Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Toxicol Environ Health A ; 76(20): 1151-67, 2013.
Article in English | MEDLINE | ID: mdl-24279816

ABSTRACT

Studies incorporating both toxicokinetic and dynamic factors provide insight into chemical sensitivity differences across the life span. Tissue (brain, plasma, liver) levels of the N-methyl carbamate carbaryl, and its metabolite 1-naphthol, were determined and related to brain and RBC cholinesterase (ChE) inhibition in the same animals. Dose-response (3, 7.5, 15, or 22.5 mg/kg, 40-45 min postdosing) and time course (3 or 15 mg/kg at 30, 60, 120, or 240 min postdosing) of acute effects of carbaryl (oral gavage) in preweanling (postnatal day [PND] 18) and adult male Brown Norway rats from adolescence to senescence (1, 4, 12, 24 mo) were compared. At all ages there were dose-related increases in carbaryl and 1-naphthol in the dose-response study, and the time-course study showed highest carbaryl levels at 30 min postdosing. There were, however, age-related differences in that the 1- and 4-mo rats showed the lowest levels of carbaryl and 1-naphthol, and PND18 and 24-mo rats had similar, higher levels. The fastest clearance (shortest half-lives) was observed in 1- and 4-mo rats. Carbaryl levels were generally higher than 1-naphthol in brain and plasma, but in liver, 1-naphthol levels were similar to or greater than carbaryl. Brain ChE inhibition closely tracked brain carbaryl concentrations regardless of the time after dosing, but there was more variability in the relationship between RBC ChE and plasma carbaryl levels. Within-subject analyses suggested somewhat more brain ChE inhibition at lower carbaryl levels only in the PND18 rats. These findings may reflect maturation followed by decline in kinetic factors over the life span.


Subject(s)
Aging/metabolism , Brain/enzymology , Carbaryl/metabolism , Carbaryl/toxicity , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/toxicity , Naphthols/metabolism , Administration, Oral , Age Factors , Aging/blood , Animals , Carbaryl/blood , Cholinesterase Inhibitors/blood , Cholinesterases/drug effects , Cholinesterases/metabolism , Dose-Response Relationship, Drug , Environmental Monitoring , Half-Life , Liver/chemistry , Liver/metabolism , Male , Naphthols/blood , Plasma/chemistry , Rats , Tissue Distribution
2.
Toxicol Sci ; 122(1): 134-45, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21482639

ABSTRACT

Perfluorooctanoic acid (PFOA) is an environmental contaminant that causes adverse developmental effects in laboratory animals. To investigate the low-dose effects of PFOA on offspring, timed-pregnant CD-1 mice were gavage dosed with PFOA for all or half of gestation. In the full-gestation study, mice were administered 0, 0.3, 1.0, and 3.0 mg PFOA/kg body weight (BW)/day from gestation days (GD) 1-17. In the late-gestation study, mice were administered 0, 0.01, 0.1, and 1.0 mg PFOA/kg BW/day from GD 10-17. Exposure to PFOA significantly (p < 0.05) increased offspring relative liver weights in all treatment groups in the full-gestation study and in the 1.0 mg PFOA/kg group in the late-gestation study. In both studies, the offspring of all PFOA-treated dams exhibited significantly stunted mammary epithelial growth as assessed by developmental scoring. At postnatal day 21, mammary glands from the 1.0 mg/kg GD 10-17 group had significantly less longitudinal epithelial growth and fewer terminal end buds compared with controls (p < 0.05). Evaluation of internal dosimetry in offspring revealed that PFOA concentrations remained elevated in liver and serum for up to 6 weeks and that brain concentrations were low and undetectable after 4 weeks. These data indicate that PFOA-induced effects on mammary tissue (1) occur at lower doses than effects on liver weight in CD-1 mice, an observation that may be strain specific, and (2) persist until 12 weeks of age following full-gestational exposure. Due to the low-dose sensitivity of mammary glands to PFOA in CD-1 mice, a no observable adverse effect level for mammary developmental delays was not identified in these studies.


