, Abdel-Rahman SZ, Haque AK, Nouraldeen AM, Shouman SA. 1992. Pulmonary toxicity of acrylonitrile: covalent interaction and effect on replicative and unscheduled DNA synthesis in the lung. Toxicology. 76(1):1–14.
  • Ahmed AE, Abdel-Rahman SZ, Nour-Al Deen AM. 1992. Acrylonitrile interaction with testicular DNA in rats. J Biochem Toxicol. 7(1):5–11.
  • Ahmed AE, Nouraldeen AM, Abdel-Rahman SZ, Rajaraman S. 1996. Role of glutathione modulation in acrylonitrile induced gastric DNA damage in rats. Arch Toxicol. 70(10):620–627.
  • Al-Abbasi FA, Esmat A, Mohamadin AM, Abdel-Naim AB. 2018. Report-role of prostaglandin H synthase in activation of acrylonitrile to cyanide. Pak J Pharm Sci. 31(4):1431–1435.
  • Al-Abbasi FA. 2012. Acrylonitrile-induced gastric toxicity in rats: the role of xanthine oxidase. Med Sci Monit. 18(6):BR208–14.
  • Albertini RJ, Kaden DA. 2020. Mutagenicity monitoring in humans: global versus specific origin of mutations. Mutat Res Rev Mutat Res. 786:108341.
  • Albertini RJ. 2001. Validated biomarker responses influence medical surveillance of individuals exposed to genotoxic agents. Radiat Prot Dosimetry. 97(1):47–54.
  • Alexander P. 1961. Mouse lymphoma cells with different radiosensitivities. Nature. 192:572–573.
  • Allen JW, Liang JC, Carrano AV, Preston RJ. 1986. Review of literature on chemical-induced aneuploidy in mammalian male germ cells. Mutat Res. 167(1–2):123–137.
  • Amacher E, Turner G. 1985. Tests for gene mutational activity in the L5158Y/TK assay system. Prog Mutat Res. 5:487–496.
  • Ames BN, Lee FD, Durston WE. 1973. An improved bacterial test system for the detection and classification of mutagens and carcinogens. Proc Natl Acad Sci U S A. 70(3):782–786.
  • Anderson D, Cross MF. 1985. Suitability of the P388F mouse lymphoma system for detecting potential carcinogens and mutagens. Food Chem Toxicol. 23(1):115–118.
  • Ansell M, Lewis FA. 1970. A review of cyanide concentrations found in human organs. A survey of literature concerning cyanide metabolism, 'normal’, non-fatal, and fatal body cyanide levels. J Forensic Med. 17(4):148–155.
  • Antoine JL, Arany J, Léonard A, Henrotte J, Jenar-Dubuisson G, Decat G. 1983. Lack of mutagenic activity of dimethylformamide. Toxicology. 26(3–4):207–212.
  • Arlandson M, Decker T, Roongta VA, Bonilla L, Mayo KH, MacPherson JC, Hazen SL, Slungaard A. 2001. Eosinophil peroxidase oxidation of thiocyanate. Characterization of major reaction products and a potential sulfhydryl-targeted cytotoxicity system. J Biol Chem. 276(1):215–224.
  • Arni P. 1985. Induction of various genetic effects in the yeast Saccharomvces cerevisiae strain D7. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 217–224.
  • Asakura M, Noguchi T, Sugiyama Y, Inoue M, Satake H. 1994. Research study on evaluation of mutagenicity for existing chemical substances. Research study on industrial safety and health in 1994-3. In vitro chromosomal aberration test on cultured mammalian cells. Tokyo: Testing Laboratory: Japan Industrial Safety and Health Association: Japan Bioassay Research Center. Owner company: Japan Industrial Safety and Health Association.
  • Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 1–752.
  • Singer B, Grunberger D. 1983. Molecular biology of mutagens and carcinogens. New York (NY): Springer.
  • Baker RU, Bonin AM. 1985. Tests with the Salmonella plate incorporation assay. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 177–180.
  • Barrett TJ, Hawkins CL. 2012. Hypothiocyanous acid: benign or deadly? Chem Res Toxicol. 25(2):263–273.
  • Barrett TJ, Pattison DI, Leonard SE, Carroll KS, Davies MJ, Hawkins CL. 2012. Inactivation of thiol-dependent enzymes by hypothiocyanous acid: role of sulfenyl thiocyanate and sulfenic acid intermediates. Free Radic Biol Med. 52(6):1075–1085.
  • Beal MA, Meier MJ, Williams A, Rowan-Carroll A, Gagné R, Lindsay SJ, Fitzgerald T, Hurles ME, Marchetti F, Yauk CL. 2019. Paternal exposure to benzo(a)pyrene induces genome-wide mutations in mouse offspring. Commun Biol. 2:228.
  • Benesh V, Shram R. 1969. Mutagenic activity of some pesticides in Drosophila melanogaster. Ind Med. 38:442–444.
  • Benz FW, Nerland DE, Li J, Corbett D. 1997. Dose dependence of covalent binding of acrylonitrile to tissue protein and globin in rats. Fundam Appl Toxicol. 36(2):149–156.
  • Beskid O, Dusek Z, Solanský I, Srám RJ. 2006. The effects of exposure to different clastogens on the pattern of chromosomal aberrations detected by FISH whole chromosome painting in occupationally exposed individuals. Mutat Res. 594(1–2):20–29.
  • Bigner DD, Bigner SH, Burger PC, Shelburne JD, Friedman HS. 1986. Primary brain tumours in F344 rats chronically exposed to acrylonitrile in their drinking water. Food Chem Toxicol. 24(2):129–137.
  • Bjorge C, Brunborg G, Wiger R, Holme JA, Scholz T, Dybing E, Søderlund EJ. 1996. A comparative study of chemically induced DNA damage in isolated human and rat testicular cells. Reprod Toxicol. 10(6):509–519.
  • Bonassi S, Lando C, Ceppi M, Landi S, Rossi AM, Barale R. 2004. No association between increased levels of high-frequency sister chromatid exchange cells (HFCs) and the risk of cancer in healthy individuals. Environ Mol Mutagen. 43(2):134–136.
  • Borba H, Monteiro M, Proença MJ, Chaveca T, Pereira V, Lynce N, Rueff J. 1996. Evaluation of some biomonitoring markers in occupationally exposed populations to acrylonitrile. Teratog Carcinog Mutagen. 16(4):205–218.
  • Bouzyk E, Gradzka I, Iwaneńko T, Kruszewski M, Sochanowicz B, Szumiel I. 2000. The response of L5178Y lymphoma sublines to oxidative stress: antioxidant defence, iron content and nuclear translocation of the p65 subunit of NF-kappaB. Acta Biochim Pol. 47(4):881–888.
  • Boysen G, Collins LB, Liao S, Luke AM, Pachkowski BF, Watters JL, Swenberg JA. 2010. Analysis of 8-oxo-7,8-dihydro-2'-deoxyguanosine by ultra high pressure liquid chromatography-heat assisted electrospray ionization-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 878(3–4):375–380.
  • Boysen G, Pachkowski BF, Nakamura J, Swenberg JA. 2009. The formation and biological significance of N7-guanine adducts. Mutat Res. 678(2):76–94.
  • Bradley MO. 1985. Measurement of DNA single-strand breaks by alkaline elution in rat hepatocytes. Prog Mutat Res. 5:353–357.
  • Brams A, Buchet JP, Crutzen-Fayt MC, De Meester C, Lauwerys R, Léonard A. 1987. A comparative study, with 40 chemicals, of the efficiency of the Salmonella assay and the SOS chromotest (kit procedure). Toxicol Lett. 38(1–2):123–133.
  • Brennan RJ, Swoboda BE, Schiestl RH. 1994. Oxidative mutagens induce intrachromosomal recombination in yeast. Mutat Res. 308(2):159–167.
  • Brooks TM, Gonzalez LP, Calvert R, Parry JM. 1985. The induction of mitotic gene conversion in the yeast Saccharomyces cerevisiae strain JD1. Prog Mutat Res. 5:225–228.
  • Burka LT, Sanchez IM, Ahmed AE, Ghanayem BI. 1994. Comparative metabolism and disposition of acrylonitrile and methacrylonitrile in rats. Arch Toxicol. 68(10):611–618.
  • Burlinson B, Tice RR, Speit G, Agurell E, Brendler-Schwaab SY, Collins AR, Escobar P, Honma M, Kumaravel TS, Nakajima M, et al. 2007. Fourth International workgroup on genotoxicity testing: results of the in vivo comet assay workgroup. Mutat Res. 627(1):31–35.
  • Butterworth BE, Ashby J, Bermudez E, Casciano D, Mirsalis J, Probst G, Williams G. 1987. A protocol and guide for the in vitro rat hepatocyte DNA-repair assay. Mutat Res. 189(2):113–121.
  • Butterworth BE, Eldridge SR, Sprankle CS, Working PK, Bentley KS, Hurtt ME. 1992. Tissue-specific genotoxic effects of acrylamide and acrylonitrile. Environ Mol Mutagen. 20(3):148–155.
  • Caito S, Park M, Aschner M. 2017. Resistance of mouse primary microglia and astrocytes to acrylonitrile-induced oxidative stress. Neurotoxicology. 63:120–125.
  • Caito S, Yu Y, Aschner M. 2013. Differential response to acrylonitrile toxicity in rat primary astrocytes and microglia. Neurotoxicology. 37:93–99.
  • Caito SW, Yu Y, Aschner M. 2014. Differential inflammatory response to acrylonitrile in rat primary astrocytes and microglia. Neurotoxicology. 42:1–7.
  • Carere A, Conti G, Conti L, Crebelli R. 1985. Assays in Aspergillus nidulans for the induction of forward-mutation in haploid strain 35 and for mitotic nondisjunction, haploidization and crossing-over in diploid strain P1. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers.
