Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Toxicol Sci ; 193(1): 1-17, 2023 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-36912747

RESUMO

Chronic inhalation of formaldehyde by F344 rats causes nasal squamous cell carcinoma (SCC). This outcome is well-characterized: including dose-response and time course data for SCC, mechanistic endpoints, and nasal dosimetry. Conolly et al. (Toxicol. Sci. 75, 432-447, 2003) used these resources to develop a biologically based dose-response (BBDR) model for SCC in F344 rats. This model, scaled up to humans, has informed dose-response conclusions reached by several international regulatory agencies. However, USEPA concluded that uncertainties precluded its use for cancer risk assessment. Here, we describe an updated BBDR model that addresses uncertainties through refined dosimetry modeling, revised analysis of labeling index data, and an extended dataset where both inhaled (exogenous) and endogenous formaldehyde (exogF, endoF) form DNA adducts. Further, since Conolly et al. (ibid) was published, it has become clear that, when controls from all F344 inhalation bioassays are considered, accounting for over 4000 rats, at most one nasal SCC occurred. This low spontaneous incidence constrains possible contribution of endoF to the formation of nasal SCC via DNA reactivity. Further, since both exogF and endoF form DNA adducts, this constraint also applies to exogF. The revised BBDR model therefore drives SCC formation through the cytotoxicity of high concentration exogF. An option for direct mutagenicity associated with DNA adducts is retained to allow estimation of an upper bound on adduct mutagenicity consistent with the lack of a spontaneous SCC incidence. These updates represent an iterative refinement of the 2003 model, incorporating new data and insights to reduce identified model uncertainties.


Assuntos
Carcinoma de Células Escamosas , Adutos de DNA , Ratos , Humanos , Animais , Ratos Endogâmicos F344 , Modelos Biológicos , Formaldeído/toxicidade , Nariz/patologia , Carcinoma de Células Escamosas/patologia
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.
Environ Toxicol Chem ; 42(1): 100-116, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36282016

RESUMO

To reduce the use of intact animals for chemical safety testing, while ensuring protection of ecosystems and human health, there is a demand for new approach methodologies (NAMs) that provide relevant scientific information at a quality equivalent to or better than traditional approaches. The present case study examined whether bioactivity and associated potency measured in an in vitro screening assay for aromatase inhibition could be used together with an adverse outcome pathway (AOP) and mechanistically based computational models to predict previously uncharacterized in vivo effects. Model simulations were used to inform designs of 60-h and 10-21-day in vivo exposures of adult fathead minnows (Pimephales promelas) to three or four test concentrations of the in vitro aromatase inhibitor imazalil ranging from 0.12 to 260 µg/L water. Consistent with an AOP linking aromatase inhibition to reproductive impairment in fish, exposure to the fungicide resulted in significant reductions in ex vivo production of 17ß-estradiol (E2) by ovary tissue (≥165 µg imazalil/L), plasma E2 concentrations (≥74 µg imazalil/L), vitellogenin (Vtg) messenger RNA expression (≥165 µg imazalil/L), Vtg plasma concentrations (≥74 µg imazalil/L), uptake of Vtg into oocytes (≥260 µg imazalil/L), and overall reproductive output in terms of cumulative fecundity, number of spawning events, and eggs per spawning event (≥24 µg imazalil/L). Despite many potential sources of uncertainty in potency and efficacy estimates based on model simulations, observed magnitudes of apical effects were quite consistent with model predictions, and in vivo potency was within an order of magnitude of that predicted based on in vitro relative potency. Overall, our study suggests that NAMs and AOP-based approaches can support meaningful reduction and refinement of animal testing. Environ Toxicol Chem 2023;42:100-116. © 2022 SETAC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.