Subject(s)
Caprylates/toxicity , Fluorocarbons/toxicity , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/chemically induced , Animals , Animals, Newborn , Body Weight/drug effects , Caprylates/blood , Dose-Response Relationship, Drug , Female , Fluorocarbons/blood , Gestational Age , Liver/drug effects , Male , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Mice , Mice, Inbred Strains , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Pregnancy , Sensitivity and Specificity
3.
Toxicology ; 281(1-3): 48-55, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21237237

ABSTRACT

Perfluorononanoic acid (PFNA) is a fluorinated organic chemical found at low levels in the environment, but is detectable in humans and wildlife. The present study compared the pharmacokinetic properties of PFNA in two laboratory rodent species. Male and female Sprague-Dawley rats were given a single dose of PFNA by oral gavage at 1, 3, or 10mg/kg, and blood was collected from the tail vein at 1, 2, 3, 4, 7, 16, 21, 28, 35, 42 and 50 days after treatment. In addition, livers and kidneys were collected for PFNA analysis at the terminal time point. CD-1 mice were given a single oral dose of PFNA of 1 or 10mg/kg, and 4 males and 4 females were killed at similar time intervals; trunk blood, liver and kidney were collected. Serum and tissue concentrations of PFNA were determined by LC-MS/MS. Serum elimination of PFNA is by and large linear with exposure doses in the rat; however, like PFOA, a major sex difference in the rate of elimination is observed, with an estimated half-life of 30.6 days for males and 1.4 days for females. PFNA is stored preferentially in the liver but not in the kidneys. In the mouse, the rates of PFNA serum elimination are non-linear with exposure dose and are slightly faster in females than males, with terminal estimated serum half-life of 25.8-68.4 days and 34.3-68.9 days, respectively. PFNA is also stored preferentially in the mouse liver but not in the kidneys. Hepatic uptake appears to be more efficient and storage capacity greater in male mice than in females. These data suggest that (1) PFNA is more persistent in the mouse than in the rat; (2) there is a major sex difference in the serum elimination of PFNA in the rat, but much less so in the mouse; and (3) there is a significantly higher hepatic accumulation of PFNA in male mice than in females.


Subject(s)
Fatty Acids/pharmacokinetics , Hydrocarbons, Fluorinated/pharmacokinetics , Administration, Oral , Animals , Fatty Acids/analysis , Fatty Acids/blood , Female , Fluorocarbons , Gas Chromatography-Mass Spectrometry , Half-Life , Hydrocarbons, Fluorinated/analysis , Hydrocarbons, Fluorinated/blood , Kidney/chemistry , Liver/chemistry , Male , Mice , Rats , Rats, Sprague-Dawley , Sex Factors
4.
PPAR Res ; 20102010.
Article in English | MEDLINE | ID: mdl-20936102

ABSTRACT

Perfluorononanoic acid (PFNA) is one of the perfluoroalkyl acids found in the environment and in tissues of humans and wildlife. Prenatal exposure to PFNA negatively impacts survival and development of mice and activates the mouse and human peroxisome proliferator-activated receptor-alpha (PPARα). In the current study, we used PPARα knockout (KO) and 129S1/SvlmJ wild-type (WT) mice to investigate the role of PPARα in mediating PFNA-induced in vivo effects. Pregnant KO and WT mice were dosed orally with water (vehicle control: 10 ml/kg), 0.83, 1.1, 1.5, or 2 mg/kg PFNA on gestational days (GDs) 1-18 (day of sperm plug = GD 0). Maternal weight gain, implantation, litter size, and pup weight at birth were unaffected in either strain. PFNA exposure reduced the number of live pups at birth and survival of offspring to weaning in the 1.1 and 2 mg/kg groups in WT. Eye opening was delayed (mean delay 2.1 days) and pup weight at weaning was reduced in WT pups at 2 mg/kg. These developmental endpoints were not affected in the KO. Relative liver weight was increased in a dose-dependent manner in dams and pups of the WT strain at all dose levels but only slightly increased in the highest dose group in the KO strain. In summary, PFNA altered liver weight of dams and pups, pup survival, body weight, and development in the WT, while only inducing a slight increase in relative liver weight of dams and pups at 2 mg/kg in KO mice. These results suggest that PPARα is an essential mediator of PFNA-induced developmental toxicity in the mouse.

