r nwn‘ .3 v-m. w: y.» 1: ..,.4.._ .. ~ ..I‘..... . . ..Z. H. In“).- --m 1! iii/ix/iii/iiwijijifm“iii/717W ‘ 3198 ' mos: ((995? This is to certify that the /thesis entitled Ziierrj ”F3 3,144d/‘f‘Q't’YCLC/LYZM Lot/>1 {Way/g O Vk W f’)099.4%) was purchased from AccuStandard Inc. , New Haven, CT. Liquid scintillation cocktail was purchased from Beckman Instruments, Inc., Fullerton, CA. Acetonitrile (HPLC grade) was purchased from EM Science, Gibbstown, NJ. Acetone was purchased from J. T. Baker Inc., Phillipsburg, NJ. Feed Preparation In this feeding trial, 1‘C-TCB was used to measure placental transfer of TCB as well as distribution of TCB in tissues. Two types of feed were prepared: “C-TCB feed (“C-TCB and TCB) and TCB feed. 33 34 “W Feed contained 0, 3 ppm, or 30 ppm total TCB. Purchased l“C-TCB (50 pCi/50 pl) was diluted by a factor of 20 with toluene to make a 50 pCi/1 ml stock solution. Acetone was used as a carrier to disperse l‘C-TCB and TCB in ground mouse chow 5015 (PMI Feeds, Inc., St. Louis, MO). After 9.52 pCi l‘C-TCB in 190.5 pl toluene and TCB were dissolved in 60 ml of acetone, 1 kg ground mouse chow was added and mixed. The specific radioactivity was 3.17 and 0.317 pCi/mg TCB in 3 ppm and 30 ppm “C-TCB feed, respectively. The feed was tumbled for 1 hour and air dried in a hood at room temperature for 24 hours, before being stored in glass jars in a 04°C refrigerator until use. The control feed consisted of ground mouse chow mixed with acetone and toluene and similarly tumbled and dried. Feed samples were analyzed by the laboratory of Dr. Thomas Voice, Department of Civil and Environmental Engineering, Michigan State University, at the onset of this study. 193.1524 Feed containing TCB only was prepared using the same procedure described above but without l‘C-TCB. The feed was prepared monthly. Treatments Before mating, 8 female C57BL/6J mice (F—O) at 15 weeks of age were randomly assigned to each treatment and provided with “C-TCB feed ad libitum for 2 weeks. Each female was then paired with one non-treated mature C57BL/6J male for 10 days between 7:30 PM and 7:30 AM, during which time feed was withheld to avoid exposing males to treatment feed. The presence of the vaginal plug was defined as day 1 of 35 gestation. F—O females continued on the “C-TCB treatment until parturition and were then switched to a TCB diet for the next 21 days. This study was terminated 21 days postpartum when the offspring were weaned. Sample Collection On day 19 of gestation, liver, thymus, uterus, abdominal adipose tissue, placenta, and fetus samples were collected and weighed. These same tissues, except uterus, placenta, and fetus, were also collected on day 21 of lactation. One animal from each treatment group was used for sample collection. One non-pregnant mouse from each treatment was sacrificed concurrently with the animals on day 19 of gestation and day 21 of lactation. The liver, thymus, and abdominal adipose tissue of 3-week-old male offspring were collected, weighed and stored for subsequent analysis. Sample Extraction and Analysis Each sample was prepared for TCB and radioactivity analysis by disruption in 10 ml acetonitrile with a Dismembrator Sonic (sonicator model 550, Fisher Scientific, Pittsburgh, PA) at 20% power for 10 minutes. The sample was then centrifuged at 1500 RPM for 10 minutes. Ten ml liquid scintillation cocktail was added to 5 ml supernatant for the measurement of radioactivity with a liquid scintillation counter (Model 1500 Tri- Carb, Packard Instrument Company, Meriden, CT). The concentrations of TCB equivalents in the tissues and organs were calculated from the radioactivity measured in the tissues and organs and the specific activity in feed (Appendices I and II). The term TCB equivalents refers to TCB and its metabolites. 36 M Animals Ninety C57BL/6J female mice at 13 weeks of age were used in this phase. Non- treated B6D2-Fl males were used as sperm donors for the in vitra fertilization assay. Mice were raised in the same facility as in Phase I. Chemicals TCB was purchased from AccuStandard Inc., New Haven, CT. Pregnant mare’s serum gonadotropin (PMSG), human chorionic gonadotropin (HCG), and all other chemicals were purchased from Sigma Chemical Co., St. Louis, MO. Feed Preparation The method of feed preparation was the same as described in Phase I. Treatments Female C57BL/6] mice (F-O) were fed Purina lab chow containing 0, 3 ppm, and 30 ppm TCB for 2 weeks beginning at 13 weeks of age. Each female was then paired with one non-treated mature C57BL/6J male for 10 days, between 7:30 PM and 7:30 AM, during which time feed was withheld to avoid exposing males to treatment feed. The treatment for F—O females continued through mating, gestation, and lactation. The offspring (F-l) were given the same feed as their dams received throughout the study. Females (F—l) were superovulated at 5, 6, and 7 weeks of age and egg fertilizing ability was assessed by in vitra fertilization (IVF) assay. At 7 weeks of age, 10 F-l females in each treatment group were bred with non-treated mature B6D2-F1 males for 10 days. Ten treated F-l males in each treatment group, aged 7 and 17 weeks, were used to breed 37 non-treated mature B6D2-F1 females for 10 days. After breeding, sperm from these males were collected and fertilizing ability was assessed by IVF assay. Determination of sperm concentration and sperm motion analysis were performed on sperm from 9- week-old F-l males hourly for 4.5 hours after sperm collection with the CellSoft computer-assisted digital image analysis system (CRYO Resources Inc., New York, 1986). Parameters Measured W Feed intake was recorded for F-0 females before mating, during gestation, and during lactation. The amount of feed added every day was recorded. Actual feed consumption was calculated every 3 days by subtracting the amount of feed left in the feed jars from the total feed provided over the 3-day period. W Body weights of F—O females were recorded at the initiation of the study (day 1), before breeding (day 14), on day 19 of gestation (day 33), and on day 21 of lactation (day 54). When the offspring were 4 days, 1 week, and 2 weeks of age, body weights were recorded by litters. The average weight of offspring in a litter was used for statistical analysis. After weaning, individual weights were recorded weekly. Might Liver and thymus weights of F-O females were recorded for animals on day 19 of gestation and on day 21 of lactation. Liver and thymus weights of El females were recorded at 5, 6, and 7 weeks of age. Liver, thymus, and testis weights of F-l males were determined at 3, 9, and 19 weeks of age. WWW Fecundity. litter sizes, sex ratios, and 4-day and 21-day survival indices were recorded for both F-0 and F-l 38 generations. Sex of pups was determined by anogenital distances at day 4 of age or at death if they died before day 4 of age. If pups died before sex could be determined, they were not included. A 4—day survival index is defined as 100 x [number of pups viable at day 4 of age]/[number of viable pups born]. A 21—day survival index is defined as 100 x [number of pups viable at day 21 of age]/[number of pups retained at day 4 of age] (Thomas, 1991). W Treated F-l females were superovulated with 8 IU PMSG, and then 8 [U HCG 48-50 hours later. Thirteen hours after HCG injection, the females were killed by cervical dislocation. Oviducts and distal portions of the uteri were excised. The eggs were collected by gentle teasing from the oviducts and incubated 5-15 minutes prior to insemination. Epididymal sperm from non-treated B6D2-F1 males were collected and incubated for 1.5 hours before insemination. Eggs in each petri dish, collected from one female, were inseminated with 50 pl sperm suspension (approximately 6 x 10’ sperm) and incubated for 24 hours. Incubation occurred in modified Tyrodes medium at 37°C and 5 % CO2 after collection and insemination. Twenty-four hours after insemination, 50 pl of a 35 pM solution of bisBenzimide (Hoechst 33258) were added to the petri dish. It was incubated for another 30 minutes, and then observed under a Nikon Optiphot microscope equipped with a 100w mercury bulb, 365/10 nm excitation filter, 400 nm dichroic mirror, and 400 nm barrier filter. Eggs at the 2-cell stage or eggs containing a second polar body and two pronuclei were considered fertilized. Degenerated eggs were considered non-fertilized. The IVF assay was also used to evaluate sperm fertilizing ability of treated males. 39 The method was the same as described above. Epididymal sperm from treated males at 9 and 19 weeks of age were collected and used to inseminate eggs from non-treated B6D2-F1 mice. Culture Medium Modified Tyrodes media contained 99.23 mM sodium chloride, 2.68 mM potassium chloride, 1.80 mM calcium chloride, 0.36 mM sodium phosphate monobasic anhydrous, 0.49 mM magnesium chloride hexahydrate, 25.0 mM sodium bicarbonate, 52.0 mM sodium lactate, 0.25 mM sodium pyruvate, 5.56 mM D-glucose, 100 IU/ml sodium penicillin G., and 100 IU/ml streptomycin sulphate. 3mm Conoontgation and Motion Aoalysis Twenty pl of the sperm suspension (approximately 2.4 x 10’ sperm/ml) were placed on a CellSoft 20 pm chamber and analyzed with the CellSoft computer-assisted digital image analysis system. A minimum of 100 sperm cells were analyzed to obtain measurements of concentration, motility, velocity, linearity, mean amplitude of lateral head (ALH) displacement, and beat/cross frequency. Motility is the percentage of sperm that travel more than 20 pm/sec. Velocity is the average distance (pm) traveled by motile sperm in one second. Linearity is a measure of whether or not the sperm move in a straight line. It is calculated by dividing the length of the straight line distance by the actual track distance. This fraction is then multipled by 10. Linearity, therefore, ranges from 0 to 10. The straighter the actual cell track, the higher the linearity value such that a value of 10 indicates a perfectly straight line and 0 indicates a circular track. ALH displacement is a measure of the displacement of the sperm head from a computer-calculated curval mean of its track. The maximum ALH displacement is measured from the curval mean for every 4O cycle of the cell’s track, then multipled by 2. For the individual cell data, the mean ALH displacement is the mean of the maximum ALH displacement for the cycles of the track. For the summary data, the mean ALH displacement is an average of the mean ALH displacement for each cell. High ALH displacement appears to be linked with the process of capacitation. The beat/cross frequency (Hz) is the number of beats (or crosses) per second. Every time the sperm cell crosses the computer-calculated curval mean, the computer counts that crossing as one beat. Statistical Analysis Fecundity, survival indices, fertilization rate, degeneration rate, and sperm motility were evaluated with the Chi square test. Body weight, organ weight, feed intake, and ALH displacement were initially tested with One Way Analysis of Variance (ANOVA). Log transformation was performed for the discrete quantitative parameters and ratios (i.e., litter size, number of eggs ovulated, sperm concentration, beat/cross frequency, sperm linearity, sex ratios, and relative organ weights) before ANOVA. Relative organ weights refer to organ weights as percentages of body weights. Those data passing homogeneity of variance and normality tests were analyzed with the Student- Newman-Keuls multiple pairwise comparison. The Kruskal-Wallis Test was performed to evaluate the non-Gaussian data. These analyses were performed with Sigmastat" (Kuo et al., 1992). RESULTS Lime! Results of feed analysis are shown in Table 1. TCB concentrations were approximately 80% of expected concentrations in Phase I and over 90% in Phase II. After 5 weeks of exposure, TCB equivalents in the liver, adipose tissue, uterus, and fetus from the 30 ppm group were 3-6 fold higher than the levels in the 3 ppm group, except for the uterus in the non-pregnant groups (Table 2). Concentrations of TCB equivalents were similar in the uterus, placenta, and fetus in the 30 ppm group (Table 2). No placenta data was available in the 3 ppm group. After 8 weeks of I‘C—TCB treatment, TCB equivalents were still accumulating in the liver and adipose tissue of non-pregnant mice, in both the low and high dose groups (Table 3). TCB equivalents in the thymus of lactating mice were higher than those in the non-pregnant mice, regardless of dose (Table 3). TCB equivalents in the thymus of 30 ppm- treated lactating mice were over 40 times higher than equivalents in the 3 ppm- treated lactating mice (Table 3). In the 3 ppm-treated group, approximately 15 % of the concentrations of TCB equivalents that had accumulated in the liver and adipose tissue before parturition were detected after termination of lactation (Tables 2 and 3). However, in the 30 ppm-treated group, approximately 56% and 4% , respectively, of the concentrations of TCB 41 42 equivalents that had accumulated in the adipose tissue and liver before parturition were observed after termination of lactation (Tables 2 and 3). After weaning, radioactivity was only detected in the liver and adipose tissue, not in the thymus of the offspring (Table 4). Similar concentrations of TCB equivalents were detected in the adipose tissue and liver of the 3 ppm-treated offspring. However, in the 30 ppm group, concentrations of TCB equivalents in the adipose tissue were 14.6 times higher than concentrations in the liver (Fable 4). Although the pups were nursed by the same dam in the 30 ppm group, large variations in the concentrations of TCB equivalents were observed (Table 4). 