Assuntos
Cyprinidae , Ovário , Humanos , Animais , Feminino , Aromatase/genética , Aromatase/metabolismo , Fadrozol/toxicidade , Ecotoxicologia , Ecossistema , Estradiol/metabolismo , Cyprinidae/fisiologia , Vitelogeninas/metabolismo
4.
Toxics ; 10(11)2022 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-36422908

RESUMO

Humans are exposed to persistent organic pollutants, such as dioxin-like compounds (DLCs), as mixtures. Understanding and predicting the toxicokinetics and thus internal burden of major constituents of a DLC mixture is important for assessing their contributions to health risks. PBPK models, including dioxin models, traditionally focus on one or a small number of compounds; developing new or extending existing models for mixtures often requires tedious, error-prone coding work. This lack of efficiency to scale up for multi-compound exposures is a major technical barrier toward large-scale mixture PBPK simulations. Congeners in the DLC family, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), share similar albeit quantitatively different toxicokinetic and toxicodynamic properties. Taking advantage of these similarities, here we reported the development of a human PBPK modeling framework for DLC mixtures that can flexibly accommodate an arbitrary number of congeners. Adapted from existing TCDD models, our mixture model contains the blood and three diffusion-limited compartments-liver, fat, and rest of the body. Depending on the number of congeners in a mixture, varying-length vectors of ordinary differential equations (ODEs) are automatically generated to track the tissue concentrations of the congeners. Shared ODEs are used to account for common variables, including the aryl hydrocarbon receptor (AHR) and CYP1A2, to which the congeners compete for binding. Binary and multi-congener mixture simulations showed that the AHR-mediated cross-induction of CYP1A2 accelerates the sequestration and metabolism of DLC congeners, resulting in consistently lower tissue burdens than in single exposure, except for the liver. Using dietary intake data to simulate lifetime exposures to DLC mixtures, the model demonstrated that the relative contributions of individual congeners to blood or tissue toxic equivalency (TEQ) values are markedly different than those to intake TEQ. In summary, we developed a mixture PBPK modeling framework for DLCs that may be utilized upon further improvement as a quantitative tool to estimate tissue dosimetry and health risks of DLC mixtures.

5.
Curr Opin Toxicol ; 16: 49-57, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31768481

RESUMO

As systems biology expands its multi-omic spectrum to increasing resolutions, distinguishing cells based on single-cell profiles becomes feasible. Unlike traditional bulk assays that average cellular responses and blur the distinct identities of responsive cells, single-cell technologies enable sensitive detection of small cellular changes and precise identification of those cells perturbed by toxicants. Among the suite of omic technologies that continue to expand and become affordable, single-cell RNA sequencing (scRNA-seq) is at the cutting edge and leading the way to transform systems toxicology. Single-cell systems toxicology can provide a wealth of information to elucidate cell-specific alterations and response trajectories, detect points-of-departure, map and develop dynamical models of toxicity pathways.

6.
Environ Sci Technol ; 53(18): 11002-11012, 2019 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-31436975

RESUMO

Exposure to environmental contaminants can lead to adverse outcomes in both human and nonhuman receptors. The Aggregate Exposure Pathway (AEP) and Adverse Outcome Pathway (AOP) frameworks can mechanistically inform cumulative risk assessment for human health and ecological end points by linking together environmental transport and transformation, external exposure, toxicokinetics, and toxicodynamics. This work presents a case study of a hypothetical contaminated site to demonstrate a quantitative approach for implementing the AEP framework and linking this framework to AOPs. We construct an AEP transport and transformation model and then quantify external exposure pathways for humans, fishes, and small herbivorous mammals at the hypothetical site. A Monte Carlo approach was used to address parameter variability. Source apportionment was quantified for each species, and published pharmacokinetic models were used to estimate internal target site exposure from external exposures. Published dose-response data for a multispecies AOP network were used to interpret AEP results in the context of species-specific effects. This work demonstrates (1) the construction, analysis, and application of a quantitative AEP model, (2) the utility of AEPs for organizing mechanistic exposure data and highlighting data gaps, and (3) the advantages provided by a source-to-outcome construct for leveraging exposure data and to aid transparency regarding assumptions.


