579
Views
12
CrossRef citations to date
0
Altmetric
Review Article

Genotoxic and mutagenic studies of teratogens in developing rat and mouse

ORCID Icon & ORCID Icon
Pages 409-429 | Received 08 Jan 2018, Accepted 13 Apr 2018, Published online: 10 May 2018

References

  • Adhikari, N. and Grover, I.S., 1988. Genotoxic effects of some systemic pesticides: in vivo chromosomal aberrations in bone marrow cells in rats. Environmental and Molecular Mutagenesis, 12, 235–242.
  • Adler, I.D., 1983. New approaches to mutagenicity studies in animals for carcinogenic and mutagenic agents. II. Clastogenic effects determined in transplacentally treated mouse embryos. Teratogenesis, Carcinogenesis, and Mutagenesis, 3, 321–334.
  • Aguilar-Mahecha, A., Hales, B.F., and Robaire, B., 2005. Effects of acute and chronic cyclophosphamide treatment on meiotic progression and the induction of DNA double-strand breaks in rat spermatocytes. Biology of Reproduction, 72, 1297–1304.
  • Albertini, R.J., et al., 2000. IPCS guidelines for the monitoring of genotoxic effects of carcinogens in humans. International Programme on Chemical Safety. Mutation Research, 463, 111–172.
  • Al-Harbi, M.M., 1993. Effect of captopril on the cytological and biochemical changes induced by adriamycin. Food and Chemical Toxicology, 31, 209–212.
  • Alkan, F.U., and Anlas, C., 2015. Genotoxicity tests and genotoxic poisons. Turkish Clinics Journal of Veterinary Sciences – Pharmacology and Toxicology – Special Topics, 1, 69–74.
  • Al-Shebaily, M.M., 1999. Evaluation of the effects of pregnyl on pituitary-ovarian hormones and biochemical markers of tissue injury in female Swiss albino mice. Research Communications in Molecular Pathology and Pharmacology, 105, 155–171.
  • Alvarez-González, I., et al., 2001. Inhibitory effect of naringin on the micronuclei induced by ifosfamide in mouse, and evaluation of its modulatory effect on the Cyp3a subfamily. Mutation Research, 480–481, 171–178.
  • Alvarez-González, I., Madrigal-Bujaidar, E., and Sánchez-García, V.Y., 2010. Inhibitory effect of grapefruit juice on the genotoxic damage induced by ifosfamide in mouse. Plant Foods for Human Nutrition (Dordrecht, Netherlands), 65, 369–373.
  • Alzahrani, H.A., 2012. Sister chromatid exchanges and sperm abnormalities produced by antidepressant drug fluoxetine in mouse treated in vivo. European Review for Medical and Pharmacological Sciences, 16, 2154–2161.
  • Amador, R.R., et al., 2012. Metformin (dimethyl-biguanide) induced DNA damage in mammalian cells. Genetics and Molecular Biology, 35, 153–158.
  • Amer, S.M. and Aly, F.A., 2001. Genotoxic effect of 2,4-dichlorophenoxy acetic acid and its metabolite 2,4-dichlorophenol in mouse. Mutation Research, 494, 1–12.
  • Anderson, D., 1988. Human biomonitoring. Mutation Research, 204, 353–541.
  • Antunes, L.M., et al., 2000. Effects of selenium pretreatment on cisplatin-induced chromosome aberrations in wistar rats. Teratogenesis, Carcinogenesis, and Mutagenesis, 20, 341–348.
  • Ashby, J., et al., 1994. Potent clastogenicity of the human carcinogen etoposide to the mouse bone marrow and mouse lymphoma L5178Y cells: comparison to Salmonella responses. Environmental and Molecular Mutagenesis, 24, 51–60.
  • Ashby, J., et al., 1997. Lack of activity of estradiol in rodent bone marrow micronucleus assays. Mutation Research, 395, 83–88.
  • Ashby, J. and Beije, B., 1985. Concomitant observations of UDS in the liver and micronuclei in the bone marrow of rats exposed to cyclophosphamide or 2-acetylaminofluorene. Mutation Research, 150, 383–392.
  • Atienzar, F.A. and Jha, A.N., 2006. The random amplified polymorphic DNA (RAPD) assay and related techniques applied to genotoxicity and carcinogenesis studies: a critical review. Mutation Research, 613, 76–102.
  • Attia, S., 2012. Modulation of irinotecan-induced genomic DNA damage by theanine. Food and Chemical Toxicology, 50, 1749–1754.
  • Attia, S.M., 2010. The impact of quercetin on cisplatin-induced clastogenesis and apoptosis in murine marrow cells. Mutagenesis, 25, 281–288.
  • Attia, S.M., 2012. Dominant lethal mutations of topoisomerase II inhibitors etoposide and merbarone in male mice: a mechanistic study. Archives of Toxicology, 86, 725–731.
  • Attia, S.M., et al., 2013. Germ cell mutagenicity of topoisomerase I inhibitor topotecan detected in the male mouse-dominant lethal study. Food and Chemical Toxicology, 62, 470–474.
  • Attia, S.M. and Bakheet, S.A., 2013. Citalopram at the recommended human doses after long-term treatment is genotoxic for male germ cell. Food and Chemical Toxicology, 53, 281–285.
  • Attia, S.M., Helal, G.K., and Alhaider, A.A., 2009. Assessment of genomic instability in normal and diabetic rats treated with metformin. Chemico-Biological Interactions, 180, 296–304.
  • Au, W.W. and Hsu, T.C., 1980. The genotoxic effects of adriamycin in somatic and germinal cells of the mouse. Mutation Research, 79, 351–361.
  • Aydemir, N. and Bilaloglu, R., 2003. Genotoxicity of two anticancer drugs, gemcitabine and topotecan, in mouse bone marrow in vivo. Mutation Research, 537, 43–51.
  • Bach, J., et al., 2014. Ogg1 genetic background determines the genotoxic potential of environmentally relevant arsenic exposures. Archives of Toxicology, 88, 585–596.
  • Barcellona, P.S., et al., 1987. Correlations between embryotoxic and genotoxic effects of phenytoin in mice. Teratogenesis, Carcinogenesis, and Mutagenesis, 7, 159–168.
  • Barnett, L.B., et al., 1992. Ethylene dibromide: negative results with the mouse dominant lethal assay and the electrophoretic specific-locus test. Mutation Research, 282, 127–133.
  • Barnett, L.B. and Lewis, S.E., 2003. Chlornaphazine and chlorambucil induce dominant lethal mutations in male mice. Mutation Research, 543, 145–154.
  • Barton, T.S., et al., 2003. Numerical chromosomal abnormalities in rat epididymal spermatozoa following chronic cyclophosphamide exposure. Biology of Reproduction, 69, 1150–1157.
  • Basler, A., Thesis, I., and Röhrborn, G., 1979. Cytogenetic effects of busulfan in vivo on bone marrow cells and oocytes of adult mice and liver cells of transplacentally exposed embryos. Environmental Mutagenesis, 1, 233–238.
  • Baumann, A., et al., 1996. Use of rat and human liver slices for the detection of steroid hormone-induced DNA-adducts in vitro by means of the (32)P-postlabeling technique. Pharmacology & Toxicology, 78, 214–223.
  • Bayram, S., et al., 2016. Genotoxicity and cytotoxicity of copper oxychloride in cultured human lymphocytes using cytogenetic and molecular tests. Cytotechnology, 68, 2027–2036.
  • Becerril, C., et al., 1999. Detection of mitomycin C-induced genetic damage in fish cells by use of RAPD. Mutagenesis, 14, 449–456.
  • Benford, D.J., et al., 1994. Investigations of the genotoxicity and cell proliferative activity of dichlorvos in mouse forestomach. Toxicology, 92, 203–215.
  • Bentley, K.S. and Working, P.K., 1988. Activity of germ-cell mutagens and nonmutagens in the rat spermatocyte UDS assay. Mutation Research, 203, 135–142.
  • Bishop, J.B. and Wassom, J.S., 1986. Toxicological review of busulfan (Myleran). Mutation Research, 168, 15–45.
  • Bjørge, C., et al., 1996. A comparative study of chemically induced DNA damage in isolated human and rat testicular cells. Reproductive Toxicology (Elmsford, N.Y.), 10, 509–519.
  • Bodell, W.J., et al., 2003. Formation of DNA adducts and induction of lacI mutations in Big Blue Rat-2 cells treated with temozolomide: implications for the treatment of low-grade adult and pediatric brain tumors. Cancer Epidemiology, Biomarkers & Prevention, 12, 545–551.
  • Bolzán, A.D. and Bianchi, M.S., 2002. Genotoxicity of streptozotocin. Mutation Research, 512, 121–134.
  • Bonassi, S., et al., 2000. Chromosomal aberrations in lymphocytes predict human cancer independently of exposure to carcinogens. European Study Group on Cytogenetic Biomarkers and Health. Cancer Research, 60, 1619–1625.
  • Bonassi, S., et al., 2004. Chromosomal aberrations and risk of cancer in humans: an epidemiologic perspective. Cytogenetic and Genome Research, 104, 376–382.
  • Bonassi, S., et al., 2005. Human population studies with cytogenetic biomarkers: review of the literature and future prospectives. Environmental and Molecular Mutagenesis, 45, 258–270.
  • Bonassi, S., et al., 2007. An increased micronucleus frequency in peripheral blood lymphocytes predicts the risk of cancer in humans. Carcinogenesis, 28, 625–631.
  • Boos, G. and Stopper, H., 2000. Genotoxicity of several clinically used topoisomerase II inhibitors. Toxicology Letters, 116, 7–16.
  • Bouslimi, A., et al., 2008. Individual and combined effects of ochratoxin A and citrinin on viability and DNA fragmentation in cultured Vero cells and on chromosome aberrations in mice bone marrow cells. Toxicology, 251, 1–7.
  • Bowen, D.E., et al., 2011. Evaluation of a multi-endpoint assay in rats, combining the bone-marrow micronucleus test, the Comet assay and the flow-cytometric peripheral blood micronucleus test. Mutation Research, 722, 7–19.