43 Table 1. TCB concentrations in treatment feed. Treatments Control 3 ppm 30 ppm Concentration (ppm) Phase I‘ 0 2.36 :l: 0.27 23.49 i 1.18 (n=2) (n=3) (n=3) Phase II" 0 3.16 :l: 0.24 27.59 i: 0.70 (n=3) (n=3) (n=3) a The TCB concentrations in feed were calculated from the specific activity in feed and the radioactivity measured in the feed with a liquid scintillation counter (Appendices I and II). b TCB concentrations in Phase II were analyzed by gas chromatography. 44 Table 2. Concentrations of TCB equivalents in tissues and organs of non-pregnant and pregnant mice. Organ/ Concentration' Treatments Tissue Non-pregnantb Pregnant" 3 . 30 3 30 PM“ PM“ PM“ PPm° Adipose Tissue ppm 0.88 4.86 0.99 3.26 Liver ppm 0.94 3.23 0.92 5.71 Thymus ng TCB/ Thymus 4.4 25.2 2.5 15.8 Uterus ppm 0.29 0.21 0.12 0.54 Placenta ppm NAd NA -‘ 0.61 Fetus ppm NA NA 0.14 :l: 0.03 0.47 :I: 0.13 (n=8) (n=21) a The term equivalents refers to TCB and its metabolites. The concentrations of TCB equivalents in organs and tissues were calculated from the specific activity in feed and the radioactivity measured in the organs and tissues with a liquid scintillation counter (Appendices I and II). b Both non-pregnant and pregnant mice were treated with l‘C-TCB feed for 5 weeks. 0 Values are the means of data from 2 pregnant mice in the 30 ppm group. Table values from all other groups reflect a sample size of one. (1 Not applicable. e Missing data. 45 Table 3. Concentrations of TCB equivalents in adipose tissue and organs of non- pregnant and lactating mice. Organ/ Concentration' Treatments Tissue Non-pregnantb Lactating" 3 ppm 30 ppm 3 ppm 30 ppm Adipose Tissue ppm 1.26 8.71 0.14 1.83 Liver ppm 2.29 5.98 0. 15 0.24 Thymus ng TCB/ Thymus 0 132.1 4.8 205.2 a The term equivalents refers to TCB and its metabolites. The concentrations of TCB equivalents in organs and tissues were calculated from the specific activity in feed and the radioactivity measured in the organs and tissues with a liquid scintillation counter (Appendices I and II). b Non-pregnant mice were treated with l‘C-TCB feed for 8 weeks. lactating mice were treated with l‘C-TCB feed for 5 weeks before parturition, and TCB feed for 3 weeks after parturition. Table values reflect a sample size of one. 46 Table 4. Concentrations of TCB equivalents in adipose tissue and organs of male offspring at 3 weeks of age. Organ/ Concentration‘l Treatments Tissue 3 ppm 30 ppm (n=4) (n=5) Adipose Tissue ppm 0.15 :t 0.03 8.15 :i: 7.84 Liver ppm 0.10 :I: 0.01 0.56 :l: 0.14 Thymus ng TCB/ Thymus 0 0 a The term equivalents refers to TCB and its metabolites. The concentrations of TCB equivalents in organs and tissues were calculated from the specific activity in feed and the radioactivity measured in the organs and tissues with a liquid scintillation counter (Appendices I and II). The calculated concentrations of TCB equivalents were transferred through the placenta and milk from the dams which were treated with 1‘C-TCB feed for 5 weeks before parturition, and then TCB feed for 3 weeks after parturition. 47 Biased After the first two weeks of treatment, there were no significant differences in body weights in the F-0 mice, although mice in the 30 ppm group consumed more feed than those in the 3 ppm group (Table 5). Feed intake during gestation days 1 - 16 was also higher in the 30 ppm group than in the 3 ppm group, although there were still no significant differences in body weights between the 2 groups. On day 21 of lactation, however, the 3 ppm TCB-treated mice consumed more feed than the 30 ppm-treated mice, although they weighed less than the 30 ppm-treated mice (Table 5). On day 19 of gestation and on day 21 of lactation, relative liver weights of F-0 mice were higher in the 30 ppm treatment group than in the 3 ppm and the control groups (Table 6). These differences were not apparent in non-pregnant females. TCB treatments had no significant effect on thymus weights in F-O females (Table 6). Fecundity in F-0 females was 80%, 71%, and 47% in the control, 3 ppm, and 30 ppm groups, respectively (Table 7). The treatments had no effect on litter sizes and sex ratios. Four-day and 21-day survival indices showed a trend of dose-response relationships. Decreased survival was observed in the 30 ppm group, in which most of the mortality occurred before 4 days of age (Table 7). There were no significant differences in body weights of F-l offspring (Table 8). At 5 and 6 weeks of age, F-l females in the 30 ppm group showed an increase in liver weights, but not in relative liver weights (Table 9). A decrease in thymus weights was observed in the 30 ppm-treated F-l females between 5 and 7 weeks of age (Table 9). However, relative thymus weights decreased in the 30 ppm-treated F-l females only at 5 weeks of age. Table 5. Effects of TCB on feed intake and body weights of F-0 female mice. Treatments Control 3 ppm 30 ppm Body Weight (g)c 21.6 :1: 0.8 21.7 :1: 0.7 21.8 :I: 1.0 (13 Weeks of Age) (11: 18) (n=24) (n=46) Feed Intake (g/day)‘l 4.7 :I: 0.9‘”- 4.4 d: 0.8‘I 5.2 :1: 0.6” (13-15 Weeks of Age) (n=15) (n=15) (n=15) Body Weight (g)° 21.7 :I: 0.9 21.6 :t 0.5 22.2 i 1.1 (15 Weeks of Age) (n=18) (n=24) (n=46) Feed Intake (g/day)e 4.8 i 1.2 4.6 :l: 1.4 5.4 :I: 1.0 During Gestation (n=7) (n=7) (n=4) Body Weight (g) 36.9 :i: 2.1 37.3 i 1.2 35.6 :I: 2.9 Day 19 of Gestation (n=3) (n=3) (n=3) Feed Intake (g/day)f 8.8 :I: 2.3‘ 10.3 :I: 1.6‘ 7.7 :I: 2.3" During Lactation (n=9) (n=8) (n=10) Body Weight (g) 28.5 :I: 1.6” 26.2 :1: 0.1‘ 29.3 :1: 1.6b Day 21 of lactation (n=3) (n=3) (n=3) ab "'5 (P <0.05). Different superscripts represent significant differences within the same row Females were treated with TCB 2 weeks beginning at 13 weeks of age. Each female was then paired with one non-treated mature male for 10 days. Values represent mean feed intake :1: SD during the two weeks before mating. Values represent mean feed intake i SD from day 1 to 16 of gestation. Values represent mean feed intake :I: SD from day 1 to 21 of lactation. 49 Table 6. Effects of TCB on organ weights and relative organ weights of F-O females. Weightc Treatments Control 3 ppm 30 ppm (n=3) (n=3) (n=3) N on-pregnant _ (18 Weeks of Age) LW (g) 1.33 :I: 0.36 1.08 :1: 0.04 1.47 i 0.55 LW/BWd X 100 5.73 :l: 1.24 5.03 :I: 0.11 6.62 :l: 2.65 TMW (g) 24.4 :1: 8.4 32.4 :1: 5.3 36.8 i 9.8 TMW/BW‘I x 100 0.11 :1: 0.04 0.15 :I: 0.03 0.16 i 0.04 Day 19 of Gestation LW (g) 1.69 i 0.07 1.81 :l: 0.12 1.82 :l: 0.13 LW/BWe x 100 4.59 :I: 0.09' 4.84 :I: 0.23' 4.95 :I: 0.07" TMW (mg) 19.0 :t 9.2 25.6 :I: 7.0 18.0 :I: 1.4 TMW/BW‘ x 100 0.06 :I: 0.03 0.07 :I: 0.02 0.05 :I: 0.003 Day 21 of Lactation LW (g) 2.06 i: 0.16‘” 1.99 d: 0.11‘ 2.37 :l: 0.20b LW/BW° x 100 7.22 :1: 0.34' 7.60 :t 0.42‘I 8.10 :l: 0.32” TMW (mg) 34.4 :t 2.3 29.0 :1: 5.9 31.7 :I: 9.6 TMW/BW‘ x 100 0.12 :t 0.01 0.11 i 0.02 0.11 :l: 0.03 ab Different superscripts represent significant differences within the same row C (P<0.05). BW, LW, and TMW represent body, liver, and thymus weights, respectively. Values represent mean organ weights or organ weights as percentages of body weights :1: SD. Body weights were 23.1 :1; 1.2, 21.5 :1: 0.5, 22.2 :t 0.5 g in control, 3 ppm, 30 ppm group, respectively. Body weights were based on the data in Table 5. 50 Table 7. Effects of TCB on reproductive performance of F-0 female mice. Treatments Control 3 ppm 30 ppm Fecundity (%)c 80“ 71" 47" Litter Size 8.3 :I: 2.0 ‘ 8.3 :I: 1.3 8.2 :1: 1.2 (n=12) (n=16) (n=20) Sex Ratio (9/6) 1.11 :I: 0.51 1.09 :I: 0.72 1.05 :l: 1.00 (n=12) (n=16) (n=20) 4-Day Survival 47.4 :I: 34.7‘I 41.6 i 39.1‘l 30.9 :I: 30.9" Indexd (n = 12) (n = 16) (n =20) 21-Day Survival 96.3 :1: 11.1‘ 88.0 :1: 31.6‘ 75.8 :I: 37.7" Index° (n =9) (11 = 10) (n = 13) ab Different superscripts represent significant differences within the same row (P<0.05). c Values are expressed as percentages of females mated which became pregnant. (I Four-day survival index = 100 X [number of pups viable at day 4 of age]/ [number of viable pups born]. e Twenty-one-day survival index=100 x [number of pups viable at day 21 of age]/[number of pups retained at day 4 of age]. The litters in which all pups died before day 4 of age were not included. 51 Table 8. Effects of TCB on body weights of El mice. Age' Treatments Control 3 ppm 30 ppm BW (g) Day 4 1.8 i 0.3 1.8 :l: 0.4 1.7 :I: 0.3 (n=9) (n=10) (n=l3) Week 1 3.3 i 0.7 3.6 :t 0.4 3.1 :l: 0.6 (n=9) (n=9) (n=12) Week 2 6.9 :t 1.4 7.3 :l: 0.6 6.5 :I: 1.5 (n=9) (n=9) (n=12) Week 3 7.9 :I: 1.2 8.4 :I: 0.7 7.6 :i: 2.4 (n=45) (n=51) (n=37) Week 4 14.5 :1; 1.3 14.7 :i: 2.8 14.9 :I: 3.5 (n=42) (n=51) (n=32) Week 5 19.3 :I: 2.4 19.0 i 3.2 19.0 :I: 3.6 (n=32) (n=47) (n=29) week 6 20.0 :I: 2.5 20.1 :I: 3.2 20.6 :t 3.1 (n=41) (n=49) (n=27) Week 7 21.7 i 3.1 21.2 :t 3.5 21.3 :I: 3.3 (n=33) (n=45) (n=21) a Body weights on day 4, week 1, and week 2 represent the means :1: SD of litters. Values for week 3 through week 7 are means :1: SD of individual weights. 52 Table 9. Effects of TCB on body weights, organ weights, and relative organ weights of F-l females between 5 and 7 weeks of age. Age Treatments Control 3 ppm 30 ppm BW (g) Week 5 17.8 i 0.5 (n=7) 18.3 :1: 1.5 (n=9) 18.3 :I: 1.8 (n=8) Week 6 18.1 :I: 0.7 (n=6) 18.9 :I: 0.2 (n=3) 18.4 :t 0.1 (n=2) Week 7 19.6 :t 0.7 (n=7) 19.8 :I: 1.0 (n=10) 18.9 j: 1.2 (n=4) Relative Liver Weight (LW/BW x 100)° (LW (g))‘ Week 5 6.26 :1: 0.53 6.09 :I: 0.47 7.05 :I: 0.42 (1.11 :1; 0.08‘) (1.11 i 0.14‘) (1.30 :I: 0.17") Week 6 6.00 :I: 0.23 5.84 :I: 0.33 7.04 :l: 0.37 (1.09 i 0.07‘) (1.10 :I: 0.07‘) (1.30 :I: 0.08") Week 7 5.75 :i: 0.60 6.06 :I: 0.38 6.35 i 0.39 (1.13 :I: 0.14) (1.20 :I: 0.12) (1.26 :I: 0.15) Relative Thymus Weight (TMW/BW x 100)° (TMW 011g»d Week 5 0.44 i 0.88‘" 0.46 :l: 0.05‘ 0.36 :t 0.03" (77.7 :1: 12.0‘) (81.0 :I: 7.0‘) (60.8 :I: 5.9") Week 6 0.43 :I: 0.08 0.42 :l: 0.09 0.30 :t 0.02 (77.5 :1: 13.7') (80.1 :I: 16.9‘") (55.1 :1; 2.3") Week 7 0.39 :t 0.08 0.37 :I: 0.08 0.30 :I: 0.06 (75.2 j: 11.7‘) (67.8 i 9.8‘") (55.8 :1: 9.4") ab Different superscripts represent significant differences within the same row (P < 0.05). . c BW, LW, and TMW represent body, liver, and thymus weights, respectively. Values represent mean of organ weights as percentages of body weights :I: SD. (1 The numbers in the parentheses under relative organ weights represent organ weights :1: SD. 53 Decreases in fertilizing ability of F-l eggs in vitra were observed in the 3 ppm and 30 ppm groups when compared to the control group (Table 10). Fertilization rates were 68.4%, 47.6%, and 45.6% for the control, 3 ppm, and 30 ppm groups, respectively. The percentage of degenerated eggs in the 3 ppm and 30 ppm groups was higher than that in the control group. The number of eggs superovulated by exogenous PMSG and HCG were the same among all groups (Table 10). F-1 females at 7 weeks of age showed similar fecundity among all groups when bred with non-treated males (Table 11). Nonetheless, all offspring from 3 ppm and 30 ppm-treated F-l females died within 4 days of birth (Table 11). Testes in treated F-l males at 3 weeks of age were heavier in mice treated with 30 ppm TCB than for mice in the control group (Table 12). However, relative testis weights were higher in both 3 ppm and 30 ppm groups than in the control group. Liver weights, relative liver weights, thymus weights, and relative thymus weights were similar among all groups (Table 12). At 9 and 19 weeks of age, liver weights and relative liver weights were higher in the 30 ppm group than in the other 2 groups. However, thymus and testis weights were the same among all 3 groups (Tables 13 and 14). Treated F-l males, at 7 and 17 weeks of age, were bred with non-treated females. Fecundity, litter sizes, sex ratios, and survival of the offspring were the same among all treatment groups (Tables 15 and 16). The 4—day survival index decreased by 20% in the 3 ppm and 30 ppm groups when treated males at 60 weeks of age were paired with non-treated females (Table 17). 