Assuntos
Rotas de Resultados Adversos , Animais , Ecologia , Peixes , Humanos , Medição de Risco , Toxicocinética
7.
Toxicol Sci ; 172(1): 1-10, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31407013

RESUMO

Traditional methods for carcinogenicity testing rely heavily on the rodent bioassay as the standard for identification of tumorigenic risk. As such, identification of species-specific outcomes and/or metabolism are a frequent argument for regulatory exemption. One example is the association of tumor formation in the mouse lung after exposure to Cyp2F2 ligands. The adverse outcome pathway (AOP) framework offers a theoretical platform to address issues of species specificity that is consistent, transparent, and capable of integrating data from new approach methodologies as well as traditional data streams. A central premise of the AOP concept is that pathway progression from the molecular initiating event (MIE) implies a definable "response-response" (R-R) relationship between each key event (KE) that drives the pathway towards a specific adverse outcome (AO). This article describes an AOP for lung cancer in the mouse from an MIE of Cyp2F2-specific reactive metabolite formation, advancing through KE that include protein and/or nucleic acid adducts, diminished Club Cell 10 kDa (CC10) protein expression, hyperplasia of CC10 deficient Club cells, and culminating in the AO of mixed-cell tumor formation in the distal airways. This tumor formation is independent of route of exposure and our AOP construct is based on overlapping mechanistic events for naphthalene, styrene, ethyl benzene, isoniazid, and fluensulfone in the mouse. This AOP is intended to accelerate the explication of an apparent mouse-specific outcome and serve as a starting point for a quantitative analysis of mouse-human differences in susceptibility to the tumorigenic effects of Cyp2F2 ligands.

8.
Sci Rep ; 9(1): 145, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30644404

RESUMO

Heart rate assays in wild-type zebrafish embryos have been limited to analysis of one embryo per video/imaging field. Here we present for the first time a platform for high-throughput derivation of heart rate from multiple zebrafish (Danio rerio) embryos per imaging field, which is capable of quickly processing thousands of videos and ideal for multi-well platforms with multiple fish/well. This approach relies on use of 2-day post fertilization wild-type embryos, and uses only bright-field imaging, circumventing requirement for anesthesia or restraint, costly software/hardware, or fluorescently-labeled animals. Our original scripts (1) locate the heart and record pixel intensity fluctuations generated by each cardiac cycle using a robust image processing routine, and (2) process intensity data to derive heart rate. To demonstrate assay utility, we exposed embryos to the drugs epinephrine and clonidine, which increased or decreased heart rate, respectively. Exposure to organic extracts of air pollution-derived particulate matter, including diesel or biodiesel exhausts, or wood smoke, all complex environmental mixtures, decreased heart rate to varying degrees. Comparison against an established lower-throughput method indicated robust assay fidelity. As all code and executable files are publicly available, this approach may expedite cardiotoxicity screening of compounds as diverse as small molecule drugs and complex chemical mixtures.


Assuntos
Frequência Cardíaca/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Animais , Cardiotoxicidade , Avaliação Pré-Clínica de Medicamentos/métodos , Embrião não Mamífero , Processamento de Imagem Assistida por Computador , Material Particulado/toxicidade , Peixe-Zebra/embriologia
9.
Front Public Health ; 6: 261, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30255008

RESUMO

Chemical toxicity testing is moving steadily toward a human cell and organoid-based in vitro approach for reasons including scientific relevancy, efficiency, cost, and ethical rightfulness. Inferring human health risk from chemical exposure based on in vitro testing data is a challenging task, facing various data gaps along the way. This review identifies these gaps and makes a case for the in silico approach of computational dose-response and extrapolation modeling to address many of the challenges. Mathematical models that can mechanistically describe chemical toxicokinetics (TK) and toxicodynamics (TD), for both in vitro and in vivo conditions, are the founding pieces in this regard. Identifying toxicity pathways and in vitro point of departure (PoD) associated with adverse health outcomes requires an understanding of the molecular key events in the interacting transcriptome, proteome, and metabolome. Such an understanding will in turn help determine the sets of sensitive biomarkers to be measured in vitro and the scope of toxicity pathways to be modeled in silico. In vitro data reporting both pathway perturbation and chemical biokinetics in the culture medium serve to calibrate the toxicity pathway and virtual tissue models, which can then help predict PoDs in response to chemical dosimetry experienced by cells in vivo. Two types of in vitro to in vivo extrapolation (IVIVE) are needed. (1) For toxic effects involving systemic regulations, such as endocrine disruption, organism-level adverse outcome pathway (AOP) models are needed to extrapolate in vitro toxicity pathway perturbation to in vivo PoD. (2) Physiologically-based toxicokinetic (PBTK) modeling is needed to extrapolate in vitro PoD dose metrics into external doses for expected exposure scenarios. Linked PBTK and TD models can explore the parameter space to recapitulate human population variability in response to chemical insults. While challenges remain for applying these modeling tools to support in vitro toxicity testing, they open the door toward population-stratified and personalized risk assessment.