  • Carls N, Schiestl RH. 1994. Evaluation of the yeast DEL assay with 10 compounds selected by the International Program on Chemical Safety for the evaluation of short-term tests for carcinogens. Mutat Res. 320(4):293–303.
  • Cerna M, Kocisova J, Kodytkova J, Kopecky J, Sram RJ. 1981. Mutagenic activity of oxiranecarbonitrile (glycidonitrile). In: Gut I, Cikrt M, Plaa GL, editors. Industrial and environmental xenobiotics. Berlin Heidelberg New York: Springer; p. 251–254.
  • Chandler JD, Day BJ. 2015. Biochemical mechanisms and therapeutic potential of pseudohalide thiocyanate in human health. Free Radic Res. 49(6):695–710.
  • Chang CM, Hsia MT, Stoner GD, Hsu IC. 1990. Acrylonitrile-induced sister-chromatid exchanges and DNA single strand breaks in adult human bronchial epithelial cells. Mutat Res. 241(4):355–360.
  • Chinchilla D, Kilheeney H, Vitello LB, Erman JE. 2014. Kinetic and equilibrium studies of acrylonitrile binding to cytochrome c peroxidase and oxidation of acrylonitrile by cytochrome c peroxidase compound I. Biochem Biophys Res Commun. 443(1):200–204.
  • Cifone MA, Myhr B, Eiche A, Bolcsfoldi G. 1987. Effect of pH shifts on the mutant frequency at the thymidine kinase locus in mouse lymphoma L5178Y TK+/- cells. Mutat Res. 189(1):39–46.
  • Clark HM, Hagedorn TD, Landino LM. 2014. Hypothiocyanous acid oxidation of tubulin cysteines inhibits microtubule polymerization. Arch Biochem Biophys. 541:67–73.
  • Clark LS, Hart DW, Vojta PJ, Harrington-Brock K, Barrett JC, Moore MM, Tindall KR. 1998. Identification and chromosomal assignment of two heterozygous mutations in the Trp53 gene in L5178Y/Tk(+/-)-3.7.2C mouse lymphoma cells. Mutagenesis. 13(5):427–434.
  • Collins AR. 2009. Investigating oxidative DNA damage and its repair using the comet assay. Mutat Res. 681(1):24–32.
  • Collins JJ, Cheng R, Buck GM, Zhang J, Klebanoff M, Schisterman EF, Scheffers T, Ohta H, Takaya K, Miyauchi H, et al. 2003. The feasibility of conducting a reproductive outcome study of Chinese acrylonitrile worker. J Environ Occup Med. 1:29–32.
  • Connor TH, Meyne J, Molina L, Legator MS. 1979. A combined testing protocol approach for mutagenicity testing. Mutat Res. 64(1):19–26.
  • Cooke MS, Evans MD, Dizdaroglu M, Lunec J. 2003. Oxidative DNA damage: mechanisms, mutation, and disease. FASEB J. 17(10):1195–1214.
  • Cote IL, Bowers A, Jaeger RJ. 1984. Effects of acrylonitrile on tissue glutathione concentrations in rat, mouse, and hamster. Res Commun Chem Pathol Pharmacol. 43(3):507–510.
  • Crawley FE, Goddard EA. 1977. Internal dose from carbon-14 labelled compounds. The metabolism of carbon-14 labelled potassium cyanide in the rat. Health Phys. 32(3):135–142.
  • Crespi CL, Ryan CG, Seixas GM, Turner TR, Penman BW. 1985. Tests for mutagenic activity using mutation assays at two loci in the human lymphoblast cell lines YK6 and AHH-1. Prog Mutat Res. 5:497–516.
  • Dahl AR, Waruszewski BA. 1989. Metabolism of organonitriles to cyanide by rat nasal tissue enzymes. Xenobiotica. 19(11):1201–1205.
  • Danford N. 1985. Tests for chromosomal aberrations and aneuploidy in the Chinese hamster fibroblast cell line CH1-L. Prog Mutat Res. 5:397–411.
  • De Groef B, Decallonne BR, Van der Geyten S, Darras VM, Bouillon R. 2006. Perchlorate versus other environmental sodium/iodide symporter inhibitors: potential thyroid-related health effects. Eur J Endocrinol. 155(1):17–25.
  • De Jesús VR, Bhandari D, Zhang L, Reese C, Capella K, Tevis D, Zhu W, Del Valle-Pinero AY, Lagaud G, Chang JT, et al. 2020. Urinary biomarkers of exposure to volatile organic compounds from the population assessment of tobacco and health study wave 1 (2013-2014). Int J Environ Res Public Health. 17(15):5408.
  • De Jesús VR, Zhang L, Bhandari D, Zhu W, Chang JT, Blount BC. 2021. Characterization of acrylonitrile exposure in the United States based on urinary n-acetyl-S-(2-cyanoethyl)-L-cysteine (2CYEMA): NHANES 2011-2016. J Expo Sci Environ Epidemiol. 31(2):377–385.
  • de Meester C, Bogaert MD-V, Lambotte-Vandepaer M, Roberfroid M, Poncelet F, Mercier M. 1979. Liver extract mediated mutagenicity of acrylonitrile. Toxicology. 13(1):7–15.
  • de Meester C, Poncelet F, Roberfroid M, Mercier M. 1978. Mutagenicity of acrylonitrile. Toxicology. 11(1):19–27.
  • De PK, Roy A, Banerjee RK. 1987. Immunological characterization of soluble peroxidases from rat tissues including preputial gland. Mol Cell Biochem. 77(2):127–134.
  • De PK. 1992. Tissue distribution of constitutive and induced soluble peroxidase in rat. Purification and characterization from lacrimal gland. Eur J Biochem. 206(1):59–67.
  • Dellarco VL. 1993. Genetic anomalies in mammalian germ cells and their significance for human reproductive and developmental risk. Environ Health Perspect. 101(Suppl 2):5–11.
  • Ding S, Lai-Ji MA, Fan W, Zhu RJ, Ying Q, Zhou YL, Jin FS. 2003. Study on mitochondrial DNA damage in peripheral blood nucleate cells of the workers exposed to acrylonitrile. Chinese J Indust Hyg Occup Dis. 21:99–101. Chinese).
  • Dong D, Wang D, Ai X, Zhang H. 1996. Unpublished study of acrylonitrile hazardous effects on workers’ reproductive system (as reported in Collins et al., 2003).
  • Douglas GR, Blakey DH, Liu-Lee VW, Bell RDL, Bayley JM. 1985. Alkaline sucrose sedimentation, sister chromatid exchange and micronucleus assays in CHO cells. Prog Mutat Res. 5:359–366.
  • Duesberg P, Li R, Fabarius A, Hehlmann R. 2006. Aneuploidy and cancer: from correlation to causation. Contrib Microbiol. 13:16–44.
  • Duverger-Van Bogaert M, Lambotte-Vandepaer M, De Meester C, Mercier M, Poncelet F. 1982a. Role of glutathione in liver-mediated mutagenicity of acrylonitrile. Toxicol Lett. 11(3–4):305–311.
  • Duverger-Van Bogaert M, Lambotte-Vandepaer M, De Meester C, Mercier M, Poncelet F. 1982b. Vinyl chloride and acrylonitrile: activation mechanism and mutagenicity. Toxicol Eur Res. 4(1):35–37.
  • Duverger-Van Bogaert M, Lambotte-Vandepaer M, de Meester C, Rollmann B, Poncelet F, Mercier M. 1981. Effect of several factors on the liver extract mediated mutagenicity of acrylonitrile and identification of four new in vitro metabolites. Toxicol Lett. 7(4–5):311–319.
  • EC [European Commission]. 2004. European union risk assessment report for acrylonitrile. Final report. European Chemicals Bureau (Existing Substances, 1st Priority List Volume 32). European Commission, JRC, 2004.
  • EC [European Commission] RAC [Risk Assessment Committee]. 2018. ANNEX 1. Background document in support of the Committee for Risk Assessment (RAC) evaluation of limit values for acrylonitrile in the workplace. ECHA/RAC/O-0000001412-86-188/F. 9 March 2018.
  • ECHA REACH. 2022. Acrylonitrile Registration Dossier: https://echa.europa.eu/registration-dossier/-/registered-dossier/15561.
  • Evans HH, Ricanati M, Horng MF. 1987. Deficiency in DNA repair in mouse lymphoma strain L5178Y-S. Proc Natl Acad Sci U S A. 84(21):7562–7566.
  • Fahmy MA. 1999. Evaluation of the genotoxicity of acrylonitrile in different tissues of male mice. Cytologia. 64(1):1–9.
  • Fan W, Wang WL, Ding S, Zhou YL, Jin FS. 2006. Application of micronucleus test of buccal mucosal cells in assessing the genetic damage of workers exposed to acrylonitrile. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi. 24(2):106–108. [Chinese]
  • Farooqui MY, Mumtaz MM. 1991. Toxicology of methacrylonitrile. Toxicology. 65(3):239–250.
  • Farooqui MY, Ahmed AE. 1983. In vivo interactions of acrylonitrile with macromolecules in rats. Chem Biol Interact. 47(3):363–371.
  • Fennell TR, Kedderis GL, Sumner SCJ. 1991. Urinary metabolites of [1,2,3-13C]acrylonitrile in rats and mice detected by carbon-13 nuclear magnetic resonance spectroscopy. Chem Res Toxicol. 4(6):678–687.
  • Ferguson LR. 1985. Petite mutagenesis in Saccharomyces cerevisiae strain D5. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 229–234.
  • Florin I, Rutberg L, Curvall M, Enzell CR. 1980. Screening of tobacco smoke constituents for mutagenicity using the Ames’ test. Toxicology. 15(3):219–232.
