Skip to main content
Dose-Response logoLink to Dose-Response
. 2006 May 1;3(3):306–331. doi: 10.2203/dose-response.003.03.003

Induction of Oxidative Stress Responses by Dioxin and other Ligands of the Aryl Hydrocarbon Receptor

John F Reichard 1, Timothy P Dalton 1, Howard G Shertzer 1, Alvaro Puga 1
PMCID: PMC2475945  PMID: 18648615

Abstract

TCDD and other polyhalogenated aromatic hydrocarbon ligands of the aryl hydrocarbon receptor (AHR) have been classically considered as non-genotoxic compounds because they fail to be directly mutagenic in either bacteria or most in vitro assay systems. They do so in spite of having repeatedly been linked to oxidative stress and to mutagenic and carcinogenic outcomes. Oxidative stress, on the other hand, has been used as a marker for the toxicity of dioxin and its congeners. We have focused this review on the connection between oxidative stress induction and the toxic effects of fetal and adult dioxin exposure, with emphasis on the large species difference in sensitivity to this agent. We examine the roles that the dioxin-inducible cytochromes P450s play in the cellular and toxicological consequences of dioxin exposure with emphasis on oxidative stress involvement. Many components of the health consequences resulting from dioxin exposure may be attributable to epigenetic mechanisms arising from prolonged reactive oxygen generation.

1. INTRODUCTION

Many polynuclear polyhalogenated aromatic hydrocarbons (PHAHs) are known or suspected environmental carcinogens, toxicants and teratogens in animals and humans (Gatmaitan et al., 1977; Talalay et al., 1988; Hebert et al., 1990; Jiang et al., 1991; Butler et al., 1992; Ralston et al., 1994; Hatae et al., 1996). 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD, dioxin) is prototypical of PHAH compounds, including polyhalogenated dibenzo-p-dioxins, dibenzofurans and coplanar biphenyls, that bind to and activate the cytosolic aryl hydrocarbon receptor (AHR). TCDD is a co-planar polychlorinated biphenyl with among the highest AHR binding affinities and agonistic activities (Poland et al., 1976a). It is this interaction of PHAHs, such as TCDD, with the AHR that mediate most if not all effects of low-concentration TCDD exposures.

The AHR is the only bona fide ligand-activated member of the PAS superfamily of proteins, named for the PER (“period” regulator of circadian rhythm), ARNT (“Ah receptor nuclear translocator”) and SIM (“single minded”, regulator of midline cell differentiation) members of helix-loop-helix transcription factors (Alsharif et al., 1994; Hassoun et al., 2003). Prior to ligand binding the AHR resides in the cytosol, associated with two molecules of HSP90 and HSP90 accessory proteins. Upon TCDD binding, the AHR is released from this cytosolic complex and is translocated into the nucleus where it forms a heterodimeric complex with ARNT (Okey et al., 1989). This complex binds to one or more aryl hydrocarbon response elements (AhRE; also known as xenobiotic response elements, XRE; and dioxin response elements, DRE) that function as cis-acting enhancers in the regulatory domains of a growing number of genes collectively known as the AHR gene battery (Nebert et al., 1993). Battery members include phase I cytochromes P450 (CYP) Cyp1a1 and Cyp1a2, Cyp1b1 and NAD(P)H quinone oxidoreductase (Nqo1), and phase II antioxidant enzymes such as UDP-glucuronosyltransferase (Ugt1a1), glutathione S-transferase (Gst1a1) and aldehyde dehydrogenase (Aldh3a1).

Ligands for the AHR include planar PHAHs and diverse classes of plant-derived chemicals. It has been hypothesized that the AHR/ARNT transcriptional complex evolved for defense against an increasingly diverse array of plant toxins and as a result it is unlikely to serve endogenous physiological functions (Gonzalez et al., 1990). More recently however, the AHR has emerged as an important regulator of physiologic and developmental processes in the absence of an apparent exogenous (xenobiotic) ligand (Fernandez-Salguero et al., 1997; Lahvis et al., 2000). The AHR represents a pivotal upstream event in the apoptosis cascade (Nebert et al., 2000; Slim et al., 2000; Dong et al., 2004), exerts an important level of influence on reproductive success (Abbott et al., 1999) and participates in cell cycle regulation (Puga et al., 2002; Marlowe et al., 2004). Further, acting through the AHR, TCDD has been shown to modulate up- or down-regulation of more than 300 known mRNAs and an equivalent number of expressed sequence tags (Puga et al., 2000b). In part, this effect can be attributed to interactions between the AHR and transcription factors other than ARNT, some of which are involved in the control of complex cellular programs, such as cell division and cell fate (Ge et al., 1998; Kolluri et al., 1999; Tian et al., 1999; Puga et al., 2000a; Elferink et al., 2001; Puga et al., 2002; Marlowe et al., 2004). In light of such studies, it is likely that the AHR has important roles in regulating cellular homeostasis that may be disrupted by environmental chemicals. The diversity of AHR ligand interactions, the complexity of the cellular transcriptome, the persistence of AHR activation, and the nature of agonist exposure determine whether the homeostatic equilibrium is maintained or perturbed.

The toxicologic responses elicited by TCDD differ widely among animal species and strains. These differences are attributable to variations in a number of molecular, tissue specific, biochemical and physiological characteristics. In making inter-model comparisons, TCDD dose can be expressed as a variety of different metrics such as administered dose, average daily dose, tissue concentration, average body burden and area under the curve (AUC). As a result, clear-cut dose-response assessments of TCDD are made difficult by the complexity of biologic responses to TCDD, the variety of tissues affected by TCDD and gaps in our understanding of the mechanisms relating exposure to toxicity. As a result, body burden rather than daily intake (administration) has been suggested as the best dose metric for interspecies comparisons and extrapolation, although the vast majority of studies describing TCDD toxicity express dose in terms of acute, subchronic and chronic exposures (DeVito et al., 1995).

2. FUNCTIONAL ALTERATIONS OF THE AHR

Like many other transcription factors, the AHR has been amenable to dissection into functional domains. The C-terminal half of the AHR, containing a glutamine-rich domain, is responsible for transactivation; whereas the N-terminal half of the AHR, consisting of a basic-region helix-loop-helix domain and two PAS domains, has overlapping functions responsible for DNA binding, ligand binding and dimerization (Hankinson, 1995). Unfortunately, the AHR peptide sequence is not particularly well conserved across species, especially the C-terminal half of the protein, thus complicating risk assessment. Polymorphisms identified within the coding region of the AHR instill differences in AHR-responsive gene induction and toxicologic responses to numerous PHAHs (Nebert, 1989; Swanson et al., 1993; Poland et al., 1994). Interspecies variation notwithstanding, the AHR has been widely studied in mice and rats, which, relative to the human AHR, have high ligand binding affinity.

In mice, differences in TCDD sensitivity have been related to polymorphisms in the AHR that give rise to the commonly studied “responsive” and “nonresponsive” strains (C57BL/6 and DBA/2, respectively). AHR polymorphisms in DBA/2 mice reduce ligand binding affinity approximately 10-fold and thereby diminish TCDD potency for acute lethality (Chapman et al., 1985; Okey et al., 1989). Several groups have sequenced the AHR alleles from inbred strains of mice. These studies have characterized four distinct alleles in mice, referred to as Ahrb-1, Ahrb-2, Ahrb-3, and Ahrd. Among these, the “responsive” phenotype in C57BL/6 mice is encoded by the autosomal dominant Ahrb-1 allele while the “nonresponsive” DBA/2 phenotype is encoded by the Ahrd allele. The four identified mouse alleles differ by 8 nucleotides in their shared open reading frame. In addition, these AHRs differ by 45 amino acids at their C-terminus as a result of a nucleotide change in the Ahrd allele that replaces the stop codon in the Ahrb allele with an arginine (Chang et al., 1993; Poland et al., 1994). Most of the amino acid changes distinguishing these strains occur within the transactivation domain and have little or no known functional consequence (Chang et al., 1993). However, a point mutation at position 375 of the DBA/2 AHR results in an ALA to VAL substitution in the second PAS domain of the C57BL/6 strain that is responsible for the difference in ligand binding affinity and transactivation (Poland et al., 1994; Maier et al., 1998). These findings have been further supported in mice with homozygous loss of functional AHR (Fernandez-Salguero et al., 1995; Schmidt et al., 1996). These Ahr−/− knockout strains were refractory to TCDD-mediated CYP1A1 induction and were highly resistant to TCDD-mediated pathologies up to 2000 μg/kg, a 10-fold higher dose than that which induce severe toxicity in functional AHR expressing mice (Fernandez-Salguero et al., 1996).

Rat strains have also been characterized with respect to their TCDD sensitivity. At the extremes of TCDD responsiveness are the “sensitive” Long-Evans rats (L-E) and the “resistant” Hans/Wistar (H/W) substrain of Wistar rats that differ by at least 1000-fold in the acute lethality of TCDD (LD50 between 10 to 20 μg/kg and >9600 μg/kg, respectively) (Pohjanvirta et al., 1994b). Inheritance studies implicate the AHR gene locus and a second uncharacterized gene B in the TCDD resistance of the H/W rats, with the AHR contributing the largest quantitative role (Tuomisto et al., 1999). Unlike the C57BL/6 and DBA/2 mice, TCDD resistance is the dominant trait in rats, segregating with autosomal inheritance (Pohjanvirta et al., 1999). Molecular analysis of the coding region of AHR cDNAs from the H/W and the L-E rat revealed a Val497Ala amino acid change in the transactivation domain and, perhaps more importantly, a single point mutation in the first nucleotide of intron 10, resulting in altered mRNA splicing (Pohjanvirta et al., 1998). The loss of this splice-donor site results in the use of the nearest upstream and two downstream consensus splice sites that yields three different molecular AHR species having either a deletion of 43 amino acids in exon 10, an extra 7-amino acid stretch encoded by intron 10, or no translated contribution from exon 11. The net effect of the exon 10 mutation is a modified AHR transactivation domain that has little or no effect on AHR accumulation, ligand binding affinity, or activation of CYP1A gene expression (Pohjanvirta et al., 1988; Unkila et al., 1993; Pohjanvirta et al., 1999), but which effectively converts the Han/Wistar rat into the most resistant naturally occurring mammals to TCDD toxicity (Simanainen et al., 2003).

3. PRINCIPLES OF TCDD SENSITIVITY

Within a single animal, tissues vary in response to TCDD-receptor binding. It is the coupling of TCDD-receptor interaction to a measured response that accounts for varying tissue sensitivities and thereby target organ toxicity. In general, the maximum response elicited by a receptor agonist for a specific endpoint is defined as the intrinsic efficacy of a ligand. Efficacy is therefore a quantitative measure of the signaling events that couples the formation of a ligand-receptor complex to a biologic response (Hestermann et al., 2000). By measuring nonlethal endpoints, differences in TCDD intrinsic efficacy between L-E and H/W rat strains have been categorized into two classes: Type I endpoints (EROD activities, thymus weight, tooth defect) that showed similar efficacy in both strains; and Type II endpoints (body weight, serum FFA and bilirubin levels, and serum ASAT activity) where the response in H/W rats was less than half that observed in L-E rats (Simanainen et al., 2003). The contribution of the AHR and the product of gene B to these endpoints was investigated by segregating the H/W resistant genes into three different rat lines, designated A, B and C, by congenic crossbreeding with inbread L-E rats (Tuomisto et al., 1999). Line A possessed the original “resistant” H/W AHR allele but with a wild-type gene B allele. Line B possessed a normal AHR allele, but was homozygous for the H/W gene B allele. Line C possessed neither of the H/W resistance alleles. These studies demonstrated that the AHR is the most important factor decreasing TCDD intrinsic efficacy, and that an uncharacterized mechanistic difference exists between type I and II effects that is linked to the altered AHR transactivation domain. Relative to the large difference in acute LD50 values between L-E and H/W rat strains, the potency of TCDD for nonlethal type I endpoints was much less affected by the H/W AHR phenotype (Tuomisto et al., 1999; Simanainen et al., 2003). In rat line B, the mutated B allele had only a minor influence on TCDD efficacy and the dose responses did not clearly fit into either Type I or Type II responses, but were clearly different from lines A and C. Thus the B allele is concluded to contribute modestly to TCDD resistance independent of the AHR (Simanainen et al., 2003).

The combination of efficacy and ligand-binding affinity determine the relative potency of TCDD (Hestermann et al., 2000), which is defined as the dose of TCDD required to achieve a specific endpoint. Both parameters contributing to potency can vary between animal species, strains and tissues to produce net sensitivities. For example experiments investigating the relative potency of various AHR ligands have shown that TCDD, PCB126, PCB156 and PCB105 all bind to the AHR with reported affinity constants equivalent to 0.76, 16, 2500 and 4600 nM respectively, relative to [3H]-TCDD binding. However, the stimulus-response relationship demonstrates that while TCDD and PCB126 have high intrinsic efficacy for CYP1A1 induction, PCB126 is much less efficient at eliciting a response after binding the AHR. PCB105, which binds the AHR at high concentrations, competes for [3H]TCDD binding while eliciting no response qualifying PCB105 as a competitive antagonist. With the exception of PCB105, each of these agents is a full agonist since each elicits the same maximal response. Therefore the potency of these AHR ligands can be expressed in terms of the effective concentration that elicit 50% maximal response for CYP1A1 induction (EC50) and have been shown to be equivalent to 0.015 nM (TCDD), 0.12 nM (PCB126) and 1900 nM (PCB156). In comparison PCB105 has no EC50 since it does not elicit a response (Hestermann et al., 2000).

