Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Birth Defects Res ; 109(16): 1301-1304, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28755433

ABSTRACT

BACKGROUND: Cervical ribs in rat fetuses have been widely reported to occur in controls as well as in response to various maternal chemical exposures. However, few evaluations of cervical ribs have been reported in rats postnatally. The available literature has indicated that the postnatal incidences of cervical ribs in control rats are no higher than in perinatal fetuses. METHODS: In a developmental toxicity study in rats conducted by the inhalation route, a control group of 44 time-mated female rats was exposed to filtered air only from gestation day (GD) 6 to 20, and divided into two cohorts. For one cohort, fetuses were removed from dams by laparohysterectomy for skeletal evaluation on GD 21. The other cohort of dams was permitted to deliver, and adult offspring were euthanized on postnatal day 65 for a subsequent postnatal skeletal evaluation of cervical ribs. RESULTS: The incidence of cervical ribs (mean percentage of affected fetuses or adults per litter) was observed to increase during postnatal development, from 1.0% on GD 21 to 12.7% on postnatal day 65. Further evaluation is ongoing to determine whether these observations were attributable to the inhalation exposure conditions used in this study. CONCLUSION: These results, while limited to the evaluation of one skeletal alteration in control rats, support the need for additional research into the area of postnatal development of skeletal abnormalities observed in developmental toxicity studies and the relevance of these skeletal observations to human risk assessment. Birth Defects Research 109:1301-1304, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Bone Development/drug effects , Cervical Rib/pathology , Abnormalities, Drug-Induced , Administration, Inhalation , Animals , Animals, Newborn/growth & development , Bone and Bones/metabolism , Dose-Response Relationship, Drug , Female , Fetus/drug effects , Maternal Exposure , Pregnancy , Rats , Reproduction , Teratogens/pharmacology
2.
Toxicology ; 340: 1-9, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26743852

ABSTRACT

Ammonium, 2,3,3,3-tetrafluoro-2-(heptafluoropropoxy)-propanoate has been developed as a processing aid used in the manufacture of fluoropolymers. The absorption, distribution, elimination, and distribution (ADME) and kinetic behavior of this substance has been evaluated in rats, mice, and cynomolgus monkeys by oral and intravenous routes of exposure and studied in both plasma and urine. The test substance is rapidly and completely absorbed in both rats and mice and both in vivo and in vitro experiments indicate that it is not metabolized. The test substance is rapidly eliminated exclusively in the urine in both rats and mice, with rats eliminating it more quickly than mice (approximately 5h elimination half-life in rats, 20 h half-life in mice). Pharmacokinetic analysis in monkeys, rats, and mice indicate rapid, biphasic elimination characterized by a very fast alpha phase and a slower beta phase. The beta phase does not contribute to potential accumulation after multiple dosing in rats or monkeys. Comparative pharmacokinetics in rats, mice, and monkeys indicates that the rat is more similar to the monkey and is therefore a more appropriate rodent model for pharmacokinetics in primates.


Subject(s)
Fluorocarbons/administration & dosage , Fluorocarbons/pharmacokinetics , Propionates/administration & dosage , Propionates/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Female , Fluorocarbons/blood , Fluorocarbons/urine , Gastrointestinal Absorption , Half-Life , Hepatocytes/metabolism , Macaca fascicularis , Male , Metabolic Clearance Rate , Mice, Inbred ICR , Models, Biological , Propionates/blood , Propionates/urine , Rats, Sprague-Dawley , Species Specificity , Tissue Distribution
3.
Free Radic Biol Med ; 80: 171-82, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25091901