54 Table 10. Fertilizing ability of eggs from F-l females exposed to TCB in utera through postnatal development. Treatments Control 3 ppm 30 ppm No. Females Tested 15 16 12 No. Exp. 6 6 6 Eggs Ovulated Per Animal (No.) 40 j; 10 29 j; 21 34 j; 22 Fertilization In Vitra (%) 68.4 :1: 15.1‘ 47.6 :I: 25.9" 45.6 :I: 18.7" Degeneration Rate (%) 6.0 :I: 4.8‘l 15.6 :I: 20.2" 13.4 :I: 7.9" ab Different superscripts represent significant differences within the same row (P < 0.05) ' 55 Table 11. Effects of TCB on reproductive performance of F -1 females at 7 weeks of age. Treatments Control 3 ppm 30 ppm No. Females Mated 10 10 10 Fecundity (%) 50 . 40 70 Litter Size 7.0 :t 2.3 5.3 :I: 1.7 7.6 i 1.9 (n=5) (n=4) (n=7) 4-Day Survival 46.4 :1; 43.0' 0" 0" Indexc (n =5) (n =4) (n =7) ab Different superscripts represent significant differences within the same row (P<0.05). c Four-day survival index = 100 x [number of pups viable at day 4 of age]/[number of viable pups born]. 56 Table 12. Effects of TCB on body weights, organ weights, and relative organ weights of F-l males at 3 weeks of age. Weightc Treatments Control 3 ppm 30 ppm (n=7) (n=7) (n=8) BW (g) 8.4 :1: 0.5 8.7 :I: 0.5 8.5 :l: 0.7 LW (g) 0.35 :I: 0.08 0.31 :1: 0.05 0.34 :t 0.04 LW/BW X 100 4.2 :l: 0.8 3.6 :I: 0.5 4.0 :I: 0.2 TMW (mg) 53.2 :t 5.0 53.3 :I: 9.8 49.4 :t 13.4 TMW/BW X 100 0.64 i 0.09 0.62 :l: 0.13 0.58 :t 0.14 'I'TW (mg) 32.9 d: 3.3‘ 40.6 :1: 5.7‘" 43.2 :1: 7.0" TTW/BW X 100 0.39 :I: 0.03‘I 0.47 i 0.05" 0.51 :t 0.10" ab Different superscripts represent significant differences within the same row (P < 0.05). BW, LW, TMW, TTW represent body, liver, thymus, and testis weights, respectively. Values represent mean organ weights or organ weights as percentages of body weights :I: SD. 57 Table 13. Effects of TCB on body weights, organ weights, and relative organ weights of F-l males at 9 weeks of age. Weightc Treatment Control 3 ppm 30 ppm (n=10) (n=10) (n=10) BW (g) 26.8 :I: 2.2 23.5 i 4.3 24.7 :1: 2.5 LW (g) 1.25 :I: 0.16‘ 1.26 :l: 0.28‘ 1.48 :I: 0.20" LW/BW X 100 5.05 :I: 0.50' 5.33 :I: 0.45'I 5.99 :i: 0.46" TMW (mg) 37.5 :1: 13.1 32.5 :t 7.2 32.6 :I: 5.0 TMW/BW X 100 0.12 i- 0.02 0.12 :I: 0.01 0.12 i 0.01 TTW (mg) 194.3 :I: 9.9 192.7 :I: 10.8 197.6 i 24.0 TTW/BW X 100 0.79 i 0.06 0.85 :i: 0.22 0.80 :t: 0.08 ab Different superscripts represent significant differences within the same row (P<0.05). BW, LW, TMW, TTW represent body, liver, thymus, and testis weights, respectively. Values represent mean organ weights or organ weights as percentages of body weights :1: SD. 58 Table 14. Effects TCB on body weights, organ weights, and relative organ weights F-l males at 19 weeks of age. Weight° Treatment Control 3 ppm 30 ppm (n=6) (n=6) (n=6) BW (g) 30.9 :1: 2.1 _ 29.4 :I: 3.5 31.3 :1: 2.0 LW (g) 1.32 i 0.14' 1.32 :I: 0.14‘ 1.67 :I: 0.16" LW/BW X 100 4.27 :1: 0.31' 4.50 :I: 0.21‘l 5.35 :1: 0.31" TMW (mg) 51.0 :1: 11.8 38.0 i 11.0 41.6 :1: 12.0 TMW/BW X 100 0.16 :I: 0.03. 0.13 :I: 0.03 0.14 21:0.04 TTW (mg) 219.4 :1; 17.5 223.5 :1: 17.8 232.4 :I: 14.6 TTW/BW X 100 0.71 :I: 0.05 0.77 :I: 0.08 0.74 :I: 0.04 ab Different superscripts represent significant differences within the same row (P<0.05). c BW, LW, TMW, TTW represent body, liver, thymus, and testis weights, respectively. Values represent mean organ weights or organ weights as percentages of body weights :1: SD. Table 15. Effects of TCB on reproductive performance of F-1 males at 7 weeks of age. Treatments Control 3 ppm 30 PM“ No. Males Mated‘ 10 10 10 Fecundity (%) 90 80 90 Litter Size 8.9 :1: 2.4 9.8 :I: 2.4 9.3 :I: 2.3 (n=9) (n=8) (n=9) Sex Ratio ((3/9) 2.0 :1; 2.3 1.4 :1; 0.8 1.3 :I: 1.0 (n=9) (n=8) (n=9) 4-Day Survival 100 100 100 Index" (n=9) (n=8) (n=9) 21—Day Survival 100 100 100 Index° (n=9) (n=8) (n=9) One non-treated female was mated with one treated male for 10 days, from 7 :30 PM to 7:30 AM. TCB treatment was withheld during breeding. of viable pups born]. Four-day survival index = 100 x [number of pups viable at day 4 of age]/ [number Twenty-one-day survival index= 100 X [number of pups viable at day 21 of age]/[number of pups retained at day 4 of age]. The litters in which all pups died before day 4 of age were not included. Table 16. Effects of TCB on reproductive performance of F-l males at 17 weeks of age. Treatments Control 3 ppm 30 ppm No. Males Matedc 6 6 6 Fecundity (%) 67 100 100 Litter Size 8.8 i 1.3 9.0 :1: 0.9 8.8 i 1.2 (n=4) (n=6) (n=6) Sex Ratio (cf/9) 1.4 i 1.0 2.4 :1: 2.8 2.1 :I: 2.9 (n=4) (n=6) (n=6) 4-Day Survival 94.7 :I: 6.1 100 100 Index‘1 (n=4) (n=6) (n=6) 21-Day Survival 100 96.3 :t 9.1 100 Index‘ (n=4) (n=6) (n=6) ab Different superscripts represent significant differences within the same row (P <0.05). c One non-treated female was mated with 1 treated male for 10 days, from 7:30 PM to 7:30 AM. TCB treatment was withheld during breeding. d Four-day survival index = 100 x [number of pups viable at day 4 of age]/[number of viable pups born]. e Twenty—one—day survival index= 100 x [number of pups viable at day 21 of age]/ [number of pups retained at day 4 of age]. pups died before day 4 of age were not included. The litters in which all 61 Table 17. Effects of TCB on reproductive performance of F—l males at 60 weeks of age. Treatments Control 3 ppm 30 ppm No. Males Matedc 5 4 5 Fecundity (%) 25 50 33 Litter Size 9.0 :1: 1.0 8.0 i 1.3 9.2 :t: 2.2 (n=3) (n=6) (n=5) Sex Ratio (6/9) 0.74 :1; 0.13 1.54 :I: 0.63 1.23 :t 0.70 (n=3) (n=6) (n=5) 4-Day Survival Index“ 100‘ 75.8 :1; 28.3" 78.5 :1; 20.9" (n=3) (n=6) (n=5) 21-Day Survival 100 100 100 Indexe (n=3) (n=6) (n=5) ab Different superscripts represent significant differences within the same row (P<0.05). 0 Three non-treated females were mated with 1 treated male for 90 days. TCB treatment was withheld during breeding. d Four-day survival index = 100 X [number of pups viable at day 4 of age]/[number of viable pups born]. e Twenty-one—day survival index=100 X [number of pups viable at day 21 of age]/[number of pups retained at day 4 of age]. The litters in which all pups died before day 4 of age were not counted. 62 Body weights of offspring sired by treated 7- and 60-week-old F-l males were not affected (Tables 18 and 20). However, body weights of offspring sired by the 30 ppm- treated 17-week-old F-l males were higher than the control (Table 19). At 9 and 19 weeks of age, epididymal sperm was collected from treated F-l males. Sperm concentration, velocity, linearity, ALH displacement, and beat/cross frequency were similar among all groups (Tables 21 and 22). Percentages of eggs fertilized by the sperm from 30 ppm-treated males was lower than the percentages for control group at 19 weeks of age, but not at 9 weeks of age (Tables 21 and 22). At 2.5 hours after sperm collection, decreased sperm motility was observed in 9-week-old 30 ppm-treated males. This was not observed at 0.25, 1.5, 3.5, and 4.5 hours after collection (Figure 1). 63 Table 18. Effects TCB on body weights of offspring sired by F-l males at 7 weeks of age. Age" Treatment Control 3 ppm 30 ppm BW (3) Day 4 2.4 i 0.3 (n=9) 2.4 :1: 0.3 (n=8) 2.3 :I: 0.2 (n=9) Week 1 4.3 :1: 0.4 (n=9) 4.3 :1: 0.4 (n=8) 4.2 :1; 0.3 (n=9) Week 2 8.2 i 0.9 (n=9) 7.9 :I: 0.9 (n=8) 7.9 :1: 0.5 (n=9) Week 3 (d‘) 10.5 :I: 1.0 (n=46) 10.3 :I: 1.0 (n=42) 10.2 :1; 0.9 (n=47) (9) 10.5 :I: 1.1 (n=23) 10.1 :t 1.0 (n=29) 10.0 :I: 1.1 (n=36) a Body weights on day 4, week 1, and week 2 represent the means :1: SD of litters. Values for week 3 are means :1: SD of individual weights. 64 Table 19. Effects of TCB on body weights of offspring sired by F—l males at 17 weeks of age. Age" Treatments Control 3 ppm 30 ppm BW (3) Day 4 2.0 :I: 0.3 (n=4) 2.1 :t 0.1 (n=6) 2.5 :1: 0.4 (n=6) Week 1 3.7 :1: 0.4 (n=4) 4.0 :t 0.2 (n=6) 4.4 :I: 0.5 (n=6) Week 2 7.6 :I: 0.7 (n=4) 7.8 :t: 0.9 (n=6) 8.0 :1; 0.7 (n=6) Week 3 (d) 9.8 i 1.6" (n=17) 10.5 :I: 1.1‘" (n=30) 10.7 :1; 1.0" (n=27) (9) 9.4 :I: 1.2" (n=16) 10.2 :1: 0.9" (n=22) 10.5 :I: 1.0b (n=25) Body weights on day 4, week 1, and week 2 represent the means :1; SD of litters. Values for week 3 are means :1: SD of individual weights. 65 Table 20. Effects of TCB on body weights of offspring sired by F-l males at 60 weeks of age. Age" Treatments" Control 3 ppm 30 1’1““ BW (3) Week 1 4.0 :I: 0.3 (n=3) 3.5 :I: 0.6 (n=6) 3.5 :I: 1.0 (n=4) Week 2 7.8 i 0.7 (n=3) 7.4 :1: 1.3 (n=6) 8.0 :I: 1.5 (n=4) Week 3 (d) 10.2 :1: 1.2 (n=12) 10.3 :I: 1.3 (n=20) 10.5 :I: 1.6 (n=17) (9) 10.2 :I: 1.2 (n=16) 10.0 :1: 1.9 (n=12) 9.7 :1: 1.1 (n=13) a Three non-treated females were mated with 1 treated male for 90 days. TCB treatment was withheld during breeding. b Body weights on week 1 and week 2 represent the means :1: SD of litters. Values for week 3 are means j: SD of individual weights. 66 Table 21. Effects of TCB on fertilization rates in vitra and sperm motion analysis for F-l males at 9 weeks of age. Parameters" Treatments Control 3 ppm 30 ppm (n=10) (n=10) (n=10) Concentration (x 10"/ml) 12.4 :1: 3.1 12.7 :1: 5.8 12.1 :1: 3.8 Motility (%)" 70.9 :1: 10.2 69.2 :1: 11.7 72.9 :1: 7.0 Velocity (pm/sec)c 246.9 :1: 59.9 210.8 :1: 92.9 260.2 i 67.3 Linearity“ 5.4 i 0.8 4.6 :1: 1.8 5.6 :1: 0.9 Amplitude of Lateral Head Displacement (pm)° 7.3 i 1.9 5.9 :1; 2.8 7.6 :1: 3.4 Beat/Cross Freq. (Hz)f 12.7 :1: 1.3 11.2 :1: 4.3 12.6 :1: 1.6 Fertilization In Vitra (%) 66.1 :1: 27.4 68.9 :1: 28.1 63.3 :1: 21.6 a In vitra fertilization and sperm motion analysis were performed 1.5 hours after sperm collection. b Percentage of sperm that travel more than 20 pm/sec. c Average distance traveled by motile sperm. (1 A calculation of the length of the straight line distance divided by the track distance covered by sperm, multipled by 10. This is expressed on a scale of 0 to 10; 10 indicates a perfectly straight line and 0 indicates a circular track. e A measure of the displacement of the sperm head from a computer-calculated curval mean of its track. f The number of beats (or crosses) per second. Every time the sperm cell crosses the computer-calculated curval mean, the computer counts that crossing as one beat. 67 Table 22. Effects of TCB on fertilization rates in vitra and sperm motion analysis for F-l males at 19 weeks of age. Parameters" Treatments Control 3 ppm 30 ppm (n=6) (n=6) (n=6) Concentration (x lO‘lml) 13.6 :1: 2.8 11.6 :1: 3.3 11.0 :1: 2.0 Motility (%)“ 63.5 :1: 9.5 60.6 :1: 13.2 66.5 i 10.3 Velocity (pm/sec)e 208.9 :I: 35.4 190.8 :1: 24.0 200.9 :1: 11.8 Linearity‘ 4.9 :1: 0.2 4.8 :t 1.2 4.7 :1: 0.5 Amplitude of Lateral Head Displacement (pm)‘ 5.7 :1: 1.5 5.5 i 1.1 5.5 :l: 1.9 Beat/Cross Freq. (Hz)" 11.5 :1: 1.9 12.7 :1: 2.3 12.6 :1: 1.0 Fertilization In Vitra (%) 57.9 :1: 12.6" 57.2 :1: 19.7" 46.7 :1: 11.5" ab Different superscripts represent significant differences within the same row. c In vitra fertilization and sperm motion analysis were performed 1.5 hours after sperm collection. d Percentage of sperm that travel more than 20 pm/sec. e Average distance traveled by motile sperm. f A calculation of the length of the straight line distance divided by the track distance covered by sperm, multipled by 10. This is expressed on a scale of 0 to 10; 10 indicates a perfectly straight line and 0 indicates a circular track. g A measure of the displacement of the sperm head from a computer-calculated curval mean of its track. h The number of beats (or crosses) per second. Every time the sperm cell crosses the computer-calculated curval mean, the computer counts that crossing as one beat. Motility (%) T j l j I 15 90 1 50 21 0 270 Time (min) Figure l. Motility of sperm taken from 9-week-old F-l males. Females (F-O) were treated with 0, 3, and 30‘ ppm TCB 2 weeks prior to breeding. Treatments continued through mating, gestation, and lactation. Ofi‘spring (F-l) were given the ’ same diet as their dams received. Each value reflects the mean of data from 7 mice. Different superscripts represent significant differences 1 (P<0.05). -o- Control -a— 3 ppm +30 ppm DISCUSSION Ritual Results of this study demonstrated. that accumulation of TCB in liver and adipose tissue is not arithmetically proportional to exposure concentrations. After 5 weeks of exposure, concentrations of TCB equivalents in the liver, adipose tissue, uterus, and fetus of mice in the 30 ppm group were only 3-6 times higher than concentrations in mice in the 3 ppm group. Greater induction of hepatic microsomal monooxygenases has been observed in pregnant rabbits treated with 10 mg Aroclor 1254/kg bw than in those treated with 1.0 mg/kg (Villeneuve and Grant, 1971). Thus, it is possible that in the present study, hepatic microsomal monooxygenases in the 30 ppm-treated mice were also induced to a greater extent than in 3 ppm-treated mice, resulting in an increase in metabolism and elimination of TCB in the high dose group. After 5 