This same principle has been utilized to describe the biologic responses observed between different animal species and strains. For example, the efficacy of TCDD for lethality ranges among rodent species from the guinea pig (LD50 = 1 μg/kg) to H/W rat (LD50 >9600 μg/kg) (Henck et al., 1981; Poland et al., 1982; Pohjanvirta et al., 1994a). This range of sensitivity has been attributed to a restructured transactivation domain in the hamster AHR, presumably producing a much less responsive ligand-receptor complex (Korkalainen et al., 2004). In comparison, experiments investigating TCDD resistance in mice have found that sensitivity correlates with binding affinity because the dose-response curve is shifted to the right without a reduction in response magnitude (Poland et al., 1976b). Thus, both intrinsic efficacy and ligand-binding contribute to the manifestation of TCDD toxicity and the tremendous variability in dose response that has been reported between animal species.

In vitro modeling of the TCDD dose-response using CYP1A1 induction as a biomarker has demonstrated that TCDD need only occupy a fraction of AHR receptors to elicit maximal response. In PLHC-1 cells, a hepatocellular carcinoma cell line derived from the teleost Poeciliopsis lucida, only 1.9 % of available receptor sites were required to be occupied for 50% maximal response while 28% saturation produced 95% maximal response. These data establish a “spare” receptor relationship for the high-intrinsic efficacy AHR ligands such as TCDD, which contrasts with the low-intrinsic activity of ortho-substituted PCB congeners that fail to elicit maximal response even with AHR saturation (Hestermann et al., 2000).

In addition to the individual contribution of intrinsic efficacy and ligand binding to the wide range of observed species and strain TCDD susceptibilities, receptor density also modulates the response. Regulation of receptor expression levels by its own ligand is a common pharmacologic observation and receptor theory predicts that changes in AHR levels will influence both the potency and the maximal response of TCDD. Up-regulation of a receptor's presence increases the potency of its ligand and is referred to as “sensitization”, while down regulation results in “desensitization” and subsequent tolerance. In such a manner, AHR expression is significantly influenced by dose and duration of TCDD exposure (Pollenz, 2002). Following short-term in vitro exposure of cultured Hepa-1 cells to 2 nM TCDD, AHR levels are reduced to less than 20% of original levels within 6 hours following treatment, and this desensitization persists for at least 72 hours (Giannone et al., 1998). In vivo studies, however, have failed to consistently demonstrate a prominent physiologic effect caused AHR by ligand binding. In Sprague Dawley rats, a single oral dose of 10 or 50 μg/kg produced a pronounced initial reduction of liver and cytosol AHR concentrations, though, in the case of the former, depletion persisted for 14 days, while in the latter, depletion was followed by AHR induction (Pollenz et al., 1998; Franc et al., 2001a). A similar effect was also reported for Hans/Wistar rats, though these rats had lower liver AHR concentrations than either SD or L-E rats regardless of TCDD treatment (Franc et al., 2001a). In contrast to high-dose TDCC-mediated loss of the AHR, acute and chronic low-dose TCDD administration produced either an increase, or no change in AHR concentrations (Sloop et al., 1987; Franc et al., 2001a; Franc et al., 2001b). In addition, when increased receptor presence was observed it did not translate into sensitization as determined by CYP1A1 induction (Franc et al., 2001a; Franc et al., 2001b). These reports suggest that low-dose TCDD, such as a typical environmental exposure, is not likely to produce either TCDD tolerance or sensitivity, while higher doses appear to be associated with a transient desensitization. They also demonstrate that receptor density does not contribute to the variation in TCDD responsiveness associated with the L-E and H/W rat strains.

4. EFFECTS OF GENDER AND SEX HORMONES IN THE TCDD DOSE-RESPONSE

In long-term bioassays, TCDD increased the incidence of liver tumors in female, but not male, rats (Kociba et al., 1978; Huff et al., 1991; Sawyer et al., 1999). In general, female rats have been shown to be more susceptible to TCDD-induced oxidative stress (Stohs, 1990), oxidative DNA damage (Wyde et al., 2001b) and hepatocarcinogenesis (Huff et al., 1994). These TCDD-mediated effects are, at least in part, dependent on the presence of estrogen (Jana et al., 2000; Coumoul et al., 2001; Lai et al., 2004), though the role of the estrogen receptor remains equivocal (Wyde et al., 2000; Wyde et al., 2001a). In an initiation-promotion model, ovariectomy inhibited TCDD-induced preneoplastic foci and reduced TCDD-induced liver tumor formation (Lucier et al., 1991) suggesting involvement of estrogen which was later supported by the observation that supplemental 17β-estradiol (E2) restored tumorogenic sensitivity of ovariectomized females (Wyde et al., 2001b). The presumption that estrogen mediates oxidative stress and carcinogenesis was confirmed in Syrian hamsters, which show 100% kidney tumor incidence following the administration of 17β-estradiol or estrone (Liehr, 1997). Likewise estradiol was also found to produce oxidative DNA damage in hamster tissues and other biological model systems (Han et al., 1995; Tritscher et al., 1996; Wyllie et al., 1997; Hodgson et al., 1998; Cavalieri et al., 2000; Liehr, 2001; Wyde et al., 2001b).

Oxidative estrogen metabolism results in the formation of two estrogen catechols, 2-hydroxylated and 4-hydroxylated estradiol. Under circumstances where these catechols are excessively produced, or where their metabolism is impaired, catalytic oxidation to semiquinones and quinones can occur. Of particular importance is formation of 4-hydroxyestradiol, the oxidized quinone of which has been associated with oxidative DNA damage and increased cancer risk (Bradlow et al., 1985; Bradlow et al., 1986; Telang et al., 1992; Nebert, 1993; Bradlow et al., 1995; Telang et al., 1997; Liehr, 1999; Jefcoate et al., 2000; Cavalieri et al., 2000). In contrast, 2-hydroxyestradiol formation has not been associated with either DNA damage or increased cancer risk (Bradlow et al., 1996; Telang et al., 1997; Cavalieri et al., 2000). Toxicity of 4-hydroxyestradiol results from two types of reactions; a one electron redox cycling reaction that occurs when 4-hydroxyestradiol is oxidized to estrone 3,4-quinone, and a two electron electrophilic addition reaction (Liehr, 2000; Cavalieri et al., 2000). Redox cycling generates superoxide and ultimately the highly reactive genotoxic hydroxyl radical (Roy et al., 1991; Han and Liehr, 1995). Superoxide produced in this way may further enhance redox cycling by mobilizing iron from ferritin, increasing cellular Fenton chemistry (Wyllie and Liehr, 1997; Liehr et al., 2001). Rearrangement of estrone 3,4-quinone produces a strongly electrophilic carbonium cation that may undergo a Michael addition reaction with cellular sulfhydrals such as (i.e. glutathione, protein thiols) or by electrophilic addition to DNA purine bases resulting in depurinating adducts (Cavalieri et al., 2000), and ultimately procarcinogenic mutations (Liehr, 2001; Embrechts et al., 2003). The relative contributions of redox cycling and electrophilic interactions in the oxidative stress response and toxicity have not been firmly established; however, limited evidence suggests that covalent sulfhydral modification by electrophiles is likely to be a greater cytotoxic hazard than transient quinone formation that facilitates disposal from the cell (Buffinton et al., 1989).

Xenobiotics acting through the AHR may alter the metabolic profile of E2 and therefore its estrogenic and toxicological profile. Metabolism of E2 to 2-hydroxyestradiol is predominantly catalyzed by cytochrome P450 CYP1A1 (Roy et al., 1992; Spink et al., 1998) with some contribution by members of the CYP3A family (Hammond et al., 1997), while metabolism of E2 to the 4-hydroxyestradiol is mainly a result of CYP1B1 activity (Spink et al., 1994; Hayes et al., 1996; Jefcoate et al., 2000). In liver, TCDD increases the levels of CYP1A1 and CYP1A2 relative to CYP1B1 (Walker et al., 1999), hence 2-hydroxylation predominates over 4-hydroxylation. Similar results have been reported in several breast epithelial tumor and nontumor cell lines where TCDD strongly induced CYP1A1 activity with resultant 2-hydroxyestradiol formation as the major E2 metabolite (Spink et al., 1998). In this regard, increased production of 2-hydroxyestradiol relative to 4-hydroxyestradiol and 16α-hydroxyestrone has been observed after exposure to indole 3-carbinol, a dietary micronutrient and AHR proligand. This finding is of potential clinical importance for cancer, since indole carbinols, that bind AHR as acid condensation products, are in clinical trials as cancer chemoprotective agents (Gillner et al., 1985; Malloy et al., 1997; Michnovicz et al., 1997; Telang et al., 1997; Rosen et al., 1998; Yuan et al., 1999; Bell et al., 2000) (reviewed by (Shertzer et al., 2000)).

5. EFFECTS OF TCDD EXPOSURE ON DEVELOPMENT

AHR in development

In the mammalian fetus and in fish larvae, the AHR plays prominent roles in both resolving vascular structures and mediating cardiovascular toxicities of TCDD (Lahvis et al., 2000; Bello et al., 2004). In mammals, the importance of functional AHR is demonstrated in AHR-null mice by a failure of a fetal vascular structure, the ductus venosus, to close, thus permitting blood from the portal vein to bypass the liver by shunting to the inferior vena cava. Functional AHR is also required for normal vascular “pruning” during fetal development, the absence of which results in the propagation into maturity the highly anastomotic vasculature architecture of the liver, eye and kidney that that are characteristically neonatal (Lahvis et al., 2000).

Because of the involvement of the AHR in resolving fetal vascular structures, it is not surprising that the cardiovascular system has been shown to be an important target of TCDD-mediated toxicity (Jokinen et al., 2003; Karyala et al., 2004). Although the mechanisms underlying cardiovascular risks are undetermined, it has been postulated that TCDD interferes with cardiovascular development by sequestering the AHR or displacing an as yet unidentified ligand, thereby preventing the AHR from carrying out its normal endogenous activity. In support of this hypothesis, knockdown of the TCDD-responsive AHR2 in zebrafish with morpholino-substituted oligonucleotides has specifically demonstrated that TCDD retardation of common cardinal vein (CCV) regression is AHR dependent (Bello et al., 2004). That knockdown of AHR2 expression itself did not inhibit CCV regression in a manner similar to that of the Ahr-null mouse is attributable to the fact that zebrafish possess a second, TCDD refractory ahr locus (ahr1) that may compensate for the loss of ahr2 (Bello et al., 2004). In separate experiments, fish have provided evidence that the vascular endothelium is also a sensitive target for TCDD toxicity, and this may prove important with regards to the human health effects of TCDD. In fish TCDD elicits increased vascular permeability, which in lake trout manifests as yolk sac edema, and in zebrafish as extravascular accumulation of serum proteins in mesencephalic brain tissues (Guiney et al., 2000; Dong et al., 2004). Although the precise nature of this microvascular leakage remains to be determined, decreased cardiac output, increased endothelial vacuolation (Guiney et al., 2000), disruption of peripheral vascular beds (Henry et al., 1997) and/or disruption of angiogenic signaling (Bello et al., 2004) have been suggested. Whether TCDD mediates developmental vascular defects in fish by the same mechanism as those observed in mammals following gestational exposure remains to be determined; however, two separate reports demonstrate that loss of the AHR protects against the teratogenic effects of TCDD (Mimura et al., 1997; Peters et al., 1999b).

In humans there are few reliable studies linking maternal exposure to TCDD and related compounds (e.g., other dioxins, furans, and dioxinlike PCBs) with impaired fetal development. A number of epidemiologic studies have been confounded by the use of indirect estimates of TCDD exposure, such as local soil levels (Stockbauer et al., 1988), estimates of dietary consumption (Svensson et al., 1991; Rylander et al., 2000) and correlation with residential location (Revich et al., 2001). Only a few studies have used biologic measures of exposure, such as dioxin or PCB concentrations in breast milk and serum (Patandin et al., 1998; Eskenazi et al., 2003). In both of these studies birth weight and gestational age did not differ between mothers with higher exposure levels relative to controls, though these findings are somewhat offset by reports that birth weight was negatively correlated with cord plasma PCB and dioxin levels (Patandin et al., 1998; Vartiainen et al., 1998). One mechanistic explanation for the equivocal association between maternal exposure and teratogenesis may relate to the low affinity human AHR, comparable to the nonresponsive DBA/J2 mouse strain (Ramadoss et al., 2004). In experiments with AHR-null mice, oral exposure of pregnant dams (40 μg/kg TCDD) was sufficient to produce cleft palate and hydronephrosis in nearly all wild-type fetuses while no mice with the homologous AHR knockout were sensitive to the teratogenic effects (Mimura et al., 1997; Peters et al., 1999a). This possibility is further supported by the use of humanized mice expressing human AHR rather than mouse AHR. These studies demonstrated that mice expressing human AHR had a weaker response to TCDD than resistant DAB/2 mice, and that the humanized AHR phenotype protected against cleft palate (Moriguchi et al., 2003).

