首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Prenatal exposure to TCDD interferes with fetal development at doses lower than those causing overt toxicity in adult animals. Exposure to TCDD during development produces alterations in the reproductive system of the developing pups- delayed puberty and reduced sperm counts in males and malformations in the external genitalia of females. The objectives of this study were to determine maternal and fetal tissue concentrations of TCDD after acute exposure and whether these tissue concentrations can be used to estimate the intensity of the developmental abnormalities reported by other laboratories. Pregnant Long Evans rats received a single, oral dose of 0.05, 0.20, 0.80, or 1.0 microg [3H]-TCDD/kg on gestation day (GD) 15, and maternal and fetal tissue concentrations of TCDD were measured on GD16 and GD21. On GD16, maternal liver contained the greatest amount of TCDD (30-47% administered dose). One day after administration of 0.20 microg TCDD/kg on GD15, there were 13.2 pg TCDD/g present in an individual fetus. This concentration is associated with delayed puberty and decreased epididymal sperm counts in male pups as well as malformations in the external genitalia of females. For the responses studied, tissue concentration measured during a critical period of gestation adequately predicts the intensity of the response. In addition, there was a strong correlation between fetal body burden and maternal body burden on GD16. A dose of 0.05 microg TCDD/kg resulted in maternal body burdens of 30.6+/-3.1 and 26.6+/-3.1 ng TCDD/kg on GD16 and GD21, respectively. In conclusion, low-level TCDD exposure during the perinatal stage of life can produce adverse effects within the developing pups and that tissue concentration measured during a critical period is the appropriate dose metric to predict adverse reproductive and developmental effects.  相似文献   

2.
Prenatal exposue to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)interfereswith fetal development at doses lower than those causing overttoxicity in adult animals. In a multigeneration study (Murrayet al., 1979), female rats that were administered 0.01 µgTCDD/kg/day in their diet did not experience reduced fertility;however, reduced fertility was seen in the F1 and F2 generations.Exposure to TCDD during development produces alterations inthe reproductive system of the developing pups, such as delayedpuberty and reduced sperm counts in males (Mably et al., 1992a;Gray et al., 1995) and malformations in the external genitaliaof females (Gray and Ostby, 1995). Therefore, the objectivesof this study were to determine maternal and fetal tissue concentrationsof TCDD that are associated with the adverse reproductive effectsseen by Gray and co-workers. Pregnant Long Evans rats receiveda single oral dose of 1.15 µg [3H]TCDD/kg on GestationDay (GD) 8 and maternal as well as fetal tissue concentrationsof TCDD were measured on GD9, GD16, and GD21. On GD9, the highestlevel of TCDD localized in the maternal liver (25.1% dose).In addition, the amount reaching all the embryos on GD9 was0.01% of the administered dose, which resulted in a concentrationof 0.02% dose/g . The amount of TCDD reaching the fetal compartment(fetuses + placentas) increased to 0.12% dose/tissue on GD16and 0.71% by GD21. The concentration of TCDD within the fetalcompartment (0.01% dose/g) on GD16 was comparable to that foundin the maternal blood and spleen. Concentrations of TCDD ina single embryo/fetus were 39.6, 18.1, and 22.1 pg/g on GD9,GD16, and GD21, respectively. Estimates of hepatic half-lifeof elimination in pregnant rats suggested that TCDD may be eliminatedfaster in pregnant LE rats. Therefore, measurements of biliaryelimination were made in pregnant and nonpregnant LE rats tocompare rates of metabolism; however, biliary elimination ofTCDD is not affected by pregnancy. In conclusion, this doseadministered during a critical period of organogenesis causesadverse effects on the developing reproductive system of rodents.This dose produced a body burden of 22.1 pg TCDD/g within asingle fetus on GD21. This indicates that low-level TCDD exposureduring the perinatal stage of life can produce adverse effectswithin the developing pups.  相似文献   

