Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Front Pharmacol ; 14: 1088011, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36909196

RESUMO

Introduction: A physiologically based pharmacokinetic (PBPK) model for 3-chloroallyl alcohol (3-CAA) was developed and used to evaluate the design of assays for the in vivo genotoxicity of 3-CAA. Methods: Model development was supported by read across from a published PBPK model for ethanol. Read across was motivated by the expectation that 3-CAA, which like ethanol is a primary alcohol, is metabolized largely by hepatic alcohol dehydrogenases. The PBPK model was used to evaluate how two metrics of tissue dosimetry, maximum blood concentration (Cmax; mg/L) and area under the curve (AUC; mg-hr/L) vary with dose of 3-CAA and with dose route (oral gavage, drinking water). Results: The model predicted that oral gavage results in a 6-fold higher Cmax than the same dose administered in drinking water, but in similar AUCs. Predicted Cmax provided the best correlation with severe toxicity (e.g., lethality) from 3-CAA, consistent with the production of a reactive metabolite. Therefore, drinking water administration can achieve higher sustained concentration without severe toxicity in vivo. Discussion: This evaluation is significant because cytotoxicity is a potential confounder of mutagenicity testing. The PBPK model can be used to ensure that studies meet OECD and USEPA test guidelines and that the highest dose used is not associated with severe toxicity. In addition, PBPK modeling provides assurance of target tissue (e.g., bone marrow) exposure even in the absence of laboratory data, by defining the relationship between applied dose and target tissue dose based on accepted principles of pharmacokinetics, relevant physiology and biochemistry of the dosed animals, and chemical-specific information.

2.
Toxicol Sci ; 191(1): 15-24, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36409013

RESUMO

Understanding the dose-response for formaldehyde-induced nasal cancer in rats is complicated by (1) the uneven distribution of inhaled formaldehyde across the interior surface of the nasal cavity and, (2) the presence of endogenous formaldehyde (endoF) in the nasal mucosa. In this work, we used computational fluid dynamics (CFD) modeling to predict flux of inhaled (exogenous) formaldehyde (exogF) from air into tissue at the specific locations where DNA adducts were measured. Experimental work has identified DNA-protein crosslink (DPX) adducts due to exogF and deoxyguanosine (DG) adducts due to both exogF and endoF. These adducts can be considered biomarkers of exposure for effects of endoF and exogF on DNA that may be part of the mechanism of tumor formation. We describe a computational model linking CFD-predicted flux of formaldehyde from air into tissue, and the intracellular production of endoF, with the formation of DPX and DG adducts. We assumed that, like exogF, endoF can produce DPX. The model accurately reproduces exogDPX, exogDG, and endoDG data after inhalation from 0.7 to 15 ppm. The dose-dependent concentrations of exogDPX and exogDG are predicted to exceed the concentrations of their endogenous counterparts at about 2 and 6 ppm exogF, respectively. At all concentrations examined, the concentrations of endoDPX and exogDPX were predicted to be at least 10-fold higher than that of their DG counterparts. The modeled dose-dependent concentrations of these adducts are suitable to be used together with data on the dose-dependence of cell proliferation to conduct quantitative modeling of formaldehyde-induced rat nasal carcinogenicity.


Assuntos
Adutos de DNA , DNA , Ratos , Animais , Ratos Endogâmicos F344 , Mucosa Nasal , Formaldeído/toxicidade , Desoxiguanosina
3.
Toxicol Sci ; 191(2): 212-226, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36453847

RESUMO

In earlier physiologically based pharmacokinetic (PBPK) models for manganese (Mn), the kinetics of transport of Mn into and out of tissues were primarily driven by slow rates of association and dissociation of Mn with tissue binding sites. However, Mn is known to show rapidly reversible binding in tissues. An updated Mn model for primates, following similar work with rats, was developed that included rapid association/dissociation processes with tissue Mn-binding sites, accumulation of free Mn in tissues after saturation of these Mn-binding sites and rapid rates of entry into tissues. This alternative structure successfully described Mn kinetics in tissues in monkeys exposed to Mn via various routes including oral, inhalation, and intraperitoneal, subcutaneous, or intravenous injection and whole-body kinetics and tissue levels in humans. An important contribution of this effort is showing that the extension of the rate constants for binding and cellular uptake established in the monkey were also able to describe kinetic data from humans. With a consistent model structure for monkeys and humans, there is less need to rely on cadaver data and whole-body tracer studies alone to calibrate a human model. The increased biological relevance of the Mn model structure and parameters provides greater confidence in applying the Mn PBPK models to risk assessment. This model is also well-suited to explicitly incorporate emerging information on the role of transporters in tissue disposition, intestinal uptake, and hepatobiliary excretion of Mn.


