74
Views
3
CrossRef citations to date
0
Altmetric
Review

In-vitro screening of the antiestrogenic activity of chemicals

&
Pages 605-617 | Published online: 18 May 2008

Bibliography

  • Colborn T, Vom Saal FS, Soto AM. Developmental effects of endocrine-disrupting chemicals in wildlife and humans. Environ Health Perspect 1993;101:378-84
  • Couse JF, Korach KS. Estrogen receptor null mice: What have we learned and where will they lead us? Endocr Rev 1999;20:358-417
  • Pettersson K, Gustafsson J-Å. Role of estrogen receptor β in estrogen action. Annu Rev Physiol 2001;63:165-92
  • Matthews J, Gustafsson J-Å. Estrogen signalling: a subtle balance between ER alpha and ER beta. Mol Interv 2003;3:281-92
  • Safe S. Modulation of gene expression and endocrine response pathways by 2,3,7,8-tetrachlorodibenzo-p-dioxin and related compounds. Pharmac Ther 1995;67:247-81
  • Navas JM, Segner H. Antiestrogenic activity of anthropogenic and natural chemicals. Environ Sci Pollut Res 1998;5:75-82
  • McMaster ME, Van der Kraak GJ, Munkitrick KR. Exposure to bleached kraft pulp mill effluent reduces the steroid biosynthetic capacity of white sucker ovarian follicles. Comp Biochem Physiol 1995;112(C):169-78
  • Hilscherova K, Jones PD, Gracia T, et al. Assessment of the effects of chemicals on the expression of ten steroidogenic genes in the H295R cell line using real time PCR. Toxicol Sci 2004;81:78-89
  • Hecker M, Hollert H, Cooper R, et al. The OECD validation program of the H295R steroidogenesis assay for the identification of in vitro inhibitors and inducers of testosterone and estradiol production. Phase 2: inter-laboratory pre-validation studies. Env Sci Pollut Res 2007;14(Special Issue 1):23-30
  • Sánchez R, Nguyen D, Rocha W, White JH, Mader S. Diversity in the mechanisms of gene regulation by estrogen receptors. BioEssays 2002;24:244-54
  • Barkhem T, Carlsson B, Nilsson Y, et al. Differential response of estrogen receptor α and estrogen receptor β to partial estrogen agonists/antagonists. Mol Pharmacol 1998;54:105-12
  • Hall JM, McDonnell DP. The estrogen receptor β-isoform (ERβ) of the human estrogen receptor modulates ERα transcriptional activity and is a key regulator of the cellular response to estrogens and antiestrogens. Endocrinology 1999;140:5566-78
  • Cheskis BJ, Greger JG, Nagpal S, Freedman LP. Signalling by estrogens. J Cell Physiol 2007;213:610-7
  • Heldring N, Pike A, Andersson S, et al. Estrogen receptors: how do they signal and what are their targets? Physiol Rev 2007;87:905-31
  • Pettersson K, Grandien K, Kuiper GG, Gustafsson J-Å. Mouse estrogen receptor β forms estrogen response element-binding heterodimers with estrogen receptor α. Mol Endocrinol 1997;11:1486-96
  • Ogawa S, Inoue S, Watanabe T, et al. The complete primary structure of human estrogen receptor β (hERβ) and its heterodimerization with ER α in vivo and in vitro. Biochem Biophys Res Commun 1998;243:122-6
  • Tsai MJ, O'Malley BW. Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Annu Rev Biochem 1994;63:451-86
  • Björnström L, Sjöberg M. Mechanisms of estrogen receptor signaling: convergence of genomic and nongenomic actions on target genes. Mol Endocrinol 2005;19:833-42
  • Coleman KM, Smith CL. Intracellular signalling pathways: nongenomic actions of estrogens and ligand-independent activation of estrogen receptors. Front Biosci 2001;6:D1379-91