3.
The maintenance of the placental vasculature is essential for sustaining normal fetal growth. On the basis of our previous observation that fetal death was accompanied by placental hypoxia upon exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (R. Ishimura et al., 2002a, Toxicol. Appl. Pharmacol. 185, 197-206), we here investigated the effects of TCDD on the placenta, focusing on the development of the labyrinth zone. Holtzman rats were administered a single oral dose of 1.6 mug of TCDD/kg body weight or an equivalent volume of vehicle (control) on gestation day (GD) 15, and placental tissues were analyzed on GD20. Immunohistochemical staining showed that the exposure to TCDD decreased the size of maternal blood sinusoids and caused the constriction of fetal capillaries in the placenta. In contrast, we found that vascular remodeling occurred in the labyrinth zone of normal rat placenta; that is, the vascular development in the normal labyrinth zone during the late gestation (from GD16 to GD20) showed dilated maternal blood sinusoids and fetal capillaries accompanied by a decrease in thickness and the apoptosis of trophoblasts. The present results demonstrate that this remodeling is suppressed by TCDD, which is further supported by the decreased expression level of Tie2 mRNA, the gene which is associated with vascular remodeling, and also by the decrease in the number of apoptotic trophoblasts in TCDD-exposed rats. The present study provided a new finding on the development of the vasculature in the labyrinth zone during the late gestation under normal conditions and showed the inhibition of vascular remodeling in TCDD-exposed rats.  相似文献   

4.
The effects of in utero and lactational 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the reproductive system of male rat offspring (F1) and the sex ratio of the subsequent generation (F2) were examined. Female Holtzman rats were gavaged with an initial loading dose of 400 ng/kg TCDD prior to mating, followed by weekly maintenance doses of 80 ng/kg during mating, pregnancy, and the lactation period. Maternal exposure to TCDD had no significant effects on fetus/pup (F1) mortality, litter size, or sex ratio on gestation day (GD) 20 or postnatal day (PND) 2. The TCDD concentration in maternal livers and adipose tissue on GD20 was 1.21 and 1.81 ng/kg, respectively, and decreased at weaning to 0.72 in the liver and 0.84 in the adipose tissue. In contrast, the TCDD concentration in pup livers was 1.32 ng/kg on PND2 and increased to 1.80 ng/kg at weaning. Ventral prostate weight of male offspring was significantly decreased by TCDD exposure on PND28 and 120 compared with that of controls. Weight of the testes, cauda epididymides, and seminal vesicle, and sperm number in the cauda epididymis were not changed by TCDD exposure at PND120. TCDD- or vehicle-exposed male offspring were mated with unexposed females. The sex ratio (percentage of male pups) of F2 offspring was significantly reduced in the TCDD-exposed group compared with controls. These results suggest that in utero and lactational TCDD exposures affect the development of male gonads in offspring (F1), leading to changes in the sex ratio of the subsequent generation (F2).  相似文献   

5.
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is teratogenic in mice, inducing cleft palate and hydronephrosis. After exposure in vivo, TCDD specifically alters differentiation of embryonic palatal medial epithelial cells. In this study, the palatal epithelial cell response to TCDD is determined in vitro. C57BL/6N palatal shelves were placed in organ culture on gestation day (GD) 12 in Richter's improved modified Eagle's medium:Ham's F12 medium (1:1) with 1% fetal bovine serum for 3 or 4 days. Medium contained 0.1% dimethylsulfoxide and TCDD at 0, 10(-13), 10(-12), 10(-11), 10(-10), and 10(-9) M, with some doses at 5 x 10(-11), 7.5 x 10(-11), and 5 x 10(-12) M. Epithelial cell responses to TCDD occurred over a narrow range of concentrations, with maximal response at 5 x 10(-11) M. Cytotoxicity was detected at 1 x 10(-10) M. At a stage when control medial cells ceased proliferation and EGF receptors were not detected immunohistochemically. TCDD-exposed medial cells incorporated [3H]thymidine and high levels of epidermal growth factor receptors were detected. TCDD prevented programmed cell death of medial peridermal cells, and induced a shift in the differentiation of medial cells toward an oral-like phenotype. The responses to TCDD observed after exposure in vitro were indistinguishable from previously reported effects observed after exposure in vivo. In the present study, the distribution of TCDD in the fetus after exposure in vivo was examined. The levels of exposure to TCDD are similar for in vitro and in vivo exposure routes. The levels of TCDD in 1 x 10(-11) to 1 x 10(-10) M solutions (3 to 32 pg/ml) were comparable to levels observed in fetal tissues after in vivo exposure on GD 11 to 30 microns/kg [3H]TCDD, where the palatal shelf contained 1.4 to 3.5. pg TCDD, representing 0.0003% of the total dose. In vivo, TCDD was detected in the GD 11 embryo 3 hr postexposure and the TCDD was equally distributed between the embryonic head and body. At 72 hr postexposure, 0.035% of the total dose was in fetal tissues, and 1% of the TCDD in the fetus was found in the palatal shelf. The present study shows that the palatal epithelium responds to TCDD in vitro in a manner comparable to that observed after in vivo exposure, and that the response occurs at a concentration comparable to in vivo levels in the fetus. The availability of an in vitro system will facilitate studies of TCDD toxicity that are difficult or impossible to perform in vivo, such as comparisons of TCDD effects between species, including human tissues.  相似文献   