  • Boyer, I.J., 1989. Toxicity of dibutyltin, tributyltin and other organotin compounds to humans and to experimental animals. Toxicology, 55, 253–298.
  • Brambilla, G. and Martelli, A., 2002. Are some progestins genotoxic liver carcinogens? Mutation Research, 512, 155–163.
  • Bremer, S. and Hartung, T., 2004. The use of embryonic stem cells for regulatory developmental toxicity testing in vitro-the current status of test development. Current Pharmaceutical Design, 10, 2733–2747.
  • Brown, N.A., et al., 1995. Screening chemicals for reproductive toxicity: the current alternatives. Alternatives to Laboratory Animals, 23, 868–882.
  • Brunborg, G., et al., 1988. An automated alkaline elution system: DNA damage induced by 1,2-dibromo-3-chloropropane in vivo and in vitro. Analytical Biochemistry, 174, 522–536.
  • Brunborg, G., et al., 1990. Organ-specific genotoxic effects of chemicals: the use of alkaline elution to detect DNA damage in various organs of in vivo-exposed animals. Progress in Clinical and Biological Research, 340, 43–52.
  • Brusick, D., et al., 1989. Assessment of the genotoxicity of calcium cyclamate and cyclohexylamine. Environmental and Molecular Mutagenesis, 14, 188–199.
  • Buenaventura, S.K., et al., 1984. Induction of sister chromatid exchange by diethylstilbestrol in metabolically competent hepatoma cell lines but not in fibroblasts. Cancer Research, 44, 3851–3855.
  • Calviello, G., et al., 2006. DNA damage and apoptosis induction by the pesticide Mancozeb in rat cells: involvement of the oxidative mechanism. Toxicology and Applied Pharmacology, 211, 87–96.
  • Cammerer, Z., Elhajouji, A., and Suter, W., 2007a. In vivo micronucleus test with flow cytometry after acute and chronic exposures of rats to chemicals. Mutation Research, 626, 26–33.
  • Cammerer, Z., et al., 2007b. Comparison of the peripheral blood micronucleus test using flow cytometry in rat and mouse exposed to aneugens after single-dose applications. Mutagenesis, 22, 129–134.
  • Campos, P.B., et al., 2012. Cycle arrest and aneuploidy induced by zidovudine in murine embryonic stem cells. Mutagenesis, 27, 431–436.
  • Carrano, A.V., et al., 1978. Sister chromatid exchanges as an indicator of mutagenesis. Nature, 271, 551–553.
  • Carrano, A.V. and Natarajan, A.T., 1988. Consideration for population monitoring using cytogenetic techniques. Mutation Research, 204, 379–406.
  • Carvalho, I.C., et al., 2016. High doses of alcohol during pregnancy cause DNA damages in osteoblasts of newborns rats. Birth Defects Research Part A, Clinical and Molecular Teratology, 106, 122–132.
  • Caspary, W.J., et al., 1987. Mutagenic activity of fluorides in mouse lymphoma cells. Mutation Research, 187, 165–180.
  • Cebral, E., Abrevaya, X.C., and Mudry, M.D., 2011. Male and female reproductive toxicity induced by sub-chronic ethanol exposure in CF-1 mice. Cell Biology and Toxicology, 27, 237–248.
  • Chapin, R.E., Sloane, R.A., and Haseman, J.K., 1997. The relationships among reproductive endpoints in Swiss mice, using the reproductive as-sessment by continuous breeding database. Toxicological Sciences, 38, 129–142.
  • Charles, J.M., et al., 1999. Ames assays and unscheduled DNA synthesis assays on 2, 4-dichlorophenoxyacetic acid and its derivatives. Mutation Research, 444, 207–216.
  • Cheng, T.J.1., et al., 1996. Increased micronucleus frequency in lymphocytes from smokers with lung cancer. Mutation Research, 349, 43–50.
  • Cheng, M., Conner, M.K., and Alaria, Y., 1981. Potency of some carbamates as multiple tissue sister chromatid exchanges in Bloom’s Syndrome lymphocytes. Cancer Research, 71, 4508–4512.
  • Chinnasamy, N., et al., 1988. Modulation of O 6 -alkylating agent induced clastogenicity by enhanced DNA repair capacity of bone marrow cells. Mutation Research, 416, 1–10.
  • Chłopkiewicz, B. and Gruber, B., 1997. The in vitro study on genotoxic activity of adriamycin and bleomycin in cells of mice with different catalase and superoxide dismutase activity. Acta Poloniae Pharmaceutica, 54, 437–441.
  • Choudhury, R.C., et al., 2002. Spermatogonial cytogenetic toxicity of vincristine and its transmission in the germline cells of Swiss mice. Journal of Environmental Pathology, Toxicology and Oncology, 21, 249–257.
  • Ciaravino, V., et al., 1995. The genotoxicity profile of atorvastatin, a new drug in the treatment of hypercholesterolemia. Mutation Research, 343, 95–107.
  • Cicchetti, R., Bari, M., and Argentin, G., 1999. Induction of micronuclei in bone marrow by two pesticides and their differentiation with CREST staining: an in vivo study in mice. Mutation Research, 439, 239–248.
  • Clive, D., 1973. Recent developments with the L5178Y TK heterozygote mutagen assay system. Environmental Health Perspectives, 6, 119–125.
  • Clive, D., et al., 1990. Molecular aspects of chemical mutagenesis in L5178Y/tk +/- mouse lymphoma cells. Mutagenesis, 5, 191–197.
  • Clive, D., Turner, N., and Krehl, R., 1988. Procarbazine is a potent mutagen at the heterozygous thymidine kinase (tk +/-) locus of mouse lymphoma assay. Mutagenesis, 3, 83–87.
  • Coelho, V.R., et al., 2016. Genotoxic and mutagenic effects of vigabatrin, a γ-aminobutyric acid transaminase inhibitor, in Wistar rats submitted to rotarod task. Human & Experimental Toxicology, 35, 958–965.
  • Coffing, S., et al., 2011. The rat gut micronucleus assay: a good choice for alternative in vivo genetic toxicology testing strategies. Environmental and Molecular Mutagenesis, 52, 269–279.
  • Cohen, A.M., and Prabhakar, H., 1983. Carcinogen induced DNA damage in isolated rat liver nuclei. Cancer Letters, 18, 163–167.
  • Cole, J., Muriel, W.J., and Bridges, B.A., 1986. The mutagenicity of sodium fluoride to L5178Y [wild-type and TK+/- (3.7.2c)] mouse lymphoma cells. Mutagenesis, 1, 157–167.
  • Cook, C.S., et al., 1988. Oppermann Difference in metabolic profile of potassium canrenoate and spironolactone in the rat: mutagenic metabolites unique to potassium canrenoate. Archives of Toxicology, 61, 201–212.
  • Cooper, G.M. and Hausman, R.E., 2004. The cell: a molecular approach. Washington (DC): ASM Press [Turkish edition Sakizli M, Atabey N (2006) Hucre: molekuler yaklasim. Izmir, Turkey: İzmir Tıp Kitabevi].
  • Dallegrave, E., et al., 2003. The teratogenic potential of the herbicide glyphosate-Roundup in Wistar rats. Toxicology Letters, 142, 45–52.
  • Dasgupta, Y., et al., 2016. Normal ABL1 is a tumor suppressor and therapeutic target in human and mouse leukemias expressing oncogenic ABL1 kinases. Blood, 127, 2131–2143.
  • Davis, A., et al., 1987. Evaluation of the genetic and embryotoxic effects of bis(tri-n-butyltin)oxide (TBTO), a broad-spectrum pesticide, in multiple in vivo and in vitro short-term tests. Mutation Research, 188, 65–95.
  • Dearfield, K.L., et al., 1985. In vitro assays of in vivo exposure to cyclophosphamide: induction of sister-chromatid exchanges in peripheral lymphocytes, bone-marrow cells and in cultured cells exposed to plasma. Mutation Research, 158, 97–104.
  • Deknudt, G., et al., 1986. In vivo studies in male mice on the mutagenic effects of inorganic arsenic. Mutagenesis, 1, 33–39.
  • Dellarco, V.L., Mavournin, K.H., and Tice, R.R., 1985. Aneuploidy and health risk assessment: current status and future directions. Environmental Mutagenesis, 7, 405–424.
  • DeMarini, D.M., et al., 1987. Mutagenicity and clastogenicity of teniposide (VM-26) in L5178Y/TK +/− -3.7.2C mouse lymphoma cells. Mutation Research, 187, 141–149.
  • DeMarini, D.M., et al., 1987. Mutagenicity of actinomycin D in mammalian cells due to clastogenic effects. Mutation Research, 192, 151–155.
  • Deng, D.J., 1993. Modulation of mutagenic drug-induced unscheduled DNA synthesis (UDS) in primary rat hepatocytes by diallyl trisulfide. Zhonghua Zhong Liu Za Zhi, 15, 423–426.
  • Dertinger, S.D., et al., 2014. Pig-a gene mutation and micronucleated reticulocyte induction in rats exposed to tumorigenic doses of the leukemogenic agents chlorambucil, thiotepa, melphalan, and 1,3-propane sultone. Environmental and Molecular Mutagenesis, 55, 299–308.
  • Dhillon, V.S., et al., 1995. In vitro and in vivo genotoxicity of hormonal drugs. VI. Fluoxymesterone. Mutation Research, 342, 103–111.
  • Dhillon, V.S. and Dhillon, I.K., 1995. Genotoxicity evaluation of estradiol. Mutation Research, 345, 87–95.
  • Dhillon, V.S. and Dhillon, I.K., 1996. Genotoxicity evaluation of norethisterone acetate. Mutation Research, 367, 1–10.
  • Di Renzo, F., et al., 2011. Is the amphibian X. laevis WEC a good alternative method to rodent WEC teratogenicity assay? The example of the three triazole derivative fungicides Triadimefon, Tebuconazole, Cyproconazole. Reproduction Toxicology, 32, 220–226.
