Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Appl Toxicol ; 36(9): 1095-111, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26649444

RESUMEN

As a result of its presence in water as a volatile disinfection byproduct, bromodichloromethane (BDCM), which is mutagenic, poses a potential health risk from exposure via oral, dermal and inhalation routes. We developed a refined human physiologically based pharmacokinetic (PBPK) model for BDCM (including new chemical-specific human parameters) to evaluate the impact of BDCM exposure during showering and bathing on important measures of internal dose compared with oral exposure. The refined model adequately predicted data from the published literature for oral, dermal and bathing/showering exposures. A liter equivalency approach (L-eq) was used to estimate BDCM concentration in a liter of water consumed by the oral route that would be required to produce the same internal dose of BDCM resulting from a 20-min bath or a 10-min shower in water containing 10 µg l(-1) BDCM. The oral liter equivalent concentrations for the bathing scenario were 605, 803 and 5 µg l(-1) BDCM for maximum venous blood concentration (Cmax), the area under the curve (AUCv) and the amount metabolized in the liver per hour (MBDCM), respectively. For a 10-min showering exposure, the oral L-eq concentrations were 282, 312 and 2.1 µg l(-1) for Cmax, AUC and MBDCM, respectively. These results demonstrate large contributions of dermal and inhalation exposure routes to the internal dose of parent chemical reaching the systemic circulation, which could be transformed to mutagenic metabolites in extrahepatic target tissues. Thus, consideration of the contribution of multiple routes of exposure when evaluating risks from water-borne BDCM is needed, and this refined human model will facilitate improved assessment of internal doses from real-world exposures. Published 2015. This article has been contributed to by US Government employees and their work is in the public domain in the USA.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Administración Cutánea , Administración Oral , Adolescente , Adulto , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Exposición a Riesgos Ambientales/análisis , Femenino , Humanos , Exposición por Inhalación/efectos adversos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Modelos Biológicos , Medición de Riesgo , Sensibilidad y Especificidad , Pruebas de Toxicidad , Trihalometanos/toxicidad , Contaminantes Químicos del Agua/análisis , Contaminantes Químicos del Agua/toxicidad , Adulto Joven
2.
Toxicol Mech Methods ; 26(8): 620-626, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27595344

RESUMEN

A rate for hepatic metabolism (Vmax) determined in vitro must be scaled for in vivo use in a physiologically based pharmacokinetic (PBPK) model. This requires the use of scaling factors such as mg of microsomal protein per gram of liver (MPPGL) and liver mass (FVL). Variation in MPPGL and FVL impacts variation in Vmax, and hence PBPK model-derived estimates of internal dose used in dose response analysis. The impacts of adult human variation in MPPGL and FVL on estimates of internal dose were assessed using a human PBPK model for bromodichloromethane (BDCM), a water disinfection byproduct, for multiple internal dose metrics for two exposure scenarios (single 0.25 liter drink of water or 10 min shower) under plausible (5 µg/L) and high level (20 µg/L) water concentrations. For both concentrations, all internal dose metrics were changed less than 5% for the showering scenario (combined inhalation and dermal exposure). In contrast, a 27-fold variation in area under the curve (AUC) for BDCM in venous blood was observed at both oral exposure concentrations, whereas total amount of BDCM metabolized in liver was relatively unchanged. This analysis demonstrates that variability in the scaling factors used for in vitro to in vivo extrapolation (IVIVE) for metabolic rate parameters can have a significant route-dependent impact on estimates of internal dose under environmentally relevant exposure scenarios. This indicates the need to evaluate both uncertainty and variability for scaling factors used for IVIVE.


Asunto(s)
Exposición a Riesgos Ambientales/análisis , Hígado/efectos de los fármacos , Modelos Biológicos , Contaminantes Químicos del Agua/administración & dosificación , Administración Cutánea , Administración Oral , Relación Dosis-Respuesta a Droga , Exposición a Riesgos Ambientales/efectos adversos , Humanos , Exposición por Inhalación/efectos adversos , Exposición por Inhalación/análisis , Hígado/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Método de Montecarlo , Distribución Tisular , Trihalometanos/administración & dosificación , Trihalometanos/sangre , Trihalometanos/farmacocinética , Contaminantes Químicos del Agua/sangre , Contaminantes Químicos del Agua/farmacocinética
3.
J Pharmacokinet Pharmacodyn ; 42(6): 591-609, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26194069