6.
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent developmental toxicant in rodents, and these effects occur at exposures similar to background human body burdens. A physiologically based pharmacokinetic (PBPK) model can aid in quantitatively describing the relationship between exposure, dose, and response. The aim of this work was the development a PBPK model to describe the relationship between maternal TCDD exposure and fetal TCDD concentrations during critical windows of susceptibility in the rat. This PBPK model is a modification of an eight-compartment model that describes the adult female rat. The modified model reduces the compartments from eight to four maternal compartments (liver, fat, placenta and rest of the body). Activation of the placental compartment and a separate fetal compartment occurs during gestation. The systemic circulation connects the maternal compartments. The physiological and biochemical parameters were obtained from the literature. The model validation used experimental data from acute and subchronic exposures prior to and during gestation. The simulations predict the TCDD tissue concentrations of the maternal compartments within the standard deviation of the experimental data. The model overestimates the fetal concentrations by approximately a factor of two at low subchronic exposures, but does predict the fetal tissue concentrations within the range of the experimental data at the higher exposures. This model may provide a framework for the development of a human PBPK model to estimate fetal TCDD concentrations in human health risk assessments.  相似文献   

7.
Timed-pregnant CD-1 outbred albino Swiss mice received eithermethacrylamlde (MAC; 0, 60, 120, or 180 mg/kg/day) or N,N'-methylenebisacrylamide(BAC; 0, 3, 10, or 30 mg/kg/day) po in distilled water on gestationaldays (GD) 6 through 17. Maternal clinical status was monitoreddaily. At termination (GD 17), confirmed-pregnant females (27–30per group, MAC; 24–25 per group, BAC) were evaluated forclinical status and gestational outcome; live fetuses were examinedfor external, visceral, and skeletal malformations. For MAC,no treatment-related maternal mortality was observed. Maternalbody weight on GD 17, maternal weight gain during treatmentand gestation, and corrected maternal weight gain were reducedat the high dose. Relative maternal food and water intake wasnot adversely affected; neurotoxicity was not observed. Relativematernal liver weight was increased at a 120 mg/kg/day; graviduterine weight was decreased at 180 mg/kg/day. The maternalno-observed adverse effect level (NOAEL) was 60 mg/kg/day. TheNOAEL for developmental toxicity was also 60 mg/kg/day. At 120 mg/kg/day, mean fetal body weight was reduced. At 180 mg/kg/day,increased postimplantation death per litter was observed. Morphologicaldevelopment was not affected. The maternal NOAEL for BAC was10 mg/kg/day. At 30 mg/kg/day, decreased maternal body weighton GD 17, maternal body weight change during treatment and gestation,corrected maternal body weight, and gravid uterine weight wereobserved. Relative maternal liver weight increased at 30 mg/kg/day.The developmental NOAEL was 3 mg/kg/day BAC. Mean fetal bodyweight was reduced at 30 mg/kg/day. At 10 mg/kg/day, an increasedincidence of fetal variations (extra rib) was observed, althoughfetal malformation rate was unaffected. MAC and BAC were notteratogenic to Swiss mice at the doses tested. BAC was morepotent than MAC in causing adverse maternal and developmentaleffects.  相似文献   