  • Foureman P, Mason JM, Valencia R, Zimmering S. 1994. Chemical mutagenesis testing in Drosophila. X. Results of 70 coded chemicals tested for the National Toxicology Program. Environ Mol Mutagen. 23(3):208–227. I don’t see ACN in this paper. Should this be: Foureman P, Mason JM, Valencia R, Zimmering S. Chemical mutagenesis testing in Drosophila. IX. Results of 50 coded compounds tested for the National Toxicology Program. Environ Mol Mutagen. 1994;23(1):51–63.
  • Friedman MA, Beliles RP. 2002. Three-generation reproduction study of rats receiving acrylonitrile in drinking water. Toxicol Lett. 132(3):249–261.
  • Gallagher GT, Maull EA, Kovacs K, Szabo S. 1988. Neoplasms in rats ingesting acrylonitrile for two years. J Am Coll Toxicol. 7(5):603–615.
  • Galloway SM, Deasy DA, Bean CL, Kraynak AR, Armstrong MJ, Bradley MO. 1987. Effects of high osmotic strength on chromosome aberrations, sister-chromatid exchanges and DNA strand breaks, and the relation to toxicity. Mutat Res. 189(1):15–25.
  • García-Mengual E, Triviño JC, Sáez-Cuevas A, Bataller J, Ruíz-Jorro M, Vendrell X. 2019. Male infertility: establishing sperm aneuploidy thresholds in the laboratory. J Assist Reprod Genet. 36(3):371–381.
  • Garcia-Rodriguez A, de la Casa M, Serrano M, Gosálvez J, Roy Barcelona R. 2018. Impact of polymorphism in DNA repair genes OGG1 and XRCC1 on seminal parameters and human male infertility. Andrologia. 50(10):e13115.
  • Gargas ML, Andersen ME, Teo SK, Batra R, Fennell TR, Kedderis GL. 1995. A physiologically based dosimetry description of acrylonitrile and cyanoethylene oxide in the rat. Toxicol Appl Pharmacol. 134(2):185–194.
  • Garner RC, Campbell J. 1985. Tests for the induction of mutations to ouabain or 6-thioguanine resistance in mouse lymphoma L5178Y cells. Prog Mutat Res. 5:525–529.
  • Ghanayem BI, Nyska A, Haseman JK, Bucher JR. 2002. Acrylonitrile is a multisite carcinogen in male and female B6C3F1 mice. Toxicol Sci. 68(1):59–68.
  • Glauert HP, Kennan WS, Sattler GL, Pitot HC. 1985. Assays to measure theinduction of unscheduled DNA synthesis in cultured hepatocytes. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 371–373.
  • Guengerich FP, Geiger LE, Hogy LL, Wright PL. 1981. In vitro metabolism of acrylonitrile to 2-cyanoethylene oxide, reaction with glutathione, and irreversible binding to proteins and nucleic acids. Cancer Res. 41(12 Pt 1):4925–4933.
  • Gulati DK, Sabharwal PS, Shelby MD. 1985. Tests for the induction of chromosomal aberrations and sister chromatid exchanges in cultured Chinese hamster ovary (CHO) cells. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 413–426.
  • Gut I, Nerudová J, Stiborová A, Kopecký J, Frantík E. 1985. Acrylonitrile inhalation in rats: II. Excretion of thioethers and thiocyanate in urine. J Hyg Epidemiol Microbiol Immunol. 29(1):9–13.
  • Hachiya N. 1987. Evaluation of chemical genotoxicity by a series of short-term tests. Akita Med. 14:269–292.
  • Hachiya N, Sato M, Takizawa Y. 1984. Detection of DNA damage in mutagen-treated mammalian tissues by alkaline elution assay. Mutat Res. 130(5):363.
  • Hachiya N, Tanaka N, Takizawa Y. 1986. DNA damages in mammalian tissues, II. DNA single-strand breaks and alkali-liable sites detected by alkaline elution assay. Mutat Res. 164(4):266.
  • Hagio S, Tsuji N, Furukawa S, Takeuchi K, Hayashi S, Kuroda Y, Honma M, Masumura K. 2021. Effect of sampling time on somatic and germ cell mutations induced by acrylamide in gpt delta mice. Genes Environ. 43(1):4.
  • Hakura A, Shimada H, Nakajima M, Sui H, Kitamoto S, Suzuki S, Satoh T. 2005. Salmonella/human S9 mutagenicity test: a collaborative study with 58 compounds. Mutagenesis. 20(3):217–228.
  • Hamdy NM, Al-Abbasi FA, Alghamdi HA, Tolba MF, Esmat A, Abdel-Naim AB. 2012. Role of neutrophils in acrylonitrile-induced gastric mucosal damage. Toxicol Lett. 208(2):108–114.
  • Hartung R. 1982. Cyanides and nitriles. In: Clayton GD, Clayton E, editors. Patty’s industrial hygiene and toxicology. Vol. 2C. New York (NY): John Wiley & Sons; p. 4845–4900.
  • Hartwig A, Arand M, Epe B, Guth S, Jahnke G, Lampen A, Martus HJ, Monien B, Rietjens IMCM, Schmitz-Spanke S, et al. 2020. Mode of action-based risk assessment of genotoxic carcinogens. Arch Toxicol. 94(6):1787–1877. Erratum in: arch Toxicol. 2020 Sep;94(9):3347.
  • Heflich RH, Johnson GE, Zeller A, Marchetti F, Douglas GR, Witt KL, Gollapudi BB, White PA. 2020. Mutation as a toxicological endpoint for regulatory decision-making. Environ Mol Mutagen. 61(1):34–41.
  • Himwich WA, Saunders JP. 1948. Enzymatic conversion of cyanide to thiocyanate. Am J Physiol. 153(2):348–354.
  • Hoffmann D, Hoffmann I. 1997. The changing cigarette, 1950-1995. J Toxicol Environ Health. 50(4):307–364.
  • Hogy LL, Guengerich FP. 1986. In vivo interaction of acrylonitrile and 2-cyanoethylene oxide with DNA in rats. Cancer Res. 46(8):3932–3938.
  • Hunter J, Maxwell JD, Carrella M, Stewart DA, Williams R. 1971. Urinary d-glucaric acid excretion as a measure of hepatic enzyme induction in man. Clin Sci. 40(3):10P.
  • IARC. 2004. Tobacco smoke and involuntary smoking. IARC Monogr Eval Carcinog Risks Hum. 83:1–1438.
  • IARC (International Agency for Research on Cancer). 1999. Predictive value of rodent forestomach and gastric neuroendocrine tumours in evaluating carcinogenic risks to humans, views and expert opinions of an IARC Working Group. Technical Publication No. 39. Lyon (France): International Agency for Research on Cancer.
  • Inge-Vechtomov SG, Pavlov YI, Noskov VN, Repnevskaya MV, Karpova TS, Khromov-Borisov NN, Chekuolene J, Chitavichus D. 1985. Tests for genetic activity in the yeast Saccharomyces cerevisiae: study of forward and reverse mutation, mitotic recombination and illegitimate mating induction. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 243–255.
  • Ishidate M, Jr, Sofuni T. 1985. The in vitro chromosomal aberration test using Chinese hamster lung (CHL) fibroblast cells in culture. Prog Mutat Res. 5:427–432.
  • Ishidate M, Jr, Sofuni T, Yoshikawa K. 1981. Chromosomal aberration tests in vitro as a primary screening tool for environmental mutagens and/or carcinogens. Gann. 27:95–108.
  • Jacob S, Ahmed AE. 2003. Acrylonitrile-induced neurotoxicity in normal human astrocytes: oxidative stress and 8-hydroxy-2'-deoxyguanosine formation. Toxicol Mech Methods. 13(3):169–179.
  • Jalil RA. 1994. Concentrations of thiocyanate and hypothiocyanite in the saliva of young adults. J Nihon Univ Sch Dent. 36(4):254–260.
  • Jiang J, Xu Y, Klaunig JE. 1998. Induction of oxidative stress in rat brain by acrylonitrile (ACN). Toxicol Sci. 46(2):333–341.
  • Johannsen FR, Levinskas GJ. 2002a. Comparative chronic toxicity and carcinogenicity of acrylonitrile by drinking water and oral intubation to Spartan Sprague-Dawley rats. Toxicol Lett. 132(3):197–219.
  • Johannsen FR, Levinskas GJ. 2002b. Chronic toxicity and oncogenic dose-response effects of lifetime oral acrylonitrile exposure to F344 rats. Toxicol Lett. 132(3):221–247.
  • Johnston RV, Schwetz BA, Middleton J, Humiston JCG, Lisowe RW. 1978. Unpublished report from the Dow Chemical Company (as reported in WHO, 1983)
  • Jung R, Engelhart G, Herbolt B, Jäckh R, Müller W. 1992. Collaborative study of mutagenicity with Salmonella typhimurium TA102. Mutat Res. 278(4):265–270.
  • Junge B. 1985. Changes in serum thiocyanate concentration on stopping smoking. Br Med J (Clin Res Ed). 291(6487):22.
  • Kailasam S, Rogers KR. 2007. A fluorescence-based screening assay for DNA damage induced by genotoxic industrial chemicals. Chemosphere. 66(1):165–171.
  • Kamendulis LM, Jiang J, Zhang H, deFeijter-Rupp H, Trosko JE, Klaunig JE. 1999. The effect of acrylonitrile on gap junctional intercellular communication in rat astrocytes. Cell Biol Toxicol. 15(3):173–183.
  • Kamendulis LM, Jiang J, Xu Y, Klaunig JE. 1999. Induction of oxidative stress and oxidative damage in rat glial cells by acrylonitrile. Carcinogenesis. 20(8):1555–1560.
