Effects of Postnatal Exposure to Mixtures of Non-ortho-PCBs, PCDDs, and PCDFs in Prepubertal Female Rats

D. Desaulniers*,1, K. Leingartner*, B. Musicki{dagger}, A. Yagminas*, G.-H. Xiao*, J. Cole*, L. Marro*, M. Charbonneau{ddagger} and B. K. Tsang{dagger}

* Health Canada, Environmental and Occupational Toxicology Division, Tunney’s Pasture, A.L:0803D, Ottawa, Ontario, Canada K1A 0L2; {dagger} Reproductive Biology Unit and Division of Reproductive Medicine, Departments of Obstetrics and Gynaecology and Cellular and Molecular Medicine, University of Ottawa; Hormones, Growth and Development Program, Ottawa Health Research Institute, 725 Parkdale Avenue, Ottawa, Ontario, Canada K1Y 4E9; and {ddagger} INRS-Institut Armand Frappier, Université du Québec, 245, Boulevard Hymus, Pointe-Claire, Québec, Canada H9R 1G6

Received April 1, 2003; accepted June 27, 2003


    ABSTRACT
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
There are concerns that postnatal exposure to organochlorines present in breast milk could lead to adverse health effects. We reconstituted four mixtures of aryl-hydrocarbon receptor (AhR) agonists (3 non-ortho polychlorinated biphenyls [PCBs], 6 polychlorinated dibenzodioxins [PCDDs], 7 polychlorinated dibenzofurans [PCDFs], or all 16 chemicals together [referred to as AhRM]) based on their concentrations in breast milk, and examined their effects following exposure by gavage from day 1 until day 20 of age. Female neonates received dosages of AhRM equivalent to 1, 10, 100, or 1000 times the amount consumed by an infant over the first 24 days of life. Other groups received the PCBs, the PCDDs, or the PCDFs at the 1000x level. All rats were sacrificed at 21 days of age. Changes in ethoxyresorufin-o-deethylase hepatic activity, thymus and body weights, and serum thyroxin were linked to the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) toxic equivalents (TEQ) of the four mixtures (1000x-AhRM > PCDDs > PCBs > PCDFs). To test for AhRM antiestrogenicity, two additional groups received 1.5 µg/kg of 17{alpha}-ethynyl estradiol (EE) with or without the 1000x-AhRM. The AhRM had no effect on uterine weight or EE-stimulated uterine growth. The actions of the combined EE and AhRM treatments suggest additive effects in decreasing pentoxyresorufin-o-deethylase activity and spleen weight, but nonadditive/antagonistic effects on adrenal weight and serum thyroxin. In conclusion, (1) 10x-AhRM had no detectable effects, (2) TEQ values relate to observed toxicities, even when testing complex mixtures of AhR agonists, and (3) indications of tissue-specific additive and nonadditive/antagonistic effects, but no synergism, were observed when doses of AhRM were increased, or combined with EE.

Key Words: uterotrophic bioassay; prepubertal; rat; mixture; CYP 2B; toxic equivalent; polychlorinated biphenyls; polychlorinated dibenzodioxins; polychlorinated dibenzofurans; breast milk.


    INTRODUCTION
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Organochlorines are ubiquitous environmental contaminants for which human exposure starts at the gamete stage (Bush et al., 1986Go; Jarrell et al., 1993Go; Pauwels et al., 2000Go; Schecter et al., 1996Go), followed by in utero (Feeley and Brouwer, 2000Go; Foster et al., 2000Go), and the postnatal period. There is concern that exposure to chemicals during the developmentally sensitive in utero and postnatal periods may have long-term adverse health consequences, including increased risk of developing cancers, reproductive problems, learning difficulties, and immune system and thyroid function deficiencies (Aoki, 2001Go; Golden et al., 1998Go). During the postnatal period, exposure to organochlorines is more important in breastfed than nonbreastfed infants, since breast milk is an important route for excretion of accumulated lipophylic contaminants (Haines et al., 1998Go; Lorber and Phillips, 2002Go). There is limited data available concerning risks associated with exposure to breast milk contaminants (Landrigan et al., 2002Go; Pronczuk et al., 2002Go). The beneficial effects of breastfeeding are well recognized, and it has recently been suggested that neurological impairments originating from in utero exposure to organochlorines may be counteracted by breastfeeding (Ribas-Fito et al., 2003Go). Nevertheless, a better understanding of possible adverse effects resulting from exposure in utero, during the postnatal period, or from the combined periods, is essential to ensure appropriate risk assessment and management, and for regulatory agencies to develop chemical testing strategies adequately predicting outcomes later in life.

Non-ortho polychlorinated biphenyls (PCBs), polychlorinated dibenzodioxins (PCDDs), and polychlorinated dibenzofurans (PCDFs) are organochlorines present as contaminants in breast milk (Brouwer et al., 1998aGo; Dewailly et al., 1992Go; Lorber and Phillips, 2002Go). These chemicals exert most of their toxic effects through activation of the aryl-hydrocarbon receptor (AhR), and are referred to as AhR agonists (Denison et al., 2002Go; Safe, 2001Go). Based on their toxic effects in the liver, thymus gland, and body weight (Safe, 2001Go), they can be considered among the most toxic organochlorines. The effectiveness of these chemicals to bind the AhR and induce these toxic effects was used to set up the Toxic Equivalent (TEQ) system. This system provides indices of toxicity for AhR agonists relative to TCDD, and simplifies risk assessment of mixtures (Safe, 1994Go; van Leeuwen et al., 2000Go). In addition, AhR agonists have the potential to promote cancer and affect the immune, nervous, reproductive, and endocrine systems (Aoki, 2001Go; Baccarelli et al., 2002Go; Petroff et al., 2001bGo; Tian et al., 2002Go; Weiss, 2002Go). Among endocrine effects, modulation of the estrogen receptor (ER) and thyroid hormone systems are being extensively studied. AhR agonists have been shown to act as antiestrogens, not by competing for the ligand domain of the ER, but by downregulating ER, increasing the rate of estradiol-17ß metabolism, inhibiting various growth factors and cell cycle enzymes (Chen et al., 2001Go), mainly through crosstalk between AhR and ER pathways (Reen et al., 2002Go; Safe, 1999Go). Liganded AhR complexes interact with DNA dioxin response elements near promoter regions activated by ER and thus induce antiestrogenic effects (reviewed by Safe, 1999Go). They could also interact with unliganded ER to induce estrogenic effects (Ohtake et al., 2003Go). AhR agonists affect multiple components of the thyroid system, including displacement of thyroid hormones from the carrier protein transthyretin to decrease deiodinase and increase glucuronidase enzyme activities (Brouwer et al., 1998bGo; Khan et al., 2002Go; Vansell and Klaassen, 2002aGo,bGo; Wade et al., 2002Go).

Very few studies have tested the effects of reconstituted breast milk contaminant mixtures in neonates, and at exposure levels similar to humans. Parkinson et al.(1980)Go observed that a reconstituted breast milk mixture of 13 ortho-PCBs was seven times more potent for stimulating hepatic aryl hydroxylase activity, compared to the more complex commercial PCB mixture Kanechlor 500. We found that postnatal exposure to high doses of the most abundant breast milk contaminants (PCBs, p,p'-dichlorodiphenyltrichloroethane [DDT], p,p'-dichlorodiphenyldichloroethene [DDE], or to TCDD), promoted the development of methylnitrosourea-induced mammary lesions in the rat (Desaulniers et al., 2001Go). To continue testing reconstituted mixtures of breast milk contaminants, our objective for the current study is to describe dose-response effects in 21-day-old female Sprague-Dawley rats following postnatal exposure to mixtures of non-ortho PCBs, PCDDs, and PCDFs. The mixtures were prepared according to the amount of these contaminants present in breast milk (Brouwer et al., 1998aGo; Dewailly et al., 1992Go; Lorber and Phillips, 2002Go). As classical indicators for effects of AhR agonists, we assessed changes in organ weights, three reference points to the TEQ system (hepatic cytochrome P450 [CYP] 1A1 induction, thymus atrophy, and reduction in body weight [Safe, 2001Go]) and effects on CYP2B and 3A as other indicators of hepatic exposure. In contrast to CYP1A1, which is regulated through the AhR, the expression of CYP2B and 3A are regulated through heterodimerization of the retinoid X receptor with either the nuclear orphan constitutively active receptor (CAR), or the pregnane X receptor, respectively (Cai et al., 2002Go; Masahiko and Honkakoski, 2000Go; Sueyoshi and Negishi, 2001Go; Ueda et al., 2002Go; Zelko and Negishi, 2000Go). Endocrine effects were assessed by measuring serum corticosterone, pituitary luteinizing hormone (LH), serum thyroxin and the pituitary thyroid stimulating hormone (TSH) as indicators of thyroid function (Brouwer et al., 1998bGo; Khan et al., 2002Go; Vansell and Klaassen, 2002aGo,bGo; Wade et al., 2002Go). In addition, the antiestrogenic potential of the AhR agonist mixture (AhRM) was investigated by testing its effect on uterine growth and its ability to counteract the uterotrophic effect of 17{alpha}-ethynyl-estradiol (EE). The ability of uterine cells to proliferate in response to estrogens is the basis for the rodent uterotrophic bioassay. This test, although not very sensitive, is considered the "gold standard" test for estrogenicity, because it takes into account the effects of metabolism, serum binding, and pharmacokinetics (Gray et al., 1997Go; Odum et al., 1997Go). It is intended to be used by regulatory agencies to identify the in vivo activities of suspected estrogen agonists or antagonists (Kanno et al., 2001Go; Padilla-Banks et al., 2001Go).


