Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Crit Rev Toxicol ; 54(5): 315-329, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38808643

RESUMEN

To accurately characterize human health hazards, human, animal, and mechanistic data must be integrated and the relevance to the research question of all three lines of evidence must be considered. Mechanistic data are often critical to the full integration of animal and human data and to characterizing relevance and uncertainty. This novel evidence integration framework (EIF) provides a method for synthesizing data from comprehensive, systematic, quality-based assessments of the epidemiological and toxicological literature, including in vivo and in vitro mechanistic studies. It organizes data according to both the observed human health effects and the mechanism of action of the chemical, providing a method to support evidence synthesis. The disease-based component uses the evidence of human health outcomes studied in the best quality epidemiological literature to organize the toxicological data according to authors' stated purpose, with the pathophysiology of the disease determining the potential relevance of the toxicological data. The mechanism-based component organizes the data based on the proposed mechanisms of effect and data supporting events leading to each endpoint, with the epidemiological data potentially providing corroborating information. The EIF includes a method to cross-classify and describe the concordance of the data, and to characterize its uncertainty. At times, the two methods of organizing the data may lead to different conclusions. This facilitates identification of knowledge gaps and shows the impact of uncertainties on the strength of causal inference.


Asunto(s)
Sustancias Peligrosas , Humanos , Medición de Riesgo/métodos , Animales , Sustancias Peligrosas/toxicidad
2.
Toxicol Sci ; 193(1): 1-17, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-36912747

RESUMEN

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.


Asunto(s)
Carcinoma de Células Escamosas , Aductos de ADN , Ratas , Humanos , Animales , Ratas Endogámicas F344 , Modelos Biológicos , Formaldehído/toxicidad , Nariz/patología , Carcinoma de Células Escamosas/patología
3.
Toxicol Sci ; 191(2): 212-226, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36453847

RESUMEN

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.


Asunto(s)
Manganeso , Modelos Biológicos , Humanos , Ratas , Animales , Haplorrinos , Transporte Biológico , Administración por Inhalación
4.
Toxicol Sci ; 191(1): 15-24, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36409013

RESUMEN

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.


Asunto(s)
Aductos de ADN , ADN , Ratas , Animales , Ratas Endogámicas F344 , Mucosa Nasal , Formaldehído/toxicidad , Desoxiguanosina
5.
Toxicology ; 461: 152893, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34425169

RESUMEN

Evidence from both in vivo and in vitro studies suggests that gene expression changes from long-term exposure to arsenite evolve markedly over time, including reversals in the direction of expression change in key regulatory genes. In this study, human uroepithelial cells from the ureter segments of 4 kidney-donors were continuously treated in culture with arsenite at concentrations of 0.1 or 1 µM for 60 days. Gene expression at 10, 20, 30, 40, and 60 days was determined using Affymetrix human genome microarrays and signal pathway analysis was performed using GeneGo Metacore. Arsenic treated cells continued to proliferate for the full 60-day period, whereas untreated cells ceased proliferating after approximately 30 days. A peak in the number of gene changes in the treated cells compared to untreated controls was observed between 30 and 40 days of exposure, with substantially fewer changes at 10 and 60 days, suggesting remodeling of the cells over time. Consistent with this possibility, the direction of expression change for a number of key genes was reversed between 20 and 30 days, including CFOS and MDM2. While the progression of gene changes was different for each subject, a common pattern was observed in arsenic treated cells over time, with early upregulation of oxidative stress responses (HMOX1, NQ01, TXN, TXNRD1) and down-regulation of immune/inflammatory responses (IKKα). At around 30 days, there was a transition to increased inflammatory and proliferative signaling (AKT, CFOS), evidence of epithelial-to-mesenchymal transition (EMT), and alterations in DNA damage responses (MDM2, ATM). A common element in the changing response of cells to arsenite over time appears to involve up-regulation of MDM2 by inflammatory signaling (through AP-1 and NF-κB), leading to inhibition of P53 function.


