Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Theriogenology ; 84(2): 208-16, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25892340

RESUMO

The influence of the central circadian clock on reproductive timing is well established. Much less is known about the role of peripheral oscillators such as those in the ovary. We investigated the influence of photoperiod and timing of the LH surge on expression of circadian clock genes and genes involved in steroidogenesis in ovine ovarian stroma. Seventy-two Suffolk cross ewes were divided into two groups, and their estrous cycles were synchronized. Progestagen sponge removal was staggered by 12 hours between the groups such that expected LH peak would occur midway through either the light or dark phase of the photoperiodic cycle. Four animals from each group were killed, and their ovaries were harvested beginning 36 hours after sponge removal, at 6-hour intervals for 48 hours. Blood was sampled every 3 hours for the period 24 to 48 hours after sponge removal to detect the LH surge. The interval to peak LH did not differ between the groups (36.2 ± 1.2 and 35.6 ± 1.1 hours, respectively). There was an interaction between group and the time of sponge removal on the expression of the core clock genes ARNTL, PER1, CRY1, CLOCK, and DBP (P < 0.01, P < 0.05, P < 0.01, P < 0.01, and P < 0.01, respectively). As no significant interaction between group and time of day was detected, the datasets were combined. Statistically significant rhythmic oscillation was observed for ARNTL, CLOCK, CRY1 (P < 0.01, respectively), PTGS2, DBP, PTGER2, and CYP17A1 (P < 0.05, respectively), confirming the existence of a time-sensitive functionality within the ovary, which may influence steroidogenesis and is independent of the ovulatory cycle.


Assuntos
Ritmo Circadiano/fisiologia , Ovário/fisiologia , Fotoperíodo , Ovinos/fisiologia , Animais , Ritmo Circadiano/genética , Feminino , Expressão Gênica , Hormônio Luteinizante/sangue , Ovulação/fisiologia , Radioimunoensaio/veterinária , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Fatores de Tempo
2.
Food Chem Toxicol ; 58: 506-21, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23707562

RESUMO

Estimates of internal dosimetry for acrylamide (AA, 2-propenamide; CASRN: 79-06-1) and its active metabolite glycidamide (GA) were compared using either biomarkers of internal exposure (hemoglobin adduct levels in rats and humans) or a PBTK model (Sweeney et al., 2010). The resulting impact on the human equivalent dose (HED, oral exposures), the human equivalent concentration (HEC, inhalation), and final reference values was also evaluated. Both approaches yielded similar AA HEDs and HECs for the most sensitive noncancer effect of neurotoxicity, identical oral reference doses (RfD) of 2×10(-3) mg AA/kg bw/d, and nearly identical inhalation reference concentrations (RfC=0.006 mg/m(3) and 0.007 mg/m(3), biomarker and PBTK results, respectively). HED and HEC values for carcinogenic potential were very similar, resulting in identical inhalation unit risks of 0.1/(mg AA/m(3)), and nearly identical oral cancer slope factors (0.4 and 0.5/mg AA/kg bw/d), biomarker and PBTK results, respectively. The concordance in estimated HEDs, HECs, and reference values from these two diverse methods increases confidence in those values. Advantages and specific application of each approach are discussed. (Note: Reference values derived with the PBPK model were part of this research, and do not replace values currently posted on IRIS: http://www.epa.gov/iris/toxreviews/0286tr.pdf.).


Assuntos
Acrilamida/administração & dosagem , Biomarcadores/metabolismo , Modelos Biológicos , Acrilamida/farmacocinética , Animais , Área Sob a Curva , Feminino , Humanos , Masculino , Ratos
3.
Int J Toxicol ; 28(6): 498-509, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19966142

RESUMO

This report describes a physiologically based pharmacokinetic model for cyclohexane and its use in comparing internal doses in rats and volunteers following inhalation exposures. Parameters describing saturable metabolism of cyclohexane are measured in rats and used along with experimentally determined partition coefficients. The model is evaluated by comparing predicted blood and brain concentrations to data from studies in rats and then allometrically scaling the results to humans. Levels of cyclohexane in blood and exhaled air are measured in human volunteers and compared with model values. The model predicts that exposure of volunteers to cyclohexane at levels of 4100 mg/m(3) ( approximately 1200 ppm) will result in brain levels similar to those in rats exposed to 8000 mg/m(3) (the no-effect level for acute central nervous system effects). There are no acute central nervous system effects in humans exposed to 860 mg/m(3), consistent with model predictions that current occupational exposure levels for cyclohexane protect against acute central nervous system effects.