ABSTRACT

This review examines oxidative protein folding within the mammalian endoplasmic reticulum (ER) from an enzymological perspective. In protein disulfide isomerase-first (PDI-first) pathways of oxidative protein folding, PDI is the immediate oxidant of reduced client proteins and then addresses disulfide mispairings in a second isomerization phase. In PDI-second pathways the initial oxidation is PDI-independent. Evidence for the rapid reduction of PDI by reduced glutathione is presented in the context of PDI-first pathways. Strategies and challenges are discussed for determination of the concentrations of reduced and oxidized glutathione and of the ratios of PDI(red):PDI(ox). The preponderance of evidence suggests that the mammalian ER is more reducing than first envisaged. The average redox state of major PDI-family members is largely to almost totally reduced. These observations are consistent with model studies showing that oxidative protein folding proceeds most efficiently at a reducing redox poise consistent with a stoichiometric insertion of disulfides into client proteins. After a discussion of the use of natively encoded fluorescent probes to report the glutathione redox poise of the ER, this review concludes with an elaboration of a complementary strategy to discontinuously survey the redox state of as many redox-active disulfides as can be identified by ratiometric LC-MS-MS methods. Consortia of oxidoreductases that are in redox equilibrium can then be identified and compared to the glutathione redox poise of the ER to gain a more detailed understanding of the factors that influence oxidative protein folding within the secretory compartment.


Subject(s)
Disulfides/chemistry , Endoplasmic Reticulum/metabolism , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Protein Disulfide-Isomerases/chemistry , Protozoan Proteins/chemistry , Sulfhydryl Compounds/chemistry , Animals , Disulfides/metabolism , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/ultrastructure , Eukaryotic Cells/cytology , Eukaryotic Cells/enzymology , Glutathione/chemistry , Glutathione/metabolism , Glutathione Disulfide/chemistry , Glutathione Disulfide/metabolism , Humans , Kinetics , Models, Molecular , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Folding , Protozoan Proteins/metabolism , Sulfhydryl Compounds/metabolism , Trypanosoma brucei brucei/cytology , Trypanosoma brucei brucei/enzymology
4.
Free Radic Biol Med ; 69: 129-35, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24468475

ABSTRACT

A sensitive new plate-reader assay has been developed showing that adult mammalian blood serum contains circulating soluble sulfhydryl oxidase activity that can introduce disulfide bonds into reduced proteins with the reduction of oxygen to hydrogen peroxide. The activity was purified 5000-fold to >90% homogeneity from bovine serum and found by mass spectrometry to be consistent with the short isoform of quiescin-sulfhydryl oxidase 1 (QSOX1). This FAD-dependent enzyme is present at comparable activity levels in fetal and adult commercial bovine sera. Thus cell culture media that are routinely supplemented with either fetal or adult bovine sera will contain this facile catalyst of protein thiol oxidation. QSOX1 is present at approximately 25 nM in pooled normal adult human serum. Examination of the unusual kinetics of QSOX1 toward cysteine and glutathione at low micromolar concentrations suggests that circulating QSOX1 is unlikely to significantly contribute to the oxidation of these monothiols in plasma. However, the ability of QSOX1 to rapidly oxidize conformationally mobile protein thiols suggests a possible contribution to the redox status of exofacial and soluble proteins in blood plasma. Recent proteomic studies showing that plasma QSOX1 can be utilized in the diagnosis of pancreatic cancer and acute decompensated heart failure, together with the overexpression of this secreted enzyme in a number of solid tumors, suggest that the robust QSOX assay developed here may be useful in the quantitation of enzyme levels in a wide range of biological fluids.


Subject(s)
Disulfides/blood , Oxidoreductases Acting on Sulfur Group Donors/isolation & purification , Protein Isoforms/isolation & purification , Amino Acid Motifs , Amino Acid Sequence , Animals , Catalysis , Cattle , Humans , Kinetics , Oxidoreductases Acting on Sulfur Group Donors/blood , Oxidoreductases Acting on Sulfur Group Donors/chemistry , Protein Conformation , Protein Isoforms/blood , Protein Isoforms/chemistry , Proteomics , Rats
5.
Biochemistry ; 52(46): 8323-32, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24147449