  • Kawachi T, Yahagi T, Kada T, Tazima Y, Ishidate M, Sasaki M, Sugiyama T. 1980. Cooperative programme on short-term assays for carcinogenicity in Japan. IARC Sci Publ. (27):323–330.
  • Kedderis GL, Batra R. 1993. Species differences in the hydrolysis of 2-cyanoethylene oxide, the epoxide metabolite of acrylonitrile. Carcinogenesis. 14(4):685–689.
  • Kedderis GL, Batra R, Koop DR. 1993. Epoxidation of acrylonitrile by rat and human cytochromes P450. Chem Res Toxicol. 6(6):866–871.
  • Kedderis GL, Batra R, Turner MJ. Jr. 1995. Conjugation of acrylonitrile and 2-cyanoethylene oxide with hepatic glutathione. Toxicol Appl Pharmacol. 135(1):9–17.
  • Kedderis GL, Sumner SC, Held SD, Batra R, Turner MJ, Roberts AE, Fennell TR. 1993. Dose-dependent urinary excretion of acrylonitrile metabolites by rats and mice. Toxicol Appl Pharmacol. 120(2):288–297.
  • Khudoley VV, Mizgireuv I, Pliss GB. 1987. The study of mutagenic activity of carcinogens and other chemical agents with Salmonella typhimurium assays: testing of 126 compounds. Arch Geschwulstforsch. 57:453–462.
  • Kirman CR, Sweeney LM, Gargas ML, Strother DE, Collins JJ, Deskin R. 2008. Derivation of noncancer reference values for acrylonitrile. Risk Anal. 28(5):1375–1394.
  • Klaunig JE, Forney RB. 2010. Nongenotoxic Mechanisms of Acrylonitrile Carcinogenicity Study I: Antioxidants screening [unpublished]. Oakland (CA): Testing Laboratory: Center for Environmental Health, USA. Owner company: The AN group Inc.
  • Kobets T, Iatropoulos MJ, Williams GM. 2022. Acrylonitrile induction of rodent neoplasia: potential mechanism of action and relevance to humans. Toxicol Res Appl. 6:239784732110553.
  • Kodama Y, Boreiko CJ, Skopek TR, Recio L. 1989. Cytogenetic analysis of spontaneous and 2-cyanoethylene oxideinduced tk-/- mutants in TK6 human lymphoblastoid cultures. Environ Mol Mutagen. 14(3):149–154.
  • Kolenda-Roberts HM, Harris N, Singletary E, Hardisty JF. 2013. Immunohistochemical characterization of spontaneous and acrylonitrile-induced brain tumors in the rat. Toxicol Pathol. 41(1):98–108.
  • Koutros S, Lubin JH, Graubard BI, Blair A, Stewart PA, Beane Freeman LE, Silverman DT. 2019. Extended mortality follow-up of a cohort of 25,460 workers exposed to acrylonitrile. Am J Epidemiol. 188(8):1484–1492.
  • Krajewska B, Lutz W, Piłacik B. 1998. Determination of blood serum oncoprotein NEU and antioncoprotein p-53–molecular biomarkers in various types of occupational exposure. Int J Occup Med Environ Health. 11(4):343–348.
  • Kroetz DL, Kerr BM, McFarland LV, Loiseau P, Wilensky AJ, Levy RH. 1993. Measurement of in vivo microsomal epoxide hydrolase activity in white subjects. Clin Pharmacol Ther. 53(3):306–315.
  • Kamendulis LM, Siesky AM, Park JJ, Ren B, Xu Y, Klaunig JE. 2001. Examination of DNA synthesis and oxidative stress following treatment with acrylonitrile in the mouse. Toxicologist. Abstract 1366.
  • Lakhanisky T, Hendrickx B. 1985. Induction of DNA single-strand breaks in CHO cells in culture. Prog Mutat Res. 5:367–370.
  • Lambert IB, Singer TM, Boucher SE, Douglas GR. 2005. Detailed review of transgenic rodent mutation assays. Mutat Res. 590(1–3):1–280.
  • Lambotte-Vandepaer M, Duverger-van Bogaert M, de Meester C, Poncelet F, Mercier M. 1980. Mutagenicity of urine from rats and mice treated with acrylonitrile. Toxicology. 16(1):67–71.
  • Lambotte-Vandepaer M, Bogaert M-V, de Meester C, Rollmann B, Poncelet F, Mercier M. 1981. Identification of two urinary metabolites of rats treated with acrylonitrile; influence of several inhibitors on the mutagenicity of those urines. Toxicol Lett. 7(4–5):321–327.
  • Laugesen M, Fowles J. 2005. Scope for regulation of cigarette smoke toxicity according to brand differences in published toxicant emissions. N Z Med J. 118(1213):U1401.
  • Lee CG, Webber TD. 1985. The induction of gene mutations in the mouse lymphoma L5178Y/TK+/- assay and the Chinese hamster V79/HGPRT assay. Prog Mutat Res. 5:547–554.
  • Lefkowitz DL, Lefkowitz SS. 2008. Microglia and myeloperoxidase: a deadly partnership in neurodegenerative disease. Free Radic Biol Med. 45(5):726–731.
  • Leonard A, Garny V, Poncelet F, Mercier M. 1981. Mutagenicity of acrylonitrile in mouse. Toxicol Lett. 7(4–5):329–334.
  • Li XJ, Li B, Huang JS, Shi JM, Wang P, Fan W, Zhou YL. 2014. Effects of acrylonitrile on lymphocyte lipid rafts and RAS/RAF/MAPK/ERK signaling pathways. Genet Mol Res. 13(3):7747–7756.
  • Li Z. 1996. Survey of reproductive system of female workers exposed to acrylonitrile. China Ind Med Mag. 9:33–39.
  • Liang JC, Sherron DA, Johnston D. 1986. Lack of correlation between mutagen-induced chromosomal univalency and aneuploidy in mouse spermatocytes. Mutat Res. 163(3):285–297.
  • Liber HL. Jr, 1985. Mutation tests with salmonella using 8-azaguanine resistance as the genetic marker. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 213–216.
  • Lijinsky W, Andrews AW. 1980. Mutagenicity of vinyl compounds in Salmonella typhimurium. Teratog Carcinog Mutagen. 1(3):259–267.
  • Lin CY, Lee HL, Sung FC, Su TC. 2018. Investigating the association between urinary levels of acrylonitrile metabolite N-acetyl-S-(2-cyanoethyl)-L-cysteine and the oxidative stress product 8-hydroxydeoxyguanosine in adolescents and young adults. Environ Pollut. 239:493–498.
  • Litton Bionetics. 1976. Mutagenicity evaluation of MCA 730. Final report, LBI Project No. 2548, prepared for the Manufacturing Chemists Association. Kensington (MD): Litton Bionetics.
  • Litton Bionetics. 1980. Three-generation reproduction study of rats receiving acrylonitrile in drinking water. Unpublished report. Kensington (MD): Litton Bionetics.
  • Loft S, Høgh Danielsen P, Mikkelsen L, Risom L, Forchhammer L, Møller P. 2008. Biomarkers of oxidative damage to DNA and repair. Biochem Soc Trans. 36(Pt 5):1071–1076.
  • Loprieno N, Boncristiani G, Forster R, Goldstein B, Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH. 1985. Assays for forward mutation in Schizosaccharomyces pombe strain P1. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 297–306.
  • Luetjens CM, Rolf C, Gassner P, Werny JE, Nieschlag E. 2002. Sperm aneuploidy rates in younger and older men. Hum Reprod. 17(7):1826–1832.
  • Luijten MNH, Lee JXT, Crasta KC. 2018. Mutational game changer: chromothripsis and its emerging relevance to cancer. Mutat Res Rev Mutat Res. 777:29–51.
  • Lundquist P, Kågedal B, Nilsson L. 1995. An improved method for determination of thiocyanate in plasma and urine. Eur J Clin Chem Clin Biochem. 33(6):343–349.
  • Luo JC, Liu HT, Cheng TJ, Du CL, Wang JD. 1999. Plasma p53 protein and anti-p53 antibody expression in vinyl chloride monomer workers in Taiwan. J Occup Environ Med. 41(6):521–526.
  • Luo YS, He QK, Sun MX, Qiao FX, Liu YC, Xu CL, Xu ZR, Zhao SC, Wang HL, Qi ZQ, et al. 2022. Acrylonitrile exposure triggers ovarian inflammation and decreases oocyte quality probably via mitochondrial dysfunction induced apoptosis in mice. Chem Biol Interact. 360:109934.
  • Madle S, Dean SW, Andrae U, Brambilla G, Burlinson B, Doolittle DJ, Furihata C, Hertner T, McQueen CA, Mori H. 1994. Recommendations for the performance of UDS tests in vitro and in vivo. Mutat Res. 312(3):263–285. 10.1016/0165-1161(94)00013-1.
  • Mahalakshmi K, Pushpakiran G, Anuradha CV. 2003. Taurine prevents acrylonitrile-induced oxidative stress in rat brain. Pol J Pharmacol. 55:1037–1043.
  • Major J, Hudák A, Kiss G, Jakab MG, Szaniszló J, Náray M, Nagy I, Tompa A. 1998. Follow-up biological and genotoxicological monitoring of acrylonitrile- and dimethylformamide-exposed viscose rayon plant workers. Environ Mol Mutagen. 31(4):301–310.
  • Major J, Jakab MG, Tompa A. 1999. The frequency of induced premature centromere division in human populations occupationally exposed to genotoxic chemicals. Mutat Res. 445(2):241–249.
  • Maltoni C, Ciliberti A, Cotti G, Perino G. 1988. Long-term carcinogenicity bioassays on acrylonitrile administered by inhalation and by ingestion to Sprague-Dawley rats. Ann N Y Acad Sci. 534:179–202.