5.
PPAR Res ; 20102010.
Article in English | MEDLINE | ID: mdl-20936131

ABSTRACT

Perfluorooctane sulfonate (PFOS) is a perfluoroalkyl acid (PFAA) and a persistent environmental contaminant found in the tissues of humans and wildlife. Although blood levels of PFOS have begun to decline, health concerns remain because of the long half-life of PFOS in humans. Like other PFAAs, such as, perfluorooctanoic acid (PFOA), PFOS is an activator of peroxisome proliferator-activated receptor-alpha (PPARα) and exhibits hepatocarcinogenic potential in rodents. PFOS is also a developmental toxicant in rodents where, unlike PFOA, its mode of action is independent of PPARα. Wild-type (WT) and PPARα-null (Null) mice were dosed with 0, 3, or 10 mg/kg/day PFOS for 7 days. Animals were euthanized, livers weighed, and liver samples collected for histology and preparation of total RNA. Gene profiling was conducted using Affymetrix 430_2 microarrays. In WT mice, PFOS induced changes that were characteristic of PPARα transactivation including regulation of genes associated with lipid metabolism, peroxisome biogenesis, proteasome activation, and inflammation. PPARα-independent changes were indicated in both WT and Null mice by altered expression of genes related to lipid metabolism, inflammation, and xenobiotic metabolism. Such results are similar to studies done with PFOA and are consistent with modest activation of the constitutive androstane receptor (CAR), and possibly PPARγ and/or PPARß/δ. Unique treatment-related effects were also found in Null mice including altered expression of genes associated with ribosome biogenesis, oxidative phosphorylation, and cholesterol biosynthesis. Of interest was up-regulation of Cyp7a1, a gene which is under the control of various transcription regulators. Hence, in addition to its ability to modestly activate PPARα, PFOS induces a variety of PPARα-independent effects as well.

6.
Reprod Toxicol ; 27(3-4): 278-288, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19429403

ABSTRACT

Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are environmental contaminants found in the tissues of humans and wildlife. They are activators of peroxisome proliferator-activated receptor-alpha (PPAR alpha) and exhibit hepatocarcinogenic potential in rats. PFOS and PFOA are also developmental toxicants in rodents and PFOS has been shown to induce pulmonary deficits in rat offspring. Pregnant CD-1 mice were dosed with 0, 5, or 10mg/kg PFOS from gestation days 1-17. Transcript profiling was conducted on the fetal liver and lung. Results were contrasted to data derived from a previous PFOA study. PFOS-dependent changes were primarily related to activation of PPAR alpha. No remarkable differences were found between PFOS and PFOA. Given that PPAR alpha signaling is required for neonatal mortality in PFOA-treated mice but not those exposed to PFOS, the neonatal mortality observed for PFOS may reflect functional deficits related to the physical properties of the chemical rather than to transcript alterations.


Subject(s)
Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Fluorocarbons/toxicity , Gene Expression Profiling , Liver/metabolism , Lung/metabolism , Alkanesulfonic Acids/pharmacology , Animals , Caprylates/pharmacology , Dose-Response Relationship, Drug , Female , Fetus/metabolism , Fluorocarbons/pharmacology , Maternal Exposure , Mice , Mice, Inbred Strains , Microarray Analysis , Pregnancy
7.
Reprod Toxicol ; 27(3-4): 258-265, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18595657

ABSTRACT

Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are members of a family of perfluorinated compounds. Both are environmentally persistent and found in the serum of wildlife and humans. PFOS and PFOA are developmentally toxic in laboratory rodents. Exposure to these chemicals in utero delays development and reduces postnatal survival and growth. Exposure to PFOS on the last 4 days of gestation in the rat is sufficient to reduce neonatal survival. PFOS and PFOA are weak agonists of peroxisome proliferator activated receptor-alpha (PPAR alpha). The reduced postnatal survival of neonatal mice exposed to PFOA was recently shown to depend on expression of PPAR alpha. This study used PPAR alpha knockout (KO) and 129S1/SvlmJ wild type (WT) mice to determine if PPAR alpha expression is required for the developmental toxicity of PFOS. After mating overnight, the next day was designated gestation day (GD) 0. WT females were weighed and dosed orally from GD15 to 18 with 0.5% Tween-20, 4.5, 6.5, 8.5, or 10.5mg PFOS/kg/day. KO females were dosed with 0.5% Tween-20, 8.5 or 10.5mg PFOS/kg/day. Dams and pups were observed daily and pups were weighed on postnatal day (PND) 1 and PND15. Eye opening was recorded from PND12 to 15. Dams and pups were killed on PND15, body and liver weights recorded, and serum collected. PFOS did not affect maternal weight gain or body or liver weights of the dams on PND15. Neonatal survival (PND1-15) was significantly reduced by PFOS in both WT and KO litters at all doses. WT and KO pup birth weight and weight gain from PND1 to 15 were not significantly affected by PFOS exposure. Relative liver weight of WT and KO pups was significantly increased by the 10.5mg/kg dose. Eye opening of PFOS-exposed pups was slightly delayed in WT and KO on PND13 or 14, respectively. Because results in WT and KO were comparable, it is concluded that PFOS-induced neonatal lethality and delayed eye opening are not dependent on activation of PPAR alpha.