A variety of human epidemiologic studies have suggested a link between TCDD exposure and cardiovascular morbidity following occupational exposure (Bertazzi et al., 1989; Flesch-Janys et al., 1995; Vena et al., 1998; Pesatori et al., 1998; Pesatori et al., 2003). Retrospective analysis of 1189 chemical plant workers exposed to dioxin and furans reported a highly significant 2.5-fold (95% confidence interval-1.3–4.7) increase in relative-risk of death from heart disease due to dioxin exposure (Flesch-Janys et al., 1995; Pelclova et al., 2002). The body burden at which these effects were seen ranges from 110 to 4000 ng/kg of TCDD in blood fat, well below the body burden of TCDD shown to induce cancer in rodents (100–140000 ng/kg) (DeVito et al., 1995). These observations were later supported in hyperlipidemic mice subchronicly treated with TCDD (150 ng/kg, 3 times weekly), resulting in increased blood pressure and atherogenic lipids; the two most important clinical risk factors for atherosclerotic plaque formation. Further, TCDD exposed animals had a trend towards earlier onset and increased severity of atherosclerotic plaques compared to vehicle treated mice (Dalton et al., 2001). Similarly, in female Sprague-Dawley rats treated 5 days per week with up to 100 ng/kg/day TCDD for 2 years, cardiomyopathy and chronic active arteritis increased in a dose dependent manner. However the severity of cardiomyopathy did not increase in a dose-responsive manner and only became evident in the later treatment groups (Jokinen et al., 2003)

6. ROLE OF CYTOCHROME P450 ENZYMES IN TCDD TOXICITY

Several cytochrome P450 genes under the control of the AHR, notably those in the CYP1 family (CYP1A1, CYP1A2 and CYP1B1) have been suggested to contribute to TCDD-induced toxicity (Andersen et al., 1998; Nebert et al., 2004). Because TCDD-associated toxicities are slow to develop, requiring days to weeks, it is likely that the transcriptional events elicited by TCDD-mediated AHR activation must be persistent. Therefore, we believe that the persistent changes in gene expression induced by TCDD disrupt signal transduction homeostasis leading to the accumulation of toxicants (i.e reactive oxygen species, lipid peroxidation products) that in turn lead to pathology. In the liver, one such gene circuit involves the Cyp1a monooxygenase subfamily. Studies utilizing Cyp1a1−/− knockout mice from a C57BL/6J background demonstrate that a single high dose of TCDD (200 μg/kg) is highly lethal to Cyp1a1+/+ males but not to Cyp1a1−/− males or to females of either genotype. This protective effect conveyed by gender, however, is quite limited compared to the protective effect afforded by Ahr knockout, that protected against TCDD doses of up to 2 mg/kg (Fernandez-Salguero et al., 1996). Further, Cyp1a1−/− mice are resistant to TCDD-induced wasting syndrome, which is manifested by weight loss or poor weight gain in conjunction with marked increases in serum AST levels, reflecting rhabdomyolysis. Glycogen depletion and down regulation of phospho-enol-pyruvate carboxykinase, combined with SRC oncoprotein action have been suggested to play a role in this process (Dunlap et al., 2002). Cyp1a1−/− mice, regardless of gender, are more resistant to hepatocyte hypertrophy; likewise, Cyp1a1−/− mice experience decreased accumulation of microvesicular and interstitial lipid accumulation for reasons that are not yet clear. It is of interest to note that the H/W rat, which is resistant to TCDD toxicity for reasons already discussed, shows normal acute induction of CYP1 family genes and uroporphyria following TCDD exposure (Simanainen et al., 2003; Niittynen et al., 2003; Uno et al., 2004; Korkalainen et al., 2004).

The other member of the cytochrome P4501A family, CYP1A2, is emerging as a monooxygenase that dichotomously contributes to both protective and sensitizing effects to TCDD toxicity. The protective effects result from pharmacokinetic and antioxidant activities associated with CYP1A2 expression. Pharmacokinetically, CYP1A2 is the primary hepatic TCDD-binding protein, capable of sequestering significant quantities of dioxin; this is not true of CYP1A1 (Diliberto et al., 1997; Uno et al., 2004). Further, CYP1A2 is stabilized by TCDD extending its half-life and therefore augmenting its phramacokinetic effect (Andersen et al., 1997; Diliberto et al., 1997). Presumably by sequestering TCDD, CYP1A2 acts to reduce the free fraction of TCDD available to mediate gene induction through AHR interaction. Pharmacokinetic studies have shown that levels of both CYP1A1 and CYP1A2 must be considered in predicting tissue concentrations of TCDD from the administered doses (Wang et al., 1997a; Wang et al., 1997b; Santostefano et al., 1998). In terms of antioxidant protection, CYP1A2 enzyme activity is associated with decreased microsomal H2O2 production, possibly by acting as an electron transport pathway or electron sink for uncoupled electron transfer by CYP2E1 or other microsomal enzyme systems.

Contrasting with these apparent beneficial effects, CYP1A2, and to some extent CYP1A1, has been demonstrated in mice to mediate the uroporphyrinogenic effect of TCDD. In brief, uroporphyria results from dysfunction of uroporpheryninogen decarboxylase (UROD) during hepatic heme synthesis, leading to significant hepatocellular uroporphyrin accumulation and possibly liver injury (Pohjanvirta and Tuomisto, 1994a; Smith et al., 1998). In TCDD-mediated uroporphyria, CYP1A2 is necessary and sufficient to inhibit UROD metabolism, resulting in accumulation of uroporphyrin isomers (Smith et al., 1998). Knockout mouse experiments have shown that loss of CYP1A2 completely, and CYP1A1 partially, protects against TCDD-mediated uroporphyrin accumulation (Smith et al., 2001; Uno et al., 2004).

In the course of the reaction catalyzed by monooxygenase P450 enzymes, two electrons are sequentially transferred from NADPH-dependent cytochrome P450 oxidoreductase to each atom of bound oxygen, resulting in the production of oxygenated substrate and water (Guengerich et al., 1985; Poulos et al., 1992). This reaction is reversible, a process that is perhaps toxicologically important, since physiologically-derived peroxides can metabolize various xenobiotics, particularly carcinogenic arylamines, via the peroxidase activity of CYP1A2 (Anari et al., 1997). Although tight coupling normally exists between oxygen reduction and monooxygenation, some reactive oxygen may be released as either superoxide or H2O2 in the course of electron transfer. The monooxygenase-dependent production of ROS in liver microsomes, supported by NADPH, is a well-known phenomenon (Gillette et al., 1957) that clearly contributes to the total cellular production of reactive oxygen in rat liver, without necessitating enzyme induction (Bondy et al., 1994). Even in the absence of exogenous xenobiotic substrates, endogenous substrates, such as any one of the many arachidonic acid metabolites, may stimulate ROS production (Capdevila et al., 1988; Rifkind et al., 1990; Nakai et al., 1992). In this regard, lipoxin A4, a metabolite of arachidonic acid, may act as an inducing ligand for the AHR (Schaldach et al., 1999). Further, substrate independent ROS production, due to inefficient microsomal electron coupling, has been demonstrated for CYP2E (Ekstrom et al., 1986; Dai et al., 1993), CYP2B, and CYP3A (Ahmed et al., 1995).

While xenobiotic AHR ligands, such as TCDD, can induce microsomal CYP1 expression and ROS production, suppression of CYP1A1 activity has been reported with high dose exposure to several PHAH. This phenomenon has been studied in fish and rodent liver microsomes using the AHR inducing compounds 3,3′,4,4′-tetrachlorodiphenyl (TCB) and 3,3′,4,4′,5-pentachlorobiphenyl (PeCB) (Schlezinger et al., 2001). CYP1A1 enzyme activity was strongly inhibited even though treatment with the halogenated biphenyls increased cyp1a1 mRNA. Since these compounds are poorly metabolized, inhibition by product could not explain the results. The loss of CYP1A1 activity was attributed to the ability of TCB and PeCB to accelerate CYP1A1 electron flow with concomitantly increased ROS production. Although some reactive oxygen species are released by enzyme uncoupling, ROS scavengers were unable to prevent the loss of CYP1A1 activity indicating that the chemistry involved occurs entirely within the enzyme active site. TCB also stimulated ROS production in microsomes from insect cells expressing human CYP1A1, but not in microsomes from cells expressing human CYP1A2 (Schlezinger et al., 1999). These results may explain the previous observation in mice that TCDD produced a sustained elevation of hepatic CYP1A2 activity, while CYP1A1 showed a transient increase, followed by a rapid loss (Shertzer et al., 1998).

7. TCDD-MEDIATED PERTURBATION OF REDOX HOMEOSTASIS

In addition to its involvement in normal physiological processes and signal transduction, the AHR appears to mediate toxicological effects through oxidative stress. As used here, the term oxidative stress refers to any condition that produces an oxidative stress response through an increase in the cellular oxidation state. An oxidative shift in cellular redox homeostasis generally results from increased production of reactive oxygen species relative to cellular antioxidant defenses. Although oxidative stress does not necessarily result in toxicity, it is an important mechanistic component of many toxicologic processes. In this regard, TCDD-mediated activation of the AHR shifts the cellular redox balance to produce an oxidative stress response (Hassoun et al., 1998; Shertzer et al., 1998; Slezak et al., 2000; Senft et al., 2002a; Senft et al., 2002b). For this reason it has become widely hypothesized that the toxicity induced by TCDD involves an oxidative stress component; an observation that has been reported by several laboratories (Stohs, 1990; Alsharif et al., 1994; Shertzer et al., 1998; Slezak et al., 2000).

Several mechanisms have been proposed to explain TCDD-mediated oxidative stress including reduction in expression levels of protective antioxidant enzyme systems (Latchoumycandane et al., 2003) and perturbation of cytochrome P450 levels (Nebert et al., 2000; Lee et al., 2002). The incomplete reduction of O2 by several enzyme systems, in particular the cytochrome P450 enzymes that are induced by TCDD, is known to result in the generation of superoxide and hydrogen peroxide through poor coupling of electron flow. TCDD has been implicated in the formation of the superoxide anion in rat brain (Hassoun et al., 2003) and hydrogen peroxide in mouse liver (Shertzer et al., 1998; Senft et al., 2002a), with resultant generation of lipid peroxides in rat brain, mouse liver and rat testis (Shertzer et al., 1998; Hassoun et al., 2003; Latchoumycandane et al., 2003). Several lines of evidence support the AHR as a mediator of oxidative stress. It has been observed that peritoneal lavage cells from C57BL/6 mice, which carry the high-affinity Ahrb1 allele, demonstrated considerably greater production of superoxide anion in response to TCDD relative to cells from low-affinity DBA/2 mice (Alsharif et al., 1994). Likewise, hepatic lipid peroxidation induced by TCDD occurred at low doses (500 ng/kg) in C57BL/6 mice and only at higher doses (5 μg/kg) in DBA/2 mice (Mohammadpour et al., 1988). In addition, inactivation of aconitase activity, a reliable measure of oxidative stress (Pantopoulos et al., 1995), was documented in C57BL/6 but not in DBA/2 mice following TCDD treatment (Smith et al., 1998).

It should be noted that TCDD dose and tissue concentration do not necessarily correlate with ROS production; the pattern of TCDD exposure also has a prominent effect on ROS production. In liver, an acute oral dose of TCDD (10 and 100 μg/kg) administered to C57BL/6 mice produced a sustained increase in liver superoxide anion and thiobarbituric acid reactive substance (TBARS), attaining to hepatic TCDD concentrations of 55 and 321 ng/g respectively at 13 weeks following exposure. In comparison, subchronic TCDD administration (0.15 to 150 ng/kg; 5 days/week for 13 weeks; po) produced increased superoxide and TBARS only with the highest (150 ng/kg/day) exposure level, corresponding to a hepatic TCDD concentration of 12 ng/g of liver. These data suggest that higher tissue TCDD concentrations are required to elicit oxidative stress following acute exposure than with subchronic TCDD exposure (Slezak et al., 2000). For this reason it is clear that uncharacterized factors remain that can contribute to tissue responses during TCDD exposure.

Enzyme systems catalyzing O2 reduction must be in balance within the cell because partially reduced oxygen species can be more reactive and deleterious than the parent molecule. Such is the case with hydrogen peroxide, which is generated during superoxide detoxification. Detoxification of superoxide to H2O requires the sequential action of SOD with catalase or glutathione peroxidase. Three- to six-fold overexpression of Cu/Zn SOD in transgenic mice results in increased production of H2O2 and hydroxyl radicals, which accompany enhanced apoptosis of thymocytes and bone marrow cells (Peled-Kamar et al., 1995). This is similar in nature to the enhanced neurotoxicity of kainic acid by SOD overexpression that also occurs through the generation of superoxide (Bar-Peled et al., 1996). Therefore, the consequence of TCDD-induced changes in antioxidant enzyme expression is uncertain, as illustrated by the fact that up-regulation of SOD does not necessarily dictate a decrease in cellular ROS.