  • Maltoni C, Ciliberti A, Di MV. 1977. Carcinogenicity bioassays on rats of acrylonitrile administered by inhalation and by ingestion. Med Lav. 68(6):401–411.
  • Manso JA, Camacho IFC, Calle E, Casado J. 2011. Potential of α,β-unsaturated compounds. Org Biomol Chem. 9(18):6226–6233.
  • Marchetti F, Aardema MJ, Beevers C, van Benthem J, Godschalk R, Williams A, Yauk CL, Young R, Douglas GR. 2018. Identifying germ cell mutagens using OECD test guideline 488 (transgenic rodent somatic and germ cell gene mutation assays) and integration with somatic cell testing. Mutat Res Genet Toxicol Environ Mutagen. 832–833:7–18. Erratum in: Mutat Res Genet Toxicol Environ Mutagen. 2019;844:70–71.
  • Marchetti P, Fovez Q, Germain N, Khamari R, Kluza J. 2020. Mitochondrial spare respiratory capacity: mechanisms, regulation, and significance in non-transformed and cancer cells. FASEB J. 34(10):13106–13124.
  • Marnett LJ, Riggins JN, West JD. 2003. Endogenous generation of reactive oxidants and electrophiles and their reactions with DNA and protein. J Clin Invest. 111(5):583–593.
  • Marsh GM, Kruchten AT. 2023. A reevaluation of selected mortality risks in the updated NCI/NIOSH acrylonitrile cohort study. Front Public Health. 11:1122346.
  • Marsh GM, Zimmerman SD. 2015. Mortality among chemical plant workers exposed to acrylonitrile: 2011 follow-up. J Occup Environ Med. 57(2):134–145.
  • Martin CN, Campbell J. 1985. Tests for the induction of unscheduled DNA repair synthesis in HeLa cells. Prog Mutat Res. 5:375–379.
  • Masumura K, Ando T, Toyoda-Hokaiwado N, Ukai A, Nohmi T, Honma M. 2021. Comparison of the frequencies of ENU-induced point mutations in male germ cells and inherited germline mutations in their offspring. Genes Environ. 43(1):43.
  • Masumura K, Toyoda-Hokaiwado N, Ukai A, Gondo Y, Honma M, Nohmi T. 2016. Estimation of the frequency of inherited germline mutations by whole exome sequencing in ethyl nitrosourea-treated and untreated gpt delta mice. Genes Environ. 38:10.
  • Mathews A, Ohsawa K, Buckland ME, Kesavadas C, Ratheesan K, Kusumakumary P, Burger PC, Kohsaka S, Graeber MB. 2016. Microglioma in a child – a further case in support of the microglioma entity and distinction from histiocytic sarcoma. Clin Neuropathol. 35(5):302–313.
  • Matsushima T, Muramatsu M, Haresaku M. 1985. Mutation tests on Salmonella tvohimurium by the preincubation method. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 181–186.
  • Matsushita H, Goto S. 1980. Study for the screening technology and development of carcinogen focused on mutagenicity. Contract research on occupational health in 1980: 3) mutation assays of 30 chemical substances on Salmonella typhimurium and Escherichia coli. Tokyo (Japan): Testing laboratory: The Institute of Public Health. Owner company: Ministry of Labor.
  • Matthews EJ, DelBalzo T, Rundell JO. 1985. Assays for morphological transformation and mutation to ouabain resistance of Balb/c-3T3 cells in culture. Prog Mutat Res. 5:639–650.
  • McMahon RE, Cline JC, Thompson CZ. 1979. Assay of 855 test chemicals in ten tester strains using a new modification of the Ames test for bacterial mutagens. Cancer Res. 39(3):682–693.
  • Meek ME, Bucher JR, Cohen SM, Dellarco V, Hill RN, Lehman-McKeeman LD, Longfellow DG, Pastoor T, Seed J, Patton DE. 2003. A framework for human relevance analysis of information on carcinogenic modes of action. Crit Rev Toxicol. 33(6):591–653.
  • Mehrotra J, Khanna VK, Husain R, Seth PK. 1988. Biochemical and developmental effects in rats following in utero exposure to acrylonitrile: a preliminary report. Ind Health. 26(4):251–255.
  • Mehta RD Jr., von Borstel RC. 1985. Tests for genetic activity in the yeast Saccharomyces cerevisiae using strains D7-144, XV185-14C, and RM52. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 271–284.
  • Meijer GA. 2005. Chromosomes and cancer, Boveri revisited. Cell Oncol. 27(5–6):273–275.
  • Milvy P, Wolff M. 1977. Mutagenic studies with acrylonitrile. Mutat Res. 48(3–4):271–278.
  • Mohamadin AM, Abdel-Naim AB. 2003. In vitro activation of dibromoacetonitrile to cyanide: role of xanthine oxidase. Arch Toxicol. 77(2):86–93.
  • Mohamadin AM. 2001. Possible role of hydroxyl radicals in the oxidation of dichloroacetonitrile by Fenton-like reaction. J Inorg Biochem. 84(1–2):97–105.
  • Moore MM, Doerr CL. 1990. Comparison of chromosome aberration frequency and small-colony TK-deficient mutant frequency in L5178Y/TK(+/-)-3.7.2C mouse lymphoma cells. Mutagenesis. 5(6):609–614.
  • Moore RR, Hardisty JF. 2014. Immunohistochemical characterization of brain tumors: a two-year toxicity and oncogenicity study with acrylonitrile following inhalation exposure in rats (Quast et al. 1980). EPL Project No. 694-004. Final Pathology Report, November 4, 2014.
  • Morita T, Asano N, Awogi T, Sasaki YF, Sato S, Shimada H, Sutou S, Suzuki T, Wakata A, Sofuni T, et al. 1997. Evaluation of the rodent micronucleus assay in the screening of IARC carcinogens (groups 1,2A and 2B): the summary report of the 6th collaborative study by CSGMT/JEMS MMS. Collaborative study of the micronucleus group test. Mammalian Mutagenicity Study Group. Mutat Res. 389(1):3–122.
  • Morris SM, Manjanatha MG, Shelton SD, Domon OE, McGarrity LJ, Casciano DA. 1996. A mutation in the p53 tumor suppressor gene of AHH-1 tk+/- human lymphoblastoid cells. Mutat Res. 356(2):129–134.
  • Mortelmans K, Zeiger E. 2000. The Ames Salmonella/microsome mutagenicity assay. Mutat Res. 455(1–2):29–60.
  • Müller W, Engelhart G, Herbold B, Jäckh R, Jung R. 1993. Evaluation of mutagenicity testing with Salmonella typhimurium TA102 in three different laboratories. Environ Health Perspect. 101(Suppl 3):33–36.
  • Murray FJ, Schwetz BA, Nitschke KD, John JA, Norris JM, Gehring PJ. 1978. Teratogenicity of acrylonitrile given to rats by gavage or by inhalation. Food Cosmet Toxicol. 16(6):547–551.
  • Mutlu E, Jeong YC, Collins LB, Ham AJ, Upton PB, Hatch G, Winsett D, Evansky P, Swenberg JA. 2012. A new LC-MS/MS method for the quantification of endogenous and vinyl chloride-induced 7-(2-Oxoethyl)guanine in sprague-dawley rats. Chem Res Toxicol. 25(2):391–399.
  • Myhr B, Bowers L, Kaspary WJ. 1985. Assays for the induction of gene mutations at the thymidine kinase locus in L5178Y mouse lymphoma cells in culture. Prog Mutat Res. 5:555–568.
  • Nakagawa Y, Toyoizumi T, Sui H, Ohta R, Kumagai F, Usumi K, Saito Y, Yamakage K. 2015. In vivo comet assay of acrylonitrile, 9-aminoacridine hydrochloride monohydrate and ethanol in rats. Mutat Res Genet Toxicol Environ Mutagen. 786–788:104–113.
  • Nakamura SI, Oda Y, Shimada T, Oki I, Sugimoto K. 1987. SOS-inducing activity of chemical carcinogens and mutagens in Salmonella typhimurium TA1535-psk-1002: examination with 151 chemicals. Mutat Res. 192(4):239–246.
  • Nasralla SN, Ghoneim AI, Khalifa AE, Gad MZ, Abdel-Naim AB. 2009. Lactoperoxidase catalyzes in vitro activation of acrylonitrile to cyanide. Toxicol Lett. 191(2–3):347–352.
  • Natarajan AT, Bussmann CJM, van Kesteren-van Leeuwen AC, Meijers M, Van Rijn JLS. 1985. Tests for chromosomal aberrations and sister chromatid exchanges in cultured Chinese hamster ovary (CHO) cells. Prog Mutat Res. 5:433–437.
  • Neal BH, Collins JJ, Strother DE, Lamb JC. 2009. Weight-of-the-evidence review of acrylonitrile reproductive and developmental toxicity studies. Crit Rev Toxicol. 39(7):589–612.
  • Nemec MD, Kirkpatrick DT, Sherman J, Van Miller JP, Pershing ML, Strother DE. 2008. Two-generation reproductive toxicity study of inhaled acrylonitrile vapors in Crl: CD(SD) rats. Int J Toxicol. 27(1):11–29.
  • Nesterova EV, Durnev AD, Seredenin SB. 1999. Tsitogeneticheskie éffekty akrilamida, akrilonitrila i ikh kombinatsii s verapamilom in vivo [Cytogenetic effects of acrylamide, acrylonitrile and their combination with verapamil in vivo]. Biull Eksp Biol Med. 128(12):684–689.
  • NHANES. 2015–2016. Laboratory data. [accessed 2022 Aug 1]. https://wwwn.cdc.gov/nchs/nhanes/Default.aspx.