Asunto(s)
Arsenitos/toxicidad , Células Epiteliales/efectos de los fármacos , Proteínas Proto-Oncogénicas c-mdm2/genética , Urotelio/efectos de los fármacos , Adulto , Arsenitos/administración & dosificación , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genómica , Humanos , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Uréter/citología , Uréter/efectos de los fármacos , Urotelio/citología , Adulto Joven
6.
Toxicol Sci ; 177(2): 325-333, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32735340

RESUMEN

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.


Asunto(s)
Aductos de ADN , Formaldehído/toxicidad , Guanina , Animales , ADN , Cinética , Mucosa Nasal , Ratas
7.
Risk Anal ; 40(2): 294-318, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31524302

RESUMEN

ß-Chloroprene is used in the production of polychloroprene, a synthetic rubber. In 2010, Environmental Protection Agency (EPA) published the Integrated Risk Information System "Toxicological Review of Chloroprene," concluding that chloroprene was "likely to be carcinogenic to humans." This was based on findings from a 1998 National Toxicology Program (NTP) study showing multiple tumors within and across animal species; results from occupational epidemiological studies; a proposed mutagenic mode of action; and structural similarities with 1,3-butadiene and vinyl chloride. Using mouse data from the NTP study and assuming a mutagenic mode of action, EPA calculated an inhalation unit risk (IUR) for chloroprene of 5 × 10-4 per µg/m3 . This is among the highest IURs for chemicals classified by IARC or EPA as known or probable human carcinogens and orders of magnitude higher than the IURs for carcinogens such as vinyl chloride, benzene, and 1,3-butadiene. Due to differences in pharmacokinetics, mice appear to be uniquely responsive to chloroprene exposure compared to other animals, including humans, which is consistent with the lack of evidence of carcinogenicity in robust occupational epidemiological studies. We evaluated and integrated all lines of evidence for chloroprene carcinogenicity to assess whether the 2010 EPA IUR could be scientifically substantiated. Due to clear interspecies differences in carcinogenic response to chloroprene, we applied a physiologically based pharmacokinetic model for chloroprene to calculate a species-specific internal dose (amount metabolized/gram of lung tissue) and derived an IUR that is over 100-fold lower than the 2010 EPA IUR. Therefore, we recommend that EPA's IUR be updated.


Asunto(s)
Pruebas de Carcinogenicidad , Carcinógenos , Cloropreno/efectos adversos , Administración por Inhalación , Animales , Cricetinae , Humanos , Cinética , Ratones , Modelos Animales , Ratas , Medición de Riesgo , Estados Unidos , United States Environmental Protection Agency
8.
Regul Toxicol Pharmacol ; 106: 210-223, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31059732

RESUMEN

Anticipating the need to evaluate and integrate scientific evidence to inform new risk assessments or to update existing risk assessments, the Formaldehyde Panel of the American Chemistry Council (ACC), in collaboration with the University of North Carolina, convened a workshop: "Understanding Potential Human Health Cancer Risk - From Data Integration to Risk Evaluation" in October 2017. Twenty-four (24) invited-experts participated with expertise in epidemiology, toxicology, science integration and risk evaluation. Including members of the organizing committee, there were 29 participants. The meeting included eleven presentations encompassing an introduction and three sessions: (1) "integrating the formaldehyde science on nasal/nasopharyngeal carcinogenicity and potential for causality"; (2) "integrating the formaldehyde science on lymphohematopoietic cancer and potential for causality; and, (3) "formaldehyde research-data suitable for risk assessment". Here we describe key points from the presentations on epidemiology, toxicology and mechanistic studies that should inform decisions about the potential carcinogenicity of formaldehyde in humans and the discussions about approaches for structuring an integrated, comprehensive risk assessment for formaldehyde. We also note challenges expected when attempting to reconcile divergent results observed from research conducted within and across different scientific disciplines - especially toxicology and epidemiology - and in integrating diverse, multi-disciplinary mechanistic evidence.