  • Brzozowski AM, Pike AC, Dauter Z, et al. Molecular basis of agonism and antagonism on the oestrogen receptor. Nature 1997;389:753-8
  • McDonnell DP. The molecular pharmacology of SERMs. Trends Endocrinol Metab 1999;10:301-11
  • Katzenellenbogen BS, Choi I, Delage-Mourroux R, et al. Molecular mechanisms of estrogen action: selective ligands and receptor pharmacology. J Steroid Biochem Mol Biol 2000;74:279-85
  • Frasor J, Stossi S, Danes JM, et al. Selective estrogen receptor modulators: discrimination of agonistic versus antagonistic activities by gene expression profiling in breast cancer cells. Cancer Res 2004;64:1522-33
  • Shang Y, Brown M. Molecular determinants for the tissue specificity of SERMs. Science 2002;295:2465-8
  • Orti E, Bodwell JE, Munck A. Phosphorylation of steroid hormone receptors. Endocr Rev 1992;13:105-28
  • Kato S, Endoh H, Masuhiro Y, et al. Activation of the estrogen receptor through phosporylation by mitogen-activated protein kinase. Science 1995;270:1491-4
  • Le Goff P, Montano MM, Schodin DJ, Katzenellenbogen BS. Phosphorylation of the human estrogen receptor: Identification of hormone-regulated sites and examination of their influence on transcriptional activity. J Biol Chem 1994;269:4458-66
  • Weigel NL. Steroid hormone receptors and their regulation by phosphorylation. Biochem J 1996;319:657-67
  • Aronica SM, Katzenellenbogen BS. Stimulation of estrogen receptor-mediated transcription and alteration in the phosphorylation state of the rat uterine estrogen receptor by estrogen, cyclic adenosine monophosphate, and insulin-like growth factor-I. Mol Endocrinol 1993;7:743-52
  • Davila DR, Mounho BJ, Burchiel SW. Toxicity of polyaromatic hydrocarbons to the human immune system: models and mechanisms. Toxicol Ecotoxicol News 1997;4:5-9
  • Enan E, El-Sabeawy F, Moran F, et al. Interruption of estradiol signal transduction by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) through disruption of the protein phosphorylation pathway in adipose tissues from immature and mature female rats. Biochem Pharmacol 1998;55:1077-90
  • Katzenellenbogen JA, O'Malley BW, Katzenellenbogen BS. Tripartite steroid hormone receptor pharmacology: Interaction with multiple effector sites as a basis for the cell- and promoter-specific action of these hormones. Mol Endocrinol 1996;10:119-31
  • Bollag W, Peck R, Frey JR. Inhibition of proliferation by retinoids, cytokines and their combination in four transformed epithelial cell lines. Cancer Lett 1992;62:167-72
  • Segars JH, Marks MS, Hirschfeld S, et al. Inhibition of estrogen-responsive gene activation by the retinoid X receptor β: Evidence for multiple inhibitory pathways. Mol Cell Biol 1993;4:2258-68
  • Pottinger TG, Pickering AD. The effect of cortisol administration on hepatic and plasma estradiol binding capacity in immature female rainbow trout. Gen Comp Endocrinol 1990;80:264-73
  • Bemanian V, Male R, Goksoyr A. The aryl hydrocarbon receptor-mediated disruption of vitellogenin synthesis in the fish liver: cross-talk between AHR- and ERα-signaling pathways. Comp Hepatol 2004;3:2
  • Cheshenko K, Pakdel F, Segner H, et al. Interference of endocrine disrupting chemicals with aromatase CYP19 expression or activity, and consequences for reproduction of teleost fish. Gen Comp Endocrinol 2008;155:31-62
  • Navas JM, Segner H. Antiestrogenicity of β-naphthoflavone and PAHs in cultured rainbow trout hepatocytes: evidence for a role of the arylhydrocarbon receptor. Aquat Toxicol 2000;51:79-92
  • Smeets JMW, Van Holsteijn I, Giesy JP, Van den Berg M. The anti-estrogenicity of Ah receptor agonists in carp (Cyprinus carpio) hepatocytes. Toxicol Sci 1999;52:178-88
  • Safe S, Wormke M. Inhibitory aryl hydrocarbon receptor-estrogen receptor a cross-talk and mechanisms of action. Chem Res Toxicol 2003;16:807-16
  • Ohtake F, Takeyama K, Matsumoto T, et al. Modulation of oestrogen receptor signalling by association with the activated dioxin receptor. Nature 2003;423:545-50
  • Brunnberg S, Pettersson K, Rydin E, et al. The basic helix-loop-helix-PAS protein ARNT functions as a potent coactivator of estrogen receptor-dependent transcription. Proc Natl Acad Sci USA 2003;100:6517-22
  • Hahn ME. Aryl hydrocarbon receptors: diversity and evolution. Chem Biol Interact 2002;141:131-60
  • Nebert DW, Puga A, Vasiliou V. Role of the Ah receptor and the dioxin-inducible toxicity, cancer, and signal transduction. Ann NY Acad Sci 1993;685:624-40
  • Denison MS, Nagy SR. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol 2003;43:309-34
  • Navas JM, Chana A, Herradón B, Segner H. Induction of cytochrome P4501A (CYP1A) by clotrimazole, a nonplanar aromatic compound. Computational studies on structural features of clotrimazole and related imidazole derivatives. Life Sci 2004;76:699-714
  • Casado S, Alonso M, Herradón B, et al. Activation of the aryl hydrocarbon receptor by carbaryl: Computational evidence of the ability of carbaryl to assume a planar conformation. Environ Toxicol Chem 2006;25:3141-7
  • Babín M, Casado S, Chana A, et al. Cytochrome P4501A induction caused by the imidazole derivative Prochloraz in a rainbow trout cell line. Toxicol in vitro 2005;19:899-902
  • Denison MS, Whitlock JP Jr. Xenobiotic-inducible transcription of cytochrome P450 genes. J Biol Chem 1995;270:18175-8
  • Rowlands JC, Gustafsson JA. Aryl hydrocarbon receptor-mediated signal transduction. Crit Rev Toxicol 1997;27:109-34
  • Stegeman JJ, Hahn ME. Biochemistry and molecular biology of monooxygenases: Current perspectives on forms, functions, and regulation of cytochrome P450 in aquatic species. In: Malins DC, Ostrander GK, editors, Aquat toxicol. Boca Raton, FL: CRC; 1994. p. 87-206
  • Spink DC, Lincoln DW, Dickerman HW, Gierthy JF. 2,3,7,8-tetrachlorodibenzo-p-dioxin causes an extensive alteration of 17β-estradiol metabolism in human breast cancer cells. Proc Natl Acad Sci USA 1990;87:6971-21
  • Pocar P, Fischer B, Klonisch T, Hombach-Klonisch S. Molecular interactions of the aryl hydrocarbon receptor and its biological and toxicological relevance for reproduction Reproduction 2005;129:379-89
  • Ricci MS, Toscano DG, Mattingly CJ, Toscano WA Jr. Estrogen receptor reduces CYP1A1 induction in cultured human endometrial cells. J Biol Chem 1999;274:3430-8
  • Callige M, Richard-Foy H. Ligand-induced estrogen receptor alpha degradation by the proteasome: new actors? Nucl Recept Signal 2006;4:e004
  • Wormke M, Stoner M, Saville B, et al. The aryl hydrocarbon receptor mediates degradation of estrogen receptor alpha through activation of proteasomes. Mol Cell Biol 2003;23:1843-55
  • Wijayaratne AL, McDonnell DP. The human estrogen receptor-α is an ubiquitinated protein whose stability is affected differentially by agonists, antagonists, and selective estrogen receptor modulators. J Biol Chem 2001;276:35684-92
  • Wang F, Samudio I, Safe S. Transcriptional activation of cathepsin D gene expression by 17β-estradiol: mechanism of aryl hydrocarbon receptor-mediated inhibition. Mol Cell Endocrinol 2001;172:91-103
  • Klinge CM, Bowers JL, Kulakosky PC, et al. The aryl hydrocarbon receptor (AHR)/AHR nuclear translocator (ARNT) heterodimer interacts with naturally occurring estrogen response elements. Mol Cell Endocrinol 1999;157:105-19
  • Krishnan V, Porter W, Santostefano M, et al. Molecular mechanism of inhibition of estrogen-induced cathepsin D gene expression by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in MCF-7 cells. Mol Cell Endocrinol 1995;15:6710-9
  • Mortensen AS, Arukwe A. Interactions between estrogen- and Ah-Receptor signalling pathways in primary culture of salmon hepatocytes exposed to nonylphenol and 3,3′,4,4′-tetrachlorobiphenyl (PCB 77). Comp Hepatol 2007;6:1-14. Available from: www.comparative-hepatology.com/content/6/1/2 [Last accessed 20 February 2008]
  • Navas JM, Segner H. Estrogen-mediated suppression of cytochrome P4501A (CYP1A) expression in rainbow trout hepatocytes: Role of estrogen receptor. Chem Biol Interac 2001;138:285-98
  • Abdelrahim M, Ariazi F, Kim K, et al. 3-methylcholanthrene and other aryl hydrocarbon receptor agonists directly activate estrogen receptor alpha. Cancer Res 2006;66:2459-60
  • Lind PM, Eriksen EF, Sahlin L, et al. Effects of the antiestrogenic environmental pollutant 3,3′,4,4′,5-pentachlorobiphenyl (PCB #126) in rat bone and uterus: diverging effects in ovariectomized and intact animals. Toxicol Appl Pharmacol 1999;154:236-44
  • Charles GD, Bartels MJ, Gennings C, et al. Incorporation of S-9 activation into an ER-α transactivation assay. Reprod Toxicol 2000;14:207-16
  • Sumida K, Ooe N, Nagahori H, et al. An in vitro reporter gene assay method incorporating metabolic activation with human and rat S9 or liver microsomes. Biochem Biophys Res Comm 2001;280:85-91
  • Mueller SO, Kling M, Firzani PA, et al. Activation of estrogen receptor a and estrogen receptor β by 4-methylbenzylidene-camphor in human and rat cells: comparison with phyto- and xenoestrogens. Tox Lett 2003;142:89-101
  • Coecke S, Ahr H, Blaauboer B, et al. Metabolism: A bottleneck in in vitro toxicological test development. ATLA 2006;34:49-84
  • Bernauer U, Glatt HR, Heinrich-Hirsch B, et al. Heterologous expression of mouse cytochrome P450 2E1 in V79 cells: construction and characterisation of the cell line and comparison with V79 cell lines stably expressing rat P450 2E1 and human P450 2E1. ATLA 2003;31:21-30
  • Zacharewski T. in vitro bioassays for assessing estrogenic substances. Environ Sci Technol 1997;31:613-23
  • Charles GD. in vitro models in endocrine disruptor screening. ILAR J 2004;45:494-501
  • Matthews JB, Fertuck KC, Celius T, et al. Ability of structurally diverse natural products and synthetic chemicals to induce gene expression mediated by estrogen receptors from various species. J Steroid Biochem Mol Biol 2002;82:181-94
  • McDonnell DP, Clemm DL, Hermann T, et al. Analysis of estrogen receptor function in vitro reveals three distinct classes of antiestrogens. Mol Endocrinol 1995;9:659-69
  • Bolger R, Wiese TE, Ervin K, et al. Rapid screening of environmental chemicals for estrogen receptor. Environ Health Perspect 1998;106:551-7
  • Mueller SO. Overview of in vitro tools to assess the estrogenic and antiestrogenic activity of phytoestrogens. J Chromatogr B 2002;777:155-65
  • Takeuchi S, Lida M, Kobayashi S, et al. Differential effects of phthalate esters on transcriptional activities via human estrogen receptors alpha and beta, and androgen receptor. Toxicology 2005;210:223-33
  • Kojima H, Katsura E, Takeuchi S, et al. Screening for estrogen and androgen receptor activities in 200 pesticides by in vitro reporter gene assays using Chinese hamster ovary cells. Environ Health Persp 2003;122:524-31
  • Eggen RIL, Segner H. The potential of mechanism-based bioanalytical tools in ecotoxicological exposure and effect assessment. Anal Bioanal Chem 2003;377:386-96
  • Petit F, Le Goff P, Cravédi J-P, et al. Two complementary bioassays for screening the estrogenic potency of xenobiotics: recombinant yeast for trout estrogen receptor and trout hepatocyte cultures. J Mol Endocrinol 1997;19:321-35
  • Rutishauser BV, Pesonen M, Escher BI, et al. Comparative analysis of estrogenic activity in sewage treatment plant effluents involving three in vitro assays and chemical analysis of steroids. Environ Toxicol Chem 2004;23:857-64
  • Routledge EJ, Sumpter JP. Estrogenic activity of surfactants and some of their degradation products assessed using a recombinant yeast screen. Environ Toxicol Chem 1996;15:241-8
  • Zhang QH, Xu Y, Schramm KW, et al. Antiestrogenic and antiprogesteronic activity of tire extracts with yeast-based steroid hormone receptor gene transcription assay. Bull Environ Contam Toxicol 2002;69:863-8
  • Le Guével R, Petit FG, Le Goff P, et al. Inhibition of rainbow trout (Oncorhynchus mykiss) estrogen receptor activity by cadmium. Biol Reprod 2000;63:259-66
  • Tran DQ, Ide CF, McLachlan JA, Arnold SF. The anti-estrogenic activity of selected polynuclear aromatic hydrocarbons in yeast expressing human estrogen receptor. Biochem Biophys Res Comm 1996;229:102-8
  • Harris CA, Routledge EJ, Schaffner C, et al. Benzotriazole is antiestrogenic in vitro but not in vivo. Environ Toxicol Chem 2007;26:2367-72
  • Shiau S-P, Glasebrook A, Hardikar SD, et al. Activation of the human estrogen receptor by estrogenic and antiestrogenic compounds in Saccharomyces cerevisiae: a positive selection system. Gene 1996;179:205-10
  • Yamasaki K, Takeyoshi M, Yakabe Y, et al. Comparison of reporter gene assay and immature rat Uterotrophic assay of twenty-three chemicals. Toxicology 2002;170:21-30
  • Takeyoshi M, Yamasaki K, Sawaki M, et al. The efficacy of endocrine disruptor screening tests in detecting anti-estrogenic effects downstream of receptor-ligand interactions. Toxicol Lett 2002;126:91-8
  • Sonneveld E, Riteco JA, Jansen HJ, et al. Comparison of in vitro and in vivo screening models for androgenic and estrogenic activities. Toxicol Sci 2006;1:173-87
  • Draft report of pre-validation and inter-laboratory validation for stably transfected transciptional activation (TA) assay to detect estrogenic activity. OECD. Available from: www.oecd.org/dataoecd/7/27/37504278.pdf [Last accessed 21 February 2008]
  • Soto AM, Sonnenschein C, Chung KL, et al. The E-SCREEN assay as a tool to identify estrogens: an update on estrogenic environmental pollutants. Environ Health Perspect 1995;103:113-22
  • Olea N, Pulgar R, Pérez P, et al. Estrogenicity of resin-based composites and sealants used in dentistry. Environ Health Perspect 1996;104:298-305
  • Scrimshaw MD, Lester JN. in vitro assays for determination of oestrogenic activity. Anal Bional Chem 2004;378:576-81
  • Ueng T-H, Wang H-W, Huang Y-P, Hung C-C. Antiestrogenic effects of motorcycle exhaust particulate in MCF-7 human breast cancer cells and immature female rats. Arch Environ Contam Toxicol 2004;46:454-62
  • Han D-H, Tachibana H, Yamada K. Inhibition of environmental estrogen-induced proliferation of human breast carcinoma MCF-7 cells by flavonoids. in vitro Cell Dev Biol Anim 2001;37:275-82
  • Oh S-M, Park K, Chung K-H. Combination of in vitro bioassays encompassing different mechanisms to determine the endocrine-disrupting effects of river water. Sci Total Environ 2006;354:252-64
  • Gierthy JF, Bennett JA, Bradley LM, Cutler DS. Correlation of in vitro and in vivo growth suppression of MCF-7 human breast cancer by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Cancer Res 1993;53:3149-53
  • Merchant M, Krishnan V, Safe S. Mechanism of action of a-naphthoflavone as an Ah receptor antagonist in MCF-7 human breast cancer cells. Toxicol Appl Pharmacol 1993;120:179-85
  • Arcaro KF, O'Keefe PW, Yang Y, et al. Antiestrogenicity of environmental polyclcyclic aromatic hydrocarbons in human breast cancer cells. Toxicology 1999;13:115-27
  • Horwitz KB, Costlow ME, McGuire WL. MCF-7; a human breast cancer cell line with estrogen, androgen, progesterone, and glucocorticoid receptors. Steroids 1975;26:785-95
  • Kuil CW, Brouwer A, Van der Saag PT, Van der Burg B. Interference between progesterone and dioxin signal transduction pathways. J Biol Chem 1998;273:8829-34
  • Gierthy JF, Lincoln DW, Roth KE, et al. Estrogen-stimulation of postconfluent cell accumulation and foci formation of human MCF-7 breast cancer cells. J Cell Biochem 1991;45:177-87
  • Baker PR, Wilton JC, Jones CE, et al. Bile acids influence the growth, oestrogen receptor and oestrogen-regulated proteins of MCF-7 human breast cancer cells. Br J Cancer 1992;65:566-72
  • Weisz A, Rosales R. Identification of an estrogen response element upstream of the human c-fos gene that binds the estrogen receptor and the AP-1 transcription factor. Nucl Acids Res 1990;18:5097-106
  • Anderson MJ, Miller MR, Hinton DE. in vitro modulation of 17β-estradiol-induced vitellogenin synthesis: Effects of cytochrome P4501A1 inducing compounds on rainbow trout (Oncorhynchus mykiss) liver cells. Aquat Toxicol 1996;34:327-50
  • Navas JM, Segner H. Vitellogenin synthesis in primary cultures of fish liver cells as endpoint for in vitro screening of the (anti) estrogenic activity of chemical substances. Aquat Toxicol 2006;80:1-22
  • Bergink E, Wallace RA. Precursor-product relationship between amphibian vitellogenin and yolk proteins, LV and phosvitin. J Biol Chem 1974;249:2897-903
  • Mommsen TP, Walsh PJ. Vitellogenesis and oocyte assembly. In: Hoar WS, Randall DJ, editors. Fish physiology, Volume XIA. New York: Academic Press; 1988. p. 347-406
  • Pelissero C, Flouriot G, Foucher JL, et al. Vitellogenin synthesis in cultured hepatocytes; an in vitro test for the estrogenic potency of chemicals. J Steroid Biochem Molec Biol 1993;44:263-72
  • Combes RD. Endocrine disruptors: A critical review of in vitro and in vivo testing strategies for assessing their toxic hazards to humans. ATLA 2000;28:81-118
  • Zacharewski T. Identification and assessment of endocrine disruptors: Limitations of in vivo and in vitro assays. Environ Health Persp 1998;106(Suppl 2):577-82

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.