Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Planta Med ; 90(3): 219-242, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38198805

RESUMO

In September 2022, the 3rd International Workshop on pyrrolizidine alkaloids (PAs) and related phytotoxins was held on-line, entitled 'Toxins in botanical drugs and plant-derived food and feed - from science to regulation'. The workshop focused on new findings about the occurrence, exposure, toxicity, and risk assessment of PAs. In addition, new scientific results related to the risk assessment of alkenylbenzenes, a distinct class of herbal constituents, were presented. The presence of PAs and alkenylbenzenes in plant-derived food, feed, and herbal medicines has raised health concerns with respect to their acute and chronic toxicity but mainly related to the genotoxic and carcinogenic properties of several congeners. The compounds are natural constituents of a variety of plant families and species widely used in medicinal, food, and feed products. Their individual occurrence, levels, and toxic properties, together with the broad range of congeners present in nature, represent a striking challenge to modern toxicology. This review tries to provide an overview of the current knowledge on these compounds and indicates needs and perspectives for future research.


Assuntos
Plantas Medicinais , Alcaloides de Pirrolizidina , Alcaloides de Pirrolizidina/toxicidade
2.
Arch Toxicol ; 97(3): 685-696, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36436016

RESUMO

Poly- and perfluoroalkyl substances (PFASs) are omnipresent in the environment and have been shown to accumulate in humans. Most PFASs are not biotransformed in animals and humans, so that elimination is largely dependent on non-metabolic clearance via bile and urine. Accumulation of certain PFASs in humans may relate to their reabsorption from the pre-urine by transporter proteins in the proximal tubules of the kidney, such as URAT1 and OAT4. The present study assessed the in vitro transport of 7 PFASs (PFHpA, PFOA, PFNA, PFDA, PFBS, PFHxS and PFOS) applying URAT1- or OAT4-transfected human embryonic kidney (HEK) cells. Virtually no transport of PFASs could be measured in URAT1-transfected HEK cells. All PFASs, except PFBS, showed clear uptake in OAT4-transfected HEK cells. In addition, these in vitro results were further supported by in silico docking and molecular dynamic simulation studies assessing transporter-ligand interactions. Information on OAT4-mediated transport may provide insight into the accumulation potential of PFASs in humans, but other kinetic aspects may play a role and should also be taken into account. Quantitative information on all relevant kinetic processes should be integrated in physiologically based kinetic (PBK) models, to predict congener-specific accumulation of PFASs in humans in a more accurate manner.


Assuntos
Ácidos Alcanossulfônicos , Fluorocarbonos , Transportadores de Ânions Orgânicos , Animais , Humanos , Rim/metabolismo , Túbulos Renais Proximais/metabolismo , Proteínas de Transporte/metabolismo , Fluorocarbonos/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Ácidos Alcanossulfônicos/metabolismo
3.
Arch Toxicol ; 96(12): 3407-3419, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36063173

RESUMO

With an increasing need to incorporate new approach methodologies (NAMs) in chemical risk assessment and the concomitant need to phase out animal testing, the interpretation of in vitro assay readouts for quantitative hazard characterisation becomes more important. Physiologically based kinetic (PBK) models, which simulate the fate of chemicals in tissues of the body, play an essential role in extrapolating in vitro effect concentrations to in vivo bioequivalent exposures. As PBK-based testing approaches evolve, it will become essential to standardise PBK modelling approaches towards a consensus approach that can be used in quantitative in vitro-to-in vivo extrapolation (QIVIVE) studies for regulatory chemical risk assessment based on in vitro assays. Based on results of an ECETOC expert workshop, steps are recommended that can improve regulatory adoption: (1) define context and implementation, taking into consideration model complexity for building fit-for-purpose PBK models, (2) harmonise physiological input parameters and their distribution and define criteria for quality chemical-specific parameters, especially in the absence of in vivo data, (3) apply Good Modelling Practices (GMP) to achieve transparency and design a stepwise approach for PBK model development for risk assessors, (4) evaluate model predictions using alternatives to in vivo PK data including read-across approaches, (5) use case studies to facilitate discussions between modellers and regulators of chemical risk assessment. Proof-of-concepts of generic PBK modelling approaches are published in the scientific literature at an increasing rate. Working on the previously proposed steps is, therefore, needed to gain confidence in PBK modelling approaches for regulatory use.


