586
Views
41
CrossRef citations to date
0
Altmetric
Review Article

Reproductive toxicants have a threshold of adversity

, , , , , & show all
Pages 545-554 | Received 14 Oct 2010, Accepted 11 Jan 2011, Published online: 24 May 2011

References

  • Ashby J, Tinwell H, Haseman J. (1999). Lack of effects for low dose levels of bisphenol A and diethylstilbestrol on the prostate gland of CF1 nice exposed in utero. Regul Toxicol Pharmacol 30:156–166.
  • Ashby J, Tinwell H, Odum J, Lefevre P. (2004). Natural variability and the influence of concurrent control values on the detection and interpretation of low-dose or weak endocrine toxicities. Environ Health Perspect 112:847–853.
  • Beckman DA, Brent RL (1992). Basic principles of teratology. In: Reece EA, ed. Medicine of the Fetus and Mother. Philadelphia: J. B. Lippincott, 293–299.
  • Boobis AR, Daston GP, Preston RJ, Olin SS. (2009). Application of key events analysis to chemical carcinogens and noncarcinogens. Crit Rev Food Sci Nutr 49:690–707.
  • Brent RL. (1999). Utilization of developmental basic science principles in the evaluation of reproductive risks from pre- and postconception environmental radiation exposures. Teratology 59:182–204.
  • Cagen SZ, Waechter JM Jr, Dimond SS, Breslin WJ, Butala JH, Jekat FW, Joiner RL, Shiotsuka RN, Veenstra GE, Harris LR. (1999). Normal reproductive organ development in Wistar rats exposed to bisphenol A in the drinking water. Regul Toxicol Pharmacol 30:130–139.
  • Campbell JL Jr, Smith MA, Fisher JW, Warren DA. (2004). Dose-response for retinoic acid-induced forelimb malformations and cleft palate: A comparison of computerized image analysis and visual inspection. Birth Defects Res B Dev Reprod Toxicol 71:289–295.
  • Campo-Paysaa F, Marlétaz F, Laudet V, Schubert M. (2008). Retinoic acid signaling in development: Tissue-specific functions and evolutionary origins. Genesis 46:640–656.
  • Castoldi AF, Onishchenko N, Johansson C, Coccini T, Roda E, Vahter M, Ceccatelli S, Manzo L. (2008). Neurodevelopmental toxicity of methylmercury: Laboratory animal data and their contribution to human risk assessment. Regul Toxicol Pharmacol 51:215–229.
  • Chabbert Buffet N, Djakoure C, Maitre SC, Bouchard P. (1998). Regulation of the human menstrual cycle. Front Neuroendocrinol 19:151–186.
  • Choi BH, Lapham LW, Amin-Zaki L, Saleem T. (1978). Abnormal neuronal migration, deranged cerebral cortical organization and diffuse white matter astrocytosis of human fetal brain: A major effect of methylmercury poisoning in utero. J Neuropathol Exp Neurol 37:719–733.
  • Cox C, Marsh D, Myers G, Clarkson T. (1995). Analysis of data on delayed development from the 1971–72 outbreak of methylmercury poisoning in Iraq: Assessment of influential points. Neurotoxicology 16:727–730.
  • Crump KS. (1984). A new method for determining allowable daily intakes. Fundam Appl Toxicol 4:854–871.
  • Curle DC, Ray M, Persaud TV. (1983). Methylmercury toxicity: In vivo evaluation of teratogenesis and cytogenetic changes. Anat Anz 153:69–82.
  • Degen GH, Bolt HM. (2000). Endocrine disruptors: Update on xenoestrogens. Int Arch Occup Environ Health 73:433–441.
  • Doak SH, Jenkins GJ, Johnson GE, Quick E, Parry EM, Parry JM. (2007). Mechanistic influences for mutation induction curves after exposure to DNA-reactive carcinogens. Cancer Res 67:3904–3911.
  • European Commission. (1996). European Workshop on the Impact of Endocrine Disrupters on Human Health and Wildlife. Report reference EUR 17549. Brussels, Belgium: European Commission, DGXII.