weeks, the concentrations of TCB equivalents in the liver of pregnant mice treated with 30 ppm TCB in the feed was 1.8 times higher than those in non- pregnant mice. This may be due, in part, to an increase in the amount of fat in the liver during pregnancy, causing a redistribution of TCB and its metabolites from adipose tissues to liver. This hypothesis is also evident in the low concentrations of TCB equivalents in the adipose tissue of pregnant mice, which was 2/3 of that in non-pregnant mice. Increases in deposits of body fat cause dilution of TCB concentrations and may 69 70 also contribute to lower concentrations of TCB equivalents in pregnant mice than in the non-pregnant mice. Nonetheless, when two groups of mice were exposed to a single injection of 100 mg l“C-6-CB/kg bw, and one group was then mated while one group remained virgin, radioactivity in the kidney, adipose tissue, and liver of pregnant mice was higher than that in the virgins sacrificed concurrently (Vodicnik and Lech, 1980). The differences in adipose tissue radioactivity in the two studies could be attributed to different administration doses and different congeners. Induction of hepatic microsomal ethoxycoumarin-O—deethylase (ECOD) by 4-CB was less in pregnant mice than in virgins over a 4-day period (V odicnik, 1986). A slower rate of elimination of 4-CB equivalents was observed in pregnant mice than in virgin mice. Results in the present study also indicated that dosage affected distribution of TCB in the liver and adipose tissue. The concentrations of TCB equivalents in the liver and adipose tissue of the 3 ppm-treated mice were the same in pregnant and non-pregnant mice. Any differences in body burden between pregnant and non-pregnant mice can not be explained, since PCBs in the fetus only aeoount for approximately 0.003 % of maternal body burden of pregnant rats (Takagi et al., 1976). In this study, concentrations of TCB equivalents, calculated from radioactivity, were similar in the uteri, placentas, and fetuses of treated mice. Darnerud et al. (1986) reported that TCB metabolites, but no parent TCB, were detected in the fetus when pregnant mice were gavaged with 25 mg TCB/kg bw on day 13 of gestation and sacrificed on day 17 of gestation. Therefore, it is likely that the radioactivity detected in the fetal tissue in the current study is from TCB metabolites. 71 Results from this study also point to redistribution of TCB equivalents to the thymus from other tissues during lactation in the 30 ppm group. Although “C—TCB feed was changed to TCB feed at parturition, radioactivity in the thymus increased during lactation. However, only background radioactivity values were detected in the thymus of the 3 ppm-treated mice. Although TCB redistribution into the thymus was observed in the high dose group during lactation, the mechanisms by which this occurs is unknown. The redistribution of radioactivity in pregnant and lactating mice is likely to be associated with the increase of very low density lipoprotein (VLDL), the primary carrier of 6—CB in viva which occurs during gestation (Spindler-Vomachka and Vodicnik, 1984). VLDL is a major substrate for mammary gland lipoprotein lipase which is elevated during late gestation and lactation (Spindler-Vomachka and Vodicnik, 1984). More than 70% of circulating 6-CB was associated with VLDL during late gestation in female mice treated with 6-CB two weeks prior to mating (Gallenberg and Vodicnik, 1987). Continuous exposure to varying doses of TCB results in differences in TCB redistribution between organs and tissues. In the 3 ppm-treated group, approximately 85% of the concentrations of TCB equivalents disappeared from both liver and adipose tissue during lactation. However, in the 30 ppm group, 96% disappeared from the liver and only 44% disappeared from the adipose tissue. It is possible that circulating lipoproteins in the blood were "saturated" with TCB equivalents in the 30 ppm group, which decreased the redistribution of TCB from adipose tissue to liver. This phenomenon,"saturable kinetics", was observed in the fetal compartment when dams were pretreated with TCB 72 (12.5-50 mg/kg BW) and followed 4 hours later by l‘C-TCB (10 mg/kg) (Darnerud et al., 1986). Radioactivity in fetal brain, liver, thymus, skin, and muscle in the pretreatment groups was less than half of that observed in the group without TCB pretreatment (Darnerud et a1 . , 1986). Different doses affect the distribution of TCB in the liver and adipose tissue of offspring. In the offspring of the 30 ppm treatment group, concentrations of TCB equivalents in the adipose tissue were 14.6 times higher than those in the liver. However, concentrations of TCB equivalents in the adipose tissue were only 1.5 times higher than those in the liver of 3 ppm-exposed offspring. In addition, although the pups were nursed by the same dam in the 30 ppm group, large variations in concentrations of TCB equivalents were observed. These results are due to large variations of offspring body weights and fat deposits in the 30 ppm-exposed offspring. Variations of offspring body weights were smaller in the 3 ppm group than those in the 30 ppm group. This preliminary study indicates that accumulation of TCB equivalents in the adipose tissue and liver of mice is not arithmetically proportional to exposure. This is probably due to the greater induction of hepatic microsomal monooxygenases in 30 ppm- treated mice than in the 3 ppm-treated mice, resulting in an increase in metabolism and excretion of TCB. A similar accumulation of TCB equivalents between 3 ppm-treated non-pregnant and pregnant mice suggests similar activities of hepatic microsomal monooxygenases under differing physiological conditions. Different doses result in variations of TCB redistribution during late gestation and lactation. 73 13m Feed Intake and Body Weight Based on feed intake, F-0 females in the 30 ppm treatment group consumed approximately 7.5, 5.9, and 8.3 mg TCB/kg bw/day before breeding, from day 1 to day 16 of gestation, and during lactation, respectively. Body weights were not significantly different among F-0 treatment groups before breeding and during gestation. During lactation, body weights in the 30 ppm group were higher than those in the 3 ppm group. However, body weights in the control group were not different from TCB treated groups. As Yoshimura et al. (1979) reported, body weight gains in weanling rats over a 4-day period were not affected by a single i.p. injection of 50 mg TCB/kg bw. In immature male rats, ED50 for body weight loss caused by TCB was approximately 213 mg/kg bw in a single i.p. injection (Leece et al., 1985). Reduced body weight gains were reported by Sanders et al. (1974) in rats after 2 weeks of 62.5 or 1000 ppm of Aroclor 1254 exposure through the feed. These doses are much higher than the high dose tested in this study which was 7.5 mg/kg bw/day prior to breeding. High feed intake observed in the 3 ppm-treated mice during lactation was in all likelihood a compensation response to the low feed intake before breeding and during gestation. Changes in body weights of offspring treated with PCBs perinatally were dependent on exposure period, dose, and on the congener or commercial mixture. F-l body weights at weaning in the 3 ppm group were numerically greater than those in the 30 ppm group. The increase in F-l weaning weights in the 3 ppm group could be the result of increase in milk production of the dams which had high feed intake. As Gellert 74 and Wilson (1979) reported, no effects on body weights of female rat offspring were observed when dams were gavaged with 30 mg Aroclors 1242 and 1260/kg bw daily on days 14-20 of gestation. Offspring body weights, however, increased when dams were treated with Aroclor 1221 (Gellert and Wilson, 1979). On the other hand, decreased body weights of young mice were observed when parents were dosed with 10 mg Aroclor 1254/kg bw throughout gestation and lactation (Linzey, 1988), although these litters did not differ in body weights at birth. Similar regimens using different PCBs and doses produced diverse results. The relative growth rates of young male mice from dams administered orally 0.005 mg 2,4’,5-trichlorobiphenyl or 6-CB/day, from day 5 of gestation to day 22 of lactation, were higher than those of male offspring in the control group (Orberg, 1978b). The differences in offspring body weights at weaning are mostly diminished after maturity. For example, decreased body weight gains during lactation followed by increased body weight gains after weaning were observed in rat offspring when dams were treated on days 1, 3, 5, 7, and 9 postpartum with a dose of 32 or 64 mg Aroclor 1254/kg bw (Sager et al., 1991). Weight gains were similar among treatment groups after those offspring reached 5 months of age. In the present study, the body weights of offspring of treated F-l males are dependent on the exposure period. No differences were observed in body weights of offspring born to non-treated females bred with 7-week-old 30 ppm-treated F-l males. However, body weights of offspring from non-treated females bred with 17-week-old 30 ppm-treated F-l males increased compared to control. These increases occurred at 3 weeks of age and were independent of the body weights of treated F-l males and dams 75 at weaning. Since only sperm from the treated males could have contributed to the increases in body weights,TCB exposure must have altered some characteristic of pre- ejaculated sperm. Hepatotoxicity Liver enlargement has been observed by others in TCB-treated rats (Yoshimura et al. , 1979; Clarke et al., 1984). Increased numbers of peroxisomes and lipid droplets, proliferated smooth endoplasmic reticulum, abnormal mitochondria (Harris and Bradshaw, 1984; MacLellan et al., 1994), and distended cisternae of the rough endoplasmic reticulum (Harris and Bradshaw, 1984) are the most overt signs of toxicity in the livers of rats treated with TCB. Total liver lipid content, including cholesterol, phospholipid and neutral lipid, increased in mature rats exposed to 50 or 500 ppm Aroclor 1254 for 3 weeks (Garthoff et al., 1977). Total protein was also increased by the 500 ppm treatment, while the relative total protein (mg/g wet weight) decreased. The increases in lipid content were associated with increases in the activity of 3-hydroxy-3- methylglutaryl coenzyme A reductase, the rate-limiting enzyme of cholesterol synthesis, as well as increases in three NADPH-generating enzymes: malic enzyme, glucose-6- phosphate dehydrogenase, and 6-phosphogluconate dehydrogenase (Kato and Yoshida, 1980; Hitomi et al., 1993). NADPH is essential for biosynthesis of cholesterol, lipid, and several amino acids (Voet and Voet, 1990). This study demonstrated that the effects of TCB on liver weights depends on the physiological condition and age of the animal. Increases in liver weights were apparent in F-0 pregnant and lactating mice, but not in non-pregnant mice. large variations in 76 liver weights of non-pregnant mice may contribute to the lack of statistical significance of the data. Notably, F—l females treated with 30 ppm TCB showed increases in liver weights at 5 and 6 weeks of age, but not in relative liver weights. At 7 weeks of age, while there was an increase in liver weights and relative liver weights in the 30 ppm group, the differences were not statistically significant. Differences in exposure period and inducibility of hepatic microsomal enzymes may have contributed to the differences in offspring liver weights before and after puberty. Both liver weights and relative liver weights increased in the 30 ppm group in F-l males at both 9 and 19 weeks of age, but not at 3 weeks of age. A significant induction of uridine diphosphoglucuronic acid glucuronyltransferase (UDPGT), an important microsomal glucuronide conjugation enzyme, was observed in the TCB treated rats (3 mg/kg bw/day prenatally) at 3 weeks of age (Harris and Bradshaw, 1984). However, this induction diminished after 3 weeks of age. In addition, a higher-fold induction of hepatic microsomal monooxygenases above the control level was observed in the weanlings than in the adults (Chen and DuBois, 1973). This high induction of hepatic microsomal enzymes enhances biotransformation and excretion of TCB in males at 3 weeks of age. Thymic Atrophy Thymic atrophy is a consistently observed effect of TCDD and PCBs in all animals thus far examined (Faith and Moore, 1977 ; McConnell, 1989; Smialowicz et al. , 1989). Microscopically, the reduction in size is reflected almost entirely as a loss of cortical lymphocytes (McConnell et al., 1978). Although Clarke et al. (1984) reported 77 that 5 mg TCB/kg bw/day for 3 weeks caused thymic atrophy in young female rats, the present study showed no such changes