    MATERIALS AND METHODS
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Animal treatment.
Four mixtures were prepared and included 3 non-ortho-chlorinated PCBs, 6 PCDDs, 7 PCDFs, or all 16 of these chemicals together, which we will refer to as AhRM, respectively (Table 1Go). The amounts of each chemical were calculated to achieve, over the exposure period, a cumulative dose representing the amount that a human baby would have consumed over its first 24 days of life. The calculations were based on milk fat concentrations of each chemical present in breast milk samples collected in 1989–1990 from Caucasian women in southern Quebec (Dewailly et al., 1992Go), a newborn consumption rate of 120 ml breast milk/kg body weight per day (Ayotte et al., 1995Go), and a milk fat content of 3.7% (Frank et al., 1988Go).


View this table:
[in this window]
[in a new window]
 
TABLE 1 Concentrations of PCB, PCDF, and PCDD Isomers in the 1000x AhRM
 
At one day of age, female Sprague-Dawley rats (Charles River, St-Constant, QC) were randomly assigned to 18 fostering dams with five to eight female neonates per dam. To randomize the litter effect, the neonates within a litter were assigned to different treatment groups (10 to 13 neonates per group). The experiment included nine separate treatment groups: water, corn oil (vehicle), AhRM at dosages equivalent to 1, 10, 100, or 1000 times the amount that an infant would have consumed over its first 24 days of life, and the three chemical family components of the 1000x-AhRM which include the non-ortho PCBs, PCDDs, and PCDFs. The neonates were identified and exposed to the vehicle or the mixtures by gavages at ages 1, 5, 10, 15, and 20 days, each dose (5 ml of mixture per kg bw) representing 5 days of ingestion (Fig. 1Go).



View larger version (11K):
[in this window]
[in a new window]
 
FIG. 1. Experimental design. Female neonates were exposed to the mixtures by gavage (5 ml/kg bw) at day 1 of age (a cumulative dose for days 0, 1, 2, 3, and 4), day 5 (for days 5, 6, 7, 8, and 9), day 10 (for days 10, 11, 12, 13, 14), day 15 (for days 15, 16, 17, 18, 19), and 20 days (for days 20, 21, 22, 23, and 24). In addition, between 0800–0900 h on day 18, 19, and 20 of age, two groups (oil, 1000X) were weighed and injected sc with 17{alpha}-ethynyl estradiol (EE; 0.5 µg/kg). All rats were sacrificed at 21 days of age.

 
Two additional groups were used to test for antiestrogenic effects of the 1000X-AhRM. The first group (n = 10) received corn oil (the vehicle for the AhRM) at 1, 5, 10, 15, and 20 days of age and a daily dose of 17{alpha}-ethynyl estradiol (EE; CAS No. 57–63–6; 0.5 µg/kg, corn oil as vehicle) injected sc in the morning on days 18, 19 and 20 of age. The second group included eleven rats receiving the same doses of EE plus AhRM as described previously (Fig. 1Go). All rats were sacrificed at 21 days of age.

Radioimmunoassays.
Serum thyroxin and corticosterone levels were determined using commercial I125 radioimmunoassay (RIA) kits (ICN Biomedicals, Costa Mesa, CA), while pituitary LH and TSH contents were determined by RIA as previously described (Desaulniers et al., 1999Go). Briefly, the pituitary glands were sonicated in 200 µl of 0.05 M phosphate-buffered saline, 1% BSA and protease inhibitors (1 mM EDTA, 1.0 µM pepstatin A, 10 µM E-64, 17 µM 2,3-dehydro-2-deoxy-n-acetylneuraminic acid and 1.5 µg/ml aprotinin; Sigma Chemicals, St. Louis, MO) and the homogenates were centrifuged (1300 x g, 30 min, 4°C), to recuperate the supernatant. The sonication/centrifugation procedure was repeated twice, resulting in a final volume of 0.6 ml from which hormones were measured. Pituitary hormone preparations for iodination, standard curves, and primary antisera were rLH-I-9 (AFP-10250C), rLH-RP-3 (AFP-7187B), and rLH-S-11 for LH and rTSH-I-9 (AFP-11542B), rTSH-RP3 (AFP-5512B), and rTSH-RIA-6 for TSH, respectively (all provided by the National Hormone and Pituitary Program, the National Institute of Diabetes and Digestive and Kidney Diseases). The sensitivity of the LH, TSH, thyroxin, and corticosterone assays were 0.02 ng/tube, 0.04 ng/tube, 0.63 µg/dl and 25 ng/ml, respectively, and the interassay coefficients of variation were 6.9, 6.2, 7.4, and 13%, respectively.

Hepatic cytochrome P450 enzyme assays.
Hepatic CYP1A1 and CYP2B1/2 activities in 10,000x g supernatants were examined by determining the activities of ethoxyresorufin-o-deethylase (EROD), pentoxyresorufin-o-deethylase (PROD), and benzyloxyresorufin-o-deethylase (BROD), according to the method of Burke et al.(1985)Go. While PROD and BROD activities have classically been considered indicators of CYP2B1/2 and 3A activities (Burke et al., 1985Go; Chen and Eaton, 1991Go; Namkung et al., 1988Go), respectively, they have been shown to predominantly reflect CYP2B activities (Nerurkar et al., 1993Go). CYP enzyme activities were calculated from the slope of the linear portion of the reaction.

Analysis of hepatic cytochrome P450 protein content by Western blot.
Liver homogenates, prepared in 2.5 volumes of cold Tris-KCl (0.5M /1.15%; pH 7.4) were centrifuged (10,000 x g, 20 min) to obtain a post mitochrondrial fraction (PMF) that was further centrifuged (105,000 x g, 1 h). The pellets were resuspended in 0.5M Tris/HCl (pH 6.8) and centrifuged again (105,000 x g, 1 h) to obtain the microsomal preparations. The protein concentration of the microsomal solution was measured by the method of Bradford (1976)Go. Microsomal preparations were diluted 1:1 with loading buffer (0.5M Tris/HCl pH 6.8, 10% glycerol, 0.5% sodium dodecyl sulfate [SDS]), and the proteins were resolved (50 min at 150V) on 4–20% gradient precast gels (ICN Biomedicals, Aurora, OH). The proteins were electro-transferred (100 V at 0EC) onto nitrocellulose membranes in transfer buffer (25 mM Tris, 192 mM Glycine, 20% Methanol, pH 8.3). The membranes were treated with SuperBlock (Pierce Chemical Co., Rockford, IL) and 0.1% Tween20 in phosphate-buffered saline (PBS) to block nonspecific binding sites and incubated with the primary antibody (1 h, RT; all from Gentest Corporation, Woburn, MA). Blots were then washed with 0.1% Tween20 in PBS, incubated with an alkaline phosphatase conjugated secondary antibody and visualized using nitroblue tetrazolium/5-bromo-4-chloro-3-indolylphosphate (Sigma-Aldrich Ltd., Oakville, ON, Canada). Signal intensity of the samples and standards (CYP1A1, In Vitro Technologies, Inc. Baltimore, MD; CYP3A2, and CYP2B1, Gentest Corporation, Woburn, MA) were quantified using an imaging densitometry system (BioRad Model GS-670; Molecular Analyst/PC software, BioRad Laboratories Inc., Mississauga, ON, Canada). The results were corrected for the amount of protein loaded on the gel and are expressed as the ratio of sample density/standard density.

Determination of hepatic CYP2B1, CYP3A2, and ER{alpha} mRNA abundance.
The hepatic mRNA abundance of CYP2B1, CYP3A2, and ER{alpha} was determined by quantitative real-time reverse transcription (RT)-PCR method. Total RNA was isolated from rat liver tissue samples using the RNeasy mini kit and the RNase-free DNase set (Qiagen, Mississauga, ON, Canada). RT of the total RNA was performed at 37°C for 1 h in 20 µl of the reaction mixture containing 2 µg of total RNA, 0.5 mM dNTPs, 10 units of recombinant RNase inhibitor, 1 µM oligo (dT) primer and 4 units reverse transcriptase (Qiagen Omniscript Reverse Transcriptase kit) and inactivated by heating (95°C, 5 min). To test for residual DNA contamination, negative control samples did not include the reverse transcriptase. The resulting cDNA was amplified by quantitative real-time PCR analysis.

The quantitative real-time PCR (iCycler iQ system, BioRad, Mississauga, ON, Canada) was performed in 25 µl of reaction mixture containing QuantiTect SYBR Green PCR Master Mix (1X, BioRad, Mississauga, ON, Canada), 4 µl of RT products (80 ng total RNA), and 0.3 µM primers (forward and reverse). The temperature program included 15 min at 95°C, then 40 cycles of 30 s at 95°C, 30 s at 55°C, and 1 min at 72°C. The PCR primers (forward and reverse), designed in accordance with published rat gene sequences (Fujii-Kuriyama et al., 1982Go; Miyata et al., 1994Go; Nudel et al., 1983Go; Spreafico et al., 1992Go), were CAATGAGCTGCGTGTGGC and GGGTCATCTTTTCACGGTTGG for ß-actin, TTCAGCTCTCACACTGGAA and AAAGGCAGAGGTTTTGGC for cyp3A2, TGATCTTTGCCAATGGGGAAC and CCGTTCTTCCACACTCCTCT for cyp2B1, and GCGCCGCCTACGAGTTCA and GACCGTAAGTGATGCTCGACTG for ER{alpha}. The amplification of a single specific DNA band of the correct size was verified by gel electrophoresis, and the presence of a single melting curve. The amount of PCR product was quantified by comparing the threshold cycle of each sample to the standard curve established with threshold cycles of serially diluted quantities of purified cDNA of the same genes. The results were then normalized relative to the abundance of ß-actin mRNA present in each sample.