  • ECB. (2005). Draft risk assessment report: Vinyl acetate. ECB R059_0505_env Draft of 04.05.2005.
  • ECHA. (2008). Guidance for the implementation of REACH. Guidance on information requirements and chemical safety assessment Chapter R.8: Characterisation of dose [concentration]-response for human health. Available at: http://guidance.echa.europa.eu/docs/guidance_document/information_requirements_en.htm#A Accessed 20 September 2010.
  • ECHA. (2009). Introductory Guidance on Regulation (EC) No 1272/2008 on classification, labelling and packaging (CLP) of substances and mixtures. ECHA-09-G-01-EN.
  • Eckhoff C, Löfberg B, Chahoud I, Bochert G, Nau H. (1989). Transplacental pharmacokinetics and teratogenicity of a single dose of retinol (vitamin A) during organogenesis in the mouse. Toxicol Lett 48:171–184.
  • EFSA. (2005). Opinion of the Scientific Committee on a request from EFSA related to a harmonised approach for risk assessment of substances which are both genotoxic and carcinogenic. EFSA J 282:1–31.
  • EFSA. (2008). Toxicokinetics of bisphenol A. EFSA J 759:1–10.
  • EFSA. (2009). European Food Safety Authority. Guidance of the scientific committee on use of the benchmark dose approach in risk assessment. EFSA J 1150:1–72.
  • Ekino S, Susa M, Ninomiya T, Imamura K, Kitamura T. (2007). Minamata disease revisited: An update on the acute and chronic manifestations of methyl mercury poisoning. J Neurol Sci 262:131–144.
  • Eluma FO, Sucheston ME, Hayes TG, Paulson RB. (1984). Teratogenic effects of dosage levels and time of administration of carbamazepine, sodium valproate, and diphenylhydantoin on craniofacial development in the CD-1 mouse fetus. J Craniofac Genet Dev Biol 4:191–210.
  • EU. (2006). Regulation (EC) No. 1907/2006 of the European Parliament and of the Council of 18 December 2006 concerning the Registration, Evaluation, Authorisation and Restriction of Chemicals (REACH), establishing a European Chemicals Agency, amending Directive, 1999/45/EC and repealing Council Regulation (EEC) no. 793/93 and Commission Regulation (EC) No. 1488/94 as well as Council Directive 76/769 EEC and Commission Directive 91/155/EEC, 93/67/EEC, 93/105EC and 2000/21/EC.
  • Fearon ER, Vogelstein B. (1990). A genetic model for colorectal tumorigenesis. Cell 61:759–767.
  • Friedman JM. (2010). The principles of teratology: Are they still true? Birth Defects Res A Clin Mol Teratol 88:766–768.
  • Fritz H, Müller D, Hess R. (1976). Comparative study of the teratogenicity of phenobarbitone, diphenylhydantoin and carbamazepine in mice. Toxicology 6:323–330.
  • Fukushima SH, Wanibuchi K, Morimura S, Iwai D, Nakae H, Kishida H, Tsuda N, Uehara K, Imaida T, Shirai M, Tatematsu T, Tsukamoto M, Furukawa F. (2004). Existence of a threshold for induction of aberrant crypt foci in the rat colon with low doses of 2-amino-1-methyl-6-phenolimidazo[4,5-b]pyridine. Toxicol Sci 80:109–114.
  • Fukushima S, Wanibuchi H, Morimura K, Nakae D, Tsuda H, Imaida K, Shirai T, Tatematsu M, Tsukamoto T, Hirose M, et al. (2005). Lack of potential of low dose N-nitrosodimethylamine to induce preneoplastic lesions, glutathione S-transferase placental form-positive foci, in rat liver. Cancer Lett 222:11–15.
  • Gan EH, Quinton R. (2010). Physiological significance of the rhythmic secretion of hypothalamic and pituitary hormones. Prog Brain Res 181:111–126.