in F-0 female mice after 5 weeks of treatment. The difference between the two observations may be explained by species variation or the differences in maturity of the test animals. Rats in the study by Clarke et al. weighed only half of their mature body weight, while the F-0 females in the present study had reached their mature body weight. Decreases in thymus/body weight ratios in the offspring of dams exposed to TCDD during gestation and lactation were more apparent than those only exposed to TCDD postnatally (Faith and Moore, 1977). At 5, 6 and 7 weeks of age, F-l females treated with 30 ppm TCB showed decreases in thymus weights, however, TCB did not induce thymic atrophy in F-l males at 3, 9, and 19 weeks of age in the present study. Gender appears to be the primary factor contributing to the differences of thymic effects between the F-1 males and females. Female Reproductive Toxicity In our study, fecundity in 30 ppm—exposed females (F-0) was reduced by 40%, while the fecundity in treated F-l females was the same as in the control group. The decrease in the F-0 females could be the result of many reproductive dysfunctions from mating to fertilization, to embryo and fetal development. Existing information points to a decrease in mating of PCB-treated females. In rats, the percent of females mated, judged by vaginal plugs, decreased by 40% when those females were dosed with 7.5 mg Aroclor 1242/kg bw/day for 36 weeks (Jonsson et al., 1976). Similar results have been reported by Brezner et al. (1984) who stated that sexual receptivity was decreased by 20% in rats exposed orally to 30 mg Aroclor 1254/kg bw for 1 month. This sexual 78 receptivity was determined by the presence of sperm in the vaginal smear on the day after mating. Small sample sizes of F-l females may, in part, have prevented the detection of differences in fecundity among treatment groups. Mortality of offspring mostly occurred within 4 days postpartum. Litter sizes of treated F-0 females and F—l offspring were similar among all 3 groups at parturition. These results reflect the observations by Rands et al. (1982) that the mortality during parturition was the same in TCB treated rats as in controls. Four-day and 21-day survival indices only decreased in the 30 ppm group in F-0 females, while all pups born to TCB treated F-l females of both dose groups died within 4 days of age. In rats, a high incidence of neonatal mortality was also observed after prenatal exposure to TCB at 3 mg/kg bw/day from day 6 through day 18 of gestation (Rands et al., 1982; Harris and Bradshaw, 1984). The reasons for the mortality of the progeny are likely associated with the behavioral changes of dams (Pantaleoni et al., 1988), or direct toxicity caused by TCB and its metabolites (Darnerud et al., 1986), or some combination of these factors. In addition, there is a low percentage of fat in the young which is one of the major tissues where PCBs accumulate. This heightens the amount of absorbed PCBs distributed to target organs and tissues in the body. Postnatal mortality is also related to toxicity of TCB and its metabolites in embryos and fetuses. Accumulations of 2-hydroxy-TCB and methylsulphonyl- tetrachlorobiphenyl, two TCB metabolites, in fetal tissue and uterine fluid were observed after a single intravenous injection of 3.5 mg TCB/kg bw in pregnant mice (Darnerud et al., 1986). Hemorrhage in intestinal mucosa of fetuses was observed when pregnant 79 rats were treated with 3 mg TCB/kg bw on days 6—18 of gestation (Rands et al., 1982). Embryonic death in mice increased after a single oral dose of 25 mg TCB/kg bw on day 11 of gestation (d’Argy et al. , 1987). Since the parent TCB was not found in fetal tissue (Darnerud et al., 1986), the metabolites of TCB play a very important role in embryotoxicity. The in vitra fertilization assay showed that egg fertilizing ability decreased in F-l females exposed to 3 ppm and 30 ppm TCB. The poor egg quality was also demonstrated by the high percentage of degenerated eggs after collection and culture. Similar results were observed in a study in which sperm and eggs were incubated in medium containing PCBs. After 1.5 hours of incubation, sperm were added to the petri dish where superovulated eggs were treated with the following PCBs: TCB, Aroclor 1221, and Aroclor 1268. Fertilization rates decreased at a concentration of 1 pg/ml for these PCBs tested (Kholkute et al., 1994). In the present study, it is suggested that impaired egg quality is associated with decreased estrogen levels in the follicles. Preovulatory follicular growth is dependent on the interaction of estradiol, FSH, and LH (Richards, 1980). The ability of healthy eggs with germinal vesicles to resume meiosis in vitra is highly correlated with the concentration of estradiol in antral fluid (McNatty et al., 1979). Theca interna, an androgen biosynthesizing site in the ovary in all species, is differentiated from stroma cells. The synthesized androgens are then transferred to the basement membrane of the follicle and on to the granulosas where they are aromatized to estrogens (Peters, 1979; Erickson et al. , 1985; Ryan, 1988). Characteristic changes in the ovarian stroma cells, 80 including a spindling of ovarian cells accompanied by looseness of cellular arrangement and reduction of follicle numbers, were observed in 150 ppm Aroclor 1242-treated rats (Jonsson et a1 . , 1976). The stroma changes may affect differentiation of theca interna and decrease androgen and estrogen biosynthesis. In addition, metabolism and elimination of estrogen can be increased by hepatic microsomal monooxygenases induced by PCBs or other chemicals (e.g., phenobarbital) (Risebrough and Brodine, 1970; Chen et al. , 1993). Environmental estrogens, including PCBs, have received much attention lately (Hileman, 1994; Stone, 1994). Several commercial PCB mixtures and congeners related to estrogenic activity have been investigated (Bitman and Cecil, 1970; Ecobichon and MacKenzie, 1974; Gellert, 1978; Korach et al., 1987). In general, the lesser chlorinated PCBs have higher estrogenic activity than the more highly chlorinated PCBs, based on the glycogen response of the immature rat uterus. It has been suggested that uterotropic activity of PCBs is dependent upon the formation of metabolites (Ecobichon and MacKenzie, 1974). The lesser chlorinated PCBs can be more easily biotransformed to hydroxylated metabolites than the more highly chlorinated PCBs. In addition, for the hydroxylated congeners tested, those compounds with strong affinities to estrogen receptors possess either single or multiple artha-chlorine substitutions (Korach et al. , 1987). No artha-substituted TCB metabolites were detected in rats (Yoshimura and Yamamoto, 1974; Yoshimura et al., 1987; Koga et al., 1989) and mice (Darnerud et al. , 1986; Klasson Wehler et al., 1989). Although TCB metabolites show relatively little estrogenic activity, the role they play in reproductive and developmental toxicity can not 8 1 be ignored. In the present study, eggs superovulated by exogenous PMSG and HCG were similar in all groups. Similar results were observed by Brezner et al. (1984) in that the number of ovulations was not affected in mature rats exposed to Aroclor 1254 at a dose of 10 mg/kg bw/day for 1 month. In contrast, both decreases and increases in germ cells have been reported in TCB-exposed mouse offspring (Ronnback, 1991; Ronnback and de Rooij, 1994). The different results appear to be dependent on exposure regimens in the 2 studies. F-O females in the Ronnback (1991) study were only injected intraperitoneally on day 13 of gestation with 1.5-15 mg TCB/kg bw. However, in the study by Ronnback and de Rooij (1994), F0 females were gavaged with TCB at 9 or 15 mg TCB/kg bw weekly for 2 weeks prior to mating. The treatment continued through gestation and lactation for a total of 7 doses. The increases in germ cells in F—l females were associated with the inhibition of atresia (Ronnback and de Rooij, 1994). Male Reproductive Toxicity In this study, testis weights increased in F-l males at 3 weeks of age, but not at 9 and 19 weeks of age. Similar results were also observed in albino mice (Johansson, 1987). Increases in relative testis weights (mg/ 100 g bw) were noted in mice treated perinatally with 6-CB at 200 or 400 mg/kg bw doses, but not in mice treated in the pubertal period (Johansson, 1987). Testis weights were not affected in studies where adult rats were exposed to 500 ppm Aroclor 1254 (Garthoff et al. , 1977) or in adult mice treated with 200 or 1000 ppm Aroclor 1254 (Sanders et al., 1974, 1977). In contrast, increased testis weights of offspring were observed in pubertal rats when dams were 82 treated with 32 mg Aroclor 1254/kg bw on days 1, 3, 5, 7, and 9 of lactation (Sager et al. , 1983). Existing knowledge suggests two possible mechanisms for these effects. One is the development of a blood-testis barrier which may play a role in decreasing testis toxicity in adults. The blood-testis barrier develops at the time of puberty and just prior to the onset of spermatogenesis (Sever and Hessol, 1985; Thomas, 1991). In rats, the barriers between pairs of Sertoli cells form at approximately 18 days of age (Gilula et al. , 1976). A second possible mechanism is the induction of hepatic microsomal enzymes. Induction of N-demethylase in treated weanling rats was 3.5-4.2 times that of the control group, while treated adults had induction of 2.4-2.8 times that of control group. This pattern also exists with 0-ethyl 0-(4-nitrophenyl)phenyl phosphonothioate (EPN) detoxification activity (Chen and DuBois, 1973). Uridine diphosphoglucuronic acid glucuronyltransferase (UDPGT), one of the major enzymes in the second phase of biotransformation, converts both exogenous and endogenous compounds to polar and water-soluble compounds (Sipes and Gandolfi, 1991). When pregnant rats were orally administered 3 mg TCB/kg bw daily on days 8 - 18 of gestation, the hepatic UDPGT activity in the offspring increased at 3 weeks of age, but diminished by 8 weeks of age (Lucier and McDaniel, 1979). The increased induction of hepatic enzymes may enhance the metabolism of testosterone in weanlings. The enlargement of the testis may have been compensation for the enhanced testosterone metabolism by PCB-induced hepatic microsomal enzymes (Risebrough and Brodine, 1970; Haake-McMillan and Safe, 1991). Testicular toxicity may result in impairment of spermatogenesis. In this study, 83 epididymal sperm concentration was similar among the treatment groups at both 9 and 19 weeks of age. To the contrary, Sanders et al. (1977) reported that 200 ppm Aroclor 1254 in the feed of adult albino male mice significantly reduced the number of sperm cells per milligram of testis. A similar effect of Aroclor 1254 was observed in adult white-footed male mice (Sanders and Kirkpatrick, 1975). Exposure to 400 ppm Aroclor 1254 for 2 weeks reduced the number of sperm per testis. There was no effect on the number of sperm cells at 100 or 200 ppm (Sanders and Kirkpatrick, 1975). Compared with those observations, the doses which affected sperm production were much higher than the high dose in the present study. The sites used to measure the number of sperm may have led to the diversity of results. The authors measured the number of sperm in the testis (Sanders and Kirkpatrick, 1975; Sanders et al. , 1977), however, in the present study, the epididymal sperm concentration was measured. Although enlarged testes were observed at 3 weeks of age, epididymal sperm concentration was not affected when mice were at 9 and 19 weeks. Decreases in egg fertilizing ability in vitra were observed with sperm from 30 ppm-treated F-l males at 19 weeks of age, but not at 9 weeks of age. In another study, a decrease in the normal number of embryos was observed as a result of the mating of non-treated female rats with treated males dosed with 8 mg Aroclor 1254/kg bw on days 1, 3, 5, 7, and 9 of lactation (Sager et al., 1987, 1991). Based on the existing information, there are two possible mechanisms associated with this decreased sperm quality. The first one is testicular oxidative stress. Two of the antioxidant enzymes, superoxide dismutase (SOD) and catalase, decreased in mature rats after a single i.p. 34 injection of 100 mg Clophen A 50/kg bw (Peltola et al., 1994). In addition, testicular mRNAs for NADPH-generating enzymes decreased by 23-32% in rats fed 200 ppm Aroclor 1254 for 3 days (Hitomi et al., 1993). NADPH, generated primarily by the reactions catalyzed by malic enzyme (ME), glucose-6-phosphate dehydrogenase (G6PD), and 6-phosphogluconate dehydrogenase (6PGD) (V oet and Voet, 1990), is necessary for the reduction of glutathione disulfide to glutathione. Secondly, administration of TCB throughout gestation and