Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Health Perspect ; 115(9): 1298-305, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17805419

RESUMO

BACKGROUND: The presence of perfluorooctanesulfonate (PFOS), perfluorohexanesulfonate (PFHS), and perfluorooctanoate (PFOA) has been reported in humans and wildlife. Pharmacokinetic differences have been observed in laboratory animals. OBJECTIVE: The purpose of this observational study was to estimate the elimination half-life of PFOS, PFHS, and PFOA from human serum. METHODS: Twenty-six (24 male, 2 female) retired fluorochemical production workers, with no additional occupational exposure, had periodic blood samples collected over 5 years, with serum stored in plastic vials at -80 degrees C. At the end of the study, we used HPLC-mass spectrometry to analyze the samples, with quantification based on the ion ratios for PFOS and PFHS and the internal standard (18)O(2)-PFOS. For PFOA, quantitation was based on the internal standard (13)C(2)-PFOA. RESULTS: THE ARITHMETIC MEAN INITIAL SERUM CONCENTRATIONS WERE AS FOLLOWS: PFOS, 799 ng/mL (range, 145-3,490); PFHS, 290 ng/mL (range, 16-1,295); and PFOA, 691 ng/mL (range, 72-5,100). For each of the 26 subjects, the elimination appeared linear on a semi-log plot of concentration versus time; therefore, we used a first-order model for estimation. The arithmetic and geometric mean half-lives of serum elimination, respectively, were 5.4 years [95% confidence interval (CI), 3.9-6.9] and 4.8 years (95% CI, 4.0-5.8) for PFOS; 8.5 years (95% CI, 6.4-10.6) and 7.3 years (95% CI, 5.8-9.2) for PFHS; and 3.8 years (95% CI, 3.1-4.4) and 3.5 years (95% CI, 3.0-4.1) for PFOA. CONCLUSIONS: Based on these data, humans appear to have a long half-life of serum elimination of PFOS, PFHS, and PFOA. Differences in species-specific pharmacokinetics may be due, in part, to a saturable renal resorption process.


Assuntos
Ácidos Alcanossulfônicos/sangue , Caprilatos/sangue , Fluorocarbonos/sangue , Exposição Ocupacional , Ácidos Sulfônicos/sangue , Idoso , Indústria Química , Feminino , Meia-Vida , Humanos , Masculino , Pessoa de Meia-Idade , Aposentadoria
2.
Drug Metab Dispos ; 34(8): 1406-10, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16720684

RESUMO

N-Alkylperfluorooctanesulfonamides have been used in a range of industrial and commercial applications. Perfluorooctanesulfonamide (FOSA) is a major metabolite of N-alkylperfluorooctanesulfonamides and has a long half-life in animals and in the environment and is biotransformed to FOSA N-glucuronide. The objective of this study was to identify and characterize the human and experimental animal liver UDP-glucuronosyltransferases (UGTs) that catalyze the N-glucuronidation of FOSA. The results showed that pooled human liver and rat liver microsomes had high N-glucuronidation activities. Expressed rat UGT1.1, UGT2B1, and UGT2B12 in HK293 cells catalyzed the N-glucuronidation of FOSA but at rates that were lower than those observed in rat liver microsomes. Of the 10 expressed human UGTs (1A1, 1A3, 1A4, 1A6, 1A9, 2B4, 2B7, 2B15, and 2B17) studied, only hUGT2B4 and hUGT2B7 catalyzed the N-glucuronidation of FOSA. The kinetics of N-glucuronidation of FOSA by rat liver microsomes and by hUGT2B4/7 was consistent with a single-enzyme Michaelis-Menten model, whereas human liver microsomes showed sigmoidal kinetics. These data show that rat liver UGT1.1, UGT2B1, and UGT2B12 catalyze the N-glucuronidation of FOSA, albeit at low rates, and that hUGT2B4 and hUGT2B7 catalyze the N-glucuronidation of FOSA.


Assuntos
Fluorocarbonos/metabolismo , Glucuronídeos/metabolismo , Glucuronosiltransferase/metabolismo , Sulfonamidas/metabolismo , Animais , Cães , Humanos , Técnicas In Vitro , Cinética , Macaca mulatta , Masculino , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Ratos
3.
Crit Rev Toxicol ; 34(4): 351-84, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15328768

RESUMO

PFOA is a peroxisome proliferator (PPAR agonist) and exerts morphological and biochemical effects characteristic of PPAR agonists. These effects include increased beta-oxidation of fatty acids, increases in several cytochrome P-450 (CYP450)-mediated reactions, and inhibition of the secretion of very low-density lipoproteins and cholesterol from the liver. These effects on lipid metabolism and transport result in a reduction of cholesterol and triglycerides in serum and an accumulation of lipids in the liver. The triad of tumors observed (liver, Leydig cell, and pancreatic acinar-cell) is typical of many PPAR agonists and is believed to involve nongenotoxic mechanisms. The hepatocellular tumors observed in rats are likely to have been the result of the activation of the peroxisome proliferator activated receptor alpha (PPARalpha). The tumors observed in the testis (Leydig-cell) have been hypothesized to be associated with an increased level of serum estradiol in concert with testicular growth factors. The mechanism responsible for the acinar-cell tumors of the pancreas in rats remains the subject of active investigation. The mechanism resulting in the hepatocellular tumors in rats (PPARalpha activation) is not likely to be relevant to humans. Similarly, the proposed mechanism for Leydig-cell tumor formation is of questionable relevance to humans. Acinar tumors of the pancreas are rare in humans, and the relevance of the these tumors, as found in rats, to humans is uncertain. Epidemiological investigations and medical surveillance of occupationally exposed workers have not found consistent associations between PFOA exposure and adverse health effects.


Assuntos
Caprilatos/toxicidade , Fluorocarbonos/toxicidade , Tumor de Células de Leydig/induzido quimicamente , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Pancreáticas/induzido quimicamente , Proliferadores de Peroxissomos/agonistas , Neoplasias Testiculares/induzido quimicamente , Animais , Caprilatos/sangue , Caprilatos/farmacocinética , Feminino , Fluorocarbonos/sangue , Fluorocarbonos/farmacocinética , Humanos , Masculino , Testes de Mutagenicidade , Ratos , Pele/efeitos dos fármacos , Distribuição Tecidual
4.
Chem Res Toxicol ; 17(6): 767-75, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15206897

RESUMO

Perfluorooctanesulfonic acid (PFOS) and its derivatives have been used in a range of industrial and commercial applications, including the manufacture of surfactants, adhesives, anticorrosion agents, and insecticides. PFOS is found at detectable concentrations in human and wildlife tissues and in the global environment. N-Substituted perfluorooctanesulfonamides are believed to be degraded to PFOS and, therefore, contribute to the accumulation of PFOS in the environment. N-Ethyl-N-(2-hydroxyethyl)perfluorooctanesulfonamide (N-EtFOSE) is converted to PFOS in experimental animals. The objective of this study was to elucidate the pathways for the biotransformation of N-EtFOSE, which is a major precursor and component of PFOS-based compounds. N-EtFOSE and several putative metabolites were incubated with liver microsomes and cytosol and with liver slices from male Sprague-Dawley rats. Microsomal fractions fortified with NADPH catalyzed the N-deethylation of N-EtFOSE to give N-(2-hydroxyethyl)perfluorooctanesulfonamide (FOSE alcohol) and of FOSE alcohol to give perfluorooctanesulfonamide (FOSA). These N-dealkylation reactions were catalyzed mainly by male rat P450 2C11 and P450 3A2 and by human P450 2C19 and 3A4/5. Rat liver microsomal fractions incubated with UDP-glucuronic acid catalyzed the O-glucuronidation of N-EtFOSE and FOSE alcohol and the N-glucuronidation of FOSA. Cytosolic fractions incubated with NAD(+) catalyzed the oxidation of FOSE alcohol to perfluooctanesulfonamidoacetate (FOSAA). The oxidation of N-EtFOSE to N-ethylperfluorooctanesulfonamidoacetate (N-EtFOSAA) was observed in liver slices but not in cytosolic fractions. FOSA was biotransformed in liver slices to PFOS, albeit at a low rate. These results show that the major pathway for the biotransformation of N-EtFOSE is N-dealkylation to give FOSA. The biotransformation of FOSA to PFOS explains the observation that PFOS is found in animals given N-EtFOSE.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Fluorocarbonos/metabolismo , Fígado/enzimologia , Microssomos Hepáticos/enzimologia , Sulfonamidas/metabolismo , Animais , Biotransformação , Cromatografia Líquida , Citosol/enzimologia , Humanos , Masculino , Espectrometria de Massas , Ratos , Ratos Sprague-Dawley , Frações Subcelulares
5.
Toxicol Sci ; 80(1): 151-60, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15071170

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors that activate target genes involved in lipid metabolism, energy homeostasis, and cell differentiation in response to diverse compounds, including environmental chemicals. The liver-expressed receptor PPARalpha mediates peroxisome proliferative responses associated with rodent hepatocarcinogenesis. Previous studies have established that certain perfluorooctanesulfonamide-based chemicals (PFOSAs) alter lipid metabolism, are hepatic peroxisome proliferators, and induce hepatocellular adenoma formation in rodents, suggesting that they activate PPARalpha. The present study investigates this question and characterizes the activation of mouse and human PPARalpha by PFOSAs. Perfluorooctanesulfonate (PFOS), an end-stage metabolite common to several PFOSAs, was found to activate both mouse and human PPARalpha in a COS-1 cell-based luciferase reporter trans-activation assay. Half-maximal activation (EC50) occurred at 13-15 microM PFOS, with no significant difference in the responsiveness of mouse and human PPARalpha. Mouse and human PPARalpha were activated by perfluorooctanesulfonamide (FOSA) over a similar concentration range; however, cellular toxicity precluded an accurate determination of EC50 values. Studies of 2-N-ethylperfluorooctanesulfonamido ethanol were less informative due to its insolubility. These findings were verified in an FAO rat hepatoma cell line that stably expresses PPARalpha, where the endogenous PPARalpha target genes peroxisomal bifunctional enzyme and peroxisomal 3-ketoacyl-CoA thiolase were activated up to approximately 10-20-fold by PFOS and FOSA. The interactions of PPARalpha with PFOS and FOSA, and the potential of these chemicals for activation of unique sets of downstream target genes, may help explain the diverse biological effects exhibited by PFOSAs and may aid in the evaluation of human and environmental risks associated with exposure to this important class of fluorochemicals.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Fluorocarbonos/toxicidade , PPAR alfa/biossíntese , Proliferadores de Peroxissomos/farmacologia , Sulfonamidas/toxicidade , Ácidos Alcanossulfônicos/química , Animais , Western Blotting , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Fluorocarbonos/química , Regulação da Expressão Gênica , Genes Reporter , Humanos , Hidrocarbonetos Fluorados , Luciferases/genética , Camundongos , PPAR alfa/genética , Proliferadores de Peroxissomos/química , Reação em Cadeia da Polimerase , RNA Mensageiro/biossíntese , Solubilidade , Sulfonamidas/química , Ativação Transcricional
6.
Toxicology ; 183(1-3): 117-31, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12504346

RESUMO

Perfluorooctanesulfonate (PFOS) is a widely disseminated persistent compound found at low (part-per-billion) concentrations in serum and liver samples from humans and fish-eating wildlife. This study investigated the hypotheses that early hepatocellular peroxisomal proliferation and hepatic cellular proliferation are factors in chronic liver response to dietary dosing, that lowering of serum total cholesterol is an early clinical measure of response to treatment, and that liver and serum PFOS concentrations are proportional to dose and cumulative dose after sub-chronic treatment. PFOS was administered in diet as the potassium salt at 0, 0.5, 2.0, 5.0, and 20 parts per million (ppm) to Sprague Dawley rats for 4 or 14 weeks. At 4 weeks, effects included decreased serum glucose and an equivocal (

Assuntos
Ácidos Alcanossulfônicos/toxicidade , Fluorocarbonos/toxicidade , Alanina Transaminase/sangue , Ácidos Alcanossulfônicos/administração & dosagem , Animais , Análise Química do Sangue , Peso Corporal , Colesterol/sangue , Relação Dose-Resposta a Droga , Feminino , Fluorocarbonos/administração & dosagem , Testes Hematológicos , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/metabolismo , Fígado/patologia , Masculino , Nível de Efeito Adverso não Observado , Tamanho do Órgão , Oxirredutases/biossíntese , Oxirredutases/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Ratos Sprague-Dawley , Urinálise
7.
Toxicology ; 176(3): 175-85, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12093614

RESUMO

Liver-fatty acid binding protein (L-FABP) is an abundant intracellular lipid-carrier protein. The hypothesis that perfluorooctanesulfonate (PFOS), perfluorooctanoate (PFOA), and certain related perfluorooctanesulfonamide-based fluorochemicals (PFOSAs) can interfere with the binding affinity of L-FABP for fatty acids was tested. The relative effectiveness of PFOA, PFOS, N-ethylperfluorooctanesulfonamide (N-EtFOSA), N-ethylperfluorooctanesulfonamido ethanol (N-EtFOSE), and of the strong peroxisome proliferator Wyeth-14643 (WY) to inhibit 11-(5-dimethylaminonapthalenesulphonyl)-undecanoic acid (DAUDA) binding to-L-FABP was determined. The dissociation constant (Kd) of the DAUDA-L-FABP complex was 0.47 nM. PFOS exhibited the highest level of inhibition of DAUDA-L-FABP binding in the competitive binding assays, followed by N-EtFOSA, WY, and, with equal IC(50)s, N-EtFOSE and PFOA. The in vitro data presented in this study support the hypothesis that these fluorochemicals may interfere with the binding of fatty acids or other endogenous ligands to L-FABP. Furthermore, this work provides evidence to support the hypothesis that displacement of endogenous ligands from L-FABP may contribute to toxicity in rodents fed these fluorochemicals.


Assuntos
Proteínas de Transporte/química , Fluorocarbonos/química , Fígado/química , Proteínas de Neoplasias , Proteínas do Tecido Nervoso , Algoritmos , Animais , Ligação Competitiva/efeitos dos fármacos , Eletroforese em Gel de Poliacrilamida , Proteína 7 de Ligação a Ácidos Graxos , Proteínas de Ligação a Ácido Graxo , Ácidos Graxos/metabolismo , Corantes Fluorescentes , Masculino , Proliferadores de Peroxissomos/farmacologia , Ligação Proteica/efeitos dos fármacos , Pirimidinas/farmacologia , Ratos , Espectrometria de Fluorescência , Espectrometria de Massas por Ionização por Electrospray
8.
Toxicol Sci ; 68(1): 249-64, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12075127

RESUMO

This study was conducted to determine the earliest measurable response of primates to low-level perfluorooctanesulfonate (PFOS) exposure and to provide information to reduce uncertainty in human health risk assessment. Groups of male and female monkeys received 0, 0.03, 0.15, or 0.75 mg/kg/day potassium PFOS orally for 182 days. Recovery animals from each group, except the 0.03 mg/kg/day dose group, were monitored for one year after treatment. Significant adverse effects occurred only in the 0.75 mg/kg/day dose group and included compound-related mortality in 2 of 6 male monkeys, decreased body weights, increased liver weights, lowered serum total cholesterol, lowered triiodothyronine concentrations (without evidence of hypothyroidism), and lowered estradiol levels. Decreased serum total cholesterol occurred in the 0.75 mg/kg/day dose group at serum PFOS levels > 100 ppm. Hepatocellular hypertrophy and lipid vacuolation were present at term in the 0.75 mg/kg/day dose group. No peroxisomal (palmitoyl CoA oxidase) or cell proliferation (proliferating cell nuclear antigen immunohistochemistry) was detected. Complete reversal of clinical and hepatic effects and significant decreases in serum and liver PFOS occurred within 211 days posttreatment. Liver-to-serum PFOS ratios were comparable in all dose groups, with a range of 1:1 to 2:1. Serum concentrations associated with no adverse effects (0.15 mg/kg/day) were 82.6 +/- 25.2 ppm for males and 66.8 +/- 10.8 ppm for females. Comparison of serum PFOS concentrations associated with no adverse effect in this study to those reported in human blood samples (0.028 +/- 0.014 ppm) indicated an adequate margin of safety.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Fluorocarbonos/toxicidade , Macaca fascicularis , Testes de Toxicidade/métodos , Administração Oral , Ácidos Alcanossulfônicos/administração & dosagem , Ácidos Alcanossulfônicos/farmacocinética , Animais , Peso Corporal/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Fluorocarbonos/administração & dosagem , Fluorocarbonos/farmacocinética , Imuno-Histoquímica , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Longevidade/efeitos dos fármacos , Masculino , Tamanho do Órgão/efeitos dos fármacos , Peroxissomos/efeitos dos fármacos , Peroxissomos/enzimologia , Peroxissomos/ultraestrutura , Antígeno Nuclear de Célula em Proliferação/metabolismo , Indução de Remissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...