Subject(s)
Alkanesulfonic Acids/toxicity , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , PPAR alpha/metabolism , Alkanesulfonic Acids/agonists , Animals , Birth Weight , Dose-Response Relationship, Drug , Environmental Pollutants/agonists , Eye/drug effects , Eye/growth & development , Female , Fluorocarbons/agonists , Liver/drug effects , Liver/growth & development , Male , Maternal Exposure , Mice , Mice, Inbred Strains , Mice, Knockout , Organ Size/drug effects , Sex Factors , Survival Analysis
8.
Toxicol Sci ; 105(1): 173-81, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18511431

ABSTRACT

Perfluorobutyrate (PFBA) is a perfluoroalkyl acid (PFAA) found in the environment. Previous studies have indicated developmental toxicity of PFAAs (perfluorooctane sulfonate [PFOS] and perfluorooctanoate [PFOA]); the current study examines that of PFBA. PFBA/NH4(+) was given to timed-pregnant CD-1 mice by oral gavage daily from gestational day (GD) 1 to 17 at 35, 175, or 350 mg/kg (chosen to approximate the developmentally toxic doses of PFOA); controls received water. At GD 18, serum levels of PFBA were 3.8, 4.4, and 2.5 microg/ml, respectively, in the three treated groups. PFBA did not significantly affect maternal weight gain, number of implantations, fetal viability, fetus weight, or incidence of fetal malformations. Incidence of full-litter loss was significantly greater in the 350 mg/kg group, and maternal liver weights were significantly increased in the 175 and 350 mg/kg groups. In contrast to PFOA and PFOS, PFBA exposure during pregnancy did not adversely affect neonatal survival or postnatal growth. Liver enlargement was detected in the PFBA-exposed pups on postnatal day (PD) 1, but not by PD 10. Expression of selected hepatic genes in PFBA-exposed pups at PD 1 did not reveal any significant changes from controls. A significant delay in eye-opening in offspring was detected in all three PFBA groups, and slight delays in the onset of puberty were noted in the 175 and 350 mg/kg groups. These data suggest that exposure to PFBA during pregnancy in the mouse did not produce developmental toxicity comparable to that observed with PFOA, in part, due to rapid elimination of the chemical.


Subject(s)
Fetus/drug effects , Fluorocarbons/toxicity , Abnormalities, Drug-Induced , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Female , Fluorocarbons/blood , Gene Expression/drug effects , Growth/drug effects , Liver/drug effects , Liver/metabolism , Mice , Organ Size/drug effects , Pregnancy
9.
Toxicol Pathol ; 36(4): 592-607, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18467677

ABSTRACT

Health concerns have been raised because perfluorooctanoic acid (PFOA) is commonly found in the environment and can be detected in humans. In rodents, PFOA is a carcinogen and a developmental toxicant. PFOA is a peroxisome proliferator-activated receptor alpha (PPARalpha) activator; however, PFOA is capable of inducing heptomegaly in the PPARalpha-null mouse. To study the mechanism associated with PFOA toxicity, wild-type and PPARalpha-null mice were orally dosed for 7 days with PFOA (1 or 3 mg/kg) or the PPARalpha agonist Wy14,643 (50 mg/kg). Gene expression was evaluated using commercial microarrays. In wild-type mice, PFOA and Wy14,643 induced changes consistent with activation of PPARalpha. PFOA-treated wild-type mice deviated from Wy14,643-exposed mice with respect to genes involved in xenobiotic metabolism. In PFOA-treated null mice, changes were observed in transcripts related to fatty acid metabolism, inflammation, xenobiotic metabolism, and cell cycle regulation. Hence, a component of the PFOA response was found to be independent of PPARalpha. Although the signaling pathways responsible for these effects are not readily apparent, overlapping gene regulation by additional PPAR isoforms could account for changes related to fatty acid metabolism and inflammation, whereas regulation of xenobiotic metabolizing genes is suggestive of constitutive androstane receptor activation.