lactation is likely to disrupt hormonal balance during postnatal development, thus affecting spermatogenesis. Decreases in plasma testosterone were observed in 5- and lO-week-old male offspring of dams gavaged with 3 mg TCB/kg bw/day on day 6 through 18 of gestation (Vincent et al., 1992). Stem cells differentiate into type A spermatogonia during early postpartum. The number of type A spermatogonia continues to increase until 35 days of age, and decreases thereafter in mice (Foster, 1989; Vergouwen et al., 1993). Rapidly dividing and differentiating spermatogonia are the cells most susceptible to chemical toxicity during spermatogenesis (Meistrich, 1986). Sensitivity of stem cells to toxic materials in this critical period explains, in part, the affected sperm quality in this study and in those studies by Sager et al. (1987 and 1991). Exposure during the critical postnatal period (approximately one week) may affect subsequent reproductive performance. In the Sager studies, lactating dams were treated orally on days 1, 3, 5, 7, and 9 postpartum. Reduced incidences of implantation were observed in non-treated females mated with treated males. The lowest observable adverse effect level was the maternal oral dose of 8 mg Aroclor 1254/kg bw every 2 days during days 1-9 of lactation. However, no effect 85 on fertility was observed by Kihlstrom et al. (1975) when male mice offspring exposed to PCBs during suckling were mated to non-treated females. In that study, those offspring were exposed to PCBs only from dams injected with 50 mg Clophen A 60/kg bw subcutaneously on the day of parturition and repeated weekly for three successive weeks (Kihlstrom et al., 1975). The frequency of administration during the critical postnatal period in the Sager studies is higher than that in the study by Kihlstrom et al. (1975). This may account for their different results. Parameters assessed by sperm motion analysis were similar among all treatment groups, except for the decreased motility observed in the 30 ppm-treated males at 2.5 hours after collection. These parameters included velocity, linearity, ALH displacement, beat/cross frequency, and motility. The decreased sperm motility was probably associated with inhibition of ATPase inhibition (Ia Rocca and Carlson, 1979). A significant decrease in total ATPase activity of liver, kidney, and brain tissue was observed in rats treated with 25 mg Aroclor 1242/kg bw for 7 days (Ia Rocca and Carlson, 1979). It is well established that ATPase causes the hydrolysis of ATP to ADP with the release of energy. This is critical for sperm motility. Fecundity and litter sizes were the same in non-treated females bred with treated F-l males at 7 and 17 weeks of age. The 21-day survival index was decreased in the offspring of treated F-l males at 60 weeks of age in the 3 ppm and 30 ppm groups. Sperm from the treated males was the only factor resulting in decreased survival of offspring. It is, therefore, hypothesized that TCB exposure must have altered some characteristic of sperm. This conclusion should be taken conservatively due to the small sample sizes in this observation. Only 86 1-2 males in each treatment group (4-5 males/group) at 60 weeks of age were fertile after 3 months of breeding with non-treated females. Sexual Differentiation Androgenic deficiency caused by exposure to TCDD and PCBs during prenatal and early postnatal periods can impair male reproductive function by disrupting the development of sex organs and impairing sexual differentiation of the central nervous system. Hormones regulate sexual differentiation in the critical periods during both prenatal and early postnatal stages (George and Wilson, 1988; Hadley 1992). Testosterone and DHT are responsible for the differentiation of male internal genital ducts and adult secondary sex characteristics, and external genitalia, respectively (Hadley, 1992). When testosterone in the brain is transformed into estradiol by aromatase, the estradiol controls the differentiation of male hypothalamus (Hadley 1992). Sex ratios were similar throughout this study in both treated males and females. In contrast, the sex ratios (6/ 9) of neonates decreased when pregnant rats were treated by gavage with a single dose of 3 mg TCB/kg bw from day 6 to day 18 of gestation (the exact day was not reported) (Simmons et al. , 1984). The high dose used in the present study was approximately 7.5 and 5.9 mg TCB/kg bw/day before breeding and from day 1 to day 16 of gestation, respectively. It is likely that the induction of hepatic microsomal monooxygenases biotransformed the highly toxic TCB into less toxic metabolites when dams were treated continuously in the present study. However, in the study by Simmons et al., TCB was administered during the critical period when organogenesis occurred. Large variations of sex ratios were observed in all groups 87 which made it difficult to establish a statistical difference in sex ratios among treatment groups. A decrease in testosterone concentrations was observed in the fetus on days 18 through 21, and shortly after birth when dams were treated with 1.0 pg TCDD/kg bw on day 15 of gestation (Mably et al., 1992). Anogenital distances, measured 1-5 days after parturition, were shorter in the TCDD exposed neonates than those in the controls. Thus, TCDD and PCBs may cause feminization by disrupting the hormonal balance, especially that of testosterone and DHT. CONCLUSIONS Liver weights increased in the 30 ppm-treated lactating F-0 mice and in all F-l mice except for the weanling males. Exposure period and induction of hepatic microsomal enzymes may explain the differences of liver weights between F-l male weanlings and adults. Thymus weights only decreased in the F—1 females between 5 and 7 weeks of age. In this study, thymic effects were shown to be age and sex dependent. Fecundity only decreased in the 30 ppm-treated F-0 group, but not in the F-1 mice. Unaffected fecundity in F-l mice can be attributed to small sample sizes. Survival of the offspring of treated F-0 and F-l females decreased. This increased mortality is probably associated with either the behavioral changes of dams or direct toxicity caused by TCB and its metabolites, or any combination of the two factors. Increased testis weights were only observed in the F-1 weanlings, not in the pubertal and mature mice. Development of blood-testis junction and hepatic microsomal enzymes may contribute to this result. Decreased gamete fertilizing ability was observed in both treated F-l males and females. Reproductive performance and gamete integrity were impaired by TCB exposure. Since testosterone, DHT, and estrogen are critical for organogenesis and gametogenesis, these effects are in all likelihood predominantly associated with the hormonal imbalance caused by TCB treatment. 88 APPENDIX APPENDIX I Appendix I. Specific activity of l‘C-TCB in feed 1 kg of 3 ppm TCB fgog Specific activity of l‘C-TCB = “C-TCB/total TCB = 9.52 pCi/3 mg TCB = 3.17 pCi/mg TCB 1 kg of 30 pom TCB foeo Specific activity of l‘C-TCB= I‘C-TCB/total TCB = 9.52 pCi/30 mg TCB = 0.317 pCi/mg TCB 89 APPENDIX II Appendix II. Calculation of concentrations of TCB equivalents in the sample. DPM = CPM/efficiency of liquid scintillation counter for 1“C = CPM/0.90 Radioactivity (pCi/g) = 2{[(DPM-background DPM)/(2.22 X 10")]/sample weight} Concentrations of TCB equivalents = Radioactivity/specific activity of l‘C-TCB in feed BIBLIOGRAPHY BIBLIOGRAPHY Abdel-Hamid, F. M., J. A. Moore, and H. B. Matthews. 1981. Comparative study of 3,4,3’,4’-tetrachlorobiphenyl in male and female rats and female monkeys. J. Toxicol. Environ. Hlth. 7: 181-191. .Agrawal, A. K., H. A. Tilson, and S. C. Bondy. 1981. 3,4,3’,4’-tetrachlorobiphenyl given to mice prenatally produces long-term decreases in striatal dopamine and receptor binding sites in the caudate nucleus. Toxicol. Lett. 7: 417-424. Allen, J. R., D. A. Barsotti, L. K. Iambrecht, and J. P. Van Miller. 1979. Reproductive effects of halogenated aromatic hydrocarbons on nonhuman primates. Ann. N. Y. Acad. Sci. 320: 419-425. Anon. 1966. Report of a new chemical hazard. New Scientist. 32: 612. Atlas, E.,‘ T. Bidleman, and C. S. Giam. 1986. Atmospheric transport of PCBs to the oceans. In: PCBs and The Environment. Volume I. pp. 79-100. Ed. J. S. Wald. CRC Press, Boca Raton, FL. Aulerich, R. J., S. J. Bursian, W. J. Breslin, B. A. Olson, and R. K. Ringer. 1985. Toxic2ological manifestations of 2,4,5,2’,4’,5’-, 2,3,6,2’,3’,6’-, and 3,4,5,3’,4’,5’-hexachlorobiphenyl and Aroclor 1254 in mink. J. Toxicol. Environ. Hlth. 15: 63-79. Bandiera, S., S. Safe, and A. B. Okey. 1982. Binding of polychlorinated biphenyls classified as either phenobarbitone-, 3-methylcholanthrene-, or mixed-type inducers to cytosolic Ah receptor. Chem. Biol. Interact. 39: 259-277. Barsotti, D. A., R. J. Marlar, and J. R. Allen. 1976. Reproductive dysfunction in rhesus monkeys exposed to low levels of polychlorinated biphenyls (Aroclor 1248). Fd. Cosmet. Toxicol. 14: 99-103. Bates, M. N., D. J. Hannah, S. J. Buckland, J. A. Taucher, and T. v. Maanen. 1994. Chlorinated organic contaminants in breast milk of New Zealand women. Environ. Hlth. Persp. Supple. 102(Supple 1): 211-217. 91 92 Bitman, J. and H. C. Cecil. 1970. Estrogenic activity of DDT analogs and polychlorinated biphenyls. J. Agr. Food Chem. 18(6): 1108-1112. Bleavins, M. R., W. J. Breslin, R. J. Aulerich, and R. K. Ringer. 1984. Placental and mammary transfer of a polychlorinated biphenyl mixture (Aroclor 1254) in the European ferret (Mustela putarius fura). Environ. Toxicol. Chem. 3: 637-644. Brandt, 1., P. O. Darnerud, A. Bergman, and Y. Larsson. 1982. Metabolism of 2,4’,5- trichlorobiphenyl: Enrichment of hydroxylated and methyl sulphone metabolites in the uterine luminal fluid of pregnant mice. Chem-Biol. Interact. 40: 45-56. Brezner, E., J. Terkel and A. S. Perry. 1984. The effects of Aroclor 1254 (PCB) on the physiology of reproduction in the female rat-I. Comp. Biochem. Physiol. 770(1): 65-70. Burse, V. W., R. D. Kimbrough, E. C. Villaneuva, R. W. Jennings, R. E. Linder, and G. W. Sovocool. 1974. Polychlorinated biphenyls storage, distribution, excretion, and recovery: Liver morphology after prolonged dietary ingestion. Arch. Environ. Hlth. 29: 301-307. Chen, P. H., K. T. Chang, and Y. D. Lu. 1981. Polychlorinated biphenyls and polychlorinated dibenzofurans in the toxic rice-bran oil that caused PCB poisoning in Taichung. Bull. Environ. Contam. Toxicol. 26: 489-495. Chen, P. H. and S.-T. Hsu. 1989. PCB poisoning from toxic rice-bran oil in Taiwan. In: PCBs and The Environment. Volume 111. pp. 27-38. Ed. J. S. Waid. CRC Press, Boca Raton, FL. Chen, S.-W., B. Magnus Francis, and P. J. Dziuk. 1993. Effects of concentration of mixed-function oxidase on concentration of estrogen, rate of egg lay, eggshell thickness, and plasma calcium in laying hens. J. Anim. Sci. 71: 2700-2707. Chen, T. S. and K. P. Dubois. 1973. Studies of the enzyme inducing effect of polychlorinated biphenyls. Toxicol. Appl. Pharmacol. 26:504-512. Clarke, D. W., J. F. Brien, W. J. Racz, K. Nakatsu, and G. S. Marks. 1984. The disposition and the liver and thymus gland toxicity of 3,3’,4,4’- tetrachlorobiphenyl in the female rat. Can. J. Physiol. Pharmacol. 62: 1253-1260. Curley, A., V. W. Burse, and M. E. Grim. 1973. Polychlorinated biphenyls: Evidence of transplacental passage in the Sherman rat. Fd. Cosmet. Toxicol. 11: 471—476. d’Argy, R., L. Denker, E. Klasson-Wehler, A. Bergman, P. O. Darnerud, and I. Brandt. 1987. 3,3’,4,4’-Tetrachlorobiphenyl in pregnant mice: Embryotoxicity, 93 teratogenicity, and toxic effects on the cultured embryonic thymus. Pharmacol. Toxicol. 61: 53-57. Darnerud, P. 0., I. Brandt, E. Klasson-Wehler, A. Bergman, R. d’Argy, L. Dencker, and G. O. Sperber. 1986. 3,3’,4,4’-Tetrachloro[14C]biphenyl in pregnant mice: Enrichment of phenol and methyl sulphone metabolites in late gestational fetuses. Xenobiotica 16(4): 295-306. Dencker, L. and R. M. Pratt. 1981. Association between the presence of the Ah receptor in embryonic murine tissues and sensitivity to TCDD-induced cleft palate. Teratog. Carcinog. Mutag. 1: 399—406. Duax, W. L. and J. F. Griffin. 1985. Structur-activity relationships of estrogenic chemicals. In: Estrogens in The Environment 11: Influence on Development. PP. 15-23. Ed. J. A. McIachlan. Elsevier Science Publishing Co., Inc. New York, NY. Ecobichon, D. J. and D. O. MacKenzie. 1974. The uterotropic activity of commercial and isomerically-pure chlorobiphenyl in the rat. Res. Commun. Chem. Pathol. Pharmacol. 9(1): 85-95. Erickson, G. F., D. A. Magoffin, C. A. Dyer, and C. Hofeditz. 1985. The ovarian androgen producing cells: A review of structure/function relationships. Endocri. Rev. 6(3): 371-399. Faith, R. E. and J. A. Moore. 