Assuntos
Modelos Biológicos , Animais , Cinética , Medição de Risco/métodos
4.
Chem Res Toxicol ; 34(2): 460-472, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33382582

RESUMO

The goal of the present study was to develop an online web-based toolbox that contains generic physiologically based kinetic (PBK) models for rats and humans, including underlying calculation tools to predict plasma protein binding and tissue:plasma distribution, to be used for quantitative in-vitro-to-in-vivo extrapolations (QIVIVE). The PBK models within the toolbox allow first estimations of internal plasma and tissue concentrations of chemicals to be made, based on the logP and pKa of the chemicals and values for intestinal uptake and intrinsic hepatic clearance. As a case study, the toolbox was used to predict oral equivalent doses of in vitro ToxCast bioactivity data for the food additives methylparaben, propyl gallate, octyl gallate, and dodecyl gallate. These oral equivalent doses were subsequently compared with human exposure estimates, as a low tier assessment allowing prioritization for further assessment. The results revealed that daily intake levels of especially propyl gallate can lead to internal plasma concentrations that are close to in vitro biological effect concentrations, particularly with respect to the inhibition of human thyroid peroxidase (TPO). Estrogenic effects were not considered likely to be induced by the food additives, as daily exposure levels of the different compounds remained 2 orders of magnitude below the oral equivalent doses for in vitro estrogen receptor activation. Overall, the results of the study show how the toolbox, which is freely accessible through www.qivivetools.wur.nl, can be used to obtain initial internal dose estimates of chemicals and to prioritize chemicals for further assessment, based on the comparison of oral equivalent doses of in vitro biological activity data with human exposure levels.


Assuntos
Aditivos Alimentares/análise , Ensaios de Triagem em Larga Escala , Internet , Testes de Toxicidade , Animais , Aditivos Alimentares/administração & dosagem , Humanos , Cinética
5.
Chem Res Toxicol ; 34(2): 300-312, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33253545

RESUMO

The intention of this study was to determine the utility of high-throughput screening (HTS) data, as exemplified by ToxCast and Tox21, for application in toxicological read-across in food-relevant chemicals. Key questions were addressed on the extent to which the HTS data could provide information enabling (1) the elucidation of underlying bioactivities associated with apical toxicological outcomes, (2) the closing of existing toxicological data gaps, and (3) the definition of the boundaries of chemical space across which bioactivity could reliably be extrapolated. Results revealed that many biological targets apparently activated within the chemical groupings lack, at this time, validated toxicity pathway associations. Therefore, as means of providing proof-of-principle, a comparatively well-characterized end point-estrogenicity-was selected for evaluation. This was facilitated through the preparation of two exploratory case studies, focusing upon groupings of paraben-gallates and pyranone-type compounds (notably flavonoids). Within both, the HTS data were seen to reflect estrogenic potencies in a manner which broadly corresponded to established structure-activity group relationships, with parabens and flavonoids displaying greater estrogen receptor affinity than benzoate esters and alternative pyranone-containing molecules, respectively. As such, utility in the identification of out-of-domain compounds was demonstrated, indicating potential for application in addressing point (3) as detailed above.


Assuntos
Flavonoides/efeitos adversos , Ensaios de Triagem em Larga Escala , Piranos/efeitos adversos , Testes de Toxicidade , Humanos , Estrutura Molecular , Medição de Risco , Relação Estrutura-Atividade
6.
J Appl Toxicol ; 40(12): 1647-1660, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33034907

RESUMO

Aristolochic acid I (AAI) is a well-known genotoxic kidney carcinogen. Metabolic conversion of AAI into the DNA-reactive aristolactam-nitrenium ion is involved in the mode of action of tumor formation. This study aims to predict in vivo AAI-DNA adduct formation in the kidney of rat, mouse and human by translating the in vitro concentration-response curves for AAI-DNA adduct formation to the in vivo situation using physiologically based kinetic (PBK) modeling-based reverse dosimetry. DNA adduct formation in kidney proximal tubular LLC-PK1 cells exposed to AAI was quantified by liquid chromatography-electrospray ionization-tandem mass spectrometry. Subsequently, the in vitro concentration-response curves were converted to predicted in vivo dose-response curves in rat, mouse and human kidney using PBK models. Results obtained revealed a dose-dependent increase in AAI-DNA adduct formation in the rat, mouse and human kidney and the predicted DNA adduct levels were generally within an order of magnitude compared with values reported in the literature. It is concluded that the combined in vitro PBK modeling approach provides a novel way to define in vivo dose-response curves for kidney DNA adduct formation in rat, mouse and human and contributes to the reduction, refinement and replacement of animal testing.