Subject(s)
Caprylates/toxicity , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Gene Expression Profiling , Gene Expression/drug effects , Liver/drug effects , PPAR alpha/metabolism , Animals , Caprylates/pharmacokinetics , Environmental Pollutants/pharmacokinetics , Fluorocarbons/pharmacokinetics , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , PPAR alpha/genetics , Pyrimidines/toxicity , Reverse Transcriptase Polymerase Chain Reaction
10.
Toxicol Pathol ; 36(4): 632-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18467680

ABSTRACT

Perfluorooctanoic acid (PFOA) is a chemical used in the production of fluoropolymers. Its persistence in the environment and presence in humans and wildlife has raised health concerns. Liver tumor induction by PFOA is thought to be mediated in rodents by PPAR-alpha. A recent US EPA scientific advisory board questioned the contribution of PPAR-alpha in PFOA-induced liver tumors. Liver response in CD-1, SV/129 wild-type (WT), and PPAR-alpha knockout (KO) SV/129 mice was evaluated after seven daily treatments of PFOA-NH4(+) (1, 3, or 10 mg/kg, p.o.) or the prototype PPARalpha-agonist Wyeth 14,643 (WY, 50 mg/kg). Livers were examined by light and electron microscopy. Proliferation was quantified after PCNA immunostaining. PFOA treatment induced a dose-dependent increase in hepatocyte hypertrophy and labeling index (LI) similar to WY in WT mice. Ultrastructural alterations of peroxisome proliferation were similar between WY-treated and 10 mg/kg PFOA-treated WT mice. KO mice had a dose-dependent increase in hepatocyte vacuolation but increased LI only at 10 mg PFOA/kg. WY-treated KO mice were not different from KO control. These data suggest that PPAR-alpha is required for WY- and PFOA-induced cellular alterations in WT mouse liver. Hepatic enlargement observed in KO mice may be due to an accumulation of cytoplasmic vacuoles that contain PFOA.


Subject(s)
Caprylates/toxicity , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Liver/drug effects , PPAR alpha/physiology , Pyrimidines/toxicity , Animals , Caprylates/blood , Caprylates/pharmacokinetics , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Environmental Pollutants/blood , Environmental Pollutants/pharmacokinetics , Fluorocarbons/blood , Fluorocarbons/pharmacokinetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Liver/metabolism , Liver/ultrastructure , Mice , Mice, Knockout , Organ Size/drug effects , PPAR alpha/agonists , PPAR alpha/genetics , Pyrimidines/blood , Pyrimidines/pharmacokinetics
11.
Toxicology ; 239(1-2): 15-33, 2007 Sep 24.
Article in English | MEDLINE | ID: mdl-17681415

ABSTRACT

Perfluorooctanoic acid (PFOA) is a stable perfluoroalkyl acid used to synthesize fluoropolymers during the manufacture of a wide variety of products. Concerns have been raised over the potential health effects of PFOA because it is persistent in the environment and can be detected in blood and other tissues of many animal species, including humans. PFOA has also been shown to induce growth deficits and mortality in murine neonates. To better understand the mechanism of PFOA induced developmental toxicity, lung and liver gene expression profiling was conducted in PFOA-exposed full-term mouse fetuses. Thirty timed-pregnant CD-1 mice were orally dosed from gestation days 1-17 with either 0, 1, 3, 5, or 10mg/(kgday) PFOA in water. At term, fetal lung and liver were collected, total RNA prepared, and samples pooled from three fetuses per litter. Five biological replicates consisting of individual litter samples were then evaluated for each treatment group using Affymetrix mouse 430_2 microarrays. The expression of genes related to fatty acid catabolism was altered in both the fetal liver and lung. In the fetal liver, the effects of PFOA were robust and also included genes associated with lipid transport, ketogenesis, glucose metabolism, lipoprotein metabolism, cholesterol biosynthesis, steroid metabolism, bile acid biosynthesis, phospholipid metabolism, retinol metabolism, proteosome activation, and inflammation. These changes are consistent with transactivation of PPARalpha, although, with regard to bile acid biosynthesis and glucose metabolism, non-PPARalpha related effects were suggested as well. Additional studies will be needed to more thoroughly address the role of PPARalpha, and other nuclear receptors, in PFOA mediated developmental toxicity.