Statistical analysis.
The effects of treatments on body weight were analyzed using the software SAS (release 8.02; SAS Institute Inc., Cary, NC, 2001) with the following repeated measure model:


where Ti represents the ith treatment group, Aj(i) is the jth animal nested in the ith treatment group, Pk is the kth time period, and yijk is the weight of the jth animal in the ith treatment group at time k. The animal term (Aj(i)) is the error term used for testing treatment differences, and {varepsilon}k(ij) represents error associated with period differences as well as the interaction between period and treatment differences. All other statistical analyses were performed with the software JMP (SAS Institute Inc., 1998Go) by applying one-way ANOVA followed by the Tukey-Kramer test to identify significantly different means. The analysis of data which tested the effects of EE and 1000X-AhRM were performed using two way ANOVA considering the effects of the mixture, EE, and their interaction. Homogeneity of variances and normality of the data were initially verified by O’Brien, Brown-Forsyth and Shapiro-Wilk Goodness-of-fit tests (SAS Institute Inc., 1998Go). The nonparametric Wilcoxon/Kruskal Wallis tests were used when normality of data and/or equal variances were not reached. In all cases, p <= 0.05 was considered to be a significant effect, whereas p <= 0.1 was considered as a tendency of effect.


    RESULTS
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
The effects of treatment on body weight were not statistically significant, but they were inversely related to the administered TEQ doses for the different groups (Tables 2Go and 3Go). The liver/body weight ratio was significantly increased with 1000x-AhRM, PCB, and PCDD treatments (Tables 2Go and 3Go), while only the 1000x-AhRM treatment decreased thymus and adrenal weights (Table 2Go). The 1000x-AhRM had no effects on brain weights, but it increased the brain/body weight ratios (Table 2Go). Serum thyroxin concentrations were significantly lower in the 1000x-AhRM group (Tables 2Go and 3Go), although none of the mixture treatments affected serum corticosterone levels, pituitary LH and TSH contents.


View this table:
[in this window]
[in a new window]
 
TABLE 2 TCDD-TEQ Doses for the AhRM and Dose-Response Effects on Organ Weights and Serum Thyroxin
 

View this table:
[in this window]
[in a new window]
 
TABLE 3 TCDD-TEQ Doses for the PCB, PCDD, and PCDF Components of the 1000x AhRM and Their Effects on Organ Weights and Serum Thyroxin
 
A significant increase in EROD activity was detected in the 100x group (p < 0.05), while the largest response was observed in the 1000x-AhRM group (Fig. 2Go). The PCB, PCDD, and PCDF treatments increased EROD activity with the smallest induction observed in the PCDF group. Although the PCDD group received a TCDD-TEQ dose 55% lower than that of the 1000x-AhRM group, there were no significant differences in EROD activities between the two groups (Fig. 2AGo). PROD activity in the 1000x-AhRM group was reduced below the control level (p = 0.007, Fig. 2BGo). BROD activity was significantly increased in the 100X AhRM, PCDF, and PCDD groups (p < 0.05, Fig. 2CGo). In light of the large coefficients of variation for PROD and BROD data at high dosages, we verified the effects of 1000x-AhRM by measuring the hepatic CYP1A1, CYP2B1, and CYP3A2 protein and mRNA levels (Table 4Go). Protein and mRNA analyses confirmed the suppression of CYP2B1 by 1000x-AhRM. This effect on CYP2B1 mRNA was also detectable at the 100x dose (p < 0.05). The 1000x-AhRM significantly increased messenger RNA abundance and protein content of CYP1A1, but not of CYP3A2. There were no significant correlations between enzyme activities and associated protein levels. The hepatic ER{alpha} mRNA abundance was reduced by 1000x-AhRM.



View larger version (22K):
[in this window]
[in a new window]
 
FIG. 2. Hepatic ethoxy (EROD) (A), pentoxy (PROD) (B), and benzyloxy resorufin-o-deethylase (BROD) (C) activities (n = 10 to 13) following postnatal oral exposure to water, oil (vehicle), AhRM in increasing doses (1x, 10x, 100x, and 1000x), and its chemical family components (non-ortho PCBs, PCDDs, and PCDFs) at the 1000x level. Means with different letters are significantly different (p < 0.05). *Significantly smaller, or ** greater, than oil (t-test, p < 0.05). Coefficients of variation are provided within parenthesis.

 

View this table:
[in this window]
[in a new window]
 
TABLE 4 Effects of the AhR Agonist Mixture on Hepatic CYP1A1, CYP2B1, and CYP3A2, Protein and mRNA Levels, and ER{alpha} mRNA Levels (Mean ± SE)
 
Figure 3Go shows a regression analysis between the TCDD-TEQ doses of the AhRM relative to hepatic EROD activities. TCDD-TEQ values were derived by multiplying the TCDD-TEQ of each mixture by the cumulative volume of solution administered to each rat from day 1 until day 20. The body weight at day 20 was used to transform the TCDD-TEQ dose on a per kg basis. The expected EROD activity for the PCDD group, calculated using the regression and the average TCDD-TEQ dose for that group, is significantly lower (p < 0.0001) than the experimental EROD activity (Fig. 2Go). Perhaps this is an indication of increased nonadditive/antagonistic effects among chemicals of the more complex AhRM. A lowest observable effect level (LOEL) of 15 ng/kg AhRM was calculated based on EROD activity at the 95% upper bound level (0.0799 nmol/min/mg) of the no observable effect level (NOEL, based on the 1x-AhRM group). Note that the 95% upper bound level for the combined water, corn oil, and 1x-AhRM groups is similar (0.0793 nmol/min/mg).



View larger version (20K):
[in this window]
[in a new window]
 
FIG. 3. Regression analysis between EROD hepatic activities and TCDD-TEQs in AhRM treated rats [r2 = 0.8, p < 0.0001, Log EROD = -1.27 + (-0.35 Log TCDD-TEQ) + 0.16 (Log TCDD-TEQ)2]. Using the regression equation and the average TCDD-TEQ dose (507 ng/kg) of the PCDD group (scattered diagram), the calculated EROD activity (0.61 nmol/min/mg) is significantly lower than the experimental mean obtained for the PCDD group (p < 0.0001). The dotted lines suggest a LOEL at 15 ng/kg based on EROD activity at the 95% upper bound level (0.079 nmol/min/mg) of the NOEL.

 
The antiestrogenicity of 1000x-AhRM was studied by testing its ability to attenuate the EE-induced uterotophic effect (Table 5Go). This test also studied the combined effects of EE and 1000x-AhRM treatments on other endpoints. As expected the EE treatment increased uterine weight, but the 1000x-AhRM did not reduce this effect. The EE treatment slightly increased pituitary and brain weights. The results from Table 5Go confirmed those of Tables 2Go and 3Go, which indicated that AhRM increased liver weight, but decreased the weight of the spleen and thymus. Interestingly, there are indications of additive (spleen) and nonadditive/antagonistic effects (kidneys, adrenals, serum thyroxin; Table 5Go, Fig. 4CGo). The 1000x-AhRM reduced the weight of the spleen, which was further reduced by the EE treatment. While 1000x-AhRM attenuated the EE-induced reduction in kidney weight, it decreased adrenal weight but this effect was prevented by EE. The decrease in adrenal weight induced by 1000X-AhRM was not associated with significant changes in serum corticosterone level (control: 72 ± 11 ng/ml; EE: 50 ± 8 ng/ml; 1000X-AhRM, 78 ± 13 ng/ml; 1000X-AhRM + EE, 61 ± 13 ng/ml). Serum thyroxin was reduced by 1000x-AhRM, but this effect was counteracted by EE (Fig. 4CGo). In addition, EE slightly lowered pituitary TSH, but significantly decreased pituitary LH, hepatic PROD and BROD activities (Fig. 4Go).


View this table:
[in this window]
[in a new window]
 
TABLE 5 Effects of 1000x-AhRM and 17{alpha}-Ethynyl-Estradiol (EE) on Organ Weights
 


View larger version (21K):
[in this window]
[in a new window]
 
FIG. 4. Pituitary gland content in LH (A), and TSH (B), serum thyroxin (C), hepatic BROD (D), and PROD (E) activities, following exposure to 1000x-AhRM with or without 17{alpha}-ethynyl estradiol (EE; cumulative dose of 1.5 µg/kg bw). Mean ± SE. p values were derived using two-way ANOVA.

 

    DISCUSSION
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
The dosages of organochlorines administered in this study were based on the concentrations found in Canadian breast milk (Dewailly et al., 1992Go), and fortunately produced relatively small responses at the dose 100x. These results are consistent with our findings from an experiment conducted concurrently showing no long term effects of 1000x-AhRM on time to vaginal opening, length of estrous cycle, mammary gland development, and methylnitrosourea-induced mammary tumor development (Desaulniers et al., manuscript in preparation).