8.
The disposition and toxicity of inhaled elemental mercury (Hg0) vapor for pregnant Long-Evans rats, and potential adverse effects on reproductive outcome were investigated. Rats were exposed to 0, 1, 2, 4, or 8 mg Hg0/m(3) for 2 h/day from gestation day (GD) 6 through GD 15. Maternal toxicity occurred primarily in rats exposed to 4 and 8 mg/m(3) and was manifested as a concentration-related decrease in body weight gain and mild nephrotoxicity. Control rats gained about 13% of their initial body weight during the 10-day exposure. Rats exposed to 4 mg/m(3) Hg0 gained about 7% less than controls, and rats exposed to 8 mg/m(3) Hg0 lost about 17% of their initial body weight during the 10-day exposure period. Maternal kidney weights were significantly increased in the 4 and 8 mg/m(3) concentration groups, and urinalysis revealed increased levels of protein and alkaline phosphatase activity in urine of all Hg0-exposed rats. Dams exposed to 8 mg/m(3) were euthanized in moribund condition on postnatal day (PND) 1. There was no histopathological evidence of toxicity in maternal lung, liver, or kidney of exposed rats at GD 6, GD 15, or PND 1. The incidence of resorptions was significantly increased, litter size and PND 1 neonatal body weights were significantly decreased only in the 8-mg/m(3) group. Total Hg concentrations in maternal tissues increased with increasing number of exposure days and concentration. In general, approximately 70% of Hg was eliminated from maternal tissues during the week following the last exposure (GD 15 to PND 1). Elimination of Hg from maternal brain and kidney was slower than in other tissues, possibly due to higher levels of metallothionein. Total Hg concentrations in fetal tissues increased with increasing number of exposure days and concentration, demonstrating that a significant amount of Hg crossed the placenta. One week after the last exposure, significant amounts of Hg were still present in brain, liver, and kidney of PND 1 neonates. Metallothionein levels in neonatal tissues were not significantly increased by exposure to 4 mg/m(3) Hg0. The total amount of Hg in neonatal brain (ng/brain) continued to increase after termination of inhalation exposure, suggesting a redistribution of Hg from the dam to neonatal brain. These data demonstrate that inhaled Hg0 vapor is distributed to all maternal and fetal tissues in a dose-dependent manner. Adverse effects of Hg on developmental outcome occurred only at a concentration that caused maternal toxicity.  相似文献   

9.
The effect of single maternal subcutaneous (s.c.) injection of 0.12 mg/kg diphenyl ditelluride, (PhTe)2, diluted in canola oil at days 6, 10 or 17 of gestation were evaluated in Wistar rats. The reduction of body weight gain was statistically significant at GD9, for the dams that received (PhTe)2, at GD6; at GD13, for the dams that received (PhTe)2, at GD10, and at GD20, for the dams that received (PhTe)2, at GD17, when compared to respective control groups. External and internal fetal soft tissues examination was performed on day 20 of gestation. Single maternal injection at day 10 of gestation resulted in appearance of malformation in fore- and hind-limbs, absent or short tail, subcutaneous blood clots, exophthalmia, hydrocephalus and absence of the cranial bone and cutaneous tissue in fetuses on day 20 of gestation. Besides, (PhTe)2 reduced fetal body and cerebral weight, kidney length, measurements of body dimension and provoked 73% of fetal mortality. Subcutaneous administration of (PhTe)2 on day 17 of gestation was associated with 94% mortality, hydrocephalus and edema. Histological evaluations of fetal brain demonstrated displaced brain tissue with absence of the cranial bone and cutaneous tissue when diphenyl ditelluride was administered in GD10. Histological evaluation of fetal head exposed at GD17 revealed a decrease of the brain volume with consequent dilation of the lateral ventricles and the adjacent tissues were thinner than that of control group tissues. No fetal changes were observed after administration of (PhTe)2 at day 6 of gestation. Thus, (PhTe)2 can be teratogenic to rat fetuses and toxic for dams. The late fetal stages of rat prenatal development appeared uniquely sensitive to organic tellurium exposure.  相似文献   