  • Dobrovolsky, V.N., Shaddock, J.G., and Heflich, R.H., 2002. Mutagenicity of gamma-radiation, mitomycin C, and etoposide in the Hprt and Tk genes of Tk(+/-) mice. Environmental and Molecular Mutagenesis, 39, 342–347.
  • Doherty, A.T., et al., 2012. Chromosome aberration frequency in rat peripheral lymphocytes increases with repeated dosing with hexamethylphosphoramide or cyclophosphamide. Mutagenesis, 27, 533–539.
  • Duffaud, F., et al., 1997. Comparison between micronucleated lymphocytes rates observed in healthy subject and cancer patients. Mutagenesis, 12, 227–231.
  • Dunipace, A.J., et al., 1989. Genotoxic evaluation of chronic fluoride exposure: micronucleus and sperm morphology studies. Journal of Dental Research, 68, 1525–1528.
  • Duran, B., et al., 2006. In vivo evaluation of the genotoxic effects of clomiphene citrate on rat reticulocytes: a micronucleus genotoxicity. Gynecologic and Obstetric Investigation, 61, 228–231.
  • Dusman, E., et al., 2014. Cytotoxicity and mutagenicity of fluoxetine hydrochloride (Prozac), with or without vitamins A and C, in plant and animal model systems. Genetics and Molecular Research, 13, 578–589.
  • Edelweiss, M.I., et al., 1995. Clastogenic effect of cisplatin on Wistar rat bone marrow cells. Brazilian Journal of Medical and Biological Research, 28, 679–683.
  • El-Ashmawy, I.M., el-Nahas, A.F., and Bayad, A.E., 2011. Teratogenic and cytogenetic effects of ivermectin and its interaction with P-glycoprotein inhibitor. Research in Veterinary Science, 90, 116–123.
  • El-Shershaby, A.F., et al., 2014. In utero exposure to itraconazole during different gestational periods of rats. Toxicology Mechanisms and Methods, 24, 50–59.
  • Epstein, S.S., et al., 1972. Detection of chemical mutagens by the dominant lethal assay in the mouse. Toxicology and Applied Pharmacology, 23, 288–325.
  • Etebari, M., et al., 2016. Genotoxicity evaluation of hydroalcoholic and aqueous extracts of Dorema aucheri by the comet assay. Advanced Biomedical Research, 5, 199.
  • Evans, H.J., 1984. Human peripheral blood lymphocytes for the analysis of chromosome aberrations in mutagen tests. In: B.J. Kilbey, M. Legator, W. Nichols, and C. Ramel, eds. Handbook of mutagenicity test procedures. 2nd ed. Amsterdam: Elsevier, 405–427.
  • Ezzi, L., et al., 2016. Histopathological and genotoxic effects of chlorpyrifos in rats. Environmental Science and Pollution Research International, 23, 4859–4867.
  • Farag, A.T., El Okazy, A.M., and El-Aswed, A.F., 2003. Developmental toxicity study of chlorpyrifos in rats. Reproductive Toxicology (Elmsford, N.Y.), 17, 203–208.
  • FDA. 2017a. US Food and Drug Administration, Drug Classes. Available from: https://www.drugs.com/drug-class [Accessed 25 Feb 2017].
  • FDA. 2017b. US Food and Drug Administration, Content and Format of Labeling for Human Prescription Drug and Biological Products; Requirements for Pregnancy and Lactation Labeling. Available from: https://www.federalregister.gov/documents/2008/05/29/E8-11806/content-and-format-of-labeling-for-human-prescription-drug-and-biological-products-requirements-for [Accesssed 29 Mar 2017].
  • Fenech, M., 2000. The in vitro micronucleus technique. Mutation Research, 455, 81–95.
  • Fenech, M., et al., 1999. The human Micronucleus project-An international collaborative study on the use of the micronucleus technique for measuring DNA damage in humans. Mutation Research, 428, 271–283.
  • Ferjani, H., et al., 2016. Combination of tacrolimus and mycophenolate mofetil induces oxidative stress and genotoxicity in spleen and bone marrow of Wistar rats. Mutation Research, 810, 48–55.
  • Flajs, D. and Peraica, M., 2009. Toxicological properties of citrinin. Arhiv Za Higijenu Rada i Toksikologiju, 60, 457–464.
  • Fluoride Action Network, 2017. Teratogenic Fluorinated and Fluoride Pesticides. Available from: https://www.fluoridealert.org/wp-content/pesticides/effects.teratogenic.htm [Accessed 21 Mar 2017].
  • Foote, R.H. and Carney, E.W., 2000. The rabbit as a model for reproductive and developmental toxicity studies. Reproductive Toxicology (Elmsford, N.Y.), 14, 477–493.
  • Fucic, A., et al., 2008. Developmental and transplacental genotoxicology: fluconazole. Mutation Research, 657, 43–47.
  • Fujioka, M., et al., 2016. Examination of in vivo mutagenicity of sodium arsenite and dimethylarsinic acid in gpt delta rats. Journal of Environmental Sciences, 49, 125–130.
  • Garriott, M.L., et al., 1995. The in vivo rat micronucleus test: integration with a 14-day study. Mutation Research, 342, 71–76.
  • Gautam, D.C. and Kapoor, L., 1991. Genotoxic effects of dithane M-45 on the bone marrow cells of mice in vivo. Experientia, 47, 280–282.
  • Gebel, T., et al., 1997. In vivo genotoxicity of selected herbicides in the mouse bone-marrow micronucleus test. Archives of Toxicology, 71, 193–197.
  • Gebel, T.W., 2001. Genotoxicity of arsenical compounds. International Journal of Hygiene and Environmental Health, 203, 249–262.
  • Geiger, H., et al., 2006. Mutagenic potential of temozolomide in bone marrow cells in vivo. Blood, 107, 3010–3011.
  • Georgieva, V., et al., 1990. Genotoxic activity of benomyl in different test systems. Environmental and Molecular Mutagenesis, 16, 32–36.
  • Giurgiovich, A.J., et al., 1996. Cisplatin-DNA adduct formation in maternal and fetal rat tissues after transplacental cisplatin exposure. Carcinogenesis, 17, 1665–1669.
  • Gold, B. and Brunk, G., 1988. The effect of pyrazole, phenobarbital, ethanol and 3-methylcholanthrene pretreatment on the in vivo and in vitro genotoxicity of N-nitrosopyrrolidine. Carcinogenesis, 9, 1001–1005.
  • Goldoni, A., et al., 2014. DNA damage in Wistar rats exposed to dithiocarbamate pesticide mancozeb. Folia Biologica, 60, 202–204.
  • Goldstein, L.S., 1987–1988 Mutagenesis in murine spermatogonia by MOPP therapy. Reproductive Toxicology (Elmsford, N.Y.), 1, 99–103.
  • Gollapudi, B.B., et al., 1999. Evaluation of the genotoxicity of 2,4-dichlorophenoxyacetic acid and its derivatives in mammalian cell cultures. Mutation Research, 444, 217–225.
  • Gollapudi, B.B., Mendrala, A.L., and Linscombe, V.A., 1995. Evaluation of the genetic toxicity of the organophosphate insecticide chlorpyrifos. Mutation Research, 342, 25–36.
  • Gómez-Meda, B.C., et al., 2016. Micronucleated erythrocytes in peripheral blood from neonate rats exposed by breastfeeding to cyclophosphamide, colchicine, or cytosine-arabinoside. BioMed Research International, 2016, 9161648.
  • Grover, I.S. and Malhi, P.K., 1985. Genotoxic effects of some organophosphorous pesticides. I. Induction of micronuclei in bone marrow cells in rat. Mutation Research, 155, 131–134.
  • Guérard, M., et al., 2013. Assessment of the genotoxic potential of azidothymidine in the comet, micronucleus, and Pig-a assay. Toxicological Sciences, 135, 309–316.
  • Guérard, M., et al., 2017. Dose-response relationship of temozolomide, determined by the Pig-a, comet, and micronucleus assay. Archives of Toxicology, 91, 2443–2453.
  • Gupta, R.S., et al., 1987. Etoposide (VP16) and teniposide (VM26): novel anticancer drugs, strongly mutagenic in mammalian but not prokaryotic test systems. Mutagenesis, 2, 179–186.
  • Habas, K., Brinkworth, M.H., and Anderson, D., 2017. In vitro responses to known in vivo genotoxic agents in mouse germ cells. Environmental and Molecular Mutagenesis, 58, 99–107.
  • Hacker-Klom, U.B., Meistrich, M.L., and Göhde, W., 1986. Effect of doxorubicin and 4'-epi-doxorubicin on mouse spermatogenesis. Mutation Research, 160, 39–46.
  • Hagio, S., et al., 2014. Repeated dose liver micronucleus assay using adult mice with multiple genotoxicity assays concurrently performed as a combination test. The Journal of Toxicological Sciences, 39, 437–445.
  • Hagmar, L., et al., 1994. Cancer risk in human predicted by increased levels of chromosomal aberrations in lymphocytes: Nordic Study Group on the Health Risk of Chromosome Damage. Cancer Research, 54, 2919–2922.
  • Hamilelikte ilac kategorileri-teratojen ilaclar (The drug categories during pregnancy-Teratogenic drugs) 2017. Available from: http://www.gebelik.org/dosyalar/ilaclar/teratojen2.html [Accessed 26 Feb 2017].
  • Harrouk, W., et al., 2000. Paternal exposure to cyclophosphamide induces DNA damage and alters the expression of DNA repair genes in the rat preimplantation embryo. Mutation Research, 461, 229–241.
  • Haynes, P., Lambert, T.R., and Mitchell, I.D., 1996. Comparative in-vivo genotoxicity of antiviral nucleoside analogues; penciclovir, acyclovir, ganciclovir and the xanthine analogue, caffeine, in the mouse bone marrow micronucleus assay. Mutation Research - Genetic Toxicology and Environment, 369, 65–74.