Assuntos
Ácidos Aristolóquicos/toxicidade , Adutos de DNA/metabolismo , Rim/efeitos dos fármacos , Modelos Biológicos , Alternativas aos Testes com Animais , Animais , Cromatografia Líquida , Relação Dose-Resposta a Droga , Humanos , Rim/metabolismo , Rim/patologia , Células LLC-PK1 , Camundongos , Ratos , Espectrometria de Massas por Ionização por Electrospray , Suínos , Espectrometria de Massas em Tandem , Toxicocinética
7.
Arch Toxicol ; 93(7): 1941-1953, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31111190

RESUMO

The goal of the present study was to obtain an in vivo relevant prioritization method for the endocrine potencies of different polycarbonate monomers, by combining in vitro bioassay data with physiologically based kinetic (PBK) modelling. PBK models were developed for a selection of monomers, including bisphenol A (BPA), two bisphenol F (BPF) isomers and four different bisphenol A diglycidyl ethers (BADGEs), using in vitro input data. With these models, the plasma concentrations of the compounds were simulated, providing means to estimate the dose levels at which the in vitro endocrine effect concentrations are reached. The results revealed that, whereas the in vitro relative potencies of different BADGEs (predominantly anti-androgenic effects) can be up to fourfold higher than BPA, the estimated in vivo potencies based on the oral equivalent doses are one to two orders of magnitude lower than BPA because of fast detoxification of the BADGEs. In contrast, the relative potencies of 2,2-BPF and 4,4-BPF increase when accounting for the in vivo availability. 4,4-BPF is estimated to be fivefold more potent than BPA in humans in vivo in inducing estrogenic effects and both 2,2-BPF and 4,4-BPF are estimated to be, respectively, 7 and 11-fold more potent in inducing anti-androgenic effects. These relative potencies were considered to be first-tier estimates, particularly given that the potential influence of intestinal metabolism on the in vivo availability was not accounted for. Overall, it can be concluded that both 2,2-BPF and 4,4-BPF are priority compounds.


Assuntos
Compostos Benzidrílicos/administração & dosagem , Compostos de Epóxi/administração & dosagem , Modelos Biológicos , Fenóis/administração & dosagem , Antagonistas de Androgênios/administração & dosagem , Antagonistas de Androgênios/farmacocinética , Antagonistas de Androgênios/toxicidade , Compostos Benzidrílicos/farmacocinética , Compostos Benzidrílicos/toxicidade , Células CACO-2 , Linhagem Celular , Relação Dose-Resposta a Droga , Compostos de Epóxi/farmacocinética , Compostos de Epóxi/toxicidade , Estrogênios/administração & dosagem , Estrogênios/farmacocinética , Estrogênios/toxicidade , Humanos , Fenóis/farmacocinética , Fenóis/toxicidade
8.
Chem Res Toxicol ; 31(5): 285-286, 2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29641185

RESUMO

Quantitative predictions of in vivo chemical levels based on in vitro data will become a cornerstone of next generation nonanimal risk evaluations. Both regulatory and scientific experience with quantitative toxicokinetics must increase now for this transition to happen.


Assuntos
Alternativas aos Testes com Animais , Testes de Toxicidade , Toxicocinética , Animais , Humanos , Medição de Risco
9.
J Nanobiotechnology ; 16(1): 70, 2018 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-30219059

RESUMO

BACKGROUND: Upon ingestion, nanoparticles can interact with the intestinal epithelial barrier potentially resulting in systemic uptake of nanoparticles. Nanoparticle properties have been described to influence the protein corona formation and subsequent cellular adhesion, uptake and transport. Here, we aimed to study the effects of nanoparticle size and surface chemistry on the protein corona formation and subsequent cellular adhesion, uptake and transport. Caco-2 intestinal cells, were exposed to negatively charged polystyrene nanoparticles (PSNPs) (50 and 200 nm), functionalized with sulfone or carboxyl groups, at nine nominal concentrations (15-250 µg/ml) for 10 up to 120 min. The protein coronas were analysed by LC-MS/MS. RESULTS: Subtle differences in the protein composition of the two PSNPs with different surface chemistry were noted. High-content imaging analysis demonstrated that sulfone PSNPs were associated with the cells to a significantly higher extent than the other PSNPs. The apparent cellular adhesion and uptake of 200 nm PSNPs was not significantly increased compared to 50 nm PSNPs with the same surface charge and chemistry. Surface chemistry outweighs the impact of size on the observed PSNP cellular associations. Also transport of the sulfone PSNPs through the monolayer of cells was significantly higher than that of carboxyl PSNPs. CONCLUSIONS: The results suggest that the composition of the protein corona and the PSNP surface chemistry influences cellular adhesion, uptake and monolayer transport, which might be predictive of the intestinal transport potency of NPs.