Asunto(s)
Formaldehído/efectos adversos , Comunicación Interdisciplinaria , Animales , Humanos , Medición de Riesgo
9.
Toxicol Appl Pharmacol ; 372: 1-10, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30978397

RESUMEN

Previously, we developed a series of physiologically based pharmacokinetic (PBPK) models for manganese (Mn) in which saturable tissue binding and dose-dependent increases in biliary excretion captured key aspects of Mn homeostasis biology. These models reproduced the non-linear behavior of Mn kinetics in different tissues, accounting for dose-dependent changes in Mn kinetics. The original model construct had relatively slow association and dissociation rate constants for Mn binding in tissues. In this updated model, both rates of entry into tissue and the interaction of Mn with binding sites are rapid, and the step limiting Mn accumulation is the saturation of tissue binding sites. This binding reflects general cellular requirements for Mn with high affinity but rapid exchange between bound and free forms, which we captured using a dissociation constant (KD) of ~ 0.5 µM across tissues while maintaining different maximum binding capacities in each tissue. Variability in the binding capacities accounted for different background levels of Mn in particular tissues. This alternative structure successfully described Mn kinetics in tissues in adult rats exposed to Mn either in their diet or by inhalation, indicating that both the original and the present models capture the dose-dependent and tissue-specific kinetic behavior of Mn in adult rats. Although the published models that emphasize the role of smaller tissue binding rate constants in non-linear behaviors capture all relevant dose-dependent kinetic behaviors of this metal, increasing biological relevance of the model structure and parameters should provide greater confidence in applying the Mn PBPK models to risk assessment.


Asunto(s)
Manganeso/farmacocinética , Modelos Biológicos , Animales , Transporte Biológico , Relación Dosis-Respuesta a Droga , Eliminación Hepatobiliar , Homeostasis , Humanos , Manganeso/toxicidad , Dinámicas no Lineales , Unión Proteica , Medición de Riesgo , Distribución Tisular , Toxicocinética
10.
Crit Rev Toxicol ; 49(1): 36-84, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30932726

RESUMEN

The biologic effects of inorganic arsenic predominantly involve reaction of the trivalent forms with sulfhydryl groups in critical proteins in target cells, potentially leading to various toxicologic events including cancer. This mode of action is a threshold process, requiring sufficient concentrations of trivalent arsenic to disrupt normal cellular function. Nevertheless, cancer risk assessments for inorganic arsenic have traditionally utilized various dose-response models that extrapolate risks from high doses assuming low-dose linearity without a threshold. We present here an approach for a cancer risk assessment for inorganic arsenic in drinking water that involves considerations of this threshold process. Extensive investigations in mode of action analysis, in vitro studies (>0.1 µM), and in animal studies (>2 mg/L in drinking water or 2 mg/kg of diet), collectively indicate a threshold basis for inorganic arsenic-related cancers. These studies support a threshold for the effects of arsenic in humans of 50-100 µg/L in drinking water (about 65 µg/L). We then evaluate the epidemiology of cancers of the urinary bladder, lung, and skin and non-cancer skin changes for consistency with this calculated value, focusing on studies involving low-level exposures to inorganic arsenic primarily in drinking water (approximately <150 µg/L). Based on the relevant epidemiological studies with individual-level data, a threshold level for inorganic arsenic in the drinking water for these cancers is estimated to be around 100 µg/L, with strong evidence that it is between 50 and 150 µg/L, consistent with the value calculated based on mechanistic, in vitro and in vivo investigations. This evaluation provides an alternative mode of action-based approach for assessing health-protective levels for oral arsenic exposure based on the collective in vitro, in vivo, and human evidence rather than the use of a linear low-dose extrapolation based on default assumptions and theories.