  • Heddle, J.A., et al., 1991. Micronuclei as an index of cytogenetic damage: past, present, and future. Environmental and Molecular Mutagenesis, 18, 277–291.
  • Helleday, T., 2003. Pathways for mitotic homologous recombination in mammalian cells. Mutation Research, 532, 103–115.
  • Hewitt, M.J., Mutch, P., and Pratten, M.K., 2005. Potential teratogenic effects of benomyl in rat embryos cultured in vitro. Reproductive Toxicology (Elmsford, N.Y.), 20, 271–280.
  • Hirsimäki, P., Aaltonen, A., and Mäntylä, E., 2002. Toxicity of antiestrogens. The Breast Journal, 8, 92–96.
  • Hoffmann, S.H., et al., 1987. Dominant lethal study of ribavirin in male rats. Mutation Research, 188, 29–34.
  • Holden, H.E., Studwell, D., and Majeska, J.B., 1999. Oxymetholone: I. Evaluation in a comprehensive battery of genetic toxicology and in vitro transformation assays. Toxicologic Pathology, 27, 501–506.
  • Holme, J.A., et al., 1989. Comparative genotoxicities of procarbazine and two deuterated analogs in mammalian cells in vitro and in vivo. Mutagenesis, 4, 355–360.
  • Hooser, S.B., et al., 2000. Cisplatin-DNA adduct formation in rat spermatozoa and its effect on fetal development. Cancer Letters, 151, 71–80.
  • Hosseinimehr, S.J. and Karami, M., 2005. Chemoprotective effects of captopril against cyclophosphamide-induced genotoxicity in mouse bone marrow cells. Archives of Toxicology, 79, 482–486.
  • Hosseinimehr, S.J., Mahmoudzadeh, A., and Akhlagpour, S., 2007. Captopril protects mice bone marrow cells against genotoxicity induced by gamma irradiation. Cell Biochemistry and Function, 25, 389–394.
  • Huang, P.H. and McBride, W.G., 1990. Thalidomide induced alteration in secondary structure of rat embryonic DNA in vivo. Teratogenesis, Carcinogenesis, and Mutagenesis, 10, 281–294.
  • Hundal, B.S., Dhillon, V.S., and Sidhu, I.S., 1997. Genotoxic potential of estrogens. Mutation Research, 389, 173–181.
  • Imanishi, S., et al., 2013. Prenatal exposure to permethrin influences vascular development of fetal brain and adult behavior in mice offspring. Environmental Toxicology, 28, 617–629.
  • Jemilev, Z.A., et al., 1981. Chromosomal analysis of baboons and their mothers, following application to mothers of potentially post-ovulation fertility-inhibiting steroids. Zentralblatt Fur Gynakologie, 103, 1215–1219.
  • Jeswal, P., 1996. Citrinin-induced chromosomal abnormalities in the bone marrow cells of Mus musculus. Cytobios, 86, 29–33.
  • Jirsová, K. and Mandys, V., 1996. Carboplatin-induced micronuclei formation in non-neuronal cells of rat foetal dorsal root ganglia cultured in vitro and comparison with another anticancer drug-cisplatin. Sbornik Lekarsky, 97, 331–342.
  • Kaina, B., 2003. DNA damage-triggered apoptosis: critical role of DNA repair, double-strand breaks, cell proliferation and signaling. Biochemical Pharmacology, 66, 1547–1554.
  • Kallio, M. and Lähdetie, J., 1993. Analysis of micronuclei induced in mouse early spermatids by mitomycin C, vinblastine sulfate or etoposide using fluorescence in situ hybridization. Mutagenesis, 8, 561–567.
  • Karia, B., Martinez, J.A., and Bishop, A.J., 2013. Induction of homologous recombination following in utero exposure to DNA-damaging agents. DNA Repair (Repair), 12, 912–921.
  • Kesari, V.P., Kumar, A., and Khan, P.K., 2012. Genotoxic potential of arsenic at its reference dose. Ecotoxicology and Environmental Safety, 80, 126–131.
  • Khalil, A.M., 1995. Chromosome aberrations in cultured rat bone marrow cells treated with inorganic fluorides. Mutation Research, 343, 67–74.
  • Khalil, A.M. and Da'dara, A.A., 1994. The genotoxic and cytotoxic activities of inorganic fluoride in cultured rat bone marrow cells. Archives of Environmental Contamination and Toxicology, 26, 60–63.
  • Khan, P.K., Kesari, V.P., and Kumar, A., 2013. Mouse micronucleus assay as a surrogate to assess genotoxic potential of arsenic at its human reference dose. Chemosphere, 90, 993–997.
  • Kim, S.Y., et al., 2006. Antiestrogens and the formation of DNA damage in rats: a comparison. Chemical Research in Toxicology, 19, 852–858.
  • Kimoto, T., 2014. Introduction to a special issue on the Pig-a gene mutation assay. Genes and Environment, 36, 167–168.
  • Kindig, D., et al., 1992. Diphenylhydantoin is not genotoxic in a battery of short-term cytogenetic assays. Teratogenesis, Carcinogenesis, and Mutagenesis, 12, 43–50.
  • Kirsch-Volders, M., et al., 1997. The in vitro micronucleus test: A multi-endpoint assay to detect simultaneously mitotic delay, apoptosis, chromosome breakage, chromosome loss and non-disjunction. Mutation Research, 392, 19–30.
  • Kligerman, A.D., et al., 2000. Cytogenetic studies of three triazine herbicides. II. In vivo micronucleus studies in mouse bone marrow. Mutation Research, 471, 107–112.
  • Kligerman, A.D., et al., 2003. Methylated trivalent arsenicals as candidate ultimate genotoxic forms of arsenic: induction of chromosomal mutations but not gene mutations. Environmental and Molecular Mutagenesis, 42, 192–205.
  • Kligerman, A.D., Erexson, G.L., and Wilmer, J.L., 1985. Induction of sister-chromatid exchange (SCE) and cell-cycle inhibition in mouse peripheral blood B lymphocytes exposed to mutagenic carcinogens in vivo. Mutation Research, 157, 181–187.
  • Kosinska, W., et al., 2002. Comparative mutagenicities of bleomycin and ferric-nitrilotriacetate in lacZ mice. Cancer Letters, 187, 41–46.
  • Kraynak, A.R., et al., 2015. Alkaline comet assay in liver and stomach, and micronucleus assay in bone marrow, from rats treated with 2-acetylaminofluorene, azidothymidine, cisplatin, or isobutyraldehyde. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 786-788, 77–86.
  • Krishna, G., et al., 1985. In vivo cytogenetic studies on mice exposed to ethylene dibromide. Mutation Research, 158, 81–87.
  • Krishna, G. and Theiss, J.C., 1995. Concurrent analysis of cytogenetic damage in vivo: a multiple endpoint-multiple tissue approach. Environmental and Molecular Mutagenesis, 25, 314–320.
  • Kumar, S.G., et al., 2006. Dacarbazine induces genotoxic and cytotoxic germ cell damage with concomitant decrease in testosterone and increase in lactate dehydrogenase concentration in the testis. Mutation Research, 607, 240–252.
  • Kuroiwa, Y., et al., 2007. Lack of in vivo mutagenicity and oxidative DNA damage by flumequine in the livers of gpt delta mice. Archives of Toxicology, 81, 63–69.
  • Lähdetie, J., et al., 1994. Etoposide (VP-16) is a potent inducer of micronuclei in male rat meiosis: spermatid micronucleus test and DNA flow cytometry after etoposide treatment. Environmental and Molecular Mutagenesis, 24, 192–202.
  • Lambert, B. and Eriksson, G., 1979. Effects of cancer chemotherapeutic agents on testicular DNA synthesis in the rat. Evaluation of a short-term test for studies of the genetic toxicity of chemicals and drugs in vivo. Mutation Research, 68, 275–289.
  • Larsson, K.S., et al., 1976. Studies of teratogenic effects of the dithiocarbamates maneb, mancozeb, and propineb. Teratology, 14, 171–183.
  • Leite Ade, L., et al., 2007. Absence of DNA damage in multiple organs (blood, liver, kidney, thyroid gland and urinary bladder) after acute fluoride exposure in rats. Human & Experimental Toxicology, 26, 435–440.
  • Lents, N.H., et al., 2008. Identification and characterization of a novel Mdm2 splice variant acutely induced by the chemotherapeutic agents adriamycin and actinomycin D. Cell Cycle (Georgetown, Texas), 7, 1580–1586.
  • Léonard, A., Hantson, P., and Gerber, G.B., 1995. Mutagenicity, carcinogenicity and teratogenicity of lithium compounds. Mutation Research, 339, 131–137.
  • Léonard, A. and Léonard, E.D., 1978. Cytogenetic effects of myleran in vivo on bone-marrow cells from male mice. Mutation Research, 56, 329–333.
  • Li, Y., Dunipace, A.J., and Stookey, G.K., 1987. Lack of genotoxic effects of fluoride in the mouse bone-marrow micronucleus test. Journal of Dental Research, 66, 1687–1690.
  • Li, A.P. and Long, T.J., 1988. An evaluation of the genotoxic potential of glyphosate. Fundamental and Applied Toxicology, 10, 537–546.
  • Liegibel, U., et al., 1992. Clastogenicity to the mouse bone marrow of the mouse germ cell genotoxin Streptozotocin. Mutagenesis, 7, 471–474.
  • Ligumsky, M., et al., 2005. Effects of 6-mercaptopurine treatment on sperm production and reproductive performance: a study in male mice. Scandinavian Journal of Gastroenterology, 40, 444–449.
  • London, S.N., et al., 2000. Clomiphene citrate-induced perturbations during meiotic maturation and cytogenetic abnormalities in mouse oocytes in vivo and in vitro. Fertility and Sterility, 73, 620–626.
  • Lorke, D. and Machemer, L., 1975. Influence of several weeks' treatment of male and female mice with saccharin, cyclamate or cyclohexylamine sulfate on fertility and dominant lethal effects. Humangenetik, 26, 199–205.