Two sites of TCDD-induced reactive oxygen production have been proposed: the microsomes and the mitochondria. Microsomal reactive oxygen production in mouse liver is regulated by at least three forms of cytochrome P450s (Uno et al., 2004; Shertzer et al., 2004b) that clearly contribute to the total cellular production of reactive oxygen in rat liver (Dai et al., 1993; Bondy and Naderi, 1994). CYP1A1 and CYP2E1 generate reactive oxygen in liver microsomes, while CYP1A2 diminishes reactive oxygen production. The stoichiometric ratios of NADPH and O2 utilized relative to H2O2 produced indicate that the pathway of electron flow is short-circuited by TCDD-mediated microsome induction, resulting in increased H2O2 production (Shertzer et al., 2004a). CYP1A2 contributes to the time course of the oxidative stress response elicited by AHR ligands by reducing the microsomal oxidative stress response, including lipid peroxidation and decreased membrane fluidity, which is observed following TCDD treatment in mice. CYP1A2 appears to act as an electron sink by accepting electrons generated by CYP1A1 and CYP2E1, preventing the generation of H2O2 and the oxidation of microsomal membrane lipids (Shertzer et al., 2004b).

Mitochondria appear to be the major site for reactive oxygen production (Senft et al., 2002b; Latchoumycandane et al., 2003) and as such may represent a target for TCDD-dependent injury. One proposed mechanism by which TCDD may contribute to increased mitochondrially-derived reactive oxygen is inhibition of electron transport at complex III, producing a persistent increase in succinate-dependent superoxide and hydrogen peroxide production (Senft et al., 2002b). Respiratory chain-derived reactive oxygen can result from a decrease in cytochrome c oxidase (complex IV) activity, coupled with an increase in succinate-cytochrome c reductase (complex II), resulting in an increase in the reduction state of cytochrome bc1 complex (complex III) to facilitate univalent reduction of oxygen (Senft et al., 2002a; Senft et al., 2002b). Mechanistically, TCDD-dependent electron flow through complex III results in increased electron pressure and increase redox cycling of Coenzyme Q or Fe-S proteins. The physiologic relevance of this mechanism however is not well established, since the concentration of TCDD necessary to act in this manner is far greater than would be expected to be found in naturally-occurring exposures.

Following TCDD treatment, and associated with increases in the production of reactive oxygen, both GSH and GSSG increase in the cytosol and in the mitochondria. However, in the mitochondria, GSH increases to a greater extent relative to the cytosol, while GSSG increases to a lesser extent. These differences resulted in shifts in equilibrium for both type 1 (protein mixed disulfides) and type 2 (protein disulfides) thiol-disulfide switches (Schafer et al., 2001). In the cytosol, TCDD produces an increase in oxidation state, with decreases in type 1 and type 2 switches, as well as an increase (more positive) in the reduction potential (ΔE) of GSSG/2GSH. In sharp contrast, mitochondria display an increase in reduction state, with increases in type 1 and type 2 thiol redox switches, as well as a decrease (more negative) in the ΔE of GSSG/2GSH half reaction (Dalton et al., 2004). These results from the authors' labs are consistent with the hypothesis that TCDD mediates an increase in mitochondrial reactive oxygen result from an overall increase in the reduction state of the mitochondria. As such, the mitochondrial generation of reactive oxygen by TCDD may be considered a form of reductive stress, rather that the clearly defined oxidative stress response that occurs in the cytoplasm.

Although TCDD is not genotoxic in the Ames test, one suggested pathway by which it produces toxic effects involves oxidative DNA damage and increased mutation frequency. A strong relationship has been established between oxidative damage to DNA and chemical carcinogenesis (Cairns et al., 1991). Oxidation of DNA at the 8-position deoxyguanosine produces 8-hydroxydeoxyguanosine (8-OHdG), which represents the major promutagenic lesion produced during oxidative stress. When guanosine base modification is followed by DNA replication G→T and A→C transversions can be produced. In addition, reactive oxygen-induced DNA damage activates error-prone polymerase DNA repair that may in turn produce base mispairing (Cairns et al., 1991). Although exonucleases and glycosylases can repair such oxidative DNA damage, the probability of mutation fixation increases with the duration of exposure to a mutagen and with increases in the mitotic rate (Kasai et al., 1986; Cheng et al., 1992; Aronica et al., 1993; Kamiya et al., 1995), which, given the biologic persistence of TCDD, increase the total probability of a mutational event to a level comparable with that of stronger mutagens. An increase in 8-OHdG that persisted 8 weeks after treatment with TCDD was observed in the urine of C57BL/6 mice (Shen et al., 1995; Shertzer et al., 1998) and in the tissue culture medium of hepatoma Hepa-1c1c7 cells treated with TCDD (Park et al., 1996).

Though TCDD has been repeatedly linked to oxidative stress and the oxidative stress response to mutagenesis, TCDD has not been shown to be directly mutagenic in either bacterial or most in vitro assay systems (Giri, 1986). In this regard, an important negative finding has been that, at a dosing regimen capable of producing an oxidative stress response, TCDD did not alter the mutation frequency or the mutation spectrum of the lacI transgene in male or female Big Blue rats (Thornton et al., 2001). Since oxidative stress and some oxidative stress response genes are induced by TCDD in vivo, it can be concluded with caution that TCDD-mediated oxidative damage may not be a prominent cause of mutations. For this reason, an alternate pathway for enhancing cell proliferation and malignant conversion appears likely. Despite the ability of TCDD to generate oxidative base products (Park et al., 1996), the health implications of such findings must be questioned.

8. CONCLUDING REMARKS

In all likelihood, there are several interdependent AHR-dependent pathways that lead to the increased generation of ROS and to the decreased ability to defend against their action. For example, AHR activation may lead to an increase in superoxide production through increased expression of xanthine dehydrogenase/xanthine oxidase and monooxygenases; to an increase in capacity for superoxide reduction through increases in CuZnSOD; and to inhibition of glutathione peroxidases through the generation of J series prostaglandins. The net effect of all these changes would be an increased production of H2O2 and a decreased capacity to detoxify it. Studies aimed at understanding AHR-mediated toxicity have led to the discovery of AHR variants that appear to maintain physiological function and yet confer greatly diminish toxicity; perhaps this is due to the remarkable structural plasticity of the AHR, as shown by work in inbred mouse strains and in rats. The human AHRs thus far studied demonstrate ligand binding affinity characteristics similar to those of the low-affinity mouse strains. This is likely to be an important factor explaining why PHAHs show relatively low toxicity in humans. It is also intriguing to speculate that AHR variants may exist in the human population that confer sensitivity to PHAH pollutants because they behave more like the high-affinity rodent AHR variants. Indeed the studies in the rat and hamster suggest that poorly understood functions of the AHR transactivation domain contribute to toxicity.

One of the greater challenges in Ah receptor research is to identify the connection between toxicity and exposure. While the use of various model systems do serve to elucidate mechanism of TCDD toxicity, it remains difficult to draw broad conclusions given the wide variations in TCDD responses associated with species and strain susceptibilities, exposure models and response endpoints. Are changes in the redox state of cells exposed to PHAHs adaptive or toxic? Are the effects of these changes cumulative? The extent to which the AHR ligands elicit oxidative stress may depend on the duration and nature of the exposure, as well as on the properties of the agonist. At times AHR activation may be so transient that it causes modest and largely unnoticed perturbations to the cellular redox status. At other times or with other AHR ligands, the effects might be much more significant and harmful because of the severity and length of the oxidative stress response. We believe that many of the health consequences resulting from TCDD exposure may likely result from epigenetic mechanisms, including those exerted by cytosolic and mitochondrial reactive oxygen production. In this scenario, resultant toxicity would be related to the non-physiological persistent activation of AHR-dependent signaling pathways due to the long biological half-life of the compound.

ACKNOWLEDGEMENTS

Preparation of this review and the work done in our labs cited herein were supported in part by NIH Grants RO1 ES06273, R01 ES10807, R01 ES10133 and P30 ES06096. J.F.R. is supported by NIEHS Training Grant T32 ES07250.