10.
This study tested whether maternal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) may disrupt the development of neuroendocrine system of their offspring during the perinatal period. TCDD (0.2 or 0.4 μg/kg body weight) was orally administered to pregnant rats from gestation day (GD) 1 to lactation day (LD) 30. Potential effects on neuroendocrine function were evaluated by measuring serum thyroid hormone levels in pregnant rats and their offspring and measuring some biochemical parameters in cerebellum of these offspring on GD 16 and 19, and LD 10, 20, and 30. In both treated groups, a decrease in serum thyroxine (T4), triiodothyronine (T3) and increase in thyrotropin (TSH) levels were noticed during the tested days in dams and offspring, as well as GH levels were decreased in offspring with respect to control group. In cerebellum of control offspring, the levels of monoamines, γ-aminobutyric acid (GABA) and acetylcholinesterase (AchE) were found to be increased from GD 16 to LD 30. The hypothyroid conditions due to both maternal administrations of TCDD produced inhibitory effects on monoamines and AchE, and stimulatory actions on GABA in cerebellum of offspring. These alterations were dose and age dependent. Overall, these results suggest that TCDD may act as neuroendocrine disruptor.  相似文献   

11.
We compared the effects of a single acute dose, or chronic fetal exposure, to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the male reproductive system of the Wistar(Han) rat. Tissue samples were taken from dams on gestation day (GD)16 and GD21, and from offspring on postnatal days (PND)70 and 120. Steady-state concentration of TCDD was demonstrated in the chronic study: body burdens were comparable in both studies. Fetal TCDD concentrations were comparable after acute and chronic exposure, and demonstrate more potent toxicity after chronic versus acute dosing. In maternal liver, cytochrome P450 (CYP)1A1 and CYP1A2 RNA were induced. In fetus, there was induction of both CYP1A1 and CYP1A2 RNA at medium and high doses, but inadequate evidence for induction at low dose in either study. The low level induction of CYP1A1 RNA at low dose in fetus argues against AhR activation in fetus as a mechanism of toxicity of TCDD in causing delay in balanopreputial separation (BPS), and the greater induction of CYP1A1 RNA in PND70 offspring liver from chronically-dosed dams suggests that lactational transfer of TCDD is crucial to this toxicity. These data characterize the maternal and fetal disposition of TCDD, induction of CYP1A1 RNA as a measure of AhR activation, and suggest that lactational transfer of TCDD determines the difference in delay in BPS between the two studies.  相似文献   

12.
8-2 Fluorotelomer alcohol (FTOH) and its metabolites, perfluorooctanoic acid (PFOA) and perfluorononanoic acid (PFNA), are developmental toxicants but metabolism and distribution during pregnancy are not known. To examine this, timed-pregnant mice received a single gavage dose (30 mg 8-2 FTOH/kg body weight) on gestational day (GD) 8. Maternal and neonatal serum and liver as well as fetal and neonatal homogenate extracts were analyzed using gas chromatography coupled with mass spectrometry. During gestation (GD9 to GD18), maternal serum and liver concentrations of PFOA decreased from 789 +/- 41 to 668 +/- 23 ng/ml and from 673 +/- 23 to 587 +/- 55 ng/g, respectively. PFOA was transferred to the developing fetuses as early as 24-h posttreatment with concentrations increasing from 45 +/- 9 ng/g (GD10) to 140 +/- 32 ng/g (GD18), while PFNA was quantifiable only at GD18 (31 +/- 4 ng/g). Post-partum, maternal serum PFOA concentrations decreased from 451 +/- 21 ng/ml postnatal day (PND) 1 to 52 +/- 19 ng/ml (PND15) and PFNA concentrations, although fivefold less, exhibited a similar trend. Immediately after birth, pups were cross-fostered with dams that had been treated during gestation with 8-2 FTOH (T) or vehicle (C) resulting in four treatment groups in which the first letter represents in utero (fetal) exposure and the second represents lactational (neonatal) exposure: C/C, T/C, C/T, T/T. On PND1, neonatal whole-body homogenate concentrations of PFOA from T/T and T/C groups averaged 200 +/- 26 ng/g, decreased to 149 +/- 19 ng/g at PND3 and this decreasing trend was seen in both neonatal liver and serum from PND3 to PND15. Based on detectible amounts of PFOA in neonatal serum in the C/T group on PND3 (57 +/- 11 ng/ml) and on PND15 (58 +/- 3 ng/ml), we suggest that the neonates were exposed through lactation. In conclusion, exposure of neonates to PFOA and PFNA occurs both pre- and postnatally following maternal 8-2 FTOH exposure on GD8.  相似文献   

13.
14.
Naturally mated female New Zealand White (NZW) rabbits (24/group) received formamide (35, 70, or 140 mg/kg/day) or vehicle (1 ml/kg deionized/distilled water) by gavage on gestational days (GD) 6 through 29. The study was conducted using a 2-replicate design. Maternal food consumption (absolute and relative), body weight, and clinical signs were monitored at regular intervals throughout gestation. One and four maternal deaths occurred at the low and high doses, respectively. Abortions or early deliveries were noted in 0, 2, 2, and 8 females in the 0, 35, 70, and 140-mg/kg/day dose groups, respectively. Other clinical signs associated with formamide exposure were minimal: primarily reduced or absent fecal output at the high dose (2-13 animals/day). Also at the high dose, maternal body weight was significantly depressed on GD 21, 24, and 27 (87-90% of the control value); maternal body weight gain was significantly reduced for GD 12 to 15, 18 to 21, and 21 to 24 (treated animals gained less than 1 g, or lost up to 100 g). In addition, maternal body weight gain was reduced at the middle dose for GD 18 to 21. Maternal body weight gain, corrected for gravid uterine weight, was unaffected. Relative maternal food consumption in the high-dose group was 34-59% of control intake from GD 12 through GD 24, but was comparable to controls thereafter. At termination (GD 30), confirmed-pregnant females (9-20 per group) were evaluated for clinical status, liver weights, and gestational outcome; live fetuses were examined for external, visceral, and skeletal malformations and variations. Maternal liver weight (absolute or relative to body weight) was unaffected by treatment, but gravid uterine weight at the high dose was 71% of the control value. A significantly increasing trend was noted for the percent non-live implants per litter. In addition, although not statistically significant from the control group, the values for the percent late fetal deaths per litter and percent non-live implants per litter in the 140-mg/kg/day group were higher than maximum historical values, suggesting an increase in late gestational deaths in the surviving high-dose animals. Formamide decreased the mean number of live fetuses per litter at the high dose to 66% of the control value. Mean fetal body weight per litter for males and the sexes combined was significantly decreased at the high dose; mean female fetal body weight was also decreased, although the difference did not reach statistical significance. There was no effect of treatment on the incidence of external, visceral, or skeletal malformations or variations in animals surviving to scheduled necropsy. In summary, the no-observed-adverse-effect level (NOAEL) for maternal toxicity was 70 mg/kg/day and the lowest-observed-adverse-effect level (LOAEL) was 140 mg/kg/day under the conditions of this study. Similarly, the NOAEL for developmental toxicity was 70 mg/kg/day and the LOAEL was 140 mg/kg/day.  相似文献   

15.
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and related chemicals are potent cardiovascular teratogens in developing piscine and avian species. In the present study we investigated the effects of TCDD on murine cardiovascular development. Pregnant mice (C57Bl6N) were dosed with 1.5-24 microg TCDD/kg on gestation day (GD) 14.5. At GD 17.5, fetal mice exhibited a dose-related decrease in heart-to-body weight ratio that was significantly reduced at a maternal dose as low as 3.0 microg TCDD/kg. In addition, cardiocyte proliferation was reduced in GD 17.5 fetal hearts at the 6.0-microg TCDD/kg maternal dose. To determine if this reduction in cardiac weight was transient, or if it continued after birth, dams treated with control or 6.0 microg TCDD/kg were allowed to deliver, and heart weight of offspring was determined on postnatal days (P) 7 and 21. While no difference was seen on P 7, on P 21 pups from TCDD-treated litters showed an increase in heart-to-body weight ratio and in expression of the cardiac hypertrophy marker atrial natriuretic factor. Additionally, electrocardiograms of P 21 offspring showed that the combination of in utero and lactational TCDD exposure reduced postnatal heart rate but did not alter cardiac responsiveness to isoproterenol stimulation of heart rate. These results demonstrate that the fetal murine heart is a sensitive target of TCDD-induced teratogenicity, resembling many of TCDD-induced effects observed in fish and avian embryos, including reduced cardiocyte proliferation and altered fetal heart size. Furthermore, the combination of in utero and lactational TCDD exposure can induce cardiac hypertrophy and bradycardia postnatally, which could increase the risk of cardiovascular disease development.  相似文献   

16.
A single maternal dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on gestation day (GD) 13 can greatly impair ventral prostate, dorsolateral prostate, anterior prostate, and seminal vesicle development in wild-type C57BL/6 mice. The developmental stages at which these organs are most sensitive to TCDD exposure have now been investigated. Pregnant mice were dosed orally with 5 micro g TCDD/kg or vehicle on GD 13, and their pups were fostered at birth to dams of the same treatment or cross-fostered to dams of the opposite treatment. Additional males had in utero and lactational TCDD exposure following maternal dosing on GD 16. Organ weights and secretory protein, cytokeratin 8, and cyclophilin mRNA expression were determined at 35 days of age. Effects of TCDD on ventral prostate development were due primarily to in utero exposure; the critical window was between GD 13 and birth. Dorsolateral prostate development was inhibited about equally by in utero or lactational exposure, and vulnerability did not begin until GD 16. Anterior prostate development was also affected by both in utero and lactational TCDD exposure, particularly the former. Vulnerability began before GD 16 and continued into postnatal life. Seminal vesicle development was essentially unaffected by in utero or lactational exposure alone but was significantly inhibited by combined exposure, regardless of whether dams were dosed on GD 13 or 16. In summary, the time during which each organ was most vulnerable to TCDD exposure varied from one organ to another. These findings provide insights into the developmental processes that TCDD inhibits in each organ, and demonstrate that TCDD inhibits ventral prostate development before this organ first appears, presumably via effects on the urogenital sinus. The observation that in utero TCDD exposure (alone) inhibited development of each prostate lobe is significant because previous studies have shown that little TCDD is transmitted to mice before birth.  相似文献   

17.
The present study of subchronic low exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) at or near steady-state levels tries to emulate the most probable mode for human exposure, dietary consumption. This study is the first and most intensive pharmacokinetic study to be reported with repeated dosing, multiple times, and multiple doses examining disposition of TCDD-derived radioactivity and CYP1A activities in mice. For time-course relationships, animals were dosed (daily, Monday-Friday) with 0, 1.5, or 150 ng [3H]TCDD/kg for 4, 8, 13, or 17 weeks and also for 13 weeks followed by 4 weeks with no dosing. For dose-response relationships, animals were dosed for 13 weeks (daily, Monday-Friday) with 0, 0.15, 0.45, 1.5, 4.5, 15, 45, 150, or 450 ng [3H]TCDD/kg. Additional animals dosed for 13 weeks (daily, Monday-Friday) with 1.5 or 150 ng [(3)H]TCDD/kg were housed in metabolism cages. Time- and dose-dependencies of TCDD were confirmed in all measured tissues. Liver/fat (L/F) concentration ratios ranged from 0.2-3.4 (low to high dose). Hepatic CYP1A1 enzymatic activity increased (p < 0.05) starting at 0.15 ng/kg/day with L/F of 0.2 and body burden of 2.8 ng TCDD/kg body weight. By examining TCDD exposures at or near steady state, this study reports for the first time and provides direct evidence of low-dose effects on a measured reversible response at body burdens that are within background levels of the general human population. In addition, this study emphasizes cumulative effects of daily dosing and suggests the importance of tissue dosimetry or body burden for a persistent chemical such as TCDD.  相似文献   