  • L'vova, T.S., 1984. Mutagenic action of 5 prospective pesticides on mouse bone marrow, in a culture of human peripheral blood lymphocytes and on saccharomycete yeasts. Tsitologiia Genetika, 18, 455–457.
  • Madrigal-Bujaidar, E., Hernández-Ceruelos, A., and Chamorro, G., 2001. Induction of sister chromatid exchanges by 2,4-dichlorophenoxyacetic acid in somatic and germ cells of mice exposed in vivo. Food and Chemical Toxicology, 39, 941–946.
  • Maeda, A., et al., 2016. Evaluation of red blood cell Pig-a assay and PIGRET assay in rats using chlorambucil. Mutation Research, 811, 91–96.
  • Maier, P. and Schmid, W., 1976. Ten model mutagens evaluated by the micronucleus test. Mutation Research, 40, 325–337.
  • Mailander, P.C., et al., 2006. Induction of A.T to G.C mutations by erroneous repair of depurinated DNA following estrogen treatment of the mammary gland of ACI rats. The Journal of Steroid Biochemistry and Molecular Biology, 101, 204–215.
  • Mailhes, J.B. and Aardema, M.J., 1992. Benomyl-induced aneuploidy in mouse oocytes. Mutagenesis, 7, 303–309.
  • Makita, T., Hashimoto, Y., and Noguchi, T., 1973. Mutagenic, cytogenetic and teratogenic studies on thiophanate-methyl. Toxicology and Applied Pharmacology, 24, 206–215.
  • Malhi, P.K. and Grover, I.S., 1987. Genotoxic effects of some organophosphorus pesticides. II. In vivo chromosomal aberration bioassay in bone marrow cells in rat. Mutation Research, 188, 45–51.
  • Manandhar, M., et al., 1986. Genetic toxicology profile of the new antineoplastic drug mitoxantrone in the mammalian test systems. Arzneimittel-Forschung, 36, 1375–1379.
  • Mañas, F., et al., 2009. Genotoxicity of glyphosate assessed by the comet assay and cytogenetic tests. Environmental Toxicology and Pharmacology, 28, 37–41.
  • Manjanatha, M.G., et al., 2014. Genotoxicity of doxorubicin in F344 rats by combining the comet assay, flow-cytometric peripheral blood micronucleus test, and pathway-focused gene expression profiling. Environmental and Molecular Mutagenesis, 55, 24–34.
  • Manson, J.M., Brown, T., and Baldwin, D.M., 1984. Teratogenicity of nitrofen (2,4-dichloro-4'-nitrodiphenyl ether) and its effects on thyroid function in the rat. Toxicology and Applied Pharmacology, 73, 323–335.
  • Maron, D.M. and Ames, B.N., 1983. Revised method for the Salmonella mutagenicity test. Mutation Research, 113, 173–215.
  • Martelli, A., et al., 1987. Induction of DNA fragmentation and DNA repair synthesis in human and rat hepatocytes by diethylstilbestrol. Cancer Letters, 36, 19–27.
  • Martelli, A., et al., 1991. Genotoxicity testing of chloramphenicol in rodent and human cells. Mutation Research, 260, 65–72.
  • Martelli, A., et al., 1996. Induction of micronuclei and initiation of enzyme-altered foci in the liver of female rats treated with cyproterone acetate, chlormadinone acetate, or megestrol acetate. Carcinogenesis, 17, 551–554.
  • Martelli, A., et al., 1998. Induction of micronuclei and of enzyme-altered foci in the liver of female rats exposed to progesterone and three synthetic progestins. Mutation Research, 419, 33–41.
  • Martelli, A., et al., 1999. Genotoxicity testing of potassium canrenoate in cultured rat and human cells. Mutagenesis, 14, 463–472.
  • Martelli, A., et al., 2000. Evaluation of flutamide genotoxicity in rats and in primary human hepatocytes. Pharmacology & Toxicology, 86, 129–134.
  • Martelli, A., et al., 2002. DNA damage in tissues of rat treated with potassium canrenoate. Toxicology, 171, 95–103.
  • Martelli, A., et al., 2003. Species, sex and inter-individual differences in DNA repair induced by nine sex steroids in primary cultures of rat and human hepatocytes. Mutation Research, 536, 69–78.
  • Martin, G.R., et al., 1979. Lack of cytogenetic effects in mice or mutations in Salmonella receiving sodium fluoride. Mutation Research, 66, 159–167.
  • Matheson, D., Brusick, D., and Carrano, R., 1978. Comparison of the relative mutagenic activity for eight antineoplastic drugs in the Ames Salmonella/microsome and TK+/− mouse lymphoma assays. Drug and Chemical Toxicology, 1, 277–304.
  • Mathew, G., Rahiman, M.A., and Vijayalaxmi, K.K., 1990. In vivo genotoxic effects in mice of Metacid 50, an organophosphorus insecticide. Mutagenesis, 5, 147–149.
  • Matsushita, K., et al., 2013. Development of a medium-term animal model using gpt delta rats to evaluate chemical carcinogenicity and genotoxicity. Journal of Toxicologic Pathology, 26, 19–27.
  • Matthiesen, T., et al., 1998. The experimental toxicology of tramadol: an overview. Toxicology Letters, 95, 63–71.
  • Mattioli, F., et al., 2004. DNA fragmentation, DNA repair and apoptosis induced in primary rat hepatocytes by dienogest, dydrogesterone and 1,4,6-androstatriene-17beta-ol-3-one acetate. Mutation Research, 564, 21–29.
  • Matveeva, V.G., 1982. Mutagenic activity of synthetic interferon inducers in laboratory mice. Voprosy Virusologii, 27, 28–31.
  • McGregor, D.B., et al., 1988. Responses of the L5178Y tk+/tk- mouse lymphoma cell forward mutation assay: III. 72 coded chemicals. Environmental and Molecular Mutagenesis, 12, 85–154.
  • McGregor, D.B., et al., 1991. Responses of the L5178Y mouse Lymphoma cell forward mutation assay. V: 27 coded chemicals. Environmental and Molecular Mutagenesis, 17, 196–219.
  • McGregor, D.B., et al., 1996. Mutagenic responses of L5178Y mouse cells at the tk and hprt loci. Toxicology In Vitro, 10, 643–647.
  • Mehta, M. and Hundal, S.S., 2014. Assessment of genotoxic potential of arsenic in female albino rats at permissible dose levels. Toxicology International, 21, 24–28.
  • Mehta, A., Verma, R.S., and Srivastava, N., 2008. Chlorpyrifos-induced DNA damage in rat liver and brain. Environmental and Molecular Mutagenesis, 49, 426–433.
  • Meireles, J.R., et al., 2013. Genotoxic and cytotoxic effects of testosterone cypionate (deposteron(®)). Mutation Research, 753, 72–75.
  • Meistrich, M.L., et al., 1990. Low levels of chromosomal mutations in germ cells derived from doxorubicin-treated stem spermatogonia in the mouse. Cancer Research, 50, 370–374.
  • Melek, F.R., et al., 2015. Three further triterpenoid saponins from Gleditsia caspica fruits and protective effect of the total saponin fraction on cyclophosphamide-induced genotoxicity in mice. Zeitschrift Fur Naturforschung C, Journal of Biosciences, 70, 31–37.
  • Miura, D., 2014. The in vivo Pig-A gene mutation assay. Genes and Environment, 36, 169–173.
  • Molinari, G., Soloneski, S., and Larramendy, M.L., 2010. New ventures in the genotoxic and cytotoxic effects of macrocyclic lactones, abamectin and ivermectin. Cytogenetic and Genome Research, 128, 37–45.
  • Montes de Oca-Luna, R., et al., 1984. The effect of diphenylhydantoin on the frequency of micronuclei in bone-marrow polychromatic erythrocytes of mice. Mutation Research, 141, 183–187.
  • Moore, F.R., et al., 1995. An in vivo/in vitro method for assessing micronucleus and chromosome aberration induction in rat bone marrow and spleen. 2. Studies with chlorambucil and mitomycin C. Mutation Research, 335, 201–206.
  • Moore, M.M., et al., 1987. Mutagenicity and clastogenicity of adriamycin in L5178Y/TK(+/-)-3.7.2C mouse lymphoma cells. Mutation Research, 191, 183–188.
  • Moore, M.M., Harrington-Brock, K., and Doerr, C.L., 1997. Relative genotoxic potency of arsenic and its methylated metabolites. Mutation Research, 386, 279–290.
  • Morales-Ramírez, P., et al., 2004. In vivo kinetics of micronuclei induction by bifunctional alkylating antineoplastics. Mutagenesis, 19, 207–213.
  • Mughal, A., et al., 2010. Micronucleus and comet assay in the peripheral blood of juvenile rat: establishment of assay feasibility, time of sampling and the induction of DNA damage. Mutation Research, 700, 86–94.
  • Murkunde, Y.V., et al., 2012. Transplacental genotoxicity evaluation of cypermethrin using alkaline comet assay. Human & Experimental Toxicology, 31, 185–192.
  • Nagao, T. and Fujikawa, K., 1990. Genotoxic potency in mouse spermatogonial stem cells of triethylenemelamine, mitomycin C, ethylnitrosourea, procarbazine, and propyl methanesulfonate as measured by F1 congenital defects. Mutation Research, 229, 123–128.
  • Nakanishi, Y. and Schneider, E.L., 1979. In vivo sister-chromatid exchange: a sensitive measure of DNA damage. Mutation Research, 60, 329–337.
  • Nakanomyo, H., Hiraoka, M., and Shiraya, M., 1986. Mutagenicity tests of etoposide and teniposide. The Journal of Toxicological Sciences, 11, 301–310.
  • Narayana, K., D'Souza, U.J., and Seetharama Rao, K.P., 2002. The genotoxic and cytotoxic effects of ribavirin in rat bone marrow. Mutation Research, 521, 179–185.