In the current study, effects on EROD activity, body weight, serum thyroxin, and liver weight followed additive patterns with the most important changes induced by 1000x-AhRM, and to a lesser extent by the PCDD, PCB, and PCDF mixtures. These results confirm that the TEQ system permits the derivation of TEQ values that relate to the observed toxicity, even when testing complex mixtures of AhR agonists. The PCDD mixture, however, induced more EROD activity than what could have been predicted from the AhRM (Fig. 3Go). This suggests that there may be nonadditive/antagonistic interactions among AhR agonists, and an overestimation of risks based on the TEQ derived from complex mixtures. Similarly, Parkinson et al.(1980)Go found that a reconstituted breast milk mixture of 13 ortho-PCBs was seven times more potent in stimulating hepatic aryl hydroxylase activity than the more complex commercial PCB mixture Kanechlor 500. No synergistic effects were observed in our study. The number of cellular pathways altered by exposure to chemicals, and pharmacokinetic interactions, are factors determining if effects are additive, antagonistic, or synergistic (Chu et al., 2001Go; Lee et al., 2002Go; Petrulis and Bunce, 2000Go; van Birgelen et al., 1994aGo,bGo). Supra-additive (Burgin et al., 2002Go) or synergistic effects are infrequent findings, which usually result from exposure to mixtures of unrelated chemicals with different modes of action that may regulate the same endpoint (Abbott et al., 1994Go; Jensen et al., 2000Go; van Birgelen et al., 1996aGo,bGo). In this context, the TCDD-TEQ system, based mostly on interactions with a single receptor (i.e., AhR), has recognized limitations (Safe, 2001Go) and cannot correlate with all toxic effects such as estrogenicity, neurotoxicity, oxidative stress, and reproductive and developmental effects (Burgin et al., 2002Go; Li and Hansen, 1996Go). The importance of this system should, however, not be minimized as it clearly provides reliable indicators of toxicity, as shown in the present investigations.

In contrast to EROD induction, PROD and BROD activities showed no clear additive effects among the PCB, the PCDD, and the PCDF groups. Moreover, the 1000x-AhRM decreased PROD activities, CYP2B protein content, and mRNA abundance below the control levels. Others observed antagonistic effects, but of lower magnitude than in the current study, on PROD and/or BROD activities following coadministration of TCDD (or other AhR agonists) with nondioxin like PCBs or mixtures in prepubertal and adult female rats (Chu et al., 2001Go; van Birgelen et al., 1994bGo; van der Kolk et al., 1992Go), mice (De Jongh et al., 1993Go), and rabbits (Serabjit-Singh et al., 1983Go). Collectively, these data suggest that the suppressive effect is a high dose phenomenon associated with the presence of AhR agonists. The suppression of CYP2B might be attributed to its complex regulatory mechanisms. In contrast to CYP1A1, which is regulated through the AhR, the expression of CYP2B and 3A are regulated through heterodimerization of the retinoid X receptor with either the nuclear orphan CAR, or the pregnane X receptor, respectively (Cai et al., 2002Go; Masahiko and Honkakoski, 2000Go; Sueyoshi and Negishi, 2001Go; Ueda et al., 2002Go; Zelko and Negishi, 2000Go). Since protein and mRNA levels of CYP2B, but not of CYP3A2, were significantly reduced by the treatment (Table 4Go), it may be speculated that this suppression was mediated through the CAR pathway.

In addition to enzyme suppression, large coefficients of variation characterized BROD and PROD activities among the high dose groups (Fig. 2Go). Dynamic hormonal and metabolic changes occurring during development may be responsible for some variability of the results in the prepubertal rat. In fact, there is a gradual development in the expression of CYP enzymes in the fetal (Hakkola et al., 2001Go), and immature rat liver (Hines and McCarver, 2002Go; Larsen-Su et al., 2001Go), associated with dynamic endocrine and metabolic changes (Li and Hansen, 1997Go). While reasons for these differences in BROD and PROD activities among individuals are yet to be determined, some variability might be attributed to gene polymorphisms, the lack of substrate specificity (Iba et al., 2000Go), the presence of competitive metabolizable substrate, or suicide substrates which bind to the cytochromes and inhibit their catalytic activities (Voorman and Aust, 1987Go). Note that EE, a suicide substrate for CYP2B enzymes (Kent et al., 2002Go), decreased BROD and PROD activities, particularly in the presence of high concentrations of AhR agonists (Fig. 4Go).

Our studies have shown that AhRM does not affect uterine weight at 21 days of age, or the EE induced uterotrophic effect. The absence of antiestrogenic effects of AhRM on the uterus are consistent with the results from White et al.(1995)Go showing an absence of antiestrogenic effects of TCDD (even at high doses) on uterine tissues in prepubertal female rats. Others have reported antiestrogenic activity of numerous AhR agonists in slightly older (23–26 day old) prepubertal female rats (Chen et al., 2001Go; Jansen et al., 1993Go; Ohtake et al., 2003Go; Safe, 1999Go). Reasons for the controversial findings are yet to be explained, but they may be related to specific AhR agonists or dosages being tested, to the selection of the potent EE instead of estradiol-17ß as estrogenic agent, to the circulating level of estradiol-17ß that is known to differ even in age-matched prepubertal rats, and to developmental delays prior to the establishment of crosstalk between the AhR and ER. It has been demonstrated that the antiestrogenic activity of AhR agonists is largely mediated through crosstalk pathways between the ER and AhR (Chen et al., 2001Go; Safe, 1999Go; Zacharewski and Safe, 1998Go). The regulation of AhR is tissue-specific, with a developmental down-regulation of the AhR protein in the rat prostate occurring from postnatal day 7 to 21 (Sommer et al., 1999Go). We are not aware of similar observations made from the prepubertal female reproductive tract. However, the estrogenic environment, which is at the nadir in the 21-day-old female rat (Li and Hansen, 1997Go), is likely to influence the expression of the AhR. Chaffin et al.(2000)Go showed changes in the expression of the AhR in the liver and the ovary during the reproductive cycle of Sprague-Dawley rats, with high levels being present during the estrogenic phase (early proestrus), but suggested that estradiol-17ß was not the key regulator of this process. Others have reported that estradiol-17ß enhances AhR expression and CYP 1A1 induction by TCDD in the immature (Petroff et al., 2001aGo) and adult female rat (Sarkar et al., 2000Go). The sensitivity of the uterine response to detect antiestrogenic compounds should also be investigated in adult ovariectomized rats (Kanno et al., 2001Go) or immature mice (Newbold et al., 2001Go; Ohtake et al., 2003Go; Padilla-Banks et al., 2001Go).

Despite the lack of antiestrogenic effects detected in uterine tissues from 21-day-old female rats, the 1000x-AhRM reduced ER{alpha} mRNA levels by 87% in the liver (Table 4Go), but had no effects in the uterine tissue (oil: n = 6, 1.12 ± 0.11; 1000x: n = 7, 1.31 ± 0.11 attoM ER{alpha}/fentoM ß-actin mRNA; data added during revision). Similar observations were made in adult rats seven days after exposure to TCDD, in which the density of the ER protein decreased by 92% in the liver, but only slightly in the uterus (Hruska and Olson, 1989Go). Perhaps the lower sensitivity of the uterus to the effects of the AhRM may be attributed to pharmacodynamic differences between the 21-day-old uterus and the liver.

The results from testing the effects of 1000x-AhRM with EE provided indications of additive and nonadditive/antagonist effects. AhR agonists are known to induce immunotoxicity (Lin et al., 2001Go; Tryphonas et al., 2001Go) and, as expected, the spleen and thymus weights were slightly reduced in AhRM treated rats (Tables 2Go, 3Go, and 5Go). Coadministration of AhRM with EE elicited additive effects and further decreased spleen weight, an observation supported by the immunomodulatory role of endogenous (Ito et al., 2002Go; Karpuzoglu-Sahin et al., 2001Go) and exogenous estrogens (genistein, methoxychlor; Guo et al., 2002Go). This additive effect contrasts with the reported antiestrogenic effects of AhR agonists, and highlights organ differences in chemical interactions. Although the AhRM reduced adrenal weight and EE counteracted this response, these treatments had no effect on serum corticosterone. However, TCDD has been reported to inhibit the adrenal cytochrome P450 side-chain cleavage (Harvey et al., 1999Go) and adrenal steroidogenesis (Goldman and Yawetz, 1992Go). In our experiment, all necropsies were performed in the morning, a time at which corticosterone levels are at their lowest. It is possible that collecting serum samples at a later time of the day, when corticosterone levels peak, might have facilitated the detection of treatment effects. The compensatory effect of EE on the AhR-induced adrenal weight loss might be explained by the fact that estradiol stimulates adrenocorticotropin secretion (Harvey et al., 1999Go), increases adrenal blood flow and lipid accumulation, and thus adrenal weight (Hinson and Raven, 1999Go). The 1000x-AhRM and EE treatments reduced kidney weights (Table 5Go), in line with the observations that estrogens exacerbate nephrotoxicity and even induce kidney tumorigenesis (Devanesan et al., 2001Go; Healing, 1999Go; Nakamura et al., 2001Go). Adrenals and kidneys interact through the renin-angiotensin system (Healing, 1999Go), and perhaps interference with this system is linked to changes in both organ weights. Brain weight was slightly increased by EE (Table 5Go), perhaps as a consequence of direct beneficial actions of estrogens on the nervous system (Mattson, 1999Go), and/or indirectly via estrogen stimulation of the thyroid system (Croissandeau et al., 1996Go; Kimura et al., 1994Go; Lisboa et al., 1997Go; Schomburg and Bauer, 1997Go), which is a regulator of normal brain development (Howdeshell, 2002Go; Zoeller et al., 2002Go). Stimulation of the thyroid system by EE is supported by our results showing that 1000x-AhRM reduced serum thyroxin, but this effect was attenuated by EE (Fig. 4Go). A reduction in serum thyroxin induced by the AhRM was expected since multiple components of the thyroid system are vulnerable to the effects of AhR agonists (Brouwer et al., 1998bGo; Khan et al., 2002Go; Vansell and Klaassen, 2002aGo,bGo).