Assuntos
Manganês , Modelos Biológicos , Humanos , Ratos , Animais , Haplorrinos , Transporte Biológico , Administração por Inalação
5.
Sci Rep ; 12(1): 1091, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35058535

RESUMO

Physiologically based pharmacokinetic (PBPK) modeling can be a useful tool for characterizing nicotine pharmacokinetics (PK) from use of tobacco products. We expand a previously published PBPK model to simulate a nicotine PK profile, following single or multiple use of various tobacco products [cigarettes, smokeless tobacco, and electronic nicotine delivery systems, or a nicotine inhaler (NICOTROL)] The uptake route in the model was designed to allow for three uptake compartments: buccal cavity (BC), upper respiratory tract (URT) (conducting and transitional airways) and lower respiratory tract (alveolar region). Within each region, the model includes product-specific descriptions of the flux of nicotine into plasma, as well as the flux of nicotine from the BC and URT to the gastrointestinal tract. These descriptions are based on regional deposition and diffusion models of nicotine into plasma, which depends on the product type. Regional deposition flux combined with regional differences in physiological parameters (e.g., blood perfusion ratio and tissue thickness) play a key role in the product-specific PK profile of nicotine. The current model describes the slower flux of nicotine into plasma across the BC and URT, as well as the rapid flux known to occur in the alveolar region. Overall, the addition of the BC and respiratory tract compartments to the nicotine model provided simulation results that are comparable to the nicotine time-course plasma concentrations reported from clinical studies for the four product categories simulated.


Assuntos
Nicotina/administração & dosagem , Nicotina/farmacocinética , Uso de Tabaco/fisiopatologia , Fumar Cigarros , Biologia Computacional/métodos , Simulação por Computador , Sistemas Eletrônicos de Liberação de Nicotina , Humanos , Modelos Biológicos , Produtos do Tabaco/efeitos adversos , Uso de Tabaco/efeitos adversos , Tabaco sem Fumaça
6.
Toxicol Appl Pharmacol ; 417: 115463, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33631232

RESUMO

By extending our Paraquat (PQ) work to include primates we have implemented a modelling and simulation strategy that has enabled PQ pharmacokinetic data to be integrated into a single physiologically based pharmacokinetic (PBPK) model that enables more confident extrapolation to humans. Because available data suggested there might be differences in PQ kinetics between primates and non-primates, a radiolabelled study was conducted to characterize pharmacokinetics and excretion in Cynomolgus monkeys. Following single intravenous doses of 0.01 or 0.1 mg paraquat dichloride/kg bw, plasma PQ concentration-time profiles were dose-proportional. Excretion up to 48 h (predominantly urinary) was 82.9%, with ca. 10% remaining unexcreted. In vitro blood binding was similar across Cynomolgus monkeys, humans and rat. Our PBPK model for the rat, mouse and dog, employing a single set of PQ-specific parameters, was scaled to Cynomolgus monkeys and well represented the measured plasma concentration-time profiles over 14 days. Addition of a cartilage compartment to the model better captured the percent remaining in the monkeys at 48 h, whilst having negligible effect on model predictions for the other species. The PBPK model performed well for all four species, demonstrating there is little difference in PQ kinetics between non-primates and primates enabling a more confident extrapolation to humans. Scaling of the PBPK model to humans, with addition of a human-specific dermal submodel based on in vitro human dermal absorption data, provides a valuable tool that could be employed in defining internal dosimetry to complement human health risk assessments.