10.
Toxicol Sci ; 163(2): 430-439, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29471466

RESUMO

Controlled human exposure to the oxidant air pollutant ozone causes decrements in lung function and increased inflammation as evidenced by neutrophil influx into the lung and increased levels of proinflammatory cytokines in the airways. Here we describe a targeted metabolomics evaluation of human bronchoalveolar lavage fluid (BALF) following controlled in vivo exposure to ozone to gain greater insight into its pulmonary effects. In a 2-arm cross-over study, each healthy adult human volunteer was randomly exposed to filtered air (FA) and to 0.3 ppm ozone for 2 h while undergoing intermittent exercise with a minimum of 4 weeks between exposures. Bronchoscopy was performed and BALF obtained at 1 (n = 9) or 24 (n = 23) h postexposure. Metabolites were detected using ultrahigh performance liquid chromatography-tandem mass spectroscopy. At 1-h postexposure, a total of 28 metabolites were differentially expressed (DE) (p < .05) following ozone exposure compared with FA-exposure. These changes were associated with increased glycolysis and antioxidant responses, suggesting rapid increased energy utilization as part of the cellular response to oxidative stress. At 24-h postexposure, 41 metabolites were DE. Many of the changes were in amino acids and linked with enhanced proteolysis. Changes associated with increased lipid membrane turnover were also observed. These later-stage changes were consistent with ongoing repair of airway tissues. There were 1.37 times as many metabolites were differentially expressed at 24 h compared with 1-h postexposure. The changes at 1 h reflect responses to oxidative stress while the changes at 24 h indicate a broader set of responses consistent with tissue repair. These results illustrate the ability of metabolomic analysis to identify mechanistic features of ozone toxicity and aspects of the subsequent tissue response.


Assuntos
Líquido da Lavagem Broncoalveolar/química , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ozônio/toxicidade , Pneumonia/induzido quimicamente , Adulto , Aminoácidos/metabolismo , Estudos Cross-Over , Ácidos Graxos/metabolismo , Voluntários Saudáveis , Humanos , Inflamação , Exposição por Inalação/efeitos adversos , Pulmão/imunologia , Metabolômica , Pneumonia/imunologia , Pneumonia/metabolismo , Espectrometria de Massas em Tandem
11.
Environ Sci Technol ; 52(2): 839-849, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29236470

RESUMO

Cumulative risk assessment (CRA) methods promote the use of a conceptual site model (CSM) to apportion exposures and integrate risk from multiple stressors. While CSMs may encompass multiple species, evaluating end points across taxa can be challenging due to data availability and physiological differences among organisms. Adverse outcome pathways (AOPs) describe biological mechanisms leading to adverse outcomes (AOs) by assembling causal pathways with measurable intermediate steps termed key events (KEs), thereby providing a framework for integrating data across species. In this work, we used a case study focused on the perchlorate anion (ClO4-) to highlight the value of the AOP framework for cross-species data integration. Computational models and dose-response data were used to evaluate the effects of ClO4- in 12 species and revealed a dose-response concordance across KEs and taxa. The aggregate exposure pathway (AEP) tracks stressors from sources to the exposures and serves as a complement to the AOP. We discuss how the combined AEP-AOP construct helps to maximize the use of existing data and advances CRA by (1) organizing toxicity and exposure data, (2) providing a mechanistic framework of KEs for integrating data across human health and ecological end points, (3) facilitating cross-species dose-response evaluation, and (4) highlighting data gaps and technical limitations.


Assuntos
Rotas de Resultados Adversos , Ecologia , Humanos , Modelos Teóricos , Medição de Risco
12.
Environ Sci Technol ; 51(8): 4661-4672, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28355063

RESUMO

A quantitative adverse outcome pathway (qAOP) consists of one or more biologically based, computational models describing key event relationships linking a molecular initiating event (MIE) to an adverse outcome. A qAOP provides quantitative, dose-response, and time-course predictions that can support regulatory decision-making. Herein we describe several facets of qAOPs, including (a) motivation for development, (b) technical considerations, (c) evaluation of confidence, and (d) potential applications. The qAOP used as an illustrative example for these points describes the linkage between inhibition of cytochrome P450 19A aromatase (the MIE) and population-level decreases in the fathead minnow (FHM; Pimephales promelas). The qAOP consists of three linked computational models for the following: (a) the hypothalamic-pitutitary-gonadal axis in female FHMs, where aromatase inhibition decreases the conversion of testosterone to 17ß-estradiol (E2), thereby reducing E2-dependent vitellogenin (VTG; egg yolk protein precursor) synthesis, (b) VTG-dependent egg development and spawning (fecundity), and (c) fecundity-dependent population trajectory. While development of the example qAOP was based on experiments with FHMs exposed to the aromatase inhibitor fadrozole, we also show how a toxic equivalence (TEQ) calculation allows use of the qAOP to predict effects of another, untested aromatase inhibitor, iprodione. While qAOP development can be resource-intensive, the quantitative predictions obtained, and TEQ-based application to multiple chemicals, may be sufficient to justify the cost for some applications in regulatory decision-making.


Assuntos
Inibidores da Aromatase/toxicidade , Fadrozol/toxicidade , Animais , Cyprinidae , Estradiol/metabolismo , Modelos Teóricos , Valor Preditivo dos Testes , Vitelogeninas/metabolismo
13.
Inhal Toxicol ; 29(12-14): 586-597, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29405084

RESUMO

Study of the mode of action (MOA) relating exposure to a given chemical with an associated adverse outcome is an iterative process with each iteration driven by new understandings of the relevant biology. Here, we revisit a previously described, MOA-based clonal growth model of the human respiratory tract cancer risk associated with formaldehyde inhalation. Changes reflect a better understanding of populations of cells at risk of carcinogenic transformation in the pharynx, larynx and respiratory bronchiolar portions of the human respiratory tract and inclusion of basal cells in the pool of cells at risk. The focus of this report is not on cancer risk per se, but rather on the sensitivity of model parameters and predicted risks to alternative descriptions of the fraction of cells at risk for carcinogenic transformation. For a population of formaldehyde-exposed nonsmokers, revised specification of cells at risk resulted in changes in both parameter estimates and in predicted risks. Compared to our previous assessment, predicted additional risks were up to 87% greater at exposure levels ≤1 ppm, but up to about 130% lower at high exposure levels (2-5 ppm). While this work should not be considered an update to MOA-based risk assessments for formaldehyde described previously, it illustrates the sensitivity of parameter estimates and risk predictions to the quantitative specification of cells at risk of carcinogenic transformation and, therefore, the motivation for describing the relevant biology as accurately as possible.


Assuntos
Carcinogênese/induzido quimicamente , Formaldeído/toxicidade , Modelos Biológicos , Mucosa Respiratória/efeitos dos fármacos , Sistema Respiratório/efeitos dos fármacos , Carcinogênese/patologia , Células Cultivadas , Desinfetantes/toxicidade , Humanos , Exposição por Inalação/efeitos adversos , Mucosa Respiratória/patologia , Sistema Respiratório/patologia , Fatores de Risco
14.
Artigo em Inglês | MEDLINE | ID: mdl-26875912

RESUMO

There is international concern about chemicals that alter endocrine system function in humans and/or wildlife and subsequently cause adverse effects. We previously developed a mechanistic computational model of the hypothalamic-pituitary-gonadal (HPG) axis in female fathead minnows exposed to a model aromatase inhibitor, fadrozole (FAD), to predict dose-response and time-course behaviors for apical reproductive endpoints. Initial efforts to develop a computational model describing adaptive responses to endocrine stress providing good fits to empirical plasma 17ß-estradiol (E2) data in exposed fish were only partially successful, which suggests that additional regulatory biology processes need to be considered. In this study, we addressed short-comings of the previous model by incorporating additional details concerning CYP19A (aromatase) protein synthesis. Predictions based on the revised model were evaluated using plasma E2 concentrations and ovarian cytochrome P450 (CYP) 19A aromatase mRNA data from two fathead minnow time-course experiments with FAD, as well as from a third 4-day study. The extended model provides better fits to measured E2 time-course concentrations, and the model accurately predicts CYP19A mRNA fold changes and plasma E2 dose-response from the 4-d concentration-response study. This study suggests that aromatase protein synthesis is an important process in the biological system to model the effects of FAD exposure.


Assuntos
Aromatase/metabolismo , Cyprinidae/fisiologia , Disruptores Endócrinos/toxicidade , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Modelos Biológicos , Ovário/efeitos dos fármacos , Animais , Aromatase/química , Aromatase/genética , Inibidores da Aromatase/administração & dosagem , Inibidores da Aromatase/toxicidade , Biologia Computacional , Cyprinidae/sangue , Cyprinidae/crescimento & desenvolvimento , Relação Dose-Resposta a Droga , Disruptores Endócrinos/administração & dosagem , Estradiol/sangue , Fadrozol/administração & dosagem , Fadrozol/toxicidade , Feminino , Proteínas de Peixes/agonistas , Proteínas de Peixes/antagonistas & inibidores , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Ovário/enzimologia , Ovário/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Reprodutibilidade dos Testes , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testes de Toxicidade/métodos , Poluentes Químicos da Água/administração & dosagem , Poluentes Químicos da Água/toxicidade
15.
Environ Health Perspect ; 122(12): 1261-70, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25117432

RESUMO

BACKGROUND: Increasingly, there is a move toward using in vitro toxicity testing to assess human health risk due to chemical exposure. As with in vivo toxicity testing, an important question for in vitro results is whether there are thresholds for adverse cellular responses. Empirical evaluations may show consistency with thresholds, but the main evidence has to come from mechanistic considerations. OBJECTIVES: Cellular response behaviors depend on the molecular pathway and circuitry in the cell and the manner in which chemicals perturb these circuits. Understanding circuit structures that are inherently capable of resisting small perturbations and producing threshold responses is an important step towards mechanistically interpreting in vitro testing data. METHODS: Here we have examined dose-response characteristics for several biochemical network motifs. These network motifs are basic building blocks of molecular circuits underpinning a variety of cellular functions, including adaptation, homeostasis, proliferation, differentiation, and apoptosis. For each motif, we present biological examples and models to illustrate how thresholds arise from specific network structures. DISCUSSION AND CONCLUSION: Integral feedback, feedforward, and transcritical bifurcation motifs can generate thresholds. Other motifs (e.g., proportional feedback and ultrasensitivity)produce responses where the slope in the low-dose region is small and stays close to the baseline. Feedforward control may lead to nonmonotonic or hormetic responses. We conclude that network motifs provide a basis for understanding thresholds for cellular responses. Computational pathway modeling of these motifs and their combinations occurring in molecular signaling networks will be a key element in new risk assessment approaches based on in vitro cellular assays.


Assuntos
Exposição Ambiental , Poluentes Ambientais/toxicidade , Modelos Biológicos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Poluentes Ambientais/administração & dosagem , Humanos , Testes de Toxicidade
16.
Toxicol Sci ; 138(2): 412-24, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24385418

RESUMO

Formaldehyde is a nasal carcinogen in rodents at high doses and is an endogenous compound that is present in all living cells. Due to its high solubility and reactivity, quantitative risk estimates for inhaled formaldehyde have relied on internal dose estimates in the upper respiratory tract. Dosimetry calculations are complicated by the presence of endogenous formaldehyde concentrations in the respiratory mucosa. Anatomically accurate computational fluid dynamics (CFD) models of the rat, monkey, and human nasal passages were used to simulate uptake of inhaled formaldehyde. An epithelial structure was implemented in the nasal CFD models to estimate formaldehyde absorption from air:tissue partitioning, species-specific metabolism, first-order clearance, DNA binding, and endogenous formaldehyde production. At an exposure concentration of 1 ppm, predicted formaldehyde nasal uptake was 99.4, 86.5, and 85.3% in the rat, monkey, and human, respectively. Endogenous formaldehyde in nasal tissues did not significantly affect wall mass flux or nasal uptake predictions at exposure concentrations > 500 ppb; however, reduced nasal uptake was predicted at lower exposure concentrations. At an exposure concentration of 1 ppb, predicted nasal uptake was 17.5 and 42.8% in the rat and monkey; net desorption of formaldehyde was predicted in the human model. The nonlinear behavior of formaldehyde nasal absorption will affect the dose-response analysis and subsequent risk estimates at low exposure concentrations. Updated surface area partitioning of nonsquamous epithelium and average flux values in regions where DNA-protein cross-links and cell proliferation rates were measured in rats and monkeys are reported for use in formaldehyde risk models of carcinogenesis.


Assuntos
Carcinógenos/toxicidade , Formaldeído/toxicidade , Exposição por Inalação/análise , Modelos Biológicos , Mucosa Nasal/efeitos dos fármacos , Mucosa Nasal/metabolismo , Animais , Carcinógenos/metabolismo , Biologia Computacional/métodos , Adutos de DNA/metabolismo , Relação Dose-Resposta a Droga , Formaldeído/metabolismo , Humanos , Exposição por Inalação/efeitos adversos , Macaca mulatta , Modelos Anatômicos , Cavidade Nasal/anatomia & histologia , Valor Preditivo dos Testes , Ratos , Ratos Endogâmicos F344 , Especificidade da Espécie
17.
Regul Toxicol Pharmacol ; 66(2): 234-40, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23567188

RESUMO

The NRC report Science and Decisions: Advancing Risk Assessment made several recommendations to improve chemical risk assessment, with a focus on in-depth chronic dose-response assessments conducted by the U.S. Environmental Protection Agency. The recommendations addressed two broad elements: improving technical analysis and utility for decision making. To advance the discussions in the NRC report, in three multi-stakeholder workshops organized by the Alliance for Risk Assessment, available and evolving risk assessment methodologies were considered through the development and application of case studies. A key product was a framework (http://www.allianceforrisk.org/Workshop/Framework/ProblemFormulation.html) to guide risk assessors and managers to various dose-response assessment methods relevant to a range of decision contexts ranging from priority setting to full assessment, as illustrated by case studies. It is designed to facilitate selection of appropriate methodology for a variety of problem formulations and includes a variety of methods with supporting case studies, for areas flagged specifically by the NRC committee for consideration--e.g., susceptible sub-populations, population variability and background. The framewok contributes to organization and communication about methodologies for incorporating increasingly biologically informed and chemical specific knowledge into dose-response analysis, which is considered critical in evolving fit-for-purpose assessment to address relevant problem formulations.


Assuntos
Relação Dose-Resposta a Droga , Animais , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Medição de Risco/métodos
18.
Toxicol Sci ; 133(2): 234-47, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23475784

RESUMO

Endocrine-disrupting chemicals can affect reproduction and development in humans and wildlife. We developed a computational model of the hypothalamic-pituitary-gonadal (HPG) axis in female fathead minnows to predict dose-response and time-course (DRTC) behaviors for endocrine effects of the aromatase inhibitor, fadrozole (FAD). The model describes adaptive responses to endocrine stress involving regulated secretion of a generic gonadotropin (LH/FSH) from the hypothalamic-pituitary complex. For model development, we used plasma 17ß-estradiol (E2) concentrations and ovarian cytochrome P450 (CYP) 19A aromatase mRNA data from two time-course experiments, each of which included both an exposure and a depuration phase, and plasma E2 data from a third 4-day study. Model parameters were estimated using E2 concentrations for 0, 0.5, and 3 µg/l FAD exposure concentrations, and good fits to these data were obtained. The model accurately predicted CYP19A mRNA fold changes for controls and three FAD doses (0, 0.5, and 3 µg/l) and plasma E2 dose response from the 4-day study. Comparing the model-predicted DRTC with experimental data provided insight into how the feedback control mechanisms in the HPG axis mediate these changes: specifically, adaptive changes in plasma E2 levels occurring during exposure and "overshoot" occurring postexposure. This study demonstrates the value of mechanistic modeling to examine and predict dynamic behaviors in perturbed systems. As this work progresses, we will obtain a refined understanding of how adaptive responses within the vertebrate HPG axis affect DRTC behaviors for aromatase inhibitors and other types of endocrine-active chemicals and apply that knowledge in support of risk assessments.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Alternativas aos Testes com Animais , Inibidores da Aromatase/toxicidade , Simulação por Computador , Antagonistas de Estrogênios/toxicidade , Fadrozol/toxicidade , Ovário/efeitos dos fármacos , Animais , Cyprinidae/fisiologia , Relação Dose-Resposta a Droga , Estradiol/sangue , Feminino , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/enzimologia , Masculino , Ovário/enzimologia , Valor Preditivo dos Testes , Fatores de Tempo
19.
Toxicol Sci ; 133(2): 225-33, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23492810

RESUMO

Adaptive or compensatory responses to chemical exposure can significantly influence in vivo concentration-duration-response relationships. This study provided data to support development of a computational dynamic model of the hypothalamic-pituitary-gonadal axis of a model vertebrate and its response to aromatase inhibitors as a class of endocrine active chemicals. Fathead minnows (Pimephales promelas) were either exposed to the aromatase inhibitor fadrozole (0.5 or 30 µg/l) continuously for 1, 8, 12, 16, 20, 24, or 28 days or exposed for 8 days and then held in control water (no fadrozole) for an additional 4, 8, 12, 16, or 20 days. The time course of effects on ovarian steroid production, circulating 17ß-estradiol (E2) and vitellogenin (VTG) concentrations, and expression of steroidogenesis-related genes in the ovary was measured. Exposure to 30 µg fadrozole/l significantly reduced plasma E2 and VTG concentrations after just 1 day and those effects persisted throughout 28 days of exposure. In contrast, ex vivo E2 production was similar to that of controls on day 8-28 of exposure, whereas transcripts coding for aromatase and follicle-stimulating hormone receptor were elevated, suggesting a compensatory response. Following cessation of fadrozole exposure, ex vivo E2 and plasma E2 concentrations exceeded and then recovered to control levels, but plasma VTG concentrations did not, even after 20 days of depuration. Collectively these data provide several new insights into the nature and time course of adaptive responses to an aromatase inhibitor that support development of a computational model (see companion article).


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Inibidores da Aromatase/toxicidade , Cyprinidae/fisiologia , Antagonistas de Estrogênios/toxicidade , Fadrozol/toxicidade , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Ovário/efeitos dos fármacos , Alternativas aos Testes com Animais , Animais , Inibidores da Aromatase/análise , Estradiol/sangue , Antagonistas de Estrogênios/análise , Fadrozol/análise , Feminino , Sistema Hipotálamo-Hipofisário/enzimologia , Masculino , Ovário/enzimologia , Valor Preditivo dos Testes , Fatores de Tempo , Vitelogeninas/sangue
20.
Toxicol Appl Pharmacol ; 268(1): 17-26, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23357550

RESUMO

Many environmental contaminants can disrupt the adaptive immune response. Exposure to the ubiquitous aryl hydrocarbon receptor (AhR) ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and other agonists suppresses the antibody response. The underlying pathway mechanism by which TCDD alters B cell function is not well understood. The present study investigated the mechanism of AhR-mediated pathways and mode of suppression by which TCDD perturbs terminal differentiation of B cells to plasma cells and thereby impairs antibody production. An integrated approach combining computational pathway modeling and in vitro assays with primary mouse B cells activated by lipopolysaccharide was employed. We demonstrated that suppression of the IgM response by TCDD occurs in an all-or-none (binary) rather than graded mode: i.e., it reduces the number of IgM-secreting cells in a concentration-dependent manner without affecting the IgM content in individual plasma cells. The mathematical model of the gene regulatory circuit underpinning B cell differentiation revealed that two previously identified AhR-regulated pathways, inhibition of signaling protein AP-1 and activation of transcription factor Bach2, could account for the all-or-none mode of suppression. Both pathways disrupt the operation of a bistable-switch circuit that contains transcription factors Bcl6, Prdm1, Pax5, and Bach2 and regulates B cell fate. The model further predicted that by transcriptionally activating Bach2, TCDD might delay B cell differentiation and increase the likelihood of isotype switching, thereby altering the antibody repertoire. In conclusion, the present study revealed the mode and specific pathway mechanisms by which the environmental immunosuppressant TCDD suppresses B cell differentiation.


Assuntos
Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Modelos Imunológicos , Dibenzodioxinas Policloradas/toxicidade , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/imunologia , Animais , Linfócitos B/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/antagonistas & inibidores , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Simulação por Computador , Feminino , Citometria de Fluxo , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Receptores de Hidrocarboneto Arílico/imunologia , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...