The lowest level of exposure achieved in the current experiment (AhRM-1x) represents the average exposure level of a breast fed infant during its first 24 days of life (Table 1Go). This was based on the average milk fat levels of AhR agonists present in Caucasian breast milk from southern Quebec during 1989–1990 (Dewailly et al., 1992Go). In general, these levels were similar to those reported in studies from the U.S., Europe (Dewailly et al., 1996Go; Schecter and Piskac, 2001Go; WHO, 1996Go), and Canada (Newsome and Ryan, 1999Go), but 2 to 10 times smaller than those measured from Inuit women living in Arctic Québec (Dewailly et al., 1992Go). The use of the current study for risk assessment should consider three factors that may contribute to overestimating possible risks in the general population. First, the levels of AhR agonists in breast milk have been declining (Craan and Haines, 1998Go; Norén and Meironyté, 2000Go; Solomon and Weiss, 2002Go; WHO, 1996Go), possibly by half every 9.6 years (WHO, 1996Go). Second, dosages were based on mean concentrations (Table 1Go) and this data is inherently not normally distributed, which implies that a large proportion of samples had much lower residue levels than the mean. Finally, there is a wide range of beneficial effects of breastfeeding (American Academy of Pediatrics, 1997Go; Haller and Simpser, 1999Go; McVea et al., 2000Go; Oddy, 2001Go; Pletta et al., 2000Go), and it is suspected to counteract adverse neurological effects presumed to occur from in utero exposure to organochlorines (Ribas-Fito et al., 2003Go).

In summary, the highlights of this article are (1) the AhRM administered by gavage during the postnatal period had no effects in the rat at a dose ten times the amount consumed by an infant over its first 24 days of life; (2) despite reports that AhR agonists are antiestrogenic, the AhRM had no effects on basal or EE-stimulated uterine growth; (3) while the AhRM increased EROD activity, it suppressed CYP2B activity, protein, and mRNA levels; (4) there were indications of agonistic and nonadditive/antagonistic interactions between the AhRM and EE; and (5) changes in AhR-dependent endpoints (EROD, body weights, serum thyroxin) were associated with TCDD-TEQ doses (1000x-AhRM > PCDDs > PCBs > PCDFs), even when testing complex mixtures.


    ACKNOWLEDGMENTS
 
The authors are grateful to Zahra Allidina, Lijun Shao, Karen Pazterko, Yulian Li, Korian Soumano, Lorraine Casavant, and Tanya Odorozzi for their excellent assistance; to the National Hormone and Pituitary Program, the National Institute of Diabetes and Digestive and Kidney Diseases, and Dr. A. F. Parlow for providing radioimmunoassay reagents; and to Dr. Raymond Poon for carefully reviewing this manuscript. This study was funded by the Toxic Substance Research Initiative and Health Canada.


    NOTES
 
1 To whom correspodence should be addressed. Fax: (613) 957-8800. Email:daniel_desaulniers{at}hc-sc.gc.ca. Back


    REFERENCES
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Abbott, B. D., Perdew, G. H., Buckalew, A. R., and Birnbaum, L. S. (1994). Interactive regulation of Ah and glucocorticoid receptors in the synergistic induction of cleft palate by 2,3,7,8-tetrachlorodibenzo-p-dioxin and hydrocortisone. Toxicol. Appl. Pharmacol. 128, 138–150.[CrossRef][ISI][Medline]

American Academy of Pediatrics (1997). Breastfeeding and the use of human milk. Workgroup on Breastfeeding, American Academy of Pediatrics. Pediatrics 100, 1035–1039.[Abstract/Free Full Text]

Aoki, Y. (2001). Polychlorinated biphenyls, polychlorinated dibenzo-p-dioxins, and polychlorinated dibenzofurans as endocrine disrupters—what we have learned from Yusho disease. Environ. Res. 86, 2–11.[CrossRef][ISI][Medline]

Ayotte, P., Dewailly, E., Bruneau, S., Careau, H., and Vezina, A. (1995). Arctic air pollution and human health: What effects should be expected? Sci. Total Environ. 160/161, 529–537.[CrossRef]

Baccarelli, A., Mocarelli, P., Patterson, D. G., Jr., Bonzini, M., Pesatori, A. C., Caporaso, N., and Landi, M. T. (2002). Immunologic effects of dioxin: New results from Seveso and comparison with other studies. Environ. Health Perspect. 110, 1169–1173.[ISI][Medline]

Bradford, M. M. (1976). A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 72, 248–254.[CrossRef][ISI][Medline]

Brouwer, A., Ahlborg, U. G., van Leeuwen, F. X., and Feeley, M. M. (1998a). Report of the WHO working group on the assessment of health risks for human infants from exposure to PCDDs, PCDFs and PCBs. Chemosphere 37, 1627–1643.[CrossRef][ISI][Medline]

Brouwer, A., Morse, D. C., Lans, M. C., Schuur, A. G., Murk, A. J., Klasson-Wehler, E., Bergman, A., and Visser, T. J. (1998b). Interactions of persistent environmental organohalogens with the thyroid hormone system: Mechanisms and possible consequences for animal and human health. Toxicol. Indust. Health 14, 59–84.[ISI][Medline]

Burgin, D. E., Diliberto, J. J., Derr-Yellin, E. C., Kannan, N., Kodavanti, P. R., and Birnbaum, L. S. (2002). Differential effects of two lots of aroclor 1254 on enzyme induction, thyroid hormones, and oxidative stress. Environ. Health Perspect. 109, 1163–1168.[ISI]

Burke, M. D., Thompson, S., Elcombe, C. R., Halpert, J., Haaparanta, T., and Mayer, R. T. (1985). Ethoxy-, pentoxy- and benzyloxyphenoxazones and homologues: A series of substrates to distinguish between different induced cytochromes P-450. Biochem. Pharmacol. 34, 3337–3345.[CrossRef][ISI][Medline]

Bush, B., Snow, J., and Bennett, A. H. (1986). Polychlorobiphenyl congeners, p-p'-DDE, and sperm function in humans. Arch. Environ. Contam. Toxicol. 15, 333–341.[ISI][Medline]

Cai, Y., Konishi, T., Han, G., Campwala, K. H., French, S. W., and Wan, Y. J. Y. (2002). The role of hepatocyte RXR alpha in xenobiotic-sensing nuclear receptor-mediated pathways. Eur. J. Pharm. Sci. 15, 89–96.[CrossRef][ISI][Medline]

Chaffin, C. L., Trewin, A. L., and Hutz, R. J. (2000). Estrous cycle-dependent changes in the expression of aromatic hydrocarbon receptor (AHR) and AHR-nuclear translocator (ARNT) mRNAs in the rat ovary and liver. Chem. Biol. Interact. 124, 205–216.[CrossRef][ISI][Medline]

Chen, I., Hsieh, T., Thomas, T., and Safe, S. (2001). Identification of estrogen-induced genes downregulated by AhR agonists in MCF-7 breast cancer cells using suppression subtractive hybridization. Gene 262, 207–214.[CrossRef][ISI][Medline]

Chen, Z. Y., and Eaton, D. L. (1991). Differential regulation of cytochrome(s) P450 2B1/2 by phenobarbital in hepatic hyperplastic nodules induced by aflatoxin B1 or diethylnitrosamine plus 2-acetylaminofluorene in male F344 rats. Toxicol. Appl. Pharmacol. 111, 132–144.[ISI][Medline]

Chu, I., Lecavalier, P., Hakansson, H., Yagminas, A., Valli, V. E., Poon, R., and Feeley, M. (2001). Mixture effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin and polychlorinated biphenyl congeners in rats. Chemosphere 43, 807–814.[CrossRef][ISI][Medline]

Craan, A. G., and Haines, D. A. (1998). Twenty-five years of surveillance for contaminants in human breast milk. Arch. Environ. Contam. Toxicol. 35, 702–710.[CrossRef][ISI][Medline]

Croissandeau, G., Schussler, N., Grouselle, D., Pagesy, P., Rauch, C., Bayet, M. C., Peillon, F., and Le Dafniet, M. (1996). Evidence of thyrotropin-releasing hormone (TRH) gene expression in rat anterior pituitaries and modulation by estrogens of TRH-like immunoreactivity and TRH-elongated peptide contents. J. Endocrinol. 151, 87–96.[Abstract]

De Jongh, J., Wondergem, F., Seinen, W., and van den Berg, M. (1993). Toxicokinetic interactions between chlorinated aromatic hydrocarbons in the liver of the C57BL/6J mouse: I. Polychlorinated biphenyls (PCBs). Arch. Toxicol. 67, 453–460.[ISI][Medline]

Denison, M. S., Pandini, A., Nagy, S. R., Baldwin, E. P., and Bonati, L. (2002). Ligand binding and activation of the Ah receptor. Chem. Biol. Interact. 141, 3–24.[CrossRef][ISI][Medline]

Desaulniers, D., Leingartner, K., Russo, J., Perkins, G., Chittim, B. G., Archer, M. C., Wade, M., and Yang, J. (2001). Modulatory effects of neonatal exposure to TCDD, or a mixture of PCBs, p,p'-DDT and p. p'-DDE, on methylnitrosourea-induced mammary tumor development in the rat. Environ. Health Perspect. 109, 739–747.