1977. Impairment of thymus-dependent immune functions by exposure of the developing immune system to 2,3,7,8-tetrachlorodibenzo-p- dioxin (TCDD). J. Toxicol. Environ. Hlth. 3: 451-464. Fishbein L. 1974. Toxicity of chlorinated biphenyls. Annu. Rev. Pharmacol. 14: 139- 156. Foster, P. M. D. 1989. Testicular structure and physiology: A toxicologist’s view. In: Toxicology of The Male and Female Reproductive systems. pp. 1-14. Ed. P. K. Working. Hemisphere Publishing Corporation, New York, NY. Freeman, H. C., G. Sangalang, and B. Flemming. 1982. The sublethal effects of a polychlorinated biphenyl (Aroclor 1254) diet on the Atlantic cod (Gadus morhua). Sci. Total Environ. 24: 1-11. Friend, M. and D. 0. Trainer. 1970. Polychlorinated biphenyl: Interaction with duck hepatitis virus. Science 170: 1314. Gage, J. C. and S. Holm. 1976. The influence of molecular structure of the retention and 94 excretion of polychlorinated biphenyls by the mouse. Toxicol. Appl. Pharmacol. 36: 555-560. Gallenberg, L. A. and M. J. Vodicnik. 1987. Potential mechanism for redistribution of polychlorinated biphenyls during pregnancy and lactation. Xenobiotica 17(3): 299- 3 10. Garthoff, L. H., L. Friedman, T. M. Farber, K. K. Locke, T. J. Sobotka, S. Green, N. E. Hurley, E. L. Peters, G. E. Story, F. M. Moreland, C. H. Graham, J. E. Keys, M. J. Taylor, J. V. Scalera, J. E. Rothlein, E. M. Marks, F. E. Cerra, S. B. Rodi, E. M. Sporn. 1977. Biochemical and cytogenetic effects in rats caused by short-term ingestion of Aroclor 1254 or Firemaster BP6. J. Toxicol. Environ. Hlth. 3: 769-796. Gellert, R. J. 1978. Uterotrophic activity of polychlorinated biphenyls (PCB) and induction of precocious reproductive aging in neonatally treated female rats. Environ. Res. 16: 123-130. Gellert, R. J. and C. Wilson. 1979. Reproductive function in rats exposed prenatally to pesticides and polychlorinated biphenyls (PCB). Environ. Res. 18: 437-443. George, F. W. and J. D. Wilson. 1988. Sex determination and differentiation. In: The Physiology of Reproduction. pp. 3-26. Eds. E. Knobil, J. D. Neill, L. L. Ewing, G. S. Greenwald, C. L. Markert, and D. W. Pfaff. Raven Press, New York, NY. Golub, M. S., J. M. Donald, and J. A. Reyes. 1991. Reproductive toxicity of commercial PCB mixtures: LOAELs and NOAELs from animal studies. Environ. Hlth. Persp. 94: 245-253. Gilula, N. B., W. Fawcett, and A. Aoki. 1976. The Sertoli cell occluding junctions and gap junctions in mature and developing mammalian testes. Dev. Biol. 50: 142- 168. Guo, Y. L., C. J. Lin, W. J. Yao, J. J. Ryan, and C. C. Hsu. 1994. Musculoskeletal changes in children prenatally exposed to polychlorinated biphenyls and related compounds (Yu-Cheng children). J. Toxicol. Environ. Hlth. 41: 83-93. Haake-McMillan, J. M. and S. H. Safe. 1991. Neonatal exposure to Aroclor 1254: Effects on adult hepatic testosterone hydroxylase activities. Xenobiotica 21: 481- 489. Hadley, M. E. 1992. In: Endocrinology. 606 pp. Ed. M. E. Hadley. Pretice-Hall Inc., Englewood Cliffs, NJ. 95 Hansen, L. G. 1987. Food chain modification of the composition and toxicity of polychlorinated biphenyl (PCB) residues. Rev. Environ. Toxicol. 3: 149-212. Harris, C. and W. S. Bradshaw. 1984. Alterations in liver ultrastructure and induction of UDP-glucuronyltransferase in the rat following prenatal exposure to 3,4,3’,4’- tetrachlorobiphenyl. Arch. Environ. Contam. Toxicol. 13: 715-721. Harris, G. W. 1964. Sex hormones, brain development and brain function. Endocri. 75:627-648. Hemminki, K. and P. Vineis. 1985. Extrapolation of the evidence on teratogenicity of chemicals between humans and experimental animals: Chemicals other than drugs. Teratogen. Carcino. Mutagen. 5: 251-318. Hileman, B. 1994. Environmental estrogens linked to reproductive abnormalities, cancer. C & EN. January. 19-23. Hirayama, C. 1976. Clinical aspects of PCB poisoning. In: PCB Poisoning and Pollution. Ed. K. Higuchi. pp. 87- 104. Academic Press, New York. Hitomi, Y., M. Wakayama, H. Oda, and A. Yoshida. 1993. Liver-specific induction of N ADPH-generating enzymes by polychlorinated biphenyls in rats. Biosci. Biotech. Biochem. 57(7): 1134-1136. Jacobson, S. W., J. L. Jacobson, P. M. Schwartz, and G. G. Fein. 1983. Intrauterine exposure of human newborns to PCBs: Measures of exposure. In: PCBs: Human and Environmental Hazards. pp. 311-343. Eds. F. M. D’ltri and M. A. Kamrin. Butterworth Publisher, Woburn, MA. Jonsson, H. T. Jr., J. E. Keil, R. G. Gaddy, C. B. Loadholt, G. R. Hennigar, and E. M. Walker, Jr. 1976. Prolonged ingestion of commercial DDT and PCB: Effects on progesterone levels and reproduction in the mature female rat. Arch. Environ. Contam. 3: 479-490. Johansson, B. 1987. Lack of effects of polychlorinated biphenyls on testosterone synthesis in mice. Pharmacol. Toxicol. 61: 220-223. Kashimoto, T. and H. Miyata. 1986. Differences between Yusho and other kinds of poisoning involving only PCBs. In: PCBs and The Environment. Vol. 111. pp. 1- 26. Ed. J. S. Waid. CRC Press, Boca Raton, FL. Kato, N. and A. Yoshida. 1980. Effect of dietary PCB on hepatic cholesterogenesis in rats. Nutr. Rep. Int. 21: 107-112. 96 Kholkute, S. D., J. Rodriguez, and W. R. Dukelow. 1994. Effects of polychlorinated biphenyls (PCBs) on in vitro fertilization in the mouse. Reprod. Toxicol. 8(1): 69-73. Kihlstrom, J. E., C. Lundberg, J. Orberg, P. O. Danielsson, and J. Sydhoff. 1975. Sexual functions of mice neonatally exposed to DDT or PCB. Environ. Physiol. Biochem. 5: 54-57. Kikuchi, M. and Y. Masuda. 1976. The pathology of Yusho. In: PCB Poisoning and Pollution. pp. 69-86. Ed. K. Higuchi. Kodansha Ltd. Tokyo. Kilburn, K. H., R. H. Warsaw, and M. G. Shields. 1989. Neurobehavioral dysfunction in fireman exposed to polychlorinated biphenyls (PCBs): Possible improvement after detoxification. Arch. Environ. Hlth. 44(6): 345-350. Klasson Wehler, E., A. Bergman, I. Brandt, P. O. Darnerud, and C. A. Wachtmeister. 1989. 3,3’ ,4,4’-tetrachlorobiphenyl excretion and tissue retention of hydroxylated metabolites in the mouse. Drug Metab. Dispos. 17(4): 441-448. Klasson Wehler, E., B. Brunstrom, U. Rannug, and A. Beggman. 1990. 3,3’,4,4’- Tetrachlorobiphenyl: Metabolism by the chick embryo in ovo and toxicity of hydroxylated metabolites. Chem-Biol. Interact. 73: 121-132. Koga, N., M. Beppu, C. Ishida, and H. Yoshimura. 1989. Further studies on metabolism in viva of 3,4,3’,4’-tetrachlorobiphenyl in rats: Identification of minor metabolites in rat faeces. Xenobiotica. 19(11): 1307-1318. Koga, N., M. Beppu, and H. Yoshimura. 1990. Metabolism in viva of 3,4,5,3’,4’- pentachlorobiphenyl and toxicological assessment of the metabolite in rats. J. Pharmacobio-Dyn. 13: 497-506. Korach, K. S., P. Sarver, K. Chae, J. A. Mclachlan, and J. D. Mckinney. 1987. Estrogen receptor binding activity of polychlorinated hydroxybiphenyls: Conformationally restricted structural probes. Mol. Pharmacol. 33: 120-126. Kuo, J., E. Fox, and S. McDonald. 1992. In: SigmaStat: User’s Manual. 13-60 pp. Jandel Scientific. San Rafael, CA. La Rocca, P. T. and G. P. Carlson. 1979. The effect of polychlorinated biphenyls on adenosine triphosphatase activity. Toxicol. Appl. Pharmacol. 48: 185-192. Leece, B., M. A. Denomme, R. Towners, S. M. Angela Li, and S. Safe. 1985. Polychlorinated biphenyls: Correlation between in vivo and in vitro quantitative structure-activity relationships (QSARs). J. Toxicol. Environ. Hlth. 16: 379-388. 97 Lincer, J. L. and D. B. Peakall. 1970. Metabolic effects of polychlorinated biphenyls in the American kestrel. Nature 228: 783-784. Linzey, A. V. 1987. Effects of chronic polychlorinated biphenyls exposure on reproductive success of white-footed mice (Peromyscus leucopus). Arch. environ. Contam. Toxicol. 16: 455-460. Linzey, A. V. 1988. Effects of chronic polychlorinated biphenyls on growth and reproduction of second generation white-footed mice (Peromyscus leucopus). Arch. Environ. Contam. Toxicol. 17: 39-45. Lucier, G. W., O. S. McDaniel, C. M. Schiller, and H. B. Matthews. 1978. Structural requirements for the accumulation of chlorinated biphenyl metabolites in the fetal rat intestine. Drug Metab. Dispos. 6(5): 584-590. Lucier, G. W. and O. S. McDaniel. 1979. Developmental toxicology of the halogenated aromatics: Effects on enzyme development. Ann. N. Y. Acad. Sci. 320: 449-457. Lutz, R. J., R. L. Dedrick, H. B. Matthews, T. E. Eling, and M. W. Anderson. 1977. A preliminary pharmacokinetic model for several chlorinated biphenyls in the rat. Drug Metab. Dispos. 5: 386-396. Lutz, R. J. and R. L. Dedrick. 1987. Physiologic pharmacokinetic modeling of polychlorinated biphenyls. In: Polychlorinated Biphenyls (PCBs) .° Mammalian and Environmental Toxicology. pp. 111-131. Ed. S. Safe. Splinger-Verlag Press, Berlin. Mably, T. A., R. W. Moore, and R. E. Peterson. 1992. In utero and lactational exposure of male rats to 2,3,7,8-tetrachlorodibenzo-p-dioxin. 1. Effects on androgenic status. Toxicol. Appl. Pharmacol. 114: 97-107. MacLellan, K., A. Singh, 1. Chu, and D. C. Villeneuve. 1994. Subchronic toxicity of 3,3’,4,4’-tetrachlorobiphenyl in the rat liver: A electric microscope study. Histol. Histopath. 9:453-459. Maliwal, B. P. and F. Guthrie. 1982. In vitro uptake and transfer of chlorinated hydrocarbons among human lipoproteins. J. Lipid Res. 23: 474-479. Marks, T. A., G. L. Kimmel, and R. E. Staples. 1981. Influence of symmetrical polychlorinated biphenyl isomers on embryo and fetal development in mice. Toxicol. Appl. Pharmacol. 61: 269-276. Marks, T. A. and R. E. Staples. 1980. Teratogenic evaluation of the symmetrical isomers of hexachlorobiphenyl (HCB) in the mouse. In: Prac. 20th Annu. Meet. 98 Teratal. Sac. p. 54A. Matthews, H. B. and D. B. Tuey. 1980. The effect of chlorine position on the distribution and excretion of four hexachlorobiphenyl isomers. Toxicol. Appl. Pharmacol. 53: 377-388. Matthews, H. B. and M. W. Anderson. 1975. Effects of chlorination on the distribution and excretion of polychlorinated biphenyls. Drug. Metab. Dispos. 3: 371- 380. Matthews, H. B. and R. L. Dedrick. 1984. Pharmacokinetics of PCBs. Ann. Rev. Pharmacol. Toxicol. 24: 85-103. McConnell, E. E., J. A. Moore, J. K. Haseman, and M. W. Harris. 1978. The comparative Toxicity of chlorinated dibenzo—p—dioxin in mice and guinea pigs. Toxicol. Appl. Pharmacol. 44: 335-356. McConnell, E. E. and J. A. Moore. 1979. Toxicopathology characteristics of the halogenated aromatics. Ann. N. Y. Acad. Sci. 320: 138-150. McConnell, E. E. 1985. Comparative toxicity of PCBs and related compounds in various species of animals. Environ. Hlth. Persp. 60: 29—33. McConnell, E. E. 1989. Acute and chronic toxicity and carcinogenesis in animals. In: Halagenated Biphenyls, Terphenyls, Naphthalenes, Dibenzodiaxins and Related Products. pp. 161-193. Eds. R. D. Kimbrough and A. A. Jensen. Elsevier, New York, NY. Mclachlan, M. S. 1993. Digestive tract absorption of polychlorinated dibenzo-p-dioxin, dibenzofurans, and biphenyls in a nursing infant. Toxicol. Appl. Pharmacol. 123: 68-72. McNatty, K. P., D. M. Smith, A. Makris, R. Osathanondh, and K. J. Ryan. 1979. The microenvironment of the human antral follicle: Interrelationships among the steroid levels in antral fluid, the population of granulosa cells, and the status of the oocyte in vivo and in vitro. J. Cli. Endocri. Meta. 49(6): 851-860. Meistrich, M. L. 1986. Critical components of testicular function and sensitivity to disruption. Biol. Reprod. 34: 17-28. Merson, M. H. and R. L. Kirkpatrick. 1975. Effects of dietary exposure to PCB on . testicular characteristics, organ weights, and plasma corticoids of male white- footed mice. The Virginia Journal of Science 26: 58 (Abstract). 99 Mizutani, T., K. Hidaka, T. Ohe, and M. Matsumoto. 1977. A comparative study on accumulation and elimination of tetrachlorobiphenyl isomers in mice. Bull. Environ. Contam. Toxicol. 18(4): 452-461. Morrissey, R. E. and B. A. Schwetz. 1989. Reproductive and developmental toxicity in animals. In: Halagenated Biphenyls, Terphenyls, Naphthalenes, Dibenzodioxins and Related Products. pp. 195-225. Eds. R. D. Kimbrough and A. A. Jensen. Elsevier, New York. Neal, R. A. 1985. Mechanisms of the. biological effects of PCBs, polychlorinated dibenzo-p—dioxins and polychlorinated dibenzofurans in experimental animals. Environ. Hltll. Persp. 60: 41-46. Nebert, D. W. 1989. The Ah locus: Genetic differences in toxicity, cancer, mutation, and birth defects. Critical Rev. Toxicol. 20(3): 137-152. Orberg, J. and J. E. Kihlstrom. 1973. Effects of long-term feeding of polychlorinated biphenyls (PCB, Clophen A 60) on the length of the oestrous cycle and on the frequency of implanted ova in the mouse. Environ. Res. 6: 176-179. Orberg, J. 1978a. Effects of pure chlorobiphenyls (2,4’,5-trichlorobiphenyl and 2,2’,4,4’,5,5’-hexachlorobiphenyl) on the reproductive capacity in female mice. Acta. Pharmacol. et. Toxicol. 