  • Gao Y, Ding Y, Shi R, Tian Y. (2008). Effects of methylmercury on postnatal neurobehavioral development in mice. Neurotoxicol Teratol 30:462–467.
  • Geyer MA, Butcher RE, Fite K. (1985). A study of startle and locomotor activity in rats exposed prenatally to methylmercury. Neurobehav Toxicol Teratol 7:759–765.
  • Gocke E, Muller L. (2009). In vivo studies in the mouse to define a threshold for the genotoxicity of EMS and ENU. Mutat Res 678:101–107.
  • Greim HA. (2004). The endocrine and reproductive system: Adverse effects of hormonally active substances? Pediatrics 113(4 Suppl):1070–1075.
  • Gutendorf B, Westendorf J. (2001). Comparison of an array of in vitro assays for the assessment of the estrogenic potential of natural and synthetic estrogens, phytoestrogens and xenoestrogens. Toxicology 166:79–89.
  • Herbst AL, Ulfelder H, Poskanzer DC. (1971). Adenocarcinoma of the vagina. Association of maternal stilbestrol therapy with tumor appearance in young women. N Engl J Med 284:878–881.
  • Hoshi M, Morimura K, Wanibuchi H, Wei M, Okochi E, Ushijima T, Takaoka K, Fukushima S. (2004). No-observed effect levels for carcinogenicity and for in vivo mutagenicity of a genotoxic carcinogen. Toxicol Sci 81:273–279.
  • Janssen P, Speijers GAB. (1997). Guidance on the derivation of maximum permissible risk levels for human intake of soil contaminants. RIVM report no. 711701006. Bilthoven, The Netherlands: National Institute of Public Health and the Environment.
  • Kaufmann WK. (2007). Initiating the uninitiated: Replication of damaged DNA and carcinogenesis. Cell Cycle 6:1460–1467.
  • Kavlock RJ. (1997). Recent advances in mathematical modeling of developmental abnormalities using mechanistic information [review]. Reprod Toxicol 11:423–434.
  • Ladyman SR, Augustine RA, Grattan DR. (2010). Hormone interactions regulating energy balance during pregnancy. J Neuroendocrinol 22:805–817.
  • Langman LJ, Kapur BM. (2006). Toxicology: Then and now. Clin Biochem 39:498–510.
  • Lapham LW, Cernichiari E, Cox C, Myers GJ, Baggs RB, BrewerR Shamlaye, CF, Davidson PW, Clarkson TW. (1995). An analysis of autopsy brain tissue from infants prenatally exposed to methymercury. Neurotoxicology 16:689–704.
  • Lau C, Setzer RW. (2000). Biologically based risk assessment models for developmental toxicity. Methods Mol Biol 136:271–281.
  • Lenz W. (1988). A short history of thalidomide embryopathy. Teratology 38:203–215.
  • Lewandowski TA, Ponce RA, Charleston JS, Hong S, Faustman EM. (2003). Effect of methylmercury on midbrain cell proliferation during organogenesis: Potential cross-species differences and implications for risk assessment. Toxicol Sci 75:124–133.
  • Loeb LA, Harris CC. (2008). Advances in chemical carcinogenesis: A historical review and prospective. Cancer Res 68:6863–6872.
  • Luo T, Sakai Y, Wagner E, Dräger UC. (2006). Retinoids, eye development, and maturation of visual function. J Neurobiol 66:677–686.
  • Lutz WK, Gaylor DW, Conolly RB, Lutz RW. (2005). Nonlinearity and thresholds in dose-response relationships for carcinogenicity due to sampling variation, logarithmic dose scaling, or small differences in individual susceptibility. Toxicol Appl Pharmacol 207(2 Suppl):565–569.
  • Maden M, Holder N. (1992). Retinoic acid and development of the central nervous system. Bioessays 14:431–438.
  • Matthews JB, Twomey K, Zacharewski TR. (2001). In vitro and in vivo interactions of bisphenol A and its metabolite, bisphenol A glucuronide, with estrogen receptors alpha and beta. Chem Res Toxicol 14:149–157.