Desaulniers, D., Leingartner, K., Wade, M., Fintelman, E., Yagminas, A., and Foster, W. G. (1999). Effects of acute exposure to PCB 126 and 153 on anterior pituitary and thyroid hormones and FSH isoforms in adult Sprague Dawley male rats. Toxicol. Sci. 47, 158–169.[Abstract]

Devanesan, P., Todorovic, R., Zhao, J., Gross, M. L., Rogan, E. G., and Cavalieri, E. L. (2001). Catechol estrogen conjugates and DNA adducts in the kidney of male Syrian golden hamsters treated with 4-hydroxyestradiol: Potential biomarkers for estrogen-initiated cancer. Carcinogenesis 22, 489–497.[Abstract/Free Full Text]

Dewailly, E., Ayotte, P., Laliberte, C., Weber, J. P., Gingras, S., and Nantel, A. J. (1996). Polychlorinated biphenyl (PCB) and dichlorodiphenyl dichloroethylene (DDE) concentrations in the breast milk of women in Quebec. Am. J. Public Health 86, 1241–1246.[Abstract]

Dewailly, E., Nantel, A., Bruneau, S., Laliberte, C., Ferron, L., and Gingras, S. (1992). Breast milk contamination by PCDDs, PCDFs and PCBs in arctic Quebec: A preliminary assessment. Chemosphere 25, 1245–1249.[CrossRef][ISI]

Feeley, M., and Brouwer, A. (2000). Health risks to infants from exposure to PCBs, PCDDs and PCDFs. Food Addit. Contam. 17, 325–333.[CrossRef][ISI][Medline]

Foster, W., Chan, S., Platt, L., and Hughes, C. (2000). Detection of endocrine disrupting chemicals in samples of second trimester human amniotic fluid. J. Clin. Endocrinol. Metab. 85, 2954–2957.[Abstract/Free Full Text]

Frank, R., Rasper, J., Smout, M. S., and Braun, H. E. (1988). Organochlorine residues in adipose tissues, blood and milk from Ontario residents, 1976–1985. Can. J. Public Health 79, 150–158.[ISI][Medline]

Fujii-Kuriyama, Y., Mizukami, Y., Kawajiri, K., Sogawa, K., and Muramatsu, M. (1982). Primary structure of a cytochrome P-450: Coding nucleotide sequence of phenobarbital-inducible cytochrome P-450 cDNA from rat liver. Proc. Natl. Acad. Sci. U.S.A. 79, 2793–2797.[Abstract]

Golden, R. J., Noller, K. L., Titus-Ernstoff, L., Kaufman, R. H., Mittendorf, R., Stillman, R., and Reese, E. A. (1998). Environmental endocrine modulators and human health: An assessment of the biological evidence. Crit. Rev. Toxicol. 28, 109–227.[ISI][Medline]

Goldman, D., and Yawetz, A. (1992). The interference of polychlorinated biphenyls (Aroclor 1254) with membrane regulation of the activities of cytochrome P-450C21 and P-450(17) alpha lyase in guinea-pig adrenal microsome. J. Steroid Biochem. Mol. Biol. 42, 37–47.[CrossRef][ISI][Medline]

Gray, L. E., Kelce, W. R., Wiese, T., Tyl, R., Gaido, K., Cook, J., Klinefelter, G., Desaulniers, D., Wilson, E., Zacharewski, T., et al. (1997). Endocrine screening methods workshop report: Detection of estrogenic and androgenic, hormonal and antihormonal activity, for chemicals that act via receptor or sterodogenic enzyme mechanisms. Reprod. Toxicol. 11, 719–750.[CrossRef][ISI][Medline]

Guo, T. L., Zhang, X. L., Bartolucci, E., McCay, J. A., White, K. L. J., and You, L. (2002). Genistein and methoxychlor modulate the activity of natural killer cells and the expression of phenotypic markers by thymocytes and splenocytes in F0 and F1 generations of Sprague-Dawley rats. Toxicology 172, 205–215.[CrossRef][ISI][Medline]

Haines, D., Laube, V., Jordan, S., Mortimer, J., Gupta, S., and Arvanitakis, G. (1998). Persistent Environmental Contaminants and the Great Lakes Basin Population: An Exposure Assessment, pp. 1–358. Minister of Health, Ottawa, Canada.

Hakkola, J., Raunio, H., Purkunen, R., Saarikoski, S., Vahakangas, K., Pelkonen, O., Edward, R. J., Boobis, A. R., and Pasanen, M. (2001). Cytochrome P450 3A expression in the human fetal liver: Evidence that CYP3A5 is expressed in only a limited number of fetal livers. Biol. Neonate 80, 193–201.[CrossRef][ISI][Medline]

Haller, C. A., and Simpser, E. (1999). Breastfeeding: 1999 perspective. Curr. Opin. Pediatr. 11, 379–383.[CrossRef][Medline]

Harvey, P. W., Malarkey, P., and Cockburn, A. (1999). The endocrine-disrupting properties of dioxin (TCDD): The potential role of the adrenal cortex and glucocorticoid receptor in contributing to the dioxin toxicity syndrome. In Endocrine and Hormonal Toxicology (P. W. Harvey, K. C. Rush, and A. Cockburn, Eds.), pp. 535–554. John Wiley & Sons, Toronto, Canada.

Healing, G. (1999). Hormonal mechanisms in kidney toxicity. In Endocrine and Hormonal Toxicology (P. W. Harvey, K. C. Rush, and A. Cockburn, Eds.), pp. 291–307. John Wiley & Sons, Toronto, Canada.

Hines, R. N., and McCarver, D. G. (2002). The ontogeny of human drug-metabolizing enzymes: Phase I oxidative enzymes. J. Pharmacol. Exp. Ther. 300, 355–360.[Abstract/Free Full Text]

Hinson, J. P., and Raven, P. W. (1999). Adrenal toxicology. In Endocrine and Hormonal Toxicology (P. W. Harvey, K. C. Rush, and A. Cockburn, Eds.), pp. 67–89. John Wiley & Sons, Toronto, Canada.

Howdeshell, K. L. (2002). A model of the development of the brain as a construct of the thyroid system. Environ. Health Perspect. 110(Suppl. 3), 337–348.

Hruska, R. E., and Olson, J. R. (1989). Species differences in estrogen receptors and in the response to 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure. Toxicol. Lett. 48, 289–299.[CrossRef][ISI][Medline]

Iba, M. M., Fung, J., Cooper, K. R., Thomas, P. E., Wagner, G. C., and Park, Y. (2000). Effect of gestational and lactational 2,3,7, 8-tetrachlorodibenzo-p-dioxin exposure on the level and catalytic activities of hepatic microsomal CYP1A in prepubertal and adult rats. Biochem. Pharmacol. 59, 1147–1154.[CrossRef][ISI][Medline]

Ito, A., Buenafe, A. C., Matejuk, A., Zamora, A., Silverman, M., Dwyer, J., Vandenbark, A. A., and Offner, H. (2002). Estrogen inhibitis systemic T cell expression of TNF-alpha and recruitment of TNF-alpha(+) T cells and macrophages into the CNS of mice developing experimental encephalomyelitis. Clin. Immunol. 102, 275–282.[CrossRef][ISI][Medline]

Jansen, H. T., Cooke, P. S., Porcelli, J., Liu, T.-C., and Hansen, L. G. (1993). Estrogenic and antiestrogenic actions of PCBs in the female rat: In vitro and in vivo studies. Reprod. Toxicol. 7, 237–248.[CrossRef][ISI][Medline]

Jarrell, J. F., Villeneuve, D., Franklin, C., Bartlett, S., Wrixon, W., Kohut, J., and Zouves, C. G. (1993). Contamination of human ovarian follicular fluid and serum by chlorinated organic compounds in three Canadian cities. CMAJ 148, 1321–1327.[Abstract]

Jensen, K. G., Wiberg, K., Klasson-Wehler, E., and Onfelt, A. (2000). Induction of aberrant mitosis with PCBs: Particular efficiency of 2, 3,3',4,4'-pentachlorobiphenyl and synergism with triphenyltin. Mutagenesis 15, 9–15.[Abstract/Free Full Text]

Kanno, J., Onyon, L., Haseman, J., Fenner-Crisp, P., Ashby, J., and Owens, W. (2001). The OECD program to validate the rat uterotrophic bioassay to screen compounds for in vivo estrogenic responses: Phase 1. Environ. Health Perspect. 109, 785–794.[ISI][Medline]

Karpuzoglu-Sahin, E., Hissong, B. D., and Ansar-Ahmed, S. (2001). Interferon-gamma levels are upregulated by 17-beta-estradiol and diethylstilbestrol. J. Reprod. Immunol. 52, 113–127.[CrossRef][ISI][Medline]

Kent, U. M., Mills, D. E., Rajnarayanan, R. V., Alworth, W. L., and Hollenberg, P. F. (2002). Effect of 17-{alpha}-ethynylestradiol on activities of cytochrome P450 2B (P450 2B) enzymes: Characterization of inactivation of P450s 2B1 and 2B6 and identification of metabolites. J. Pharmacol. Exp. Ther. 300, 549–558.[Abstract/Free Full Text]

Khan, M. A., Lichtensteiger, C. A., Faroon, O., Mumtaz, M., Schaeffer, D. J., and Hansen, L. G. (2002). The hypothalamo-pituitary-thyroid (HPT) axis: A target of nonpersistent ortho-substituted PCB congeners. Toxicol. Sci. 65, 52–61.[Abstract/Free Full Text]