Assuntos
Mucosa Intestinal/metabolismo , Nanopartículas/metabolismo , Poliestirenos/metabolismo , Coroa de Proteína/análise , Coroa de Proteína/metabolismo , Transporte Biológico , Células CACO-2 , Adesão Celular , Sobrevivência Celular , Humanos , Nanopartículas/química , Nanopartículas/ultraestrutura , Tamanho da Partícula , Poliestirenos/química , Eletricidade Estática , Propriedades de Superfície , Espectrometria de Massas em Tandem
10.
Regul Toxicol Pharmacol ; 92: 145-151, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29196030

RESUMO

Inclusion of alternative methods that replace, reduce, or refine (3R) animal testing within regulatory safety evaluations of chemicals generally faces many hurdles. The goal of the current work is to i) collect responses from key stakeholders involved in food safety evaluations on what they consider the most relevant factors that influence the acceptance and use of 3R methods and to ii) use these responses to formulate activities needed to increase the acceptance and use of 3R methods, particularly for kinetics. The stakeholders were contacted by e-mail for their opinions, asking the respondents to write down three barriers and/or drivers and scoring these by distributing 5 points over the three factors. The main barriers that obtained the highest aggregated scores were i) uncertain predictability 3R methods/lack of validation, ii) insufficient guidance regulators/industry and iii) insufficient harmonization of legislation. The major driver identified was the possibility of 3R methods to provide more mechanistic information. Based on the results, recommendations are given to enhance the acceptance and application of 3R toxicokinetic methods in food safety evaluations. These include steering of regulatory data requirements as well as creating (funding) opportunities for development and validation of alternative methods for kinetics and development of guidances.


Assuntos
Alternativas aos Testes com Animais/métodos , Alternativas aos Testes com Animais/normas , Prova Pericial/normas , Inocuidade dos Alimentos/métodos , Animais , Humanos , Cinética , Testes de Toxicidade/métodos , Testes de Toxicidade/normas
11.
Arch Toxicol ; 91(5): 2119-2133, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27815601

RESUMO

Toxicity outcomes derived in vitro do not always reflect in vivo toxicity values, which was previously observed for a series of phenols tested in the embryonic stem cell test (EST). Translation of in vitro data to the in vivo situation is therefore an important, but still limiting step for the use of in vitro toxicity outcomes in the safety assessment of chemicals. The aim of the present study was to translate in vitro embryotoxicity data for a series of phenols to in vivo developmental toxic potency values for the rat by physiologically based kinetic (PBK) modelling-based reverse dosimetry. To this purpose, PBK models were developed for each of the phenols. The models were parameterised with in vitro-derived values defining metabolism and transport of the compounds across the intestinal and placental barrier and with in silico predictions and data from the literature. Using PBK-based reverse dosimetry, in vitro concentration-response curves from the EST were translated into in vivo dose-response curves from which points of departure (PoDs) were derived. The predicted PoDs differed less than 3.6-fold from PoDs derived from in vivo toxicity data for the phenols available in the literature. Moreover, the in vitro PBK-based reverse dosimetry approach could overcome the large disparity that was observed previously between the in vitro and the in vivo relative potency of the series of phenols. In conclusion, this study shows another proof-of-principle that the in vitro PBK approach is a promising strategy for non-animal-based safety assessment of chemicals.