Subject(s)
Caprylates/toxicity , Environmental Pollutants/toxicity , Fetal Development/drug effects , Fluorocarbons/toxicity , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Liver/drug effects , Lung/drug effects , Administration, Oral , Animals , Dose-Response Relationship, Drug , Fatty Acids/metabolism , Female , Fetal Development/physiology , Liver/embryology , Liver/metabolism , Lung/embryology , Lung/metabolism , Maternal Exposure , Mice , Mice, Inbred Strains , Oligonucleotide Array Sequence Analysis , Pregnancy , RNA, Messenger/metabolism
12.
Toxicol Sci ; 98(2): 571-81, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17488742

ABSTRACT

Perfluorooctanoic acid (PFOA) is a member of a family of perfluorinated chemicals that have a variety of applications. PFOA persists in the environment and is found in wildlife and humans. In mice, PFOA is developmentally toxic producing mortality, delayed eye opening, growth deficits, and altered pubertal maturation. PFOA activates peroxisome proliferators-activated receptor-alpha (PPARalpha), a pathway critical to the mode of induction of liver tumors in rodents. The present study uses 129S1/SvlmJ wild-type (WT) and PPARalpha knockout (KO) mice to determine if PPARalpha mediates PFOA-induced developmental toxicity. Pregnant mice were dosed orally from gestation days 1-17 with water or 0.1, 0.3, 0.6, 1, 3, 5, 10, or 20 mg PFOA/kg. PFOA did not affect maternal weight, embryonic implantation, number, or weight of pups at birth. At 5 mg/kg, the incidence of full litter resorptions increased in both WT and KO mice. In WT, but not KO, neonatal survival was reduced (0.6 mg/kg) and eye opening was delayed (1 mg/kg). There was a trend across dose for reduced pup weight (WT and KO) on several postnatal days (PND), but only WT exposed to 1 mg/kg were significantly different from control (PND7-10 and 22). Maternal factors (e.g., background genetics) did not contribute to differences in postnatal mortality, as PFOA induced postnatal mortality in heterozygous pups born to WT or KO dams. In conclusion, early pregnancy loss was independent of PPARalpha expression. Delayed eye opening and deficits in postnatal weight gain appeared to depend on PPARalpha expression, although other mechanisms may contribute. PPARalpha was required for PFOA-induced postnatal lethality and expression of one copy of the gene was sufficient to mediate this effect.


Subject(s)
Caprylates/toxicity , Embryo Loss/chemically induced , Fluorocarbons/toxicity , PPAR alpha/genetics , Animals , Caprylates/blood , Caprylates/pharmacokinetics , Embryo Loss/blood , Embryo Loss/genetics , Eye/drug effects , Eye/growth & development , Female , Fluorocarbons/blood , Fluorocarbons/pharmacokinetics , Liver/drug effects , Liver/growth & development , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Organ Size/drug effects , PPAR alpha/deficiency , Pregnancy
13.
Toxicol Sci ; 96(1): 133-44, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17132714

ABSTRACT

Perfluorooctanoic acid (PFOA), with diverse and widespread commercial and industrial applications, has been detected in human and wildlife sera. Previous mouse studies linked prenatal PFOA exposure to decreased neonatal body weights (BWs) and survival in a dose-dependent manner. To determine whether effects were linked to gestational time of exposure or to subsequent lactational changes, timed-pregnant CD-1 mice were orally dosed with 5 mg PFOA/kg on gestation days (GD) 1-17, 8-17, 12-17, or vehicle on GD 1-17. PFOA exposure had no effect on maternal weight gain or number of live pups born. Mean pup BWs on postnatal day (PND) 1 in all PFOA-exposed groups were significantly reduced and decrements persisted until weaning. Mammary glands from lactating dams and female pups on PND 10 and 20 were scored based on differentiation or developmental stages. A significant reduction in mammary differentiation among dams exposed GD 1-17 or 8-17 was evident on PND 10. On PND 20, delays in normal epithelial involution and alterations in milk protein gene expression were observed. All exposed female pups displayed stunted mammary epithelial branching and growth at PND 10 and 20. While control litters at PND 10 and 20 had average scores of 3.1 and 3.3, respectively, all treated litters had scores of 1.7 or less, with no progression of duct epithelial growth evident over time. BW was an insignificant covariate for these effects. These findings suggest that in addition to gestational exposure, abnormal lactational development of dams may play a role in early growth retardation of developmentally exposed offspring.


Subject(s)
Caprylates/toxicity , Cell Differentiation/drug effects , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Mammary Glands, Animal/drug effects , Prenatal Exposure Delayed Effects , Administration, Oral , Animals , Animals, Newborn , Body Weight/drug effects , Caprylates/administration & dosage , Caprylates/blood , Caprylates/metabolism , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/blood , Environmental Pollutants/metabolism , Female , Fluorocarbons/administration & dosage , Fluorocarbons/blood , Fluorocarbons/metabolism , Gene Expression Regulation/drug effects , Gestational Age , Liver/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Maternal Exposure , Mice , Milk Proteins/metabolism , Pregnancy , RNA, Messenger , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...