ABSTRACT

Augmenter of liver regeneration is a member of the ERV family of small flavin-dependent sulfhydryl oxidases that contain a redox-active CxxC disulfide bond in redox communication with the isoalloxazine ring of bound FAD. These enzymes catalyze the oxidation of thiol substrates with the reduction of molecular oxygen to hydrogen peroxide. This work studies the catalytic mechanism of the short, cytokine form of augmenter of liver regeneration (sfALR) using model thiol substrates of the enzyme. The redox potential of the proximal disulfide in sfALR was found to be approximately 57 mV more reducing than the flavin chromophore, in agreement with titration experiments. Rapid reaction studies show that dithiothreitol (DTT) generates a transient mixed disulfide intermediate with sfALR signaled by a weak charge-transfer interaction between the thiolate of C145 and the oxidized flavin. The subsequent transfer of reducing equivalents to the flavin ring is relatively slow, with a limiting apparent rate constant of 12.4 s(-1). However, reoxidation of the reduced flavin by molecular oxygen is even slower (2.3 s(-1) at air saturation) and thus largely limits turnover at 5 mM DTT. The nature of the charge-transfer complexes observed with DTT was explored using a range of simple monothiols to mimic the initial nucleophilic attack on the proximal disulfide. While ß-mercaptoethanol is a very poor substrate of sfALR (∼0.3 min(-1) at 100 mM thiol), it rapidly generates a mixed disulfide intermediate allowing the thiolate of C145 to form a strong charge-transfer complex with the flavin. Unlike the other monothiols tested, glutathione is unable to form charge-transfer complexes and is an undetectable substrate of the oxidase. These data are rationalized on the basis of the stringent steric requirements for thiol-disulfide exchange reactions. The inability of the relatively bulky glutathione to attain the in-line geometry required for efficient disulfide exchange in sfALR may be physiologically important in preventing the oxidase from catalyzing the potentially harmful oxidation of intracellular glutathione.


Subject(s)
Cytochrome Reductases/metabolism , Oxidoreductases/metabolism , Catalysis , Cytochrome Reductases/genetics , Disulfides/chemistry , Flavins/chemistry , Flavins/metabolism , Humans , Mercaptoethanol/metabolism , Models, Molecular , Oxidation-Reduction , Oxidoreductases/genetics , Oxidoreductases Acting on Sulfur Group Donors , Oxygen/chemistry
6.
PLoS One ; 6(3): e18187, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21479176

ABSTRACT

Under the shell of a chicken egg are two opposed proteinaceous disulfide-rich membranes. They are fabricated in the avian oviduct using fibers formed from proteins that are extensively coupled by irreversible lysine-derived crosslinks. The intractability of these eggshell membranes (ESM) has slowed their characterization and their protein composition remains uncertain. In this work, reductive alkylation of ESM followed by proteolytic digestion led to the identification of a cysteine rich ESM protein (abbreviated CREMP) that was similar to spore coat protein SP75 from cellular slime molds. Analysis of the cysteine repeats in partial sequences of CREMP reveals runs of remarkably repetitive patterns. Module a contains a C-X(4)-C-X(5)-C-X(8)-C-X(6) pattern (where X represents intervening non-cysteine residues). These inter-cysteine amino acid residues are also strikingly conserved. The evolutionarily-related module b has the same cysteine spacing as a, but has 11 amino acid residues at its C-terminus. Different stretches of CREMP sequences in chicken genomic DNA fragments show diverse repeat patterns: e.g. all a modules; an alternation of a-b modules; or an a-b-b arrangement. Comparable CREMP proteins are found in contigs of the zebra finch (Taeniopygia guttata) and in the oviparous green anole lizard (Anolis carolinensis). In all these cases the long runs of highly conserved modular repeats have evidently led to difficulties in the assembly of full length DNA sequences. Hence the number, and the amino acid lengths, of CREMP proteins are currently unknown. A 118 amino acid fragment (representing an a-b-a-b pattern) from a chicken oviduct EST library expressed in Escherichia coli is a well folded, highly anisotropic, protein with a large chemical shift dispersion in 2D solution NMR spectra. Structure is completely lost on reduction of the 8 disulfide bonds of this protein fragment. Finally, solid state NMR spectra suggest a surprising degree of order in intact ESM fibers.