  • NTP (National Toxicology Program). 2001. Toxicology and carcinogenesis studies of acrylonitrile (CAS No. 107- 13-1) in B6C3F1 mice (gavage studies). Public Health Service, U.S. Department of Health and Human Services; NTP TR506. Available from the National Institute of Environmental Health Sciences, Research Triangle Park, NC. [accessed 2009 Apr 21]. http://ntp.niehs.nih.gov/ntpweb/index.cfm?objectid=D16D6C59-F1F6-975E-7D23D1519B8CD7A5#tr500.
  • NTP [National Toxicology Program]. 2021. 15th ROC [report on carcinogens] acrylonitrile. https://ntp.niehs.nih.gov/whatwestudy/assessments/cancer/roc/index.html
  • Obe G, Hille A, Jonas R, Schmidt S, Thenhaus U. 1985. Tests for the induction ofsister-chromatid exchanges in human peripheral lymphocytes in culture. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 439–442.
  • Oberly TJ, Bewsey BJ, Probst GS. 1984. An evaluation of the L5178Y TK+/- mouse lymphoma forward mutation assay using 42 chemicals. Mutat Res. 125(2):291–306.
  • Oberly TJ, Hoffman WP, Garriott ML. 1996. An evaluation of the twofold rule for assessing a positive response in the L5178Y TK(+/-) mouse lymphoma assay. Mutat Res. 369(3–4):221–232.
  • OECD. 1997. Test no. 486: unscheduled DNA synthesis (UDS) test with mammalian liver cells in vivo. OECD guidelines for the testing of chemicals, Section 4. Paris: OECD Publishing.
  • Osgood C, Bloomfield M, Zimmering S. 1991. Aneuploidy in drosophila. IV. Inhalation studies on the induction of aneuploidy by nitriles. Mutat Res. 259(2):165–176.
  • Page BD, Charbonneau CF. 1983. Determination of acrylonitrile in foods by headspace gas-liquid chromatography with nitrogen-phosphorus detection. J Assoc off Anal Chem. 66(5):1096–1105. 6630123.
  • Page BD, Charbonneau CF. 1985. Improved procedure for determination of acrylonitrile in foods and its application to meat. J Assoc off Anal Chem. 68(3):606–608.
  • Parent RA, Casto BC. 1979. Effect of acrylonitrile on primary Syrian golden hamster embryo cells in culture: transformation and DNA fragmentation. J Natl Cancer Inst. 62(4):1025–1029.
  • Parry JM. 1985. Summary report on the performance of the yeast and Aspergillus assays. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 25–46.
  • Parry JM, Eckardt F. 1985a. The detection of mitotic gene conversion, point mutation and mitotic segregation using the yeast Saccharomyces cerevisiae strain D7. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. New York: Elsevier; p. 261–269.
  • Parry JM, Eckardt F. 1985b. The induction of mitotic aneuploidy, point mutation and mitotic crossing-over in the yeast Saccharomyces ceretqsiae D61.M and D6. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 285–295.
  • Pattison DI, Davies MJ, Hawkins CL. 2012. Reactions and reactivity of myeloperoxidase-derived oxidants: differential biological effects of hypochlorous and hypothiocyanous acids. Free Radic Res. 46(8):975–995.
  • Pegg AE. 1990. Properties of mammalian O6-alkylguanine-DNA transferases. Mutat Res. 233(1–2):165–175.
  • Perocco P, Pane G, Bolognesi S, Zannotti M. 1982. Increase of sister chromatid exchange and unscheduled synthesis of deoxyribonucleic acid by acrylonitrile in human lymphocytes in vitro. Scand J Work Environ Health. 8(4):290–293.
  • Peter H, Appel KE, Berg R, Bolt HM. 1983. Irreversible binding of acrylonitrile to nucleic acids. Xenobiotica. 13(1):19–25.
  • Peter H, Schwarz M, Mathiasch B, Appel KE, Bolt HM. 1983. A note on synthesis and reactivity towards DNA of glycidonitrile, the epoxide of acrylonitrile. Carcinogenesis. 4(2):235–237.
  • Petrikovics I, Thompson DE, Rockwood GA, Logue BA, Martin S, Jayanna P, Yu JC. 2011. Organ-distribution of the metabolite 2-aminothiazoline-4-carboxylic acid in a rat model following cyanide exposure. Biomarkers. 16(8):686–690.
  • Priston RJ, Dean BJ, et al. 1985. Tests for the induction of chromosomeaberrations, polyploidy and sister chromatid exchanges in rat liver (RL) cells. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 387–395.
  • Priston RAJ, Dean BJ. 1985. Tests for the induction of chromosomal aberrations, polyploidy and sister chromatid exchanges in rat liver (RL4) cells. Prog Mutat Res. 5:387–395.
  • Probst GS, Hill LE. 1985. Tests for the induction of DNA-repair synthesis in primary cultures of adult rat hepatocytes. Prog Mutat Res. 5:381–386.
  • Prokopczyk B, Bertinato P, Hoffman D. 1988. Cyanoethylation of DNA in vivo in 3-(methylnitrosamino)-propionitrile, an Areca-derived carcinogen. Cancer Res. 48(23):6780–6784.
  • Pu X, Wang Z, Klaunig JE. 2015. Alkaline comet assay for assessing DNA damage in individual cells. Curr Protoc Toxicol. 65:3.12.1–3.12.11.
  • Pu X, Kamendulis LM, Klaunig JE. 2009. Acrylonitrile-induced oxidative stress and oxidative DNA damage in male Sprague-Dawley rats. Toxicol Sci. 111(1):64–71.
  • Pu X, Kamendulis LM, Klaunig JE. 2006. Acrylonitrile-induced oxidative DNA damage in rat astrocytes. Environ Mol Mutagen. 47(8):631–638.
  • Puppel K, Kapusta A, Kuczyńska B. 2015. The etiology of oxidative stress in the various species of animals, a review. J Sci Food Agric. 95(11):2179–2184.
  • Quast JF. 2002. Two-year toxicity and oncogenicity study with acrylonitrile incorporated in the drinking water of rats. Toxicol Lett. 132(3):153–196.
  • Quast JF, Schuetz DJ, Balmer MF, Gushow TS, Park CN, McKenna MJ. 1980. A two-year toxicity and oncogenicity study with acrylonitrile following inhalation exposure of rats. Midland (MI): Dow Chemical Co., Toxicology Research Laboratory.
  • Quast JF, Wade C, Humiston C, Gushow TS, Park CN, McKenna MJ. 1980. Two-year toxicity and oncogenicity study with acrylonitrile incorporated in the drinking water of rats. Midland (MI): Dow Chemical Co., Toxicology Research Laboratory.
  • Rabello-Gay MN, Ahmed AE. 1980. Acrylonitrile: in vivo cytogenetic studies in mice and rats. Mutat Res. 79(3):249–255.
  • Rao P, Singh P, Yadav SK, Gujar NL, Bhattacharya R. 2013. Acute toxicity of some synthetic cyanogens in rats: time-dependent cyanide generation and cytochrome oxidase inhibition in soft tissues after sub-lethal oral intoxication. Food Chem Toxicol. 59:595–609.
  • Recio L, Simpson D, Cochrane J, Liber H, Skopek TR. 1990. Molecular analysis of hprt mutants induced by 2-cyanoethylene oxide in human lymphoblastoid cells. Mutat Res. 242(3):195–208.
  • Recio L, Skopek TR. 1988. The cellular and molcular analysis of acrylonitrile-induced mutations in human cells. CIIT Activities. 8:1–6.
  • Rexroat MA, Probst GS, et al. 1985. Mutation tests with Salmonella using the plate-incorporation assay. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 201–212.
  • Rice GC, Hoy C, Schimke RT. 1986. Transient hypoxia enhances the frequency of dihydrofolate reductase gene amplification in Chinese hamster ovary cells. Proc Natl Acad Sci U S A. 83(16):5978–5982.
  • Rioux KL, Delaney S. 2020. 1,N6-ethenoadenine: from molecular to biological consequences. Chem Res Toxicol. 33(11):2688–2698.
  • Rizzi R, Chiesara E, Cova D, Mattioli M, Di Lernia R. 1984. Acrylonitrile: mutagenicity in yeasts and genotoxicity in HeLa cells. Mut Res Environ Mutagen Relat Subj. 130(3):223.
  • Robbiano L, Allavena A, Bagarolo C, Martelli A, Brambilla G. 1994. Comparison in human and rat hepatocytes of the DNA-damaging activity of five chemicals probably carcinogenic to humans. Toxicol in Vitro. 8(1):131–137.
  • Roberfroid M, Poncelet F, Lambotte-Vandepaer M, Duverger-Van Bogaert M, de Meester C, Mercier M. 1978. Acute biotoxic effect of styrene on rat liver. Correlation with enzyme-mediated mutagenicity of benzpyrene and acrylonitrile. Scand J Work Environ Health. 4(2):163–168.
  • Roberts AE, Kedderis GL, Turner MJ, Rickert DE, Swenberg JA. 1991. Species comparison of acrylonitrile epoxidation by microsomes from mice, rats and humans: relationship to epoxide concentrations in mouse and rat blood. Carcinogenesis. 12(3):401–404.
  • Rössner P, Jr, Binková B, Chvátalová I, Srám RJ. 2002. Acrylonitrile exposure: the effect on p53 and p21(WAF1) protein levels in the blood plasma of occupationally exposed workers and in vitro in human diploid lung fibroblasts. Mutat Res. 517(1–2):239–250.
  • Roy P, Kulkarni AP. 1999. Co-oxidation of acrylonitrile by soybean lipoxygenase and partially purified human lung lipoxygenase. Xenobiotica. 29(5):511–531.