42: 323-327. Orberg, J. 1978b. Effects of pure chlorobiphenyls (2,4’,5-trichlorobiphenyl and 2,2’ ,4,4’,5,5’-hexachlorobiphenyl) on the post-natal growth. Acta. Pharmacol. et. Toxicol. 42: 275-279. Overrnann, S. R., J. Kostas, L. R. Wilson, W. Shain, and B. Bush. 1987. Neurobehavioral and somatic effects of perinatal PCB exposure in rats. Environ. Res. 44: 56—70. Pantaleoni, (3., D. Fanini, A. M. Sponta, G. Palumbo, R. Giorgi, and P. M. Adams. 1988. Effects of maternal exposure to polychlorobiphenyls (PCBs) on F1 generation behavior in the rat. Fundam. Appl. Toxicol. 11: 440-449. Parkinson, A. and S. Safe. 1981. Aryl hydrocarbon hydroxylase induction and its relationship to the toxicity of halogenated aryl hydrocarbons. Toxicol. Environ. Chem. Rev. 4: 1-45. Parkinson, A., S. H. Safe, L. W. Robertson, P. E. Thomas, D. B. Ryan, L. M. Reik, and W. Levin. 1983. Immunochemical quantitation of cytochrome P-450 isozymes and epoxide hydroxylase in liver microsomes from polychlorinated or polychlorinated biphenyl-treated rats. J. Biol. Chem. 258(9): 5967-5976. 100 Parkinson, A. and S. Safe. 1987. Mammalian biologic and toxic effects of PCBs. In: Polychlorinated Biphenyls (PCBs) .° Mammalian and Environmental Toxicology. pp. 49-75. Ed. S. Safe. Splinger-Verlag Press, Berlin. Peltola, V., E. Mantyla, I. Huhtaniemi, and M. Ahotupa. 1994. Lipid peroxidation and antioxidant enzyme activities in the rat testis after cigarette smoke inhalation or administration of polychlorinated biphenyls or polychlorinated naphthalenes. J. Androl. 15(4): 353-361. Peters, H. 1979. Some aspects of early follicular development. In: Ovarian Follicular DevelopmentandFunction. pp. 1-15. Eds. A. R. Midgley, Jr. andW. A. S.adler Plenum, New York, NY. Peterson, R. E., H. M. Theobald, and G. L. Kimmel. 1993. Developmental and reproductive toxicity of dioxins and related compounds: Cross-species comparisons. Crit. Rev. Toxicol. 23(3): 283-335. Phillips, D. J. H. 1986. Use of organism to quantify PCBs in marine and estuarine environments. In: PCBs and The Environment. Vol. II. pp. 127-181. Ed. J. S. Waid. CRC Press, Boca Raton, FL. Platonow, N. S. and H. S. Funnell. 1971. Anti-androgenic-like effects of polychlorinated biphenyls in cockerels. Vet. Rec. 89:109-110. Poland, A. and J. Knutson. 1982. 2,3,7,8—tetrachlorodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: Examination of the mechanism of toxicity. Ann. Rev. Pharmacol. Toxicol. 22: 517-554. Rands, P. L., R. D. White, M. W. Carter, S. D. Allen, and W. S. Bradshaw. 1982. Indicators of developmental toxicity following prenatal administration of hormonally active compounds in the rat. I. Gestational length. Teratol. 25: 37-43. Richards, J. S. 1980. Maturation of ovarian follicles: Actions and interactions of pituitary and ovarian hormones on follicular cell differentiation. Physiol. Rev. 60(1): 51- 89. Richardson B. J. and J. S. Waid. 1982. Polychlorinated Biphenyls (PCBs): An Australian viewpoint on a global problem. Search 13 (1-2): 17-25. Ringer, R. K., R. J. Aulerich, and M. Zabik. 1972. Effect of dietary polychlorinated biphenyls on growth and reproduction of mink. Proceedings of the 164th National Meeting of the American Chemical Society. P. WATR41. Risebrough, R. and V. Brodine. 1970. More letters in the wind. Environ. 12(1): 16—27. 101 Rogan, W. J. 1982. PCBs and cola-colored babies: Japan, 1968, and Taiwan, 1979. Teratol. 26: 259-261. Rogan, W. J., B. C. Gladen, K.-L. Hung, S.-L. Koong, L.-Y. Shih, J. S. Taylor, Y.-C. Wu, D. Yang, N. B. Ragan, and C.-C. Hsu. 1988. Congenital poisoning by polychlorinated biphenyls and their contaminants in Taiwan. Science 241: 334-336. Ronnback, C. 1991. Effects of 3,3’,4,4’-tetrachlorobiphenyl (TCB) on ovaries of foetal mice. Pharmacol. Toxicol. 69:340-345. Ronnback, C. and D. G. de Rooij. 1994. Effects of 3,3’,4,4’-tetrachlorobiphenyl on foetal germ cells in two mouse strains after repeated treatment of the dams during and after pregnancy. Pharmacol. Toxicol. 74: 287-293. Ryan, K. J. 1988. Endocrine function of the ovary. In: Endocrinology: People and Ideas. pp. 201-213. Ed. 8. M. McCann. Waverly Press, Inc., Baltimore, MD. Safe, S. 1984. Polychlorinated biphenyls (PCBs) and polybrominated biphenyls (PBBs): Biochemistry, toxicology, and mechanism of action. CRC Crit. Rev. Toxicol. 13(4): 319-395. Safe, S. 1989. Polyhalogenated aromatics: Uptake, disposition and metabolism. In: Halagenated Piphenyls, Terphenyls, Naphthalenes, Dibenzodiaxins and Related Products. pp. 131-159. Eds. R. D. Kimbrough and A. A. Jensen. Elsevier, New York, NY. Safe, S. 1990. Polychlorinated biphenyls (PCBs), dibenzo-p-dioxins (PCDDs), dibenzofurans (PCDFs), and related compounds: Environmental and mechanistic considerations which support the development of toxic equivalency factors (TEFs). CRC Crit. Rev. Toxicol. 21: 51-88. Safe, 8., L. Safe, and M. Mullin. 1987. Polychlorinated biphenyls: Environmental occurrence and analysis. In: Polychlorinated Biphenyls (PCBs): Mammalian and Environmental Toxicology. pp. 1-13. Eds. S. Safe and O. Hutzinger. Springer- Verlag Press, Berlin. Safe, S. H. 1994. Polychlorinated biphenyls (PCBs): environmental impact, biochemical and toxic responses, and implications for risk assessment. Crit. Rev. Toxicol. 24(2): 87-149. Sager, D. B. 1983. Effect of postnatal exposure to polychlorinated biphenyl on adult male reproductive function. Environ. Res. 31: 76-94. 102 Sager, D. B., W. Shih-Schroeder, and D. Girard. 1987. Effects of early postnatal exposure to polychlorinated biphenyls (PCBs) on fertility in male rats. Bull. Environ. Contam. Toxicol. 38: 946-953. Sager D., D. Girard, and D. Nelson. 1991. Early postnatal exposure to PCBs: Sperm function in rats. Environ. Toxicol. Chem. 10:737-746. Sanders, O. T. and R. L. Kirkpatrick. 1975. Effects of a polychlorinated biphenyl (PCB) on sleeping times, plasma corticosteroids, and testicular activity of white-footed mice. Environ. Physiol. Biochem. 5: 308-313. Sanders, O. T., R. L. Kirkpatrick, and P. E. Scanlon. 1977. Polychlorinated biphenyls and nutritional restriction: Their effects and interactions on endocrine and reproductive characteristics of male white mice. Toxicol. Appl. Pharmacol. 40: 91-98. Sanders, O. T., R. L. Zepp, and R. L. Kirkpatrick. 1974. Effects of PCB ingestion on sleeping times, organ weights, food consumption, serum corticosterone and survival of albino mice. Bull. Environ. Contam. Toxicol. 12 (4): 394-399. Sawhney, B. L. 1986. Chemistry and properties of PCBs in relation to environmental effects. In: PCBs and The Environment. Vol. I. pp. 47-64. Ed. J. S. Waid. CRC Press, Boca Raton, FL. Sawyer, T. and S. Safe. 1982. PCB isomers and congeners: Induction of aryl hydrocarbon hydroxylase and ethoxyresorufin-O—deethylase enzyme activities in rat hepatoma cells. Toxicol. Lett. 13: 87-94. Schecter, A., J. Stanley, K. Boggess, Y. Masuda, J. Mes, M. Wolff, P. Furst, C. Furst, K. Wilson-Yang, and B. Chisholm. 1994. Polychlorinated biphenyl levels in the tissues of exposed and nonexposed humans. Environ. Hlth. Persp. Supple. 102(Supple 1): 149-158. Schlebusch, H., U. Wagner, H. van der Ven, S. AI-Hasani, K. Diedrich, and D. Krebs. 1989. Polychlorinated biphenyls: The occurrence of the main congeners in follicular and sperm fluids. J. Clin. Chem. Clin. Biochem. 27: 663-667. Sever, L. E. and N. A. Hessol. 1985. Toxic effects of occupational and environmental chemicals on the testes. In: Endocrine Toxicology. pp. 211-248. Eds. J. A. Thomas, K. S. Korach, and J. A. McIachlan. Raven Press, New York, NY. Shaw, G. R. and D. W. Connell. 1990. Factors controlling bioaccumulation of PCBs. In: PCBs and TheEnviranment. Vol. I. pp. 135-141. Ed. J. S. Waid. CRC Press, Boca Raton, FL. 103 Simmons, D. L., D. M. Valentine, and W. S. Bradshaw. 1984. Different patterns of developmental toxicity in the rat following prenatal administration of structurally adverse chemicals. J. Toxicol. Environ. Hlth. 14: 121-136. Sipes, I. G. and A. J. Gandolfi. 1991. Biotransformation of toxicants In: Casarett and Doull’s Toxicology: The Basic Science of Poisons. pp. 88-126. Eds. M. O. Amdur, J. Doull, and C. D. Klaassen. Pergamon Press, Elmsford, NY. Sipes, I. G. and R. G. Schnellmann. 1987. Biotransformation of PCBs: Metabolic pathways and metabol isms. In: Polychlorinated Biphenyls (PCBs): Mammalian and Environmental Toxicology. pp. 97-110. Ed. 8. Safe. Splinger-Verlag Press, Berlin. Smialowicz, R. J., J. E. Andrews, M. M. Riddle, R. R. Rogers, R. W. Luebke, and C. B. Copeland. 1989. Evaluation of the immunotoxicity of low level PCB exposure in the rat. Toxicol. 56: 197-211. Spindler-Vomachka, M. and M. J. Vodicnik. 1984. Distribution’of 2,4,5,2’,4’,5’- hexachlorobiphenyl among lipoproteins during pregnancy and lactation in the rat. J. Pharmacol. Exp. Ther. 230(2): 263-268. Stone, R. 1994. Environmental estrogens stir debate. Science. 265: 308-310. Takagi, Y., S. Aburada, 1K. Hashimoto, and T. Kitaura. 1986. Transfer and distribution of accumulation (“C) polychlorinated biphenyls from maternal to fetal and suckling rats. Arch. Environ. Contam. Toxicol. 15: 709-715. Takagi, Y., T. Otake, M. Kataoka, Y. Murata, S. Aburada, S. Akasaka, K. Hashimoto, H. Uda, and T. Kitaura. 1976. Studies on the transfer and distribution of [“C] polychlorinated biphenyls from maternal to fetal and suckling rats. Toxicol. Appl. Pharmacol. 38: 549-558. Tanabe, S. , Y. Nakagawa, and R. Tatsukawa. 1981. Absorption efficiency and biological half-life of individual chlorobiphenyls in rats treated with Kanechlor products. Agric. Biol. Chem. 45: 717-726. Tanabe, S. 1988. PCB problems in the future: Foresight from current knowledge. Environ. Pollut. 50: 5-28. Thomas, J. A. 1991. Toxic responses of the reproductive system. In: Casarett and Daull’s Toxicology: The Basic Science of Poisons. pp. 484-520. Eds. M. O. Amdur, J. Doull, and C. D. Klaassen. Pergamon Press. New York, NY. Tilson, H. A., G. J. Davis, J. A. Mclachlan, and G. W. Lucier. 1979. The effects of 104 polychlorinated biphenyls given prenatally on the neurobehavioral development of mice. Environ. Res. 18: 466-474. Topham, J. C. 1980. Do induced sperm-head abnormalities in mice specifically identify mammalian mutagens rather than carcinogens? Mutation Res. 74: 379-387. Truelove, J. F., J. R. Tanner, I. A. Ianglois, R. A. Stapley, D. L. Arnold, and J. C. Mes. 1990. Effect of polychlorinated biphenyls on several endocrine reproductive parameters in the female rhesus monkey. Arch. Environ. Contam. Toxicol. 19: 939-943. ven den Berg, K. J ., C. Zurcher, A. Brouwer, and D. W. van Bekkum. 1988. Chronic toxicity of 3,3’,4,4’-tetrachlorobiphenyl in the marmoset monkey (Callithrix jacchus). Toxicol. 48: 209-224. Vergouwen, R. P. F. A., R. Huiskamp, R. J. Bas, H. L. Roepers-Gajadien, J. A. G. Davis, and D. G. de Rooij. 1993. Postnatal development of testicular cell populations in mice. J. Reprod. Fert. 99: 479-485. Villeneuve, D. C. and D. L. Grant. 1971. Effects of PCB administration on microsomal enzyme activity in pregnant rabbits. Bull. Environ. Contam. Toxicol. 6(2): 120- 128. Vincent, D. R., W. S. Bradshaw, G. M. Booth, R. E. Seegmiller, and S. D. Allen. 1992. Effect of PCB and DES on rat monoamine oxidase, acetylcholinesterase, testosterone and estradiol ontogeny. Bull. Environ. Contam. Toxicol. 48: 884-893. Vodicnik, M. J. and J. J. Lech. 1980. The transfer of 2,4,5,2’,4’,5’-hexachlorobiphenyl to fetuses and nursing offspring. Toxicol. Appl. Pharmacol. 54: 293-300. Vodicnik, M. J. 1986. The effect of pregnancy and lactation on the disposition of [2,4,2’,4’-“C]tetrachlorobiphenyl in the mouse. Fundam. Appl. Toxicol. 6: 53- 61. Voet, D. and J. G. Voet. 1990. In: Biochemistry. 1223 pp. Eds. D. Voet and J. G. Voet. John. Wiley and Sons, New York, NY. Vos, J. G. and R. B. Beems. 1971. Dermal Toxicity studies of technical polychlorinated biphenyls and fractions thereof in rabbits. Toxicol. Appl. Pharmacol. 19: 617- 633. Ward, J. M. 1985. Proliferative lesions of the glandular stomach and liver in F334 rats fed diets containing Aroclor 1254. Environ. Hlth. Persp. 60: 89-95. 105 Yoshimura, H. and H. Yamamoto. 1974. Metabolic studies on polychlorinated biphenyls. IV. Biotransformation of 3,4,3’,4’-tetrachlorobiphenyl, one of the major components of Kanechlor-400. Fukuoka Acta Med. 65(1): 5-11. Yoshimura, H., S. Yoshihara, N. Ozawa, and M. Miki. 1979. Possible correlation between induction modes of hepatic enzymes by PCBs and their toxicity in rats. Ann. N. Y. Acad. Sci. 320: 179-192. Yoshimura, H., Y. Yonemoto, H. Yamada, N. Koga, K. Oguri, and S. Saeki. 1987. Metabolism in viva of 3,3’,4,4’-tetrachlorobiphenyl and toxicological assessment of the metabolites in rats. Xenobiotica. 17(8): 897-910. nrcuran STATE UNIV. LIBRARIES llHIHWIWIWW11111111W1111“11111111111111 31293014203198