Assuntos
Relação Dose-Resposta a Droga , Desenvolvimento Embrionário/efeitos dos fármacos , Modelos Teóricos , Fenóis/toxicidade , Animais , Células CACO-2 , Simulação por Computador , Células-Tronco Embrionárias/efeitos dos fármacos , Feminino , Humanos , Intestinos/efeitos dos fármacos , Camundongos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Fenóis/administração & dosagem , Fenóis/farmacocinética , Placenta/efeitos dos fármacos , Gravidez , Ratos
12.
Chem Res Toxicol ; 29(4): 659-68, 2016 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-26952143

RESUMO

Estragole is a known hepatocarcinogen in rodents at high doses following metabolic conversion to the DNA-reactive metabolite 1'-sulfooxyestragole. The aim of the present study was to model possible levels of DNA adduct formation in (individual) humans upon exposure to estragole. This was done by extending a previously defined PBK model for estragole in humans to include (i) new data on interindividual variation in the kinetics for the major PBK model parameters influencing the formation of 1'-sulfooxyestragole, (ii) an equation describing the relationship between 1'-sulfooxyestragole and DNA adduct formation, (iii) Monte Carlo modeling to simulate interindividual human variation in DNA adduct formation in the population, and (iv) a comparison of the predictions made to human data on DNA adduct formation for the related alkenylbenzene methyleugenol. Adequate model predictions could be made, with the predicted DNA adduct levels at the estimated daily intake of estragole of 0.01 mg/kg bw ranging between 1.6 and 8.8 adducts in 10(8) nucleotides (nts) (50th and 99th percentiles, respectively). This is somewhat lower than values reported in the literature for the related alkenylbenzene methyleugenol in surgical human liver samples. The predicted levels seem to be below DNA adduct levels that are linked with tumor formation by alkenylbenzenes in rodents, which were estimated to amount to 188-500 adducts per 10(8) nts at the BMD10 values of estragole and methyleugenol. Although this does not seem to point to a significant health concern for human dietary exposure, drawing firm conclusions may have to await further validation of the model's predictions.


Assuntos
Anisóis/metabolismo , Carcinógenos/metabolismo , Adutos de DNA/metabolismo , Fígado/metabolismo , Sulfonas/metabolismo , Adolescente , Adulto , Idoso , Derivados de Alilbenzenos , Pré-Escolar , Simulação por Computador , Feminino , Humanos , Lactente , Cinética , Fígado/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Método de Monte Carlo , NAD/metabolismo , Oxirredução , Adulto Jovem
13.
Toxicol Appl Pharmacol ; 283(2): 117-26, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25549870

RESUMO

The present study aims at predicting the level of formation of the ultimate carcinogenic metabolite of methyleugenol, 1'-sulfooxymethyleugenol, in the human population by taking variability in key bioactivation and detoxification reactions into account using Monte Carlo simulations. Depending on the metabolic route, variation was simulated based on kinetic constants obtained from incubations with a range of individual human liver fractions or by combining kinetic constants obtained for specific isoenzymes with literature reported human variation in the activity of these enzymes. The results of the study indicate that formation of 1'-sulfooxymethyleugenol is predominantly affected by variation in i) P450 1A2-catalyzed bioactivation of methyleugenol to 1'-hydroxymethyleugenol, ii) P450 2B6-catalyzed epoxidation of methyleugenol, iii) the apparent kinetic constants for oxidation of 1'-hydroxymethyleugenol, and iv) the apparent kinetic constants for sulfation of 1'-hydroxymethyleugenol. Based on the Monte Carlo simulations a so-called chemical-specific adjustment factor (CSAF) for intraspecies variation could be derived by dividing different percentiles by the 50th percentile of the predicted population distribution for 1'-sulfooxymethyleugenol formation. The obtained CSAF value at the 90th percentile was 3.2, indicating that the default uncertainty factor of 3.16 for human variability in kinetics may adequately cover the variation within 90% of the population. Covering 99% of the population requires a larger uncertainty factor of 6.4. In conclusion, the results showed that adequate predictions on interindividual human variation can be made with Monte Carlo-based PBK modeling. For methyleugenol this variation was observed to be in line with the default variation generally assumed in risk assessment.