  • Mattingly RF, Stafl A. (1976). Cancer risk in diethylstilbestrol-exposed offspring. Am J Obstet Gynecol 126:543–548.
  • McLachlan JA. (1979). Transplacental effects of diethylstilbestrol in mice. Natl Cancer Inst Monogr 51:67–72.
  • Myers JP, vom Saal FS, Akingbemi BT, Arizono K, Belcher S, Colborn T, Chahoud I, Crain DA, Farabollini F, Guillette LJ Jr, Hassold T, Ho SM, Hunt PA, Iguchi T, Jobling S, Kanno J, Laufer H, Marcus M, McLachlan JA, Nadal A, Oehlmann J, Olea N, Palanza P, Parmigiani S, Rubin BS, Schoenfelder G, Sonnenschein C, Soto AM, Talsness CE, Taylor JA, Vandenberg LN, Vandenbergh JG, Vogel S, Watson CS, Welshons WV, Zoeller RT. (2009). Why public health agencies cannot depend on good laboratory practices as a criterion for selecting data: The case of bisphenol A. Environ Health Perspect 117:309–315.
  • Mulder GB, Manley N, Grant J, Schmidt K, Zeng W, Eckhoff C, Maggio-Price L. (2000). Effects of excess vitamin A on development of cranial neural crest-derived structures: A neonatal and embryologic study. Teratology 62:214–226.
  • Muller L, Gocke E, Lave T, Pfister T. (2009). Ethyl methanesulfonate toxicity in Viracept—A comprehensive human risk assessment based on threshold data for genotoxicity. Toxicol Lett 190:317–329.
  • Nagar S, Remmel RP. (2006). Uridine diphosphoglucuronosyltransferase pharmacogenetics and cancer. Oncogene 25:1659–1672.
  • Nagel SC, vom Saal FS, Thayer KA, Dhar MG, Boechler M, Welshons WV. (1997). Relative binding affinity-serum modified access (RBA-SMA) assay predicts the relative in vivo bioactivity of the xenoestrogens bisphenol A and octylphenol. Environ Health Perspect 105:70–76.
  • Nau H. (1985). Teratogenic valproic acid concentrations: Infusion by implanted minipumps vs conventional injection regimen in the mouse. Toxicol Appl Pharmacol 80:243–250.
  • Neubert D. (2002). Reproductive toxicology: The science today. Teratog Carcinog Mutagen 22:159–174.
  • Neumann HG. (2009). Risk assessment of chemical carcinogens and thresholds. Crit Rev Toxicol 39:449–461.
  • Nishimura H, Hirota S, Tanaka O, Ueda M, Uno T. (1974). Normal mercury level in human embryos and fetuses. Biol Neonate 24:197–205.
  • OECD. (2001). OECD TG 414. Prenatal Developmental Toxicity Study. http://www.oecd.org/dataoecd/18/15/1948482.pdf.
  • OECD. (2001). OECD TG 416. Two-Generation Reproductive Toxicity Study. http://www.oecd.org/dataoecd/18/13/1948466.pdf.
  • Owens JW, Chaney JG. (2005). Weighing the results of differing ‘low dose’ studies of the mouse prostate by Nagel, Cagen, and Ashby: Quantification of experimental power and statistical results. Regul Toxicol Pharmacol 43:194–202, Sept 17, 2010.
  • Peters PWJ, Garbis-Berkvens JM. (1996). General reproductive toxicology. In: Niesink RJM, de Vries J, Hollinger MA, eds. Toxicology. Boca Raton, FL: CRC Press, 948–947, Sept 17, 2010.
  • Piersma AH, Verhoef A, te Biesebeek JD, Pieters MN, Slob W. (2000). Developmental toxicity of butylbenzylphthalate in the rat using a multiple dose study design. Reprod Toxicol 14:417–425.
  • Pratt I, Barlow S, Kleiner J, Larsen JC. (2009). The influence of thresholds on the risk assessment of carcinogens in food. Mutat Res 678:113–117.