Kimura, N., Arai, K., Sahara, Y., and Suzuki, H. (1994). Estradiol transcriptionally and posttranscriptionally up-regulates thyrotropin-releasing hormone receptor messenger ribonucleic acid in rat pituitary cells. Endocrinology 134, 432–440.[Abstract]

Landrigan, P. J., Sonawane, B., Mattison, D., McCally, M., and Garg, A. (2002). Chemical contaminants in breast milk and their impacts on children’s health: An overview. Environ. Health Perspect. 110, A313–A315.[ISI][Medline]

Larsen-Su, S. A., and Williams, D. E. (2001). Transplacental exposure to indole-3-carbinol induces sex-specific expression of CYP1A1 and CYP1B1 in the liver of Fischer 344 neonatal rats. Toxicol. Sci. 64, 162–168.[Abstract/Free Full Text]

Lee, S. K., Ou, Y. C., and Yang, R. S. H. (2002). Comparison of pharmacokinetic interactions and physiologically based pharmacokinetic modeling of PCB 153 and PCB 126 in nonpregnant mice, lactating mice, and suckling pups. Toxicol. Sci. 65, 26–34.[Abstract/Free Full Text]

Li, M.-H., and Hansen, L. G. (1996). Enzyme induction and acute endocrine effects in prepubertal female rats receiving environmental PCB/PCDF/PCDD mixtures. Environ. Health Perspect. 104, 712–722.[ISI][Medline]

Li, M.-H., and Hansen, L. G. (1997). Consideration of enzyme and endocrine interactions in the risk assessment of PCBs. Rev. Toxicol. 1, 71–156.

Lin, T. M., Ko, K., Moore, R. W., Buchanan, D. L., Cooke, P. S., and Pederson, R. E. (2001). Role of the aryl hydrocarbon receptor in the development of control and 2,3,7,8-tetrachlorodibenzo-p-dioxin-exposed male mice. J. Toxicol. Environ. Health 64, 327–342.[CrossRef][ISI]

Lisboa, P. C., Curty, F. H., Moreina, R. M., and Pazos-Moura, C. C. (1997). Effects of estradiol benzoate on 5'-iodothyronine deiodinase activities in female rat anterior pitiutary gland, liver and thyroid gland. Braz. J. Med. Biol. Res. 30, 1479–1484.[ISI][Medline]

Lorber, M., and Phillips, L. (2002). Infant exposure to dioxin-like compounds in breast milk. Environ. Health Perspect. 110, A325–A332.[ISI][Medline]

Masahiko, N., and Honkakoski, P. (2000). Induction of drug metabolism by nuclear receptor CAR: Molecular mechanisms and implications for drug research. Eur. J. Pharm. Sci. 11, 259–264.[CrossRef][ISI][Medline]

Mattson, M. P. (1999). Hormonal mechanisms in neurotoxicity. In Endocrine and Hormonal Toxicology (P. W. Harvey, K. C. Rush, and A. Cockburn, Eds.), pp. 209–225. John Wiley & Sons Ltd, Toronto, Canada.

McVea, K. L., Turner, P. D., and Peppler, D. K. (2000). The role of breastfeeding in sudden infant death syndrome. J. Hum. Lact. 16, 13–20.[Abstract/Free Full Text]

Miyata, M., Nagata, K., Shimada, M., Yamazoe, Y., and Kato, R. (1994). Structure of a gene and cDNA of a major constitutive form of testosterone 6 beta-hydroxylase (P450/6 beta A) encoding CYP3A2: Comparison of the cDNA with P450PCN2. Arch. Biochem. Biophys. 314, 351–359.[CrossRef][ISI][Medline]

Nakamura, M., Ikeda, Y., Mine, M., Tomiyoshi, Y., and Sakemi, T. (2001). Somatostatin analogue attenuates estrogen-induced augmentation of glomerular injury in spontaneous hypercholesterolemic female Imai rats. Nephron 89, 448–454.[CrossRef][ISI][Medline]

Namkung, M. J., Yang, H. L., Hulla, J. E., and Junchau, M. R. (1988). On the substrate specificity of cytochrome P450IIIA1. Mol. Pharmacol. 34, 628–637.[Abstract]

Nerurkar, P. V., Park, S. S., Thomas, P. E., Nims, R. W., and Lubet, R. A. (1993). Methoxyresorufin and benzyloxyresorufin: Substrates preferentially metabolized by cytochromes P4501A2 and 2B, respectively, in the rat and mouse. Biochem. Pharmacol. 46, 933–943.[CrossRef][ISI][Medline]

Newbold, R. R., Jefferson, W. N., Padilla-Banks, E., Walker, V. R., and Pena, D. S. (2001). Cell response endpoints enhance sensitivity of the immature mouse uterotropic assay. Reprod. Toxicol. 15, 245–252.[CrossRef][ISI][Medline]

Newsome, W. H., and Ryan, J. J. (1999). Toxaphene and other chlorinated compounds in human milk from northern and southern Canada: A comparison. Chemosphere 39, 519–526.[CrossRef][ISI][Medline]

Norén, K., and Meironyté, D. (2000). Certain organochlorine and organobromine contaminants in Swedish human milk in perspective of past 20–30 years. Chemosphere 40, 1111–1123.[CrossRef][ISI][Medline]

Nudel, U., Zakut, R., Shani, M., Neuman, S., Levy, Z., and Yaffe, D. (1983). The nucleotide sequence of the rat cytoplasmic beta-actin gene. Nucleic Acids Res. 11, 1759–1771.[Abstract]

Oddy, W. H. (2001). Breastfeeding protects against illness and infection in infants and children: A review of the evidence. Breastfeed. Rev. 9, 11–18.[Medline]

Odum, J., Lefevre, P. A., Tittensor, S., Paton, D., Routledge, E. J., Beresford, N. A., Sumpter, J. P., and Ashby, J. (1997). The rodent uterotrophic assay: Critical protocol features, studies with nonyl phenols, and comparison with a yeast estrogenicity assay. Regul. Toxicol. Pharmacol. 25, 176–188.[CrossRef][ISI][Medline]

Ohtake, F., Takeyama, K., Matsumoto, T., Kitagawa, H., Yamamoto, Y., Nohara, K., Tohyama, C., Krust, A., Mimura, J., Chambon, P., et al. (2003). Modulation of oestrogen receptor signalling by association with the activated dioxin receptor. Nature 423, 545–550.[CrossRef][ISI][Medline]

Padilla-Banks, E., Jefferson, W. N., and Newbold, R. R. (2001). The immature mouse is a suitable model for detection of estrogenicity in the uterotropic bioassay. Environ. Health Perspect. 109, 821–826.[ISI][Medline]

Parkinson, A., Robertson, L., and Safe, S. (1980). Reconstituted human breast milk PCBs as potent inducers of aryl hydrocarbon hydroxylase. Biochem. Biophys. Res. Comm. 96, 882–889.[ISI][Medline]

Pauwels, A., Cenijn, P. H., Schepens, P. J., and Brouwer, A. (2000). Comparison of chemical-activated luciferase gene expression bioassay and gas chromatography for PCB determination in human serum and follicular fluid. Environ. Health Perspect. 108, 553–557.[ISI][Medline]

Petroff, B. K., Gao, X., Rozman, K. K., and Terranova, P. F. (2001a). The effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on weight gain and hepatic ethoxyresorufin-o-deethylase (EROD) induction vary with ovarian hormonal status in the immature gonadotropin-primed rat model. Reprod. Toxicol. 15, 269–274.[CrossRef][ISI][Medline]

Petroff, B. K., Roby, K. F., Gao, X., Son, D., Williams, S., Johnson, D., Rozman, K. K., and Terranova, P. F. (2001b). A review of mechanisms controlling ovulation with implications for the anovulatory effects of polychlorinated dibenzo-p-dioxins in rodents. Toxicology 158, 91–107.[CrossRef][ISI][Medline]

Petrulis, J. R., and Bunce, N. J. (2000). Competitive behavior in the interactive toxicology of halogenated aromatic compounds. J. Biochem. Mol. Toxicol. 14, 73–81.[CrossRef][ISI][Medline]

Pletta, K. H., Eglash, A., and Choby, K. (2000). Benefits of breastfeeding: A review for physicians. Wis. Med. J. 99, 55–58.

Pronczuk, J., Akre, J., Moy, G., and Vallenas, C. (2002). Global perspectives in breast milk contamination: Infectious and toxic hazards. Environ. Health Perspect. 110, A349–A351.[ISI][Medline]

Reen, R. K., Cadwallader, A., and Perdew, G. H. (2002). The subdomains of the transactivation domain of the aryl hydrocarbon receptor (AhR) inhibit AhR and estrogen receptor transcriptional activity. Arch. Biochem. Biophys. 408, 93–102.[CrossRef][ISI][Medline]

Ribas-Fito, N., Cardo, E., Sala, M., Eulalia, D. M., Mazon, C., Verdu, A., Kogevinas, M., Grimalt, J. O., and Sunyer, J. (2003). Breastfeeding, exposure to organochlorine compounds, and neurodevelopment in infants. Pediatrics 111, e580–e585.[Abstract/Free Full Text]

Safe, S. (1994). Polychlorinated biphenyls (PCBs): Environmental impact, biochemical and toxic responses, and implications for risk assessment. C.R.C. Crit. Rev. Toxicol. 24, 87–149.

Safe, S. (2001). PCBs as aryl hydrocarbon receptor agonists: Implication for risk assessment. In PCBs: Recent Advances in Environmental Toxicology and Health Effects (L. W. Robertson and L. G. Hansen, Eds.), pp. 171–177. The University Press of Kentucky, Lexington, KY.