REFERENCES

  1. Abbott BD, Schmid JE, Pitt JA, Buckalew AR, Wood CR, Held GA, Diliberto JJ. Adverse reproductive outcomes in the transgenic Ah receptor-deficient mouse. Toxicol Appl Pharmacol. 1999;155:62–70. doi: 10.1006/taap.1998.8601. [DOI] [PubMed] [Google Scholar]
  2. Ahmed SS, Napoli KL, Strobel HW. Oxygen radical formation during cytochrome P450-catalyzed cyclosporine metabolism in rat and human liver microsomes at varying hydrogen ion concentrations. Mol Cell Biochem. 1995;151:131–140. doi: 10.1007/BF01322335. [DOI] [PubMed] [Google Scholar]
  3. Alsharif NZ, Lawson T, Stohs SJ. Oxidative stress induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin is mediated by the aryl hydrocarbon (Ah) receptor complex. Toxicology. 1994;92:39–51. doi: 10.1016/0300-483x(94)90166-x. [DOI] [PubMed] [Google Scholar]
  4. Anari MR, Khan S, Jatoe SD, O'Brien PJ. Cytochrome P450 dependent xenobiotic activation by physiological hydroperoxides in intact hepatocytes. Eur J Drug Metab Pharmacokinet. 1997;22:305–310. doi: 10.1007/BF03190961. [DOI] [PubMed] [Google Scholar]
  5. Andersen ME, Barton HA. The use of biochemical and molecular parameters to estimate dose-response relationships at low levels of exposur. Environ Health Perspect. 1998;106(Suppl 1):349–355. doi: 10.1289/ehp.98106s1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Andersen ME, Birnbaum LS, Barton HA, Eklund CR. 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. 1997;144:145–155. doi: 10.1006/taap.1996.8067. [DOI] [PubMed] [Google Scholar]
  7. 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;7:743–752. doi: 10.1210/mend.7.6.7689695. [DOI] [PubMed] [Google Scholar]
  8. Bar-Peled O, Korkotian E, Segal M, Groner Y. Constitutive overexpression of Cu/Zn superoxide dismutase exacerbates kainic acid-induced apoptosis of transgenic-Cu/Zn superoxide dismutase neurons. Proc Natl Acad Sci U S A. 1996;93:8530–8535. doi: 10.1073/pnas.93.16.8530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bell MC, Crowley-Nowick P, Bradlow HL, Sepkovic DW, Schmidt-Grimminger D, Howell P, Mayeaux EJ, Tucker A, Turbat-Herrera EA, Mathis JM. Placebo-controlled trial of indole-3-carbinol in the treatment of CIN. Gynecol Oncol. 2000;78:123–129. doi: 10.1006/gyno.2000.5847. [DOI] [PubMed] [Google Scholar]
  10. Bello SM, Heideman W, Peterson RE. 2,3,7,8-Tetrachlorodibenzo-p-Dioxin Inhibits Regression of the Common Cardinal Vein in Developing Zebrafish. Toxicol Sci. 2004;78:258–266. doi: 10.1093/toxsci/kfh065. [DOI] [PubMed] [Google Scholar]
  11. Bertazzi PA, Zocchetti C, Pesatori AC, Guercilena S, Sanarico M, Radice L. Mortality in an area contaminated by TCDD following an industrial incident. Med Lav. 1989;80:316–329. [PubMed] [Google Scholar]
  12. Bondy SC, Naderi S. Contribution of hepatic cytochrome P450 systems to the generation of reactive oxygen species. Biochem Pharmacol. 1994;48:155–159. doi: 10.1016/0006-2952(94)90235-6. [DOI] [PubMed] [Google Scholar]
  13. Bradlow HL, Davis DL, Lin G, Sepkovic D, Tiwari R. Effects of pesticides on the ratio of 16 alpha/2-hydroxyestrone: a biologic marker of breast cancer risk. Environ Health Perspect. 1995;103(Suppl 7):147–150. doi: 10.1289/ehp.95103s7147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bradlow HL, Hershcopf R, Martucci C, Fishman J. 16α-hydroxylation of estradiol: A possible risk marker for breast cancer. Ann New York Acad Sci. 1986;464:138–151. doi: 10.1111/j.1749-6632.1986.tb16001.x. [DOI] [PubMed] [Google Scholar]
  15. Bradlow HL, Hershcopf RJ, Martucci CP, Fishman J. Estradiol 16 alpha-hydroxylation in the mouse correlates with mammary tumor incidence and presence of murine mammary tumor virus: a possible model for the hormonal etiology of breast cancer in humans. Proc Natl Acad Sci USA. 1985;82:6295–6299. doi: 10.1073/pnas.82.18.6295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bradlow HL, Telang NT, Sepkovic DW, Osborne MP. 2-hydroxyestrone: the ‘good’ estrogen. J Endocrinol. 1996;150(Suppl):S259–S265. [PubMed] [Google Scholar]
  17. Buffinton GD, Ollinger K, Brunmark A, Cadenas E. DT-diaphorase-catalysed reduction of 1,4-naphthoquinone derivatives and glutathionyl-quinone conjugates. Effect of substituents on autoxidation rates. Biochem J. 1989;257:561–571. doi: 10.1042/bj2570561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Butler MA, Lang NP, Young JF, Caporaso NE, Vineis P, Hayes RB, Teitel CH, Massengill JP, Lawsen MF, Kadlubar FF. Determination of CYP1A2 and acetyltransferase phenotype in human populations by analysis of caffeine urinary metabolites. Pharmacogenetics. 1992;2:116–127. doi: 10.1097/00008571-199206000-00003. [DOI] [PubMed] [Google Scholar]
  19. Cairns W, Cairns C, Pongratz I, Poellinger L, Okret S. Assembly of a glucocorticoid receptor complex prior to DNA binding enhances its specific interaction with a glucocorticoid response element. J Biol Chem. 1991;266:11221–11226. [PubMed] [Google Scholar]
  20. Capdevila J, Gil L, Orellana M, Marnett LJ, Mason JI, Yadagiri P, Falck JR. Inhibitors of cytochrome P-450-dependent arachidonic acid metabolism. Arch Biochem Biophys. 1988;261:257–263. doi: 10.1016/0003-9861(88)90340-2. [DOI] [PubMed] [Google Scholar]
  21. Cavalieri E, Frenkel K, Liehr JG, Rogan E, Roy D. Estrogens as endogenous genotoxic agents—DNA adducts and mutations. J Natl Cancer Inst Monogr. 2000:75–93. doi: 10.1093/oxfordjournals.jncimonographs.a024247. [DOI] [PubMed] [Google Scholar]
  22. Chang C, Smith DR, Prasad VS, Sidman CL, Nebert DW, Puga A. Ten nucleotide differences, five of which cause amino acid changes, are associated with the Ah receptor locus polymorphism of C57BL/6 and DBA/2 mice. Pharmacogenetics. 1993;3:312–321. doi: 10.1097/00008571-199312000-00005. [DOI] [PubMed] [Google Scholar]
  23. Chapman DE, Schiller CM. Dose-related effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in C57BL/6J and DBA/2J mice. Toxicol Appl Pharmacol. 1985;78:147–157. doi: 10.1016/0041-008x(85)90314-x. [DOI] [PubMed] [Google Scholar]
  24. Cheng KC, Cahill DS, Kasai H, Nishimura S, Loeb LA. 8-Hydroxyguanine, an abundant form of oxidative DNA damage, causes G->T and A->C substitutions. J Biol Chem. 1992;267:166–172. [PubMed] [Google Scholar]
  25. Coumoul X, Diry M, Robillot C, Barouki R. Differential regulation of cytochrome P450 1A1 and 1B1 by a combination of dioxin and pesticides in the breast tumor cell line MCF-7. Cancer Res. 2001;61:3942–3948. [PubMed] [Google Scholar]
  26. Dai Y, Rashba-Step J, Cederbaum AI. Stable expression of human cytochrome P4502E1 in HepG2 cells: characterization of catalytic activities and production of reactive oxygen intermediates. Biochemistry. 1993;32:6928–6937. doi: 10.1021/bi00078a017. [DOI] [PubMed] [Google Scholar]
  27. Dalton TP, Kerzee JK, Wang B, Miller M, Dieter MZ, Lorenz JN, Shertzer HG, Nerbert DW, Puga A. Dioxin exposure is an environmental risk factor for ischemic heart disease. Cardiovasc Toxicol. 2001;1:285–298. doi: 10.1385/ct:1:4:285. [DOI] [PubMed] [Google Scholar]
  28. Dalton TP, Chen Y, Schneider SN, Nerbert DW, Shertzer HG. Genetically altered mice to evaluate glutathione homeostasis in health and disease. Free Radic Biol Med. 2004;37:1511–1526. doi: 10.1016/j.freeradbiomed.2004.06.040. [DOI] [PubMed] [Google Scholar]
  29. DeVito MJ, Birnbaum LS, Farland WH, Gasiewicz TA. Comparisons of estimated human body burdens of dioxinlike chemicals and TCDD body burdens in experimentally exposed animals. Environ Health Perspect. 1995;103:820–831. doi: 10.1289/ehp.95103820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Diliberto JJ, Burgin D, Birnbaum LS. Role of CYP1A2 in hepatic sequestration of dioxin: Studies using CYP1A2 knock-out mice. Biochem Biophys Res Comm. 1997;236:431–433. doi: 10.1006/bbrc.1997.6973. [DOI] [PubMed] [Google Scholar]
  31. Dong W, Teraoka H, Tsujimoto Y, Stegeman JJ, Hiraga T. Role of aryl hydrocarbon receptor in mesencephalic circulation failure and apoptosis in zebrafish embryos exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Sci. 2004;77:109–116. doi: 10.1093/toxsci/kfh023. [DOI] [PubMed] [Google Scholar]
  32. Dunlap DY, Ikeda I, Nagashima H, Vogel CF, Matsumura F. Effects of src-deficiency on the expression of in vivo toxicity of TCDD in a strain of c-src knockout mice procured through six generations of backcrossings to C57BL/6 mice. Toxicology. 2002;172:125–141. doi: 10.1016/s0300-483x(02)00006-9. [DOI] [PubMed] [Google Scholar]
  33. Ekstrom G, Cronholm T, Ingelman-Sundberg M. Hydroxyl-radical production and ethanol oxidation by liver microsomes isolated from ethanol-treated rats. Biochem J. 1986;233:755–761. doi: 10.1042/bj2330755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Elferink CJ, Ge NL, Levine A. Maximal Aryl Hydrocarbon Receptor Activity Depends on an Interaction with the Retinoblastoma Protein. Mol Pharmacol. 2001;59:664–673. doi: 10.1124/mol.59.4.664. [DOI] [PubMed] [Google Scholar]
  35. Embrechts J, Lemiere F, Van Dongen W, Esmans EL, Buytaert P, Van Marck E, Kockx M, Makar A. Detection of estrogen DNA-adducts in human breast tumor tissue and healthy tissue by combined nano LC-nano ES tandem mass spectrometry. J Am Soc Mass Spectrom. 2003;14:482–491. doi: 10.1016/S1044-0305(03)00130-2. [DOI] [PubMed] [Google Scholar]
  36. Eskenazi B, Wyrobek AJ, Sloter E, Kidd SA, Moore L, Young S, Moore D. The association of age and semen quality in healthy men. Hum Reprod. 2003;18:447–454. doi: 10.1093/humrep/deg107. [DOI] [PubMed] [Google Scholar]
  37. Fernandez-Salguero P, Pineau T, Hilbert DM, McPhail T, Lee SS, Kimura S, Nebert DW, Rudikoff S, Ward JM, Gonzalez FJ. Immune system impairment and hepatic fibrosis in mice lacking the dioxin-binding Ah receptor. Science. 1995;268:722–726. doi: 10.1126/science.7732381. [DOI] [PubMed] [Google Scholar]
  38. Fernandez-Salguero PM, Hilbert DM, Rudikoff S, Ward JM, Gonzalez FJ. Aryl-hydrocarbon receptor-deficient mice are resistant to 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced toxicity. Toxicol Appl Pharmacol. 1996;140:173–179. doi: 10.1006/taap.1996.0210. [DOI] [PubMed] [Google Scholar]
  39. Fernandez-Salguero PM, Ward JM, Sundberg JP, Gonzalez FJ. Lesions of aryl-hydrocarbon receptor-deficient mice. Vet Pathol. 1997;34:605–614. doi: 10.1177/030098589703400609. [DOI] [PubMed] [Google Scholar]
  40. Flesch-Janys D, Berger J, Gurn P, Manz A, Nagel S, Waltsgott H, Dwyer JH. Exposure to polychlorinated dioxins and furans (PCDD/F) and mortality in a cohort of workers from a herbicide-producing plant in Hamburg, Federal Republic of Germany. Am J Epidemiol. 1995;142:1165–1175. doi: 10.1093/oxfordjournals.aje.a117575. [DOI] [PubMed] [Google Scholar]
  41. Franc MA, Pohjanvirta R, Tuomisto J, Okey AB. 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. 2001;62:1565–1578. doi: 10.1016/s0006-2952(01)00820-6. [DOI] [PubMed] [Google Scholar]
  42. Franc MA, Pohjanvirta R, Tuomisto J, Okey AB. Persistent, low-dose 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure: effect on aryl hydrocarbon receptor expression in a dioxin-resistance model. Toxicol Appl Pharmacol. 2001;175:43–53. doi: 10.1006/taap.2001.9222. [DOI] [PubMed] [Google Scholar]
  43. Gatmaitan Z, Lewis S, Turchin H, Arias IM. Premature development of ligandin (GSH transferase B) in mice with an inherited defect in endoplasmic reticulum-golgi structure and function. Biochem Biophys Res Comm. 1977;75:337–337. doi: 10.1016/0006-291x(77)91047-6. [DOI] [PubMed] [Google Scholar]
  44. Ge NL, Elferink CJ. A direct interaction between the aryl hydrocarbon receptor and retinoblastoma protein. Linking dioxin signaling to the cell cycle. J Biol Chem. 1998;273:22708–22713. doi: 10.1074/jbc.273.35.22708. [DOI] [PubMed] [Google Scholar]
  45. Giannone JV, Li W, Probst M, Okey AB. Prolonged depletion of AH receptor without alteration of receptor mRNA levels after treatment of cells in culture with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Biochem Pharmacol. 1998;55:489–497. doi: 10.1016/s0006-2952(97)00493-0. [DOI] [PubMed] [Google Scholar]
  46. Gillette JR, Brodie BB, LaDu BN. The oxidation of drugs by liver microsomes: On the role of TPNH and oxygen. J Pharmacol Exp Ther. 1957;119:532–540. [PubMed] [Google Scholar]
  47. Gillner M, Bergman J, Cambillau C, Fernström B, Gustafsson J. Interactions of indoles with specific binding sites for 2,3,7,8-tetrachlorodibenzo-p-dioxin in rat liver. Mol Pharmacol. 1985;28:357–363. [PubMed] [Google Scholar]
  48. Giri AK. Mutagenic and genotoxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin, a review. Mut Res. 1986;168:241–248. doi: 10.1016/0165-1110(86)90022-9. [DOI] [PubMed] [Google Scholar]
  49. Gonzalez FJ, Nebert DW. Evolution of the P450 gene superfamily: animal-plant ‘warfare’, molecular drive and human genetic differences in drug oxidation. Trends Genet. 1990;6:182–186. doi: 10.1016/0168-9525(90)90174-5. [DOI] [PubMed] [Google Scholar]
  50. Guengerich FP, Lieber DC. Enzymatic activation of chemicals to toxic metabolites. CRC Critical Reviews in Toxicology. 1985;14:259–307. doi: 10.3109/10408448509037460. [DOI] [PubMed] [Google Scholar]
  51. Guiney PD, Walker MK, Spitsbergen JM, Peterson RE. Hemodynamic dysfunction and cytochrome P4501A mRNA expression induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin during embryonic stages of lake trout development. Toxicol Appl Pharmacol. 2000;168:1–14. doi: 10.1006/taap.2000.8999. [DOI] [PubMed] [Google Scholar]
  52. Hammond DK, Zhu BT, Wang MY, Ricci MJ, Liehr JG. Cytochrome P450 metabolism of estradiol in hamster liver and kidney. Toxicol Appl Pharmacol. 1997;145:54–60. doi: 10.1006/taap.1997.8167. [DOI] [PubMed] [Google Scholar]
  53. Han X, Liehr JG. Microsome-mediated 8-hydroxylation of guanine bases of DNA by steroid estrogens: correlation of DNA damage by free radicals with metabolic activation to quinones. Carcinogenesis. 1995;16:2571–2574. doi: 10.1093/carcin/16.10.2571. [DOI] [PubMed] [Google Scholar]
  54. Hankinson O. The aryl hydrocarbon receptor complex. Annu Rev Pharmacol Toxicol. 1995;35:307–340. doi: 10.1146/annurev.pa.35.040195.001515. [DOI] [PubMed] [Google Scholar]
  55. Hassoun EA, Al Ghafri M, Abushaban A. The role of antioxidant enzymes in TCDD-induced oxidative stress in various brain regions of rats after subchronic exposure. Free Radic Biol Med. 2003;35:1028–1036. doi: 10.1016/s0891-5849(03)00458-1. [DOI] [PubMed] [Google Scholar]
  56. Hassoun EA, Wilt SC, DeVito MJ, Van Birgelen A, Alsharif NZ, Birnbaum LS, Stohs SJ. Induction of oxidative stress in brain tissues of mice after subchronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Sci. 1998;42:23–27. doi: 10.1006/toxs.1997.2411. [DOI] [PubMed] [Google Scholar]
  57. Hatae T, Hara S, Yokoyama C, Yabuki T, Inoue H, Ullrich V, Tanabe T. Site-directed mutagenesis of human prostacyclin synthase: Alteration of Cys441 of the Cys-pocket, and Glu347 and Arg350 of the EXXR motif. FEBS Lett. 1996;389:268–272. doi: 10.1016/0014-5793(96)00600-x. [DOI] [PubMed] [Google Scholar]
  58. Hayes CL, Spink DC, Spink BC, Cao JQ, Walker NJ, Sutter TR. 17 beta-estradiol hydroxylation catalyzed by human cytochrome P450 1B1. Proc Natl Acad Sci USA. 1996;93:9776–9781. doi: 10.1073/pnas.93.18.9776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hebert CD, Harris MW, Elwell MR, Birnbaum LS. Relative toxicity and tumor-promoting ability of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), 2,3,4,7,8-pentachlorodibenzofuran (PCDF), and 1,2,3,4,7,8-hexachlorodibenzofuran (HCDF) in hairless mice. Toxicol Appl Pharmacol. 1990;102:362–377. doi: 10.1016/0041-008x(90)90033-q. [DOI] [PubMed] [Google Scholar]
  60. Henck JM, New MA, Kociba RJ, Rao KS. 2,3,7,8-tetrachlorodibenzo-p-dioxin: acute oral toxicity in hamsters. Toxicol Appl Pharmacol. 1981;59:405–407. doi: 10.1016/0041-008x(81)90212-x. [DOI] [PubMed] [Google Scholar]
  61. Henry TR, Spitsbergen JM, Hornung MW, Abnet CC, Peterson RE. Early life stage toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in zebrafish (Danio rerio) Toxicol Appl Pharmacol. 1997;142:56–68. doi: 10.1006/taap.1996.8024. [DOI] [PubMed] [Google Scholar]
  62. Hestermann EV, Stegeman JJ, Hahn ME. Relative contributions of affinity and intrinsic efficacy to aryl hydrocarbon receptor ligand potency. Toxicol Appl Pharmacol. 2000;168:160–172. doi: 10.1006/taap.2000.9026. [DOI] [PubMed] [Google Scholar]
  63. Hodgson AV, Ayala-Torres S, Thompson EB, Liehr JG. Estrogen-induced microsatellite DNA alterations are associated with Syrian hamster kidney tumorigenesis. Carcinogenesis. 1998;19:2169–2172. doi: 10.1093/carcin/19.12.2169. [DOI] [PubMed] [Google Scholar]
  64. Huff J, Lucier G, Tritscher A. Carcinogenicity of TCDD: experimental, mechanistic, and epidemiologic evidence. Annu Rev Pharmacol Toxicol. 1994;34:343–372. doi: 10.1146/annurev.pa.34.040194.002015. [DOI] [PubMed] [Google Scholar]
  65. Huff JE, Salmon AG, Hooper NK, Zeise L. Long-term carcinogenesis studies on 2,3,7,8-tetrachlorodibenzo-p-dioxin and hexachlorodibenzo-p-dioxins. Cell Biol Toxicol. 1991;7:67–94. doi: 10.1007/BF00121331. [DOI] [PubMed] [Google Scholar]
  66. Jana NR, Sarkar S, Ishizuka M, Yonemoto J, Tohyama C, Sone H. Comparative effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on MCF-7, RL95-2, and LNCaP cells: role of target steroid hormones in cellular responsiveness to CYP1A1 induction. Mol Cell Biol Res Commun. 2000;4:174–180. doi: 10.1006/mcbr.2001.0275. [DOI] [PubMed] [Google Scholar]
  67. Jefcoate CR, Liehr JG, Santen RJ, Sutter TR, Yager JD, Yue W, Santner SJ, Tekmal R, Demers L, Pauley R, Naftolin F, Mor G, Berstein L. Tissue-specific synthesis and oxidative metabolism of estrogens. J Natl Cancer Inst Monogr. 2000:95–112. doi: 10.1093/oxfordjournals.jncimonographs.a024248. [DOI] [PubMed] [Google Scholar]
  68. Jiang CK, Epstein HS, Tomic M, Freedberg IM, Blumenberg M. Functional comparison of the upstream regulatory DNA sequences of four human epidermal keratin gene. J Invest Dermatol. 1991;96:162–167. doi: 10.1111/1523-1747.ep12460939. [DOI] [PubMed] [Google Scholar]
  69. Jokinen MP, Walker NJ, Brix AE, Sells DM, Haseman JK, Nyska A. Increase in cardiovascular pathology in female sprague-dawley rats following chronic treatment with 2,3,7,8-tetrachlorodibenzop- dioxin and 3,3′,4,4′,5-pentachlorobiphenyl. Cardiovasc Toxicol. 2003;3:299–310. doi: 10.1385/CT:3:4:299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Kamiya H, Miura H, Murata-Kamiya N, Ishikawa H, Sakaguchi T, Inoue H, Sasaki T, Masutani C, Hanaoka F, Nishimura S. 8-Hydroxyadenine (7,8-dihydro-8-oxoadenine) induces mis-incorporation in in vitro DNA synthesis and mutations in NIH 3T3 cells. Nucleic Acids Res. 1995;23:2893–2899. doi: 10.1093/nar/23.15.2893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Karyala S, Guo J, Sartor M, Medvedovic M, Kann S, Puga A, Ryan P, Tomlinson CR. Different Global Gene Expression Profiles in Benzo[a]Pyrene- and Dioxin-Treated Vascular Smooth Muscle Cells of AHR-Knockout and Wild-Type Mice. Cardiovasc Toxicol. 2004;4:47–74. doi: 10.1385/ct:4:1:47. [DOI] [PubMed] [Google Scholar]
  72. Kasai H, Crain PF, Kuchino Y, Nishimura S, Ootsuyama A, Tanooka H. Formation of 8-hydroxyguanine moiety in cellular DNA by agents producing oxygen radicals and evidence for its repair. Carcinogenesis. 1986;7:1849–1851. doi: 10.1093/carcin/7.11.1849. [DOI] [PubMed] [Google Scholar]
  73. Kociba RJ, Keyes DG, Beyer JE, Carreon RM, Wade CE, Dittenber DA, Kalnins RP, Frauson LE, Park CN, Barnard SD, Hummel RA, Humiston CG. Results of a two-year chronic toxicity and oncogenicity study of 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats. Toxicol Appl Pharmacol. 1978;46:279–303. doi: 10.1016/0041-008x(78)90075-3. [DOI] [PubMed] [Google Scholar]
  74. Kolluri SK, Weiss C, Koff A, Gottlicher M. p27(Kip1) induction and inhibition of proliferation by the intracellular Ah receptor in developing thymus and hepatoma cells. Genes Dev. 1999;13:1742–1753. doi: 10.1101/gad.13.13.1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Korkalainen M, Tuomisto J, Pohjanvirta R. Primary structure and inducibility by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) of aryl hydrocarbon receptor repressor in a TCDD-sensitive and a TCDD-resistant rat strain. Biochem Biophys Res Commun. 2004;315:123–131. doi: 10.1016/j.bbrc.2004.01.028. [DOI] [PubMed] [Google Scholar]
  76. Lahvis GP, Lindell SL, Thomas RS, McCuskey RS, Murphy C, Glover E, Bentz M, Southard J, Bradfield CA. Portosystemic shunting and persistent fetal vascular structures in aryl hydrocarbon receptor-deficient mice. Proc Natl Acad Sci USA. 2000;97:10442–10447. doi: 10.1073/pnas.190256997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Lai KP, Wong MH, Wong CK. Modulation of AhR-mediated CYP1A1 mRNA and EROD activities by 17beta-estradiol and dexamethasone in TCDD-induced H411E cells. Toxicol Sci. 2004;78:41–49. doi: 10.1093/toxsci/kfh045. [DOI] [PubMed] [Google Scholar]
  78. Latchoumycandane C, Chitra KC, Mathur PP. 2,3,7,8-tetrachlorodibenzop- dioxin (TCDD) induces oxidative stress in the epididymis and epididymal sperm of adult rats. Arch Toxicol. 2003;77:280–284. doi: 10.1007/s00204-003-0439-x. [DOI] [PubMed] [Google Scholar]
  79. Lee YS, Jin DQ, Park SH, Han SY, Kim HS, Jeong TC, Huh K, Kim JA. 2,3,7,8-tetrachlorobenzo-p-dioxin inhibits proliferation of SK-N-SH human neuronal cells through decreased production of reactive oxygen species. Free Radic Res. 2002;36:1283–1289. doi: 10.1080/1071576021000016517. [DOI] [PubMed] [Google Scholar]
  80. Liehr JG. Hormone-associated cancer: mechanistic similarities between human breast cancer and estrogen-induced kidney carcinogenesis in hamsters. Environ Health Perspect. 1997;105(Suppl 3):565–569. doi: 10.1289/ehp.97105s3565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Liehr JG. 4-hydroxylation of oestrogens as a marker for mammary tumours. Biochem Soc Trans. 1999;27:318–323. doi: 10.1042/bst0270318. [DOI] [PubMed] [Google Scholar]
  82. Liehr JG. Role of DNA adducts in hormonal carcinogenesis. Regul Toxicol Pharmacol. 2000;32:276–282. doi: 10.1006/rtph.2000.1432. [DOI] [PubMed] [Google Scholar]
  83. Liehr JG. Genotoxicity of the steroidal oestrogens oestrone and oestradiol: possible mechanism of uterine and mammary cancer development. Hum Reprod Update. 2001;7:273–281. doi: 10.1093/humupd/7.3.273. [DOI] [PubMed] [Google Scholar]
  84. Liehr JG, Jones JS. Role of iron in estrogen-induced cancer. Curr Med Chem. 2001;8:839–849. doi: 10.2174/0929867013372931. [DOI] [PubMed] [Google Scholar]
  85. Lucier GW, Tritscher A, Goldsworthy T, Foley J, Clark G, Goldstein J, Maronpot R. 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. 1991;51:1391–1397. [PubMed] [Google Scholar]
  86. Maier A, Micka J, Miller K, Denko T, Chang C-Y, Nebert DW, Puga A. Aromatic hydrocarbon receptor (AHR) polymorphism: development of new methods to correlate genotype with phenotype. Environ Health Perspect. 1998;106:421–426. doi: 10.1289/ehp.106-1533118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Malloy VL, Bradlow HL, Orentreich N. Interaction between a semisynthetic diet and indole-3-carbinol on mammary tumor incidence in Balb/cfC3H mice. Anticancer Res. 1997;17:4333–4337. [PubMed] [Google Scholar]
  88. Marlowe JL, Knudsen ES, Schwemberger S, and Puga A. 2004. The aryl hydrocarbon receptor displaces p300 from E2F-dependent promoters and represses S-phase specific gene expression. J Biol Chem E-Pub. [DOI] [PubMed]
  89. Michnovicz JJ, Adlercreutz H, Bradlow HL. Changes in levels of urinary estrogen metabolites after oral indole-3- carbinol treatment in humans. J Natl Cancer Inst. 1997;89:718–723. doi: 10.1093/jnci/89.10.718. [DOI] [PubMed] [Google Scholar]
  90. Mimura J, Yamashita K, Nakamura K, Morita M, Takagi TN, Nakao K, Ema M, Sogawa K, Yasuda M, Katsuki M, Fujii-Kuriyama Y. Loss of teratogenic response to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in mice lacking the Ah (dioxin) receptor. Genes Cells. 1997;2:645–654. doi: 10.1046/j.1365-2443.1997.1490345.x. [DOI] [PubMed] [Google Scholar]
  91. Mohammadpour H, Murray WJ, Stohs SJ. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD)-induced lipid peroxidation in genetically responsive and non-responsive mice. Arch Environ Contam Toxicol. 1988;17:645–650. doi: 10.1007/BF01055833. [DOI] [PubMed] [Google Scholar]
  92. Moriguchi T, Motohashi H, Hosoya T, Nakajima O, Takahashi S, Ohsako S, Aoki Y, Nishimura N, Tohyama C, Fujii-Kuriyama Y, Yamamoto M. Distinct response to dioxin in an arylhydrocarbon receptor (AHR)-humanized mouse. Proc Natl Acad Sci U S A. 2003;100:5652–5657. doi: 10.1073/pnas.1037886100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Nakai K, Ward AM, Gannon M, Rifkind AB. -Naphthoflavone induction of a cytochrome P-450 arachidonic acid epoxygenase in chick embryo liver distinct from the aryl hydrocarbon hydroxylase and from phenobarbital-induced arachidonate epoxygenase. J Biol Chem. 1992;267:19503–19512. [PubMed] [Google Scholar]
  94. Nebert DW. The Ah locus: genetic differences in toxicity, cancer, mutation, and birth defects. Crit Rev Toxicol. 1989;20:153–174. doi: 10.3109/10408448909017908. [DOI] [PubMed] [Google Scholar]
  95. Nebert DW. Elevated estrogen 16 alpha-hydroxylase activity: is this a genotoxic or nongenotoxic biomarker in human breast cancer risk? Journal National Cancer Institute. 1993;85:1888–1891. doi: 10.1093/jnci/85.23.1888. [DOI] [PubMed] [Google Scholar]
  96. Nebert DW, Dalton TP, Okey AB, and Gonzalez FJ. 2004. Role of aryl hydrocarbon receptor-mediated induction of the CYP1 enzymes in environmental toxicity and cancer. J Biol Chem E-Pub. [DOI] [PubMed]
  97. Nebert DW, Puga A, Vasiliou V. Role of the Ah receptor and the dioxin-inducible [Ah] gene battery in toxicity, cancer, and signal transduction. Ann N Y Acad Sci. 1993;685:624–640. doi: 10.1111/j.1749-6632.1993.tb35928.x. [DOI] [PubMed] [Google Scholar]
  98. Nebert DW, Roe AL, Dieter MZ, Solis WA, Yang Y, Dalton TP. Role of the aromatic hydrocarbon receptor and [Ah] gene battery in the oxidative stress response, cell cycle control, and apoptosis. Biochem Pharmacol. 2000;59:65–85. doi: 10.1016/s0006-2952(99)00310-x. [DOI] [PubMed] [Google Scholar]
  99. Niittynen M, Tuomisto JT, Auriola S, Pohjanvirta R, Syrjala P, Simanainen U, Viluksela M, Tuomisto J. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced accumulation of biliverdin and hepatic peliosis in rats. Toxicol Sci. 2003;71:112–123. doi: 10.1093/toxsci/71.1.112. [DOI] [PubMed] [Google Scholar]
  100. Okey AB, Vella LM, Harper PA. Detection and characterization of a low affinity form of cytosolic Ah receptor in livers of mice nonresponsive to induction of cytochrome P1-450 by 3-methylcholanthrene. Mol Pharmacol. 1989;35:823–830. [PubMed] [Google Scholar]
  101. Pantopoulos K, Hentze MW. Rapid responses to oxidative stress mediated by iron regulatory protein. EMBO J. 1995;14:2917–2924. doi: 10.1002/j.1460-2075.1995.tb07291.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Park JY, Shigenaga MK, Ames BN. Induction of cytochrome P4501A1 by 2,3,7,8- tetrachlorodibenzop- dioxin or indolo(3,2-b)carbazole is associated with oxidative DNA damage. Proc Natl Acad Sci USA. 1996;19:2322–2327. doi: 10.1073/pnas.93.6.2322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Patandin S, Koopman-Esseboom C, de Ridder MA, Weisglas-Kuperus N, Sauer PJ. Effects of environmental exposure to polychlorinated biphenyls and dioxins on birth size and growth in Dutch children. Pediatr Res. 1998;44:538–545. doi: 10.1203/00006450-199810000-00012. [DOI] [PubMed] [Google Scholar]
  104. Pelclova D, Fenclova Z, Preiss J, Prochazka B, Spacil J, Dubska Z, Okrouhlik B, Lukas E, Urban P. Lipid metabolism and neuropsychological follow-up study of workers exposed to 2,3,7,8- tetrachlordibenzop- dioxin. Int Arch Occup Environ Health. 2002;75(Suppl):S60–S66. doi: 10.1007/s00420-002-0350-4. [DOI] [PubMed] [Google Scholar]
  105. Peled-Kamar M, Lotem J, Okon E, Sachs L, Groner Y. Thymic abnormalities and enhanced apoptosis of thymocytes and bone marrow cells in transgenic mice overexpressing Cu/Zn-superoxide dismutase: implications for Down syndrome. EMBO J. 1995;14:4985–4993. doi: 10.1002/j.1460-2075.1995.tb00181.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Pesatori AC, Consonni D, Bachetti S, Zocchetti C, Bonzini M, Baccarelli A, Bertazzi PA. Short- and long-term morbidity and mortality in the population exposed to dioxin after the “Seveso accident”. Ind Health. 2003;41:127–138. doi: 10.2486/indhealth.41.127. [DOI] [PubMed] [Google Scholar]
  107. Pesatori AC, Zocchetti C, Guercilena S, Consonni D, Turrini D, Bertazzi PA. Dioxin exposure and non-malignant health effects: a mortality study. Occup Environ Med. 1998;55:126–131. doi: 10.1136/oem.55.2.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Peters JM, Narotsky MG, Elizondo G, Fernandez-Salguero PM, Gonzalez FJ, Abbott BD. Amelioration of TCDD-induced teratogenesis in aryl hydrocarbon receptor (AhR)-null mice. Toxicol Sci. 1999;47:86–92. doi: 10.1093/toxsci/47.1.86. [DOI] [PubMed] [Google Scholar]
  109. Peters JM, Narotsky MG, Elizondo G, Fernandez-Salguero PM, Gonzalez FJ, Abbott BD. Amelioration of TCDD-induced teratogenesis in aryl hydrocarbon receptor (AhR)-null mice. Toxicol Sci. 1999;47:86–92. doi: 10.1093/toxsci/47.1.86. [DOI] [PubMed] [Google Scholar]
  110. Pohjanvirta R, Juvonen R, Karenlampi S, Raunio H, Tuomisto J. Hepatic Ah-receptor levels and the effect of 2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD) on hepatic microsomal monooxygenase activities in a TCDD-susceptible and -resistant rat strain. Toxicol Appl Pharmacol. 1988;92:131–140. doi: 10.1016/0041-008x(88)90235-9. [DOI] [PubMed] [Google Scholar]
  111. Pohjanvirta R, Tuomisto J. Short-term toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in laboratory animals: effects, mechanisms, and animal models. Pharmacol Rev. 1994;46:483–549. [PubMed] [Google Scholar]
  112. Pohjanvirta R, Tuomisto J. Short-term toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in laboratory animals: effects, mechanisms, and animal models. Pharmacol Rev. 1994;46:483–549. [PubMed] [Google Scholar]
  113. Pohjanvirta R, Viluksela M, Tuomisto JT, Unkila M, Karasinska J, Franc MA, Holowenko M, Giannone JV, Harper PA, Tuomisto J, Okey AB. Physicochemical differences in the AH receptors of the most TCDD-susceptible and the most TCDD-resistant rat strains. Toxicol Appl Pharmacol. 1999;155:82–95. doi: 10.1006/taap.1998.8565. [DOI] [PubMed] [Google Scholar]
  114. Pohjanvirta R, Wong JM, Li W, Harper PA, Tuomisto J, Okey AB. Point mutation in intron sequence causes altered carboxyl-terminal structure in the aryl hydrocarbon receptor of the most 2,3,7,8-tetrachlorodibenzo-p-dioxin-resistant rat strain. Mol Pharmacol. 1998;54:86–93. doi: 10.1124/mol.54.1.86. [DOI] [PubMed] [Google Scholar]
  115. Poland A, Glover E, Kende AS. Stereospecific, high affinity binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin by hepatic cytosol. Evidence that the binding species is receptor for induction of aryl hydrocarbon hydroxylase. J Biol Chem. 1976;251:4936–4946. [PubMed] [Google Scholar]
  116. Poland A, Glover E, Kende AS. Stereospecific, high affinity binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin by hepatic cytosol. Evidence that the binding species is receptor for induction of aryl hydrocarbon hydroxylase. J Biol Chem. 1976;251:4936–4946. [PubMed] [Google Scholar]
  117. Poland A, Knutson JC. 2,3,7,8,-tetrachlorodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: examination of the mechanisms of toxicity. Annu Rev Pharmacol Toxicol. 1982;22:517–554. doi: 10.1146/annurev.pa.22.040182.002505. [DOI] [PubMed] [Google Scholar]
  118. Poland A, Palen D, Glover E. Analysis of the four alleles of the murine aryl hydrocarbon receptor. Mol Pharmacol. 1994;46:915–921. [PubMed] [Google Scholar]
  119. Pollenz RS. The mechanism of AH receptor protein down-regulation (degradation) and its impact on AH receptor-mediated gene regulation. Chem Biol Interact. 2002;141:41–61. doi: 10.1016/s0009-2797(02)00065-0. [DOI] [PubMed] [Google Scholar]
  120. Pollenz RS, Santostefano MJ, Klett E, Richardson VM, Necela B, Birnbaum LS. Female Sprague-Dawley rats exposed to a single oral dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin exhibit sustained depletion of aryl hydrocarbon receptor protein in liver, spleen, thymus, and lung. Toxicol Sci. 1998;42:117–128. doi: 10.1006/toxs.1998.2439. [DOI] [PubMed] [Google Scholar]
  121. Poulos TL, Raag R. Cytochrome P450cam: crystallography, oxygen activation, and electron transfer. FASEB J. 1992;6:674–679. doi: 10.1096/fasebj.6.2.1537455. [DOI] [PubMed] [Google Scholar]
  122. Puga A, Barnes SJ, Dalton TP, Chang C, Knudsen ES, Maier MA. Aromatic hydrocarbon receptor interaction with the retinoblastoma protein potentiates repression of E2F-dependent transcription and cell cycle arrest. J Biol Chem. 2000;275:2943–2950. doi: 10.1074/jbc.275.4.2943. [DOI] [PubMed] [Google Scholar]
  123. Puga A, Maier A, Medvedovic M. The transcriptional signature of dioxin in human hepatoma HepG2 cells. Biochem Pharmacol. 2000;60:1129–1142. doi: 10.1016/s0006-2952(00)00403-2. [DOI] [PubMed] [Google Scholar]
  124. Puga A, Xia Y, Elferink C. Role of the aryl hydrocarbon receptor in cell cycle regulation. Chem Biol Interact. 2002;141:117–130. doi: 10.1016/s0009-2797(02)00069-8. [DOI] [PubMed] [Google Scholar]
  125. Ralston SL, Lau HHS, Seidel A, Luch A, Platt KL, Baird WM. The potent carcinogen dibenzo[a,l]pyrene is metabolically activated to fjord-region 11,12-diol 13,14-epoxides in human mammary carcinoma MCF-7 cell cultures. Cancer Res. 1994;54:887–890. [PubMed] [Google Scholar]
  126. Ramadoss P, Petrulis JR, Hollingshead BD, Kusnadi A, Perdew GH. Divergent roles of hepatitis B virus X-associated protein 2 (XAP2) in human versus mouse Ah receptor complexes. Biochemistry. 2004;43:700–709. doi: 10.1021/bi035827v. [DOI] [PubMed] [Google Scholar]
  127. Revich B, Aksel E, Ushakova T, Ivanova I, Zhuchenko N, Klyuev N, Brodsky B, Sotskov Y. Dioxin exposure and public health in Chapaevsk, Russia. Chemosphere. 2001;43:951–966. doi: 10.1016/s0045-6535(00)00456-2. [DOI] [PubMed] [Google Scholar]
  128. Rifkind AB, Gannon M, Gross SS. Arachidonic metabolism by dioxin-induced cytochrome P-450: a new hypothesis on the role of P-450 in dioxin toxicity. Biochem Biophys Res Comm. 1990;172:1180–1188. doi: 10.1016/0006-291x(90)91573-b. [DOI] [PubMed] [Google Scholar]
  129. Rosen CA, Woodson GE, Thompson JW, Hengesteg AP, Bradlow HL. Preliminary results of the use of indole-3- carbinol for recurrent respiratory papillomatosis. Otolaryngol Head Neck Surg. 1998;118:810–815. doi: 10.1016/S0194-5998(98)70274-8. [DOI] [PubMed] [Google Scholar]
  130. Roy D, Bernhardt A, Strobel HW, Liehr JG. Catalysis of the oxidation of steroid and stilbene estrogens to estrogen quinone metabolites by the beta-naphthoflavone-inducible cytochrome P450 IA family. Arch Biochem Biophys. 1992;296:450–456. doi: 10.1016/0003-9861(92)90596-o. [DOI] [PubMed] [Google Scholar]
  131. Roy D, Strobel HW, Liehr JG. Cytochrome b5-mediated redox cycling of estrogen. Arch Biochem Biophys. 1991;285:331–338. doi: 10.1016/0003-9861(91)90368-s. [DOI] [PubMed] [Google Scholar]
  132. Rylander L, Hagmar L. Medical and psychometric examinations of conscripts born to mothers with a high intake of fish contaminated with persistent organochlorines. Scand J Work Environ Health. 2000;26:207–212. doi: 10.5271/sjweh.533. [DOI] [PubMed] [Google Scholar]
  133. Santostefano MJ, Wang X, Richardson VM, Ross DG, DeVito MJ, Birnbaum LS. A pharmacodynamic analysis of TCDD-induced cytochrome P450 gene expression in multiple tissues: Dose- and time-dependent effects. Toxicol Appl Pharmacol. 1998;151:294–310. doi: 10.1006/taap.1998.8466. [DOI] [PubMed] [Google Scholar]
  134. Sawyer DE, Van Houten B. Repair of DNA damage in mitochondria. Mutat Res. 1999;434:161–176. doi: 10.1016/s0921-8777(99)00027-0. [DOI] [PubMed] [Google Scholar]