Assuntos
Cicloexanos/farmacocinética , Cicloexanos/toxicidade , Solventes/farmacocinética , Solventes/toxicidade , Algoritmos , Animais , Encéfalo/metabolismo , Interpretação Estatística de Dados , Humanos , Masculino , Modelos Estatísticos , Nível de Efeito Adverso não Observado , Exposição Ocupacional/efeitos adversos , Exposição Ocupacional/normas , Farmacocinética , Alvéolos Pulmonares/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Wistar , Especificidade da Espécie , Temperatura , Distribuição Tecidual , Adulto Jovem
4.
Food Chem Toxicol ; 47(4): 760-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19166901

RESUMO

Acrylamide in cooked foods results in wide-spread, low-level human exposure. Potential risks from dietary intake remain unclear due to apparent conflicting results from cancer bioassays conducted in rats that reported tumors and epidemiology studies that are suggestive but provide little or no evidence of increased cancer. Risk estimation often includes two common assumptions: (1) tumor response rates in test species can be extrapolated systematically to estimate human response rates and (2) tumor rates observed following high-dose exposures can be linearly extrapolated to predict response rates following low-dose exposures. The validity of these assumptions was evaluated for acrylamide based upon the examination of relevant toxicokinetic and toxicodynamic differences between humans and rats, including sources of nonlinearity that modify high to low dose extrapolation of cancer incidence. Important species differences and sources of nonlinearity are identified, and recommendations for addressing them within the quantitative framework of a PBTK/TD model are discussed. These differences are likely to estimate risk levels up to several orders of magnitude lower in humans than in rats. Quantitative inclusion of these TK/TD factors will more closely estimate actual human cancer risk derived from high-dose rodent studies, since detoxification processes for acrylamide and glycidamide appear adequately protective against toxicity from human dietary doses.


Assuntos
Acrilamida/toxicidade , Medição de Risco , Acrilamida/farmacocinética , Animais , Proteínas Sanguíneas/metabolismo , Sistema Enzimático do Citocromo P-450/fisiologia , Dano ao DNA , Relação Dose-Resposta a Droga , Epóxido Hidrolases/fisiologia , Humanos , Modelos Biológicos , Ligação Proteica , Especificidade da Espécie
5.
Risk Anal ; 28(5): 1375-94, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18761732

RESUMO

Dose-response assessments were conducted for the noncancer effects of acrylonitrile (AN) for the purposes of deriving subchronic and chronic oral reference dose (RfD) and inhalation reference concentration (RfC) values. Based upon an evaluation of available toxicity data, the irritation and neurological effects of AN were determined to be appropriate bases for deriving reference values. A PBPK model, which describes the toxicokinetics of AN and its metabolite 2-cyanoethylene oxide (CEO) in both rats and humans, was used to assess the dose-response data in terms of an internal dose measure for the oral RfD values, but could not be used in deriving the inhalation RfC values. Benchmark dose (BMD) methods were used to derive all reference values. Where sufficient information was available, data-derived uncertainty factors were applied to the points of departure determined by BMD methods. From this assessment, subchronic and chronic oral RfD values of 0.5 and 0.05 mg/kg/day, respectively, were derived. Similarly, subchronic and chronic inhalation RfC values of 0.1 and 0.06 mg/m(3), respectively, were derived. Confidence in the reference values derived for AN was considered to be medium to high, based upon a consideration of the confidence in the key studies, the toxicity database, dosimetry, and dose-response modeling.


Assuntos
Acrilonitrila/administração & dosagem , Carcinógenos/administração & dosagem , Acrilonitrila/farmacocinética , Acrilonitrila/toxicidade , Administração por Inalação , Administração Oral , Experimentação Animal , Animais , Carcinógenos/farmacocinética , Carcinógenos/toxicidade , Ensaios Clínicos como Assunto , Relação Dose-Resposta a Droga , Humanos , Ratos , Valores de Referência
6.
J Toxicol Environ Health A ; 70(21): 1838-48, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17934956

RESUMO

A physiologically based pharmacokinetic (PBPK) model was developed for inhaled ethylbenzene (EB) in B6C3F1 mice. The mouse physiological parameters were obtained from the literature, but the blood:air and tissue:air partition coefficients were determined by vial equilibration technique. The maximal velocity for hepatic metabolism (Vmax) obtained from a previously published rat study was increased by a factor of approximately 3 to account for enzyme induction during repeated exposures. The Michaelis affinity constant (Km) for hepatic metabolism of EB, obtained from a previously published rat PBPK modeling study, was kept unchanged during single and repeated exposure scenarios. Hepatic metabolism alone could not adequately describe the clearance of EB from mouse blood. Additional metabolism was assumed to be localized in the lung. The parameters for pulmonary metabolism were obtained by optimization of PBPK model fits to kinetic data collected following exposures to 75-1000 ppm. The PBPK model successfully predicted all available blood and tissue concentration data in mice exposed to 75 or 750 ppm EB. Overall, the results indicate that the rate of EB clearance is markedly higher in B6C3F1 mice than rats or humans and exceeds the hepatic metabolism capacity. Available biochemical evidence is consistent with a significant role for pulmonary metabolism; however, the extent to which the extrahepatic metabolism is localized in the lung is unclear. Overall, the PBPK model developed for the mouse adequately simulated the blood and tissue kinetics of EB by accounting for its high rate of clearance.


Assuntos
Derivados de Benzeno/farmacocinética , Fígado/metabolismo , Pulmão/metabolismo , Modelos Biológicos , Administração por Inalação , Animais , Derivados de Benzeno/administração & dosagem , Derivados de Benzeno/sangue , Débito Cardíaco , Feminino , Masculino , Taxa de Depuração Metabólica , Camundongos
7.
Risk Anal ; 25(2): 271-84, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15876203

RESUMO

Reference values, including an oral reference dose (RfD) and an inhalation reference concentration (RfC), were derived for propylene glycol methyl ether (PGME), and an oral RfD was derived for its acetate (PGMEA). These values were based on transient sedation observed in F344 rats and B6C3F1 mice during a two-year inhalation study. The dose-response relationship for sedation was characterized using internal dose measures as predicted by a physiologically-based pharmacokinetic (PBPK) model for PGME and its acetate. PBPK modeling was used to account for changes in rodent physiology and metabolism due to aging and adaptation, based on data collected during Weeks 1, 2, 26, 52, and 78 of a chronic inhalation study. The peak concentration of PGME in richly perfused tissues (i.e., brain) was selected as the most appropriate internal dose measure based on a consideration of the mode of action for sedation and similarities in tissue partitioning between brain and other richly perfused tissues. Internal doses (peak tissue concentrations of PGME) were designated as either no-observed-adverse-effect levels (NOAELs) or lowest-observed-adverse-effect levels (LOAELs) based on the presence or the absence of sedation at each time point, species, and sex in the two-year study. Distributions of the NOAEL and LOAEL values expressed in terms of internal dose were characterized using an arithmetic mean and standard deviation, with the mean internal NOAEL serving as the basis for the reference values, which was then divided by appropriate uncertainty factors. Where data were permitting, chemical-specific adjustment factors were derived to replace default uncertainty factor values of 10. Nonlinear kinetics, which was predicted by the model in all species at PGME concentrations exceeding 100 ppm, complicate interspecies, and low-dose extrapolations. To address this complication, reference values were derived using two approaches that differ with respect to the order in which these extrapolations were performed: (1) default approach of interspecies extrapolation to determine the human equivalent concentration (PBPK modeling) followed by uncertainty factor application, and (2) uncertainty factor application followed by interspecies extrapolation (PBPK modeling). The resulting reference values for these two approaches are substantially different, with values from the latter approach being seven-fold higher than those from the former approach. Such a striking difference between the two approaches reveals an underlying issue that has received little attention in the literature regarding the application of uncertainty factors and interspecies extrapolations to compounds where saturable kinetics occur in the range of the NOAEL. Until such discussions have taken place, reference values based on the former approach are recommended for risk assessments involving human exposures to PGME and PGMEA.


Assuntos
Propilenoglicóis/química , Propilenoglicóis/farmacocinética , Administração por Inalação , Administração Oral , Fatores Etários , Envelhecimento , Animais , Relação Dose-Resposta a Droga , Feminino , Hipnóticos e Sedativos/farmacologia , Cinética , Masculino , Camundongos , Modelos Químicos , Modelos Teóricos , Dinâmica não Linear , Perfusão , Coelhos , Ratos , Ratos Endogâmicos F344 , Padrões de Referência , Medição de Risco , Fatores Sexuais , Fatores de Tempo
8.
Risk Anal ; 24(5): 1165-83, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15563286

RESUMO

Ethylene oxide (EO) has been identified as a carcinogen in laboratory animals. Although the precise mechanism of action is not known, tumors in animals exposed to EO are presumed to result from its genotoxicity. The overall weight of evidence for carcinogenicity from a large body of epidemiological data in the published literature remains limited. There is some evidence for an association between EO exposure and lympho/hematopoietic cancer mortality. Of these cancers, the evidence provided by two large cohorts with the longest follow-up is most consistent for leukemia. Together with what is known about human leukemia and EO at the molecular level, there is a body of evidence that supports a plausible mode of action for EO as a potential leukemogen. Based on a consideration of the mode of action, the events leading from EO exposure to the development of leukemia (and therefore risk) are expected to be proportional to the square of the dose. In support of this hypothesis, a quadratic dose-response model provided the best overall fit to the epidemiology data in the range of observation. Cancer dose-response assessments based on human and animal data are presented using three different assumptions for extrapolating to low doses: (1) risk is linearly proportionate to dose; (2) there is no appreciable risk at low doses (margin-of-exposure or reference dose approach); and (3) risk below the point of departure continues to be proportionate to the square of the dose. The weight of evidence for EO supports the use of a nonlinear assessment. Therefore, exposures to concentrations below 37 microg/m3 are not likely to pose an appreciable risk of leukemia in human populations. However, if quantitative estimates of risk at low doses are desired and the mode of action for EO is considered, these risks are best quantified using the quadratic estimates of cancer potency, which are approximately 3.2- to 32-fold lower, using alternative points of departure, than the linear estimates of cancer potency for EO. An approach is described for linking the selection of an appropriate point of departure to the confidence in the proposed mode of action. Despite high confidence in the proposed mode of action, a small linear component for the dose-response relationship at low concentrations cannot be ruled out conclusively. Accordingly, a unit risk value of 4.5 x 10(-8) (microg/m3)(-1) was derived for EO, with a range of unit risk values of 1.4 x 10(-8) to 1.4 x 10(-7) (microg/m3)(-1) reflecting the uncertainty associated with a theoretical linear term at low concentrations.


Assuntos
Carcinógenos/toxicidade , Óxido de Etileno/toxicidade , Mutagênicos/toxicidade , Neoplasias/induzido quimicamente , Animais , Carcinógenos/administração & dosagem , Relação Dose-Resposta a Droga , Óxido de Etileno/administração & dosagem , Feminino , Humanos , Leucemia/induzido quimicamente , Masculino , Camundongos , Modelos Biológicos , Mutagênicos/administração & dosagem , Dinâmica não Linear , Ratos , Medição de Risco
9.
Regul Toxicol Pharmacol ; 38(3): 345-67, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14623485

RESUMO

The performance of allometric scaling of dose as a power of body weight under a variety of extrapolation conditions with respect to species, route, exposure intensity, and mechanism/mode of action, remains untested in many cases. In this paper, animal-human internal dose ratio comparisons have been developed for 12 chemicals (benzene, carbon tetrachloride, chloroform, diisopropylfluorophosphate, ethanol, ethylene oxide, methylene chloride, methylmercury, styrene, tetrachloroethene, trichloroethene, and vinyl chloride). This group of predominantly volatile and lipophilic chemicals was selected on the basis that their kinetics have been well-studied and can be predicted in mice, rats, and humans using physiologically based pharmacokinetic (PBPK) models. PBPK model predictions were compared to the allometric scaling predictions for interspecies extrapolation. Recommendations for the application of the allometric scaling are made with reference to internal dose measure (mode of action) and concentration level. The results of this assessment generally support the use of scaling factors recommended in the published literature, which includes scaling factors of 1.0 for risk assessments in which toxicity is attributed to the parent chemical or stable metabolite, and -0.75 for dose-response assessments in which toxicity is attributed to the formation of a reactive metabolite from an inhaled compound. A scaling factor of 0.75 is recommended for dose-response assessments of orally administered compounds in which toxicity is attributed to the parent chemical or stable metabolite and 1.0 for risk assessments in which toxicity is attributed to the formation of a reactive metabolite from a compound administered by the oral route. A dose-dependency in the results suggests that the scaling factors appropriate at high exposures may differ from those at low exposures, primarily due to the impact of saturable metabolism.


Assuntos
Antropometria/métodos , Relação Dose-Resposta a Droga , Inativação Metabólica/fisiologia , Medição de Risco/métodos , Administração por Inalação , Administração Oral , Animais , Benzeno/administração & dosagem , Benzeno/farmacocinética , Avaliação Pré-Clínica de Medicamentos , Etanol/administração & dosagem , Etanol/farmacocinética , Óxido de Etileno/administração & dosagem , Óxido de Etileno/farmacocinética , Humanos , Hidrocarbonetos Clorados/administração & dosagem , Hidrocarbonetos Clorados/farmacocinética , Isoflurofato/administração & dosagem , Isoflurofato/farmacocinética , Compostos de Metilmercúrio/administração & dosagem , Compostos de Metilmercúrio/farmacocinética , Camundongos , Modelos Biológicos , Ratos , Estireno/administração & dosagem , Estireno/farmacocinética
10.
Toxicol Sci ; 62(1): 124-39, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11399800

RESUMO

Methoxyethanol (ethylene glycol monomethyl ether, EGME), ethoxyethanol (ethylene glycol monoethyl ether, EGEE), and ethoxyethyl acetate (ethylene glycol monoethyl ether acetate, EGEEA) are all developmental toxicants in laboratory animals. Due to the imprecise nature of the exposure data in epidemiology studies of these chemicals, we relied on human and animal pharmacokinetic data, as well as animal toxicity data, to derive 3 occupational exposure limits (OELs). Physiologically based pharmacokinetic (PBPK) models for EGME, EGEE, and EGEEA in pregnant rats and humans have been developed (M. L. Gargas et al., 2000, Toxicol. Appl. Pharmacol. 165, 53-62; M. L. Gargas et al., 2000, Toxicol. Appl. Pharmacol. 165, 63-73). These models were used to calculate estimated human-equivalent no adverse effect levels (NAELs), based upon internal concentrations in rats exposed to no observed effect levels (NOELs) for developmental toxicity. Estimated NAEL values of 25 ppm for EGEEA and EGEE and 12 ppm for EGME were derived using average values for physiological, thermodynamic, and metabolic parameters in the PBPK model. The uncertainties in the point estimates for the NOELs and NAELs were estimated from the distribution of internal dose estimates obtained by varying key parameter values over expected ranges and probability distributions. Key parameters were identified through sensitivity analysis. Distributions of the values of these parameters were sampled using Monte Carlo techniques and appropriate dose metrics calculated for 1600 parameter sets. The 95th percentile values were used to calculate interindividual pharmacokinetic uncertainty factors (UFs) to account for variability among humans (UF(h,pk)). These values of 1.8 for EGEEA/EGEE and 1.7 for EGME are less than the default value of 3 for this area of uncertainty. The estimated human equivalent NAELs were divided by UF(h,pk) and the default UFs for pharmacodynamic variability among animals and among humans to calculate the proposed OELs. This methodology indicates that OELs (8-h time-weighted average) that should protect workers from the most sensitive adverse effects of these chemicals are 2 ppm EGEEA and EGEE (11 mg/m(3) EGEEA, 7 mg/m(3) EGEE) and 0.9 ppm (3 mg/m(3)) EGME. These recommendations assume that dermal exposure will be minimal or nonexistent.


Assuntos
Etilenoglicóis/farmacocinética , Exposição por Inalação , Modelos Biológicos , Método de Monte Carlo , Exposição Ocupacional , Níveis Máximos Permitidos , Administração por Inalação , Animais , Área Sob a Curva , Relação Dose-Resposta a Droga , Etilenoglicóis/administração & dosagem , Humanos , Nível de Efeito Adverso não Observado , Especificidade da Espécie
11.
Chem Biol Interact ; 135-136: 303-22, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11397398

RESUMO

Potential health effects of human exposure to 1,3-butadiene (BD) are of concern due to the use of BD in industry and its low-level presence throughout the environment. Physiologically based toxicokinetic (PBTK) models of BD in rodents have been developed by multiple research groups in an effort to explain species differences in toxicity (especially carcinogenic potency) through toxicokinetics. PBTK modeling of dose metrics related to a non-cancer endpoint, ovotoxicity in experimental animals, was conducted. The cumulative area under the blood concentration vs. time curve (AUC) for the metabolite diepoxybutane (butadiene diepoxide, DEB) was found to be consistent with ovotoxicity in mice and rats exposed to BD by inhalation or epoxybutene (butadiene monoepoxide, EB) or DEB by intraperitoneal injection. This suggests that cumulative DEB AUC may also be an appropriate metric for possible human risk. A preliminary human PBTK model was assembled for the eventual assessment of reproductive risk to humans and for prioritizing the determination of model parameters. The preliminary model accurately predicted published data on exhaled breath BD concentrations in a human volunteer exposed to BD by inhalation. The fit was relatively insensitive to the rate constant for BD epoxidation. Sensitivity analyses were conducted on this human PBTK model. Using a range of published rate constants, human blood DEB was found to be sensitive to rates of epoxidation of EB to DEB and hydrolysis of EB and DEB, but not BD epoxidation. Because of the large ranges of rates measured in vitro for these reactions, different combinations of in-vitro rates produce varying predictions of blood DEB concentration. Thus, validation of a human PBTK model with human biomonitoring data will be essential to produce a PBTK model that can be applied to risk assessment.


Assuntos
Butadienos/metabolismo , Butadienos/toxicidade , Modelos Biológicos , Animais , Compostos de Epóxi/metabolismo , Feminino , Humanos , Camundongos , Ratos , Medição de Risco , Sensibilidade e Especificidade , Especificidade da Espécie
12.
Toxicol Appl Pharmacol ; 165(1): 53-62, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10814553

RESUMO

Exposures to sufficiently high doses of ethylene glycol monomethyl ether (2-methoxyethanol, 2-ME) have been found to produce developmental effects in rodents and nonhuman primates. The acetic acid metabolite of 2-ME, 2-methoxyacetic acid (2-MAA), is the likely toxicant, and, as such, an understanding of the kinetics of 2-MAA is important when assessing the potential risks to humans associated with 2-ME. A previously described physiologically based pharmacokinetic (PBPK) model of 2-ME/2-MAA kinetics for rats exposed via oral or iv administration was extended and validated to inhalation exposures. Pregnant Sprague-Dawley rats were exposed for 5 days (gestation days 11-15), 6 h/day, to 2-ME vapor at 10 and 50 ppm. Validation consisted of comparing model output to maternal blood and fetal 2-ME and 2-MAA concentrations during and following 5 days of exposure (gestation days 11-15). These concentrations correspond to a known no observed effect level (NOEL) and a lowest observed effect level (LOEL) for developmental effects in rats. The rat PBPK model for 2-ME/2-MAA was scaled to humans and the model (without the pregnancy component) was used to predict data collected by other investigators on the kinetics of 2-MAA excretion in urine following exposures to 2-ME in human volunteers. The partially validated human model (with the pregnancy component) was used to predict equivalent human exposure concentrations based on 2-MAA dose measures (maximum blood concentration, C(max), and average daily area under the 2-MAA blood concentration curve, AUC, during pregnancy) that correspond to the concentrations measured at the rat NOEL and LOEL exposure concentrations. Using traditional PBPK scale-up techniques, it was calculated that pregnant women exposed for 8 h/day, 5 days/week, for the duration of pregnancy would need to be exposed to 12 or 60 ppm 2-ME to produce maternal 2-MAA blood concentrations (C(max) or average daily AUC) equivalent to those in rats exposed to the NOEL (10 ppm) or LOEL (50 ppm), respectively.


Assuntos
Etilenoglicóis/farmacocinética , Teratogênicos/farmacocinética , Acetatos/farmacocinética , Acetatos/urina , Animais , Etilenoglicóis/toxicidade , Etilenoglicóis/urina , Feminino , Humanos , Imunossupressores/farmacocinética , Imunossupressores/urina , Exposição por Inalação , Modelos Biológicos , Valor Preditivo dos Testes , Gravidez , Planejamento da Radioterapia Assistida por Computador , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Teratogênicos/toxicidade
13.
Toxicol Appl Pharmacol ; 165(1): 63-73, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10814554

RESUMO

The solvents ethylene glycol monoethyl ether acetate (EGEEA) and ethylene glycol monoethyl ether (EGEE), at sufficiently high doses, are known to be rodent developmental toxicants, exerting their toxic effects through the action of their metabolite 2-ethoxyacetic acid (2-EAA). Thus risks associated with exposure to these compounds are best evaluated based on a measure of the internal dose of 2-EAA. The goals of the work reported here were to develop physiologically based pharmacokinetic (PBPK) models of EGEEA and EGEE for pregnant rats and humans. These models were used to identify human exposure levels (ppm in air) equivalent to the rat no observed effect level (NOEL) and lowest observed effect level (LOEL) for developmental effects (Hanley et al., 1984). We exposed pregnant Sprague-Dawley rats to concentrations of EGEEA corresponding to the NOEL and LOEL. Maternal blood, urine, and fetal tissue concentrations of EGEE and 2-EAA measured in these experiments were used to validate the rat EGEEA and EGEE models. Data collected by other researchers were used to validate the capabilities of the rodent EGEEA and EGEE models to predict the kinetics in humans. The models for estimating circulating blood concentrations of 2-EAA were considered valid based on the ability of the model to accurately predict 2-EAA concentrations in rat blood, urine, and fetal tissue. The human inhaled concentration equivalent to the rat NOEL for EGEEA (50 ppm) was predicted to be 25 ppm using the maternal blood average daily area under the curve (AUC) and 40 ppm using the maximum concentration achieved in maternal blood (C(max)). The human inhaled concentration equivalent to the rat LOEL for EGEEA (100 ppm) was determined to be 55 ppm using the maternal blood average daily AUC and 80 ppm using the maternal blood C(max).


Assuntos
Etilenoglicóis/farmacocinética , Teratogênicos/farmacocinética , Animais , Exposição Ambiental , Etilenoglicóis/toxicidade , Feminino , Humanos , Exposição por Inalação , Modelos Biológicos , Exposição Ocupacional , Valor Preditivo dos Testes , Gravidez , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Teratogênicos/toxicidade
14.
Chem Biol Interact ; 104(2-3): 103-15, 1997 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-9212778

RESUMO

1,2-Epoxybutene (BMO) and diepoxybutane (BDE) are metabolic products of 1,3-butadiene in rodents. Both BMO and BDE are suspect in the development of tumors in rats and mice. To understand the distribution and elimination of these compounds in the absence of the rate-limiting production from butadiene, the pharmacokinetics of BMO and BDE in blood were determined in adult male Sprague-Dawley rats following intravenous administration. All animals were dually cannulated in these studies. For the BMO studies, rats were dosed with 71, 143, or 286 mumol/kg BMO (n = 3 for each dose group). For the BDE studies, rats were dosed with 523 mumol/kg BDE (n = 3). All animals tolerated the BMO and BDE doses without grossly observable adverse effects. Blood was drawn at predetermined time points and extracted in methylene chloride. BDE and BMO concentrations were quantitated by gas chromatography or gas chromatography/mass spectrometry. The BMO distribution half-lives were short and ranged from 1.4 min at the lowest dose to 1.8 min at the highest dose. Volume of distribution at steady state ranged from 0.53 +/- 0.17 to 0.59 +/- 0.31 l/kg. Systemic clearances ranged from 67 +/- 17 to 114 +/- 20 ml/min per kg. The terminal elimination half-lives were also short and ranged from 5.7 to 8.5 min among the doses. The pharmacokinetic parameters after an i.v. dose of 523 mumol/kg BDE were a distribution half-life of 2.7 min, terminal elimination T1/2 of 14 min, volume of distribution at steady state of 0.73 +/- 0.06 l/kg, and systemic clearance of 76 +/- 8 ml/min per kg. These pharmacokinetic parameters demonstrate the similarity between disposition of the two epoxides in rats, that include a rapid distribution after i.v. administration into a small extravascular body compartment as well as a rapid elimination from blood. These pharmacokinetic data provide useful blood clearance information for assessing the critical physiological and biochemical determinants underlying the disposition of butadiene epoxides.


Assuntos
Carcinógenos/farmacocinética , Compostos de Epóxi/farmacocinética , Mutagênicos/farmacocinética , Animais , Área Sob a Curva , Carcinógenos/administração & dosagem , Cromatografia Gasosa , Relação Dose-Resposta a Droga , Compostos de Epóxi/administração & dosagem , Compostos de Epóxi/sangue , Cromatografia Gasosa-Espectrometria de Massas , Meia-Vida , Hematócrito , Injeções Intravenosas , Masculino , Mutagênicos/administração & dosagem , Ratos , Ratos Sprague-Dawley , Padrões de Referência , Software
15.
Carcinogenesis ; 18(4): 611-25, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9111190

RESUMO

1,3-Butadiene (BD) is a more potent tumor inducer in mice than in rats. BD also shows striking differences in metabolic activation, with substantially higher blood concentrations of 1,2:3,4-diepoxybutane (butadiene diepoxide; BDE) in BD-exposed mice than in similarly exposed rats. The objective of this study was to develop a single mechanistic model structure capable of describing BD disposition in both species. To achieve this objective, known pathways of 1,2-epoxy-3-butene (butadiene monoepoxide; BMO) and BDE metabolism were incorporated into a physiologically based pharmacokinetic model by scaling rates determined in vitro. With this model structure, epoxide clearance was underestimated for both rats and mice. Improved simulation of blood epoxide concentrations was achieved by addition of first-order metabolism in the slowly perfused tissues, verified by simulation of data on the time course for BMO elimination after i.v. injection of BMO. Blood concentrations of BD were accurately predicted for mice and rats exposed by inhalation to constant concentrations of BD. However, if all BD was assumed to be metabolized to BMO, blood concentrations of BMO were overpredicted. By assuming that only a fraction of BD metabolism produces BMO, blood concentrations of BMO could be predicted over a range of BD exposure concentrations for both species. In vitro and in vivo studies suggest an alternative cytochrome P-450-mediated pathway for BD metabolism that does not yield BMO. Including an alternative pathway for BD metabolism in the model also gave accurate predictions of blood BDE concentrations after inhalation of BD. Blood concentrations of BMO and BDE observed in both mice and rats are best explained by the existence of an alternative pathway for BD metabolism which does not produce BMO.


Assuntos
Butadienos/farmacocinética , Compostos de Epóxi/farmacocinética , Administração por Inalação , Animais , Butadienos/sangue , Butadienos/toxicidade , Compostos de Epóxi/sangue , Compostos de Epóxi/toxicidade , Injeções Intravenosas , Camundongos , Modelos Biológicos , Ratos , Especificidade da Espécie
16.
Drug Chem Toxicol ; 20(4): 401-9, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9433667

RESUMO

Effective management of human health and ecological hazards in the manufacturing and maintenance environment can be achieved by focusing on the risks associated with these operations. The NDCEE Industrial Health Risk Assessment (IHRA) Program is developing a comprehensive approach to risk analysis applied to existing processes and used to evaluate alternatives. The IHRA Risk-Based Tiered Approach (RBTASM) builds on the American Society for Testing and Materials (ASTM) Risk-Based Corrective Action (RBCA) effort to remediate underground storage tanks. Using readily available information, a semi-quantitative ranking of alternatives based on environmental, safety, and occupational health criteria was produced. A Rapid Screening Assessment of alternative corrosion protection products was performed on behalf of the Joint Group on Acquisition Pollution Prevention (JG-APP). Using the RBTASM in pollution prevention alternative selection required higher tiered analysis and more detailed assessment of human health risks under site-specific conditions. This example illustrates the RBTASM for a organic finishing line using three different products (one conventional spray and two alternative powder coats). The human health risk information developed using the RBTASM is considered along with product performance, regulatory, and cost information by risk managers downselecting alternatives for implementation or further analysis.


Assuntos
Ecologia , Poluição Ambiental/prevenção & controle , Medição de Risco , Animais , Exposição Ambiental/prevenção & controle , Humanos , Indústrias , Saúde Ocupacional , Estados Unidos
17.
Toxicology ; 113(1-3): 318-21, 1996 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-8901917

RESUMO

In vitro and in vivo butadiene (BD) metabolism data from laboratory animals were integrated into a rodent physiologically based pharmacokinetic (PBPK) model with flow- and diffusion-limited compartments. The resulting model describes experimental data from multiple sources under scenarios such as closed chamber inhalation and nose-only flow-through inhalation exposures. Incorporation of diurnal glutathione (GSH) variation allows accurate simulation of GSH changes observed in air control nose-only exposures and BD exposures. An isolated tissue model based on rate parameters determined in vitro predicts the decrease in epoxide concentrations in intact animals during the time lag between exsanguination and tissue removal for tissues capable of epoxide biotransformation, providing a better indication of in vivo dosimetry. Further refinements of the model are required relative to model predictions of an important BD metabolite, diepoxybutane.


Assuntos
Carcinógenos/farmacocinética , Compostos de Epóxi/farmacocinética , Modelos Biológicos , Animais , Glutationa/metabolismo , Camundongos , Ratos
18.
Ann Biomed Eng ; 24(2): 305-20, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8678360

RESUMO

Naphthalene is a toxicant with unusual species and tissue specificity that has been the subject of in vitro studies. We describe a preliminary physiologically based pharmacokinetic (PBPK) model for naphthalene constructed solely from in vitro data for comparison to animal data without the use of adjustable parameters. The prototypical PBPK model containing five lumped tissue compartments was developed to describe the uptake and metabolism of naphthalene by mice and rats dosed intraperitoneally (i.p.) and orally (po). The model incorporates circulation and biotransformation of the semistable reactive intermediate, naphthalene oxide, as well as the parent compound naphthalene. Circulation is included because the toxic action of naphthalene has been proposed to be caused by the formation of a reactive metabolite in one organ (liver) and its circulation to another organ (lung) being adversely affected by the metabolite. The model allows conversion of naphthalene oxide into dihydrodiol, glutathione (GSH) conjugates, 1-naphthol (non-enzymatically) and covalently bound adducts with proteins. Model simulations are compared with previously reported in vivo measurements of glutathione depletion, mercapturic acid formation, and covalently bound protein formation. The mouse model predicts accurately the amount of mercapturates excreted, the effect of various pretreatments, and the extent of covalent binding in the lung and liver resulting from ip administration, including the sharp increase in binding between 200 and 400 mg/kg.


Assuntos
Modelos Biológicos , Naftalenos/farmacocinética , Absorção , Administração Oral , Animais , Disponibilidade Biológica , Compartimentos de Líquidos Corporais , Células Cultivadas , Glutationa/metabolismo , Injeções Intraperitoneais , Fígado/metabolismo , Fígado/patologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Tamanho do Órgão , Ratos , Análise de Regressão
19.
Toxicol In Vitro ; 9(3): 307-16, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20650092

RESUMO

The difficulties of large-scale animal testing of compounds has spurred development of in vitro testing methods and physiologically based pharmacokinetic models (PBPK). In existing in vitro methods, tissue interactions occurring in vivo are not reproduced accurately and in PBPKs the a priori prediction of metabolism is difficult. Through development of a multicompartmental, multiple cell type bioreactor system these limitations can be circumvented. A cell culture analogue (CCA) of a PBPK was developed. The CCA contains multiple chambers, each of which represents a tissue or group of similar tissues as specified in the PBPK. Proof-of-concept experiments were done using naphthalene as a model. Naphthalene is converted into naphthalene oxide and the circulation of this reactive metabolite from the liver to lung is a possible mechanism for lung injury. A CCA with liver, lung and other tissue compartments was constructed. This system was used in conjunction with cultured H4IIE rat hepatoma cells and L2 rat lung cells to study the importance of circulated naphthalene metabolites (presumably naphthalene oxides) on lung cell toxicity in rodents. By increasing the number of cells and/or inducing cytochrome P-450 activity in the liver compartment, lung cell mortality was increased. Glutathione depletion in the lung and liver cells was also observed. These results indicate that the CCA is a potentially useful concept for studying the action of compounds with reactive metabolites.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...