Assuntos
Herbicidas/farmacocinética , Modelos Biológicos , Paraquat/farmacocinética , Animais , Simulação por Computador , Herbicidas/administração & dosagem , Herbicidas/sangue , Herbicidas/toxicidade , Humanos , Infusões Intravenosas , Eliminação Intestinal , Macaca fascicularis , Paraquat/administração & dosagem , Paraquat/sangue , Paraquat/toxicidade , Ratos , Eliminação Renal , Medição de Risco , Absorção Cutânea , Especificidade da Espécie , Distribuição Tecidual , Toxicocinética
7.
Toxicol Appl Pharmacol ; 417: 115462, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33631233

RESUMO

Paraquat dichloride (PQ) is a non-selective herbicide which has been the subject of numerous toxicology studies over more than 50 years. This paper describes the development of a physiologically-based pharmacokinetic (PBPK) model of PQ kinetics for the rat, mouse and dog, firstly to aid the interpretation of studies in which no kinetic measurements were made, and secondly to enable the future extension of the model to humans. Existing pharmacokinetic data were used to develop a model for the rat and mouse. Simulations with this preliminary model were then used to identify key data gaps and to design a new blood binding study to reduce uncertainty in critical aspects of the model. The new data provided evidence to support the model structure, and its predictive performance was then assessed against dog and rat datasets not used in model development. The PQ-specific model parameters are the same for all three species, with only the physiological parameters varying between species. This consistency across species provides a strong basis for extrapolation to other species, as demonstrated here for the dog. The model enables a wide range of PQ data to be linked together to provide a broad understanding of PQ pharmacokinetics in rodents and the dog, showing that the key aspects of PQ kinetics in these species are understood and adequately encapsulated within the model.


Assuntos
Herbicidas/farmacocinética , Modelos Biológicos , Paraquat/farmacocinética , Animais , Simulação por Computador , Cães , Herbicidas/sangue , Herbicidas/toxicidade , Eliminação Intestinal , Camundongos , Paraquat/sangue , Paraquat/toxicidade , Ligação Proteica , Ratos , Eliminação Renal , Medição de Risco , Especificidade da Espécie , Distribuição Tecidual , Toxicocinética
8.
Int J Environ Health Res ; 31(8): 951-962, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31850798

RESUMO

A physiologically based pharmacokinetic (PBPK) model was developed to described uptake, disposition and clearance of bromate in the rat using published experimental data in rat. The rodent bromate model was extrapolated to human using species-specific physiological parameters and standard interspecies scaling of rate constants. The bromate model is kinetically linear (i.e. AUC and Cmax) across the range of drinking water concentrations used in the cancer bioassays (15 to 500 ppm). This is likely the result of the poor oral bioavailability of bromate due to high reduction rates in the intestinal tract. The bromate PBPK model was used to assess the human equivalent drinking water concentration (HEC) consistent with average plasma concentrations in the rodent bioassays. At drinking water concentrations <500 mg/L, the predicted HEC was two to three fold lower than the bioassay concentration and was dependent on the reported drinking water intake reported in the bioassay.


Assuntos
Bromatos/farmacocinética , Água Potável/química , Poluentes Químicos da Água/farmacocinética , Animais , Disponibilidade Biológica , Bromatos/análise , Simulação por Computador , Exposição Dietética/análise , Feminino , Humanos , Modelos Biológicos , Ratos , Poluentes Químicos da Água/análise
9.
Toxicol Sci ; 177(2): 325-333, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32735340

RESUMO

Although formaldehyde is a normal constituent of tissues, lifetime inhalation exposures at 6 h/day, 5 days/week at concentrations ≥6 ppm caused a nonlinear increase in nasal tumors in rats with incidence reaching close to 50% at 15 ppm. Studies with heavy isotope labeled [13CD2]-formaldehyde permit quantification of both the mass-labeled exogenous and endogenous DNA-formaldehyde reaction products. An existing pharmacokinetic model developed initially to describe 14C-DNA-protein crosslinks (DPX) provided a template for describing the time course of mass-labeled adducts. Published datasets included both DPX and N2-HO13CD2-dG adducts measured after a single 6-h exposure to 0.7, 2, 6, 9, 10, or 15 ppm formaldehyde, after multi-day exposures to 2 ppm for 6 h/day, 7 days/week with interim sacrifices up to 28 days, and after 28-day exposures for 6 h/day, 7 days/week to 0.3, 0.03, or 0.001 ppm. The existing kinetic model overpredicted endogenous adducts in the nasal epithelium after 1-day [13CD2]-formaldehyde exposure, requiring adjustment of parameters for rates of tissue metabolism and background formaldehyde. After refining tissue formaldehyde parameters, we fit the model to both forms of adducts by varying key parameters and optimizing against all 3 studies. Fitting to all these studies required 2 nonlinear pathways-one for high-exposure saturation of clearance in the nasal epithelial tissues and another for extracellular clearance that restricts uptake into the epithelial tissue for inhaled concentrations below 0.7 ppm. This refined pharmacokinetic model for endogenous and exogenous formaldehyde acetal adducts can assist in updating biologically based dose-response models for formaldehyde carcinogenicity.


Assuntos
Adutos de DNA , Formaldeído/toxicidade , Guanina , Animais , DNA , Cinética , Mucosa Nasal , Ratos
10.
J Toxicol Environ Health A ; 83(19-20): 631-648, 2020 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-32757748

RESUMO

A physiologically based pharmacokinetic (PBPK) model for di-isononyl phthalate (DiNP) was developed by adapting the existing models for di(2-ethylhexyl) phthalate (DEHP) and di-butylphthalate (DBP). Both pregnant rat and human time-course plasma and urine data were used to address the hydrolysis of DiNP in intestinal tract, plasma, and liver as well as hepatic oxidative metabolism and conjugation of the monoester and primary oxidative metabolites. Data in both rats and humans were available to inform the uptake and disposition of mono-isononyl phthalate (MiNP) as well as the three primary oxidative metabolites including hydroxy (7-OH)-, oxo (7-OXO)-, and carboxy (7-COX)-monoisononyl phthalate in plasma and urine. The DiNP model was reliable over a wide range of exposure levels in the pregnant rat as well as the two low exposure levels in humans including capturing the nonlinear behavior in the pregnant rat after repeated 750 mg/kg/day dosing. The presented DiNP PBPK model in pregnant rat and human, based upon an extensive kinetic dataset in both species, may provide a basis for assessing human equivalent exposures based upon either rodent or in vitro points of departure.


Assuntos
Poluentes Ambientais/farmacocinética , Ácidos Ftálicos/farmacocinética , Plastificantes/farmacocinética , Animais , Feminino , Humanos , Intestinos , Fígado/metabolismo , Desintoxicação Metabólica Fase II , Modelos Animais , Oxirredução , Plasma/metabolismo , Gravidez , Ratos
11.
Toxicol In Vitro ; 66: 104855, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32278033

RESUMO

Advancements in measurement and modeling capabilities are providing unprecedented access to estimates of chemical exposure and bioactivity. With this influx of new data, there is a need for frameworks that help organize and disseminate information on chemical hazard and exposure in a manner that is accessible and transparent. A case study approach was used to demonstrate integration of the Adverse Outcome Pathway (AOP) and Aggregate Exposure Pathway (AEP) frameworks to support cumulative risk assessment of co-exposure to two phthalate esters that are ubiquitous in the environment and that are associated with disruption of male sexual development in the rat: di(2-ethylhexyl) phthalate (DEHP) and di-n-butyl phthalate (DnBP). A putative AOP was developed to guide selection of an in vitro assay for derivation of bioactivity values for DEHP and DnBP and their metabolites. AEPs for DEHP and DnBP were used to extract key exposure data as inputs for a physiologically based pharmacokinetic (PBPK) model to predict internal metabolite concentrations. These metabolite concentrations were then combined using in vitro-based relative potency factors for comparison with an internal dose metric, resulting in an estimated margin of safety of ~13,000. This case study provides an adaptable workflow for integrating exposure and toxicity data by coupling AEP and AOP frameworks and using in vitro and in silico methodologies for cumulative risk assessment.


Assuntos
Dibutilftalato , Dietilexilftalato , Exposição Ambiental/efeitos adversos , Poluentes Ambientais , Modelos Biológicos , Rotas de Resultados Adversos , Animais , Dibutilftalato/farmacocinética , Dibutilftalato/farmacologia , Dibutilftalato/toxicidade , Dietilexilftalato/farmacocinética , Dietilexilftalato/farmacologia , Dietilexilftalato/toxicidade , Poluentes Ambientais/farmacocinética , Poluentes Ambientais/farmacologia , Poluentes Ambientais/toxicidade , Humanos , Masculino , Ratos , Desenvolvimento Sexual/efeitos dos fármacos
12.
Environ Res ; 182: 109017, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31865168

RESUMO

Exposure to perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) has been associated with the occurrence of thyroid disease in some epidemiologic studies. We hypothesized that in a specific epidemiologic study based on the National Health and Nutrition Examination Survey, the association of subclinical thyroid disease with serum concentration of PFOA and PFOS was due to reverse causality. Thyroid hormone affects glomerular filtration, which in turn affects excretion of PFOA and PFOS. We evaluated this by linking a model of thyroid disease status over the lifetime to physiologically based pharmacokinetic models of PFOA and PFOS. Using Monte Carlo methods, we simulated the target study population and analyzed the data using multivariable logistic regression. The target and simulated populations were similar with respect to age, estimated glomerular filtration rate, serum concentrations of PFOA and PFOS, and prevalence of subclinical thyroid disease. Our findings suggest that in the target study the associations with subclinical hypothyroidism were overstated and the results for subclinical hyperthyroidism were, in general, understated. For example, for subclinical hypothyroidism in men, the reported odds ratio per ln(PFOS) increase was 1.98 (95% CI 1.19-3.28), whereas in the simulated data the bias due to reverse causality gave an odds ratio of 1.19 (1.16-1.23). Our results provide evidence of bias due to reverse causality in a specific cross-sectional study of subclinical thyroid disease with exposure to PFOA and PFOS among adults.


Assuntos
Ácidos Alcanossulfônicos , Poluentes Ambientais , Fluorocarbonos , Doenças da Glândula Tireoide , Adulto , Caprilatos , Estudos Transversais , Poluentes Ambientais/sangue , Poluentes Ambientais/toxicidade , Feminino , Fluorocarbonos/sangue , Fluorocarbonos/toxicidade , Humanos , Masculino , Inquéritos Nutricionais , Doenças da Glândula Tireoide/induzido quimicamente
13.
Inhal Toxicol ; 31(13-14): 468-483, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31992090

RESUMO

Objective: To develop a physiologically based pharmacokinetic (PBPK) model for chloroprene in the mouse, rat and human, relying only on in vitro data to estimate tissue metabolism rates and partitioning, and to apply the model to calculate an inhalation unit risk (IUR) for chloroprene.Materials and methods: Female B6C3F1 mice were the most sensitive species/gender for lung tumors in the 2-year bioassay conducted with chloroprene. The PBPK model included tissue metabolism rate constants for chloroprene estimated from results of in vitro gas uptake studies using liver and lung microsomes. To assess the validity of the PBPK model, a 6-hr, nose-only chloroprene inhalation study was conducted with female B6C3F1 mice in which both chloroprene blood concentrations and ventilation rates were measured. The PBPK model was then used to predict dose measures - amounts of chloroprene metabolized in lungs per unit time - in mice and humans.Results: The mouse PBPK model accurately predicted in vivo pharmacokinetic data from the 6-hr, nose-only chloroprene inhalation study. The PBPK model was used to conduct a cancer risk assessment based on metabolism of chloroprene to reactive epoxides in the lung, the target tissue in mice. The IUR was over100-fold lower than the IUR from the EPA Integrated Risk Information System (IRIS), which was based on inhaled chloroprene concentration. The different result from the PBPK model risk assessment arises from use of the more relevant tissue dose metric, amount metabolized, rather than inhaled concentrationDiscussion and conclusions: The revised chloroprene PBPK model is based on the best available science, including new test animal in vivo validation, updated literature review and a Markov-Chain Monte Carlo analysis to assess parameter uncertainty. Relying on both mouse and human metabolism data also provides an important advancement in the use of quantitative in vitro to in vivo extrapolation (QIVIVE). Inclusion of the best available science is especially important when deriving a toxicity value based on species extrapolation for the potential carcinogenicity of a reactive metabolite.


Assuntos
Poluentes Atmosféricos/farmacocinética , Cloropreno/farmacocinética , Exposição por Inalação/efeitos adversos , Pulmão/metabolismo , Modelos Biológicos , Poluentes Atmosféricos/sangue , Poluentes Atmosféricos/toxicidade , Animais , Cloropreno/sangue , Cloropreno/toxicidade , Feminino , Humanos , Exposição por Inalação/análise , Pulmão/efeitos dos fármacos , Cadeias de Markov , Camundongos , Método de Monte Carlo , Pletismografia , Valor Preditivo dos Testes , Ratos , Medição de Risco , Especificidade da Espécie , Distribuição Tecidual
15.
Environ Int ; 113: 91-99, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29421411

RESUMO

A higher body mass index (BMI) has been positively associated with the rate of excretion of di-2-ethylhexyl phthalate (DEHP) metabolites in urine in data from the National Health and Nutrition Examination Survey (NHANES), suggesting an association between DEHP exposure and BMI. The association, however, may be due to the association between body mass maintenance and higher energy intake, with higher energy intake being accompanied by a higher intake of DEHP. To examine this hypothesis, we ran a Monte Carlo simulation with a DEHP physiologically-based pharmacokinetic (PBPK) model for adult humans. A realistic exposure sub-model was used, which included the relation of body weight to energy intake and of energy intake to DEHP intake. The model simulation output, when compared with urinary metabolite data from NHANES, supported good model validity. The distribution of BMI in the simulated population closely resembled that in the NHANES population. This indicated that the simulated subjects and DEHP exposure model were closely aligned with the NHANES population of interest. In the simulated population, the ordinary least squares regression coefficient for log(BMI) as a function of log(DEHP nmol/min) was 0.048 (SE 0.001), as compared with the reported value of 0.019 (SE 0.005). In other words, given our model structure, the higher energy intake in the overweight and obese, and the concomitant higher DEHP exposure, describes the reported relationship between BMI and DEHP.


Assuntos
Peso Corporal/fisiologia , Dietilexilftalato/urina , Ingestão de Energia/fisiologia , Obesidade , Adulto , Índice de Massa Corporal , Simulação por Computador , Dietilexilftalato/farmacocinética , Exposição Ambiental , Humanos , Método de Monte Carlo , Inquéritos Nutricionais , Obesidade/metabolismo , Obesidade/urina , Sobrepeso/metabolismo , Sobrepeso/urina
16.
Toxicol Lett ; 279 Suppl 1: 125-135, 2017 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-28408153

RESUMO

The multi-compound, and multi-dose (MC-MD) route physiologically based pharmacokinetic (PBPK) model for cyclic siloxanes reported by McMullin et al. (2016) brought together the series of models for octamethylcyclotetrasiloxane (D4) and decamethylcyclopentasiloxane (D5) in rat and human into a unified code structure that would allow simulation of both compounds following the inhalation and dermal routes of exposure. The refined MC-MD PBPK model presented here expands upon this effort to include representation of rat kinetic data in plasma, tissues and exhaled breath for the parent compounds after oral bolus administration. Additional refinements were made with regards to hepatic induction of metabolism in the liver and allometric scaling of rate constants for the deep tissue compartments which will allow the MC-MD model to be used in uncertainty analysis. Overall, the refined MC-MD model was able to reproduce both parent D4 and D5 kinetic data in rat and human after inhalation exposure (rat and human) or dermal exposure (human). The inclusion of sequestered (i.e., lipid associated) oral absorption into plasma after oral bolus dosing successfully described the lack of exhalation as well as the initial distribution of siloxane to the liver which was higher than simple partitioning from plasma would allow. The refined MC-MD PBPK model presented here can be incorporated into uncertainty and variability analysis and cross-species dosimetry for both D4 and D5.


Assuntos
Exposição Ambiental , Siloxanas/metabolismo , Administração Oral , Adulto , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Exposição por Inalação , Masculino , Modelos Biológicos , Ratos , Siloxanas/toxicidade
17.
Toxicol Appl Pharmacol ; 332: 149-158, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28392392

RESUMO

Dichloromethane (DCM) is a lung and liver carcinogen in mice at inhalation exposures≥2000ppm. The modes of action (MOA) of these responses have been attributed to formation of genotoxic, reactive metabolite(s). Here, we examined gene expression in lung and liver from female B6C3F1 mice exposed to 0, 100, 500, 2000, 3000 and 4000ppm DCM for 90days. We also simulated dose measures - rates of DCM oxidation to carbon monoxide (CO) in lung and liver and expected blood carboxyhemoglobin (HbCO) time courses with a PBPK model inclusive of both conjugation and oxidation pathways. Expression of large numbers of genes was altered at 100ppm with maximal changes in the numbers occurring by 500 or 2000ppm. Most changes in genes common to the two tissues were related to cellular metabolism and circadian clock. At the lower concentrations, the changes in metabolism-related genes were discordant - up in liver and down in lung. These processes included organelle biogenesis, TCA cycle, and respiratory electron transport. Changes in circadian cycle genes - primarily transcription factors - showed strong concentration-related response at higher concentrations (Arntl, Npas2, and Clock were down-regulated; Cry2, Wee1, Bhlhe40, Per3, Nr1d1, Nr1d2 and Dbp) were up-regulated with similar directionality in both tissues. Overall, persistently elevated HbCO from DCM oxidation appears to cause extended periods of hypoxia, leading to altered circadian coupling to cellular metabolism. The dose response for altered circadian processes correlates with the cancer outcome. We found no evidence of changes in genes indicative of responses to cytotoxic, DNA-reactive metabolites.


Assuntos
Ritmo Circadiano , Hipóxia/genética , Fígado/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Cloreto de Metileno/toxicidade , Transcriptoma , Animais , Carboxihemoglobina/genética , Carboxihemoglobina/metabolismo , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/genética , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica , Hipóxia/induzido quimicamente , Hipóxia/patologia , Exposição por Inalação/efeitos adversos , Fígado/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos , Farmacocinética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
Toxicol Sci ; 150(2): 441-53, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26794140

RESUMO

The previously-published physiologically based pharmacokinetic model for atrazine (ATZ), deisopropylatrazine (DIA), deethylatrazine (DEA), and diaminochlorotriazine (DACT), which collectively comprise the total chlorotriazines (TCT) as represented in this study, was modified to allow for scaling to humans. Changes included replacing the fixed dose-dependent oral uptake rates with a method that represented delayed absorption observed in rats administered ATZ as a bolus dose suspended in a methylcellulose vehicle. Rate constants for metabolism of ATZ to DIA and DEA, followed by metabolism of DIA and DEA to DACT were predicted using a compartmental model describing the metabolism of the chlorotriazines by rat and human hepatocytesin vitro Overall, the model successfully predicted both the 4-day plasma time-course data in rats administered ATZ by bolus dose (3, 10, and 50 mg/kg/day) or in the diet (30, 100, or 500 ppm). Simulated continuous daily exposure of a 55-kg adult female to ATZ at a dose of 1.0 µg/kg/day resulted in steady-state urinary concentrations of 0.6, 1.4, 2.5, and 6.0 µg/L for DEA, DIA, DACT, and TCT, respectively. The TCT (ATZ + DEA + DIA + DACT) human urinary biomonitoring equivalent concentration following continuous exposure to ATZ at the chronic point of departure (POD = 1.8 mg/kg/day) was 360.6 µg/L.


Assuntos
Atrazina/farmacocinética , Hepatócitos/metabolismo , Modelos Biológicos , Triazinas/farmacocinética , Absorção Fisiológica , Administração Oral , Animais , Atrazina/sangue , Atrazina/urina , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Hepatócitos/efeitos dos fármacos , Humanos , Ratos Sprague-Dawley , Especificidade da Espécie , Distribuição Tecidual , Triazinas/sangue , Triazinas/urina
19.
Toxicol Sci ; 150(2): 269-82, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26794141

RESUMO

The risk of human exposure to total chlorotriazines (TCT) in drinking water was evaluated using a physiologically based pharmacokinetic (PBPK) model. Daily TCT (atrazine, deethylatrazine, deisopropylatrazine, and diaminochlorotriazine) chemographs were constructed for 17 frequently monitored community water systems (CWSs) using linear interpolation and Krieg estimates between observed TCT values. Synthetic chemographs were created using a conservative bias factor of 3 to generate intervening peaks between measured values. Drinking water consumption records from 24-h diaries were used to calculate daily exposure. Plasma TCT concentrations were updated every 30 minutes using the PBPK model output for each simulated calendar year from 2006 to 2010. Margins of exposure (MOEs) were calculated (MOE = [Human Plasma TCTPOD] ÷ [Human Plasma TCTEXP]) based on the toxicological point of departure (POD) and the drinking water-derived exposure to TCT. MOEs were determined based on 1, 2, 3, 4, 7, 14, 28, or 90 days of rolling average exposures and plasma TCT Cmax, or the area under the curve (AUC). Distributions of MOE were determined and the 99.9th percentile was used for risk assessment. MOEs for all 17 CWSs were >1000 at the 99.9(th)percentile. The 99.9(th)percentile of the MOE distribution was 2.8-fold less when the 3-fold synthetic chemograph bias factor was used. MOEs were insensitive to interpolation method, the consumer's age, the water consumption database used and the duration of time over which the rolling average plasma TCT was calculated, for up to 90 days. MOEs were sensitive to factors that modified the toxicological, or hyphenated appropriately no-observed-effects level (NOEL), including rat strain, endpoint used, method of calculating the NOEL, and the pharmacokinetics of elimination, as well as the magnitude of exposure (CWS, calendar year, and use of bias factors).


Assuntos
Atrazina/farmacocinética , Água Potável/química , Monitoramento Ambiental/métodos , Modelos Biológicos , Poluentes Químicos da Água/farmacocinética , Atrazina/análise , Atrazina/sangue , Atrazina/toxicidade , Água Potável/normas , Monitoramento Ambiental/estatística & dados numéricos , Humanos , Probabilidade , Medição de Risco , Poluentes Químicos da Água/análise , Poluentes Químicos da Água/sangue , Poluentes Químicos da Água/toxicidade
20.
Toxicology ; 332: 67-76, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25839974

RESUMO

Parabens have been reported as potential endocrine disrupters and are widely used in consumer projects including cosmetics, foods and pharmaceuticals. We report on the development of a PBPK model for methyl-, propyl-, and butylparaben. The model was parameterized through a combination of QSAR for tissue solubility and quantitative in vitro to in vivo extrapolation (IVIVE) for hydrolysis in portals of entry including intestine and skin as well as in the primary site of metabolism, the liver. Overall, the model provided very good agreement with published time-course data in blood and urine from controlled dosing studies in rat and human, and demonstrates the potential value of quantitative IVIVE in expanding the use of human biomonitoring data in safety assessment. An in vitro based cumulative margin of safety (MOS) was calculated by comparing the effective concentrations from an in vitro assay of estrogenicity to the free paraben concentrations predicted by the model to be associated with the 95th percentile urine concentrations reported in NHANES (2009-2010 collection period). The calculated MOS for adult females was 108, whereas the MOS for males was 444.


Assuntos
Disruptores Endócrinos/farmacocinética , Disruptores Endócrinos/toxicidade , Poluentes Ambientais/farmacocinética , Modelos Biológicos , Parabenos/farmacocinética , Parabenos/toxicidade , Testes de Toxicidade/métodos , Adulto , Animais , Biotransformação , Células Cultivadas , Disruptores Endócrinos/sangue , Disruptores Endócrinos/química , Poluentes Ambientais/sangue , Poluentes Ambientais/química , Poluentes Ambientais/toxicidade , Feminino , Genes Reporter , Humanos , Hidrólise , Modelos Lineares , Masculino , Inquéritos Nutricionais , Parabenos/química , Ratos Sprague-Dawley , Ratos Wistar , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Medição de Risco , Fatores de Risco , Solubilidade , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...