  135. Schafer FQ, Buettner GR. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med. 2001;30:1191–1212. doi: 10.1016/s0891-5849(01)00480-4. [DOI] [PubMed] [Google Scholar]
  136. Schaldach CM, Riby J, Bjeldanes LF. Lipoxin A4: a new class of ligand for the Ah receptor. Biochemistry. 1999;38:7594–7600. doi: 10.1021/bi982861e. [DOI] [PubMed] [Google Scholar]
  137. Schlezinger JJ, Stegeman JJ. Induction and suppression of cytochrome P450 1A by 3,3′,4,4′,5-pentachlorobiphenyl and its relationship to oxidative stress in the marine fish scup (Stenotomus chrysops) Aquat Toxicol. 2001;52:101–115. doi: 10.1016/s0166-445x(00)00141-7. [DOI] [PubMed] [Google Scholar]
  138. Schlezinger JJ, White RD, Stegeman JJ. Oxidative inactivation of cytochrome P-450 1A (CYP1A) stimulated by 3,3′,4,4′-tetrachlorobiphenyl: production of reactive oxygen by vertebrate CYP1As. Mol Pharmacol. 1999;56:588–597. doi: 10.1124/mol.56.3.588. [DOI] [PubMed] [Google Scholar]
  139. Schmidt JV, Su GH-T, Reddy JK, Simon MC, Bradfield CA. Characterization of a murine Ahr null allele: involvement of the Ah receptor in hepatic growth and development. Proc Natl Acad Sci USA. 1996;93:6731–6736. doi: 10.1073/pnas.93.13.6731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Senft AP, Dalton TP, Nebert DW, Genter MB, Hutchinson RJ, Shertzer HG. Dioxin increases reactive oxygen production in mouse liver mitochondria. Toxicol Appl Pharmacol. 2002;178:15–21. doi: 10.1006/taap.2001.9314. [DOI] [PubMed] [Google Scholar]
  141. Senft AP, Dalton TP, Nebert DW, Genter MB, Puga A, Hutchinson RJ, Kerzee JK, Uno S, Shertzer HG. Mitochondrial reactive oxygen production is dependent on the aromatic hydrocarbon receptor. Free Radic Biol Med. 2002;33:1268–1278. doi: 10.1016/s0891-5849(02)01014-6. [DOI] [PubMed] [Google Scholar]
  142. Shen H-M, Ong C-N, Lee B-L, Shi C-Y. Aflatoxin B{-1}-induced 8-hydroxydeoxyguanosine formation in rat hepatic DNA. Carcinogenesis. 1995;16:419–422. doi: 10.1093/carcin/16.2.419. [DOI] [PubMed] [Google Scholar]
  143. Shertzer HG, Clay CD, Genter MB, Chames MC, Schneider SN, Oakley GG, Nebert DW, Dalton TP. Uncoupling-mediated generation of reactive oxygen by halogenated aromatic hydrocarbons in mouse liver microsomes. Free Radic Biol Med. 2004;36:618–631. doi: 10.1016/j.freeradbiomed.2003.11.014. [DOI] [PubMed] [Google Scholar]
  144. Shertzer HG, Clay CD, Genter MB, Schneider SN, Nebert DW, Dalton TP. Cyp1a2 protects against reactive oxygen production in mouse liver microsomes. Free Radic Biol Med. 2004;36:605–617. doi: 10.1016/j.freeradbiomed.2003.11.013. [DOI] [PubMed] [Google Scholar]
  145. Shertzer HG, Nebert DW, Puga A, Ary M, Sonntag D, Dixon K, Robinson LJ, Cianciolo E, Dalton TP. Dioxin causes a sustained oxidative stress response in the mouse. Biochem Biophys Res Comm. 1998;253:44–48. doi: 10.1006/bbrc.1998.9753. [DOI] [PubMed] [Google Scholar]
  146. Shertzer HG, Senft AP. The micronutrient indole-3- carbinol: implications for disease and chemoprevention. Drug Metabol Drug Interact. 2000;17:159–188. doi: 10.1515/dmdi.2000.17.1-4.159. [DOI] [PubMed] [Google Scholar]
  147. Simanainen U, Tuomisto JT, Tuomisto J, Viluksela M. Dose-response analysis of short-term effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin in three differentially susceptible rat lines. Toxicol Appl Pharmacol. 2003;187:128–136. doi: 10.1016/s0041-008x(02)00068-6. [DOI] [PubMed] [Google Scholar]
  148. Slezak BP, Hatch GE, DeVito MJ, Diliberto JJ, Slade R, Crissman K, Hassoun E, Birnbaum LS. Oxidative stress in female B6C3F1 mice following acute and subchronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) Toxicol Sci. 2000;54:390–398. doi: 10.1093/toxsci/54.2.390. [DOI] [PubMed] [Google Scholar]
  149. Slim R, Toborek M, Robertson LW, Lehmler HJ, Hennig B. Cellular glutathione status modulates polychlorinated biphenyl-induced stress response and apoptosis in vascular endothelial cells. Toxicol Appl Pharmacol. 2000;166:36–42. doi: 10.1006/taap.2000.8944. [DOI] [PubMed] [Google Scholar]
  150. Sloop TC, Lucier GW. Dose-dependent elevation of Ah receptor binding by TCDD in rat liver 1. Toxicol Appl Pharmacol. 1987;88:329–337. doi: 10.1016/0041-008x(87)90208-0. [DOI] [PubMed] [Google Scholar]
  151. Smith AG, Clothier B, Carthew P, Childs NL, Sinclair PR, Nebert DW, Dalton TP. Protection of the Cyp1a2(−/−) null mouse against uroporphyria and hepatic injury following exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol. 2001;173:89–98. doi: 10.1006/taap.2001.9167. [DOI] [PubMed] [Google Scholar]
  152. Smith AG, Clothier B, Robinson S, Scullion MJ, Carthew P, Edwards R, Luo J, Lim CK, Toledano M. Interaction between iron metabolism and 2,3,7,8-tetrachlorodibenzo-p-dioxin in mice with variants of the Ahr gene: a hepatic oxidative mechanism. Mol Pharmacol. 1998;53:52–61. doi: 10.1124/mol.53.1.52. [DOI] [PubMed] [Google Scholar]
  153. Spink DC, Hayes CL, Young NR, Christou M, Sutter TR, Jefcoate CR, Gierthy JF. The effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on estrogen metabolism in MCF-7 breast cancer cells: evidence for induction of a novel 17 beta-estradiol 4-hydroxylase. J Steroid Biochem Mol Biol. 1994;51:251–258. doi: 10.1016/0960-0760(94)90037-x. [DOI] [PubMed] [Google Scholar]
  154. Spink DC, Spink BC, Cao JQ, DePasquale JA, Pentecost BT, Fasco MJ, Li Y, Sutter TR. Differential expression of CYP1A1 and CYP1B1 in human breast epithelial cells and breast tumor cells. Carcinogenesis. 1998;19:291–298. doi: 10.1093/carcin/19.2.291. [DOI] [PubMed] [Google Scholar]
  155. Stockbauer JW, Hoffman RE, Schramm WF, Edmonds LD. Reproductive outcomes of mothers with potential exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Am J Epidemiol. 1988;128:410–419. doi: 10.1093/oxfordjournals.aje.a114981. [DOI] [PubMed] [Google Scholar]
  156. Stohs SJ. Oxidative stress induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) Free Radic Biol Med. 1990;9:79–90. doi: 10.1016/0891-5849(90)90052-k. [DOI] [PubMed] [Google Scholar]
  157. Svensson BG, Nilsson A, Hansson M, Rappe C, Akesson B, Skerfving S. Exposure to dioxins and dibenzofurans through the consumption of fish. N Engl J Med. 1991;324:8–12. doi: 10.1056/NEJM199101033240102. [DOI] [PubMed] [Google Scholar]
  158. Swanson HI, Bradfield CA. The AH-receptor: genetics, structure and function. Pharmacogenetics. 1993;3:213–230. doi: 10.1097/00008571-199310000-00001. [DOI] [PubMed] [Google Scholar]
  159. Talalay P, De Long MJ, Prochaska HJ. Identification of a common chemical signal regulating the induction of enzymes that protect against chemical carcinogenesis. Proc Natl Acad Sci USA. 1988;85:8261–8265. doi: 10.1073/pnas.85.21.8261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Telang NT, Katdare M, Bradlow HL, Osborne MP, Fishman J. Inhibition of proliferation and modulation of estradiol metabolism: novel mechanisms for breast cancer prevention by the phytochemical indole-3- carbinol. Proc Soc Exp Biol Med. 1997;216:246–252. doi: 10.3181/00379727-216-44174. [DOI] [PubMed] [Google Scholar]
  161. Telang NT, Suto A, Wong GY, Osborne MP, Bradlow HL. Induction by estrogen metabolite 16α-hydroxyestrone of genotoxic damage and aberrant proliferation in mouse mammary epithelial cells. Journal of the National Cancer Institute. 1992;84:634–638. doi: 10.1093/jnci/84.8.634. [DOI] [PubMed] [Google Scholar]
  162. Thornton AS, Oda Y, Stuart GR, Glickman BW, de Boer JG. Mutagenicity of TCDD in Big Blue transgenic rats. Mutat Res. 2001;478:45–50. doi: 10.1016/s0027-5107(01)00105-1. [DOI] [PubMed] [Google Scholar]
  163. Tian Y, Ke S, Denison MS, Rabson AB, Gallo MA. Ah receptor and NF-kappaB interactions, a potential mechanism for dioxin toxicity. J Biol Chem. 1999;274:510–515. doi: 10.1074/jbc.274.1.510. [DOI] [PubMed] [Google Scholar]
  164. Tritscher AM, Seacat AM, Yager JD, Groopman JD, Miller BD, Bell D, Sutter TR, Lucier GW. Increased oxidative DNA damage in livers of 2,3,7,8-tetrachlorodibenzo-p-dioxin treated intact but not ovariectomized rats. Cancer Letters. 1996;98:219–225. [PubMed] [Google Scholar]
  165. Tuomisto JT, Viluksela M, Pohjanvirta R, Tuomisto J. The AH receptor and a novel gene determine acute toxic responses to TCDD: segregation of the resistant alleles to different rat lines. Toxicol Appl Pharmacol. 1999;155:71–81. doi: 10.1006/taap.1998.8564. [DOI] [PubMed] [Google Scholar]
  166. Unkila M, Tuomisto JT, Pohjanvirta R, MacDonald E, Tuomisto L, Koulu M, Tuomisto J. Effect of a single lethal dose of TCDD on the levels of monoamines, their metabolites and tryptophan in discrete brain nuclei and peripheral tissues of Long-Evans rats. Pharmacol Toxicol. 1993;72:279–285. doi: 10.1111/j.1600-0773.1993.tb01650.x. [DOI] [PubMed] [Google Scholar]
  167. Uno S, Dalton TP, Sinclair PR, Gorman N, Wang B, Smith AG, Miller ML, Shertzer HG, Nebert DW. Cyp1a1(−/−) male mice: protection against high-dose TCDD-induced lethality and wasting syndrome, and resistance to intrahepatocyte lipid accumulation and uroporphyria. Toxicol Appl Pharmacol. 2004;196:410–421. doi: 10.1016/j.taap.2004.01.014. [DOI] [PubMed] [Google Scholar]
  168. Vartiainen T, Jaakkola JJ, Saarikoski S, Tuomisto J. Birth weight and sex of children and the correlation to the body burden of PCDDs/PCDFs and PCBs of the mother. Environ Health Perspect. 1998;106:61–66. doi: 10.1289/ehp.9810661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Vena J, Boffetta P, Becher H, Benn T, Bueno-de-Mesquita HB, Coggon D, Colin D, Flesch-Janys D, Green L, Kauppinen T, Littorin M, Lynge E, Mathews JD, Neuberger M, Pearce N, Pesatori AC, Saracci R, Steenland K, Kogevinas M. Exposure to dioxin and nonneoplastic mortality in the expanded IARC international cohort study of phenoxy herbicide and chlorophenol production workers and sprayers. Environ Health Perspect. 1998;106(Suppl 2):645–653. doi: 10.1289/ehp.98106645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Walker NJ, Portier CJ, Lax SF, Crofts FG, Li Y, Lucier GW, Sutter TR. 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. 1999;154:279–286. doi: 10.1006/taap.1998.8595. [DOI] [PubMed] [Google Scholar]
  171. Wang X, Santostefano MJ, DeVito MJ, Birnbaum LS. Extrapolation of a previous PBPK model for TCDD across routes of exposure, gender, and from rats to mice. Toxicological Sciences. 1997:38–42. [Google Scholar]
  172. Wang X, Santostefano MJ, Evans MV, Richardson VM, Diliberto JJ, Birnbaum LS. Determination of parameters responsible for pharmacokinetic behavior of TCDD in female Sprague-Dawley rats. Toxicol Appl Pharmacol. 1997;147:151–168. doi: 10.1006/taap.1997.8242. [DOI] [PubMed] [Google Scholar]
  173. Wyde ME, Eldridge SR, Lucier GW, Walker NJ. Regulation of 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced tumor promotion by 17 beta-estradiol in female Sprague-Dawley rats. Toxicol Appl Pharmacol. 2001;173:7–17. doi: 10.1006/taap.2001.9166. [DOI] [PubMed] [Google Scholar]
  174. Wyde ME, Seely J, Lucier GW, Walker NJ. Toxicity of chronic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin in diethylnitrosamine-initiated ovariectomized rats implanted with subcutaneous 17 beta-estradiol pellets. Toxicol Sci. 2000;54:493–499. doi: 10.1093/toxsci/54.2.493. [DOI] [PubMed] [Google Scholar]
  175. Wyde ME, Wong VA, Kim AH, Lucier GW, Walker NJ. Induction of hepatic 8-oxodeoxyguanosine adducts by 2,3,7,8- tetrachlorodibenzo-p-dioxin in Sprague-Dawley rats is female-specific and estrogen-dependent. Chem Res Toxicol. 2001;14:849–855. doi: 10.1021/tx000266j. [DOI] [PubMed] [Google Scholar]
  176. Wyllie S, Liehr JG. Release of iron from ferritin storage by redox cycling of stilbene and steroid estrogen metabolites: a mechanism of induction of free radical damage by estrogen. Arch Biochem Biophys. 1997;346:180–186. doi: 10.1006/abbi.1997.0306. [DOI] [PubMed] [Google Scholar]

Articles from Dose-Response are provided here courtesy of SAGE Publications

RESOURCES