RESUMEN

Any statistical model should be identifiable in order for estimates and tests using it to be meaningful. We consider statistical analysis of physiologically-based pharmacokinetic (PBPK) models in which parameters cannot be estimated precisely from available data, and discuss different types of identifiability that occur in PBPK models and give reasons why they occur. We particularly focus on how the mathematical structure of a PBPK model and lack of appropriate data can lead to statistical models in which it is impossible to estimate at least some parameters precisely. Methods are reviewed which can determine whether a purely linear PBPK model is globally identifiable. We propose a theorem which determines when identifiability at a set of finite and specific values of the mathematical PBPK model (global discete identifiability) implies identifiability of the statistical model. However, we are unable to establish conditions that imply global discrete identifiability, and conclude that the only safe approach to analysis of PBPK models involves Bayesian analysis with truncated priors. Finally, computational issues regarding posterior simulations of PBPK models are discussed. The methodology is very general and can be applied to numerous PBPK models which can be expressed as linear time-invariant systems. A real data set of a PBPK model for exposure to dimethyl arsinic acid (DMA(V)) is presented to illustrate the proposed methodology.


Asunto(s)
Ácido Cacodílico/farmacocinética , Exposición a Riesgos Ambientales , Contaminantes Ambientales/farmacocinética , Modelos Biológicos , Modelos Estadísticos , Animales , Teorema de Bayes , Biotransformación , Ácido Cacodílico/efectos adversos , Ácido Cacodílico/orina , Simulación por Computador , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes Ambientales/efectos adversos , Contaminantes Ambientales/orina , Humanos , Modelos Lineales , Metilaminas/farmacocinética , Ratones , Medición de Riesgo
4.
Toxicology ; 465: 153046, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34813904

RESUMEN

Short-term biomarkers of toxicity have an increasingly important role in the screening and prioritization of new chemicals. In this study, we examined early indicators of liver toxicity for three reference organophosphate (OP) chemicals, which are among the most widely used insecticides in the world. The OP methidathion was previously shown to increase the incidence of liver toxicity, including hepatocellular tumors, in male mice. To provide insights into the adverse outcome pathway (AOP) that underlies these tumors, effects of methidathion in the male mouse liver were examined after 7 and 28 day exposures and compared to those of two other OPs that either do not increase (fenthion) or possibly suppress liver cancer (parathion) in mice. None of the chemicals caused increases in liver weight/body weight or histopathological changes in the liver. Parathion decreased liver cell proliferation after 7 and 28 days while the other chemicals had no effects. There was no evidence for hepatotoxicity in any of the treatment groups. Full-genome microarray analysis of the livers from the 7 and 28 day treatments demonstrated that methidathion and fenthion regulated a large number of overlapping genes, while parathion regulated a unique set of genes. Examination of cytochrome P450 enzyme activities and use of predictive gene expression biomarkers found no consistent evidence for activation of AhR, CAR, PXR, or PPARα. Parathion suppressed the male-specific gene expression pattern through STAT5b, similar to genetic and dietary conditions that decrease liver tumor incidence in mice. Overall, these findings indicate that methidathion causes liver cancer by a mechanism that does not involve common mechanisms of liver cancer induction.


Asunto(s)
Transformación Celular Neoplásica/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Genómica , Insecticidas/toxicidad , Neoplasias Hepáticas/genética , Hígado/efectos de los fármacos , Compuestos Organofosforados/toxicidad , Transcriptoma/efectos de los fármacos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Receptor de Androstano Constitutivo/agonistas , Receptor de Androstano Constitutivo/genética , Receptor de Androstano Constitutivo/metabolismo , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Fentión/toxicidad , Perfilación de la Expresión Génica , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Compuestos Organotiofosforados/toxicidad , PPAR alfa/agonistas , PPAR alfa/genética , PPAR alfa/metabolismo , Paratión/toxicidad , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo
5.
Toxicology ; 457: 152809, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33965444

RESUMEN

Arsenic (As) poses unique challenges in PBTK model development and risk analysis applications. Arsenic metabolism is complex, adequate information to attribute specific metabolites to particular adverse effects in humans is sparse, and measurement of relevant metabolites in biological media can be difficult. Multiple As PBTK models have been published and used or adapted for use in various exposure and risk analysis applications. These applications illustrate the broad utility of PBTK models for exposure and dose-response analysis, particularly for arsenic where multi-pathway, multi-route exposures and multiple toxic effects are of concern. Arsenic PBTK models have been used together with exposure reconstruction and dose-response functions to estimate risk of specific adverse health effects due to drinking water exposure and consumption of specific foodstuffs (e.g. rice, seafood), as well as to derive safe exposure levels and develop consumption advisories. Future refinements to arsenic PBTK models can enhance the confidence in such analyses. Improved estimates for methylation biotransformation parameters based on in vitro to in vivo extrapolation (IVIVE) methods and estimation of interindividual variability in key model parameters for specific toxicologically relevant metabolites are two important areas for consideration.