  • National Cancer Institute [NCI], 1968. Evaluation of carcinogenic, teratogenic and mutagenic activities of selected pesticides and industrial chemicals. In: Carcinogenic study. National Institutes of Health Report Number NCI-DCCP-CG-1973-1-1. Available from: https://ntrl.ntis.gov/NTRL/dashboard/searchResults.xhtml?searchQuery=PB223159 [Accessed 28 Mar 2017].
  • Nehéz, M., Tóth, C., and Dési, I., 1994. The effect of dimethoate, dichlorvos, and parathion-methyl on bone marrow cell chromosomes of rats in subchronic experiments in vivo. Ecotoxicology and Environmental Safety, 29, 365–371.
  • Nersesyan, A., et al., 2003. Genotoxic action of cycloplatam, a new platinum antitumor drug, on mammalian cells in vivo and in vitro. Chemotherapy, 49, 132–137.
  • NIOSH, 2016. NIOSH list of antineoplastic and other hazardous drugs in healthcare settings. By Connor TH, MacKenzie BA, DeBord DG, Trout DB, O’Callaghan JP. Cincinnati OH: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health. DHHS (NIOSH) Publication Number 2016-161 (Supersedes 2014-138). Available from: https://www.cdc.gov/niosh/topics/antineoplastic/pdf/hazardous-drugs-list_2016-161.pdf [Accessed 20 Mar 2017].
  • Ni, Z., et al., 1993. Induction of micronucleus by organophosphorus pesticides both in vivo and in vitro. Hua Xi Yi Ke Da Xue Xue Bao, 24, 82–86.
  • Norppa, H., et al., 2006. Chromosomal aberrations and SCEs as biomarkers of cancer risk. Mutation Research, 600, 37–45.
  • Norppa, H., and Falck, G.C.M., 2003. What do human micronuclei contain? Mutagenesis, 18, 221–233.
  • Noshy, M.M., Hussien, N.A., and El-Ghor, A.A., 2013. Evaluation of the role of the antioxidant silymarin in modulating the in vivo genotoxicity of the antiviral drug ribavirin in mice. Mutation Research, 752, 14–20.
  • NTP, 1989a. National Toxicology Program, Toxicology and carcinogenesis studies of tetracycline hydrochloride (CAS No. 64-75-5) in F344/N rats and B6C3F1 mice (feed studies). National Toxicology Program Technical Report Series, 344, 1–172.
  • NTP, 1989b. National Toxicology Program, Toxicology and carcinogenesis studies of Dichlorvos (CAS No. 62-73-7) in F344/N rats and B6C3F1 mice (gavage studies). National Toxicology Program Technical Report Series, 342, 1–208.
  • NTP, 1993. National Toxicology Program, Toxicology and carcinogenesis studies of 5,5-diphenylhydantoin (CAS No. 57-41-0) (Phenytoin) in F344/N rats and B6C3F1 mice (feed studies). National Toxicology Program Technical Report Series, 404, 1–303.
  • Nurulain, S.M. and Shafiullah, M., 2012. Teratogenicity and embryotoxicity of organophosphorus compounds in animal models – a short review. Military Medical Science Letters, 81, 16–26.
  • Oberly, T.J., et al., 1993. A comparison of the CHO/HGPRT + and the L5178Y/TK+/- mutation assays using suspension treatment and soft agar cloning: results for 10 chemicals. Cell Biology and Toxicology, 9, 243–257.
  • OECD, 1986. Genetic toxicology: in vitro sister chromatid exchange assay in mammalian cells. In: Guideline for the testing of chemicals. Available from: http://www.oecd-ilibrary.org/environment/test-no-479-genetic-toxicology-in-vitro-sister-chromatid-exchange-assay-in-mammalian-cells_9789264071384-en [Accessed 11 Mar 2017].
  • OECD, 1997a. In vitro mammalian chromosome aberration. In: Guideline for the testing of chemicals. Available from: http://www.oecd.org/chemicalsafety/risk-assessment/1948434.pdf [Accessed 11 Mar 2017].
  • OECD, 1997b. Mammalian bone marrow chromosome aberration test. In: Guideline for the testing of chemicals. Available from: http://www.oecd-ilibrary.org/environment/test-no-475-mammalian-bone-marrow-chromosome-aberration-test_9789264071308-en [Accessed 12 Mar 2017].
  • Ojha, A. and Srivastava, N., 2014. In vitro studies on organophosphate pesticides induced oxidative DNA damage in rat lymphocytes. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 761, 10–17.
  • Oliveira, R.J., et al., 2014. Compounds used to produce cloned animals are genotoxic and mutagenic in mammalian assays in vitro and in vivo. Brazilian Journal of Medical and Biological Research, 47, 287–298.
  • Osterman Golkar, S., et al., 2013. Intracellular deoxyribonucleotide pool imbalance and DNA damage in cells treated with hydroxyurea, an inhibitor of ribonucleotide reductase. Mutagenesis, 28, 653–660.
  • Padmanabhan, S., et al., 2008. Cytotoxic and genotoxic effects of methotrexate in germ cells of male Swiss mice. Mutation Research, 655, 59–67.
  • Pandey, N., et al., 1990. Studies on the genotoxicity of endosulfan, an organochlorine insecticide, in mammalian germ cells. Mutation Research, 242, 1–7.
  • Pandita, T.K., 1983. Mutagenic studies on the insecticide Metasystox-R with different genetic systems. Mutation Research, 124, 97–102.
  • Parodi, S., et al., 1981. DNA-damaging activity in vivo and bacterial mutagenicity of sixteen hydrazine derivatives as related quantitatively to their carcinogenicity. Cancer Research, 41, 1469–1482.
  • Patlolla, A.K., et al., 2012. Arsenic-induced biochemical and genotoxic effects and distribution in tissues of Sprague-Dawley rats. Microchemical Journal, Devoted to the Application of Microtechniques in All Branches of Science, 105, 101–107.
  • Patlolla, A.K. and Tchounwou, P.B., 2005. Cytogenetic evaluation of arsenic trioxide toxicity in Sprague-Dawley rats. Mutation Research, 587, 126–133.
  • Perry, P. and Evans, H.J., 1975. Cytological detection of mutagen-carcinogen exposure by sister chromatid exchange. Nature, 258, 121–125.
  • Perry, P.E. and Thompson, E.J., 1984. The methodology of sister chromatid exchanges. In: B.J. Kilbey, M. Legator, W. Nichols, and C. Ramel, eds. Handbook of mutagenicity test procedures. 2nd ed. Amsterdam: Elsevier, p. 495–529.
  • Philipose, B., et al., 1997. Comparative mutagenic and genotoxic effects of three propionic acid derivatives ibuprofen, ketoprofen and naproxen. Mutation Research, 393, 123–131.
  • Phillips, M.D., et al., 1991. Induction of micronuclei in mouse bone marrow cells: an evaluation of nucleoside analogues used in the treatment of AIDS. Environmental and Molecular Mutagenesis, 18, 168–183.
  • Picada, J.N., et al., 2003. DNA damage in brain cells of mice treated with an oxidized form of apomorphine. Brain Research/Molecular Brain Research, 114, 80–85.
  • Picada, J.N., Roesler, R., and Henriques, J.A., 2005. Genotoxic, neurotoxic and neuroprotective activities of apomorphine and its oxidized derivative 8-oxo-apomorphine. Brazilian Journal of Medical and Biological Research, 38, 477–486.
  • Piña-Guzmán, B., et al., 2006. Genetic damage caused by methyl-parathion in mouse spermatozoa is related to oxidative stress. Toxicology and Applied Pharmacology, 216, 216–224.
  • Pino, A., Maura, A., and Grillo, P., 1988. DNA damage in stomach, kidney, liver and lung of rats treated with atrazine. Mutation Research, 209, 145–147.
  • Prasad, S., et al., 2009. Clastogenic effects of glyphosate in bone marrow cells of swiss albino mice. Journal of Toxicology, 2009, 308985.
  • PubMed, 2017. US National Library of Medicine, National Institutes of Health. Available from: https://www.ncbi.nlm.nih.gov/pubmed [Accessed 01 Mar–03 Apr 2017].
  • Pylkkänen, L. and Salonen, I., 1987. Concomitant mutagenicity of ethanol and x-ray irradiation in the mouse male germ cells. Alcohol (Fayetteville, N.Y.), 4, 401–404.
  • Rajaniemi, H., et al., 1998. DNA binding of tamoxifen and its analogues: identification of the tamoxifen-DNA adducts in rat liver. Toxicology Letters, 102–103, 453–457.
  • Rajaniemi, H., Mantyla, E., and Hemminki, K., 1998. DNA adduct formation by tamoxifen and structurally-related compounds in rat liver. Chemico-Biological Interactions, 113, 145–159.
  • Rank, J., et al., 1993. Genotoxicity testing of the herbicide Roundup and its active ingredient glyphosate isopropylamine using the mouse bone marrow micronucleus test, Salmonella mutagenicity test, and Allium anaphase-telophase test. Mutation Research, 300, 29–36.
  • Rannug, U., 1980. Genotoxic effects of 1,2-dibromoethane and 1,2-dichloroethane. Mutation Research, 76, 269–295.
  • Rao, K.P. and Rahiman, M.A., 1989. Cytogenetic effects of ribavirin on mouse bone marrow. Mutation Research, 224, 213–218.
  • Recio, L., et al., 2010. Dose-response assessment of four genotoxic chemicals in a combined mouse and rat micronucleus (MN) and Comet assay protocol. The Journal of Toxicological Sciences, 35, 149–162.
  • Reddy, M.V., 1987. Randerath 32P-analysis of DNA adducts in somatic and reproductive tissues of rats treated with the anticancer antibiotic, mitomycin C. Mutation Research, 179, 75–88.
  • Reimann, R., Kalweit, S., and Lang, R., 1996. Studies for a genotoxic potential of some endogenous and exogenous sex steroids. II. Communication: examination for the induction of cytogenetic damage using the chromosomal aberration assay on human lymphocytes in vitro and the mouse bone marrow micronucleus test in vivo. Environmental and Molecular Mutagenesis, 28, 133–144.