  • Renwick AG. (2005). Structure-based thresholds for toxicological concern—Guidance for application to substances present at low levels in the diet. Toxicol Appl Pharmacol 207:S585–S591
  • Rivas A, McKinnell C, Fisher JS, Atanassova N, Williams K, Sharpe RM. (2003). Neonatal coadministration of testosterone with diethylstilbestrol prevents diethylstilbestrol induction of most reproductive tract abnormalities in male rats. J Androl 24:557–567.
  • Rosa FW. (1986). Retinoic acid embryopathy. N Engl J Med 315:262.
  • Rubin BS, Murray MK, Damassa DA, King JC, Soto AM. (2001). Perinatal exposure to low doses of bisphenol A affects body weight, patterns of estrous cyclicity, and plasma LH levels. Environ Health Perspect 109:675–680.
  • Scadding SR, Maden M. (1994). Retinoic acid gradients during limb regeneration. Dev Biol 162:608–617.
  • Sharpe RM. (2010). Is it time to end concerns over the estrogenic effects of bisphenol A? Toxicol Sci 114:1–4.
  • Sheweita SA, Tilmisany AK. (2003). Cancer and phase II drug-metabolizing enzymes. Curr Drug Metab 4:45–58.
  • Shin JH, Moon HJ, Kang IH, Kim TS, Kim IY, Park IS, Kim HS, Jeung EB, Han SY. (2006). Repeated 28-day oral toxicity study of ketoconazole in rats based on the draft protocol for the “Enhanced OECD Test Guideline No. 407” to detect endocrine effects. Arch Toxicol 80:797–803.
  • Singh MS, Michael M. (2009). Role of xenobiotic metabolic enzymes in cancer epidemiology. Methods Mol Biol 472:243–264.
  • Slob W. (2002). Dose-response modeling of continuous endpoints. Toxicol Sci 66:298–312.
  • Slob W, Pieters MN. (1998). A probabilistic approach for deriving acceptable human intake limits and human health risks from toxicological studies: General framework. Risk Anal 18:787–798.
  • Slob W, Moerbeek M, Rauniomaa E, Piersma AH. (2005). A statistical evaluation of toxicity study designs for the estimation of the benchmark dose in continuous endpoints. Toxicol Sci 84:167–185.
  • Snow ET, Sykora P, Durham TR, Klein CB. (2005). Arsenic, mode of action at biologically plausible low doses: What are the implications for low dose cancer risk? Toxicol Appl Pharmacol 207(2 Suppl):557–564.
  • Sulik KK, Dehart DB, Rogers JM, Chernoff N. (1995). Teratogenicity of low doses of all-trans retinoic acid in presomite mouse embryos. Teratology 51:398–403.
  • Szymczak W. (2009). [Quantitative methods of cancer risk assessment in exposure to chemicals] (Polish). Med Pr 60:215–221.
  • Thurnham DI, Northrop-Clewes CA. (1999). Optimal nutrition: Vitamin A and the carotenoids. Proc Nutr Soc 58:449–457.
  • Tyl RW. (2009). Basic exploratory research versus guideline-compliant studies used for hazard evaluation and risk assessment: Bisphenol A as a case study. Environ Health Perspect 117:1644–1651.
  • Tyl RW, Myers CB, Marr MC, Thomas BF, Keimowitz AR, Brine DR, Veselica MM, Fail PA, Chang TY, Seely JC, Joiner RL, Butala JH, Dimond SS, Cagen SZ, Shiotsuka RN, Stropp GD, Waechter JM. (2002). Three-generation reproductive toxicity study of dietary bisphenol A in CD Sprague-Dawley rats. Toxicol Sci 68:121–146.
  • Tyl RW, Myers CB, Marr MC, Sloan CS, Castillo NP, Veselica MM, Seely JC, Dimond SS, Van Miller JP, Shiotsuka RN, Beyer D, Hentges SG, Waechter JM Jr. (2008). Two-generation reproductive toxicity study of dietary bisphenol A in CD-1 (Swiss) mice. Toxicol Sci 104:362–384, Sept 17, 2010.