Subject(s)
Avian Proteins/metabolism , Chickens/metabolism , Disulfides/metabolism , Egg Shell/metabolism , Membrane Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Avian Proteins/chemistry , Avian Proteins/isolation & purification , Conserved Sequence/genetics , Cysteine/metabolism , Dictyosteliida/metabolism , Egg Shell/cytology , Evolution, Molecular , Finches , Lizards , Magnetic Resonance Spectroscopy , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Membranes , Microscopy, Fluorescence , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Substrate Specificity
7.
Drug Metab Dispos ; 39(7): 1288-93, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21493824

ABSTRACT

In vitro metabolism of 1,2,3,3,3-pentafluoropropene (PFP) was investigated in the present study. PFP was metabolized via cytochrome P450-catalyzed oxidative dehalogenation in liver microsomes and glutathione transferase (GST)-catalyzed conjugation in liver microsomes and cytosol. Two oxidation products, 2,3,3,3-tetrafluoropropionaldehyde (TPA) and 3,3,3-trifluoropyruvaldehyde (TFPA), and two GSH conjugates, S-(2,3,3,3-tetrafluoropropenyl)-GSH (TFPG) and S-(1,2,3,3,3-pentafluoropropyl)-GSH (PFPG) were identified. Enzyme kinetic parameters for the formation of TFPA, TFPG, and PFPG were obtained in male and female rat, mouse, dog, and human liver microsomes and cytosol and were confirmed using freshly isolated male rat hepatocytes. For the TFPA pathway, dog microsomes exhibited much larger K(m) values than rat, mouse, and human microsomes. Sex differences in the rates of metabolism within a given species were minor and generally were less than 2-fold. Across the species, liver microsomes were the primary subcellular fraction for GSH S-conjugation and the apparent reaction rates for the formation of TFPG were much greater than those for PFPG in liver microsomes. PFPG was unstable and had a half-life of approximately 3.9 h in a phosphate buffer (pH 7.4 and 37°C). The intrinsic clearance values for the formation of TFPA were much greater than those for the formation of GSH S-conjugates, suggesting that cytochrome P450-mediated oxidation is the primary pathway for the metabolism of PFP at relatively low PFP concentrations. Because saturation of the GST-mediated reactions was not reached at the highest possible PFP concentration, GSH S-conjugation may become a much more important pathway at higher PFP concentrations (relative to the K(m) for TFPA).


Subject(s)
Cytosol/metabolism , Fluorocarbons/metabolism , Glutathione/metabolism , Hepatocytes/metabolism , Microsomes, Liver/metabolism , Animals , Dogs , Female , Humans , Kinetics , Magnetic Resonance Spectroscopy , Male , Mice , Oxidation-Reduction , Rats
8.
Toxicology ; 283(1): 55-62, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21349313

ABSTRACT

The absorption, tissue distribution, elimination, and metabolism of [1-¹4C]-PFHx in rats and mice dosed orally at 2 or 100 mg/kg was evaluated following a single dose or after 14 consecutive doses. Absorption was rapid in rats as evidenced by a short time to maximum concentration (C(max)) of 30 min in male rats and 15 min in female rats at both the 2 and 100mg/kg dose level. The plasma elimination half-life was somewhat longer in males (1.5-1.7 h) than in females (0.5-0.7 h). Absorption in the mouse was also rapid with the maximum plasma concentration occurring between 15 and 30 min after dosing. The maximum concentration was not appreciably different between male and female mice (8 µg equiv./g at 2 mg/kg; ~350 µg equiv./g at 100 mg/kg). The primary route of elimination was via the urine. PFHx was not metabolized in rat or mouse hepatocytes, nor were any metabolites observed after oral dosing in either rodent species. Essentially 100% of the dose was eliminated in urine within 24 h demonstrating that PFHx is readily absorbed and bioavailability approaches 100%, even at a dose as high as 100 mg/kg. The route and extent of elimination was unchanged after 14 days of daily dosing. Tissues were collected at three time points (rat: 0.5, 2, and 24 h; mice: 0.25, 1, and 24 h) after dosing to investigate the tissue clearance kinetics of PFHx following a single dose at 2 or 100 mg/kg. In all tissues except skin, PFHx was not quantifiable 24 h after dosing in both sexes of the two species.


Subject(s)
Caproates/pharmacokinetics , Fluorocarbons/pharmacokinetics , Animals , Area Under Curve , Caproates/blood , Caproates/urine , Carbon Radioisotopes , Female , Fluorocarbons/blood , Fluorocarbons/urine , Guinea Pigs , Half-Life , Hepatocytes/metabolism , Intestinal Absorption , Male , Mice , Rats , Rats, Sprague-Dawley , Tissue Distribution
9.
Inhal Toxicol ; 21(6): 552-82, 2009 May.
Article in English | MEDLINE | ID: mdl-19519155

ABSTRACT

Methyl iodide (MeI) has been proposed as an alternative to methyl bromide as a pre-plant soil fumigant that does not deplete stratospheric ozone. In inhalation toxicity studies performed in animals as part of the registration process, three effects have been identified that warrant consideration in developing toxicity reference values for human risk assessment: nasal lesions (rat), acute neurotoxicity (rat), and fetal loss (rabbit). Uncertainties in the risk assessment can be reduced by using an internal measure of target tissue dose that is linked to the likely mode of action (MOA) for the toxicity of MeI, rather than the external exposure concentration. Physiologically based pharmacokinetic (PBPK) models have been developed for MeI and used to reduce uncertainties in the risk assessment extrapolations (e.g. interspecies, high to low dose, exposure scenario). PBPK model-derived human equivalent concentrations comparable to the animal study NOAELs (no observed adverse effect levels) for the endpoints of interest were developed for a 1-day, 24-hr exposure of bystanders or 8 hr/day exposure of workers. Variability analyses of the PBPK models support application of uncertainty factors (UF) of approximately 2 for intrahuman pharmacokinetic variability for the nasal effects and acute neurotoxicity.


Subject(s)
Hydrocarbons, Iodinated/pharmacokinetics , Models, Biological , Adipose Tissue/metabolism , Adult , Aged , Animals , Brain/metabolism , Child , Female , Humans , Infant , Kidney/metabolism , Male , Nasal Mucosa/metabolism , Pregnancy , Rabbits , Rats , Rats, Sprague-Dawley , Respiratory Physiological Phenomena
10.
Food Chem Toxicol ; 46(6): 2023-34, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18329151

ABSTRACT

N-acetyl-l-aspartic acid (NAA) is a constituent of the mammalian central nervous system (CNS) that has been identified in a number of commonly consumed foods. The current study reports the outcome of acute and repeated dose oral toxicology studies conducted with NAA in Sprague-Dawley (SD) rats. No mortalities or evidence of adverse effects were observed in SD rats following acute oral administration of 2000mg/kg NAA. In a separate study, NAA was added to the diets of SD rats (n=10/sex group) at concentrations corresponding to daily doses of 10, 100, or 1000mg/kg/day for 14 consecutive days and 100, 500, and 1000mg/kg/day for another 14 days. All rats survived until scheduled sacrifice and no differences in body weights, feed consumption values, or clinical signs were observed in any of the treatment groups. No biologically significant differences were observed in functional observational battery (FOB), motor activity evaluations, ophthalmologic examinations, hematology, coagulation, clinical chemistry, or organ weights of any of the NAA treatment groups. Further, no test substance-related gross or microscopic changes were observed in NAA exposure groups. Based on these results, NAA was not considered acutely toxic following oral exposure to 2000mg/kg and the no-observed-adverse-effect-level (NOAEL) for systemic toxicity from repeated dose dietary exposure to NAA is 1000mg/kg/day.


Subject(s)
Aspartic Acid/analogs & derivatives , Administration, Oral , Animals , Aspartic Acid/analysis , Aspartic Acid/toxicity , Behavior, Animal/drug effects , Blood Cell Count , Blood Chemical Analysis , Blood Coagulation/drug effects , Body Weight/drug effects , Diet , Eating/drug effects , Eye/pathology , Female , Food Analysis , Male , Motor Activity/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley
11.
Drug Chem Toxicol ; 31(2): 189-216, 2008.
Article in English | MEDLINE | ID: mdl-18330782

ABSTRACT

8-2 fluorotelomer alcohol is a fluorinated chemical intermediate used to manufacture specialty polymers and surfactants. The potential subchronic toxicity and the reversibility of the effects of this chemical were evaluated following approximately 90 days of oral gavage dosing to Crl:CD(SD)IGS BR rats. A complete toxicological profile, including neurobehavioral assessments and hepatic beta-oxidation, were conducted at selected intervals and a group of rats was included for a 90-day postdosing recovery period. Dose levels tested were 0 (control), 1, 5, 25, and 125 mg/kg. No test-substance-related mortality occurred at any dose level. Rats at 125 mg/kg developed striated teeth, such that these animals were switched to ground chow at 77 days. No treatment-related alterations in body weight, food consumption, neurobehavioral parameters, or hematology/clinical chemistry were found. Hepatic beta-oxidation was increased in males at 125 mg/kg and in females at 25 and 125 mg/kg. In both males and females, plasma fluorine levels were increased at 125 mg/kg and urinary fluorine was elevated at > or =5 mg/kg. Degeneration/disorganization of enamel organ ameloblast cells was observed at 125 mg/kg in males, but not females. Liver weight increases accompanied by focal hepatic necrosis were observed at both 25 and 125 mg/kg, and chronic progressive nephrotoxicity occurred in female rats at 125 mg/kg. With the exception of hepatocellular necrosis in males at 125 mg/kg and the increased incidence and severity of chronic progressive nephropathy in females at 125 mg/kg, all other changes showed evidence of reversibility. The no-observed-adverse-effect level was 5 mg/kg.


Subject(s)
Behavior, Animal/drug effects , Hydrocarbons, Fluorinated/toxicity , Liver/drug effects , Administration, Oral , Ameloblasts/drug effects , Ameloblasts/metabolism , Animals , Dose-Response Relationship, Drug , Female , Fluorine/blood , Fluorocarbons , Hydrocarbons, Fluorinated/administration & dosage , Kidney/drug effects , Kidney/pathology , Liver/metabolism , Liver/pathology , Male , Necrosis/chemically induced , No-Observed-Adverse-Effect Level , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Sex Factors , Time Factors , Toxicity Tests, Chronic
12.
Drug Metab Dispos ; 34(6): 1019-29, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16540590

ABSTRACT

Geranyl nitrile (GN) and citronellyl nitrile (CN) are fragrance components used in consumer and personal care products. Differences in the clastogenicity of these two terpenes are postulated to result from differential biotransformation, presumably involving the conjugated nitrile moiety. The metabolic clearance and biotransformation of GN and CN were compared in primary hepatocytes from mice, rats, and humans. For determination of intrinsic clearance, GN and CN were incubated with hepatocytes in sealed vials, and the headspace was sampled periodically by solid-phase microextraction and analyzed by gas chromatography/mass spectrometry. For metabolite identification, GN and CN were incubated with hepatocytes from each species for 60 min, and reaction mixtures were extracted and analyzed by mass spectroscopy. Both GN and CN were rapidly metabolized in hepatocytes from all species (T1/2, 0.7-11.6 min). Within a species, intrinsic clearance was similar for both compounds and increased in the order human < rat << mouse. Major common pathways for biotransformation of GN and CN involved 1) epoxidation of the 6-alkenyl moiety followed by conjugation with glutathione, 2) hydroxylation of the terminal methyl group(s) followed by direct conjugation with glucuronic acid in rodents or further oxidation to the corresponding acid in human cells, and 3) hydroxylation of the allylic C5 position. No evidence for either phase I or phase II metabolism of the conjugated nitrile moiety was obtained. Thus, the presumed metabolic basis for differences in genotoxicity remains elusive.


Subject(s)
Hepatocytes/metabolism , Monoterpenes/metabolism , Mutagens/metabolism , Nitriles/metabolism , Animals , Biotransformation , Chromatography, Gas , Cosmetics , Glucuronides/metabolism , Humans , In Vitro Techniques , Kinetics , Male , Mass Spectrometry , Mice , Perfume , Rats , Rats, Sprague-Dawley
13.
Toxicol Sci ; 91(2): 341-55, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16543293

ABSTRACT

The absorption, distribution, metabolism, and elimination of [3-14C] 8-2 fluorotelomer alcohol (8-2 FTOH, C7F1514CF2CH2CH2OH) following a single oral dose at 5 and 125 mg/kg in male and female rats have been determined. Following oral dosing, the maximum concentration of 8-2 FTOH in plasma occurred by 1 h postdose and cleared rapidly with a half-life of less than 5 h. The internal dose to 8-2 FTOH, as measured by area under the concentration-time curve to infinity, was similar for male and female rats and was observed to increase in a dose-dependent fashion. The majority of the 14C 8-2 FTOH (> 70%) was excreted in feces, and 37-55% was identified as parent. Less than 4% of the administered dose was excreted in urine, which contained low concentrations of perfluorooctanoate (approximately 1% of total 14C). Metabolites identified in bile were principally composed of glucuronide and glutathione conjugates, and perfluorohexanoate was identified in excreta and plasma, demonstrating the metabolism of the parent FTOH by sequential removal of multiple CF2 groups. At 7 days postdose, 4-7% of the administered radioactivity was present in tissues, and for the majority, 14C concentrations were greater than whole blood with the highest concentration in fat, liver, thyroid, and adrenals. Distribution and excretion of a single 125-mg/kg [3-14C] 8-2 FTOH dermal dose following a 6-h exposure in rats was also determined. The majority of the dermal dose either volatilized from the skin (37%) or was removed by washing (29%). Following a 6-h dermal exposure and a 7-day collection period, excretion of total radioactivity via urine (< 0.1%) and feces (< 0.2%) was minor, and radioactivity concentrations in most tissues were below the limit of detection. Systemic availability of 8-2 FTOH following dermal exposure was negligible.


Subject(s)
Fatty Alcohols/pharmacokinetics , Absorption , Administration, Cutaneous , Administration, Oral , Animals , Bile/chemistry , Cells, Cultured , Fatty Alcohols/administration & dosage , Fatty Alcohols/blood , Fatty Alcohols/urine , Feces/chemistry , Female , Hepatocytes/metabolism , Male , Metabolic Clearance Rate , Rats , Rats, Inbred Strains , Tissue Distribution
14.
Toxicology ; 211(1-2): 139-48, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15863257

ABSTRACT

This study was conducted to develop a quantitative understanding of the potential for gestational and lactational transfer of perfluorooctanoate (PFOA) in the rat. Time-mated female rats were dosed by oral gavage once daily at concentrations of 3, 10, or 30 mg/kg/day of the ammonium salt of PFOA (APFO) starting on gestation (G) day 4 and continuing until sacrifice. On days 10, 15, and 21G, five rats per dose level were sacrificed and blood samples were collected 2h post-dose. Embryos were collected on day 10G, amniotic fluid, placentas, and embryos/fetuses were collected on days 15 and 21G, and fetal blood samples were collected on day 21G. Five rats per dose level were allowed to deliver and nurse their litters, and on days 3, 7, 14, and 21 post-partum (PP) milk and blood samples of maternal and pup were collected 2h post-dose. All samples were analyzed by high-performance liquid chromatography-mass spectrometry (HPLC-MS) for PFOA concentration. Concentrations of PFOA in maternal plasma and milk attained steady state during the sampling interval. The steady-state concentrations in maternal plasma were 10-15, 25-30, and 60-75 microg/mL in rats receiving 3, 10, and 30 mg/kg, respectively. Steady-state concentrations in milk were approximately 10 times less than those in maternal plasma. The concentration of PFOA in fetal plasma on day 21G was approximately half the steady-state concentration in maternal plasma. The milk concentrations appeared to be generally comparable to the concentrations in pup plasma. Pup plasma concentrations decreased from day 3PP to day 7PP, and were similar on days 7, 14, and 21PP at all dose levels. PFOA was detected in placenta (days 15 and 21G), amniotic fluid (days 15 and 21G), embryo (days 10 and 15G), and fetus (day 21G). These pharmacokinetics allow estimation of the dose to developing and nursing rat offspring following maternal exposure.


Subject(s)
Caprylates/pharmacokinetics , Fluorocarbons/pharmacokinetics , Lactation/physiology , Placenta/metabolism , Animals , Biotransformation , Body Weight/drug effects , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Female , Fetus/metabolism , Litter Size/drug effects , Mass Spectrometry , Maternal-Fetal Exchange , Milk/metabolism , Pregnancy , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...