Asunto(s)
Arsénico/farmacocinética , Arsénico/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Modelos Biológicos , Animales , Agua Potable/administración & dosificación , Agua Potable/efectos adversos , Alimentos/efectos adversos , Humanos , Medición de Riesgo , Toxicocinética
6.
J Toxicol Environ Health A ; 73(4): 301-18, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20077299

RESUMEN

Aging adults are a growing segment of the U.S. population and are likely to exhibit increased susceptibility to many environmental toxicants. However, there is little information on the susceptibility of the aged to toxicants. The toxicity of toluene has been well characterized in young adult rodents but there is little information in the aged. Three approaches were used: (1) pharmacokinetic (PK), (2) cardiac biomarkers, and (3) whole-animal physiology to assess whether aging increases susceptibility to toluene in the Brown Norway (BN) rat. Three life stages, young adult, middle aged, and aged (4, 12, and 24 mo, respectively), were administered toluene orally at doses of 0, 0.3, 0.65, or 1 g/kg and subjected to the following: terminated at 45 min or 4 h post dosing, and blood and brain toluene concentration were measured; terminated at 4 h post dosing, and biomarkers of cardiac function were measured; or monitor heart rate (HR), core temperature (Tc), and motor activity (MA) by radiotelemetry before and after dosing. Brain toluene concentration was significantly elevated in aged rats at 4 h after dosing with either 0.3 or 1 g/kg. Blood toluene concentrations were unaffected by age. There were various interactions between aging and toluene-induced effects on cardiac biomarkers. Most notably, toluene exposure led to reductions in mRNA markers for oxidative stress in aged but not younger animals. Toluene also produced a reduction in cardiac endothelin-1 in aged rats. Higher doses of toluene led to tachycardia, hypothermia, and a transient elevation in MA. Aged rats were less sensitive to the tachycardic effects of toluene but showed a prolonged hypothermic response. Elevated brain levels of toluene in aged rats may be attributed to their suppressed cardiovascular and respiratory responses. The expression of several cardiac biochemical markers of toluene exposure in the aged may also reflect differential susceptibility to this toxicant.


Asunto(s)
Envejecimiento/fisiología , Tolueno/farmacocinética , Tolueno/toxicidad , Animales , Biomarcadores , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Corazón/efectos de los fármacos , Masculino , Miocardio/metabolismo , Ratas , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Tolueno/sangre
7.
Toxicol In Vitro ; 69: 105002, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32946980

RESUMEN

Metabolic rate parameters estimation using in vitro data is necessary due to numbers of chemicals for which data are needed, trend towards minimizing laboratory animal use, and limited opportunity to collect data in human subjects. We evaluated how well metabolic rate parameters derived from in vitro data predict overall in vivo metabolism for a set of environmental chemicals for which well validated and established methods exist. We compared values of VmaxC derived from in vivo vapor uptake studies with estimates of VmaxC scaled up from in vitro hepatic microsomal metabolism studies for VOCs for which data were available in male F344 rats. For 6 of 7 VOCs, differences between the in vivo and scaled up in vitro VmaxC estimates were less than 2.6-fold. For bromodichloromethane (BDCM), the in vivo derived VmaxC was approximately 4.4-fold higher than the in vitro derived and scaled up VmaxC. The more rapid rate of BDCM metabolism estimated based in vivo studies suggests other factors such as extrahepatic metabolism, binding or other non-specific losses making a significant contribution to overall clearance. Systematic and reliable utilization of scaled up in vitro biotransformation rate parameters in PBPK models will require development of methods to predict cases in which extrahepatic metabolism and binding as well as other factors are likely to be significant contributors.


Asunto(s)
Compuestos Alílicos/farmacocinética , Hidrocarburos Clorados/farmacocinética , Propano/análogos & derivados , Compuestos Orgánicos Volátiles/farmacocinética , Animales , Masculino , Tasa de Depuración Metabólica , Modelos Biológicos , Propano/farmacocinética , Ratas Endogámicas F344
8.
Toxicol Sci ; 167(2): 347-359, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30252107