  • United Nations. (2009). Globally Harmonized System of Classification and Labelling of Chemicals (GHS). Available at: http://www.unece.org/trans/danger/publi/ghs/ghs_rev03/03files_e.html.
  • US EPA. (1986). U.S. Environmental Protection Agency: Guidelines for carcinogen risk assessment. Available at: http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=54933.
  • US EPA. (1999). U.S. Environmental Protection Agency: Guidelines for carcinogen risk assessment. Risk Assessment Forum. SAB Review draft. Available at: http://epa.gov/raf/publications/pdfs/ec15.pdf.
  • van der Ven LT, van de Kuil T, Leonards PE, Slob W, Lilienthal H, Litens S, Herlin M, Håkansson H, Cantón RF, van den Berg M, Visser TJ, van Loveren H, Vos JG, Piersma AH. (2009). Endocrine effects of hexabromocyclododecane (HBCD) in a one-generation reproduction study in Wistar rats. Toxicol Lett 185:51–62.
  • Van Leeuwen CG, Vermeire TG, eds. (2007). Risk Assessment of Chemicals: An Introduction. Dordrecht, The Netherlands: Springer Verlag.
  • Vogelstein B, Kinzler KW. (1993). The multistep nature of cancer. Trends Genet 9:138–141.
  • Vom Saal FS, Cooke PS, Buchanan DL, Palanza P, Thayer KA, Nagel SC, Parmigiani S, Welshons WV. (1998). A physiologically based approach to the study of bisphenol A and other estrogenic chemicals on the size of the reproductive organs, daily sperm production, and behavior. Toxicol Ind Health 14:239–260.
  • Vom Saal FS, Richter CA, Ruhlen RR, Nagel SC, Timms BG, Welshons WV. (2005). The importance of appropriate controls, animal feed, and animal models in interpreting results from low-dose studies of bisphenol A. Birth Defects Res A Clin Mol Teratol 73:140–145.
  • Vorhees CV. (1987). Teratogenicity and developmental toxicity of valproic acid in rats. Teratology 35:195–202.
  • Vorhees CV, Acuff KD, Weisenburger WP, Minck DR. (1990). Teratogenicity of carbamazepine in rats. Teratology 41:311–317.
  • VROM. (1989). Memorandum “Dealing with risks” (in Dutch: “Omgaan met risico’s”). Appendix no. 5 of the National Plan for the Environment of 1989. Ministry of Housing, Spatial Planning and the Environment. Lower House of the Dutch Parliament, meeting year 1988/1989, document 21.137 no. 5. The Hague, The Netherlands: SDU Publishers.
  • Waddell WJ, Fukushima S, Williams GM. (2006). Concordance of thresholds for carcinogenicity of N-nitrosodiethylamine. Arch Toxicol 80:305–309.
  • Wei M, Hori T-a Ichihara, T, Wanibuchi H, Morimura K, Kang JS, Puatanachokchai R, Fukushima S. (2006). Existence of no-observed effect levels for 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline on hepatic preneoplastic lesion development in BN rats. Cancer Lett 231:304–308.
  • Wheeler MW, Bailer AJ. (2007). Properties of model-averaged BMDLs: A study of model averaging in dichotomous response risk estimation. Risk Anal 27:659–670.
  • Williams GM, Iatropoulos MJ, Jeffrey AM. (2004). Thresholds for the effects of 2-acetylaminofluorene in rat liver. Toxicol Pathol 32(Suppl 2):85–91.
  • Wilson J. (1977). Current status of teratology: General principles and mechanisms derived from animal studies. In: Wilson J, Fraser F, eds. Handbook of Teratalogy. 4 Vols. New York: Plenum Press, Volume 1: 47–74.
  • Woo GH, Shibutani M, Ichiki T, Hamamura M, Lee KY, Inoue K, Hirose M. (2007). A repeated 28-day oral dose toxicity study of nonylphenol in rats, based on the ‘Enhanced OECD Test Guideline 407′ for screening of endocrine-disrupting chemicals. Arch Toxicol 81:77–88.

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.