  • Ren, L., et al., 1993. A sequential study on the use of the cytokinesis-block micronucleus method in mouse splenocytes. Mutation Research, 301, 223–227.
  • Rerko, R.M., et al., 1992. Photofrin II photosensitization is mutagenic at the tk locus in mouse L5178Y cells. Photochemistry and Photobiology, 55, 75–80.
  • Rinchik, E.M., Flaherty, L., and Russell, L.B., 1993. High-frequency induction of chromosomal rearrangements in mouse germ cells by the chemotherapeutic agent chlorambucil. Bioessays: News and Reviews in Molecular, Cellular and Developmental Biology, 15, 831–836.
  • Rjiba-Touati, K., et al., 2012. Induction of DNA fragmentation, chromosome aberrations and micronuclei by cisplatin in rat bone-marrow cells: protective effect of recombinant human erythropoietin. Mutation Research, 747, 202–206.
  • Roloff, B.D., Belluck, D.A., and Meisner, L.F., 1992. Cytogenetic studies of herbicide interactions in vitro and in vivo using atrazine and linuron. Archives of Environmental Contamination and Toxicology, 22, 267–271.
  • Roma, G.C., et al., 2012. Genotoxic and mutagenic effects of permethrin in mice: micronuclei analysis in peripheral blood erythrocytes. Microscopy Research and Technique, 75, 1732–1736.
  • Ross, J.A. and Leavitt, S.A., 2010. Analysis of the mutations induced by conazole fungicides in vivo. Mutagenesis, 25, 231–234.
  • Rossi, A.M., et al., 1986. Chromosome aberrations in rat liver cells and bone marrow cells following treatment in vivo with mitomycin C. Mutagenesis, 1, 335–338.
  • Roux, C., Emerit, I., and Taillemite, J.L., 1971. Chromosomal breakage and teratogenesis. Teratology, 4, 303–315.
  • Röhrborn, G. and Basler, A., 1977. Cyto-enetic investigations of mammals. Comparison of the genetic activity of cytostatics in mammals. Archives of Toxicology, 38, 35–43.
  • Russell, L.B., et al., 1998. Unlike other chemicals, etoposide (a topoisomerase-II inhibitor) produces peak mutagenicity in primary spermatocytes of the mouse. Mutation Research, 400, 279–286.
  • Russell, L.B., et al., 2000. Bleomycin, unlike other male-mouse mutagens, is most effective in spermatogonia, inducing primarily deletions. Mutation Research, 469, 95–105.
  • Russell, L.B., Hunsicker, P.R., and Shelby, M.D., 1996. Chlorambucil and bleomycin induce mutations in the specific-locus test in female mice. Mutation Research, 358, 25–35.
  • Ruzicska, P., Péter, S., and Czeizel, A., 1975. Proceedings: Studies on the chromosomal mutagenic effect of Benomyl in rats and humans. Mutation Research, 29, 201.
  • Sanada, M., et al., 2004. DNA Repair (Amsterdam), 3, 413–420.
  • Sanderson, B.J., Ferguson, L.R., and Denny, W.A., 1996. Mutagenic and carcinogenic properties of platinum-based anticancer drugs. Mutation Research, 355, 59–70.
  • Sandhu, S.S. and Waters, M.D., 1980. Mutagenicity evaluation of chemical pesticides. Journal of Environmental Science and Health, Part B, Pesticides, Food Contaminants, and Agricultural Wastes, 15, 929–948.
  • Sargent, L.M., et al., 1996. Induction of hepatic aneuploidy in vivo by tamoxifen, toremifene and idoxifene in female Sprague-Dawley rats. Carcinogenesis, 17, 1051–1056.
  • Sarty, K.I., Cowie, A., and Martyniuk, C.J., 2017. The legacy pesticide dieldrin acts as a teratogen and alters the expression of dopamine transporter and dopamine receptor 2a in zebrafish (Danio rerio) embryos. Comparative Biochemistry and Physiology, Toxicology & Pharmacology, 194, 37–47.
  • Sasaki, Y.F., et al., 1986. Mutagenicity of 1,2-dibromo-3-chloropropane (DBCP) in the mouse spot test. Mutation Research, 174, 145–147.
  • Sasaki, Y.F., et al., 1998. Detection of in vivo genotoxicity of haloalkanes and haloalkenes carcinogenic to rodents by the alkaline single cell gel electrophoresis (comet) assay in multiple mouse organs. Mutation Research, 419, 13–20.
  • Savage, J.R.K., 1993. Update on target theory as applied to chromosomal aberrations. Environmental and Molecular Mutagenesis, 22, 198–207.
  • Seiler, J.P., 1976. The mutagenicity of benzimidazole and benzimidazole derivatives. VI. Cytogenetic effects of benzimidazole derivatives in the bone marrow of the mouse and the Chinese hamster. Mutation Research, 40, 339–347.
  • Sen, A.K., Karakas, E., and Bilaloglu, R., 2010. Genotoxic effect of epirubicin in mouse bone marrow in vivo. Zeitschrift Fur Naturforschung C, Journal of Biosciences, 65, 211–217.
  • Sgura, A., et al., 2005. Use of chromosome painting for detecting stable chromosome aberrations induced by melphalan in mice. Environmental and Molecular Mutagenesis, 45, 419–426.
  • Sherman, H., Culik, R., and Jackson, R.A., 1975. Reproduction, teratogenic, and mutagenic studies with benomyl. Toxicology and Applied Pharmacology, 32, 305–315.
  • Shimada, K., et al., 2015a. Prolonged rest period enables the detection of micronucleated hepatocytes in the liver of young adult rats after a single dose of diethylnitrosamine or mitomycin C. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 791, 38–41.
  • Shimada, K., et al., 2015b. Repeated-dose liver micronucleus assay of mitomycin C in young adult rats. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 780–781, 85–89.
  • Shreder, O.V., et al., 2011. Association of genotoxic and teratogenic effects induced by cyclophosphamide and their modification with afobazole. Gigiena Sanitariia, 5, 64–88.
  • Shyama, S.K. and Abdul Rahiman, M., 1993. Progestin (norethisterone)-induced genetic damage in mouse bone marrow. Mutation Research, 300, 215–221.
  • Sicardi, S.M., Martiarena, J.L., and Iglesias, M.T., 1991. Mutagenic and analgesic activities of aniline derivatives. Journal of Pharmaceutical Sciences, 80, 761–764.
  • Siddique, Y.H. and Afzal, M., 2004. Induction of chromosomal aberrations and sister chromatid exchanges by chlormadinone acetate in human lymphocytes: a possible role of reactive oxygen species. Indian Journal of Experimental Biology, 42, 1078–1083.
  • Silva, C.R., Antunes, L.M., and Bianchi, M.L., 2001. Antioxidant action of bixin against cisplatin-induced chromosome aberrations and lipid peroxidation in rats. Pharmacological Research, 43, 561–566.
  • Simula, A.P. and Priestly, B.G., 1992. Species differences in the genotoxicity of cyclophosphamide and styrene in three in vivo assays. Mutation Research, 271, 49–58.
  • Singh, K.P. and DuMond, J.W., Jr2007. Genetic and epigenetic changes induced by chronic low dose exposure to arsenic of mouse testicular Leydig cells. International Journal of Oncology, 30, 253–260.
  • Sitarek, K., 2001. Embryolethal and teratogenic effects of carbendazim in rats. Teratogenesis, Carcinogenesis, and Mutagenesis, 21, 335–340.
  • Skare, J.A. and Schrotel, K.R., 1985. Validation of an in vivo alkaline elution assay to detect DNA damage in rat testicular cells. Environmental Mutagenesis, 7, 563–576.
  • Skorski, T., 2008. BCR/ABL, DNA damage and DNA repair: implications for new treatment concepts. Leukemia & Lymphoma, 49, 610–614.
  • Smith, C.C., et al., 1999. Analysis of gene mutations and clastogenicity following short-term treatment with azathioprine in MutaMouse. Environmental and Molecular Mutagenesis, 34, 131–189.
  • Sonoda, E., et al., 1999. Sister chromatid exchanges are mediated by homologous recombination in vertebrate cells. Molecular and Cellular Biology, 19, 5166–5169.
  • Spencer, D.L., Hines, K.C., and Caspary, W.J., 1994. An in situ protocol for measuring the expression of chemically-induced mutations in mammalian cells. Mutation Research, 312, 85–97.
  • Spielmann, H. and Vogel, R., 1993. Genotoxic and embryotoxic effects of gonadotropin hyperstimulated ovulation on murine oocytes, preimplantation embryos and term fetuses. Annali Dell'istituto Superiore Di Sanita, 29, 35–39.
  • Steiblen, G., et al., 2005. Comparison of the relative sensitivity of human lymphocytes and mouse splenocytes to two spindle poisons. Mutation Research, 588, 143–151.
  • Storer, R.D., et al., 1996. Revalidation of the in vitro alkaline elution/rat hepatocyte assay for DNA damage: improved criteria for assessment of cytotoxicity and genotoxicity and results for 81 compounds. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 368, 59–155.
  • Subramanyam, S., Laxminarayana, D., and Helen, K.D., 1984. Evaluation of genotoxic potential of vincristine from multiple parameters. Mutation Research, 138, 55–62.
  • Sudman, P.D., et al., 1992. Bleomycin: female-specific dominant lethal effects in mice. Mutation Research, 296, 143–156.
  • Surrallés, J., et al., 1995. The Suitability of the micronucleus assay in human lymphocytes as a new biomarker of excision repair. Mutation Research, 342, 43–59.
  • Suzuki, T., et al., 1999. Procarbazine genotoxicity in the MutaMouse; strong clastogenicity and organ-specific induction of lacZ mutations. Mutation Research, 444, 269–281.
  • Takahashi, M., et al., 1986. Effects of ethanol, potassium metabisulfite, formaldehyde and hydrogen peroxide on gastric carcinogenesis in rats after initiation with N-methyl-N-nitro-N-nitrosoguanidine. Japanese Journal of Cancer Research, 77, 118–124.