18.
This study tested the hypothesis that prenatal ethanol exposure during the last third of gestation, including the brain growth spurt (BGS), in the guinea pig produces neurobehavioural teratogenicity, manifesting as brain growth restriction and hyperactivity. Pregnant guinea pigs (term, about gestational day (GD) 68) received oral administration of ethanol (2 g/kg maternal body weight per day on GD 43 and/or GD 44 and then 4 g/kg maternal body weight per day from GD 45 to GD 62), isocaloric-sucrose/pair-feeding, or water. Maternal blood ethanol concentration (BEC) on GD 57 or 58, at 1 h after the daily dose, was 340+/-76 mg/dl (n=8). Ethanol treatment decreased brain, cerebral cortical, hippocampal, and cerebellar weights at GD 63 (P<0.05), and decreased brain and cerebral cortical weights at postnatal day 10 (P<0.05), with no effect on body weight and no apparent effect on spontaneous locomotor activity. The data demonstrate that, in the guinea pig, prenatal ethanol exposure during the last third of gestation, including the BGS, decreases brain weight that persists into postnatal life, which is associated with growth restriction of the cerebral cortex. However, this prenatal ethanol exposure regimen, including the BGS, does not increase spontaneous locomotor activity in contrast to the persistent hyperactivity that occurs after chronic ethanol exposure throughout gestation.  相似文献   

19.
Exposure to a low dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) produces a variety of toxic manifestations, including fetal death. In order to evaluate the effects of low-dose TCDD on placental function in this study, pregnant Holtzman rats were given a single oral dose of 800 or 1600 ng TCDD/kg body wt or an equivalent volume of vehicle (control) on gestation day (GD) 15 and the results were observed on GD16 and GD20. The number of fetal deaths increased in the animals exposed to TCDD. Although fetal and placental weight did not differ significantly between the control group and the TCDD groups, histological differences from the control rats were clearly observed in the junctional zone (JZ) of the placentas of the TCDD-exposed rats. In the control placenta, glycogen cells occupied the majority of the JZ on GD16, but then decreased in number and almost disappeared by GD20, whereas on GD20 the placenta of the TCDD-exposed rats exhibited a larger area occupied by the glycogen cells and cysts filled with eosinophilic material surrounded by glycogen cells in the JZ than that of the control group. Glycogen assay revealed that the glycogen content of the placentas from the TCDD-exposed rats was higher than in the control rats. Semiquantitative RT-PCR analysis was performed to assess the expression of glucose transporter 1 (GLUT1) and GLUT3, the two major placental glucose transporter isoforms. On GD20 the level of expression of GLUT1 mRNA in the placentas was not different between the control and TCDD groups, whereas the level of expression of GLUT3 mRNA approximately doubled in both the 800 and 1600 ng/kg TCDD groups. GLUT3 mRNA expression was restricted to the labyrinth zone of placenta, where zone-specific expression of mRNA arylhydrocarbon receptor and induction of cytochrome P450 1A1 mRNA by TCDD were observed, and none was detected in the JZ. These results, including the increase of glycogen content and GLUT3 mRNA level in TCDD-exposed placentas, provide the first evidence of alteration of glucose kinetics in the placenta by TCDD.  相似文献   

20.
Previous studies have shown that maternal doses of 1 microg/kg or less of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in late gestation can demasculinize and feminize reproductive behavior in male rats. However, it was not known whether coplanar polychlorinated biphenyls (PCBs) had similar effects, or whether non-reproductive sexually dimorphic behaviors such as saccharin preference behavior were also altered. We determined the effects of TCDD or coplanar PCBs on saccharin consumption and saccharin preference in male and female rats. Sprague-Dawley rats were dosed with 3,3',4, 4'-tetrachlorobiphenyl (PCB 77; 2 or 8 mg/kg/day), 3,3',4,4', 5-pentachlorobiphenyl (PCB 126; 0.25 or 1.0 microg/kg/day), TCDD (0. 025 or 0.10 microg/kg/day), or corn oil vehicle on days 10-16 of gestation. Maternal exposure to TCDD or coplanar PCBs did not change saccharin consumption or saccharin preference in male rats. However, TCDD and coplanar PCB-exposed females showed decreased saccharin consumption and saccharin preference. The results indicate that saccharin consumption is masculinized in female rats exposed to TCDD or coplanar PCBs during perinatal development. This effect could be related to the anti-estrogenic actions of these chemicals.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号