  • Rudd CJ. 1983. L5178Y tk+/? mouse lymphoma forward mutation assay of acrylonitrile. Prepared by SRI International, Menlo Park, CA, for the Environmental Research Center, U.S. Environmental Protection Agency, Research Triangle Park, NC; SRI Project LSU-3447, EPA Contract No. 68-02-3703.
  • Saillenfait A, Bonnet P, Guenier J, Deceaurriz J. 1993. Relative developmental toxicities of inhaled aliphatic mononitriles in rats. Fundam Appl Toxicol. 20(3):365–375.
  • Saillenfait AM, Sabaté JP. 2000. Comparative development toxicities of aliphatic nitriles: in vivo and in vitro observations. Toxicol Appl Pharmacol. 163(2):149–163.
  • Sakurai H, Onodera M, Utsunomiya T. 1978. Health effects of acrylonitrile in acrylic fibre factories. Br J Ind Med. 35(3):219–225.
  • Sasaki M, Sugimura K, Yoshida MA, et al. 1980. Cytogenetic effects of 60 chemicals on cultured human and Chinese hamster cells. La Kromosomo II. 20:574–584.
  • Savage JRK. 2011. An introduction to chromosomal aberrations. Atlas of genetics and cytogenetics in oncology and haematology. https://atlasgeneticsoncology.org/deep-insight/20001/an-introduction-to-chromosomal-aberrations/
  • Schettgen T, Bertram J, Kraus T. 2012. Accurate quantification of the mercapturic acids of acrylonitrile and its genotoxic metabolite cyanoethylene-epoxide in human urine by isotope-dilution LC-ESI/MS/MS. Talanta. 98:211–219.
  • Schiestl RH, Gietz RD, Mehta RD, Hastings PJ. 1989. Carcinogens induce intrachromosomal recombination in yeast. Carcinogenesis. 10(8):1445–1455.
  • Schiestl RH, Reynolds P, Prakash S, Prakash L. 1989. Cloning and sequence analysis of the Saccharomyces cerevisiae RAD9 gene and further evidence that its product is required for cell cycle arrest induced by DNA damage. Mol Cell Biol. 9(5):1882–1896.
  • Schneider J, Presek P, Braun A, Woitowitz HJ. 1999. Serum levels of pantropic p53 protein and EGF-receptor, and detection of anti-p53 antibodies in former uranium miners (SDAG Wismut). Am J Ind Med. 36(6):602–609.
  • Schulz V, Bonn R, Kindler J. 1979. Kinetics of elimination of thiocyanate in 7 healthy subjects and in 8 subjects with renal failure. Klin Wochenschr. 57(5):243–247.
  • Sekihashi K, Yamamoto A, Matsumura Y, Ueno S, Watanabe-Akanuma M, Kassie F, Knasmüller S, Tsuda S, Sasaki YF. 2002. Comparative investigation of multiple organs of mice and rats in the comet assay. Mutat Res. 517(1–2):53–75.
  • Serota DG, Giles HD, Coyne JM, Hogan DB. 1996. Sub-chronic toxicity study in B6C3F1 mice. Birmingham (AL): Testing Laboratory: Southern Research Institute. Report no: SRI-LIF-95- 593-8618-I.
  • Sharief Y, Brown AM, Backer LC, Campbell JA, Westbrook-Collins B, Stead AG, Allen JW. 1986. Sister chromatid exchange and chromosome aberration analyses in mice after in vivo exposure to acrylonitrile, styrene, or butadiene monoxide. Environ Mutagen. 8(3):439–448.
  • Silver EH, Szabo S. 1982. Possible role of lipid peroxidation in the actions of acrylonitrile on the adrenals, liver and gastrointestinal tract. Res Commun Chem Pathol Pharmacol. 36:33–43.
  • Skopek TR, Liber HL, Kaden DA, Thilly WG. 1978. Relative sensitivities of forward and reverse mutation assays in Salmonella typhimurium. Proc Natl Acad Sci U S A. 75(9):4465–4469.
  • Smith CC, Adkins DJ, Martin EA, O'Donovan MR. 2008. Recommendations for design of the rat comet assay. Mutagenesis. 23(3):233–240. Erratum in: Mutagenesis. 2009;24(1):95. Dosage error in article text.
  • Smith CC, O'Donovan MR, Martin EA. 2006. hOGG1 recognizes oxidative damage using the comet assay with greater specificity than FPG or ENDOIII. Mutagenesis. 21(3):185–190.
  • Solomon JJ, Cote IL, Wortman M, Decker K, Segal A. 1984. In vitro alkylation of calf thymus DNA by acrylonitrile. Isolation of cyanoethyl-adducts of guanine and thymidine and carboxyethyl-adducts of adenine and cytosine. Chem Biol Interact. 51(2):167–190.
  • Solomon JJ, Singh US, Segal A. 1993. In vitro reactions of 2-cyanoethylene oxide with calf thymus DNA. Chem Biol Interact. 88(2–3):115–135.
  • Speit G, Vasquez M, Hartmann A. 2009. The comet assay as an indicator test for germ cell genotoxicity. Mutat Res. 681(1):3–12.
  • Speit G, Schütz P, Bonzheim I, Trenz K, Hoffmann H. 2004. Sensitivity of the FPG protein towards alkylation damage in the comet assay. Toxicol Lett. 146(2):151–158.
  • Srám RJ, Beskid O, Binkova B. 2004. Cytogenetic analysis using fluorescence in situ hybridization (FISH) to evaluate occupational exposure to carcinogens. Toxicol Lett. 149:335–344.
  • Stankowski LF, Jr, Lallier B, Tydrick C, Baba R, Tincher S, Callupe J, Kwok VK. 2019. Re-evaluation of discordant results in related OECD TG741 tester strains. https://www.criver.com/sites/default/files/resource-files/SP-EMGS-18-re-evaluation-of-discordant-results-in-related-OECD-TG471-tester-strains.pdf.
  • Strum JM, Shear CR. 1982. Harderian glands in mice: fluorescence, peroxidase activity and fine structure. Tissue Cell. 198214(1):135–148.
  • Styles JA, Clay P, Cross MF. 1985. Assays for the induction of gene mutations at the thymidine kinase and the Na+/K + ATPase loci in two different mouse lymphoma cell lines in culture. Prog Mutat Res. 5:587–596.
  • Sumner SC, Selvaraj L, Nauhaus SK, Fennell TR. 1997. Urinary metabolites from F344 rats and B6C3F1 mice coadministered acrylamide and acrylonitrile for 1 or 5 days. Chem Res Toxicol. 10(10):1152–1160.
  • Sumner SC, Fennell TR, Moore TA, Chanas B, Gonzalez F, Ghanayem BI. 1999. Role of cytochrome P450 2E1 in the metabolism of acrylamide and acrylonitrile in mice. Chem Res Toxicol. 12(11):1110–1116.
  • Swaen GMH, Bloemen LJN, Twisk J, Scheffers T, Slangen JJM, Collins JJ, ten Berge WFJP. 2004. Mortality update of workers exposed to acrylonitrile in the Netherlands. J Occup Environ Med. 46(7):691–698.
  • Swenberg JA, Lu K, Moeller BC, Gao L, Upton PB, Nakamura J, Starr TB. 2011. Endogenous versus exogenous DNA adducts: their role in carcinogenesis, epidemiology, and risk assessment. Toxicol Sci. 120(Suppl 1):S130–S145.
  • Symons JM, Kreckmann KH, Sakr CJ, Kaplan AM, Leonard RC. 2008. Mortality among workers exposed to acrylonitrile in fiber production: an update. J Occup Environ Med. 50(5):550–560.
  • Szumiel I. 2005. L5178Y sublines: a look back from 40 years. Part 1: general characteristics. Int J Radiat Biol. 81(5):339–352.
  • Tandon R, Saxena DK, Chandra SV, Seth PK, Srivastava SP. 1988. Testicular effects of acrylonitrile in mice. Toxicol Lett. 42(1):55–63.
  • Tavares R, Borba H, Monteiro M, Proença MJ, Lynce N, Rueff J, Bailey E, Sweetman GM, Lawrence RM, Farmer PB, et al. 1996. Monitoring of exposure to acrylonitrile by determination of N-(2-cyanoethyl)valine at the N-terminal position of haemoglobin. Carcinogenesis. 17(12):2655–2660.
  • Thier R, Balkenhol H, Lewalter J, Selinski S, Dommermuth A, Bolt HM. 2001. Influence of polymorphisms of the human glutathione transferases and cytochrome P450 2E1 enzyme on the metabolism and toxicity of ethylene oxide and acrylonitrile. Mutat Res. 482(1–2):41–46. 6.
  • Thier R, Lewalter J, Kempkes M, Selinski S, Brüning T, Bolt HM. 1999. Haemoglobin adducts of acrylonitrile and ethylene oxide in acrylonitrile workers, dependent on polymorphisms of the glutathione transferases GSTT1 and GSTM1. Arch Toxicol. 73(4–5):197–202.
  • Thiess AM, Fleig I. 1978. Analysis of chromosomes of workers exposed to acrylonitrile. Arch Toxicol. 41(2):149–152.
  • Tonacchera M, Pinchera A, Dimida A, Ferrarini E, Agretti P, Vitti P, Santini F, Crump K, Gibbs J. 2004. Relative potencies and additivity of perchlorate, thiocyanate, nitrate, and iodide on the inhibition of radioactive iodide uptake by the human sodium iodide symporter. Thyroid. 14(12):1012–1019.
  • TRL (Toxicological Research Laboratory). 1975. Results of a reproduction study in rats maintained on drinking water containing acrylonitrile. Unpublished report. Midland (MI): Dow Chemical Company, Health and Environmental Research.
  • Trosko JE. 2001. Commentary: Is the concept of “tumor promotion” a useful paradigm? Mol Carcinog. 30(3):131–137.