  • Takasawa, H., et al., 2010. Evaluation of a liver micronucleus assay in young rats (IV): a study using a double-dosing/single-sampling method by the Collaborative Study Group for the Micronucleus Test (CSGMT)/Japanese Environmental Mutagen Society (JEMS)-Mammalian Mutagenicity Study Group (MMS). Mutation Research, 698, 24–29.
  • Teo, S., et al., 2000. Assessment of the in vitro and in vivo genotoxicity of Thalomid (thalidomide). Teratogenesis, Carcinogenesis, and Mutagenesis, 20, 301–311.
  • Terra, D.G., et al., 2016. Low dose of methyltestosterone in ovariectomised rats improves baroreflex sensitivity without geno- and cytotoxicity. Fundamental & Clinical Pharmacology, 30, 316–326.
  • TGA, 2017. Australian Goverment, Therapeutiq Goods Adminitration, Prescribing medicine in pregnancy database. Available from: https://www.tga.gov.au/prescribing-medicines-pregnancy-database [Accessed 28 Mar 2017].
  • Tinwell, H., Stephens, S.C., and Ashby, J., 1991. Arsenite as the probable active species in the human carcinogenicity of arsenic: mouse micronucleus assays on Na and K arsenite, orpiment, and Fowler's solution. Environmental Health Perspectives, 95, 205–210.
  • Topaktas, M. and Rencuzogullari, E., 2010. Sitogenetik (Cytogenetic). Ankara, Turkey: Nobel Yayınları.
  • Topaktas, M., Rencuzogullari, E., and Ila, H.B., 1996. In vivo chromosomal aberrations in bone marrow cells of rats treated with marshal. Mutation Research, 371, 259–264.
  • Topinka, J., et al., 1995. DNA-damaging activity of the cyproterone acetate analogues chlormadinone acetate and megestrol acetate in rat liver. Carcinogenesis, 16, 1483–1487.
  • Topinka, J., et al., 2004a. No-effect level in the mutagenic activity of the drug cyproterone acetate in rat liver. Part II. Multiple dose treatment. Mutation Research, 550, 101–108.
  • Topinka, J., et al., 2004b. No-effect level in the mutagenic activity of the drug cyproterone acetate in rat liver. Part I. Single dose treatment. Mutation Research, 550, 89–99.
  • Torres, S.M., et al., 2007. Mutagenicity of zidovudine, lamivudine, and abacavir following in vitro exposure of human lymphoblastoid cells or in utero exposure of CD-1 mice to single agents or drug combinations. Environmental and Molecular Mutagenesis, 48, 224–238.
  • Tucker, J.D., et al., 1993. Sister-chromatid exchange: second report of the Gene-Tox Program. Mutation Research, 297, 101–180.
  • Umeda, M., Tsutsui, T., and Saito, M., 1977. Mutagenicity and inducibility of DNA single-strand breaks and chromosome aberrations by various mycotoxins. Gan, 68, 619–625.
  • United States Environmental Protection Agency [US EPA], 2017. Available from: https://cfpub.epa.gov/ncea/iris/search/index.cfm?keyword=teratogen [Accessed 25 Mar 2017].
  • van Gelder, M.M., et al., 2010. Teratogenic mechanisms of medical drugs. Human Reproduction Update, 16, 378–394.
  • van Went, G.F., 1979. Investigation into the mutagenic activity of azathioprine (Imuran) in different test systems. Mutation Research, 68, 153–162.
  • Vanparys, P., et al., 1992. Sampling times in micronucleus testing. Mutation Research, 282, 191–196.
  • Várnagy, L., et al., 2000. Interaction of dithane M-45 (mancozeb) and lead acetate during a teratogenicity test in rats. Acta Veterinaria Hungarica, 48, 113–124.
  • Vashishta, A. and Hetman, M., 2014. Inhibitors of histone deacetylases enhance neurotoxicity of DNA damage. Neuromolecular Medicine, 16, 727–741.
  • Velazquez-Guadarrama, N., et al., 2005. Genotoxic evaluation of sodium fluoride and sodium perborate in mouse bone marrow cells. Bulletin of Environmental Contamination and Toxicology, 74, 566–572.
  • Vijayalaxmi, K.K. and Rai, S.P., 1996. Studies on the genotoxicity of tamoxifen citrate in mouse bone marrow cells. Mutation Research, 368, 109–114.
  • Vikram, A., et al., 2007. Evaluation of streptozotocin genotoxicity in rats from different ages using the micronucleus assay. Regulatory Toxicology and Pharmacology, 49, 238–244.
  • Von Tungeln, L.S., et al., 2002. Frequency of Tk and Hprt lymphocyte mutants and bone marrow micronuclei in B6C3F(1)/Tk+/- mice treated neonatally with zidovudine and lamivudine. Carcinogenesis, 23, 1427–1432.
  • Voronova, O.L., et al., 2003. Cytogenetic effects of antibodies to gamma-interferon in ultralow doses. Bulletin of Experimental Biology and Medicine, 135, 65–66.
  • Wang, Y.J. and Zhang, L.S., 2006. Evaluation of the genotoxicity of vincristine and colchicine using mouse lymphoma tk mutation assay. Wei Sheng Yan Jiu, 35, 179–181.
  • Wangenheim, J. and Bolcsfoldi, G., 1988. Mouse lymphoma L5178Y thymidine kinase locus assay of 50 compounds. Mutagenesis, 3, 193–205.
  • Wedebye, E.B., et al., 2015. QSAR screening of 70,983 REACH substances for genotoxic carcinogenicity, mutagenicity and developmental toxicity in the ChemScreen project. Reproductive Toxicology (Elmsford, N.Y.), 55, 64–72.
  • Weiner, M.L., et al., 1990. Genotoxicity evaluation of lithium hypochlorite. Toxicology, 65, 1–22.
  • Werner, S., et al., 1997. Formation of DNA adducts by cyproterone acetate and some structural analogues in primary cultures of human hepatocytes. Mutation Research, 395, 179–187.
  • Whittaker, P., et al., 2001. Genotoxicity of iron chelators in L5178Y mouse lymphoma cells. Environmental and Molecular Mutagenesis, 38, 347–356.
  • Williams, G.M., et al., 1998. Genotoxicity studies with the antiestrogen toremifene. Drug and Chemical Toxicology, 21, 449–476.
  • Wilson, J.G., 1973. Environment and birth defects. By Aston SA, Published: 2014-06-21. Available from: https://embryo.asu.edu/pages/environment-and-birth-defects-1973-james-g-wilson [Accessed 14 Mar 2017].
  • Witt, K.L., et al., 2008. Comparison of flow cytometry- and microscopy-based methods for measuring micronucleated reticulocyte frequencies in rodents treated with nongenotoxic and genotoxic chemicals. Mutation Research, 649, 101–113.
  • Woodburn, K.W., et al., 2011. Genotoxic assessment and toxicity evaluation of peginesatide in CByB6F1 hybrid mice. Drug and Chemical Toxicology, 34, 240–249.
  • Working, P.K. and Butterworth, B.E., 1984. An assay to detect chemically induced DNA repair in rat spermatocytes. Environmental Mutagenesis, 6, 273–286.
  • Wu, Y.G., et al., 2007. The establishment of hypospadias rat model and embryoteratogenic test of Atrazine. Zhonghua Zheng Xing Wai Ke Za Zhi, 23, 340–343.
  • Wubah, J. A., et al., 1996. Teratogen-induced eye defects mediated by p53-dependent apoptosis. Current Biology, 6, 60–69.
  • Wutzler, P. and Thust, R., 2001. Genetic risks of antiviral nucleoside analogues – a survey. Antiviral Research, 49, 55–74.
  • Yaduvanshi, S.K., et al., 2012. Evaluation of micronuclei induction capacity and mutagenicity of organochlorine and organophosphate pesticides. Drug Metabolism Letters, 6, 187–197.
  • Yavuz Kocaman, A., et al., 2008. The genotoxic effect of potassium metabisulfite using chromosome aberration, sister chromatid exchange, micronucleus tests in human lymphocytes and chromosome aberration test in bone marrow cells of rats. Environmental and Molecular Mutagenesis, 49, 276–282.
  • Yavuz Kocaman, A., Rencuzogullari, E., and Topaktas, M., 2014. In vitro investigation of the genotoxic and cytotoxic effects of thiacloprid in cultured human peripheral blood lymphocytes. Environmental Toxicology, 29, 631–641.
  • Yilmaz, H.R., et al., 2010. Anticlastogenic effect of caffeic acid phenethyl ester on cisplatin-induced chromosome aberrations in rat bone marrow cells. Toxicology and Industrial Health, 26, 33–37.
  • Yilmaz, S., Gonenc, I.M., and Yilmaz, E., 2014. Genotoxicity of the some selective estrogen receptor modulators: a review. Cytotechnology, 66, 533–541.
  • Yoshida, I., et al., 2016. Red blood cell Pig-a assay and PIGRET assay in rats with azathioprine. Mutation Research, 811, 60–64.
  • Yuzbasioglu, D., et al., 2007. Genotoxicity testing of fluconazole in vivo and in vitro. Mutation Research, 649, 155–160.
  • Zambrano, M.A., Targa, H.J., and Nazareth Rabello-Gay, M., 1982. Evaluation of the action of clomiphene citrate on Mouse chromosomes by the metaphase and micronucleus tests. Brazilian Journal of Genetics, 2, 339–344.
  • Zhang, Y. and Sun, K., 1992. Unscheduled DNA synthesis induced by the antitumor drug vincristine in germ cells of male mice. Mutation Research, 281, 25–29.
  • Zhou, C., et al., 2013. Comparison of three-colour flow cytometry and slide-based microscopy for the scoring of micronucleated reticulocytes in rat bone-marrow and peripheral blood. Mutation Research, 758, 12–17.
  • Zúñiga-González, G.M., et al., 2003. Induction of micronucleated erythrocytes in mouse peripheral blood after cutaneous application of 5-fluorouracil. Archives of Medical Research, 34, 141–144.

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.