Safe, S. H. (1999). 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related environmental antiestrogens: Characterization and mechanism of action. In Endocrine Disruptors: Effects on Male and Female Reproductive Systems (R. K. Naz, Ed.), pp. 187–221. CRC Press, New York.

Sarkar, S., Jana, N. R., Yonemoto, J., Tohyama, C., and Sone, H. (2000). Estrogen enhances induction of cytochrome P-4501A1 by 2,3,7, 8-tetrachlorodibenzo-p-dioxin in liver of female Long-Evans rats. Int. J. Oncol. 16, 141–147.[ISI][Medline]

SAS Institute (1998). JMP Statistics and Graphics Guide, Version 3, pp. 1–593. SAS Institute, Cary, NC.

Schecter, A., McGee, H., Stanley, J. S., Boggess, K., and Brandt-Rauf, P. (1996). Dioxins and dioxin-like chemicals in blood and semen of American Vietnam veterans from the state of Michigan. Am. J. Ind. Med. 30, 647–654.[CrossRef][ISI][Medline]

Schecter, A. J., and Piskac, A. L. (2001). PCBs, dioxins, and dibenzofurans: Measured levels and toxic equivalents in blood, milk and food from various countries. In PCBs: Recent Advances in Environmental Toxicology and Health Effects (L. W. Robertson and L. G. Hansen, Eds.), pp. 161–168. The University Press of Kentucky, Lexington, KY.

Schomburg, L., and Bauer, K. (1997). Regulation of the adenohypophyseal thyrotropin-releasing hormone-degrading ectoenzyme by estradiol. Endocrinology 138, 3587–3593.[Abstract/Free Full Text]

Serabjit-Singh, C. J., Albro, P. W., Robertson, I. G. C., and Philpot, R. M. (1983). Interactions between xenobiotics that increase or decrease the levels of cytochrome P-450 isozymes in rabbit lung and liver. J. Biol. Chem. 258, 12827–12834.[Abstract/Free Full Text]

Solomon, G. M., and Weiss, P. M. (2002). Chemical contaminants in breast milk: Time trends and regional variability. Environ. Health Perspect. 110, A339–A347.[ISI][Medline]

Sommer, R. J., Sojka, K. M., Pollenz, R. S., Cooke, P. S., and Peterson, R. E. (1999). Ah receptor and ARNT protein and mRNA concentrations in rat prostate: Effects of stage of development and 2,3,7,8-tetrachlorodibenzo-p-dioxin treatment. Toxicol. Appl. Pharmacol. 155, 177–189.[CrossRef][ISI][Medline]

Spreafico, E., Bettini, E., Pollio, G., and Maggi, A. (1992). Nucleotide sequence of estrogen receptor cDNA from Sprague-Dawley rat. Eur. J. Pharmacol. 227, 353–356.[CrossRef][Medline]

Sueyoshi, T., and Negishi, M. (2001). Phenobarbital response elements of cytochrome P450 genes and nuclear receptors. Annu. Rev. Pharmacol. Toxicol. 41, 123–143.[CrossRef][ISI][Medline]

Tian, Y., Rabson, A. B., and Gallo, M. A. (2002). Ah receptor and NF-kappaB interactions: Mechanisms and physiological implications. Chem. Biol. Interact. 141, 97–115.[CrossRef][ISI][Medline]

Tryphonas, H., and Feeley, M. (2001). Polychlorinated biphenyl-induced immunomodulation and human health effects. In: PCBs: Recent advances in environmental toxicology and health effects (L. W. Robertson and L. G. Hansen, Eds.), pp. 193–209. The University Press of Kentucky, Lexington, KY.

Ueda, A., Hamadeh, H. K., Webb, H. K., Yamamoto, Y., Sueyoshi, T., Afshari, C. A., Lehmann, J. M., and Negishi, M. (2002). Diverse roles of the nuclear orphan receptor CAR in regulating hepatic genes in response to phenobarbital. Mol. Pharmacol. 61, 1–6.[Abstract/Free Full Text]

van Birgelen, A. P. J. M., DeVito, M. J., Akins, J. M., Ross, D. G., Diliberto, J. J., and Birnbaum, L. S. (1996a). Relative potencies of polychlolrinated dibenzo-p-dioxins, dibenzofurans, and biphenyls derived from hepatic porphyrin accumulation in mice. Toxicol. Appl. Pharmacol. 138, 98–109.[CrossRef][ISI][Medline]

van Birgelen, A. P. J. M., Fase, K. M., van der Kolk, J., Poiger, H., Brouwer, A., Seinen, W., and van den Berg, M. (1996b). Synergistic effect of 2,2',4,4',5,5'-hexachlorobiphenyl and 2,3,7,8-tetrachlorodibenzo-p-dioxin on hepatic porphyrin levels in the rat. Environ. Health Perspect. 104, 550–557.[ISI][Medline]

van Birgelen, A. P. J. M., van der Kolk, J., Fase, K. M., Bol, I., Poiger, H., Brouwer, A., and Van der Berg, M. (1994a). Toxic potency of 3,3',4,4',5-pentachlorobiphenyl relative to and in combination with 2,3,7,8-tetrachlorodibenzo-p-dioxin in a subchronic feeding study in the rat. Toxicol. Appl. Pharmacol. 127, 209–221.[CrossRef][ISI][Medline]

van Birgelen, A. P. J. M., van der Kolk, J., Fase, K. M., Bol, I., Poiger, H., van den Berg, M., and Brouwer, A. (1994b). Toxic potency of 2,3,3',4,4',5-hexachlorobiphenyl relative to and in combination with 2,3,7,8-tetrachlorodibenzo-p-dioxin in a subchronic feeding study in the rat. Toxicol. Appl. Pharmacol. 126, 202–213.[CrossRef][ISI][Medline]

Van den Berg, M., Birnbaum, L., Bosveld, A. T. C., Brunstrom, B., Cook, P., Feeley, M., Giesy, J. P., Hanberg, A., Hasegawa, R., Kennedy, S. W., et al. (1998). Toxic equivalency factors (TEFs) for PCBs, PCDDs, PCDFs for humans and wildlife. Environ. Health Perspect. 106, 775–792.[ISI][Medline]

van der Kolk, J., van Birgelen, A. P. J. M., Poiger, H., and Schlatter, C. (1992). Interactions of 2,2',4,4',5,5'-hexachlorobiphenyl and 2,3,7,8-tetrachlorodibenzo-p-dioxin in a subchronic feeding study in the rat. Chemosphere 25, 2023–2027.[CrossRef][ISI]

van Leeuwen, F. X. R., Feeley, M., Schrenk, D., Larsen, J. C., Farland, W. H., and Younes, M. (2000). Dioxins: WHO’s tolerable daily intake (TDI) revisited. Chemosphere 40, 1095–1101.[CrossRef][ISI][Medline]

Vansell, N. R., and Klaassen, C. D. (2002a). Effect of microsomal enzyme inducers on the biliary excretion of triiodothyronine (T3) and its metabolites. Toxicol. Sci. 65, 184–191.[Abstract/Free Full Text]

Vansell, N. R., and Klaassen, C. D. (2002b). Increase in rat liver UDP-glucuronosyltransferase mRNA by microsomal enzyme inducers that enhance thyroid hormone glucuronidation. Drug Metab. Dispos. 30, 240–246.[Abstract/Free Full Text]

Voorman, R., and Aust, S. D. (1987). Specific binding of polyhalogenated aromatic hydrocarbon inducers of cytochrome P-450d to the cytochrome and inhibition of its estradiol 2-hydroxylase activity. Toxicol. Appl. Pharmacol. 90, 69–78.[ISI][Medline]

Wade, M. G., Parent, S., Finnson, K. W., Foster, W., Younglai, E., McMahon, A., Cyr, D. G., and Hughes, C. (2002). Thyroid toxicity due to subchronic exposure to a complex mixture of 16 organochlorines, lead, and cadmium. Toxicol. Sci. 67, 207–218.[Abstract/Free Full Text]

Weiss, B. (2002). Sexually dimorphic nonreproductive behaviors as indicators of endocrine disruption. Environ. Health Perspect. 110(Suppl. 3), 387–391.

White, T. E. K., Rucci, G., Liu, Z., and Gasiewicz, T. A. (1995). Weanling female Sprague-Dawley rats are not sensitive to the antiestrogenic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol. Appl. Pharmacol. 133, 313–320.[CrossRef][ISI][Medline]

World Health Organization (WHO) (1996). Environmental Health in Europe No. 3. Levels of PCBs, PCDDs and PCDFs in human milk. Second round of WHO-coordinated exposure study, pp. 1–121. Copenhagen, Denmark.

Zacharewski, T., and Safe, S. (1998). Antiestrogenic activity of TCDD and related compounds. In Reproductive and Developmental Toxicology (K. S. Korach, Ed.), pp. 431–448. Marcel Dekker, New York.

Zelko, I., and Negishi, M. (2000). Phenobarbital-elicited activation of nuclear receptor CAR in induction of cytochrome P450 genes. Biochem. Biophys. Res. Commun. 277, 1–6.[CrossRef][ISI][Medline]

Zoeller, R. T., Dowling, A. L. S., Herzig, C. T. A., Iannacone, E. A., Gauger, K. J., and Bansal, R. (2002). Thyroid hormone, brain development, and the environment. Environ. Health Perspect. 110(Suppl. 3), 355–361.