  • Turker MS, Gage BM, Rose JA, Elroy D, Ponomareva ON, Stambrook PJ, Tischfield JA. 1999. A novel signature mutation for oxidative damage resembles a mutational pattern found commonly in human cancers. Cancer Res. 59(8):1837–1839.
  • USEPA. 2011. Toxicological review of acrylonitrile (external review draft). Archived.
  • Valko M, Rhodes CJ, Moncol J, Izakovic M, Mazur M. 2006. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact. 160(1):1–40.
  • Ved Brat S, Williams GM. 1982. Hepatocyte-mediated production of sister chromatid exchange in co-cultured cells by acrylonitrile: evidence for extra cellular transport of a stable reactive intermediate. Cancer Lett. 17(2):213–216.
  • Venitt S. 1978. Letter on acrylonitrile mutagenicity. Mutat Res. 57(1):107–113.
  • Venitt S, Bushell CT, Osborne M. 1977. Mutagenicity of acrylonitrile (cyanoethylene) in Escherichia coli. Mutat Res. 45(2):283–288.
  • Vodicka P, Gut I, Frantík E. 1990. Effects of inhaled acrylic acid derivatives in rats. Toxicology. 65(1–2):209–221.
  • Wakata A, Miyamae Y, Sato S-i, Suzuki T, Morita T, Asano N, Awogi T, Kondo K, Hayashi M. 1998. Evaluation of the rat micronucleus test with bone marrow and peripheral blood: summary of the 9th collaborative study by CSGMT/JEMS.MMS. Environ Mol Mutagen. 32(1):84–100.
  • Walker VE, Fennell TR, Walker DM, Bauer MJ, Upton PB, Douglas GR, Swenberg JA. 2020. Analysis of DNA adducts and mutagenic potency and specificity in rats exposed to acrylonitrile. Chem Res Toxicol. 33(7):1609–1622.
  • Walker VE, Walker DM, Ghanayem BI, Douglas GR. 2020. Analysis of biomarkers of DNA Damage and mutagenicity in mice exposed to acrylonitrile. Chem Res Toxicol. 33(7):1623–1632.
  • Wallace SS. 2002. Biological consequences of free radical-damaged DNA bases. Free Radic Biol Med. 33(1):1–14.
  • Wang CY, Liu LN, Zhao ZB. 2013. The role of ROS toxicity in spontaneous aneuploidy in cultured cells. Tissue Cell. 45(1):47–53.
  • Wang Z, Li Z, Wei X. 1995. Study on acrylonitrile to teratogenesis of rat sperm. J Lanzhou Med Coll. 21:3.
  • Wang H, Chanas B, Ghanayem BI. 2002. Cytochrome P450 2E1 (CYP2E1) is essential for acrylonitrile metabolism to cyanide: comparative studies using CYP2E1-null and wild-type mice. Drug Metab Dispos. 30(8):911–917.
  • Warneke C, Roberts JM, Veres P, Gilman J, Kuster WC, Burling I, Yokelson R, de Gouw JA. 2011. VOC identification and inter-comparison from laboratory biomass burning using PTR-MS and PIT-MS. Int J Mass Spectrom. 303(1):6–14.
  • White WE, Jr, Pruitt KM, Mansson-Rahemtulla B. 1983. Peroxidase-thiocyanate-peroxide antibacterial system does not damage DNA. Antimicrob Agents Chemother. 23(2):267–272.
  • Whittaker SG, Zimmermann FK, Dicus B, Piegorsch WW, Resnick MA, Fogel S. 1990. Detection of induced mitotic chromosome loss in Saccharomyces cerevisiae–an interlaboratory assessment of 12 chemicals. Mutat Res. 241(3):225–242.
  • Whysner J, Steward RE, Chen D, Conaway CC, Verna LK, Richie JP, Ali N, Williams GM. 1998. Formation of 8-oxodeoxyguanosine in brain DNA of rats exposed to acrylonitrile. Arch Toxicol. 72(7):429–438.
  • Willhite CC, Ferm VH, Smith RP. 1981a. Teratogenic effects of aliphatic nitriles. Teratology. 23(3):317–323.
  • Willhite CC, Marin-Padilla M, Ferm VH, Smith RP. 1981. Morphogenesis of axial skeletal (dysraphic) disorders induced by aliphatic nitriles. Teratology. 23(3):325–333.
  • Williams BJ, Ballenger CA, Malter HE, Bishop F, Tucker M, Zwingman TA, Hassold TJ. 1993. Non-disjunction in human sperm: results of fluorescence in situ hybridization studies using two and three probes. Hum Mol Genet. 2(11):1929–1936.
  • Williams GM, Kobets T, Duan JD, Iatropoulos MJ. 2017. Assessment of DNA binding and oxidative DNA damage by acrylonitrile in two rat target tissues of carcinogenicity: implications for the mechanism of action. Chem Res Toxicol. 30(7):1470–1480.
  • Williams GM, Tong CV, Brat S. 1985. Tests with the rat hepatocyte primary culture/DNA-repair test. Prog Mutat Res. 5:341–345.
  • Wilson DM, Thompson LH. 2007. Molecular mechanisms of sister-chromatid exchange. Mutat Res. 616(1–2):11–23.
  • Wilson RH, McCormick WE. 1949. Acrylonitrile; its physiology and toxicology. Ind Med Surg. 18(6):243–245.
  • Wolff J. 1998. Perchlorate and the thyroid gland. Pharmacol Rev. 50(1):89–105.
  • Working PK, Bentley KS, Hurtt ME, Mohr KL. 1987. Comparison of the dominant lethal effects of acrylonitrile and acrylamide in male F344 rats. Mutagenesis. 2(3):215–220.
  • Wu WK, Su J, Huang MY. 1994. Epidemiological investigation of reproductive outcomes in acrylonitrile-exposed male workers. J Clio Ind Hygiene Occup Med. 12:261–263.
  • Wu X, Jin T. 2000. An overview of the study of acrylonitrile on reproductive toxicology. J Labour Med. 17(1):51–52.
  • Wu W, Su J, Huang M. 1995. An epidemiological study on reproductive effects in female workers exposed to acrylonitrile. Zhonghua Yu Fang Yi Xue Za Zhi (China Prevent Med Mag). 29:83–85. (Chinese).
  • Xu D-X, Zhu Q-X, Zheng L-K, Wang Q-N, Shen H-M, Deng L-X, Ong C-N. 2003. Exposure to acrylonitrile induced DNA strand breakage and sex chromosome aneuploidy in human spermatozoa. Mutat Res. 537(1):93–100.
  • Yang B, Zhao W, Yin C, Bai Y, Wang S, Xing G, Li F, Bian J, Aschner M, Cai J, et al. 2021. Acute acrylonitrile exposure inhibits endogenous H2S biosynthesis in rat brain and liver: the role of CBS/3-MPST-H2S pathway in its astrocytic toxicity. Toxicology. 451:152685.
  • Yang J, Duerksen-Hughes P. 1998. A new approach to identifying genotoxic carcinogens: p53 induction as an indicator of genotoxic damage. Carcinogenesis. 19(6):1117–1125.
  • Yates JM, Fennell TR, Turner MJ, Recio L, Sumner SC. Jr 1994. Characterization of phosphodiester adducts produced by the reaction of cyanoethylene oxide with nucleotides. Carcinogenesis. 15(2):277–283.,
  • Yates JM, Summer SC, Turner MJ, Recio L, Fennell TR. 1993. Characterization of an adduct and its degradation product produced by the reaction of cyanoethylene oxide with deoxythymidine and DNA. Carcinogenesis. 14(7):1363–1369.
  • Yauk CL, Aardema MJ, van Benthem J, Bishop JB, Dearfield KL, DeMarini DM, Dubrova YE, Honma M, Lupski JR, Marchetti F, et al. 2015. Approaches for identifying germ cell mutagens: report of the 2013 IWGT workshop on germ cell assays(⋆). Mutat Res Genet Toxicol Environ Mutagen. 783:36–54.
  • Yokelson RJ, Karl T, Artaxo P, Blake DR, Christian TJ, Griffith David WT, Guenther A, Hao WM. 2007. The tropical forest and fire emissions experiment: overview and airborne fire emission factor measurements. Chemistry and Biochemistry Faculty Publications. 3. https://scholarworks.umt.edu/chem_pubs/3.
  • Zeiger E, Haworth S. 1985. Tests with a preincubation modification ofthe Salmonella/microsome assay. In: Ashby J, de Serres FJ, Draper M, Ishidate M, Jr, Margolin BH, Matter BE, Shelby MD, editors. Progress in mutation research: evaluation of short-term tests for carcinogens; report of the international programme on chemical safety’s collaborative study on in vitro assays. Vol. 5. Amsterdam: Elsevier Science Publishers; p. 187–199.
  • Zhang H, Kamendulis LM, Klaunig JE. 2002. Mechanisms for the induction of oxidative stress in Syrian hamster embryo cells by acrylonitrile. Toxicol Sci. 67(2):247–255.
  • Zhang H, Kamendulis LM, Jiang J, Xu Y, Klaunig JE. 2000. Acrylonitrile-induced morphological transformation in Syrian hamster embryo cells. Carcinogenesis. 21(4):727–733.
  • Zhurkov VS, Shram RY, Dugan AM. 1983. Analysis of the mutagenic activity of acrylonitrile. Gig Sanit. 1:71–72.
  • Zimmermann FK, Mayer VW, Scheel I, Resnick MA. 1985. Acetone, methyl ethyl ketone, ethyl acetate, acetonitrile and other polar aprotic solvents are strong inducers of aneuploidy in Saccharomyces cerevisiae. Mutat Res. 149(3):339–351.