Asunto(s)
Arsénico/análisis , Agua Potable/química , Exposición a Riesgos Ambientales/estadística & datos numéricos , Neoplasias/epidemiología , Contaminantes Químicos del Agua/análisis , Relación Dosis-Respuesta a Droga , Agua Potable/análisis
11.
Risk Anal ; 39(7): 1441-1464, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30925210

RESUMEN

We review approaches for characterizing "peak" exposures in epidemiologic studies and methods for incorporating peak exposure metrics in dose-response assessments that contribute to risk assessment. The focus was on potential etiologic relations between environmental chemical exposures and cancer risks. We searched the epidemiologic literature on environmental chemicals classified as carcinogens in which cancer risks were described in relation to "peak" exposures. These articles were evaluated to identify some of the challenges associated with defining and describing cancer risks in relation to peak exposures. We found that definitions of peak exposure varied considerably across studies. Of nine chemical agents included in our review of peak exposure, six had epidemiologic data used by the U.S. Environmental Protection Agency (US EPA) in dose-response assessments to derive inhalation unit risk values. These were benzene, formaldehyde, styrene, trichloroethylene, acrylonitrile, and ethylene oxide. All derived unit risks relied on cumulative exposure for dose-response estimation and none, to our knowledge, considered peak exposure metrics. This is not surprising, given the historical linear no-threshold default model (generally based on cumulative exposure) used in regulatory risk assessments. With newly proposed US EPA rule language, fuller consideration of alternative exposure and dose-response metrics will be supported. "Peak" exposure has not been consistently defined and rarely has been evaluated in epidemiologic studies of cancer risks. We recommend developing uniform definitions of "peak" exposure to facilitate fuller evaluation of dose response for environmental chemicals and cancer risks, especially where mechanistic understanding indicates that the dose response is unlikely linear and that short-term high-intensity exposures increase risk.


Asunto(s)
Neoplasias/inducido químicamente , Neoplasias/epidemiología , Medición de Riesgo/métodos , Acrilonitrilo , Contaminantes Atmosféricos/análisis , Benceno , Exposición a Riesgos Ambientales , Estudios Epidemiológicos , Óxido de Etileno , Formaldehído , Humanos , Leucemia/inducido químicamente , Linfoma/inducido químicamente , Cloruro de Metileno , Neoplasias/prevención & control , Estireno , Tricloroetileno , Estados Unidos , United States Environmental Protection Agency
12.
Regul Toxicol Pharmacol ; 103: 237-252, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30707931

RESUMEN

The United States Environmental Protection Agency's (USEPA) 2017 report, "Draft Report: Proposed Approaches to Inform the Derivation of a Maximum Contaminant Level Goal for Perchlorate in Drinking Water", proposes novel approaches for deriving a Maximum Contaminant Level Goal (MCLG) for perchlorate using a biologically-based dose-response (BBDR) model. The USEPA (2017) BBDR model extends previously peer-reviewed perchlorate models to describe the relationship between perchlorate exposure and thyroid hormone levels during early pregnancy. Our evaluation focuses on two key elements of the USEPA (2017) report: the plausibility of BBDR model revisions to describe control of thyroid hormone production in early pregnancy and the basis for linking BBDR model results to neurodevelopmental outcomes. While the USEPA (2017) BBDR model represents a valuable research tool, the lack of supporting data for many of the model assumptions and parameters calls into question the fitness of the extended BBDR model to support quantitative analyses for regulatory decisions on perchlorate in drinking water. Until more data can be developed to address uncertainties in the current BBDR model, USEPA should continue to rely on the RfD recommended by the NAS (USEPA, 2005) when considering further regulatory action.


Asunto(s)
Agua Potable/química , Percloratos/análisis , Contaminantes Químicos del Agua/análisis , Relación Dosis-Respuesta a Droga , Humanos , Medición de Riesgo , Estados Unidos , United States Environmental Protection Agency
13.
Inhal Toxicol ; 31(13-14): 468-483, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31992090

RESUMEN

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.


Asunto(s)
Contaminantes Atmosféricos/farmacocinética , Cloropreno/farmacocinética , Exposición por Inhalación/efectos adversos , Pulmón/metabolismo , Modelos Biológicos , Contaminantes Atmosféricos/sangre , Contaminantes Atmosféricos/toxicidad , Animales , Cloropreno/sangre , Cloropreno/toxicidad , Femenino , Humanos , Exposición por Inhalación/análisis , Pulmón/efectos de los fármacos , Cadenas de Markov , Ratones , Método de Montecarlo , Pletismografía , Valor Predictivo de las Pruebas , Ratas , Medición de Riesgo , Especificidad de la Especie , Distribución Tisular
14.
Toxicol Appl Pharmacol ; 359: 70-81, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30243844

RESUMEN

Due to concerns for enhanced absorption of manganese (Mn) from drinking water compared to diet, bioavailability of Mn from drinking water remains a major data gap in understanding Mn kinetics. In this study, PBPK models for adult rats and humans were updated with a drinking water exposure route and were used to assess the homeostatic control of Mn uptake, excretion and tissue kinetics between the two different ingestion modes. Drinking water model parameters were estimated from tissue kinetic data from a drinking water study in rats. The published study included a 10 ppm-Mn diet with additional Mn added to drinking water to give a total ingested Mn dose equivalent to that from a 200 ppm diet. The 200 ppm diet and equivalent mixed drinking water/diet exposures provided Mn concentrations for brain (striatum, olfactory bulb, and cerebellum), liver and bone after 7 and 61 days of Mn exposure. Modeling of these data sets indicated that (1) the oral Mn bioavailability is similar for diet or drinking water and (2) homeostatic control of gut uptake of Mn occurs with either drinking water or dietary ingestion. This updated description for absorption and distribution of Mn from gut was added to a human Mn-PBPK model to simulate Mn exposure from multiple routes of exposure (i.e. dietary intake, drinking water, and inhalation). This increases the utility of the Mn PBPK model by allowing for the simulation of multiple Mn exposure scenarios, including variable daily food and drinking water exposures in a human population.


Asunto(s)
Dieta , Agua Potable , Manganeso/farmacocinética , Adolescente , Animales , Disponibilidad Biológica , Peso Corporal/efectos de los fármacos , Huesos/metabolismo , Encéfalo/metabolismo , Niño , Femenino , Análisis de los Alimentos , Tracto Gastrointestinal/metabolismo , Humanos , Exposición por Inhalación , Absorción Intestinal , Hígado/metabolismo , Masculino , Modelos Biológicos , Ratas , Distribución Tisular
15.
J Toxicol Environ Health A ; 81(18): 893-912, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30230972

RESUMEN

The aim of this study was to establish a process for deriving a chemical-specific mode of action (MOA) from chemical-agnostic adverse outcome pathway (AOPs), using inorganic arsenic (iAs) as a case study. The AOP developed for this case study are related to disruption of cellular signaling by chemicals that strongly bind to vicinal dithiols in cellular proteins, leading to disruption of inflammatory and oxidative stress signaling along with inhibition of the DNA damage responses. The proposed MOA for iAs incorporates this AOP, overlaid on a background of increasing oxidative stress and/or co-exposure to mutagenic chemicals or radiation. The most challenging aspect of developing a MOA from AOP is the incorporation of metabolism and dose-response, neither of which may be considered in the development of an AOP. The cellular responses to relatively low concentrations (below 100 parts per billion) of iAs in drinking water appear to be secondary to binding of trivalent arsenite and its trivalent metabolite, monomethyl arsenous acid to key cellular vicinal dithiols in target tissues, resulting in a co-carcinogenic MOA. The proposed AOP may also be applied to non-cancer endpoints, enabling an integrated approach to conducting a risk assessment for iAs.