RESUMEN

Biotransformation rates extrapolated from in vitro data are used increasingly in human physiologically based pharmacokinetic (PBPK) models. This practice requires use of scaling factors, including microsomal content (mg of microsomal protein/g liver, MPPGL), enzyme specific content, and liver mass as a fraction of body weight (FVL). Previous analyses indicated that scaling factor variability impacts pharmacokinetic (PK) outcomes used in adult population dose-response studies. This analysis was extended to pediatric populations because large inter-individual differences in enzyme ontogeny likely would further contribute to scaling factor variability. An adult bromodichloromethane (BDCM) model (Kenyon, E. M., Eklund, C., Leavens, T. L., and Pegram, R. A. (2016a). Development and application of a human PBPK model for bromodichloromethane (BDCM) to investigate impacts of multi-route exposure. J. Appl. Toxicol. 36, 1095-1111) was re-parameterized for neonates, infants, and toddlers. Monte Carlo analysis was used to assess the impact of pediatric scaling factor variation on model-derived PK outcomes compared with adult findings. BDCM dose metrics were estimated following a single 0.05-liter drink of water or a 20-min bath, under typical (5 µg/l) and plausible higher (20 µg/l) BDCM concentrations. MPPGL, CYP2E1, and FVL values reflected the distribution of reported pediatric population values. The impact of scaling factor variability on PK outcome variation was different for each exposure scenario, but similar for each BDCM water concentration. The higher CYP2E1 expression variability during early childhood was reflected in greater variability in predicted PK outcomes in younger age groups, particularly for the oral exposure route. Sensitivity analysis confirmed the most influential parameter for this variability was CYP2E1, particularly in neonates. These findings demonstrate the importance of age-dependent scaling factor variation used for in vitro to in vivo extrapolation of biotransformation rates.


Asunto(s)
Exposición a Riesgos Ambientales/análisis , Hígado/efectos de los fármacos , Modelos Biológicos , Contaminantes Químicos del Agua/farmacocinética , Biotransformación , Peso Corporal/fisiología , Preescolar , Exposición a Riesgos Ambientales/efectos adversos , Humanos , Lactante , Recién Nacido , Hígado/metabolismo , Hígado/patología , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Microsomas Hepáticos/patología , Método de Montecarlo , Tamaño de los Órganos/fisiología , Distribución Tisular , Trihalometanos/farmacocinética
9.
Toxicol Appl Pharmacol ; 232(3): 359-68, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18687352

RESUMEN

Quantitative biologically-based models describing key events in the continuum from arsenic exposure to the development of adverse health effects provide a framework to integrate information obtained across diverse research areas. For example, genetic polymorphisms in arsenic metabolizing enzymes can lead to differences in target tissue dosimetry for key metabolites causative in toxic and carcinogenic response. This type of variation can be quantitatively incorporated into pharmacokinetic (PK) models and used together with population-based modeling approaches to evaluate the impact of genetic variation in methylation capacity on dose of key metabolites to target tissue. The PK model is an essential bridge to the pharmacodynamic (PD) models. A particular benefit of PD modeling for arsenic is that alternative models can be constructed for multiple proposed modes of action for arsenicals. Genomics data will prove useful for identifying the key pathways involved in particular responses and aid in determining other types of data needed for quantitative modeling. These models, when linked with PK models, can be used to better understand and explain dose- and time-response behaviors. This in turn assists in prioritizing modes of action with respect to their risk assessment relevance and future research. This type of integrated modeling approach can form the basis for a highly informative mode-of-action directed risk assessment for inorganic arsenic (iAs). This paper will address both practical and theoretical aspects of integrating PK and PD data in a modeling framework, including practical barriers to its application.


Asunto(s)
Arsénico/farmacocinética , Arsénico/toxicidad , Modelos Biológicos , Medición de Riesgo , Relación Dosis-Respuesta a Droga , Variación Genética , Humanos , Matemática , Metilación , Estado Nutricional , Factores Sexuales
10.
J Toxicol Environ Health A ; 71(4): 249-65, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18253891

RESUMEN

Toluene is found in petroleum-based fuels and used as a solvent in consumer products and industrial applications. The critical effects following inhalation exposure involve the brain and nervous system in both humans and experimental animals, whether exposure duration is acute or chronic. The goals of this physiologically based pharmacokinetic (PBPK) model development effort were twofold: (1) to evaluate and explain the influence of feeding status and activity level on toluene pharmacokinetics utilizing our own data from toluene-exposed Long Evans (LE) rats, and (2) to evaluate the ability of the model to simulate data from the published literature and explain differing toluene kinetics. Compartments in the model were lung, slowly and rapidly perfused tissue groups, fat, liver, gut, and brain; tissue transport was blood-flow limited and metabolism occurred in the liver. Chemical-specific parameters and initial organ volumes and blood flow rates were obtained from the literature. Sensitivity analysis revealed that the single most influential parameter for our experimental conditions was alveolar ventilation; other moderately influential parameters (depending upon concentration) included cardiac output, rate of metabolism, and blood flow to fat. Based on both literature review and sensitivity analysis, other parameters (e.g., partition coefficients and metabolic rate parameters) were either well defined (multiple consistent experimental results with low variability) or relatively noninfluential (e.g. organ volumes). Rats that were weight-maintained compared to free-fed rats in our studies could be modeled with a single set of parameters because feeding status did not have a significant impact on toluene pharmacokinetics. Heart rate (HR) measurements in rats performing a lever-pressing task indicated that the HR increased in proportion to task intensity. For rats acclimated to eating in the lab during the day, both sedentary rats and rats performing the lever-pressing task required different alveolar ventilation rates to successfully predict the data. Model evaluation using data from diverse sources together with statistical evaluation of the resulting fits revealed that the model appropriately predicted blood and brain toluene concentrations with some minor exceptions. These results (1) emphasize the importance of experimental conditions and physiological status in explaining differing kinetic data, and (2) demonstrate the need to consider simulation conditions when estimating internal dose metrics for toxicity studies in which kinetic data were not collected.


Asunto(s)
Conducta Alimentaria/fisiología , Modelos Biológicos , Actividad Motora/fisiología , Solventes/farmacocinética , Tolueno/farmacocinética , Animales , Encéfalo/metabolismo , Condicionamiento Operante , Frecuencia Cardíaca , Masculino , Ratas , Ratas Long-Evans , Tolueno/sangre
11.
J Pharmacokinet Pharmacodyn ; 35(1): 31-68, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17943421

RESUMEN

A physiologically-based pharmacokinetic (PBPK) model was developed to estimate levels of arsenic and its metabolites in human tissues and urine after oral exposure to arsenate (As(V)), arsenite (As(III)) or organoarsenical pesticides. The model consists of interconnected individual PBPK models for inorganic arsenic (As(V) and As(III)), monomethylarsenic acid (MMA(V)), and, dimethylarsenic acid (DMA(V)). Reduction of MMA(V) and DMA(V) to their respective trivalent forms also occurs in the lung, liver, and kidney including excretion in urine. Each submodel was constructed using flow limited compartments describing the mass balance of the chemicals in GI tract (lumen and tissue), lung, liver, kidney, muscle, skin, heart, and brain. The choice of tissues was based on physiochemical properties of the arsenicals (solubility), exposure routes, target tissues, and sites for metabolism. Metabolism of inorganic arsenic in liver was described as a series of reduction and oxidative methylation steps incorporating the inhibitory influence of metabolites on methylation. The inhibitory effects of As(III) on the methylation of MMA(III) to DMA, and MMA(III) on the methylation of As(III) to MMA were modeled as noncompetitive. To avoid the uncertainty inherent in estimation of many parameters from limited human data, a priori independent parameter estimates were derived using data from diverse experimental systems with priority given to data derived using human cells and tissues. This allowed the limited data for human excretion of arsenicals in urine to be used to estimate only parameters that were most sensitive to this type of data. Recently published urinary excretion data, not previously used in model development, are also used to evaluate model predictions.


Asunto(s)
Arsénico/farmacocinética , Ácido Cacodílico/metabolismo , Modelos Biológicos , Adulto , Arseniatos/metabolismo , Arseniatos/farmacocinética , Arsénico/metabolismo , Arsenitos/metabolismo , Arsenitos/farmacocinética , Femenino , Humanos , Riñón/metabolismo , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Metilación , Persona de Mediana Edad , Adulto Joven
12.
Toxicol Sci ; 100(1): 146-55, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17698514

RESUMEN

The behavioral and neurophysiological effects of acute exposure to toluene are the most thoroughly explored of all the hydrocarbon solvents. Behavioral effects have been experimentally studied in humans and other species, for example, rats. The existence of both rat and human dosimetric data offers the opportunity to quantitatively compare the relative sensitivity to acute toluene exposure. The purpose of this study was to fit dose-effect curves to existing data and to estimate the dose-equivalence equation (DEE) between rats and humans. The DEE gives the doses that produce the same magnitude of effect in the two species. Doses were brain concentrations of toluene estimated from physiologically based pharmacokinetic models. Human experiments measuring toluene effects on choice reaction time (CRT) were meta-analyzed. Rat studies employed various dependent variables: amplitude of visual-evoked potentials (VEPs), signal detection (SIGDET) accuracy (ACCU) and reaction time (RT), and escape-avoidance (ES-AV) behaviors. Comparison of dose-effect functions showed that human and rat sensitivity was practically the same for those two task regimens that exerted the least control over the behaviors being measured (VEP in rats and CRT in humans) and the sensitivity was progressively lower for SIGDET RT, SIGDET ACCU, and ES-AV behaviors in rats. These results suggested that the sensitivity to impairment by toluene depends on the strength of control over the measured behavior rather than on the species being tested. This interpretation suggests that (1) sensitivity to toluene would be equivalent in humans and rats if both species performed behaviors that were controlled to the same extent, (2) the most sensitive tests of neurobehavioral effects would be those in which least control is exerted on the behavior being measured, and (3) effects of toluene in humans may be estimated using the DEEs from rat studies despite differences in the amount of control exerted by the experimental regimen or differences in the behaviors under investigation.


Asunto(s)
Conducta Animal/efectos de los fármacos , Conducta de Elección/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Solventes/toxicidad , Tolueno/toxicidad , Enfermedad Aguda , Animales , Reacción de Prevención/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Reacción de Fuga/efectos de los fármacos , Potenciales Evocados Visuales/efectos de los fármacos , Humanos , Modelos Biológicos , Ratas , Tiempo de Reacción/efectos de los fármacos , Medición de Riesgo , Detección de Señal Psicológica/efectos de los fármacos , Solventes/farmacocinética , Especificidad de la Especie , Tolueno/farmacocinética
13.
Toxicol Sci ; 99(1): 181-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17548890

RESUMEN

Knowledge of the appropriate metric of dose for a toxic chemical facilitates quantitative extrapolation of toxicity observed in the laboratory to the risk of adverse effects in the human population. Here, we utilize a physiologically based toxicokinetic (PBTK) model for toluene, a common volatile organic compound (VOC), to illustrate that its acute behavioral effects in rats can be quantitatively predicted on the basis of its concentration in the brain. Rats previously trained to perform a visual signal detection task for food reward performed the task while inhaling toluene (0, 1200, 1600, 2000, and 2400 ppm in different test sessions). Accuracy and speed of responding were both decreased by toluene; the magnitude of these effects increased with increasing concentration of the vapor and with increasing duration of exposure. Converting the exposure conditions to brain toluene concentration using the PBTK model yielded a family of overlapping curves for each end point, illustrating that the effects of toluene can be described quantitatively by its internal dose at the time of behavioral assessment. No other dose metric, including inhaled toluene concentration, duration of exposure, the area under the curve of either exposure (ppm h), or modeled brain toluene concentration (mg-h/kg), provided unambiguous predictions of effect. Thus, the acute behavioral effects of toluene (and of other VOCs with a similar mode of action) can be predicted for complex exposure scenarios by simulations that estimate the concentration of the VOC in the brain from the exposure scenario.


Asunto(s)
Conducta Animal/efectos de los fármacos , Exposición por Inhalación/efectos adversos , Solventes/toxicidad , Tolueno/toxicidad , Administración por Inhalación , Animales , Área Bajo la Curva , Atención/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Aprendizaje/efectos de los fármacos , Masculino , Modelos Biológicos , Ratas , Ratas Long-Evans , Tiempo de Reacción/efectos de los fármacos , Detección de Señal Psicológica/efectos de los fármacos , Solventes/farmacocinética , Tolueno/farmacocinética
14.
Toxicol Sci ; 99(2): 572-81, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17623699

RESUMEN

Acute exposure to toluene was assessed in two experiments to determine the relationship between brain toluene concentration and changes in neurophysiological function. The concentration of toluene in brain tissue at the time of assessment was estimated using a physiologically based pharmacokinetic model. Brain neurophysiological function was measured using pattern-elicited visual evoked potentials (VEP) recorded from electrodes located over visual cortex of adult male Long-Evans rats. In the first experiment, VEPs were recorded before and during exposure to control air or toluene at 1000 ppm for 4 h, 2000 ppm for 2 h, 3000 ppm for 1.3 h, or 4000 ppm for 1 h. In the second experiment, VEPs were recorded during and after exposure to clean air or 3000 or 4000 ppm toluene. In both experiments, the response amplitude of the major spectral component of the VEP (F2 at twice the stimulus rate in steady-state responses) was reduced by toluene. A logistic function was fit to baseline-adjusted F2 amplitudes from the first experiment that described a significant relationship between brain toluene concentration and VEP amplitude deficits. In the second experiment, 3000 ppm caused equivalent VEP deficits during or after exposure as a function of estimated brain concentration, but 4000 ppm showed a rapid partial adaptation to the acute effects of toluene after exposure. In general, however, the neurophysiological deficits caused by acute toluene exposure could be described by estimates of the momentary concentration of toluene in the brain at the time of VEP evaluation.


Asunto(s)
Encéfalo/metabolismo , Potenciales Evocados Visuales/efectos de los fármacos , Tolueno/toxicidad , Animales , Masculino , Modelos Biológicos , Ratas , Ratas Long-Evans , Tolueno/farmacocinética , Tricloroetileno/toxicidad
15.
Toxicol Sci ; 160(1): 15-29, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28973534

RESUMEN

Current strategies for predicting carcinogenic mode of action for nongenotoxic chemicals are based on identification of early key events in toxicity pathways. The goal of this study was to evaluate short-term key event indicators resulting from exposure to androstenedione (A4), an androgen receptor agonist and known liver carcinogen in mice. Liver cancer is more prevalent in men compared with women, but androgen-related pathways underlying this sex difference have not been clearly identified. Short-term hepatic effects of A4 were compared with reference agonists of the estrogen receptor (ethinyl estradiol, EE) and glucocorticoid receptor (prednisone, PRED). Male B6C3F1 mice were exposed for 7 or 28 days to A4, EE, or PRED. EE increased and PRED suppressed hepatocyte proliferation, while A4 had no detectable effects. In a microarray analysis, EE and PRED altered >3000 and >670 genes, respectively, in a dose-dependent manner, whereas A4 did not significantly alter any genes. Gene expression was subsequently examined in archival liver samples from male and female B6C3F1 mice exposed to A4 for 90 days. A4 altered more genes in females than males and did not alter expression of genes linked to activation of the mitogenic xenobiotic receptors AhR, CAR, and PPARα in either sex. A gene expression biomarker was used to show that in female mice, the high dose of A4 activated the growth hormone-regulated transcription factor STAT5b, which controls sexually dimorphic gene expression in the liver. These findings suggest that A4 induces subtle age-related effects on STAT5b signaling that may contribute to the higher risk of liver cancer in males compared with females.


Asunto(s)
Androstenodiona/toxicidad , Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/química , Transformación Celular Neoplásica/genética , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/genética , Hígado/efectos de los fármacos , Animales , Biomarcadores de Tumor/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Relación Dosis-Respuesta a Droga , Etinilestradiol/toxicidad , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Fenotipo , Prednisona/toxicidad , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Factores Sexuales , Factores de Tiempo , Transcriptoma
16.
Toxicol Mech Methods ; 16(1): 27-36, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-20021038

RESUMEN

Realistic simulation of environmental exposure scenarios requires dynamic methods in which exposures and human activities vary continuously as a function of time. Simulation of such complex scenarios is, with conventional physiologically based methods, a complex and programming-intensive task. The goal of the present effort was to simplify this task by combining a commercially available general whole-body human physiological model (QCP2004) with a slightly extended physiologically based toxicokinetic (PBTK) model from the literature. The QCP2004 model is a differential equation-based model similar to PBTK models except that normal organ function is simulated and the body organs are appropriately interlinked. Here QCP2004 provided estimates of physiological parameters required by the PBTK model. These were updated as the model was iteratively executed appropriate to the varying activity of the human subject. The combined general physiological model and the PBTK model was called a general physiological and toxicokinetic (GPAT) model. The GPAT model was tested and (within the constraints of available toluene exposure experiments in the literature) found to predict toluene blood concentrations, even in dynamic situations. A model of the structure used in the present work is capable of expansion as new knowledge is developed and greater detail is desired. Similarly, multiple toxicant PBTK models can be developed and incorporated for applications to mixtures risk assessment. Additionally, toxicant effects on organ systems can be achieved by altering organ function during a simulation as a function of the internal dose of toxicants. By cumulatively adding detail to the model as new physiological and chemical-specific information becomes available, the model can become a repository of knowledge for increasingly sophisticated risk-assessment applications.

18.
Toxicol Sci ; 149(2): 312-25, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26519955

RESUMEN

Current strategies for predicting adverse health outcomes of environmental chemicals are centered on early key events in toxicity pathways. However, quantitative relationships between early molecular changes in a given pathway and later health effects are often poorly defined. The goal of this study was to evaluate short-term key event indicators using qualitative and quantitative methods in an established pathway of mouse liver tumorigenesis mediated by peroxisome proliferator-activated receptor alpha (PPARα). Male B6C3F1 mice were exposed for 7 days to di (2-ethylhexyl) phthalate (DEHP), di-n-octyl phthalate (DNOP), and n-butyl benzyl phthalate (BBP), which vary in PPARα activity and liver tumorigenicity. Each phthalate increased expression of select PPARα target genes at 7 days, while only DEHP significantly increased liver cell proliferation labeling index (LI). Transcriptional benchmark dose (BMDT) estimates for dose-related genomic markers stratified phthalates according to hypothetical tumorigenic potencies, unlike BMDs for non-genomic endpoints (relative liver weights or proliferation). The 7-day BMDT values for Acot1 as a surrogate measure for PPARα activation were 29, 370, and 676 mg/kg/day for DEHP, DNOP, and BBP, respectively, distinguishing DEHP (liver tumor BMD of 35 mg/kg/day) from non-tumorigenic DNOP and BBP. Effect thresholds were generated using linear regression of DEHP effects at 7 days and 2-year tumor incidence values to anchor early response molecular indicators and a later phenotypic outcome. Thresholds varied widely by marker, from 2-fold (Pdk4 and proliferation LI) to 30-fold (Acot1) induction to reach hypothetical tumorigenic expression levels. These findings highlight key issues in defining thresholds for biological adversity based on molecular changes.


Asunto(s)
Neoplasias Hepáticas Experimentales/inducido químicamente , PPAR alfa/fisiología , Animales , Benchmarking , Peso Corporal/efectos de los fármacos , Proliferación Celular , Dietilhexil Ftalato/toxicidad , Relación Dosis-Respuesta a Droga , Modelos Lineales , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Estrés Oxidativo , Ácidos Ftálicos/toxicidad , Reacción en Cadena de la Polimerasa
19.
Toxicology ; 206(3): 389-401, 2005 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-15588929

RESUMEN

Heme oxygenase (HO) is the rate-limiting enzyme in heme degradation and its activity has a significant impact on intracellular heme pools. Rat studies indicate that HO induction is a sensitive, dose-dependent response to arsenite (As(III)) exposure in both liver and kidney. The objective of this study was to evaluate the relationship of HO induction to administered As(III) dose, and concentrations of inorganic arsenic (iAs) in tissues and urine. Levels of iAs, mono- (MMA) and dimethylated arsenic (DMA) as well as HO activity were determined in liver, lung and kidney over time in female B6C3F1 mice given a single oral dose of 0, 1, 10, 30 or 100 micromol/kg As(III). Increased HO activity was a time and dose-dependent response in liver and kidney, but not in lung. Activity peaked in the 4-6 h time range in liver and kidney with the responsiveness in liver being approximately 2- to 3-fold greater than kidney. The lowest observed effect levels (LOELs) in this study for HO induction are 30 and 100 micromol/kg, respectively, in liver and kidney. The predominant form of arsenic (As) was iAs in liver at all doses, whereas DMA was the predominant form of As in kidney at all doses. Three- to four-fold higher levels of iAs were achieved in liver compared to kidney. MMA was the least abundant form of As in liver and kidney, never exceeding more than 20% of the total As present. The concentration of iAs in tissue or urine demonstrated the strongest correlation with HO activity in both liver and kidney. Results of this study suggest that HO induction is a biomarker of effect that is specific for tissue iAs because a high, but nontoxic, acute dose of DMA (5220 micromol/kg) did not induce HO in mice. Thus, HO induction has potential for use as a biomarker of effect for inorganic arsenic exposure and may be used as an indicator response to further the development of a biologically-based dose response model for As.


Asunto(s)
Arsenitos/farmacología , Hemo Oxigenasa (Desciclizante)/biosíntesis , Animales , Arsenicales/farmacocinética , Arsenicales/farmacología , Arsenitos/sangre , Arsenitos/farmacocinética , Arsenitos/orina , Ácido Cacodílico/farmacocinética , Ácido Cacodílico/farmacología , Cruzamientos Genéticos , Relación Dosis-Respuesta a Droga , Inducción Enzimática/efectos de los fármacos , Femenino , Riñón/efectos de los fármacos , Riñón/enzimología , Hígado/efectos de los fármacos , Hígado/enzimología , Pulmón/efectos de los fármacos , Pulmón/enzimología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL
20.
Toxicol Sci ; 79(1): 28-37, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14976335

RESUMEN

beta-Chloroprene (2-chloro-1,3-butadiene; CD), which is used in the synthesis of polychloroprene, caused significant incidences of several tumor types in B6C3F1 mice and Fischer rats, but not in Wistar rats or Syrian hamsters. This project investigates the relevance of the bioassay lung tumor findings to human health risk by developing a physiologically based toxicokinetic (PBTK) model and exploring a tissue specific exposure-dose-response relationship. Key steps included identification of the plausible genotoxic mode of action, experimental quantification of tissue-to-air partition coefficients, scaling of in vitro parameters of CD metabolism for input into the PBTK model, comparing the model with in vivo experimental gas uptake data, selecting an appropriate tissue dosimetric, and predicting a corresponding human exposure concentration. The total daily milligram amount of CD metabolized per gram of lung was compared with the animal bioassay response data, specifically combined bronchiolar adenoma/carcinoma. The faster rate of metabolism in mouse lung agreed with the markedly greater incidence of lung tumors compared with the other rodent species. A lung tissue dose was predicted for the combined rodent lung tumor bioassay data at a 10% benchmark response. A human version of the PBTK model predicted that the lung tissue dose in humans would be equivalent to continuous lifetime daily exposure of 23 ppm CD. PBTK model sensitivity analysis indicated greater dependence of model predictions of dosimetry on physiological than biochemical parameters. The combined analysis of lung tumor response across species using the PBTK-derived internal dose provides an improved alternative to default pharmacokinetic interspecies adjustments for application to human health risk assessment.


Asunto(s)
Cloropreno/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Administración por Inhalación , Animales , Cámaras de Exposición Atmosférica , Cloropreno/química , Cloropreno/farmacología , Cricetinae , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Predicción , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Cinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Mesocricetus , Ratones , Ratones Endogámicos , Modelos Biológicos , Músculos/efectos de los fármacos , Músculos/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Wistar , Distribución Tisular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA