879
Views
63
CrossRef citations to date
0
Altmetric
Review Article

Mode of action and dose–response framework analysis for receptor-mediated toxicity: The aryl hydrocarbon receptor as a case study

, , , , , , , , , , , , & show all
Pages 83-119 | Received 07 Jan 2013, Accepted 14 Aug 2013, Published online: 19 Nov 2013

References

  • Abdelrahim M, Smith R 3rd, Safe S. (2003). Aryl hydrocarbon receptor gene silencing with small inhibitory rna differentially modulates ah-responsiveness in mcf-7 and hepg2 cancer cells. Mol Pharmacol, 63, 1373–81
  • Abel J, Haarmann-Stemmann T. (2010). An introduction to the molecular basics of aryl hydrocarbon receptor biology. Biol Chem, 391, 1235–48
  • Abraham K, Geusau A, Tosun Y, et al. (2002). Severe 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) intoxication: insights into the measurement of hepatic cytochrome p450 1a2 induction. Clin Pharmacol Ther, 72, 163–74
  • Afford S, Randhawa S. (2000). Demystified… apoptosis. Mol Pathol, 53, 55–63
  • Ahmed S, Valen E, Sandelin A, Matthews J. (2009). Dioxin increases the interaction between aryl hydrocarbon receptor and estrogen receptor alpha at human promoters. Toxicol Sci, 111, 254–66
  • Akahoshi E, Yoshimura S, Uruno S, Ishihara-Sugano M. (2009). Effect of dioxins on regulation of tyrosine hydroxylase gene expression by aryl hydrocarbon receptor: a neurotoxicology study. Environ Health, 8, 24
  • Akhtar FZ, Garabrant DH, Ketchum NS, Michalek JE. (2004). Cancer in us air force veterans of the vietnam war. J Occup Environ Med, 46, 123–36
  • Akintobi AM, Villano CM, White LA. (2007). 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) exposure of normal human dermal fibroblasts results in AhR-dependent and -independent changes in gene expression. Toxicol Appl Pharmacol, 220, 9–17
  • Alison MR. (2005). Liver stem cells: implications for hepatocarcinogenesis. Stem Cell Rev, 1, 253–60
  • Alison MR, Islam S, Lim S. (2009). Stem cells in liver regeneration, fibrosis and cancer: the good, the bad and the ugly. J Pathol, 217, 282–98
  • Aly HA, Domenech O. (2009). Cytotoxicity and mitochondrial dysfunction of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in isolated rat hepatocytes. Toxicol Lett, 191, 79–87
  • Ambolet-Camoit A, Bui LC, Pierre S, et al. (2010). 2,3,7,8-Tetrachlorodibenzo-p-dioxin counteracts the p53 response to a genotoxicant by upregulating expression of the metastasis marker agr2 in the hepatocarcinoma cell line hepg2. Toxicol Sci, 115, 501–12
  • Anandatheerthavarada HK, Vijayasarathy C, Bhagwat SV, et al. (1999). Physiological role of the n-terminal processed p4501a1 targeted to mitochondria in erythromycin metabolism and reversal of erythromycin-mediated inhibition of mitochondrial protein synthesis. J Biol Chem, 274, 6617–25
  • Andersen ME, Barton HA. (1999). Biological regulation of receptor-hormone complex concentrations in relation to dose-response assessments for endocrine-active compounds. Toxicol Sci, 48, 38–50
  • Andersen ME, Birnbaum LS, Barton HA, Eklund CR. (1997). Regional hepatic cyp1a1 and cyp1a2 induction with 2,3,7,8-tetrachlorodibenzo-p-dioxin evaluated with a multicompartment geometric model of hepatic zonation. Toxicol Appl Pharmacol, 144, 145–55
  • Andersen ME, Conolly RB. (1998). Mechanistic modeling of rodent liver tumor promotion at low levels of exposure: an example related to dose-response relationships for 2,3,7,8-tetrachlorodibenzo-p-dioxin. Hum Exp Toxicol, 17, 683–90
  • Andersen ME, Mills JJ, Gargas ML, et al. (1993). Modeling receptor-mediated processes with dioxin: implications for pharmacokinetics and risk assessment. Risk Anal, 13, 25–36
  • Andersen ME, Mills JJ, Jirtle RL, Greenlee WF. (1995). Negative selection in hepatic tumor promotion in relation to cancer risk assessment. Toxicology, 102, 223–37
  • Andersen ME, Preston RJ, Maier A, et al. (2013). Dose-response approaches for nuclear receptor-mediated modes of action for liver carcinogenicity: results of a workshop. Crit Rev Toxicol, in press
  • Andersson P, McGuire J, Rubio C, et al. (2002). A constitutively active dioxin/aryl hydrocarbon receptor induces stomach tumors. Proc Natl Acad Sci USA, 99, 9990–5
  • Apte U, Thompson MD, Cui S, et al. (2008). Wnt/beta-catenin signaling mediates oval cell response in rodents. Hepatology, 47, 288–95
  • Badawi AF, Cavalieri EL, Rogan EG. (2000). Effect of chlorinated hydrocarbons on expression of cytochrome P450 1A1, 1A2 and 1B1 and 2- and 4-hydroxylation of 17beta-estradiol in female Sprague-Dawley rats. Carcinogenesis, 21, 1593–9
  • Baker TK, Kwiatkowski AP, Madhukar BV, Klaunig JE. (1995). Inhibition of gap junctional intercellular communication by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in rat hepatocytes. Carcinogen, 16, 2321–6
  • Bars RG, Elcombe CR. (1991). Dose-dependent acinar induction of cytochromes P450 in rat liver. Evidence for a differential mechanism of induction of P450IA1 by beta-naphthoflavone and dioxin. Biochem J, 277, 577–80
  • Bauman JW, Goldsworthy TL, Dunn CS, Fox TR. (1995). Inhibitory effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on rat hepatocyte proliferation induced by 2/3 partial hepatectomy. Cell Prolif, 28, 437–51
  • Behari J. (2010). The Wnt/β-catenin signaling pathway in liver biology and disease. Expert Rev Gastroenterol Hepatol, 4, 745–56
  • Bock KW, Kohle C. (2005). Ah receptor- and TCDD-mediated liver tumor promotion: clonal selection and expansion of cells evading growth arrest and apoptosis. Biochem Pharmacol, 69, 1403–8
  • Bock KW, Kohle C. (2006). Ah receptor: dioxin-mediated toxic responses as hints to deregulated physiologic functions. Biochem Pharmacol, 72, 393–404
  • Boffetta P, Mundt KA, Adami HO, et al. (2011). TCDD and cancer: a critical review of epidemiologic studies. Crit Rev Toxicol, 41, 622–36
  • Boobis AR, Cohen SM, Dellarco V, et al. (2006). Ipcs framework for analyzing the relevance of a cancer mode of action for humans. Crit Rev Toxicol, 36, 781–92
  • 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
  • Boutros PC, Yao CQ, Watson JD, et al. (2011). Hepatic transcriptomic responses to TCDD in dioxin-sensitive and dioxin-resistant rats during the onset of toxicity. Toxicol Appl Pharmacol, 251, 119–29
  • Boverhof DR, Burgoon LD, Tashiro C, et al. (2005). Temporal and dose-dependent hepatic gene expression patterns in mice provide new insights into TCDD-mediated hepatotoxicity. Toxicol Sci, 85, 1048–63
  • Boverhof DR, Burgoon LD, Tashiro C, et al. (2006). Comparative toxicogenomic analysis of the hepatotoxic effects of TCDD in Sprague DawleyDawley rats and c57bl/6 mice. Toxicol Sci, 94, 398–416
  • Braeuning A. (2009). Regulation of cytochrome P450 expression by Ras- and beta-catenin-dependent signaling. Curr Drug Metab, 10, 138–58
  • Braeuning A, Schwarz M. (2010). beta-Catenin as a multilayer modulator of zonal cytochrome P450 expression in mouse liver. Biol Chem, 391, 139–48
  • Braeuning A, Ittrich C, Köhle C, et al. (2009). Differential gene expression in periportal and perivenous mouse hepatocytes. FEBS J, 273, 5051–61
  • Brix AE, Nyska A, Haseman JK, et al. (2005). Incidences of selected lesions in control female harlan Sprague-DawleyDawley rats from two-year studies performed by the national toxicology program. Toxicol Pathol, 33, 477–83
  • Brunnberg S, Andersson P, Lindstam M, et al. (2006). The constitutively active ah receptor (ca-AHR) mouse as a potential model for dioxin exposure – effects in vital organs. Toxicology, 224, 191–201
  • Buchmann A, Stinchcombe S, Korner W, et al. (1994). Effects of 2,3,7,8-tetrachloro- and 1,2,3,4,6,7,8-heptachlorodibenzo-p-dioxin on the proliferation of preneoplastic liver cells in the rat. Carcinogenesis, 15, 1143–50
  • Buckley DB, Klaassen CD. (2009). Induction of mouse udp-glucuronosyltransferase mrna expression in liver and intestine by activators of aryl-hydrocarbon receptor, constitutive androstane receptor, pregnane x receptor, peroxisome proliferator-activated receptor alpha, and nuclear factor erythroid 2-related factor 2. Drug Metab Dispos, 37, 847–56
  • Budinsky RA, LeCluyse EL, Ferguson SS, et al. (2010). Human and rat primary hepatocyte cyp1a1 and 1a2 induction with 2,3,7,8-tetrachlorodibenzo-p-dioxin, 2,3,7,8-tetrachlorodibenzofuran, and 2,3,4,7,8-pentachlorodibenzofuran. Toxicol Sci, 118, 224–35
  • Budinsky RA, Paustenbach D, Fontaine D, et al. (2006). Recommended relative potency factors for 2,3,4,7,8-pentachlorodibenzofuran: the impact of different dose metrics. Toxicol Sci, 91, 275–85
  • Bunger MK, Glover E, Moran SM, et al. (2008). Abnormal liver development and resistance to 2,3,7,8-tetrachlorodibenzo-p-dioxin toxicity in mice carrying a mutation in the DNA-binding domain of the aryl hydrocarbon receptor. Toxicol Sci, 106, 83–92
  • Bunger MK, Moran SM, Glover E, et al. (2003). Resistance to 2,3,7,8-tetrachlorodibenzo-p-dioxin toxicity and abnormal liver development in mice carrying a mutation in the nuclear localization sequence of the aryl hydrocarbon receptor. J Biol Chem, 278, 17767–74
  • Cain K, Freathy C. (2001). Liver toxicity and apoptosis: role of TGF-β1, cytochrome c and the apoptosome. Toxicol Lett, 120, 307–15
  • Calvert GM, Hornung RW, Sweeney MH, et al. (1992). Hepatic and gastrointestinal effects in an occupational cohort exposed to 2,3,7,8-tetrachlorodibenzo-para-dioxin. JAMA, 267, 2209–14
  • Cardinale V, Semeraro R, Torrice A, et al. (2010). Intra-hepatic and extra-hepatic cholangiocarcinoma: new insight into epidemiology and risk factors. World J Gastrointest Oncol, 2, 407–16
  • Carlson EA, McCulloch C, Koganti A, et al. (2009). Divergent transcriptomic responses to aryl hydrocarbon receptor agonists between rat and human primary hepatocytes. Toxicol Sci, 112, 257–72
  • Carney SA, Peterson RE, Heideman W. (2004). 2,3,7,8-Tetrachlorodibenzo-p-dioxin activation of the aryl hydrocarbon receptor/aryl hydrocarbon receptor nuclear translocator pathway causes developmental toxicity through a CYP1A-independent mechanism in zebrafish. Mol Pharmacol, 66, 512–21
  • Cavalieri E, Frenkel K, Liehr JG, et al. (2000). Estrogens as endogenous genotoxic agents – DNA adducts and mutations. J Natl Cancer Inst Monogr, 2000, 75–93
  • Chang H, Wang YJ, Chang LW, Lin P. (2005). A histochemical and pathological study on the interrelationship between TCDD-induced AHR expression, AHR activation, and hepatotoxicity in mice. J Toxicol Environ Health A, 68, 1567–79
  • Chang JT, Chang H, Chen PH, et al. (2007). Requirement of aryl hydrocarbon receptor overexpression for CYP1B1 up-regulation and cell growth in human lung adenocarcinomas. Clin Cancer Res, 13, 38–45
  • Chapman DE, Schiller CM. (1985). Dose-related effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in c57bl/6j and dba/2j mice. Toxicol Appl Pharmacol, 78, 147–57
  • Cheng H, Aylward L, Beall C, et al. (2006). Tcdd exposure-response analysis and risk assessment. Risk Anal, 26, 1059–71
  • Chiaro CR, Patel RD, Marcus CB, Perdew GH. (2007). Evidence for an aryl hydrocarbon receptor-mediated cytochrome p450 autoregulatory pathway. Mol Pharmacol, 72, 1369–79
  • Cho YC, Zheng W, Jefcoate CR. (2004). Disruption of cell–cell contact maximally but transiently activates AHR-mediated transcription in 10t1/2 fibroblasts. Toxicol Appl Pharmacol, 199, 220–38
  • Chopra M, Dharmarajan AM, Meiss G, Schrenk D. (2009). Inhibition of UV-c light-induced apoptosis in liver cells by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Sci, 111, 49–63
  • Chopra M, Gahrs M, Haben M, et al. (2010). Inhibition of apoptosis by 2,3,7,8-tetrachlorodibenzo-p-dioxin depends on protein biosynthesis. Cell Biol Toxicol, 26, 391–401
  • Chopra M, Schrenk D. (2011). Dioxin toxicity, aryl hydrocarbon receptor signaling, and apoptosis-persistent pollutants affect programmed cell death. Crit Rev Toxicol, 41, 292--320
  • Christensen JG, Gonzales AJ, Cattley RC, Goldsworthy TL. (1998). Regulation of apoptosis in mouse hepatocytes and alteration of apoptosis by nongenotoxic carcinogens. Cell Growth Differ, 9, 815–25
  • Christoffels VM, Sassi H, Ruijter JM, et al. (1999). A mechanistic model for the development and maintenance of portocentral gradients in gene expression in the liver. Hepatology, 29, 1180–92
  • Chute JP, Ross JR, McDonnell DP. (2010). Minireview: nuclear receptors, hematopoiesis, and stem cells. Mol Endocrinol, 24, 1–10
  • Coenraads PJ, Olie K, Tang NJ. (1999). Blood lipid concentrations of dioxins and dibenzofurans causing chloracne. Br J Dermatol, 141, 694--7
  • Cohen SM. (1998). Cell proliferation and carcinogenesis. Drug Metab Rev, 30, 339–57
  • Cohen SM, Klaunig J, Meek ME, et al. (2004). Evaluating the human relevance of chemically induced animal tumors. Toxicol Sci, 78, 181–6
  • Cohen SM, Meek ME, Klaunig JE, et al. (2003). The human relevance of information on carcinogenic modes of action: overview. Crit Rev Toxicol, 33, 581–9
  • Cole P, Trichopoulos D, Pastides H, et al. (2003). Dioxin and cancer: a critical review. Regul Toxicol Pharmacol, 38, 378–88
  • Collins JJ, Bodner K, Aylward LL, et al. (2009a). Mortality rates among trichlorophenol workers with exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Am J Epidemiol, 170, 501–6
  • Collins JJ, Bodner K, Aylward LL, et al. (2009b). Mortality rates among workers exposed to dioxins in the manufacture of pentachlorophenol. J Occup Environ Med, 51, 1212–9
  • Congiu M, Mashford ML, Slavin JL, Desmond PV. (2009). Coordinate regulation of metabolic enzymes and transporters by nuclear transcription factors in human liver disease. J Gastroenterol Hepatol, 24, 1038–44
  • Connor KT, Aylward LL. (2006). Human response to dioxin: aryl hydrocarbon receptor (AHR) molecular structure, function, and dose-response data for enzyme induction indicate an impaired human AHR. J Toxicol Environ Health B Crit Rev, 9, 147–71
  • Connor KT, Harris MA, Edwards MR, et al. (2008). Ah receptor agonist activity in human blood measured with a cell-based bioassay: evidence for naturally occurring AH receptor ligands in vivo. J Expo Sci Environ Epidemiol, 18, 369–80
  • Conolly RB, Andersen ME. (1997). Hepatic foci in rats after diethylnitrosamine initiation and 2,3,7,8-tetrachlorodibenzo-p-dioxin promotion: evaluation of a quantitative two-cell model and of cyp 1a1/1a2 as a dosimeter. Toxicol Appl Pharmacol, 146, 281–93
  • Consonni D, Pesatori AC, Zocchetti C, et al. (2008). Mortality in a population exposed to dioxin after the Seveso, Italy, accident in 1976: 25 years of follow-up. Am J Epidemiol, 167, 847–58
  • Crump KS, Chen C, Chiu WA, et al. (2010). What role for biologically based dose-response models in estimating low-dose risk? Environ Health Perspect, 118, 585–8
  • Darwiche H, Petersen BE. (2010). Biology of the adult hepatic pregenitor cell: “ghosts in the machine”. Prog Mol Biol Transl Sci, 97, 229–49
  • Davarinos NA, Pollenz RS. (1999). Aryl hydrocarbon receptor imported into the nucleus following ligand binding is rapidly degraded via the cytosplasmic proteasome following nuclear export. J Biol Chem, 274, 28708–15
  • Degner SC, Papoutsis AJ, Selmin O, Romagnolo DF. (2009). Targeting of aryl hydrocarbon receptor-mediated activation of cyclooxygenase-2 expression by the indole-3-carbinol metabolite 3,3′-diindolylmethane in breast cancer cells. J Nutr, 139, 26–32
  • Della Porta G, Dragani TA, Sozzi G. (1987). Carcinogenic effects of infantile and long-term 2,3,7,8-tetrachlorodibenzo-p-dioxin treatment in the mouse. Tumori, 73, 99--107
  • Dellarco VL, Baetcke K. (2005). A risk assessment perspective: application of mode of action and human relevance frameworks to the analysis of rodent tumor data. Toxicol Sci, 86, 1–3
  • Denison MS, Nagy SR. (2003). Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol, 43, 309–34
  • Dietrich C, Kaina B. (2010). The aryl hydrocarbon receptor (AHR) in the regulation of cell-cell contact and tumor growth. Carcinogenesis, 31, 1319–28
  • DiNatale BC, Schroeder JC, Francey LJ, et al. (2010). Mechanistic insights into the events that lead to synergistic induction of interleukin 6 transcription upon activation of the aryl hydrocarbon receptor and inflammatory signaling. J Biol Chem, 285, 24388–97
  • Dong W, Matsumura F, Kullman SW. (2010). TCDD induced pericardial edema and relative COX-2 expression in medaka (Oryzias Latipes) embryos. Toxicol Sci, 118, 213–23
  • Dragan YP, Schrenk D. (2000). Animal studies addressing the carcinogenicity of TCDD (or related compounds) with an emphasis on tumour promotion. Food Addit Contam, 17, 289–302
  • Dragan YP, Xu XH, Goldsworthy TL, et al. (1992). Characterization of the promotion of altered hepatic foci by 2,3,7,8-tetrachlorodibenzo-p-dioxin in the female rat. Carcinogenesis, 13, 1389–95
  • Dragin N, Shi Z, Madan R, et al. (2008). Phenotype of the cyp1a1/1a2/1b1-/- triple-knockout mouse. Mol Pharmacol, 73, 1844–56
  • Drahushuk AT, McGarrigle BP, Larsen KE, et al. (1998). Detection of cyp1a1 protein in human liver and induction by TCDD in precision-cut liver slices incubated in dynamic organ culture. Carcinogenesis, 19, 1361–8
  • Drahushuk AT, McGarrigle BP, Slezak BP, et al. (1999). Time- and concentration-dependent induction of cyp1a1 and cyp1a2 in precision-cut rat liver slices incubated in dynamic organ culture in the presence of 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol, 155, 127–38
  • Drahushuk AT, McGarrigle BP, Tai HL, et al. (1996). Validation of precision-cut liver slices in dynamic organ culture as an in vitro model for studying cyp1a1 and cyp1a2 induction. Toxicol Appl Pharmacol, 140, 393–403
  • Du L, Neis MM, Ladd PA, Keeney DS. (2006). Differentiation-specific factors modulate epidermal CYP1-4 gene expression in human skin in response to retinoic acid and classic aryl hydrocarbon receptor ligands. J Pharmacol Exp Ther, 319, 1162–71
  • Duncan AW, Taylor MH, Hickey RD, et al. (2010). The ploidy conveyor of mature hepatocytes as a source of genetic variation. Nature, 467, 707–10
  • Eagle H, Levine EM. (1967). Growth regulatory effects of cellular interaction. Nature, 213, 1102–6
  • Enan E, Liu PC, Matsumura FJ. (1992). 2,3,7,8-Tetrachlorodibenzo-p-dioxin causes reduction of glucose transporting activities in the plasma membranes of adipose tissue and pancreas from the guinea pig. Biol Chem, 267, 19785–91
  • Eckle VS, Buchmann A, Bursch W, et al. (2004). Immunohistochemical detection of activated caspases in apoptotic hepatocytes in rat liver. Toxicol Pathol, 32, 9–15
  • Elferink CJ, Ge NL, Levine A. (2001). Maximal aryl hydrocarbon receptor activity depends on an interaction with the retinoblastoma protein. Mol Pharmacol, 59, 664–73
  • Ema M, Ohe N, Suzuki M, et al. (1994). Dioxin binding activities of polymorphic forms of mouse and human arylhydrocarbon receptors. J Biol Chem, 269, 27337–43
  • Fan F, Yan B, Wood G, et al. (1997). Cytokines (IL-1beta and TNFalpha) in relation to biochemical and immunological effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in rats. Toxicology, 116, 9–16
  • Fan Y, Boivin GP, Knudsen ES, et al. (2010). The aryl hydrocarbon receptor functions as a tumor suppressor of liver carcinogenesis. Cancer Res, 70, 212–20
  • Fattore E, Trossvik C, Hakansson H. (2000). Relative potency values derived from hepatic vitamin Areduction in male and female Sprague-DawleyDawley rats following subchronic dietary exposure to individual polychlorinated dibenzo-p-dioxin and dibenzofuran congeners and a mixture thereof. Toxicol Appl Pharmacol, 165, 184–94
  • Faust D, Vondráček J, Krčmář P, et al. (2013). AhR-mediated changes in global gene expression in rat liver progenitor cells. Arch Toxicol, 87, 681–98
  • Fausto N. (2004). Liver regeneration and repair: hepatocytes, progenitor cells, and stem cells. Hepatology, 39, 1477–87
  • Fernandez-Salguero PM, Hilbert DM, Rudikoff S, et al. (1996). Aryl-hydrocarbon receptor-deficient mice are resistant to 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced toxicity. Toxicol Appl Pharmacol, 140, 173–9
  • Fingerhut MA, Halperin WE, Marlow DA, et al. (1991). Cancer mortality in workers exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. N Engl J Med, 324, 212–8
  • Flaveny CA, Murray IA, Chiaro CR, Perdew GH. (2009). Ligand selectivity and gene regulation by the human aryl hydrocarbon receptor in transgenic mice. Mol Pharmacol, 75, 1412–20
  • Flaveny CA, Murray IA, Perdew GH. (2010). Differential gene regulation by the human and mouse aryl hydrocarbon receptor. Toxicol Sci, 114, 217–25
  • Fletcher N, Wahlstrom D, Lundberg R, et al. (2005). 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) alters the mrna expression of critical genes associated with cholesterol metabolism, bile acid biosynthesis, and bile transport in rat liver: a microarray study. Toxicol Appl Pharmacol, 207, 1–24
  • Forgacs AL, Kent MN, Makley MK, et al. (2012). Comparative metabolomic and genomic analyses of TCDD-elicited metabolic disruption in mouse and rat liver. Toxicol Sci, 125, 41–55
  • Fox TR, Best LL, Goldsworthy SM, et al. (1993). Gene expression and cell proliferation in rat liver after 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure. Cancer Res, 53, 2265–71
  • Franc MA, Moffat ID, Boutros PC, et al. (2008). Patterns of dioxin-altered mrna expression in livers of dioxin-sensitive versus dioxin-resistant rats. Arch Toxicol, 82, 809–30
  • Franc MA, Pohjanvirta R, Tuomisto J, Okey AB. (2001). In vivo up-regulation of aryl hydrocarbon receptor expression by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in a dioxin-resistant rat model. Biochem Pharmacol, 62, 1565–78
  • Friedman SL. (2008). Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev, 88, 125–72
  • Furness SG, Whelan F. (2009). The pleiotropy of dioxin toxicity – xenobiotic misappropriation of the aryl hydrocarbon receptor’s alternative physiological roles. Pharmacol Ther, 124, 336–53
  • Furuyama K, Kawaguchi Y, Akiyama H, et al. (2011). Continuous cell supply from a sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet, 43, 34–41
  • Gaikwad, NW, Yang L, Muti P, et al. (2008). The molecular etiology of breast cancer: evidence from biomarkers of risk. Int J Cancer, 122, 1949–57
  • Gasiewicz TA, Henry EC, Collins LL. (2008). Expression and activity of aryl hydrocarbon receptors in development and cancer. Crit Rev Eukaryot Gene Expr, 18, 279–321
  • Gasiewicz TA, Singh KP, Casado FL. (2010). The aryl hydrocarbon receptor has an important role in the regulation of hematopoiesis: implications for benzene-induced hematopoietic toxicity. Chem Biol Interact, 184, 246–51
  • Gaudio E, Carpino G, Cardinale V, et al. (2009). New insights into liver stem cells. Dig Liver Dis, 41, 455–62
  • Ge NL, Elferink CJ. (1998). A direct interaction between the aryl hydrocarbon receptor and retinoblastoma protein: linking dioxin signaling to the cell cycle. J Biol Chem, 273, 22708–13
  • Geusau A, Khorchide M, Mildner M, et al. (2005). 2,3,7,8-tetrachlorodibenzo-p-dioxin impairs differentiation of normal human epidermal keratinocytes in a skin equivalent model. J Invest Dermatol, 124, 275–7
  • Ghezzi I, Cannatelli P, Assennato G, et al. (1982). Potential 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure of seveso decontamination workers: a controlled prospective study. Scand J Work Environ Health, 8, 176–9
  • Giera S, Braeuning A, Köhle C, et al. (2010). Wnt/beta-catenin signaling activates and determines hepatic zonal expression of glutathione S-transferases in mouse liver. Toxicol Sci, 115, 22–33
  • Gil R, Callaghan R, Boix J, et al. (1988). Morphometric and cytophotometric nuclear analysis of altered hepatocyte foci induced by N-nitrosomorpholine (NNM) and aflatoxin B1 (AFB1) in liver of Wistar rats. Virchows Arch B Cell Pathol Incl Mol Pathol, 54, 341–9
  • Glaser SS, Gaudio E, Miller T, et al. (2009). Cholangiocyte proliferation and liver fibrosis. Expert Rev Mol Med, 11, e7, 1--7
  • Goetz ME, Luch A. (2008). Reactive species: a cell damaging rout assisting to chemical carcinogens. Cancer Lett, 266, 73–83
  • Goldfarb S, Pugh TD, Koen H, He YZ. (1983). Preneoplastic and neoplastic progression during hepatocarcinogenesis in mice injected with diethylnitrosamine in infancy. Environ Health Perspect, 50, 149–61
  • Gomez-Duran A, Carvajal-Gonzalez JM, Mulero-Navarro S, et al. (2009). Fitting a xenobiotic receptor into cell homeostasis: how the dioxin receptor interacts with TGF-β signaling. Biochem Pharmacol, 77, 700–12
  • Gonzalez FJ, Fernandez-Salguero P. (1998). The aryl hydrocarbon receptor: studies using the AHR-null mice. Drug Metab Dispos, 26, 1194–8
  • Gonzalez FJ, Fernandez-Salguero P, Lee SS, et al. (1995). Xenobiotic receptor knockout mice. Toxicol Lett, 82–3, 117–21
  • Gonzalez FJ, Fernandez-Salguero P, Ward JM. (1996). The role of the aryl hydrocarbon receptor in animal development, physiological homeostasis and toxicity of TCDD. J Toxicol Sci, 21, 273–7
  • Goodman DG, Sauer RM. (1992). Hepatotoxicity and carcinogenicity in female Sprague-DawleyDawley rats treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD): a pathology working group reevaluation. Regul Toxicol Pharmacol, 15, 245–52
  • Graham MJ, Lucier GW, Linko P, et al. (1988). Increases in cytochrome p-450 mediated 17 beta-estradiol 2-hydroxylase activity in rat liver microsomes after both acute administration and subchronic administration of 2,3,7,8-tetrachlorodibenzo-p-dioxin in a two-stage hepatocarcinogenesis model 1. Carcinogenesis, 9, 1935–41
  • Greenbaum LE, Wells RG. (2011). The role of stem cells in liver repair and fibrosis. Int J Biochem Cell Biol, 43, 222–9
  • Gupta S. (2000). Hepatic polyploidy and liver growth control. Semin Cancer Biol, 10, 161–71
  • Guzelian P, Quattrochi L, Karch N, et al. (2006). Does dioxin exert toxic effects in humans at or near current background body levels?: an evidence-based conclusion. Hum Exp Toxicol, 25, 99–105
  • Haarmann-Stemmann T, Bothe H, Abel J. (2009). Growth factors, cytokines and their receptors as downstream targets of arylhydrocarbon receptor (AHR) signaling pathways. Biochem Pharmacol, 77, 508–20
  • Haarmann-Stemmann T, Bothe H, Kohli A, et al. (2007). Analysis of the transcriptional regulation and molecular function of the aryl hydrocarbon receptor repressor in human cell lines. Drug Metab Dispos, 35, 2262–9
  • Hahn ME, Allan LL, Sherr DH. (2009). Regulation of constitutive and inducible AHR signaling: complex interactions involving the AHR repressor. Biochem Pharmacol, 77, 485–97
  • Hahn ME, Karchner SI, Evans BR, et al. (2006). Unexpected diversity of aryl hydrocarbon receptors in non-mammalian vertebrates: insights from comparative genomics. J Exp Zool A Comp Exp Biol, 305, 693–706
  • Hailey JR, Walker NJ, Sells DM, et al. (2005). Classification of proliferative hepatocellular lesions in harlan Sprague-DawleyDawley rats chronically exposed to dioxin-like compounds. Toxicol Pathol, 33, 165–74
  • Hakansson H, Hanberg A. (1989). The distribution of [14c]-2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and its effect on the vitamin A content in parenchymal and stellate cells of rat liver. J Nutr, 119, 573–80
  • Hanahan D, Weinberg RA. (2011). Hallmarks of cancer: the next generation. Cell, 144, 646–74
  • Hankinson O. (2005). Role of coactivators in transcriptional activation by the aryl hydrocarbon receptor. Arch Biochem Biophys, 433, 379–86
  • Hankinson O. (2009). Repression of aryl hydrocarbon receptor transcriptional activity by epidermal growth factor. Mol Interv, 9, 116–8
  • Harada T, Maronpot RR, Morris RW, et al. (1989). Morphological and stereological characterization of hepatic foci of cellular alteration in control fischer 344 rats. Toxicol Pathol, 17, 579–93
  • Hassoun EA, Li F, Abushaban A, Stohs SJ. (2000). The relative abilities of TCDD and its congeners to induce oxidative stress in the hepatic and brain tissues of rats after subchronic exposure. Toxicology, 145, 103–13
  • Hattis D. (1996). Human interindividual variability in susceptibility to toxic effects: from annoying detail to a central determinant of risk. Toxicology, 111, 5–14
  • Hayes JD, Dinkova-Kostova AT, McMahon M. (2009). Cross-talk between transcription factors AHR and nrf2: lessons for cancer chemoprevention from dioxin. Toxicol Sci, 111, 199–201
  • Head JL, Lawrence BP. (2009). The aryl hydrocarbon receptor is a modulator of anti-viral immunity. Biochem Pharmacol, 77, 642–53
  • Hemming H, Bager Y, Flodstrom S, et al. (1995). Liver tumour promoting activity of 3,4,5,3′,4′-pentachlorobiphenyl and its interaction with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Eur J Pharmacol, 292, 241–9
  • Hernandez-Ochoa I, Karman BN, Flaws JA. (2009). The role of the aryl hydrocarbon receptor in the female reproductive system. Biochem Pharmacol, 77, 547–59
  • Hill AB. (1965). The environment and disease: association or causation? Proc R Soc Med, 58, 295–300
  • Himmelstein MW, Boogaard PJ, Cadet J, et al. (2009). Creating context for the use of DNA adduct data in cancer risk assessment: II. Overview of methods of identification and quantitation of DNA damage. Crit Rev Toxicol, 39, 679–94
  • Hiraku Y, Yamashita N, Nishiguchi M, Kawanishi S. (2001). Catechol estrogens induce oxidative DNA damage and estradiol enhances cell proliferation. Int J Cancer, 92, 333–7
  • Holsapple MP, Pitot HC, Cohen SM, et al. (2006). Mode of action in relevance of rodent liver tumors to human cancer risk. Toxicol Sci, 89, 51–6
  • Huang G, Elferink CJ. (2005). Multiple mechanisms are involved in ah receptor-mediated cell cycle arrest. Mol Pharmacol, 67, 88–96
  • Hushka DR, Greenlee WF. (1995). 2,3,7,8-Tetrachlorodibenzo-p-dioxin inhibits DNA synthesis in rat primary hepatocytes. Mutat Res, 333, 89–99
  • Ichihara S, Yamada Y, Gonzalez FJ, et al. (2009). Inhibition of ischemia-induced angiogenesis by benzo[a]pyrene in a manner dependent on the aryl hydrocarbon receptor. Biochem Biophys Res Commun, 381, 44–9
  • Ishida M, Mikami S, Kikuchi E, et al. (2010). Activation of the aryl hydrocarbon receptor pathway enhances cancer cell invasion by upregulating the MMP expression and is associated with poor prognosis in upper urinary tract urothelial cancer. Carcinogenesis, 31, 287–95
  • Ishimura R, Kawakami T, Ohsako S, Tohyama C. (2009). Dioxin-induced toxicity on vascular remodeling of the placenta. Biochem Pharmacol, 77, 660–9
  • JEFCA, Joint FAO/WHO Food Standards Programme. (2003). Position Paper On Dioxins And Dioxin-Like PCB. CX/FAC 04/36/32
  • Jefcoate CR, Liehr JG, Santen RJ, et al. (2000). Tissue-specific synthesis and oxidative metabolism of estrogens. J Natl Cancer Inst Monogr, 2000, 95–112
  • Jarabek AM, Pottenger LH, Andrews LS, et al. (2009). Creating context for the use of DNA adduct data in cancer risk assessment: I. Data organization. Crit Rev Toxicol, 39, 659–78
  • Jepsen P, Vilstrup H, Tarone RE, et al. (2007). Incidence rates of intra- and extrahepatic cholangiocarcinomas in Denmark from 1978 through 2002. J Natl Cancer Inst, 99, 895–7
  • Jones G, Greig JB. (1975). Pathological changes in the liver of mice given 2,3,7,8-tetrachlorodibenzo-p-dioxin. Experientia, 31, 1315–7
  • Judson RS, Kavlock RJ, Setzer RW, et al. (2011). Estimating toxicity-related biological pathway altering doses for high-throughput chemical risk assessment. Chem Res Toxicol, 24, 451–62
  • Julien E, Boobis AR, Olin SS. (2009). The key events dose-response framework: a cross-disciplinary mode-of-action based approach to examining dose-response and thresholds. Crit Rev Food Sci Nutr, 49, 682–9
  • Kawajiri K, Kobayashi Y, Ohtake F, et al. (2009). Aryl hydrocarbon receptor suppresses intestinal carcinogenesis in ApcMin/+ mice with natural ligands. Proc Natl Acad Sci USA, 106, 13481–6
  • Keates AC, Castagliuolo I, Cruickshank WW, et al. (2000). Interleukin 16 is up-regulated in Crohn’s disease and participates in TNBS colitis in mice. Gastroenterology, 119, 972–82
  • Kelley SK, Nilsson CB, Green MH, et al. (2000). Mobilization of vitamin A stores in rats after administration of 2,3, 7,8-tetrachlorodibenzo-p-dioxin: a kinetic analysis. Toxicol Sci, 55, 478–84
  • Kerkvliet NI. (2009). AHR-mediated immunomodulation: the role of altered gene transcription. Biochem Pharmacol, 77, 746–60
  • Kerkvliet NI, Steppan LB, Vorachek W, et al. (2009). Activation of aryl hydrocarbon receptor by TCDD prevents diabetes in nod mice and increases foxp3+ t cells in pancreatic lymph nodes. Immunotherapy, 1, 539–47
  • Kim AH, Kohn MC, Nyska A, Walker NJ. (2003). Area under the curve as a dose metric for promotional responses following 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure. Toxicol Appl Pharmacol, 191, 12–21
  • Kim DW, Gazourian L, Quadri SA, et al. (2000). The rela nf-kappab subunit and the aryl hydrocarbon receptor (AHR) cooperate to transactivate the c-myc promoter in mammary cells. Oncogene, 19, 5498–506
  • Kim JH, Stallcup MR. (2004). Role of the coiled-coil coactivator (cocoa) in aryl hydrocarbon receptor-mediated transcription. J Biol Chem, 279, 49842–8
  • Kim S, Dere E, Burgoon LD, et al. (2009). Comparative analysis of AHR-mediated TCDD-elicited gene expression in human liver adult stem cells. Toxicol Sci, 112, 229–44
  • Kinyamu HK, Chen J, Archer TK. (2005). Linking the ubiquitin-proteasome pathway to chromatin remodeling/modification by nuclear receptors. J Mol Endocrinol, 34, 281–97
  • Kitchin KT, Woods JS. (1979). 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) effects on hepatic microsomal cytochrome p-448-mediated enzyme activities. Toxicol Appl Pharmacol, 47, 537–46
  • Klimisch HJ, Andreae M, Tillmann U. (1997). A systematic approach for evaluating the quality of experimental toxicological and ecotoxicological data. Regul Toxicol Pharmacol, 25, 1–5
  • Kmieć Z. (2001). Cooperation of liver cells in health and disease. Adv Anat Embryol Cell Biol, 161III–XIII, 1–151
  • Knerr S, Schaefer J, Both S, et al. (2006). 2,3,7,8-tetrachlorodibenzo-p-dioxin induced cytochrome p450s alter the formation of reactive oxygen species in liver cells. Mol Nutr Food Res, 50, 378–84
  • Knerr S, Schrenk D. (2006). Carcinogenicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in experimental models. Mol Nutr Food Res, 50, 897–907
  • Knight TR, Choudhuri S, Klaassen CD. (2008). Induction of hepatic glutathione S-transferases in male mice by prototypes of various classes of microsomal enzyme inducers. Toxicol Sci, 106, 329–38
  • Kociba RJ, Keyes DG, Beyer JE, et al. (1978). Results of a two-year chronic toxicity and oncogenicity study of 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats. Toxicol Appl Pharmacol, 46, 279–303
  • Kogevinas M, Becher H, Benn T, et al. (1997). Cancer mortality in workers exposed to phenoxy herbicides, chlorophenols, and dioxins. An expanded and updated international cohort study. Am J Epidemiol, 145, 1061–75
  • Kohle C, Bock KW. (2006). Activation of coupled AH receptor and nrf2 gene batteries by dietary phytochemicals in relation to chemoprevention. Biochem Pharmacol, 72, 795–805
  • Kohle C, Schwarz M, Bock KW. (2008). Promotion of hepatocarcinogenesis in humans and animal models. Arch Toxicol, 82, 623–31
  • Kollara A, Brown TJ. (2006). Functional interaction of nuclear receptor coactivator 4 with aryl hydrocarbon receptor. Biochem Biophys Res Commun, 346, 526–34
  • Kolluri SK, Weiss C, Koff A, Gottlicher M. (1999). P27(kip1) induction and inhibition of proliferation by the intracellular ah receptor in developing thymus and hepatoma cells. Genes Dev, 13, 1742–53
  • Kuhlmann WD, Peschke P. (2006). Hepatic progenitor cells, stem cells, and AFP expression in models of liver injury. Int J Exp Pathol, 87, 343–59
  • Kumar MB, Ramadoss P, Reen RK, et al. (2001). The q-rich subdomain of the human ah receptor transactivation domain is required for dioxin-mediated transcriptional activity. J Biol Chem, 276, 42302–10
  • Kundu JK, Surh YJ. (2008). Inflammation: gearing the journey to cancer. Mutat Res, 659, 15–30
  • Lambert GH, Needham LL, Turner W, et al. (2006). Induced cyp1a2 activity as a phenotypic biomarker in humans highly exposed to certain PCBs/pcdfs. Environ Sci Technol, 40, 6176–80
  • Le Vee M, Jouan E, Fardel O. (2010). Involvement of aryl hydrocarbon receptor in basal and 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced expression of target genes in primary human hepatocytes. Toxicol In Vitro, 24, 1775–81
  • Lefkowitch JH. (2009). Recent developments in liver pathology. Hum Pathol, 40, 445–55
  • Liebermann DA, Hoffman B, Steinman RA. (1995). Molecular controls of growth arrest and apoptosis: P53-dependent and independent pathways. Oncogene, 11, 199–210
  • Lindros KO, Oinonen T, Johansson I, Ingelman-Sundberg M. (1997). Selective centrilobular expression of the aryl hydrocarbon receptor in rat liver. J Pharmacol Exp Ther, 280, 506–11
  • Lindros KO, Oinonen T, Kettunen E, et al. (1998). Aryl hydrocarbon receptor-associated genes in rat liver: regional coinduction of aldehyde dehydrogenase 3 and glutathione transferase Ya. Biochem Pharmacol, 55, 413–21
  • Loertscher JA, Sattler CA, Allen-Hoffmann BL. (2001). 2,3,7,8-Tetrachlorodibenzo-p-dioxin alters the differentiation pattern of human keratinocytes in organotypic culture. Toxicol Appl Pharmacol, 175, 121–9
  • Lu F, Zahid M, Saeed M, et al. (2007). Estrogen metabolism and formation of estrogen-DNA adducts in estradiol-treated MCF-10F cells. The effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin induction and catechol-O-methyltransferase inhibition. J Steroid Biochem Mol Biol, 105, 150–8
  • Lucier GW, Tritscher A, Goldsworthy T, et al. (1991). Ovarian hormones enhance 2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated increases in cell proliferation and preneoplastic foci in a two-stage model for rat hepatocarcinogenesis. Cancer Res, 51, 1391–7
  • Luebeck EG, Buchmann A, Stinchcombe S, et al. (2000). Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on initiation and promotion of GST-p-positive foci in rat liver: a quantitative analysis of experimental data using a stochastic model. Toxicol Appl Pharmacol, 167, 63–73
  • Lutz WK. (2001). Susceptibility differences in chemical carcinogenesis linearize the dose-response relationship: threshold doses can be defined only for individuals. Mutat Res, 482, 71–6
  • Lutz WK, Gaylor DW. (2008). Dose-response relationships for cancer incidence reflect susceptibility distributions. Chem Res Toxicol, 21, 971–2; author reply 972–3
  • Lutz WK, Lutz RW, Andersen ME. (2006). Dose-incidence relationships derived from superposition of distributions of individual susceptibility on mechanism-based dose responses for biological effects. Toxicol Sci, 90, 33–8
  • Ma Q, Lu AY. (2007). Cyp1a induction and human risk assessment: an evolving tale of in vitro and in vivo studies. Drug Metab Dispos, 35, 1009–16
  • Malik R, Selden C, Hodgson H. (2002). The role of non-parenchymal cells in liver growth. Semin Cell Dev Biol, 13, 425–31
  • Mannetje A, McLean D, Cheng S, et al. (2005). Mortality in New Zealand workers exposed to phenoxy herbicides and dioxins. Occup Environ Med, 62, 34–40
  • Mantovani A, Allavena P, Sica A, Balkwill F. (2008). Cancer-related inflammation. Nature, 454, 436–44
  • Marlowe JL, Puga A. (2005). Aryl hydrocarbon receptor, cell cycle regulation, toxicity, and tumorigenesis. J Cell Biochem, 96, 1174–84
  • Maronpot RR, Foley JF, Takahashi K, et al. (1993). Dose response for TCDD promotion of hepatocarcinogenesis in rats initiated with den: histologic, biochemical, and cell proliferation endpoints. Environ Health Perspect, 101, 634–42
  • Marshall NB, Kerkvliet NI. (2010). Dioxin and immune regulation: emerging role of aryl hydrocarbon receptor in the generation of regulatory t cells. Ann N Y Acad Sci, 1183, 25–37
  • Matsumura F. (2009). The significance of the nongenomic pathway in mediating inflammatory signaling of the dioxin-activated Ah receptor to cause toxic effects. Biochem Pharmacol, 77, 608–26
  • Matthews J, Gustafsson JA. (2006). Estrogen receptor and aryl hydrocarbon receptor signaling pathways. Nucl Recept Signal, 25, 4e016
  • Matthews J, Wihlen B, Thomsen J, Gustafsson JA. (2005). Aryl hydrocarbon receptor-mediated transcription: ligand-dependent recruitment of estrogen receptor alpha to 2,3,7,8-tetrachlorodibenzo-p-dioxin-responsive promoters. Mol Cell Biol, 25, 5317–28
  • McBride DI, Collins JJ, Humphry NF, et al. (2009). Mortality in workers exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin at a trichlorophenol plant in new zealand. J Occup Environ Med, 51, 1049–56
  • McGrath LF, Cooper KR, Georgopoulos P, Gallo MA. (1995). Alternative models for low dose-response analysis of biochemical and immunological endpoints for tetrachlorodibenzo-p-dioxin. Regul Toxicol Pharmacol, 21, 382–96
  • McMartin DN, Sahota PS, Gunson DE, et al. (1992). Neoplasms and related proliferative lesions in control Sprague-DawleyDawley rats from carcinogenicity studies. Historical data and diagnostic considerations. Toxicol Pathol, 20, 212–25
  • Meek ME. (2008). Recent developments in frameworks to consider human relevance of hypothesized modes of action for tumours in animals. Environ Mol Mutagen, 49, 110–16
  • Meek ME, Bucher JR, Cohen SM, et al. (2003). A framework for human relevance analysis of information on carcinogenic modes of action. Crit Rev Toxicol, 33, 591–653
  • Mills JJ, Andersen ME. (1993). Dioxin hepatic carcinogenesis: biologically motivated modeling and risk assessment. Toxicol Lett, 68, 177–89
  • Milstone LM, LaVigne JF. (1984). 2,3,7,8-tetrachlorodibenzo-p-dioxin induces hyperplasia in confluent cultures of human keratinocytes. J Invest Dermatol, 82, 532–4
  • Mitchell KA, Elferink CJ. (2009). Timing is everything: consequences of transient and sustained AHR activity. Biochem Pharmacol, 77, 947–56
  • Mitchell KA, Lockhart CA, Huang G, Elferink CJ. (2006). Sustained aryl hydrocarbon receptor activity attenuates liver regeneration. Mol Pharmacol, 70, 163–70
  • Mizukami S, Ichimura R, Kemmochi S, et al. (2010). Induction of gst-p-positive proliferative lesions facilitating lipid peroxidation with possible involvement of transferrin receptor up-regulation and ceruloplasmin down-regulation from the early stage of liver tumor promotion in rats. Arch Toxicol, 84, 319–31
  • Mocarelli P, Marocchi A, Brambilla P, et al. (1986). Clinical laboratory manifestations of exposure to dioxin in children. A six-year study of the effects of an environmental disaster near seveso, italy. JAMA, 256, 2687–95
  • Mocarelli P, Needham LL, Marocchi A, et al. (1991). Serum concentrations of 2,3,7,8-tetrachlorodibenzo-p-dioxin and test results from selected residents of seveso, italy. J Toxicol Environ Health, 32, 357–66
  • Moennikes O, Loeppen S, Buchmann A, et al. (2004). A constitutively active dioxin/aryl hydrocarbon receptor promotes hepatocarcinogenesis in mice. Cancer Res, 64, 4707–10
  • Moolgavkar SH, Luebeck EG, Buchmann A, Bock KW. (1996). Quantitative analysis of enzyme-altered liver foci in rats initiated with diethylnitrosamine and promoted with 2,3,7,8-tetrachlorodibenzo-p-dioxin or 1,2,3,4,6,7,8-heptachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol, 138, 31–42
  • Moriguchi T, Motohashi H, Hosoya T, et al. (2003). Distinct response to dioxin in an arylhydrocarbon receptor (AHR)-humanized mouse. Proc Natl Acad Sci USA, 100, 5652–7
  • Mroue RM, El-Sabban ME, Talhouk RS. (2011). Connexins and the gap in context. Integr Biol (Camb), 3, 255–66
  • Mukai M, Tischkau SA. (2007). Effects of tryptophan photoproducts in the circadian timing system: searching for a physiological role for aryl hydrocarbon receptor. Toxicol Sci, 95, 172–81
  • Mukai R, Shirai Y, Saito N, et al. (2010). Suppression mechanisms of flavonoids on aryl hydrocarbon receptor-mediated signal transduction. Arch Biochem Biophys, 501, 134–41
  • Mulholland DJ, Dedhar S, Coetzee GA, Nelson CC. (2005). Interaction of nuclear receptors with the Wnt/beta-catenin/Tcf signaling axis: What you like to know? Endocr Rev, 26, 898–915
  • Munzel P, Bock-Hennig B, Schieback S, et al. (1996). Growth modulation of hepatocytes and rat liver epithelial cells (wb-f344) by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Carcinogenesis, 17, 197–202
  • Murray IA, Morales JL, Flaveny CA, et al. (2010). Evidence for ligand-mediated selective modulation of aryl hydrocarbon receptor activity. Mol Pharmacol, 77, 247–54
  • Murray IA, Reen RK, Leathery N, et al. (2005). Evidence that ligand binding is a key determinant of ah receptor-mediated transcriptional activity. Arch Biochem Biophys, 442, 59–71
  • Murrell JA, Portier CJ, Morris RW. (1998). Characterizing dose-response: I: critical assessment of the benchmark dose concept. Risk Anal, 18, 13–26
  • Nakata A, Urano D, Fujii-Kuriyama Y, et al. (2009). G-protein signalling negatively regulates the stability of aryl hydrocarbon receptor. EMBO Rep, 10, 622–8
  • NAS (National Academies of Science). (2007). Toxicity testing in the 21st century: a vision and a strategy. The National Academies Press, National Research Council, ISBN-10: 0-309-15173-2. Available from: http://www.nap.edu/catalog.php?record_id=11970 [last accessed 9 May 2013]
  • Nawaz Z, O’Malley BW. (2004). Urban renewal in the nucleus: is protein turnover by proteasomes absolutely required for nuclear receptor-regulated transcription? Mol Endocrinol, 18, 493–9
  • Nebert DW, Puga A, Vasiliou V. (1993). Role of the ah receptor and the dioxin-inducible [AH] gene battery in toxicity, cancer, and signal transduction. Ann N Y Acad Sci, 685, 624–40
  • Neuman MG. (2001). Apoptosis in diseases of the liver. Crit Rev Clin Lab Sci, 38, 109–66
  • Newsholme SJ, Fish CJ. (1994). Morphology and incidence of hepatic foci of cellular alteration in Sprague-DawleyDawley rats. Toxicol Pathol, 22, 524–7
  • Nguyen LK, Kulasiri D. (2009). On the functional diversity of dynamical behaviour in genetic and metabolic feedback systems. BMC Syst Biol, 3, 51
  • Nguyen NT, Kimura A, Nakahama T, et al. (2010). Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc Natl Acad Sci USA, 107, 19961–6
  • Niittynen M, Simanainen U, Syrjala P, et al. (2007). Differences in acute toxicity syndromes of 2,3,7,8-tetrachlorodibenzo-p-dioxin and 1,2,3,4,7,8-hexachlorodibenzo-p-dioxin in rats. Toxicol, 235, 39–51
  • Nishimura N, Miyabara Y, Suzuki JS, et al. (2001). Induction of metallothionein in the livers of female Sprague-Dawley rats treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Life Sci, 69, 1291–303
  • Nishiumi S, Yoshida K, Ashida H. (2007). Curcumin suppresses the transformation of an aryl hydrocarbon receptor through its phosphorylation. Arch Biochem Biophys, 466, 267–73
  • NTP. (1980). Bioassay of a mixture of 1,2,3,6,7,8-hexachlorodibenzo-p-dioxin and 1,2,3,7,8,9-hexachlorodibenzo-p-dioxin (gavage) for possible carcinogenicity (CAS no. 57653-85-7, CAS no. 19408-74-3). Natl Toxicol Program Tech Rep Ser, 198, 1–187
  • NTP. (1982). Carcinogenesis bioassay of 2,3,7,8-tetrachlorodibenzo-p-dioxin (CAS no. 1746-01-6) in osborne-mendel rats and b6c3f1 mice (gavage study). Natl Toxicol Program Tech Rep Ser, 209, 1–195
  • NTP. (2006a). Ntp toxicology and carcinogenesis studies of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (CAS no. 1746-01-6) in female harlan Sprague-Dawley rats (gavage studies). Natl Toxicol Program Tech Rep Ser, 1, Jan, 4-246
  • NTP. (2006b). Ntp toxicology and carcinogenesis studies of 2,3,4,7,8-pentachlorodibenzofuran (pecdf) (CAS no. 57117-31-4) in female harlanSprague-Dawley rats (gavage studies). Natl Toxicol Program Tech Rep Ser, 525, 1–198
  • NTP. (2006c). Ntp toxicology and carcinogenesis studies of 3,3′,4,4′,5-pentachlorobiphenyl (PCB 126) (CAS no. 57465-28-8) in female harlan Sprague-Dawley rats (gavage studies). Natl Toxicol Program Tech Rep Ser, 520, 4–256
  • NTP. (2006d). Ntp toxicology and carcinogenesis studies of a mixture of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (CAS no. 1746-01-6), 2,3,4,7,8-pentachlorodibenzofuran (pecdf) (CAS no. 57117-31-4), and 3,3′,4,4′,5-pentachlorobiphenyl (PCB 126) (CAS no. 57465-28-8) in female harlanSprague-Dawley rats (gavage studies). Natl Toxicol Program Tech Rep Ser, 526, 1–180
  • NTP. (2010). Toxicology and carcinogenesis studies of 2,3′,4,4′,5-pentachlorobiphenyl (PCB 118) (CAS no. 31508-00-6) in female harlan Sprague-DawleyDawley rats (gavage studies). Natl Toxicol Program Tech Rep Ser, 559, 1–174
  • Oesch-Bartlomowicz B, Huelster A, Wiss O, et al. (2005). Aryl hydrocarbon receptor activation by camp vs. Dioxin: divergent signaling pathways. Proc Natl Acad Sci USA, 102, 9218–23
  • Oesch-Bartlomowicz B, Oesch F. (2009). Role of camp in mediating AHR signaling. Biochem Pharmacol, 77, 627–41
  • Oh SHH, Petersen BE. (2003). Hepatic oval ‘stem’ cell in liver regeneration. Semin Cell Dev Biol, 14, 405–9
  • Oinonen T, Lindros KO. (1998). Zonation of hepatic cytochrome P-450 expression and regulation. Biochem J, 329, 17–35
  • Onozuka D, Yoshimura T, Kaneko S, Furue M. (2009). Mortality after exposure to polychlorinated biphenyls and polychlorinated dibenzofurans: a 40-year follow-up study of yusho patients. Am J Epidemiol, 169, 86–95
  • Opitz CA, Litzenburger UM, Sahm F, et al. (2011). An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature, 478, 197–203
  • Ott MG, Zober A. (1996). Cause specific mortality and cancer incidence among employees exposed to 2,3,7,8-TCDD after a 1953 reactor accident. Occup Environ Med, 53, 606–12
  • Ovando BJ, Vezina CM, McGarrigle BP, Olson JR. (2006). Hepatic gene downregulation following acute and subchronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Sci, 94, 428–38
  • Paajarvi G, Viluksela M, Pohjanvirta R, et al. (2005). Tcdd activates mdm2 and attenuates the p53 response to DNA damaging agents. Carcinogenesis, 26, 201–8
  • Paku S, Schnur J, Nagy P, Thorgeirsson SS. (2001). Origin and structural evolution of the early proliferating oval cells in rat liver. Am J Pathol, 158, 1313–23
  • Partch CL, Card PB, Amezcua CA, Gardner KH. (2009). Molecular basis of coiled coil coactivator recruitment by the aryl hydrocarbon receptor nuclear translocator (ARNT). J Biol Chem, 284, 15184–92
  • Patel RD, Kim DJ, Peters JM, Perdew GH. (2006). The aryl hydrocarbon receptor directly regulates expression of the potent mitogen epiregulin. Toxicol Sci, 89, 75–82
  • Pesatori AC, Consonni D, Rubagotti M, et al. (2009). Cancer incidence in the population exposed to dioxin after the “seveso accident”: twenty years of follow-up. Environ Health, 8, 1--11
  • Petersen BE. (2001). Hepatic “stem” cells: coming full circle. Blood Cells Mol Dis, 27, 590–600
  • Peterson S, Schwarz Y, Li SS, et al. (2009). CYP1A2, GSTM1, and GSTT1 polymorphisms and diet effects on CYP1A2 activity in a crossover feeding trial. Cancer Epidemiol Biomarkers Prev, 18, 3118–25
  • Pintilie DG, Shupe TD, Oh SH, et al. (2010). Hepatic stellate cells’ involvement in progenitor-mediated liver regeneration. Lab Invest, 90, 1199–208
  • Pitot HC, Goldsworthy TL, Moran S, et al. (1987). A method to quantitate the relative initiating and promoting potencies of hepatocarcinogenic agents in their dose-response relationships to altered hepatic foci 11. Carcinogenesis, 8, 1491–9
  • Pocchiari F, Silano V, Zampieri A. (1979). Human health effects from accidental release of tetrachlorodibenzo-p-dioxin (TCDD) at seveso, italy. Ann N Y Acad Sci, 320, 311–20
  • Pohjanvirta R, Viluksela M, Tuomisto JT, et al. (1999). Physicochemical differences in the AH receptors of the most TCDD-susceptible and the most TCDD-resistant rat strains. Toxicol Appl Pharmacol, 155, 82–95
  • Poland A, Glover E. (1979). An estimate of the maximum in vivo covalent binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin to rat liver protein, ribosomal RNA, and DNA. Cancer Res, 39, 3341–4
  • Poland A, Palen D, Glover E. (1994). Analysis of the four alleles of the murine aryl hydrocarbon receptor. Mol Pharmacol, 46, 915–21
  • Pollenz RS. (2002). The mechanism of ah receptor protein down-regulation (degradation) and its impact on ah receptor-mediated gene regulation. Chem Biol Interact, 141, 41–61
  • Pollenz RS, Barbour ER. (2000). Analysis of the complex relationship between nuclear export and aryl hydrocarbon receptor-mediated gene regulation. Mol Cell Biol, 20, 6095–104
  • Popp JA, Scortichini BH, Garvey LK. (1985). Quantitative evaluation of hepatic foci of cellular alteration occurring spontaneously in fischer-344 rats. Fundam Appl Toxicol, 5, 314–9
  • Portier C, Tritscher A, Kohn M, et al. (1993). Ligand/receptor binding for 2,3,7,8-TCDD: implications for risk assessment. Fundam Appl Toxicol, 20, 48–56
  • Portier CJ, Sherman CD, Kohn M, et al. (1996). Modeling the number and size of hepatic focal lesions following exposure to 2,3,7,8-TCDD. Toxicol Appl Pharmacol, 138, 20–30
  • Puga A, Barnes SJ, Dalton TP, et al. (2000). Aromatic hydrocarbon receptor interaction with the retinoblastoma protein potentiates repression of e2f-dependent transcription and cell cycle arrest. J Biol Chem, 275, 2943–50
  • Puga A, Ma C, Marlowe JL. (2009). The aryl hydrocarbon receptor cross-talks with multiple signal transduction pathways. Biochem Pharmacol, 77, 713–22
  • Puga A, Marlowe J, Barnes S, et al. (2002). Role of the aryl hydrocarbon receptor in cell cycle regulation. Toxicology, 181–2, 171–7
  • Qu Z, Vondriska TM. (2009). The effects of cascade length, kinetics and feedback loops on biological signal transduction dynamics in a simplified cascade model. Phys Biol, 6, 1--21
  • Randerath K, Putman KL, Randerath E, et al. (1990). Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on i-compounds in hepatic DNA of Sprague-Dawley rats: sex-specific effects and structure-activity relationships. Toxicol Appl Pharmacol, 103, 271–80
  • Rao MS, Subbarao V, Prasad JD, Scarpelli DG. (1988). Carcinogenicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in the Syrian golden hamster. Carcinogenesis, 9, 1677–9
  • Reggiani G. (1980). Acute human exposure to TCDD in seveso, italy. J Toxicol Environ Health, 6, 27–43
  • Rela L, Szczupak L. (2004). Gap junctions: their importance for the dynamics of neural circuits. Mol Neurobiol, 30, 341–57
  • Reichard JF, Dalton TP, Shertzer HG, Puga A. (2005). Induction of oxidative stress responses by dioxin and other ligands of the aryl hydrocarbon receptor. Dose Response, 3, 306–31
  • Rhomberg LR, Goodman JE, Haber LT, et al. (2011). Linear low-dose extrapolation for noncancer heath effects is the exception, not the rule. Crit Rev Toxicol, 41, 1–19
  • Rifkind AB. (2006). CYP1A in TCDD toxicity and in physiology-with particular reference to CYP dependent arachidonic acid metabolism and other endogenous substrates. Drug Metab Rev, 38, 291–355
  • Roberts BJ, Whitelaw ML. (1999). Degradation of the basic helix-loop-helix/per-arnt-sim homology domain dioxin receptor via the ubiquitin/proteasome pathway. J Biol Chem, 274, 36351–6
  • Roberts RA, Ganey PE, Ju C, et al. (2007). Role of the Kupffer cell in mediating hepatic toxicity and carcinogenesis. Toxicol Sci, 96, 2–15
  • Roberts RA, Nebert DW, Hickman JA, et al. (1997). Perturbation of the mitosis/apoptosis balance: a fundamental mechanism in toxicology. Fundam Appl Toxicol, 38, 107–15
  • Roman AC, Carvajal-Gonzalez JM, Rico-Leo EM, Fernandez-Salquero PM. (2009). Dioxin receptor deficiency impairs angiogenesis by a mechanism involving VEGF-A depletion in the endothelium and transforming growth factor-beta overexpression in the stroma. J Biol Chem, 284, 25135–48
  • Roskams TA, Theise ND, Balabaud C, et al. (2004). Nomenclature of the finer branches of the biliary tree: canals, ductules, and ductular reactions in human livers. Hepatology, 39, 1739–45
  • Rotroff DM, Wetmore BA, Dix DJ, et al. (2010). Incorporating human dosimetry and exposure into high-throughput in vitro toxicity screening. Toxicol Sci, 117, 348–58
  • Roy D, Cai Q, Felty Q, Narayan S. (2007). Estrogen-induced generation of reactive oxygen and nitrogen species, gene damage, and estrogen-dependent cancers. J Toxicol Environ Health B Crit Rev, 10, 235–57
  • Sahin MB, Schwartz RE, Buckley SM, et al. (2008). Isolation and characterization of a novel population of progenitor cells from unmanipulated rat liver. Liver Transpl, 14, 333–45
  • Sand S, Fletcher N, von Rosen D, et al. (2010). Quantitative and statistical analysis of differences in sensitivity between long-evans and han/wistar rats following long-term exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Regul Toxicol Pharmacol, 57, 136–45
  • Santostefano MJ, Richardson VM, Walker NJ, et al. (1999). Dose-dependent localization of TCDD in isolated centrilobular and periportal hepatocytes. Toxicol Sci, 52, 9–19
  • Sarafoff M, Rabes HM, Dormer P. (1986). Correlations between ploidy and initiation probability determined by DNA cytophotometry in individual altered hepatic foci. Carcinogenesis, 7, 1191–6
  • Sargent L, Xu YH, Sattler GL, et al. (1989). Ploidy and karyotype of hepatocytes isolated from enzyme-altered foci in two different protocols of multistage hepatocarcinogenesis in the rat. Carcinogenesis, 10, 387–91
  • Sarkar P, Shiizaki K, Yonemoto J, Sone H. (2006). Activation of telomerase in BeWo cells by estrogen and 2,3,7,8-tetrachlorodibenzo-p-dioxin in co-operation with c-Myc. Int J Oncol, 28, 43–51
  • Sartor MA, Schnekenburger M, Marlowe JL, et al. (2009). Genomewide analysis of aryl hydrocarbon receptor binding targets reveals an extensive array of gene clusters that control morphogenetic and developmental programs. Environ Health Perspect, 117, 1139–46
  • Schecter A, Ryan JJ, Päpke O. (1998). Decrease in levels and body burdens od dioxins, dibenzofurans, PCBS, DDE, HCB in blood and milk in a mother nursing twins over a thirty-eight month period. Chemosphere, 37, 1807–16
  • Schmidt CK, Hoegberg P, Fletcher N, et al. (2003). 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) alters the endogenous metabolism of all-trans-retinoic acid in the rat. Arch Toxicol, 77, 371–83
  • Schrenk D, Buchmann A, Dietz K, et al. (1994). Promotion of preneoplastic foci in rat liver with 2,3,7,8-tetrachlorodibenzo-p-dioxin, 1,2,3,4,6,7,8-heptachlorodibenzo-p-dioxin and a defined mixture of 49 polychlorinated dibenzo-p-dioxins. Carcinogenesis, 15, 509–15
  • Schrenk D, Lipp HP, Wiesmuller T, et al. (1991). Assessment of biological activities of mixtures of polychlorinated dibenzo-p-dioxins: comparison between defined mixtures and their constituents. Arch Toxicol, 65, 114–8
  • Schrenk D, Schmitz HJ, Bohnenberger S, et al. (2004). Tumor promoters as inhibitors of apoptosis in rat hepatocytes. Toxicol Lett, 149, 43–50
  • Schrenk D, Stuven T, Gohl G, et al. (1995). Induction of cyp1a and glutathione S-transferase activities by 2,3,7,8-tetrachlorodibenzo-p-dioxin in human hepatocyte cultures. Carcinogenesis, 16, 943–6
  • Schroeder JC, Dinatale BC, Murray IA, et al. (2010). The uremic toxin 3-indoxyl sulfate is a potent endogenous agonist for the human aryl hydrocarbon receptor. Biochemistry, 49, 393–400
  • Schulz VJ, Smit JJ, Willemsen KJ, et al. (2011). Activation of the aryl hydrocarbon receptor suppresses sensitization in a mouse peanut allergy model. Toxicol Sci, 123, 491–500
  • Schwarz M, Buchmann A, Stinchcombe S, et al. (2000). Ah receptor ligands and tumor promotion: survival of neoplastic cells. Toxicol Lett, 112–3, 69–77
  • Scott RJ, Chakraborty S, Sell S, et al. (1989). Change in the ploidy state of rat liver cells during chemical hepatocarcinogenesis and its relationship to the increased expression of alpha-fetoprotein. Cancer Res, 49, 6085–90
  • Seed J, Carney EW, Corley RA, et al. (2005). Overview: using mode of action and life stage information to evaluate the human relevance of animal toxicity data. Crit Rev Toxicol, 35, 664–72
  • Seefeld MD, Keesey RE, Peterson RE. (1984). Body weight regulation in rats treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol, 76, 526–36
  • Seglen PO. (1997). DNA ploidy and autophagic protein degradation as determinants of hepatocellular growth and survival. Cell Biol Toxicol, 13, 301–15
  • Sell S, Dunsford HA. (1989). Evidence for the stem cell origin of hepatocellular carcinoma and cholangiocarcinoma. Am J Pathol, 134, 1347–63
  • Sell S, Leffert HL. (2008). Liver cancer stem cells. J Clin Oncol, 26, 2800–5
  • Sempoux C, Jibara G, Ward SC, et al. (2011). Intrahepatic cholangiocarcinoma: new insights in pathology. Semin Liver Dis, 31, 49–60
  • Senft AP, Dalton TP, Nebert DW, et al. (2002). Dioxin increases reactive oxygen production in mouse liver mitochondria. Toxicol Appl Pharmacol, 178, 15–21
  • Senoo H, Yoshikawa K, Morii M, et al. (2010). Hepatic stellate cell (vitaminA-storing cell) and its relative – past, present and future. Cell Biol Int, 34, 1247–72
  • Shah I, Houck K, Judson RS, et al. (2011). Using nuclear receptor activity to stratify hepatocarcinogens. PLOS One, 6, 1–11
  • Shao K, Small MJ. (2011). Potential uncertainty reduction in model-averaged benchmark dose estimates informed by an additional dose study. J Risk Anal, 31, 1561-1575
  • Sheikh-Bahaei S, Maher JJ, Anthony Hunt C. (2010). Computational experiments reveal plausible mechanisms for changing patterns of hepatic zonation of xenobiotic clearance and hepatoxicity. J Theor Biol, 265, 718–33
  • Shen D, Dalton TP, Nebert DW, Shertzer HG. (2005). Glutathione redox state regulates mitochondrial reactive oxygen production. J Biol Chem, 280, 25305–12
  • Shertzer HG, Genter MB, Shen D, et al. (2006). Tcdd decreases atp levels and increases reactive oxygen production through changes in mitochondrial ff-atp synthase and ubiquinone. Toxicol Appl Pharmacol, 217, 363–74
  • Shimamoto K, Dewa Y, Ishii Y, et al. (2011). Indole-3-carbinol enhances oxidative stress responses resulting in the induction of preneoplastic liver cell lesions in partially hepatectomized rats initiated with diethylnitrosamine. Toxicology, 283, 109–17
  • Shin EK, Kim DH, Lim H, et al. (2010). The anti-inflammatory effects of a methanolic extract from Radix Isatidis in murine macrophages and mice. Inflammation, 33, 110–8
  • Shmarakov I, Fleshman MK, D’Ambrosio DN, et al. (2010). Hepatic stellate cells are an important cellular site for beta-carotene conversion to retinoid. Arch Biochem Biophys, 504, 3–10
  • Silkworth JB, Carlson EA, McCulloch C, et al. (2008). Toxicogenomic analysis of gender, chemical, and dose effects in livers of TCDD- or aroclor 1254-exposed rats using a multifactor linear model. Toxicol Sci, 102, 291–309
  • Silkworth JB, Koganti A, Illouz K, et al. (2005). Comparison of TCDD, PCB CYP1A induction sensitivities in fresh hepatocytes from human donors, Sprague-Dawley rats, and rhesus monkeys and HepG2 cells. Toxicol Sci, 87, 508–19
  • Simon T. (2010). Just who is at risk? The ethics of environmental regulation. Hum Exp Toxicol, 30, 795--819
  • Simon T, Aylward LL, Kirman CR, et al. (2009). Estimates of cancer potency of 2,3,7,8-tetrachlorodibenzo(p)dioxin using linear and nonlinear dose-response modeling and toxicokinetics. Toxicol Sci, 112, 490–506
  • Simones T, Shepherd DM. (2011). Consequences of AHR activation in steady-state dendritic cells. Toxicol Sci, 119, 293–307
  • Singh KP, Casado FL, Opanashuk LA, Gasiewicz TA. (2009). The aryl hydrocarbon receptor has a normal function in the regulation of hematopoietic and other stem/progenitor cell populations. Biochem Pharmacol, 77, 577–87
  • Song J, Clagett-Dame M, Peterson RE, et al. (2002). A ligand for the aryl hydrocarbon receptor isolated from lung. Proc Natl Acad Sci USA, 99, 14694–9
  • Sonich-Mullin C, Fielder R, Wiltse J, et al. (2001). Ipcs conceptual framework for evaluating a mode of action for chemical carcinogenesis. Regul Toxicol Pharmacol, 34, 146–52
  • Steenland K, Piacitelli L, Deddens J, et al. (1999). Cancer, heart disease, and diabetes in workers exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. J Natl Cancer Inst, 91, 779–86
  • Stinchcombe S, Buchmann A, Bock KW, Schwarz M. (1995). Inhibition of apoptosis during 2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated tumour promotion in rat liver. Carcinogenesis, 16, 1271–5
  • Sutter CH, Yin H, Li Y, et al. (2009). EGF receptor signaling blocks aryl hydrocarbon receptor-mediated transcription and cell differentiation in human epidermal keratinocytes. Proc Natl Acad Sci USA, 106, 4266–71
  • Suzuki T, Nohara K. (2007). Regulatory factors involved in species-specific modulation of arylhydrocarbon receptor (AHR)-dependent gene expression in humans and mice. J Biochem, 142, 443–52
  • Swenberg JA, Fryar-Tita E, Jeong YC, et al. (2008). Biomarkers in toxicology and risk assessment: informing critical dose-response relationships. Chem Res Toxicol, 21, 253–65
  • Swenberg JA, Lu K, Moeller BC, et al. (2011). Endogenous versus exogenous DNA adducts: their role in carcinogenesis, epidemiology, and risk assessment. Toxicol Sci, 120, S130–45
  • Takeuchi A, Takeuchi M, Oikawa K, et al. (2009). Effects of dioxin on vascular endothelial growth factor (VEGF) production in the retina associated with choroidal neovascularization. Invest Ophthalmol Vis Sci, 50, 3410–6
  • Tamano S, Merlino GT, Ward JM. (1994). Rapid development of hepatic tumors in transforming growth factor alpha transgenic mice associated with increased cell proliferation in precancerous hepatocellular lesions initiated by n-nitrosodiethylamine and promoted by phenobarbital. Carcinogenesis, 15, 1791–8
  • Tanaka J, Sugimoto K, Shiraki K, et al. (2010). Functional cell surface expression of toll-like receptor 9 promotes cell proliferation and survival in human hepatocellular carcinomas. Int J Oncol, 37, 805–14
  • Tang NJ, Liu J, Coenraads PJ, et al. (2008). Expression of AhR, CYP1A1, GSTA1, c-fos and TGF-alpha in skin lesions from dioxin-exposed humans with chloracne. Toxicol Lett, 177, 182--7
  • Tappenden DM, Lynn SG, Crawford RB, et al. (2011). The aryl hydrocarbon receptor interacts with ATP5α1, a subunit of the ATP synthase complex, and modulates mitochondrial function. Toxicol Appl Pharmacol, 254, 299–310
  • Taylor RT, Wang F, Hsu EL, Hankinson O. (2009). Roles of coactivator proteins in dioxin induction of cyp1a1 and cyp1b1 in human breast cancer cells. Toxicol Sci, 107, 1–8
  • Teeguarden JG, Dragan YP, Singh J, et al. (1999). Quantitative analysis of dose- and time-dependent promotion of four phenotypes of altered hepatic foci by 2,3,7,8-tetrachlorodibenzo-p-dioxin in female Sprague-Dawley rats. Toxicol Sci, 2, 211–23
  • TERA. (2010). Dose-response approaches for nuclear receptor-mediated modes of action. Available from: http://www.tera.org/peer/nuclearreceptor/ [last accessed 9 Jun 2013]
  • Theise ND, Saxena R, Portmann BC, et al. (1999). The canals of hering and hepatic stem cells in humans. Hepatology, 30, 1425–33
  • Thomas RS, Clewell HJ 3rd, Allen BC, et al. (2011). Application of transcriptional benchmark dose values in quantitative cancer and noncancer risk assessment. Toxicol Sci, 120, 194–205
  • Tian Y. (2009). Ah receptor and nf-kappab interplay on the stage of epigenome. Biochem Pharmacol, 77, 670–80
  • Tian Y, Ke S, Denison MS, et al. (1999). Ah receptor and nf-kappab interactions, a potential mechanism for dioxin toxicity. J Biol Chem, 274, 510–5
  • Tijet N, Boutros PC, Moffat ID, et al. (2006). Aryl hydrocarbon receptor regulates distinct dioxin-dependent and dioxin-independent gene batteries. Mol Pharmacol, 69, 140–53
  • Toyoshiba H, Walker NJ, Bailer AJ, Portier CJ. (2004). Evaluation of toxic equivalency factors for induction of cytochromes p450 cyp1a1 and cyp1a2 enzyme activity by dioxin-like compounds. Toxicol Appl Pharmacol, 194, 156–68
  • Tritscher AM, Clark GC, Sewall C, et al. (1995). Persistence of TCDD-induced hepatic cell proliferation and growth of enzyme altered foci after chronic exposure followed by cessation of treatment in den initiated female rats. Carcinogenesis, 16, 2807–11
  • Tritscher AM, Goldstein JA, Portier CJ, et al. (1992). Dose-response relationships for chronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin in a rat tumor promotion model: quantification and immunolocalization of cyp1a1 and cyp1a2 in the liver. Cancer Res, 52, 3436–42
  • Tsai SP, Ahmed FS, Wendt JK, et al. (2007). A 56-year mortality follow-up of texas petroleum refinery and chemical employees, 1948-2003. J Occup Environ Med, 49, 557–67
  • Tsao MS, Smith JD, Nelson KG, Grisham JW. (1984). A diploid epithelial cell line from normal adult rat liver with phenotypic properties of ‘oval’ cells. Exp Cell Res, 154, 38–52
  • Tsuji S, Ogawa K, Takasaka H, et al. (1988). Clonal origin of gamma-glutamyl transpeptidase-positive hepatic lesions induced by initiation-promotion in ornithine carbamoyltransferase mosaic mice. Jpn J Cancer Res, 79, 148–51
  • Turgeon JL, McDonnell DP, Martin KA, Wise PM. (2004). Hormone therapy: physiological complexity belies therapeutic simplicity. Science, 304, 1269–73
  • Turner R, Lozoya O, Wang Y, et al. (2011). Human hepatic stem cell and maturational liver lineage biology. Hepatology, 53, 1035–45
  • Turteltaub KW, Felton JS, Gledhill BL, et al. (1990). Accelerator mass spectrometry in biomedical dosimetry: relationship between low-level exposure and covalent binding of heterocyclic amine carcinogens to DNA. Proc Natl Acad Sci USA, 87, 5288–92
  • Umannová L, Zatloukalová J, Machala M, et al. (2007). Tumor necrosis factor-alpha modulates effects of aryl hydrocarbon receptor ligands on cell proliferation and expression of cytochrome P450 enzymes in rat liver “stem-like” cells. Toxicol Sci, 99, 79–89
  • Uno S, Endo K, Ishida Y, et al. (2009). Cyp1a1 and cyp1a2 expression: comparing ‘humanized’ mouse lines and wild-type mice; comparing human and mouse hepatoma-derived cell lines. Toxicol Appl Pharmacol, 237, 119–26
  • USEPA. (2001). Toxicological review of chloroform (CAS no. 67-66-3). United States Environmental Protection Agency. Available from: http://www.epa.gov/iris/toxreviews/0025tr.pdf [last accessed 9 May 2013]
  • USEPA. (2003). Exposure and human health reassessment of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds
  • USEPA. (2005). Guidelines for carcinogen risk assessment. EPA/630/P-03/001B March 2005
  • USEPA. (2010) EPA’s Reanalysis of Key Issues Related to Dioxin Toxicity and Response to NAS Comments (External Review Draft). U.S. EPA/600/R-10/038A, 2010
  • Van den Berg M, Birnbaum LS, Denison M, et al. (2006). The 2005 world health organization reevaluation of human and mammalian toxic equivalency factors for dioxins and dioxin-like compounds. Toxicol Sci, 93, 223–41
  • Vanden-Heuvel JP, Clark GC, Kohn MC, et al. (1994). Dioxin-responsive genes: examination of dose-response relationships using quantitative reverse transcriptase-polymerase chain reaction. Cancer Res, 54, 62–8
  • Vickers AE, Lucier GW. (1996). Estrogen receptor levels and occupancy in hepatic sinusoidal endothelial and kupffer cells are enhanced by initiation with diethylnitrosamine and promotion with 17alpha-ethinylestradiol in rats. Carcinogenesis, 17, 1235–42
  • Vickers AE, Nelson K, McCoy Z, Lucier GW. (1989). Changes in estrogen receptor, DNA ploidy, and estrogen metabolism in rat hepatocytes during a two-stage model for hepatocarcinogenesis using 17 alpha-ethinylestradiol as the promoting agent. Cancer Res, 49, 6512–20
  • Villano CM, Murphy KA, Akintobi A, White LA. (2006). 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces matrix metalloproteinase (MMP) expression and invasion in A2058 melanoma cells. Toxicol Appl Pharmacol, 210, 212–24
  • Viluksela M, Bager Y, Tuomisto JT, et al. (2000). Liver tumor-promoting activity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in TCDD-sensitive and TCDD-resistant rat strains. Cancer Res, 60, 6911–20
  • Viluksela M, Stahl BU, Birnbaum LS, Rozman KK. (1997). Subchronic/chronic toxicity of 1,2,3,4,6,7,8-heptachlorodibenzo-p-dioxin (hpcdd) in rats. Part ii. Biochemical effects. Toxicol Appl Pharmacol, 146, 217–26
  • Viluksela M, Stahl BU, Birnbaum LS, Rozman KK. (1998). Subchronic/chronic toxicity of a mixture of four chlorinated dibenzo-p-dioxins in rats. Ii. Biochemical effects. Toxicol Appl Pharmacol, 151, 70–8
  • Waern F, Flodstrom S, Busk L, et al. (1991). Relative liver tumour promoting activity and toxicity of some polychlorinated dibenzo-p-dioxin- and dibenzofuran-congeners in female Sprague-Dawley rats. Pharmacol Toxicol, 69, 450–8
  • Wahba ZZ, Lawson TW, Murray WJ, Stohs SJ. (1989). Factors influencing the induction of DNA single strand breaks in rats by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicology, 58, 57–69
  • Walisser JA, Glover E, Pande K, et al. (2005). Aryl hydrocarbon receptor-dependent liver development and hepatotoxicity are mediated by different cell types. Proc Natl Acad Sci U S A, 102, 17858–63
  • Walker NJ, Miller BD, Kohn MC, et al. (1998). Differences in kinetics of induction and reversibility of TCDD-induced changes in cell proliferation and cyp1a1 expression in female Sprague-Dawley rat liver. Carcinogenesis, 19, 1427–35
  • Walker NJ, Portier CJ, Lax SF, et al. (1999). Characterization of the dose-response of cyp1b1, cyp1a1, and cyp1a2 in the liver of female Sprague-Dawley rats following chronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol, 154, 279–86
  • Walker NJ, Wyde ME, Fischer LJ, et al. (2006). Comparison of chronic toxicity and carcinogenicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in 2-year bioassays in female Sprague-Dawley rats. Mol Nutr Food Res, 50, 934–44
  • Wambaugh J, Shah I. (2010). Simulating microdosimetry in a virtual hepatic lobule. PLoS Comput Biol, 6, e1000756
  • Wang JH, Hinrichsen LI, Whitacre CM, et al. (1990). Nuclear DNA content of altered hepatic foci in a rat liver carcinogenesis model. Cancer Res, 50, 7571–6
  • Wang S, Ge K, Roeder RG, Hankinson O. (2004). Role of mediator in transcriptional activation by the aryl hydrocarbon receptor. J Biol Chem, 279, 13593–600
  • Ward JM, Henneman JR. (1990). Naturally-occurring age-dependent glutathione S-transferase pi immunoreactive hepatocytes in aging female f344 rat liver as potential promotable targets for non-genotoxic carcinogens. Cancer Lett, 52, 187–95
  • Wassom JS, Huff JE, Loprieno N. (1977). A review of the genetic toxicology of chlorinated dibenzo-p-dioxins. Mutat Res, 47, 141–60
  • Watabe Y, Nazuka N, Tezuka M, Shimba S. (2010). Aryl hydrocarbon receptor functions as a potent coactivator of e2f1-dependent trascription activity. Biol Pharm Bull, 33, 389–97
  • Weiss C, Faust D, Schreck I, et al. (2008). Tcdd deregulates contact inhibition in rat liver oval cells via ah receptor, jund and cyclin a. Oncogene, 27, 2198–207
  • Weiss C, Kolluri SK, Kiefer F, Gottlicher M. (1996). Complementation of ah receptor deficiency in hepatoma cells: negative feedback regulation and cell cycle control by the ah receptor. Exp Cell Res, 226, 154–63
  • Welzel TM, Graubard BI, El-Serag HB, et al. (2007). Risk factors for intrahepatic and extrahepatic cholangiocarcinoma in the United States: a population-based case-control study. Clin Gastroenterol Hepatol, 5, 1221–8
  • Westerink WM, Stevenson JC, Schoonen WG. (2008). Pharmacologic profiling of human and rat cytochrome p450 1a1 and 1a2 induction and competition. Arch Toxicol, 82, 909–21
  • Whysner J, Williams GM. (1996). 2,3,7,8-tetrachlorodibenzo-p-dioxin mechanistic data and risk assessment: gene regulation, cytotoxicity, enhanced cell proliferation, and tumor promotion. Pharmacol Ther, 71, 193–223
  • Wicha MS, Liu S, Dontu G. (2006). Cancer stem cells: an old idea – a paradigm shift. Cancer Res, 66, 1883–90
  • Wihlen B, Ahmed S, Inzunza J, Matthews J. (2009). Estrogen receptor subtype- and promoter-specific modulation of aryl hydrocarbon receptor-dependent transcription. Mol Cancer Res, 7, 977–86
  • Wincent E, Amini N, Luecke S, et al. (2009). The suggested physiologic aryl hydrocarbon receptor activator and cytochrome p4501 substrate 6-formylindolo[3,2-b]carbazole is present in humans. J Biol Chem, 284, 2690–6
  • Worner W, Schrenk D. (1996). Influence of liver tumor promoters on apoptosis in rat hepatocytes induced by 2-acetylaminofluorene, ultraviolet light, or transforming growth factor beta 1. Cancer Res, 56, 1272–8
  • Wu Q, Suzuki JS, Zaha H, et al. (2008). Differences in gene expression and benzo[a]pyrene-induced DNA adduct formation in the liver of three strains of female mice with identical AHRb2 genotype treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin and/or benzo[a]pyrene. J Appl Toxicol, 28, 724–33
  • Wyde ME, Eldridge SR, Lucier GW, Walker NJ. (2001a). Regulation of 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced tumor promotion by 17 beta-estradiol in female Sprague–Dawley rats. Toxicol Appl Pharmacol, 173, 7–17
  • Wyde ME, Wong VA, Kim AH, et al. (2001b). Induction of hepatic 8-oxo-deoxyguanosin adducts by 2,3,7,8-tetrachlorodibenzo-p-dioxin in Sprague-Dawley rats is female-specific and estrogen-dependent. Chem Res Toxicol, 14, 849–55
  • Xu L, Li AP, Kaminski DL, Ruh MF. (2000). 2,3,7,8 tetrachlorodibenzo-p-dioxin induction of cytochrome p4501a in cultured rat and human hepatocytes. Chem Biol Interact, 124, 173–89
  • Yager JDJ, Yager R. (1980). Oral contraceptive steroids as promoters of hepatocarcinogenesis in female Sprague-Dawley rats. Cancer Res, 40, 3680–5
  • Yang W, Yan HX, Chen L, et al. (2008). Wnt/beta-catenin signaling contributes to activation of normal and tumorigenic liver progenitor cells. Cancer Res, 68, 4287–95
  • Yeager RL, Reisman SA, Aleksunes LM, Klaassen CD. (2009). Introducing the “TCDD-inducible AHR-NRF2 gene battery”. Toxicol Sci, 111, 238–46
  • Yoon CY, Park M, Kim BH, et al. (2006). Gene expression profile by 2,3,7,8-tetrachlorodibenzo-p-dioxin in the liver of wild-type (AHR+/+) and aryl hydrocarbon receptor-deficient (AHR−/−) mice. J Vet Med Sci, 68, 663–8
  • Zeytun A, McKallip RJ, Fisher M, et al. (2002). Analysis of 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced gene expression profile in vivo using pathway-specific cdna arrays. Toxicology, 178, 241–60
  • Zhang L, Zheng W, Jefcoate CR. (2003). Ah receptor regulation of mouse cyp1b1 is additionally modulated by a second novel complex that forms at two AHR response elements. Toxicol Appl Pharmacol, 192, 174–90
  • Zhang Q, Andersen ME. (2007). Dose–response relationship in anti-stress gene regulatory networks. PLoS Comput Biol, 3, e24
  • Zhang ZY, Pelletier RD, Wong YN, et al. (2006). Preferential inducibility of cyp1a1 and cyp1a2 by TCDD: differential regulation in primary human hepatocytes versus transformed human cells. Biochem Biophys Res Commun, 341, 399–407

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.