Asunto(s)
Rutas de Resultados Adversos , Arsenicales/efectos adversos , Intoxicación por Arsénico/metabolismo , Arsenicales/metabolismo , Humanos , Medición de Riesgo/métodos
16.
Regul Toxicol Pharmacol ; 95: 371-384, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29614342

RESUMEN

Quantitative risk assessment (QRA), a scientific, evidence-based analytical process that combines chemical and biological data to quantify the probability and potential impact of some defined risk, is used by regulatory agencies for decision-making. Thus, in tobacco product regulation, specifically in substantial equivalence (SE) evaluations, QRA can provide a useful, practical, and efficient approach to address questions that might arise regarding human health risk and potential influence on public health. In SE reporting, when differences in product characteristics may necessitate the determination of whether a new product raises different questions of public health, the results from QRA are a valuable metric. An approach for QRA in this context is discussed, which is modeled after the methodology for assessment of constituent mixtures by the US Environmental Protection Agency for environmental Superfund site assessment. Given the intent in both cases is an assessment of the public health impact resulting from the totality of exposure to a mixture of constituents, the application is appropriate. Although some uncertainties in the information incorporated may exist, relying on the most appropriate of the available data increases the confidence and decreases the uncertainty in the risk characterization using this data-driven methodology.


Asunto(s)
Productos de Tabaco/toxicidad , Adulto , Animales , Exposición a Riesgos Ambientales/efectos adversos , Humanos , Neoplasias , Medición de Riesgo
19.
Regul Toxicol Pharmacol ; 92: 472-490, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29158043

RESUMEN

Shortly after the International Agency for Research on Cancer (IARC) determined that formaldehyde causes leukemia, the United States Environmental Protection Agency (EPA) released its Draft IRIS Toxicological Review of Formaldehyde ("Draft IRIS Assessment"), also concluding that formaldehyde causes leukemia. Peer review of the Draft IRIS Assessment by a National Academy of Science committee noted that "causal determinations are not supported by the narrative provided in the draft" (NRC 2011). They offered recommendations for improving the Draft IRIS assessment and identified several important research gaps. Over the six years since the NRC peer review, significant new science has been published. We identify and summarize key recommendations made by NRC and map them to this new science, including extended analysis of epidemiological studies, updates of earlier occupational cohort studies, toxicological experiments using a sensitive mouse strain, mechanistic studies examining the role of exogenous versus endogenous formaldehyde in bone marrow, and several critical reviews. With few exceptions, new findings are consistently negative, and integration of all available evidence challenges the earlier conclusions that formaldehyde causes leukemia. Given formaldehyde's commercial importance, environmental ubiquity and endogenous production, accurate hazard classification and risk evaluation of whether exposure to formaldehyde from occupational, residential and consumer products causes leukemia are critical.


Asunto(s)
Formaldehído/toxicidad , Leucemia/inducido químicamente , Leucemia/etiología , Animales , Médula Ósea/efectos de los fármacos , Humanos , Exposición Profesional/efectos adversos , Medición de Riesgo , Estados Unidos , United States Environmental Protection Agency
20.
Crit Rev Toxicol ; 47(7): 592-602, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28462599

RESUMEN

Several cross-sectional studies of a single population of workers exposed to formaldehyde at one of two factories using or producing formaldehyde-melamine resins in China have concluded that formaldehyde exposure induces damage to hematopoietic cells that originate in the bone marrow. Moreover, the investigators interpret observed differences between groups as evidence that formaldehyde induces myeloid leukemias, although the mechanisms for inducing these diseases are not obvious and recently published scientific findings do not support causation. Our objective was to evaluate hematological parameters and aneuploidy in relation to quantitative exposure measures of formaldehyde. We obtained the study data for the original study (Zhang et al. 2010 ) and performed linear regression analyses. Results showed that differences in white blood cell, granulocyte, platelet, and red blood cell counts are not exposure dependent. Among formaldehyde-exposed workers, no association was observed between individual average formaldehyde exposure estimates and frequency of aneuploidy, suggested by the original study authors to be indicators of myeloid leukemia risk.


Asunto(s)
Formaldehído/efectos adversos , Formaldehído/toxicidad , Sustancias Peligrosas/toxicidad , Exposición Profesional/estadística & datos numéricos , Hipersensibilidad Respiratoria/epidemiología , Adulto , Aneuploidia , Cromosomas , Estudios Transversales , Humanos , Leucemia , Células Progenitoras Mieloides
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...