Assuntos
Carcinógenos/farmacocinética , Eugenol/análogos & derivados , Modelos Biológicos , Método de Monte Carlo , Carcinógenos/toxicidade , Sistema Enzimático do Citocromo P-450/metabolismo , Relação Dose-Resposta a Droga , Avaliação de Medicamentos/métodos , Eugenol/farmacocinética , Eugenol/toxicidade , Humanos , Cinética , Redes e Vias Metabólicas/efeitos dos fármacos , Redes e Vias Metabólicas/fisiologia
14.
Arch Toxicol ; 87(9): 1709-23, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23943240

RESUMO

In vitro assays are often used for the hazard characterisation of compounds, but their application for quantitative risk assessment purposes is limited. This is because in vitro assays cannot provide a complete in vivo dose-response curve from which a point of departure (PoD) for risk assessment can be derived, like the no observed adverse effect level (NOAEL) or the 95 % lower confidence limit of the benchmark dose (BMDL). To overcome this constraint, the present study combined in vitro data with a physiologically based kinetic (PBK) model applying reverse dosimetry. To this end, embryotoxicity of phenol was evaluated in vitro using the embryonic stem cell test (EST), revealing a concentration-dependent inhibition of differentiation into beating cardiomyocytes. In addition, a PBK model was developed on the basis of in vitro and in silico data and data available from the literature only. After evaluating the PBK model performance, effective concentrations (ECx) obtained with the EST served as an input for in vivo plasma concentrations in the PBK model. Applying PBK-based reverse dosimetry provided in vivo external effective dose levels (EDx) from which an in vivo dose-response curve and a PoD for risk assessment were derived. The predicted PoD lies within the variation of the NOAELs obtained from in vivo developmental toxicity data from the literature. In conclusion, the present study showed that it was possible to accurately predict a PoD for the risk assessment of phenol using in vitro toxicity data combined with reverse PBK modelling.


Assuntos
Alternativas ao Uso de Animais , Desinfetantes/toxicidade , Células-Tronco Embrionárias/efeitos dos fármacos , Modelos Biológicos , Fenol/toxicidade , Teratogênicos/toxicidade , Animais , Biotransformação , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Citosol/enzimologia , Citosol/metabolismo , Desinfetantes/metabolismo , Desinfetantes/farmacocinética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Humanos , Cinética , Fígado/enzimologia , Fígado/metabolismo , Masculino , Camundongos , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Especificidade de Órgãos , Fenol/metabolismo , Fenol/farmacocinética , Ratos , Medição de Risco/métodos , Especificidade da Espécie , Teratogênicos/metabolismo , Teratogênicos/farmacocinética
15.
ALTEX ; 40(2): 237­247, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35901496

RESUMO

In vitro toxicokinetic data are critical in meeting an increased regulatory need to improve chemical safety evaluations towards a better understanding of internal human chemical exposure and toxicity. In vitro intrinsic hepatic clearance (CLint), the fraction unbound in plasma (fup), and the intestinal apparent permeability (Papp) are important parameters as input in a physiologically based kinetic (PBK) model to make first estimates of internal exposure after oral dosing. In the present study we explored the experimental variation in the values for these parameters as reported in the literature. Furthermore, the impact that this experimental variation has on PBK model predictions of maximum plasma concentration (Cmax) and the area under the concentration time curve (AUC0-24h) was determined. As a result of the experimental variation in CLint, Papp, and fup, the predicted variation in Cmax for individual compounds ranged between 1.4- to 28-fold, and the predicted variation in AUC0-24h ranged between 1.4- and 23-fold. These results indicate that there are still some important steps to take to achieve robust data that can be used in regulatory applications. To gain regulatory acceptance of in vitro kinetic data and PBK models based on in vitro input data, the boundaries in experimental conditions as well as the applicability domain and the use of different in vitro kinetic models need to be described in guidance documents.


Assuntos
Fígado , Modelos Biológicos , Humanos , Intestinos , Cinética
16.
Toxins (Basel) ; 15(2)2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36828409

RESUMO

Safrole, a 162.2 Da natural compound belonging to the alkenylbenzenes class, is classified as a possible carcinogen to humans by IARC (group IIB) and has proven to be genotoxic and carcinogenic to rodents. Despite its use as a food or feed additive, it is forbidden in many countries due to its documented toxicity; yet, it is still broadly present within food and feed and is particularly abundant in spices, herbs and essential oils. Specifically, safrole may exert its toxicity upon bioactivation to its proximate carcinogen 1'-hydroxy-safrole via specific members of the cytochrome P450 protein family with a certain inter/intra-species variability. To investigate this variability, an in-silico workflow based on molecular modelling, docking and molecular dynamics has been successfully applied. This work highlighted the mechanistic basis underpinning differences among humans, cats, chickens, goats, sheep, dogs, mice, pigs, rats and rabbits. The chosen metric to estimate the likeliness of formation of 1'-hydroxy-safrole by the species-specific cytochrome P450 under investigation allowed for the provision of a knowledge-based ground to rationally design and prioritise further experiments and deepen the current understanding of alkenylbenzenes bioactivation and CYPs mechanics. Both are crucial for a more informed framework of analysis for safrole toxicity.


Assuntos
Derivados de Alilbenzenos , Safrol , Ratos , Animais , Camundongos , Humanos , Cães , Coelhos , Ovinos , Suínos , Safrol/metabolismo , Galinhas/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Carcinógenos/metabolismo
17.
Artigo em Inglês | MEDLINE | ID: mdl-36395382

RESUMO

Calibration of a kinetic model for the transfer of PCDD/Fs and dl-PCBs from feed to the hen's body and eggs was thus far restricted to the total TEQ concentration, i.e. the summed concentrations of PCDD/Fs and dl-PCBs expressed in terms of equivalents of 2,3,7,8-TCDD. However, this approach may lead to over- or underestimation of the transfer if the mixture contains congeners with kinetic characteristics which differ considerably from those used in such a model. This paper extends a previous transfer model of PCDD/Fs and dl-PCBs from feed to egg yolk fat and abdominal fat of high production laying hens, based on the total TEQ approach, to the level of individual congeners. Both modelling approaches are compared and the new approach is presented as a webtool application. This congener-specific approach enabled the calibration of 25 of the 29 relevant PCDD/F and dl-PCB congeners with respect to their individual transfer characteristics to body fat and egg yolk fat and their clearance from the body. Limitations of the available experimental data prevented the calibration of 1,2,3,4,6,7,8-HpCDD, OCDD, OCDF and PCB 123. The fraction transferred to egg yolk fat after long-term daily intake of contaminated feed was found to be at least 0.78 for 2,3,7,8-TCDD, 0.75 for PeCDD, 0.42-0.61 for HxCDDs, 0.70 for 2,3,7,8-TCDF, 0.71 for PeCDF, 0.54-0.60 for HxCDFs, 0.18-0.24 for HpCDFs and 0.89-1.00 for dl-PCBs. Various experimental and feed incident mixtures were used to compare the total TEQ- model with the congener-specific approach. An overestimation of the transfer by the total TEQ method was shown in particular for mixtures with a substantial contribution of hexa-, hepta- and octa-PCDD/Fs to the total TEQ level.


Assuntos
Benzofuranos , Dioxinas , Bifenilos Policlorados , Dibenzodioxinas Policloradas , Feminino , Animais , Dibenzofuranos , Galinhas , Dibenzofuranos Policlorados
18.
Toxicol Appl Pharmacol ; 260(3): 271-84, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22445790

RESUMO

This study defines a physiologically based kinetic (PBK) model for methyleugenol (ME) in human based on in vitro and in silico derived parameters. With the model obtained, bioactivation and detoxification of methyleugenol (ME) at different doses levels could be investigated. The outcomes of the current model were compared with those of a previously developed PBK model for methyleugenol (ME) in male rat. The results obtained reveal that formation of 1'-hydroxymethyleugenol glucuronide (1'HMEG), a major metabolic pathway in male rat liver, appears to represent a minor metabolic pathway in human liver whereas in human liver a significantly higher formation of 1'-oxomethyleugenol (1'OME) compared with male rat liver is observed. Furthermore, formation of 1'-sulfooxymethyleugenol (1'HMES), which readily undergoes desulfonation to a reactive carbonium ion (CA) that can form DNA or protein adducts (DA), is predicted to be the same in the liver of both human and male rat at oral doses of 0.0034 and 300 mg/kg bw. Altogether despite a significant difference in especially the metabolic pathways of the proximate carcinogenic metabolite 1'-hydroxymethyleugenol (1'HME) between human and male rat, the influence of species differences on the ultimate overall bioactivation of methyleugenol (ME) to 1'-sulfooxymethyleugenol (1'HMES) appears to be negligible. Moreover, the PBK model predicted the formation of 1'-sulfooxymethyleugenol (1'HMES) in the liver of human and rat to be linear from doses as high as the benchmark dose (BMD10) down to as low as the virtual safe dose (VSD). This study shows that kinetic data do not provide a reason to argue against linear extrapolation from the rat tumor data to the human situation.


Assuntos
Simulação por Computador , Eugenol/análogos & derivados , Microssomos Hepáticos/metabolismo , Modelos Biológicos , Administração Oral , Animais , Adutos de DNA/metabolismo , Relação Dose-Resposta a Droga , Eugenol/administração & dosagem , Eugenol/farmacocinética , Eugenol/toxicidade , Feminino , Humanos , Masculino , Ratos , Especificidade da Espécie
19.
Chem Res Toxicol ; 25(12): 2630-41, 2012 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-22978292

RESUMO

trans-2-Hexenal (2-hexenal) is an α,ß-unsaturated aldehyde that occurs naturally in a wide range of fruits, vegetables, and spices. 2-Hexenal as well as other α,ß-unsaturated aldehydes that are natural food constituents or flavoring agents may raise a concern for genotoxicity due to the ability of the α,ß-unsaturated aldehyde moiety to react with DNA. Controversy remains, however, on whether α,ß-unsaturated aldehydes result in significant DNA adduct formation in vivo at realistic dietary exposure. In this study, a rat physiologically based in silico model was developed for 2-hexenal as a model compound to examine the time- and dose-dependent detoxification and DNA adduct formation of this selected α,ß-unsaturated aldehyde. The model was developed based on in vitro and literature-derived parameters, and its adequacy was evaluated by comparing predicted DNA adduct formation in the liver of rats exposed to 2-hexenal with reported in vivo data. The model revealed that at an exposure level of 0.04 mg/kg body weight, a value reflecting estimated daily human dietary intake, 2-hexenal is rapidly detoxified predominantly by conjugation with glutathione (GSH) by glutathione S-transferases. At higher dose levels, depletion of GSH results in a shift to 2-hexenal oxidation and reduction as the major pathways for detoxification. The level of DNA adduct formation at current levels of human dietary intake was predicted to be more than 3 orders of magnitude lower than endogenous DNA adduct levels. These results support that rapid detoxification of 2-hexenal reduces the risk arising from 2-hexenal exposure and that at current dietary exposure levels, DNA adduct formation is negligible.


Assuntos
Aldeídos/farmacocinética , Adutos de DNA , Aromatizantes/farmacocinética , Modelos Biológicos , Aldeído Desidrogenase/metabolismo , Aldeídos/toxicidade , Animais , Simulação por Computador , Reparo do DNA , Aromatizantes/toxicidade , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Inativação Metabólica , Intestino Delgado/metabolismo , Masculino , Mitocôndrias Hepáticas/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Wistar
20.
Chem Res Toxicol ; 25(11): 2352-67, 2012 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-22992039

RESUMO

The present study describes physiologically based kinetic (PBK) models for the alkenylbenzene elemicin (3,4,5-trimethoxyallylbenzene) in rat and human, based on the PBK models previously developed for the structurally related alkenylbenzenes estragole, methyleugenol, and safrole. Using the newly developed models, the level of metabolic activation of elemicin in rat and human was predicted to obtain insight in species differences in the bioactivation of elemicin and read across to the other methoxy allylbenzenes, estragole and methyleugenol. Results reveal that the differences between rat and human in the formation of the proximate carcinogenic metabolite 1'-hydroxyelemicin and the ultimate carcinogenic metabolite 1'-sulfoxyelemicin are limited (<3.8-fold). In addition, a comparison was made between the relative importance of bioactivation for elemicin and that of estragole and methyleugenol. Model predictions indicate that compound differences in the formation of the 1'-sulfoxymetabolites are limited (<11-fold) in rat and human liver. The insights thus obtained were used to perform a risk assessment for elemicin using the margin of exposure (MOE) approach and read across to the other methoxy allylbenzene derivatives for which in vivo animal tumor data are available. This reveals that elemicin poses a lower priority for risk management as compared to its structurally related analogues estragole and methyleugenol. Altogether, the results obtained indicate that PBK modeling provides an important insight in the occurrence of species differences in the metabolic activation of elemicin. Moreover, they provide an example of how PBK modeling can facilitate a read across in risk assessment from compounds for which in vivo toxicity studies are available to a compound for which only limited toxicity data have been described, thus contributing to the development of alternatives for animal testing.


Assuntos
Modelos Biológicos , Pirogalol/análogos & derivados , Animais , Humanos , Cinética , Masculino , Microssomos/química , Microssomos/metabolismo , Estrutura Molecular , Pirogalol/síntese química , Pirogalol/química , Pirogalol/metabolismo , Ratos , Ratos Sprague-Dawley , Medição de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA